Synthesis of Novel Chloroquine Derivatives and Insights Into The Interactions of A Resistance Protein
Synthesis of Novel Chloroquine Derivatives and Insights Into The Interactions of A Resistance Protein
By
A thesis submitted to McGill University in partial fulfillment of the requirements of the degree of
Doctor of Philosophy
Department of Chemistry
November 2020
II
Abstract
The human toll caused by malaria remains devastating. In 2018 alone, there were 228 million
cases, which resulted in 405,000 deaths. Malaria is caused by the Plasmodium parasite transmitted
by the Anopheles mosquito. There are five species: P. malariae, P.falciparum, P. vivax, P. ovale
and P. knowlesi, in which P. falciparum is the most common and most deadly. An antimalarial
drug called chloroquine (CQ) was first introduced in the 1940s and is considered the gold-standard
antimalarial drug due to its high efficacy, low-cost, and low-toxicity. It is listed under the World
Health Organization (WHO) essential medicines and showed great promise in their endeavor to
eradicate malaria.
Unfortunately, after two decades of use resistance began to arise, which led to its gradual
withdrawal for P. falciparum cases. The WHO has reported an increase in the number of malaria
cases over the last six years (214, 216, 228 million cases in 2014, 2016 and 2018, respectively),
this is alarming as it greatly hampers the effects of eradication. Therefore, it remains crucial to
invest in the research and development of new antimalarial drug candidates. Recent studies have
reported the re-sensitization of chloroquine in countries where it was once completely banned,
such as Malawi, Zambia and certain regions of The Ivory Coast. This indicates that resistance is
reversible because it requires a fitness deficit. When drug pressure is removed, the frequency of
resistance alleles is reduced but not eliminated. Therefore, the reintroduction of drug pressure will
cause a rapid reemergence of resistance. As CQ was the ideal drug, many derivatives with varying
sidechains and aryl substituents such as amodiaquine and piperaquine have emerged as
commercial drugs. Aminoquinolines (AQs) continue to be prepared and tested, and two AQs were
III
recently undergoing phase II clinical trials, demonstrating the robustness of the AQ core and their
future potential.
My research centres around the repurposing of chloroquine via modification at the 3-position of
the quinoline ring. In chapter 2 I describe the synthesis and optimization of 3-aminochloroquine
(3-NH2CQ). The synthesis of this derivative allows for expansion at the 3-position for the synthesis
In chapter 3, I describe the synthesis of novel substituted chloroquine analogs. The structure-
activity relationship (SAR) was determined as I synthesized a library of compounds with varying
steric and electronic effects. It was determined that electron-withdrawing substituents at the para-
position gave the best antimalarial activity. Although the synthesized compounds were not as
effective as CQ, they may be candidates for combination therapy with chloroquine.
In chapter 4, I describe the synthesis of a CQ photoaffinity label with minor modifications. An aryl
azide is installed at the 3rd-position on the quinoline ring, making it the first AQ photoaffinity label
in the literature with the least number of modifications whilst retaining all features of the parent
compound, to our knowledge. Labelling studies were then carried out to determine the lability of
Lastly, the protein responsible for CQ resistance, Plasmodium falciparum chloroquine resistance
transporter (PfCRT), has been isolated for the first time. In chapter 5, I describe binding studies
carried out with PfCRT. In collaboration with Prof. Fidock's group at Columbia University,
IV
fluorescence and UV-Vis binding studies were carried out to determine binding affinities of heme
V
Résumé
Le bilan humain causé par le paludisme reste dévastateur. Rien qu'en 2018, il y a eu 228 millions
de cas, entraînant 405000 décès. Le paludisme est causé par le parasite Plasmodium transmis par
knowlesi, dans lesquelles P. falciparum est le plus commun et le plus mortel. Un médicament
antipaludique appelé chloroquine (CQ) a été introduit pour la première fois dans les années 1940
son faible coût et sa faible toxicité. Il est répertorié dans la liste des médicaments essentiels de
l'Organisation Mondiale de la Santé (OMS) et s'est montré très prometteur dans leurs efforts pour
éradiquer le paludisme.
cela a conduit à son retrait progressif pour les cas de P. falciparum. L'OMS a signalé une
augmentation du nombre de cas de paludisme au cours des 6 dernières années (214, 216, 228
millions de cas en 2014, 2016 et 2018 respectivement), ce qui est alarmant car cela entrave
considérablement les effets de l'éradication. Par conséquent, il reste crucial d'investir dans la
fait état d’un regain de l’efficacité de la chloroquine dans des pays où elle était autrefois
complètement interdite, comme le Malawi, la Zambie et certaines régions de Côte d'Ivoire. Cela
indique que la résistance est réversible car elle nécessite un déficit de forme physique. Lorsque la
pression médicamenteuse est supprimée, la fréquence des allèles de résistance est réduite mais pas
VI
avec des chaînes latérales et des substituants aryle variables tels que l'amodiaquine et la
pipéraquine ont émergé en tant que médicaments commerciaux. Les aminoquinoléines (AQ)
continuent d'être préparées et testées sous forme de deux AQ actuellement en phase II d'essais
cliniques: AQ-13 et ferroquine. Démontrer la robustesse du noyau d'AQ et leur potentiel futur.
Mes recherches sont centrées sur la réutilisation de la chloroquine via une modification en position
structure-activité a été déterminée lorsque j'ai synthétisé une bibliothèque de composés avec divers
effets stériques et électroniques. Il a été déterminé que les substituants attracteurs d'électrons en
position para donnaient la meilleure activité antipaludique. Bien que les composés synthétisés ne
soient pas aussi efficaces que la CQ, ils peuvent être des candidats pour une thérapie combinée
avec la chloroquine.
Dans le chapitre 4, je décris la synthèse d'un marqueur de photoaffinité CQ avec des modifications
mineures. Un aryl azide est installé en position 3, 3-azidochloroquine, ce qui en fait le premier
à notre connaissance toutes les caractéristiques du composé parent. Des études de marquage ont
VII
Enfin, la protéine responsable de la résistance à la CQ Plasmodium falciparum chloroquine
résistance transporter (PfCRT) a été isolée pour la première fois. Dans le chapitre 5, je décris les
études de liaison réalisées avec PfCRT. En collaboration avec le groupe Prof. Fidock de
l'Université de Columbia, des études de fluorescence et de liaison UV-Vis ont été réalisées pour
VIII
Acknowledgement
I am very thankful to my supervisor, Prof. Scott Bohle, who has taught me so much about the
beauty of chemistry through his love for science. Thank you for challenging me, for all the
I want to thank my research committee members Dr. Elias Georges, Prof. Karine Auclair, Prof.
Hanadi Sleiman and Prof. Gonzalo Cosa, for their interest in my research and input over the years.
I would also like to thank Dr. Robin Stein and Dr. Tara Sprules for their help with NMR
spectroscopy and Dr. Nadim K. Saadeh and Dr. Alex Wahba for carrying out High-Resolution
Mass Spectroscopy.
I thank all the group members along the way: Cassidy VanderSchee, Dr. Danae Guerra, Dr. David
Kuter, Harrison Cassidy, Dr. Ivor Wharf, Dr. Kristopher Rosadiuk, Dr. Munendra Yadav and
Quentin Gaydon for the interesting conversations, solicited distractions, and valuable input. I’d
also like to thank my friends Allie Domínguez, Jane Eiyegbenin, Ifenna Mbaezue, and Martin
Sichinga for their support and laughs along the way. Working alongside such a talented group of
The decision to pursue a doctorate and to see it through is not an easy one. It requires self-
motivation, determination, mental strength, and plenty of support. The love and joy received from
IX
I thank the Kapuku clan for being the best family members I could have ever wished for. Thank
you for a lifetime of unforgettable moments and for helping shape the person that I am today. My
success is our success, and this thesis is for all the Kapuku's.
I thank my sisters Nadine and Gloria for being there to listen. Thank you for the words of
encouragement and for always keeping me smiling. Thank you for being my ride-or-die sisters and
I thank my brothers Jeremie, Sam and Medi, for being my hype men. Thank you for the energy
and joy that you bring into my life. I can always rely on you guys for a fun time and the most
interesting conversations.
I thank my partner Hubert for being my cheerleader. Thank you for listening to all the rants and
sitting through all the practice presentations. Thank you for the motivational speeches and your
infectious smile. Thank you for keeping me grounded and for being the sugar in my ice-cream.
Finally, none of this would have been possible without my amazing parents' undying love and
support. I thank my mother, Espérance, and father, Jean-Pierre, for teaching me the values of hard
work and perseverance. Thank you for the endless prayers, words of wisdom and for being my
rocks. Thank you for all the sacrifices made so that I could live my best life. Thank you for
believing in me even when I did not believe in myself. I am so grateful to have been loved and to
be loved by you.
X
Contribution of Authors
The author carried out all experimental design, compound synthesis, structural analysis, binding
studies and all the work detailed in the thesis under the supervision of Prof. Scott Bohle.
