Metabolomics and Its Impact On Health and Diseases
Metabolomics and Its Impact On Health and Diseases
Veronica Ghini
Kathleen A. Stringer
Claudio Luchinat Editors
Metabolomics
and Its Impact
on Health and
Diseases
Handbook of Experimental Pharmacology
Volume 277
Editor-in-Chief
Martin C. Michel, Dept of Pharmacology, Johannes Gutenberg Universität, Mainz,
Germany
Claudio Luchinat
Department of Chemistry
University of Florence
Sesto Fiorentino, Florence, Italy
# The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature Switzerland
AG 2023
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether
the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and
transmission or information storage and retrieval, electronic adaptation, computer software, or by
similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication
does not imply, even in the absence of a specific statement, that such names are exempt from the relevant
protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in this book
are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the
editors give a warranty, expressed or implied, with respect to the material contained herein or for any
errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
This Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Preface
v
vi Preface
of this remarkable series, and to the past and current editorial board members who
have dedicated time and effort into establishing this series as one of the most
recognized publications in pharmacology.
Contents
1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2 A Short History of NMR-Based Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3 Practical Aspects of NMR-Based Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.1 Sample Preparation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.2 Choosing the Right Pulse Sequence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
3.3 Spectral Acquisition and Processing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4 Data Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
4.1 Data Analysis for Targeted Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
4.2 Data Analysis for Untargeted Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
4.3 Targeted or Untargeted? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
5 Biological Interpretation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
6 Conclusion and Future Prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
D. S. Wishart (✉)
Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
Department of Computing Science, University of Alberta, Edmonton, AB, Canada
Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
e-mail: [email protected]
M. Rout · B. L. Lee · M. Berjanskii · M. LeVatte
Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
e-mail: [email protected]; [email protected]; [email protected]; [email protected]
M. Lipfert
Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
Reference Standard Management & NMR QC, Lonza Group AG, Visp, Switzerland
e-mail: [email protected]
Abstract
While NMR-based metabolomics is only about 20 years old, NMR has been a key
part of metabolic and metabolism studies for >40 years. Historically, metabolic
researchers used NMR because of its high level of reproducibility, superb instru-
ment stability, facile sample preparation protocols, inherently quantitative char-
acter, non-destructive nature, and amenability to automation. In this chapter, we
provide a short history of NMR-based metabolomics. We then provide a detailed
description of some of the practical aspects of performing NMR-based
metabolomics studies including sample preparation, pulse sequence selection,
and spectral acquisition and processing. The two different approaches to
metabolomics data analysis, targeted vs. untargeted, are briefly outlined. We
also describe several software packages to help users process NMR spectra
obtained via these two different approaches. We then give several examples of
useful or interesting applications of NMR-based metabolomics, ranging from
applications to drug toxicology, to identifying inborn errors of metabolism to
analyzing the contents of biofluids from dairy cattle. Throughout this chapter, we
will highlight the strengths and limitations of NMR-based metabolomics. Addi-
tionally, we will conclude with descriptions of recent advances in NMR hard-
ware, methodology, and software and speculate about where NMR-based
metabolomics is going in the next 5–10 years.
Keywords
Applications · Experimental methods · NMR spectroscopy · Targeted
metabolomics · Untargeted metabolomics
1 Introduction
are encoded for and controlled by genes. Therefore, metabolites are exquisitely
sensitive indicators of problems in the genome. Indeed, a single base change in a
gene can lead to a 10,000-fold change in the concentrations of certain metabolites
(Wishart et al. 2007). This remarkable sensitivity of metabolite levels to genetic
variations led to one of the most common and widespread medical testing
initiatives – newborn screening. For more than 100 years, metabolite testing has
been used to identify and detect genetic diseases and inborn errors of metabolism or
IEMs, such as phenylketonuria or alkaptonuria (Levy 2010). Metabolites are not
only sensitive to genome-related processes, but also to what happens in the environ-
ment. In particular, metabolite concentrations are influenced by nutrition, exposure
to workplace or household chemicals, physical activity, the time of day, or even the
outside temperature (Bassini and Cameron 2014; Brown 2016).
Because metabolites are affected by what happens intra-cellularly (via the
genome) and extra-cellularly (via the environment), metabolomics provides a
detailed view of the gene–environment interactions. Metabolomics is therefore an
ideal route for scientists to access and measure an organism’s “chemical phenotype”
(Fiehn 2002). This represents an important advantage of metabolomics over geno-
mics analyses. While the genome can suggest what might happen, the metabolome
actually indicates what is happening.
Continued advances in analytical chemistry and computational data analysis have
made the study of metabolomics more accessible to a wider range of scientific
disciplines. These advances have led to metabolomics being routinely used in
disease screening, drug discovery, food and nutritional analysis, veterinary studies,
crop assessment, biomaterial production, and environmental monitoring (Holmes
et al. 2008; Viant 2008; Wishart 2008a, 2016; Kim et al. 2016). Indeed,
metabolomics research has grown exponentially since 1999 which reported just
two metabolomics papers to nearly 9,000 papers published in 2020.
Metabolomics experiments are relatively simple to perform. The general
workflow to collect metabolomics data is shown in Fig. 1. The experiments begin
with a biological sample which can be a biofluid or a tissue. For tissues, the
metabolites must first be extracted or homogenized to produce a fluid. Once an
appropriate metabolite extract or biofluid has been obtained, the liquid sample must
be analyzed by one or more analytical chemistry platforms. The most popular
platforms are liquid chromatography mass spectrometry (LC-MS) and nuclear
magnetic resonance (NMR) spectroscopy. These analytical platforms are ultimately
responsible for helping to identify and/or quantify the chemicals in the different
biological mixtures. With the help of specialized software and carefully developed
databases of compounds, the data generated from these platforms can be used to
identify hundreds of compounds in the biological samples.
While LC-MS methods account for >70% of published metabolomics studies to
date, NMR-based methods still garner considerable interest among metabolomics
researchers. For instance, more than 1,200 NMR-based metabolomics papers were
published in 2020, the most ever published in any given year. This suggests that
NMR-based metabolomics is growing, and it still has plenty to offer to the
metabolomics community. NMR has some unique advantages compared to other
4 D. S. Wishart et al.
Fig. 1 A simplified workflow for metabolomics. Tissue samples may be obtained and
homogenized, or biofluids such as urine or blood may be collected. Spectra of the liquid portion
of the samples can be acquired using NMR or mass spectrometry, and the resulting data used for
analysis
help users process NMR spectra obtained via these two different approaches. We
also illustrate several examples of where and how NMR-based metabolomics has
been successfully implemented. Finally, we speculate on the future of NMR-based
metabolomics and the potential areas of growth for this field.
For the past 40 years, NMR has played a central role in the understanding of
metabolism and metabolomic processes. The first example demonstrating how
NMR could be used in metabolic studies was published in 1974 when a 13C
isotope-tracer analysis was combined with NMR studies to decipher specific details
of ethanol metabolism (Wilson and Burlingame 1974). Since then, steady
improvements in NMR technology, NMR field strength, and other advancements
have increased the popularity of NMR for many applications in biochemistry and
metabolism. Indeed, NMR quickly became the tool of choice for many metabolism
research areas, particularly those studying drug metabolism. During the 1970s and
1980s, NMR spectroscopy was used in studies to explore drug kinetics, drug
metabolism, and the identification of drug metabolites (Midgley and Hawkins
1978; Williams et al. 1979). These studies were complemented by more traditional
metabolic studies, focusing on cellular, microbial, plant, and animal metabolism
using in vivo or in vitro 1H, 13C, and 31P NMR techniques (Cohen et al. 1979;
Weiner et al. 1989; Rothman et al. 2003). The widespread use of NMR for “classi-
cal” metabolic studies combined with its exceptional capacity to handle complex
metabolomic mixtures made NMR the preferred analytical chemistry platform for
launching the new field of metabolomics. Indeed, the very first metabolomics
(or metabonomics) studies were conducted using NMR (Bock 1982; Yoshikawa
et al. 1982; Bales et al. 1984a, b). By the late 1980s, NMR-based metabolomics
studies of human plasma led to the identification of several putative biomarkers for
cancer and coronary artery diseases (Fossel et al. 1986; Otvos et al. 1991). These
studies were complemented by pioneering studies by Nicholson and colleagues who
used NMR spectra from urine to characterize inborn errors of metabolism (IEM) and
drug toxicity. These early studies proved that urine, a very complex biofluid, can be
successfully analyzed by NMR (Bales et al. 1984a, b; Nicholson et al. 1984a, b).
The greatest challenge facing these early metabolomics researchers was the sheer
complexity of the NMR spectra they were collecting from biofluids and tissue
extracts. A metabolically rich biofluid such as urine can contain up to 5,000
detectable proton resonances in countless variations depending on the pH or
concentrations of salts in the sample (Nicholson and Wilson 2003). This complexity
of the spectral output led researchers to develop two different approaches for
collecting, processing, analyzing, and interpreting metabolomics NMR data. One
approach called “targeted metabolomics” uses spectral deconvolution software to
identify and quantify fluid-specific or known metabolites in individual NMR spectra.
The second approach called “untargeted metabolomics,” or statistical spectroscopy,
uses spectral alignment, spectral binning, and multivariate statistical analysis to
6 D. S. Wishart et al.
identify spectral features of interest. Once the key features are identified, the
corresponding compounds and metabolites may or may not be identified. Both
targeted and untargeted approaches have their advantages and disadvantages.
While targeted metabolomics is more precise, highly quantitative, and much more
reproducible, it is more time-consuming, more limited in scope and it does not allow
one to identify novel chemicals. Untargeted metabolomics is faster, relatively open
ended, and more useful for identification of novel compounds but it is not quantita-
tive nor is it particularly reproducible. Given the importance of targeted and
untargeted approaches in NMR-based metabolomics and given their requirements
for different types of data analysis techniques, we will discuss them in more detail in
Sect. 4.
This section will provide an overview of the practical aspects and consensus
recommendations for conducting liquid-state NMR metabolomics studies with a
primary focus on 1H NMR of biofluids or fluidized tissue extracts (McKay,
“Metabolomics using NMR – avoiding the black box”; Raftery, “Quantitative
NMR methods in metabolomics”). Readers interested in obtaining precise protocols
or information about solid-state NMR or magic angle sample spinning (MAS) NMR
should refer to other excellent reviews and book chapters (Weber et al. 2012; Wolak
et al. 2012; Nagana Gowda and Raftery 2014; Nagana Gowda et al. 2015; Zhang
et al. 2016; Mazzei and Piccolo 2017; Tilgner et al. 2019).
are extracted with cold methanol or cold methanol/water or with chloroform (Wolak
et al. 2012; Nagana Gowda and Raftery 2014). The use of an organic solvent, in
addition to extracting the metabolites of interest, quenches unwanted metabolism by
denaturing and precipitating almost all proteins/enzymes within the sample. The
organic solvent extract is then centrifuged to separate the precipitated proteins from
the sample. The sample then must be dried (by freeze-drying/lyophilization) and
then re-dissolved in water or appropriate NMR solvents prior to NMR analysis.
For biofluids, the extraction protocols are much simpler. Cell-free biofluids such
as serum, plasma, saliva, growth media, cell extracts, plant sap, or fecal water
typically require ultrafiltration through a 3–5 kDa molecular weight cut-off filter
that removes higher molecular weight proteins and enzymes (Psychogios et al. 2011;
Nagana Gowda and Raftery 2014). Since no organic solvent is used, no lyophiliza-
tion or evaporation is required, and volatile metabolites, such as formate, acetate,
methanol and ethanol, can be easily detected by NMR. However, ultrafiltration can
cause chemical contamination as the filters contain glycerol or other humectants as
preservatives. These filters must be washed multiple times to remove any traces of
these agents. Ultrafiltration may also artificially lower the concentrations of some
metabolites, such as benzoic acid or tryptophan, which appear to bind to the filter or
are bound to proteins that are removed by the filter (Psychogios et al. 2011; Nagana
Gowda and Raftery 2014). Urine and cerebrospinal fluid samples (as well as other
fluids, such as juice, wine, or beer, that are largely sterile and protein-free) are often
simply filter-sterilized by passing the fluid through a 0.22 μm filter to remove any
cells or organic debris. Often researchers also add a small amount of sodium azide
(an NMR-invisible salt) to the sample to destroy any residual microbial activity.
An alternative method to ultrafiltration or filter-sterilization is organic solvent
extraction. In general, methanol or acetonitrile (for hydrophilic compounds) and
chloroform or methyl-tert-butyl ether (for hydrophobic compounds) can be added to
a sample and used to simultaneously extract metabolites, sterilize the sample, and
precipitate proteins. This method may be applied to serum, plasma, or urine
(Beckonert et al. 2007; Nagana Gowda and Raftery 2014). Solvent extraction may
also enrich for a particular class of chemical compounds (hydrophobic or hydro-
philic compounds) depending on the choice of solvent and/or extraction protocol. As
a general rule, methanol extraction works best for most NMR samples (Lin et al.
2007). However, as organic solvent extraction involves an evaporation step to
remove the organic solvent (which is time consuming), volatile metabolites such
as formic acid, ethanol, or acetic acid may be volatilized and no longer detectable.
Another method to enrich or concentrate particular classes of metabolites uses
stable isotope chemical derivatization. Although more commonly used in MS-based
metabolomics (Gowda et al. 2010), this technique is less widely utilized in
NMR-based metabolomics. Stable isotope labeling can enhance the sensitivity and
resolution by enabling heteronuclear NMR spectroscopy. It can also help to enrich
certain classes of metabolites that contain a specific reactive chemical group (i.e.,
amines or carboxylate groups). Two isotopic tags have gained some popularity in the
NMR community, 15N-ethanolamine and 15N-cholamine (see Fig. 2). Both react
selectively with carboxyl groups (Ye et al. 2009; Tayyari et al. 2013). These isotope
8 D. S. Wishart et al.
Fig. 2 The chemical structures of 15N-labeled ethanolamine and cholamine (top). The amine group
can react with carboxylic acid groups on metabolites, using DMT-MM (4-(4,6-dimethoxy[1,3,5]
triazin-2-yl)-4-methylmorpholinium-chloride) as a catalyst (bottom). 2D 15N-1H HSQC spectra can
then be acquired to detect the tagged metabolites
tags can be used to enhance the detection of organic acids and amino acids and can
also be used in MS-based metabolomics studies.
After the extraction and/or enrichment step is complete, it is critical to use the
right buffer system to adjust the salt concentrations and pH of the sample. For
untargeted approaches, sample uniformity is absolutely required. Ideally all samples
should have identical pH values and identical salt concentrations to ensure uniform
chemical shifts among all metabolites. This uniformity greatly enhances the spectral
alignment. For targeted metabolomics studies, pH and salt concentrations are not as
critical. Nonetheless, buffering the sample with a 50–150 mM potassium phosphate
buffer, maintaining the sample temperature at a constant value (say 25°C), adding a
small amount of D2O as a lock solvent, and ensuring the pH is near 7.0 are all
recommended for targeted profiling with software tools such as Bayesil or Chenomx
(to be discussed later) which have well-defined sample collection conditions
(Mercier et al. 2011; Ravanbakhsh et al. 2015).
Sample preparation for NMR analysis always requires the addition of a chemical
shift reference compound. The International Union of Pure and Applied Chemistry
(IUPAC) and the International Union of Biochemistry and Molecular Biology
(IUBMB) recommend the use of 4,4-dimethyl-4-silapentane-1-sulfonic acid (DSS)
as an internal chemical shift standard for aqueous samples (Wishart et al. 1995;
Harris et al. 2002). Usually DSS is used in its deuterated form DSS-d6. As the DSS
peak at 0.00 ppm is usually well resolved and easily detected, it may also be used as
an internal quantification reference. Trimethylsilylpropanoic acid (TSP) is also
commonly used as a chemical shift standard but is not recommended for
NMR-based metabolomics applications as the chemical shift varies substantially
with pH. Both DSS and TSP can bind to macromolecules such as lipids or proteins
(found in unfiltered serum or plasma), resulting in a broadened signal if these
molecules are present in large quantities. If this occurs, the signal cannot be used
for quantification. An alternative to the internal standard is an electronic reference
signal, often called ERETIC (Electronic REference To access In vivo
Concentrations) (Akoka et al. 1999). The ERETIC method is particularly appealing
since the signal is electronically generated and can be placed in any position in the
Practical Aspects of NMR-Based Metabolomics 9
Once the sample is loaded into the NMR spectrometer, the NMR spectrum
(or spectra) can be acquired. Most NMR-based metabolomics studies use 1D 1H
NMR because of its speed and simplicity with which spectra can be acquired,
processed, and interpreted. Two types of pulse sequences or experiments are used
for 1D NMR-based metabolomics: the metnoesy or 1D NOESY (Nuclear
Overhauser Effect Spectroscopy) experiment and the Carr-Purcell-Meiboom-Gill
(CPMG) experiment. The metnoesy experiment is a simple 1D NOESY pulse
sequence that provides solvent suppression before the experiment and during the
mixing time without the use of gradients (see Fig. 3). This simple pulse sequence can
be used with almost any NMR probe (Mckay 2011). The metnoesy pulse sequence
can be modified with more advanced water suppression techniques that use gradients
(i.e., watergate, excitation sculpting) for more robust and effective solvent removal.
With such modifications, more dilute samples can be recorded in less time (McKay
2009). Another advantage of the metnoesy sequence is that shaped pulses can be
used to suppress not only water but additional other strong signals such as ethanol in
wine and beer samples or organic solvents used in the extraction process. As most
databases and deconvolution programs for NMR-based metabolomics were built
using this simplistic metnoesy pulse sequence, this pulse sequence predominates
most metabolomics studies with 1D 1H-NMR.
The second most commonly used 1D NMR pulse sequence is the CPMG experi-
ment (Fig. 4). This pulse sequence can spectroscopically remove signals of large
molecules, such as proteins or lipoproteins, from the spectrum without the need for
ultrafiltration or solvent extraction (Beckonert et al. 2007). The CPMG experiment
takes advantage of the fact that small molecule metabolites and macromolecules
(such as proteins) have different T2 relaxation times. The T2 relaxation time of
macromolecules is very short (milliseconds) while the T2 of metabolites is longer
(seconds). With the CPMG sequence, all molecules with a short T2 are suppressed,
whereas those with a large T2 are unaffected. Under ideal conditions, the CPMG
pulse sequence would eliminate the need for solvent extraction and ultrafiltration,
reducing time and resources required for sample preparation. However, the CPMG
Fig. 3 The pulse sequence for the metnoesy or 1D NOESY experiment. This pulse sequence has a
delay (D1) with a low power presaturation pulse, followed by two successive 90° pulses followed
by a defined mixing time. The pulse sequence concludes with a final 90° pulse followed by the
acquisition period
Practical Aspects of NMR-Based Metabolomics 11
Fig. 4 The pulse sequence for the Carr-Purcell-Meiboom-Gill (CPMG) experiment. This experi-
ment uses repeated pulses of 180° to select signals with long T2 relaxation times and remove signals
with short T2 times
experiment is not perfect and all protein signals are not suppressed, increasing the
time required to properly phase, process, and compare multiple CPMG spectra. As
very few CPMG reference spectra are available in the NMR databases, it makes the
CMPG pulse sequence very difficult to use for spectral deconvolution in targeted
NMR-based metabolomics studies.
More recent developments in NMR-based metabolomics use “pure-shift” pulse
sequences (Moutzouri et al. 2017; Lopez et al. 2019) to simplify overcrowded 1D
1
H-NMR spectra. Pure-shift NMR aims to convert all signals into singlets by
refocusing homonuclear couplings and collapsing multiplet peaks into singlets,
thereby reducing the overlap between compound peaks. However, this class of
experiments are much less sensitive than standard 1H experiments due to the use
of spatial or frequency selective techniques. They also contain artifacts that result
from the pseudo-2D method of data acquisition. The reduced sensitivity, the
increased complexity, and the greater hardware requirements over more conven-
tional 1D NMR experiments have likely prevented their widespread use in
metabolomics. Despite these drawbacks, the improved resolution provided by
these methods has seen their successful application in metabolite profiling (Lopez
et al. 2019).
1D NMR pulse sequences are not the only experiments available to metabolomics
researchers. With 2D NMR, multidimensional data can be recorded from the same
type of nuclei (homonuclear) or different nuclei (heteronuclear). For metabolically
complex samples such as urine with hundreds of different and variable metabolites,
it can be advantageous to use 2D NMR experiments. Indeed homonuclear 2D
experiments, such as 2D 1H COSY (COrrelated SpectroscopY), 2D 1H INADE-
QUATE (Incredible Natural Abundance DoublE QUAntum Transfer Experiment),
or 2D J-resolved experiments have been widely used in NMR-based metabolomics
studies (Martineau et al. 2011; Bingol and Brüschweiler 2014). 2D NMR spectra
allow researchers to more easily identify unknown compounds, characterize novel
compounds, and deconvolute overlapping peaks, which could be problematic for 1D
spectral deconvolution programs. Several tools and databases are available to inter-
pret 2D homonuclear metabolomics data (Bingol et al. 2014, 2016). For complex
mixtures such as those found in metabolomics studies, the 2D J-resolved (JRES)
experiment is particularly attractive due to its simplicity and relatively short
12 D. S. Wishart et al.
acquisition time relative to other 2D experiments (Ludwig and Viant 2010). The
JRES experiment results in singlets in the 1H dimension of the spectrum, effectively
providing a robust, broadband decoupled 1H spectrum, similar to the “pure-shift”
experiments mentioned above. This provides a way to resolve the complex
overlapping signals from a 1D 1H spectrum. The multiplet patterns, however, are
retained in the second dimension of the JRES spectrum, which can further aid in the
identification of the metabolites. The JRES experiment can also be used for metabo-
lite quantification. Further enhancement of metabolite identification can be achieved
using heteronuclear 2D experiments, such as 2D 1H-15N HSQC (Heteronuclear
Single Quantum Coherence Spectroscopy) and 1H-13C HSQC experiments. These
experiments provide additional chemical shift information in the second dimension
(15N/13C) and offer important structural detail and connectivity information (Lewis
et al. 2007; Ye et al. 2009). Similar to the homonuclear 2D metabolomics data,
spectral databases and programs are also available to facilitate the interpretation of
2D heteronuclear metabolomics data (Bingol et al. 2015, 2016).
While 2D NMR experiments offer tremendous advantages, there are at least three
major disadvantages to using 2D or multidimensional NMR for metabolomics. The
first disadvantage is the time required to collect, process, and interpret the data
(hours compared to minutes for the 1D experiments). Second, 2D NMR experiments
are less sensitive, with a lower limit of metabolite detection that is 5-10X higher than
1D NMR (often >50 μM compared to 10 μM). And lastly, obtaining robust absolute
quantification of metabolites from 2D spectra is particularly challenging – although
improvements are being made (Martineau and Giraudeau 2019; Martineau et al.
2020; Hansen et al. 2021).
Since the informational density in 2D NMR experiments is higher than 1D
experiments, there is a considerable interest in developing techniques to reduce the
time required to acquire 2D spectra. A number of different approaches are being
investigated to reduce 2D spectral scanning time (Le Guennec et al. 2014). The first
approach utilizes spectral folding or aliasing to reduce the spectral width. With this
technique, peaks can be folded in empty spaces of the 2D spectrum. The reduction of
the spectral width means less data points are required, shortening the overall
acquisition time. However, additional NMR experiments have to be performed to
determine the true chemical shift of folded peaks (Foroozandeh and Jeannerat 2010).
If the sample pool is the same (such as urine), this only needs to be performed once
with a single representative sample.
A second approach shortens the acquisition time by reducing the delay between
scans or shortening the relaxation time. These experiments are known as band-
selective optimized flip angle short transient (SOFAST) and band-selective excita-
tion short transient (BEST) methods. However, as these experiments rely on spin
diffusion as an effective relaxation mechanism (which is only true for
macromolecules or small molecules in viscous solvents), SOFAST and BEST
experiments can only be used in studies involving living cells, which have a viscous
cytoplasm (Motta et al. 2010).
A third approach is available that dramatically shortens 2D NMR data acquisition
time by combining multiple pulse sequences (such as COSY, NOESY, HSQC) into
Practical Aspects of NMR-Based Metabolomics 13
one supersequence (Kupče and Claridge 2017; Hansen et al. 2021). An example of
such a supersequence is called NOAH (NMR by Ordered Acquisition using 1H
detection). This pulse sequence has enabled 2D data collection in a single measure-
ment and has been used to comprehensively characterize multiple metabolites within
metabolically complex biofluids such as mouse urine.
A fourth method to shorten the experimental time of 2D NMR experiments uses
non-uniform sampling (NUS). With this method, all data points in the indirect
dimension are not recorded. Instead, the recorded data points are randomized and
differentially weighted across the indirect dimension(s). The missing data points are
reconstructed after the data is collected. With the NUS technique, the acquisition
time of 2D and 3D experiments can be reduced by up to 75% while still retaining the
same spectral resolution as a full-time multidimensional experiment. The quality of
the spectrum mainly depends on the algorithm used to reconstruct it (Kazimierczuk
et al. 2010).
The last technique is the ultrafast (UF) 2D NMR. This technique utilizes the
sample height (or length) and generates slices which correlate with different time
points in the second dimension (called spatial encoding). With the UF-NMR tech-
nique, a 2D spectrum can be acquired in a single scan, making it the fastest 2D NMR
technique available. However, using this experiment, a compromise between spec-
tral widths, resolution, and sensitivity is often necessary. Fortunately, the spectral
width limitations of UF-NMR can be addressed by the folding/aliasing method
(mentioned above) and the sensitivity can be improved by increasing the number
of scans (Shrot and Frydman 2009; Tal and Frydman 2010; Pathan et al. 2011).
Compared to 1D NMR, 2D NMR techniques have a number of limitations with
respect to metabolite quantification. Because 2D NMR techniques use more pulses,
they are more sensitive to pulse imperfections. This may result in inconsistencies in
peak intensity and peak volume, limiting the reliability of quantification. Further-
more, for heteronuclear NMR, the 13C or 15N nuclei for most metabolites have a very
broad chemical shift range. An equal excitation of the 13C or 15N complete spectral
region is difficult for higher field (>500 MHz) spectrometers. To achieve equal
excitation, one could use complex pulses or pulse sequences, such as adiabatic
pulses or shaped pulses. However, the use of these pulses leads to inconsistencies
in spectral peak intensities and volumes, which makes quantification challenging. In
addition, highly variable coupling and relaxation times also alter the peak volume in
hard-to-predict ways, thereby limiting the use of 2D techniques for quantitative
analysis. To overcome these limitations, large numbers of 2D spectral calibration
curves must be collected for each type of 2D NMR experiment in order to use them
for accurate metabolite quantification. However, gathering this kind of data is
tedious, time-consuming, and difficult due to the long experimental acquisition
times (Lewis et al. 2007).
In recent years, it has been shown that 13C-HSQC data can be recorded in a way
that is inherently quantitative. This can be done by extrapolation of the signal back to
the initial excitation (time point zero), known as the HSQC0 experiment (which
requires three separate HSQC experiments) or with a quantitative sequence or the
Q-HSQC experiment (which requires four times more scans to achieve the same
14 D. S. Wishart et al.
Fig. 5 A simple illustration of the effects of shimming on an NMR signal on the DSS reference
peak. Poor shimming can cause peaks to appear unsymmetric or even appear like multiple peaks
spectrometers. This ensures that the digital resolution will typically be less than
0.25 Hz in the 1H dimension on most high-resolution instruments (see Eq. 1; sweep
widths (sw) range from 6,000 to 10,000 Hz):
sw
Resolution = 2 × ð1Þ
np
Good digital resolution ensures sharp resonances, but good S/N ensures good
sensitivity. NMR is not known to be a particular sensitive technique. Thus,
metabolomics researchers are always looking for ways to improve NMR sensitivity
to extend the lowest detectable metabolite concentration. Increased sensitivity can be
achieved by increasing the magnetic field strength (the higher the better), using
cryogenically cooled probes (which have 2–4 times better S/N compared to room-
temperature probes via the reduction of electronic noise), concentrating the sample,
increasing sample volume, or optimizing the excitation flip angle. However, one of
the simplest approaches to increasing the S/N and lowering the limit of detection is
increasing the number of scans (ns) (as shown in Eq. 2):
S pffiffiffiffiffi
= ns ð2Þ
N
Using Eq. 2, one can see that increasing the scans does not increase the S/N
linearly. With four scans, the sensitivity (S/N) increases just twofold. With
100 scans, the sensitivity increases only 10-fold. The number of scans collected in
a given NMR experiment must be tempered by the time it takes to collect each of
those scans. That time is determined by the repetition period between scans, also
known as relaxation delay. This delay is defined as the sum of the acquisition time
and acquisition delay prior to the next scan. The relaxation delay should be five times
longer than the longitudinal relaxation time (also known as T1). For metabolites, T1
is typically 2–3 s (Bloembergen et al. 1948). Therefore, a relaxation delay of about
15 s is usually enough time for a complete relaxation of all resonances (and
consequently full recovery of signal intensity) between scans. However, employing
such long delays would make data collection incredibly inefficient and severely limit
the number of scans that could be collected. Therefore, shorter repetition times of
16 D. S. Wishart et al.
2–4 s are often used for the majority of NMR-based metabolomics studies
(Beckonert et al. 2007). These shorter relaxation delays represent a reasonable
compromise between trying to maximize signal recovery and maximizing the
number of scans. However, this compromise means that spectral deconvolution
algorithms used in targeted metabolomics cannot use “idealized” or theoretical
reference NMR spectra but must, instead, use reference spectra that have been
experimentally recorded using exactly the same type of acquisition parameters and
short relaxation delays as used in the actual metabolomics experiment (Mercier et al.
2011; Worley and Powers 2014; Ravanbakhsh et al. 2015).
After an NMR spectrum has been collected, it must be properly phased. Phasing
is an NMR spectral adjustment process that is designed to maximize the absorptive
character of NMR peaks over all regions of an NMR spectrum. There are two types
of phasing: zero-order phase correction (frequency-independent) and first-order
(frequency-dependent) phase correction. While zero-order phase correction is inde-
pendent of the position of the peaks, first-order phasing increases linearly with the
offset from the carrier frequency. Both types of phase correction are usually needed
to obtain symmetric, purely absorptive peaks. Accurate phase correction is an
important step in spectral processing of metabolomics data as even small phase
errors can lead to a cascade of problems in downstream spectral processing and post-
spectral analysis affecting targeted or untargeted metabolomics techniques (Emwas
et al. 2018). Several algorithms have been published for automated phasing of NMR
spectra (Chen et al. 2002; de Brouwer 2009; Binczyk et al. 2015; Zorin et al. 2017;
Steimers et al. 2020). Some of these routines have already been implemented in the
operating systems of many modern NMR spectrometers. However, additional man-
ual phasing is often required in NMR-based metabolomics studies since auto-
phasing routines may have difficulty with more spectrally crowded metabolite
spectra. Furthermore, auto-phasing programs can sometimes end up distorting the
entire NMR spectrum while attempting to correct for the residual water signal.
Despite these caveats, auto-phasing is still widely used in the NMR metabolomics
community because it is fast, reasonably reliable and it avoids operator bias.
Baseline correction is another important step in NMR spectral data processing.
Baseline correction yields a more pleasant looking NMR spectrum where signal-free
regions appear as completely flat lines with zero intensity. While baseline correction
is relatively easy for simpler NMR spectra with just a few peaks, it is much more
difficult for NMR spectra containing thousands of peaks with large differences in
peak intensities and peak widths. High-quality baseline correction is critical for
proper spectral alignment (in untargeted metabolomics) and proper quantification
or peak integration (in targeted metabolomics). Like phase correction, small errors in
the baseline correction can lead to significant errors in the quantification of low
abundance metabolites.
There are two general approaches to baseline correction: one involves correction
in the time domain and the other involves correction in the frequency domain.
Baseline correction through the time domain removes corrupted data in the free
induction decay (FID) to decrease the effect of low frequencies. This can be done by
discarding some of the initial data points and recreating them by a technique called
Practical Aspects of NMR-Based Metabolomics 17
Fig. 6 An example of how good shimming, water suppression, phasing, baseline correction, and
chemical shift referencing can make a significant difference to the quality and usability of a 1D
NMR spectrum of a biofluid. The same sample and experimental parameters were used for both
spectra, except shimming and water suppression were adjusted away from optimal for the left
spectrum. The receiver gain was adjusted automatically
back-prediction (Heuer and Haeberlen 1989; Halamek et al. 1994). Baseline correc-
tion in the frequency domain involves selecting the valley or signal-absent regions of
the spectra (either automatically or manually) and fitting these regions with a
polynomial spline function. The corresponding baseline offset values based on this
spline function are then subtracted from the spectrum to yield a corrected baseline
(Golotvin and Williams 2000; Xi and Rocke 2008). Combining both frequency and
time domain methods further improves the quality of the baseline. Baseline correc-
tion routines are available in the operating systems of most modern NMR
instruments, although often the best baseline correction routines for complex spectra
are found in spectral deconvolution tools designed specifically for NMR-based
metabolomics (Weljie et al. 2006; Mercier et al. 2011; Worley and Powers 2014;
Ravanbakhsh et al. 2015).
The final step of NMR data processing is spectra alignment. In this step, the ppm
scale of all the experiments is calibrated by adjusting the position of the internal
standard (DSS) to 0 ppm. Then, the intensities of all the peaks are normalized using
the height of the internal standard peak. An example of a 1D NMR spectrum before
and after proper chemical shift referencing, shimming, phasing, solvent removal, and
baseline correction is shown in Fig. 6.
18 D. S. Wishart et al.
4 Data Analysis
After collecting a set of NMR spectra for a metabolomics study, the next step is data
analysis. The type of data analysis one undertakes depends on the experimental
design, the choice of the metabolomics experiment (targeted vs. untargeted), and the
type of multivariate statistical techniques or software available to the researcher.
Many excellent reviews have been written on experimental design and multivari-
ate statistics for metabolomics and readers who are interested in these topics should
refer to these publications (Madsen et al. 2010; Smolinska et al. 2012; Saccenti et al.
2014; Ebbels et al. 2019; Percival et al. 2020). We would encourage new users or
readers to explore a software tool called MetaboAnalyst to better understand and
visualize multivariate statistics as used in metabolomics (Xia et al. 2009).
MetaboAnalyst is an easy-to-use, web-based tool that is routinely used by approxi-
mately half of the global metabolomics community. It supports a wide range of
downstream data analysis applications with extensive graphics support including
principal component analysis (PCA), analysis of variance (ANOVA), partial least-
squares discriminant analysis (PLS-DA), heat mapping and clustering, biomarker
identification, pathway analysis, power analysis, and time series analysis (Verpoorte,
“Natural products drug discovery: on silica or in-silico?”; Millet, “Prospective
metabolomic studies in precision medicine. The AKRIBEA project”). A screenshot
of the MetaboAnalyst package is shown in Fig. 7. MetaboAnalyst is configured to
work with both targeted and untargeted metabolomics data, as well as with NMR and
MS data. It is also quite unique in that it has a number of freely available NMR
metabolomic data sets that users can test or explore.
For targeted metabolomics studies, the NMR spectra need to be analyzed using
specialized peak fitting software. This software fits a reference set of NMR spectra
obtained from pure compounds to the NMR spectra of the mixture of compounds
found in the biofluid or extract. By matching the positions and intensities (or area) of
peak clusters, the software identifies each reference spectrum and accurately
determines their concentrations in the biofluid. This approach is often called spectral
deconvolution. An illustration of how spectral deconvolution works is shown in
Fig. 8. Once the compounds are identified and their concentrations have been
precisely quantified, this information can then be compared against known or normal
concentrations for that biofluid or further analyzed to detect significant differences
between one sample (or group of samples) and the next.
The concepts underlying targeted NMR metabolomics emerged independently on
three separate occasions – for three different applications. Targeted metabolomics
was first described in 1991, as a novel approach to perform serum lipid and
lipoprotein particle profiling (Otvos et al. 1991). The protocol uses a reference
spectral library of different lipoprotein subclasses, to identify and quantify 15 differ-
ent subcategories of VLDL (very low density lipoprotein), LDL (low density
Practical Aspects of NMR-Based Metabolomics 19
Fig. 7 A screenshot montage of the MetaboAnalyst website. More than a dozen statistical modules
are available for analyzing metabolomics data
lipoprotein), and HDL (high density lipoprotein) particles (Jeyarajah et al. 2006).
Several variations of the method have been described in the literature, with some
methods using only 11 lipoprotein categories or others using more sophisticated
wavelet deconvolution algorithms (Serrai et al. 1998; Ala-Korpela et al. 2007).
Nevertheless, the method has proven to be particularly simple, fast, and robust
compared to traditional methods of lipoprotein profiling which are less accurate,
provide less information, and require time-consuming, manually intensive, multi-
step separations (McNamara et al. 2006).
Targeted NMR metabolomics emerged for a second time in 1993, when it was
used to identify brain metabolites from localized in vivo NMR spectroscopy
(Provencher 1993). However it was not until 2001 that the method, known as
LCModel, was made generally available (Provencher 2001). The central concept
behind the LCModel approach for metabolite identification is to fit the broad peaks
20 D. S. Wishart et al.
Fig. 8 A schematic illustration of the principles behind spectral deconvolution. The biofluid
spectrum at the top contains a combination of the three individual reference spectra below it. The
challenge in spectral deconvolution is to determine which combination of reference spectra, their
scaling, and positioning, best produces the observed spectrum
obtained from in vivo NMR data to spectral libraries of pure metabolites (called
in vitro basis sets) in which the spectra are artificially broadened and otherwise
mathematically modified to look like those seen for in vivo NMR spectra. LCModel
uses a constrained regularization method to handle differences in phase, baseline,
and line shapes between the in vitro and in vivo spectra. It is able to identify between
10 and 15 metabolites and can accurately estimate the metabolite concentrations and
their uncertainties (Simister et al. 2003; Marliani et al. 2007).
The third occasion in which targeted metabolomics emerged was in 2001, when it
was specifically implemented for small molecule identification and quantification in
biofluids and tissue extracts (i.e., traditional metabolomics) (Wishart et al. 2001).
While similar in principle to the LCModel and lipoprotein characterization software
mentioned above, the actual implementation is somewhat different. This is because
spectral deconvolution of high-resolution NMR spectra consisting of dozens of
small molecules requires the fitting of not just 15–20 broad peaks, but the fitting
of hundreds to thousands of very sharp peaks. Small variations in position, line
width, shape, or intensity due to pH or matrix effects can make the spectral fitting
problem particularly challenging and “ill-conditioned.” Likewise, because of spin-
coupling effects, the NMR spectrum of a small molecule collected at 500 MHz often
differs substantially from one collected at 800 MHz. To address these problems,
thousands of reference NMR spectra must be collected for hundreds of reference
compounds at different pH values and at different NMR spectrometer frequencies.
Additionally, very sophisticated curve fitting programs that robustly handle sparse
matrices and potential singularities must be employed.
Fortunately, a variety of software tools for small molecule NMR spectral
deconvolution have been developed over the past 10 years that make this spectral
fitting process relatively painless. These include commercial tools, such as the
Chenomx NMR Suite (Mercier et al. 2011), Bruker’s JuiceScreener (Monakhova
et al. 2014), WineScreener (Spraul et al. 2015), FoodScreener (https://www.bruker.
Practical Aspects of NMR-Based Metabolomics 21
com/en/products-and-solutions/mr/nmr-food-solutions/food-screener.html), and
in vitro diagnostic research system (IVDr). Bruker’s IVDr system expands the utility
of automated metabolite quantification to several biofluids (urine, cerebrospinal
fluid, plasma, and serum) by standardizing sample analyses through the
incorporation of standard hardware, standard operating procedures, and automated
sample handling and analysis (Bruker Corporation-Bruker 2013). The Bruker “X”-
-Screener and IVDr systems are normally bundled with specific NMR instruments or
licensed on a per-sample basis, making them relatively expensive. A cheaper
alternative to the commercial deconvolution and automated quantification tools are
a number of freely available, non-commercial tools, such as Batman (Hao et al.
2014), Bayesil (Ravanbakhsh et al. 2015), an automated quantification algorithm
(AQuA) (Röhnisch et al. 2018), an automatic method for identification and quantifi-
cation of metabolites (ASICS) (Tardivel et al. 2017) and rDolphin (Cañueto et al.
2018). A screenshot of the Bayesil web server and its standard output is shown in
Fig. 9. As can be seen in this figure, most deconvolution programs provide lists of
compound identities and estimated concentrations along with an interactive display
showing the fit between the observed NMR spectrum and the reference library NMR
spectra. Some of these deconvolution tools are almost fully automated (such as
Bayesil, Chenomx, and the Bruker products) while others require a fair bit of manual
manipulation.
In addition to tools such as Bayesil, Chenomx, and the Bruker “X”-Screener and
IVDr products, which identify and quantify organic compounds, there are now
NMR-based tools for identifying and quantifying inorganic compounds. In many
fields of metabolomics, metal ions are considered to be important metabolites and
this subdiscipline of studying metal ions is called “metallomics.” Normally metal
ions are measured via inductively coupled plasma mass spectrometry (ICP-MS).
However, it is also possible to identify and quantify metal ions and other inorganic
ions from 1H-NMR spectra. This can be done by taking advantage of the effect of
dissolved inorganic ions (invisible by NMR) on organic compound chemical shifts
(visible by NMR). This approach was described in detail by Takis et al. (2017). The
technique required the measurement of 4,000 variable synthetic urine mixtures
containing differing amounts of the most common organic compounds (> 90%
occurrence) and differing amounts of the most abundant inorganic ions in urine
and acquiring 1H-NMR spectra of each of these mixtures. From these spectra, Takis
et al. created an algorithm, called the Urine Shift Predictor, which is able to predict
the concentration-dependent, inorganic ion-induced chemical shifts on different
organic compounds. By measuring these chemical shift changes it is possible to
estimate inorganic ion and metal ion concentrations from 1H urine spectra.
Regardless of whether one measures organic or inorganic metabolite data
(or both), once the list of metabolites and their concentrations has been obtained
from a targeted metabolomics study, it is relatively easy to apply standard multivari-
ate statistics such as PCA or PLS-DA (and other tools found in packages such as
MetaboAnalyst) to identify significantly changed metabolites or to detect specific
sets of metabolites as biomarkers.
22 D. S. Wishart et al.
Fig. 9 A series of screenshots taken from the Bayesil NMR server. Bayesil is a freely available
spectral deconvolution package that can take high-resolution NMR spectra of biofluids, such as
serum, and automatically identify and quantify the compounds in that biofluid. The top image (a)
shows the Bayesil home page. The middle image (b) shows the interactive spectral viewer. The
bottom image (c) shows a selection of the compounds identified from the spectrum
Untargeted NMR metabolomics first emerged in the early 1990s when the complex-
ity of high-resolution NMR spectra collected on biofluids seemed to be too daunting
to allow routine compound identification (Gartland et al. 1990). Rather than
attempting to identify compounds via spectral deconvolution as is done with targeted
metabolomics, untargeted metabolomics exploits a field of science known as
chemometrics to simplify, classify, and interpret groups of NMR spectra. More
specifically, chemometrics is a branch of information science that uses mathematical
and statistical methods to identify patterns and extract information from large data
Practical Aspects of NMR-Based Metabolomics 23
Fig. 10 An example of how spectral alignment of NMR spectra can be performed. Typically,
multiple NMR spectra are collected, compared, and then aligned. The top figure shows multiple
superimposed 1D 1H NMR spectra of human serum. The glutamine peak at 2.427 ppm shows a
larger sensitivity to sample conditions between spectra compared to other nearby peaks. The bottom
figure shows the same spectra aligned using a spectral alignment algorithm (Savorani et al. 2010).
The glutamine peak is now better aligned between the spectra
sets collected on analytical instruments, such as UV, IR, and NMR spectrometers.
When chemometrics is applied to NMR data, it is essential to have many NMR
spectra already collected (generally dozens to hundreds of spectra, including both
cases and controls). These spectra must then be aligned and binned using specially
developed statistical approaches. A simple illustration of how spectral alignment is
done is shown in Fig. 10. After the alignment step has been completed, the spectra
must be scaled or normalized so that they can be easily compared. Once the scaling
and normalization are complete, multivariate statistical techniques such as PCA and
PLS-DA can be used to identify interesting spectral regions or clusters of peaks that
differentiate one group of spectra from another (Beckonert et al. 2007; Lindon et al.
2007; Barton et al. 2008).
A method called statistical total correlation spectroscopy or STOCSY has also
been developed which generates a pseudo-2D NMR spectrum representing the
correlation among the peaks in a set of NMR spectra (Cloarec et al. 2005). STOCSY
allows for the identification of peaks from the same compound or sets of compounds
that co-vary among the spectra. In an untargeted analysis, sometimes only a set of
statistically important spectral peaks or features is presented as the final result,
without compound identification. In other cases, compound identification occurs
only on the peaks which show the most significant changes in a particular study. The
final peak identification step may use spectral deconvolution, compound spike-in
methods, or peak look-up tables (Martínez-Arranz et al. 2015). A variety of software
packages for NMR statistical spectroscopy have been developed over the past
24 D. S. Wishart et al.
10–15 years to make this kind of analysis more routine. These include MVAPack
(Worley and Powers 2014), Automics (Wang et al. 2009), and KIMBLE (Verhoeven
et al. 2018).
For relatively simple biofluids with fewer than 60–70 NMR-detectable compounds
(such as serum, plasma, cerebrospinal fluid, fecal water, juice, or other fruit extracts)
targeted NMR metabolomics techniques that use spectral deconvolution appear to
work very well (Ravanbakhsh et al. 2015). Most of these methods focus on
analyzing 1D 1H NMR spectra, although methods have also been developed to
analyze 2D 1H NMR spectra and 2D heteronuclear spectra (Lewis et al. 2009; Bingol
et al. 2014, 2015). Extensive spectral libraries now exist for essentially all the
NMR-detectable compounds found in these biofluids and a number of the
deconvolution software tools are becoming almost fully automated. Indeed, some
software packages can be extremely fast and robust with >95% of the known
compounds in a given biofluid being identified and accurately quantified within a
few minutes (Mercier et al. 2011; Zheng et al. 2011; Hao et al. 2014; Ravanbakhsh
et al. 2015).
On the other hand, for very complex biofluids, such as cell growth media, cell
lysates, and urine, the corresponding NMR spectra are often too complex for any
existing spectral deconvolution packages. These biofluids can contain between
70–150 NMR-detectable compounds and often less than 50% of the known
compounds can be confidently identified or quantified using spectral deconvolution.
Furthermore, the quality of the annotation is often highly dependent on the skill or
experience of the operator (Sokolenko et al. 2013). Therefore, for complex biofluids,
such as urine, statistical spectroscopy techniques or untargeted NMR approaches
appear to offer the best option for spectral interpretation. These approaches allow
useful results to be obtained with relatively little manual effort.
5 Biological Interpretation
The ultimate goal of acquiring and analyzing NMR metabolomics data is to use the
results to reveal something about the biology of the system under study. This may
involve the identification of biomarkers or biomarker panels, the tabulation of
reference metabolite concentrations for specific samples or sample types, the char-
acterization of activated or deactivated biological pathways, or the acquisition of
insights into the underlying molecular metabolic or biochemical mechanisms
associated with a particular biological condition. Rather than describing the software
or methods used to perform biological interpretation, here we will focus on some of
the more successful applications of NMR-based metabolomics toward biological
interpretation.
Practical Aspects of NMR-Based Metabolomics 25
(Capitani et al. 2017; Sobolev et al. 2019), and to the composition and quality of
foods, such as wine, beer, oil, juice, milk, honey, and fruit (Wishart 2008b; Melzer
et al. 2013; Kim et al. 2016). NMR-based methods have also been applied to
environmental metabolomics. In particular, NMR-based metabolomics has been
used to explore how organisms respond to environmental changes, pollution, and
climate change (Williams et al. 2009; Simpson and Bearden 2013; Sumner et al.
2015).
The field of metabolomics has been around for 20 years. However, NMR has been a
key part of metabolic studies for more than 40 years. The popularity of NMR as a
metabolomics platform is largely due to its high instrument stability, reproducibility,
simple non-destructive sample preparation, ease of quantification, and its amenabil-
ity to automation. However, NMR is not without its limitations. Compared to MS,
NMR has relatively poor sensitivity, a large instrument footprint, and high up-front
and maintenance costs. NMR also lags behind MS in terms of available
metabolomics libraries, easy-to-use data processing and analysis tools, and easy-
to-use sample preparation kits.
However, progress is being made on almost all the fronts where NMR lags behind
MS. To address the cost and space issues of NMR instrumentation,
non-superconducting, bench-top NMR instruments with field strengths of up to
60 MHz are coming into routine use (Percival et al. 2019; Izquierdo-Garcia et al.
2020) and ones approaching or exceeding 200 MHz (Blümich and Singh 2018) are
being developed. However, these lower field instruments have lower sensitivity and
less spectral resolution than higher field instruments. These disadvantages could be
compensated by adopting 2D UF-NMR techniques (Giraudeau and Frydman 2014)
on bench-top instruments.
At the other extreme, the recent introduction of 1.2 GHz NMR spectrometers
promises to greatly improve the sensitivity and resolution of many NMR-based
metabolomics studies (Schwalbe 2017; Luchinat et al. 2021). These super-high field
instruments will lower the limits of metabolite detection to the high nanomolar range
(compared to 5 μM with lower field NMR instruments) and will likely double the
number of metabolites detectable in biofluids such as serum and urine.
Developments in NMR probe technology are also leading to some exciting
improvements in sensitivity and compound structural elucidation. For instance, the
recently developed 13C-optimized 1.5-mm cryoprobe and a 1H-13C dual-optimized
NMR probe permit 2D 1H-13C HSQC experiments to be collected at natural abun-
dance (Ramaswamy et al. 2013, 2016; Clendinen et al. 2014, 2015).
Another route to improving the sensitivity of NMR is hyperpolarization where
sensitivity enhancements of several thousand-fold (allowing detection of metabolites
in the nM range) are technically possible. While hyperpolarization has been used for
13
C-labeled metabolites (Ardenkjaer-Larsen et al. 2003; Keshari et al. 2010; Lumata
et al. 2015; Dey et al. 2020), the need for specialized equipment and sample
28 D. S. Wishart et al.
which no reference NMR (or MS) spectra exist. On the other hand, “unknown
unknowns” are compounds that are completely novel and have never been described
before. To identify “known unknowns,” a targeted NMR approach is often possible.
For instance, one can use standard 2D NMR approaches to determine the unknown’s
molecular constituents or atomic connectivities which might lead to a possible match
to an existing/known structure. Alternately, one can use a “suspect screening”
approach where intelligent guesses (guided by chemical shift similarities) and the
spiking in of pure standard of the suspected compound into the sample can help
confirm its identity.
Obviously, the most appropriate solution to identifying “known unknowns” is
adding more high-quality, experimentally collected, fully assigned reference NMR
data for many more metabolites to NMR databases such as the HMDB, BMRB, or
nmrshiftdb (McAlpine et al. 2019). Unfortunately, the pace at which reference
experimental NMR spectra are being added to these databases has slowed consider-
ably. In the absence of reference experimental NMR data, the use of predicted NMR
spectra to identify “known unknowns” may be possible. Certainly, continued
advancements in computing power along with improvements to the accuracy of
quantum mechanical and machine learning based predictions of NMR chemical
shifts and NMR coupling constants suggest that computational approaches could
be an appealing new route (Borges et al. 2021).
To identify truly novel compounds (i.e., “unknown unknowns”), de novo struc-
ture determination would be required. De novo structure determination often
requires partial or complete purification of the compound of interest, followed by a
combination of mass spectrometry and 2D NMR analysis of the purified compound
(Garcia-Perez et al. 2020). The process of characterizing novel compounds, how-
ever, can be particularly time consuming and also often limited by the low
concentrations of many unknown compounds. Computational approaches for the
analysis of spectral data and the proposal of candidate structures (through techniques
such as computer-aided structure elucidation – also known as CASE) can accelerate
the de novo structure determination process (Boiteau et al. 2018; Leggett et al.
2019).
The computational and experimental innovations being applied to the identifica-
tion of unknowns by NMR is typical of the remarkable inventiveness and impressive
creativity often seen in the NMR community. No doubt many other innovations in
NMR instrumentation, in NMR spectral collection, or in NMR data analysis that will
benefit NMR-based metabolomics are on the horizon. Certainly, for anyone who has
been in the NMR field for more than a few years, one quickly learns to never
underestimate the potential of NMR spectroscopists to come up with some remark-
able, paradigm-changing innovations. It is probably fair to expect that the next
20 years of NMR-based metabolomics will be as interesting and fruitful as the
past 20 years.
Acknowledgments The authors wish to thank Genome Alberta (a division of Genome Canada),
The Canadian Institutes of Health Research (CIHR), Western Economic Diversification (WED),
and Alberta Innovates Health Solutions (AIHS) for financial support.
30 D. S. Wishart et al.
Glossary
Baseline correction A spectral processing technique that yields a more pleasant looking
NMR spectrum where signal-free regions appear as completely flat
lines with zero intensity
BEST Band-Selective Excitation Transient
Chemometrics A branch of information science that uses mathematical and statistical
methods to identify patterns and extract information from large data
sets collected on analytical instruments, such as UV, IR, and NMR
spectrometers
CPMG Carr-Purcell-Meiboom-Gill
CPMG experiment A pulse that can filter out the signals arising from large molecules,
such as proteins or lipoproteins, from the spectrum (without the need
for ultrafiltration or solvent extraction)
COSY COrrelated SpectroscopY
DSS 4,4-dimethyl-4-silapentane-1-sulfonic acid, a chemical shift reference
compound
ERETIC Electronic REference To access In vivo Concentrations, an electronic
reference signal
HSQC Heteronuclear Single Quantum Coherence Spectroscopy
IEM Inborn error of metabolism. They are rare genetic disorders
characterized by significant changes (several-fold increase or absence)
in the concentration of specific metabolites that result from
disturbances in normal metabolism
INADEQUATE Incredible Natural Abundance DoublE QUAntum Transfer
Experiment
IUPAC The International Union of Pure and Applied Chemistry
IUBMB The International Union of Biochemistry and Molecular Biology
Metabolomics A branch of analytical chemistry that comprehensively characterizes
the molecules in various biofluids and tissues
Metabolites The chemical constituents of the metabolome
Metabolome The complete collection of all chemicals or metabolites found within
cells, biofluids, organs, or organisms
Metnoesy experiment A simple 1D NOESY pulse sequence that provides solvent suppres-
sion before the experiment and during the mixing time without the use
of gradients
NOESY Nuclear Overhauser Effect Spectroscopy
Phasing An NMR spectral adjustment process that is designed to maximize the
absorptive character of NMR peaks over all regions of an NMR
spectrum
Relaxation delay The sum of the acquisition time and acquisition delay prior to the
next scan
SABRE-SHEATH Signal Amplification by Reversible Exchange in SHield Enables
Alignment Transfer to Heteronuclei
SOFAST Band-Selective Optimized Flip Angle Short Transient
STOCSY Statistical total correlation spectroscopy
T1 Longitudinal relaxation time
T2 Transverse relaxation time
Targeted metabolomics A metabolomics technique that uses spectral deconvolution software
to identify and quantify fluid-specific or targeted metabolites in indi-
vidual spectra
Practical Aspects of NMR-Based Metabolomics 31
References
Akoka S, Barantin L, Trierweiler M (1999) Concentration measurement by proton NMR using the
ERETIC method. Anal Chem 71(13):2554–2557. https://doi.org/10.1021/ac981422i
Ala-Korpela M et al (2007) The inherent accuracy of 1H NMR spectroscopy to quantify plasma
lipoproteins is subclass dependent. Atherosclerosis 190(2):352–358. https://doi.org/10.1016/j.
atherosclerosis.2006.04.020
Ardenkjaer-Larsen JH et al (2003) Increase in signal-to-noise ratio of >10,000 times in liquid-state
NMR. Proc Natl Acad Sci 100(18):10158–10163. https://doi.org/10.1073/pnas.1733835100
Bales JR, Higham DP et al (1984a) Use of high-resolution proton nuclear magnetic resonance
spectroscopy for rapid multi-component analysis of urine. Clin Chem 30(3):426–432
Bales JR, Sadler PJ et al (1984b) Urinary excretion of acetaminophen and its metabolites as studied
by proton NMR spectroscopy. Clin Chem 30(10):1631–1636
Barton RH et al (2008) High-throughput 1H NMR-based metabolic analysis of human serum and
urine for large-scale epidemiological studies: validation study. Int J Epidemiol 37(Suppl 1):i31–
i40. https://doi.org/10.1093/ije/dym284
Bassini A, Cameron LC (2014) Sportomics: building a new concept in metabolic studies and
exercise science. Biochem Biophys Res Commun 445(4):708–716. https://doi.org/10.1016/j.
bbrc.2013.12.137
Bathe OF et al (2011) Feasibility of identifying pancreatic cancer based on serum metabolomics.
Cancer Epidemiol Biomarkers Prev 20(1):140–147. https://doi.org/10.1158/1055-9965.EPI-
10-0712
Beckonert O et al (2007) Metabolic profiling, metabolomic and metabonomic procedures for NMR
spectroscopy of urine, plasma, serum and tissue extracts. Nat Protoc 2(11):2692–2703. https://
doi.org/10.1038/nprot.2007.376
Bernini P et al (2011) Standard operating procedures for pre-analytical handling of blood and urine
for metabolomic studies and biobanks. J Biomol NMR 49(3):231–243. https://doi.org/10.1007/
S10858-011-9489-1
Binczyk F, Tarnawski R, Polanska J (2015) Strategies for optimizing the phase correction
algorithms in nuclear magnetic resonance spectroscopy. Biomed Eng Online 14(2):S5. https://
doi.org/10.1186/1475-925X-14-S2-S5
Bingol K, Brüschweiler R (2014) Multidimensional approaches to NMR-based metabolomics. Anal
Chem 86(1):47–57. https://doi.org/10.1021/ac403520j
Bingol K et al (2014) Customized metabolomics database for the analysis of NMR 1H-1H TOCSY
and 13C-1H HSQC-TOCSY spectra of complex mixtures. Anal Chem 86(11):5494–5501.
https://doi.org/10.1021/ac500979g
Bingol K et al (2015) Unified and isomer-specific NMR metabolomics database for the accurate
analysis of 13 C – 1 H HSQC spectra. ACS Chem Biol 10(2):452–459. https://doi.org/10.1021/
cb5006382
Bingol K et al (2016) Comprehensive metabolite identification strategy using multiple
two-dimensional NMR spectra of a complex mixture implemented in the COLMARm web
server. Anal Chem 88(24):12411–12418. https://doi.org/10.1021/acs.analchem.6b03724
Blankenberg FG et al (1997) Quantitative analysis of apoptotic cell death using proton nuclear
magnetic resonance spectroscopy. Blood 89(10):3778–3786
32 D. S. Wishart et al.
Cohen SM, Ogawa S, Shulman RG (1979) 13C NMR studies of gluconeogenesis in rat liver cells:
utilization of labeled glycerol by cells from euthyroid and hyperthyroid rats. Proc Natl Acad Sci
U S A 76(4):1603–1609. https://doi.org/10.1073/pnas.76.4.1603
Dame ZT et al (2015) The human saliva metabolome. Metabolomics 11(6):1864–1883. https://doi.
org/10.1007/s11306-015-0840-5
de Brouwer H (2009) Evaluation of algorithms for automated phase correction of NMR spectra. J
Magn Reson 201(2):230–238. https://doi.org/10.1016/j.jmr.2009.09.017
Dey A et al (2020) Hyperpolarized NMR metabolomics at natural 13C abundance. Anal Chem
92(22):14867–14871. https://doi.org/10.1021/acs.analchem.0c03510
Ebbels TMD, De Iorio M, Stephens DA (2019) Statistical methods in metabolomics. In: Balding D,
Moltke I, Marioni J (eds) Handbook of statistical genomics. Wiley, pp 949–975. https://doi.org/
10.1002/9781119487845.ch34
Eisner R et al (2013) A machine-learned predictor of colonic polyps based on urinary
metabolomics. Biomed Res Int 2013:1–11. https://doi.org/10.1155/2013/303982
Embade N et al (2019) NMR-based newborn urine screening for optimized detection of inherited
errors of metabolism. Sci Rep 9(1):13067. https://doi.org/10.1038/s41598-019-49685-x
Emwas A-H et al (2015) Standardizing the experimental conditions for using urine in NMR-based
metabolomic studies with a particular focus on diagnostic studies: a review. Metabolomics
11(4):872–894. https://doi.org/10.1007/S11306-014-0746-7
Emwas A-H et al (2018) Recommended strategies for spectral processing and post-processing of
1D 1H-NMR data of biofluids with a particular focus on urine. Metabolomics 14(3):31. https://
doi.org/10.1007/s11306-018-1321-4
Fan TW-M, Lane AN (2011a) Erratum to: NMR-based stable isotope resolved metabolomics in
systems biochemistry. J Biomol NMR 49:325. https://doi.org/10.1007/s10858-011-9503-7
Fan TWM, Lane AN (2011b) NMR-based stable isotope resolved metabolomics in systems
biochemistry. J Biomol NMR 49(3–4):267–280. https://doi.org/10.1007/s10858-011-9484-6
Fan TWM, Lane AN (2016) Applications of NMR spectroscopy to systems biochemistry. Prog
Nucl Magn Reson Spectrosc 92–93:18–53. https://doi.org/10.1016/j.pnmrs.2016.01.005
Farshidfar F et al (2012) Serum metabolomic profile as a means to distinguish stage of colorectal
cancer. Genome Med 4(5):42. https://doi.org/10.1186/gm341
Fiehn O (2002) Metabolomics – the link between genotypes and phenotypes. Plant Mol Biol
48(1–2):155–171. https://doi.org/10.1023/A:1013713905833
Fiehn O et al (2007) The metabolomics standards initiative (MSI). Metabolomics 3(3):175–178.
https://doi.org/10.1007/S11306-007-0070-6
Fong MY, McDunn J, Kakar SS (2011) Identification of metabolites in the normal ovary and their
transformation in primary and metastatic ovarian cancer. PLoS One 6(5):e19963. https://doi.
org/10.1371/journal.pone.0019963
Foroozandeh M, Jeannerat D (2010) Deciphered chemical shifts in aliased spectra recorded with
two slightly different narrow windows or differential chemical shift evolution. ChemPhysChem
11(12):2503–2505. https://doi.org/10.1002/cphc.201000421
Foroutan A et al (2019) Chemical composition of commercial cow’s milk. J Agric Food Chem
67(17):4897–4914. https://doi.org/10.1021/acs.jafc.9b00204
Foroutan A et al (2020) The bovine metabolome. Metabolites 10(6):233. https://doi.org/10.3390/
metabo10060233
Fossel ET, Carr JM, McDonagh J (1986) Detection of malignant tumors. Water-suppressed proton
nuclear magnetic resonance spectroscopy of plasma. N Engl J Med 315(22):1369–1376. https://
doi.org/10.1056/NEJM198611273152201
Garcia-Perez I et al (2020) Identifying unknown metabolites using NMR-based metabolic profiling
techniques. Nat Protoc 15(8):2538–2567. https://doi.org/10.1038/s41596-020-0343-3
Gartland KP et al (1990) Pattern recognition analysis of high resolution 1H NMR spectra of urine. A
nonlinear mapping approach to the classification of toxicological data. NMR Biomed 3(4):
166–172. https://doi.org/10.1002/nbm.1940030404
34 D. S. Wishart et al.
Ghini V et al (2019) NMR for sample quality assessment in metabolomics. N Biotechnol 52:25–34.
https://doi.org/10.1016/J.NBT.2019.04.004
Giraudeau P, Frydman L (2014) Ultrafast 2D NMR: an emerging tool in analytical spectroscopy.
Annu Rev Anal Chem 7(1):129–161. https://doi.org/10.1146/annurev-anchem-071213-020208
Goldansaz SA et al (2017) Livestock metabolomics and the livestock metabolome: a systematic
review. PLoS One 12(5):e0177675. https://doi.org/10.1371/journal.pone.0177675
Golotvin S, Williams A (2000) Improved baseline recognition and modeling of FT NMR spectra. J
Magn Reson 146(1):122–125. https://doi.org/10.1006/jmre.2000.2121
Gowda GAN et al (2010) Quantitative analysis of blood plasma metabolites using isotope enhanced
NMR methods. Anal Chem 82(21):8983–8990. https://doi.org/10.1021/ac101938w
Griffiths RD, Edwards RH (1987) Magnetic resonance spectroscopy in the recognition of metabolic
disease. J Inherit Metab Dis 10(Suppl 1):147–158
Hailemariam D et al (2014) Identification of predictive biomarkers of disease state in transition
dairy cows. J Dairy Sci 97(5):2680–2693. https://doi.org/10.3168/jds.2013-6803
Halamek J, Vondra V, Kasal M (1994) The elimination of baseline distortions induced by audio
filters. J Magn Reson Ser 110(2):194–197. https://doi.org/10.1006/jmra.1994.1204
Hanahan D, Weinberg R (2000) The hallmarks of cancer. Cell 100(1):57–70. https://doi.org/10.
1016/S0092-8674(00)81683-9
Hansen DF (2019) Using deep neural networks to reconstruct non-uniformly sampled NMR
spectra. J Biomol NMR 73(10–11):577–585. https://doi.org/10.1007/s10858-019-00265-1
Hansen AL et al (2021) 2D NMR-based metabolomics with HSQC/TOCSY NOAH
supersequences. Anal Chem 93(15):6112–6119. https://doi.org/10.1021/acs.analchem.0c05205
Hao J et al (2014) Bayesian deconvolution and quantification of metabolites in complex 1D NMR
spectra using BATMAN. Nat Protoc 9(6):1416–1427. https://doi.org/10.1038/nprot.2014.090
Harris RK et al (2002) NMR nomenclature: nuclear spin properties and conventions for chemical
shifts. IUPAC recommendations 2001. Solid State Nucl Magn Reson 22(4):458–483. https://
doi.org/10.1006/snmr.2002.0063
Heuer A, Haeberlen U (1989) A new method for suppressing baseline distortions in FT NMR. J
Magn Reson (1969) 85(1):79–94. https://doi.org/10.1016/0022-2364(89)90322-3
Holmes E, Wilson ID, Nicholson JK (2008) Metabolic phenotyping in health and disease. Cell
134(5):714–717. https://doi.org/10.1016/j.cell.2008.08.026
Hu K, Westler WM, Markley JL (2011) Simultaneous quantification and identification of individual
chemicals in metabolite mixtures by two-dimensional extrapolated time-zero 1H-13C HSQC
(HSQC 0). J Am Chem Soc 133(6):1662–1665. https://doi.org/10.1021/ja1095304
Iles RA et al (1984) Rapid screening of metabolic diseases by proton NMR. Lancet 2(8413):
1221–1222. https://doi.org/10.1016/S0140-6736(84)92783-1
Izquierdo-Garcia JL et al (2020) Discovery and validation of an NMR-based metabolomic profile in
urine as TB biomarker. Sci Rep 10(1):22317. https://doi.org/10.1038/s41598-020-78999-4
Jeyarajah EJ, Cromwell WC, Otvos JD (2006) Lipoprotein particle analysis by nuclear magnetic
resonance spectroscopy. Clin Lab Med 26(4):847–870. https://doi.org/10.1016/j.cll.2006.
07.006
Kaddurah-Daouk R (2006) Metabolic profiling of patients with schizophrenia. PLoS Med 3(8):
e363. https://doi.org/10.1371/journal.pmed.0030363
Karamanos TK et al (2015) Mechanisms of amyloid formation revealed by solution NMR. Prog
Nucl Magn Reson Spectrosc 88–89:86–104. https://doi.org/10.1016/j.pnmrs.2015.05.002
Karu N et al (2018) A review on human fecal metabolomics: methods, applications and the human
fecal metabolome database. Anal Chim Acta 1030:1–24. https://doi.org/10.1016/j.aca.2018.
05.031
Kazimierczuk K et al (2010) Random sampling in multidimensional NMR spectroscopy. Prog Nucl
Magn Reson Spectrosc 57(4):420–434. https://doi.org/10.1016/j.pnmrs.2010.07.002
Keshari KR et al (2010) Hyperpolarized (13)C spectroscopy and an NMR-compatible bioreactor
system for the investigation of real-time cellular metabolism. Magn Reson Med 63(2):322–329.
https://doi.org/10.1002/mrm.22225
Practical Aspects of NMR-Based Metabolomics 35
Kim S et al (2016) Food metabolomics: from farm to human. Curr Opin Biotechnol 37:16–23.
https://doi.org/10.1016/j.copbio.2015.09.004
Kim E et al (2019) Urine-NMR metabolomics for screening of advanced colorectal adenoma and
early stage colorectal cancer. Sci Rep 9(1):4786. https://doi.org/10.1038/s41598-019-41216-y
Kork F et al (2012) A biomarker for severity of Alzheimer’s disease: 1H-NMR resonances in
cerebrospinal fluid correlate with performance in mini-mental-state-exam. Biomarkers 17(1):
36–42. https://doi.org/10.3109/1354750X.2011.635806
Kostidis S, Mikros E (2015) NMR studies of inborn errors of metabolism. In: eMagRes. Wiley,
Chichester, pp 57–68. https://doi.org/10.1002/9780470034590.emrstm1400
Kupče Ē, Claridge TDW (2017) NOAH: NMR supersequences for small molecule analysis and
structure elucidation. Angew Chem Int Ed 56(39):11779–11783. https://doi.org/10.1002/anie.
201705506
Lane AN et al (2011) Stable isotope-resolved metabolomics (SIRM) in cancer research with clinical
application to nonsmall cell lung cancer. OMICS 15(3):173–182. https://doi.org/10.1089/omi.
2010.0088
Le Guennec A, Giraudeau P, Caldarelli S (2014) Evaluation of fast 2D NMR for metabolomics.
Anal Chem 86(12):5946–5954. https://doi.org/10.1021/ac500966e
Leggett A et al (2019) Identification of unknown metabolomics mixture compounds by combining
NMR, MS, and cheminformatics. Methods Enzymol 615:407–422. https://doi.org/10.1016/BS.
MIE.2018.09.003
Levy PA (2010) An overview of newborn screening. J Dev Behav Pediatr 31(7):622–631. https://
doi.org/10.1097/DBP.0b013e3181eedf01
Lewis IA et al (2007) Method for determining molar concentrations of metabolites in complex
solutions from two-dimensional 1H-13C NMR spectra. Anal Chem 79(24):9385–9390. https://
doi.org/10.1021/ac071583z
Lewis IA, Schommer SC, Markley JL (2009) rNMR: open source software for identifying and
quantifying metabolites in NMR spectra. Magn Reson Chem 47(Suppl 1):S123. https://doi.org/
10.1002/mrc.2526
Lin CY et al (2007) Evaluation of metabolite extraction strategies from tissue samples using NMR
metabolomics. Metabolomics 3(1):55–67. https://doi.org/10.1007/s11306-006-0043-1
Lindon JC et al (2005) The consortium for metabonomic toxicology (COMET): aims, activities and
achievements. Pharmacogenomics 6(7):691–699. https://doi.org/10.2217/14622416.6.7.691
Lindon JC, Holmes E, Nicholson JK (2007) Metabonomics in pharmaceutical R & D. FEBS J
274(5):1140–1151. https://doi.org/10.1111/j.1742-4658.2007.05673.x
Lopez JM, Cabrera R, Maruenda H (2019) Ultra-clean pure shift 1 H-NMR applied to
metabolomics profiling. Sci Rep 9(1):6900. https://doi.org/10.1038/s41598-019-43374-5
Luchinat E et al (2021) Protein in-cell NMR spectroscopy at 1.2 GHz. J Biomol 75(2–3):97–107.
https://doi.org/10.1007/s10858-021-00358-w
Ludwig C, Viant M (2010) Two-dimensional J-resolved NMR spectroscopy: review of a key
methodology in the metabolomics toolbox. Phytochem Anal 21(1):22–32. https://doi.org/10.
1002/PCA.1186
Lumata L et al (2015) Hyperpolarized 13C magnetic resonance and its use in metabolic assessment
of cultured cells and perfused organs. Methods Enzymol 561:73–106. https://doi.org/10.1016/
bs.mie.2015.04.006
Lutz N, Sweedler J, Wevers R (eds) (2013) Methodologies for metabolomics: experimental
strategies and techniques. Cambridge University Press, Cambridge. https://doi.org/10.1017/
CBO9780511996634
Madsen R, Lundstedt T, Trygg J (2010) Chemometrics in metabolomics – a review in human
disease diagnosis. Anal Chim Acta 659(1–2):23–33. https://doi.org/10.1016/j.aca.2009.11.042
Mak CM et al (2013) Inborn errors of metabolism and expanded newborn screening: review and
update. Crit Rev Clin Lab Sci 50(6):142–162. https://doi.org/10.3109/10408363.2013.847896
36 D. S. Wishart et al.
Nagana Gowda GA, Gowda YN, Raftery D (2015) Expanding the limits of human blood metabolite
quantitation using NMR spectroscopy. Anal Chem 87(1):706–715. https://doi.org/10.1021/
ac503651e
Namer IJ et al (2011) Metabolomic characterization of ovarian epithelial carcinomas by HRMAS-
NMR spectroscopy. J Oncol 2011:174019. https://doi.org/10.1155/2011/174019
Nicholson JK, Wilson ID (2003) Opinion: understanding “global” systems biology: metabonomics
and the continuum of metabolism. Nat Rev Drug Discov 2(8):668–676. https://doi.org/10.1038/
nrd1157
Nicholson JK et al (1984a) Monitoring metabolic disease by proton NMR of urine. Lancet 2(8405):
751–752. https://doi.org/10.1016/S0140-6736(84)92656-4
Nicholson JK et al (1984b) Proton-nuclear-magnetic-resonance studies of serum, plasma and urine
from fasting normal and diabetic subjects. Biochem J 217(2):365–375. https://doi.org/10.1042/
bj2170365
Nicholson JK et al (2002) Metabonomics: a platform for studying drug toxicity and gene function.
Nat Rev Drug Discov 1(2):153–161. https://doi.org/10.1038/nrd728
Oliver S et al (1998) Systematic functional analysis of the yeast genome. Trends Biotechnol 16(9):
373–378. https://doi.org/10.1016/S0167-7799(98)01214-1
Otvos JD, Jeyarajah EJ, Bennett DW (1991) Quantification of plasma lipoproteins by proton
nuclear magnetic resonance spectroscopy. Clin Chem 37(3):377–386
Palmnas MSA, Vogel HJ (2013) The future of NMR metabolomics in cancer therapy: towards
personalizing treatment and developing targeted drugs? Metabolites 3(2):373–396. https://doi.
org/10.3390/metabo3020373
Paruzzo F, Bruderer S, Janjar Y, Bjoern Heitmann B, Bolliger C (2020) Automatic signal region
detection in 1H NMR spectra using deep learning [White paper]. Switzerland
Pathan M et al (2011) “Multi-scan single shot” quantitative 2D NMR: a valuable alternative to fast
conventional quantitative 2D NMR. Analyst 136(15):3157–3163. https://doi.org/10.1039/
c1an15278e
Percival BC et al (2019) Low-field, benchtop NMR spectroscopy as a potential tool for point-of-
care diagnostics of metabolic conditions: validation, protocols and computational models. High-
Throughput 8(1):2. https://doi.org/10.3390/ht8010002
Percival B et al (2020) Chapter 1: univariate and multivariate statistical approaches to the analysis
and interpretation of NMR-based metabolomics datasets of increasing complexity. In: Compu-
tational techniques for analytical chemistry and bioanalysis. Royal Society of Chemistry, pp
1–40. https://doi.org/10.1039/9781788015882-00001
Peterson DJ, Loening NM (2007) QQ-HSQC: a quick, quantitative heteronuclear correlation
experiment for NMR spectroscopy. Magn Reson Chem 45(11):937–941. https://doi.org/10.
1002/mrc.2073
Provencher SW (1993) Estimation of metabolite concentrations from localized in vivo proton NMR
spectra. Magn Reson Med 30(6):672–679. https://doi.org/10.1002/mrm.1910300604
Provencher SW (2001) Automatic quantitation of localized in vivo 1H spectra with LCModel.
NMR Biomed 14(4):260–264. https://doi.org/10.1002/nbm.698
Psychogios N et al (2011) The human serum metabolome. PLoS One 6(2):e16957. https://doi.org/
10.1371/journal.pone.0016957
Ramaswamy V et al (2013) Development of a 13C-optimized 1.5-mm high temperature
superconducting NMR probe. J Magn Reson 235:58–65. https://doi.org/10.1016/j.jmr.2013.
07.012
Ramaswamy V et al (2016) Development of a 1H-13C dual-optimized NMR probe based on
double-tuned high temperature superconducting resonators. IEEE Trans Appl Supercond
26(3):1–5. https://doi.org/10.1109/TASC.2016.2522302
Ravanbakhsh S et al (2015) Accurate, fully-automated NMR spectral profiling for metabolomics.
PLoS One 10(5):1–15. https://doi.org/10.1371/journal.pone.0124219
38 D. S. Wishart et al.
Spicer R, Salek R, Steinbeck C (2017) A decade after the metabolomics standards initiative it’s time
for a revision. Sci Data 4(1):170138. https://doi.org/10.1038/sdata.2017.138
Spraul M et al (2015) Wine analysis to check quality and authenticity by fully-automated
1 H-NMR. BIO Web Conf 5(23):02022. https://doi.org/10.1051/bioconf/20150502022
Steimers E et al (2020) Application of a new method for simultaneous phase and baseline correction
of NMR signals (SINC). Magn Reson Chem 58(3):260–270. https://doi.org/10.1002/mrc.4964
Sumner LW et al (2015) Modern plant metabolomics: advanced natural product gene discoveries,
improved technologies, and future prospects. Nat Prod Rep 32(2):212–229. https://doi.org/10.
1039/C4NP00072B
Sundekilde UK et al (2014) Association between the bovine milk metabolome and rennet-induced
coagulation properties of milk. J Dairy Sci 97(10):6076–6084. https://doi.org/10.3168/jds.
2014-8304
Takis PG et al (2017) Deconvoluting interrelationships between concentrations and chemical shifts
in urine provides a powerful analysis tool. Nat Commun 8(1):1662. https://doi.org/10.1038/
s41467-017-01587-0
Tal A, Frydman L (2010) Single-scan multidimensional magnetic resonance. Prog Nucl Magn
Reson Spectrosc 57(3):241–292. https://doi.org/10.1016/j.pnmrs.2010.04.001
Tardivel P et al (2017) ASICS: an automatic method for identification and quantification of
metabolites in complex 1D 1H NMR spectra. Metabolomics 13(10):109. https://doi.org/10.
1007/s11306-017-1244-5
Tasic L et al (2017) Metabolomics and lipidomics analyses by1H nuclear magnetic resonance of
schizophrenia patient serum reveal potential peripheral biomarkers for diagnosis. Schizophr Res
185:182–189. https://doi.org/10.1016/j.schres.2016.12.024
Tayyari F et al (2013) 15N-Cholamine – a smart isotope tag for combining NMR- and MS-based
metabolite profiling. Anal Chem 85(18):8715–8721. https://doi.org/10.1021/ac401712a
Teahan O et al (2011) Metabolic signatures of malignant progression in prostate epithelial cells. Int
J Biochem Cell Biol 43(7):1002–1009. https://doi.org/10.1016/j.biocel.2010.07.003
Teng Q et al (2009) A direct cell quenching method for cell-culture based metabolomics.
Metabolomics 5(2):199–208. https://doi.org/10.1007/s11306-008-0137-z
Theis T et al (2016) Direct and cost-efficient hyperpolarization of long-lived nuclear spin states on
universal 15N2-diazirine molecular tags. Sci Adv 2(3):e1501438. https://doi.org/10.1126/
sciadv.1501438
Tilgner M et al (2019) High-resolution magic angle spinning (HRMAS) NMR methods in
metabolomics. Methods Mol Biol 2037:49–67. https://doi.org/10.1007/978-1-4939-9690-2_4
Truong ML et al (2015) 15N hyperpolarization by reversible exchange using SABRE-SHEATH. J
Phys Chem 119(16):8786–8797. https://doi.org/10.1021/acs.jpcc.5b01799
Verhoeven A, Giera M, Mayboroda OA (2018) KIMBLE: a versatile visual NMR metabolomics
workbench in KNIME. Anal Chim Acta 1044:66–76. https://doi.org/10.1016/j.aca.2018.07.070
Viant MR (2008) Recent developments in environmental metabolomics. Mol Biosyst 4(10):
980–986. https://doi.org/10.1039/b805354e
Vuckovic D (2012) Current trends and challenges in sample preparation for global metabolomics
using liquid chromatography-mass spectrometry. Anal Bioanal Chem 403(6):1523–1548.
https://doi.org/10.1007/s00216-012-6039-y
Wang T et al (2009) Automics: an integrated platform for NMR-based metabonomics spectral
processing and data analysis. BMC Bioinformatics 10(1):83. https://doi.org/10.1186/1471-
2105-10-83
Warburg O (1956) On the origin of cancer cells. Science 123(3191):309–314. https://doi.org/10.
1126/SCIENCE.123.3191.309
Watanabe R et al (2016) Quantitative nuclear magnetic resonance spectroscopy based on PULCON
methodology: application to quantification of invaluable marine toxin, okadaic acid. Toxins
8(10):294. https://doi.org/10.3390/toxins8100294
40 D. S. Wishart et al.
Weber F et al (2012) 31P and 13C solid-state NMR spectroscopy to study collagen synthesis and
biomineralization in polymer-based bone implants. NMR Biomed 25(3):464–475. https://doi.
org/10.1002/nbm.1649
Weiner MW et al (1989) Clinical magnetic resonance spectroscopy of brain, heart, liver, kidney,
and cancer. A quantitative approach. NMR Biomed 2(5–6):290–297. https://doi.org/10.1002/
nbm.1940020519
Weljie AM et al (2006) Targeted profiling: quantitative analysis of 1H NMR metabolomics data.