Chapter 3: Antiplasmodial assays were carried out by our collaborators Dr. Fadi Baakdah, under
Chapter 5: PfCRT isoform 7G8, GtrB and empty nanodisc were gifted from Jonathan Kim, under
XI
Table of Contents
Abstract ....................................................................................................................................... III
Résumé......................................................................................................................................... VI
Acknowledgement....................................................................................................................... IX
XII
1.5.4.1 Ferroquine.................................................................................................................. 48
1.5.4.2 AQ-13 ........................................................................................................................ 50
1.5.5 Aryl aminoalcohols .......................................................................................................... 51
1.5.5.1 Quinine ...................................................................................................................... 51
1.5.5.2 Mefloquine ................................................................................................................ 52
1.5.6 Non-quinoline antimalarials ........................................................................................... 53
XIII
3.3.3 Isomerization ................................................................................................................. 115
3.3.4 Fluoride coupling in 1H NMR ...................................................................................... 116
3.3.5 Synthesis of Sulfonamides and Schiff-base derivatives ............................................... 117
3.3.6 pKa studies ...................................................................................................................... 119
XIV
5.3.2 UV-Vis Studies ............................................................................................................... 206
5.3.2.1 UV-Vis Studies with heme ...................................................................................... 206
XV
List of Figures
Figure 1.1. Plasmodium life cycle. Illustration adapted from Wirth.7 ................................... 27
Figure 1.2. Catabolism of Hb and heme detoxification mechanism. Illustration adapted
from Rosenthal and Meshnick.14 ........................................................................................ 29
Figure 1.3. Structure of Fe(III)PPIX. Figure adapted from Egan.18 ...................................... 31
Figure 1.4. 8-Aminoquinolines. .................................................................................................. 34
Figure 1.5. Acridines. .................................................................................................................. 36
Figure 1.6. 4-aminoquinolines. ................................................................................................... 36
Figure 1.7. CQ mechanism of action. ......................................................................................... 38
Figure 1.8. Ga(III)PPIX and CQ complex formed in the presence of methanol.42 ............... 39
Figure 1.9. CQ Structure-Activity Relationship. ...................................................................... 41
Figure 1.10. ADQ analogs. .......................................................................................................... 47
Figure 1.11. Antimalarial drug candidates in clinical trials. ................................................... 48
Figure 1.12. a) Hydrogen-bonding in FQ and b) the structure of methyl FQ. ...................... 49
Figure 1.13. Aryl aminoalcohols................................................................................................. 51
Figure 1.14. Non-quinoline antimalarial drugs. ....................................................................... 53
Figure 1.15. Structure of PfCRT 7G8 isoform. A) Topology showing inverted antiparallel
TM helices. B) Surface representation of the central cavity’s electrostatic potential
with red and blue, indicating negative and positive residues, respectively. Figures are
taken from Kim et al.83 ........................................................................................................ 55
Figure 1.16. K76T mutation removes a positive charge, which allows CQ2+ efflux from its
active site. ............................................................................................................................. 56
Figure 1.17 Structure of pfmdr1 with highlighted mutations: N86Y, Y184F, S1034C,
N1042D, D1246Y and two nucleotide-binding domains. Illustration adapted from
Valderramos and Fidock.91 ................................................................................................. 58
Figure 2.1. Simple AQs that do not inhibit hemozoin formation. ........................................... 74
Figure 2.2. AQs that do not inhibit hemozoin formation. ........................................................ 75
Figure 2.3. Proposed pathways for Cu(I)/ Cu(III) Ullmann-type amination catalytic cycle.
............................................................................................................................................... 89
Figure 2.4. Proposed Cu(I)/ Cu(II) SRN1 mechanism. .............................................................. 90
Figure 2.5. Proposed p-complexation Ullmann-type mechanism............................................ 91
Figure 3.1. Craig Plot groups substituents with similar electronic and hydrophobic effects.
Highlighted are synthesized compounds containing substituents at various locations on
the phenyl ring. .................................................................................................................. 107
Figure 3.2. Synthesized compounds. Colours: grey: the reference compound, blue: electron-
withdrawing and hydrophilic, orange: electron-withdrawing and hydrophobic, purple:
electron-donating and hydrophilic, green: electron-donating and hydrophobic. ....... 108
Figure 3.3. Possible benzamidoCQ conformations. ................................................................ 110
Figure 3.4. Possible fluorobenzamidoCQ conformations. ..................................................... 112
Figure 3.5. Possible chlorobenzamidoCQ conformations. ..................................................... 113
Figure 3.6. Trans and Cis benzamidoCQ isomers. ................................................................. 115
Figure 3.7. Fluorine and hydrogen coupling. .......................................................................... 116
Figure 3.8. Structure of sulfonamide and Schiff-base derivatives ........................................ 117
Figure 3.9. Possible sulfonamide conformations. ................................................................... 118
XVI
Figure 4.1. Known photoaffinity labels. .................................................................................. 139
Figure 4.2. Structure of novel 3-Azidomethylchloroquine in which all important CQ
features are present. .......................................................................................................... 141
Figure 4.3. Overlaying spectra of 3-NH2MeCQ, 3-N3MeCQ and CQ.2H3PO4 for
comparison. ........................................................................................................................ 155
Figure 4.4. Photolysis of 3-N3MeCQ at 254 nm excitation. ................................................... 156
Figure 4.5. Photolysis of 42.97 µM N3 in H2O and 35.57 µM N3 in MeOH at 365 nm. ....... 157
Figure 4.6. Photolysis of 3-N3MeCQ at 0 and 16 minutes in MeOD-d4. ............................... 157
Figure 4.7. Photolysis of 3-N3MeCQ in MeOD-d4 over 16 minutes. ..................................... 158
Figure 4.8. Possible irradiated 3-N3MeCQ products formed. ............................................... 159
Figure 4.9. Photolysis of 21.49 µM 3-N3MeCQ with 22.32 µM GlyGlyGly and 4.29 µM 3-
N3MeCQ with 40.18 µM GlyGlyGly at 365 nm excitation over five minutes. ............. 160
Figure 4.10. Photolysis of 19.56 µM 3-N3MeCQ with 18.77 µM maleimide, 7.11 µM 3-
N3MeCQ with 33.37 µM maleimide and 3.56 µM 3-N3MeCQ and 37.54 µM maleimide
at 365 nm excitation over 5 minutes. ............................................................................... 161
Figure 4.11. Photolysis of 26.7mM 3-N3MeCQ and 26.6mM maleimide at 365 nm in d4-
MeOD.................................................................................................................................. 162
Figure 4.12. Comparison between activated and unactivated N-phenylmaleimide. ........... 163
Figure 4.13. a) Desired product v possible observed photolysis product, b) NMR of
photolysis product formation. .......................................................................................... 164
Figure 5.1. Structure of PfCRT 7G8 isoform. A) Topology showing inverted antiparallel
TM helices. B) Surface representation of the electrostatic potential of the central
cavity with red and blue indicating negative and positive residues, respectively.
Figures adapted from Kim et al.3 ..................................................................................... 189
Figure 5.2. Effect of 4 equivalent of CQ on the emission spectrum of PfCRT at pH 7,
PfCRT at pH 8, GtrB and nanodisc. ................................................................................ 192
Figure 5.3. Stern-Volmer plots for PfCRT, GtrB and nanodisc quenched with up to 4
equivalents of CQ (each dot represents a 0.2 molar addition). ..................................... 194
Figure 5.4. Modified Stern-Volmer plots for PfCRT, GtrB and nanodisc quenched with up
to 4 equivalents of CQ (each dot represents a 0.2 molar addition). .............................. 195
Figure 5.5. Concentration-dependent response of PfCRT to CQ. ........................................ 197
Figure 5.6. Binding plot to determine the number of CQ binding sites and binding constant.
............................................................................................................................................. 199
Figure 5.7. Effect of heme on fluorescence spectrum of PfCRT at pH 7 and 8, GtrB and
nanodisc. ............................................................................................................................. 200
Figure 5.8. Stern-Volmer plot for PfCRT, GtrB and nanodisc quenched with up to 4
equivalents of Heme (each dot represents a 0.2 molar addition). ................................. 202
Figure 5.9. Modified Stern-Volmer plots for PfCRT, GtrB and nanodisc quenched with up
to 4 equivalents of heme (each dot represents a 0.2 molar addition). ........................... 203
Figure 5.10. Binding plot to determine the number of heme binding sites and binding
constant. .............................................................................................................................. 205
Figure 5.11. Changes to MPLME with up to 2 molar addition of heme. ............................. 207
XVII
List of Schemes
XVIII
List of Tables
XIX
List of Abbreviations
20
DMF Dimethylformamide
DMSO Dimethyl sulfoxide
DV Digestive vacuole
equiv. Equivalent
ESI Electrospray ionization
EtOAc Ethyl acetate
EtOH Ethanol
FAQ 4-fluoroamodiaquine
Fe(III)PPIX Iron protoporphyrin IX
FQ Ferroquine
g Gram(s)
G6PD Glucose-6-phosphate dehydrogenase
Gtr glycosyltransferase
H2 O Water
Hb Hemoglobin
HCQ Hydroxychloroquine
Hex Hexanes
HMBC Heteronuclear multiple bond correlation spectroscopy
HRMS High-resolution mass spectroscopy
HSQC Heteronuclear single quantum coherence spectroscopy
Hz Hertz
HZ Hemozoin
IC50 Half maximal inhibitory concentration
J Coupling constant
K2CO3 Potassium carbonate
KSV Stern-Volmer quenching constant
L Litre
LiAlH4 Lithium aluminium hydride
M Molar
MeCN Acetonitrile
MeCQ Methyl chloroquine
MeOH Methanol
mg Milligram(s)
MgSO4 Magnesium sulfate
MHz Megahertz
mL Millilitre
mL Microliter
MP Mepacrine
MPLME Membrane proteins and lipid membrane ensemble
MQ Mefloquine
21
MW Microwave
N2 Nitrogen gas
Na2CO3 Sodium carbonate
NaHCO3 Sodium bicarbonate
NaN3 Sodium azide
NaOH Sodium hydroxide
NBD Nucleotide-binding domain
NBS N-bromosuccinimide
NBu4N3 Tetrabutylammoniumazide
NEt3 Triethylamine
NH4OH Ammonium hydroxide
nM Nanomolar
nm Nanometer
NMR Nuclear magnetic resonance
NOSEY Nuclear overhauser effect spectroscopy
PAL Photoaffinity label
PfCRT Plasmodium falciparum chloroquine resistant transporter
pfmdr1 Plasmodium falciparum multidrug resistance protein 1
PN Pyronaridine
ppm Parts per million
PPQ Piperaquine
PQ Primaquine
QD Quinidine
QN Quinine
RT Room temperature
SAR Structure-activity relationship
SET Single-electron transfer
SNAR Nucleophilic aromatic substitution
SQ Sontoquine
SRN1 Unimolecular radical nucleophilic substitution
tBuOK Potassium tert-butoxide
TCNE Tetracyanoethylene
TEMPO (2,2,6,6-Tetramethylpiperidin-1-yl)oxyl
TFQ Tafenoquine
THF Tetrahydrofuran
TLC Thin-layer chromatography
TM Transmembrane
TMSN3 Azidotrimethyl silane
Trp Tryptophan
UV-Vis Ultraviolet-visible spectroscopy
22
VP Verapamil
WHO World health organization
23
Chapter 1
1.1 Introduction
1.2 Malaria
Malaria is a devastating disease in which over 3 billion people are at risk of contracting the disease.
The most recent report by the World Health Organization (WHO) reported 228 million cases in
2018. Of those, there were 405,000 deaths. Africa bears 93% of all cases, followed by South-East
Asia and the Eastern Mediterranean. Children under the age of 5 are the most vulnerable,
accounting for 67% of deaths globally.1 Pregnant women are also at high risk. Despite the WHO’s
sustained efforts, the number of malaria cases has increased year-over-year from 2015-2019 from
Malaria is a vector-borne disease carried by the female Anopheles spp. mosquito. It transmits
parasites of the Plasmodium genus. Five Plasmodium species are known to infect humans: P.
falciparum, P. vivax, P. ovale, P. malariae and P. knowlesi. Plasmodium falciparum is the most
The parasitic life cycle begins during a blood meal of an infected female Anopheles mosquito,
Figure 1.1. The mosquito transmits parasites in their sporozoite stage from its saliva into the hosts’
bloodstream while feeding. The sporozoites travel to and enter the liver within 60 minutes of
entering the host to evade the hosts’ immune system. The liver-stage is asymptomatic and can last
between 5-16 days. Within the liver, sporozoites develop into a tissue schizont containing several
thousand merozoites.4 The asexual erythrocytic cycle begins when mature tissue schizonts rupture,
25
Chapter 1 Development of Antimalarial Drugs
resulting in thousands of merozoites released into the bloodstream. These merozoites then enter
red blood cells (RBCs), where they develop into blood schizonts. Once matured, the blood schizont
ruptures, releasing up to 32 merozoites that can re-enter RBCs to continue the cycle.5 This constant
invasion and rupture of RBCs cause clinical symptoms to appear, such as fever, fatigue, and chills.
These symptoms usually appear within one week after the commencement of the asexual cycle.
The cycle ends at the intervention of the immune system, administration of prophylactic
medication, or the host’s death. The differentiation of merozoites into hypnozoites occurs only in
P. vivax and P. ovale. Hypnozoites are parasites that can remain dormant in the host hepatocytes
for months or years and are the cause of recrudescence of malaria in cured patients.5
Once in the bloodstream, some merozoites within infected erythrocytes differentiate into male and
female gametocytes, which marks the beginning of the sexual cycle. These gametocytes travel to
skin capillaries where they can be taken up during another blood meal. They remain in the
bloodstream but are non-pathogenic; however, it is unclear how they evade the immune system.
The beginning of sexual reproduction occurs within the mosquito’s midgut, where gametes form
a zygote, which develops into an ookinete and eventually forms an oocyst outside the midgut.
Within the oocyst, sporozoites are formed, and once mature, the oocyst ruptures, releasing
sporozoites, which concentrate in the mosquito salivary glands, ready to restart the life cycle. The
26
Chapter 1 Development of Antimalarial Drugs
During the trophozoite stage of the asexual cycle, the parasite degrades RBCs as it requires room
to grow and to obtain nutrients via the catabolism of hemoglobin (Hb). Hb has a cytosolic
27
Chapter 1 Development of Antimalarial Drugs
concentration of 5 mM, and it accounts for 95% of parasitic cytosolic proteins.8 Hb is transported
into the acidic digestive vacuole (DV), which has a pH of 5.0 – 5.6, where it is catabolized.9–11 Hb
catabolism releases Fe2+ and globin. Globin is hydrolyzed into amino acids as it is the primary
source of amino acids for the parasite. The parasite incorporates Hb amino acids with its proteins
as a source of nutrients and for de novo synthesis of proteins required for replication. However,
the parasite does not solely rely on Hb amino acids as Hb is deficient in isoleucine, methionine,
glutamic acid, and glutamine.8 Therefore, it is a dual requirement to have host amino acids and the
ability to synthesize amino acids as needed. Studies have shown that when Hb proteolysis is
Hb proteolysis occurs in the DV. It is during the early trophozoite stage where hemozoin (HZ)
formation becomes microscopically visible; however, HZ is present in most of the parasite life
cycle.13 Plasmepsins and falcipains are the main proteases involved in Hb degradation. More
specifically, cysteine and aspartic proteases are the primary enzymes involved in Hb proteolysis
as they account for 20-40% and 60-80% of globin degrading activity, respectively.8
Hb is denatured before heme is released from the peptide.14 Two aspartic proteases, plasmepsin I
and II, initiate Hb hydrolysis, Figure 1.2. Plasmepsin I and II initiate Hb cleavage by cleaving the
hinges that hold the tetramer together when oxygen is bound. Once the tertiary structure falls apart,
the plasmepsins further cleave Hb.15 Plasmepsin I is primarily synthesized in the early ring stage
but also synthesized in the trophozoite stage. Plasmepsin II is synthesized primarily in the
trophozoite stage. Therefore, plasmepsin I is more involved in Hb degradation than plasmepsin II.
28
Chapter 1 Development of Antimalarial Drugs
Studies show that blocking plasmepsin I leads to parasite death.8 Plasmepsin II is predominantly
active in the hydrolysis of the disassembled Hb units.16 The cysteine protease involved in globin
hydrolysis is the 28 kDa protein falcipain, synthesized in the trophozoite stage. Plasmepsins
hydrolysis Hb into large fragments of globin. Falcipain catabolizes globin into smaller peptide
fragments. Assays in which cysteine protease inhibitors were incubated with trophozoite stage
falcipains in parasite survival.14 There are two primary cysteine proteases that the parasite utilizes
falcipain-2 and falcipain-3. Falcipain-2 exerts 90% of falcipain activity and is predominant in the
early trophozoite stage. Falcipain-3 is only present in the late trophozoite stage, and both cleave
native Hb and denatured globin.17 Amino acids generated from globin hydrolysis are incorporated
Figure 1.2. Catabolism of Hb and heme detoxification mechanism. Illustration adapted from
29
Chapter 1 Development of Antimalarial Drugs
Once Hb has been disassembled by the aspartic and cysteine proteases, heme is released into the
cytosol of the DV. The released free heme is very reactive and toxic to the parasite. Free heme can
cause the release of reactive oxygen species, which leads to oxidative stress, membrane rupture,
and eventual death of the parasite. To circumvent this damage, the parasite transforms free heme
into HZ. Studies have shown that 95% of released heme is converted to HZ.18 Pagola et al. have
shown that HZ is an insoluble biomineral that is identical to synthetic b-hematin. X-ray structures
show that HZ is dimerized Fe(III)PPIX where a propionate group of each molecule coordinates
with the adjacent central Fe(III), and the dimers are linked via hydrogen bonding with the
It has been reported that HZ is completely encapsulated with a neutral lipid body upon its
formation, which is comprised of mono-, di- and tri-acylglycerols. Specifically, neutral lipids
monopalmitoyl- and monostearoyl glycerol promote HZ formation.20 It is believed that these lipid
bodies originate from the inner membrane of endocytotic transport vesicles that transport Hb to
the DV.18 Histidine-rich proteins have also been associated with HZ formation.
30
Chapter 1 Development of Antimalarial Drugs
OH H O
O O N
N
N N FeIII
FeIII N N
N N
O
O
OH
O
O
Cyclic dimer
Free heme
O
N N
FeIII
H O
OH N N
X O
O
O O H N N
N N FeIII
FeIII N N
N N
O
O
OH
O
O
Hematin O
X=OH/H2O
N N
FeIII
N N
O
O H
Hemozoin
(β-Hematin or hematin anhydride)
Powdered cinchona tree bark has been used to treat malaria chills for centuries in South America.
It was later discovered that the cinchona sp. contains several alkaloids, and the active ingredient
for treating malaria was quinine (QN) and quinidine (QD), Figure 1.13, first discovered in 1820
and 1833, respectively. QN was first isolated in 1820, and its first total synthesis was reported in
1944; however, its total synthesis is more laborious than its extraction. Depending on the species,
a cinchona bark contains 6 - 10 % alkaloids, of which 2-8 % is QN, and 0.2 % is QD. The tree also
contains smaller quantities of over 30 other alkaloids.21,22 Despite its effectiveness, there are many
31
Chapter 1 Development of Antimalarial Drugs
concerns about QNs toxicity profile; therefore, its use has been limited for treating severe malaria
in combination with doxycycline when other front-line medications are not available.23
The discovery of QN led to the synthesis of different classes of antimalarial drugs, mostly still in
use today. In an attempt to synthesize QN in 1856, William Henry Perkins accidentally synthesized
the first synthetic dye methylene blue. In 1891 Paul Ehrlich cured malaria patients with the dye
after noticing the dye was selectively taken up by the parasite. Methylene blue inhibits glutathione
reductase, disturbing redox homeostasis of the parasite.24 Modification of methylene blue led to
the synthesis of 3 classes of antimalarial drug families: 8-aminoquinolines (8-AQs), acridines, and
4-AQs.5
1.5.1 8-Aminoquinolines
Modification of methylene blue led to the synthesis 8-AQs, which are used as a radical cure against
malaria. The first 8-AQ, Pamaquine was synthesized in 1925 by the German pharmaceutical
company Bayer but displayed many side effects; therefore, it was not widely used.5 Modification
via the removal of the diethylamino sidechain of pamaquine for an unsubstituted amine gave
1.5.1.1 Primaquine
PQ, synthesized in 1946, has a much-improved safety profile compared to pamaquine and is
currently used in clinics.25 It is effective against hypnozoites in P. vivax and P. ovale and the liver
recently, PQ was the only approved drug for the treatment of liver-stage parasites. It has a short
32
Chapter 1 Development of Antimalarial Drugs
metabolites that are directly responsible for its toxicity (quinone toxicity is discussed in section
1.5.3.4.1). PQ is not safe for pregnant women or children under the age of 4.26 Due to its weak
1.5.1.2 Tafenoquine
Tafenoquine (TFQ), developed by GlaxoSmithKline and Medicines for Malaria Venture, is a more
lipophilic derivative of PQ that has a longer elimination half-life of 2 weeks. It was first developed
in 1978 by the Walter Reed Army Institute of Research. In 2018 TFQ was approved by the USA’s
Food and Drug Administration as a prophylactic treatment of P. vivax.29 Due to its long elimination
half-life TFQ is administered orally at 200 mg once a day for three days and then 200 mg once a
week for eight weeks.30 TFQ is effective against liver-stage, asexual stage and sexual stage parasite
in P. vivax and P. falciparum. Like PQ, TFQ is highly effective against hypnozoites. Although it
is better tolerated, TFQ cannot be given to patients with a G6PD deficiency or pregnant women.