Anal Chem 78(13):4430–4442. https://doi.org/10.1021/ac060209g
Weljie AM et al (2011) 1H NMR metabolomics identification of markers of hypoxia-induced
metabolic shifts in a breast cancer model system. J Biomol NMR 49(3–4):185–193. https://doi.
org/10.1007/s10858-011-9486-4
Williams TH et al (1979) Novel application of proton nuclear magnetic resonance spectroscopy in
the identification of 2’-chloronordiazepam metabolites in the dog. J Med Chem 22(4):436–440.
https://doi.org/10.1021/jm00190a016
Williams TD et al (2009) Hepatic transcriptomic and metabolomic responses in the stickleback
(Gasterosteus aculeatus) exposed to environmentally relevant concentrations of
dibenzanthracene. Environ Sci Technol 43(16):6341–6348. https://doi.org/10.1021/es9008689
Wilson DM, Burlingame AL (1974) Deuterium and carbon-13 tracer studies of ethanol metabolism
in the rat by 2H, 1H-decoupled 13C nuclear magnetic resonance. Biochem Biophys Res
Commun 56(3):828–835. https://doi.org/10.1016/0006-291X(74)90680-9
Wishart DS (2005) Metabolomics: the principles and potential applications to transplantation. Am J
Transplant 5(12):2814–2820. https://doi.org/10.1111/j.1600-6143.2005.01119.x
Wishart DS (2008a) Applications of metabolomics in drug discovery and development. Drugs R D
9(5):307–322. https://doi.org/10.2165/00126839-200809050-00002
Wishart DS (2008b) Metabolomics: applications to food science and nutrition research. Trends
Food Sci Technol 19(9):482–493. https://doi.org/10.1016/j.tifs.2008.03.003
Wishart DS (2015) Is cancer a genetic disease or a metabolic disease? EBioMedicine 2(6):478–479.
https://doi.org/10.1016/j.ebiom.2015.05.022
Wishart D (2016) Emerging applications of metabolomics in drug discovery and precision medi-
cine. Nat Rev Drug Discov 15(7):473–484. https://doi.org/10.1038/nrd.2016.32
Wishart DS et al (1995) 1H, 13C and 15N chemical shift referencing in biomolecular NMR. J
Biomol NMR 6(2):135–140. https://doi.org/10.1007/BF00211777
Wishart DS et al (2001) Magnetic resonance diagnostics: a new technology for high-throughput
clinical diagnostics. Clin Chem 47(10):1918–1921
Wishart DS et al (2007) HMDB: the human metabolome database. Nucleic Acids Res 35(Suppl 1):
D521–D526. https://doi.org/10.1093/nar/gkl923
Wishart DS et al (2008) The human cerebrospinal fluid metabolome. J Chromatogr B Analyt
Technol Biomed Life Sci 871(2):164–173. https://doi.org/10.1016/j.jchromb.2008.05.001
Wishart DS et al (2018) HMDB 4.0: the human metabolome database for 2018. Nucleic Acids Res
46(D1):D608–D617. https://doi.org/10.1093/nar/gkx1089
Wolak J et al (2012) Noninvasive fluxomics in mammals by nuclear magnetic resonance
spectroscopy. In: Fan TW-M, Lane A, Higashi R (eds) The handbook of metabolomics.
Methods in pharmacology and toxicology. Humana Press, Totowa, pp 321–392. https://doi.
org/10.1007/978-1-61779-618-0_11
Worley B, Powers R (2014) MVAPACK: a complete data handling package for NMR
metabolomics. ACS Chem Biol 9(5):1138–1144. https://doi.org/10.1021/cb4008937
Wu J et al (2016) NMR analysis of the CSF and plasma metabolome of rigorously matched
amyotrophic lateral sclerosis, Parkinson’s disease and control subjects. Metabolomics 12(6):
101. https://doi.org/10.1007/s11306-016-1041-6
Wu K et al (2021) Improvement in signal-to-noise ratio of liquid-state NMR spectroscopy via a
deep neural network DN-Unet. Anal Chem 93(3):1377–1382. https://doi.org/10.1021/acs.
analchem.0c03087
Practical Aspects of NMR-Based Metabolomics 41
Contents
1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
2 Step 1: Study Design: Define Compound Identification Goals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
3 Step 2: Acquire Data – Experimental Strategies to Identify Compounds
in Metabolomics Data . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
4 Step 3: Computational Strategies for Data Cleaning and Feature Annotation . . . . . . . . . . . . . . 54
5 Step 4: MS/MS Libraries and Compound Identification Using Library Search . . . . . . . . . . . . 57
6 Step 5: Assess and Report Identification Confidence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
7 Conclusion: Pharmacology-Focused Compound Identification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
Abstract
The metabolome is composed of a vast array of molecules, including endogenous
metabolites and lipids, diet- and microbiome-derived substances,
pharmaceuticals and supplements, and exposome chemicals. Correct identifica-
tion of compounds from this diversity of classes is essential to derive biologically
relevant insights from metabolomics data. In this chapter, we aim to provide a
practical overview of compound identification strategies for mass spectrometry-
based metabolomics, with a particular eye toward pharmacologically-relevant
studies. First, we describe routine compound identification strategies applicable
to targeted metabolomics. Next, we discuss both experimental (data
Keywords
Compound identification · Identification confidence · LC-MS · Metabolomics ·
Molecular formula assignment · MS/MS search
1 Introduction
The first step of any metabolomics experiment is to define its goals; this also applies
to compound identification. Is the project intended to perform routine identification
and quantitation of a moderate number of well-known metabolic intermediates or a
46 R. Hissong et al.
drug and its known metabolites in human urine? Or, is it to detect and identify
biomarkers of a disease in human plasma, including unknown compounds? Or, to
determine if a particular metabolite profile predicts responsiveness to a treatment?
All these scenarios, and many other uses of metabolomics, rely on robust compound
identification. However, the way compound identification is best achieved is likely
to differ depending on study design. One of the first choices that must be made is
whether the metabolomics workflow to be used should be targeted, untargeted, or
use a combination of both approaches. The distinctions between targeted and
untargeted metabolomics are described in other chapters of this book and have
been reviewed extensively (Patti et al. 2012; Cajka and Fiehn 2016; Fiehn 2016a;
Schrimpe-Rutledge et al. 2016); Saigusa et al. describe advantages and
disadvantages of targeted and untargeted methods in the context of
pharmacometabolomics and drug discovery (Saigusa et al. 2021). Here, we briefly
consider commonalities and distinctions of compound identification in targeted and
untargeted metabolomics.
Once a study design has been selected and goals for compound identification have
been established, the next step is to perform experimental analysis. Here, we
describe major steps of a metabolomics experiment relevant to compound identifi-
cation, pointing out specific strategies that can be employed to improve the number
and quality of identifications that can be obtained from the data.
chloride and benzoyl chloride react quantitatively with amines, improving ionization
efficiency, chromatographic retention, and MS/MS fragmentation (Guo and Li 2009;
Wong et al. 2016). The compound 3-nitrophenylhydrazine can be used to derivatize
fatty acids, other carboxylic acids, and phosphoryl metabolites (Meng et al. 2021).
Using a combination of derivatization techniques has been proposed as a strategy to
facilitate characterization of the chemical composition of unknowns by revealing the
presence of key functional groups and allowing focused analysis of these subsets of
the metabolome (Zhao and Li 2020). Derivatization also has potential disadvantages:
(1) derivatization of unknowns may not necessarily result in easier characterization
of their structure, and (2) derivatization reactions may not always proceed to
completion, resulting in a mixture of partially derivatized molecules and hence a
more complex sample to analyze, and (3) some metabolites may not derivatize at all.
Stable Isotopes Compounds enriched with one or more stable isotopes of common
elements (e.g., 2H,13C, 15N, or 18O) can be easily distinguished from their unlabeled
counterparts by mass spectrometry, while their chemical properties remain essen-
tially identical to the unlabeled compound. In addition to their applications as
internal standards and as tracers to help quantitate metabolite flux, stable isotopes
can also be used to aid compound identification in metabolomics. One approach
using stable isotopes is termed isotope ratio outlier analysis (IROA), which uses
paired samples isotopically labeled with either 5% or 95% 13C and uses characteris-
tic isotope patterns to help differentiate biological signals from artifacts and con-
strain potential molecular formulas assigned to unknowns (Clendinen et al. 2015).
Another application of stable isotopes is to investigate the metabolic fate of individ-
ual metabolites or drugs (Kempa, “Advancements in pulsed stable isotope re-solved.
Metabolomics”; Günther, “Metabolomics in cell biology”). In one example, Chen
et al. injected mice with 400 mg/kg of [acetyl-2H3]- or [2,3,5,6-2H4]-acetaminophen
and used untargeted feature detection, followed by MS/MS-based structural elucida-
tion, to identify three novel urinary acetaminophen metabolites potentially
associated with its toxicity in overdose (Beyoğlu et al. 2018). Another potential
use of stable isotopes is the use of hydrogen-deuterium exchange to help determine
the chemical structure of unknown compounds (Majuta et al. 2019).
principal advantage of higher mass accuracy is that it reduces the number of possible
molecular formulas that can be assigned to a feature; although, as demonstrated by
Fiehn et al. even 0.1 ppm mass accuracy is not sufficient to unambiguously assign a
molecular formula for species <500 Da, much less for larger compounds (Kind and
Fiehn 2007). Spectral resolution does not directly determine the accuracy of molec-
ular formula determination. Instead, higher resolution reduces the possibility of
interference from co-eluting nearly isobaric species. The isotope distribution of an
unknown is another feature of MS data that aids compound identification; if
measured accurately, it is very effective in constraining molecular formula assign-
ment (Böcker et al. 2009).
influenced by the MS1 data. Details regarding these approaches and their advantages
and disadvantages have been described in detail elsewhere (Guo and Huan
2020a, b).
It is good practice to acquire MS/MS data during any substantial untargeted
metabolomics project. This allows both automated database searching and follow-
up analysis to attempt identification or classification of features of interest, without
returning to the instrument to acquire new data. In many cases, MS/MS data need not
be acquired for each individual sample, but can be generated using representative
pooled samples from the study. To acquire high-quality MS/MS spectra of as many
features as possible, recent data acquisition software packages enable automatic
generation of precursor ion exclusion lists that prevent serial LC–MS/MS runs of
the same sample from acquiring MS/MS data on ions already fragmented in previous
runs. This method, sometimes termed iterative DDA, is a powerful strategy to obtain
deeper MS/MS coverage of detected features than is possible in a standard DDA
workflow (Anderson et al. 2021; Koelmel et al. 2017).
Certain mass analyzers are capable of multiple stages of fragmentation, in which
fragment ions are further fragmented. This is termed MSn analysis; it enables
construction of fragmentation trees for unknown compounds (Vaniya and Fiehn
2015). MSn data are less well cataloged and interpretation of MSn data is less widely
supported by databases and software tools compared to MS/MS data. Nevertheless,
under many circumstances MSn data provide useful information to characterize and
annotate unknown compounds (Vinaixa et al. 2016; Ridder et al. 2012).
and detected. More complex ion formation processes are frequently observed,
including multiply charged ions, ions with multiple charge carriers, solvent adducts,
adducted fragment ions, and dimers, multimers and heterodimers. (Mahieu and Patti
2017; Mahieu et al. 2016; Kachman et al. 2019; Nelson et al. 2022) Collectively,
such features can be termed “degenerate” signals.
To avoid investing effort to identify features that are degenerate signals produced
by other already-identified compounds, data clean-up steps including adduct and
fragment annotation and/or removal are necessary. For GC-EI-MS, spectral
deconvolution software such as AMDIS (Davies 1998) or MS-DIAL (Tsugawa
et al. 2015) can help separate spectra of features that are only partially resolved by
chromatography. For LC–MS, the process of adduct annotation can be at least
partially automated by either instrument vendor or open-source data analysis soft-
ware such as CAMERA, MZmine, or MS-DIAL (Tsugawa et al. 2015; Kuhl et al.
2012; Pluskal et al. 2010). The most rigorous approaches for degeneracy annotation
also consider correlation of intensity for co-eluting features; those with high correla-
tion are more likely to represent degenerate features (Kachman et al. 2019;
Broeckling et al. 2014). Application of these clean-up steps reduces compound
identification workload by decreasing the number of features that must be subjected
to additional computational analysis and/or manual review.
Table 1 Selected small molecule compound databases. Table adapted from Blaženović et al.
(2018)
Free web
Additional compound ID-relevant access/free
Database Contents features download?
PubChem (Bolton All small Structure similarity search tool Y/Y
et al. 2008) molecules
ChemSpider All small Y/N
(Pence and molecules
Williams 2010)
ChEBI (Hastings Small Focus on compounds of biological Y/ Y
et al. 2016) molecules interest
KEGG (Kanehisa Metabolites Curated pathway maps Y/N
et al. 2006)
MetaCyc (Caspi Metabolites Curated pathway maps Y/
et al. 2008) noncommercial
HMDB (Wishart Human Physiological concentration data, Y/Y
et al. 2022) metabolites/ MS/MS spectra, text-mined literature
exposome context
Metlin (Guijas Metabolites Y (MS1 only)/
et al. 2018) N
RefMet (Fahy and Metabolites Name conversion tool Y/Y
Subramaniam
2020)
ChEMBL (Davies Bioactive Y/Y
et al. 2015) drug-like
molecules
DrugBank Known drugs Y/
(Wishart et al. noncommercial
2006)
Compound Databases Once degeneracy removal has been performed, and prefer-
ably after a molecular formula has been assigned, a feature’s m/z or neutral mass can
be searched against a compound database. Although m/z alone is by no means
sufficient to identify an unknown metabolite, databases help provide a list of
candidate compounds that can be evaluated using other means. Small molecule
compound databases vary in their scope and application; major examples are listed
in Table 1.
Databases range from those which focus on known endogenous metabolites to
those which attempt to cover all known and plausible small molecule chemical
structures. It is desirable to begin by querying the most specific applicable database
first. Small molecule databases focused on pharmaceutical compounds, such as
DrugBank (Wishart et al. 2018) and ChEMBL (Papadatos and Overington 2014;
Mendez et al. 2019), are of particular use for putative annotation of drugs and their
metabolites in pharmacometabolomics data. Likewise, using organism-specific
databases or constraining taxonomy to the sample type being analyzed will generate
Compound Identification Strategies in Mass Spectrometry-Based. . . 57
Table 2 Widely used spectral libraries. Table adapted from Blaženović et al. (2018)
Free web
Additional compound access/free
Library Data type Type ID-relevant features download?
NIST EI-MS, Experimental Highly curated, includes N/N
(Stein CID-MS/ search software, available from
1999) MS multiple vendors
Wiley EI-MS, Experimental Largest collection of EI-MS N/N
(Solutions CID-MS/ data, available from multiple
2022) MS vendors
METLIN CID-MS/ Experimental Developed for Q-ToF N/N
(Guijas MS instruments, licensed annually
et al. 2018)
MoNA EI, Experimental, Community database, Y/Y
(Fiehn MS/MS, user- automated curation
2016b) MSn contributed
MassBank Metabolites Experimental, Community database Y/Y
(Horai et al. user-
2010) contributed
mzCloud MS/MS, Experimental Most complete MSn database Y/N
(LLC, H MSn
2022)
GNPS MS/MS Experimental, Integrated with molecular Y/Y
(Wang user- networking tools
et al. 2016) contributed
LipidBlast Bioactive Computational Fully computational Y/Y
(Kind et al. drug-like lipidomics database
2013) molecules
hits may be retained for user review. Many different scoring functions exist; among
these, probability-based matching (PBM) was one of the first introduced scoring
functions for EI-MS spectra (Stauffer et al. 1985), and the classic “dot-product” and
“reverse dot-product” scoring algorithms have been widely used for MS/MS
searching (Stein and Scott 1994). Many other scoring functions have also been
evaluated, including a recent spectral entropy-based algorithm that demonstrated
superior performance to dot-product scores (Li et al. 2021). Additional constraints,
such as a narrow mass window for precursor ion match and a precursor ion isotope
pattern match, can serve to further shorten a list of candidates or even select a single
probable compound identity.
Many software tools exist to automate spectral library search. Most instrument
manufacturers incorporate library search tools into their data analysis software; some
also provide access to proprietary libraries searchable only using these tools. Among
cross-platform tools, NIST MS Search is one of the best known (Stein 1999); it
allows searching individual spectra against the NIST library and other user-loaded
libraries with visualization of spectral matches using head-to-tail or difference plots.
To enable much more rapid search of many spectra, a useful alternative is
MSPepSearch (Zhang et al. 2018). Originally designed for peptide spectra, it has
been adopted and extensively used for small molecule analysis; it uses a similar
scoring algorithm but generates output in tabular form. Progenesis QI is a commer-
cial data analysis tool that enables cross-vendor MS/MS search in addition to general
data analysis. MS-DIAL is a free, open-source alternative that integrates feature
finding, alignment, and spectral search in a unified workflow (Tsugawa et al. 2015).
Other widely used untargeted metabolomics data analysis tools, including XCMS
(Smith et al. 2006) and MZmine (Pluskal et al. 2010), also have some MS/MS search
capabilities, though they are implemented in a less visual manner than in MS-DIAL.
SIRIUS (Böcker et al. 2009; Dührkop et al. 2019) and GNPS (Wang et al. 2016),
described later in this chapter, also use spectral search heavily in their workflow but
due to other features are classified separately from typical library search tools.
It is important to note that while spectral searching provides a fast and often
accurate means of assigning chemical structures to features in metabolomics data, it
does not provide an objective means to assess the probability that these
identifications are correct. Manual review of spectra, including matches beyond
the top hit, can sometimes help clarify ambiguous assignments and determine
appropriate score thresholds for identification. Well-defined strategies for assessing
and reporting identification confidence are important to data analysis and are
described in “Step 5” of our workflow.
Level 1:
IDENTIFIED Time (min)
Time (min)
Spectral
Level 2: Search Decoy
PUTATIVE ID m/z Score(s) Library search for
m/z
Feature data Feature data FDR
esmaon
Level 3: ID with
Manual COMPOUND RT
Predicon confidence
Review CLASS ID esmate
m/z Score
Time (min)
Level 4: E. coli
Spectral
UNKNOWN In silico ? ? ?
Search
Fragmentaon Biological
Dri me
Score(s)
Score Probability
Assessment
RT
Collisional Cross Secon Score
Fig. 2 Current typical current metabolite identification and reporting strategy compared to a
hypothetical multi-input method for improving identification accuracy and assessing and reporting
identification confidence
proposed and tested for small molecules, ranging from randomizing all peaks found
in a typical library to approaches based on rearrangement of fragmentation trees
(Scheubert et al. 2017; Wang et al. 2018; Alka et al. 2022). Decoy libraries have
been used to help select spectral search score thresholds and other criteria to achieve
a desired FDR. However, these attempts have demonstrated that decoy libraries are
not very helpful to assess FDR for some classes of metabolites, particularly for
compounds that produce very few fragment ions and those that have multiple
structural isomers that produce similar fragmentation spectra. Thus, FDR estimation
strategies for metabolomics need further refinement and will certainly require sup-
plementation with strategies beyond decoy library searching before compound
identification in metabolomics can achieve the level of automation and accuracy
enjoyed by other omics sciences.
7 Conclusion: Pharmacology-Focused
Compound Identification
used. Researchers should also consider biological factors that may affect both
compound identification and quantitation, such as sex-specific responses to drug
metabolism (Soldin et al. 2011; Chary et al. 2022). Finally, the application of
metabolomics to pharmacological studies is still a developing area of study. While
at present most of the data relevant to compound identification in metabolomics is
found in the analytical chemistry and bioinformatics literature, resources specific to
the challenges of pharmacology can be expected to develop and strengthen over
future years.
References
Abdelhameed A et al (2014) Pseudo-MS3 approach using electrospray mass spectrometry
(ESI-MS/MS) to characterize certain (2E)-2-[3-(1H-Imidazol-1-yl)-1-phenylpropylidene]
hydrazinecarboxamide derivatives. J Chem 10 p. https://doi.org/10.1155/2014/386301
Alka O et al (2022) DIAMetAlyzer allows automated false-discovery rate-controlled analysis for
data-independent acquisition in metabolomics. Nat Commun 13:1347. https://doi.org/10.1038/
s41467-022-29006-z
Alley WR, Mechref Y, Novotny MV (2009) Characterization of glycopeptides by combining
collision-induced dissociation and electron-transfer dissociation mass spectrometry data.
Rapid Commun Mass Spectrom 23:161–170. https://doi.org/10.1002/rcm.3850
Anderson BG, Raskind A, Habra H, Kennedy RT, Evans CR (2021) Modifying chromatography
conditions for improved unknown feature identification in untargeted metabolomics. Anal
Chem 93:15840–15849. https://doi.org/10.1021/acs.analchem.1c02149
Baba T, Campbell J, Le Blanc Y, Baker P, Ikeda K (2018) Quantitative structural multi-class
lipidomics using differential mobility-electron impact excitation of ions from organics (EIEIO)
mass spectrometry. J Lipid Res 59:jlr.D083261. https://doi.org/10.1194/jlr.D083261
Balogh MP (2004) Debating resolution and mass accuracy in mass spectrometry. Spectroscopy
(Santa Monica) 19:34–39
Beyoğlu D, Zhou Y, Chen C, Idle JR (2018) Mass isotopomer-guided decluttering of metabolomic
data to visualize endogenous biomarkers of drug toxicity. Biochem Pharmacol 156:491–500.
https://doi.org/10.1016/j.bcp.2018.09.022
Blaženović I, Kind T, Ji J, Fiehn O (2018) Software tools and approaches for compound identifica-
tion of LC-MS/MS data in metabolomics. Metabolites 8. https://doi.org/10.3390/
metabo8020031
Böcker S, Letzel MC, Lipták Z, Pervukhin A (2009) SIRIUS: decomposing isotope patterns for
metabolite identification. Bioinformatics 25:218–224
Bolton EE, Wang Y, Thiessen PA, Bryant SH (2008) Annual reports in computational chemistry,
vol 4. Elsevier, pp 217–241
Bonini P, Kind T, Tsugawa H, Barupal DK, Fiehn O (2020) Retip: retention time prediction for
compound annotation in untargeted metabolomics. Anal Chem 92:7515–7522. https://doi.org/
10.1021/acs.analchem.9b05765
Borges RM et al (2021) Quantum chemistry calculations for metabolomics. Chem Rev 121:5633–
5670. https://doi.org/10.1021/acs.chemrev.0c00901
Broeckling CD, Afsar FA, Neumann S, Ben-Hur A, Prenni JE (2014) RAMClust: a novel feature
clustering method enables spectral-matching-based annotation for metabolomics data. Anal
Chem 86:6812–6817. https://doi.org/10.1021/ac501530d
Bruce SJ et al (2009) Investigation of human blood plasma sample preparation for performing
metabolomics using ultrahigh performance liquid chromatography/mass spectrometry. Anal
Chem 81:3285–3296. https://doi.org/10.1021/ac8024569
Compound Identification Strategies in Mass Spectrometry-Based. . . 65
Cajka T, Fiehn O (2016) Toward merging untargeted and targeted methods in mass spectrometry-
based metabolomics and Lipidomics. Anal Chem 88:524–545. https://doi.org/10.1021/acs.
analchem.5b04491
Caspi R et al (2008) The MetaCyc database of metabolic pathways and enzymes and the BioCyc
collection of pathway/genome databases. Nucleic Acids Res 36:D623–D631. https://doi.org/10.
1093/nar/gkm900
Chaleckis R, Meister I, Zhang P, Wheelock CE (2019) Challenges, progress and promises of
metabolite annotation for LC-MS-based metabolomics. Curr Opin Biotechnol 55:44–50.
https://doi.org/10.1016/j.copbio.2018.07.010
Chary S, Amrein K, Mahmoud SH, Lasky-Su JA, Christopher KB (2022) Sex-specific catabolic
metabolism alterations in the critically ill following high dose vitamin D. Metabolites 12. https://
doi.org/10.3390/metabo12030207
Clendinen CS et al (2015) An overview of methods using 13C for improved compound identifica-
tion in metabolomics and natural products. Front Plant Sci 6. https://doi.org/10.3389/fpls.2015.
00611
Colby SM et al (2019) ISiCLE: a quantum chemistry pipeline for establishing in silico collision
cross section libraries. Anal Chem 91:4346–4356. https://doi.org/10.1021/acs.analchem.
8b04567
Contrepois K et al (2018) Cross-platform comparison of untargeted and targeted Lipidomics
approaches on aging mouse plasma. Sci Rep 8:17747. https://doi.org/10.1038/s41598-018-
35807-4
Cooper BT et al (2019) Hybrid search: a method for identifying metabolites absent from tandem
mass spectrometry libraries. Anal Chem 91:13924–13932. https://doi.org/10.1021/acs.
analchem.9b03415
Davies AN (1998) The new Automated Mass Spectrometry Deconvolution and Identification
System (AMDIS). Spectrosc Eur 10(3):22–26
Davies M et al (2015) ChEMBL web services: streamlining access to drug discovery data and
utilities. Nucleic Acids Res 43:W612–W620. https://doi.org/10.1093/nar/gkv352
Dona AC et al (2016) A guide to the identification of metabolites in NMR-based metabonomics/
metabolomics experiments. Comput Struct Biotechnol J 14:135–153. https://doi.org/10.1016/j.
csbj.2016.02.005
Ducati AO et al (2021) Improved metabolite characterization by liquid chromatography – Tandem
mass spectrometry through electron impact type fragments from adduct ions. Anal Chim Acta
1150:338207. https://doi.org/10.1016/j.aca.2021.338207
Dührkop K, Shen H, Meusel M, Rousu J, Böcker S (2015) Searching molecular structure databases
with tandem mass spectra using CSI:FingerID. Proc Natl Acad Sci 112:12580–12585. https://
doi.org/10.1073/pnas.1509788112
Dührkop K et al (2019) SIRIUS 4: a rapid tool for turning tandem mass spectra into metabolite
structure information. Nat Methods 16:299–302. https://doi.org/10.1038/s41592-019-0344-8
Elias JE, Gygi SP (2010) Target-decoy search strategy for mass spectrometry-based proteomics.
Methods Mol Biol 604:55–71. https://doi.org/10.1007/978-1-60761-444-9_5
Emwas A-H et al (2019) NMR spectroscopy for metabolomics research. Metabolites 9. https://doi.
org/10.3390/metabo9070123
Fahy E, Subramaniam S (2020) RefMet: a reference nomenclature for metabolomics. Nat Methods
17:1173–1174. https://doi.org/10.1038/s41592-020-01009-y
Fiehn O (2016a) Metabolomics by gas chromatography-mass spectrometry: combined targeted and
untargeted profiling. Curr Protoc Mol Biol 114:30.34.31–30.34.32. https://doi.org/10.1002/
0471142727.mb3004s114
Fiehn O (2016b) Mass resolution and resolving power. https://fiehnlab.ucdavis.edu/projects/seven-
golden-rules/mass-resolution
Gowda GAN, Djukovic D (2014) Overview of mass spectrometry-based metabolomics:
opportunities and challenges. Methods Mol Biol 1198:3–12. https://doi.org/10.1007/978-1-
4939-1258-2_1
66 R. Hissong et al.
Griffiths WJ et al (2010) Targeted metabolomics for biomarker discovery. Angew Chem Int Ed 49:
5426–5445. https://doi.org/10.1002/anie.200905579
Guijas C et al (2018) METLIN: a technology platform for identifying knowns and unknowns. Anal
Chem 90:3156–3164. https://doi.org/10.1021/acs.analchem.7b04424
Guntner AS, Thalhamer B, Klampfl C, Buchberger W (2019) Collision cross sections obtained with
ion mobility mass spectrometry as new descriptor to predict blood-brain barrier permeation by
drugs. Sci Rep 9:19182. https://doi.org/10.1038/s41598-019-55856-7
Guo J, Huan T (2020a) Comparison of full-scan, data-dependent, and data-independent acquisition
modes in liquid chromatography–mass spectrometry based untargeted metabolomics. Anal
Chem 92:8072–8080. https://doi.org/10.1021/acs.analchem.9b05135
Guo J, Huan T (2020b) Evaluation of significant features discovered from different data acquisition
modes in mass spectrometry-based untargeted metabolomics. Anal Chim Acta 1137:37–46.
https://doi.org/10.1016/j.aca.2020.08.065
Guo K, Li L (2009) Differential (12)C/(13)C-isotope dansylation labeling and fast liquid chroma-
tography/mass spectrometry for absolute and relative quantification of the metabolome. Anal
Chem 81:3919–3932. https://doi.org/10.1021/ac900166a
Habra H et al (2021) metabCombiner: paired untargeted LC-HRMS metabolomics feature matching
and concatenation of disparately acquired data sets. Anal Chem 93:5028–5036. https://doi.org/
10.1021/acs.analchem.0c03693
Halket JM et al (2005) Chemical derivatization and mass spectral libraries in metabolic profiling by
GC/MS and LC/MS/MS. J Exp Bot 56:219–243. https://doi.org/10.1093/jxb/eri069
Han J, Lin K, Sequeira C, Borchers CH (2015) An isotope-labeled chemical derivatization method
for the quantitation of short-chain fatty acids in human feces by liquid chromatography-tandem
mass spectrometry. Anal Chim Acta 854:86–94. https://doi.org/10.1016/j.aca.2014.11.015
Hastings J et al (2016) ChEBI in 2016: improved services and an expanding collection of
metabolites. Nucleic Acids Res 44:D1214–D1219. https://doi.org/10.1093/nar/gkv1031
Health in Northern Ireland (2022) National Center for Advancing Translational Sciences Com-
pound Management. https://ncats.nih.gov/preclinical/core/compound.
Hines KM, Ross DH, Davidson KL, Bush MF, Xu L (2017) Large-scale structural characterization
of drug and drug-like compounds by high-throughput ion mobility-mass spectrometry. Anal
Chem 89:9023–9030. https://doi.org/10.1021/acs.analchem.7b01709
Horai H et al (2010) MassBank: a public repository for sharing mass spectral data for life sciences. J
Mass Spectrom 45:703–714. https://doi.org/10.1002/jms.1777
Ichou F et al (2014) Comparison of the activation time effects and the internal energy distributions
for the CID, PQD and HCD excitation modes. J Mass Spectrom 49:498–508. https://doi.org/10.
1002/jms.3365
Kachman M et al (2019) Deep annotation of untargeted LC-MS metabolomics data with Binner.
Bioinformatics 36:1801–1806. https://doi.org/10.1093/bioinformatics/btz798
Kanehisa M et al (2006) From genomics to chemical genomics: new developments in KEGG.
Nucleic Acids Res 34:D354–D357
Kantae V et al (2017) Integration of pharmacometabolomics with pharmacokinetics and pharma-
codynamics: towards personalized drug therapy. Metabolomics 13. https://doi.org/10.1007/
s11306-016-1143-1
Kanu AB, Dwivedi P, Tam M, Matz L, Hill HH (2008) Ion mobility-mass spectrometry. J Mass
Spectrom 43:1–22. https://doi.org/10.1002/jms.1383
Kind T, Fiehn O (2007) Seven golden rules for heuristic filtering of molecular formulas obtained by
accurate mass spectrometry. BMC Bioinformatics 8:105. https://doi.org/10.1186/1471-2105-
8-105
Kind T, Fiehn O (2010) Advances in structure elucidation of small molecules using mass spec-
trometry. Bioanal Rev 2:23–60. https://doi.org/10.1007/s12566-010-0015-9
Kind T et al (2009) FiehnLib: mass spectral and retention index libraries for metabolomics based on
quadrupole and time-of-flight gas chromatography/mass spectrometry. Anal Chem 81:10038–
10048. https://doi.org/10.1021/ac9019522
Compound Identification Strategies in Mass Spectrometry-Based. . . 67
Kind T et al (2013) LipidBlast in silico tandem mass spectrometry database for lipid identification.
Nat Methods 10:755–758. https://doi.org/10.1038/nmeth.2551
Koek MM, Muilwijk B, van Stee LL, Hankemeier T (2008) Higher mass loadability in comprehen-
sive two-dimensional gas chromatography-mass spectrometry for improved analytical perfor-
mance in metabolomics analysis. J Chromatogr A 1186:420–429. https://doi.org/10.1016/j.
chroma.2007.11.107
Koelmel JP et al (2017) Expanding lipidome coverage using LC-MS/MS data-dependent acquisi-
tion with automated exclusion list generation. J Am Soc Mass Spectrom 28:908–917
Kuhl C, Tautenhahn R, Böttcher C, Larson TR, Neumann S (2012) CAMERA: an integrated
strategy for compound spectra extraction and annotation of LC/MS data sets. Anal Chem 84:
283–289. https://doi.org/10.1021/ac202450g
Kvitvang HFN, Andreassen T, Adam T, Villas-Bôas SG, Bruheim P (2011) Highly sensitive
GC/MS/MS method for quantitation of amino and nonamino organic acids. Anal Chem 83:
2705–2711. https://doi.org/10.1021/ac103245b
Lai Z, Fiehn O (2018) Mass spectral fragmentation of trimethylsilylated small molecules. Mass
Spectrom Rev 37:245–257. https://doi.org/10.1002/mas.21518
Lee HB, Peart TE, Svoboda ML (2005) Determination of endocrine-disrupting phenols, acidic
pharmaceuticals, and personal-care products in sewage by solid-phase extraction and gas
chromatography-mass spectrometry. J Chromatogr A 1094:122–129. https://doi.org/10.1016/j.
chroma.2005.07.070
Lenz EM et al (2007) HPLC-NMR with severe column overloading: fast-track metabolite identifi-
cation in urine and bile samples from rat and dog treated with [14C]-ZD6126. J Pharm Biomed
Anal 43:1065–1077. https://doi.org/10.1016/j.jpba.2006.09.010
Li Y et al (2021) Spectral entropy outperforms MS/MS dot product similarity for small-molecule
compound identification. Nat Methods 18:1524–1531. https://doi.org/10.1038/s41592-021-
01331-z
Lisec J, Schauer N, Kopka J, Willmitzer L, Fernie AR (2006) Gas chromatography mass
spectrometry-based metabolite profiling in plants. Nat Protoc 1:387–396. https://doi.org/10.
1038/nprot.2006.59
LLC, H (2022) mzCloud advanced mass spectral database. https://www.mzcloud.org/
Lorenz MA, Burant CF, Kennedy RT (2011) Reducing time and increasing sensitivity in sample
preparation for adherent mammalian cell metabolomics. Anal Chem 83:3406–3414. https://doi.
org/10.1021/ac103313x
Lu W, Bennett BD, Rabinowitz JD (2008) Analytical strategies for LC-MS-based targeted
metabolomics. J Chromatogr B Analyt Technol Biomed Life Sci 871:236–242. https://doi.
org/10.1016/j.jchromb.2008.04.031
Ludwig M et al (2019) ZODIAC: database-independent molecular formula annotation using Gibbs
sampling reveals unknown small molecules
Mahieu NG, Patti GJ (2017) Systems-level annotation of a metabolomics data set reduces 25,000
features to fewer than 1000 unique metabolites. Anal Chem 89:10397–10406. https://doi.org/
10.1021/acs.analchem.7b02380
Mahieu NG, Spalding JL, Gelman SJ, Patti GJ (2016) Defining and detecting complex peak
relationships in mass spectral data: the Mz.Unity algorithm. Anal Chem 88:9037–9046.
https://doi.org/10.1021/acs.analchem.6b01702
Majuta SN et al (2019) Rapid solution-phase hydrogen/deuterium exchange for metabolite com-
pound identification. J Am Soc Mass Spectrom 30:1102–1114. https://doi.org/10.1007/s13361-
019-02163-0
Matyash V, Liebisch G, Kurzchalia TV, Shevchenko A, Schwudke D (2008) Lipid extraction by
methyl-tert-butyl ether for high-throughput lipidomics. J Lipid Res 49:1137–1146. https://doi.
org/10.1194/jlr.D700041-JLR200
Maurer HH (1998) Liquid chromatography mass spectrometry in forensic and clinical toxicology. J
Chromatogr B 713:3–25. https://doi.org/10.1016/s0378-4347(97)00514-8
68 R. Hissong et al.
McCann MR, George De la Rosa MV, Rosania GR, Stringer KA (2021) L-carnitine and
acylcarnitines: mitochondrial biomarkers for precision medicine. Metabolites 11. https://doi.
org/10.3390/metabo11010051
Mendez D et al (2019) ChEMBL: towards direct deposition of bioassay data. Nucleic Acids Res 47:
D930–d940. https://doi.org/10.1093/nar/gky1075
Meng X et al (2021) Simultaneous 3-nitrophenylhydrazine derivatization strategy of carbonyl,
carboxyl and phosphoryl submetabolome for LC-MS/MS-based targeted metabolomics with
improved sensitivity and coverage. Anal Chem 93:10075–10083. https://doi.org/10.1021/acs.
analchem.1c00767
Misra BB, Olivier M (2020) High resolution GC-Orbitrap-MS metabolomics using both electron
ionization and chemical ionization for analysis of human plasma. J Proteome Res 19:2717–
2731. https://doi.org/10.1021/acs.jproteome.9b00774
Nelson AB, Chow LS, Hughey CC, Crawford PA, Puchalska P (2022) Artifactual fatty acid dimers
mimic FAHFA signals in untargeted metabolomics pipelines. J Lipid Res 100201. https://doi.
org/10.1016/j.jlr.2022.100201
Papadatos G, Overington JP (2014) The ChEMBL database: a taster for medicinal chemists. Future
Med Chem 6:361–364. https://doi.org/10.4155/fmc.14.8
Parker CE et al (2014) In: Tang NLS, Poon T (eds) Chemical diagnostics: from bench to bedside
vol. 336 topics in current chemistry-series. pp 117–137
Patti GJ, Yanes O, Siuzdak G (2012) Metabolomics: the apogee of the omics trilogy. Nat Rev Mol
Cell Biol 13:263–269. https://doi.org/10.1038/nrm3314
Pence H, Williams A (2010) ChemSpider: an online chemical information resource. J Chem Educ
87:1123–1124. https://doi.org/10.1021/ED100697W
Plante P-L et al (2019) Predicting ion mobility collision cross-sections using a deep neural network:
DeepCCS. Anal Chem 91:5191–5199. https://doi.org/10.1021/acs.analchem.8b05821
Pluskal T, Castillo S, Villar-Briones A, Oresic M (2010) MZmine 2: modular framework for
processing, visualizing, and analyzing mass spectrometry-based molecular profile data. BMC
Bioinformatics 11:395. https://doi.org/10.1186/1471-2105-11-395
Prakash C, Shaffer CL, Nedderman A (2007) Analytical strategies for identifying drug metabolites.
Mass Spectrom Rev 26:340–369. https://doi.org/10.1002/mas.20128
Rainville PD et al (2017) Ion mobility spectrometry combined with ultra performance liquid
chromatography/mass spectrometry for metabolic phenotyping of urine: effects of column
length, gradient duration and ion mobility spectrometry on metabolite detection. Anal Chim
Acta 982:1–8. https://doi.org/10.1016/j.aca.2017.06.020
Ridder L et al (2012) Substructure-based annotation of high-resolution multistage MSn spectral
trees. Rapid Commun Mass Spectrom 26:2461–2471. https://doi.org/10.1002/rcm.6364
Roberts LD, Souza AL, Gerszten RE, Clish CB (2012) Targeted metabolomics. Curr Protoc Mol
Biol. Chapter 30, Unit 30.32.31-24. https://doi.org/10.1002/0471142727.mb3002s98
Ruttkies C, Schymanski EL, Wolf S, Hollender J, Neumann S (2016) MetFrag relaunched:
incorporating strategies beyond in silico fragmentation. J Chem 8:3. https://doi.org/10.1186/
s13321-016-0115-9
Saigusa D, Matsukawa N, Hishinuma E, Koshiba S (2021) Identification of biomarkers to diagnose
diseases and find adverse drug reactions by metabolomics. Drug Metab Pharmacokinet 37:
100373. https://doi.org/10.1016/j.dmpk.2020.11.008
Scheubert K et al (2017) Significance estimation for large scale metabolomics annotations by
spectral matching. Nat Commun 8:1494. https://doi.org/10.1038/s41467-017-01318-5
Schrimpe-Rutledge AC, Codreanu SG, Sherrod SD, McLean JA (2016) Untargeted metabolomics
strategies – challenges and emerging directions. J Am Soc Mass Spectrom 27:1897–1905.
https://doi.org/10.1007/s13361-016-1469-y
Schymanski EL et al (2014) Identifying small molecules via high resolution mass spectrometry:
communicating confidence. Environ Sci Technol 48:2097–2098. https://doi.org/10.1021/
es5002105
Compound Identification Strategies in Mass Spectrometry-Based. . . 69
Scott DR (1992) Rapid and accurate method for estimating molecular weights of organic
compounds from low resolution mass spectra. Chemom Intel Lab Syst 16:193–202. https://
doi.org/10.1016/0169-7439(92)80037-5
Shackleton C, Pozo OJ, Marcos J (2018) GC/MS in recent years has defined the Normal and
clinically disordered Steroidome: will it soon be surpassed by LC/tandem MS in this role? J
Endocr Soc 2:974–996. https://doi.org/10.1210/js.2018-00135
Smith CA, Want EJ, O'Maille G, Abagyan R, Siuzdak G (2006) XCMS: processing mass spec-
trometry data for metabolite profiling using nonlinear peak alignment, matching, and identifica-
tion. Anal Chem 78:779–787. https://doi.org/10.1021/ac051437y
Soldin OP, Chung SH, Mattison DR (2011) Sex differences in drug disposition. J Biomed
Biotechnol 2011:187103. https://doi.org/10.1155/2011/187103
Solutions WS (2022) Wiley registry 12th edition/NIST 2020 mass spectral library. https://
sciencesolutions.wiley.com/solutions/technique/gc-ms/wiley-registry-12th-edition-nist-2020/
Stanstrup J, Neumann S, Vrhovšek U (2015) PredRet: prediction of retention time by direct
mapping between multiple chromatographic systems. Anal Chem 87:9421–9428. https://doi.
org/10.1021/acs.analchem.5b02287
Stauffer DB, McLafferty FW, Ellis RD, Peterson DW (1985) Probability-based-matching algorithm
with forward searching capabilities for matching unknown mass spectra of mixtures. Anal Chem
57:1056–1060. https://doi.org/10.1021/ac00283a021
Stein SE (1999) An integrated method for spectrum extraction and compound identification from
gas chromatography/mass spectrometry data. J Am Soc Mass Spectrom 10:770–781
Stein SE, Scott DR (1994) Optimization and testing of mass spectral library search algorithms for
compound identification. J Am Soc Mass Spectrom 5:859–866. https://doi.org/10.1016/1044-
0305(94)87009-8
Steuer AE, Brockbals L, Kraemer T (2019) Metabolomic strategies in biomarker research-new
approach for indirect identification of drug consumption and sample manipulation in clinical
and forensic toxicology? Front Chem 7:319. https://doi.org/10.3389/fchem.2019.00319
Strehmel N, Hummel J, Erban A, Strassburg K, Kopka J (2008) Retention index thresholds for
compound matching in GC-MS metabolite profiling. J Chromatogr B Analyt Technol Biomed
Life Sci 871:182–190. https://doi.org/10.1016/j.jchromb.2008.04.042
Sumner LW et al (2007) Proposed minimum reporting standards for chemical analysis chemical
analysis working group (CAWG) metabolomics standards initiative (MSI). Metabolomics 3:
211–221. https://doi.org/10.1007/s11306-007-0082-2
Taylor T (2015) Understanding electron ionization processes for GC–MS. LCGC N Am 33:290
Theodoridis GA, Gika HG, Want EJ, Wilson ID (2012) Liquid chromatography-mass spectrometry
based global metabolite profiling: a review. Anal Chim Acta 711:7–16. https://doi.org/10.1016/
j.aca.2011.09.042
Tiwary S et al (2019) High-quality MS/MS spectrum prediction for data-dependent and data-
independent acquisition data analysis. Nat Methods 16:519–525. https://doi.org/10.1038/
s41592-019-0427-6
Tolić N et al (2017) Formularity: software for automated formula assignment of natural and other
organic matter from ultrahigh-resolution mass spectra. Anal Chem 89:12659–12665. https://doi.
org/10.1021/acs.analchem.7b03318
Tsugawa H et al (2015) MS-DIAL: data-independent MS/MS deconvolution for comprehensive
metabolome analysis. Nat Methods 12:523–526. https://doi.org/10.1038/nmeth.3393
Tsugawa H et al (2016) Hydrogen rearrangement rules: computational MS/MS fragmentation and
structure elucidation using MS-FINDER software. Anal Chem 88:7946–7958. https://doi.org/
10.1021/acs.analchem.6b00770
van der Laan T et al (2021) Fractionation platform for target identification using off-line directed
two-dimensional chromatography, mass spectrometry and nuclear magnetic resonance. Anal
Chim Acta 1142:28–37. https://doi.org/10.1016/j.aca.2020.10.054
70 R. Hissong et al.
Vaniya A, Fiehn O (2015) Using fragmentation trees and mass spectral trees for identifying
unknown compounds in metabolomics. Trends Anal Chem 69:52–61. https://doi.org/10.1016/
j.trac.2015.04.002
Vinaixa M et al (2016) Mass spectral databases for LC/MS- and GC/MS-based metabolomics: state
of the field and future prospects. TrAC Trends Anal Chem 78:23–35. https://doi.org/10.1016/j.
trac.2015.09.005
Wang H et al (2015) Systematic optimization of long gradient chromatography mass spectrometry
for deep analysis of brain proteome. J Proteome Res 14:829–838. https://doi.org/10.1021/
pr500882h
Wang M et al (2016) Sharing and community curation of mass spectrometry data with global
natural products social molecular networking. Nat Biotechnol 34:828–837. https://doi.org/10.
1038/nbt.3597
Wang X et al (2018) Target-decoy-based false discovery rate estimation for large-scale metabolite
identification. J Proteome Res 17:2328–2334. https://doi.org/10.1021/acs.jproteome.8b00019
Wang S, Kind T, Tantillo DJ, Fiehn O (2020) Predicting in silico electron ionization mass spectra
using quantum chemistry. J Chem 12:63. https://doi.org/10.1186/s13321-020-00470-3
Watson DG (2013) A rough guide to metabolite identification using high resolution liquid chroma-
tography mass spectrometry in metabolomic profiling in metazoans. Comput Struct Biotechnol J
4:e201301005. https://doi.org/10.5936/csbj.201301005
Whiley L et al (2019) Systematic isolation and structure elucidation of urinary metabolites
optimized for the analytical-scale molecular profiling laboratory. Anal Chem 91:8873–8882.
https://doi.org/10.1021/acs.analchem.9b00241
Willoughby PH, Jansma MJ, Hoye TR (2014) A guide to small-molecule structure assignment
through computation of (1H and 13C) NMR chemical shifts. Nat Protoc 9:643–660. https://doi.
org/10.1038/nprot.2014.042
Wishart DS (2009) Computational strategies for metabolite identification in metabolomics.
Bioanalysis 1:1579–1596. https://doi.org/10.4155/bio.09.138
Wishart DS et al (2006) DrugBank: a comprehensive resource for in silico drug discovery and
exploration. Nucleic Acids Res 34:D668–D672. https://doi.org/10.1093/nar/gkj067
Wishart DS et al (2018) DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic
Acids Res 46:D1074–d1082. https://doi.org/10.1093/nar/gkx1037
Wishart DS et al (2022) HMDB 5.0: the human metabolome database for 2022. Nucleic Acids Res
50:D622–D631. https://doi.org/10.1093/nar/gkab1062
Wong JM et al (2016) Benzoyl chloride derivatization with liquid chromatography-mass spectrom-
etry for targeted metabolomics of neurochemicals in biological samples. J Chromatogr A 1446:
78–90. https://doi.org/10.1016/j.chroma.2016.04.006
Wu Q et al (2019) Enhancing coverage in LC-MS-based untargeted metabolomics by a new sample
preparation procedure using mixed-mode solid-phase extraction and two derivatizations. Anal
Bioanal Chem 411:6189–6202. https://doi.org/10.1007/s00216-019-02010-x
Xue J et al (2020) Enhanced in-source fragmentation annotation enables novel data independent
acquisition and autonomous METLIN molecular identification. Anal Chem 92:6051–6059.
https://doi.org/10.1021/acs.analchem.0c00409
Yoo HJ, Liu HC, Hakansson K (2007) Infrared multiphoton dissociation and electron-induced
dissociation as alternative MS/MS strategies for metabolite identification. Anal Chem 79:7858–
7866. https://doi.org/10.1021/ac071139w
Zaitsu K, Hayashi Y, Kusano M, Tsuchihashi H, Ishii A (2016) Application of metabolomics to
toxicology of drugs of abuse: a mini review of metabolomics approach to acute and chronic
toxicity studies. Drug Metab Pharmacokinet 31:21–26. https://doi.org/10.1016/j.dmpk.2015.
10.002
Zhang Z et al (2018) Reverse and random decoy methods for false discovery rate estimation in high
mass accuracy peptide spectral library searches. J Proteome Res 17:846–857. https://doi.org/10.
1021/acs.jproteome.7b00614
Compound Identification Strategies in Mass Spectrometry-Based. . . 71
Ryan T. McKay
Contents
1 Objectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 74
2 Brief History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 75
2.1 Potential . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
2.1.1 A Warning . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
2.2 Definitions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
2.2.1 Metabolome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
2.2.2 Metabo -Nomics or -Lomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
2.2.3 Metabolites . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
3 Expectations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
3.1 Reality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
3.1.1 Key Considerations for NMR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
4 NMR and MS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
4.1 Complementation: Can We Just Skip to the End? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
4.2 Metabolomics via Mass Spectrometry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 80
4.3 Metabolomics by NMR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81
5 Nuclear Magnetic Resonance Spectroscopy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81
5.1 NMR Experiment for Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81
5.1.1 First Impressions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
5.2 Liquids NMR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
5.2.1 Progression to Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
5.3 Samples . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
5.3.1 Preparation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
5.3.2 NMR Tubes, Spinners, and Side-Bands . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
5.3.3 Gathering and Handling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
5.3.4 In the Tube . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
5.4 NMR Spectrometer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90
5.4.1 Probe . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90
5.4.2 Console . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92
R. T. McKay (✉)
Department Chemistry, College of Natural and Applied Sciences, University of Alberta, Edmonton,
AB, Canada
e-mail: [email protected]
Abstract
The purpose of this manuscript will be to convince the reader to dive deeper into
NMR spectroscopy and prevent the technique from being just another “black-
box” in the lab. We will try to concisely highlight interesting topics and supply
additional references for further exploration at each stage. The advantages of
delving into the technique will be shown. The secondary objective, i.e., avoiding
common problems before starting, will hopefully then become clear. Lastly, we
will emphasize the spectrometer information needed for manuscript reporting to
allow reproduction of results and confirm findings.
Keywords
Automation · Biochemistry · Liquids · Metabolites · Metabolomics · NMR ·
Nuclear magnetic resonance · Quantitation · Small molecule · Solvent
suppression · Spectrometry
1 Objectives
The purpose of this manuscript will be to convince the reader to dive deeper into
NMR spectroscopy and prevent the technique from being just another “black-box”
in the lab. We will try to concisely highlight interesting topics and supply additional
references for further exploration at each stage. The advantages of delving into the
technique will be shown. The secondary objective, i.e., avoiding common problems
Metabolomics and NMR 75
before starting, will hopefully then become clear. Lastly, we will emphasize the
spectrometer information needed for manuscript reporting to allow reproduction of
results and confirm findings.
2 Brief History
2.1 Potential
Metabolomics has been utilized now for several decades (see (Emwas et al. 2020;
Finco et al. 2016; Heather et al. 2013; Kenny et al. 2010; Li et al. 2012; Psychogios
et al. 2011; Rasmussen et al. 2012) and references therein) but has not reached the
promised potential for applicable personalized/precision medicine. While there are
certainly specifically applied findings, there has not been the explosion of the
predicted novel medical treatments. Very recently mRNA vaccine developments
have introduced new concerns regarding the possible over-production of proteins/
metabolites (see review (Pardi et al. 2018)) and highlighted the application of
metabolomics for monitoring.
2.1.1 A Warning
It is important to remind the reader that the promise of marketability drove much of
the initial science. Metabolomics starting in the early 2000s, was envisioned as a
novel tool with exceedingly high profitability that would be: rapid, reliable, repro-
ducible, using easily acquired samples (e.g., urine) without extensive sample manip-
ulation (Bingol et al. 2016; Tayyari et al. 2013), and contain directly interpretable,
widely applicable, and useful results. Companies quickly jockeyed to be the first to
sell and inhabiting their NMR spectrometers (and now perhaps benchtop units
(Izquierdo-Garcia et al. 2020)) throughout clinical testing facilities across the
globe. For any new marketable technique to survive it must either be unique and
advantageous (i.e., provide a novel result) or be extremely competitively priced
when compared to existing technology (i.e., faster, cheaper, etc.). The corporate
emphasis was on finding unique biomarkers for dramatic high-profile diseases (e.g.,
cardiovascular disease, cancers, etc.) as quickly as possible and thus establish
lucrative patents and contracts. This did not encourage systematic, calm evaluation,
nor retesting, and that rush may have inadvertently hurt the entire field. Subsequent
validation studies (Emwas et al. 2020; Lacy et al. 2014; Sokolenko et al. 2013;
Markley et al. 2017) have begun to fill in the gaps, however questions regarding the
cross-validation of NMR data persists, e.g., site to site and/or study to study along
with assessment (Rocca-Serra et al. 2016; Cassiède et al. 2017).
2.2 Definitions
There has been ambiguity in the literature regarding key definitions and to avoid
confusion we will quickly define our working interpretation below.
2.2.1 Metabolome
The metabolome is commonly defined as all small molecules in the mass range of
50 to 1,500 Da (not a hard limit as certainly lipids can quickly exceed), associated
with a particular organism (Dunn et al. 2011; Psychogios et al. 2011; Wolfender
et al. 2013; Zulyniak and Mutch 2011). This includes all the various complexes,
sizes, and repetitive units of amino acids, lipids, carbohydrates, and other organic
Metabolomics and NMR 77
molecules collectively termed “metabolites” and are involved in all stages of metab-
olism both from naturally internal (endogenous) starting points, and those
originating from external (exogenous) sources. External include ingested foods or
pharmaceuticals, and the gastrointestinal metagenome along with subsequent
by-products, more colloquially referred to as the “gut flora and fauna” with these
microbes apparently playing a diverse role as our understanding expands.
More “-Omics”?
For a review, one cannot simply ignore the “-omics” flood as it now also includes
subtopics of metabolomics. Logically we start with the genome, then onto the
regulation of transcription with subsequent modifications. Then we consider effec-
tive production/regulation/degradation (e.g., protein turn-over), and finally the basic
building block level resulting in functional metabolism. These levels have an
expanding and/or encompassing “-omics” (Ragguett and McIntyre 2020) associated
with their study (i.e., genomics, transcriptomics, proteomics, metabolomics), and
even more recent approaches such as pharmacogenomics or pharmacometabolomics
(Van Der Wouden et al. 2020; Emwas et al. 2021; Lasky-Su et al. 2021; Vignoli
et al. 2019; Sherlock and Mok 2019).
There are many new subgroupings of of “omics” including Lipidomics (see a
recent full Nature review1), Foodomics (Balkir et al. 2021; Picone et al. 2022; Valdés
et al. 2021), and Elemental Metabolomics (Andersson et al. 2021; Edison et al. 2020;
Niziol et al. 2021; Zhang et al. 2018). In case readers have not had enough “-omics,”
there is a relatively new though well established and pertinent sub-group focusing on
the changes of metabolites over time called Fluxomics, see the comprehensive
review by Giraudeau (2020) and also (Emwas et al. 2020) for additional references.
Of key interest is the use of specific NMR labelling (Xu et al. 1999) in metabolites.
1
https://www.nature.com/subjects/lipidomics.
78 R. T. McKay
2.2.3 Metabolites
To properly address the term metabolites, we will use the excellent definition
provided by Tenori et al. in their concise review (Tenori et al. 2020).
Metabolites are the small molecules produced by, or taking part in, the chemical reactions
due to biochemical activities (i.e., the metabolism) in living organisms, and their levels can
change according to pathophysiological or environmental factors.
3 Expectations
3.1 Reality
While DNA is a single chemical class comprising only four nucleotides, immense
molecular stability, and a second strand as an immediately available backup copy for
error correction, moving downstream to the metabolites suddenly expands to
thousands if not tens of thousands of arguably critical molecules. These downstream
molecules can be quickly and substantially impacted by even small changes at the
DNA or transcription level. Metabolite concentrations can normally and
dynamically range by several orders of magnitude (Bouatra et al. 2013). Immedi-
ately the reader can see that while there may indeed be amplification, i.e., a
downstream effect any single “snap-shot” acquisition of metabolites (regardless of
how detailed and accurate) may have limited practical applicability to any disease
detection. This gets even more complicated regarding any practical translation to
personalized-medicine (“bedside”) treatment. Also metabolites have an immense
range of molecular variability in terms of primary/secondary structure, function,
modification, and lifespan.
4 NMR and MS
2
This overly simplified one-molecule/one-marker situation is highly unlikely.
80 R. T. McKay
The author approaches mass spectrometry (MS) with admittedly little practical
experience surrendering any in-depth evaluation of MS to the experts, e.g., see
highly recommended reviews (Wang and Li 2020; Alseekh et al. 2021)and
references therein. While mass spectrometry has undeniably superior solute sensi-
tivity (e.g., microlitre or sub-microlitre volumes with relatively low concentrations
(Li et al. 2020) per unit instrument time, it does come with a literal financial cost and
a cost in terms of total experimental length, monitoring/compensation for separation
technique(s), necessity of quality/control sampling, and finally the destruction of the
sample. It is important to note that MS instrumentation has a substantially smaller
initial instrument cost, along with smaller maintenance costs (e.g., cryogens). A
critical distinction is that MS usually requires some form of sample separation
(Petrović et al. 2005; Korfmacher 2005; Alseekh et al. 2021): chromatographic
(liquid or gas), electrophoretic, or based on ion mobility that can perturb the types
and quantities of measured metabolites. The separation efficacy changes over time
requiring continuous monitoring, evaluation, and correction during analysis, e.g.,
separatory columns degrade nonuniformly over time requiring a calibration of
resulting metabolites. Sample separation and detection limitations (Wang and Li
2020) can also be linked to the particular detector such as3: Fourier transform ion
cyclotron resonance (Marshall et al. 1998, 2007; Nikolaev et al. 2016), ion trap
(Todd and March 1999), Orbitrap and linear ion (Perry et al. 2008), time-of-flight
(Boesl 2017), and quadrupole (Linge and Jarvis 2009) to name some of the most
common. Evaluation of the potential and realized application has been extensively
3
Detector order is alphabetical only, not intended to imply frequency nor capabilities.
Metabolomics and NMR 81
covered and readers are directed to representative articles (Trifonova et al. 2021;
Alseekh et al. 2021; Lasky-Su et al. 2021; Wang and Li 2020). In the end MS has
become an extremely successful method accurately detecting thousands of
molecules and at much lower concentrations that can be practicably detected
by NMR.
Metabolomics by NMR has been vastly reviewed (Tenori et al. 2020; Giraudeau
2020; Giraudeau et al. 2014, 2015; Tavares et al. 2015; Halabalaki et al. 2014;
Bingol and Brüschweiler 2014; Bouatra et al. 2013; Wolfender et al. 2013; Lubbe
et al. 2013; Ellinger et al. 2013; Heather et al. 2013; Smolinska et al. 2012; Dunn
et al. 2011; Macel et al. 2010; Lindon et al. 2007; Beckonert et al. 2007; Emwas et al.
2018, 2019; Stringer et al. 2016). There is also a highly recommended NMR book
with an entire chapter dedicated to the practical aspects of metabolomics and NMR
sample handling/data/processing (Teng 2012).
Metabolomics by NMR contains all the common instrumental concerns (e.g.,
consistency, detection, assignment of signals, etc.), but a unique aspect of NMR is
that each atom, even in the same molecule, can have a specific resonance. This is
both the strength, i.e., the ability to resolve an atom based on its magnetic environ-
ment, and weakness. Every atom resonating by itself provides little cumulative
signal. Each relevant atomic signal must be detected, assigned, and then analyzed.
All of this with the massive assumption that no unexpected external change(s), other
than the central hypothesis of the study will perturb the measurement.
We will address that perturbation assumption and more below.
We will highlight key points in the subsections below to detail important information
from our experience to hopefully improve the reader’s future research studies. The
points will use a special format indicated by Note.
To start, a metabolomics NMR experiment (i.e., the “pulse sequence” itself) ideally
must be easy to acquire, i.e., any required hardware is commonplace, minimal setup,
robust, reliable, and reproducible. The instrument is assumed to be constantly
maintained, tested, and repaired be experienced operators to ensure predictable
performance. Next there are literally hundreds of NMR experiments (Berger and
Braun 2004; Braun et al. 1998), and the number of individual and inter-related NMR
parameters for each experiment is often overwhelming. The initial selection, testing,
and maintenance can be a full-time occupation for facilities (Reynolds and Enriquez
82 R. T. McKay
2002). The person contemplating a metabolomics NMR project must either acquire
equipment and become this, or already have the equipment and technical personnel
available.
Note
While the accepted “NMR standard” is 0.1% ethylbenzene with 0.01% TMS
in deuterated chloroform, Benchtop NMR manufacturers have created their
own standard using 1% ethylbenzene instead. Therefore, the reported bench-
top 1H NMR signal-to-noise ratios must be either divided by a factor of 10 to
compare to previously published standards and/or the user must recognize a
factor of 100 for required experiment time to achieve comparable results.
While it is fun to teach and delve into the world of NMR theory (e.g., providing
useful analogies describing each atomic nuclei as a little bar magnet spinning at its
own frequency depending on the magnetic field strength etc.), we simply do not have
the space in this review; especially considering the wealth of previously published
Metabolomics and NMR 83
Note
It is important to note that much of the background NMR material does not
address complex or “strong” 1H-1H coupling regarding metabolomics
assignments (see sections below).
Once the reader goes beyond the simple spin-1/2-coupling “tree” diagrams into
the higher order (also called second order, complex, or strong coupling depending on
the terminology used), we discover spectrometer/magnetic field-dependent patterns
(Foroozandeh et al. 2014; Bain et al. 1994). This is important for software selection,
analysis, and anyone aiming to assign biomarkers (Mercier et al. 2011; Tredwell
et al. 2011; Weljie et al. 2006). Using a reference database that does not take this into
account and/or have information for the magnetic field used will be more difficult to
use and require operators to be more experienced (Lacy et al. 2014; Mercier et al.
2011; Tredwell et al. 2011; Weljie et al. 2006).
4
No implied order, and naming a few of the author’s personal favourites. There are many others
certainly worth the reader’s attention and the provided list is not exhaustive.
5
Please forgive the puns.
84 R. T. McKay
hydrogen from 400 to 800 MHz (i.e., 9.39 to 18.8 Tesla static magnetic field
strength) and offered from a range of vendors.6
While concerns about data processing and analysis are equally important, there
are many publications and reviews focusing on issues involving time domain to
frequency signals such as signal enhancement (apodization), deconvolution, integra-
tion, and then reliably identifying and assigning signals or signal components
(Schönberger et al. 2015) to particular metabolite resonances. Readers seeking
even more information are directed to a representative set of papers and references
therein (Bartel et al. 2013; Beckonert et al. 2007; Bingol et al. 2016; Dudzik et al.
2018; Dunn et al. 2011; Eghbalnia et al. 2017; Ellinger et al. 2013; Emwas et al.
2016, 2018, 2019; Wang and Li 2020; Kohl et al. 2012; Krishnamurthy 2013; Lacy
et al. 2014; Parsons et al. 2009). We will now focus on the practical aspects of
sample and NMR instrumentation, e.g., see Chap. 3 of reference (Cavanagh et al.
2006) and Chap. 2 of reference (Zerbe and Jurt 2013).
Minimizing Problems
The detection and elimination of “confounders,” i.e., signals or influences on signals
that are not dependent on the hypothesized change (e.g., disease vs. healthy) but
instead are artifacts and/or errors in sample preparation and/or instrumentation, must
be a major focus for everyone involved in metabolomics. We wish to remove (or at
least minimize) as many variables as possible prior to the acquisition of NMR data
(Athersuch et al. 2013; Gibney et al. 2005; Meissner et al. 2014; Staab et al. 2010;
Zulyniak and Mutch 2011). This requires prior awareness, continued attention,
precise planning throughout, and finally careful preparation, and this leads us
directly to the sections below regarding sample and instrumentation details and
recommendations.
5.3 Samples
5.3.1 Preparation
Sample selection, preparation, consideration for repetitive sampling, and handling
consistency are all incredibly important. This is only the first step, but the easiest to
get wrong, and sometimes without being determinable until very late in the study.
Note
The Brian Sykes’ First rule of NMR applies here, i.e., “Garbage” in equals
“Garbage” out. Essentially no matter how good the operator nor expensive the
instrumentation, if your sample is poor, your results are poor.
6
Unfortunately in 2014, Agilent (who had purchased Varian Inc. in 2010) exited the NMR market.
This has added a level of uncertainty for those still needing repair/support/parts for their massive
equipment investments, especially in the ever-expanding austerity environments facing publicly
funded academic institutions.
Metabolomics and NMR 85
Note
The key point here is absolute consistency, from everyone. Despite careful
planning all the group members that will be handling samples must understand
the stringent necessity of consistent sample treatment.
7
https://blogs.umass.edu/weiguoh/?cat=81121.
8
X and Y based shims (termed the non-spin shims) could be optimized more easily and the overall
spectral line shape was normally much improved.
86 R. T. McKay
Note
In our experience, the performance gains from spinning samples do not make
up for the variability and problems associated with spinning side-bands. We
do not use spinning nor recommend for metabolomics.
Additionally
The NMR tube manufacturing common practice of putting a magnetic field
strength recommendation (e.g., eco tube for 400 MHz or below, or precision
NMR tube for 600–800 MHz) assumes spinning of samples and is not
necessary for non-spun and high throughput studies. In our experience, basic
economical NMR tubes are sufficient in nearly all sample applications.
Volunteers
First, can enough volunteers be found? How does one know how many or even how
to appropriately attract volunteers for a study? While seemingly a trivial point it is no
easy task with modern privacy laws, e.g., how do you legally even approach people
without prior permission? A definite catch-22 situation. Our experience was this
severely slowed down our study almost to the point of cancellation regardless of
successful funding.
Metabolomics and NMR 87
Note
How to get study volunteers? This is under jurisdictional control so carefully
check your local requirements. We had to have research assistants set up tables
on campus with signs and wait for individuals to approach. Privacy rules
prevented pretty much every other idea we came up with (e.g., emails, offering
prizes/incentives, telephone, etc.).
9
This is a loaded word, i.e., depending on study type, number of participants, metabolic reaction
time, etc. just to name a few likely variables.
88 R. T. McKay
very precise details, e.g., how long does it take (and what temperature do samples
reach?) for samples to transfer from long-term storage to “in the instrument.” Prior
agreement between individuals is key for consistency.
Note
While the final data acquisition and analysis (Schönberger et al. 2015) is often
strongly emphasized in publications, we recommend focusing more on the
initial preparation, handling, and organization as this will facilitate analysis.
Examples from practical experience; does one take all samples out of -80°C
storage at the same time? We did choose to remove all samples at once, but
immediately stored them in a normal +5°C refrigerator to thaw slowly and consis-
tently (i.e., samples on the outside of the group did not thaw first etc.). We also had to
address the preparation time difference between the first and last handled sample.
Specifically, how long does the last sample sit in the spectrometer robot sample
handling system waiting while all the previous samples are acquired, e.g., 7–8 min
per sample with 100 submitted samples therefore the last sample sits ~800 min
longer at room temperature than the first sample. Assuming bacterial inhibitors
(Bernini et al. 2011; Lauridsen et al. 2007) are used, any chemical reaction and/or
oxidation within the sample will undoubtedly be temperature and time sensitivity.
So there are really three key areas to consider. The first, what is the long-term
storage available? Second, conditions (time, temp, etc.) from thawing to analysis.
The last, how many times can the sample be frozen/re-frozen either for re-analysis or
due to unforeseen delays (Pinto et al. 2014; Saude et al. 2006; Saude and Sykes
2007; Rist et al. 2013). The first concern is usually dealt with by storing at a
consistent -80°C, and this has become the standard.
Note
Do not forget -80°C ultra-low temperature freezers are expensive, can have
additional electrical requirements, produce a lot of residual room heating, and
do fail. This means extensive infrastructure pre-planning and monitoring.
The second issue has a relatively simple solution, i.e., prep samples just prior to
acquiring data and use lower ambient temperatures during prep. Specifically we
recommend keeping samples at 5°C until just before instrument acquisition (and
depending on your latitude/location you may also need to include humidity control).
This includes appropriately selected robotic sample controls, and a consideration of
total time waiting for acquisition (Saude and Sykes 2007) such as limiting the size
and using a batch method for sample preparation. Preparing 40 samples at a time
instead of 400 reduces the time differences, however this substantially increases the
labor involved as more frequent/smaller batches must be prepared reducing lab
efficiency and increasing costs. It was our experience that 20–30 samples per
Metabolomics and NMR 89
batch, with two batches a day worked well allowing a single spectrometer to be
utilized efficiently. This was especially important as we were restricted to weekend
access and therefore had to maximize usage.
Note
While lowered temperature control while waiting on the instrument itself is
beneficial, it adds complications for the NMR instrumentation. Thermal equi-
librium (usually at room temperature) must be reached for the entire NMR
sample prior to data acquisition in the spectrometer receiver coil.
Here one does not gain “mass sensitivity” through concentrating into smaller
diameter tubes. In these cases, a wider diameter sample tube physically allows
more atoms (albeit at a lower “concentration” via a greater number of overall
molecules) to be placed inside the observing receiver coils, and results in an
improved signal. Conversely if solubility is not a limiting factor and the same
molar mass of material can be concentrated into a smaller volume and smaller
NMR tube, e.g., a 1.7 mm diameter NMR tube versus the larger 5 mm tube, then
the atoms are physically closer to the receiver NMR coils and produce a larger
relative signal. Smaller tubes also become advantageous with high salt samples and
cryogenically cooled (i.e., cold) NMR probes. The S/N improvements of cold-
probes are quickly lost (Kelly et al. 2002; Nausner et al. 2010; Xiao et al. 2009) as
the ionic concentrations rise, but this phenomenon can be compensated for with
narrower or specially shaped tubes.
Note
One substantial advantage for 5 mm NMR tubes is the increased ease of
removing/recovering samples for further study and/or storage also the
increased ability to wash and re-use the NMR tubes.
Groups may opt to dispose of 3 mm or thinner NMR tubes after each use due to
cleaning/recovery difficulties and therefore the costs must be taken into consider-
ation for large-scale samples studies.
5.4.1 Probe
The NMR probe is critical. The probe inductively delivers relatively high-power
electromagnetic energy to the sample and influences the sample’s atomic state(s),
therefore performing the NMR experiment (called a “pulse sequence”). The probe is
also responsible for detecting the sample’s subsequent response(s), i.e., very low
power inductive signals from the precessing atoms in the sample, due to the presence
of a strong static external magnetic field (i.e., the NMR magnet).
One can easily imagine that the probe experiences a light impact from each
sample and spinner as the air pressure is decreased and sample raised/lowered.
Also contamination from any materials passed from the operator’s hands to the
NMR sample, material (e.g., dust) from the air as the sample lowers, NMR tube
breakage (e.g., multiple samples inserted without removal of the previous10) and
wear due to high-power electromagnetic induction during pulse sequences to name
but a few challenges. There are also physical moving electronic components (see
10
For high throughout facilities under manual (i.e., non-robotic) sample handling, this problem
occurs more often than you might think.
Metabolomics and NMR 91
Sect. 5.4.8 below) (Halliday et al. 2013; Derome 2013) with the temperature, ionic
strength, and dielectric constant of the solvent all coming into play. These aspects are
important and cannot simply be considered “constant.” Even if the instrument is
solely dedicated to the metabolomics user and not a multi-user facility (more
common), the NMR instrumentation experiences wear-and-tear over time.
An improperly setup probe will not yield the expected signal amplitudes and may
even rapidly devolve into relaxation and off-resonance effects, and far more compli-
cated spectra outcomes.11 On the receiver side, the resulting signal intensity will also
suffer at best decreasing certainty of measurement, or worse preventing observation
of the signal. Early metabolomics studies may not have had access to robotic sample
handling that included automated tune/match (nor even regulated temperatures for
samples awaiting data acquisition). Early studies assumed that the tune and match
could be optimized for the first sample and would not deviate perceptively from
sample to sample. In our experience, this assumption is not correct especially with
human urine samples where the salt concentrations and therefore the dielectric
matching conditions can change substantially with every sample.
Note
The practical result of improper tune/match (incorrect sample impedance
matching) is inconsistent and inefficient energy transfer/excitation to the
sample. Remember that metabolomics is all about consistency. It is our
recommendation that any study include infrastructure capable of optimizing
the tune/match for every entering NMR data acquisition.
11
For those wishing a more detailed and mathematical description, see Chap. 23 especially Sect.
5 in (Brown 2016).
92 R. T. McKay
Note
Cryogenically cooled probes are often a logical and cost-effective upgrade to
existing systems. However, probes and the application to magnets along with
annual maintenance costs and supporting infrastructure requirements are not
trivial so beware.
5.4.2 Console
The NMR console is the heart of the NMR spectrometer containing frequency
generation, timing control, power supply, input/output controls, amplifiers, band
selective equipment, relays, pneumatic controllers, thermal regulation, pulse field
gradient generation, shim controls, etc. all the components necessary to perform the
desired NMR experiments. The console, just like the NMR probe, requires constant
monitoring, calibration, and inevitably some repair and/or replacement of parts.
These must be done with full understanding of all consequences regarding instru-
ment performance for metabolomics studies.
Note
We strongly recommend a professional staff person be utilized to initially
calibrate and monitor the equipment at regular intervals to ensure consistency
in the console performance.
Note
Cursory comparisons of pulse sequence basics between versions or vendors
will often miss internal phase cycle details. Even experienced NMR users can
be caught unaware of the complexities involved.
94 R. T. McKay
5.4.5 Parameters
There are dozens to hundreds of parameters involved various pulse sequences. For
example, even the simplest 1D “Pulse-Read” experiment (see Appendix A) requires
the carrier position, power level, and pulse duration to be properly set. In addition,
the length of the acquisition period and dwell time (i.e., time between sample-
induced voltage readings in the receiver coil) will need to be properly selected so
that required information is properly represented. Even this is a huge oversimplifi-
cation. The more complex the manipulation of spins during the sequence the more
parameters are involved, and the incorporation of multiple nuclei and/or multiple
dimensions of indirectly detected dimensions will amplify the number of parameters
exponentially.
We will focus on the most concise list of NMR parameters that need to be
addressed for each sample involved in metabolomics NMR experiments. These
are also necessary for testing and reproduction and should be included for publica-
tion of manuscript data.
We will first briefly address the practical outcome of parameters.
Note
The phase cycle is used to cancel any general imperfections in the spectrome-
ter receiver path, i.e., cyclops basic phase cycle. The higher orders of the phase
cycle will usually attempt to remove smaller and smaller artifacts so one can
usually use a subset or portion of the entire cycle. Consistency is again key,
and one must determine the best selection prior to study start.
Note
We are concerned with comparing data from instrument to instrument, but
even if a single unchanging/dedicated instrument is used for the entire study,
one would only have precision (i.e., consistent error). This may not be
reproducible later even with the same spectrometer so relying on precision
may not be sufficient. We highly recommend directly calibrating each instru-
ment prior to study start, and after any hardware changes using referenced
techniques (Holz et al. 2000; Karschin et al. 2022; Raiford et al. 1979).
Regarding sample temperature, Saude et al. (2006, 2007; Saude and Sykes 2007)
published detailed tests of human urine stability versus storage time and temperature
and we highly recommend interested readers consider this type of preparation for
their samples to establish utmost consistency.
Note
To speed up data acquisition, we used a two-stage sample cooler. The first
stage was long-term storage at ~5°C. The second stage was (using a golf
analogy) termed “on-deck” and was a slightly higher temperature than the
equilibrium temperature. See below for details.
5.4.8 Tune/Match
Frequency “tuning” and impedance “matching” of the sample and the spectrometer
transmit/receive electronic circuit is critical for optimization. This ensures that the
inductive moment or energy transfer going to, and the subsequent signal coming
from the sample is as efficient as possible.
The concepts involved in impedance involve the resistors, frequencies, inductors,
capacitance, mutual or self-inductance, etc. and goes far beyond this chapter (e.g.,
Metabolomics and NMR 97
see Chaps. 32 and 33 dealing with these topics (Halliday et al. 2013)) and the authors
capabilities. However in terms of practical NMR tuning and matching are still vitally
important, and it is usually sufficient to use the common analogy that impedance
matching involves adjusting the spectrometer circuitry to “match” that of the sample
and the sample’s dielectric constant of the solvent. Each solvent/sample can have
different impendence characteristics (depending on temperature, salts, solvent(s),
type of glass, ions in solution, etc.), so the probe must have an electronic range of
capabilities for the nuclei to be observed and appropriate for the sample itself. Probes
are usually designed with either manually manipulated, or automated movement of
physical rods connecting deep inside the probe head. These rods move specific
electronic components changing the transmitter and receiver’s electronic circuit
configuration.12 A maximum transfer of power occurs when the two circuits (i.e.,
probe and sample) are identical in resistance, or in this case, due to alternating
current, their impedance (Wilson 2007).
Note
With a poor “matching” condition, there can be substantial energy produced
by the NMR console that does not enter the sample, but instead reflects,
returning up the pathway and potentially damaging equipment.
12
Capacitors and inductors (trying not to get confused with inductance nor impedance) of different
sizes, types, and changeable physical positions are used to control analog frequency band widths
and the subsequent matching of the circuitry.
13
I had the pleasure of attending one of Dr. Hoult’s talks as he was an invited speaker for the Alberta
Cancer Foundation at the University of Alberta in 2015. His talk was amazing and the first time I
really started to understand the difference between true NMR magnetic inductance of signal
transmission versus the standard NMR analogy of radio signals and antennas.
98 R. T. McKay
For the simplest 1D-1H direct detection NMR pulse sequence experiment (aka
pulse-read) formed from a recovery delay, excitation pulse, and observation/data
acquisition period, the only pulse power/duration consideration is the excitation
pulse. As the reader may be aware, especially from the referenced literature, most
spectroscopists use the maximum power (or close to it) that the probe can withstand.
Therefore only the duration of the pulse application can be altered safely by the user.
Consistent determination and application of the optimal excitation (i.e., movement
of the bulk magnetization vector fully into the XY plane for detection by the receiver
NMR coil in the probe) are critical for multi-pulse experiments, but the story can be
more complex for the simplest pulse-read experiment. Examples of optimization are
provided in these references (Bodenhausen et al. 1984; Breton and Reynolds 2013;
Burrow et al. 2014; Reynolds and Enriquez 2002; Reynolds and Burns 2012). While
some articles (Schönberger et al. 2014) recommend using only a 90° excitation
pulse, there are complications due to relaxation. The “Ernst Angle”(Cavanagh et al.
2006; Freeman 1997; Keeler 2010; Levitt 2001) can be applied for simple
experiments or more complex ones (Ogg et al. 1994; Zhang et al. 2000). The Ernst
angle was originally developed to achieve the greatest S/N per unit time available. In
our facility, the primary purpose of using less than a 90° pulse is to improve the
accuracy of integration over all regions of the molecule. Different regions/atoms/
functional-groups may have different relaxation rates, and therefore additional scans
(to improve the S/N) will result in different signal intensities/integrations based not
just on the number of nuclei, but also their relative atomic and molecular mobility.
Using a smaller pulse angle (e.g., 45°) reduces the time needed to fully relax, thus
restoring the integrity of integration but with an S/N cost. For multi-pulse, e.g.,
multi-dimensional/multi-nuclear experiments involving subsequent magnetization
precession and evolution with time, the optimal full 90° pulse is critical. Otherwise,
there is a cumulative and compounding loss for each “imperfect” pulse.
Note
For metabolomics using the first increment of the 2D-1H,1H-NOESY experi-
ment (aka metnoesy (McKay 2011)) which is standard for the Chenomx
metabolomics software and database, it is essential that the calibrated 90°
pulse is used consistently for all samples. Unless the solvent is highly consis-
tent, unlikely with human urine/metabolomics samples, the 90° pulse needs to
be determined for each sample prior to data acquisition.
5.4.10 Gain
Gain is essentially the volume control on the signal receiver. While we do not have
time for the fundamental radio/receiver electronic aspects, suffice to say that too low
a gain negatively impacts the experiment with poor S/N, while setting the gain too
high risks overloading the receiver coil and damaging the FID data. Damaged data is
not something that can be corrected later, and the experiment is forfeited. Overloads
Metabolomics and NMR 99
occur when the voltage reading from the transverse recorded magnetization exceeds
the electronic capability, and this is most often caused by a disproportionately strong
solvent signal (e.g., water) or sample component (e.g., urea). Careful and consistent
setting must be used to insure proper acquisition over a long-term metabolomics
study.
Note
While it is not relevant to report the instrumental value of gain in the manu-
script, it is important to note for the manuscript reviewer and readers how an
appropriate gain was determined and if consistently used for all samples
ensuring that receiver and/or analog-to-digital converter (ADC) errors were
avoided.
14
A possible solution is to apply backwards linear prediction to the first few points, thus replacing
the damaged data with realistic predictions based on the later non-distorted information. Not ideal
but sometimes useful.
100 R. T. McKay
sees a large distortion on peak shape. Chenomx software uses the shape of the DSS
methyl group to back-calculate corrections for any small shim imperfections. A
distortion of the DSS methyl reference peak, however, can dramatically affect
resonance integrations and the subsequent metabolite identification stage. There is
no way to correct for PFG dispersive changes in the reference signal once collected
(i.e., you cannot phase the distortion out without negatively impacting other
resonances).
Note
For metabolomics, it is our experience that PFGs cause far more problems than
they solve, and we highly recommend not utilizing pulse sequences
with PFGs.
The easiest and therefore most common method is to remove all PFGs from the
metabolomics NMR pulse sequence. This has been the case for nearly two decades
now. While the use of alternating directional gradients can be used to reduce the
artifacts (Nguyen et al. 2007; Sokolenko et al. 2013), validation of NMR
metabolomics databases has not been done with any of the proposed pulse sequence
changes.
Note
Without the 90° excitation pulse duration (often termed pulse width) or that a
90° pulse was used and stating the induced field strength(s) for the various
pulses, we cannot reproduce the experiment. No reviewer should allow a paper
into publication that does not have at least that minimum information.
Note
The key point here is consistency of relaxation times within the study. The
number of scans, and other experimental parameters must be maintained for all
the samples in the study.
The pulse sequence cannot be swapped out nor altered halfway through the study
(e.g., choice of water suppression), as any change will affect the relaxation, water
suppression, degree of excitation, etc. which will certainly be detected later in the
processing/analysis phase.
Of equal importance is that one must conform to the pulse sequence delays
required by the database(s) being used to determine sample content (see Sect.
5.4.6). For example, Chenomx Inc. (Mercier et al. 2011; Tredwell et al. 2011;
Weljie et al. 2006) relies on the user running a 10 ms recovery time, with 990 ms
solvent saturation at a controlled level (i.e., ~80 Hz gammaB1 effective induced field
strength see previous Sect. 5.4.9) precisely on resonance, followed by the excitation
pulses, a 100 ms “mix” where saturation is turned back on, and finally the last 90°
pulse and 4 s of acquisition. Total experiment time for a single scan is 5 s. Deviation
from this will move the user away from accurate/useful integrations when comparing
to the Chenomx database (Lacy et al. 2014; Mercier et al. 2011; Tredwell et al. 2011;
Weljie et al. 2006).
102 R. T. McKay
Note
We have found that a great deal of problems can be avoided by using line
broadening, e.g., 0.25–0.5 Hz.
Small shimming errors can be easily averaged or approximated out by the slight
change. This must be included in the reported parameters for comparison and
evaluation.
15
e.g. alfa and rof2 on older Varian/Agilent spectrometers with ddrtc coming into play on newer.
Metabolomics and NMR 103
Note
Utmost effort should be taken to optimize NMR pulse sequence delays prior to
the start of NMR acquisition. This care along with knowledgeable lock
settings/setup should minimize the necessity of phasing and the introduction
of possible user error.
5.5.5 Referencing
Referencing the zero point on the spectra is incredibly important for metabolomics.
The databases typically use the chemical shift position of a peak to begin all auto-
assignments. Even peak pattern recognition usually starts with the chemical shift of a
unique resonance and tries to build out from there the possible coupling profiles. If
the sample is referenced incorrectly the software will have infinitely more difficulty
assigning peaks, let alone correctly.
As mentioned, the Chenomx software has several internal reference standards
available checking the reference peak line shape for application of corrections to the
rest of the spectra.
Note
The known amount of reference intensity also provides quantitative informa-
tion for the integrations.
6 Future Ideas
While metabolomics has been predominantly only liquids likely due to a focus on
easily acquired samples (e.g., blood, urine, sputum, etc.), there is a growing amount
of research into solids NMR utilizing high-resolution magic angle spinning
(HRMAS) for metabolomics (Cheng 2007) and specifically Chap. 4 (Tilgner et al.
2019). We direct interested readers to the many excellent reviews of solid-state
NMR (Laws et al. 2002; Ashbrook and Sneddon 2014; Reif et al. 2021) and
references therein, solid-state cold-probes (Matsuki et al. 2015), and the relatively
new field of dynamic nuclear polarization (DNP) enhancement (Albert et al. 2017;
van Bentum et al. 2016; Matsuki et al. 2015). This is an exciting new area, and we
look forward to developments.
104 R. T. McKay
One of the biggest potential new developments involves the PureShift style/family of
NMR pulse sequences (Kiraly et al. 2018, 2021; Dumez 2018; Foroozandeh et al.
2018; Moutzouri et al. 2017; Castañar 2017; Mishra and Suryaprakash 2017; Kew
et al. 2017; Zangger 2015; Aguilar et al. 2015; Foroozandeh et al. 2014, 2015;
Mauhart et al. 2015; Reinsperger and Luy 2014; Kaltschnee et al. 2014; Paudel et al.
2013; Meyer and Zangger 2013; Aguilar et al. 2012; Aguilar et al. 2010; Zangger
et al. 2001). There are many variations, iterations, and subsequent improvements,
but all focus on removing the dipolar “coupling” or crosstalk between neighboring
hydrogen atoms (as previously mentioned). PureShift sequences attempt to remove
(or at least minimize) the couplings and complexity of the spectra. The variations
attempt to compensate for the inherent weaknesses of the pulse sequence, i.e., signal-
to-noise, and distortions in tightly coupled systems.
While these couplings are often crucial for organic chemistry and basic molecular
identification in traditional NMR usage, for metabolomics they create greater spec-
tral complexity with a multitude of overlapping congested spectral information.
Chenomx software checks each peak position, relative integration, and coupling
pattern when it attempts identification, and these have been validated for each
16
“Krish” to friends and colleagues is an amazing resource for the NMR community and we
gratefully acknowledge years of benefiting from this person’s contributions.
Metabolomics and NMR 105
7 Conclusion
The most important point we can make is that there are many aspects requiring
attention prior to initiating a metabolomics study via NMR. The information and
experiences detailed above will hopefully help the reader avoid some of the common
pitfalls that we encountered. We also hope that we have convinced new users that
every single metabolomics study can benefit from having experienced knowledge-
able spectroscopists involved, especially in the earliest planning stages. The chances
of acquiring reliable/reproducible data increase exponentially when including the
NMR operators. Unfortunately, these problems are often unrealized until the statis-
tical analysis stage, when the errors due to confounders can exceed the actual data.
The entire study may thus be ruined.
Consistency in every aspect is paramount. From sample acquisition, storage,
preparation, handling, data acquisition, storage and/or retesting, to processing and
analysis. There are many steps that can go astray, making the data more difficult to
assess. However, organized from the start, carefully monitored throughout, and
diligently systematically checking while acquiring the data will give the research
group their best chance of having a useful data set at the end. Spectrometer
consistency is paramount and mentioned above extensively, therefore users may
want to explore the possibility of establishing quality control checks are regular
intervals in their study.
Finally, we hope that the excellent referenced publications and books referenced
will be as useful to your study as they have been to our education, though we are sure
there are many others we have not yet discovered. Best of luck.
Acknowledgments The author would like to thank Professor Vladimir Michaelis (Department of
Chemistry, Univ. of Alberta) for critical reading and suggestions on the writing of this document.
106 R. T. McKay
12.5us 12.5us
12.5us 100.0ms
3.999sec
990.0ms
Tx 10.0ms 0.0
H1 d1 satdly pw pw hst mix-hst
pw
A B C D
Experimental Section
NMR Samples
NMR samples were dissolved in 90% D2O (~600 μL in volume) and sample data
acquired in 5 mm XXXmodel NMR tubes purchased from XXXmanufacturer. All
NMR solvents were purchased from XXX. NMR tubes were washed using three
rinses of 95% ethanol with a single final D2O rinse, and then inverted to air dry
overnight.
NMR Spectroscopy
NMR experiments were collected on either a 14.1T (600 MHz) Varian/Agilent
VNMRS with an Agilent 7,620 automatic sample handling system, or a 9.39T
Metabolomics and NMR 107
(400 MHz) Varian Inova NMR spectrometer, both at 27°C (calibrated using metha-
nol17). On the 600 MHz spectrometer, an HCN BioNMR probe (i.e., direct detect 1H
inner coil) was used, while on the 400 MHz instrument an AutoDB broadband (i.e.,
broadband inner coil) 5 mm probe was available. All spectra were run “locked” on
the 2H resonance signal and chemical shifts were referenced using the residual
proton 1HOD signal position18 (i.e., 4.7 ppm) prior to saturation. One dimensional
1
H data was acquired using either presaturation19 followed by a single excitation
pulse/acquire spectrometer sequence, or the first dimension of a -2D-1H,1H-NOESY
(metnoesy). The metnoesy (see Appendix A) uses a recovery delay of 10 ms, then
990 ms of presaturation followed by two 90° pulses, a mixing time of 100 ms with
saturation, a final 90° pulse and lastly a 4 s acquisition period. The saturation pulse
and carrier position were manually optimized and placed on the water resonance.
Saturation was applied with a gammaB1 induced field strength of 100 Hz (600) or
30 Hz (400) depending on water suppression efficacy and to avoid receiver
overloads. For the simple 1D-1H experiments, the duration of the saturation pulse
was 2 s on both instruments. Parameter settings for all experiments were: sweep
width of 7,183 Hz, acquisition time 2 s, with 28,736 real plus imaginary acquired
(600), or a sweep width of 4,801 Hz. For the 1D-1H, an acquisition time of 3 s with
28,812 real and imaginary points for the 400 MHz NMR was utilized, and an ~30°
(i.e., 3.4 μs) excitation pulse angle (applied at ~24 kHz gammaB1) was used
following the concept of the improved integration and reduced relaxation times
(commonly known as the “Ernst Angle”),20 however on some of the initial highly
concentrated samples an extremely short pulse length was needed to avoid receiver
overflows (e.g., 1us excitation pulse).
For processing of all NMR data, the acquired points were zero-filled to twice the
number of acquired points, and a line-broadening apodization function of 0.5 Hz was
17
D. S. Raiford, C. L. Fisk, E. D. Becker, Anal. Chem. 51, 2050 (2002).
18
Wishart, D., Bigam, C., Yao, J., Abildgaard, F., Dyson, H. J., Oldfield, E., Markley, J., and Sykes,
B. (1995) 1H, 13C and 15N chemical shift referencing in biomolecular NMR, J Biomol NMR 6,
135–140.
Trainor, K., Palumbo, J. A., MacKenzie, D. W. S., and Meiering, E. M. (2020) Temperature
dependence of NMR chemical shifts: Tracking and statistical analysis, Protein Science 29,
306–314.
19
Hoult, D. I. (1976). Solvent Peak Saturation with SIngle Phase and Quadrature Fourier Transfor-
mation. Journal of Magnetic Resonance, 21, 337–347.
Campbell, I. D., Dobson, C. M., Jeminet, G., & Williams, R. J. P. (1974). Pulsed NMR methods
for the observation and assignment of exchangeable hydrogens: Application to bacitracin. FEBS
Letters, 49(1), 115–119.
20
Lauridsen, M., Maher, A. D., Keun, H., Lindon, J. C., Nicholson, J. K., Nyberg, N. T. et al.
(2008). Application of the FLIPSY pulse sequence for increased sensitivity in 1H NMR-based
metabolic profiling studies. Anal Chem, 80(9), 3365-3371.
Waugh, J. S. (1970). Sensitivity in Fourier transform NMR spectroscopy of slowly relaxing
systems. Journal of Molecular Spectroscopy, 35(2), 298–305.
Ernst, R. R., & Anderson, W. A. (1966). Application of Fourier Transform Spectroscopy to
Magnetic Resonance. Review of Scientific Instruments, 37(1), 93–102.
108 R. T. McKay
then applied. Manual phasing and referencing to the solvent peak were used to
confirm referencing based on the lock solvent (when available) and previously
determined carrier position. Spectra were analyzed using VNMRJ 4.2 patch110
software.
References
Aguilar JA et al (2010) Pure shift 1H NMR: a resolution of the resolution problem. Angew Chem
Int Ed Engl 49(23):3901–3903
Aguilar JA et al (2012) Decoupling two-dimensional NMR spectroscopy in both dimensions: pure
shift NOESY and COSY. Angew Chem Int Ed Engl 51(26):6460–6463
Aguilar JA et al (2015) Minimising research bottlenecks by decluttering NMR spectra. Chemistry
21(17):6623–6630
Albert BJ et al (2017) Instrumentation for cryogenic magic angle spinning dynamic nuclear
polarization using 90L of liquid nitrogen per day. J Magn Reson 283:71–78
Alseekh S et al (2021) Mass spectrometry-based metabolomics: a guide for annotation, quantifica-
tion and best reporting practices. Nat Methods 18(7):747–756
Amoureux JP et al (2008) Rapid analysis of isotopically unmodified amino acids by high-resolution
(14)N-edited (1)H-(13)C correlation NMR spectroscopy. Chem Commun (Camb) 48:6525–
6527
Andersson ER et al (2021) Identifying metabolic alterations associated with coral growth anomalies
using 1H NMR metabolomics. Coral Reefs 40(4):1195–1209
Ashbrook SE, Sneddon S (2014) New methods and applications in solid-state NMR spectroscopy
of quadrupolar nuclei. J Am Chem Soc 136(44):15440–15456
Athersuch TJ et al (2013) Evaluation of 1H NMR metabolic profiling using biofluid mixture design.
Anal Chem 85(14):6674–6681
Bain AD, Burton IW, Reynolds WF (1994) Artifacts in two-dimensional NMR. Prog Nucl Magn
Reson Spectrosc 26:59–89
Bakhmutov VI (2005) Practical nuclear magnetic resonance relaxation for chemists. Wiley, p 216
Balkir P, Kemahlioglu K, Yucel U (2021) Foodomics: a new approach in food quality and safety.
Trends Food Sci Technol 108:49–57
Ban D et al (2017) Recent advances in measuring the kinetics of biomolecules by NMR relaxation
dispersion spectroscopy. Arch Biochem Biophys 628:81–91
Bartel J, Krumsiek J, Theis FJ (2013) Statistical methods for the analysis of high-throughput
metabolomics data. Comput Struct Biotechnol J 4:e201301009
Barton RH et al (2008) High-throughput 1H NMR-based metabolic analysis of human serum and
urine for large-scale epidemiological studies: validation study. Int J Epidemiol 37 Suppl 1:i31–
i40
Beckonert O et al (2007) Metabolic profiling, metabolomic and metabonomic procedures for NMR
spectroscopy of urine, plasma, serum and tissue extracts. Nat Protoc 2(11):2692–2703
Bendet-Taicher E, Müller N, Jerschow A (2014) Dependence of NMR noise line shapes on tuning,
matching, and transmission line properties. Concepts Magn Reson B Magn Reson Eng 44(1):
1–11
Berger S, Braun S (2004) 200 and more NMR experiments: a practical course. Wiley-VCH, p 838
Bernard GM et al (2017) Methylammonium lead chloride: a sensitive sample for an accurate NMR
thermometer. J Magn Reson 283:14–21
Bernini P et al (2011) Standard operating procedures for pre-analytical handling of blood and urine
for metabolomic studies and biobanks. J Biomol NMR 49(3–4):231–243
Bingol K, Brüschweiler R (2014) Multidimensional approaches to NMR-based metabolomics. Anal
Chem 86(1):47–57
Metabolomics and NMR 109
Laws DD, Bitter HM, Jerschow A (2002) Solid-state NMR spectroscopic methods in chemistry.
Angew Chem Int Ed Engl 41(17):3096–3129
Lehnert W, Hunkler D (1986) Possibilities of selective screening for inborn errors of metabolism
using high-resolution 1H-FT-NMR spectrometry. Eur J Pediatr 145(4):260–266
Lepre CA (2011) Practical aspects of NMR-based fragment screening. Methods Enzymol 493:219–
239
Lesar CT et al (2011) Report on the analysis of common beverages spiked with gamma-
hydroxybutyric acid (GHB) and gamma-butyrolactone (GBL) using NMR and the PURGE
solvent-suppression technique. Forensic Sci Int 212(1–3):e40–e45
Levitt MH (2001) Spin dynamics: basics of nuclear magnetic resonance
Li K et al (2012) Establishment of a blunt impact-induced brain injury model in rabbits. Chin J
Traumatol 15(2):100–104
Li Y et al (2020) Sub-nanoliter metabolomics via mass spectrometry to characterize volume-limited
samples. Nat Commun 11(1):1–16
Lindon JC, Nicholson JK, Everett JR (1999) NMR spectroscopy of biofluids. In: Annual reports on
NMR spectroscopy: annual reports on NMR spectroscopy volume 38. Elsevier, pp 1–88
Lindon JC, Holmes E, Nicholson JK (2007) Metabonomics in pharmaceutical R&D. FEBS J
274(5):1140–1151
Linge KL, Jarvis KE (2009) Quadrupole ICP-MS: introduction to instrumentation, measurement
techniques and analytical capabilities. Geostand Geoanal Res 33(4):445–467
Link M et al (2014) Food analysis to check quality, safety and authenticity by full-automated
1H-NMR. ll1.workcast.net
Liu G et al (2014) Fast 3D gradient shimming by only 2×2 pixels in XY plane for NMR-solution
samples. J Magn Reson 248:13–18
Lubbe A et al (2013) NMR-based metabolomics analysis. In: Lämmerhofer M, Weckwerth W (eds)
Metabolomics in practice, Metabolomics in practice: successful strategies to generate and
analyze metabolic data. Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, pp 209–238
Macel M, Van Dam NM, Keurentjes JJ (2010) Metabolomics: the chemistry between ecology and
genetics. Mol Ecol Resour 10(4):583–593
Markley JL et al (1998) Recommendations for the presentation of NMR structures of proteins and
nucleic acids. IUPAC-IUBMB-IUPAB inter-union task group on the standardization of data
bases of protein and nucleic acid structures determined by NMR spectroscopy. J Biomol NMR
12(1):1–23
Markley JL et al (2017) The future of NMR-based metabolomics. Curr Opin Biotechnol 43:34–40
Marshall AG, Hendrickson CL, Jackson GS (1998) Fourier transform ion cyclotron resonance mass
spectrometry: a primer. Mass Spectrom Rev 17(1):1–35
Marshall AG et al (2007) Fourier transform ion cyclotron resonance: state of the art. Eur J Mass
Spectrom 13(1):57–59
Matsuki Y et al (2015) Closed-cycle cold helium magic-angle spinning for sensitivity-enhanced
multi-dimensional solid-state NMR. J Magn Reson 259:76–81
Maudsley AA, Simon HE, Hilal SK (1984) Magnetic field measurement by NMR imaging. J Phys E
Sci Instrum 17(3):216–220
Mauhart J et al (2015) Faster and cleaner real-time pure shift NMR experiments. J Magn Reson 259:
207–215
McClung RED (1999) Coherence transfer pathways and phase cycles: the decoding of a pulse
sequence. Concepts Magn Reson 11(1):1–28
McGrath BM et al (2007) Unlike lithium, anticonvulsants and antidepressants do not alter rat brain
myo-inositol. Neuroreport 18(15):1595–1598
McKay RT (2009) Recent advances in solvent suppression for solution NMR: a practical
reference. In: Webb G (ed) Annual reports on NMR spectroscopy. Elsevier, pp 33–76
McKay RT (2011) How the 1D-NOESY suppresses solvent signal in metabonomics NMR spec-
troscopy: an examination of the pulse sequence components and evolution. Concepts Magn
Reson A 38A(5):197–220
Metabolomics and NMR 113
Tayyari F et al (2013) 15N-cholamine – a smart isotope tag for combining NMR- and MS-based
metabolite profiling. Anal Chem 85(18):8715–8721
Teng Q (2012) Structural biology: practical NMR applications. Springer Science & Business Media
Tenori L, Turano P, Luchinat C (2020) Metabolic profiling by NMR. eMagRes 9:199–204
Tilgner M et al (2019) High-resolution magic angle spinning (HRMAS) NMR methods in
metabolomics. In: NMR-based metabolomics. Springer, pp 49–67
Todd JFJ, March RE (1999) A retrospective review of the development and application of the
quadrupole ion trap prior to the appearance of commercial instruments. Int J Mass Spectrom
190:9–35
Torchia DA (2009) Slight mistuning of a cryogenic probe significantly perturbs the water 1H
precession frequency. J Biomol NMR 45(3):241–244
Tredwell GD et al (2011) Between-person comparison of metabolite fitting for NMR-based
quantitative metabolomics. Anal Chem 83(22):8683–8687
Trifonova OP et al (2021) Mass spectrometry-based metabolomics diagnostics–myth or reality.
Expert Rev Proteomics 18(1):7–12
Valdés A et al (2021) Foodomics: analytical opportunities and challenges. Anal Chem 94(1):
366–381
van Bentum J et al (2016) Perspectives on DNP-enhanced NMR spectroscopy in solutions. J Magn
Reson 264:59–67
van der Sar SA et al (2015) Ethanol contamination of cerebrospinal fluid during standardized
sampling and its effect on (1)H-NMR metabolomics. Anal Bioanal Chem
van Zijl PCM (1987) The use of deuterium as a nucleus for locking, shimming, and measuring
NMR at high magnetic fields. J Magn Reson (1969) 75(2):335–344
van Zijl PC, Hurd RE (2011) Gradient enhanced spectroscopy. J Magn Reson 213(2):474–476
Van Zijl PCM et al (1994) Optimized shimming for high-resolution NMR using three-dimensional
image-based field mapping. J Magn Reson 111:203–207
Van QN, Chmurny GN, Veenstra TD (2003) The depletion of protein signals in metabonomics
analysis with the WET-CPMG pulse sequence. Biochem Biophys Res Commun 301(4):
952–959
Vignoli A et al (2019) High-throughput metabolomics by 1D NMR. Angew Chem Int Ed 58(4):
968–994
Visconti A et al (2019) Interplay between the human gut microbiome and host metabolism. Nat
Commun 10(1):1–10
Vögeli B (2014) The nuclear Overhauser effect from a quantitative perspective. Prog Nucl Magn
Reson Spectrosc 78:1–46
Wang X, Li L (2020) Mass spectrometry for metabolome analysis. Mass Spectrom Lett 11(2):
17–24
Wang H et al (2010) Metabolomics and detection of colorectal cancer in humans: a systematic
review. Future Oncol 6(9):1395–1406
Webb AG (2006) Advances in probe design for protein NMR. In: Annual reports on NMR
spectroscopy. Elsevier, pp 1–50
Weljie AM et al (2006) Targeted profiling: quantitative analysis of 1H NMR metabolomics data.
Anal Chem 78(13):4430–4442
Wilson MJ (2007) The ARRL operating manual for radio amateurs. American Radio Relay League
Wolfender JL et al (2013) Plant metabolomics: from holistic data to relevant biomarkers. Curr Med
Chem 20(8):1056–1090
Wüthrich K (1986) NMR of proteins and nucleic acids, vol 17. Wiley, New York, pp 1–292
Xiao C et al (2009) An optimized buffer system for NMR-based urinary metabonomics with
effective pH control, chemical shift consistency and dilution minimization. Analyst 134(5):
916–925
Xu R et al (1999) Chemical ligation of folded recombinant proteins: segmental isotopic labeling of
domains for NMR studies. Proc Natl Acad Sci 96(2):388–393
Zangger K (2015) Pure shift NMR. Prog Nucl Magn Reson Spectrosc 86–87:1–20
116 R. T. McKay
Contents
1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 118
2 Chemical Profiling Techniques in Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
3 Statistical Methods to Correlate Between Chemical Profiles and Bioactivity . . . . . . . . . . . . . 125
3.1 Correlation with a Simplified Dataset: Discriminant Analysis . . . . . . . . . . . . . . . . . . . . . . . 127
3.2 Correlation Analysis with Non-Discriminant Variables . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
4 Application of Metabolomics-Based Bioactivity to Various Disease Models . . . . . . . . . . . . . 130
4.1 Antimicrobial Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 130
4.2 Anticancer Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 131
4.3 Anti-inflammatory Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
5 Conclusions and Perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138
Abstract
Natural products have been the most important source for drug development
throughout the human history. Over time, the formulation of drugs has evolved
from crude drugs to refined chemicals. In modern drug discovery, conventional
natural products lead-finding usually uses a top-down approach, namely
bio-guided fractionation. In this approach, the crude extracts are separated by
# The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 117
V. Ghini et al. (eds.), Metabolomics and Its Impact on Health and Diseases,
Handbook of Experimental Pharmacology 277, https://doi.org/10.1007/164_2022_611
118 H. K. Kim et al.
Keywords
1 Introduction
Since ancient times mankind has searched for medicinal plants in nature, in early
times very likely by simple trial and error which eventually evolved into complex
systems that included the documentation of the accrued knowledge which allowed
its organized transmission from generation to generation. This is known as tradi-
tional medicine, some outstanding examples of which are the traditional Chinese
Medicine (TCM) and Ayurveda in India. The traditional medicine of the Mediterra-
nean region became the basis of Western medicine (Leonti and Verpoorte 2017).
Thanks to the technological advances in processing and the accumulation of clinical
information, the application of natural products developed from the direct use of a
medicinal plant to the use of extracts, and then to pure compounds. Processing steps
including grinding, drying, fermentation, extraction, heating, and the addition of
other plants or materials (e.g., milk, butter, honey, sugar) have been, and still are,
worldwide common practices in traditional medicine. This information is important
for the transformation of the transmitted traditional knowledge into novel leads for
medicines.
Natural Products Drug Discovery: On Silica or In-Silico? 119
degradation. But the greatest limitation of this method is its intrinsic ignorance of
synergistic effects and prodrugs. The single target-single compound paradigm
appears to contradict the base of traditional medicines that often relies on complex
mixtures of plants in which every plant has a different function. That means that the
multitarget-multicomponent approach is the key difference between traditional and
modern drug development. It is obvious that such a systemic approach to drug
discovery demands different tools. In the case of therapeutic activity it means a
multitarget-testing system, i.e. the use of living organisms. For the identification of
active compounds, a holistic approach such as metabolomics can provide the type of
information required to identify correlations between activities and metabolites,
thanks to the application of chemometric tools (see below).
Metabolomics aims at profiling all the metabolites in an organism. To these ends,
it provides an overview of the metabolic profiles of the subject of interest in an
untargeted manner, that is, ultimately, a systems biology approach (Wang et al.
2005). This systems biology approach using omics tools gained importance in the
quest for biomarkers for diseases and other situations that require the identification
of distinctive markers, e.g. quality standards. The experimental design for this
approach includes processing a large number of representative samples (e.g., plant
extracts or fractions) to obtain both metabolomics and activity data of each one.
Numerous chemometric methods such as multivariate data analysis have been
developed to identify potential correlations between all observations and identify
indicators (e.g., NMR signals, LC-MS or GC-MS peaks) that correlate with a given
activity. In this approach, any variable that has been measured can be used to test for
possible correlations. Metabolomics, eventually in combination with proteomics,
transcriptomics, and/or genomics, can also be used to identify potential targets in the
tested organisms used in bioassays, e.g. cell lines or test organisms such as mice
(Parng et al. 2002), zebra fish (Danio rerio) (Mushtaq et al. 2013; Akhtar et al.
2016), Caenorhabditis elegans (Salzer and Witting 2021), and brine shrimp
(Artemia salina) (Ntungwe et al. 2020), it is possible to gain better insight into the
possible mode(s) of action of an extract or a pure compound. This would be a
molecular follow-up of the classic Hippocratic screening. The method must be
validated by measuring the effect of various classes of drugs on the metabolome
and a database of the changes in the metabolome can then be used to compare with
the effects of extracts or novel compounds.
Finally, there is also the possibility of coupling a bioassay with the separation of
extracts. For example, inhibition of acetylcholine esterase activity can be coupled
with HPLC (Ingkaninan et al. 2000) or TLC (Rhee et al. 2003). Particularly in the
case of TLC, its coupling with various bioactivity screening methods has shown to
be a powerful tool for nano-scale identification of biologically active compounds
(Klingelhofer et al. 2021; Morlock 2021; Morlock et al. 2021; Schreiner and
Morlock 2021). In this case, though, the use of a single compound, single target
approach, makes it somewhat less attractive for the study of complex traditional
medicines, where synergy most likely plays a predominant role (Verpoorte et al.
2018). As seen in the study of Artemisia annua, the antimalarial activity observed in
tea extract cannot be explained only by the already-known active compound
Natural Products Drug Discovery: On Silica or In-Silico? 121
Lead
Clinical studies
Production
Synthesis
New medicine Formulation
Quality control
Biotechnology
Network pharmacology
Fig. 1 Different steps in drug discovery and examples of metabolomics applied in this process.
Adapted from Cuperlovic-Culf and Culf (2016)
artemisinin, as its concentration is too low in the tea extract to be responsible for the
activity. The possible explanation is the presence of other active compounds such as
flavonoids or saponins which could have a synergistic effect with artemisinin
(De Donno et al. 2012). The limitation of this fully “on-silica” approach is that it
is based on the single target-single compound approach, so any activity involving
synergy and other interactions between molecules will not be detected, whereas the
“in-silico” approach can reveal synergistic effects on other types of interaction-based
mechanisms. The “in-silico” tools require the building of databases for fast identifi-
cation of known compounds in extracts, and metabolomics databases of the changes
caused by a standard set of major drugs in the in-vivo test systems. Network
pharmacology would be useful to support such databases. Network pharmacology
brings together all information on the effect of known drugs on the network of
metabolism, signaling and diseases, on the level of genome, transcriptome, prote-
ome, and metabolome. It can be used to obtain an insight into the mode of action of
medicines and evaluate the differences and similarities of new drugs with existing
ones (Boezio et al. 2017; Hopkins 2007, 2008; Jiang et al. 2021; Li and Zhang 2013;
Ye et al. 2016). In Fig. 1 the various aspects of drug development are summarized,
and the applications of metabolomics in these fields are highlighted. There are,
basically, two types of applications. One applies metabolomics to various test
organisms and patients aiming to identify markers for diseases and for the effects
of (novel) medicines on their evolution. In the other case, metabolomics is used to
identify active compounds in complex mixtures, e.g. traditional medicines. The
systems biology approach opens new windows into the interactions between
mixtures of active molecules and the test organisms providing even new insights
122 H. K. Kim et al.
Chromatography became a major game changer in life sciences in the 1960s (TLC
and GC) and 1970s (HPLC). An example of metabolomics avant-la-lettre is a paper
by Baerheim Svendsen and Karlsen (1967) on the GC analysis of the essential oils of
three different plants. However, in the 1980s this was considered to be like
“collecting stamps.” Fifteen years later, this sort of analysis was called metabolomics
and included under the umbrella of “omics” technologies, in which the understand-
ing of the function of genes was studied by the integration of genomics (DNA),
transcriptomics (RNA), proteomics (proteins), and metabolomics data. The DNA
and RNA molecules share similar physicochemical properties, enabling highly
reproducible analyses based on strict robust extraction protocols. This is not the
case of proteins that have a wide range of properties. In proteomics analysis, proteins
are divided into two major classes, according to their solubility in water in hydro-
philic and hydrophobic proteins and have thus different extraction protocols. In
metabolomics, the situation is even more complex due to the great difference in
physicochemical properties of metabolites in general. These can stem from their
molecular size (e.g., polysaccharides, lignin) or even hydro/lipophilicity in the case
of the small molecules. Among the “small” molecules there are primary metabolites,
ubiquitous compounds that are found in all living cells but there are also specialized
metabolites that are generally species-specific and related to the survival of an
organism in its ecosystem. Altogether some 350,000 natural products are known
from various sources (e.g., plants, microbes, insects) (Banerjee et al. 2015).
Estimations of the number of specialized metabolites are mere speculations, but
knowing that the number of species of living organisms in the world is somewhere
between 10 and 100 million (Pimm et al. 1995) and assuming that every species
produces one unique compound, there should be between 10 and100 million
metabolites in the metabolome of the Earth. The total chemical space of natural
products is thus very much larger than that covered by our present knowledge. There
is still so much to be discovered!
This brings us to the problem of how to define a metabolome. We have just
referred to the metabolome of the planet earth, i.e., the sum of all the metabolomes of
all the organisms (dead or alive) and their environment. Each plant species has its
own metabolome, which is the sum of the metabolomes of all its organs, i.e., roots,
stem, and leaves, etc. These metabolomes correspond to that of the many different
cells in the plant. Even within the cell the different organelles have different
metabolomes. The metabolome of a species might be best defined as the measurable
Natural Products Drug Discovery: On Silica or In-Silico? 123
set of metabolites that an organism is able to produce, i.e. some compounds might
not always be present in certain plant parts, but their presence depends on the
existence of certain circumstances. The metabolome of a species depends on the
species’ genome. Obviously the single cell analysis at all omics levels is the holy
grail of the omics.
Clearly, any reference to metabolomics requires a clear documentation of the
source of the “metabolome.” The ultimate goal of metabolomics is to describe the
changes in the metabolome under well-defined conditions. Eventually the fluxes in
the organism through the various metabolic pathways should be analyzed
(fluxomics), as changes in the homeostasis of an organism are connected with
changes in fluxes.
To attain the goal of metabolomics, all applications have the same requirements,
i.e. both qualitative and quantitative information about all metabolites in the
biological sample must be generated. However, as per the discussion above it is
clear that the requirements vary according to the application and so will the method
needed to obtain the information. Firstly, when searching for biomarkers of diseases
or bioactivities, it is essential to be able to count on a large database with
metabolomic data of the test organisms to be used or material that has the desired
activity, e.g. extracts from a specific plant species with an interesting activity. This
database should ideally provide information on the effect of all biological variables
on the studied material, for example, the diurnal variation of the metabolome.
Existing databases with all kinds of information from previous experiments that
provide abundant background information can help in the identification of
compounds from their spectra and/or chromatographic data. However,
bioprospecting requires the screening of large numbers of plants that are
characterized by a highly variable and species-specific metabolome, particularly in
the case of specialized metabolism, a situation that is clearly different from the case
of mammalian cells, for example, in which most compounds are well-known
primary metabolites.
It is now generally accepted that no single analytical tool can reveal the real
metabolome (Wishart 2008; Emwas et al. 2019). At present, analytical chemists are
moving in two possible directions to circumvent this limitation, i.e., improving the
quality of individual techniques and/or integrating data from multiple methods.
In the case of NMR, the main limiting issues are its low sensitivity (μmol for
detection limit) and complexity of signals. Sensitivity has been increased using cryo
(cold)-probes and reducing the diameter of NMR tubes (capillary-, micro-, or nano-
tubes). Nevertheless, the sensitivity is still far below that of MS-based methods
(up to 10 times for conventional NMR probe). Generally speaking, NMR is consid-
ered to be the most powerful analytical tool for structure elucidation of pure
compounds but not necessarily in the case of mixture analysis. The most attractive
advantage of NMR as a metabolomics tool is its ease of quantitation. The height or
area of 1H NMR signals is directly proportional to the molar concentration of
analytes, i.e. with a single internal standard all signals can be easily integrated and
quantified. It is in this aspect that NMR has a clear edge over all other analytical
techniques.
124 H. K. Kim et al.
It should also be noted that NMR is currently the only metabolomics method that
does not include a separation step.
In MS-MS, the separation is in the first MS dimension, based on molecular mass,
after which the individual signals are subject to a second MS step in which the
molecules are subject to fragmentation. In hyphenated chromatography-MS
techniques, the retention time, molecular mass, and the fragmentation pattern can
be used to search databases for already-known compounds. For example, it is
possible to identify unknown peaks with high-resolution mass measurements in
combination with the “seven golden rules” or other “dereplication” approaches.
Though (stereo) isomers will be difficult to identify with 100% certainty, unfortu-
nately too often the identity of a compound is accepted ignoring the inherent level of
uncertainty of the used method. For a full identification of new or rare compounds,
their UV, MS, 1H NMR, 13C NMR, and various multidimensional NMR spectral
information must be analyzed. The 1D-NMR data alone are insufficient. Most cases
require multidimensional NMR methods such as COSY, TOCSY, HSQC, and
HMBC spectroscopy to determine distances and interactions between protons,
e.g. to confirm stereochemistry. Known compounds can be identified with a reason-
able certainty if isolated, but the full structural elucidation of new specialized
metabolites from natural products mixtures is still enormously challenging. The
technical limitations of the instrumental NMR analysis were partially solved by a
statistical signal correlation of 1H NMR resonances, the so-called statistical total
correlation spectroscopy (STOCSY) that has been used to identify novel metabolites
in urine, enabling the selection of signals characteristic of one molecule; there are, as
well, a multitude of deconvolution methods for NMR data (Cloarec et al. 2005). As
mentioned before, most of the chemical diversity of metabolites includes specialized
metabolites of which there are no reliable comprehensive NMR databases. However,
there are a number of available NMR spectra databases of primary metabolites
(Human Metabolome Database (HMDB, http://www.hmdb.ca), Biological Mag-
netic Resonance Bank (BMRB, http://www.bmrb.wisc.edu/metabolomics/),
NMRshiftDB (http://nmrshiftdb.ice.mpg.de/).
Mass spectrometry (MS) is a highly sensitive method (pmol level) and is also
selective due to the high resolution and level of accuracy of the determined molecu-
lar mass and the different fragmentation patterns of the metabolites. The number of
detected signals in MS-based platforms is 10–100-fold that of NMR. In terms of
identification, however, it has inherent limitations. The mass spectrometer only
detects ions formed in the ion source, but there can be a large variation in the
sensitivity for the formation of ions. That means that absolute quantitation is only
possible by running calibration curves for every single compound within certain
ranges of concentrations. Identification is possible by comparison of the exact
molecular mass of the compounds with databases and comparison of fragmentation
patterns with possible candidates and closely related compounds. The MS data is
insufficient for the structural elucidation of new compounds. For low molecular
mass values there are scores of isomers, thus for the elucidation of the full structure
of a molecule, including its stereochemistry, further spectral data are needed (see
above).
Natural Products Drug Discovery: On Silica or In-Silico? 125
The key step of OMICS-based chemical profiling is data mining using statistical
analysis. The metabolomics datasets generated by NMR or MS are vast, requiring
chemometrics to extract any useful information from the data. The chemometrics
methods usually include a statistics-focused approach and/or a computer-dominant
approach such as machine learning (Wishart 2008). Paul and de Boves Harrington
(2021) summarized the basic concepts and applications of those methods very
clearly.
In this section, we will focus on the statistical approach which is generally used to
detect the active compounds in natural products extracts. This approach uses two
different methods to link signals of compounds with activity: discriminant analysis
or correlation analysis.
126 H. K. Kim et al.
Discriminant analysis is nowadays the most popular approach for the identification
of active compounds in complex mixtures. Partial least squares discriminant analysis
(PLS-DA) is frequently applied to detect differences by making qualitative
classifications, such as active and non-active samples; it is based on the definition
of preferably two classes, and any type of classes can be formulated. PLS-DA, a
supervised method, models the variance within the dataset by statistically discrimi-
nating groups of observations. Orthogonal PLS (OPLS) was developed to improve
the correlation power of PLS by orthogonalizing non-related variables through
orthogonal signal correction (OSC filtering). This facilitates the interpretation of
the model because the variables (metabolome) are related to the targeted property
(e.g., high or low bioactivity).
A good example of this application, published by Cardoso-Taketa et al. (2008), is
the correlation of the sedative effect of Galphimia glauca Cav. and galphimine, a
triterpenoid already known to occur in this plant. Working on six different
collections of G. glauca they found two collections that were highly active, while
the rest did not show much activity. Applying PLS-DA to the NMR metabolomic
data and the sedative effects found in the animal model, they were able to correlate
the biological activity with galphimine.
A similar approach was applied to identify biologically active metabolites
obtained with different extraction methods and solvents from Ocotea odorifera
using MS-based metabolomics. Alcântara et al. (2021) investigated the anti-
inflammatory activity of a decoction of Ocotea odorifera and different fractions
from its ethanolic extracts using dual inhibition of edema and neurophil recruitment.
The chemical profiling data obtained by UPLC-HRMS or GC-MS were correlated
with the anti-inflammatory activity by PLS-DA, resulting in the identification of
S-(+)-reticuline as an active principle. Its activity was confirmed by testing the pure
compound after isolation.
Other examples of the successful application of this approach are the studies of
the antimicrobial activity of essential oils (Maree et al. 2014) and the identification of
an antitussive active compound from Tussilago farfara L. (Lia et al. 2013).
The work of Maree et al. (2014) showcases the strength of the chemometrics and
in particular of the OPLS-DA method. Based on the data obtained from the analysis
of 158 different essential oils by GC-MS and their antimicrobial activities on several
different strains of microorganisms, two classes were defined: active (MIC <2 mg/
ml) and non-active (MIC >2 mg/ml). This allowed the detection of eugenol as a
putative marker for activity, and an apparent synergistic effect with geraniol.
α-Pinene, limonene, and sabinene, other components of the essential oils correlated
with no activity. The study also revealed both antagonistic and synergistic antimi-
crobial effects between monoterpenes and eugenol on some microbes.
The correlation approach is clearly more efficient than conventional bioactivity-
guided fractionation, to identify active compounds in the initial stage of drug
development. The limitation of this application, however, lies in a tendency to
overfit if the number of variables considerably exceeds the number of samples
128 H. K. Kim et al.
was pumped through the ground dry plant material mixed with kieselguhr (7:1),
yielding between 20 and 30 fractions. Each fraction was profiled by 1H NMR and its
potential anti-obesity activity was assessed based on an adenosine A1 receptor
binding assay. Examination of the loading plots of the PLS and OPLS models led
to the identification of two active methoxyflavonoids, 4′,5,6,7-tetramethoxyflavone
(tetramethyl scutellarein) and 3′,4′,5,6,7- pentamethoxyflavone (sinensetin).
Another example is the work of Ali et al. (2013), who applied PLS and OPLS
analyses to correlate the metabolome of sponges obtained using NMR spectroscopy
with their effect in the adenosine A1 receptor binding activity assay. The
metabolome of over 200 sponge samples was obtained using 1H NMR, and their
adenosine A1 receptor binding activity was measured. OPLS analysis was
performed with two datasets. From the loading plots several signals were selected
as activity-associated. Most signals were unknown or difficult to identify only by
1
H-NMR, requiring the isolation of compounds. Having isolated several compounds
which matched well with activity-associated signals, these active metabolites were
then identified as the sesterterpenes, halisulfate -1, -3, -4, -5, and suvanine.
De Melo et al. (2020) studied the spasmolytic activity of Cissampelos
sympodialis Eichler leaf extract in a trachea preparation. While warifteine, a
bisbenzylisoquilonine alkaloid, is allegedly the main bioactive substance in this
species, its low solubility in polar solvents suggested the presence of another
bioactive compound in the aqueous extracts. To investigate this alternative, PLS
modeling was used to study the correlation of the metabolome obtained by NMR of
diverse polar extracts with spasmolytic activity. The PLS model showed that the
signals from flavonoids were positively correlated with the activity. Therefore, they
concluded that identified flavonoids such as kaempferol and quercetin might be the
important contributors to the activity.
Many studies have applied successfully the combination of metabolomics and
bioassays with chemometrics in the identification of biologically active compounds
in complex mixtures. Researchers can identify active metabolites from the crude
extracts without isolation of single compound with this approach. However, from a
practical point of view, the metabolomics approach itself is not enough to identify
active molecules. Ultimately, the bioactivity of a compound must be confirmed with
tests on the pure compound. In any case, and particularly in case of novel
compounds, proper identification requires the determination of the full set of physi-
cochemical properties and spectral data. Nonetheless, the advantages of the
metabolomics approach are clear. To begin with, it allows the fast dereplication in
case of already-known active compounds. Secondly, there is no need for large-scale
isolation of the active compounds, nor bioassays and, if necessary, fractionation can
be guided by the NMR or (LC- or GC-) MS spectra of the fractions. Particularly in
the case of in-vivo bioassays (including clinical trials) only one first round of
bioassays is needed, after which chemistry-guided fractionation is sufficient to
isolate the active compound. In many cases this may have the additional advantage
of reducing the number of animal experiments.
130 H. K. Kim et al.
Most infectious diseases are easy to diagnose and humans have been tirelessly
searching for medicines in nature to treat infections since ancient times. With the
discovery of penicillin in 1928 by Fleming, microorganisms have been targeted for
the discovery of other novel antibiotics. After a period of reduced interest, in recent
years there is a revival in antibiotic bioprospecting, mainly because of the rapid
spread of antibiotic-resistant pathogenic microorganisms and the failure to develop
active derivatives of the existing ones. The conventional lead-finding of antimicro-
bial drugs is bioactivity-guided fractionation much like for other disease models.
Aided by chemometrics, diverse in-silico methods have been recently introduced
into this field, most of which are applied in the post lead-finding steps such as hit
characterization and hit optimization (Xu et al. 2009). The application of
metabolomics in the process of lead-finding involves:
were negatively correlated with each other in terms of activity, suggesting antago-
nism of those compounds.
Streptomyces species are the most studied Actinomycetes for the production of
antibiotics. Wu et al. (2015a) applied NMR-based metabolomics to Actinomycetes
and rapidly discovered novel antibiotics. The metabolomic comparison of wild-type
and streptomycin-resistant actinomycetes strains revealed a stronger antimicrobial
activity of the latter. Using PLS analysis, 7-prenylisatin was identified as the
bioactive compound. The antimicrobial activity was confirmed by studies with the
pure isolated compound (Fig. 2).
Because of bacterial resistance to current antibiotics, it is extremely important to
find active compounds with a different mode of action to that of the current
antibiotics. Thus, the mode of action should be identified in an early stage of lead-
finding. A metabolomics approach in which the effect on the pathogenic
microorganisms of known antibiotics and novel candidates is compared could
provide an insight into the mode of action (Hoerr et al. 2016). Clearly, metabolomics
is an important tool that should speed up the process of finding novel antibiotics.
While not directly connected to the correlation between chemical ingredients and
antibiotic activity, the possibility of the induction of new antibiotics in a co-culture
system (Streptomyces and Aspergillus) was probed using an analogous approach.
Though well-known as a source of antibiotics, the Streptomyces metabolome is very
sensitive to a number of external conditions. By co-culturing two different species,
the interactions between the organisms may induce the production of novel
compounds in either species. Moreover, catabolism of the compounds formed may
add even further chemodiversity. Wu et al. (2015b) showed that the co-culture of
Streptomyces with Aspergillus produced significant modifications in their individual
biosynthetic processes yielding many new metabolites which could not be detected
in the individual organisms.
Natural products and their derivatives have a tremendous potential for the develop-
ment of anticancer drugs. More than 50% of all anticancer drugs approved for
therapeutic use in the past 70 years are either natural product or semi-synthetic
analogs (Butler et al. 2014). Metabolomics has been used not only for drug devel-
opment, but also in many fields related to cancer research including cancer progno-
sis, diagnosis, and treatment efficacy. The in-silico approach described above has
been applied to the search of new lead compounds in many natural resources. An
interesting example of its application in cancer research is reported by Graziani et al.
(2018). Extracts of 14 legumes were screened against an array of human colorectal
cancer cell lines in the search for bioactive natural products. Two plant extracts were
selected for their strong antiproliferative activity using PCA and Hierarchical Cluster
Analysis (HCA) (Fig. 3). The 2D NMR analysis of these plant extracts allowed the
identification of two putative active compounds: a cycloartane glycoside and a
protodioscin derivative. To confirm their bioactivity, these two compounds were
132 H. K. Kim et al.
Fig. 2 Example of multivariate data analysis. PLS score plot (a) shows a good separation between
wild-type Streptomyces sp. (blue) and its two mutant derivatives (MBT28-30: light blue and
MBT28-91: red). The corresponding loading plot (b) presents the NMR signals (dashed circles)
which contribute mostly to the separation of the samples and their bioactivity. The arrow refers to
the characteristic proton signal at δ 7.03 (t, J = 7.2 Hz). X, primary variable of the chemical shift; Y,
bioactivity. (Adopted from Wu et al. 2015a)
Natural Products Drug Discovery: On Silica or In-Silico? 133
Fig. 3 Data of cell growth percentage were analyzed by HCA dendrogram (a) and PCA (b).
Control and colon cancer cell lines which were treated with the plant extracts were distinguished.
These analyses enabled classification of the species into 3 subsets; Groups I – the active species
134 H. K. Kim et al.
isolated and tested positively for antiproliferative activity against colon cancer cells.
These results are clear evidence of the efficiency of metabolomics as a tool to speed
up the lead-finding process, since the active compounds are already identified in the
crude extracts avoiding the time-consuming on-silica isolation procedure.
Using a similar approach, Bao et al. (2018) applied UPLC-MS-based
metabolomics to identify anticancer compounds in Forsythiae fructus (from For-
sythia suspensa Vahl). Crude extracts obtained with diverse solvents were profiled
and the results were correlated with their anticancer activity against murine mela-
noma B16-F10 cell lines. OPLS-DA was applied to maximize the discrimination of
extracts with different degrees of anticancer activity resulting in the selection of
betulinic acid as a potential anticancer compound in Forsythiae fructus.
Gao et al. (2010) performed antiproliferative activity tests against human lung
cell line SK-MES-1 on Scutellaria baicalensis Georgi root extracts. The data
provided by the HPLC-UV and 1H NMR analysis of the extracts were combined
with PCA and PLS analysis for more complete metabolomics data, allowing the
identification of baicalin, baicalein, and wogonin as the compounds responsible for
the cell growth inhibition activity of the extracts.
Tawfike et al. (2019) adopted a different approach to study anticancer activity of
the endophytic fungus Aspergillus flocculus, isolated from the stem of the medicinal
plant Markhamia lutea (Benth.) K.Schum. (Markhamia platycalyx is a synonym). Its
in-vitro culture was found to yield high levels of anticancer compounds active
against the chronic myelogenous leukemia cell line K562. Several fractions of the
fungal culture were analyzed by LC-MS. A combination of a molecular interaction
network and OPLS-DA of the chemical data resulted in the identification of five
active metabolites, namely, cis-4-hydroxymellein, 5-hydroxymellein, diorcinol,
botryoisocoumarin A, and mullein. This study clearly shows the advantage of
combining different forms of in-silico analysis prior to any purification attempts.
Another successful study using this approach was recently published by Ory et al.
(2019) involving an anti-breast cancer compound (tested on MCF-7 cells) from a
marine-derived Penicillium chrysogenum extract. Using different correlation and
discriminant analysis including PLS-DA and PLS modeling, ergosterol was found to
have anticancer activity with an antiproliferative activity on MCF-7 cells with an
IC50 at 0.10 mM concentration. An interesting aspect of this report is the description
of a workflow that allows the combination of both the chemical and biological data, a
key step in this sort of application. Sometimes, a metabolomics approach alone is not
sufficient to identify active metabolites in the crude extracts due not only to the
complexity of the mixture but also to the low concentration levels of potentially
interesting metabolites. In that case, conventional bio-guided fractionation can be
useful as a supplementary tool. The work of Graziani et al. (2021) is a good example
of this. They evaluated the anticancer activity of two plant species, Ononis diffusa
Fig. 3 (continued) (black), group II – active only at the highest tested doses (gray), and group III –
no significant effect (white). Adapted from Graziani et al. (2018)
Natural Products Drug Discovery: On Silica or In-Silico? 135
Ten. and O. variegata L. using colorectal cancer cell lines. When the NMR-based
metabolomics data were correlated with the anticancer activity, most of the poten-
tially interesting activity related signals were concentrated in the aliphatic region of
the NMR spectra, a region which is difficult to annotate. The partial purification of
the extracts using column chromatography and the application of different 2D NMR
techniques led to the identification of a mixture of oxylipins as putative bioactive
compounds. However, the report provides no information on activity confirmation of
pure isolated compounds.
Gao et al. (2010) reported the application of metabolomics to study the mode of
action of (-)-5-hydroxyequol, an isoflavone metabolite obtained by microbial
biotransformation known to affect lung cancer cells through an unknown mechanism
of action. Applying 1H NMR-based metabolomics it was possible to observe rapid
changes in the metabolism in human lung cancer cells, especially in the
glycometabolism.
References
Akhtar MHT, Mushtaq MY, Verpoorte R et al (2016) Zebrafish as a model for systems medicine
R&D: rethinking the metabolic effects of carrier solvents and culture buffers determined by 1H
NMR metabolomics. OMICS 20:42–52. https://doi.org/10.1089/omi.2015.0119
Alcântara BGV, Oliveira FP, Katchborian-Neto A et al (2021) Confirmation of
ethnopharmacological anti-inflammatory properties of Ocotea odorifera and determination of
its main active compounds. J Ethnopharmacol 264:113378. https://doi.org/10.1016/j.jep.2020.
113378
Ali K, Iqbal M, Yuliana ND et al (2013) Identification of bioactive metabolites against adenosine
A1 receptor using NMR-based metabolomics. Metabolomics 9:778–785. https://doi.org/10.
1007/s11306-013-0498-9
Allard PM, Peŕesse T, Bisson J et al (2016) Integration of molecular networking and in-silico
MS/MS fragmentation for natural products dereplication. Anal Chem 88:3317–3323. https://
doi.org/10.1021/acs.analchem.5b04804
Baerheim Svendsen A, Karlsen J (1967) Gaschromatographie von Monoterpenkohlenwasserstoffen
aus Aetherischen Oelen an gepackten Trennsaulen mit niedrigem Gehalt an fluessiger
stationaere Phase. Planta Med 15:1–5. https://doi.org/10.1055/s-0028-1099949
Banerjee P, Erehman J, Gohlke BO et al (2015) Super natural II – a database of natural products.
Nucleic Acids Res 43:D935–D939. https://doi.org/10.1093/nar/gku886
Bao JL, Ding RB, Jia XJ et al (2018) Fast identification of anticancer constituents in Forsythiae
Fructus based on metabolomics approaches. J Pharm Biomed Anal 154:312–320. https://doi.
org/10.1016/j.jpba.2018.03.020
Boezio B, Audouze K, Ducrot P et al (2017) Network-based approaches in pharmacology. Mol
Informatics 36:1700048. https://doi.org/10.1002/minf.201700048
Butler MS, Robertson AAB, Cooper MA (2014) Natural product and natural product derived drugs
in clinical trials. Nat Prod Rep 31:1612–1661. https://doi.org/10.1039/c4np00064a
Cardoso-Taketa AT, Pereda-Miranda R, Choi YH et al (2008) Metabolic profiling of the Mexican
anxiolytic and sedative plant Galphimia glauca using nuclear magnetic resonance spectroscopy
and multivariate data analysis. Planta Med 74:1295–1301. https://doi.org/10.1055/s-
2008-1074583
Chen L, Deng H, Cui H et al (2017) Inflammatory responses and inflammation-associated diseases
in organs. Oncotarget 9:7204–7218. https://doi.org/10.18632/oncotarget.23208
Cloarec O, Dumas ME, Craig A et al (2005) Statistical total correlation spectroscopy: an explor-
atory approach for latent biomarker identification from metabolic 1H NMR data sets. Anal Chem
77:1282–1289. https://doi.org/10.1021/ac048630x
Cragg GM, Newman DJ (2005) Plants as a source of anti-cancer agents. J Ethnopharmacol 100:72–
79. https://doi.org/10.1016/j.jep.2005.05.011
Cuperlovic-Culf M, Culf AS (2016) Applied metabolomics in drug discovery. Expert Opin Drug
Discovery 11(8):759–770. https://doi.org/10.1080/17460441.2016.1195365
De Donno A, Grassi T, Idolo A et al (2012) First-time comparison of the in vitro antimalarial
activity of Artemisia annua herbal tea and artemisinin. Trans R Soc Trop Med Hyg 106:696–
700. https://doi.org/10.1016/j.trstmh.2012.07.008
de Melo ICAR, de Souza ILL, Vasconcelos LHC et al (2020) Metabolomic fingerprinting of
Cissampelos sympodialis Eichler leaf extract and correlation with its spasmolytic activity. J
Ethnopharmacol 253:112678. https://doi.org/10.1016/j.jep.2020.112678
Natural Products Drug Discovery: On Silica or In-Silico? 139
Domingos OD, Alcântara BGV, Santos MFC et al (2019) Anti-inflammatory derivatives with dual
mechanism of action from the metabolomic screening of Poincianella pluviosa. Molecules
24:4375. https://doi.org/10.3390/molecules24234375
Donlan RM, Costerton JW (2002) Biofilms: survival mechanisms of clinically relevant
microorganisms. Clin Microb Rev 15:167–193. https://doi.org/10.1128/CMR.15.2.167-193.
2002
dos Santos FA, Sousa IP, Furtado NAJC et al (2018) Combined OPLS-DA and decision tree as a
strategy to identify antimicrobial biomarkers of volatile oils analyzed by gas chromatography–
mass spectrometry. Rev Bras Farm 28:647–653. https://doi.org/10.1016/j.bjp.2018.08.006
Emwas AH, Roy R, McKay RT, Tenori L, Saccenti E, Gowda GAN, Raftery D, Alahmari F,
Jaremko L, Jaremko M, Wishart DS (2019) NMR spectroscopy for metabolomics research.
Metabolites 9(7):123. https://doi.org/10.3390/metabo9070123
Gao JY, Zhao HY, Hylands PJ et al (2010) Secondary metabolite mapping identifies Scutellaria
inhibitors of human lung cancer cells. J Pharm Biomed Anal 53:723–728. https://doi.org/10.
1016/j.jpba.2010.04.019
Graziani V, Scognamiglio M, Belli V et al (2018) Metabolomic approach for a rapid identification
of natural products with cytotoxic activity against human colorectal cancer cells. Sci Rep 8:
5309. https://doi.org/10.1038/s41598-018-23704-9
Graziani V, Potenza N, D’Abrosca B et al (2021) NMR profiling of Ononis diffusa identifies
cytotoxic compounds against cetuximab-resistant colon cancer cell lines. Molecules 26:3266.
https://doi.org/10.3390/molecules26113266
Gromski PS, Muhamadali H, Di E et al (2015) A tutorial review: metabolomics and partial least
squares-discriminant analysis – a marriage of convenience or a shotgun wedding. Anal Chim
Acta 879:10–23. https://doi.org/10.1016/j.aca.2015.02.012
Hoerr V, Duggan GE, Zbytnuik L et al (2016) Characterization and prediction of the mechanism of
action of antibiotics through NMR metabolomics. BMC Microbiol 16:82. https://doi.org/10.
1186/s12866-016-0696-5
Hopkins AL (2007) Network pharmacology: network biology illuminates our understanding of
drug action. Nat Biotechnol 25:1110–1111. https://doi.org/10.1038/nbt1007-1110
Hopkins AL (2008) Network pharmacology: the next paradigm in drug discovery. Nat Chem Biol
4:682–690. https://doi.org/10.1038/nchembio.118
Ingkaninan K, de Best CM, van der Heijden R et al (2000) HPLC with on-line coupled UV, mass
spectrometric and biochemical detection for identification of acetylcholinesterase inhibitors
from natural products. J Chromatogr A 872:61–73. https://doi.org/10.1016/s0021-9673(99)
01292-3
Jeffryes JG, Colastani RL, Elbadawi-Sidhu M et al (2015) MINEs: open access databases of
computationally predicted enzyme promiscuity products for untargeted metabolomics. J
Chem Inform 7:44. https://doi.org/10.1186/s13321-015-0087-1
Jiang H, Hu C, Chen M (2021) The advantages of connectivity map applied in traditional Chinese
medicine. Front Pharmacol 12:474267. https://doi.org/10.3389/fphar.2021.474267
Kim GD, Lee JY, Auh JH (2019) Metabolomic screening of anti-inflammatory compounds from the
leaves of Actinidia arguta (Siebold & Zucc.) Planch. ex Miq. (Hardy Kiwi). Foods 8:47. https://
doi.org/10.3390/foods8020047
Klingelhofer I, Ngoc LP, van der Burg B et al (2021) A bioimaging system combining human
cultured reporter cells and planar chromatography to identify novel bioactive molecules. Anal
Chim Acta 1183:338956. https://doi.org/10.1016/j.aca.2021.338956
Leonti M, Verpoorte R (2017) Traditional mediterranean and European herbal medicines. J
Ethnopharmacol 199:161–167. https://doi.org/10.1016/j.jep.2017.01.052
Li S, Zhang B (2013) Traditional Chinese medicine network pharmacology: theory, methodology
and application. Chin J Nat Med 11:0110–0120. https://doi.org/10.1016/S1875-5364(13)
60037-0
140 H. K. Kim et al.
Lia ZY, Zhi HJ, Zhang FS et al (2013) Metabolomic profiling of the antitussive and expectorant
plant Tussilago farfara L. by nuclear magnetic resonance spectroscopy and multivariate data
analysis. J Pharm Biomed Anal 75:158–164. https://doi.org/10.1016/j.jpba.2012.11.023
Malone MH, Robichaud RC (1962) A Hippocratic screen for pure or crude drug materials. Lloydia
25:320–332
Maree J, Kamatou G, Gibbons S et al (2014) The application of GC–MS combined with
chemometrics for the identification of antimicrobial compounds from selected commercial
essential oils. Chemom Intel Lab Syst 130:172–181. https://doi.org/10.1016/j.chemolab.2013.
11.004
Morlock GE (2021) High-performance thin-layer chromatography combined with effect directed
assays and high-resolution mass spectrometry as an emerging hyphenated technology: a tutorial
review. Anal Chim Acta 1180:338644. https://doi.org/10.1016/j.aca.2021.338644
Morlock GE, Drotleff L, Brinkmann S (2021) Miniaturized all-in-one nanoGIT(+active) system for
on-surface metabolization, separation and effect imaging. Anal Chim Acta 1154:33830. https://
doi.org/10.1016/j.aca.2021.338307
Mushtaq MY, Verpoorte R, Kim HK (2013) Zebrafish as a model for systems biology. Biotechnol
Gen Engin Rev 29:187–205. https://doi.org/10.1080/02648725.2013.801238
Newman DJ, Cragg GM (2007) Natural products as sources of new drugs over the last 25 years. J
Nat Prod 70:461–477. https://doi.org/10.1021/np068054v
Newman DJ, Cragg GM (2020) Natural products as sources of new drugs over the nearly four
decades from 01/1981 to 09/2019. J Nat Prod 83:770–803. https://doi.org/10.1021/acs.jnatprod.
9b01285
Ntungwe NE, Dominguez-Martin EM, Roberto A et al (2020) Artemia species: an important tool to
screen general toxicity samples. Curr Pharm Design 26:2892–2908. https://doi.org/10.2174/
1381612826666200406083035
Ory L, Nazih EH, Daoud S et al (2019) Targeting bioactive compounds in natural extracts –
development of a comprehensive workflow combining chemical and biological data. Anal Chim
Acta 1070:29–42. https://doi.org/10.1016/j.aca.2019.04.038
Parng C, Seng WL, Semino C et al (2002) Zebrafish: a preclinical model for drug screening. Assay
Drug Dev Technol 1:41–48. https://doi.org/10.1089/154065802761001293
Paul A, de Boves Harrington P (2021) Chemometric applications in metabolomic studies using
chromatography-mass spectrometry. Trends Anal Chem 135:116165. https://doi.org/10.1016/j.
trac.2020.116165
Pimm SL, Russell GJ, Gittleman JL, Brooks TM (1995) The future of biodiversity. Science
269:347–350. https://doi.org/10.1126/science.269.5222.347
Raŕova L, Ncube B, Van Staden J et al (2019) Identification of narciclasine as an in vitro anti-
inflammatory component of Cyrtanthus contractus by correlation-based metabolomics. J Nat
Prod 82:1372–1376. https://doi.org/10.1021/acs.jnatprod.8b00973
Rhee IK, van Rijn RM, Verpoorte R (2003) Qualitative determination of false-positive effects in the
acetylcholinesterase assays using thin layer chromatography. Phytochem Anal 14:127–131.
https://doi.org/10.1002/pca.675
Salzer L, Witting M (2021) Quo Vadis Caenorhabditis elegans metabolomics-a review of current
methods and applications to explore metabolism in the nematode. Metabolites 11:284. https://
doi.org/10.3390/metabo11050284
Samuelsson G, Bohlin L (eds) (2009) Drugs of natural origin: a treatise of pharmacognosy. Swedish
Academy of Pharmaceutical Sciences, Stockholm
Schreiner T, Morlock GE (2021) Non-target bioanalytical eight-dimensional hyphenation including
bioassay, heart-cut trapping, online desalting, orthogonal separations and mass spectrometry. J
Chromatogr 1647:462154. https://doi.org/10.1016/j.chroma.2021.462154
Szymańska E, Saccenti E, Smilde AK et al (2012) Double-check: validation of diagnostic statistics
for PLS-DA models in metabolomics studies. Metabolomics 8:3–16. https://doi.org/10.1007/
s11306-011-0330-3
Natural Products Drug Discovery: On Silica or In-Silico? 141
Contents
1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144
2 Quantitation Approaches Using NMR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 146
2.1 Internal Reference Standards for Absolute Quantitation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 146
2.2 Alternative Reference Standards for Absolute Quantitation . . . . . . . . . . . . . . . . . . . . . . . . . 149
2.3 Quantitation of Metabolites Using Intact Samples . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149
2.3.1 Intact Serum and Plasma Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
2.3.2 Intact Urine Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
2.3.3 Intact Tissue Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 151
2.4 Metabolite Quantitation Using Processed Samples . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152
2.4.1 Analysis of Aqueous Metabolites . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152
2.4.2 Analysis of Coenzymes and Antioxidants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 154
2.4.3 Analysis of Lipids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 156
2.5 Quantitation Methods Using Stable Isotope Labeling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 157
2.5.1 Isotope Labeling Focused on Metabolic Fluxes and Pathways . . . . . . . . . . . . . . 157
2.5.2 Isotope Labeling Focused on Metabolite Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . 157
3 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 158
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 159
D. Raftery (✉)
Northwest Metabolomics Research Center, University of Washington, Seattle, WA, USA
Mitochondria and Metabolism Center, Anesthesiology and Pain Medicine, University of
Washington, Seattle, WA, USA
Fred Hutchinson Cancer Research Center, Seattle, WA, USA
e-mail: [email protected]
# The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 143
V. Ghini et al. (eds.), Metabolomics and Its Impact on Health and Diseases,
Handbook of Experimental Pharmacology 277, https://doi.org/10.1007/164_2022_612
144 G. A. Nagana Gowda and D. Raftery
Abstract
Keywords
Fast NMR methods · Isotope tagging · Metabolomics · Nuclear magnetic
resonance (NMR) · Quantitation
1 Introduction
The key steps involved in almost all metabolomics investigations include metab-
olite detection, unknown peak identification, and quantification. Relative or absolute
concentrations of metabolites thus obtained are then subjected to statistical and/or
metabolic pathway analysis focused on a wide variety of applications in the areas of
basic and medical sciences. Typically, metabolite data are analyzed using univariate
and multivariate statistical analysis focused on the discovery and validation of
putative metabolite biomarkers. Alternatively, metabolite levels or isotope labeled
metabolites are used for identifying the perturbed metabolic pathways, which pro-
vide mechanistic understanding of cellular functions including information on drug
targets for therapeutic development.
Many compounds (>25) have been evaluated as potential internal standards for
applications in numerous areas including organic chemistry, natural product chem-
istry, agriculture, drug discovery, and pharmaceuticals (Maniara et al. 1998;
Holzgrabe 2010; Pauli et al. 2012; Rundlöf et al. 2010; Salem and Mossa 2012).
These compounds exhibit favorable physical characteristics, such as unique chemi-
cal shift, purity, stability, solubility, and suitability for accurate gravimetry.
Quantitative NMR Methods in Metabolomics 147
However, most of these are not suitable for metabolomics due to aqueous solubility
concerns or chemical shift overlap. Chemical shift reference compounds such as TSP
(trimethylsilylpropionic acid) and DSS (trimethylsilylpropanesulfonic acid) have
been used as internal standards for absolute quantitation of metabolites. It was
realized some years ago, however, that these compounds are unsuitable for quantita-
tion owing to their peak suppression arising from the interaction with proteins. One
alternative, formic acid, was evaluated as an alternative to TSP for quantitation of
metabolites in intact serum many years ago (Kriat et al. 1992). However, formic acid
is unsuitable as a reference since it is an endogenous metabolite; the endogenous
concentration in serum varies significantly from person to person (~40 to 350 μM)
and hence, it interferes with externally added formic acid (Kubáň and Boček 2013;
d'Alessandro et al. 1994; Kapur et al. 2007). In another study, DSA (4,4-dimethyl-4-
silapentane-1-ammonium trifluoroacetate), which is a derivative of DSS, was
evaluated as a potential internal standard using intact rat plasma (Alum et al.
2008). However, it is also unsuitable as a reliable internal standard since several
factors including the increased line broadening by a factor of >2 at pH 7.4 relative to
pH 3.0 indicate that DSA interacts with sample matrix. One remedy for analysis of
metabolites in samples such as blood serum/plasma that contain copious
macromolecules is to remove the macromolecules effectively by ultrafiltration; in
such a case, TSP or DSS can still be used as standards for absolute quantitation
(Psychogios et al. 2011; Barding et al. 2012; Simón-Manso et al. 2013). The
challenge with ultrafiltration, however, is that it attenuates many metabolite peaks
(Nagana Gowda and Raftery 2014b), requires larger sample volumes, and is cum-
bersome for large-scale studies. In addition, ultrafiltration cannot be used for analysis
of samples such as tissue and whole blood. Ultrafiltration is also incompatible with
MS analysis, the other major analytical platform used in metabolomics, since MS
analysis invariably employs protein precipitation to remove macromolecules, prior
to analysis (Nagana Gowda et al. 2018).
Protein precipitation that removes macromolecules from samples provides an
alternative approach to quantitate metabolites and is well suited for large-scale
studies. However, even in such samples, peaks from the traditional internal
standards, TSP and DSS, are attenuated by up to 35% and hence they are unsuitable
as internal standards. More recently, two compounds, maleic acid and fumaric acid,
were evaluated for their utility as potential internal standards for quantitation of
metabolites since both provide a single peak in NMR spectrum and their peaks do
not overlap with peaks from bio-specimen spectra (Nagana Gowda et al. 2021)
(Fig. 1). It was shown that fumaric acid is a robust standard for protein precipitated
serum, plasma, and whole blood; and maleic acid is suitable for plasma and serum,
but it overlaps with coenzyme peaks in whole blood samples. These findings provide
new opportunities for improved and accurate quantitation of metabolites in human
plasma, serum, and whole blood using NMR spectroscopy. The potential utility of
maleic acid and fumaric acid as internal standards may be extended to other
biological specimens, as long as they do not overlap with bio-specimen peaks.
148 G. A. Nagana Gowda and D. Raftery
TSP
Maleic acid
Fumaric acid
Serum
Plasma
Whole Blood
Fig. 1 Typical 800 MHz 1H NMR spectra of a protein precipitated sample of human (a) whole
blood, (b) plasma, and (c) serum, solubilized in D2O buffer containing a mixture of three internal
standards (TSP, 238 μM; maleic acid, 350 μM; fumaric acid, 293 μM). Each spectrum is overlaid
with a spectrum from the blank buffer consisting of the same three standards (spectrum shown in
red) to enable the visualization of peak heights for the three internal standards; the spectra of the
bio-specimens are slightly right shifted relative to the blank spectrum for clarity. Peaks from the
blank are marked with asterisks. Heights for the fumaric acid peaks from the bio-specimen
and blank are approximately matched; however, a significant attenuation of the TSP peak in all
three bio-specimens spectra is noticeable. TSP: Trimethylsilylpropionic acid-d4. (reproduced with
permission from Nagana Gowda et al. 2021)
Quantitative NMR Methods in Metabolomics 149
The ability to analyze intact samples with no need for sample preparation or
separation using NMR is an important characteristic that continues to drive
NMR-based metabolomics. Initially, widely used bio-specimens including blood
serum and plasma were used only in their intact forms and this approach continues to
be widely used. In the following sections, analyses of intact bio-specimens that are
most widely used such as serum/plasma, urine, and tissue are described. The
methods presented here are also applicable for other specimen types.
150 G. A. Nagana Gowda and D. Raftery
22 22
2:1 MeOH:Serum 2:1 ACN:Serum
20 20
18
3:1 MeOH:Serum 18 3:1 ACN:Serum
16 4:1 MeOH:Serum 16 4:1 ACN:Serum
14 14
(a) (e)
12 12
10 10
8 8
6 6
4 4
2 2
0 0
100 100
90 90
80 (b) 80 (f
70 70
60 60
50 50
40 40
30 30
20 20
10 10
0 0
450 450
400 400
350 (c) 350 (g)
300 300
250 250
200 200
150 150
100 100
50 50
0 0
Fig. 2 Comparison of absolute concentrations (in μM) of metabolites detected in pooled human
blood serum and quantitated using 800 MHz NMR spectroscopy after protein precipitation using
methanol (MeOH) (a, b, c and d) or acetonitrile (ACN) (e, f, g and h) at a solvent to serum ratios of
2:1, 3:1, and 4:1. Methanol performs most optimally over a wide range and a methanol to serum
ratio of 2:1 provides the best performance (reproduced with permission from Nagana Gowda et al.
2015)
154 G. A. Nagana Gowda and D. Raftery
(c)
NAD+ Acetyl CoA (b)
ATP CoA
NADH CoA-S-S-G
AMP
Acetyl-CoA NADPH
NADP+
CoA ADP
Gluta-
mate
Glutamine
Ace-tate
TSP
α-Glucose
Aromatic amino acids
β-Glucose
Lactate
BCCA
Fumarate
A
(a)
(e)
GSH (d)
Cr
GSSG
PCr
Fig. 3 (a) Typical 800 MHz 1H NMR spectrum of a mouse heart tissue extract with labeling of
some of the metabolites: BCCA: branched chain amino acids; TSP: reference peak; (b–e) expanded
spectral regions highlighting characteristic peaks for (b) coenzyme A (CoA), acetyl coenzyme A
(acetyl-CoA), and coenzyme A glutathione disulfide (CoA-S-S-G); (c) CoA, acetyl-CoA, oxidized
nicotinamide adenine dinucleotide (NAD+), oxidized nicotinamide adenine dinucleotide phosphate
(NADP+), reduced nicotinamide adenine dinucleotide (NADH), reduced nicotinamide adenine
dinucleotide phosphate (NADPH), adenosine triphosphate (ATP), adenosine diphosphate (ADP),
and adenosine monophosphate (AMP); (d) reduced glutathione (GSH) and oxidized glutathione
(GSSG); and (e) creatine (Cr) and phosphocreatine (PCr) (reproduced with permission from Nagana
Gowda et al. 2019)
156 G. A. Nagana Gowda and D. Raftery
additional avenues for metabolite identification (Chikayama et al. 2008; Bingol et al.
2012).
An altogether different approach is to label different classes of metabolites based
on the specific functional group (Shanaiah et al. 2007; Desilva et al. 2009; Ye et al.
2009). Chemical derivatization of metabolites using a substrate that contains isotope
such as 13C, 15N, or 31P offers both sensitivity and resolution enhancement, owing to
the high isotopic abundance and wide chemical shift dispersion imparted by the
incorporated isotope. The 1H decoupled 1D or 2D NMR spectrum involving the
isotope labeled heteronuclei provides a single peak for each metabolite, which
further adds to the sensitivity and resolution. Metabolite classes including amines,
carboxylic acids, and hydroxyls have thus been tagged with isotopes and analyzed
using 1D or 2D NMR (Shanaiah et al. 2007; Desilva et al. 2009; Ye et al. 2009;
Vicente-Muñoz et al. 2021). Owing to its high natural abundance, 31P signals from
metabolites, however, can show up as strong background peaks in the 31P enriched
experiments, unlike the other nuclei. Incorporation of a “smart isotope tag” such as
15
N-cholamine enables analysis of the carboxylic acid class of metabolites using
both NMR and MS methods (Tayyari et al. 2013). The smart isotope tag possesses
an NMR sensitive isotope (15N) that offers good chemical shift dispersion and a
permanent positive charge that improves MS sensitivity and enables quantitation of
metabolites more accurately by both NMR and MS. Such analysis allows direct
comparison of NMR and MS data, which is an important characteristic for biomarker
discovery and biological interpretation in the metabolomics field.
3 Conclusion
Acknowledgments The authors gratefully acknowledge the financial support from the NIH grants
R01GM138465, R01GM131491, P30CA015704, and P30DK035816.
Quantitative NMR Methods in Metabolomics 159
References
Airoldi C, Tripodi F, Guzzi C, Nicastro R, Coccetti P (2015) NMR analysis of budding yeast
metabolomics: a rapid method for sample preparation. Mol Biosyst 11(2):379–383
Akoka S, Barantin L, Trierweiler M (1999) Concentration measurement by proton NMR using the
ERETIC method. Anal Chem 71(13):2554–2557
Albrecht B, Voronina E, Schipke C, Peters O, Parr MK, Díaz-Hernández MD, Schlörer NE (2020)
Pursuing experimental reproducibility: an efficient protocol for the preparation of cerebrospinal
fluid samples for NMR-based metabolomics and analysis of sample degradation. Metabolites
10(6):251
Alum MF, Shaw PA, Sweatman BC, Ubhi BK, Haselden JN, Connor SC (2008) 4,4-Dimethyl-4-
silapentane-1-ammonium trifluoroacetate (DSA), a promising universal internal standard for
NMR-based metabolic profiling studies of biofluids, including blood plasma and serum.
Metabolomics 4:122–127
Anderson JR, Phelan MM, Rubio-Martinez LM, Fitzgerald MM, Jones SW, Clegg PD, Peffers MJ
(2020) Optimization of synovial fluid collection and processing for NMR metabolomics and
LC-MS/MS proteomics. J Proteome Res 19(7):2585–2597
Bala L, Ghoshal UC, Ghoshal U, Tripathi P, Misra A, Nagana Gowda GA, Khetrapal CL (2006)
Malabsorption syndrome with and without small intestinal bacterial overgrowth: a study on
upper-gut aspirate using 1H NMR spectroscopy. Magn Reson Med 56(4):738–744
Barding GA Jr, Salditos R, Larive CK (2012) Quantitative NMR for bioanalysis and metabolomics.
Anal Bioanal Chem 404(4):1165–1179
Barrilero R, Gil M, Amigó N, Dias CB, Wood LG, Garg ML et al (2018) LipSpin: a new
bioinformatics tool for quantitative 1 H NMR lipid profiling. Anal Chem 90:2031–2040
Beckonert O, Keun HC, Ebbels TM, Bundy J, Holmes E, Lindon JC, Nicholson JK (2007)
Metabolic profiling, metabolomic and metabonomic procedures for NMR spectroscopy of
urine, plasma, serum and tissue extracts. Nat Protoc 2(11):2692–2703
Bell JD, Brown JC, Kubal G, Sadler PJ (1988) NMR-invisible lactate in blood plasma. FEBS Lett
235:81–86
Bingol K, Zhang F, Bruschweiler-Li L, Brüschweiler R (2012) Carbon backbone topology of the
metabolome of a cell. J Am Chem Soc 134:9006–9011
Bingol K, Zhang F, Bruschweiler-Li L, Brüschweiler R (2013) Quantitative analysis of metabolic
mixtures by two-dimensional 13C constant-time TOCSY NMR spectroscopy. Anal Chem 85:
6414–6420
Bligh EG, Dyer WJ (1959) A rapid method of total lipid extraction and purification. Can J Biochem
Physiol 37(8):911–917
Bouatra S, Aziat F, Mandal R, Guo AC, Wilson MR, Knox C, Bjorndahl TC et al (2013) The human
urine metabolome. Plos One 8(9):e73076
Chatham JC, Forder JR (1999) Lactic acid and protein interactions: implications for the NMR
visibility of lactate in biological systems. Biochim Biophys Acta 1426(1):177–184
Chen W, Lu S, Wang G, Chen F, Bai C (2017) Staging research of human lung cancer tissues by
high-resolution magic angle spinning proton nuclear magnetic resonance spectroscopy
(HRMAS 1 H NMR) and multivariate data analysis. Asia Pac J Clin Oncol 13(5):e232–e238
Chikayama E, Suto M, Nishihara T, Shinozaki K, Kikuchi J (2008) Systematic NMR analysis of
stable isotope labeled metabolite mixtures in plant and animal systems: coarse grained views of
metabolic pathways. PLoS One 3:e3805
Choi JS, Baek HM, Kim S, Kim MJ, Youk JH, Moon HJ, Kim EK et al (2012) HR-MAS MR
spectroscopy of breast cancer tissue obtained with core needle biopsy: correlation with prog-
nostic factors. PLoS One 7:e51712
Cruz M, Wang M, Frisch-Daiello J, Han X (2016) Improved butanol-methanol (BUME) method by
replacing acetic acid for lipid extraction of biological samples. Lipids 51(7):887–896
160 G. A. Nagana Gowda and D. Raftery
d'Alessandro A, Osterloh JD, Chuwers P, Quinlan PJ, Kelly TJ, Becker CE (1994) Formate in
serum and urine after controlled methanol exposure at the threshold limit value. Environ Health
Perspect 102(2):178–181
Daykin CA, Foxall PJ, Connor SC et al (2002) The comparison of plasma deproteinization methods
for the detection of low-molecular-weight metabolites by (1)H nuclear magnetic resonance
spectroscopy. Anal Biochem 304(2):220–230
DeSilva MA, Shanaiah N, Nagana Gowda GA, Raftery MA, Hainline BE, Raftery D (2009)
Application of 31P NMR spectroscopy and chemical derivatization for metabolite profiling of
lipophilic compounds in human serum. Magn Reson Chem 47Suppl 1:S74–S80
Dinges SS, Vandergrift LA, Wu S, Berker Y, Habbel P, Taupitz M, Wu C-L et al (2019)
Metabolomic prostate cancer fields in HRMAS MRS-profiled histologically benign tissue
vary with cancer status and distance from cancer. NMR Biomed 32(10):e4038
Djukovic D, Raftery D, Nagana Gowda GA (2020) Chapter 16 – Mass spectrometry and NMR
spectroscopy based quantitative metabolomics. In: Issaq HJ, Veenstra TD (eds) Proteomic and
metabolomic approaches to biomarker discovery, 2nd edn. Academic Press, pp 289–311
Edison S, Colonna M, Gouveia GJ, Holderman NR, Judge MT, Shen X, Zhang S (2021) NMR:
unique strengths that enhance modern metabolomics research. Anal Chem 93:478
Emwas AH, Luchinat C, Turano P et al (2015) Standardizing the experimental conditions for using
urine in NMR-based metabolomic studies with a particular focus on diagnostic studies: a
review. Metabolomics 11(4):872–894
Emwas AH, Roy R, McKay RT, Ryan D, Brennan L, Tenori L, Luchinat C, Gao X, Zeri AC,
Nagana Gowda GA, Raftery D, Steinbeck C, Salek RM, Wishart DS (2016) Recommendations
and standardization of biomarker quantification using NMR-based metabolomics with particular
focus on urinary analysis. J Proteome Res 15(2):360–373
Engel KM, Baumann S, Rolle-Kampczyk U, Schiller J, von Bergen M, Grunewald S (2019)
Metabolomic profiling reveals correlations between spermiogram parameters and the
metabolites present in human spermatozoa and seminal plasma. PLoS One 14(2):e0211679
Evans C, Bogan KL, Song P, Burant CF, Kennedy RT, Brenner C (2010) NAD+ metabolite levels
as a function of vitamins and calorie restriction: evidence for different mechanisms of longevity.
BMC Chem Biol 10:2
Fan TW (2012) In: Fan TW, Higashi RM, Lane AN (eds) The handbook of metabolomics, methods
in pharmacology and toxicology, vol 2012. Springer, New York, pp 7–27
Fan TW, Lane AN, Higashi RM, Farag MA, Gao H, Bousamra M, Miller DH (2009) Altered
regulation of metabolic pathways in human lung cancer discerned by (13)C stable isotope-
resolved metabolomics (SIRM). Mol Cancer 8:41
Folch J, Lees M, Stanley G (1957) A simple method for the isolation and purification of total lipides
from animal tissues. J Biol Chem 226:497–509
Gil M, Samino S, Barrilero R, Correig X (2019) Lipid profiling using 1 H NMR spectroscopy.
Methods Mol Biol 2037:35–47
Goldman A, Khiste S, Freinkman E, Dhawan A, Majumder B, Mondal J, Pinkerton AB, Eton E,
Medhi R, Chandrasekar V, Rahman MM, Ichimura T, Gopinath KS, Majumder P, Kohandel M,
Sengupta S (2019) Targeting tumor phenotypic plasticity and metabolic remodeling in adaptive
crossdrug tolerance. Sci Signal 12(595):eaas8779. https://doi.org/10.1126/scisignal.aas8779
Goldoni L, Beringhelli T, Rocchia W, Realini N, Piomelli D (2016) A simple and accurate protocol
for absolute polar metabolite quantification in cell cultures using quantitative nuclear magnetic
resonance. Anal Biochem 501:26–34
Hernandez ME, Lopez AC, Calatayud AG et al (2001) Vesical uric acid lithiasis in a child with
renal hypouricemia. An Esp Pediatr 55(3):273–276
Holzgrabe U (2010) Quantitative NMR spectroscopy in pharmaceutical applications. Prog Nucl
Magn Reson Spectrosc 57(2):229–240
Hoult DI (2000) The principle of reciprocity in signal strength calculations – a mathematical guide.