TFQ is more potent than PQ against blood-stage parasites, but its activity is low; therefore, it is
33
Chapter 1 Development of Antimalarial Drugs
1.5.1.3 Bulaquine
Bulaquine (BQ) is a PQ prodrug approved outside of North America. It was developed by the
Central Drug Research Institute in India for the treatment of P. vivax,32 and it is given in
combination with chloroquine (CQ). BQ prevents relapse in P. vivax cases and has
gametocytocidal activity. BQ can be used as a safe alternative to PQ. It is currently under Phase II
O O O
N N O N
HN HN NH2
NH2 OH
F3C O
O O
O
N O
N O
HN HN
NH2 N
Tafenoquine Bulaquine
1.5.2 Acridines
Another class of antimalarial drugs that arose from methylene blue modifications is acridines,
Figure 1.5. Acridines were first synthesized in the 1930s, and their primary mode of action is on
blood-stage parasites.5 However, their use was short-lived because shortly after their introduction,
CQ was synthesized. As CQ was a significantly better drug, due to its low toxicity and high
34
Chapter 1 Development of Antimalarial Drugs
potency, acridine use was reduced. When CQ resistance became more widespread in the 1960s,
1.5.2.1 Mepacrine
Mepacrine (MP) was first synthesized in 1930; however, its use has not been widespread due to
its many side effects. It accumulates in the liver, kidney, and spleen and has a long elimination
half-life of up to 14 days. MP (also known as Quinacrine) has also been shown to accumulate in
lysosomes and bind to nucleic acids. It is an effective inhibitor of parasitic nucleic acid synthesis,
and it has been shown that low concentrations of MP inhibited the incorporation of 80% of P32
into erythrocytic DNA and RNA of rat P. berghei.33 Another study showed MP inhibits the
causes skin and tissue discolouration, dermatitis, diarrhea, and vertigo, to name a few. For these
1.5.2.2 Pyronaridine
Pyronaridine (PN) was synthesized in China in 1970 by the Institute of Parasite Diseases. Similar
two Mannich base side chains. Its mechanism of action remains unknown, but it is speculated that
it interferes with HZ formation. It is highly effective against CQ resistant (CQR) and CQ sensitive
(CQS) parasite strains. There is a possibility of cross-resistance / susceptibility between PN, and
CQ.35 PN is well tolerated; however, it can be metabolized to the quinone imine, a compound toxic
to neutrophils, Figure 1.5. It is currently administered as combination therapy with the artemisinin
35
Chapter 1 Development of Antimalarial Drugs
N N
OH O
N N N
HN HN HN
O N O N O
Cl N Cl N Cl N
Mepacrine Pyronaridine Pyronaridine quinone imine
1.5.3 4-Aminoquinolines
The most important class of compounds to arise from methylene blue modifications are the 4-AQs,
Figure 1.6. There have been countless derivatives and reviews on this group of compounds, which
gave rise to the most crucial antimalarial ever synthesized, CQ, considered the gold standard of
OH
N N N
HN HN HN
Cl N Cl N Cl N
Chloroquine Hydroxychloroquine Sontoquine
OH N N
N
HN N N
Cl N Cl N N Cl
Amodiaquine Piperaquine
36
Chapter 1 Development of Antimalarial Drugs
1.5.3.1 Chloroquine
CQ was first synthesized in 1934 by Bayer Pharmaceuticals. However, its first safety profile
determined that it was too toxic. Interest in CQ was reignited in 1945 when its development was
handed over to the USA.36 15 years after its introduction, there was a high occurrence of CQR
parasite strains, and in 2001 the molecular biomarker of CQ resistance was discovered. It is a
months.37
CQ is a lipophilic, weak diprotic base, with pKas of the quinoline nitrogen and the dialkylamino
side chain of 8.1 and 10.2, respectively. CQ accumulates in the DV, where it is primarily active.
Unprotonated CQ is membrane permeable and easily enters the DV down its pH gradient. The DV
has a pH of ~ 5.2. Once CQ is within the vacuole, it becomes diprotonated and membrane-
impermeable, thus trapped within its target organelle.38 Once within the DV, it has been reported
that CQ interferes with the parasite’s detoxification mechanism of HZ formation, Figure 1.7. CQ
forms a complex with Fe(III)PPIX, thus preventing the propagation of HZ.39 This leads to the
37
Chapter 1 Development of Antimalarial Drugs
CQ inhibits b-hematin formation by forming a complex with Fe(III)PPIX. This complex may
inhibit crystal nucleation, block the fastest growing face of the HZ crystal,19 or essentially bind to
any stage of HZ formation to block propagation of growth.40 It has been suggested that this
complex is driven by coordination to the Fe(III) centre, H-bonding, and p – p interactions between
the porphyrin and the quinoline ring. The association constant between the porphyrin and quinoline
ring is dependent on the quinoline ring substituents. Lipophilic, electron-withdrawing groups form
There has not yet been a universal consensus reached on the structure of the Fe(III)PPIX - CQ
complex formed. With the use of a gallium protoporphyrin IX (Ga(III)PPIX) model, Dodd and
Bohle were able to mimic the potential complex formed both in solution and in the solid-state,42
38
Chapter 1 Development of Antimalarial Drugs
Figure 1.8. They show that CQ forms a distinct, well-defined complex with [Ga(III)PPIX]2 and
highlights the significance of alkoxide coordination and H-bonding as a stabilizing feature. Other
publications focus on !-oxo dimer formation and the reliance on " – " stacking interactions to
explain complex formation.12,43,44 However, there is no evidence of the presence of !-oxo dimer
formation in vivo, though it is expected to form under slightly acidic conditions pH 5.5 – 6.0.
Lower pH’s promote the aggregation of the dimer and may accelerate HZ formation.12,44
CQ activity is highly dependent on each component of its structure, Figure 1.9. Structure-Activity
Relationship (SAR) analyses have been carried out to determine the role of each structural
component. As mentioned, CQ accumulates within the DV via the protonation of the two basic
amines. The removal of the quinolinium amine reduces the accumulation of the compound 100 –
39
Chapter 1 Development of Antimalarial Drugs
fold compared to compounds containing a basic side chain.45 As accumulation within the active
which the only modification was the absence of the tertiary alkylamine and found that it is 4 – fold
less potent than CQ in CQS strain NF54.46 Egan et al. were able to demonstrate that CQ analogs
lacking a basic side chain retained their ability to inhibit b-hematin formation (discussed in chapter
2); however, they displayed poor antiplasmodial activity, with IC50 values between 3.8 – 5 µM.45
It has been shown that the length of the side chain has a significant impact on the antiplasmodial
activity of the compound. Shortening the chain appears to increase potency, whereas increasing
the length has a negative effect on activity. This is evident in the antimalarial drug candidate
currently undergoing clinical trials, which contains a shortened chain, AQ13, Figure 1.11.
Furthermore, the length of the side chain has an impact on the stability of the quinoline-
Fe(III)PPIX complex formed. Increasing the length by more than three carbons may force the
chain outside the Fe(III)PPIX rim. A chain too short may lower van der Waals contributions,
Figure 1.8.47
A key feature of CQ’s activity is its ability to accumulate within the DV and inhibit b-hematin
formation. The nature of the substituent at the 7-position determines the compound’s ability to
inhibit b-hematin. This is because substituents on the quinoline ring will affect the compound’s
lipophilicity and the pKa of the quinoline nitrogen, which determines the association constant with
Fe(III)PPIX and its ability to accumulate with the DV. Studies have shown that the 7-chloro
lipophilic groups such as Cl, CF3, and NO2 lowers the pKa of the quinolinium nitrogen and
40
Chapter 1 Development of Antimalarial Drugs
increases the compound’s lipophilicity its association constant. Thus, the Cl moiety shows the
highest inhibition of b-hematin formation.48 The use of electron-donating groups such as NH2 and
OCH3 raises the quinolinium pKa, decreases the lipophilicity of the compound and its association
constant relative to CQ. Substituents on the quinoline ring also affect the pKa of the tertiary
alkylamine; however, the effects on the side chain vary with varying side chain lengths. Hawley
et al. reported the correlation between drug accumulation and b-hematin inhibition,49 and Egan et
al. have reported a linear correlation between b-hematin inhibition and antiplasmodial activity.
Therefore, CQ activity is dependent on its accumulation within the DV and its ability to inhibit b-
hematin formation, which are both influenced by the nature of the substituent at the 7-position.
Antiplasmodial activity
N NH
41
Chapter 1 Development of Antimalarial Drugs
1.5.3.1.4 Metabolism
N HN H 2N
NH NH NH NH2
Cl N Cl N Cl N Cl N
Chloroquine Desethylchloroquine Bisdesethylchloroquine 4-amino-7-
chloroquinoline
extent 4-amino-7-CQ by the kidney and liver, as shown in Scheme 1.1. DesethylCQ is the major
metabolite that is found in high concentrations in the blood and plasma.50 DesethylCQ is as active
against CQS strains as CQ but is less active than CQ in CQR strains.47,51 Furthermore, CQ is
CQ is well tolerated; it is safe for use in children and pregnant women. However, it does have
some side effects, which include nausea, dizziness, and headaches. CQ can cross the blood-retina
wall and deposit into melanin-rich tissue. Long-term use of over five years can cause retinopathy,
leading to blurred vision to complete vision impairment.53,54 Retinopathy is irreversible, but early
The K+ ion channel encoded by the human ether-a-go-go related gene (hERG), which regulates
cardiac action potential, is inhibited by CQ. Inhibition of this channel can cause prolonged QTc
intervals, arrhythmia and, in some cases, heart failure.41,55 Therefore, it is crucial to identify
42
Chapter 1 Development of Antimalarial Drugs
1.5.3.2 Hydroxychloroquine
Hydroxychloroquine (HCQ) was first synthesized in 1946. It is a more polar derivative with a
hydroxy group on the alkyl side chain. The pKa of the quinolinium nitrogen is similar to that of
CQ; however, tertiary amine salt has a lowered pKa of 9.7.56 HCQ is less lipophilic therefore does
not easily diffuse across cell membranes, resulting in less accumulation in the DV. HCQ is 1.6 –
fold less potent than CQ in CQS strains and 8.8 - fold less potent than CQ in CQR strains.56 For
HCQ is the safer alternative to CQ as it does not readily cross the blood-retina barrier; therefore,
it deposits less onto melanin-rich tissue. Thus, retinopathy is only observed after continuous long-
term usage of HCQ. HCQ can also cross the placental as levels in maternal cells are equivalent to
that of the fetus. Interestingly, to date, there has been no evidence of toxicity to the fetus, no
congenital abnormalities, and no harm to newborns via breastfeeding. Therefore, HCQ is safe for
pregnant and breastfeeding women.57 The presence of the hydroxy group facilitates phase II
conjugation allowing for bile excretion.52 HCQ causes immunomodulatory effects such as
inhibition of phagocytosis and proteolysis, interference with lysosomal acidification, and toll-like
receptor signalling inhibition. In the USA, HCQ is commonly used to treat systemic lupus
1.5.3.3 Sontoquine
The 3-methyl CQ analogue sontoquine (SQ) was synthesized in 1936 by Bayer Pharmaceuticals.
Its synthesis was fueled by the initial incorrect classification of CQ as too toxic for use. SQ is
slightly less potent than CQ in CQS and CQR strains and was relatively safe. Initial human trials
began in 1937 in Cameroon to treat P. vivax and P. falciparum. This led to further trials in 1942
43
Chapter 1 Development of Antimalarial Drugs
in Tunisia by German and French scientists for the development of SQ. In 1943 research data was
handed over to the USA for analysis, which led to the resynthesis of CQ.36 The rediscovery of CQ
showed that it was non-toxic and more potent antimalarial. SQ development was discontinued in
favour of CQ development.