Concepts Magn Reson 12:173–187
Quantitative NMR Methods in Metabolomics 161
Hoult DI, Richards RE (1976) The signal-to-noise ratio of the nuclear magnetic resonance experi-
ment. J Magn Reson 24:71–85
Ivanisevic J, Zhu ZJ, Plate L, Tautenhahn R, Chen S, O'Brien PJ, Johnson CH, Marletta MA, Patti
GJ, Siuzdak G (2013) Toward 'omic scale metabolite profiling: a dual separation-mass spec-
trometry approach for coverage of lipid and central carbon metabolism. Anal Chem 85(14):
6876–6884
Jeyarajah EJ, Cromwell WC, Otvos JD (2006) Lipoprotein particle analysis by nuclear magnetic
resonance spectroscopy. Clin Lab Med 26:847–870
Jiménez B, Holmes E, Heude C, Tolson RF, Harvey N, Lodge SL, Chetwynd AJ et al (2018)
Quantitative lipoprotein subclass and low molecular weight metabolite analysis in human serum
and plasma by 1H NMR spectroscopy in a multilaboratory trial. Anal Chem 90(20):
11962–11971
Johnson CS (1999) Diffusion ordered nuclear magnetic resonance spectroscopy: principles and
applications. Prog Nucl Magn Reson Spectrosc 34:203–256
Johnson CH, Ivanisevic J, Siuzdak G (2016) Metabolomics: beyond biomarkers and towards
mechanisms. Nat Rev Mol Cell Biol 17:451–459
Kaess B, Fischer M, Baessler A, Stark K, Huber F, Kremer W, Kalbitzer HR et al (2008) The
lipoprotein subfraction profile: heritability and identification of quantitative trait loci. J Lipid
Res 49:715–723
Kalfe A, Telfah A, Lambert J, Hergenröder R (2015) Looking into living cell systems: planar
waveguide microfluidic NMR detector for in vitro metabolomics of tumor spheroids. Anal
Chem 87(14):7402–7410
Kapur BM, Vandenbroucke AC, Adamchik Y, Lehotay DC, Carlen PL (2007) Formic acid, a novel
metabolite of chronic ethanol abuse, causes neurotoxicity, which is prevented by folic acid.
Alcohol Clin Exp Res 31(12):2114–2120
Kodama M, Oshikawa K, Shimizu H, Yoshioka S, Takahashi M, Izumi Y, Bamba T, Tateishi C,
Tomonaga T, Matsumoto M, Nakayama KI (2020) A shift in glutamine nitrogen metabolism
contributes to the malignant progression of cancer. Nat Commun 11:1320
Kriat M, Confort-Gouny S, Vion-Dury J, Sciaky M, Viout P, Cozzone PJ (1992) Quantitation of
metabolites in human blood serum by proton magnetic resonance spectroscopy. A comparative
study of the use of formate and TSP as concentration standards. NMR Biomed 5(4):179–184
Kubáň P, Boček P (2013) Direct analysis of formate in human plasma, serum and whole blood by
in-line coupling of microdialysis to capillary electrophoresis for rapid diagnosis of methanol
poisoning. Anal Chim Acta 768:82–89
Lane AN, Fan TW, Bousamra M II, Higashi RM, Yan J, Milleret DM (2011) Stable isotope-
resolved metabolomics (SIRM) in cancer research with clinical application to NonSmall cell
lung cancer OMICS. J Integr Biol 15:173–182
Lane AN, Tan J, Wang Y et al (2017) Probing the metabolic phenotype of breast cancer cells by
multiple tracer stable isotope resolved metabolomics. Metab Eng 43(Pt B):125–136
Lin P, Lane AN, Fan TW (2019) Stable isotope-resolved metabolomics by NMR. Methods Mol
Biol 2037:151–168
Locasale JW, Grassian AR, Melman T, Lyssiotis CA, Mattaini KR, Bass AJ, Heffron G et al (2011)
Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis. Nat
Genet 43:869–874
Löfgren L, Ståhlman M, Forsberg GB, Saarinen S, Nilsson R, Hansson GI (2012) The BUME
method: a novel automated chloroform-free 96-well total lipid extraction method for blood
plasma. J Lipid Res 53:1690–1700
Lohavanichbutr P, Zhang Y, Wang P, Gu H, Nagana Gowda GA, Djukovic D, Buas MF, Raftery D,
Chen C (2018) Salivary metabolite profiling distinguishes patients with oral cavity squamous
cell carcinoma from normal controls. PLoS One 13(9):e0204249
Lucas LH, Larive CK, Wilkinson PS, Huhn S (2005) Progress toward automated metabolic
profiling of human serum: comparison of CPMG and gradient-filtered NMR analytical methods.
J Pharm Biomed Anal 39(1–2):156–163
162 G. A. Nagana Gowda and D. Raftery
Lussu M, Camboni T, Piras C, Serra C, Del Carratore F, Griffin J, Atzori L, Manzin A (2017) 1H
NMR spectroscopy-based metabolomics analysis for the diagnosis of symptomatic E. coli-
associated urinary tract infection (UTI). BMC Microbiol 17(1):201
Mallol R, Rodriguez MA, Brezmes J, Masana L, Correig X (2013) Human serum/plasma lipopro-
tein analysis by NMR: application to the study of diabetic dyslipidemia. Prog Nucl Magn Reson
Spectrosc 70:1–24
Mallol R, Amigó N, Rodríguez MA, Heras M, Vinaixa M, Plana N, Rock E et al (2015) Liposcale: a
novel advanced lipoprotein test based on 2D diffusion-ordered 1H NMR spectroscopy. J Lipid
Res 56(3):737–746
Maniara G, Rajamoorthi K, Rajan S, Stockton GW (1998) Method performance and validation for
quantitative analysis by 1H and 31P NMR spectroscopy. Applications to analytical standards and
agricultural chemicals. Anal Chem 70(23):4921–4928
Maniscalco M, Cutignano A, Paris D, Melck DJ, Molino A, Fuschillo S, Motta A (2020)
Metabolomics of exhaled breath condensate by nuclear magnetic resonance spectroscopy and
mass spectrometry: a methodological approach. Curr Med Chem 27(14):2381–2399
Mo H, Raftery D (2008) Solvent signal as an NMR concentration reference. Anal Chem 80(24):
9835–9839
Nagana Gowda GA (2011) NMR spectroscopy for discovery and quantitation of biomarkers of
disease in human bile. Bioanalysis 3(16):1877–1890
Nagana Gowda GA (2018) Profiling redox and energy coenzymes in whole blood, tissue and cells
using NMR spectroscopy. Metabolites 8(2):32
Nagana Gowda GA, Raftery D (2014a) Advances in NMR based metabolomics. In: Simó C,
Cifuentes A, García-Cañas V (eds) Fundamentals of advanced Omics technologies: from
genes to metabolites, comprehensive analytical chemistry, vol 63. Elsevier, New York, pp
187–211
Nagana Gowda GA, Raftery D (2014b) Quantitating metabolites in protein precipitated serum using
NMR spectroscopy. Anal Chem 86(11):5433–5440
Nagana Gowda GA, Raftery D (2015) Can NMR solve some significant challenges in
metabolomics? J Magn Reson 260:144–160
Nagana Gowda GA, Raftery D (2017a) Recent advances in NMR-based metabolomics. Anal Chem
89(1):490–510
Nagana Gowda GA, Raftery D (2017b) Whole blood metabolomics by 1H NMR spectroscopy
provides a new opportunity to evaluate coenzymes and antioxidants. Anal Chem 89(8):
4620–4627
Nagana Gowda GA, Raftery D (eds) (2019) NMR based metabolomics: methods and protocols,
Methods in molecular biology, vol 2037. Humana Press/Springer Science, New York
Nagana Gowda GA, Gowda YN, Raftery D (2015) Expanding the limits of human blood metabolite
quantitation using NMR spectroscopy. Anal Chem 87(1):706–715
Nagana Gowda GA, Abell L, Lee CF, Tian R, Raftery D (2016) Simultaneous analysis of major
coenzymes of cellular redox reactions and energy using ex vivo (1)H NMR spectroscopy. Anal
Chem 88(9):4817–4824
Nagana Gowda GA, Djukovic D, Bettcher LF, Gu H, Raftery D (2018) NMR-guided mass
spectrometry for absolute quantitation of human blood metabolites. Anal Chem 90(3):
2001–2009
Nagana Gowda GA, Abell L, Tian R (2019) Extending the scope of 1H NMR spectroscopy for the
analysis of cellular coenzyme a and acetyl coenzyme A. Anal Chem 91(3):2464–2471
Nagana Gowda GA, Hong NN, Raftery D (2021) Evaluation of Fumaric acid and maleic acid as
internal standards for NMR analysis of protein precipitated plasma, serum, and whole blood.
Anal Chem 93(6):3233–3240
Nguyen TTM, An YJ, Cha JW, Ko YJ, Lee H, Chung CH, Jeon SM et al (2020) Real-time
in-organism NMR metabolomics reveals different roles of AMP-activated protein kinase cata-
lytic subunits. Anal Chem 92(11):7382–7387
Quantitative NMR Methods in Metabolomics 163
Nicholson JK, Gartland KP (1989) 1H NMR studies on protein binding of histidine, tyrosine and
phenylalanine in blood plasma. NMR Biomed 2(2):77–82
Nicholson JK, Foxall PJD, Spraul M, Farrant RD, Lindon JC (1995) 750 MHz 1H and 1H-13C
NMR spectroscopy of human blood plasma. Anal Chem 67:793–811
Orczyk-Pawilowicz M, Jawien E, Deja S, Hirnle L, Zabek A, Mlynarz P (2016) Metabolomics of
human amniotic fluid and maternal plasma during normal pregnancy. PLoS One 11(4):
e0152740
Paul A, Kumar S, Raj A, Sonkar AA, Jain S, Singhai A, Roy R (2018) Alteration in lipid
composition differentiates breast cancer tissues: a 1 H HRMAS NMR metabolomic study.
Metabolomics 14(9):119
Pauli GF, Gödecke T, Jaki BU, Lankin DC (2012) Quantitative 1H NMR. Development and
potential of an analytical method: an update. J Nat Prod 75(4):834–851
Psychogios N, Hau DD, Peng J, Guo AC, Mandal R, Bouatra S, Sinelnikov I et al (2011) The
human serum metabolome. PLoS One 6(2):e16957
Raftery D (ed) (2014) Mass spectrometry in metabolomics: methods and protocols, Methods in
molecular biology, vol 1198. Humana Press/Springer Science, New York
Rundlöf T, Mathiasson M, Bekiroglu S, Hakkarainen B, Bowden T, Arvidsson T (2010) Survey and
qualification of internal standards for quantification by 1H NMR spectroscopy. J Pharm Biomed
Anal 52(5):645–651
Rylander R, Remer T, Berkemeyer S, Vormann J (2006) Acid–base status affects renal magnesium
losses in healthy, elderly persons. J Nutr 136(9):2374–2377
Salem AA, Mossa HA (2012) Method validation and determinations of levofloxacin, metronidazole
and sulfamethoxazole in an aqueous pharmaceutical, urine and blood plasma samples using
quantitative nuclear magnetic resonance spectrometry. Talanta 88:104–114
Shanaiah N, Desilva MA, Nagana Gowda GA et al (2007) Class selection of amino acid metabolites
in body fluids using chemical derivatization and their enhanced 13C NMR. Proc Natl Acad Sci
U S A 104(28):11540–11544
Simón-Manso Y, Lowenthal MS, Kilpatrick LE, Sampson ML, Telu KH, Rudnick PA, Mallard WG
et al (2013) Metabolite profiling of a NIST standard reference material for human plasma (SRM
1950): GC-MS, LC-MS, NMR, and clinical laboratory analyses, libraries, and web-based
resources. Anal Chem 85(24):11725–11731
Sitter B, Bathen TF, Singstad TE, Fjøsne HE, Lundgren S, Halgunset J, Gribbestad IS (2010)
Quantification of metabolites in breast cancer patients with different clinical prognosis using HR
MAS MR spectroscopy. NMR Biomed 23(4):424–431
Soininen P, Kangas AJ, Wurtz P, Tukiainen T, Tynkkynen T, Laatikainen R, Jarvelin MR et al
(2009) High-throughput serum NMR metabonomics for cost-effective holistic studies on
systemic metabolism. Analyst 134:1781–1785
Soininen P, Kangas AJ, Würtz P, Suna T, Ala-Korpela M (2015) Quantitative serum nuclear
magnetic resonance metabolomics in cardiovascular epidemiology and genetics. Circ
Cardiovasc Genet 8(1):192–206
Takis PG, Schäfer H, Spraul M, Luchinat C (2017) Deconvoluting interrelationships between
concentrations and chemical shifts in urine provides a powerful analysis tool. Nat Commun
8(1):1662
Tayyari F, Nagana Gowda GA, Gu H, Raftery D (2013) 15N-cholamine – a smart isotope tag for
combining NMR- and MS-based metabolite profiling. Anal Chem 85(18):8715–8721
Tilgner M, Vater TS, Habbel P, Cheng LL (2019) High-resolution magic angle spinning (HRMAS)
NMR methods in metabolomics. Methods Mol Biol 2037:49–67
Tiziani S, Emwas AH, Lodi A, Ludwig C, Bunce CM, Viant MR, Güntheret UL (2008) Optimized
metabolite extraction from blood serum for 1H nuclear magnetic resonance spectroscopy. Anal
Biochem 377(1):16–23
Trammell SA, Brennera C (2013) Targeted, LCMS-based metabolomics for quantitative measure-
ment of NAD(+) metabolites. Comput Struct Biotechnol J 4:e201301012
164 G. A. Nagana Gowda and D. Raftery
Contents
1 Introduction: A Brief History of Isotopic Labelling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166
2 Time- and Isotope-Resolved Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168
2.1 Stable Isotope-Resolved Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168
2.2 Time Resolved Isotope Labelling Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169
2.3 Isotope Tracing at the Crossroad of Metabolic Pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172
2.4 The Application of Ultra-High Resolution Mass Spectrometry Allows the Tracing
of Different Isotopic Species . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174
3 Applications and Future Perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 176
3.1 Applications of pSIRM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 176
3.2 Perspective Towards Single Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 178
Abstract
# The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 165
V. Ghini et al. (eds.), Metabolomics and Its Impact on Health and Diseases,
Handbook of Experimental Pharmacology 277, https://doi.org/10.1007/164_2022_621
166 M. Forbes et al.
The most important metabolic pathways are encompassed in the central carbon
metabolism (CCM). According to the Kyoto Encyclopedia of Genes and
Genomes (KEGG), the central carbon metabolism “is the most basic aspect of
life”. It includes all metabolites and enzymatic reactions within: glycolysis and
gluconeogenesis, pentose phosphate pathway (PPP), tricarboxylic acid (TCA)
cycle, oxidative phosphorylation (OXPHOS), amino acids and nucleotide meta-
bolic pathways. Some molecules are at the crossroad of metabolic pathways,
interconnecting diverse metabolic and therefore functional outcomes. Labelling
these nodal metabolites and analysing their isotopic composition allows the
precise determination of the metabolic flow within the biochemical networks
that they are in.
Application of stable isotope labelled substrates allows the measurement of
metabolic flux through a biochemical pathway. The rapid turnover of metabolites
in pathways requires pulse-feeding cells with a labelled substrate. This method
allows for the determination of different cell states. For example, the action of a
drug from immediate impact until the compensatory response of the metabolic
system (cell, organs, organisms). Pulsed labelling is an elegant way to analyse the
action of small molecules and drugs and enables the analysis of regulatory
metabolic processes in short time scales.
Keywords
The use of isotope labelled molecules in metabolic research began within the first
decades of the last century. In the early days of metabolism research, radioactive
isotopes were used to investigate the metabolic flow in bacteria, plants and animals.
In order to investigate the structure of biochemical pathways and specifically
metabolic cycles, e.g. tricarboxylic acid cycle, reverse tricarboxylic acid cycle or
Calvin-Benson cycle, isotopes were applied in a time resolved manner. Time
resolution allowed for the elucidation of the consecutive order of chemical reactions
within the investigated pathways. The use of radioactive isotopes was of paramount
importance to decipher the flow of carbon within cells and organisms.
In 1910, English chemist Frederick Soddy observed that “elements of different
atomic weights may possess identical (chemical) properties” and therefore belong to
the same position in the periodic table. This included not only radioactive elements
but also stable isotopes of atoms, i.e. atoms with the same number of protons but
different numbers of neutrons in their nucleus. We can refer to these elements as
“hot” or “cold” isotopes of an atom in dependency of their radio- or
non-radioactivity. The presence of radioactive isotopes was discovered by black
spots occurring on photosensitive emulsions, as the decaying radioactive element
Advancements in Pulsed Stable Isotope-Resolved Metabolomics 167
produced traceable radiation. This phenomenon gave rise to years of research into
the isotopes of the periodic table and their many uses in a broad range of analytical
fields, including: chemistry, geology, biology, physics and medicine (Wilikinson
2018).
Georg Charles de Hevesy was a Hungarian radio-chemist and Nobel Prize in
Chemistry laureate, recognised in 1943 for his achievements in the development of
radioactive tracers in the study of metabolism in animals. Hevesy is considered the
first to use radioisotopes to measure metabolic flux in biological systems. One such
(sadly unpublished) experiment of Hevesy and Rutherford describes the practices of
a shrewd landlady in 1911, Manchester. Hevesy was convinced that his landlady was
recycling food, much to the denial of such practice. In order to thwart the thrifty
landlady, Hevesy “spiked” a portion of leftover meat with a tiny amount of a
radioactive material. A few days later he brought an electroscope to the table to
demonstrate to the indignant landlady that the food served that day was radioactive.
Hevesy is thought to have enjoyed fresh hot meals as a result of his radioactive
tracing experiment (Myers 1996). Hevesy continued his endeavours into the usage of
tracers in biological systems. In 1934, Hevesy and Hofer first used tracers in
medicine by using an enriched stable isotope to determine the rate of elimination
of water from the human body. Hevesy and Hofer drank dilute deuterated water and
assayed the isotopic dilution of the deuterium in their urine. From their results they
concluded the average time a water molecule spent in their bodies was 13 ± 1.5 days
(Hevesy and Hofer 1934).
The accounts of Hevesy are far more than comical anecdotes. They provide an
understandable example for the use of radioisotopes in analytical chemistry.
Through the development of mass spectrometry, stable isotopes replaced
radioisotopes in tracing experiments. In mass spectrometry, a metabolite can be
identified by the mass spectrum of its fragments, i.e. the intensity of the fragment’s
peaks at a defined mass-to-charge (m/z) ratio. The additional neutron in the atomic
nucleus of isotopes makes the atom 1 Da heavier and increases the m/z ratio. The
incorporation of isotopes into a metabolite changes the atomic composition of this
intermediate and induces a shift in the respective fragment’s mass spectrum. Specifi-
cally, stable isotopes of carbon (13C), nitrogen (15N), oxygen (18O) and hydrogen
(2H) can be introduced into organic compounds. By tracing the incorporation of the
isotope, the metabolic fate of these compounds within biological systems can be
characterised (Wilikinson 2018). The incorporation of these isotopes can be resolved
in molecules such as sugars, amino acids or nucleotides. This method of isotope
labelling was used to describe the effect of oncogenes on cancer cell metabolism. For
example, Le et al. applied 13C-Glucose and 13C,15N-Glutamine to trace glucose and
glutamine metabolism when the MYC oncogene was induced in P493 cancer cells.
The authors showed that glutamine plays an essential role in the cells’ proliferation
and survival, highlighting targets of glutamine metabolism for cancer therapy
(Le et al. 2012). The usage of isotope labelling provides insights into the dynamics
and kinetics of metabolism, as a function of time and cell state.
168 M. Forbes et al.
In order to measure the metabolite flow, or more precisely the metabolic flux through
a biological system, substrates of the system are labelled with stable isotopes. These
substrates may be, glucose or glutamine which provide carbon and nitrogen to the
central metabolic pathways (Fig. 1). The usage of these substrates by the cell leads to
the incorporation of the isotopes into downstream intermediates. After isotopically
labelling a biological system and extracting the cellular metabolites, mass spectrom-
etry is then employed to analyse the number of heavy atoms incorporated
(isotopologues) and their positions (isotopomers) in detected metabolites (Bruntz
et al. 2017). Isotopologues can be identified by increasing mass shifts, as every
heavy atom incorporated rises the m/z ratio by one. Isotopomers, on the other hand,
can be distinguished by the heavy atom incorporation visible in different fragments
Fig. 1 Scheme of central metabolic pathways indicating carbon and nitrogen flow. The scheme
displays main central metabolic pathways: glycolysis, pentose phosphate pathway, nucleotide
metabolism, tricarboxylic acid cycle and glutaminolysis. The different colours depict the contribu-
tion of the distinct carbon and nitrogen atoms stemming from glucose or glutamine, respectively
Advancements in Pulsed Stable Isotope-Resolved Metabolomics 169
of the same metabolite. In summary, the usage of isotope labelled substrates allows
for the follow-up of interconnected anabolic and catabolic pathways. The tracing of
isotope labels in metabolites, and the atomic position in which the label resides, is
referred to as stable isotope-resolved metabolomics (SIRM).
Cellular metabolism is a rapid, dynamic process, in which both anabolism and
catabolism of metabolites produce energy, build macromolecules for biomass and
generate intermediates involved in intra- and inter-cellular signalling. The under-
standing of biochemical processes of metabolism is inferred through the concentra-
tion of intermediates and the rate of metabolite conversion. However, the
measurement of metabolite concentrations does not give a full representation of
this dynamic system. In order to understand the kinetics of metabolism, it must be
described and quantified in terms of metabolite flow as a function of time (Sauer
2006; Pietzke et al. 2014; Buescher et al. 2015; Jang et al. 2018).
Fig. 2 Modelling the kinetics of metabolite labelling. As cells are incubated with a labelled
substrate for a period of time, the quantity of label incorporated into metabolites increases. The
rate of reaction, or the percentage of label in the metabolites relative to the unlabelled fraction, is
dependent on the position of the metabolite in the pathway and the efficiency of associated
enzymes. In order to understand the rate and kinetics of metabolic processes, we aim to observe
labelled metabolites in the instationary phase (Fig. 2, left (I)). On the contrary, if all metabolites
are saturated with label after a given period of time (Fig. 2, left (II)), then no kinetic information
can be derived
Fig. 3 pSIRM data in 2D. Each point represents one single metabolite of the CCM. On the x-axis
fold-changes of metabolite concentrations and on the y-axis changes of isotope incorporation are
shown. For this experiment cancer cells were incubated for a total time of 15 min with BrPyr and
metabolites were measured using the pSIRM technology (see Pietzke et al. (2014)). The graph
shows that in some metabolites a quantitative change in metabolite concentration can be observed.
However, the effect on glycolytic inhibition can only be observed in the label incorporation of
metabolites downstream of the GAPDH reaction
We have described the use of isotope labelled substrates for investigating the rate
of reactions in metabolic pathways. One may also investigate the direction of
metabolic pathways or deduce the relative dominance of a pathway by using
substrates labelled at specific carbon positions. For example, the pentose phosphate
pathway (PPP) – which links glycolysis to nucleotide synthesis – generates ribose-5-
phosphate (R5P) from two reactions. The oxidative PPP utilises glucose-6-phos-
phate to produce 6-phosphogluconate which is de-carboxylated to ribulose-6-phos-
phate (Ru5P). In turn Ru5P is isomerised to R5P. The non-oxidative PPP cycles
carbons from fructose-6-phosphate, glyceraldehyde-3-phosphate and erythrose-4-
phosphate to produce xyulose-5-phosphate and R5P. This branch does not contain a
de-carboxylation event and carbon number is maintained in the process. Therefore,
we may employ a labelling strategy to decipher the ratio of the oxidative and
non-oxidative branches by assessing the state of the de-carboxlyation event in the
oxidative branch. By using 1,2-[13C]-Glc as the labelling substrate we can measure
the ratio of R5P present in the sample which contains a single (m/z + 1) or a double
(m/z + 2) label. R5P which is derived from the oxidative PPP will only contain a
single label as the labelled carbon is lost during the de-carboxylation event (Fig. 4).
Oxythiamine is an inhibitor of the non-oxidative PPP enzyme, transketolase
(TKT). Boros et al. (1997) showed that oxythiamine inhibits Mia pancreatic adeno-
carcinoma cell growth by 39%. By using 1,2-[13C]-Glc labelling the authors
172 M. Forbes et al.
Fig. 4 Utilising 1,2-[13C]-Glc in the analysis of the pentose phosphate pathway. The pentose
phosphate pathway is comprised of the oxidative and non-oxidative branches. The oxidative branch
observes a de-carboxylation event which removes a labelled carbon from glucose-6-phosphate. In
the non-oxidative branch, labelled carbons are maintained as de-carboxylation does not occur. The
ratio of single to double labelled R5P shows the relative dominance of the branches. One may
observe the single or double labelled R5P in a mass spectrum generated by MS, by the relative
abundance of R5P (m/z + 1) and R5P (m/z + 2). This is depicted in the simulated MS spectrum on
the right
discovered that this tumour cell line generated R5P predominantly through
transketolase and the non-oxidative PPP (85%). It was deduced that oxythiamine
was an effective anti-tumour inhibitor due to the dependency of R5P synthesis, and
downstream nucleotide synthesis, on the non-oxidative PPP (Boros et al. 1997). In
summary, differential labelling of substrates permits not only the analysis of the
kinetics of metabolism but also gives information on the relative dependencies of
parallel reactions in different cell lines for targeted therapy development.
The most important metabolic pathways are encompassed in the central carbon
metabolism (CCM). According to the Kyoto Encyclopedia of Genes and Genomes
(KEGG), the central carbon metabolism “is the most basic aspect of life”. It includes
all enzymatic reactions within: glycolysis and gluconeogenesis, pentose phosphate
pathway (PPP), tricarboxylic acid (TCA) cycle and oxidative phosphorylation
(OXPHOS), amino acids and nucleotide metabolism pathways. Additionally, the
CCM includes six known carbon fixation pathways (reductive pentose phosphate
cycle (Calvin cycle), reductive citrate cycle, 3-hydroxypropionate bi-cycle, two
variants of 4-hydroxybutyrate pathway and reductive acetyl-coenzyme A (CoA)
pathway) as well as some pathways of methane metabolism, all not relevant in
animal cells (Qiu 2013). Some molecules are at the crossroad of metabolic pathways,
Advancements in Pulsed Stable Isotope-Resolved Metabolomics 173
Fig. 5 13C and 15N label integration into pathways of the central carbon metabolism. Pictogram of
glycolysis, TCA cycle, pentose phosphate pathway, as well as de novo purine and pyrimidine
biosynthesis. Red circles represent glucose-derived 13C atoms. Blue represent the 13C (dark blue) or
15
N (light blue) atoms derived from glutamine. Unlabelled C and N atoms are displayed in white
m/z + 5 in citrate are the result of anaplerotic fuelling from glutamine into the TCA
cycle and oxidative or reductive cycling, respectively.
Other examples of pathway interconnection are pyruvate and glutamine, as both
can be metabolised into very different intermediates and play a role in distinct
cellular functions. Pyruvate-derived 13C-atoms can integrate, as described above,
into citrate or oxaloacetate, but also into alanine or lactate. Thereby, metabolic flow
through pyruvate merges amino acid metabolism, anaerobic glycolysis and TCA
cycle. Glutamine, on the other hand, is an important nitrogen donor and is involved
in amino acids metabolism and de novo biosynthesis of purine and pyrimidine
nucleotides (Bayram et al. 2020). Additionally, via glutamic acid, the carbon
backbone of glutamine feeds into TCA cycle, as well as polyamine and glutathione
synthesis. The example of glutamine highlights the necessity of dual carbon and
nitrogen labelling in order to greatly increase our understanding of pathway connec-
tivity and metabolic fate. Combining 13C- and 15N-labelling allows the follow-up of
glutamine usage, while distinguishing between the amino group, amido group and
carbon backbone utilisation.
Using stable isotopes of different elements allows to determine the utilisation of two
metabolic precursors (e.g. glucose and glutamine) in a single experiment simulta-
neously, or to follow the fate of different atoms from the same precursors (e.g. C and
N from glutamine) into the downstream metabolic network.
For a long time such experiments have not been possible, since mass analysers
with nominal mass resolution are not able to distinguish the mass increase due to the
presence of different elemental isotopes. For example, the difference in the mass
increase given by one 13C or one 15N is only 0.00632 Da (Fig. 6). According to the
IUPAC definition (McNaught and Wilkinson 2008), the resolution needed to sepa-
rate such mass difference for two molecules with nominal mass of 400 Da is around
60,000, far beyond the resolution of conventional quadrupole or ion trap analysers
and barely reached by Time-of-Flight instruments. Only with the advent of (ultra)
high resolution Fourier transform mass spectrometry mass analysers it became
possible to analyse the isotopic fine structure of labelled compounds (Werner et al.
2008; Marshall and Hendrickson 2008).
One implementation of (ultra) high resolution MS for isotope tracing is the direct
infusion of the metabolites in an Fourier-transform ion cyclotron resonance mass
spectrometer (Le et al. 2012; Yang et al. 2017). This approach offers excellent results
in terms of achievable resolution and accuracy in the determination of the ratios
between different isotopes, because the ion signal in the ion cyclotron resonance cell
can be averaged for long times. However, in direct infusion highly abundant analytes
can suppress the ionisation of low abundant ones (Han et al. 2008). In addition, in
absence of another mean of separation, isomers cannot be distinguished because they
have the same accurate m/z ratio, like G6P and F6P. MS/MS experiments can help
Advancements in Pulsed Stable Isotope-Resolved Metabolomics
Fig. 6 MS spectrum of 15N labelled Glutamine. The ion at m/z 271.16635 corresponds to the unlabelled fragment of TBDMS-derivatised glutamine with
molecular formula C12H27ON2Si2. It contains 4 carbon and all nitrogen atoms of glutamine. Insert shows the magnification of the m/z + 1 peaks, revealing the
separation of peaks due to the incorporation of 15N or 13C. The peak of 29Si isotope, present in the derivatisation agent, is also resolved. The masses reported in
the insert are the Δm relative to the monoisotopic peak
175
176 M. Forbes et al.
In line with the general “OMICS” concept, metabolomics aims to measure all
metabolic components of a biological system at once in a quantitative manner.
Unfortunately that’s not possible so far. The chemical space of biomolecules regard-
ing polarity and size as well as their quantities – ranging from a few molecules up to
millimolar concentrations – is hindering its simultaneous detection. One step further
is the measurement of the dynamics of metabolism. The combination of stable
isotope labelling and mass spectrometric detection made a big contribution in this
regard. More and more methods allow the simultaneous measurement of metabolite
concentrations and isotope incorporation (Pietzke et al. 2014). Such integrated
methods are a prerequisite when metabolic dynamics in vivo or in tissues will be
analysed in a clinical context. Often a consecutive sampling and parallel quantifica-
tion in additional samples is not possible. The application of pSIRM together with
the present knowledge of the biochemical network allows to define certain metabolic
nodes that contain superior information of the usage of metabolic pathways,
e.g. citrate. Future method development may focus on such nodes, allowing for
more directed pSIRM applications with tailored isotopically labelled substrates.
In the last years, pSIRM has been applied to successfully analyse metabolic changes
within short and defined windows, e.g. during differentiation of cells. Delp et al.
found that immature and mature neurons rely on different fuels. Precursor cells were
found to be mainly glycolytic and strongly dependent on glutamine. During differ-
entiation, however, they lost their glutamine dependency while gaining flexibility in
energy production (Delp et al. 2018). By analysing the glutamine-derived carbon
utilisation in high salt-treated differentiating macrophages a specific down-
regulation of the succinate to fumarate conversion could be revealed. This observa-
tion led to the identification of sodium as strong regulator of complex II activity
(Geisberger et al. 2021).
pSIRM can also be applied in vivo as well as ex vivo in tissue slices or organs. By
administering 13C6-glucose into the peritoneum of a hepatocellular carcinoma mouse
model, Berndt et al. described enhanced glycolytic rates in tumours compared to
normal liver. These data contributed to the creation of individualised metabolic
profiles of tumours and modelling predictions of the efficacy of drug therapies
Advancements in Pulsed Stable Isotope-Resolved Metabolomics 177
(Berndt et al. 2020). Similarly, application of stable isotopes in situ to cancerous and
non-cancerous lung tissue revealed higher activity of glycolysis and the Krebs cycle
in the tumour tissue (Fan et al. 2009). In brain tissue slices from naked mole rats,
e.g. labelling with 13C6-fructose uncovered the ability of these animals to metabolise
fructose as fuel for neuronal tissues under hypoxic conditions (Park et al. 2017).
Beside the application of stable isotope labelled organic compounds, inorganic
compounds such as CO2 can be used for labelling experiments. For example,
Rohwer et al. used 13CO2 to demonstrate the reductive carboxylation in gastric
cancer cells (Rohwer et al. 2016). These are only few examples of the wide range
of applications pSIRM; a very versatile technique to describe metabolism in function
of quantity and time.
The advent of single cell “OMICS” has allowed for the analysis of the architecture of
heterogenous cell samples at single cell resolution. Currently, single cell proteomics,
genomics and transcriptomics dominate the single cell technology landscape, while
single cell metabolomics is still in its infancy. The discrepancy of single cell
metabolomics is mainly due to the hardware of mass spectrometry being incompati-
ble with single cell resolution. A mammalian cell contains roughly 1 pL of analytical
volume. Due to this extremely small sample volume GC-MS methods often use
around two million cells per analysis. To further add complexity, metabolism is
highly dynamic – temporally and spatially – in tissue. Therefore, acquisition of
single cells and their processing presents many challenges in the context of meta-
bolic profiling.
Firstly, we will address the process of sampling single cells. The sorting of cells
from a sample is essential to single cell analysis. Fluorescence-activated cell sorting
(FACS) employs fluorescent labels to sort cells of specified origin. FACS may be
coupled to mass spectrometry methods to assess the profiles of sorted cells. How-
ever, the FACS process may interfere with metabolic profiles and therefore FACS
coupled with MS is more suited to proteomic analysis (Bandura et al. 2009). To
minimise the sampling time methods such as matrix-assisted laser desorption/
ionisation (MALDI) employ a laser to ablate cells and their metabolites in situ.
Following laser ablation the ionised metabolites are analysed via MS. Advances
towards single cell resolution have been reported. By using a combination of
computational imaging techniques and nuclei staining, the points of laser ablation
during MALDI-MS analysis can be inferred as metabolite acquisition from a single
cell (Rappez et al. 2021). However, this method is not quantitative and is biased to
high abundant metabolites and proteins.
Current technologies are aiming to sample single cells through microfluidic
trapping coupled with acquisition of intracellular metabolites through micro
capillaries. However, efforts to move to a microfluidic environment present their
own complications, mainly due to maintaining homeostatic environment in a
miniaturised platform (Ali et al. 2019).
178 M. Forbes et al.
References
Ali A, Abouleila Y, Shimizu Y, Hiyama E, Emara S, Mashaghi A, Hankemeier T (2019) Single-cell
metabolomics by mass spectrometry: advances, challenges, and future applications. TrAC
Trends Anal Chem 120:115436., ISSN 0165-9936. https://doi.org/10.1016/J.TRAC.2019.
02.033
Bandura DR, Baranov VI, Ornatsky OI, Antonov A, Kinach R, Lou X, Pavlov S, Vorobiev S, Dick
JE, Tanner SD (2009) Mass cytometry: technique for real time single cell multitarget immuno-
assay based on inductively coupled plasma time-of-flight mass spectrometry. Anal Chem
81(16):6813–6822., ISSN 00032700. https://pubs.acs.org/doi/full/10.1021/ac901049w. https://
doi.org/10.1021/AC901049W
Bayram S, Fürst S, Forbes M, Kempa S (2020) Analysing central metabolism in ultra-high
resolution: at the crossroads of carbon and nitrogen. Mol Metab 33:38–47., ISSN 2212-8778.
https://doi.org/10.1016/J.MOLMET.2019.12.002
Berndt N, Egners A, Mastrobuoni G, Vvedenskaya O, Fragoulis A, Dugourd A, Bulik S, Pietzke M,
Bielow C, van Gassel R, Damink SW, Erdem M, Saez-Rodriguez J, Holzhütter HG, Kempa S,
Cramer T (2020. ISSN 15321827. https://www.nature.com/articles/s41416-019-0659-3)
Kinetic modelling of quantitative proteome data predicts metabolic reprogramming of liver
cancer. Br J Cancer 122(2):233–244. https://doi.org/10.1038/s41416-019-0659-3
Boros LG, Puigjaner J, Cascante M, Lee WNP, Brandes JL, Bassilian S, Yusuf FI, Williams RD,
Muscarella P, Melvin WS, Schirmer WJ (1997) Oxythiamine and dehydroepiandrosterone
inhibit the nonoxidative synthesis of ribose and tumor cell proliferation. Cancer Res. ISSN
00085472
Bruntz RC, Lane AN, Higashi RM, Fan TW-M (2017) Exploring cancer metabolism using stable
isotope-resolved metabolomics (SIRM). J Biol Chem 292(28):11601–11609., ISSN 0021-9258.
http://www.jbc.org/article/S0021925820370289/fulltext. http://www.jbc.org/article/S002192
5820370289/abstract. https://doi.org/10.1074/JBC.R117.776054
Buescher JM, Antoniewicz MR, Boros LG, Burgess SC, Brunengraber H, Clish CB, DeBerardinis
RJ, Feron O, Frezza C, Ghesquiere B, Gottlieb E, Hiller K, Jones RG, Kamphorst JJ, Kibbey
RG, Kimmelman AC, Locasale JW, Lunt SY, Maddocks OD, Malloy C, Metallo CM, Meuillet
EJ, Munger J, Nöh K, Rabinowitz JD, Ralser M, Sauer U, Stephanopoulos G, St-Pierre J,
Tennant DA, Wittmann C, Heiden MGV, Vazquez A, Vousden K, Young JD, Zamboni N,
Fendt SM (2015) A roadmap for interpreting 13C metabolite labeling patterns from cells. Curr
Opin Biotechnol 34:189–201. ISSN 0958-1669. https://doi.org/10.1016/J.COPBIO.2015.
02.003
Delp J, Gutbier S, Cerff M, Zasada C, Niedenführ S, Zhao L, Smirnova L, Hartung T,
Borlinghaus H, Schreiber F, Bergemann J, Gätgens J, Beyss M, Azzouzi S, Waldmann T,
Kempa S, Nöh K, Leist M (2018) Stage-specific metabolic features of differentiating neurons:
implications for toxicant sensitivity. Toxicol Appl Pharmacol 354:64–80. ISSN 1096-0333.
https://pubmed.ncbi.nlm.nih.gov/29278688/. https://doi.org/10.1016/J.TAAP.2017.12.013
Fan TW, Lane AN, Higashi RM, Farag MA, Gao H, Bousamra M, Miller DM (2009) Altered
regulation of metabolic pathways in human lung cancer discerned by 13C stable isotope-
resolved metabolomics (SIRM). Mol Cancer 8(1):1–19. ISSN 14764598. https://molecular-
cancer.biomedcentral.com/articles/10.1186/1476-4598-8-41. https://doi.org/10.1186/1476-
4598-8-41/TABLES/6
Geisberger S, Bartolomaeus H, Neubert P, Willebrand R, Zasada C, Bartolomaeus T, McParland V,
Swinnen D, Geuzens A, Maifeld A, Krampert L, Vogl M, Mähler A, Wilck N, Markó L, Tilic E,
Advancements in Pulsed Stable Isotope-Resolved Metabolomics 179
Sauer U (2006) Metabolic networks in motion: 13C-based flux analysis. Mol Syst Biol 2(1):62.,
ISSN 1744-4292. https://onlinelibrary.wiley.com. https://doi.org/10.1038/MSB4100109
Werner E, Heilier J, Ducruix C, Ezan E, Junot C, Tabet J (2008. ISSN 1570-0232. https://pubmed.
ncbi.nlm.nih.gov/18672410/) Mass spectrometry for the identification of the discriminating
signals from metabolomics: current status and future trends. J Chromatogr B Analyt Technol
Biomed Life Sci 871(2):143–163. https://doi.org/10.1016/J.JCHROMB.2008.07.004
Wilikinson D (2018) Historical and contemporary stable isotope tracer approaches to studying
mammalian protein metabolism. Mass Spectrom Rev 37(1):57–80. ISSN 1098-2787. https://
pubmed.ncbi.nlm.nih.gov/27182900/
Yang Y, Fan T, Lane A, Higashi RM (2017) Chloroformate derivatization for tracing the fate of
amino acids in cells and tissues by multiple stable isotope resolved metabolomics (mSIRM).
Anal Chim Acta 976:63–73. ISSN 1873-4324. https://pubmed.ncbi.nlm.nih.gov/28576319/.
https://doi.org/10.1016/J.ACA.2017.04.014
Metabolomics in Cell Biology
Contents
1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182
1.1 Omics Context . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182
1.2 History of Metabolomics Using Cellular Samples . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 183
2 Metabolomics Approaches: Targeted vs. Untargeted . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184
2.1 Sample Types . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186
2.2 Preparation of Cell Extracts for Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186
2.3 Metabolomics Technologies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 188
2.3.1 NMR-Based Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 189
2.3.2 MS-Based Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
3 Case Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193
3.1 Examples of Cancer Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193
3.2 Real-Time Measurements of Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
3.3 Metabolic Studies in Mycobacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 197
3.4 Metabolic Fluxes by NMR and GC-MS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 198
3.5 Examples for Fluxes in Cancer Cells by GC-MS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199
3.6 Fluxes and Metabolic Modelling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 201
3.7 Metabolic Fluxes in Liver Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 201
Z. Eraslan
Department of Dermatology, Weill Cornell Medicine, New York, NY, USA
M. Cascante
Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona,
Spain
Institute of Biomedicine of University of Barcelona (IBUB), University of Barcelona, Barcelona,
Spain
CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII),
Madrid, Spain
U. L. Günther (✉)
Institute of Chemistry and Metabolomics, University of Lübeck, Lübeck, Germany
e-mail: [email protected]
# The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 181
V. Ghini et al. (eds.), Metabolomics and Its Impact on Health and Diseases,
Handbook of Experimental Pharmacology 277, https://doi.org/10.1007/164_2022_619
182 Z. Eraslan et al.
4 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203
Abstract
Metabolomics has long been used in a biomedical context. The most typical
samples are body fluids in which small molecules can be detected and quantified
using technologies such as Nuclear Magnetic Resonance (NMR) and Mass
Spectrometry (MS). Many studies, in particular in the wider field of cancer
research, are based on cellular models. Different cancer cells can have vastly
different ways of regulating metabolism and responses to drug treatments depend
on specific metabolic mechanisms which are often cell type specific. This has led
to a series of publications using metabolomics to study metabolic mechanisms.
Cell-based metabolomics has specific requirements and allows for interesting
approaches where metabolism is followed in real-time. Here applications of
metabolomics in cell biology have been reviewed, providing insight into specific
technologies used and showing exemplary case studies with an emphasis towards
applications which help to understand drug mechanisms.
Keywords
Cell biology · Metabolomics
1 Introduction
Besides the use of metabolomics in a medical context, often with the intent to find
biomarkers, there are an increasing number of applications in biological studies with
the goal to identify specific mechanisms in cells. When looking at such applications
of metabolomics in cellular systems the boundaries between targeted and
non-targeted analysis are often more fluid. As a matter of fact, some of the earliest
applications of what would now be called metabolomics were conducted in cellular
systems. As early as 1978, Ugurbil and Shulman used NMR to study metabolic
turnover in Escherichia coli cells (E. coli), first by 31P-NMR and soon afterwards by
13
C-NMR. The first report used 31P-NMR to measure glycolysis rates in E. coli, and
also looked at the effect of ATPase inhibition (Ugurbil et al. 1978a). Even at a
spectrometer with 360 MHz proton frequency, a 2 min time resolution was achieved.
In a second publication, E. coli cells were incubated with [1-13C]-labelled glucose in
an NMR tube and followed the turnover of glucose via fructose bisphosphate into
amino acids (alanine and valine) along with lactate, succinate and acetate using
13
C-NMR spectra (Ugurbil et al. 1978b). Upon oxygenation, the authors also
observed glutamate, labelled at the C-4, C-3 and C-2 positions. Time-courses were
reported over up to 3 h with a time resolution of 1 min, which is remarkable
considering that the NMR instrumentation in 1978 was at 90 MHz proton frequency.
This early work demonstrates the advantage of NMR to identify site-specific label
incorporation. In a subsequent publication from Shulman’s group, rat hepatocytes
were used to quantitatively assess the metabolic flux of glycerol in gluconeogenesis,
showing label incorporation in several glycolytic intermediates and products (Cohen
et al. 1979) and to understand whether or to what extent hyperthyroid cells (after
pre-treatment of rats with triiodothyronine hormone (T3)) increases the rate of
glycerol consumption and glucose formation. In this work not only signal intensities
were used but also scalar couplings between 13C nuclei to quantitatively assess label
incorporation. In hyperthyroid cells labelling was also reported in lactate, alanine,
aspartate, glutamate and ketone bodies. By analysing time-courses of label
incorporation in different positions of glucose from differently labelled glycerol
precursors pentose phosphate pathway activity could be determined, along with
activities of transaldolase and transketolase. These early findings which provide a
historical background for cell-based metabolic studies are confirmed by more recent
work taking similar approaches to measure fluxes by NMR (Jin et al. 2013).
Soon afterwards, Jardetzky and co-workers described a system for continuous-
flow monitoring of metabolism in mammalian cells (Chinese hamster ovarian cells)
184 Z. Eraslan et al.
(Gonzalez-Mendez et al. 1982). These seminal experiments laid the ground for using
NMR to look at metabolism at a time when nobody thought about metabolomics.
They exemplify the virtues of NMR for examining metabolism in cellular systems,
which include real-time analysis of intact cells, using isotopic labels as tracers and
the ability to observe site-specific label incorporation in molecules. Although any
analysis that uses labelled precursors is by definition not untargeted, NMR was
always used to simultaneously detect many metabolites in one NMR spectrum.
Another milestone in this development has come from Szyperski et al. (Szyperski
1995; Szyperski et al. 1996) in 1995, using 1H-13C_HSQC spectra to analyse cellular
label incorporation. Today HSQC spectra are often preferred as they offer higher
sensitivity than13C-observed spectra. Szyperski was able to make sense of highly
complex JCC couplings patterns which arise from mixtures of isotopomers. Today’s
most advanced NMR systems work at 1.2 GHz, commonly used metabolomics
systems work at 600 MHz. Cryoprobes and other improvements of NMR technology
have boosted the sensitivity of NMR experiments by at least an order of magnitude.
There are now cryoprobes available that have been optimised for mass limited
samples which provide immense sensitivity advantages for cell-based studies
(Saborano et al. 2019). Microprobes hold great promise to work with very few
cells (Finch et al. 2016).
Here we review approaches of metabolomics in cells and present the methods
used, with a specific focus on advanced NMR methods. Exemplary case studies are
presented which demonstrate how metabolomics can be used in cell-based
investigations in the wider context of drug discovery.
Metabolomics can be carried out at different levels and using very different
approaches (Fig. 1). Methods are typically classified as targeted or non-targeted. In
early metabolomics non-targeted fingerprinting was commonly used where
“features” or raw spectra were employed without specific assignments, often using
multivariate statistical approaches to identify the most relevant features for given
classes (Nicholson et al. 1999). The general advantage of fingerprinting is that data
can be left unassigned. For NMR most initial work has used raw spectra for
subsequent statistical, often multivariate analyses (Nicholson et al. 1999), although
this approach is becoming increasingly uncommon as metabolites are now easily
assigned in many types of samples, including cell extracts where it is extremely well-
known which signals are unique identifiers for individual metabolites. In the case of
GC-MS, the assignments of the signals are usually also well-known thus favouring
targeted approaches. This has included tracer-based approaches for which GC-MS is
equally well suited (Bruntz et al. 2017). Employing typical LC-MS methods,
thousands of features can be detected which can either be used in univariate or
multivariate statistical analyses in an untargeted manner. Alternatively in targeted
approaches features are assigned and only fully assigned signals are used for further
Metabolomics in Cell Biology 185
When working with cellular systems or organelles, there is a wide array of possible
samples. Primary cultures are directly taken from organisms and can be subject to
investigation for at least a short time. Cell lines arise from primary cells which have
been immortalized. Cell cultures are available from public resources, such as ATCC
(http://www.atcc.org), Coriell (http://ccr.coriell.org), DSMZ (http://www.dsmz.de),
ECACC (https://www.culturecollections.org.uk), JCRB (https://cellbank.nibiohn.
go.jp) or RIKEN Bioresource centre (https://web.brc.riken.jp/en), for which robust
quality control protocols have been established (Yu et al. 2015). Most studies use
immortalised cell lines which can be readily grown to obtain sufficient amounts of
metabolites. Cell extracts represent the most common type of sample, but it is also
possible and often reasonable to measure metabolite concentrations in the medium in
which cells were grown, which can be used to calculate turnover for individual
nutrients. Media-sampling can also be carried out in bioreactors where metabolic
turnover can be directly detected. Moreover, NMR is also suitable for non-invasive
analysis of metabolism in living cells and organelles which has led to several
interesting studies focussed on specific metabolic mechanisms.
Wash cells 2-3 times with ice cold PBS to Centrifuge at 1500rpm, for 5 minutes, at
remove residual media 21° C and remove supernatant
Scrape cells off the flask and transfer to a Vortex cells in methanol and transfer to a
glass vial glass vial
Add 200 μL of chloroform and vortex for Add 325 μL of water and 400 μL of
30 seconds chloroform
Extraction
addition of methanol as tests with plastic vials showed compounds leaching from the
plastic with methanol and chloroform. More recently plastic tubes have become
available that are supposed to be chloroform proof.
Cell extracts recapitulate metabolic levels inside cells at one particular point of
time. Such cell extracts can be examined by NMR or by MS. If NMR is used, a
one-dimensional spectrum allows to reliably identify and quantify 30–40
188 Z. Eraslan et al.
metabolites using a 600 MHz spectrometer with a cryoprobe. The number of cells
required for such measurements depends on the cell size. For reference,
haematological cancer cells such as HL60 or K562, approximately 1–10 million
cells are required for one-dimensional spectra, depending on the specific type of
NMR experiments. For tracer-based flux experiments the number of cells required is
larger and should be at least 10 million for 1H-13C-HSQC spectra or isotope-filtered
spectra. For directly observed 13C-spectra even higher concentrations of metabolites
are required, although recently developed 13C-optimised micro-cryoprobes offer
huge sensitivity advantages with significant potential for metabolomics in cell-
based systems (Ramaswamy et al. 2013; Clendinen et al. 2014; Thomas et al.
2021). Most MS studies of metabolic mechanisms used GC-MS which is somewhat
more sensitive than NMR, >1 million cells are typically needed. LC-MS is much
more sensitive and can work with very small numbers of cells, very much depending
on the specific version of mass spectrometer that is used.
As already introduced, the main technologies behind metabolomics are NMR and
MS, each with specific advantages (Verpoorte, “Natural products drug discovery: on
silica or in-silico?”; Evans, “Compound Identification Strategies in Mass
Spectrometry-Based Metabolomics and Pharmacometabolomics”; Raftery, “Quanti-
tative NMR Methods in Metabolomics; Wishart, “Practical Aspects of NMR-Based
Metabolomics”; McKay, “Metabolomics using NMR- avoiding the “Black-Box””.).
NMR methods are well established in drug discovery, mainly for studying the
interaction of proteins with small molecules (Meyer and Peters 2003; Becker et al.
2018; Ludwig and Guenther 2009), and there is a range of methods to study cells.
Table 1 lists the overall advantages of NMR and MS approaches along with specific
sensitivities. For biologically driven users, it is particularly relevant to understand
the level of application in a cellular context, translated into accessible metabolite
concentrations, cell numbers and types of samples that can be studied. NMR requires
millions of cells (assuming haematological cancer cells such as HL60 and K562 as a
reference) whereas MS based methods are substantially more sensitive. NMR has a
significant advantage for quantification and to detect small but relevant changes.
Other advantages of NMR are the possibility to look a living cells non-invasively
enabling the measurement of time-courses, and to identify site-specific label
incorporation in tracer-based experiments. On the other hand, MS offers several
orders higher sensitivity, enabling even single-cell metabolomics and providing
access to a much wider range of metabolites.
MS can now be applied on a single-cell level, even combined with imaging
(Rappez et al. 2021). Table 1 provides an overview over the different analytical
methods that can be used providing information, their applicability, advantages and
limitations. GC-MS has commonly preferred for tracer-based approaches and there
are well-established protocols for this (Cascante and Marin 2008).
Metabolomics in Cell Biology 189
1D-spectra can also be used for absolute quantification of metabolites and this in a
highly reproducible manner. This can either be achieved by using an internal
standard such as Trimethylsilylpropanoic acid (TMSP) or by using an electronic or
software-generated reference signals using technologies which are now incorporated
in most spectrometers such as ERETIC and PULCON (Akoka et al. 1999; Watanabe
et al. 2016). These methods are now commonly used in NMR protocols for the
analysis of blood samples (Dona et al. 2014).
NMR also offers a large number of alternative pulse sequences which each yield
specific spectral features (reviewed in (Vignoli et al. 2019)). J-resolved (J-RES)
spectra are frequently used, in particular in samples with significant overlap between
signals. This is more common in plant-based samples than in samples arising from
mammalian cell lines. For the assignment of J-RES spectra, the Birmingham Metab-
olite Library (Ludwig et al. 2011) provides a unique resource. J-RES spectra are
usually processed in a manner where scalar couplings are removed thus providing
considerably simplified spectra. J-RES is also a quantitative method suitable to
calculate metabolite concentrations. Recently, a sophisticated 31P-selective J-RES
experiment has been used to identify several phosphometabolites from glycolysis
(Cox et al. 2021). While this method has limited sensitivity, it can distinguish
G6P/F6P or 2PG/3PG which is not possible using MS methods.
Many other NMR pulse sequences have been used to study metabolomics
samples. The most important ones are the TOCSY experiment which links chemi-
cally connected protons and HSQC spectra which correlate 1H and 13C resonances.
The two can also be combined in 2D-HSQC-TOCSY spectra which represent
invaluable tools for the assignment of metabolites in metabolomics. Importantly,
HSQC and HSQC-TOCSY spectra have been compiled in the COLMAR database
which also offers online tools for metabolite identification (Robinette et al. 2008;
Bingol et al. 2014, 2015a; Wang et al. 2020). HSQC-TOCSY spectra have also been
used in conjunction with MS for the assignment of unknown metabolites in an
approach that involves the simulation of NMR spectra using preliminary assignment
from MS (Bingol et al. 2015b). Although generally important for metabolomics, this
approach is less relevant for mammalian cells where the same 30–40 metabolites are
observed in almost all cell extracts. Although TOCSY spectra cannot be used to
derive absolute concentrations of metabolites, they have been used extensively for
the analysis of tracer-based metabolism, mainly by Fan and Lane (2008). HSQC
spectra offer considerable deconvolution of NMR spectra although at a much lower
sensitivity than 1D spectra and with a loss of quantification. The reason for this lies
in the nature of HSQCs which depend on scalar 1H-13C couplings (1JCH) which vary
considerably between metabolites. As a consequence, the signal intensity of a HSQC
spectrum depends not only on the concentration of a particular molecule but also on
the size of the coupling constant. Wan et al. suggested to determine these coupling
constants for a large number of metabolites and calculate the effect of1JCH on the
overall HSQC signal intensities (Wan et al. 2017), but this would require a signifi-
cant effort of collecting such a spectral database.
Nevertheless, HSQC spectra have been used in metabolomics, predominantly in
tracer-based metabolism (Saborano et al. 2019). For labelled samples, the effect of
Metabolomics in Cell Biology 191
the coupling constant on the HSQC intensity can be ignored when signals are
compared for the same metabolite for samples grown under different conditions.
In such cases, ratios between signals provide the relevant information on relative
label incorporations. Alternatively specific filtering methods have been employed to
quantify label incorporation. Such filters can be designed to become largely inde-
pendent of the JCH coupling constant (Reed et al. 2019).
A key advantage of NMR is that analyses are non-invasive, i.e. can be carried out
using living cells. This has been exploited in various real-time approaches where
metabolites have been measured over extended periods of time. Two main types of
real-time experiments have been reported. High-Resolution Magic Angle Spinning
(HR-MAS) stands for a technology which eliminates line broadening due to aniso-
tropic interactions in samples (Raftery, “Quantitative NMR methods in
metabolomics”). HR-MAS technologies for metabolomics have recently been
reviewed in (Edison et al. 2021). HR-MAS requires almost no sample preparation
and is non-destructive as long as cells can tolerate the high spinning forces.
HR-MAS enables the measurement of metabolites in intact tissue samples, small
animals (C. elegans (Blaise et al. 2007; Mobarhan et al. 2017) or drosophila (Sarou-
Kanian et al. 2015)) or cells. The need for MAS arises from the limited mobility of
molecules in semi-solid samples leading to broad lines in NMR spectra, either
caused by anisotropic dipole–dipole interactions or by local variations in magnetic
susceptibility. Both effects can be eliminated by rotating samples at the “magic
angle” of 54.7° to the magnetic field. This requires placing the sample in a small
rotor spinning at circa 6,000 Hz. For a 1.4 mm rotor at this spinning rate the sample
experiences 200,000 g acceleration which induces serious stress to the sample
(Edison et al. 2021). Recent developments may help to overcome this limitation,
with rotor synchronised pulse sequences it has been possible to obtain good spectra
of earth worms with as low as 100 Hz (Mobarhan et al. 2017). Micro-sized coils for
1 mm rotors were shown to further improve mass sensitivity (Lucas-Torres and
Wong 2019). HR-MAS has been used, for example, to study bacterial cells (Righi
et al. 2013), mycobacteria (Hanoulle et al. 2005a, 2006a; Lee et al. 2005) and also for
examination of mammalian cells (Nyblom et al. 2008; Garcia-Alvarez et al. 2011;
Gogiashvili et al. 2019; Judge et al. 2019; García-Álvarez et al. 2009; Vermathen
et al. 2021) and recently a cellular fungus (Edison et al. 2021). Although such
g-forces are seen as being too large for haematological cancer cells, Edison et al.
demonstrated applicability to chronic lymphoid leukaemia cells (Edison et al. 2021).
Recent works by Edison et al. were focussed towards real-time monitoring using
HR-MAS (Edison et al. 2021). As an alternative approach, cells have been used in
flow bioreactors (Hall et al. 2016) or have been embedded in matrices such as
agarose for real-time measurements (Koczula et al. 2016; Alshamleh et al. 2020).
Examples will be shown below.
GC-MS were reported in the 1960s covering sugars, amino acids, sterols, hormones,
hydroxyl acids, fatty acids, aromatics and many other small molecules (<650 Da), as
reviewed in (Fiehn 2016), before the term metabolomics had been coined. GC-MS
requires derivatization which has been established for a large range of small
molecules and is also well suited for metabolic flux analysis. The method benefits
greatly from large data bases (e.g. the NIST Mass Spectral Library (Babushok et al.
2007) and others reviewed in (Fiehn 2016)) and has played an important role in the
study of cellular metabolism and metabolic fluxes (Fischer et al. 2004). Unlike
NMR, GC-MS is not inherently quantitative but quantification is feasible using
external or internal standards (Fiehn 2016).
Most modern applications rely on LC-MS which does not require any derivatiza-
tion and is potentially truly global, covering thousands of features. LC-MS is a
widely used technology which is now available in many bioanalytical laboratories.
Unfortunately, there is much heterogeneity between LC-MS methods, different
upfront chromatographic approaches, different ionisation sources and different MS
hardware. The advantage of LC-MS for studying metabolomics in the context of
drug discovery is well illustrated by a recent workflow which combined
metabolomics, proteomics and transcriptomics datasets of 54 cancer cells to derive
a map of metabolite–transcriptional regulator interactions which was also used to
look at drug sensitivities (Ortmayr et al. 2019). Such studies typically involve a
systems biology layer that is used to combine large amounts of data. It should be
mentioned that direct flow-injection analysis time-of-flight mass spectrometry
(FIA-TOFMS) shows a lot of potential, in particular because measurements are
very fast, allowing for >5,000 samples to be run per day (Fuhrer et al. 2011) as
illustrated by a recent study by Ortmayr et al. (2019) who screened 54 cancer cell
lines linked with transcriptomic and proteomic data. This study reports 689 putative
assignments based on HMDB (Ortmayr et al. 2019). Typically, 250–350 metabolites
can be uniquely assigned using LC-MS, although not easily quantified.
For cell-based applications, the Biocrates kits approach has become popular as it
offers excellent quantification by using isotopically labelled reference compounds
(see (Illig et al. 2010) as an early example where this technology was used).
BIOCRATES kits now cover a wide range of hydrophilic and lipophilic metabolites
(Thompson et al. 2019) and offer a simple protocol for sample preparation.
Highly specialised MS metabolomics techniques have now reached a sensitivity
where single cells become amenable to investigation, even combined with imaging
(Rappez et al. 2021). This is particularly interesting to assess cancer cell heteroge-
neity and plasticity, manifested as metabolic heterogeneity. Several studies have
attempted to characterise metabolic heterogeneity in cancers (Rappez et al. 2021;
DeVilbiss et al. 2021; Lau et al. 2020), although this remains a challenging area of
MS research.
Metabolomics in Cell Biology 193
3 Case Studies
Tiziani and Lodi started to extract cells in 2009 to profile drug responses in Acute
Myeloid Leukaemia (AML) cell lines through NMR spectroscopy (Tiziani et al.
2009). For this work, they established protocols for sample preparation and charac-
terization which yield highly reproducible NMR spectra. PCA shows narrow clusters
for HL-60, K562 and KG1a cell lines, perfectly separated from each other. Using
these AML cell lines a combination of two repurposed drugs, bezafibrate (BEZ) and
medroxyprogesterone acetate (MPA) was tested. This work has established the
mechanism of action of these drugs to be in part related to the generation of reactive
oxygen species (ROS) with a profound effect on cell viability. The mechanism
behind this is that high ROS causes chemically driven decarboxylation of ɑ-keto-
acids, converting ɑ-ketoglutarate into succinate, pyruvate to acetate and oxaloacetate
into malonate, a mechanism that has recently also been observed in acute myeloid
leukaemia cells interacting with stromal cells (Vilaplana-Lopera et al. 2021).
Tiziani et al. later developed this approach into a high-content drug screening tool
(Tiziani et al. 2011). For this, he developed a new protocol which combined growing
cells in 96-well plates with in situ lysing and quenching of metabolism via SDS
addition and sonication. The cell extracts were subsequently analysed by J-RES-
NMR spectra. Using this protocol, several inhibitors could be detected by monitor-
ing changes of the lactate/pyruvate ratio. Validation in cell lines and in primary
cancer cells was demonstrated. The key advantage of this approach is that it is not
based on a single read-out but rather a high-content metabolic measurement
reflecting increasingly well-understood metabolic changes in cells. Similarly, Lodi
showed in a cellular model how a combinatorial treatment using phytochemical
combinations affects prostate cancer cells (Lodi et al. 2017). In this work, flux
measurements were used to monitor changes in glutamine metabolism in response
to treatment.
Eraslan et al. have conducted metabolomic studies to reveal therapeutic metabolic
targets for germinal centre-derived Burkitt lymphoma (BL) and Diffuse large B-cell
lymphoma (DLBCL) by applying various 1D and 2D NMR techniques (Eraslan
et al. 2021). A principal component analysis (PCA) that was performed on the 1D
NMR spectra of the media showed a clear separation between the BL cells and
DLBCL (Fig. 3a). The loading plot of the corresponding PCA analysis depicted that
the separation of BL from DLBCL mostly derived from the difference in extracellu-
lar asparagine level of BL and DLBL (Fig. 3b, c). 13C-tracer-based NMR metabolic
analysis that was carried out with cell extracts derived from BL and DLBCL cells
cultured in a medium with or no asparagine containing 13C-stable-isotope labelled
tracers depicted that asparagine regulates the synthesis of serine from glucose
(Fig. 3d) and serine uptake (Fig. 3e) in both BL and DLBCL cells. By combining
a metabolomic platform with a transcriptomic platform, they uncovered that BL cells
express the genes involved in serine biosynthesis at a higher level than DLBCL cells
do (Fig. 3f) and defined a new treatment model which solely works for BL. They
194 Z. Eraslan et al.
Fig. 3 The role of extracellular asparagine in regulation of serine metabolism in BL and DLBCL.
(a) Principal Component Analysis (PCA) for 1D 1H-NMR for media samples of Burkitt lymphoma
cell lines (red) and diffuse large B-cell lymphoma cell lines (blue) (different symbols for different
types of cells). (b) Representation of corresponding loadings plot showing metabolites which
mostly contributed to separation of BL cells from DLBCL cells. (c) Relative 1D 1H-NMR peak
intensities for asparagine in the growth media of DLBCL and BL cells. (d) 2D 13C-NMR analysis of
the cell extraction of Glor and Farage cells cultured in a medium with or without asparagine cultured
with [U-13C]-glucose. (e) 2D 13C-NMR analysis of the cell extraction of Glor and Farage cells
cultured in a medium with or without asparagine cultured with [U-13C]-serine. (f) Heatmap of
statistically significant altered genes (FDR <0.01) associated with serine metabolism from differ-
ential expression analysis. (g) Analysis of 2D 13C-NMR spectra of serine extracted from Glor and
Farage cells cultured in a medium cultured [U-13C]-glucose treated with the PHGDH inhibitor
NCT-503 at 10 μM. (h) The viability of BL31 (BL cell line) and SUDHL6 (DLBCL cell line) cells
after treatment with ASNase at 0.1 U/ml and ASNase at 0.1 U/ml plus NCT-503 for 24, 48 and 72 h
in BL cells while no synergistic effect on the viability of DLBCL cells (Fig. 3h).
Thus, the integrated multi-omic approach with metabolomics and transcriptomic
data has suggested an attractive new treatment model for BL.
To the best of our knowledge, the first application of HR-MAS NMR spectros-
copy to study cells is a publication by Nyblom et al. studying fatty acid formation
from [1-13C]glucose in insulinoma cell lines (Nyblom et al. 2008)the effect of
an antimitotic glycoside in glioma cells (García-Álvarez et al. 2009). Righi has
demonstrated the applicability of HR-MAS for Pseudomonas aeruginosa, a patho-
genic gram-negative bacterium (Righi et al. 2013). 25 metabolites could be assigned
in 1D and 2D spectra. In an elegant experiment, Hanoulle et al. showed the activa-
tion of the anti-tuberculosis prodrug ethionamide in mycobacteria (Hanoulle et al.
2005b, 2006b). Judge has demonstrated the use of HR-MAS for human multiple
myeloma cells showing that spectra can be obtained in 4 min thus allowing to
measure time-courses for continuous in vivo monitoring of metabolism. In multiple
myeloma cells, employing 13C-labelled ɑ-keto-isovalerate, conversion into valine
could be monitored over a time-course of 60 min (Fig. 4) (Judge et al. 2018). The
production of branched chain amino acids is an essential process for multiple
myeloma cells which have a high demand for amino acids needed for protein
production. For multicellular Neurospora crassa fungal cells, time-courses for
central carbon metabolism, amino acids, TCA cycle intermediates, energy storage
molecules and lipid and cell wall precursors were monitored for up to 10 h (Judge
et al. 2018).
Vermathen et al. have used non-small cell lung cancer (NSCLC) cells to study
Cisplatin (cisPt)-resistance metabolic adaptations employing HR-MAS (Vermathen
et al. 2021). High-quality spectra allowed the detection of 53 metabolites. PCA
showed close clustering of replicates and a clear separation with increasing resis-
tance against cisPt. De-induced cells showed similar behaviour as cisPt-resistant
cells, indicating a long-term memory after cisPt treatment. Metabolites predomi-
nantly changed in cisPt-resistant cells and their de-induced counterparts include
glutathione and taurine (Vermathen et al. 2021). This study clearly demonstrates
how HR-MAS NMR can be used to explain metabolic adaptations during drug
resistance.
Koczula et al. described experiments where chronic lymphoid leukaemia (CLL) cells
were embedded in a low-density agarose matrix to monitor metabolism over several
hours yielding time-courses as shown in Fig. 5 (Koczula et al. 2016). The agarose
matrix first helped to maintain cells afloat in a homogenous matrix and enable
measurements with small numbers of cells (0.5–1.0 million) which would otherwise
only have covered 1–2 mm at the bottom of the NMR tube. The concentration of
cells in this experiment determines the overall turnover rate. Limited diffusion of the
matrix caused oxygen depletion after circa 2 h which causes an abrupt increase of
lactate, alanine and glutamate production along with changes in the rate of glutamine
196 Z. Eraslan et al.
Fig. 4 Targeted isotopic CIVM-NMR measurements of metabolic flux in human leukaemia cells.
(a) 13C-labelled keto-isovalerate (KIV) was converted to valine. (b) 13C-labelled valine was not
converted to KIV, confirming unidirectional flux in ML cells. (c, d) Relative concentrations over
time of 13C-labelled KIV (orange) and 13 C-labelled valine (purple) compared to baseline noise
(grey), from spectral intensities within each region of the representative experiments in (a, b),
respectively, for 3 independent replicates. Reproduced from (Judge et al. 2018)
Glucose
21
15 60 Lactate 25 110
Oxygen [%]
14
50 100
10 20
7 40 90
Oxygen [%]
5
15
0 0 30 80
0 2 4 6 10
20 70
5
10 60
0 0 50
0 5 10 15 20 0 5 10 15 20
Intensity [AU]
0,15 0,9
0,04
0,1 0,7
0,03
0,05 0,5
0 0,3 0,02
0 5 10 15 20 0 5 10 15 20 0 5 10 15 20
Time [hours]
Fig. 5 Metabolic adaptation of CLL cells to hypoxia involves HIF-1α. Representative NMR time-
course data for a CLL pre-treated for 3 h with either 0, 20 or 100 nM chetomin, before transferring
into NMR for a further 24 h. Dashed lines on the lactate graph show oxygen levels inside the NMR
tube. The top left panel shows an expanded view of lactate kinetics during the first 6 h with a visible
shift after oxygen depletion which is inhibited by CTM. Data shown are representative of a
minimum of N = 3 CLL samples. Reproduced from (Koczula et al. 2016)
Fig. 6 (a) Light microscopy images of three spheroids aligned to the spatially resolved intensity
profiles of NMR detected metabolites shown in (b) and enlarged (y-axis) in (c). Reproduced with
permission from (Knitsch et al. 2021)
cell-based models. NMR is very well suited to study such systems in real time as
illustrated and spacially resolved across the speroid.
Both NMR and GC-MS have been used to study metabolic fluxes. Cascante has
established GC-MS based fluxomics using mainly GC-MS, first by establishing the
theoretically possible distribution of isotopes arising from selectively labelled glu-
cose, comparing [1-13C]glucose or [1,2-13C]glucose (Cascante and Marin 2008).
One of the key discoveries arising from this work has been the possibility to assess
pentose phosphate pathway activity from the label distribution in pyruvate and
Metabolomics in Cell Biology 199
lactate (Boros et al. 1998). Chong et al. have shown how the complementary nature
of GC-MS and NMR can be utilised to experimentally elucidate metabolic fluxes in
mammalian cells (Chong et al. 2017). While GC-MS yields mass increments for
each 13C-atom that is incorporated into a metabolite, without positional information,
NMR offers exactly the opposite, site-specific information of label incorporation.
For this, high-resolution HSQC NMR spectra were used, which resolve adjacent
13 13
C- C couplings. These couplings can become rather complex in mixtures of
metabolites with different label incorporations. An algorithm termed Combined
Analysis of NMR and MS Spectra (CANMS) has been described to simulate such
line shapes and use complementary GC-MS data to determine the exact label
incorporation for different atoms in glycolytic or TCA cycle metabolites. Examples
are shown for lactate and glutamate, for [1,2-13C]glucose as a metabolic precursor.
This approach is somewhat cumbersome but yields highly accurate site-specific label
incorporation.
MYC addition has long been understood to be major driver for many cancers,
associated with a strong glycolytic metabolism. Tarrado-Castellarnau et al. have
used a tracer-based GC-MS and transcriptomics analysis to study metabolic
consequences arising from the inhibition of CDK4/6 (Tarrado-Castellarnau et al.
2017). Cyclin-dependent kinases (CDK) are rational cancer therapeutic targets
owing to their essential role in the regulation of cell cycle progression at the G1
restriction point. Upon inhibition of CDK4/6, an accumulation of MYC was
observed. As a consequence, glutamine metabolism is upregulated, the mTOR
pathway gets activated while HIF1α-mediated responses to hypoxia get disturbed.
These MYC-driven adaptations to CDK4/6 inhibition render cancer cells highly
sensitive to inhibitors of MYC, glutaminase or mTOR and to hypoxia. This targeted
analysis was used to explore one avenue of a synergistic strategy by also inhibiting
GLS1 demonstrating strong antiproliferative effects. This work exemplifies how
metabolomics can be efficiently used in drug discovery to identify vulnerabilities to
develop new synergistic treatment regimes.
Moonira et al. used GC-MS metabolomics to study the mechanism of the anti-
diabetic drug metformin in hepatocytes (Moonira et al. 2020). They next used
[1,2-13C2]glucose to measure partitioning of flux between glycolysis (via PFK1)
and the pentose pathway by looking at mass isotopologues of lactate. Metabolism of
[1,2-13C2]glucose by the pentose pathway generates m1 and m0 lactate, whereas
glycolysis generates m2 and m0 lactate. Metformin increased the fraction of lactate
derived from glucose and increased the m2/m1 ratio. An AMPK activator decreased
the m2/m1 ratio. Cumulatively, metformin stimulates glycolysis via PFK1 in abso-
lute terms and relative to flux via the pentose pathway, whereas the AMPK activator
had the converse effect on the m2/m1 ratio. Metformin lowered cell G6P but not
ATP and decreased G6pc mRNA at high glucose. In contrast, direct allosteric
activators of AMPK were shown to have opposite effects compared to metformin
200 Z. Eraslan et al.
on glycolysis, gluconeogenesis and cell G6P. Overall this study showed how
metformin acts in hepatocytes by lowering G6P when challenged with glucose by
stimulation of glycolysis by an AMP-activated protein kinase-independent mecha-
nism through changes in allosteric effectors of phosphofructokinase-1 and fructose
bisphosphatase-1.
Selivanov has recently developed computational workflows based on their previ-
ously published software ISODYN (Selivanov et al. 2006) for the estimation of
quantitative dynamic flux maps using stable-isotope resolved metabolomics (SIRM)
(Selivanov et al. 2020) involving the extraction of mass spectra for metabolites of
interest, correction of the spectra for natural isotope abundance and the evaluation of
fluxes by simulation of mass spectra using a kinetic model based on ordinary
differential equations (ODEs) for isotopomers of metabolites. The biochemical
network is considered to derive a dynamic flux map. Foguet et al. has also extended
a 13C MFA-approach to seamlessly incorporate gene expression with metabolic flux
data (Foguet et al. 2019).
Metabolic flux maps used for 13C-MFA typically include less than 10% of the
reactions contained in human cells Genome-Scale Metabolic Models (GSMMs) used
to predict cell-level metabolic flux maps using human metabolic network
reconstructions and constraint-based methods (Nilsson and Nielsen 2017). In recent
years, it has been shown that GSMMs provide an advantageous platform for the
integration of transcriptomics and metabolomics data to study the mechanisms
underlying cell metabolic reprogramming and to identify new drug targets for cancer
therapy (Karakitsou et al. 2021). Recently, Cascante’s team developed genome-scale
metabolic flux maps for colon cancer cell lines integrating multi-omics data on
RECON2 human metabolic network reconstruction (Tarragó-Celada et al. 2021).
In brief, transcriptomic data, growth rates, respiration parameters, rates of metabolite
uptake and secretion, 13C resolved metabolomics and targeted metabolomics have
been used to parametrize the GSMMs. From the inferred metabolic flux maps,
cystine transport xCT system and methylenetetrahydrofolate dehydrogenase
1 (MTHFD1) have been identified and validated as new potential targets for meta-
static colorectal cancer.
Two studies from recent years exemplify how large-scale metabolomics screen-
ing can be used to map the larger landscape of cancer cell line metabolism. Li et al.
have profiled 225 metabolites in 928 cell lines from more than 20 cancer types from
the Cancer Cell Line Encyclopedia (CCLE) using LC-MS. Interrogation of metabo-
lite/gene associations reiterated the well-known link between IDH1 mutations and
the onco-metabolite 2-hydrogxyglutarate, which was also found in renal cell carci-
noma cell lines that did not have a mutated IDH1. Interestingly, by combining a
CRISPR–Cas9 knockout dataset with the Dependency Map (DepMap) exhibited that
the reduced asparagine levels in cells are associated with hypermethylation of the
promoter region of asparagine synthetase (ASNS). Further experiments showed that
asparagine starvation depletes cells with lowered ASNS expression. This is in
agreement with the targeted specific results of Eraslan et al. (2021) who showed
that some Burkitt’s lymphoma cell lines depend on asparagine for serine synthesis
(needed for one-carbon metabolism). Both studies allow the conclusion that
Metabolomics in Cell Biology 201
A second excellent example for high-throughput screening of cell lines used time-
of-flight mass spectrometry (FIA-TOFMS) to obtain metabolic profiles of 54 cancer
cell lines which were correlated with transcriptomic and proteomics data
highlighting the role of transcription regulators (Ortmayr et al. 2019).
FIA-TOFMS is by far the fastest metabolomics method which requires only 1 min
per sample (Fuhrer et al. 2011). The disadvantage is that it is somewhat limited to
fingerprinting, as it can yield multiple putative assignments for each feature. This
particular study has used HMDB (Wishart et al. 2009) and RECON2 (Thiele et al.
2013) for the annotation of metabolites. The authors constructed a global network
model incorporating the transcriptome, the proteome and the metabolome to predict
regulatory associations with central metabolic pathways. One key finding was a
global coordination between glucose and one-carbon metabolism, predicting a
selective sensitivity to antifolate drugs in cell lines with low glucose uptake as a
potential diagnostic marker for cancer cells that are more likely to respond to folate
synthesis inhibitors.
Such high-throughput metabolomics approaches generally require a high level
of computational support considering that the identification of metabolites is
difficult.
Fig. 7 NMR analysis of perfused human livers exposed to stable isotope-labelled fructose confirms
ketohexokinase (KHK) inhibition reduces lipogenesis and glycolysis. 1H-13C-HSQC spectra from
matched controls (CT: blue) and KHK inhibited (KHKi red) donor liver samples exposed to labelled
fructose for indicated periods (0.5–3 h). Reproduced with permission from (Shepherd 2021)
inhibitor-treated livers, at a similar rate and accumulated over time. Gradual conver-
sion of fructose to glycerol and glycerate was observed in control livers, as was
conversion to labelled sorbitol. Although labelled sorbitol appeared in the KHK
inhibited samples, there was a distinct lack of labelled glycerol and glycerate (Fig. 7)
(Shepherd 2021).
4 Conclusions
Acknowledgments U. Günther and M. Cascante acknowledge the funding from the European
Commission (HaemMetabolome [EC-675790]). M. Cascante acknowledges grant PID2020-
115051RB-I00 funded by MCIN/AEI/10.13039/501100011033, AGAUR-Generalitat de
Catalunya (2017SGR1033) and ICREA foundation (ICREA-Academia-2021).
References
Akoka S, Barantin L, Trierweiler M (1999) Concentration measurement by proton NMR using the
ERETIC method. Anal Chem 71:2554–2557
Alshamleh I et al (2020) Real-time NMR spectroscopy for studying metabolism. Angew Chem Int
Ed 59:2304–2308
ÄŒuperloviÄ M (2010) Cell culture metabolomics: applications and future directions. Drug Discov
Today 15:12
Babushok VI et al (2007) Development of a database of gas chromatographic retention properties of
organic compounds. J Chromatogr A 1157:414–421
Becker W, Bhattiprolu KC, Gubensäk N, Zangger K (2018) Investigating protein-ligand
interactions by solution nuclear magnetic resonance spectroscopy. ChemPhysChem 19:895–
906
Bhinderwala F, Powers R (2019) NMR metabolomics protocols for drug discovery. In: Gowda
GAN, Raftery D (eds) NMR-based metabolomics, vol 2037. Springer, New York, pp 265–311
Bingol K, Bruschweiler-Li L, Li D-W, Brüschweiler R (2014) Customized metabolomics database
for the analysis of NMR 1H–1H-TOCSY and 13C–1H-HSQC-TOCSY spectra of complex
mixtures. Anal Chem 86:5494–5501
Bingol K et al (2015a) Unified and isomer-specific NMR metabolomics database for the accurate
analysis of 13 C– 1 H HSQC spectra. ACS Chem Biol 10:452–459
Bingol K et al (2015b) Metabolomics beyond spectroscopic databases: a combined MS/NMR
strategy for the rapid identification of new metabolites in complex mixtures. Anal Chem 87:
3864–3870
Blaise BJ et al (2007) Metabotyping of Caenorhabditis elegans reveals latent phenotypes. Proc Natl
Acad Sci U S A 104:19808–19812
Boros LG et al (1998) Nonoxidative pentose phosphate pathways and their direct role in ribose
synthesis in tumors: is cancer a disease of cellular glucose metabolism? Med Hypotheses 50:55–
59
Bruntz RC, Lane AN, Higashi RM, Fan TW-M (2017) Exploring cancer metabolism using stable
isotope-resolved metabolomics (SIRM). J Biol Chem 292:11601–11609
Cascante M, Marin S (2008) Metabolomics and fluxomics approaches. Essays Biochem 45:67–82
Chong M et al (2017) Combined analysis of NMR and MS spectra (CANMS). Angew Chem Int Ed
56:4140–4144
Clendinen CS et al (2014) 13C NMR metabolomics: applications at natural abundance. Anal Chem
86:9242–9250
Cohen SM, Ogawa S, Shulman RG (1979) 13C NMR studies of gluconeogenesis in rat liver cells:
utilization of labeled glycerol by cells from euthyroid and hyperthyroid rats. Proc Natl Acad Sci
76:1603–1607
Cox N, Millard P, Charlier C, Lippens G (2021) Improved NMR detection of phospho-metabolites
in a complex mixture. Anal Chem 93:4818–4824
DeVilbiss AW et al (2021) Metabolomic profiling of rare cell populations isolated by flow
cytometry from tissues. Elife 10:e61980
Dietmair S et al (2012) Metabolite profiling of CHO cells with different growth characteristics.
Biotechnol Bioeng 109:1404–1414
Dona AC et al (2014) Precision high-throughput proton NMR spectroscopy of human urine, serum,
and plasma for large-scale metabolic phenotyping. Anal Chem 86:9887–9894
204 Z. Eraslan et al.
Duarte IF, Lamego I, Rocha C, Gil AM (2009) NMR metabonomics for mammalian cell metabo-
lism studies. Bioanalysis 1:1597–1614
Edison AS et al (2021) NMR: unique strengths that enhance modern metabolomics research. Anal
Chem 93:478–499
Eraslan Z, Papatzikas G, Cazier J-B, Khanim FL, Günther UL (2021) Targeting asparagine and
serine metabolism in germinal centre-derived B cells non-Hodgkin lymphomas (B-NHL). Cell
10:2589
Fan TW-M, Lane AN (2008) Structure-based profiling of metabolites and isotopomers by NMR.
Prog Nucl Magn Reson Spectrosc 52:69–117
Fiehn O (2016) Metabolomics by gas chromatography–mass spectrometry: combined targeted and
untargeted profiling. Curr Protoc Mol Biol 114:30.4.1
Finch G, Yilmaz A, Utz M (2016) An optimised detector for in-situ high-resolution NMR in
microfluidic devices. J Magn Reson 262:73–80
Fischer E, Zamboni N, Sauer U (2004) High-throughput metabolic flux analysis based on gas
chromatography–mass spectrometry derived 13C constraints. Anal Biochem 325:308–316
Foguet C, Jayaraman A, Marin S, Selivanov VA, Moreno P, Messeguer R, de Atauri P, Cascante M
(2019) p13CMFA: Parsimonious 13C metabolic flux analysis. PLoS Comput Biol 15(9):
e1007310
Fuhrer T, Heer D, Begemann B, Zamboni N (2011) High-throughput, accurate mass metabolome
profiling of cellular extracts by flow injection–time-of-flight mass spectrometry. Anal Chem 83:
7074–7080
García-Álvarez I, Garrido L, Doncel-Pérez E, Nieto-Sampedro M, Fernández-Mayoralas A (2009)
Detection of metabolite changes in C6 glioma cells cultured with antimitotic Oleyl glycoside by
1
H MAS NMR. J Med Chem 52:1263–1267
Garcia-Alvarez I, Fernandez-Mayoralas A, Garrido L (2011) Effect of drugs in cells and tissues by
NMR spectroscopy. Curr Top Med Chem 11:27–42
Gogiashvili M et al (2019) HR-MAS NMR based quantitative metabolomics in breast cancer.
Metabolites 9:19
Gonzalez-Mendez R, Wemmer D, Hahn G, Wade-Jardetzky N, Jardetzky O (1982) Continuous-
flow NMR culture system for mammalian cells. Biochim Biophys Acta 720:274–280
Gowda GAN, Abell L, Lee CF, Tian R, Raftery D (2016) Simultaneous analysis of major
coenzymes of cellular redox reactions and energy using ex vivo 1H NMR spectroscopy. Anal
Chem 8
Halama A (2014) Metabolomics in cell culture – a strategy to study crucial metabolic pathways in
cancer development and the response to treatment. Arch Biochem Biophys 564:100–109
Hall AMR et al (2016) Practical aspects of real-time reaction monitoring using multi-nuclear high
resolution flow NMR spectroscopy. Cat Sci Technol 6:8406–8417
Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144:646–674
Hanoulle et al (2005a) Monitoring of the ethionamide pro-drug activation .pdf
Hanoulle X et al (2005b) Monitoring of the ethionamide pro-drug activation in mycobacteria by 1H
high resolution magic angle spinning NMR. Biochem Biophys Res Commun 331:452–458
Hanoulle et al (2006a) Selective intracellular accumulation of the major .pdf
Hanoulle X et al (2006b) Selective intracellular accumulation of the major metabolite issued from
the activation of the prodrug ethionamide in mycobacteria. J Antimicrob Chemother 58:768–
772
Illig T et al (2010) A genome-wide perspective of genetic variation in human metabolism. Nat
Genet 42:137–141
Jang C, Chen L, Rabinowitz JD (2018) Metabolomics and isotope tracing. Cell 173:822–837
Jin ES, Sherry AD, Malloy CR (2013) Evidence for transaldolase activity in the isolated heart
supplied with [U-13C3]glycerol. J Biol Chem 288:2914–2922
Judge MT et al (2018) Continuous in vivo metabolism by NMR. bioRxiv. https://doi.org/10.1101/
501577
Judge MT et al (2019) Continuous in vivo metabolism by NMR. Front Mol Biosci 6:26
Metabolomics in Cell Biology 205
Reed MAC, Roberts J, Gierth P, Kupče Ē, Günther UL (2019) Quantitative isotopomer rates in real-
time metabolism of cells determined by NMR methods. Chembiochem 20(17):2207–2211.
https://doi.org/10.1002/cbic.201900084
Righi V, Constantinou C, Kesarwani M, Rahme LG, Tzika AA (2013) Live-cell high resolution
magic angle spinning magnetic resonance spectroscopy for in vivo analysis of Pseudomonas
aeruginosa metabolomics. Biomed Rep 1:707–712
Robinette SL, Zhang F, Brüschweiler-Li L, Brüschweiler R (2008) Web server based complex
mixture analysis by NMR. Anal Chem 80:3606–3611
Saborano R et al (2019) A framework for tracer-based metabolism in mammalian cells by NMR. Sci
Rep 9(1):2520
Sarou-Kanian V et al (2015) Metabolite localization in living drosophila using high resolution
magic angle spinning NMR. Sci Rep 5:9872
Schmidt DR et al (2021) Metabolomics in cancer research and emerging applications in clinical
oncology. CA Cancer J Clin 71:333–358
Selivanov VA, Marin S, Lee PWN, Cascante M (2006) Software for dynamic analysis of tracer-
based metabolomic data: estimation of metabolic fluxes and their statistical analysis. Bioinfor-
matics 22:2806–2812
Selivanov VA et al (2020) Software supporting a workflow of quantitative dynamic flux maps
estimation in central metabolism from SIRM experimental data. In: Nagrath D (ed) Metabolic
flux analysis in eukaryotic cells, vol 2088. Springer, pp 271–298
Shepherd EL (2021) Ketohexokinase inhibition improves NASH by reducing fructose-induced
steatosis and fibrogenesis. JHEP Rep 3:12
Szyperski T (1995) Biosynthetically directed fractional 13C-labeling of Proteinogenic amino acids.
Eur J Biochem 232:433–448
Szyperski T, Bailey JE, Wüthrich K (1996) Detecting and dissecting metabolic fluxes using
biosynthetic fractional 13C labeling and two-dimensional NMR spectroscopy. Trends
Biotechnol 14:453–459
Tarrado-Castellarnau M, de Atauri P, Tarragó-Celada J, Perarnau J, Yuneva M, Thomson TM,
Cascante M (2017) De novo MYC addiction as an adaptive response of cancer cells to CDK4/6
inhibition. Mol Syst Biol 13(10):940
Tarragó-Celada J et al (2021) Cysteine and folate metabolism are targetable vulnerabilities of
metastatic colorectal cancer. Cancers 13:425
Thiele I et al (2013) A community-driven global reconstruction of human metabolism. Nat
Biotechnol 31:419–425
Thomas JN et al (2021) Progress towards a higher sensitivity 13C-optimized 1.5 mm HTS NMR
probe. IEEE Trans Appl Supercond 31:1–4
Thompson JW et al (2019) International ring trial of a high resolution targeted metabolomics and
lipidomics platform for serum and plasma analysis. Anal Chem 91:14407–14416
Tiziani S et al (2009) Metabolomic profiling of drug responses in acute myeloid Leukaemia cell
lines. PLoS One 4:e4251
Tiziani S, Kang Y, Choi JS, Roberts W, Paternostro G (2011) Metabolomic high-content nuclear
magnetic resonance-based drug screening of a kinase inhibitor library. Nat Commun 2:545
Ugurbil K, Rottenberg H, Glynn P, Shulman RG (1978a) 31P nuclear magnetic resonance studies
of bioenergetics and glycolysis in anaerobic Escherichia coli cells. Proc Natl Acad Sci 75:
2244–2248
Ugurbil K, Brown TR, Den Hollander JA, Glynn P, Shulman RG (1978b) High-resolution 13C
nuclear magnetic resonance studies of glucose metabolism in Escherichia coli. Proc Natl Acad
Sci 75:3742–3746
Vermathen M, von Tengg-Kobligk H, Hungerbühler MN, Vermathen P, Ruprecht N (2021) 1H
HR-MAS NMR based metabolic profiling of lung cancer cells with induced and De-induced
cisplatin resistance to reveal metabolic resistance adaptations. Molecules 26:6766
Vignoli A et al (2019) High-throughput metabolomics by 1D NMR. Angew Chem Int Ed 58:
968–994
Metabolomics in Cell Biology 207
Vilaplana-Lopera N et al (2021) Crosstalk between AML and stromal cells triggers acetate secretion
through the metabolic rewiring of stromal cells. 2021.01.21.427406 Preprint. https://doi.org/10.
1101/2021.01.21.427406
Wan Q, Wang Y, Tang H (2017) Quantitative 13 C traces of glucose fate in hepatitis B virus-
infected hepatocytes. Anal Chem 89:3293–3299
Wang C et al (2020) COLMAR lipids web server and ultrahigh-resolution methods for
two-dimensional nuclear magnetic resonance- and mass spectrometry-based lipidomics. J
Proteome Res 19:1674–1683
Watanabe R et al (2016) Quantitative nuclear magnetic resonance spectroscopy based on PULCON
methodology: application to quantification of invaluable marine toxin, okadaic acid. Toxins
(Basel) 8:294
Wishart DS (2016) Emerging applications of metabolomics in drug discovery and precision
medicine. Nat Rev Drug Discov 15:473–484
Wishart DS et al (2009) HMDB: a knowledgebase for the human metabolome. Nucleic Acids Res
37:D603–D610
Yu M et al (2015) A resource for cell line authentication, annotation and quality control. Nature
520:307–311
Zhang A, Sun H, Xu H, Qiu S, Wang X (2013) Cell metabolomics. OMICS 17:495–501
NMR-Based Metabolomics to Evaluate
Individual Response to Treatments
Contents
1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210
1.1 The Individual Metabolic Phenotype . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
1.2 Modulation of the Individual Metabolic Phenotype . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 212
2 Disease Fingerprinting and Individual Response to Pharmacological Therapy . . . . . . . . . . . . 214
2.1 Celiac Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 214
2.2 Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
2.2.1 Breast Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 216
2.2.2 Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219
2.2.3 Colon Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
2.3 Viral Infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
2.3.1 Hepatitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
2.3.2 COVID-19 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 229
2.4 Chronic Obstructive Pulmonary Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232
2.5 Drug Toxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 233
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
# The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 209
V. Ghini et al. (eds.), Metabolomics and Its Impact on Health and Diseases,
Handbook of Experimental Pharmacology 277, https://doi.org/10.1007/164_2022_618
210 A. Vignoli et al.
Abstract
Keywords
1
H-NMR · Cancer · Celiac disease · COPD · Drug toxicity · Fingerprinting ·
Profiling · Response to treatment · Viral infections
1 Introduction
The use of metabolomics in personalized medicine originates from two basic facts:
Despite 1H NMR features a lower sensitivity (detection limit in the order of μM)
with respect to MS analysis, which downsizes the total number of measurable
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 211
In 2008, our research group (Assfalg et al. 2008) demonstrated, for the first time, that
the NMR detectable part of the metabolome of urine contains an invariant part,
which can be considered as the chemical signature of each individual. Multivariate
statistical analysis of multiple urine samples of different individuals enables the
definition of their “metabolic space”, where the metabolomic fingerprint of each
subject can be visualized and discriminated from that of the other subjects with an
accuracy close to 100%. Over a timescale of 10 years (Ghini et al. 2015a), in the
absence of important stressful perturbations, each individual still occupies its spe-
cific region of the metabolic space; daily intra-individual variability leads to small
fluctuations inside the individual metabolic space; conversely, shifts to other distinct
regions are associated to significant changes of the individual metabolomic pheno-
type as a consequence of the occurrence of important physio-pathological conditions
(Fig. 1).
212 A. Vignoli et al.
Beside the genetic components, all endogenous and exogenous metabolites derived
from extrinsic factors such as diet, drugs, gut microflora, stressors, and pollutants
contribute to the definition of the human metabolic phenotype. Thus, several studies
have been performed to understand the effects of mild dietary interventions on the
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 213
The celiac disease (CD) originates from an aberrant adaptive immune response
against gluten. The presence of a characteristic metabolomic fingerprint of CD has
been demonstrated by multivariate analysis of the NMR spectra of serum and urine
samples from affected subjects and healthy controls (Bertini et al. 2009, 2011a;
Vignoli et al. 2019). For example, when comparing adult patients with sex- and age-
matched controls, a discrimination accuracy in the range of 75–83% was obtained in
urine, which rises to 81–94% in serum, depending on the study cohorts. The
differences originate from variations in the levels of metabolites related to three
main mechanisms: malabsorption, altered energy metabolism, and altered gut micro-
flora (Bertini et al. 2009).
In serum samples, the main reported differences between CD patients and
controls consist in lower levels of several amino acids (asparagine, isoleucine,
methionine, proline, and valine) and of pyruvate, lactate, and lipids, and in higher
levels of glucose and 3-hydroxybutyric acid. The decreased levels of pyruvate and
lactate along with higher levels of glucose are indicative of an impaired glycolysis
process. Enhanced beta-oxidation and malabsorption can instead explain the lower
levels of amino acids and lipids observed. A possible increase in the use of ketonic
bodies as a source of energy in celiac patients is consistent with the high levels of
3-hydroxybutyric acid. Energy conversion from lipids and catabolism of ketonic
bodies is far less efficient than that from glucids; consistently, celiac patients often
report symptoms of fatigue. Further, urine samples of CD patients are characterized
by high levels of some metabolites related to the gut microbiota, i.e. indoxyl sulfate,
meta-[hydroxyphenyl] propionic acid, phenylacetylglycine, and p-cresolsulfate. All
these findings are consistent with the hypothesis that in CD patients the gut
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 215
microflora of the small bowel is altered or presents peculiar microbial species with
their own metabolome.
Notably, the so-called potential celiac patients, i.e., those individuals who tested
positive for the antibodies but have no evidence of intestinal damage, have an NMR
fingerprint that is similar to that of the overt patients, suggesting that the
dysmetabolism precedes the intestinal damage (Bernini et al. 2011a, b).
Celiac disease represents one example where dietary intervention induces rele-
vant changes in the metabolomic fingerprint (Bernini et al. 2011a, b). The only
available treatment for this disorder is to follow a gluten-free diet. One of the most
interesting findings was that the metabolic fingerprint of CD patients reverts to
normality after 12 months of a strict gluten-free diet. After the gluten-free diet,
normal levels of glucose and 3-hydroxybutyric acids are restored, and the sense of
fatigue tends to be reduced.
2.2 Cancer
Fig. 2 Types of samples that can be analyzed via NMR-based metabolomics: cells, tissues, and
biofluids. In particular, in Sect. 2.2, we will focus on the analysis of biofluids (i.e. serum, plasma,
and urine) to investigate pathophysiological conditions associated with breast cancer, lung cancer,
and colon cancer
(Gianni et al. 2012; Martin et al. 2017; Slamon et al. 2011; von Minckwitz et al.
2017, 2019) and the metastatic setting (Swain et al. 2013; Verma et al. 2012).
Metabolomics can distinguish patients with BC with respect to healthy controls
with high predictive accuracies: PLS models with Q2 > 0.6 for studies on serum/
plasma (Cala et al. 2018; Lécuyer et al. 2018; Silva et al. 2019; Singh et al. 2017;
Sitter et al. 2006; Slupsky et al. 2010; Suman et al. 2018; Tayyari et al. 2018;
Wojtowicz et al. 2020; Zhou et al. 2017), and models with Q2 > 0.5 for urinary
studies (Silva et al. 2019; Slupsky et al. 2010; Zhou et al. 2017) were obtained.
Metabolomics proved useful in discriminating the plasma profiles of patients with
different BC molecular subtypes in comparison to controls (Díaz-Beltrán et al.
2021): as compared to HER2-patients, the HER2+ group showed elevated aerobic
glycolysis, gluconeogenesis, and increased fatty acid biosynthesis with reduced
Krebs cycle. ER+ patients, as compared to ER- ones, showed elevated alanine,
aspartate, and glutamate metabolism, decreased glycerol-lipid catabolism, and
enhanced purine metabolism (Fan et al. 2016).
Given these premises, in the framework of BC precision oncology, it is crucial to
identify, in each BC subtype, patients at higher risk of cancer recurrence and drug-
response profiles able to guide patients’ management (Bendinelli et al. 2021; Vignoli
et al. 2021b). The NMR-based metabolomics has shown to be a valuable prognostic
instrument and examples of its application with the abovementioned objectives are
provided below.
Response to Chemotherapy
The possible association between different metabolic profiles and response to
chemotherapy has been extensively investigated via NMR metabolomics of blood
derivatives.
Pharmacometabolomics on blood plasma/serum with the aim of predicting which
patients will benefit most from a specific treatment has provided significant results.
First in 2012, our group demonstrated that NMR-based metabolomics may play a
role in identifying the best responding patients with HER2+ metastatic breast cancer
(MBC) treated with paclitaxel plus lapatinib (Tenori et al. 2012). With a similar
NMR approach, Jiang and colleagues predicted the response to gemcitabine/
carboplatin chemotherapy (Jiang et al. 2018).
More recently, the general research attention has focused on neo-adjuvant che-
motherapy (NAC), the preferred treatment strategy for patients with large primary
tumors and/or locally advanced disease (Thompson and Moulder-Thompson 2012).
NAC offers the unquestionable benefit of downstaging disease and reducing the
tumor size prior to surgery, thus making patients with inoperable tumors candidates
for surgical resection (Debik et al. 2019; Thompson and Moulder-Thompson 2012).
Nonetheless, only less than 30% of patients overall exhibit pathological complete
response (disappearance of all invasive cancer in the breast) to NAC (Wei et al.
2013), with lower rates of response observed in endocrine receptor-positive, HER2-
disease.
In 2020, our research group, for the first time, investigated the capability of
predicting pathological complete response, using the baseline host immune
218 A. Vignoli et al.
Disease Recurrence
Despite all efforts aimed at better stratifying BC patients, there is still a significant
proportion of early breast cancer patients who are overtreated. Clinicians are looking
for prognostic tools able to distinguish early BC patients at high risk of disease
recurrence, who need to be treated with aggressive adjuvant therapies, with respect
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 219
Fig. 3 (a) The study design of the three metabolomic studies performed in our laboratory (Tenori
et al. 2015; Hart et al. 2017; McCartney et al. 2019) on serum samples of BC patients. Early BC
patients were randomly divided into a training and a validation set. Using the early BC training set
and the metastatic BC group, a random forest model was calculated on the serum NMR data. Then
the early BC validation set was used to test the hypothesis that early BC patients at high risk of
cancer recurrence could be classified as metastatic by the RF model. (b) The results of the three
studies are summarized by reporting for each of them the RF proximity plot (early BC green dots,
metastatic BC red dots) along with the RF model accuracy, and the area under the ROC curve of the
validation set used for the recurrence prediction
Response to Immunotherapy
Among all the therapeutic strategies, immunotherapy has become an attractive
treatment modality for chemo-refractory solid tumors (Postow et al. 2015).
NSCLC cells have the ability to evade the immune system by expressing on their
surfaces certain “immune checkpoint” molecules that normally protect against
autoimmunity and inflammation, such as cytotoxic T-lymphocyte antigen-4
(CTLA-4), programmed cell death protein 1 and its ligand (PD-1 and PDL-1,
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 221
Fig. 4 (a) Experimental scheme to evaluate individual response to immunotherapy. (b) O-PLS
discrimination between NSCLC responders and non-responders to immune check points inhibitors,
adapted from Ghini et al. (2020a). Score plot, PC1 vs. PC2. Each symbol in the O-PLS score plot
represents the fingerprint of the NMR spectrum of each patient recruited for the study. Red dots:
nivolumab responder subjects; blue dots: nivolumab non-responder subjects. Red crosses:
pembrolizumab subjects predicted as responders; blue crosses: pembrolizumab subjects predicted
as non-responders
disulfide) (Hatae et al. 2020). One year later, in (2021), Nie X. et al. used LC-MS
profiling of early on-treatment serum to explore predictors of clinical outcomes of
anti-PD-1 treatment in 74 Chinese NSCLC patients. Serum samples were collected
2–3 weeks after the first infusion of PD-1 inhibitor. A small metabolite panel
consisting of hypoxanthine and histidine was identified and validated as a predictor
of treatment response, and high levels of both hypoxanthine and histidine were
associated with improved progression-free survival and overall survival (Nie et al.
2021). The completely different metabolites observed in the reported investigations
and proposed as treatment-efficacy biomarkers derive from different analytical
platforms, which allow for the observation of different panels of metabolites.
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 223
The strength of our study relies on the uniqueness of the 1H NMR fingerprinting,
which takes advantage of its intrinsically untargeted nature and high reproducibility.
This approach allowed us to identify a metabolomic signature associated with ICIs
response that is independent of metabolite assignment and acts as a stronger “col-
lective” biomarker with respect to a single molecule or to a panel of a few molecules.
Disease Recurrence
Using metabolomics to determine the distinct profiles of CRCs might allow for more
personalized or informed cancer therapy adjustments, contributing to precision
medicine (Wishart 2015). Indeed, CRC shows different characteristics of clinical
onset and individual response, even at the same pathological stage. Despite 80% of
CRCs are diagnosed at early stage and immediately treated with surgery, 35% of
these treated patients develop cancer relapse within 2–3 years after surgery (Guraya
2019). The assessment of recurrence risk in colon cancer primarily relies on the
pathological stage defined by the TNM system, based on the depth of tumor invasion
(T), lymph nodes involvement (N), and distant metastases (M). Risk stratification is
of fundamental importance for the choice of adjuvant treatment; however, only a
224 A. Vignoli et al.
small portion of patients benefits from it, with the majority being already cured by
primary surgery, and others experiencing disease relapse despite having received
adjuvant therapy. Improved identification of individuals who would benefit most
from adjuvant chemotherapy is a critical aim, particularly in older patients who are
more susceptible to treatment-related toxicity.
In 2021, our group demonstrated that NMR-based metabolomics on serum
samples can improve risk stratification in elderly patients with early CRC
(Di Donato et al. 2021). In this study, 169 serum samples, taken from three distinct
clinical trials, were collected before treatment from elderly CRC patients. Of these,
94 were collected from patients with early CRC (65 relapse free and 29 relapsed) and
75 from patients with metastatic CRC (Fig. 5). The model was built using a
supervised algorithm to discriminate the serum fingerprint of the relapse-free
patients in the early CRC cohort and the patients in the metastatic CRC cohort,
yielded 70% accuracy, 71% sensitivity, and 69% specificity. Then, with the hypoth-
esis that the metabolomic fingerprint of relapsed early CRC patients would be more
similar to that of patients with metastatic CRC, the model was used to predict the
remaining early CRC who had disease recurrence. Among the early CRC patients,
69% were correctly predicted as metastatic (and therefore considered at high relapse
risk). This suggests that even in the absence of clinically visible metastatic spread,
the metabolomic fingerprint of individuals with early CRC, who may have cancer
recurrence, has a potential “metastatic signature.”
Furthermore, when the metabolomic classification of all patients with early CRC
was analyzed using Kaplan–Meier curves, a strong prognostic effect was observed,
with patients classified at high risk by metabolomics (thus with a metabolomic
profile more similar to that of metastatic CRC patients) having a significantly higher
probability of disease relapse than those with a low risk metabolomic score
(High vs. Low risk: Hazard Ratio = 3.68, p-value = 0.001). Histidine and glutamine
were shown to be significantly decreased in the serum of metastatic CRC patients.
Previous evidences already suggested an association of glutamine with cancer
progression and poor cancer-specific survival (Bertini et al. 2012; Sirniö et al.
2019). Also, the downregulation of histidine was already observed in other studies
(Tan et al. 2013; Zhu et al. 2014) and its alteration was correlated with an increased
activity of histidine decarboxylase.
Our research group, in another recent paper (Vignoli et al. 2021a), investigated
the CRC relapse in the serum samples of a group of pre-chemotherapy CRC patients
undergoing surgery. We demonstrated that several differences between post- and
pre-operative serum samples are associated with cancer recurrence, in particular an
increment of HDL-Chol subfractions coupled with a decrement of VLDL-Chol
subfractions. These results may corroborate the hypothesis that the development of
CRC disrupts the physiological equilibrium of lipids and lipoproteins, leading to
lipid metabolic disorders (Zhang et al. 2014).
Urine samples have been also used for an NMR-based metabolomic approach to
predict relapse in a group of 62 CRC patients, yielding an area under the ROC curve
of 0.650 for cancer recurrence (Dykstra et al. 2017). Interestingly, here the authors
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 225
Fig. 5 Experimental scheme (a) and results (b) extracted from Di Donato et al. (2021). Dots in the
score plots of the multivariate supervised PCA-CA kNN model represent the fingerprint of the
NMR spectra of each patient recruited for the study. Red dots code for metastatic CRC (mCRC)
patients, blue dots for early CRC (eCRC) relapse-free patients, green triangles for eCRC relapsed
patients predicted as “metastatic,” while green squares represent the CRC relapsed predicted as
early CRC free from relapse. The Kaplan–Meier plot on the bottom right was used by the authors to
estimate the outcome distribution
226 A. Vignoli et al.
used the NMR data to predict treatment delay which could depend on reaction to
chemotherapy, reaching an area under the ROC curve values of 0.750.
2.3.1 Hepatitis
Viral hepatitis is a global health issue that affects millions of individuals and is
associated with a high fatality rate. Except for the hepatitis A virus (HAV), all
hepatotropic viruses, including hepatitis B, C, D, and E viruses (HBV, HCV, HDV,
and HEV), can induce chronic infections. HBV and HCV are the most common viral
causes of liver disease. According to the updated estimate of the WHO, there are
about 300 million people suffering from chronic HBV and six million people
suffering from chronic HCV (World Health Organization 2021).
Viral hepatitis chronic infection can cause progressive liver damage leading to
fibrosis and cirrhosis. Cirrhosis is the end-stage of every chronic liver disease and is
the major risk factor for hepatocellular carcinoma (HCC). HBV and HCV are the
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 227
leading cause of HCC worldwide, accounting for a significant mortality of more than
1.3 million death per year (Ringehan et al. 2017).
Prevention campaigns are one of the main weapons to limit the incidence of these
viruses, especially for HCV for which a vaccine is not yet available. Direct-acting
antivirals (DAAs) were approved in 2014, revolutionizing HCV therapy and
allowing almost all patients to be cured. DAAs are very effective and well-tolerated
and they constitute the gold standard for the treatment of HCV chronic infection in
patients at all stages of liver disease.
Metabolomics analysis is being utilized to better understand host–pathogen
interactions and screen host biospecimen for biomarkers that are characteristic of
the viral infection. In 2019, an interesting picture emerged of the metabolomic
fingerprint of HCV infection compared to both healthy subjects (HS) and
HBV-infected patients (Meoni et al. 2019), suggesting that the two viruses exert a
different impact on human metabolism. Indeed, by the comparison of the 1H-NMR
serum profiles of HCV- and HBV-infected patients, we identified characteristic
metabolomic fingerprints of the two viral infections, obtaining an overall discrimi-
nation accuracy of 86% (OPLS-DA algorithm). As expected, the serum fingerprint
of HCV- and HBV-infected patients resulted to be extremely different also from the
serum fingerprint of HS, with a classification accuracy of 98.7% in the model built to
discriminate HCV vs. HS, and a classification accuracy of 80% in the model built of
HBV vs. HS (Fig. 6). Similarly, Godoy et al. (2010), using the 1H-NMR fingerprint-
ing approach, were able to accurately discriminate (95% predictive accuracy) the
urine samples of HCV-infected patients from those of HCV-negative subjects,
corroborating the potential of 1H-NMR fingerprinting for the fast, non-invasive
diagnosis of HCV infection using a urine sample.
The common changes we detected in the serum metabolomic profile of HBV- and
HCV-infected patients when compared to HS (e.g., increased levels of
3-hydroxybutyrate, acetate, lactate, and pyruvate) support the hypothesis that these
viruses preferentially stimulate glycolysis over oxidative phosphorylation, analo-
gously to the Warburg effect in cancer (Okuda et al. 2002). Instead, in the compari-
son of HCV- and HBV-infected patients we noted a different behavior of several
metabolites, suggesting that the perturbation could be attributable to a direct action
of the two types of viruses rather than to the host response. Interestingly, the higher
levels of 2-oxoglutarate and 3-hydroxybutyrate in HCV patients compared to HBV,
also identified as biomarkers of cardiovascular disease and ketoacidosis, could
explain why some extrahepatic manifestations, such as cardiovascular diseases and
diabetes, are common in patients with chronic HCV and not in patients with HBV
(Bernini et al. 2011a, b; Chen et al. 2014; Du et al. 2014).
Metabolomics proved useful also for disease staging and for characterizing the
response to treatment. Anti-HCV treatment has advanced significantly in recent
years, with direct-acting antivirals (DAAs) replacing pegylated interferon and riba-
virin and providing effective treatment and less adverse effects. In our work, we
characterized also the metabolomic fingerprint and the profile of HCV patients
before and after effective DAA treatment. In this case, we identified a major
contribution of the low-molecular weight molecules in characterizing the changes
228 A. Vignoli et al.
Fig. 6 Fingerprinting of the Hepatitis C and B viruses in serum samples resulting from OPLS-DA
models. (a) HCV- (red dots) vs HBV- (purple triangles) infected patients; (b) HCV-infected
patients (red dots) vs HS (sky-blue triangles); (c) HBV-infected patients (purple triangles) vs
HS (sky-blue triangles). Adapted from Meoni et al. (2019)
introduced in the individual metabolomic profile by the therapy, suggesting also that
the perturbation in lipid metabolism induced by the infection persists after viral
eradication.
According to other studies (Cano et al. 2017; Nguyen et al. 2021; Sarfaraz et al.
2016) tyrosine and formate levels increase on passing from no/mild fibrosis to severe
fibrosis. Furthermore, differences in metabolite levels between patients with higher
and lower fibrosis scores were reduced after DAAs therapy, confirming that altered
metabolites are restored, most likely due to liver damage regression after viral
eradication.
Regarding the effect of treatment on sera of HBV-infected patients, Nguyen et al.
2021 recently published a study confirming that NMR-based metabolomics is
capable of revealing in serum samples a gradual metabolic transition from
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 229
2.3.2 COVID-19
The COVID-19 pandemic has heavily reshaped research activities worldwide.
Along with an incredibly fast development of vaccines that could bring the pandemic
under control, efforts were also directed toward the development of antiviral drugs
and monoclonal antibodies. Additionally, the pandemic has led to a rush to repur-
pose existing drugs. The pandemic has also stimulated researchers operating in fields
complementary to pharmacology and vaccinology to contribute to the understanding
of the physiopathology of the disease and to the characterization of risk factors and
response to treatments. Coordinated efforts have been searched via the creation of
networks that facilitate discussion among participants and communication of the key
results prior to publication, together with the establishment of strategic transnational
collaborations. Worth mentioning in this context are: (1) the Covid19-NMR project
(https://covid19-nmr.de/), dealing with the determination of the structures of RNA
and proteins of SARS-CoV-2 to investigate their drugability by small molecules;
(2) the NMR international COVID-19 Research Network (CV19 Research Net-
work), a metabolomics-based initiative consisting of several institutions that collab-
orate, using standardized NMR procedures, to detect the infection, predict outcomes
during hospitalization, and direct efforts toward Long COVID.
Suitable biofluids from COVID-19 patients for metabolomics are serum, plasma,
and urine, thanks to their low viral load, albeit collection of research samples during
the worse phases of the COVID-19 pandemic was a further burden for the clinicians.
In this frame, we present here some activities from our lab directed toward the use of
NMR-based metabolomics and lipoproteomics to characterize the COVID-19
metabolomic fingerprints, to monitor the effect of repurposed drugs and vaccination
follow-up (Ghini et al. 2022a; Meoni et al. 2021).
We and others (Ghini et al. 2022b; Ballout et al. 2021; Baranovicova et al. 2021;
Bizkarguenaga et al. 2021; Bruzzone et al. 2020; Julkunen et al. 2021; Kimhofer
et al. 2020; Lodge et al. 2021; Masuda et al. 2021) have shown that SARS-CoV-
2 infection induces profound changes in the metabolic phenotype of the patients
(Millet, “Prospective metabolomic studies in precision medicine. The AKRIBEA
project.”; Rogers, “The Metabolomics of Critical Illness”). Accordingly, 1H NMR
spectra of plasma samples of COVID-19 patients could be strongly discriminated
from the spectra of both healthy subjects and COVID-19-recovered subjects, with a
discrimination accuracy higher than 90% in both cases. The differences originate
from significant alterations in the concentrations of several metabolites and of a
panel of lipoprotein components. The metabolites and lipoprotein parameters that
are significantly dysregulated in COVID-19 acute subjects are listed in Fig. 7.
Characteristic trends in metabolite and lipoprotein levels are also observed as a
function of the disease severity (Ghini et al. 2022b). The analysis of the specific
changes and correlations with clinical data enabled the identification of potential
230 A. Vignoli et al.
Fig. 7 Metabolites and lipoprotein parameters that are found significantly up- or down-regulated in
COVID-19 patients with respect to healthy controls according to different Metabolomics papers
(Meoni et al. 2021; Bruzzone et al. 2020; Kimhofer et al. 2020; Lodge et al. 2021; Ballout et al. 2021;
Masuda et al. 2021; Julkunen et al. 2021; Baranovicova et al. 2021; Bruzzone et al. 2020) and COMETA
project (Ghini et al. 2022b). Up-/down-regulated features are indicated by red/blue cells. (a) Metabolites;
(b) Lipoprotein Main Parameters; (c) Lipoprotein Subfractions. Adapted from (Ghini et al. 2022b)
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 231
lipoproteins that are smaller for COVID-19-recovered subjects with respect to naïve
subjects, suggesting that a previous infection reduces the vaccine modulation of the
lipoproteome composition. The differences between the two groups involve the
nature and number of affected lipoprotein parameters. Additionally, the effect of
the second dose is essentially negligible for the COVID-19-recovered subjects
(Ghini et al. 2022a).
Fig. 8 Heatmap showing correlations among metabolites quantified in the urine, serum, exhaled
breath condensate (EBC), and sputum supernatant samples of 14 patients with COPD. Correlation
values (R) are reported as different degrees of color intensity (blue, negative correlation; red,
positive correlations). The figure is adapted and reprinted with permission from Vignoli et al.
(2020b). Copyright 2020 American Chemical Society
treatment with formoterol alone. This research line was further explored with the
analysis of urine, serum and sputum supernatant, demonstrating that different
biological matrices provide complementary information on the effects of
beclometasone/formoterol administration in COPD patients, and thus their integra-
tion could be useful for elucidating the metabolic mechanism of action of inhaled
corticosteroids (Vignoli et al. 2020b). An overview of the metabolite correlation
patterns among the different biofluids is presented in Fig. 8.
required across the whole process of drug development, from preclinical studies to
clinical trials, as well as in post-approval safety surveillance. Biochemical and
histological analyses are the major approaches used for drug safety evaluation.
These approaches are effective in most cases to determine the safety profile of
drug candidates (Wang et al. 2017). However, these methods can neither provide
detailed information nor explore the mechanisms of drug toxicity (Nicholson et al.
2002). Metabolomics is ideally positioned to address the challenges of drug toxicity
(Hertz, “Chemotherapy-induced Peripheral Neuropathy”). It represents a powerful
tool for collecting mechanistic information, indicating not only the extent of a toxic
insult but also its underlying mechanisms (Ramirez et al. 2013). Several examples
exist in the literature of NMR-based metabolomic applications in toxicology and
drug safety evaluation. Just to cite few of them, drugs and toxicants like flutamide
(Choucha Snouber et al. 2013), hydrazine (Garrod et al. 2005; Lindon et al. 2003),
and gentamicin (Lenz et al. 2005) have been assessed.
A typical example of a clinically useful class of molecules that presents high rates
of therapy discontinuation due to acute side effects is represented by
V-phosphodiesterase (PDE5) inhibitors (PDE5i) (Corona et al. 2016). These
molecules are the first-line therapy for erectile dysfunction (ED), a widespread health
problem in the general population of middle-aged men (Ayta et al. 1999; Yafi et al.
2016). Administration of PDE5i proved beneficial in 60–70% of patients with
varying etiologies of sexual dysfunction (Yafi et al. 2016). However, adverse effects
are a common drawback. In a study from our research group, we retrospectively
evaluated serum and urine NMR-based metabolomic profiles to identify prognostic
biomarkers of unfavorable efficacy/safety profile of PDE5i before drug administra-
tion. To the best of the authors’ knowledge, this is the first and only NMR-based
metabolomic study focused on PDE5i toxicity. Patients who are likely to experience
adverse effects can be identified with an accuracy of 77% using pretreatment serum
samples. Adverse drug reactions showed to be associated with high levels of
LDL-lipoprotein subfractions at baseline (Rocca et al. 2020). The results of this
pilot study underline how metabolomics may help in identifying the metabolic bases
underlying efficacy/safety profile of the PDE5i therapy.
References
Agusti AG, Sauleda J, Miralles C, Gomez C, Togores B, Sala E, Batle S, Busquets X (2002)
Skeletal muscle apoptosis and weight loss in chronic obstructive pulmonary disease. Am J
Respir Crit Care Med 166:485–489. https://doi.org/10.1164/rccm.2108013
Airoldi C, Ciaramelli C, Fumagalli M, Bussei R, Mazzoni V, Viglio S, Iadarola P, Stolk J (2016) 1H
NMR To explore the metabolome of exhaled breath condensate in α1-antitrypsin deficient
patients: a pilot study. J Proteome Res 15:4569–4578. https://doi.org/10.1021/acs.jproteome.
6b00648
Andersen M-BS, Rinnan Å, Manach C, Poulsen SK, Pujos-Guillot E, Larsen TM, Astrup A,
Dragsted LO (2014) Untargeted metabolomics as a screening tool for estimating compliance
to a dietary pattern. J Proteome Res 13:1405–1418. https://doi.org/10.1021/pr400964s
Antoniewicz MR (2018) A guide to 13C metabolic flux analysis for the cancer biologist. Exp Mol
Med 50:1–13. https://doi.org/10.1038/s12276-018-0060-y
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 235
Asiago VM, Alvarado LZ, Shanaiah N, Gowda GAN, Owusu-Sarfo K, Ballas RA, Raftery D
(2010) Early detection of recurrent breast cancer using metabolite profiling. Cancer Res 70:
8309–8318. https://doi.org/10.1158/0008-5472.CAN-10-1319
Assfalg M, Bertini I, Colangiuli D, Luchinat C, Schäfer H, Schütz B, Spraul M (2008) Evidence of
different metabolic phenotypes in humans. Proc Natl Acad Sci U S A 105:1420–1424. https://
doi.org/10.1073/pnas.0705685105
Ayta IA, McKinlay JB, Krane RJ (1999) The likely worldwide increase in erectile dysfunction
between 1995 and 2025 and some possible policy consequences. BJU Int 84:50–56. https://doi.
org/10.1046/j.1464-410x.1999.00142.x
Ballout RA, Kong H, Sampson M, Otvos JD, Cox AL, Agbor-Enoh S, Remaley AT (2021) The
NIH Lipo-COVID study: a pilot NMR investigation of lipoprotein subfractions and other
metabolites in patients with severe COVID-19. Biomedicine 9:1090. https://doi.org/10.3390/
biomedicines9091090
Baranovicova E, Bobcakova A, Vysehradsky R, Dankova Z, Halasova E, Nosal V, Lehotsky J
(2021) The ability to normalise energy metabolism in advanced COVID-19 disease seems to be
one of the key factors determining the disease progression – a metabolomic NMR study on
blood plasma. Appl Sci 11:4231. https://doi.org/10.3390/app11094231
Barnes PJ (2000) Chronic obstructive pulmonary disease. N Engl J Med 343:269–280. https://doi.
org/10.1056/NEJM200007273430407
Bendinelli B, Vignoli A, Palli D, Assedi M, Ambrogetti D, Luchinat C, Caini S, Saieva C, Turano
P, Masala G (2021) Prediagnostic circulating metabolites in female breast cancer cases with low
and high mammographic breast density. Sci Rep 11:13025. https://doi.org/10.1038/s41598-
021-92508-1
Bernini P, Bertini I, Calabrò A, la Marca G, Lami G, Luchinat C, Renzi D, Tenori L (2011a) Are
patients with potential celiac disease really potential? The answer of metabonomics. J Proteome
Res 10:714–721. https://doi.org/10.1021/pr100896s
Bernini P, Bertini I, Luchinat C, Tenori L, Tognaccini A (2011b) The cardiovascular risk of healthy
individuals studied by NMR metabonomics of plasma samples. J Proteome Res 10:4983–4992.
https://doi.org/10.1021/pr200452j
Bertini I, Cacciatore S, Jensen BV, Schou JV, Johansen JS, Kruhøffer M, Luchinat C, Nielsen DL,
Turano P (2012) Metabolomic NMR fingerprinting to identify and predict survival of patients
with metastatic colorectal cancer. Cancer Res 72:356–364. https://doi.org/10.1158/0008-5472-
11-1543
Bertini I, Calabrò A, De Carli V, Luchinat C, Nepi S, Porfirio B, Renzi D, Saccenti E, Tenori L
(2009) The metabonomic signature of celiac disease. J Proteome Res 8:170–177. https://doi.org/
10.1021/pr800548z
Bertini I, Luchinat C, Miniati M, Monti S, Tenori L (2014) Phenotyping COPD by H-1 NMR
metabolomics of exhaled breath condensate. Metabolomics 10:302–311. https://doi.org/10.
1007/s11306-013-0572-3
Bizkarguenaga M, Bruzzone C, Gil-Redondo R, SanJuan I, Martin-Ruiz I, Barriales D, Palacios A,
Pasco ST, González-Valle B, Laín A, Herrera L, Azkarate A, Vesga MA, Eguizabal C, Anguita
J, Embade N, Mato JM, Millet O (2021) Uneven metabolic and lipidomic profiles in recovered
COVID-19 patients as investigated by plasma NMR metabolomics. NMR Biomed 35:e4637.
https://doi.org/10.1002/nbm.4637
Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E,
Holgado E, Barlesi F, Kohlhäufl M, Arrieta O, Burgio MA, Fayette J, Lena H, Poddubskaya E,
Gerber DE, Gettinger SN, Rudin CM, Rizvi N, Crinò L, Blumenschein GR, Antonia SJ,
Dorange C, Harbison CT, Graf Finckenstein F, Brahmer JR (2015) Nivolumab versus docetaxel
in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 373:1627–1639. https://
doi.org/10.1056/NEJMoa1507643
Brahmer JR (2014) Immune checkpoint blockade: the hope for immunotherapy as a treatment of
lung cancer? Semin Oncol 41:126–132. https://doi.org/10.1053/j.seminoncol.2013.12.014
236 A. Vignoli et al.
Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB (2019) Colorectal cancer. Lancet 394:
1467–1480. https://doi.org/10.1016/S0140-6736(19)32319-0
Di Donato S, Vignoli A, Biagioni C, Malorni L, Mori E, Tenori L, Calamai V, Parnofiello A, Di
Pierro G, Migliaccio I, Cantafio S, Baraghini M, Mottino G, Becheri D, Del Monte F, Miceli E,
McCartney A, Di Leo A, Luchinat C, Biganzoli L (2021) A serum metabolomics classifier
derived from elderly patients with metastatic colorectal cancer predicts relapse in the adjuvant
setting. Cancers 13:2762. https://doi.org/10.3390/cancers13112762
Díaz-Beltrán L, González-Olmedo C, Luque-Caro N, Díaz C, Martín-Blázquez A, Fernández-
Navarro M, Ortega-Granados AL, Gálvez-Montosa F, Vicente F, Pérez del Palacio J,
Sánchez-Rovira P (2021) Human plasma metabolomics for biomarker discovery: targeting the
molecular subtypes in breast cancer. Cancers 13:147. https://doi.org/10.3390/cancers13010147
Du Z, Shen A, Huang Y, Su L, Lai W, Wang P, Xie Z, Xie Z, Zeng Q, Ren H, Xu D (2014)
1H-NMR-based metabolic analysis of human serum reveals novel markers of myocardial
energy expenditure in heart failure patients. PLoS One 9:e88102. https://doi.org/10.1371/
journal.pone.0088102
Dykstra MA, Switzer N, Eisner R, Tso V, Foshaug R, Ismond K, Fedorak R, Wang H (2017) Urine
metabolomics as a predictor of patient tolerance and response to adjuvant chemotherapy in
colorectal cancer. Mol Clin Oncol 7:767–770. https://doi.org/10.3892/mco.2017.1407
Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) (2005) Effects of chemotherapy and
hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the
randomised trials. Lancet 365:1687–1717. https://doi.org/10.1016/S0140-6736(05)66544-0
Euceda LR, Haukaas TH, Giskeødegård GF, Vettukattil MR, Engel J, Silwal-Pandit L, Lundgren S,
Borgen E, Garred Ø, Postma G, Buydens LMC, Børresen-Dale A-L, Engebråten O, Bathen TF
(2017) Evaluation of metabolomic changes during neoadjuvant chemotherapy combined with
bevacizumab in breast cancer using MR spectroscopy. Metabolomics 13:37. https://doi.org/10.
1007/s11306-017-1168-0
Fan Y, Zhou X, Xia T-S, Chen Z, Li J, Liu Q, Alolga RN, Chen Y, Lai M-D, Li P, Zhu W, Qi L-W
(2016) Human plasma metabolomics for identifying differential metabolites and predicting
molecular subtypes of breast cancer. Oncotarget 7:9925–9938. https://doi.org/10.18632/
oncotarget.7155
Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, Domine M,
Clingan P, Hochmair MJ, Powell SF, Cheng SY-S, Bischoff HG, Peled N, Grossi F, Jennens
RR, Reck M, Hui R, Garon EB, Boyer M, Rubio-Viqueira B, Novello S, Kurata T, Gray JE,
Vida J, Wei Z, Yang J, Raftopoulos H, Pietanza MC, Garassino MC, KEYNOTE-189
Investigators (2018) Pembrolizumab plus chemotherapy in metastatic non-small-cell lung
cancer. N Engl J Med 378:2078–2092. https://doi.org/10.1056/NEJMoa1801005
Garrod S, Bollard ME, Nicholls AW, Connor SC, Connelly J, Nicholson JK, Holmes E (2005)
Integrated metabonomic analysis of the multiorgan effects of hydrazine toxicity in the rat. Chem
Res Toxicol 18:115–122. https://doi.org/10.1021/tx0498915
Gettinger SN, Horn L, Gandhi L, Spigel DR, Antonia SJ, Rizvi NA, Powderly JD, Heist RS,
Carvajal RD, Jackman DM, Sequist LV, Smith DC, Leming P, Carbone DP, Pinder-Schenck
MC, Topalian SL, Hodi FS, Sosman JA, Sznol M, McDermott DF, Pardoll DM, Sankar V,
Ahlers CM, Salvati M, Wigginton JM, Hellmann MD, Kollia GD, Gupta AK, Brahmer JR
(2015) Overall survival and long-term safety of nivolumab (anti-programmed death 1 antibody,
BMS-936558, ONO-4538) in patients with previously treated advanced non-small-cell lung
cancer. J Clin Oncol 33:2004–2012. https://doi.org/10.1200/JCO.2014.58.3708
Ghini V, Di Nunzio M, Tenori L, Valli V, Danesi F, Capozzi F, Luchinat C, Bordoni A (2017)
Evidence of a DHA signature in the lipidome and metabolome of human hepatocytes. Int J Mol
Sci 18:359. https://doi.org/10.3390/ijms18020359
Ghini V, Laera L, Fantechi B, del Monte F, Benelli M, McCartney A, Tenori L, Luchinat C,
Pozzessere D (2020a) Metabolomics to assess response to immune checkpoint inhibitors in
patients with non-small-cell lung cancer. Cancers 12:3574. https://doi.org/10.3390/
cancers12123574
238 A. Vignoli et al.
Lerner HJ, Band PR, Israel L, Leung BS (1976) Phase II study of tamoxifen: report of 74 patients
with stage IV breast cancer. Cancer Treat Rep 60:1431–1435
Li C, Li Z, Zhang T, Wei P, Li N, Zhang W, Ding X, Li J (2019) 1H NMR-based metabolomics
reveals the antitumor mechanisms of triptolide in BALB/c mice bearing CT26 Tumors. Front
Pharmacol 10:1175. https://doi.org/10.3389/fphar.2019.01175
Li Y, Wang C, Li D, Deng P, Shao X, Hu J, Liu C, Jie H, Lin Y, Li Z, Qian X, Zhang H, Zhao Y
(2017) 1H-NMR-based metabolic profiling of a colorectal cancer CT-26 lung metastasis model
in mice. Oncol Rep 38:3044–3054. https://doi.org/10.3892/or.2017.5954
Lin Y, Ma C, Bezabeh T, Wang Z, Liang J, Huang Y, Zhao J, Liu X, Ye W, Tang W, Ouyang T, Wu
R (2019) 1 H NMR-based metabolomics reveal overlapping discriminatory metabolites and
metabolic pathway disturbances between colorectal tumor tissues and fecal samples. Int J
Cancer 145:1679–1689. https://doi.org/10.1002/ijc.32190
Lin Y, Ma C, Liu C, Wang Z, Yang J, Liu X, Shen Z, Wu R (2016) NMR-based fecal metabolomics
fingerprinting as predictors of earlier diagnosis in patients with colorectal cancer. Oncotarget 7:
29454–29464. https://doi.org/10.18632/oncotarget.8762
Lindon JC, Nicholson JK, Holmes E, Antti H, Bollard ME, Keun H, Beckonert O, Ebbels TM,
Reily MD, Robertson D, Stevens GJ, Luke P, Breau AP, Cantor GH, Bible RH, Niederhauser U,
Senn H, Schlotterbeck G, Sidelmann UG, Laursen SM, Tymiak A, Car BD, Lehman-
McKeeman L, Colet J-M, Loukaci A, Thomas C (2003) Contemporary issues in toxicology
the role of metabonomics in toxicology and its evaluation by the COMET project. Toxicol Appl
Pharmacol 187:137–146. https://doi.org/10.1016/S0041-008X(02)00079-0
Lodge S, Nitschke P, Kimhofer T, Coudert JD, Begum S, Bong S-H, Richards T, Edgar D, Raby E,
Spraul M, Schaefer H, Lindon JC, Loo RL, Holmes E, Nicholson JK (2021) NMR spectroscopic
windows on the systemic effects of SARS-CoV-2 infection on plasma lipoproteins and
metabolites in relation to circulating cytokines. J Proteome Res 20:1382–1396. https://doi.org/
10.1021/acs.jproteome.0c00876
Marianne C, Walsh LB (2006) Effect of acute dietary standardization on the urinary, plasma, and
salivary metabolomic profiles of healthy humans. Am J Clin Nutr 84:531–539. https://doi.org/
10.1093/ajcn/84.3.531
Márquez J, Matés JM (2021) Tumor metabolome: therapeutic opportunities targeting cancer
metabolic reprogramming. Cancers (Basel) 13:314. https://doi.org/10.3390/cancers13020314
Martin M, Holmes FA, Ejlertsen B, Delaloge S, Moy B, Iwata H, von Minckwitz G, Chia SKL,
Mansi J, Barrios CH, Gnant M, Tomašević Z, Denduluri N, Šeparović R, Gokmen E,
Bashford A, Ruiz Borrego M, Kim S-B, Jakobsen EH, Ciceniene A, Inoue K, Overkamp F,
Heijns JB, Armstrong AC, Link JS, Joy AA, Bryce R, Wong A, Moran S, Yao B, Xu F,
Auerbach A, Buyse M, Chan A, ExteNET Study Group (2017) Neratinib after trastuzumab-
based adjuvant therapy in HER2-positive breast cancer (ExteNET): 5-year analysis of a
randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 18:1688–1700.
https://doi.org/10.1016/S1470-2045(17)30717-9
Masuda R, Lodge S, Nitschke P, Spraul M, Schaefer H, Bong S-H, Kimhofer T, Hall D, Loo RL,
Bizkarguenaga M, Bruzzone C, Gil-Redondo R, Embade N, Mato JM, Holmes E, Wist J,
Millet O, Nicholson JK (2021) Integrative modeling of plasma metabolic and lipoprotein
biomarkers of SARS-CoV-2 infection in Spanish and Australian COVID-19 patient cohorts. J
Proteome Res 20:4139–4152. https://doi.org/10.1021/acs.jproteome.1c00458
McCartney A, Vignoli A, Biganzoli L, Love R, Tenori L, Luchinat C, Di Leo A (2018)
Metabolomics in breast cancer: a decade in review. Cancer Treat Rev 67:88–96. https://doi.
org/10.1016/j.ctrv.2018.04.012
McCartney A, Vignoli A, Hart C, Tenori L, Luchinat C, Biganzoli L, Di Leo A (2017)
De-escalating and escalating treatment beyond endocrine therapy in patients with luminal breast
cancer. Breast 34:S13–S18. https://doi.org/10.1016/j.breast.2017.06.021
McCartney A, Vignoli A, Tenori L, Fornier M, Rossi L, Risi E, Luchinat C, Biganzoli L, Di Leo A
(2019) Metabolomic analysis of serum may refine 21-gene expression assay risk recurrence
stratification. NPJ Br Cancer 5:26. https://doi.org/10.1038/s41523-019-0123-9
NMR-Based Metabolomics to Evaluate Individual Response to Treatments 241
Meijer RI, van Wagensveld BA, Siegert CE, Eringa EC, Smulders YM (2011) Bariatric surgery as a
novel treatment for type 2 diabetes mellitus: a systematic review. Arch Surg 146:744–750.
https://doi.org/10.1001/archsurg.2011.134
Meoni G, Ghini V, Maggi L, Vignoli A, Mazzoni A, Salvati L, Capone M, Vanni A, Tenori L,
Fontanari P, Lavorini F, Peris A, Bartoloni A, Liotta F, Cosmi L, Luchinat C, Annunziato F,
Turano P (2021) Metabolomic/lipidomic profiling of COVID-19 and individual response to
tocilizumab. PLoS Pathog 17:e1009243. https://doi.org/10.1371/journal.ppat.1009243
Meoni G, Lorini S, Monti M, Madia F, Corti G, Luchinat C, Zignego AL, Tenori L, Gragnani L
(2019) The metabolic fingerprints of HCV and HBV infections studied by nuclear magnetic
resonance spectroscopy. Sci Rep 9:4128. https://doi.org/10.1038/s41598-019-40028-4
Monleón D, Morales JM, Barrasa A, López JA, Vázquez C, Celda B (2009) Metabolite profiling of
fecal water extracts from human colorectal cancer. NMR Biomed 22:342–348. https://doi.org/
10.1002/nbm.1345
Montuschi P, Santini G, Mores N, Vignoli A, Macagno F, Shohreh R, Tenori L, Zini G, Fuso L,
Mondino C, Di Natale C, D’Amico A, Barnes PJ, Higenbottam T (2018) BREATHOMICS for
ASSESSING the effects of treatment and withdrawal with inhaled beclomethasone/formoterol
in patients with COPD. Front Pharmacol 9:258. https://doi.org/10.3389/fphar.2018.00258
Motta A, Paris D, Melck D, de Laurentiis G, Maniscalco M, Sofia M, Montuschi P (2012) Nuclear
magnetic resonance-based metabolomics of exhaled breath condensate: methodological aspects.
Eur Respir J 39:498–500. https://doi.org/10.1183/09031936.00036411
Nannini G, Meoni G, Amedei A, Tenori L (2020) Metabolomics profile in gastrointestinal cancers:
update and future perspectives. World J Gastroenterol 26:2514–2532. https://doi.org/10.3748/
wjg.v26.i20.2514
Nannini G, Meoni G, Tenori L, Ringressi MN, Taddei A, Niccolai E, Baldi S, Russo E, Luchinat C,
Amedei A (2021) Fecal metabolomic profiles: a comparative study of patients with colorectal
cancer vs adenomatous polyps. World J Gastroenterol 27:6430–6441. https://doi.org/10.3748/
wjg.v27.i38.6430
Nguyen HTT, Wimmer R, Le VQ, Krarup HB (2021) Metabolic fingerprint of progression of
chronic hepatitis B: changes in the metabolome and novel diagnostic possibilities.
Metabolomics 17:16. https://doi.org/10.1007/s11306-020-01767-y
Nicholson JK, Connelly J, Lindon JC, Holmes E (2002) Metabonomics: a platform for studying
drug toxicity and gene function. Nat Rev Drug Discov 1:153–161. https://doi.org/10.1038/
nrd728
Nie X, Xia L, Gao F, Liu L, Yang Y, Chen Y, Duan H, Yao Y, Chen Z, Lu S, Wang Y, Yang C
(2021) Serum metabolite biomarkers predictive of response to PD-1 blockade therapy in
non-small cell lung cancer. Front Mol Biosci 8:472. https://doi.org/10.3389/fmolb.2021.678753
Okuda M, Li K, Beard MR, Showalter LA, Scholle F, Lemon SM, Weinman SA (2002) Mitochon-
drial injury, oxidative stress, and antioxidant gene expression are induced by hepatitis C virus
core protein. Gastroenterology 122:366–375. https://doi.org/10.1053/gast.2002.30983
O’Sullivan A, Gibney MJ, Brennan L (2011) Dietary intake patterns are reflected in metabolomic
profiles: potential role in dietary assessment studies. Am J Clin Nutr 93:314–321. https://doi.
org/10.3945/ajcn.110.000950
Polyak K (2011) Heterogeneity in breast cancer. J Clin Invest 121:3786–3788. https://doi.org/10.
1172/JCI60534
Postow MA, Callahan MK, Wolchok JD (2015) Immune checkpoint blockade in cancer therapy. J
Clin Oncol 33:1974–1982. https://doi.org/10.1200/JCO.2014.59.4358
Ramirez T, Daneshian M, Kamp H, Bois FY, Clench MR, Coen M, Donley B, Fischer SM, Ekman
DR, Fabian E, Guillou C, Heuer J, Hogberg HT, Jungnickel H, Keun HC, Krennrich G, Krupp
E, Luch A, Noor F, Peter E, Riefke B, Seymour M, Skinner N, Smirnova L, Verheij E, Wagner
S, Hartung T, van Ravenzwaay B, Leist M (2013) Metabolomics in toxicology and preclinical
research. ALTEX 30:209–225. https://doi.org/10.14573/altex.2013.2.209
242 A. Vignoli et al.
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F (2021) Global
cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for
36 cancers in 185 countries. CA Cancer J Clin 71:209–249. https://doi.org/10.3322/caac.21660
Swain SM, Kim S-B, Cortés J, Ro J, Semiglazov V, Campone M, Ciruelos E, Ferrero J-M,
Schneeweiss A, Knott A, Clark E, Ross G, Benyunes MC, Baselga J (2013) Pertuzumab,
trastuzumab, and docetaxel for HER2-positive metastatic breast cancer (CLEOPATRA
study): overall survival results from a randomised, double-blind, placebo-controlled, phase
3 study. Lancet Oncol 14:461–471. https://doi.org/10.1016/S1470-2045(13)70130-X
Takis PG, Ghini V, Tenori L, Turano P, Luchinat C (2019) Uniqueness of the NMR approach to
metabolomics. TRAC-Trends Anal Chem 120:115300. https://doi.org/10.1016/j.trac.2018.10.
036
Tan B, Qiu Y, Zou X, Chen T, Xie G, Cheng Y, Dong T, Zhao L, Feng B, Hu X, Xu LX, Zhao A,
Zhang M, Cai G, Cai S, Zhou Z, Zheng M, Zhang Y, Jia W (2013) Metabonomics identifies
serum metabolite markers of colorectal cancer. J Proteome Res 12:3000–3009. https://doi.org/
10.1021/pr400337b
Tayyari F, Gowda GAN, Olopade OF, Berg R, Yang HH, Lee MP, Ngwa WF, Mittal SK,
Raftery D, Mohammed SI (2018) Metabolic profiles of triple-negative and luminal A breast
cancer subtypes in African-American identify key metabolic differences. Oncotarget 9:11677–
11690. https://doi.org/10.18632/oncotarget.24433
Tenori L, Oakman C, Claudino WM, Bernini P, Cappadona S, Nepi S, Biganzoli L, Arbushites MC,
Luchinat C, Bertini I, Di Leo A (2012) Exploration of serum metabolomic profiles and outcomes
in women with metastatic breast cancer: a pilot study. Mol Oncol 6:437–444. https://doi.org/10.
1016/j.molonc.2012.05.003
Tenori L, Oakman C, Morris PG, Gralka E, Turner N, Cappadona S, Fornier M, Hudis C, Norton L,
Luchinat C, Di Leo A (2015) Serum metabolomic profiles evaluated after surgery may identify
patients with oestrogen receptor negative early breast cancer at increased risk of disease
recurrence. Results from a retrospective study. Mol Oncol 9:128–139. https://doi.org/10.1016/
j.molonc.2014.07.012
Thompson AM, Moulder-Thompson SL (2012) Neoadjuvant treatment of breast cancer. Ann Oncol
23:x231–x236. https://doi.org/10.1093/annonc/mds324
Turano P (2014) Colorectal cancer: the potential of metabolic fingerprinting. Expert Rev
Gastroenterol Hepatol 8:847–849. https://doi.org/10.1586/17474124.2014.945912
Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, Pegram M, Oh D-Y, Diéras V,
Guardino E, Fang L, Lu MW, Olsen S, Blackwell K, EMILIA Study Group (2012) Trastuzumab
emtansine for HER2-positive advanced breast cancer. N Engl J Med 367:1783–1791. https://
doi.org/10.1056/NEJMoa1209124
Vignoli A, Ghini V, Meoni G, Licari C, Takis PG, Tenori L, Turano P, Luchinat C (2019) High-
throughput metabolomics by 1D NMR. Angew Chem Int Ed 58:968–994. https://doi.org/10.
1002/anie.201804736
Vignoli A, Mori E, Di Donato S, Malorni L, Biagioni C, Benelli M, Calamai V, Cantafio S,
Parnofiello A, Baraghini M, Garzi A, Monte FD, Romagnoli D, Migliaccio I, Luchinat C,
Tenori L, Biganzoli L (2021a) Exploring serum NMR-based metabolomic fingerprint of
colorectal cancer patients: effects of surgery and possible associations with cancer relapse.
Appl Sci 11:11120. https://doi.org/10.3390/app112311120
Vignoli A, Muraro E, Miolo G, Tenori L, Turano P, Di Gregorio E, Steffan A, Luchinat C, Corona
G (2020a) Effect of Estrogen receptor status on circulatory immune and metabolomics profiles
of HER2-positive breast cancer patients enrolled for neoadjuvant targeted chemotherapy.
Cancers 12:314. https://doi.org/10.3390/cancers12020314
Vignoli A, Orlandini B, Tenori L, Biagini MR, Milani S, Renzi D, Luchinat C, Calabrò AS (2019)
Metabolic signature of primary biliary cholangitis and its comparison with celiac disease. J
Proteome Res 18:1228–1236. https://doi.org/10.1021/acs.jproteome.8b00849
244 A. Vignoli et al.
Xi Y, Xu P (2021) Global colorectal cancer burden in 2020 and projections to 2040. Transl Oncol
14:101174. https://doi.org/10.1016/j.tranon.2021.101174
Yafi FA, Jenkins L, Albersen M, Corona G, Isidori AM, Goldfarb S, Maggi M, Nelson CJ, Parish S,
Salonia A, Tan R, Mulhall JP, Hellstrom WJG (2016) Erectile dysfunction. Nat Rev Dis Primers
2:16003. https://doi.org/10.1038/nrdp.2016.3
Ząbek A, Stanimirova I, Deja S, Barg W, Kowal A, Korzeniewska A, Orczyk-Pawiłowicz M,
Baranowski D, Gdaniec Z, Jankowska R, Młynarz P (2015) Fusion of the 1H NMR data of
serum, urine and exhaled breath condensate in order to discriminate chronic obstructive
pulmonary disease and obstructive sleep apnea syndrome. Metabolomics 11:1563–1574.
https://doi.org/10.1007/s11306-015-0808-5
Zhang X, Zhao X-W, Liu D-B, Han C-Z, Du L-L, Jing J-X, Wang Y (2014) Lipid levels in serum
and cancerous tissues of colorectal cancer patients. World J Gastroenterol 20:8646–8652.
https://doi.org/10.3748/wjg.v20.i26.8646
Zhou J, Wang Y, Zhang X (2017) Metabonomics studies on serum and urine of patients with breast
cancer using 1 H-NMR spectroscopy. Oncotarget 5. https://doi.org/10.18632/oncotarget.16210
Zhu J, Djukovic D, Deng L, Gu H, Himmati F, Chiorean EG, Raftery D (2014) Colorectal cancer
detection using targeted serum metabolic profiling. J Proteome Res 13:4120–4130. https://doi.
org/10.1021/pr500494u
Pharmacometabolomics of Asthma
as a Road Map to Precision Medicine
Contents
1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 248
2 Pharmacometabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 248
3 Applications of Pharmacometabolomics to Asthma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
4 Metabolomic Responses to Asthma Therapeutics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
4.1 Steroids (Inhaled and Oral) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252
4.2 Bronchodilators (Beta-Agonists) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 254
4.3 Leukotriene Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 256
4.4 Combined Therapeutics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257
5 Therapy or Severity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257
6 Clinical Subgroups and Endotypes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 258
6.1 Clinically Derived Subgroups (The “Bottom-Up” Approach) . . . . . . . . . . . . . . . . . . . . . . . 259
6.2 Omic-Driven Endotypes (The “Top-Down” Approach) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 261
6.3 Aspirin-Exacerbated Asthma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 263
7 Prevention . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 264
8 Challenges of Pharmacometabolomics in Asthma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
9 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 267
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 268
Abstract
Pharmacometabolomics applies the principles of metabolomics to therapeutics in
order to elucidate the biological mechanisms underlying the variation in
responses to drugs between groups and individuals. Asthma is associated with
broad systemic effects and heterogeneity in treatment response and as such is
# The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 247
V. Ghini et al. (eds.), Metabolomics and Its Impact on Health and Diseases,
Handbook of Experimental Pharmacology 277, https://doi.org/10.1007/164_2022_615
248 R. S. Kelly et al.
Keywords
Asthma · Endotyping · Pharmacometabolomics · Steroids · Treatment response
1 Introduction
2 Pharmacometabolomics
elucidate the underlying biological mechanisms behind complex diseases and pro-
vide insights into how individuals respond to the drugs designed for these conditions
(Kaddurah-Daouk and Weinshilboum 2014). Through the implementation of
pharmacometabolomic techniques and methods, we can improve understanding of
why different individuals may respond differently to the same treatment (Kaddurah-
Daouk and Weinshilboum 2014).
Asthma is associated with broad systemic effects including inflammation, oxida-
tive stress, and tissue remodeling (Sahiner et al. 2011), which arise as a consequence
of the complex interplay between genetic and environmental factors. Thus, it is ideal
for the application of a systems approach, such as metabolomics and subsequently
pharmacometabolomics (Zhu et al. 2019). Pharmacometabolomics offers the poten-
tial to improve our understanding of the actions of drugs on individuals across the
spectrum of asthma and facilitate the development of novel targeted therapies with
improved efficacy for all. Pharmacometabolomics offers a compelling and novel
route to precision medicine for individuals with asthma.
Over the last two decades, there has been growing interest in the role of the
metabolome in asthma. A large number of studies have demonstrated that the
metabolome, as measured in a variety of biosamples including blood, urine, and
exhaled breath consendate (EBC) can be leveraged to predict, diagnose, and assess
the severity of asthma (Wang et al. 2021; Kelly et al. 2017; Papamichael et al. 2021).
Importantly, several of the metabolomic perturbations reported as being associated
with asthma and asthma phenotypes including pathways relating to hypoxia
response, oxidative stress, immunity, inflammation, lipid metabolism, and the tricar-
boxylic acid cycle have been independently validated (Chung et al. 2014). More
recently, a body of asthma metabolomics researchers have shifted their focus to
study how an individual’s metabolome can influence their response to therapeutic
strategies. There is a growing interest in the utility of metabolomics for the develop-
ment of more efficacious asthma treatments (McGeachie et al. 2015; Svenningsen
and Nair 2017; Kelly et al. 2017, 2019b; Kuruvilla et al. 2019). Although only a
small number of studies of asthma have specifically labelled themselves as
“pharmacometabolomic” studies, many more have focused on the integration of
asthma phenotypes, metabolomics, and therapeutics.
Pharmacometabolomic asthma studies tend to take one of the two broad
approaches (Fig. 1): (1) the assessment of the metabolomic responses of individuals
with asthma to various therapeutics; or (2) the subgrouping of individuals with
asthma. These subgrouping or disease stratification studies can be further subdivided
into those that take a “bottom-up” and those that take a “top-down” approach. The
“bottom-up” approach clusters asthmatics based on their clinical characteristics as
they relate to therapy and/or management and then investigates the metabolomic
profiles of these subgroups (Fig. 1 [2a]). The “top-down” studies stratify disease
based on their metabolome, clustering individuals with asthma into subgroups based
250 R. S. Kelly et al.
on their metabolic profile and investigating the therapeutic needs of these subgroups
(Fig. 1 [2b]). In both instances, the end goal is to understand the mechanisms and
biology underlying disease in the different subgroups in order to identify novel
treatments targeted toward those underlying mechanisms.
In this chapter, we explore current perspectives and uses of
pharmacometabolomics in the study of asthma, describe the findings of the studies
to date, and reflect upon their potential roles in optimizing the treatment of
individuals with asthma as a route toward precision medicine.
Multiple therapeutics are currently utilized in the management of asthma and its
symptoms. These include inhaled corticosteroids (ICSs), oral corticosteroids, long-
acting β2-agonists (LABAs), short-acting β2-agonists (SABAs), leukotriene
inhibitors (blockers of the CysLT receptor, specifically), long-acting muscarinic
antagonists (LAMAs), short-acting muscarinic antagonists (SAMAs), and anti-IL
and anti-IgE antibody therapies, theophylline and cromolyn (Wendell et al. 2020)
(Table 1). These treatments, alone or in combination, comprise both long-term and
quick-relief medications that aid in reducing symptoms among asthmatic patients by
targeting different biological pathways and processes. However, their efficacy varies
Pharmacometabolomics of Asthma as a Road Map to Precision Medicine 251
across populations (Larsson et al. 2020), and many asthmatics do not respond to the
“one-size-fits-all” management approach. Patients who do not respond to standard
care are responsible for most of the asthma-related economic burden (Chung et al.
2014). Researchers are now beginning to leverage pharmacometabolomics to try and
better understand the mechanisms of action of these therapeutics and the subgroups
that may benefit from these treatments.
252 R. S. Kelly et al.
Steroids reduce the airway inflammation associated with asthma and are the most
commonly prescribed drugs for this condition (Wendell et al. 2020). However, their
efficacy varies between individuals; approximately 25–35% of asthma patients
either do not respond or respond poorly to inhaled corticosteroids (ICS) (Kachroo
et al. 2021). As such, they are often prescribed in combination with other therapeu-
tics in order to optimize treatment. Identifying non- or poor-responders could help to
improve treatment efficacy and to reduce the potential side effects associated with
overtreatment (Kachroo et al. 2021), which can include weight gain, high blood
pressure, muscle weakness, osteoporosis, and an increased risk of infection, in
addition to the increasingly understood consequence of adrenal suppression
(Grennan and Wang 2019).
There are two different categories of steroids that work to reduce asthma
symptoms: long-term control and quick-relief medications (Wendell et al. 2020).
Long-term control medications are typically ICS such as beclomethasone
dipropionate, budesonide, ciclesonide, flunisolide, fluticasone furoate, mometasone
furoate, and fluticasone propionate. Quick-relief medications include both oral and
intravenous corticosteroids such as prednisolone, prednisone, and methylpredniso-
lone (Wendell et al. 2020). The main action of corticosteroids is to prohibit tran-
scription of inflammatory genes (Barnes 2006). They also induce transcription of
anti-inflammatory and immune regulation genes including beta2-adrenergic
receptors, secretory leukocyte inhibitory protein, and mitogen-activated protein
kinase phosphatase-1 through the glucocorticoid receptor-mediated transactivation,
further contributing to their anti-inflammatory properties (Newton and Giembycz
2016; Matera et al. 2020; Wendell et al. 2020).
Among the treatment options for asthma, steroids are by far the most commonly
studied in a pharmacometabolomic framework. Within this category, studies of ICS
dominate the research. Yet despite the relatively large body of literature, there is
some disagreement regarding the influences of steroids on the metabolome.
Ferraro et al. performed breathomics (metabolomic profiling of the EBC) in
26 children with asthma before and after a 10-week course of inhaled
beclomethasone dipropionate, a common corticosteroid, and in 16 children without
asthma (Ferraro et al. 2020). Although the children with asthma demonstrated
improvements in lung function and asthma control following treatment, there was
no change in the EBC metabolome, and no differences in the urinary endogenous
steroid profile. They also demonstrated that the EBC metabolome could distinguish
between children with and without asthma and that steroid treatment did not affect
this ability, with notable differences in prostaglandin, fatty acid, and
glycerophospholipid metabolites between children with and without asthma. Inter-
estingly, they identified more metabolomic differences between the asthmatic and
non-asthmatic children before the steroid treatment. This could be interpreted to
suggest that rather than causing further dysregulation to the metabolome the treat-
ment actually returned it to a state more akin to a non-asthmatic control. (Ferraro
et al. 2020). Another breathomics study investigated ethane, a product of lipid
Pharmacometabolomics of Asthma as a Road Map to Precision Medicine 253
ensure that both inflammation and smooth muscle relaxation are achieved (Wendell
et al. 2020). The most common types of bronchodilators include short-acting-beta-
agonists (SABAs) which are used when an individual needs an immediate muscle
relaxation effect during an asthma exacerbation, while long-acting-beta-agonists are
taken daily to maintain muscle relaxation over long periods of time (Wendell et al.
2020). Commonly prescribed SABAs include albuterol, epinephrine, isoproterenol,
metaproterenol, salbutamol, pirbuterol, and terbutaline. The most commonly utilized
LABAs are salmeterol, formoterol, BI-167107, vilanterol, indacaterol, and
olodaterol (Wendell et al. 2020).
To date, only a small number of studies have considered beta-agonists and asthma
in a metabolomics framework, with most investigating treatment related
metabolomic changes. Urinary levels of bromotyrosine, a marker reactive
brominating oxidants formed by eosinophil-catalyzed oxidation, have been shown
to be associated with the use of beta-agonists (Wedes et al. 2011). Similarly
increased serum levels of lactate have been significantly associated with albuterol
use in adult asthmatics into two independent studies (Lewis et al. 2014; Rodrigo and
Rodrigo 2005), which is thought to be a direct result of therapy (Rodrigo 2014).
McGeachie et al. also focused on SABAs, investigating differences in the levels
of serum metabolites between eight individuals with asthma who reported SABA
use in the last 7 days compared to 12 individuals with asthma who did not use
SABAs in the same time period (McGeachie et al. 2015). While no metabolites
survived stringent correction for multiple testing, they did observe a nominally
significant decrease in a monohydroxy derivative of arachidonic acid in those
individuals not using SABAs. Together with linoleic acid metabolism and
sphingolipid metabolism, arachidonic acid metabolism ranked as the top
dysregulated pathway with respect to SABA use as a proxy for asthma control
(McGeachie et al. 2015). They then integrated the metabolomic data with genome-
wide genotype, gene expression data and methylation data using a Conditional
Gaussian Bayesian Network (CGBN) to identify the strongest predictors of asthma
control across these omic types. Integrative over representation analyses of the top
multi-omic predictors supported the importance of these pathways in SABA
use/asthma control (McGeachie et al. 2015).
Taking a slightly different approach two studies considered the influence of
age-related changes on the beta-agonist – metabolite relationship. Kelly et al.
focused on the metric of bronchodilator response (BDR). BDR measures spirometry
before and after beta-agonists/bronchodilator treatment to establish reversibility of
airflow. As BDR is known to decline with age, this study aimed to determine
whether the BDR-age relationship was mediated by underlying metabolite levels.
They were able to identify and validate in two independent populations blood
metabolites that interacted with age in BDR. Increased levels of
2-hydroxyglutarate in the blood of an individual with asthma may exacerbate the
association between age and BDR decline, while increased levels of cholesterol
esters, GABA, and ribothymidine may attenuate the age-associated BDR decline
(Kelly et al. 2019b). These metabolites have been shown to play various roles in lung
function, inflammation, and immune function, but have not previously been directly
256 R. S. Kelly et al.
related to treatment (Kelly et al. 2019b). A second study (Sordillo et al. 2020) also
investigated whether circulating metabolites mediate age-related changes in bron-
chodilator response (BDR) for individuals with asthma. Sordillo et al. determined
that a portion of the effect of age on BDR acted indirectly through phosphatidylcho-
line plasmalogens. It is hypothesized that plasmalogens which protect against
oxidative stress may mediate lung function responses through their ability to alter
the structural properties of lung surfactants, and they are known to be enriched in the
smooth muscle that bronchodilator acts upon (Bozelli et al. 2021). As such, the
authors concluded that plasmalogens may serve as potential pharmacologic targets
for enhancement of lung function in individuals with asthma (Sordillo et al. 2020).
Cysteinyl leukotrienes (CysLTs) are a class of bioactive fatty acids associated with
airway inflammation (Wendell et al. 2020). These fatty acids are released by
eosinophils during an asthma exacerbation as proinflammatory mediators, which
contribute to the bronchoconstriction seen in asthma patients (Wendell et al. 2020).
The receptors for these leukotrienes exist on the cell surface of monocytes,
eosinophils, lung macrophages, and resident mast cells, and the interaction of the
CysLTs with their receptors has been found to induce anaphylaxis and cause the
prolonged contraction of bronchial smooth muscle, that is characteristic of asthma
(Augstein et al. 1973; Yokomizo et al. 2018). Leukotrienes further contribute to the
pathogenesis of asthma and airway obstruction by recruiting eosinophils and
neutrophils in the mucosa of the airway (Bisgaard 2001; Foster and Chan 1991;
Smith et al. 1993; Henderson et al. 1996).
There are two critical stages along the pathway at which the formation of
leukotrienes and the subsequent inflammatory response in the airways can be
prevented. The first is at the 5-LOX enzymatic step where arachidonic acid is
converted into LTA4 by 5-lipoxygenase, where Zileuton blocks this pathway
(Walter et al. 1987; Peters-Golden and Henderson 2007). The second is at the
G-protein coupled receptor (GPCR) CysLTR1 location, where LTC4 and LTD4
are the substrates for downstream signaling events (Wendell et al. 2020). Drugs
including Montelukast, Zafirlukast, and Pranlukast act as leukotriene receptor
antagonists (LTRAs) by targeting the CysLTR1 to obstruct the signaling pathway
(Peters-Golden and Henderson 2007; Wendell et al. 2020). Treatment with these
drugs therefore results in a decrease in the level of leukotrienes present in the body,
leading to fewer and less severe asthma exacerbations. Montelukast specifically was
found to significantly reduce eosinophil infiltration in asthma patients, while
Zileuton reduces the production of leukotrienes (Yokomizo et al. 2018; Bruno
et al. 2018).
The actions of these therapeutics are supported by cell-based studies (Werner
et al. 2019). Profiling of the lipid metabolome of healthy human macrophages
demonstrated that Zileuton reduces the intensity of the immune response by
blocking the pathway for the creation of the enzyme 5-LOX. It also showed a
Pharmacometabolomics of Asthma as a Road Map to Precision Medicine 257
In individuals with severe asthma, beta-agonists and ICS are commonly prescribed
in combination. To date very few studies have explored the combined metabolomic
effects of these therapeutics. In addition to the Loureiro et al. article (Loureiro et al.
2014), which was primarily concerned with steroid non-responsiveness in
individuals with asthma on inhaled corticosteroids and long-acting b-agonists,
Quan-Jun et al. investigated the impact on the metabolome of childhood asthmatics
receiving both the glucocorticosteroid budesonide and the beta-2-agonist salbutamol
(Quan-Jun et al. 2017). There was a clear difference in the serum metabolomic
profile of children on the combined therapeutic when compared to children receiving
neither, and this difference was driven by increased levels of 4-hydroxybutyrate,
lactate, cis-aconitate, 5-HIAA, taurine, trans-4-hydroxy-l-proline, tiglylglycine,
3-hydroxybutyrate, 3-methylhistidine, and glucose and decreased levels of alanine,
glycerol, arginine, glycylproline, 2-hydroxy-3-methylvalerate, creatine, citrulline,
glutamate, asparagine, 2-hydroxyvalerate, and citrate. There was no difference in the
metabolome when considering either treatment alone, suggesting it is the
combination of therapies that drives the metabolomic shift (Quan-Jun et al. 2017).
Furthermore, the authors determined there were no metabolomic effects of a further
beta-2-agonist, procaterol. Similar results were observed when considering the urine
metabolome of these children. Taken together these results suggested possible
“metabolic reprogramming” as a result of inhaled budesonide and salbutamol in
asthmatic children, that is particularly pronounced in the arginine and proline
metabolism pathway.
5 Therapy or Severity
Several studies report metabolomic changes that associate with the use of commonly
administered asthma therapeutics. However, when considering these results, the
issue of cause and effect must be considered. Treatment is inherently linked to the
disease itself, whereby more severe cases or cases with a particular phenotype are
more likely to receive therapy (confounding by indication). Considering this, many
pharmacometabolomic studies of the effect of a particular therapeutic on the
metabolome may be confounded by the clinical indication for that therapy. This is
reflected in the fact that a number of metabolites and metabolomic pathways found to
be associated with treatment in the studies previously mentioned have also been
258 R. S. Kelly et al.
linked to both the presence of asthma and its severity. For example, linoleic acid
metabolism and arachidonic acid metabolism have been associated with asthma
through their roles as lipid mediators of inflammation (Wendell et al. 2014).
A number of the asthma pharmacometabolomics studies have addressed the
question of cause versus effect directly. In Paredi et al.’s study of exhaled ethane
in asthma, they determined that ethane was higher in the non-treated cases, but that
even within this group it was highest in those with the greatest degree of lung
function. This suggests oxidative stress is correlated with severity itself and that the
observed association between ethane as a marker of oxidative stress and with steroid
use is due to the fact that treatment is reducing oxidative stress in this group (Paredi
et al. 2000). The fact that Ferraro et al. found few differences between the steroid-
treated EBC metabolome and healthy controls, as compared to the untreated
metabolome versus controls could support this hypothesis, particularly given no
differences were seen within the same individuals before and after treatment (Ferraro
et al. 2020). In further agreement McGeachie et al. remarked that in the confines of
their study “whether these metabolomic changes reflect the drug or the phenotype
cannot be determined” (McGeachie et al. 2015). It is also of interest to note that
lactate was implicated in several different therapeutics, which may suggest that
dysregulation of this metabolite and its pathways relates more to the need for
treatment than to the treatment itself.