1.5.3.4 Amodiaquine
Amodiaquine (ADQ), first synthesized in 1948, is a Mannich base 4-AQ with a 4-aminophenol. It
is a more potent and more lipophilic derivative of CQ. ADQ is a weaker base than CQ with pKas
of 7.1 and 8.1 for the quinolinium nitrogen and alkyl nitrogen, respectively. That being said, AQ
accumulation is 2- to 3- fold greater than CQ in CQS strain (HB3). Indicating that its accumulation
is energy and pH-dependent, as only 15% of ADQ accumulation can be attributed to its weak base
properties.58 Due to the hydroxy and diethylamino moieties’ proximity, ADQ can form
intramolecular hydrogen bonds at physiological pH. This potentially plays a role in its increased
potency in CQR parasites. ADQ analogues with hydrogen bonding groups display greater in vitro
1.5.3.4.1 Metabolism
ADQ is quickly metabolized in the liver and has an elimination half-life of 5.2 hours, Scheme 1.2.
Its metabolic products are slowly excreted from the body and have an elimination half-life >4 days.
P4502C8. AQ is 3 - fold more potent than DADQ, but due to its high concentration, DADQ
44
Chapter 1 Development of Antimalarial Drugs
The presence of the 4-hydroxyanilino group is a cause of toxicity to the compound. The toxic
metabolite ADQ quinone-imine (ADQQI) is formed in the presence of cytochrome P450. This
metabolite is then susceptible to nucleophilic attack by glutathione to give ADQ-GS, which is non-
toxic. However, long-term use or high dosage of ADQ leads to the depletion of glutathione levels,60
leading to liver toxicity due to the increased concentration of ADQQI. ADQQI is also susceptible
to nucleophilic attack by thiol-containing enzymes that can cause hepatotoxicity. They are also
highly immunogenic and lead to agranulocytosis. Hepatoxicity is observed within three weeks to
10 months of continuous use, and agranulocytosis is observed after five to 14 weeks of continuous
use.5 Thus, prophylactic use of ADQ is not recommended. The WHO lists ADQ use only for
HN N N
HO HO O
NH CYP2C8 NH P450 N
Cl N Cl N Cl N
Desethyl amodiaquine Amodiaquine Amodiaquine quinoneimine
P450
H 2N N
HO HO
Protein
NH S NH
Cl N Cl N
Bisdesthethyl amodiaquine Amodiaquine-protein adduct
45
Chapter 1 Development of Antimalarial Drugs
1.5.3.4.2 Detoxification
Although ADQ is effective in CQR strains, its side effects are a significant deterrent. In an attempt
to reduce its toxicity, several analogs with good antimalarial profiles have been synthesized.
Incorporating a fluorine group can lead to increased lipophilicity, increased oxidation potential,
stronger H-bonding, and changes in pKas. O’Neill et al. substituted the 4-hydroxy group for a 4-
fluoro group, Figure 1.10, to improve the stability of the aryl ring to oxidation, thus blocking
bioactivation to the quinone imine. The 4-fluoroamodiaquine (FAQ) analog shows retention of
The exchange of the methylene N-diethyl with the hydroxy group gives rise to isoquine. This
exchange completely inhibits the formation of ADQQI, and it is more potent than ADQ in both
CQS and CQR P. falciparum strains. However, the desethyl metabolic product displays
be dealkylated due to steric hindrance, and bioactivation is inhibited due to the positioning of the
hydroxy group. This compound also retains its antiplasmodial activity and is less toxic than ADQ.
It is currently in clinical trials at GlaxoSmithKline. FAQ4 contains the N-tert-butyl moiety, and
the 4-hydroxy is replaced with fluorine. Dealkylation is blocked due to steric hindrance, and there
is no observed quinone-imine formed because of the 4-fluoro group. FAQ4 is active in CQS and
CQR strains in P. falciparum. This compound is also currently under clinical trials with Medicines
46
Chapter 1 Development of Antimalarial Drugs
N HN
OH OH
F F
N N
H
NH NH NH NH
Cl N Cl N Cl N Cl N
Fluoroamodiaquine (FAQ) Isoquine Tert-butylisoquine 4’-fluoro-N-tert-butylamodiaquine
(FAQ4)
1.5.3.5 Piperaquine
Piperaquine (PPQ) was first synthesized in the 1960s by a group in China and France
CQ after the rise of resistance and used exclusively in China. It was phased out in the late 1980s
due to resistance. PPQ is a less toxic CQ analog that is equipotent to CQ in CQS strains and is 6 -
fold more active in CQR strains than CQ. It has a quinolinium pKa of 8.9, and it is highly lipid-
soluble. It has a similar elimination half-life to that of CQ of 1-2 months. PPQ is believed to exert
its activity similarly to CQ; however, there is insufficient research on its mode of action. PPQ
contains more protonation sites than CQ, which may allow for greater accumulation in CQR
strains. Molecular modelling shows that both quinoline nuclei are capable of forming a complex
with b-hematin.62
PPQ is relatively safe and well-tolerated with minor side effects such as dizziness, vomiting and
mild headache. A rare significant side effect is the increase in blood pressure. PPQ is not
recommended for pregnant women or children under the age of two, as there is insufficient
evidence of safety for these at-risk groups.63 PPQ has been reintroduced in the Chinese market as
47
Chapter 1 Development of Antimalarial Drugs
a combination therapy with the artemisinin derivative dihydroartemisinin. It is worth noting that
PPQ resistance has not been observed in high transmission areas such as Sub-Saharan Africa as it
HN N HN N
Fe
Cl N Cl N
AQ-13 Ferroquine
1.5.4.1 Ferroquine
Ferroquine (FQ) is a 4-amino-organometalo-quinoline. It was first designed in 1993, and its first
published synthesis was in 1997.64 FQ is the first organometallic antimalarial, with an elimination
half-life of 16 days, see Figure 1.11. It completed Phase II clinical trials in 2011 by Sanofi-Aventis
and has not entered Phase III trials (ClinicalTrials.gov Identifier: NCT00988507). The presence of
1,2-unsymmetrically substituted ferrocene gives the molecule planar chirality. Experiments have
shown no significant difference in activity between the enantiomerically pure S (-)-FQ and R (+)-
FQ; therefore, it is prepared and administered as a racemate. FQ is highly effective against CQS
and CQR strains of P. falciparum, and there is little chance of cross-resistance with CQ. FQ is a
weaker base than CQ with pKas of 7.0 and 8.5 for the quinolinium nitrogen and tertiary nitrogen.
Furthermore, FQ is 100 - times more lipophilic than CQ at cytosolic pH and is more lipophilic
than CQ at vacuolar pH. FQ accumulates 50 times more than CQ at pH 565 despite being a weaker
base.
48
Chapter 1 Development of Antimalarial Drugs
Unlike CQ, neutral FQ can easily form a strong intramolecular H-bond, as shown in Figure 1.12.
A 2.17 Å bond is formed between the anilino (N11) and tertiary amino (N24), resulting in a folded
the solvent. The folded conformation results in the increase of hydrophobicity and the protrusion
of the ferrocenyl moiety towards the outside. It is believed that the hydrophobic ferrocenyl moiety
interacts with lipid structures in the membrane, and the quinoline stacks with Fe(III)PPIX. This
reversible open/folded conformation allows for the transport of FQ between membranes and is
believed to facilitate FQs ability to accumulate in the DV and to evade resistance transport
system.65,66
a)
Open conformation Folded conformation
(Hydrophilic) (Hydrophobic)
H
O
H H N Fe
24
Fe N
H b) N N
O H H
N N Fe
H 2.17Å 11
Cl N
Cl N Cl N FQ-Me
H
O
H H
To determine the importance of H-bonding in FQ, a 4-methyl analog was synthesized with a
methyl group on the anilino nitrogen. FQ-Me is a more lipophilic and slightly more basic analog
with pKas of 7.1 and 8.8 for quinolinium and tertiary nitrogen, respectively. FQ-Me can inhibit β-
hematin formation; however, its antiplasmodial activity is considerably reduced in CQS and CQR
strains.67 Thus, the H-bond characteristic is required for effective antiplasmodial activity.
49
Chapter 1 Development of Antimalarial Drugs
to induce resistance under drug pressure shows a low chance of cross-resistance of FQ with CQ
and other antimalarials and a low possibility of the emergence of resistance. Due to its high
However, Phase II clinical trials for combination therapy with artefenomel were recently
1.5.4.2 AQ-13
AQ-13 synthesis was first published in 1948;68 however, its development in clinical trials was not
pursued until 1996.69 It is a CQ analog in which the isopentyl chain is replaced with a propyl chain,
Figure 1.11. AQ-13 was found to be effective against P. vivax and CQS and CQR strains of P.
falciparum. It has similar antiplasmodial activity to CQ in CQS strains and is more potent than CQ
in CQR strains. Like CQ, it is safe for human use with a low toxicity profile. AQ-13 is less
bioavailable than CQ and has a shorter elimination half-life of 13 days. The N-diethyl moieties are
susceptible to oxidative dealkylation. The metabolic products that result in mono- and di-
Side effects are similar to that of CQ: headache, nausea and diarrhea, and major side effect such
as QTc interval prolongation was observed only in high dosage patients.70 These effects are
avoided when drug concentration is reduced. AQ-13 is currently under phase II clinical trials
50
Chapter 1 Development of Antimalarial Drugs
HO N HO N HO
N
H
MeO MeO
N N N CF3
Quinine Quinidine CF3
Mefloquine
1.5.5.1 Quinine
QN is extracted from cinchona bark and was the first antimalarial discovered. It was first isolated
in 1820 by Pettetier and Caventon,71 and its first total synthesis was carried out in 1944 by
Woodward and Doering, Figure 1.13.72 As it currently stands, it is more economical to extract QN
than to synthesize it. QN was the front-line antimalarial used until the discovery and synthesis of
more potent AQ antimalarials. QN is a weaker base than CQ with the quinolinium pKa of 5.173
versus 8.2 for CQ. Therefore, QN accumulates in the DV in its monoprotonated form. QN is a fast-
acting schizonticide effective in P. falciparum and P. vivax. Moreover, it is also active against
gametocytes of P. vivax and P. malariae. It has an elimination half-life of 11-18 hours and is used
symptoms include tinnitus, nausea, headaches and slight hearing loss, which is reversible when
drug dosage is reduced. More severe symptoms include vertigo, diarrhea, vision and hearing loss.71
The WHO currently recommends that QN only be used for cases of severe malaria and in
51
Chapter 1 Development of Antimalarial Drugs
1.5.5.2 Mefloquine
Mefloquine (MQ) contains a single piperidine ring and two trifluoromethyl groups at the 2- and 8-
position, Figure 1.13. MQ was first synthesized during World War II and used as a CQ
replacement in the 1980s. Like CQ, it is a weak diprotic base, and at physiological pH, it can form
an intramolecular H-bond between the hydroxyl group and protonated tertiary amino group.24 MQ
is a blood schizonticide that is active against P. falciparum and P. vivax. Its mechanism of action
is unknown; however, it is believed to act similarly to CQ and QN. MQ can cause severe side
effects, particularly related to the central nervous system, such as neuropsychiatric effects. It is
recommended for pregnant women and children weighing less than 15 kg.74
52
Chapter 1 Development of Antimalarial Drugs
N
N
Cl
HO
OH O O OH OH
HO Cl
H2N
Cl H H
Cl O
N OH
CF3 Cl
H
O O
H
O HO
O O
H
HO O
O
O H
HO
O O
OH
O
O Cl
Cl
H2N
O
H N
N O
S
O2 H2N N NH2
N N
Sulfadoxine Pyrimethamine
We have discussed various quinoline-based antimalarial drugs as they are the most extensively
studied classes. Other classes of antimalarial drugs and their modes of action are listed in Table
1.1. These antimalarials are often used in combination with schizonticides to ensure the
53
Chapter 1 Development of Antimalarial Drugs
CQ resistance first appeared in the late ’50s and early ’60s in Southeast Asia and South America.