However, it should be noted that the long-term effects of ICS use on endogenous
cortisol and cortisone levels and on dehydroepiandrosterone sulfate does suggest
that treatments for asthma can cause metabolomic shifts independent of the disease.
Furthermore, changes in lactate levels have been directly attributed to treatment
(Kelly et al. 2019b). This suggests that whether or not therapy directly effects the
metabolome is metabolite specific and therapy specific, it may occur in some
instances but not others. Given findings from several studies that have considered
interactions, these relationships are also likely to be impacted by external factors
such as age and sex.
between metabolic profile and ICS treatment is greater than that between metabolic
profile and disease (Reinke et al. 2017).
In their study of the discriminatory ability of the urine metabolome for asthma
phenotypes, Mattarucchi et al. addressed the question of whether the observed
metabolomic changes were due to the treatment or due to the disease. They built
two models, one which sought to distinguish between children with asthma who
took controller medication and those who did not, and one which sought to distin-
guish cases from healthy controls. They determined that their controller medication
model was highly effective, with only a single misclassification in a population of
42 (Mattarucchi et al. 2012). In keeping with the findings of Reinke et al., they found
the discriminatory model was primarily driven by intermediates in the metabolism of
C21-steroid hormones urocortisone and urocortisol. Importantly, this model was
distinct from that that distinguished all 42 asthma cases from 12 age-matched
controls demonstrating that the metabolomic differences between asthma cases and
controls were not driven by consumption of controller medication (Mattarucchi et al.
2012).
Comhair et al. classified severe and non-severe asthmatics based on criteria
outlined in the proceedings of the American Thoracic Society Workshop on Refrac-
tory Asthma, which were largely based on treatment regimen (Comhair et al. 2015).
In their study all 10 severe adult asthmatics received a high dose of inhaled or oral
corticosteroids either singly or in combination with long-acting beta-agonists, while
the 10 non-severe asthmatics did not receive any corticosteroids or long-acting beta-
agonists and received inhaled beta-agonist (rescue medication) infrequently but less
than 2 times per week. A comparison of the plasma metabolomic profile of the severe
and non-severe asthma cases identified 18 significant metabolites, all but one of
which were lower in the severe asthmatics. Most of these metabolites were again
related to steroid metabolism including 1-steraroyylglycerol,
dehydroisoandrosterone sulfate, epi-androsterone sulfate, and androsterone sulfate.
These findings are consistent with the suppression of adrenal steroids as a conse-
quence of therapeutic use of corticosteroids as mentioned previously. They also
observed differences in several amino acids, including beta-alanine, between the two
groups of asthmatics, but further observed that the severe group shared many of the
same metabolomic signatures as non-severe asthmatics, who were not on
corticosteroids. The authors interpreted these findings as further evidence that
corticosteroid therapy is not the driver of differences between asthma cases and
healthy controls (Comhair et al. 2015). This hypothesis was further supported by
Dallinga et al. also who showed that the discrimination between asthma cases and
controls based on an analysis of volatile organic compound in breath samples was
not driven by medication usage (Dallinga et al. 2010). They predicated this conclu-
sion on the observation that the metabolomic products of known asthma medications
were not reflected in the principal components that distinguished between asthma
cases and controls.
Park et al. took a somewhat different approach, focusing on the metabolomic
differences between those who respond to corticosteroids as compared to those who
do not (Park et al. 2017). They performed high resolution mass spectrometry based
Pharmacometabolomics of Asthma as a Road Map to Precision Medicine 261
urinary metabolomic profiling of children with severe asthma; 15 children who did
not responded to corticosteroids, defined by persistently poor asthma control with
symptoms more than twice weekly and a less than 15% improvement in FEV1
2 weeks after systemic triamcinolone administration, and 15 children who did
respond. They identified 30 metabolomic features that differed significantly between
the two groups, representing differences in the metabolism of tyrosine, glutathione,
and the degradation of aromatic compounds. Of these five, 3,6-dihydronicotinic acid
3-methoxy-4-hydroxyphenyl(ethylene)glycol, 3,4-dihydroxy-phenylalanine,
γ-glutamylcysteine, Cys-Gly, and Flavin mononucleotide were determined to con-
tribute most to the distinction of corticosteroid responders and non-responders (Park
et al. 2017). These results point to the reduced synthesis and increased degradation
of the antioxidant glutathione, which in its thiol-reduced form (GSH) is the most
abundant antioxidant in the airway epithelial lining fluid and plays an important role
in the pathogenesis of asthma (Papamichael et al. 2021). The significant metabolites
also included constituents of cigarette smoke, which were higher in the urine of
corticosteroid-resistant children. This is consistent with evidence that smoking can
impair the efficacy of corticosteroid treatment in asthma (Chaudhuri et al. 2003).
Fitzpatrick et al. compared the plasma metabolome of mild to moderate childhood
asthmatics treated with ICS or ICS/ long-acting beta-agonists (LABA) combination
therapy to that of severe asthmatics treated with high-dose ICS and LABA
(Fitzpatrick et al. 2014). Interestingly, among the severe asthmatics they determined
no children displayed complete corticosteroid responsiveness (defined as symptoms,
lung function, bronchodilator reversibility, and exhaled nitric oxide values
normalized 2 weeks after treatment with systemic triamcinolone acetonide).
Among the severe asthmatics 89% demonstrated partial responsiveness, while the
remainder had no discernable response, taken together these children were consid-
ered a corticosteroid refractory population. Their results indicated that the cortico-
steroid refractory children demonstrated a vast array of metabolic derangements
relative to mild/moderate asthmatics. These differences again pointed to increased
lipid peroxidation and dysregulation in thiol redox balance and oxidative stress
related pathways, with dysregulation of the glycine, serine, and threonine metabo-
lism pathway and the N-acylethanolamine and N-acyltransferase pathways
observed. The results may support the hypothesis that oxidative stress is a contribu-
tory factor to corticosteroid refractory severe asthma in children. However, like
many of the previous studies the authors determined that confounding by indication
could not be ruled out, as individuals taking asthma medication for severe diseases
are also more likely to have co-morbid conditions including obesity sinus disease
and obstructive sleep apnea (Fitzpatrick et al. 2014).
lung function related differences between their endotypes were robust to additional
adjustment for medication use, suggesting medication is not driving the clusters.
However, further work incorporating longitudinal data is necessary to explore this
further.
7 Prevention
A growing body of literature is focused on the prevention of asthma. Given the early
life origins of this condition in most asthmatics much of this work is concentrated on
maternal or prenatal interventions (Gern et al. 1999). These have largely been in the
form of nutritional supplementations, such as vitamins D, C, and E (Litonjua et al.
2020), fish oil (Bisgaard et al. 2016), folic acid (Veeranki et al. 2015), pre and
probiotics (Cuello-Garcia et al. 2016), and antioxidants (Gref et al. 2017). However,
the results have been somewhat contradictory in terms of the efficacy of these
interventions in preventing asthma (Gur et al. 2017). Here again,
pharmacometabolomics, which in this instance can be expanded to include the use
of metabolomics for understanding differing responses to the same nutritional
intervention, can be leveraged to try and understand the mechanisms of success, or
otherwise.
To date, there has been little work on preventative pharmacometabolomics of
asthma. However, the existing literature does show that preventative measures
administered to mothers can influence their offspring’s metabolome (Blighe et al.
2017; Rago et al. 2019). For example, increased exposure to vitamin D in utero via
supplementation has been found to be associated with lower concentrations of fatty
acids, in particular linoleate, linolenate (18:3n–3/3n–6), myristate, oleate, palmitate,
palmitoleate (16:1n–7), and stearate (18:0) in infants (Blighe et al. 2017). Similarly,
prenatal supplementation with fish oil has been shown to result in lower levels of the
n-6 LCPUFA pathway-related metabolites and saturated and monounsaturated long-
chain fatty acids-containing compounds, lower levels of metabolites of the trypto-
phan pathway, and higher levels of metabolites in the tyrosine and glutamic acid
pathway in early life (Rago et al. 2019). It has also been demonstrated that vitamin D
and fish oil interventions can reduce the risk of asthma among offspring (Bisgaard
et al. 2016; Wolsk et al. 2017). Given it is known that many of these altered
metabolites play a direct role in the biological pathways and processes that underlie
the development and progression of asthma, such as oxidative stress and inflamma-
tion, this leads to the hypothesis that an altered offspring metabolomic milieu
resulting from a prenatal intervention may be mediating the association between
that intervention and asthma risk. Indeed, a prenatal fish oil supplementation related
metabolic profile at age 6 months was significantly associated with a reduced risk of
asthma by age 5 and this profile could explain 24% of the observed asthma-
protective effect of this supplementation (Rago et al. 2019). Similarly, the
metabolomic changes in children accompanying vitamin D supplementation of
their mothers have been linked to the reduced risk of asthma (Blighe et al. 2017).
However, these relationships can be complicated by the genetic profile of both the
mother and child. Sphingolipids, which among their multitude of roles act as
signaling molecules involved in immune response, inflammation, have been
suggested to mediate the relationship between maternal vitamin D supplementation
and child asthma risk (Kelly et al. 2019a). Vitamin D metabolites have been shown
to be capable of activating the sphingolipid pathway and decreased synthesis of
sphingolipids has been associated with increased airway hyperresponsiveness and
Pharmacometabolomics of Asthma as a Road Map to Precision Medicine 265
inflammation (Kelly et al. 2019a). Work within the VDAART (Vitamin D Antenatal
Asthma Reduction Trial) prenatal supplementation trial seemed to confirm this
relationship with one important caveat. There was no increase in child sphingolipid
levels and therefore no protective effect for asthma among children with a key
variant in a functional SNP in the region of ORMLD3, which regulates a
rate-limiting step in the de novo synthesis of sphingolipids (Kelly et al. 2019a).
Correspondingly, vitamin D supplementation conferred no protective effect for the
children of mothers with this same variant (Knihtilä et al. 2021).
Further studies are required to explore the role that pharmacometabolomics may
be able to play in the prevention of asthma.
9 Conclusions
References
Augstein J, Farmer JB, Lee TB et al (1973) Selective inhibitor of slow reacting substance of
anaphylaxis. Nat New Biol 245:215–217. https://doi.org/10.1038/newbio245215a0
Ban G-Y, Cho K, Kim S-H et al (2017) Metabolomic analysis identifies potential diagnostic
biomarkers for aspirin-exacerbated respiratory disease. Clin Exp Allergy 47:37–47. https://
doi.org/10.1111/cea.12797
Barnes PJ (2006) Corticosteroid effects on cell signalling. Eur Respir J 27:413–426. https://doi.org/
10.1183/09031936.06.00125404
Pharmacometabolomics of Asthma as a Road Map to Precision Medicine 269
Bigler J, Boedigheimer M, Schofield JPR et al (2017) A severe asthma disease signature from gene
expression profiling of peripheral blood from U-BIOPRED cohorts. Am J Respir Crit Care Med
195:1311–1320. https://doi.org/10.1164/rccm.201604-0866OC
Bisgaard H (2001) Pathophysiology of the cysteinyl leukotrienes and effects of leukotriene receptor
antagonists in asthma. Allergy 56(Suppl 66):7–11. https://doi.org/10.1034/j.1398-9995.56.s66.
2.x
Bisgaard H, Stokholm J, Chawes BL et al (2016) Fish oil-derived fatty acids in pregnancy and
wheeze and asthma in offspring. N Engl J Med 375:2530–2539. https://doi.org/10.1056/
NEJMoa1503734
Blighe K, Chawes BL, Kelly RS et al (2017) Vitamin D prenatal programming of childhood
metabolomics profiles at age 3 y. Am J Clin Nutr 106:1092–1099. https://doi.org/10.3945/
ajcn.117.158220
Bochenek G, Stachura T, Szafraniec K et al (2018) Diagnostic accuracy of urinary LTE4 measure-
ment to predict aspirin-exacerbated respiratory disease in patients with asthma. J Allergy Clin
Immunol Pract 6:528–535. https://doi.org/10.1016/j.jaip.2017.07.001
Bowler RP, Wendt CH, Fessler MB et al (2017) New strategies and challenges in lung proteomics
and metabolomics. An official American Thoracic Society workshop report. Ann Am Thorac
Soc 14:1721–1743. https://doi.org/10.1513/AnnalsATS.201710-770WS
Bozelli JC, Azher S, Epand RM (2021) Plasmalogens and chronic inflammatory diseases. Front
Physiol 12. https://doi.org/10.3389/fphys.2021.730829
Brinkman P, Wagener AH, Hekking P-P et al (2019) Identification and prospective stability of
electronic nose (eNose)-derived inflammatory phenotypes in patients with severe asthma. J
Allergy Clin Immunol 143:1811–1820.e7. https://doi.org/10.1016/j.jaci.2018.10.058
Bruno F, Spaziano G, Liparulo A et al (2018) Recent advances in the search for novel
5-lipoxygenase inhibitors for the treatment of asthma. Eur J Med Chem 153:65–72. https://
doi.org/10.1016/j.ejmech.2017.10.020
Carraro S, Giordano G, Reniero F et al (2013) Asthma severity in childhood and metabolomic
profiling of breath condensate. Allergy 68:110–117. https://doi.org/10.1111/all.12063
Carstairs JR, Nimmo AJ, Barnes PJ (1985) Autoradiographic visualization of beta-adrenoceptor
subtypes in human lung. Am Rev Respir Dis 132:541–547. https://doi.org/10.1164/arrd.1985.
132.3.541
Cazzola M, Page CP, Calzetta L, Matera MG (2012) Pharmacology and therapeutics of
bronchodilators. Pharmacol Rev 64:450–504. https://doi.org/10.1124/pr.111.004580
Chaudhuri R, Livingston E, McMahon AD et al (2003) Cigarette smoking impairs the therapeutic
response to oral corticosteroids in chronic asthma. Am J Respir Crit Care Med 168:1308–1311.
https://doi.org/10.1164/rccm.200304-503OC
Chung KF, Wenzel SE, Brozek JL et al (2014) International ERS/ATS guidelines on definition,
evaluation and treatment of severe asthma. Eur Respir J 43:343–373. https://doi.org/10.1183/
09031936.00202013
Clish CB (2015) Metabolomics: an emerging but powerful tool for precision medicine. Cold Spring
Harb Mol Case Stud 1:a000588. https://doi.org/10.1101/mcs.a000588
Comhair SAA, McDunn J, Bennett C et al (2015) Metabolomic endotype of asthma. J Immunol
195:643–650. https://doi.org/10.4049/jimmunol.1500736
Comhair SAA, Bochenek G, Baicker-McKee S et al (2018) The utility of biomarkers in diagnosis of
aspirin exacerbated respiratory disease. Respir Res 19:210. https://doi.org/10.1186/s12931-018-
0909-6
Cuello-Garcia CA, Fiocchi A, Pawankar R et al (2016) World allergy organization-McMaster
University guidelines for allergic disease prevention (GLAD-P): prebiotics. World Allergy
Organ J 9:10. https://doi.org/10.1186/s40413-016-0102-7
Dallinga JW, Robroeks CMHHT, van Berkel JJBN et al (2010) Volatile organic compounds in
exhaled breath as a diagnostic tool for asthma in children. Clin Exp Allergy 40:68–76. https://
doi.org/10.1111/j.1365-2222.2009.03343.x
270 R. S. Kelly et al.
Darveaux J, Busse WW (2015) Biologics in asthma – the next step toward personalized treatment. J
Allergy Clin Immunol Pract 3:152–160.; quiz 161. https://doi.org/10.1016/j.jaip.2014.09.014
Divekar R, Hagan J, Rank M et al (2016) Diagnostic utility of urinary LTE4 in asthma, allergic
rhinitis, chronic rhinosinusitis, nasal polyps, and aspirin sensitivity. J Allergy Clin Immunol
Pract 4:665–670. https://doi.org/10.1016/j.jaip.2016.03.004
Dona AC, Jiménez B, Schäfer H et al (2014) Precision high-throughput proton NMR spectroscopy
of human urine, serum, and plasma for large-scale metabolic phenotyping. Anal Chem 86:9887–
9894. https://doi.org/10.1021/ac5025039
Eder W, Ege MJ, von Mutius E (2006) The asthma epidemic. N Engl J Med 355:2226–2235.
https://doi.org/10.1056/NEJMra054308
Enilari O, Sinha S (2019) The global impact of asthma in adult populations. Ann Glob Health 85.
https://doi.org/10.5334/aogh.2412
Ferraro VA, Carraro S, Pirillo P et al (2020) Breathomics in asthmatic children treated with inhaled
corticosteroids. Metabolites 10. https://doi.org/10.3390/metabo10100390
Fiehn O (2002) Metabolomics – the link between genotypes and phenotypes. Plant Mol Biol 48:
155–171
Fiehn O, Kim J (2014) Metabolomics insights into pathophysiological mechanisms of interstitial
cystitis. Int Neurourol J 18:106–114. https://doi.org/10.5213/inj.2014.18.3.106
Fitzpatrick AM, Park Y, Brown LAS, Jones DP (2014) Children with severe asthma have unique
oxidative stress-associated metabolomic profiles. J Allergy Clin Immunol 133(258–61):e1–e8.
https://doi.org/10.1016/j.jaci.2013.10.012
Foster A, Chan CC (1991) Peptide leukotriene involvement in pulmonary eosinophil migration
upon antigen challenge in the actively sensitized guinea pig. Int Arch Allergy Appl Immunol 96:
279–284. https://doi.org/10.1159/000235508
Franklin S, Vondriska TM (2011) Genomes, proteomes, and the central dogma. Circ Cardiovasc
Genet 4:576. https://doi.org/10.1161/CIRCGENETICS.110.957795
GBD 2015 Chronic Respiratory Disease Collaborators (2017) Global, regional, and national deaths,
prevalence, disability-adjusted life years, and years lived with disability for chronic obstructive
pulmonary disease and asthma, 1990-2015: a systematic analysis for the Global Burden of
Disease Study 2015. Lancet Respir Med 5:691–706. https://doi.org/10.1016/S2213-2600(17)
30293-X
Gern JE, Lemanske RF, Busse WW (1999) Early life origins of asthma. J Clin Invest 104:837–843.
https://doi.org/10.1172/JCI8272
Gref A, Rautiainen S, Gruzieva O et al (2017) Dietary total antioxidant capacity in early school age
and subsequent allergic disease. Clin Exp Allergy 47:751–759. https://doi.org/10.1111/cea.
12911
Grennan D, Wang S (2019) Steroid side effects. JAMA 322:282–282. https://doi.org/10.1001/
JAMA.2019.8506
Gur M, Hakim F, Bentur L (2017) Better understanding of childhood asthma, towards primary
prevention – are we there yet? Consideration of pertinent literature. F1000Res 6(2152):
10.12688/f1000research.11601.1
Gurnell M, Heaney LG, Price D, Menzies-Gow A (2021) Long-term corticosteroid use, adrenal
insufficiency and the need for steroid-sparing treatment in adult severe asthma. J Intern Med
290:240–256. https://doi.org/10.1111/joim.13273
Haque R, White AA, Jackson DJ, Hopkins C (2021) Clinical evaluation and diagnosis of aspirin-
exacerbated respiratory disease. J Allergy Clin Immunol 148:283–291. https://doi.org/10.1016/
j.jaci.2021.06.018
Hargreave FE, Nair P (2009) The definition and diagnosis of asthma. Clin Exp Allergy 39:1652–
1658. https://doi.org/10.1111/j.1365-2222.2009.03321.x
Hastie AT, Moore WC, Meyers DA et al (2010) Analyses of asthma severity phenotypes and
inflammatory proteins in subjects stratified by sputum granulocytes. J Allergy Clin Immunol
125:1028–1036.e13. https://doi.org/10.1016/j.jaci.2010.02.008
Pharmacometabolomics of Asthma as a Road Map to Precision Medicine 271
Henderson WR, Lewis DB, Albert RK et al (1996) The importance of leukotrienes in airway
inflammation in a mouse model of asthma. J Exp Med 184:1483–1494. https://doi.org/10.1084/
jem.184.4.1483
Hunt JF, Erwin E, Palmer L et al (2002) Expression and activity of pH-regulatory glutaminase in the
human airway epithelium. Am J Respir Crit Care Med 165:101–107. https://doi.org/10.1164/
ajrccm.165.1.2104131
Jones KA, Perkins WJ, Lorenz RR et al (1999) F-actin stabilization increases tension cost during
contraction of permeabilized airway smooth muscle in dogs. J Physiol 519(Pt 2):527–538.
https://doi.org/10.1111/j.1469-7793.1999.0527m.x
Kachroo P, Sordillo JE, Lutz SM et al (2021) Pharmaco-metabolomics of inhaled corticosteroid
response in individuals with asthma. J Pers Med 11:1148. https://doi.org/10.3390/jpm11111148
Kachroo P, Stewart I, Kelly R et al (2022) The systematic use of metabolomic epidemiology,
biobanks, and electronic medical records for precision medicine initiatives in asthma: findings
suggest new guidelines to optimize treatment. https://doi.org/10.21203/rs.3.rs-268507/v1
Kaddurah-Daouk R, Weinshilboum RM (2014) Pharmacometabolomics: implications for clinical
pharmacology and systems pharmacology. Clin Pharmacol Therap 95:154–167. https://doi.org/
10.1038/clpt.2013.217
Kannisto S, Korppi M, Remes K, Voutilainen R (2001) Serum dehydroepiandrosterone sulfate
concentration as an indicator of adrenocortical suppression in asthmatic children treated with
inhaled steroids. J Clin Endocrinol Metab 86:4908–4912. https://doi.org/10.1210/jcem.86.10.
7975
Kannisto S, Laatikainen A, Taivainen A et al (2004) Serum dehydroepiandrosterone sulfate
concentration as an indicator of adrenocortical suppression during inhaled steroid therapy in
adult asthmatic patients. Eur J Endocrinol 150:687–690. https://doi.org/10.1530/eje.0.1500687
Kelly RS, Dahlin A, McGeachie MJ et al (2017) Asthma metabolomics and the potential for
integrative omics in research and the clinic. Chest 151:262–277. https://doi.org/10.1016/j.
chest.2016.10.008
Kelly RS, Chawes BL, Guo F et al (2019a) The role of the 17q21 genotype in the prevention of
early childhood asthma and recurrent wheeze by vitamin D. Eur Respir J 54. https://doi.org/10.
1183/13993003.00761-2019
Kelly RS, Sordillo JE, Lutz SM et al (2019b) Pharmacometabolomics of bronchodilator response in
asthma and the role of age-metabolite interactions. Metabolites 9. https://doi.org/10.3390/
metabo9090179
Kelly RS, Mendez KM, Huang M et al (2021) Metabo-endotypes of asthma reveal differences in
lung function: discovery and validation in two TOPMed cohorts. Am J Respir Crit Care Med.
https://doi.org/10.1164/rccm.202105-1268OC
Kersten ETG, Koppelman GH (2017) Pharmacogenetics of asthma: toward precision medicine.
Curr Opin Pulm Med 23:12–20. https://doi.org/10.1097/MCP.0000000000000335
Knihtilä HM, Kelly RS, Brustad N et al (2021) Maternal 17q21 genotype influences prenatal
vitamin D effects on offspring asthma/recurrent wheeze. Eur Respir J 58. https://doi.org/10.
1183/13993003.02012-2020
Kuruvilla ME, Lee FE-H, Lee GB (2019) Understanding asthma phenotypes, endotypes, and
mechanisms of disease. Clin Rev Allergy Immunol 56:219–233. https://doi.org/10.1007/
s12016-018-8712-1
Larsson K, Kankaanranta H, Janson C et al (2020) Bringing asthma care into the twenty-first
century. NPJ Prim Care Respir Med 30:25. https://doi.org/10.1038/s41533-020-0182-2
Lewis LM, Ferguson I, House SL et al (2014) Albuterol administration is commonly associated
with increases in serum lactate in patients with asthma treated for acute exacerbation of asthma.
Chest 145:53–59. https://doi.org/10.1378/chest.13-0930
Litonjua AA, Carey VJ, Laranjo N et al (2020) Six-year follow-up of a trial of antenatal vitamin D
for asthma reduction. N Engl J Med 382:525–533. https://doi.org/10.1056/NEJMoa1906137
Loureiro CC, Duarte IF, Gomes J et al (2014) Urinary metabolomic changes as a predictive
biomarker of asthma exacerbation. J Allergy Clin Immunol 133:261-3.e1–261-3.e5. https://
doi.org/10.1016/j.jaci.2013.11.004
272 R. S. Kelly et al.
Loza MJ, Djukanovic R, Chung KF et al (2016) Validated and longitudinally stable asthma
phenotypes based on cluster analysis of the ADEPT study. Respir Res 17:165. https://doi.org/
10.1186/s12931-016-0482-9
Matera MG, Page CP, Calzetta L et al (2020) Pharmacology and therapeutics of bronchodilators
revisited. Pharmacol Rev 72:218–252. https://doi.org/10.1124/pr.119.018150
Mattarucchi E, Baraldi E, Guillou C (2012) Metabolomics applied to urine samples in childhood
asthma; differentiation between asthma phenotypes and identification of relevant metabolites.
Biomed Chromatogr 26:89–94. https://doi.org/10.1002/bmc.1631
McGeachie MJ, Dahlin A, Qiu W et al (2015) The metabolomics of asthma control: a promising
link between genetics and disease. Immun Inflamm Dis 3:224–238. https://doi.org/10.1002/
iid3.61
Moore WC, Meyers DA, Wenzel SE et al (2010) Identification of asthma phenotypes using cluster
analysis in the severe asthma research program. Am J Respir Crit Care Med 181:315–323.
https://doi.org/10.1164/rccm.200906-0896OC
Mulvihill MM, Nomura DK (2014) Metabolomic strategies to map functions of metabolic
pathways. Am J Physiol Endocrinol Metab 307:E237–E244. https://doi.org/10.1152/ajpendo.
00228.2014
Newton R, Giembycz MA (2016) Understanding how long-acting β2 -adrenoceptor agonists
enhance the clinical efficacy of inhaled corticosteroids in asthma – an update. Br J Pharmacol
173:3405–3430. https://doi.org/10.1111/bph.13628
Pandya D, Puttanna A, Balagopal V (2014) Systemic effects of inhaled corticosteroids: an over-
view. Open Respir Med J 8:59–65. https://doi.org/10.2174/1874306401408010059
Papamichael MM, Katsardis C, Sarandi E et al (2021) Application of metabolomics in pediatric
asthma: prediction, diagnosis and personalized treatment. Metabolites 11. https://doi.org/10.
3390/metabo11040251
Paredi P, Kharitonov SA, Barnes PJ (2000) Elevation of exhaled ethane concentration in asthma.
Am J Respir Crit Care Med 162:1450–1454. https://doi.org/10.1164/ajrccm.162.4.2003064
Park YH, Fitzpatrick AM, Medriano CA, Jones DP (2017) High-resolution metabolomics to
identify urine biomarkers in corticosteroid-resistant asthmatic children. J Allergy Clin Immunol
139:1518–1524.e4. https://doi.org/10.1016/j.jaci.2016.08.018
Peluso A, Glen R, Ebbels TMD (2021) Multiple-testing correction in metabolome-wide association
studies. BMC Bioinform 22:67. https://doi.org/10.1186/s12859-021-03975-2
Peters-Golden M, Henderson WR (2007) Leukotrienes. N Engl J Med 357:1841–1854. https://doi.
org/10.1056/NEJMra071371
Quan-Jun Y, Jian-Ping Z, Jian-Hua Z et al (2017) Distinct metabolic profile of inhaled budesonide
and salbutamol in asthmatic children during acute exacerbation. Basic Clin Pharmacol Toxicol
120:303–311. https://doi.org/10.1111/bcpt.12686
Rago D, Rasmussen MA, Lee-Sarwar KA et al (2019) Fish-oil supplementation in pregnancy, child
metabolomics and asthma risk. EBioMedicine 46:399–410. https://doi.org/10.1016/j.ebiom.
2019.07.057
Reddel HK, Bacharier LB, Bateman ED et al (2021) Global initiative for asthma (GINA) strategy
2021 – executive summary and rationale for key changes. J Allergy Clin Immunol Pract. https://
doi.org/10.1016/j.jaip.2021.10.001
Reinke SN, Gallart-Ayala H, Gómez C et al (2017) Metabolomics analysis identifies different
metabotypes of asthma severity. Eur Respir J 49. https://doi.org/10.1183/13993003.01740-2016
Rodrigo GJ (2014) Serum lactate increase during acute asthma treatment: a new piece of the puzzle.
Chest 145:6–7. https://doi.org/10.1378/chest.13-2042
Rodrigo GJ, Rodrigo C (2005) Elevated plasma lactate level associated with high dose inhaled
albuterol therapy in acute severe asthma. Emerg Med J 22:404–408. https://doi.org/10.1136/
emj.2003.012039
Ruffin RE, McIntyre E, Crockett AJ et al (1982) Combination bronchodilator therapy in asthma. J
Allergy Clin Immunol 69:60–65. https://doi.org/10.1016/0091-6749(82)90089-6
Pharmacometabolomics of Asthma as a Road Map to Precision Medicine 273
Sahiner UM, Birben E, Erzurum S et al (2011) Oxidative stress in asthma. World Allergy Organ J 4:
151–158. https://doi.org/10.1097/WOX.0b013e318232389e
Sinha A, Krishnan V, Sethi T et al (2012) Metabolomic signatures in nuclear magnetic resonance
spectra of exhaled breath condensate identify asthma. Eur Respir J 39:500–502. https://doi.org/
10.1183/09031936.00047711
Sinha A, Desiraju K, Aggarwal K et al (2017) Exhaled breath condensate metabolome clusters for
endotype discovery in asthma. J Transl Med 15:262. https://doi.org/10.1186/s12967-017-
1365-7
Smith LJ, Shamsuddin M, Houston M (1993) Effect of leukotriene D4 and platelet-activating factor
on human alveolar macrophage eicosanoid and PAF synthesis. Am Rev Respir Dis 148:682–
688. https://doi.org/10.1164/ajrccm/148.3.682
Sordillo JE, Lutz SM, Kelly RS et al (2020) Plasmalogens mediate the effect of age on bronchodi-
lator response in individuals with asthma. Front Med (Lausanne) 7:38. https://doi.org/10.3389/
fmed.2020.00038
Svenningsen S, Nair P (2017) Asthma Endotypes and an overview of targeted therapy for asthma.
Front Med (Lausanne) 4:158. https://doi.org/10.3389/fmed.2017.00158
Tyler SR, Bunyavanich S (2019) Leveraging -omics for asthma endotyping. J Allergy Clin
Immunol 144:13–23. https://doi.org/10.1016/j.jaci.2019.05.015
Veeranki SP, Gebretsadik T, Mitchel EF et al (2015) Maternal folic acid supplementation during
pregnancy and early childhood asthma. Epidemiology 26:934–941. https://doi.org/10.1097/
EDE.0000000000000380
Walter MA, Linsley PS, Cox DW (1987) Apa I polymorphism of a human immunoglobin VH3
subclass locus. Nucleic Acids Res 15:4697. https://doi.org/10.1093/nar/15.11.4697
Wang et al (2021) Research progress of metabolomics in asthma. Metabolites. PMID: 34564383
Wedes SH, Wu W, Comhair SAA et al (2011) Urinary bromotyrosine measures asthma control and
predicts asthma exacerbations in children. J Pediatr 159:248–55.e1. https://doi.org/10.1016/j.
jpeds.2011.01.029
Wei R, Wang J, Su M et al (2018) Missing value imputation approach for mass spectrometry-based
metabolomics data. Sci Rep 8:663. https://doi.org/10.1038/s41598-017-19120-0
Wendell SG, Baffi C, Holguin F (2014) Fatty acids, inflammation, and asthma. J Allergy Clin
Immunol 133:1255–1264. https://doi.org/10.1016/j.jaci.2013.12.1087
Wendell SG, Fan H, Zhang C (2020) G protein-coupled receptors in asthma therapy: pharmacology
and drug action. Pharmacol Rev 72:1–49. https://doi.org/10.1124/pr.118.016899
Wenzel SE (2012) Asthma phenotypes: the evolution from clinical to molecular approaches. Nat
Med 18:716–725. https://doi.org/10.1038/nm.2678
Werner M, Jordan PM, Romp E et al (2019) Targeting biosynthetic networks of the
proinflammatory and proresolving lipid metabolome. FASEB J 33:6140–6153. https://doi.org/
10.1096/fj.201802509R
Wishart DS (2019) Metabolomics for investigating physiological and pathophysiological processes.
Physiol Rev 99:1819–1875. https://doi.org/10.1152/physrev.00035.2018
Wolsk HM, Chawes BL, Litonjua AA et al (2017) Prenatal vitamin D supplementation reduces risk
of asthma/recurrent wheeze in early childhood: a combined analysis of two randomized
controlled trials. PLoS One 12:e0186657. https://doi.org/10.1371/journal.pone.0186657
Yaghoubi M, Adibi A, Safari A et al (2019) The projected economic and health burden of
uncontrolled asthma in the United States. Am J Respir Crit Care Med 200:1102–1112. https://
doi.org/10.1164/rccm.201901-0016OC
Yokomizo T, Nakamura M, Shimizu T (2018) Leukotriene receptors as potential therapeutic
targets. J Clin Invest 128:2691–2701. https://doi.org/10.1172/JCI97946
Zhu Z, Camargo CA, Hasegawa K (2019) Metabolomics in the prevention and management of
asthma. Expert Rev Respir Med 13:1135–1138. https://doi.org/10.1080/17476348.2019.
1674650
Prospective Metabolomic Studies
in Precision Medicine: The AKRIBEA Project
Contents
1 Metabolomics as a Tool for Precision Medicine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 276
1.1 Precision Medicine and Molecular Medicine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 276
1.2 Biomarkers and Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 277
1.3 Description of the Accessible NMR Metabolic Landscape . . . . . . . . . . . . . . . . . . . . . . . . . . 278
1.4 On the Cohort Size for Applications in Precision Medicine . . . . . . . . . . . . . . . . . . . . . . . . . 282
2 Large-Cohort Sample Collection and Biobanking for Metabolomic Studies . . . . . . . . . . . . . . 283
3 Standard Operating Procedures for Metabolomics in Precision Medicine . . . . . . . . . . . . . . . . . 285
4 Artificial Intelligence and Data Mining: Data Is the Way . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286
4.1 Data Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286
4.2 Data Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288
4.3 Biological Contextualization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289
4.4 From the Laboratory to the Clinic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289
5 Applications of Precision Medicine: Non-oriented and Oriented Metabolomics
Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290
5.1 Non-oriented Metabolomics Study: Metabolic Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . 290
5.2 Oriented Metabolomics Study: COVID-19 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293
6 Concluding Remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295
Abstract
For a long time, conventional medicine has analysed biomolecules to diagnose
diseases. Yet, this approach has proven valid only for a limited number of
metabolites and often through a bijective relationship with the disease
# The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 275
V. Ghini et al. (eds.), Metabolomics and Its Impact on Health and Diseases,
Handbook of Experimental Pharmacology 277, https://doi.org/10.1007/164_2022_610
276 M. Bizkarguenaga et al.
Keywords
Biomarker · NMR metabolomics · Precision medicine
A silent revolution is steadily transforming traditional medicine into the novel field
of personalized medicine, which tailors medical treatment to the individual
characteristics of each patient (Kohler et al. 2017). Currently, medical practice is
mostly based in evidence, organized at the organ level and administering a rather
uniform treatment for a given disease. In contrast, precision medicine concentrates
on the mechanistic aspects of the disease, is organized at the cellular and molecular
levels, and aims to provide the most suitable personalized treatment for each patient.
As a consequence of this innovative strategy, personalized medicine focuses on early
detection and prospective evaluation, while canonical medical praxis is based on
statistically based retrospective analyses to provide curative and palliative care.
To achieve this goal, the concept of precision medicine, first used in 2009 in the
book The Innovator’s Prescription by Clayton Christensen (Christensen et al. 2009),
exploits the analysis of clinical and molecular data of patient subpopulations, which
tries to classify a subject into a healthy individual or a patient by contrasting
biological information with datasets from large population cohorts, enriched in
biological diversity. This strategy allows to endotype a particular disease, ultimately
leading to personalized care (Śliwczyński and Orlewska 2016). Furthermore, the
generation of prediction models for prevention and diagnosis paves the way for
application of precision medicine in a rationally designed drug treatment for geneti-
cally or metabolically stratified groups of patients (Authors 2021). In fact, this
strategy not only tailors the appropriate drug treatment for each patient (Tranvåg
Prospective Metabolomic Studies in Precision Medicine: The AKRIBEA Project 277
et al. 2021) but also provides an explanation for the non-responding population in
clinical phase trials of drugs, where patient sub-classification also actively
contributes to the better understanding of the mode-of-action of drugs (John et al.
2020; Puchades-Carrasco and Pineda-Lucena 2017).
As already mentioned, traditional medicine is rooted on medical evidence (mea-
surable symptomatology), from which only a limited fraction of it arises from
biochemical analyses (i.e. molecular medicine): glucose, cholesterol, high-density
lipoprotein/low-density lipoprotein, transaminases, or creatinine, among others, to
monitor diabetes, cardiovascular, liver, and renal disorders, respectively. Perhaps,
the most powerful diagnostic use of metabolites currently occurs in the heel test to
screen for neonatal congenital methabolopathies (Beger et al. 2016). In contrast,
precision medicine heavily relies on genomics, proteomics, and metabolomics for an
appropriate biochemical characterization of the individual. To that end, it integrates
molecular information originated by all the abovementioned “omic” techniques with
the medical records, environmental surroundings, diet, and lifestyle of the popula-
tion under consideration. Altogether, this approach can provide predictive, diagnos-
tic, and prognostic markers for a plethora of diseases, informing on their underlying
molecular mechanisms and enabling their sub-classification and the stratification of
patients based on the metabolic pathways involved (Śliwczyński and Orlewska
2016; Clish 2015).
proximal to the organic focus of the disease but, in reality, most diseases also have
significant systemic manifestations (Hartl et al. 2021). In this context, serum,
plasma, and urine are the most suitable biofluids, characterized by optimal sensitivity
to the time-dependent metabolome, disponible nature, and easy storage. Plasma and
serum metabolomes undergo limited daily variation, also providing information of
the extracellular metabolome, whereas urine is the most on-demand available
biofluid (Kohler et al. 2017).
Nowadays, metabolomic research technology enables the identification and
quantification of hundreds of distinct metabolites from biofluids, using technologies
that can be easily extrapolated to large-scale cohorts of donors and thousands of
samples. For the sake of reproducibility, Standardized Operating Procedures (SOPs)
must be implemented for sample collection, storage, handling, analytical analyses,
and data processing (Beger et al. 2016). Under these premises, research laboratories
with standardized infrastructures will generate dataset that become additive,
enabling large-scale and multicentred studies in metabolomics. Techniques that
nowadays can fulfil these requirements include Nuclear Magnetic Resonance spec-
troscopy (NMR), Liquid Chromatography coupled to Mass Spectrometry (LC-MS),
and Gas Chromatography with Mass Spectrometry (GC-MS). In here, we will focus
on NMR spectroscopy, which is able to simultaneously quantify hundreds of
metabolites in a biological sample with an acquisition time of less than 20 min.
Fig. 1 Central metabolism pathways. NMR analysis allows the characterization of certain metabolites that are spread all over the pathways. The colour code
represents the biofluid or methodology sensitive to the detected metabolite (see legend). The scheme is based on KEGG hsa01100 map. Abbreviations: (R)-3-
Hydroxybutyric acid, 3-HB; (S)-3-Hydroxy-3-methylglutaryl-CoA, HMG-CoA; (S)-3-Methyl-2-oxopentanoic acid, KMV; 1,2-Diacylglycerol, LPC;
1-Methylnicotinamide, MNA; 2,5-Dihydroxyphenylacetic acid, 4-HPP; 2-Hydroxyphenylacetic acid, HPAA; 2-Hydroxytricarballylic acid, CA;
279
2-Oxobutanoic acid; FA; 2-Phospho-D-glycerate, 2PG; 3,4-Dihydroxy-L-phenylalanine, L-DOPA; 3',5'-Cyclic AMP, NMPc; 3',5'-Cyclic GMP, NMPc;
3-Methyl-2-oxobutanoic acid, KIV; 3-Phospho-D-glycerate, 3PG; 4-Fumarylacetoacetic acid, 4-FAcAc; 4-Maleylacetoacetate, 4-MAcAc; 4-Methyl-2-
280 M. Bizkarguenaga et al.
among others (Fig. 1). The abnormal quantity of some of these metabolites can be
associated with clinical pathologies and prevalent syndromes (Bruzzone et al. 2021).
Additionally, the urine metabolome can also be scrutinized for the absence of
metabolites, to confirm that the individual does not suffer a congenital
metabolopathy, as an alternative to the newborn screening heel test (Embade et al.
2019).
Serum is the blood moiety devoid of cell and clotting factors, rich in proteins, and
has been used in numerous diagnostic tests. Serum is metabolically very homeostatic
and over 50 components can be easily identified and quantified by NMR spectros-
copy (Fig. 1). Yet, the added value of NMR-based analysis of serum is the lipopro-
tein profiling (Raftery, “Quantitative NMR methods in metablomics”). Lipoproteins
are a group of micelles consisting of lipids and proteins whose main function is to
smaller dataset of samples from other unrelated cohorts measured under the same
SOPs. There are two main applications of this large dataset (Fig. 2): (1) integral
analysis of the dataset to characterize the cohort (i.e. environmental factors and
habits, disease biomarkers for prevalent disease such as metabolic syndrome, density
of rare diseases, big data and artificial intelligence analyses oriented to medicine and
drug discovery), and (2) to compare the reference dataset to samples from smaller
cohorts of patients from the pathology under study. Such cohorts must have been
previously validated using an independent gold-standard methodology. In this
chapter, we will use the AKRIBEA project as a case study to do an in-depth
description of the required steps needed in precision medicine studies with large
cohorts. Moreover, we will present illustrative examples for the two abovementioned
exploration strategies: metabolic syndrome and COVID-19.
Mondragón Cooperative, whose mission is to promote the safety and health of the
workers of the associated companies.
In projects involving human samples, ethical and government permits are
required before starting the recollection of biofluids. It is important to plan this
step quite in advance due to the considerable associated bureaucracy. For the
AKRIBEA project, the approval was granted by the Euskadi Drug Research Ethics
Committee (CEIm-E), an independent and multidisciplinary agency whose main
purpose is to ensure the protection of the rights, safety, and well-being of people who
participate in a biomedical research project, guaranteeing the correct application of
ethical, methodological, and legal principles. At this stage, it is important to properly
specify the intended use of the data in the informed consent and whether the data will
be used in a coded or anonymized manner. It is convenient to circulate quite in
advance a summary of the project among the potential donors. This document should
describe the goals of the project and be redacted in an understandable way (i.e. an
easy-to-read leaflet, distributed with the internal correspondence of the Corporation).
Finally, a database must be created to correctly link the samples with the metadata
(De Souza and Greenspan 2013). This is of particular importance in longitudinal
studies.
Along AKRIBEA project, samples are collected using homogeneous disposable
material and under the same conditions for all the involved patients: fasting
conditions for serum and first urine in the morning. Extracted blood is conserved
in the form of serum or plasma. Serum is richer in metabolites concentration, but at
the expense of higher variability associated to the clotting procedure. Thus,
pre-analytical SOPs (Vignoli et al. 2019) become critical as they properly define
the clotting time and temperature, among other variables. In the case of plasma
extraction, the use of anticoagulant is needed but this can lead also to the presence of
contaminants in the samples (González-Domínguez et al. 2020) and, for example,
EDTA becomes largely observable in the NMR spectra. In turn, urine samples are
centrifuged to eliminate cell residuals and maintained at low temperature (4°C) for
the shortest time possible until freezing to avoid bacterial contamination and to
guarantee the stability of the metabolites present in this biofluid.
Automatization is key to minimize variability. In this line, the German National
Cohort health study (https://www.dkfz.de/en/epidemiologie-krebserkrankungen/
units/NAKO_Studienzentrum_eng/NAKO_engl.html), that will follow the long-
term medical histories of 200,000 participants over 25–30 years, has been equipped
with pipetting robots and all the aliquots are carried out automatically. In the
AKRIBEA project, the efficiency of the personnel associated to the biobank has
been instrumental to fulfil this goal.
In principle, biobanks can collect and store all kinds of human samples for
analysis on a large scale. Samples must be collected and stored in a way that
makes them suitable for metabolomic analysis, typically frozen at -80°C. Previous
studies have demonstrated that negligible changes can be observed after long-term
stored samples (Yin et al. 2015; Loo et al. 2020). Yet, a crucial point is to ensure
dividing specimens into aliquots adjusting the final volumes and number of aliquots
from every sample to the requirements of the project. This precaution will minimize
Prospective Metabolomic Studies in Precision Medicine: The AKRIBEA Project 285
the number of freezing/thawing cycles experienced by the sample, which has drastic
adverse effects in the metabolic integrity of the biofluids (Lodge et al. 2021a).
Data analysis may include unsupervised analysis (i.e. segmentation of the database
into two or more non-previously defined subgroups which can be mutually com-
pared) or supervised analysis (the segmentation groups have been previously defined
according to metadata). The type of analysis may, respectively, help to respond to
the questions whether there are metabolic differences in given classes or whether it is
possible to identify subgroups of different metabolic profiles. Such analyses can be
performed on each variable independently (univariate) or using two or more
variables together (multivariate).
Univariate analysis involves the unidimensional analysis of a single variable,
which usually results in straightforward biological interpretation. The analysis
includes effect size (i.e. directional variation of the metabolite concentration) and
the statistical significance of the variation, normally expressed as p-value. For the
sake of comparison, effect size is normally expressed in fold-change as binary
logarithm units. In univariate analysis, it is important to take precautions to control
Type I error (i.e. the false positive rate) in multiple testing and to properly account
for the confounding factors in the analysis, when necessary.
Multivariate analysis involves the concurrent consideration of several variables.
In here, the distinction between unsupervised and supervised analyses becomes even
stronger. The unsupervised multivariate analysis is often carried out by principal
component analysis (PCA), a method for dimensionality reduction that comprises
the maximum data variability in their first components. PCA also allows the visual
representation of multidimensional data, helps to identify patterns and that can be
combined with clustering algorithms (hierarchical or not) to discover groups of
individuals with similar characteristics. In turn, multivariate supervised analyses
identify the group or a specific value for each individual/sample (dependent variable)
and fit the input (independent) variables to a model that explains/predicts the
dependent variable. In metabolomics, it is common to use linear regression models
or PLS (Partial Least Squares) because of their explanatory capacity through their
coefficients. Instead, for classification problems, PLS Discriminant Analysis
(PLS-DA) or orthogonal PLS-DA (OPLS-DA) is widely used (Verpoorte, “Natural
products drug discovery: on silica or in-silico?”; Wishart, “Practical Aspects of
NMR-Based Metabolomics”). Other machine learning methods such as support
vector machines (SVM), random forest (RF), or neural networks are also
commonly used.
In supervised techniques it is important to pay attention to method performance
evaluation and tuning. Building a model is an iterative process where parameters
shall be adjusted, and results must be evaluated. Sensitivity, specificity, accuracy,
Area Under the Receiver Operating Characteristic curve (AUROC), coefficient of
determination (R2), and Root Mean Square Error (RMSE) are among the perfor-
mance metrics that can be used alone or in combination.
Prospective Metabolomic Studies in Precision Medicine: The AKRIBEA Project 289
The final goal for prospective any metabolomic study with human cohorts is to try to
adequately contextualize the observed metabolic changes within the underlying
biochemical pathways. Biological interpretation of metabolic changes is challenging
and, consequently, largely depends on knowledge and experience. Yet, nowadays,
there are databases and tools that help to support this task. They are mostly based on
network analysis, where the main representative is metabolic pathway analysis.
These computer programs use the identified metabolites (from metabolomics or
indirectly from other omics) to pinpoint and analyse the involved pathways at
both, the topological and the enrichment level. These analyses pave the way to
formulate new hypothesis based on the experimental data interpretation, which can
be further contrasted by searching for additional information in available online
databases or by designing validation experiments.
Of course, the final goal of any putative biomarker is to end up validated and being
used by clinicians for the diagnostic and/or prognostic of diseases. In general, for
application in the clinic, the results should be validated at multiple levels, including
analytical validation and using samples from many different sources. The validation
method may involve conducting large corroboratory studies by directly testing each
candidate biomarker with an independently collected new set of samples from the
target population that will comprise a minimum of three groups (control, targeted
disease group, and targeted disease group with comorbidities). Since this study
includes significantly larger set of samples, the applied analytical techniques must
have high-throughput capacity as, for instance, NMR can provide. Finally, it is worth
noting that validated biomarkers are not directly used as direct diagnostic methods,
but they can act as diagnostic tools to round-up the diagnostic.
An important technical aspect is that for a validated biomarker the methodology
used for its determination has to fulfil the In Vitro Diagnostic (IVD) and/or the In
Vitro Medical-devices Regulation (IVDR), a new harmonized regulatory framework
to ensure the safety and performance of in vitro diagnostic medical devices on the
European market. This implementation is technically complex and often requires to
obtain several international and national certifications. Such regulatory standards
will undoubtedly modify the laboratory operativity, accounting for many aspects
such as the identification of the persons responsible for regulatory compliance, the
implementation of unique device identification, and post-market surveillance, if that
is finally the case. The implementation of this procedures often requires a risk
analysis (typically a SWOT analysis, Strengths, Weaknesses, Threats, and
Opportunities) for all processes that allows anticipating and minimizing the proba-
bility of failures and/or errors in the procedures. Moreover, the laboratory has to
comply with some regulations such as ISO 31000 Risk Management standard and
ISO 14971: Risk Management System for Medical Devices.
290 M. Bizkarguenaga et al.
Unfortunately, the elaborated procedures and the difficult access to large cohort
studies have prevented an avenue of validated biomarkers. In fact, in the past years,
150,000 studies have been conducted on biomarker, but only 100 biomarkers have
been validated and implemented for clinical use.
The project “Metabotype of the active labour population of the Basque Country
through the metabolomic analysis of serum and urine by NMR” recruited serum and
urine samples from 10,000 subjects belonging to the working population of the
Basque Country. This sample repository was scrutinized to generate a metabolomic
(and to a less extent also a genomic) profiling for this community, that could set up
the basis for studying different pathologies impacting in the metabolism. In here, we
describe the application of the metabolomic analysis to investigate the Metabolic
Syndrome (MetS).
MetS is a group of metabolic abnormalities whose existence is considered a
predominant risk factor for the development of cardiovascular diseases (Day
2007). Currently, it is of great importance due to its high prevalence rate, since
35% of the adult population and 50% of the elderly population have MetS. Although
the pathogenesis of MetS is not fully understood, it is generally diagnosed after the
existence of 3 or more risk factors in an individual, altered glucose metabolism,
obesity, dyslipidaemia, and/or hypertension (Neuhauser 2005). Yet, this definition is
not adequate and is not supported at the molecular level by validated biomarkers.
Many studies have metabolically investigated MetS or its participating risk factors
(Reddy et al. 2018; Blouin et al. 2005), including studies based on NMR
metabolomics (Monnerie et al. 2020). For instance, Würtz et al. (2012) analysed
more than 7,000 serum samples from apparently healthy young adults using high-
throughput NMR to investigate diabetes prevalence. The metabolic signatures of
insulin resistance were modulated by obesity (Würtz et al. 2012) and found different
for men and women.
In a recent study (Bruzzone et al. 2021), we have investigated MetS using
NMR-based metabolomics of urine samples. The goal of the project was to investi-
gate the natural history of the syndrome, evaluating the metabolic fingerprint at each
step of the progression between apparently healthy individuals (at least devoid of
any risk factor related to MetS) up to subjects that suffer all the MetS-associated risk
factors. Indeed, the study was designed not only to investigate the contribution of
each of the previously risk factors that can be involved in the development of MetS
independently, but also to evaluate all their possible combinations between the
presence or absence of them. Four different risk factors and their possible interme-
diate conditions were considered: obesity, dyslipidaemia, hypertension, and diabetes
and all the possible intermediate conditions (Table 1). The reference AKRIBEA
Prospective Metabolomic Studies in Precision Medicine: The AKRIBEA Project 291
Table 1 MetS conditions, definitions, and number of individuals allocated to each condition in the
metabolic syndrome study (Bruzzone et al. 2021)
Total Female Male
MetS condition N = 10,792 N = 4,351 N = 6,441
0000 Apparently healthy 6,925 2,935 3,990
(64.17%) (67.46%) (61.95%)
0001 Hypertension 692 (6.41%) 276 416
(6.34%) (6.46%)
0010 Dyslipidaemia 733 (6.79%) 120 613
(2.76%) (9.52%)
0011 Obesity 170 (1.58%) 53 (1.22%) 117
(1.82%)
0100 Diabetes 504 (4.67%) 232 272
(5.33%) (4.22%)
0101 Dyslipidaemia + hypertension 310 (2.87%) 169 141
(3.88%) (2.19%)
0110 Obesity + hypertension 170 (1.58%) 37 (0.85%) 133
(2.06%)
0111 Obesity + dyslipidaemia 148 (1.37%) 62 (1.42%) 86 (1.34%)
1000 Diabetes + hypertension 282 (2.61%) 89 (2.05%) 193
(3.00%)
1001 Diabetes + dyslipidaemia 188 (1.74%) 83 (1.91%) 105
(1.63%)
1010 Diabetes + obesity 84 (0.78%) 18 (0.41%) 66 (1.02%)
1011 Obesity + dyslipidaemia + hypertension 90 (0.83%) 32 (0.74%) 58 (0.90%)
1100 Diabetes + dyslipidaemia + 92 (0.85%) 44 (1.01%) 48 (0.75%)
hypertension
1101 Diabetes + obesity + hypertension 202 (1.87%) 111 91 (1.41%)
(2.55%)
1110 Diabetes + obesity + dyslipidaemia 62 (0.57%) 17 (0.39%) 45 (0.70%)
1111 Diabetes + obesity + dyslipidaemia + 140 (1.30%) 73 (1.68%) 67 (1.04%)
hypertension
Legend: Diabetes as fasting plasma glucose >100 mg/dL, previously diagnosed type 2, impaired
fasting glucose, impaired glucose tolerance, or insulin resistance taking medication for
hyperglycaemia; obesity as BMI > 30 kg/m2; dyslipidaemia as triglycerides >150 mg/dL HDL
Cholesterol <34.75 mg/dL in men or <38.61 in women, previously diagnosed
hypercholesterolaemia, hyperlipidaemia or hypertriglyceridaemia taking medication for
dyslipidaemia; hypertension as blood pressure ≥140/90 mmHg, previously diagnosed hypertension
taking medication for hypertension
Fig. 5 Spearman correlation distances to the healthy condition for all 4 conditions. Colours
represent the distance to the apparently healthy (0000) condition and the lines connect adjacent
conditions. MetS definition according to WHO, European Group for the Study of Insulin (EGIR),
and American Association of Clinical Endocrinology (AACE) is represented by squares and
triangles; definition from National Cholesterol Education Program-Third Adult Treatment Panel
(NCEP:ATPIII) is represented by squares, triangles, and rhombus; definition by International
Diabetes Federation (IDF) is represented by squares and rhombus
This study revealed that COVID-19 patients dysregulate ApoA-I and ApoA-II
lipoproteins, which points to an increased risk of atherosclerosis. This is in addition
to a singular profile of low-molecular-weight metabolites, which include high levels
of ketone bodies (acetoacetic acid, 3-hydroxybutyric acid, and acetone) and
2-hydroxybutyric acid, related to liver damage, dyslipidaemia, and oxidative stress
(Bruzzone et al. 2020a).
A follow-up study, in collaboration with the group of Jeremy Nicholson (ANPC,
Murdoch University, Australia), reinforces the idea that COVID-19 disease
dysregulates amino acids, biogenic amines, and tryptophan pathway, a bona fide
biomolecular marker of the disease (Kimhofer et al. 2020). Also, in the acute phase
of the disease the systemic inflammation markers appear to be elevated. This is in
line with other COVID-19 studies (Kimhofer et al. 2020; Thomas et al. 2020), which
show decreased counts of T-helper cells (regulators of T lymphocytes) and elevated
inflammatory cytokines and biomarkers such as IL-2, IL-6, IL-7, macrophage
inflammatory protein 1-a, tumour necrosis factor a, C-reactive protein, ferritin, and
D-dimer.
The natural history of COVID-19 disease reveals that some people infected by
SARS-CoV-2 do not return to a normal/healthy state, establishing a new deteriorated
health-baseline (Ayres 2020). Long-term COVID-19 or post-acute COVID-19 syn-
drome patients may retain some symptoms and/or have developed unprecedented
adverse effects (Holmes et al. 2021; Nalbandian et al. 2021; Yong 2021). Known
manifestations include a wide variety of physical symptoms, such as severe fatigue,
malaise, headaches, and other more worrisome phenomena such as increased risk of
damage to the heart, lungs, and brain (López-León et al. 2021; Davis et al. 2021). It
is estimated that around 1 out of 10 COVID-19 patients will undergo symptoms up
to 12 weeks after having suffered the disease, according to a new report from the
European Observatory of Health Systems and Policies promoted by the World
Health Organization (WHO).
An important open question is whether post-acute sequela of COVID-19 are
accompanied by incomplete metabolic phenoreversion. Metabolic alterations on a
cohort of 27 recovered, non-hospitalized patients have been recently analysed using
a combination of LC-MS spectrometry and NMR spectroscopy to show that a subset
of these patients still display a biochemical pathophysiology (Holmes et al. 2021).
Consistently, our lab studied a cohort of 97 post-COVID-19 recovered patients,
6 months after the infection on average, and compared them to a matched reference
set (n = 87), created ad hoc from the AKRIBEA cohort. Our results show a bimodal
distribution, with half the patient's population still experiencing a significant meta-
bolic and lipidomic dysregulation after 6 months of the infection. Finally, a recent
study analysed blood samples of PCR-/controls, PCR+/not-hospitalized, PCR+/
hospitalized, and PCR+/intubated patients by mass spectrometry to show that
kynurenine/tryptophan ratio, C26:0-lysoPC, and pyruvic acid can discriminate
non-COVID controls from PCR+/not-hospitalized patients (López-Hernández
et al. 2021). Moreover, C10:2-lysoPC, butyric acid, and pyruvic acid could distin-
guish between PCR+/ not-hospitalized and PCR+/hospitalized and PCR+/intubated.
Altogether, these studies highlight the potential power of metabolomics for the
Prospective Metabolomic Studies in Precision Medicine: The AKRIBEA Project 295
6 Concluding Remarks
References
Authors (2021) Ultra-precision medicine. Nat Biotechnol 39:645. https://doi.org/10.1038/s41587-
021-00967-8
Ayres JS (2020) A metabolic handbook for the COVID-19 pandemic. Nat Metab 2:572–585
Beger RD et al (2016) Metabolomics enables precision medicine: “A White Paper, Community
Perspective”. Metabolomics 12
Bernardo-Seisdedos G et al (2021) Metabolic landscape of the mouse liver by quantitative 31P
nuclear magnetic resonance analysis of the phosphorome. Hepatology 74:148–163
Blouin K et al (2005) Contribution of age and declining androgen levels to features of the metabolic
syndrome in men. Metabolism 54:1034–1040
Bruzzone C et al (2020a) SARS-CoV-2 infection dysregulates the metabolomic and lipidomic
profiles of serum. iScience 23
Bruzzone C et al (2020b) 1H NMR-based urine metabolomics reveals signs of enhanced carbon and
nitrogen recycling in prostate cancer. J Proteome Res 19:2419–2428
Bruzzone C et al (2021) A molecular signature for the metabolic syndrome by urine metabolomics.
Cardiovasc Diabetol 20:155
Carrat F et al (2021) Evidence of early circulation of SARS-CoV-2 in France: findings from the
population-based “CONSTANCES” cohort. Eur J Epidemiol 36:219–222
Christensen CM, Grossman JH, Hwang J (2009) The innovator’s prescription: a disruptive solution
for health care. McGraw-Hill
Clish CB (2015) Metabolomics: an emerging but powerful tool for precision medicine. Mol Case
Stud 1:a000588
Davis HE et al (2021) Characterizing Long COVID in an international cohort: 7 months of
symptoms and their impact. SSRN Electron J. https://doi.org/10.2139/ssrn.3820561
Day C (2007) Metabolic syndrome, or what you will: definitions and epidemiology. Diab Vasc Dis
Res 4:32–38
De Souza YG, Greenspan JS (2013) Biobanking past, present and future: responsibilities and
benefits. AIDS 27:303–312
296 M. Bizkarguenaga et al.
Dona AC et al (2014) Precision high-throughput proton NMR spectroscopy of human urine, serum,
and plasma for large-scale metabolic phenotyping. Anal Chem 86:9887–9894
Embade N et al (2019) NMR-based newborn urine screening for optimized detection of inherited
errors of metabolism. Sci Rep 9:1–9
Emwas AH et al (2019) NMR spectroscopy for metabolomics research. Metabolites 9
Ghini V, Quaglio D, Luchinat C, Turano P (2019) NMR for sample quality assessment in
metabolomics. N Biotechnol 52:25–34
González-Domínguez R, González-Domínguez Á, Sayago A, Fernández-Recamales Á (2020)
Recommendations and best practices for standardizing the pre-analytical processing of blood
and urine samples in metabolomics. Metabolites 10:1–18
Hartl D et al (2021) Translational precision medicine: an industry perspective. J Transl Med 19:1–
14
Holmes E, Wist J, Masuda R, Lodge S, Nitschke P, Kimhofer T, Loo RL, Begum S, Boughton B,
Yang R, Morillon AC, Chin ST, Hall D, Ryan M, Bong SH, Gay M, Hartmut Lawler NG,
Gray N, Whiley L, Nicholson J (2021) Incomplete systemic recovery and metabolic
phenoreversion in post-acute-phase nonhospitalized COVID-19 patients: implications for
assessment of post-acute COVID-19 syndrome. J Proteome Res
Jiménez B et al (2018) Quantitative lipoprotein subclass and low molecular weight metabolite
analysis in human serum and plasma by (1)H NMR spectroscopy in a multilaboratory trial. Anal
Chem 90:11962–11971
John A, Qin B, Kalari KR, Wang L, Yu J (2020) Patient-specific multi-omics models and the
application in personalized combination therapy. Future Oncol 16:1737–1750
Kimhofer T et al (2020) Integrative modeling of quantitative plasma lipoprotein, metabolic, and
amino acid data reveals a multiorgan pathological signature of SARS-CoV-2. J Proteome Res
Kohler I, Hankemeier T, van der Graaf PH, Knibbe CAJ, van Hasselt JGC (2017) Integrating
clinical metabolomics-based biomarker discovery and clinical pharmacology to enable precision
medicine. Eur J Pharm Sci 109:S15–S21
Koppe L et al (2013) p-Cresyl sulfate promotes insulin resistance associated with CKD. J Am Soc
Nephrol 24:88–99
Koshiba S et al (2018) Omics research project on prospective cohort studies from the Tohoku
Medical Megabank Project. Genes Cells 23:406–417
Lodge S et al (2021a) Low volume in vitro diagnostic proton NMR spectroscopy of human blood
plasma for lipoprotein and metabolite analysis: application to SARS-CoV-2 biomarkers. J
Proteome Res 20:1415–1423
Lodge S et al (2021b) NMR spectroscopic windows on the systemic effects of SARS-CoV-
2 infection on plasma lipoproteins and metabolites in relation to circulating cytokines. J
Proteome Res 20:1382–1396
Loeffler M et al (2015) The LIFE-adult-study: objectives and design of a population-based cohort
study with 10,000 deeply phenotyped adults in Germany. BMC Public Health 15:1–14
Loo RL et al (2020) Quantitative in-vitro diagnostic NMR spectroscopy for lipoprotein and
metabolite measurements in plasma and serum: recommendations for analytical artifact minimi-
zation with special reference to COVID-19/SARS-CoV-2 samples. J Proteome Res 19:4428–
4441
López-Hernández Y et al (2021) Targeted metabolomics identifies high performing diagnostic and
prognostic biomarkers for COVID-19. Sci Rep 11:14732
López-León S et al (2021) More than 50 long-term effects of COVID-19: a systematic review and
meta-analysis. SSRN Electron J 1–22. https://doi.org/10.2139/ssrn.3769978
Martin SS et al (2015) HDL cholesterol subclasses, myocardial infarction, and mortality in
secondary prevention: the Lipoprotein Investigators Collaborative. Eur Heart J 36:22–30
Meoni G et al (2021) Metabolomic/lipidomic profiling of COVID-19 and individual response to
tocilizumab. PLoS Pathog 17
Mignot S et al (2021) Correlates of premature pap test screening, under 25 years old: analysis of
data from the CONSTANCES cohort study. BMC Public Health 21:1–9
Prospective Metabolomic Studies in Precision Medicine: The AKRIBEA Project 297
Monnerie S et al (2020) Metabolomic and lipidomic signatures of metabolic syndrome and its
physiological components in adults: a systematic review. Sci Rep 10:1–13
Monsonis Centelles S et al (2017) Toward reliable lipoprotein particle predictions from NMR
spectra of human blood: an interlaboratory ring test. Anal Chem 89:8004–8012
Nabi O et al (2021) Comorbidities are associated with fibrosis in NAFLD subjects: a nationwide
study (NASH-CO study). Dig Dis Sci. https://doi.org/10.1007/s10620-021-07032-z
Nalbandian A et al (2021) Post-acute COVID-19 syndrome. Nat Med 27:601–615
Neuhauser HK (2005) The metabolic syndrome. 366
Niazkar HR, Zibaee B, Nasimi A, Bahri N (2020) The neurological manifestations of COVID-19.
Prat Neurol FMC 11:145–146
Ortega-Azorín C et al (2019) Candidate gene and genome-wide association studies for circulating
leptin levels reveal population and sex-specific associations in high cardiovascular risk Medi-
terranean subjects. Nutrients 11:2751
Otvos JD, Jeyarajah EJ, Bennett DW, Krauss RM (1992) Development of a proton nuclear
magnetic resonance spectroscopic method for determining plasma lipoprotein concentrations
and subspecies distributions from a single, rapid measurement. Clin Chem 38:1632–1638
Puchades-Carrasco L, Pineda-Lucena A (2017) Metabolomics applications in precision medicine:
an oncological perspective. Curr Top Med Chem 17:2740–2751
Reddy P, Leong J, Jialal I (2018) Amino acid levels in nascent metabolic syndrome: a contributor to
the pro-inflammatory burden. J Diabetes Complications 32:465–469
Ruiz F et al (2016) High quality standards for a large-scale prospective population-based observa-
tional cohort: constances. BMC Public Health 16:1–10
San Juan I et al (2020) Abnormal concentration of porphyrins in serum from COVID-19 patients. Br
J Haematol 190:e265–e267
Śliwczyński A, Orlewska E (2016) Precision medicine for managing chronic diseases. Pol Arch
Med Wewn 126:681–687
Thomas T et al (2020) COVID-19 infection alters kynurenine and fatty acid metabolism, correlating
with IL-6 levels and renal status. JCI Insight 5
Tigchelaar EF et al (2015) Cohort profile: LifeLines DEEP, a prospective, general population cohort
study in the northern Netherlands: Study design and baseline characteristics. BMJ Open 5:1–10
Tranvåg EJ, Strand R, Ottersen T, Norheim OF (2021) Precision medicine and the principle of equal
treatment: a conjoint analysis. BMC Med Ethics 22:1–9
Vignoli A et al (2019) High-throughput metabolomics by 1D NMR. Angew Chem Int Ed 58:968–
994
Voge NV et al (2016) Metabolomics-based discovery of small molecule biomarkers in serum
associated with dengue virus infections and disease outcomes. PLoS Negl Trop Dis 10:1–27
WHO (2022). https://www.who.int/health-topics/coronavirus
WHO (2022). https://covid19.who.int/
Wilson D, Burlingame A, Cronholm T, Sjövall J (1974) Deuterium and carbon-13 tracer studies of
ethanol metabolism in the rat by 2H, 1H-decoupled 13C nuclear magnetic resonance. Biochem
Biophys Res Commun 56:828–835
Wishart DS (2019) NMR metabolomics: a look ahead. J Magn Reson 306:155–161
Wishart DS et al (2018) HMDB 4.0: the human metabolome database for 2018. Nucleic Acids Res
46:D608–D617
Würtz P et al (2012) Metabolic signatures of insulin resistance in 7,098 young adults. Diabetes 61:
1372–1380
Yin P, Lehmann R, Xu G (2015) Effects of pre-analytical processes on blood samples used in
metabolomics studies. Anal Bioanal Chem 407:4879–4892
Yong SJ (2021) Long COVID or post-COVID-19 syndrome: putative pathophysiology, risk
factors, and treatments. Infect Dis 0:1–18
Chemotherapy-Induced Peripheral
Neuropathy
Contents
1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 300
2 Review of CIPN Metabolomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302
2.1 Metabolomics Analyses Design . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302
2.2 CIPN Study Design . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303
2.3 Clinical CIPN Metabolomics Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303
2.4 Animal and Cellular CIPN Metabolomics Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 306
2.5 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308
3 Investigation of Peripheral Neuropathy Metabolomic Biomarker Pathways . . . . . . . . . . . . . . 308
3.1 Histidine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 313
3.1.1 1-Methylhistidine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315
3.1.2 Carnosine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315
3.1.3 Ergothioneine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 316
3.1.4 Histamine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 316
3.1.5 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 316
3.2 Phenylalanine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 317
3.2.1 Phenylalanine and Tyrosine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 317
3.2.2 Phenethylamine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 319
3.2.3 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 319
3.3 N-Acetylornithine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 319
3.3.1 Ornithine and Arginine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 320
3.3.2 Ornithine and Polyamines: Putrescine, Spermidine, and Spermine . . . . . . . . . . 321
3.3.3 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321
3.4 Threonine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321
3.5 Glycogen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322
3.6 Adenosine Phosphates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 323
4 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 324
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 324
# The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 299
V. Ghini et al. (eds.), Metabolomics and Its Impact on Health and Diseases,
Handbook of Experimental Pharmacology 277, https://doi.org/10.1007/164_2022_609
300 C.-S. Chen and D. L. Hertz
Abstract
Keywords
1 Introduction
CIPN symptoms can limit instrumental activities of daily living such as preparing
meals, shopping for groceries or clothes, or using the phone. CIPN symptoms persist
in about 40% of CIPN patients for at least 2 years and are irreversible in some
patients (Bao et al. 2016; Hershman et al. 2018; Mustafa Ali et al. 2017). Irreversible
CIPN diminishes long-term quality of life (Bandos et al. 2018; Mols et al. 2014),
increases risk for falls, and causes disability in severe cases (Kolb et al. 2016;
Winters-Stone et al. 2017). These life-long consequences are particularly concerning
in patients with favorable long-term prognosis, such as those with early stage breast
cancer (McGale et al. 2014).
The mechanism of CIPN is not yet fully understood and is likely to be multifac-
torial. The neurotoxicity caused by chemotherapy may involve DNA damage,
microtubule disruption, mitochondrial damage, pain detection receptor remodeling,
oxidative stress, and neuroinflammation in the neurons of the peripheral nervous
system (Chan et al. 2019; Kerckhove et al. 2017; Zajączkowska et al. 2019). The
lack of mechanistic understanding is one reason for the lack of effective strategies for
CIPN prevention or treatment; the only guideline-recommended strategy for treat-
ment of painful CIPN is duloxetine, which has limited efficacy (Loprinzi et al. 2020;
Smith et al. 2013). Otherwise, guidelines recommend delaying, decreasing, or
discontinuing neurotoxic chemotherapy treatment in patients experiencing CIPN,
which reduces treatment efficacy (Hertz et al. 2021).
Biomarkers that could be used prior to or early in treatment to predict which
patients are most at risk for severe, and perhaps irreversible, CIPN would be
extremely clinically useful. The most established CIPN risk factors are cumulative
treatment, prior neuropathy or neurotoxic treatment, age, race, diabetes, and obesity
(Gu et al. 2021; Hershman et al. 2016; Kandula et al. 2016; Schneider et al. 2017;
Seretny et al. 2014). There may be some confounding between these factors, as age,
race, and obesity are all risk factors for diabetes, which is also a cause of peripheral
neuropathy (Bao et al. 2016; Cox-Martin et al. 2017; Gu et al. 2021; Hershman et al.
2016; Petrovchich et al. 2019; Smith and Singleton 2013; Timmins et al. 2021;
Wiggin et al. 2009). Modifiable nutritional factors such as diet and nutrient supple-
mentation have also been reported to influence CIPN risk (Mongiovi et al. 2018;
Stankovic et al. 2020; Zirpoli et al. 2017). More severe CIPN has been reported in
patients with lower grain consumption, not taking vitamins or taking antioxidants
(Mongiovi et al. 2018; Stankovic et al. 2020; Zirpoli et al. 2017), deficiency of
vitamin D or saturated fatty acids (Grim et al. 2017; Jennaro et al. 2020), or a
sedentary lifestyle (Bulls et al. 2020; Duregon et al. 2018; Greenlee et al. 2017;
Kanzawa-Lee et al. 2020; Mols et al. 2015). Again, these risk factors may be due to
shared mechanistic pathways with neuropathy caused by poor nutrition, obesity,
and/or diabetes. Finally, substantial research has been undertaken to identify
inherited genetic markers, including those that affect neurotoxic drug elimination
or pharmacology, diabetes, and neuronal cell function or survival (Chan et al. 2019;
Cliff et al. 2017; Ng et al. 2014; Sucheston-Campbell et al. 2018; Terrazzino et al.
2015), however, no genetic markers have been sufficiently validated for use in
clinical practice (Chan et al. 2019).
302 C.-S. Chen and D. L. Hertz
The two most common techniques in metabolomics analyses are nuclear magnetic
resonance (NMR) and mass spectrometry (MS). Each has its own advantages and
disadvantages in profile coverage, sensitivity, stability, sample volume, sample
preparation, quantification, high-throughput automation, time, and cost (Everett
2017; Wishart 2016). MS-based methods are 10–100 times more sensitive than
NMR. Whereas a typical LC-MS study can detect more than 1,000 metabolites
with concentration of 10–100 nM in human plasma, a typical NMR study can only
detect 50–200 metabolites with concentration at the microM scale. On the other
hand, only a few hundred of the metabolites detected by MS-based methods can be
identified (Emwas et al. 2019). NMR sensitivity can be improved with digital
spectrometers, cryoprobes, and low volume probes (Everett 2017). There is a trend
toward increasing use of NMR due to its greater reproducibility and the ease of full
automation (Everett 2017; Wishart 2016). Although NMR requires larger sample
volume, sample preparation is minimal (Kirwan et al. 2018; Wishart 2016), and
NMR can provide absolute quantification of metabolite concentrations in only a few
minutes without reference standards (Wishart 2016). The start-up cost of NMR
instrumentation is expensive, but the cost per sample is low (Everett 2017; Wishart
Chemotherapy-Induced Peripheral Neuropathy 303
2016). There are two approaches to metabolite identification, targeted and untargeted
metabolomics. Targeted approaches focus on a set of defined metabolites, whereas
global untargeted approaches capture all metabolites that are present in a sample but
may not provide the exact identification of some of the metabolites due to the
limitations of databases or libraries. Untargeted metabolomics is usually qualitative,
or can provide relative quantitation, and is used for hypothesis-agnostic discovery,
while targeted metabolomics can provide absolute quantitation and is often used to
confirm results from untargeted studies or confirm hypotheses (Roberts et al. 2012).
Both NMR and MS-based methods can be used in untargeted and targeted analyses,
but MS-based methods are generally superior for untargeted metabolomics discov-
ery due to high sensitivity and for targeted metabolomics validation due to its
selectivity, while NMR is used primarily in untargeted analyses (Emwas et al. 2019).
Metabolomics studies of CIPN enroll patients with cancer who are going to receive
neurotoxic chemotherapy. CIPN data are collected from clinician or patient assess-
ment, and patients can be defined as whether or not they experienced CIPN, the
severity of their CIPN, or the cumulative dose at the time that CIPN occurred (Hertz
2019). Biosamples are collected pretreatment (baseline) and on-treatment to acquire
metabolite information. The pretreatment metabolite levels can be used to identify
metabolic signatures that predict peripheral neuropathy, whereas the on-treatment
changes from baseline can be used to find metabolic signatures that are associated
with the neurotoxic effect of the chemotherapy, which can still be predictive if they
are sufficiently early in treatment that CIPN has not yet become clinically observ-
able. Metabolic changes at the time of clinically evident CIPN are not useful for
prediction but may be useful to understand mechanisms underlying CIPN.
Two CIPN metabolomics studies have been conducted in patients receiving chemo-
therapy (Table 1). One untargeted whole blood NMR study by Sun et al. (2018)
enrolled 48 patients with stage I-III or oligometastatic breast cancer planning to
receive weekly paclitaxel and obtained metabolomics profiles prior to treatment
(baseline), at the end of the first paclitaxel infusion, and 24 h after the first infusion.
Paclitaxel-induced peripheral neuropathy was assessed by a patient-reported ques-
tionnaire at baseline and before each infusion, and CIPN severity was defined by the
maximum change from baseline of the sensory symptom subscale. Without strict
correction for multiple comparisons, lower pretreatment levels of three amino acids,
histidine, phenylalanine, and threonine, were moderately correlated with more
severe sensory neuropathy symptoms. Statistically uncorrected secondary
analyses identified many metabolites that changed from pretreatment to
post-infusion or 24-h later, however, none of these were associated with CIPN
Table 1 Human metabolomics studies of CIPN
304
Although clinical CIPN metabolomics studies are very scarce, animal and cellular
studies may assist with identifying metabolic pathways of interest (Table 2). Two
untargeted NMR studies by Ferrier et al. (2013, 2015) analyzed the metabolomics of
central nervous system tissue in rat models of oxaliplatin-induced neurotoxicity. A
single injection of oxaliplatin increased glutamine and decreased creatinine and
adenosine phosphate in spinal dorsal horn tissues in rats that developed acute pain
hypersensitivity compared to vehicle-treated rats, and these metabolic changes and
pain hypersensitivity were abrogated by a polyamine-deficient diet (Ferrier et al.
2013). In a similar study of multiple oxaliplatin injections, higher choline and
glycerophosphocholine and lower aspartate, glutamine, creatinine, and GABA
were found in various brain tissues in rats that developed painful neuropathy
compared to vehicle-treated rats (Ferrier et al. 2015). An untargeted LC-MS/MS
study by Wu et al. (2018) analyzed the plasma metabolomics of a rat model of
paclitaxel-induced peripheral neuropathy and found 19 lipids, including fatty acids,
ketones, and glycerophosphocholine, which differentiated paclitaxel-treated from
vehicle-treated rats. An untargeted study by Qin et al. (2012) used a different
metabolomics technique, capillary electrophoresis (CE-MS), which is increasingly
used in metabolomics due to its low sample volume requirement and high efficiency
Table 2 Animal and cellular metabolomics studies of CIPN
Neurotoxic Metabolomics Statistical
Subjects chemotherapy CIPN data techniques Sample type methods Findings Reference
Adult male rat Single Behavioral tests of NMR, Spinal dorsal Mann– Increased glutamine and (Ferrier
model of intraperitoneal mechanical allodynia, untargeted horn tissue, Whitney decreased creatinine and et al.
oxaliplatin- oxaliplatin pain hypersensitivity, 18 metabolites synthetic diet U tests adenosine phosphate 2013)
induced acute injection and cold available ad were found in
pain hypersensitivity libitum oxaliplatin-treated rats,
hypersensitivity and polyamine-deficient
diet prevented such
metabolic changes and
oxaliplatin-induced
acute pain
hypersensitivity
Chemotherapy-Induced Peripheral Neuropathy
Male rat model Intravenous Behavioral tests of NMR, Various brain Mann– Higher choline and (Ferrier
of oxaliplatin- oxaliplatin mechanical allodynia, untargeted tissues, food Whitney glycerophosphocholine et al.
induced painful injection twice a cold hypersensitivity 17 metabolites available ad U tests and lower aspartate, 2015)
neuropathy week for four libitum creatinine, glutamine,
and half weeks and GABA were found
in oxaliplatin-treated
rats
Female rat model Intraperitoneal Behavioral tests of LC-MS/MS, Plasma, food PLSDA Fatty acids, ketones, and (Wu et al.
of paclitaxel- paclitaxel mechanical allodynia untargeted, available ad and t tests glycerophosphocholine 2018)
induced injection on total number of libitum differentiated between
peripheral three alternate metabolites not paclitaxel-treated rats
neuropathy days reported and controls
Human neural Low-dose Immunofluorescence CE-MS, Cell culture Mann– Lower glutathione was (Qin et al.
progenitor cells thalidomide for imaging of neuronal untargeted Whitney found in thalidomide- 2012)
3 days differentiation 74 metabolites U tests treated cells during
neural differentiation
307
Mechanical allodynia is a painful sensation caused by innocuous stimuli such as light touch of clothes
308 C.-S. Chen and D. L. Hertz
for separating polar metabolites (Harada et al. 2018; Kristoff et al. 2020; Zhang and
Ramautar 2021). This study analyzed human neural progenitor cell cultures treated
with low-dose thalidomide and found, compared to vehicle-treated controls, lower
glutathione in thalidomide-treated cells during neural differentiation. Of the
metabolites found in these animal and cellular studies, only adenosine phosphate
was consistent with the metabolites identified in the CIPN clinical studies. This may
be due to the use of vehicle-treated controls, as opposed to treated rats that did not
develop CIPN, suggesting these results may reflect the direct effect of chemotherapy
and be unrelated to CIPN.
2.5 Summary
Two pioneering metabolomics studies have been conducted in patients with cancer
receiving neurotoxic chemotherapy to identify metabolic predictors of CIPN. These
studies suggest six metabolites with putative pathways that may be pretreatment or
early-in-treatment predictors of CIPN: histidine, phenylalanine, threonine (Sun et al.
2018), N-acetylornithine, glycogen, and adenosine mono/diphosphate (Verma et al.
2020). Except for adenosine phosphate, studies in animal models and neuronal
progenitor cells found different metabolites that are affected by chemotherapy
treatment, but their relationship to CIPN is unknown (Ferrier et al. 2013, 2015;
Qin et al. 2012; Wu et al. 2018).
The rest of this chapter further investigates the putative associations between these
six potential metabolomic biomarkers of CIPN, which will be listed in descending
order of the strength of evidence (Table 3). These pathways are examined for any
evidence of their involvement in human metabolomics studies of other types of
peripheral neuropathy, including diabetic neuropathy, hereditary neuropathy, and
peripheral neuropathy caused by autoimmune disorders. It can be difficult to collect
metabolic or peripheral neuropathy information before the onset of these other
etiologies of chronic peripheral neuropathy. Therefore, metabolomics studies usu-
ally compare metabolic profiles between patients with peripheral neuropathy and
controls, who are either healthy volunteers or patients who share the underlying
disease state but are not experiencing peripheral neuropathy. Some studies also
analyzed the association between metabolite levels and the severity of peripheral
neuropathy. Due to the large number of metabolites and the complex and unclear
mechanisms underlying peripheral neuropathy, many studies also adjust for clinical
covariates to try to identify the most representative metabolites.
In addition to reviewing metabolomics studies of other etiologies of peripheral
neuropathy, for each of these six potential metabolomic biomarkers of CIPN, the
putative mechanistic relationship with neuropathy is reviewed and any observational
Table 3 Evidence of association for each of the putative CIPN metabolomics biomarkers
Other
CIPN Other PN Other observational interventional Putative
Metabolites Description metabolomics metabolomics studies studies mechanisms
Histidine Essential from food Lower pretreatment Higher plasma Nominally higher Carnosine Histidine,
Precursor of plasma histidine histidine associated blood supplementation carnosine, and
histamine, which is associated with with diabetic ergothioneine in prevents oxaliplatin ergothioneine have
a neurotransmitter more severe neuropathy (Lin patients with less CIPN (Yehia et al. anti-oxidant and
Metabolites such as sensory paclitaxel- et al. 2019) sensory CIPN 2019) anti-inflammatory
carnosine and induced peripheral Lower plasma (Winkels et al. Ergothioneine properties
ergothioneine have neuropathy (Sun histidine correlated 2020) prevents platinum Ergothioneine
also been found to et al. 2018) with worse (Jong et al. 2011; inhibits oxaliplatin
be associated with cardiovascular Nishida et al. 2018; uptake in neurons
PN autonomic Song et al. 2010) Histamine
neuropathy CIPN in animals inhibitors reduce
(Mathew et al. Histamine pain detection in
Chemotherapy-Induced Peripheral Neuropathy
(continued)
Table 3 (continued)
310
Other
CIPN Other PN Other observational interventional Putative
Metabolites Description metabolomics metabolomics studies studies mechanisms
Phenylalanine Essential from food Lower pretreatment Lower skin Dietary Levodopa relieves Deficiencies of
Precursor of plasma fibroblast phenylalanine diabetic phenylalanine and
tyrosine and phenylalanine phenylalanine and restriction neuropathic pain tyrosine cause
catecholamine associated with tyrosine levels associated with (Ertas et al. 1998) sensory
neurotransmitters more severe associated with sensory neuropathy A phenethylamine neuropathy
such as levodopa sensory paclitaxel- Leber hereditary in patients derivative prevents
Precursor of induced peripheral optic neuropathy (Ludolph et al. and relieves acute
phenethylamine, neuropathy (Sun (Chao de la Barca 1992) and cats oxaliplatin-induced
which is a et al. 2018) et al. 2016; Morvan (Dickinson et al. neurotoxicity
neurotransmitter and Demidem 2004) (Durand et al.
2018) 2012) and diabetic
neuropathic pain
(Kadiroglu et al.
2008; Razazian
et al. 2014;
Rowbotham et al.
2004)
N-acetylornithine Non-essential N-acetylornithine Lower plasma Lower serum Arginine Arginine is anti-
Metabolized to levels on day ornithine correlated arginine associated supplementation oxidant
ornithine and then 29 predictive of with worse with diabetic prevents diabetic Polyamine-
polyamines vincristine-induced cardiovascular neuropathy (Ganz neuropathy pain in deficient diet
Ornithine peripheral autonomic et al. 2017) rats (Rondón et al. reduces neuronal
predominantly neuropathy (Verma neuropathy in 2018), but not excitotoxicity
from arginine et al. 2020) diabetic patients significant in
(Mathew et al. patients (Jude et al.
2019) 2010)
Higher plasma A polyamine-
N-acetylputrescine deficient diet
C.-S. Chen and D. L. Hertz
levels associated prevents pain
with peripheral hypersensitivity
neuropathy in (Rivat et al. 2008)
obese patients (Guo including
et al. 2021) oxaliplatin-induced
Lower skin acute pain
fibroblast arginine hypersensitivity in
and polyamines rats (Ferrier et al.
levels associated 2013)
with Leber
hereditary optic
neuropathy (Chao
de la Barca et al.
2016)
Threonine Essential from food Lower pretreatment Lower skin Higher glycine Glycine antagonist No mechanism
Chemotherapy-Induced Peripheral Neuropathy
Small amount plasma threonine fibroblast threonine levels in spinal cord did not improve determined
converts to glycine associated with and glycine levels tissues in mice with neuropathic pain
more severe associated with Charcot-Marie- (Wallace et al.
sensory paclitaxel- Leber hereditary Tooth type 2D 2002)
induced peripheral optic neuropathy neuropathy (Bais
neuropathy (Sun (Chao de la Barca et al. 2016)
et al. 2018) et al. 2016; Morvan
and Demidem
2018)
Higher
cerebrospinal fluid
threonine levels
found in patients
with endemic optic
neuropathy
(Gonzalez-
Quevedo et al.
311
2001)
(continued)
Table 3 (continued)
312
Other
CIPN Other PN Other observational interventional Putative
Metabolites Description metabolomics metabolomics studies studies mechanisms
Glycogen Glucose storage Glycogen levels on N/A Glycogen Anti-glycemic Glycogen
Glucose is its day 29 predictive of accumulation in agents prevent accumulation
precursor and vincristine-induced neurons or Schwan diabetic neuropathy causes neuropathy
metabolite peripheral cells of patients (El Mouhayyar Hyperglycemia
neuropathy (Verma with late-onset et al. 2020) and causes neuronal
et al. 2020) Pompe disease possibly treat CIPN injury and
(Finsterer et al. (Erdoğan et al. ischemia
2017) and diabetic 2020; Fujita et al.
neuropathy 2015; Shigematsu
(Bischoff 1980; et al. 2020)
Powell et al. 1985;
Yagihashi and
Matsunaga 1979)
Adenosine DNA and RNA Adenosine mono/ N/A Impaired adenosine An adenosine Adenosine
phosphates purine precursors diphosphate levels phosphate phosphate phosphate
Energy storage on day signaling pathway signaling pathway signaling
29 predictive of in mice with activator prevents pathways
vincristine-induced diabetic neuropathy oxaliplatin CIPN in promoting
peripheral (Roy Chowdhury patients (El-Fatatry mitochondrial
neuropathy (Verma et al. 2012) et al. 2018) and biogenesis and
et al. 2020) Patients taking an mice (Mao-Ying inhibiting
Decreased adenosine et al. 2014) autophagy and
adenosine phosphate neuroinflammation
phosphate signaling pathway
indicative of activator had lower
oxaliplatin-induced risk of diabetic
acute pain neuropathy
hypersensitivity (Pop-Busui et al.
C.-S. Chen and D. L. Hertz
and interventional studies that support the association are described. Four of these
metabolites are amino acids, which are the building blocks for protein synthesis. In
humans, there are 20 proteinogenic amino acids, of which nine essential amino acids
are not produced endogenously and must be supplied from the diet. Many amino
acids participate in neurotransmitter biosynthesis, providing an apparent mechanism
by which they may be involved in peripheral neuropathy (Dalangin et al. 2020;
Kölker 2018). The other two metabolites are glycogen and adenosine phosphate.
Glycogen plays a critical role in carbohydrate metabolism (Nordlie et al. 1999), and
adenosine phosphate is the precursor of RNA and provides energy for cellular
metabolic process (Hardie et al. 2012).
3.1 Histidine
Histidine is an essential amino acid supplied from dietary sources, such as meat.
Histidine has an imidazole function group that can scavenge reactive oxygen species
to achieve anti-oxidant and anti-inflammatory effects (Peterson et al. 1998). This
section will also discuss four of its potentially peripheral neuropathy-related
metabolites, 1-methylhistidine, carnosine, ergothioneine, and histamine (Fig. 1).
Similar to the paclitaxel-induced peripheral neuropathy metabolomics study (Sun
et al. 2018), two metabolomics studies identified that histidine was associated with
diabetic neuropathy, but the directions of associations were inconclusive, and the
associations were not significant after adjusting for clinical covariates (Lin et al.
Fig. 1 Histidine and its peripheral neuropathy-related metabolites have several putative
mechanisms (Bocca et al. 2021; Chao de la Barca et al. 2016; Jong et al. 2011; Lin et al. 2019;
Mathew et al. 2019; Nishida et al. 2018; Song et al. 2010; Sun et al. 2018; Winkels et al. 2020;
Yehia et al. 2019). Quercetin and lafutidine are not histidine metabolites, but they are histamine
inhibitors (Azevedo et al. 2013; Gao et al. 2016; Nagano et al. 2012; Tsukaguchi et al. 2013). Blue
boxes are neurotransmitters, such as histamine. Orange boxes are metabolites or compounds that
have been found to prevent or treat CIPN. Solid arrows are metabolic reactions that can happen in
humans, and dashed arrows are the ones that happen exclusively in animals. X-shaped arrows
indicate inhibition
314 C.-S. Chen and D. L. Hertz
2019; Mathew et al. 2019). A longitudinal MS-based study by Mathew et al. (2019)
measured plasma amino acids and tricarboxylic acid metabolites in patients with
type I diabetes but without any signs of microvascular complications and age- and
sex-matched healthy controls at baseline and for 3 years of observation. Correlation
coefficients between baseline metabolite levels and end-of-study electrocardiogram
parameters were calculated to identify metabolites that predict cardiovascular auto-
nomic neuropathy. Lower histidine levels were moderately correlated with worse
neuropathy but did not remain significant after adjusting for baseline clinical
covariates, including blood glucose, HbA1c, years of diabetes, body mass index,
cholesterol, estimated glomerular filtration rate, and urine microalbumin-to-creati-
nine ratio. An untargeted NMR study by Lin et al. (2019) enrolled patients with type
II diabetes with and without microangiopathy and healthy controls. Orthogonal
partial least squares discriminant analysis (OPLSDA) was used to identify predictive
metabolites and build predictive models of diabetes and diabetic microangiopathy
complications including sensory diabetic neuropathy. Higher plasma histidine was
identified in multiple models that predicted the occurrence of diabetic neuropathy,
but the association was not significant after adjusting for clinical covariates, includ-
ing age, sex, body mass index, and the use of medication for hypertension and
hyperlipidemia. The lack of association after covariate adjustment and seemingly
conflicting findings in these studies does not eliminate the possibility of histidine
being involved in the development of peripheral neuropathy, but indicates that the
causal relationship between histidine, diabetes, and peripheral neuropathy may be
complicated. For example, the two studies of diabetic neuropathy required fasting
before sample collection (Lin et al. 2019; Mathew et al. 2019), but the paclitaxel-
induced peripheral neuropathy study did not (Sun et al. 2018). Since diet can affect
CIPN (Mongiovi et al. 2018), diabetic neuropathy (Bunner et al. 2015; Oza et al.
2021), and diabetes (Neuenschwander et al. 2019; Schwingshackl et al. 2017), and
most of the histidine in the body is supplied by dietary sources, it is difficult to
determine whether lower or higher plasma histidine is causally related to peripheral
neuropathy or this relationship is due to statistical confounding. More studies are
needed to elucidate the effect of histidine on, and its potential for use as a predictive
biomarker of, peripheral neuropathy.
Several peripheral neuropathy metabolomics studies were focused on Leber
hereditary optic neuropathy (LHON), which is an inherited disease that results in
vision loss that can include other elements of peripheral neuropathy such as tremors
and movement disorders. LHON is caused by mutations of mitochondrial DNA
including MT-ND1, MT-ND4, MT-ND4L, or MT-ND6, which are critical for proper
mitochondrial function. Studies have analyzed skin fibroblast metabolomics in
patients with LHON to understand the metabolic changes caused by mitochondrial
dysfunction without the confounding of the short-term effects of diet, medication,
and circadian cycles, which complicate research of systemic metabolomics (Wilkins
et al. 2019). A targeted LC-MS/MS metabolomics study by Chao de la Barca et al.
(2016) used OPLSDA to compare skin fibroblast metabolomics between patients
with LHON and healthy controls. Levels of almost all proteinogenic amino acids,
including histidine, were significantly lower in patients with LHON (Chao de la
Chemotherapy-Induced Peripheral Neuropathy 315
Barca et al. 2016), which is consistent with the known function of mitochondria
synthesizing amino acids to release stored energy.
Histidine has not been tested for prevention or treatment of any types of periph-
eral neuropathy. However, histidine supplementation in obese women with meta-
bolic syndrome has been demonstrated in a randomized controlled trial to suppress
inflammation and oxidative stress (Feng et al. 2013), which may also protect against
CIPN. Interventional studies of histidine supplementation for peripheral neuropathy
prevention and treatment may be warranted.
3.1.1 1-Methylhistidine
1-Methylhistidine is a histidine metabolite that is found exclusively in red meat or
poultry, making it a biomarker for meat consumption (Altorf-van der Kuil et al.
2013; Dragsted 2010; Fraser et al. 2016; Khodorova et al. 2019; Kochlik et al. 2018;
Lloyd et al. 2011; Mitry et al. 2019; Sjölin et al. 1987; Yin et al. 2017), and has a
half-life about half day (Sjölin et al. 1987). Another untargeted LC-MS
metabolomics study of LHON by Bocca et al. (2021) found higher plasma
1-methylhistidine in patients than healthy controls in an OPLSDA multivariable
model. This study required fasting before sample collection (Bocca et al. 2021), so
this result is likely attributed to habitual meat consumption. However, prior studies
have not found that higher meat consumption increases CIPN (Kenkhuis et al. 2021;
Mongiovi et al. 2018). Alternatively, two small interventional studies demonstrated
that patients with diabetic neuropathy had less pain after receiving a low-fat and
plant-based diet (Bunner et al. 2015; Smith et al. 2006). Again, the causal
relationships between diet, metabolites, and neuropathy are likely complex and not
fully understood.
3.1.2 Carnosine
Carnosine is a dipeptide of histidine and alanine that is abundant in human and
animal muscle and brain tissue and is a biomarker for red meat consumption (Altorf-
van der Kuil et al. 2013; Cuparencu et al. 2019; Mitry et al. 2019). Observational
metabolomics studies of meat intake often find increased carnosine (Cheung et al.
2017; Cuparencu et al. 2019; Vázquez-Fresno et al. 2015). Similar to histidine,
carnosine has anti-oxidant and anti-inflammatory properties. In a randomized con-
trolled trial, carnosine supplementation was protective against oxaliplatin-induced
peripheral neuropathy, possibly by reducing nuclear factor erythroid-2 related
factor-2 (NRF2) and nuclear factor kappa light chain enhancer of activated B cells
(NFκB) (Yehia et al. 2019). Nrf2 is involved in the redox pathway and has been
proposed as a potential treatment target for CIPN (Miao et al. 2019; Yang et al. 2018)
and diabetic neuropathy (Ganesh Yerra et al. 2013; Gupta et al. 2021; Kumar and
Mittal 2017; Xu et al. 2013). Similarly, NFκB contributes to CIPN (Janes et al. 2014;
Li et al. 2015; Wang et al. 2017) and diabetic neuropathy (Cameron and Cotter 2008;
Dewanjee et al. 2018; Ganesh Yerra et al. 2013) through its involvement in inflam-
matory pathways.
316 C.-S. Chen and D. L. Hertz
3.1.3 Ergothioneine
The other histidine downstream metabolite of interest, ergothioneine, is synthesized
exclusively by fungi and bacteria, which makes it a biomarker for mushroom
consumption (Pallister et al. 2016). An observational study of patients with colorec-
tal cancer receiving chemotherapy found that patients with higher blood
ergothioneine had nominally, but not significantly, less sensory CIPN symptoms
(Winkels et al. 2020). Similar to histidine and carnosine, ergothioneine is a potent
anti-oxidant that may protect injured tissue (Cheah and Halliwell 2012; Halliwell
et al. 2016). In addition to its anti-oxidant properties, ergothioneine has also been
found in animal studies to reduce oxaliplatin accumulation in dorsal root ganglion
neurons via inhibition of organic cation/carnitine transporter (OCTN1)-mediated
oxaliplatin transport (Fujita et al. 2019; Gründemann 2012; Jong et al. 2011;
Nishida et al. 2018). Ergothioneine has been found to protect against cisplatin
(Song et al. 2010) and oxaliplatin-induced peripheral neuropathy in animal studies
(Jong et al. 2011; Nishida et al. 2018), but we are not aware of any interventional
studies in humans.
3.1.4 Histamine
Histidine is a precursor of histamine, which is a neurotransmitter that participates in
regulation of pain and neurogenic inflammation (Rosa and Fantozzi 2013; Yu et al.
2016). Histamine may have a role in diabetes and diabetic neuropathy (Pini et al.
2016; Wei et al. 2016), and there is a potential that histidine levels affect the nervous
system through histamine pathways, but we are not aware of any observational
peripheral neuropathy metabolomics studies that have directly assessed histamine. A
flavonoid from plants, quercetin, has been found to inhibit mast cells from releasing
antigen-induced histamine (Kimata et al. 2000; Pearce et al. 1984) and subsequently
inhibit transient receptor potential cation channel subfamily V member 1 (TRPV1)
(Gao et al. 2016), which has functions of detection and regulation of heat and pain
(Benítez-Angeles et al. 2020; Cui et al. 2016; Frias and Merighi 2016; Gouin et al.
2017; Romanovsky et al. 2009). Quercetin has been found to have anti-oxidant
properties via the nitric oxide pathways (Di Carlo et al. 1999; Gao et al. 2016) and
anti-inflammatory property via the arachidonic acid pathways (Di Carlo et al. 1999;
Ferrándiz and Alcaraz 1991) and has been found in animal studies to prevent pain
from oxaliplatin-(Azevedo et al. 2013) and paclitaxel-induced peripheral neuropathy
(Gao et al. 2016). A histamine H2 receptor antagonist, lafutidine, has been found in
small clinical trials to potentially be effective for treating taxane-induced peripheral
neuropathy (Nagano et al. 2012) and prevent bortezomib-induced peripheral neu-
ropathy (Tsukaguchi et al. 2013). The pain relieving effect of lafutidine has been
hypothesized to be similar to capsaicin, which activates and then reduces the
expression of TRPV1 (TRPV1 defunctionalization) in sensory neurons (Onodera
et al. 1995, 1999; Umeda et al. 1999; Yamamoto et al. 2001).
3.1.5 Summary
Lower histidine levels have been found to be associated with paclitaxel-induced
peripheral neuropathy (Sun et al. 2018) and LHON (Chao de la Barca et al. 2016),
Chemotherapy-Induced Peripheral Neuropathy 317
but the directions of associations in diabetic neuropathy were inconclusive (Lin et al.
2019; Mathew et al. 2019). The evidence suggests that histidine plays some role in
peripheral neuropathy. However, the causal mechanistic relationship is unclear,
which likely explains the lack of significance after adjusting for clinical covariates
(Lin et al. 2019; Mathew et al. 2019).
Metabolomics studies of LHON found higher levels of 1-methylhistidine (Bocca
et al. 2021), which has not been identified in observational dietary analyses
(Kenkhuis et al. 2021; Mongiovi et al. 2018), but suggested that reducing meat
consumption may be a possible dietary intervention of peripheral neuropathy
(Bunner et al. 2015; Smith et al. 2006). If meat consumption is demonstrated to
increase CIPN risk, 1-methylhistidine and carnosine are indicators of meat consump-
tion that could be used as CIPN biomarkers (Altorf-van der Kuil et al. 2013;
Cuparencu et al. 2019; Mitry et al. 2019).
Histidine, carnosine, and ergothioneine all have anti-oxidant and anti-
inflammatory properties that can protect injured tissues and possibly protect against
peripheral neuropathy. Carnosine supplementation may prevent oxaliplatin-induced
peripheral neuropathy through anti-oxidant and anti-inflammatory pathways (Yehia
et al. 2019), whereas ergothioneine may have an additional mechanism of reducing
oxaliplatin accumulation in neurons (Fujita et al. 2019; Gründemann 2012; Jong
et al. 2011; Nishida et al. 2018; Winkels et al. 2020).
Histidine may affect the nervous system through histamine, a neurotransmitter
that regulates pain and neurogenic inflammation (Rosa and Fantozzi 2013; Yu et al.
2016). Histamine inhibitors such as quercetin (Azevedo et al. 2013; Gao et al. 2016)
and lafutidine (Nagano et al. 2012; Tsukaguchi et al. 2013) may prevent or relieve
CIPN pain. Further experimental work is needed to confirm these mechanisms and
randomized interventional trials are needed to confirm the effectiveness for preven-
tion or treatment of CIPN.
3.2 Phenylalanine
Fig. 2 Phenylalanine and its peripheral neuropathy-related metabolites (Chao de la Barca et al.
2016; Dickinson et al. 2004; Ertas et al. 1998; Ludolph et al. 1992; Morvan and Demidem 2018;
Sun et al. 2018). Blue boxes are neurotransmitters. Orange boxes are metabolites or compounds that
have been found to relieve neuropathic pain. Solid arrows are metabolic reactions that can happen in
humans, and circle-shaped arrows indicate induction. Venlafaxine is a phenethylamine derivative
that inhibits the reuptake of norepinephrine, which leads to increased norepinephrine levels (Durand
et al. 2012; Kadiroglu et al. 2008; Razazian et al. 2014; Rowbotham et al. 2004)
disorders of the central nervous system, including headache (D’Andrea et al. 2019),
depression (Bot et al. 2020), and Alzheimer’s disease (Albrahim 2020), but that is
beyond the scope of this chapter.
Even though phenylalanine and its metabolites seem to play a role in neurotrans-
mission, only two peripheral neuropathy metabolomics studies identified differences
in phenylalanine levels. These two LHON skin fibroblast metabolomics studies
found most of the proteinogenic amino acids levels, including phenylalanine and
tyrosine, were significantly lower in patients with LHON (Chao de la Barca et al.
2016; Morvan and Demidem 2018). In a previously mentioned diabetic neuropathy
study, higher plasma tyrosine, but not phenylalanine, was identified in some multi-
variable models, but the performance of these models was not ideal, and tyrosine was
not significant after adjusting for clinical covariates (Lin et al. 2019).
There are no interventional trials of phenylalanine supplementation to prevent or
treat peripheral neuropathy, to the best of our knowledge. However, an observational
study suggests that dietary phenylalanine restriction in patients with hereditary
hyperphenylalaninemia causes sensory neuropathy (Ludolph et al. 1992). Another
study in cats also showed that restricting dietary phenylalanine and tyrosine can
cause sensory neuropathy (Dickinson et al. 2004). This strongly implicates
Chemotherapy-Induced Peripheral Neuropathy 319
3.2.2 Phenethylamine
The other phenylalanine metabolite, phenethylamine, is a brain neurotransmitter.
Human metabolomics studies of the use of methamphetamine (Boxler et al. 2017;
Kim et al. 2020), a phenethylamine derivative, have analyzed phenethylamine, but
we are not aware of any observational peripheral neuropathy metabolomics studies
that have assessed phenethylamine.
Another phenethylamine derivative, venlafaxine, is a serotonin-norepinephrine
reuptake inhibitor antidepressant that has been found to prevent and relieve acute
oxaliplatin-induced neurotoxicity (Durand et al. 2012), but not oxaliplatin-induced
peripheral neuropathy (Zimmerman et al. 2016). Venlafaxine has also been found to
reduce pain from diabetic neuropathy (Kadiroglu et al. 2008; Razazian et al. 2014;
Rowbotham et al. 2004), but is less effective than duloxetine and pregabalin for
treating CIPN (Farshchian et al. 2018) or diabetic neuropathy (Raskin et al. 2006;
Razazian et al. 2014).
3.2.3 Summary
Similar to the findings in CIPN (Sun et al. 2018), metabolomics studies in LHON
(Chao de la Barca et al. 2016; Morvan and Demidem 2018) indicate that phenylala-
nine or its metabolites may be involved in peripheral neuropathy. Two studies of
phenylalanine or tyrosine dietary restriction confirm this causes sensory peripheral
neuropathy (Dickinson et al. 2004; Ludolph et al. 1992), likely through the cate-
cholamine neurotransmitter pathway. Catecholamines have demonstrated effective-
ness in neuropathy-relevant conditions including the effectiveness of levodopa and
venlafaxine for relieving neuropathic pain caused by chemotherapy (Durand et al.
2012) or diabetes (Ertas et al. 1998; Kadiroglu et al. 2008; Razazian et al. 2014;
Rowbotham et al. 2004).
3.3 N-Acetylornithine
3.3.3 Summary
The direct association between N-acetylornithine and peripheral neuropathy was
only found in the vincristine-induced peripheral neuropathy metabolomics study
(Verma et al. 2020), but lower levels of ornithine, arginine, and polyamines have
been found in patients with other etiologies of peripheral neuropathy. The minimal
data from interventional studies of arginine supplementation trials indicate limited
efficacy. Despite the seemingly conflicting findings in observational studies of
polyamines, polyamine-deficient diet may protect against peripheral neuropathic
pain through NMDA pathways (Ferrier et al. 2013; Rivat et al. 2008). More studies
with a larger sample size are needed to explore the role of ornithine, arginine, or
polyamines in peripheral neuropathy.
3.4 Threonine
The association between lower threonine and peripheral neuropathy was found in the
paclitaxel-induced peripheral neuropathy whole blood metabolomics study (Sun
et al. 2018) Threonine is an essential amino acid found in high-protein food sources,
such as meat and cheese. Threonine is abundant in human plasma and does not seem
to be directly associated with neuropathy, but a small amount of threonine is
converted to glycine (Darling et al. 2000; Edgar 2002; Zhao et al. 1986). This
section will discuss threonine and glycine together.
322 C.-S. Chen and D. L. Hertz
3.5 Glycogen
Glycogen is a polysaccharide that is the primary storage form of glucose. Other than
the vincristine-induced peripheral neuropathy study (Verma et al. 2020), no other
peripheral neuropathy metabolomics studies have assessed glycogen. An inherited
disorder, Pompe disease, is caused by mutations of alpha glucosidase (GAA), which
leads to glycogen accumulation in lysosomes, especially in heart and skeletal
muscles. Neuropathy has been reported in patients with late-onset Pompe disease
due to glycogen accumulation in neurons or Schwan cells (Finsterer et al. 2017).
Aggregates of glycogen particles have been found in neurons and Schwann cells in
patients with diabetic neuropathy as well (Bischoff 1980; Powell et al. 1985;
Yagihashi and Matsunaga 1979). These studies strongly suggest that glycogen
accumulation is associated with peripheral neuropathy, but the role of glycogen in
CIPN is unclear.
Chronic elevation of glucose, the precursor and metabolite of glycogen, is the
main cause of diabetes, and high glucose levels in peripheral nerves is the pathogen-
esis of diabetic neuropathy (Freeman et al. 2016). Hyperglycemia causes injury to
neuronal cells and excessive glucose metabolism promotes thickening of capillary
basement membrane, which leads to neuronal ischemia (Filla and Edwards 2016;
Forbes and Cooper 2013). Numerous anti-glycemic agents have been approved for
Chemotherapy-Induced Peripheral Neuropathy 323
reducing blood glucose in patients with diabetes and is expected to reduce the risk of
diabetic microvascular complication including peripheral neuropathy (El Mouhayyar
et al. 2020), and some of these drugs also showed efficacy in treating CIPN through
glucagon-like peptide-1 signaling pathways (Erdoğan et al. 2020; Fujita et al. 2015;
Shigematsu et al. 2020). However, blood glucose varies by dietary intake, and
glycosylated hemoglobin is considered to be a more accurate long-term biomarker
of glycemic control than short-term blood glucose (Koenig et al. 1976).
4 Conclusion
Although scarce, there have been a few CIPN metabolomics biomarker studies in
clinical, animal, and cellular models. Our review of the clinical studies identified six
metabolites that may predict CIPN risk. We were able to find supportive evidence for
these associations by reviewing metabolomics, observational, and interventional
studies of CIPN and other etiologies of peripheral neuropathy. Histidine and phe-
nylalanine have clear roles in neurotransmission or neuroinflammation in peripheral
neuropathy, including CIPN. Furthermore, interventional studies of their
metabolites, including carnosine and levodopa, have shown promise in preventing
or treating CIPIN and other types of peripheral neuropathy, though definitive
interventional trials are needed. Further research is needed to discover and validate
CIPN metabolomics biomarkers in large clinical studies and to elucidate the
mechanisms and causal relationships through animal or cell culture experiments.
After retrospective validation, prospective biomarker-based interventional studies
are essential to translate the biomarker findings into clinical practice. For example,
metabolomics biomarkers could be used to identify patients at high CIPN risk before
or early in treatment, in whom CIPN monitoring could be enhanced or investiga-
tional preventive interventions could be tested. Metabolomic biomarkers can also be
indicative of effective interventions, such as nutrient supplementation, to prevent or
reduce CIPN. The ultimate goal is to improve treatment outcomes and quality of life
of patients with cancer.
References
Albrahim T (2020) The potential role of nutritional components in improving brain function among
patients with Alzheimers disease: a meta-analysis of RCT studies. Neurosciences 25:4–17.
https://doi.org/10.17712/nsj.2020.1.20190037
Altorf-van der Kuil W, Brink EJ, Boetje M, Siebelink E, Bijlsma S, Engberink MF, van’t Veer P,
Tomé D, Bakker SJ, van Baak MA, Geleijnse JM (2013) Identification of biomarkers for intake
of protein from meat, dairy products and grains: a controlled dietary intervention study. Br J
Nutr 110:810–822. https://doi.org/10.1017/s0007114512005788
Argyriou AA, Iconomou G, Kalofonos HP (2008) Bortezomib-induced peripheral neuropathy in
multiple myeloma: a comprehensive review of the literature. Blood 112:1593–1599. https://doi.
org/10.1182/blood-2008-04-149385
Azevedo MI, Pereira AF, Nogueira RB, Rolim FE, Brito GA, Wong DV, Lima-Júnior RC, de
Albuquerque RR, Vale ML (2013) The antioxidant effects of the flavonoids rutin and quercetin
inhibit oxaliplatin-induced chronic painful peripheral neuropathy. Mol Pain 9:53. https://doi.
org/10.1186/1744-8069-9-53
Backshall A, Sharma R, Clarke SJ, Keun HC (2011) Pharmacometabonomic profiling as a predictor
of toxicity in patients with inoperable colorectal cancer treated with capecitabine. Clin Cancer
Res 17:3019–3028. https://doi.org/10.1158/1078-0432.Ccr-10-2474
Bais P, Beebe K, Morelli KH, Currie ME, Norberg SN, Evsikov AV, Miers KE, Seburn KL,
Guergueltcheva V, Kremensky I, Jordanova A, Bult CJ, Burgess RW (2016) Metabolite profile
of a mouse model of Charcot-Marie-Tooth type 2D neuropathy: implications for disease
mechanisms and interventions. Biol Open 5:908–920. https://doi.org/10.1242/bio.019273
Chemotherapy-Induced Peripheral Neuropathy 325
Balayssac D, Ferrier J, Pereira B, Gillet B, Pétorin C, Vein J, Libert F, Eschalier A, Pezet D (2015)
Prevention of oxaliplatin-induced peripheral neuropathy by a polyamine-reduced diet-
NEUROXAPOL: protocol of a prospective, randomised, controlled, single-blind and
monocentric trial. BMJ Open 5:e007479. https://doi.org/10.1136/bmjopen-2014-007479
Bandos H, Melnikow J, Rivera DR, Swain SM, Sturtz K, Fehrenbacher L, Wade JL 3rd, Brufsky
AM, Julian TB, Margolese RG, McCarron EC, Ganz PA (2018) Long-term peripheral neuropa-
thy in breast cancer patients treated with adjuvant chemotherapy: NRG oncology/NSABP B-30.
J Natl Cancer Inst 110. https://doi.org/10.1093/jnci/djx162
Bao T, Basal C, Seluzicki C, Li SQ, Seidman AD, Mao JJ (2016) Long-term chemotherapy-induced
peripheral neuropathy among breast cancer survivors: prevalence, risk factors, and fall risk.
Breast Cancer Res Treat 159:327–333. https://doi.org/10.1007/s10549-016-3939-0
Benítez-Angeles M, Morales-Lázaro SL, Juárez-González E, Rosenbaum T (2020) TRPV1: struc-
ture, endogenous agonists, and mechanisms. Int J Mol Sci 21. https://doi.org/10.3390/
ijms21103421
Bischoff A (1980) Morphology of diabetic neuropathy. Horm Metab Res Suppl 9:18–28
Bocca C, Le Paih V, Barca JMC, Nzoughet JK, Amati-Bonneau P, Blanchet O, Védié B,
Géromin D, Simard G, Procaccio V, Bonneau D, Leaners G, Orssaud C, Reynier P (2021) A
plasma metabolomic signature of Leber hereditary optic neuropathy showing taurine and
nicotinamide deficiencies. Hum Mol Genet. https://doi.org/10.1093/hmg/ddab013
Bot M, Milaneschi Y, Al-Shehri T, Amin N, Garmaeva S, Onderwater GLJ, Pool R, Thesing CS,
Vijfhuizen LS, Vogelzangs N, Arts ICW, Demirkan A, van Duijn C, van Greevenbroek M, van
der Kallen CJH, Köhler S, Ligthart L, van den Maagdenberg A, Mook-Kanamori DO, de
Mutsert R, Tiemeier H, Schram MT, Stehouwer CDA, Terwindt GM, Willems van Dijk K,
Fu J, Zhernakova A, Beekman M, Slagboom PE, Boomsma DI, Penninx B (2020)
Metabolomics profile in depression: a pooled analysis of 230 metabolic markers in 5283
cases with depression and 10,145 controls. Biol Psychiatry 87:409–418. https://doi.org/10.
1016/j.biopsych.2019.08.016
Boxler MI, Liechti ME, Schmid Y, Kraemer T, Steuer AE (2017) First time view on human
metabolome changes after a single intake of 3,4-methylenedioxymethamphetamine in healthy
placebo-controlled subjects. J Proteome Res 16:3310–3320. https://doi.org/10.1021/acs.
jproteome.7b00294
Bulls HW, Hoogland AI, Small BJ, Kennedy B, James BW, Arboleda BL, Shahzad MMK,
Gonzalez BD, Jim HSL (2020) Lagged relationships among chemotherapy-induced peripheral
neuropathy, sleep quality, and physical activity during and after chemotherapy. Ann Behav
Med. https://doi.org/10.1093/abm/kaaa101
Bunner AE, Wells CL, Gonzales J, Agarwal U, Bayat E, Barnard ND (2015) A dietary intervention
for chronic diabetic neuropathy pain: a randomized controlled pilot study. Nutr Diabetes 5:e158.
https://doi.org/10.1038/nutd.2015.8
Cameron NE, Cotter MA (2008) Pro-inflammatory mechanisms in diabetic neuropathy: focus on
the nuclear factor kappa B pathway. Curr Drug Targets 9:60–67. https://doi.org/10.2174/
138945008783431718
Chan A, Hertz DL, Morales M, Adams EJ, Gordon S, Tan CJ, Staff NP, Kamath J, Oh J, Shinde S,
Pon D, Dixit N, D'Olimpio J, Dumitrescu C, Gobbo M, Kober K, Mayo S, Pang L, Subbiah I,
Beutler AS, Peters KB, Loprinzi C, Lustberg MB (2019) Biological predictors of chemotherapy-
induced peripheral neuropathy (CIPN): MASCC neurological complications working group
overview. Support Care Cancer 27:3729–3737. https://doi.org/10.1007/s00520-019-04987-8
Chao de la Barca JM, Simard G, Amati-Bonneau P, Safiedeen Z, Prunier-Mirebeau D, Chupin S,
Gadras C, Tessier L, Gueguen N, Chevrollier A, Desquiret-Dumas V, Ferré M, Bris C, Kouassi
Nzoughet J, Bocca C, Leruez S, Verny C, Miléa D, Bonneau D, Lenaers G, Martinez MC,
Procaccio V, Reynier P (2016) The metabolomic signature of Leber's hereditary optic neuropa-
thy reveals endoplasmic reticulum stress. Brain 139:2864–2876. https://doi.org/10.1093/brain/
aww222
326 C.-S. Chen and D. L. Hertz
Cheah IK, Halliwell B (2012) Ergothioneine; antioxidant potential, physiological function and role
in disease. Biochim Biophys Acta 1822:784–793. https://doi.org/10.1016/j.bbadis.2011.09.017
Chen L, Chen CS, Sun Y, Henry NL, Stringer KA, Hertz DL (2021) Feasibility of
pharmacometabolomics to identify potential predictors of paclitaxel pharmacokinetic
variability. Cancer Chemother Pharmacol. https://doi.org/10.1007/s00280-021-04300-7
Cheung W, Keski-Rahkonen P, Assi N, Ferrari P, Freisling H, Rinaldi S, Slimani N, Zamora-Ros R,
Rundle M, Frost G, Gibbons H, Carr E, Brennan L, Cross AJ, Pala V, Panico S, Sacerdote C,
Palli D, Tumino R, Kühn T, Kaaks R, Boeing H, Floegel A, Mancini F, Boutron-Ruault MC,
Baglietto L, Trichopoulou A, Naska A, Orfanos P, Scalbert A (2017) A metabolomic study of
biomarkers of meat and fish intake. Am J Clin Nutr 105:600–608. https://doi.org/10.3945/ajcn.
116.146639
Clayton TA, Baker D, Lindon JC, Everett JR, Nicholson JK (2009) Pharmacometabonomic
identification of a significant host-microbiome metabolic interaction affecting human drug
metabolism. Proc Natl Acad Sci U S A 106:14728–14733. https://doi.org/10.1073/pnas.
0904489106
Cliff J, Jorgensen AL, Lord R, Azam F, Cossar L, Carr DF, Pirmohamed M (2017) The molecular
genetics of chemotherapy-induced peripheral neuropathy: a systematic review and meta-
analysis. Crit Rev Oncol Hematol 120:127–140. https://doi.org/10.1016/j.critrevonc.2017.
09.009
Cox-Martin E, Trahan LH, Cox MG, Dougherty PM, Lai EA, Novy DM (2017) Disease burden and
pain in obese cancer patients with chemotherapy-induced peripheral neuropathy. Support Care
Cancer 25:1873–1879. https://doi.org/10.1007/s00520-017-3571-5
Cui M, Gosu V, Basith S, Hong S, Choi S (2016) Polymodal transient receptor potential vanilloid
type 1 Nocisensor: structure, modulators, and therapeutic applications. Adv Protein Chem Struct
Biol 104:81–125. https://doi.org/10.1016/bs.apcsb.2015.11.005
Cuparencu C, Rinnan Å, Dragsted LO (2019) Combined markers to assess meat intake-human
metabolomic studies of discovery and validation. Mol Nutr Food Res 63:e1900106. https://doi.
org/10.1002/mnfr.201900106
D'Andrea G, Gucciardi A, Perini F, Leon A (2019) Pathogenesis of cluster headache: from episodic
to chronic form, the role of neurotransmitters and neuromodulators. Headache 59:1665–1670.
https://doi.org/10.1111/head.13673
Dalangin R, Kim A, Campbell RE (2020) The role of amino acids in neurotransmission and
fluorescent tools for their detection. Int J Mol Sci 21. https://doi.org/10.3390/ijms21176197
Darling PB, Grunow J, Rafii M, Brookes S, Ball RO, Pencharz PB (2000) Threonine dehydroge-
nase is a minor degradative pathway of threonine catabolism in adult humans. Am J Physiol
Endocrinol Metab 278:E877–E884. https://doi.org/10.1152/ajpendo.2000.278.5.E877
Dewanjee S, Das S, Das AK, Bhattacharjee N, Dihingia A, Dua TK, Kalita J, Manna P (2018)
Molecular mechanism of diabetic neuropathy and its pharmacotherapeutic targets. Eur J
Pharmacol 833:472–523. https://doi.org/10.1016/j.ejphar.2018.06.034
Di Carlo G, Mascolo N, Izzo AA, Capasso F (1999) Flavonoids: old and new aspects of a class of
natural therapeutic drugs. Life Sci 65:337–353. https://doi.org/10.1016/s0024-3205(99)
00120-4
Dickinson PJ, Anderson PJ, Williams DC, Powell HC, Shelton GD, Morris JG, LeCouteur RA
(2004) Assessment of the neurologic effects of dietary deficiencies of phenylalanine and
tyrosine in cats. Am J Vet Res 65:671–680. https://doi.org/10.2460/ajvr.2004.65.671
Dragsted LO (2010) Biomarkers of meat intake and the application of nutrigenomics. Meat Sci 84:
301–307. https://doi.org/10.1016/j.meatsci.2009.08.028
Durand JP, Deplanque G, Montheil V, Gornet JM, Scotte F, Mir O, Cessot A, Coriat R,
Raymond E, Mitry E, Herait P, Yataghene Y, Goldwasser F (2012) Efficacy of venlafaxine
for the prevention and relief of oxaliplatin-induced acute neurotoxicity: results of EFFOX, a
randomized, double-blind, placebo-controlled phase III trial. Ann Oncol 23:200–205. https://
doi.org/10.1093/annonc/mdr045
Chemotherapy-Induced Peripheral Neuropathy 327
Fraser GE, Jaceldo-Siegl K, Henning SM, Fan J, Knutsen SF, Haddad EH, Sabaté J, Beeson WL,
Bennett H (2016) Biomarkers of dietary intake are correlated with corresponding measures from
repeated dietary recalls and food-frequency questionnaires in the adventist health study-2. J Nutr
146:586–594. https://doi.org/10.3945/jn.115.225508
Freeman OJ, Unwin RD, Dowsey AW, Begley P, Ali S, Hollywood KA, Rustogi N, Petersen RS,
Dunn WB, Cooper GJ, Gardiner NJ (2016) Metabolic dysfunction is restricted to the sciatic
nerve in experimental diabetic neuropathy. Diabetes 65:228–238. https://doi.org/10.2337/
db15-0835
Frias B, Merighi A (2016) Capsaicin, nociception and pain. Molecules 21. https://doi.org/10.3390/
molecules21060797
Fujita S, Hirota T, Sakiyama R, Baba M, Ieiri I (2019) Identification of drug transporters
contributing to oxaliplatin-induced peripheral neuropathy. J Neurochem 148:373–385. https://
doi.org/10.1111/jnc.14607
Fujita S, Ushio S, Ozawa N, Masuguchi K, Kawashiri T, Oishi R, Egashira N (2015) Exenatide
facilitates recovery from oxaliplatin-induced peripheral neuropathy in rats. PLoS One 10:
e0141921. https://doi.org/10.1371/journal.pone.0141921
Ganesh Yerra V, Negi G, Sharma SS, Kumar A (2013) Potential therapeutic effects of the
simultaneous targeting of the Nrf2 and NF-κB pathways in diabetic neuropathy. Redox Biol
1:394–397. https://doi.org/10.1016/j.redox.2013.07.005
Ganz T, Wainstein J, Gilad S, Limor R, Boaz M, Stern N (2017) Serum asymmetric
dimethylarginine and arginine levels predict microvascular and macrovascular complications
in type 2 diabetes mellitus. Diabetes Metab Res Rev 33. https://doi.org/10.1002/dmrr.2836
Gao W, Zan Y, Wang ZJ, Hu XY, Huang F (2016) Quercetin ameliorates paclitaxel-induced
neuropathic pain by stabilizing mast cells, and subsequently blocking PKCε-dependent activa-
tion of TRPV1. Acta Pharmacol Sin 37:1166–1177. https://doi.org/10.1038/aps.2016.58
Gonzalez-Quevedo A, Obregon F, Fernandez R, Santiesteban R, Serrano C, Lima L (2001) Amino
acid levels and ratios in serum and cerebrospinal fluid of patients with optic neuropathy in Cuba.
Nutr Neurosci 4:51–62. https://doi.org/10.1080/1028415x.2001.11747350
Gouin O, L'Herondelle K, Lebonvallet N, Le Gall-Ianotto C, Sakka M, Buhé V, Plée-Gautier E,
Carré JL, Lefeuvre L, Misery L, Le Garrec R (2017) TRPV1 and TRPA1 in cutaneous
neurogenic and chronic inflammation: pro-inflammatory response induced by their activation
and their sensitization. Protein Cell 8:644–661. https://doi.org/10.1007/s13238-017-0395-5
Greenlee H, Hershman DL, Shi Z, Kwan ML, Ergas IJ, Roh JM, Kushi LH (2017) BMI, lifestyle
factors and Taxane-induced neuropathy in breast cancer patients: the pathways study. J Natl
Cancer Inst 109. https://doi.org/10.1093/jnci/djw206
Grim J, Ticha A, Hyspler R, Valis M, Zadak Z (2017) Selected risk nutritional factors for
chemotherapy-induced polyneuropathy. Nutrients 9:535. https://doi.org/10.3390/nu9060535
Gründemann D (2012) The ergothioneine transporter controls and indicates ergothioneine activity--
a review. Prev Med 54 Suppl: S71–S74. doi: https://doi.org/10.1016/j.ypmed.2011.12.001
Gu J, Lu H, Chen C, Gu Z, Hu M, Liu L, Yu J, Wei G, Huo J (2021) Diabetes mellitus as a risk
factor for chemotherapy-induced peripheral neuropathy: a meta-analysis. Support Care Cancer.
https://doi.org/10.1007/s00520-021-06321-7
Guasch-Ferré M, Hruby A, Toledo E, Clish CB, Martínez-González MA, Salas-Salvadó J, Hu FB
(2016) Metabolomics in prediabetes and diabetes: a systematic review and meta-analysis.
Diabetes Care 39:833–846. https://doi.org/10.2337/dc15-2251
Guo K, Savelieff MG, Rumora AE, Alakwaa FM, Callaghan BC, Hur J, Feldman EL (2021) Plasma
metabolomics and lipidomics differentiate obese individuals by peripheral neuropathy status. J
Clin Endocrinol Metab. https://doi.org/10.1210/clinem/dgab844
Gupta A, Behl T, Sehgal A, Bhatia S, Jaglan D, Bungau S (2021) Therapeutic potential of Nrf-2
pathway in the treatment of diabetic neuropathy and nephropathy. Mol Biol Rep 48:2761–2774.
https://doi.org/10.1007/s11033-021-06257-5
Chemotherapy-Induced Peripheral Neuropathy 329
Li Y, Zhang H, Kosturakis AK, Cassidy RM, Zhang H, Kennamer-Chapman RM, Jawad AB,
Colomand CM, Harrison DS, Dougherty PM (2015) MAPK signaling downstream to TLR4
contributes to paclitaxel-induced peripheral neuropathy. Brain Behav Immun 49:255–266.
https://doi.org/10.1016/j.bbi.2015.06.003
Lin HT, Cheng ML, Lo CJ, Lin G, Lin SF, Yeh JT, Ho HY, Lin JR, Liu FC (2019) (1)H nuclear
magnetic resonance (NMR)-based cerebrospinal fluid and plasma metabolomic analysis in type
2 diabetic patients and risk prediction for diabetic microangiopathy. J Clin Med 8. https://doi.
org/10.3390/jcm8060874
Lloyd AJ, Favé G, Beckmann M, Lin W, Tailliart K, Xie L, Mathers JC, Draper J (2011) Use of
mass spectrometry fingerprinting to identify urinary metabolites after consumption of specific
foods. Am J Clin Nutr 94:981–991. https://doi.org/10.3945/ajcn.111.017921
Loprinzi CL, Lacchetti C, Bleeker J, Cavaletti G, Chauhan C, Hertz DL, Kelley MR, Lavino A,
Lustberg MB, Paice JA, Schneider BP, Lavoie Smith EM, Smith ML, Smith TJ, Wagner-
Johnston N, Hershman DL (2020) Prevention and management of chemotherapy-induced
peripheral neuropathy in survivors of adult cancers: ASCO guideline update. J Clin Oncol 38:
3325–3348. https://doi.org/10.1200/jco.20.01399
Ludolph AC, Ullrich K, Nedjat S, Masur H, Bick U (1992) Neurological outcome in 22 treated
adolescents with hyperphenylalaninemia. A clinical and electrophysiological study. Acta
Neurol Scand 85:243–248. https://doi.org/10.1111/j.1600-0404.1992.tb04039.x
Madhavi YV, Gaikwad N, Yerra VG, Kalvala AK, Nanduri S, Kumar A (2019) Targeting AMPK in
diabetes and diabetic complications: energy homeostasis, autophagy and mitochondrial health.
Curr Med Chem 26:5207–5229. https://doi.org/10.2174/0929867325666180406120051
Maekawa K, Ri M, Nakajima M, Sekine A, Ueda R, Tohkin M, Miyata N, Saito Y, Iida S (2019)
Serum lipidomics for exploring biomarkers of bortezomib therapy in patients with multiple
myeloma. Cancer Sci 110:3267–3274. https://doi.org/10.1111/cas.14178
Mao-Ying QL, Kavelaars A, Krukowski K, Huo XJ, Zhou W, Price TJ, Cleeland C, Heijnen CJ
(2014) The anti-diabetic drug metformin protects against chemotherapy-induced peripheral
neuropathy in a mouse model. PLoS One 9:e100701. https://doi.org/10.1371/journal.pone.
0100701
Mathew AV, Jaiswal M, Ang L, Michailidis G, Pennathur S, Pop-Busui R (2019) Impaired amino
acid and TCA metabolism and cardiovascular autonomic neuropathy progression in type
1 diabetes. Diabetes 68: 2035–2044. doi: https://doi.org/10.2337/db19-0145
McGale P, Taylor C, Correa C, Cutter D, Duane F, Ewertz M, Gray R, Mannu G, Peto R, Whelan T,
Wang Y, Wang Z, Darby S (2014) Effect of radiotherapy after mastectomy and axillary surgery
on 10-year recurrence and 20-year breast cancer mortality: meta-analysis of individual patient
data for 8135 women in 22 randomised trials. Lancet 383:2127–2135. https://doi.org/10.1016/
s0140-6736(14)60488-8
Miao H, Xu J, Xu D, Ma X, Zhao X, Liu L (2019) Nociceptive behavior induced by chemothera-
peutic paclitaxel and beneficial role of antioxidative pathways. Physiol Res 68:491–500. https://
doi.org/10.33549/physiolres.933939
Miltenburg NC, Boogerd W (2014) Chemotherapy-induced neuropathy: a comprehensive survey.
Cancer Treat Rev 40:872–882. https://doi.org/10.1016/j.ctrv.2014.04.004
Mitry P, Wawro N, Rohrmann S, Giesbertz P, Daniel H, Linseisen J (2019) Plasma concentrations
of anserine, carnosine and pi-methylhistidine as biomarkers of habitual meat consumption. Eur J
Clin Nutr 73:692–702. https://doi.org/10.1038/s41430-018-0248-1
Mols F, Beijers AJ, Vreugdenhil G, Verhulst A, Schep G, Husson O (2015) Chemotherapy-induced
peripheral neuropathy, physical activity and health-related quality of life among colorectal
cancer survivors from the PROFILES registry. J Cancer Surviv 9:512–522. https://doi.org/10.
1007/s11764-015-0427-1
Mols F, Beijers T, Vreugdenhil G, van de Poll-Franse L (2014) Chemotherapy-induced peripheral
neuropathy and its association with quality of life: a systematic review. Support Care Cancer 22:
2261–2269. https://doi.org/10.1007/s00520-014-2255-7
332 C.-S. Chen and D. L. Hertz
Mongiovi JM, Zirpoli GR, Cannioto R, Sucheston-Campbell LE, Hershman DL, Unger JM, Moore
HCF, Stewart JA, Isaacs C, Hobday TJ, Salim M, Hortobagyi GN, Gralow JR, Thomas Budd G,
Albain KS, Ambrosone CB, McCann SE (2018) Associations between self-reported diet during
treatment and chemotherapy-induced peripheral neuropathy in a cooperative group trial
(S0221). Breast Cancer Res 20:146. https://doi.org/10.1186/s13058-018-1077-9
Morvan D, Demidem A (2018) NMR metabolomics of fibroblasts with inherited mitochondrial
complex I mutation reveals treatment-reversible lipid and amino acid metabolism alterations.
Metabolomics 14:55. https://doi.org/10.1007/s11306-018-1345-9
Mustafa Ali M, Moeller M, Rybicki L, Moore HCF (2017) Long-term peripheral neuropathy
symptoms in breast cancer survivors. Breast Cancer Res Treat 166:519–526. https://doi.org/
10.1007/s10549-017-4437-8
Nagano H, Sanai H, Muraoka M, Takagi K (2012) Efficacy of lafutidine, a histamine H2-receptor
antagonist, for taxane-induced peripheral neuropathy in patients with gynecological
malignancies. Gynecol Oncol 127:172–174. https://doi.org/10.1016/j.ygyno.2012.06.029
Napoli E, Schneider A, Wang JY, Trivedi A, Carrillo NR, Tassone F, Rogawski M, Hagerman RJ,
Giulivi C (2019) Allopregnanolone treatment improves plasma metabolomic profile associated
with GABA metabolism in fragile X-associated tremor/ataxia syndrome: a pilot study. Mol
Neurobiol 56:3702–3713. https://doi.org/10.1007/s12035-018-1330-3
Neuenschwander M, Ballon A, Weber KS, Norat T, Aune D, Schwingshackl L, Schlesinger S
(2019) Role of diet in type 2 diabetes incidence: umbrella review of meta-analyses of prospec-
tive observational studies. BMJ 366:l2368. https://doi.org/10.1136/bmj.l2368
Ng T, Chan M, Khor CC, Ho HK, Chan A (2014) The genetic variants underlying breast cancer
treatment-induced chronic and late toxicities: a systematic review. Cancer Treat Rev 40:1199–
1214. https://doi.org/10.1016/j.ctrv.2014.10.001
Nishida K, Takeuchi K, Hosoda A, Sugano S, Morisaki E, Ohishi A, Nagasawa K (2018)
Ergothioneine ameliorates oxaliplatin-induced peripheral neuropathy in rats. Life Sci 207:
516–524. https://doi.org/10.1016/j.lfs.2018.07.006
Nordlie RC, Foster JD, Lange AJ (1999) Regulation of glucose production by the liver. Annu Rev
Nutr 19:379–406. https://doi.org/10.1146/annurev.nutr.19.1.379
O'Gorman A, Brennan L (2017) The role of metabolomics in determination of new dietary
biomarkers. Proc Nutr Soc 76:295–302. https://doi.org/10.1017/s0029665116002974
Onodera S, Shibata M, Tanaka M, Inaba N, Arai Y, Aoyama M, Lee B, Yamaura T (1999)
Gastroprotective mechanism of lafutidine, a novel anti-ulcer drug with histamine H2-receptor
antagonistic activity. Arzneimittelforschung 49:519–526. https://doi.org/10.1055/s-
0031-1300454
Onodera S, Shibata M, Tanaka M, Inaba N, Yamaura T, Ohnishi H (1995) Gastroprotective activity
of FRG-8813, a novel histamine H2-receptor antagonist, in rats. Jpn J Pharmacol 68:161–173.
https://doi.org/10.1254/jjp.68.161
Oza MJ, Laddha AP, Gaikwad AB, Mulay SR, Kulkarni YA (2021) Role of dietary modifications in
the management of type 2 diabetic complications. Pharmacol Res 168:105602. https://doi.org/
10.1016/j.phrs.2021.105602
Pallister T, Jennings A, Mohney RP, Yarand D, Mangino M, Cassidy A, MacGregor A, Spector
TD, Menni C (2016) Characterizing blood metabolomics profiles associated with self-reported
food intakes in female twins. PLoS One 11:e0158568. https://doi.org/10.1371/journal.pone.
0158568
Park JE, Lim HR, Kim JW, Shin KH (2018) Metabolite changes in risk of type 2 diabetes mellitus in
cohort studies: a systematic review and meta-analysis. Diabetes Res Clin Pract 140:216–227.
https://doi.org/10.1016/j.diabres.2018.03.045
Paschen W (1992) Polyamine metabolism in different pathological states of the brain. Mol Chem
Neuropathol 16:241–271. https://doi.org/10.1007/bf03159973
Pearce FL, Befus AD, Bienenstock J (1984) Mucosal mast cells. III. Effect of quercetin and other
flavonoids on antigen-induced histamine secretion from rat intestinal mast cells. J Allergy Clin
Immunol 73:819–823. https://doi.org/10.1016/0091-6749(84)90453-6
Chemotherapy-Induced Peripheral Neuropathy 333
Peterson JW, Boldogh I, Popov VL, Saini SS, Chopra AK (1998) Anti-inflammatory and
antisecretory potential of histidine in salmonella-challenged mouse small intestine. Lab Invest
78:523–534
Petrovchich I, Kober KM, Wagner L, Paul SM, Abrams G, Chesney MA, Topp K, Smoot B,
Schumacher M, Conley YP, Hammer M, Levine JD, Miaskowski C (2019) Deleterious effects
of higher body mass index on subjective and objective measures of chemotherapy-induced
peripheral neuropathy in cancer survivors. J Pain Symptom Manage 58:252–263. https://doi.
org/10.1016/j.jpainsymman.2019.04.029
Pini A, Obara I, Battell E, Chazot PL, Rosa AC (2016) Histamine in diabetes: is it time to
reconsider? Pharmacol Res 111:316–324. https://doi.org/10.1016/j.phrs.2016.06.021
Plasmati R, Pastorelli F, Cavo M, Petracci E, Zamagni E, Tosi P, Cangini D, Tacchetti P, Salvi F,
Bartolomei I, Michelucci R, Tassinari CA (2007) Neuropathy in multiple myeloma treated with
thalidomide: a prospective study. Neurology 69:573–581. https://doi.org/10.1212/01.wnl.
0000267271.18475.fe
Pop-Busui R, Lu J, Lopes N, Jones TL (2009) Prevalence of diabetic peripheral neuropathy and
relation to glycemic control therapies at baseline in the BARI 2D cohort. J Peripher Nerv Syst
14:1–13. https://doi.org/10.1111/j.1529-8027.2009.00200.x
Powell HC, Rosoff J, Myers RR (1985) Microangiopathy in human diabetic neuropathy. Acta
Neuropathol 68:295–305. https://doi.org/10.1007/bf00690832
Price TJ, Das V, Dussor G (2016) Adenosine monophosphate-activated protein kinase (AMPK)
activators for the prevention, treatment and potential reversal of pathological pain. Curr Drug
Targets 17:908–920. https://doi.org/10.2174/1389450116666151102095046
Qin XY, Akanuma H, Wei F, Nagano R, Zeng Q, Imanishi S, Ohsako S, Yoshinaga J, Yonemoto J,
Tanokura M, Sone H (2012) Effect of low-dose thalidomide on dopaminergic neuronal differ-
entiation of human neural progenitor cells: a combined study of metabolomics and morphologi-
cal analysis. Neurotoxicology 33:1375–1380. https://doi.org/10.1016/j.neuro.2012.08.016
Quasthoff S, Hartung HP (2002) Chemotherapy-induced peripheral neuropathy. J Neurol 249:9–17.
https://doi.org/10.1007/pl00007853
Raskin J, Smith TR, Wong K, Pritchett YL, D'Souza DN, Iyengar S, Wernicke JF (2006)
Duloxetine versus routine care in the long-term management of diabetic peripheral neuropathic
pain. J Palliat Med 9:29–40. https://doi.org/10.1089/jpm.2006.9.29
Rattray NJW, Deziel NC, Wallach JD, Khan SA, Vasiliou V, Ioannidis JPA, Johnson CH (2018)
Beyond genomics: understanding exposotypes through metabolomics. Hum Genomics 12:4.
https://doi.org/10.1186/s40246-018-0134-x
Razazian N, Baziyar M, Moradian N, Afshari D, Bostani A, Mahmoodi M (2014) Evaluation of the
efficacy and safety of pregabalin, venlafaxine, and carbamazepine in patients with painful
diabetic peripheral neuropathy. A randomized, double-blind trial. Neurosciences (Riyadh) 19:
192–198
Remmington T, Smith S (2021) Tyrosine supplementation for phenylketonuria. Cochrane Database
Syst Rev 1:Cd001507. https://doi.org/10.1002/14651858.CD001507.pub4
Rivat C, Richebé P, Laboureyras E, Laulin JP, Havouis R, Noble F, Moulinoux JP, Simonnet G
(2008) Polyamine deficient diet to relieve pain hypersensitivity. Pain 137:125–137. https://doi.
org/10.1016/j.pain.2007.08.021
Roberts LD, Souza AL, Gerszten RE, Clish CB (2012) Targeted metabolomics. Curr Protoc Mol
Biol. Chapter 30: Unit 30.2.1-24. https://doi.org/10.1002/0471142727.mb3002s98
Romanovsky AA, Almeida MC, Garami A, Steiner AA, Norman MH, Morrison SF, Nakamura K,
Burmeister JJ, Nucci TB (2009) The transient receptor potential vanilloid-1 channel in thermo-
regulation: a thermosensor it is not. Pharmacol Rev 61:228–261. https://doi.org/10.1124/pr.109.
001263
Rondón LJ, Farges MC, Davin N, Sion B, Privat AM, Vasson MP, Eschalier A, Courteix C (2018)
L-arginine supplementation prevents allodynia and hyperalgesia in painful diabetic neuropathic
rats by normalizing plasma nitric oxide concentration and increasing plasma agmatine concen-
tration. Eur J Nutr 57:2353–2363. https://doi.org/10.1007/s00394-017-1508-x
334 C.-S. Chen and D. L. Hertz
Rosa AC, Fantozzi R (2013) The role of histamine in neurogenic inflammation. Br J Pharmacol
170:38–45. https://doi.org/10.1111/bph.12266
Rowbotham MC, Goli V, Kunz NR, Lei D (2004) Venlafaxine extended release in the treatment of
painful diabetic neuropathy: a double-blind, placebo-controlled study. Pain 110:697–706.
https://doi.org/10.1016/j.pain.2004.05.010
Roy Chowdhury SK, Smith DR, Saleh A, Schapansky J, Marquez A, Gomes S, Akude E,
Morrow D, Calcutt NA, Fernyhough P (2012) Impaired adenosine monophosphate-activated
protein kinase signalling in dorsal root ganglia neurons is linked to mitochondrial dysfunction
and peripheral neuropathy in diabetes. Brain 135:1751–1766. https://doi.org/10.1093/brain/
aws097
Schneider BP, Shen F, Jiang G, O'Neill A, Radovich M, Li L, Gardner L, Lai D, Foroud T, Sparano
JA, Sledge GW Jr, Miller KD (2017) Impact of genetic ancestry on outcomes in ECOG-ACRIN-
E5103. JCO Precis Oncol 2017. https://doi.org/10.1200/po.17.00059
Schwingshackl L, Hoffmann G, Lampousi AM, Knüppel S, Iqbal K, Schwedhelm C, Bechthold A,
Schlesinger S, Boeing H (2017) Food groups and risk of type 2 diabetes mellitus: a systematic
review and meta-analysis of prospective studies. Eur J Epidemiol 32:363–375. https://doi.org/
10.1007/s10654-017-0246-y
Seretny M, Currie GL, Sena ES, Ramnarine S, Grant R, MacLeod MR, Colvin LA, Fallon M (2014)
Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: a sys-
tematic review and meta-analysis. Pain 155:2461–2470. https://doi.org/10.1016/j.pain.2014.
09.020
Shaw S, Uniyal A, Gadepalli A, Tiwari V, Belinskaia DA, Shestakova NN, Venugopala KN, Deb
PK, Tiwari V (2020) Adenosine receptor signalling: probing the potential pathways for the
ministration of neuropathic pain. Eur J Pharmacol 889:173619. https://doi.org/10.1016/j.ejphar.
2020.173619
Shigematsu N, Kawashiri T, Kobayashi D, Shimizu S, Mine K, Hiromoto S, Uchida M, Egashira N,
Shimazoe T (2020) Neuroprotective effect of alogliptin on oxaliplatin-induced peripheral
neuropathy in vivo and in vitro. Sci Rep 10:6734. https://doi.org/10.1038/s41598-020-62738-w
Shin SY, Fauman EB, Petersen AK, Krumsiek J, Santos R, Huang J, Arnold M, Erte I, Forgetta V,
Yang TP, Walter K, Menni C, Chen L, Vasquez L, Valdes AM, Hyde CL, Wang V, Ziemek D,
Roberts P, Xi L, Grundberg E, Waldenberger M, Richards JB, Mohney RP, Milburn MV, John
SL, Trimmer J, Theis FJ, Overington JP, Suhre K, Brosnan MJ, Gieger C, Kastenmüller G,
Spector TD, Soranzo N (2014) An atlas of genetic influences on human blood metabolites. Nat
Genet 46:543–550. https://doi.org/10.1038/ng.2982
Shrikanth CB, Nandini CD (2020) AMPK in microvascular complications of diabetes and the
beneficial effects of AMPK activators from plants. Phytomedicine 73:152808. https://doi.org/
10.1016/j.phymed.2018.12.031
Sjölin J, Hjort G, Friman G, Hambraeus L (1987) Urinary excretion of 1-methylhistidine: a
qualitative indicator of exogenous 3-methylhistidine and intake of meats from various sources.
Metabolism 36:1175–1184. https://doi.org/10.1016/0026-0495(87)90245-9
Smith AG, Russell J, Feldman EL, Goldstein J, Peltier A, Smith S, Hamwi J, Pollari D, Bixby B,
Howard J, Singleton JR (2006) Lifestyle intervention for pre-diabetic neuropathy. Diabetes Care
29:1294–1299. https://doi.org/10.2337/dc06-0224
Smith AG, Singleton JR (2013) Obesity and hyperlipidemia are risk factors for early diabetic
neuropathy. J Diabetes Complications 27:436–442. https://doi.org/10.1016/j.jdiacomp.2013.
04.003
Smith EM, Pang H, Cirrincione C, Fleishman S, Paskett ED, Ahles T, Bressler LR, Fadul CE,
Knox C, Le-Lindqwister N, Gilman PB, Shapiro CL (2013) Effect of duloxetine on pain,
function, and quality of life among patients with chemotherapy-induced painful peripheral
neuropathy: a randomized clinical trial. JAMA 309:1359–1367. https://doi.org/10.1001/jama.
2013.2813
Chemotherapy-Induced Peripheral Neuropathy 335
Song TY, Chen CL, Liao JW, Ou HC, Tsai MS (2010) Ergothioneine protects against neuronal
injury induced by cisplatin both in vitro and in vivo. Food Chem Toxicol 48:3492–3499. https://
doi.org/10.1016/j.fct.2010.09.030
Stankovic JSK, Selakovic D, Mihailovic V, Rosic G (2020) Antioxidant supplementation in the
treatment of neurotoxicity induced by platinum-based chemotherapeutics – a review. Int J Mol
Sci 21. https://doi.org/10.3390/ijms21207753
Subramaniam S, O'Connor MJ, Masukawa LM, McGonigle P (1994) Polyamine effects on the
NMDA receptor in human brain. Exp Neurol 130:323–330. https://doi.org/10.1006/exnr.1994.
1210
Sucheston-Campbell LE, Clay-Gilmour AI, Barlow WE, Budd GT, Stram DO, Haiman CA,
Sheng X, Yan L, Zirpoli G, Yao S, Jiang C, Owzar K, Hershman D, Albain KS, Hayes DF,
Moore HC, Hobday TJ, Stewart JA, Rizvi A, Isaacs C, Salim M, Gralow JR, Hortobagyi GN,
Livingston RB, Kroetz DL, Ambrosone CB (2018) Genome-wide meta-analyses identifies
novel taxane-induced peripheral neuropathy-associated loci. Pharmacogenet Genomics 28:49–
55. https://doi.org/10.1097/fpc.0000000000000318
Sun Y, Gao HY, Fan ZY, He Y, Yan YX (2020) Metabolomics signatures in type 2 diabetes: a
systematic review and integrative analysis. J Clin Endocrinol Metab 105. https://doi.org/10.
1210/clinem/dgz240
Sun Y, Kim JH, Vangipuram K, Hayes DF, Smith EML, Yeomans L, Henry NL, Stringer KA,
Hertz DL (2018) Pharmacometabolomics reveals a role for histidine, phenylalanine, and
threonine in the development of paclitaxel-induced peripheral neuropathy. Breast Cancer Res
Treat 171:657–666. https://doi.org/10.1007/s10549-018-4862-3
Terrazzino S, Argyriou AA, Cargnin S, Antonacopoulou AG, Briani C, Bruna J, Velasco R,
Alberti P, Campagnolo M, Lonardi S, Cortinovis D, Cazzaniga M, Santos C, Kalofonos HP,
Canonico PL, Genazzani AA, Cavaletti G (2015) Genetic determinants of chronic oxaliplatin-
induced peripheral neurotoxicity: a genome-wide study replication and meta-analysis. J
Peripher Nerv Syst 20:15–23. https://doi.org/10.1111/jns.12110
Timmins HC, Mizrahi D, Li T, Kiernan MC, Goldstein D, Park SB (2021) Metabolic and lifestyle
risk factors for chemotherapy-induced peripheral neuropathy in taxane and platinum-treated
patients: a systematic review. J Cancer Surviv. https://doi.org/10.1007/s11764-021-00988-x
Traynelis SF, Hartley M, Heinemann SF (1995) Control of proton sensitivity of the NMDA receptor
by RNA splicing and polyamines. Science 268:873–876. https://doi.org/10.1126/science.
7754371
Tsukaguchi M, Shibano M, Matsuura A, Mukai S (2013) The protective effects of lafutidine for
bortezomib induced peripheral neuropathy. J Blood Med 4:81–85. https://doi.org/10.2147/jbm.
S44127
Umeda M, Fujita A, Nishiwaki H, Takeuchi K (1999) Effect of lafutidine, a novel histamine
H2-receptor antagonist, on monochloramine-induced gastric lesions in rats: role of capsaicin-
sensitive sensory neurons. J Gastroenterol Hepatol 14:859–865. https://doi.org/10.1046/j.
1440-1746.1999.01976.x
Urpi-Sarda M, Almanza-Aguilera E, Llorach R, Vázquez-Fresno R, Estruch R, Corella D, Sorli JV,
Carmona F, Sanchez-Pla A, Salas-Salvadó J, Andres-Lacueva C (2019) Non-targeted
metabolomic biomarkers and metabotypes of type 2 diabetes: a cross-sectional study of
PREDIMED trial participants. Diabetes Metab 45:167–174. https://doi.org/10.1016/j.diabet.
2018.02.006
Vázquez-Fresno R, Llorach R, Urpi-Sarda M, Lupianez-Barbero A, Estruch R, Corella D, Fitó M,
Arós F, Ruiz-Canela M, Salas-Salvadó J, Andres-Lacueva C (2015) Metabolomic pattern
analysis after mediterranean diet intervention in a nondiabetic population: a 1- and 3-year
follow-up in the PREDIMED study. J Proteome Res 14:531–540. https://doi.org/10.1021/
pr5007894
Verma P, Devaraj J, Skiles JL, Sajdyk T, Ho RH, Hutchinson R, Wells E, Li L, Renbarger J,
Cooper B, Ramkrishna D (2020) A metabolomics approach for early prediction of vincristine-
induced peripheral neuropathy. Sci Rep 10:9659. https://doi.org/10.1038/s41598-020-66815-y
336 C.-S. Chen and D. L. Hertz
Wallace MS, Rowbotham MC, Katz NP, Dworkin RH, Dotson RM, Galer BS, Rauck RL, Backonja
MM, Quessy SN, Meisner PD (2002) A randomized, double-blind, placebo-controlled trial of a
glycine antagonist in neuropathic pain. Neurology 59:1694–1700. https://doi.org/10.1212/01.
wnl.0000036273.98213.34
Wang J, Zhang XS, Tao R, Zhang J, Liu L, Jiang YH, Ma SH, Song LX, Xia LJ (2017)
Upregulation of CX3CL1 mediated by NF-κB activation in dorsal root ganglion contributes
to peripheral sensitization and chronic pain induced by oxaliplatin administration. Mol Pain 13:
1744806917726256. https://doi.org/10.1177/1744806917726256
Wang S, Kobayashi K, Kogure Y, Yamanaka H, Yamamoto S, Yagi H, Noguchi K, Dai Y (2018)
Negative regulation of TRPA1 by AMPK in primary sensory neurons as a potential mechanism
of painful diabetic neuropathy. Diabetes 67:98–109. https://doi.org/10.2337/db17-0503
Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP, McCabe E, Lewis GD, Fox CS, Jacques PF,
Fernandez C, O'Donnell CJ, Carr SA, Mootha VK, Florez JC, Souza A, Melander O, Clish CB,
Gerszten RE (2011) Metabolite profiles and the risk of developing diabetes. Nat Med 17:448–
453. https://doi.org/10.1038/nm.2307
Wei H, Viisanen H, You HJ, Pertovaara A (2016) Spinal histamine in attenuation of mechanical
hypersensitivity in the spinal nerve ligation-induced model of experimental neuropathy. Eur J
Pharmacol 772:1–10. https://doi.org/10.1016/j.ejphar.2015.12.039
Wiggin TD, Sullivan KA, Pop-Busui R, Amato A, Sima AA, Feldman EL (2009) Elevated
triglycerides correlate with progression of diabetic neuropathy. Diabetes 58:1634–1640.
https://doi.org/10.2337/db08-1771
Wilkins J, Sakrikar D, Petterson XM, Lanza IR, Trushina E (2019) A comprehensive protocol for
multiplatform metabolomics analysis in patient-derived skin fibroblasts. Metabolomics 15:83.
https://doi.org/10.1007/s11306-019-1544-z
Winkels RM, van Brakel L, van Baar H, Beelman RB, van Duijnhoven FJB, Geijsen A, van
Halteren HK, Hansson BME, Richie JP, Sun D, Wesselink E, van Zutphen M, Kampman E,
Kok DE (2020) Are ergothioneine levels in blood associated with chronic peripheral neuropathy
in colorectal cancer patients who underwent chemotherapy? Nutr Cancer 72:451–459. https://
doi.org/10.1080/01635581.2019.1637005
Winters-Stone KM, Horak F, Jacobs PG, Trubowitz P, Dieckmann NF, Stoyles S, Faithfull S (2017)
Falls, functioning, and disability among women with persistent symptoms of chemotherapy-
induced peripheral neuropathy. J Clin Oncol 35:2604–2612. https://doi.org/10.1200/jco.2016.
71.3552
Wishart DS (2016) Emerging applications of metabolomics in drug discovery and precision
medicine. Nat Rev Drug Discov 15:473–484. https://doi.org/10.1038/nrd.2016.32
Wishart DS (2019) Metabolomics for investigating physiological and pathophysiological processes.
Physiol Rev 99:1819–1875. https://doi.org/10.1152/physrev.00035.2018
Wu FZ, Xu WJ, Deng B, Liu SD, Deng C, Wu MY, Gao Y, Jia LQ (2018) Wen-Luo-Tong
decoction attenuates paclitaxel-induced peripheral neuropathy by regulating linoleic acid and
glycerophospholipid metabolism pathways. Front Pharmacol 9:956. https://doi.org/10.3389/
fphar.2018.00956
Xu X, Luo P, Wang Y, Cui Y, Miao L (2013) Nuclear factor (erythroid-derived 2)-like 2 (NFE2L2)
is a novel therapeutic target for diabetic complications. J Int Med Res 41:13–19. https://doi.org/
10.1177/0300060513477004
Yagihashi S, Matsunaga M (1979) Ultrastructural pathology of peripheral nerves in patients with
diabetic neuropathy. Tohoku J Exp Med 129:357–366. https://doi.org/10.1620/tjem.129.357
Yamamoto H, Horie S, Uchida M, Tsuchiya S, Murayama T, Watanabe K (2001) Effects of
vanilloid receptor agonists and antagonists on gastric antral ulcers in rats. Eur J Pharmacol
432:203–210. https://doi.org/10.1016/s0014-2999(01)01481-9
Yang Y, Luo L, Cai X, Fang Y, Wang J, Chen G, Yang J, Zhou Q, Sun X, Cheng X, Yan H, Lu W,
Hu C, Cao P (2018) Nrf2 inhibits oxaliplatin-induced peripheral neuropathy via protection of
mitochondrial function. Free Radic Biol Med 120:13–24. https://doi.org/10.1016/j.
freeradbiomed.2018.03.007
Chemotherapy-Induced Peripheral Neuropathy 337
Yehia R, Saleh S, El Abhar H, Saad AS, Schaalan M (2019) L-carnosine protects against
oxaliplatin-induced peripheral neuropathy in colorectal cancer patients: a perspective on
targeting Nrf-2 and NF-κB pathways. Toxicol Appl Pharmacol 365:41–50. https://doi.org/10.
1016/j.taap.2018.12.015
Yerra VG, Areti A, Kumar A (2017) Adenosine monophosphate-activated protein kinase abates
hyperglycaemia-induced neuronal injury in experimental models of diabetic neuropathy: effects
on mitochondrial biogenesis, autophagy and neuroinflammation. Mol Neurobiol 54:2301–2312.
https://doi.org/10.1007/s12035-016-9824-3
Yin X, Gibbons H, Rundle M, Frost G, McNulty BA, Nugent AP, Walton J, Flynn A, Gibney MJ,
Brennan L (2017) Estimation of chicken intake by adults using metabolomics-derived markers.
J Nutr 147:1850–1857. https://doi.org/10.3945/jn.117.252197
Yu J, Tang YY, Wang RR, Lou GD, Hu TT, Hou WW, Yue JX, Ohtsu H, Shi LY, Zhang SH, Chen
Z (2016) A critical time window for the analgesic effect of central histamine in the partial sciatic
ligation model of neuropathic pain. J Neuroinflammation 13:163. https://doi.org/10.1186/
s12974-016-0637-0
Zajączkowska R, Kocot-Kępska M, Leppert W, Wrzosek A, Mika J, Wordliczek J (2019)
Mechanisms of chemotherapy-induced peripheral neuropathy. Int J Mol Sci 20:1451. https://
doi.org/10.3390/ijms20061451
Zhang W, Ramautar R (2021) CE-MS for metabolomics: developments and applications in the
period 2018-2020. Electrophoresis 42:381–401. https://doi.org/10.1002/elps.202000203
Zhao XH, Wen ZM, Meredith CN, Matthews DE, Bier DM, Young VR (1986) Threonine kinetics
at graded threonine intakes in young men. Am J Clin Nutr 43:795–802. https://doi.org/10.1093/
ajcn/43.5.795
Zimmerman C, Atherton PJ, Pachman D, Seisler D, Wagner-Johnston N, Dakhil S, Lafky JM,
Qin R, Grothey A, Loprinzi CL (2016) MC11C4: a pilot randomized, placebo-controlled,
double-blind study of venlafaxine to prevent oxaliplatin-induced neuropathy. Support Care
Cancer 24:1071–1078. https://doi.org/10.1007/s00520-015-2876-5
Zirpoli GR, McCann SE, Sucheston-Campbell LE, Hershman DL, Ciupak G, Davis W, Unger JM,
Moore HCF, Stewart JA, Isaacs C, Hobday TJ, Salim M, Hortobagyi GN, Gralow JR, Budd GT,
Albain KS, Ambrosone CB (2017) Supplement use and chemotherapy-induced peripheral
neuropathy in a cooperative group trial (S0221): the DELCaP study. J Natl Cancer Inst 109.
https://doi.org/10.1093/jnci/djx098
Metabolomics of Respiratory Diseases
Contents
1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 340
2 Respiratory Diseases with Metabolomic Signatures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343
2.1 Asthma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343
2.2 Chronic Obstructive Pulmonary Disease (COPD) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 349
2.3 Pneumonia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 352
2.4 Acute Lung Injury/Acute Respiratory Distress Syndrome (ARDS) . . . . . . . . . . . . . . . . . 352
2.5 Occupational and Environmental Lung Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355
3 Metabolomics of Lung Microbiome in Respiratory Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 357
3.1 Asthma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 357
3.2 Chronic Obstructive Pulmonary Disease (COPD) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 358
4 Conclusive Remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 359
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 360
Abstract
Metabolomics is an expanding field of systems biology that is gaining significant
attention in respiratory research. As a unique approach to understanding and
diagnosing diseases, metabolomics provides a snapshot of all metabolites present
in biological samples such as exhaled breath condensate, bronchoalveolar lavage,
plasma, serum, urine, and other specimens that may be obtained from patients
with respiratory diseases. In this article, we review the rapidly expanding field of
metabolomics in its application to respiratory diseases, including asthma, chronic
obstructive pulmonary disease (COPD), pneumonia, and acute lung injury, along
with its more severe form, adult respiratory disease syndrome. We also discuss
# The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 339
V. Ghini et al. (eds.), Metabolomics and Its Impact on Health and Diseases,
Handbook of Experimental Pharmacology 277, https://doi.org/10.1007/164_2022_614
340 S. Moitra et al.
Keywords
Acute lung injury · Acute respiratory distress syndrome (ARDS) · Asthma ·
Chronic obstructive pulmonary disease (COPD) · Pneumonia
1 Introduction
body today. Today, the measurement of small molecules in human samples forms
the basis of clinical chemistry, established to assist health professionals in diagnosis
of illnesses.
Most clinical chemistry tests rely on the measurement of handful of metabolites,
and often these are only qualitative (positive or negative) rather than quantitative
tests. Because most metabolites measured using clinical tests are abundant and not
specific to any one disease, their detection must always be taken into consideration
with other clinical descriptors. Thus, the focus of clinical chemistry on such a small
group of metabolites is a significant limitation that prevents the applicability of
metabolite detection in the specific diagnosis of many diseases.
The limitations of traditional clinical chemistry highlight the advantages of
metabolomics. Recent improvements in the sensitivity and specificity of metabolite
detection using metabolomics have allowed the characterization and quantification
of complex metabolic profiles resulting in concurrent analysis of hundreds of
metabolites in a single sample. Metabolomics seeks to quantitatively assess complex
metabolic patterns in patient samples and is coupled with computational
technologies to allow the interpretation of data in the context of known metabolic
pathways. The complexity of the metabolome in a patient sample is further aug-
mented by the presence of metabolites that derive from the microbiome, which is
present in almost all samples obtained from the human body. The microbiome
generates metabolites that are unique to prokaryotic organisms and may be distin-
guished from the host’s own metabolome, thus providing another possible approach
for enhancing the diagnosis and prognosis of disease.
Despite substantial investments in genome analysis in diseases, genetic mutations
that result in the manifestation of disease are rare. Only 1–2% of disease risk for a
spectrum of conditions including asthma, chronic obstructive pulmonary disease
(COPD), and acute respiratory distress syndrome (ARDS) can be explained by
genetic mutations. Transcriptomic and proteomic analysis has generated more
insight into their potential as biomarkers, but these too have not developed into
standard disease indicators. In contrast, metabolomics and clinical chemistry repro-
ducibly demonstrate that metabolites are highly predictive for a large proportion of
complex diseases (Xia et al. 2013). Samples may be used from a broad range of
sources including saliva, nasal lavage, exhaled breath condensate (EBC), bronchial
washings, sweat, blood (plasma and serum), urine, feces, among others. Examples of
established metabolic biomarkers include glucose for diabetes, as mentioned above,
creatinine to detect kidney disease, cholesterol and triglycerides to evaluate the risk
of developing cardiovascular disease, uric acid for gout detection, and thyroxine for
hypo/hyperthyroidism. There are undoubtedly other metabolites that may be used to
serve as biomarkers in a range of diseases.