Resistance is a result of sustained drug pressure in a geographical region. Resistance may also
arise due to monotherapy, incorrect dosage, self-medication, and the administration of cheap
counterfeit drugs.
In the year 2000, the primary determinant for CQ resistance was identified.80,81 PfCRT is a 48 kDa
protein located on the membrane of the DV. It is a member of the drug – metabolite transporter
(DMT) superfamily, Figure 1.15.80,82 It is a monomeric transmembrane (TM) protein that consists
of 10 TM helices and two juxtamembrane helices: one is located parallel to the parasite cytosol
54
Chapter 1 Development of Antimalarial Drugs
a) b)
Figure 1.15. Structure of PfCRT 7G8 isoform. A) Topology showing inverted antiparallel TM
helices. B) Surface representation of the central cavity’s electrostatic potential with red and blue,
indicating negative and positive residues, respectively. Figures are taken from Kim et al.83
The TM domains form 5 helical pairs, and TM1-4 and 6-9 form a central cavity of around 3,300
Å3. The point mutation K76T has become a molecular marker for resistance. This mutation
replaces the positively charged lysine with a neutral threonine at the 76 position in the
transmembrane domain.80 The presence of a neutral residue removes the cation – cation repulsion
between protonated CQ and the lysine residue, speculated to prevent CQ efflux from the DV,
Figure 1.16. Consequently, the mutation allows CQ to diffuse out of the DV down its
concentration gradient and reduces CQ accumulation in its active site.84 The cavity has a net
negative charge influenced by three aspartate residues, suggesting possible substrates for the
55
Chapter 1 Development of Antimalarial Drugs
Figure 1.16. K76T mutation removes a positive charge, which allows CQ2+ efflux from its active
site.
It has been proposed that PfCRT functions as a channel or transporter that mediates the efflux of
CQ out of the DV. In the channel model, PfCRT allows protonated CQ to passively diffuse out the
DV down its concentration gradient, Figure 1.16. However, in the transporter model, PfCRT
functions as an energy coupled transporter that actively effluxes CQ out of the DV.85,86 It is
believed that at low external concentrations of CQ, CQR parasites accumulate up to 10 – fold less
CQ than CQS parasites. This is achieved through the enhanced efflux capacity of the resistance
resistance protein (pfmdr1). pfmdr1 is a 162 kDa protein that belongs to the ABC (ATP – binding
cassette) superfamily. This protein is structurally similar to the mammalian multidrug resistance
transporter P-glycoprotein. It comprises two homologous halves, which contain six TM domains
56
Chapter 1 Development of Antimalarial Drugs
with one nucleotide-binding domain (NBD), Figure 1.17.88 pfmdr1 is localized to the DV
membrane and is believed to behave like an energy - coupled transporter that regulates drug influx
It contains point mutations N86Y, Y184F, S1034C, N1042D, and D1246Y associated with
resistance to AQs and aryl aminoalcohol antimalarial drugs. These mutations arose as a
consequence of drug pressure, and their expression varies with geographical region. For instance,
N84Y is mainly found in Southeast Asian isolates, and the remaining mutations are primarily
found in South American isolates.88 Although these mutations are associated with antimalarial
resistance, it is not believed that they are the sole cause of resistance. It has been shown that the
results in decreased sensitivity to MQ and vice versa. Cross-resistance is also observed with
halofantrine, lumefantrine and artemether. Therefore, the overexpression of pfmdr1 and the
drugs.84,91 Mutations may reduce pfmdr1s ability to transport drugs into the DV, resulting in the
57
Chapter 1 Development of Antimalarial Drugs
Figure 1.17 Structure of pfmdr1 with highlighted mutations: N86Y, Y184F, S1034C, N1042D,
D1246Y and two nucleotide-binding domains. Illustration adapted from Valderramos and
Fidock.91
1.7 Vaccines
Malaria vaccine development is a long sorry saga of disappointment. Naturally acquired immunity
from the disease arises after sustained repeated exposure to the parasite to maintain immunity.92
Vaccine development has been complicated because the immune response to P. falciparum
infection is not well understood. There are 5,300 parasitic antigens that elicit an immune response;
however, it is unclear which antigens are responsible for a protective response.92 Vaccine
development is currently prioritizing administration to children under the age of five, as they
58
Chapter 1 Development of Antimalarial Drugs
1.7.1 RTS,S
In 1985 GlaxoSmithKline and the Walter Reed Army Institute of Research began testing and
developing a pre-erythrocytic vaccine named RTS,S which protects against sporozoites. The DNA
hepatitis B surface antigen DNA to produce RTS. RTS binds hepatitis B surface antigens (S) to
form RTS,S particles. Then a proprietary adjuvant system AS01 (a mixture of deacylated
monophosphoryl lipid A and an emulsion QS21) is mixed with the particles to form a vaccine
Clinical trials carried out in Mozambique, Kenya and Tanzania on children aged 5 – 17 months
show that RTS,S/AS01 only provides partial immunity. Up to four doses reduced clinical cases by
56% and severe malaria cases by 47%.95 However, vaccine efficacy decreases over time. In a trial
involving children 6 – 12 weeks old, the vaccine has an initial efficacy of 33%.4 Gambian adults
given RTS,S with adjuvant AS02 over 15 weeks showed a 34% efficacy, but most importantly,
efficacy is high at 71% in the first nine weeks, but it drops to 0% in the following six weeks.92,93
It is believed that the vaccine’s low and unsustainable efficacy is because it does not induce a
memory T cell immune response for long-term protection.94 Despite the lack of long-term
In 2019 the Malaria Vaccine Implementation Program launched in Ghana, Malawi, and Kenya.
Children aged 6 – 24 months are given a dose at 6, 7, 9 and 24 months in Kenya and Ghana. In
Malawi children aged 5 – 22 months are given a dose at 5, 6, 7 and 22 months.96–98 In the following
59
Chapter 1 Development of Antimalarial Drugs
Over the last century, there have been significant advances in the development of antimalarial
drugs. However, the rise of resistance has hindered progress towards the eradication of the disease.
More than ever, we require more tools for treating and preventing malaria as the number of cases
and deaths increases year over year. We need to develop effective drugs that can circumvent
resistance for known targets and develop drugs for new targets. Vaccine development has been
challenging, however successful implementation of RTS,S along with ACT treatment can help
drive down infection rates. The use of insecticide-treated mosquito nets has proven successful,
therefore ensuring that a greater proportion of at-risk populations have access to these can help
reduce the spread of the disease. The road to eradicating malaria has been long and strenuous;
however, we must sustain research efforts if we are to meet this ambitious goal.
60
Chapter 1 Development of Antimalarial Drugs
1.8 References
(3) Barnes, K. I. Antimalarial Drugs and the Control and Elimination of Malaria. In Treatment
(4) Phillips, M. A.; Burrows, J. N.; Manyando, C.; van Huijsduijnen, R. H.; Van Voorhis, W.
(6) Josling, G. A.; Llinás, M. Sexual Development in Plasmodium Parasites: Knowing When
(8) Francis, S. E.; Sullivan, D. J.; Goldberg, and D. E. Hemoglobin Metabolism In The Malaria
(9) Spiller, D. G.; Bray, P. G.; Hughes, R. H.; Ward, S. A.; White, M. R. . The PH of the
1013.
61
Chapter 1 Development of Antimalarial Drugs
(12) Gorka, A. P.; de Dios, A.; Roepe, P. D. Quinoline Drug–Heme Interactions and Implications
for Antimalarial Cytostatic versus Cytocidal Activities. J. Med. Chem. 2013, 56 (13), 5231–
5246.
In Biopolymers Online; Matsumura, S., Steinbüchel, A., Eds.; Wiley-VCH Verlag GmbH
(14) Rosenthal, P.; Meshnick, S. R. Hemoglobin Catabolism and Iron Utilization by Malaria
(17) Rosenthal, P. J. Falcipains and Other Cysteine Proteases of Malaria Parasites. Adv. Exp.
(18) Egan, T. J. Haemozoin Formation. Mol. Biochem. Parasitol. 2008, 157 (2), 127–136.
(19) Pagola, S.; Stephens, P. W.; Bohle, D. S.; Kosar, A. D.; Madsen, S. K. The Structure of
(20) Casabianca, L. B.; Kallgren, J. B.; Natarajan, J. K.; Alumasa, J. N.; Roepe, P. D.; Wolf, C.;
(21) Nair, K. P. P. Cinchona (Cinchona Sp.). Agron. Econ. Important Tree Crop. Dev. World
62
Chapter 1 Development of Antimalarial Drugs
2010, 111–129.
(22) Sullivan, D. J. Cinchona Alkaloids: Quinine and Quinidine. In Treatment and Prevention
(23) World Health Organization. World Health Organization Model List of Essential Medicines.
(24) Kaur, K.; Jain, M.; Reddy, R. P.; Jain, R. Quinolines and Structurally Related Heterocycles
(25) Teixeira, C.; Vale, N.; Pérez, B.; Gomes, A.; Gomes, J. R. B.; Gomes, P. “Recycling”
Classical Drugs for Malaria. Chem. Rev. 2014, 114 (22), 11164–11220.