These findings indicate that the metabolome is a much more dynamic group of
analytes than the proteome, transcriptome, or genome, as it can change immediately
in response to environmental or physiological changes (Fig. 1) (Wishart 2005). To
appreciate the contribution that metabolomics may make to diagnosis of disease, it is
useful to compare the impact of environmental and physiological impact on proteins,
RNA, and genes. Environmental and physiological changes have negligible impact
342 S. Moitra et al.
Metabolomics
Metabolome
Environmental Influence
Physiological Influence
Proteomics
Proteome
Genomics
Genome
Metabolomics
Response
Proteomics
Response
Genomics
Time
Fig. 1 The systems biology pyramid and time scales of responses to environmental influences. (a)
Over 25,000 genes have been identified in human genomics, compared with a smaller number of
enzymes and even smaller number of metabolites. The responsiveness to physiological and
environmental insults of each of these components increases as we go from genomics to
metabolomics. (b) While metabolomics shows rapid changes in multiple metabolites in a short
period of time, proteomics shows smaller changes in abundance while genomics shows negligible
changes over the same period
on somatic gene expression, while some transcriptomic and proteomic changes have
been detected. In contrast, metabolomic changes in response to environmental and
physiological factors closely correlate with these events and can be altered within
seconds of exposure (Fig. 1b). Therefore, significant changes in metabolites may be
measurable in samples over far shorter time scales than by other systems biology
Metabolomics of Respiratory Diseases 343
2.1 Asthma
Fig. 2 Metabolites and metabolic pathways in childhood asthma. Heatmap of Spearman’s rank
correlation coefficients between metabolites associated with lowly and highly sensitized asthma (a)
and metabolic pathways of metabolites associated with atopic and non-atopic asthma (b). Red color
Metabolomics of Respiratory Diseases 345
correlation among specific metabolites with disease. Park et al. (2017) showed that
in severe asthma in children (≤15 years of age) exhibiting corticosteroid resistance,
tyrosine metabolism, degradation of aromatic compounds, and glutathione metabo-
lism were suggested to be significant pathways related to corticosteroid resistance
based on urine metabolites. A targeted LC-MS-based study for the presence of a
unique biomarker in asthmatic children showed a combination of 2-isopropylmalic
acid and betaine can classify children with asthma and controls. It was also shown in
the same study that asthmatics had lower relative concentrations of serum ascorbic
acid, 2-isopropylmalic acid, shikimate-3-phosphate, 6-phospho-D-gluconate, and
reduced glutathione. In the case of overweight children, niacin concentrations
were elevated in serum samples (Checkley et al. 2016). Loureiro et al. showed that
lipid peroxidation-related metabolites in urine samples are associated with asthma
severity and lung function, along with eosinophilic inflammation in nonobese
asthmatic patients (Loureiro et al. 2016). In other studies, it was shown that meta-
bolic pathways and pathway components like arginine, proline, taurine, hypotaurine,
glyoxylate, and dicarboxylate in serum and urine samples were closely related to
acute exacerbations of asthma as well as the choice of corticosteroid treatment
(Quan-Jun et al. 2017). It was also found that a set of 15 volatile carbon compounds
may discriminate between controlled and uncontrolled asthma and that 7 of these
compounds detected in exhaled breath samples could predict exacerbation within the
next 14 days with 88% sensitivity and 75% specificity (van Vliet et al. 2017).
Lipids have also been correlated with the diagnosis of asthma. Kang et al. showed
that certain metabolites, primarily lipid biomolecules in bronchoalveolar lavage
(BAL) fluid, could be markedly elevated in asthma compared to non-asthmatic
healthy individuals (Kang et al. 2014). This observation, supported by other studies
(Loureiro et al. 2016; Ghosh et al. 2020), indicates that lipid metabolism is altered in
asthma, potentially as a result of increased oxidative stress. Such altered lipid
metabolism was also associated with asthma severity, reduced lung function, and
higher eosinophilic inflammation in asthmatic individuals (Loureiro et al. 2016).
While asthma and obesity are known to share common systemic manifestations,
Maniscalco et al. showed that methane, pyruvate, and glyoxylate and dicarboxylate
metabolic pathways in EBC also greatly vary between obese and nonobese asthma
patients (Maniscalco et al. 2017), which indicates more complex crosstalk between
asthma and obesity than previously recognized.
Other recent studies have reported intriguing results of altered profiles of struc-
tural lipid molecules in asthma compared to healthy individuals (Kang et al. 2014;
Ghosh et al. 2020; Reinke et al. 2017; Pang et al. 2018; Jiang et al. 2021). Bian et al.
reported that some derivatives of serum arachidonic acid that serve as potential
mediators for allergic responses were significantly elevated in asthma (Bian et al.
⁄
Fig. 2 (continued) represents positive correlations; blue color represents negative correlations; red
arrow represents increase; blue arrow represents decrease. + symbol means a P-value < 0.05; +
+ symbol means a P-value < 0.01. [Reproduced from Chiu et al. (2021)]
346 S. Moitra et al.
Table 1 (continued)
Study population
Study (adult/children) Sample/method Summary of results
Pang et al. Adults Ultra-performance " Monosaccharides, LysoPC(o-18:
(2018) (29 asthmatics, liquid 0, 18:1), Retinyl ester, PC(18:1/2:
15 healthy) chromatography- 0), PC(16:0/18:1), arachidonic
tandem MS acid, PE(18:3/14:0) in asthma
(UPLC-MS) of # Glycerophosphocholine, PS(18:
serum 0/22:5), cholesterol glucuronide,
Phytosphingosine, Sphinganine,
LysoPC(p-18:1), retinols, PC(20:
4/16:1)
Jiang et al. Adults LC-MS/MS of " Phosphatidylethanolamine
(2021) (33 asthmatics, plasma (PE) (18:1p/22:6), PE (20:0/18:1),
28 healthy) PE (38:1), sphingomyelin
(SM) (d18:1/18:1), triglyceride
(TG) (16:0/16:0/18:1) in
asthmatics than healthy
# Phosphatidylinositol (PI) (16:0/
20:4), TG (17:0/18:1/18:1),
phosphatidylglycerol (PG) (44:0),
ceramide (d16:0/27:2), lysoPC
(22:4) in asthma
Chiu et al. Adults NMR of urine " Guanidoacetate
(2020, (30 asthmatics, # 1-Methylnicotinamide, allantoin
2018) 30 healthy)
Chiu et al. Children NMR of plasma " Lysine, isovalerate, histidine,
(2021) (28 asthmatics, tyrosine, glycine, citric acid,
25 healthy) ethanol, acetic acid, pyruvic acid in
asthma
Chang- Adults NMR of EBC " Lactate, formate, butyric acid,
Chien et al. (92 asthmatics, isobutyrate in asthma
(2021) 73 healthy)
Bian et al. Adults Ultra-high " 5(S)-Hydroxyeicosatetraenoic
(2017) (15 asthmatics and performance liquid acid (HETE), 8(S)-HETE, 11(S)-
15 healthy) chromatography HETE, 12(S)-HETE, 15(S)-HETE,
quadrupole time- 15(S)-Hydroxyeicosapentaenoic
of-flight (UHPLC)- acid (HEPE), prostaglandin
Q-TOF- MS of (PG)A2, PGB2, PGF1a, PGF2a,
serum PGJ2, 15-keto-PGF2a in asthma
compared to healthy
# Palmitic acid, Lauric acid in
asthma than healthy
Checkley Children Targeted liquid # Relative concentrations of serum
et al. (2016) (50 asthmatics and chromatography- ascorbic acid, 2-isopropylmalic
49 healthy between MS (LC-MS) of acid, shikimate-3-phosphate,
9 and 19 years) serum 6-phospho-D-gluconate, and
reduced glutathione in asthmatics
than healthy
(continued)
348 S. Moitra et al.
Table 1 (continued)
Study population
Study (adult/children) Sample/method Summary of results
Kelly et al. Children Targeted LC-MS • Metabolites (primarily
(2017) (380 asthmatics) (complementary glycerophospholipid, linoleic acid,
methods) of plasma and pyrimidine) were associated
with airway hyperreactivity, and
pre- and postbronchodilator FEV1/
FVC
• Distinct metabolites showed
moderate but important signatures
between disease severity
Tao et al. Children GC-MS of urine " Aspartic acid, Xanthosine,
(2019) (80 asthmatics, hypoxanthine,
29 healthy) N-acetylgalactosamine
# Stearic acid, Heptadecanoic acid,
uric acid, D-threitol
Li et al. Children GC-MS of urine " Azelaic acid, citraconic acid
(2020) (30 asthmatics, 4, D-altrose 1, D-erythro-
30 healthy) sphingosine 1, gentiobiose
2, 2-hydroxybutanoic acid,
L-allothreonine 1, leucine, stearic
acid, succinic acid, tyramine in
asthmatics than healthy
# 3,4-dihydroxycinnamic acid,
methionine 1, purine riboside,
malonic acid 1, cysteine, erythrose
1, lactamide 1, uric acid, valine in
asthma
Matysiak Children LC-MS/MS of " L-arginine, Β-alanine, Ƴ-amino-
et al. (2020) (13 asthmatics, blood N-butyric acid, L-histidine,
17 healthy) Hydroxy-L-proline in asthma
# D,L-Β-Aminoisobutyric acid,
taurine, L-tryptophan, L-valine in
asthma
Ferraro Children UPLC-MS of EBC " 9-amino-nonanoic acid,
et al. (2020) (26 asthmatics, 12-amino-dodecanoic acid, lactone
16 healthy) of PGF-MUM, N-linoleoyl taurine,
17-phenoxy trinor PGF2α ethyl
amide, lysoPC (18:2(9Z,12Z)) in
asthma
Van Vliet Children Targeted GC-TOF/ • 7 VOCs (3 aldehydes,
et al. (2017, (96 asthmatics) MS for VOCs in 1 hydrocarbon, 1 ketone,
2016) EBC 1 aromatic compound, and
1 unidentified VOC) in exhaled
breath could predict asthma
exacerbations
Metabolomics of Respiratory Diseases 349
2017). They described that some saturated fatty acids such as palmitic acid and lauric
acid levels were decreased in asthma. In addition, metabolites derived from protein
or carbohydrate metabolisms were found altered in asthma compared to
non-asthmatic healthy individuals in EBC, plasma, and urine (Chang-Chien et al.
2021; Chiu et al. 2018, 2020). However, in adults, the asthmatic response can be
caused, triggered, or aggravated by different risk factors such as allergy, environ-
mental exposures, active or passive smoking, and workplace conditions. Therefore,
more studies on different adult asthma phenotypes are required to better understand
those metabolic alterations.
On the other hand, asthma in children is mostly caused by allergic conditions or
genetic predisposition (such as parental atopy or asthma) and, to some extent,
pregnancy-related issues such as gestational smoking. In a group of asthmatic and
non-asthmatic children, Checkley et al. showed lowered relative concentrations of
serum ascorbic acid, reduced glutathione (GSH), and some carbohydrate derivatives
in asthma (Checkley et al. 2016). Kelly et al. further showed association between
certain plasma metabolites (glycerophospholipid, linoleic acid, and pyrimidine) and
airway hyperreactivity in asthmatic children (Kelly et al. 2017). They were able to
demonstrate moderate but clinically important signatures of distinct metabolites in
accordance with the disease severity (Kelly et al. 2017). Several other reports have
demonstrated distinct metabolomic profiles in asthmatic children compared to
non-asthmatic healthy individuals (Tao et al. 2019; Li et al. 2020; Matysiak et al.
2020; Chiu et al. 2021).
Recently, breath analysis has suggested some intriguing metabolic alterations in
asthma, particularly related to volatile organic compounds (VOCs) in the EBC that
could predict asthma exacerbations in children (van Vliet et al. 2017; Ferraro et al.
2020; Van Vliet et al. 2016). However, most of those analyses did not consider
potential risk factors or confounding factors as mentioned earlier. Therefore, clinical
correlations between metabolites and symptoms/severity are important to consider
while inferring those results into clinical practice.
COPD is a major lung disease worldwide that causes significant morbidity and
mortality and is among the top causes of death in many countries (Keogh and
Mark 2021). COPD is a chronic inflammatory disease of the lungs that is progressive
and irreversible in nature (Devine 2008). Although cigarette smoking is the most
common major risk factor for COPD, occupational or environmental insults are also
known to be prominent triggers for the onset and progression of this debilitating lung
condition.
Recent studies have demonstrated that COPD is a variable condition with multi-
modal phenotypic variants, particularly because of the differences in causal agents,
course, and progression of the disease. Although many metabolic alterations of
COPD were unknown until the beginning of the twenty-first century, these have
350 S. Moitra et al.
so far exhibited an intriguing panorama based on what has been discovered to date
(Fig. 3). In this section, we briefly describe some of the important findings from
metabolomic research in COPD.
Several studies have identified metabolites that are distinctive in COPD (Turano,
“NMR-based metabolomics to evaluate individual response to treatments”).
Novotna et al. (2018) examined 10 COPD patients and 10 healthy individuals and
observed that two amino acids, alanine and phenylalanine, were significantly lower
in the peripheral blood of COPD patients than healthy individuals, while
pyroglutamate level was higher in COPD patients. They also observed that the
free carnitine to acylcarnitine ratio was significantly lower in COPD patients than
the healthy individuals. Another report by Diao et al. (2019) further demonstrated
that COPD patients had reduced serum levels of creatine, glycine, histidine, and
threonine compared to non-COPD smokers. Although these findings indicate a
possible subclinical malnutrition in the context of respiratory disease, results are
still inconclusive regarding the association of these specific metabolites with COPD.
Body composition is greatly affected in COPD as the disease progresses (Schols
et al. 2005). Chronic bronchitis and emphysema are the two distinct phenotype
Metabolomics of Respiratory Diseases 351
variations of COPD and patients with these diseases have different body silhouettes,
presumably due to difference in lipid metabolic pathways. Some reports have
suggested that perturbation of lipid metabolism occurs in COPD (Chen et al. 2019;
Rafie et al. 2018). In the Subpopulations and Intermediate Outcomes in COPD Study
(SPIROMICS) cohort, Halper-Stromberg et al. (2019) observed that phosphatidyl-
ethanolamine, phosphatidylcholine, phosphatidylinositol, leucine, and lysine from
BAL fluid in COPD patients were associated with higher odds of developing
emphysema. Liang et al. (2019) identified that serum metabolites such as glutamine,
glycine, histidine, hypoxanthine, α-N-phenylacetyl-L-glutamine, L-pipecolic acid,
P-chlorophenylalanine, pseudouridine, and L-citrulline levels were markedly differ-
ent between asthma and COPD.
There is an increasing body of evidence suggesting that sphingolipids, which that
play crucial roles in the structure and function of plasma membranes and signal
transduction, also have roles in the pathogenesis of COPD, asthma, and other lung
conditions (Vlahos 2020). Lipidomic studies have shown that COPD patients have
higher plasma concentrations of very low density lipoprotein (VLDL) compared to
healthy individuals, which strongly correlates with higher central and peripheral
airway resistance (Rafie et al. 2018). Nambiar et al. (2021) found that blood
palmitoleic acid, linoleic acid, and dihydrotestosterone were lower in COPD patients
than healthy controls. Similarly, another study showed that the levels of serum
lysophosphatidylcholine (LPC) 18:3, lysophosphatidylethanolamine (LPE) 16:1,
and phosphatidylinositol (PI) 32:1 were markedly reduced in acute exacerbations
in COPD, thus highlighting the role of glycerophospholipids in the pathophysiology
of COPD (Gai et al. 2021).
Another recent report reiterated these findings in the context of disease onset
and stages in COPD where the authors observed that phosphatidylcholine and LPC
were key indicators of COPD onset and that phosphatidylserine and diacylglycerol
could potentially indicate the various COPD stages (Zhou et al. 2020). In line
with these observations, polyunsaturated acid metabolites were found to be
associated with reduced lung function and disease severity in COPD (Ran et al.
2019; Yu et al. 2019; Xue et al. 2020). Pinto-Plata et al. identified plasma lipid
metabolites that may predict survival differences in COPD patients (Pinto-Plata et al.
2019). Using the Karolinska COSMIC cohort, Naz et al. (2017) found that the
autotaxin-lysophosphatidic acid axis may be dysregulated due to oxidative stress
in COPD and that sex-regulated phenotypes are influential in the pathophysiology of
disease. However, despite several reports demonstrating associations between
metabolites with disease progression and severity in COPD, it is still not clear
whether these metabolites may influence pathophysiological mechanisms. Further-
more, there may be several residual confounders that influence the dysregulation of
metabolic pathways in disease conditions. Therefore, any interpretation and
conclusions made from these metabolic outcomes should be made cautiously (Kilk
et al. 2018).
352 S. Moitra et al.
2.3 Pneumonia
Acute lung injury and its more severe form, ARDS, is characterized by infiltration of
an inflammatory, fibrin-rich exudate into the pulmonary interstitium and alveolar
spaces (Gattinoni et al. 2014; Martin and Matute-Bello 2011; Ware and Matthay
Fig. 5 Metabolic profiles in mice infected with S. pneumoniae. An inbred strain of mice (C57BL/
6), maintained in specific virus antigen-free housing with autoclaved bedding and identical dietary
supplies, was infected intratracheally with a clinical isolate of S. pneumoniae, serotype 14. After
24 h of infection, bronchoalveolar lavage (BAL) samples were analyzed for cell counts (a) and
histology was carried out on lung sections (b) to confirm inflammation arising from infection. At the
same time, urine samples were collected from animals that were subjected to NMR analysis and a
PCA model of urinary metabolite concentrations was generated (c). Reprinted with permission from
Slupsky et al. (2009a) J. Proteome Res. 8(6):3029–3036. Copyright 2009 American Chemical
Society
2000; Li et al. 2011). This influx leads to impaired lung function and diminished gas
exchange (Ware and Matthay 2000). First described in 1967 by Ashbaugh et al.
(1967), ARDS is precipitated by many different causes, with the most common
being sepsis, pneumonia, severe trauma, and more recently, severe COVID-19
(Huang et al. 2020). ARDS is accompanied by an extraordinarily high mortality
rate (approximately 30% of patients die upon diagnosis of ARDS), and to date there
have been few effective pharmacotherapies for its treatment that mainly serve to
shorten the duration of illness rather than reverse it entirely. In addition, no effective
predictive or prognostic biomarkers are available to indicate the likelihood of a
patient developing ARDS. This has prompted a search for biomarkers of ARDS,
which has been led by genomics and proteomics, although neither field has yielded
suitable markers, and no candidate has progressed beyond the initial discovery phase
(Serkova et al. 2011; Rogers and Matthay 2014; Meyer 2013, 2014). This is likely
354 S. Moitra et al.
due to the heterogeneity of disease, and much of the variation could lie beyond the
proteome or genome, possibly in the metabolome (Serkova et al. 2011; Rogers and
Matthay 2014). Thus, metabolomics presents itself as a potentially valuable tool for
analysis in ARDS.
A challenge with understanding mechanisms associated with ARDS is that there
are no translational animal models that accurately mimic human disease (Martin and
Matute-Bello 2011; Matute-Bello et al. 2011; Matute-Bello and Downey 2013).
Despite this limitation, there have been several metabolomic studies carried out in
rodent models that demonstrate changes in metabolites (Stringer et al. 2016). In early
experimental models, mechanical ventilation-induced ARDS in rodents generated
metabolic profiles in serum, lung tissue, and BAL samples (Izquierdo-Garcia et al.
2014). Putative metabolites of ARDS were reported to be increased lactate and
decreased glucose and glycine in lung tissues, together with increased glucose,
lactate, acetate, 3-hydroxybutyrate, and creatine in BAL samples. NMR-detected
metabolites in lung samples were associated with markers of ARDS phenotype (peak
inspiratory pressure, PaO2, and lung histology), but there was no association
between these ARDS indices and serum metabolites. In one of the first studies
examining the metabolomics of experimental ARDS, a cytokine-induced lung injury
model was tested to determine the temporal association between inflammation in the
lungs and changes in lung metabolome (Serkova et al. 2008). Cytokine-induced lung
injury resulted in decreased ATP, energy balance, and energy charge levels,
suggesting a decreased energy state. Together with this there was a significant
increase in glycolytic activity, measured as elevated lactate-to-glucose levels that
normalized 24 h after the induction of injury. Collectively these findings indicate that
a shift in cell energy metabolism occurs in lung tissues in ARDS. The benefit of this
study was that it demonstrated an association between phenotypic and metabolic
changes, an important first step in biomarker discovery. To date, biomarkers have
not been found that can differentiate between the two extremes of mild interstitial
edema and extensive cellular injury in the spectrum of acute lung injury. However,
continued analysis by magnetic resonance imaging and metabolic NMR spectros-
copy may enhance the development of more robust and predictive longitudinal
processes of experimental lung injury. Other animal models have shown significant
metabolic shifts in ARDS induced by a variety of stimuli, reviewed in detail in
Stringer et al. (2016).
Few clinical studies have reported metabolomics analysis of patients with ARDS.
Several studies suggest that the use of BAL samples could provide insight into the
metabolomic profile associated with ARDS. In one study, at least 26 and 18 endoge-
nous metabolites, respectively, could be used to differentiate ARDS from healthy
BAL samples using liquid chromatography-MS analysis (Evans et al. 2014). These
included lactate and other energy metabolism-associated metabolites such as citrate,
creatine, and creatinine which are increased in the plasma of patients with ARDS
(Stringer et al. 2011). These findings demonstrate the utility of BAL as a biofluid for
metabolomics analysis.
Metabolomics of Respiratory Diseases 355
Occupational exposure is one of the major risk factors associated with respiratory
illnesses, and the incidence of occupational lung diseases is increasing due to
expanding populations and consumer needs (Moitra et al. 2015). According to the
report of the International Labour Organization, nearly two million people die each
year due to workplace accidents, of which over 30% die due to lung cancer or other
lung diseases as a result of workplace exposure (Cullinan et al. 2017). In many cases,
occupational lung diseases are improperly recorded or detected, often due to a lack
of causal evidence, all of which contributes to a significant underestimation of the
true burden of these diseases. Although several biomarker-related reports have been
published in the context of occupational exposure, metabolomic studies have been
very limited to date. We discuss some of the few studies below on occupational and
environmental lung diseases.
Among a group of workers employed in carbon-coating friction systems,
Maniscalco et al. (2018) found that the concentrations of VOCs, including
356 S. Moitra et al.
3.1 Asthma
The lung microbiota and metabolome are likely to play a pivotal role in the onset of
disease in the case of asthma (Barcik et al. 2020). It is now emerging that metaboli-
cally active microbiota that reside in the lung under normal conditions maintain a
complex network of crosstalk with the host in a symbiotic manner. In disease
conditions, however, this symbiosis is transformed into dysbiosis that can alter the
host immune response, which influences the overall lung health (Loverdos et al.
2019). The composition of normal lung microbiota consists of Bacteroidetes and
Firmicutes (the most abundant two genera), and apart from these two,
Proteobacteria, Actinobacteria, and Fusobacteria have also been found by 16S
rRNA sequencing in endobronchial brushing samples (Charlson et al. 2011; Bassis
et al. 2015). Although normal lung microbiota consists of a relatively small bacterial
population, estimated to be around 103 to 105/cm2, their intensely intricate crosstalk
is thought to be primarily responsible for the conduct of most of the host-microbiome
interplay (Charlson et al. 2011; Bassis et al. 2015; Hilty et al. 2010; Mathieu et al.
2018; Denner et al. 2016; Goleva et al. 2013).
In asthma, the bacterial pattern of the pulmonary microbiome has been
characterized in several studies. It is evident that some bacterial species become
elevated in nasopharyngeal swabs from asthmatics, such as Haemophilus influenzae,
Streptococcus pneumoniae, Staphylococcus aureus, and Moraxella catarrhalis,
compared to healthy controls. These bacteria are well-known pathogens that can
cause infectious exacerbations (Dickson et al. 2016). Interestingly, Huang et al.
showed in patients with severe asthma, Actinobacteria is present at high abundance
in correlation with elevated sputum leukocytes and eosinophils in bronchial biopsies
(Huang et al. 2015). It has been also shown that elevated eosinophil numbers in
lavage, along with reduced FEV1, correlate with bacterial α-diversity (based on
comparison of different species present in same sample) in endobronchial brushings
of asthmatic subjects. Bacterial species associated with lower airway obstruction
show distinctive features associated with FEV1 levels. For example, patients with
asthma exhibiting FEV1 < 60% had low α-diversity but high β-diversity compared
to asthma patients with FEV1 > 80% (Denner et al. 2016).
Interestingly, the gut microbiome is an important component of asthma patho-
physiology which has not been explored in detail. The human gut possesses a surface
area of 150–200 m2, which harbors 100,000 to 100 billion bacteria per mL of
sample, depending on the region of sample collection (Sender et al. 2016). A
relationship between the gut and lung was discovered upon the observation that
different lung diseases may be influenced by changes in the gut microflora and vice
versa. The microbiota in these two sites is therefore connected by a gut-lung axis that
is important in relation to asthma (Marsland et al. 2015). Among many different
metabolites produced by the gut microbiome, short-chain fatty acids (SCFAs), such
as acetate, propionate, and butyrate, have been found to regulate physiological and
immunological responses in humans. It is well known that not only do SCFAs
358 S. Moitra et al.
provide a source of energy, but they also function as signaling molecules. SCFAs
have been shown to have multiple signaling effects: they inhibit histone deacetylases
(HDACs) that increase cytokine gene expression by promoting an anti-inflammatory
cell phenotype to maintain homeostasis, suppress transcription factors (nuclear
factor κ-light-chain-enhancer of activated B cells (NF-κB)), and reduce tumor
necrosis factor-α (TNF-α) production (Durack et al. 2017; Chambers et al. 2018;
Tan et al. 2014). Depletion of SCFA-producing bacteria as a mechanistic link
between the microbiome and asthma susceptibility or severity has been suggested
by Cait et al. (2018). Hence, SCFAs derived from the metabolic activity of gut
microbiota inhibit proinflammatory responses in the lungs. Although the mechanism
underlying this pathway is unclear, the most likely mechanistic explanation is that
the hepatic system may weaken innate immune responses by SCFAs binding to G
protein-coupled receptors and/or inhibition of the mevalonate/isoprenoid pathway
through HMGCoA reductase (Young et al. 2016). The specific mediators that make
up the communication between gut and lung is still unclear, but it has been
speculated that gut epithelial cells and immune cells absorb signals from the
endothelium to form local cytokine microenvironments, and eventually this alters
the immune response in distal sites such as the lung (Budden et al. 2017).
Overall, these studies demonstrate that the gut and lung microbiome, and its
associated metabolome, have an enormous impact on patient outcomes in asthma.
Findings from these reports could contribute to the discovery of mechanisms and
novel biomarkers for asthma and its associated exacerbations.
Recent evidence suggests an association between the lung microbiome and COPD,
suggesting a contribution of the lung bacterial community to disease progression in
the form of dysbiosis (Hilty et al. 2010; Erb-Downward et al. 2011; Zakharkina et al.
2013; Pragman et al. 2012). Phylogenetic analysis of microbial populations in
samples collected from the oropharynx and bronchial brushings from COPD patients
and healthy controls showed increased populations of pathogenic Proteobacteria
(Haemophilus spp.) over Bacteroidetes (Prevotella spp.), with the latter being
especially reduced (Hilty et al. 2010). Other studies also demonstrated that healthy
individuals commonly exhibit higher populations of Firmicutes, Bacteroidetes,
Proteobacteria, Fusobacteria, and Actinobacteria, in contrast to pathogenic
Haemophilus, Streptococcus, Klebsiella, Pseudomonas, and Moraxella in COPD
patients (Wu et al. 2014; Murphy et al. 2005). In addition, several reports have
described that exposure to tobacco smoke can modify bacterial populations in the
mouth and lungs. Though studies are limited in the context of COPD, numerous
reports indicate an alteration of the oral and respiratory bacterial microbiome as an
effect of tobacco smoking (Morris et al. 2013; Zhang et al. 2018; Huang and Shi
2019). In COPD patients, commensal colonization of H. influenzae, S. pneumoniae,
Metabolomics of Respiratory Diseases 359
Pseudomonas, and Moraxella are frequently observed in the lungs (Simpson et al.
2016).
The gut-lung axis also features prominently in COPD (Young et al. 2016). Fecal
microbiota derived from COPD patients have been demonstrated to contribute to the
development of COPD in a mouse model (Li et al. 2021). The fecal microbiota of
COPD patients were found to contain lower levels of SCFAs, which could contribute
to the manifestation of COPD.
In another recent study comparing the metabolomic profiles of COPD patients
with healthy humans (Bowerman et al. 2020), it was found that COPD patients and
healthy individuals manifest significantly different sets of microbial and metabolic
signatures in fecal samples. As many as 146 bacterial species differ in between these
two groups, along with a group of the top 50 indicator metabolites that distinguished
between healthy and COPD individuals, consisting of mostly lipids (46%), amino
acids (20%), and xenobiotic compounds (20%). Hence, it can be deduced that the
intricate mechanisms associated with the gut-lung axis and the host’s microbial
community play a crucial role in the manifestation and progression of COPD.
4 Conclusive Remarks
References
Ashbaugh DG, Bigelow DB, Petty TL, Levine BE (1967) Acute respiratory distress in adults.
Lancet 2(7511):319–323
Barcik W, Boutin RCT, Sokolowska M, Finlay BB (2020) The role of lung and gut microbiota in
the pathology of asthma. Immunity 52(2):241–255
Bassis CM, Erb-Downward JR, Dickson RP, Freeman CM, Schmidt TM, Young VB, Beck JM,
Curtis JL, Huffnagle GB (2015) Analysis of the upper respiratory tract microbiotas as the source
of the lung and gastric microbiotas in healthy individuals. MBio 6(2):e00037
Bian X, Sun B, Zheng P, Li N, Wu JL (2017) Derivatization enhanced separation and sensitivity of
long chain-free fatty acids: application to asthma using targeted and non-targeted liquid
chromatography-mass spectrometry approach. Anal Chim Acta 989:59–70
Bos LDJ (2018) Diagnosis of acute respiratory distress syndrome by exhaled breath analysis. Ann
Transl Med 6(2):33
Bos LD, Weda H, Wang Y, Knobel HH, Nijsen TM, Vink TJ, Zwinderman AH, Sterk PJ, Schultz
MJ (2014) Exhaled breath metabolomics as a noninvasive diagnostic tool for acute respiratory
distress syndrome. Eur Respir J 44(1):188–197
Bowerman KL, Rehman SF, Vaughan A, Lachner N, Budden KF, Kim RY, Wood DLA, Gellatly
SL, Shukla SD, Wood LG, Yang IA et al (2020) Disease-associated gut microbiome and
metabolome changes in patients with chronic obstructive pulmonary disease. Nat Commun
11(1):5886
Budden KF, Gellatly SL, Wood DL, Cooper MA, Morrison M, Hugenholtz P, Hansbro PM (2017)
Emerging pathogenic links between microbiota and the gut-lung axis. Nat Rev Microbiol 15(1):
55–63
Cait A, Hughes MR, Antignano F, Cait J, Dimitriu PA, Maas KR, Reynolds LA, Hacker L, Mohr J,
Finlay BB, Zaph C et al (2018) Microbiome-driven allergic lung inflammation is ameliorated by
short-chain fatty acids. Mucosal Immunol 11(3):785–795
Chambers ES, Preston T, Frost G, Morrison DJ (2018) Role of gut microbiota-generated short-chain
fatty acids in metabolic and cardiovascular health. Curr Nutr Rep 7(4):198–206
Chang-Chien J, Huang HY, Tsai HJ, Lo CJ, Lin WC, Tseng YL, Wang SL, Ho HY, Cheng ML,
Yao TC (2021) Metabolomic differences of exhaled breath condensate among children with and
without asthma. Pediatr Allergy Immunol 32(2):264–272
Charlson ES, Bittinger K, Haas AR, Fitzgerald AS, Frank I, Yadav A, Bushman FD, Collman RG
(2011) Topographical continuity of bacterial populations in the healthy human respiratory tract.
Am J Respir Crit Care Med 184(8):957–963
Checkley W, Deza MP, Klawitter J, Romero KM, Klawitter J, Pollard SL, Wise RA, Christians U,
Hansel NN (2016) Identifying biomarkers for asthma diagnosis using targeted metabolomics
approaches. Respir Med 121(1532–3064 (Electronic)):59–66
Chen H, Li Z, Dong L, Wu Y, Shen H, Chen Z (2019) Lipid metabolism in chronic obstructive
pulmonary disease. Int J Chron Obstruct Pulmon Dis 14:1009–1018
Chiu CY, Lin G, Cheng ML, Chiang MH, Tsai MH, Su KW, Hua MC, Liao SL, Lai SH, Yao TC,
Yeh KW et al (2018) Longitudinal urinary metabolomic profiling reveals metabolites for asthma
development in early childhood. Pediatr Allergy Immunol 29(5):496–503
Chiu CY, Cheng ML, Chiang MH, Wang CJ, Tsai MH, Lin G (2020) Metabolomic analysis reveals
distinct profiles in the plasma and urine associated with IgE reactions in childhood asthma. J
Clin Med 9(3):887
Chiu CY, Cheng ML, Chiang MH, Wang CJ, Tsai MH, Lin G (2021) Integrated metabolic and
microbial analysis reveals host–microbial interactions in IgE-mediated childhood asthma. Sci
Rep 11(1):23407
Crowley G, Kwon S, Haider SH, Caraher EJ, Lam R, St-Jules DE, Liu M, Prezant DJ, Nolan A
(2018) Metabolomics of world trade center-lung injury: a machine learning approach. BMJ
Open Respir Res 5(1):e000274
Metabolomics of Respiratory Diseases 361
Huang C, Shi G (2019) Smoking and microbiome in oral, airway, gut and some systemic diseases. J
Transl Med 17(1):225
Huang YJ, Nariya S, Harris JM, Lynch SV, Choy DF, Arron JR, Boushey H (2015) The airway
microbiome in patients with severe asthma: associations with disease features and severity. J
Allergy Clin Immunol 136(4):874–884
Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, Cheng Z et al (2020)
Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet
395(10223):497–506
Izquierdo-Garcia JL, Naz S, Nin N, Rojas Y, Erazo M, Martinez-Caro L, Garcia A, de Paula M,
Fernandez-Segoviano P, Casals C, Esteban A et al (2014) A Metabolomic approach to the
pathogenesis of ventilator-induced lung injury. Anesthesiology 120(3):694–702
Jiang T, Dai L, Li P, Zhao J, Wang X, An L, Liu M, Wu S, Wang Y, Peng Y, Sun D et al (2021)
Lipid metabolism and identification of biomarkers in asthma by lipidomic analysis. Biochim
Biophys Acta Mol Cell Biol Lipids 1866(2):158853
Kang YP, Lee WJ, Hong JY, Lee SB, Park JH, Kim D, Park S, Park CS, Park SW, Kwon SW
(2014) Novel approach for analysis of bronchoalveolar lavage fluid (BALF) using HPLC-
QTOF-MS-based lipidomics: lipid levels in asthmatics and corticosteroid-treated asthmatic
patients. J Proteome Res 13(9):3919–3929
Kelly RS, Virkud Y, Giorgio R, Celedon JC, Weiss ST, Lasky-Su J (2017) Metabolomic profiling
of lung function in Costa-Rican children with asthma. Biochim Biophys Acta Mol basis Dis
1863(6):1590–1595
Keogh E, Mark WE (2021) Managing malnutrition in COPD: a review. Respir Med 176:106248
Kilk K, Aug A, Ottas A, Soomets U, Altraja S, Altraja A (2018) Phenotyping of chronic obstructive
pulmonary disease based on the integration of metabolomes and clinical characteristics. Int J
Mol Sci 19(3):666
Kwon S, Lee M, Crowley G, Schwartz T, Zeig-Owens R, Prezant DJ, Liu M, Nolan A (2021)
Dynamic metabolic risk profiling of world trade center lung disease: a longitudinal cohort study.
Am J Respir Crit Care Med 204(9):1035–1047
Lacy P, McKay RT, Finkel M, Karnovsky A, Woehler S, Lewis MJ, Chang D, Stringer KA (2014)
Signal intensities derived from different NMR probes and parameters contribute to variations in
quantification of metabolites. PLoS One 9(1):e85732
Li G, Malinchoc M, Cartin-Ceba R, Venkata CV, Kor DJ, Peters SG, Hubmayr RD, Gajic O (2011)
Eight-year trend of acute respiratory distress syndrome: a population-based study in Olmsted
County, Minnesota. Am J Respir Crit Care Med 183(1):59–66
Li S, Liu J, Zhou J, Wang Y, Jin F, Chen X, Yang J, Chen Z (2020) Urinary Metabolomic profiling
reveals biological pathways and predictive signatures associated with childhood asthma. J
Asthma Allergy 13:713–724
Li N, Dai Z, Wang Z, Deng Z, Zhang J, Pu J, Cao W, Pan T, Zhou Y, Yang Z, Li J et al (2021) Gut
microbiota dysbiosis contributes to the development of chronic obstructive pulmonary disease.
Respir Res 22(1):274
Liang Y, Gai XY, Chang C, Zhang X, Wang J, Li TT (2019) Metabolomic profiling differences
among asthma, COPD, and healthy subjects: a LC-MS-based metabolomic analysis. Biomed
Environ Sci 32(9):659–672
Loureiro CC, Oliveira AS, Santos M, Rudnitskaya A, Todo-Bom A, Bousquet J, Rocha SM (2016)
Urinary metabolomic profiling of asthmatics can be related to clinical characteristics. Allergy
71(9):1362–1365
Loverdos K, Bellos G, Kokolatou L, Vasileiadis I, Giamarellos E, Pecchiari M, Koulouris N,
Koutsoukou A, Rovina N (2019) Lung microbiome in asthma: current perspectives. J Clin Med
8(11):1967
Madsen R, Lundstedt T, Trygg J (2010) Chemometrics in metabolomics – a review in human
disease diagnosis. Anal Chim Acta 659(1–2):23–33
Maniscalco M, Paris D, Melck DJ, D'Amato M, Zedda A, Sofia M, Stellato C, Motta A (2017)
Coexistence of obesity and asthma determines a distinct respiratory metabolic phenotype. J
Allergy Clin Immunol 139(5):1536–1547
Metabolomics of Respiratory Diseases 363
Pragman AA, Kim HB, Reilly CS, Wendt C, Isaacson RE (2012) The lung microbiome in moderate
and severe chronic obstructive pulmonary disease. PLoS One 7(10):e47305
Quan-Jun Y, Jian-Ping Z, Jian-Hua Z, Yong-Long H, Bo X, Jing-Xian Z, Bona D, Yuan Z, Cheng
G (2017) Distinct metabolic profile of inhaled budesonide and salbutamol in asthmatic children
during acute exacerbation. Basic Clin Pharmacol Toxicol 120(3):303–311
Rafie S, Moitra S, Brashier BB (2018) Association between the serum metabolic profile and lung
function in chronic obstructive pulmonary disease. Turk Thorac J 19(1):13–18
Rahman I (2003) Oxidative stress, chromatin remodeling and gene transcription in inflammation
and chronic lung diseases. J Biochem Mol Biol 36(1):95–109
Ran N, Pang Z, Gu Y, Pan H, Zuo X, Guan X, Yuan Y, Wang Z, Guo Y, Cui Z, Wang F (2019) An
updated overview of Metabolomic profile changes in chronic obstructive pulmonary disease.
Meta 9(6):111
Reinke SN, Gallart-Ayala H, Gomez C, Checa A, Fauland A, Naz S, Kamleh MA, Djukanovic R,
Hinks TS, Wheelock CE (2017) Metabolomics analysis identifies different metabotypes of
asthma severity. Eur Respir J 49(3):1601740
Riely CA, Cohen G, Lieberman M (1974) Ethane evolution: a new index of lipid peroxidation.
Science 183(4121):208–210
Robertson DG, Watkins PB, Reily MD (2011) Metabolomics in toxicology: preclinical and clinical
applications. Toxicol Sci 120:S146–S170
Rogers AJ, Matthay MA (2014) Applying metabolomics to uncover novel biology in ARDS. Am J
Physiol Lung Cell Mol Physiol 306(11):L957–L961
Schols AM, Broekhuizen R, Weling-Scheepers CA, Wouters EF (2005) Body composition and
mortality in chronic obstructive pulmonary disease. Am J Clin Nutr 82(1):53–59
Schubert JK, Muller WP, Benzing A, Geiger K (1998) Application of a new method for analysis of
exhaled gas in critically ill patients. Intensive Care Med 24(5):415–421
Sender R, Fuchs S, Milo R (2016) Are we really vastly outnumbered? Revisiting the ratio of
bacterial to host cells in humans. Cell 164(3):337–340
Serkova NJ, Van Rheen Z, Tobias M, Pitzer JE, Wilkinson JE, Stringer KA (2008) Utility of
magnetic resonance imaging and nuclear magnetic resonance-based metabolomics for quantifi-
cation of inflammatory lung injury. Am J Physiol Lung Cell Mol Physiol 295(1):L152–L161
Serkova NJ, Standiford TJ, Stringer KA (2011) The emerging field of quantitative blood
metabolomics for biomarker discovery in critical illnesses. Am J Respir Crit Care Med
184(6):647–655
Simpson JL, Baines KJ, Horvat JC, Essilfie AT, Brown AC, Tooze M, McDonald VM, Gibson PG,
Hansbro PM (2016) COPD is characterized by increased detection of Haemophilus influenzae,
Streptococcus pneumoniae and a deficiency of Bacillus species. Respirology 21(4):697–704
Slupsky CM, Cheypesh A, Chao DV, Fu H, Rankin KN, Marrie TJ, Lacy P (2009a) Streptococcus
pneumoniae and Staphylococcus aureus pneumonia induce distinct metabolic responses. J
Proteome Res 8(6):3029–3036
Slupsky CM, Rankin KN, Fu H, Chang D, Rowe BH, Charles PG, McGeer A, Low D, Long R,
Kunimoto D, Sawyer MB et al (2009b) Pneumococcal pneumonia: potential for diagnosis
through a urinary metabolic profile. J Proteome Res 8(12):5550–5558
Stringer KA, Serkova NJ, Karnovsky A, Guire K, Paine R 3rd, Standiford TJ (2011) Metabolic
consequences of sepsis-induced acute lung injury revealed by plasma (1)H-nuclear magnetic
resonance quantitative metabolomics and computational analysis. Am J Physiol Lung Cell Mol
Physiol 300(1):L4–L11
Stringer KA, McKay RT, Karnovsky A, Quemerais B, Lacy P (2016) Metabolomics and its
application to acute lung diseases. Front Immunol 7:44
Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR, Macia L (2014) Chapter three – the
role of short-chain fatty acids in health and disease. In: Alt FW (ed) Advances in immunology,
vol 121. Academic Press, pp 91–119
Tao JL, Chen YZ, Dai QG, Tian M, Wang SC, Shan JJ, Ji JJ, Lin LL, Li WW, Yuan B (2019) Urine
metabolic profiles in paediatric asthma. Respirology 24(6):572–581
Metabolomics of Respiratory Diseases 365
Van Vliet D, Smolinska A, Jobsis Q, Rosias PP, Muris JW, Dallinga JW, van Schooten FJ,
Dompeling E (2016) Association between exhaled inflammatory markers and asthma control
in children. J Breath Res 10(1):016014
van Vliet D, Smolinska A, Jobsis Q, Rosias P, Muris J, Dallinga J, Dompeling E, van Schooten FJ
(2017) Can exhaled volatile organic compounds predict asthma exacerbations in children? J
Breath Res 11(1):016016
Veerappan A, Oskuei A, Crowley G, Mikhail M, Ostrofsky D, Gironda Z, Vaidyanathan S,
Wadghiri YZ, Liu M, Kwon S, Nolan A (2020) World trade center-cardiorespiratory and
vascular dysfunction: assessing the phenotype and metabolome of a murine particulate matter
exposure model. Sci Rep 10(1):3130
Vlahos R (2020) Lipids in chronic obstructive pulmonary disease: a target for future therapy? Am J
Respir Cell Mol Biol 62(3):273–274
Vos T, Lim SS, Abbafati C, Abbas KM, Abbasi M, Abbasifard M, Abbasi-Kangevari M,
Abbastabar H, Abd-Allah F, Abdelalim A, Abdollahi M et al (2020) Global burden of
369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis
for the global burden of disease study 2019. Lancet 396(10258):1204–1222
Ware LB, Matthay MA (2000) The acute respiratory distress syndrome. N Engl J Med 342(18):
1334–1349
Wei W, Wu X, Bai Y, Li G, Meng H, Feng Y, Li H, Li M, Guan X, Fu M, Wang C et al (2021)
Arsenic exposure and its joint effects with cigarette smoking and physical exercise on lung
function impairment: evidence from an occupational cohort study. Environ Res 196:110419
Wheelock CE, Goss VM, Balgoma D, Nicholas B, Brandsma J, Skipp PJ, Snowden S, Burg D,
D'Amico A, Horvath I, Chaiboonchoe A et al (2013) Application of 'omics technologies to
biomarker discovery in inflammatory lung diseases. Eur Respir J 42(3):802–825
Wishart DS (2005) Metabolomics: the principles and potential applications to transplantation. Am J
Transplant 5(12):2814–2820
Wu D, Hou C, Li Y, Zhao Z, Liu J, Lu X, Shang X, Xin Y (2014) Analysis of the bacterial
community in chronic obstructive pulmonary disease sputum samples by denaturing gradient
gel electrophoresis and real-time PCR. BMC Pulm Med 14(1):179
Xia J, Broadhurst DI, Wilson M, Wishart DS (2013) Translational biomarker discovery in clinical
metabolomics: an introductory tutorial. Metabolomics 9(2):280–299
Xue M, Cai C, Guan L, Xu Y, Lin J, Zeng Y, Hu H, Chen R, Wang H, Zhou L, Sun B (2020)
Exploration of n-6 and n-3 polyunsaturated fatty acids metabolites associated with nutritional
levels in patients with severe stable chronic obstructive pulmonary disease. Int J Chron Obstruct
Pulmon Dis 15:1633–1642
Young RP, Hopkins RJ, Marsland B (2016) The gut-liver-lung axis. Modulation of the innate
immune response and its possible role in chronic obstructive pulmonary disease. Am J Respir
Cell Mol Biol 54(2):161–169
Yu B, Flexeder C, McGarrah RW 3rd, Wyss A, Morrison AC, North KE, Boerwinkle E,
Kastenmuller G, Gieger C, Suhre K, Karrasch S et al (2019) Metabolomics identifies novel
blood biomarkers of pulmonary function and COPD in the general population. Meta 9(4):61
Zakharkina T, Heinzel E, Koczulla RA, Greulich T, Rentz K, Pauling JK, Baumbach J,
Herrmann M, Grunewald C, Dienemann H, von Muller L et al (2013) Analysis of the airway
microbiota of healthy individuals and patients with chronic obstructive pulmonary disease by
T-RFLP and clone sequencing. PLoS One 8(7):e68302
Zhang R, Chen L, Cao L, Li KJ, Huang Y, Luan XQ, Li G (2018) Effects of smoking on the lower
respiratory tract microbiome in mice. Respir Res 19(1):253
Zhou J, Li Q, Liu C, Pang R, Yin Y (2020) Plasma metabolomics and Lipidomics reveal perturbed
metabolites in different disease stages of chronic obstructive pulmonary disease. Int J Chron
Obstruct Pulmon Dis 15:553–565
The Metabolomics of Critical Illness
Contents
1Introduction: A Potential Role for Metabolomics in Understanding Critical Illness . . . . . 368
2Metabolic Alterations in Critical Illness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369
3Endocrine Drivers of Altered Metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369
4Adrenergic Mediators and Their Changes in Critical Illness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 371
5Inflammatory Mediators (Cytokines) and Their Contribution to Metabolism . . . . . . . . . . . . 371
6Metabolic Phases During Critical Illness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 371
7Autophagy and Mitophagy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 372
8Nutrient Changes During Critical Illness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 373
8.1 Lactate: The Original Metabolomic Biomarker . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 374
9 Metabolomics in Sepsis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 375
9.1 Diagnosis of Sepsis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 375
9.2 Prognostication in Sepsis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 376
9.3 Future Directions: Using Metabolomics to Guide Treatment . . . . . . . . . . . . . . . . . . . . . . . 377
10 Metabolomics Studies in ARDS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 377
10.1 Blood Metabolomics of ARDS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378
10.2 Using Blood Metabolomics to Identify High-Risk Subsets of ARDS Cohorts . . . 379
10.3 Pulmonary Specific Samples for Metabolomic Profiling . . . . . . . . . . . . . . . . . . . . . . . . . . 379
11 Metabolomics of Critical Illness: Future Directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 380
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 381
Abstract
Critical illness is associated with dramatic changes in metabolism driven by
immune, endocrine, and adrenergic mediators. These changes involve early
activation of catabolic processes leading to increased energetic substrate avail-
ability; later on, they are followed by a hypometabolic phase characterized by
# The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 367
V. Ghini et al. (eds.), Metabolomics and Its Impact on Health and Diseases,
Handbook of Experimental Pharmacology 277, https://doi.org/10.1007/164_2022_622
368 A. E. Pacheco-Navarro and A. J. Rogers
Keywords
Acute respiratory distress syndrome (ARDS) · Metabolism · Metabolomics ·
Prognostication · Sepsis
Each year, over four million critically ill Americans are admitted into intensive care
units (ICUs) around the country, a number that has only increased since the start of
the COVID-19 pandemic (ICU Outcomes 2022). Despite advances in medicine, ICU
mortality remains high, ranging between 8 and 20%, with approximately 500,000
American deaths per year. In particular, sepsis – a systemic inflammatory illness
caused by infection with a dysregulated host immune response – and the acute
respiratory distress syndrome (ARDS) – a syndrome of respiratory failure with a
diverse range of etiologies – are particularly challenging illnesses in the ICU, with
mortality upward of 40% (Fleischmann et al. 2016; Gaieski et al. 2013; Rudd et al.
2020).
Part of the challenge in treating these syndromes lies in their significant biological
heterogeneity. The majority of clinical trials in sepsis and ARDS have been negative,
and available treatment remains largely supportive and directed at treating underly-
ing infections, while not specifically addressing the inflammatory state. Recent
re-analyses of a number of negative trials have revealed that subgroups of patients
appear to respond to the intended therapy (Calfee et al. 2014; Puskarich et al. 2021),
underscoring the heterogeneity of these syndromes and the need for better
diagnostics in identifying high-risk patients and matching them with potential
treatments.
Given the dramatic and rapid metabolic alterations that are observed during
critical illness, metabolomics offers a powerful potential tool for better identifying
subphenotypes of critically ill patients who may differentially respond to treatments
and also identifying novel treatment pathways.
The Metabolomics of Critical Illness 369
In health, energetic needs are largely met by nutrient intake through food, rather than
catabolism of the body’s macromolecules. ATP, the main source of cellular energy,
is derived by aerobic metabolism, and anabolic and catabolic processes are in
balance.
In critical illness, however, dramatic changes in the endocrine and autonomic
nervous system alter these metabolic processes, shifting them toward a catabolic
state in which the body’s carbohydrates, lipids, and protein stores are used to meet
increased energetic needs. This imbalance is often further driven by reduced caloric
intake, as critically ill patients frequently suffer from symptoms like encephalopathy
and anorexia that curb eating. Clinically, such catabolism leads to skeletal muscle
wasting, weakness, and prolonged recovery (Casaer and Van den Berghe 2014).
Initially, acute stress such as that caused by sepsis and other forms of critical illness
leads to an inflammatory cascade which includes the release of cytokines, including
IL-1, IL-2, and IL-6 that can activate the hypothalamic-pituitary-adrenal axis. This
can occur through direct cytokine production by glial cells in the brain (Khardori and
Castillo 2012; Englert and Rogers 2016), and through free diffusion into the pitui-
tary, which is outside the blood brain barrier. This cytokine-derived activation
results in the release of corticotropin (ACTH) from the pituitary, which stimulates
production and secretion of cortisol from the adrenal glands (Englert and Rogers
2016; Cooper 2003). In addition to its increased production by the adrenal gland,
decreases in corticosteroid binding globulin during critical illness and impaired
hepatic metabolism of the hormone contribute to increased levels of free, biologi-
cally active cortisol (Wasyluk et al. 2021; Marik et al. 2008).
Cortisol acts as the primary endocrine mediator in sepsis, leading to a variety of
changes in metabolism, immune function, and cardiovascular function critical to
maintaining homeostasis (Fig. 1). Metabolically, it leads to increased blood glucose
through a variety of mechanisms: by activation of enzymes involved in gluconeo-
genesis, increasing insulin resistance, and decreasing glucose uptake by skeletal
muscle (Marik et al. 2008). It activates lipolysis in adipose tissue, leading the release
of free fatty acids (Marik et al. 2008).
In addition to increasing energy availability by increasing glucose and fatty acid
concentrations, cortisol has a major role in increasing blood pressure. In vascular
smooth muscle, it increases transcription of surface receptors for catecholamines and
angiotensin II, resulting in increased sensitivity to these vasoactive hormones.
Cortisol also plays a complex role in regulating and abrogating the immune
response, acting in a negative feedback loop that both reduces the number of
circulating immune cells and decreases their production of cytokines.
370 A. E. Pacheco-Navarro and A. J. Rogers
While increased cortisol synthesis and secretion is part of the body’s typical
response to stress, this response can be attenuated or halted in a variety of conditions
in the setting of critical illness. Head injuries, central nervous system depressants,
and pituitary infarction can inhibit the CRH and corticotropin driven portion of the
hypothalamic-pituitary-adrenal axis (Cooper 2003). At the level of the adrenal
glands, drugs such as the induction agent etomidate – frequently used during
intubation – can impair cortisol synthesis, and adrenal hemorrhage, though rare,
can occur in septic patients with coagulopathies (Cooper 2003). More commonly,
chronic intake of exogenous corticosteroids – e.g., in patients with rheumatologic
disease – can lead to adrenal atrophy and a sluggish or absent response to
corticotropin.
While absolute adrenal insufficiency is rare in critically ill patients, studies have
shown that relative adrenal insufficiency – as defined by inadequate increase in
cortisol level in response to a corticotropin challenge – is common in patients with
septic shock (Annane et al. 2002). In 2007, a consensus statement between the
Society of Critical Care Medicine and the European Society of Intensive Care
Medicine first coined the term “CIRCI” (critical illness-related corticosteroid insuf-
ficiency) to recognize that many critically ill patients have an impaired
hypothalamic-pituitary-adrenal response.
Despite this, it is unclear whether such patients benefit from hormone replace-
ment (Annane et al. 2002; Sprung et al. 2008), as a number of large randomized
controlled trials studying the use of steroids in sepsis have had conflicting results
(Annane et al. 2002, 2018; Sprung et al. 2008; Venkatesh et al. 2018). At the
The Metabolomics of Critical Illness 371
minimum, however, these studies suggest that steroid administration in critically ill
patients is safe and should be considered, particularly for patients with refractory
shock. As in the sepsis literature, the benefits of steroids are unclear in ARDS, but
recent high-quality studies (Villar et al. 2020) – as well as the evidence in severe
COVID-19 (RECOVERY Collaborative Group et al. 2021; Tomazini et al. 2020) –
suggest that they may be beneficial.
The sympathetic nervous system is rapidly activated in the setting of critical illness.
These stress signals can occur at the level of the peripheral nerves (e.g., in the case of
trauma), by chemoreceptors (in the case of hypoxemia or hypercapnia), or by
baroreceptors (in the setting of shock). These events all result in the release of
norepinephrine from post-ganglionic neurons, and secretion of epinephrine and
norepinephrine from chromaffin cells in the adrenal medulla (Preiser et al. 2014).
Along with exogenously delivered catecholamines that may be administered
therapeutically, endogenous dopamine, norepinephrine, and epinephrine play
major roles in activating catabolic pathways and increasing nutrient availability.
Like cortisol, these hormones work to increase plasma glucose concentrations by
upregulating hepatic gluconeogenesis and glycogenolysis and increasing insulin
resistance.
Sepsis and other forms of critical illness are characterized by an early stress response,
as described above, associated with increased catabolism and energy availability.
This early phase is often followed by a period of decreased metabolic activity and
organ dysfunction (Singer et al. 2004; Pool et al. 2018). Initially, the organ dysfunc-
tion and elevations in lactate that are frequently seen in sepsis were thought to result
from impaired tissue perfusion, but studies have since shown that although
malperfusion may play a role in the early phase of critical illness, tissue oxygen
372 A. E. Pacheco-Navarro and A. J. Rogers
delivery is adequate in the later phases of sepsis (Sair et al. 2001). Instead, cellular
changes in metabolism – decreased oxidative phosphorylation and greater reliance
on glycolytic pathways – termed cytopathic hypoxia – likely underly this organ
dysfunction. These changes are hypothesized to be adaptive mechanisms to preserve
energy for critical functions, prevent DNA damage, and limit additional injury.
In the heart, for example, sepsis and critical illness can result in reduced cardiac
myocyte contractility through a process called myocardial hibernation, which
preserves myocyte ATP levels at the cost of reduced function. In animal models,
this change in contractility has been linked to inhibition of the oxidative phosphory-
lation enzyme cytochrome oxygenase, and this phenotype can be rescued by deliv-
ery of exogenous cytochrome c (Piel et al. 2007). Similar mitochondrial changes
have also been implicated in animal models of sepsis-induced dysfunction of the
lung, kidney, and liver (Pool et al. 2018). In the kidney, the presence of
pro-inflammatory cytokines or lipopolysaccharide from bacteria leads to
downregulation of energy-intensive sodium and chloride channels (Schmidt et al.
2007). Despite these observations, the full spectrum of metabolic changes during the
phases of critical illness is not known. Metabolomics provides a potential avenue
through which changes in multiple metabolic pathways can be better described.
Plasma lactate is one of the most validated and frequently used biomarkers in the
intensive care unit. Lactate is a by-product of anerobic metabolic pathways, in which
glucose is broken down into two pyruvate molecules and then fermented to 2 lactate
molecules in a net reaction that produces 2 molecules of adenosine triphosphate
(ATP) and regenerates the NAD+ cofactor used in glycolysis (Fig. 2). Accordingly,
rising blood lactate levels can indicate inadequate tissue perfusion and oxygenation
or a perfusion-independent shift toward anerobic pathways as can occur in malig-
nancy (the Warburg effect) or in the later phases of sepsis.
In the ICU, lactate can be used for prognostication and also to guide resuscitation
during critical illness. From a risk stratification perspective, hospitalized patients
with infection-related admissions had a significantly higher risk of death if their
serum lactate was >4 mmol/L (as compared to those with normal concentrations),
and even modestly elevated lactates of 2.5–4 mmol were associated with increased
mortality (Shapiro et al. 2005). Similarly, in a study of septic patients in the
emergency department, intermediate and high lactate levels (2.5–3.9 mmol/L and
>4 mmol/L) were associated with increased mortality even in patients with initially
normal blood pressures and without evidence of organ dysfunction (Mikkelsen et al.
2009).
Lactate levels have also been applied to the goal-directed paradigm of sepsis
management and have been used to guide resuscitation, though its role in this
context has been less well-established. In the EMShockNet trial which compared
the use of ScvO2 to serial lactates in the goal-directed resuscitation of patients
(in which both groups also had MAP and CVP goals), there was a trend toward
reduced mortality in the lactate group, but this difference was not statistically
significant (Jones et al. 2010). Similarly, in another trial comparing serial lactate-
guided therapy to traditional early goal-directed therapy as described by Rivers et al.,
mortality trended lower in the lactate group, but was just outside of the predefined
alpha for statistical significance ( p = 0.067) (Jansen et al. 2010).
9 Metabolomics in Sepsis
Sepsis has a wide range of outcomes, from full recovery to long-term disability or
death (Singer et al. 2016). As such, it is not surprising that many studies in critical
care metabolomics are focused on prognostication. In these studies, septic patients
with worse outcomes (death or multiorgan failure) are compared to survivors or
individuals with less organ failure.
Using targeted metabolomic profiling of more than 300 metabolites, Langley
et al. evaluated the plasma metabolomes and proteomes in two independent cohorts
(Langley et al. 2014). The group used 150 adult patients from the Community
Acquired Pneumonia and Sepsis Outcome Diagnostics (CAPSOD) study, which
enrolled patients who presented to the emergency department of three US hospitals
with symptoms of sepsis (Langley et al. 2013) as a discovery cohort, and used
patients with sepsis from the Brigham and Women’s Hospital Registry of Critical
Illness (RoCI) cohort as a validation set. They found that the metabolic signatures of
survivors differed markedly from those that died. In particular, levels of proteins
involved in fatty acid transport and β-oxidation, gluconeogenesis, and the citric acid
cycle were significantly higher in those that died. The group used Support Vector
Machine analysis to create an algorithm using two clinical features and five metabo-
lite levels to predict survival in sepsis, using the RoCI data as a validation set.
Though the RoCI cohort had higher mortality and more cancer as compared to the
discovery cohort, the metabolic changes identified and the sepsis prognostication
model were validated in the independent cohort.
Rogers et al. also used the CAPSOD and RoCI cohorts to perform targeted
metabolic profiling on sepsis survivors and nonsurvivors (Rogers et al. 2014).
Although the Langley and Rogers groups identified many of the same metabolites
that differed in survivors and nonsurvivors, the Bayesian model developed by the
The Metabolomics of Critical Illness 377
Rogers group using RoCI as the training cohort was very different, using seven
completely different metabolites than those used in the Langley algorithm. This may
reflect a redundancy of metabolites found in the same metabolic pathways, but also
illustrates the evolving nature of analytic methodology in the field, in which the
optimal statistical methods for dealing with highly correlated data are not yet known.
One of the most promising applications for the use of metabolomics in critical care is
in the identification of patients who might differentially respond to therapies. Sepsis
treatment is currently focused on early antimicrobial administration and is otherwise
supportive, as no mechanism-targeted therapies are approved for use in the syn-
drome. The failure of many studies to identify effective, mechanism-based
treatments has been attributed to sepsis’s significant heterogeneity, which may
mask individual or subgroup responses to therapies in clinical trials.
Along these lines, Puskarich et al. used metabolomics to further phenotype
patients in a reanalysis of a phase II study of L-carnitine infusion for the treatment
of septic shock (Puskarich et al. 2015). The group performed metabolomics on
existing serum samples from septic patients in the study and found that three
metabolites involved in the synthesis and depredation of ketones differed in the
pre-intervention samples of L-carnitine-treated survivors and nonsurvivors. In later
work, they went on to identify acetylcarnitine as a particularly powerful biomarker
for predicting L-carnitine mortality benefit (Puskarich et al. 2021). While the
findings of retrospective reanalysis of a randomized controlled trials are likely
insufficient to change clinical practice, the hope is that metabolomics can be used
in the future to identify potential drug responders prior to their inclusion into
prospective clinical trials.
lung injury and inflammation, not a single drug had been approved to treat ARDS
prior to COVID-19 (Matthay et al. 2019). In COVID-19, numerous drug trials have
proved effective for the syndrome, likely because of increased homogeneity in the
patient population: all cases share an underlying etiology of SARS-CoV2 infection,
a typical time course of disease, and a substantial inflammatory component. Multiple
anti-inflammatory drug therapies are now available for COVID ARDS patients
(RECOVERY Collaborative Group et al. 2021; Investigators et al. 2021; Kalil
et al. 2021).
Metabolomics studies in ARDS are compelling because of the dynamic
metabolomic changes in the blood and lungs, with the potential for providing a
real-time snapshot of disease and identifying high-risk subjects who could be
targeted for trials (Millet, “Prospective metabolomic studies in precision medicine.
The AKRIBEA project.”; Turano, “NMR-based metabolomics to evaluate individ-
ual response to treatments”). Multiple studies have examined metabolomics of
ARDS; major studies are summarized below and in a thorough recent review
(Metwaly and Winston 2020).
Some of the most high-profile advances in ARDS pathogenesis in the past decade
have involved identifying high-risk subsets of patients who are at increased risk of
bad outcomes. Most of these risk markers have involved cytokines and proteins, for
example angiopoietin 2 (Calfee et al. 2012; Zinter et al. 2016), IL-18 (Rogers et al.
2019; Dolinay et al. 2012), and a hypoinflammatory/hyperinflammatory latent class
analysis (LCA) designation which can be assigned using a parsimonious model of
3 blood biomarkers (IL-6, TNF receptor alpha, and bicarbonate) (Calfee et al. 2014;
Sinha et al. 2020). The latent class hyperinflammatory subset has been particularly
well-studied and shown to be present in more than six ARDS populations (both
clinical trials and clinical cohorts) and associated with differential treatment
response in numerous ARDS clinical trials, including approach to positive
end-expiratory pressure (PEEP) level, fluid management, and statin therapy (Calfee
et al. 2014, 2018; Famous et al. 2017).
Fewer studies have focused on using metabolomics to identify high-risk subsets
of ARDS. Metwaly et al. tested whether orthogonal projections to latent structures
discriminant analysis (OPLS-DA) could differentiate previously identified subsets of
ARDS, including direct pulmonary risk factor for developing ARDS (e.g., pneumo-
nia) vs indirect risk factors (e.g., pancreatitis or non-pulmonary sepsis), and
hyperinflammatory subphenotype using latent class (Metwaly et al. 2021). Indeed,
using metabolites identified via high-VIP score for OPLS-DA, these subsets could
be separated in both testing and training sets; metabolomics outperformed protein
analyses in separating the direct and indirect classes.
The Rogers et al. study of 197 patients included 75 patients with ARDS (Rogers
et al. 2021). As discussed above, PLS-DA was not able to differentiate patients with
sepsis from ARDS. However, unbiased metabolic clustering of the entire cohort did
reveal three subsets of patients, with differential metabolites marked by lipid classes
driving the subclasses, with markedly different risks of mortality, regardless of
ARDS status.
Perhaps one of the most promising aspects of metabolomic profiling is the ability to
examine trace amounts of a lung-specific sample which could be captured without
need for a tissue biopsy to use for metabolic profiling (e.g., bronchoalveolar lavage
fluid, expired breath, or free-flowing pulmonary edema fluid). Such non-blood
samples may overcome the non-specific metabolic changes that occur with critical
illness and allow for a more lung injury-specific signature to emerge.
380 A. E. Pacheco-Navarro and A. J. Rogers
Both sepsis and ARDS are heterogenous and life-threatening clinical syndromes in
which a dysregulated immune response triggers dramatic changes in metabolism.
Identifying targeted and effective treatments for these syndromes remains a chal-
lenge. The current therapeutic paradigm hinges on treating underlying infections and
otherwise providing supportive care; no effective therapies have been found to treat
the metabolic and inflammatory derangements that drive these syndromes.
Metabolomics offers a potential tool for better identifying the biological
mechanisms underlying these syndromes and has already shown promise in prog-
nostication of both sepsis and ARDS. Further work will be needed to more cleanly
differentiate the lung-specific injury that occurs in ARDS from the frequently
overlapping changes seen in sepsis. Ultimately, metabolomics offers the opportunity
to uncover important biologic pathways that may serve as therapeutic targets and
The Metabolomics of Critical Illness 381
identify subgroups of patients in both sepsis and ARDS who may respond differen-
tially to current and future therapeutics.
References
Allingstrup M, Afshari A (2015) Selenium supplementation for critically ill adults. Cochrane
Database Syst Rev (7):CD003703. https://doi.org/10.1002/14651858.CD003703.pub3
Annane D, Sébille V, Charpentier C et al (2002) Effect of treatment with low doses of hydrocorti-
sone and fludrocortisone on mortality in patients with septic shock. JAMA 288(7):862–871.
https://doi.org/10.1001/jama.288.7.862
Annane D, Renault A, Brun-Buisson C et al (2018) Hydrocortisone plus fludrocortisone for adults
with septic shock. N Engl J Med 378(9):809–818. https://doi.org/10.1056/NEJMoa1705716
Bellani G, Laffey JG, Pham T et al (2016) Epidemiology, patterns of care, and mortality for patients
with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA 315(8):
788–800. https://doi.org/10.1001/jama.2016.0291
Besecker BY, Exline MC, Hollyfield J et al (2011) A comparison of zinc metabolism, inflammation,
and disease severity in critically ill infected and noninfected adults early after intensive care unit
admission. Am J Clin Nutr 93(6):1356–1364. https://doi.org/10.3945/ajcn.110.008417
Calfee CS, Gallagher D, Abbott J, Thompson BT, Matthay MA, Network NA (2012) Plasma
angiopoietin-2 in clinical acute lung injury: prognostic and pathogenetic significance. Crit Care
Med 40(6):1731–1737. https://doi.org/10.1097/CCM.0b013e3182451c87
Calfee CS, Delucchi K, Parsons PE et al (2014) Subphenotypes in acute respiratory distress
syndrome: latent class analysis of data from two randomised controlled trials. Lancet Respir
Med 2(8):611–620. https://doi.org/10.1016/S2213-2600(14)70097-9
Calfee CS, Delucchi KL, Sinha P et al (2018) Acute respiratory distress syndrome subphenotypes
and differential response to simvastatin: secondary analysis of a randomised controlled trial.
Lancet Respir Med 6(9):691–698. https://doi.org/10.1016/S2213-2600(18)30177-2
Casaer MP, Van den Berghe G (2014) Nutrition in the acute phase of critical illness. N Engl J Med
370(25):2450–2451. https://doi.org/10.1056/NEJMc1404896
Casaer MP, Mesotten D, Hermans G et al (2011) Early versus late parenteral nutrition in critically ill
adults. N Engl J Med 365(6):506–517. https://doi.org/10.1056/NEJMoa1102662
Cooper MS (2003) Corticosteroid Insufficiency in acutely Ill patients. N Engl J Med:8
Dolinay T, Kim YS, Howrylak J et al (2012) Inflammasome-regulated cytokines are critical
mediators of acute lung injury. Am J Respir Crit Care Med 185(11):1225–1234. https://doi.
org/10.1164/rccm.201201-0003OC
Eckerle M, Ambroggio L, Puskarich MA et al (2017) Metabolomics as a driver in advancing
precision medicine in sepsis. Pharmacotherapy 37(9):1023–1032. https://doi.org/10.1002/phar.
1974
Englert JA, Rogers AJ (2016) Metabolism, metabolomics, and nutritional support of patients with
sepsis. Clin Chest Med 37(2):321–331. https://doi.org/10.1016/j.ccm.2016.01.011
Famous KR, Delucchi K, Ware LB et al (2017) Acute respiratory distress syndrome subphenotypes
respond differently to randomized fluid management strategy. Am J Respir Crit Care Med
195(3):331–338. https://doi.org/10.1164/rccm.201603-0645OC
Ferguson ND, Fan E, Camporota L et al (2012) The Berlin definition of ARDS: an expanded
rationale, justification, and supplementary material. Intensive Care Med 38(10):1573–1582.
https://doi.org/10.1007/s00134-012-2682-1
Fleischmann C, Scherag A, Adhikari NKJ et al (2016) Assessment of global incidence and
mortality of hospital-treated sepsis. Current estimates and limitations. Am J Respir Crit Care
Med 193(3):259–272. https://doi.org/10.1164/rccm.201504-0781OC
382 A. E. Pacheco-Navarro and A. J. Rogers
Gaieski DF, Edwards JM, Kallan MJ, Carr BG (2013) Benchmarking the incidence and mortality of
severe sepsis in the United States. Crit Care Med 41(5):1167–1174. https://doi.org/10.1097/
CCM.0b013e31827c09f8
Hermans G, Casaer MP, Clerckx B et al (2013) Effect of tolerating macronutrient deficit on the
development of intensive-care unit acquired weakness: a subanalysis of the EPaNIC trial.
Lancet Respir Med 1(8):621–629. https://doi.org/10.1016/S2213-2600(13)70183-8
ICU Outcomes. Philip R. Lee Institute for Health Policy Studies Accessed 24 Apr 2022. https://
healthpolicy.ucsf.edu/icu-outcomes
Investigators RC, Gordon AC, Mouncey PR et al (2021) Interleukin-6 receptor antagonists in
critically ill patients with Covid-19. N Engl J Med 384(16):1491–1502. https://doi.org/10.1056/
NEJMoa2100433
Jansen TC, van Bommel J, Schoonderbeek FJ et al (2010) Early lactate-guided therapy in intensive
care unit patients: a multicenter, open-label, randomized controlled trial. Am J Respir Crit Care
Med 182(6):752–761. https://doi.org/10.1164/rccm.200912-1918OC
Jones AE, Shapiro NI, Trzeciak S et al (2010) Lactate clearance vs central venous oxygen saturation
as goals of early sepsis therapy: a randomized clinical trial. JAMA 303(8):739–746. https://doi.
org/10.1001/jama.2010.158
Kalil AC, Patterson TF, Mehta AK et al (2021) Baricitinib plus Remdesivir for hospitalized adults
with Covid-19. N Engl J Med 384(9):795–807. https://doi.org/10.1056/NEJMoa2031994
Khardori R, Castillo D (2012) Endocrine and metabolic changes during sepsis. Med Clin North Am
96(6):1095–1105. https://doi.org/10.1016/j.mcna.2012.09.005
Langley RJ, Tsalik EL, van Velkinburgh JC et al (2013) An integrated clinico-metabolomic model
improves prediction of death in sepsis. Sci Transl Med 5(195):195ra95. https://doi.org/10.1126/
scitranslmed.3005893
Langley RJ, Tipper JL, Bruse S et al (2014) Integrative “omic” analysis of experimental bacteremia
identifies a metabolic signature that distinguishes human sepsis from systemic inflammatory
response syndromes. Am J Respir Crit Care Med 190(4):445–455. https://doi.org/10.1164/
rccm.201404-0624OC
Manzanares W, Dhaliwal R, Jiang X, Murch L, Heyland DK (2012) Antioxidant micronutrients in
the critically ill: a systematic review and meta-analysis. Crit Care Lond Engl 16(2):R66. https://
doi.org/10.1186/cc11316
Marik PE, Pastores SM, Annane D et al (2008) Recommendations for the diagnosis and manage-
ment of corticosteroid insufficiency in critically ill adult patients: consensus statements from an
international task force by the American College of Critical Care Medicine. Crit Care Med
36(6):1937–1949. https://doi.org/10.1097/CCM.0b013e31817603ba
Masiero E, Agatea L, Mammucari C et al (2009) Autophagy is required to maintain muscle mass.
Cell Metab 10(6):507–515. https://doi.org/10.1016/j.cmet.2009.10.008
Matthay MA, Zemans RL, Zimmerman GA et al (2019) Acute respiratory distress syndrome. Nat
Rev Primer 5(1):18. https://doi.org/10.1038/s41572-019-0069-0
Metwaly SM, Winston BW (2020) Systems biology ARDS research with a focus on metabolomics.
Metabolites 10(5). https://doi.org/10.3390/metabo10050207
Metwaly S, Cote A, Donnelly SJ et al (2021) ARDS metabolic fingerprints: characterization,
benchmarking, and potential mechanistic interpretation. Am J Physiol Lung Cell Mol Physiol
321(1):L79–L90. https://doi.org/10.1152/ajplung.00077.2021
Mickiewicz B, Vogel HJ, Wong HR, Winston BW (2013) Metabolomics as a novel approach for
early diagnosis of pediatric septic shock and its mortality. Am J Respir Crit Care Med 187(9):
967–976. https://doi.org/10.1164/rccm.201209-1726OC
Mickiewicz B, Duggan GE, Winston BW et al (2014) Metabolic profiling of serum samples by 1H
nuclear magnetic resonance spectroscopy as a potential diagnostic approach for septic shock.
Crit Care Med 42(5):1140–1149. https://doi.org/10.1097/CCM.0000000000000142
Mikkelsen ME, Miltiades AN, Gaieski DF et al (2009) Serum lactate is associated with mortality in
severe sepsis independent of organ failure and shock. Crit Care Med 37(5):1670–1677. https://
doi.org/10.1097/CCM.0b013e31819fcf68
The Metabolomics of Critical Illness 383
Piel DA, Gruber PJ, Weinheimer CJ et al (2007) Mitochondrial resuscitation with exogenous
cytochrome c in the septic heart. Crit Care Med 35(9):2120–2127. https://doi.org/10.1097/01.
ccm.0000278914.85340.fe
Pool R, Gomez H, Kellum JA (2018) Mechanisms of organ dysfunction in sepsis. Crit Care Clin
34(1):63–80. https://doi.org/10.1016/j.ccc.2017.08.003
Preiser JC, Ichai C, Orban JC, Groeneveld ABJ (2014) Metabolic response to the stress of critical
illness. Br J Anaesth 113(6):945–954. https://doi.org/10.1093/bja/aeu187
Puskarich MA, Finkel MA, Karnovsky A et al (2015) Pharmacometabolomics of l-carnitine
treatment response phenotypes in patients with septic shock. Ann Am Thorac Soc 12(1):
46–56. https://doi.org/10.1513/AnnalsATS.201409-415OC
Puskarich MA, Jennaro TS, Gillies CE et al (2021) Pharmacometabolomics identifies candidate
predictor metabolites of an L-carnitine treatment mortality benefit in septic shock. Clin Transl
Sci. https://doi.org/10.1111/cts.13088
RECOVERY Collaborative Group, Horby P, Lim WS et al (2021) Dexamethasone in hospitalized
patients with Covid-19. N Engl J Med 384(8):693–704. https://doi.org/10.1056/
NEJMoa2021436
Rogers AJ, McGeachie M, Baron RM et al (2014) Metabolomic derangements are associated with
mortality in critically ill adult patients. PLoS One 9(1):e87538. https://doi.org/10.1371/journal.
pone.0087538
Rogers AJ, Contrepois K, Wu M et al (2017) Profiling of ARDS pulmonary edema fluid identifies a
metabolically distinct subset. Am J Physiol Lung Cell Mol Physiol 312(5):L703–L709. https://
doi.org/10.1152/ajplung.00438.2016
Rogers AJ, Guan J, Trtchounian A et al (2019) Association of elevated plasma interleukin-18 level
with increased mortality in a clinical trial of statin treatment for acute respiratory distress
syndrome. Crit Care Med 47(8):1089–1096. https://doi.org/10.1097/CCM.0000000000003816
Rogers AJ, Leligdowicz A, Contrepois K et al (2021) Plasma metabolites in early sepsis identify
distinct clusters defined by plasma lipids. Crit Care Explor 3(8):e0478. https://doi.org/10.1097/
CCE.0000000000000478
Rubenfeld GD, Caldwell E, Peabody E et al (2005) Incidence and outcomes of acute lung injury. N
Engl J Med 353(16):1685–1693. https://doi.org/10.1056/NEJMoa050333
Rudd KE, Johnson SC, Agesa KM et al (2020) Global, regional, and national sepsis incidence and
mortality, 1990-2017: analysis for the global burden of disease study. Lancet Lond Engl
395(10219):200–211. https://doi.org/10.1016/S0140-6736(19)32989-7
Rydzak T, Groves RA, Zhang R et al (2022) Metabolic preference assay for rapid diagnosis of
bloodstream infections. Nat Commun 13:2332. https://doi.org/10.1038/s41467-022-30048-6
Sair M, Etherington PJ, Peter Winlove C, Evans TW (2001) Tissue oxygenation and perfusion in
patients with systemic sepsis. Crit Care Med 29(7):1343–1349. https://doi.org/10.1097/
00003246-200107000-00008
Schmidt C, Höcherl K, Schweda F, Bucher M (2007) Proinflammatory cytokines cause down-
regulation of renal chloride entry pathways during sepsis. Crit Care Med 35(9):2110–2119.
https://doi.org/10.1097/01.ccm.0000281447.22966.8b
Shapiro NI, Howell MD, Talmor D et al (2005) Serum lactate as a predictor of mortality in
emergency department patients with infection. Ann Emerg Med 45(5):524–528. https://doi.
org/10.1016/j.annemergmed.2004.12.006
Singer M, De Santis V, Vitale D, Jeffcoate W (2004) Multiorgan failure is an adaptive, endocrine-
mediated, metabolic response to overwhelming systemic inflammation. Lancet 364(9433):
545–548. https://doi.org/10.1016/S0140-6736(04)16815-3
Singer M, Deutschman CS, Seymour CW et al (2016) The third international consensus definitions
for sepsis and septic shock (Sepsis-3). JAMA 315(8):801–810. https://doi.org/10.1001/jama.
2016.0287
Sinha P, Delucchi KL, McAuley DF, O’Kane CM, Matthay MA, Calfee CS (2020) Development
and validation of parsimonious algorithms to classify acute respiratory distress syndrome
384 A. E. Pacheco-Navarro and A. J. Rogers
phenotypes: a secondary analysis of randomised controlled trials. Lancet Respir Med 8(3):
247–257. https://doi.org/10.1016/S2213-2600(19)30369-8
Sprung CL, Annane D, Keh D et al (2008) Hydrocortisone therapy for patients with septic shock. N
Engl J Med 358(2):111–124. https://doi.org/10.1056/NEJMoa071366
Thompson BT, Chambers RC, Liu KD (2017) Acute respiratory distress syndrome. N Engl J Med
377(6):562–572. https://doi.org/10.1056/NEJMra1608077
Tomazini BM, Maia IS, Cavalcanti AB et al (2020) Effect of dexamethasone on days alive and
ventilator-free in patients with moderate or severe acute respiratory distress syndrome and
COVID-19: the CoDEX randomized clinical trial. JAMA 324(13):1307–1316. https://doi.org/
10.1001/jama.2020.17021
Vanhorebeek I, Gunst J, Derde S et al (2011) Insufficient activation of autophagy allows cellular
damage to accumulate in critically ill patients. J Clin Endocrinol Metab 96(4):E633–E645.
https://doi.org/10.1210/jc.2010-2563
Venkatesh B, Finfer S, Cohen J et al (2018) Adjunctive glucocorticoid therapy in patients with
septic shock. N Engl J Med 378:797. https://doi.org/10.1056/NEJMoa1705835
Villar J, Ferrando C, Martínez D et al (2020) Dexamethasone treatment for the acute respiratory
distress syndrome: a multicentre, randomised controlled trial. Lancet Respir Med 8(3):267–276.
https://doi.org/10.1016/S2213-2600(19)30417-5
Viswan A, Ghosh P, Gupta D, Azim A, Sinha N (2019) Distinct metabolic endotype mirroring acute
respiratory distress syndrome (ARDS) subphenotype and its heterogeneous biology. Sci Rep
9(1):2108. https://doi.org/10.1038/s41598-019-39017-4
Wasyluk W, Wasyluk M, Zwolak A (2021) Sepsis as a pan-endocrine illness-endocrine disorders in
septic patients. J Clin Med 10(10):2075. https://doi.org/10.3390/jcm10102075
Zinter MS, Spicer A, Orwoll BO et al (2016) Plasma angiopoietin-2 outperforms other markers of
endothelial injury in prognosticating pediatric ARDS mortality. Am J Physiol Lung Cell Mol
Physiol 310(3):L224–L231. https://doi.org/10.1152/ajplung.00336.2015