(26) Vale, N.; Moreira, R.; Gomes, P. Primaquine Revisited Six Decades after Its Discovery.
(27) Staines, H. M.; Krishna, S. Treatment and Prevention of Malaria: Antimalarial Drug
Chemistry, Action and Use. Treat. Prev. Malar. Antimalar. Drug Chem. Action Use 2012,
1–315.
(29) Tafenoquine Approved for Malaria Prophylaxis and Treatment | Travelers’ Health | CDC
https://wwwnc.cdc.gov/travel/news-announcements/tafenoquine-malaria-prophylaxis-and-
treatment.
(30) Kitchener, S.; Nasveld, P.; Edstein, M. D. Short Report: Tafenoquine for the Treatment of
Recurrent Plasmodium Vivax Malaria. Am. J. Trop. Med. Hyg. 2007, 76 (3), 494–496.
(31) Frampton, J. E. Tafenoquine: First Global Approval. Drugs 2018, 78 (14), 1517–1523.
63
Chapter 1 Development of Antimalarial Drugs
(33) Agents, A. Mechanism of Action of Antimicrobial and Antitumor Agents; Corcoran, J. W.,
Hahn, F. E., Snell, J. F., Arora, K. L., Eds.; Springer Berlin Heidelberg: Berlin, Heidelberg,
1975.
(34) Van Dyke, K.; Lantz, C.; Szustkiewicz, C. Quinacrine: Mechanisms of Antimalarial Action.
(35) Elueze, E. I.; Croft, S. L.; Warhurst, D. C. Activity of Pyronaridine and Mepacrine against
(3), 511–518.
(36) Coatney, G. R. Pitfalls in a Discovery: The Chronicle of Chloroquine *. Am. J. Trop. Med.
(39) Cohen, S. N.; Phifer, K. O.; Yielding, K. L. Complex Formation between Chloroquine and
Ferrihæmic Acid in Vitro, and Its Effect on the Antimalarial Action of Chloroquine. Nature
(40) Sullivan, D. J. Quinolines Block Every Step of Malaria Heme Crystal Growth. Proc. Natl.
(41) Nsumiwa, S.; Kuter, D.; Wittlin, S.; Chibale, K.; Egan, T. J. Structure–Activity
Aminoquinolines Using Experimental and Ab Initio Methods. Bioorg. Med. Chem. 2013,
64
Chapter 1 Development of Antimalarial Drugs
21 (13), 3738–3748.
(42) Dodd, E. L.; Bohle, D. S. Orienting the Heterocyclic Periphery: A Structural Model for
(43) Gorka, A. P.; Sherlach, K. S.; De Dios, A. C.; Roepe, P. D. Relative to Quinine and
Quinidine, Their 9-Epimers Exhibit Decreased Cytostatic Activity and Altered Heme
Binding but Similar Cytocidal Activity versus Plasmodium Falciparum. Antimicrob. Agents
(44) Alumasa, J. N.; Gorka, A. P.; Casabianca, L. B.; Comstock, E.; De Dios, A. C.; Roepe, P.
D. The Hydroxyl Functionality and a Rigid Proximal N Are Required for Forming a Novel
(45) Egan, T. J.; Hunter, R.; Kaschula, C. H.; Marques, H. M.; Misplon, A.; Walden, J.
(46) Vippagunta, S. R.; Dorn, A.; Matile, H.; Bhattacharjee, A. K.; Karle, J. M.; Ellis, W. Y.;
Related to Inhibition of Hematin Polymerization and Parasite Growth. J. Med. Chem. 1999,
42 (22), 4630–4639.
(47) Bray, P. G.; Park, B.; Asadollaly, E.; Biagini, G.; Jeyadevan, J.; Berry, N.; Ward, S.; O’
(48) Kaschula, C. H.; Egan, T. J.; Hunter, R.; Basilico, N.; Parapini, S.; Taramelli, D.; Pasini,
65
Chapter 1 Development of Antimalarial Drugs
Role of the Group at the 7-Position. J. Med. Chem. 2002, 45 (16), 3531–3539.
(49) Hawley, S. R.; Bray, P. G.; Mungthin, M.; Atkinson, J. D.; O’Neill, P. M.; Ward, S. A.
42 (3), 682–686.
and Amodiaquine: A Narrative Review. Iran. J. Med. Sci. 2017, 42 (2), 115–128.
(52) O’Neill, P. M.; Bray, P. G.; Hawley, S. R.; Ward, S. A.; Kevin Park, B. 4-Aminoquinolines
- Past, Present, and Future: A Chemical Perspective. Pharmacol. Ther. 1998, 77 (1), 29–58.
(54) Shinjo, S. K.; Maia Júnior, O. O.; Tizziani, V. A. P.; Morita, C.; Kochen, J. A. L.; Takahashi,
(55) Silva, J. A.; Silva, M. B.; Skare, T. L. Chloroquine and QTc Interval. Clin. Exp. Rheumatol.
(57) Ben-Zvi, I.; Kivity, S.; Langevitz, P.; Shoenfeld, Y. Hydroxychloroquine: From Malaria to
(58) Hawley, S. R.; Bray, P. G.; Park, B. K.; Ward, S. A. Amodiaquine Accumulation in
66
Chapter 1 Development of Antimalarial Drugs
(59) Parikh, S.; Ouedraogo, J. B.; Goldstein, J. A.; Rosenthal, P. J.; Kroetz, D. L. Amodiaquine
(60) Shimizu, S.; Atsumi, R.; Itokawa, K.; Iwasaki, M.; Aoki, T.; Ono, C.; Izumi, T.; Sudo, K.;
(61) O′Neill, P. M.; Harrison, A. C.; Storr, R. C.; Hawley, S. R.; Ward, S. A.; Park, B. K. The
(62) Warhurst, D. C.; Craig, J. C.; Adagu, I. S.; Guy, R. K.; Madrid, P. B.; Fivelman, Q. L.
(63) Davis, T. M. E.; Hung, T.-Y.; Sim, I.-K.; Karunajeewa, H. A.; Ilett, K. F. Piperaquine.
(64) Biot, C.; Glorian, G.; Maciejewski, L. A.; Brocard, J. S.; Domarle, O.; Blampain, G.; Millet,
P.; Georges, A. J.; Abessolo, H.; Dive, D.; Lebibi, J. Synthesis and Antimalarial Activity in
(23), 3715–3718.
(65) Biot, C.; Chavain, N.; Dubar, F.; Pradines, B.; Trivelli, X.; Brocard, J.; Forfar, I.; Dive, D.
67
Chapter 1 Development of Antimalarial Drugs
Evaluate the Mechanism of Action of Ferroquine. J. Organomet. Chem. 2009, 694 (6), 845–
854.
(66) Biot, C.; Taramelli, D.; Forfar-Bares, I.; Maciejewski, L. A.; Boyce, M.; Nowogrocki, G.;
Brocard, J. S.; Basilico, N.; Olliaro, P.; Egan, T. J. Insights into the Mechanism of Action
(67) Biot, C.; Pradines, B.; Dive, D. Ferroquine: A Concealed Weapon. Apicomplexan Parasites
(68) Berliner, R. W.; Earle, D. P.; Taggart, J. V.; Zubrod, C. G.; Welch, W. J.; Conan, N. J.;
Bauman, E.; Scudder, S. T.; Shannon, J. A. Studies On The Chemotherapy Of The Human
(69) Cogswell, F. B.; Krogstad, D. J.; De, D.; Krogstad, F. M. Aminoquinolines That Circumvent
Resistance in Plasmodium Falciparum in Vitro. Am. J. Trop. Med. Hyg. 1996, 55 (6), 579–
583.
(70) Mengue, J. B.; Held, J.; Kreidenweiss, A. AQ-13 - an Investigational Antimalarial Drug.
(71) Achan, J.; Talisuna, A. O.; Erhart, A.; Yeka, A.; Tibenderana, J. K.; Baliraine, F. N.;
(72) Woodward, R. B.; Doering, W. E. The Total Synthesis Of Quinine. J. Am. Chem. Soc. 1944,
66 (5), 849–849.
68
Chapter 1 Development of Antimalarial Drugs
(74) Palmer, K. J.; Holliday, S. M.; Brogden, R. N. Mefloquine. Drugs 1993, 45 (3), 430–475.
(76) Nosten, F.; Phillips-Howard, P. A.; ter Kuile, F. O. Other 4-Methanolquinolines, Amyl
(77) Gaillard, T.; Madamet, M.; Pradines, B. Tetracyclines in Malaria. Malar. J. 2015, 14 (1),
445.
(79) Mungthin, M.; Rungsihirunrat, K.; Na-Bangchang, K.; Sibley, C. H.; Hawkins, V. N.
(80) David A. Fidock; Nomura, T.; Talley, A. K.; Cooper, R. A.; Dzekunov, S. M.; Ferdig, M.
T.; Ursos, L. M. B.; Sidhu, A. bir S.; Naudé, B.; Deitsch, K. W.; Su, X.; Wootton, J. C.;
Transmembrane Protein PfCRT and Evidence for Their Role in Chloroquine Resistance.
(81) Djimdé, A.; Doumbo, O. K.; Cortese, J. F.; Kayentao, K.; Doumbo, S.; Diourté, Y.;
Coulibaly, D.; Dicko, A.; Su, X.; Nomura, T.; Fidock, D. A.; Wellems, T. E.; Plowe, C. V.
(82) Cooper, R. A.; Hartwig, C. L.; Ferdig, M. T. Pfcrt Is More than the Plasmodium Falciparum
69
Chapter 1 Development of Antimalarial Drugs
(83) Kim, J.; Tan, Y. Z.; Wicht, K. J.; Erramilli, S. K.; Dhingra, S. K.; Okombo, J.; Vendome,
J.; Hagenah, L. M.; Giacometti, S. I.; Warren, A. L.; Nosol, K.; Roepe, P. D.; Potter, C. S.;
Carragher, B.; Kossiakoff, A. A.; Quick, M.; Fidock, D. A.; Mancia, F. Structure and Drug
Resistance of the Plasmodium Falciparum Transporter PfCRT. Nature 2019, 576 (7786),
315–320.
(84) Ibraheem, Z. O.; Abd Majid, R.; Noor, S. M.; Sedik, H. M.; Basir, R. Role of Different Pfcrt
(85) Sanchez, C. P.; Stein, W. D.; Lanzer, M. Is PfCRT a Channel or a Carrier? Two Competing
(86) Sanchez, C. P.; Dave, A.; Stein, W. D.; Lanzer, M. Transporters as Mediators of Drug
(2), 163–171.
(88) Sidhu, A. B. S.; Valderramos, S. G.; Fidock, D. A. Pfmdr1 Mutations Contribute to Quinine
(89) Duraisingh, M. T.; Cowman, A. F. Contribution of the Pfmdr1 Gene to Antimalarial Drug-
(90) Koenderink, J. B.; Kavishe, R. A.; Rijpma, S. R.; Russel, F. G. M. The ABCs of Multidrug
70
Chapter 1 Development of Antimalarial Drugs
(92) Moorthy, V. S.; Good, M. F.; Hill, A. V. S. Malaria Vaccine Developments. Lancet 2004,
(93) Casares, S.; Brumeanu, T.-D.; Richie, T. L. The RTS,S Malaria Vaccine. Vaccine 2010, 28
(31), 4880–4894.
(94) Crompton, P. D.; Pierce, S. K.; Miller, L. H. Advances and Challenges in Malaria Vaccine
(95) Mehta, S. R. et al. First Results of Phase 3 Trial of RTS,S/AS01 Malaria Vaccine in African
https://www.who.int/immunization/diseases/malaria/malaria_vaccine_implementation_pro
gramme/RTSS_Kenya_Web_FINAL_August_2019.pdf.
https://www.who.int/immunization/diseases/malaria/malaria_vaccine_implementation_pro
gramme/RTS_S_Malawi_04_19.pdf.
https://www.who.int/immunization/diseases/malaria/malaria_vaccine_implementation_pro
gramme/RTS_S_Ghana_4_19.pdf.
71
Chapter 2
Chloroquine Derivatization.
72
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
2.1 Preamble
In this chapter, we explore the synthesis of CQ functionalized at the 3-position with an amine.
There are only a few reports on the syntheses of 3-substituted CQ analogs in the literature for
malaria treatment.1–4 Most reported examples of 3-substituted CQ were for Leishmania spp. and
ocular melanoma treatment.5,6 The shortage of examples of such compounds might be due to their
lack of synthetic access and preceding literature that reported a decrease in antimalarial activity
this position because previous work carried out in our lab showed promising results in the
combination therapy between CQ and 3-ICQ.7,8 Moreover, substitution at this position will also
2.2 Introduction
The quinoline scaffold has been one of the most significant in antimalarial drug history.9–12 Some
of the most important antimalarial drugs are quinolines, such as CQ, PPQ and PQ. These AQs have
two substituents on the quinoline ring: a secondary amine group and a chloro- or methoxy group.
It was proposed that modifications to the quinoline ring, such as replacing the 7-chloro group
binding affinity to b-hematin.13 The presence of an amine group at the 3-, 5-, 6- or 8-position
decreased activity, and these simple AQs do not form a significant association with b-hematin,
Figure 2.1. However, 2-aminoquinolines (AQs) form a complex with b-hematin but do not inhibit
73
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
NH2
NH2
N N N
N N
NH2
These findings deterred other research groups from synthesizing compounds with such
modifications. It is important to note that the aforementioned AQs do not include the 4-amino and
7-chloro moieties required for antimalarial activity. Tim Egan’s group has shown that the 7-chloro
group is crucial for efficient b-hematin association and inhibition.13 Furthermore, the secondary 4-
amino group is essential for antiplasmodial activity as its tautomeric form contributes to the drug’s
activity.15,16 One must also keep in mind that although modifications on the ring will impact the
compound’s association constant with b-hematin, research also shows no direct correlation
between b-hematin inhibition and antiplasmodial activity.15 Such compounds interact with b-
hematin in varying conformations. Furthermore, it is highly probable that b-hematin is not the sole
compound’s ability to accumulate in the DV and to inhibit b-hematin formation, which are both
74
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
As will be discussed in the next chapter, 3-haloCQ derivatives have been synthesized previously
in our group. These derivatives contain a halogen group at the 3-position of the quinoline ring. The
3-haloCQ derivatives are less potent than CQ in CQ-sensitive (CQS) and CQ-resistant (CQR)
parasites, they inhibit b-hematin formation but to a lesser extent than CQ. Interestingly, it was
found that of the 3-haloCQs synthesized, 3-ICQ was the most potent. When given as a combination
treatment with CQ in CQR parasites, 3-ICQ resensitizes the parasite to CQ. Thus, combination
For these reasons, we decided to explore further how functionality at the 3-position affects activity.
In this chapter, I will discuss the installation of an amino-functional group, which allows for further
derivatization to benzamides, sulfonamides and Schiff bases. To install the Csp2-N bond at this
75
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
Bromination of CQ has been previously reported in our lab using trifluoroacetic acid and N-
addition. The reaction mixture is then stirred at 80℃ for 2 hours to yield 73-81% of 3-BrCQ
following workup. However, the yield is greatly reduced when the reaction is scaled to gram scale
where the yields are consistently below 30%. Thus, we optimized this reaction to achieve
We carried the following reactions under microwave-assisted synthesis. Initially, we carried out a
small-scale reaction in 0.5 mL acetic acid, and the reaction mixture is heated at 80℃ for 30 minutes
(Table 2.1, entry 2). This results in an excellent yield of the desired product and the formation of
product. Moreover, when the reaction is then carried out on the gram scale in 4.5 mL acetic acid
at 85℃ for 40 minutes, 1H NMR analysis shows 32% consumption of starting material with 41:59
mono-: di- selectivity. An additional equiv. of NBS was added, and the reaction is heated at 80℃
for an additional 30 minutes. This results in 100% consumption of starting material with
regioselectivity of 48:52 (mono-: di-) with an isolated yield of 21% of the mono-brominated
product (entry 1). The poor yield and regioselectivity were unsatisfactory, which may be due to
the acidic environment which favours the activation of the 6-position due to the 7-chloro group
76
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
with is an ortho-director. Therefore, we carried out the reaction in acetonitrile on a small scale
1
H NMR analysis shows 64% conversion of starting material with a regioselectivity of 96:4 (mono-
: di-), and we isolated 96% of the desired mono-brominated product. The reaction is then scaled
up to 1 gram with 5 mL acetonitrile in a 5 mL microwave vial heated at 80℃ for 1 hour. After 1
hour, the TLC shows that the starting material is still present; therefore, the vial is returned to the
microwave and set at 85℃ for 40 minutes. Thereafter, 1H NMR shows 39% conversion of the
77
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
starting material with regioselectivity of 100:0 mono-: di-. An additional equiv. of NBS is added,
and the reaction runs at 100℃ for an additional 30 minutes. The 1H NMR analysis then shows
94% consumption of starting material to only the desired mono-BrCQ. However, we got an
isolated yield of 40%. The lower than expected yield may be due to side product formation
stemming from the high reaction temperature. As the yields were still low for the gram scale
reactions, we decided to carry out a gram-scale reaction with CQ diphosphate rather than the free
base (entry 5). The reaction with CQ salt is carried out at 100℃ for 1 hour to give an isolated yield
of 64%. The 1H NMR conversion and regioselectivity for this reaction were not measured before
purification; however, only one product spot is observed on the TLC. It is important to note that
the salt does not fully dissolve in acetonitrile at 100℃, and the starting material is visible in the
vial after the reaction is removed from the microwave, reducing the potential yield. To determine
the effect of temperature on the reaction, free base CQ is placed in a microwave vial with 1.5
equiv. NBS and 5 mL acetonitrile, set at 100℃ for 1 hour (entry 6). After the time elapsed, the
starting material is still present as verified by TLC and 0.5 equiv. NBS is added, and the reaction
is run at 100℃ for an additional 30 minutes. Finally, in an attempt to further push the reaction to
completion, it is run at 140℃ for 10 minutes; thus, the reaction was carried out from 100-140℃
for a total of 100 minutes. 1H NMR analysis of the crude mixture shows 36% conversion of the
starting material with regioselectivity 100: 0 (mono-: di-), and we are able to isolate 40% of the
product.
This bromination reaction works best on a small scale to form the 3-BrCQ as the major product
with a minor formation of 3,6-BrCQ. Once the reaction is scaled up in acetic acid, we obtain greater
formation of 3,6-BrCQ, with 3-BrCQ as the minor product. However, we get better regioselectivity
78
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
in acetonitrile. Similarly, small scale reactions in acetonitrile result in a high yield of our desired
product. When CQ.2H3PO4 is used, we are able to get good yields at gram scale with a yield of
64%. Finally, when higher temperatures are used for CQ free-base in acetonitrile, the reaction
As we achieved good yields with gram scale bromination, we continued to the synthesis of 3-
aminochloroquine (3-NH2CQ). The formation of the heteroatom- Csp2 bond occurs under Ullmann-
type coupling reaction conditions,17,18 the mechanism for this cross-coupling is described later in
this chapter.
3-NH2CQ can be synthesized using the linear route displayed in Scheme 2.1. We began with the
2.5. Then nitration at the 3-position with nitric acid to give 2.6, followed by chlorination with
side chain 2.8 gives 3-NO2CQ 2.9, which is then reduced to 3-NH2CQ 2.10 with stannous
chloride.19 This route involves 5 steps carried out over 5 days; thus, there was a need to create a
faster synthetic route to 3-NH2CQ. We achieved this by creating a 2-step synthesis carried out in
reaction.
79
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
Cl OH HNO3, OH
Acetic Acid, Propionic acid,
NO2
125oC, 1h 140oC, 18h
Cl N Cl N Cl N
2.4 2.5 2.6
Phosphoryl Chloride,
115oC, 18h
The Ullmann-type reaction is a coupling reaction that involves a copper catalyst and an aryl halide
with various heteroatoms to form a heteroatom - Csp2 bond, with Cu(I) as the active species in this
reaction. However, Cu(0) and Cu(II) salts are used with a base or reducing agent for in situ
reduction/oxidation to generate the active Cu(I) species.17 The use of a reducing agent such as
sodium ascorbate was required to obtain higher yields of the amine. The omission of the reducing
agent did not result in azide formation, It is desirable that we use easily accessible, cheap reagents
for this coupling reaction because antimalarial drug candidates used for treatment in developing
countries require cost-efficient production. We first began with ammonium hydroxide as the amine
source under various conditions with/ without the presence of a ligand or base, Table 2.2.
80
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
Et2N Et2N
NH Cu, Ligand, NH
Br Base NH2
NH4OH
Cl N Cl N
2.2 2.10
Reducing
Catalyst Temperature Yield
Entry Ligand [mol%] agent/base Solvent
[mol%] (oC) (%)
[mol%]
1 Cu [20] - - EtOH 95 -
L-Ascorbic 2M NH3
2 CuI [20] DMEDA [200] 95 Tracea
Acid [200] in EtOH
Cu2O Diethylenetriamine
3 KOH [200] H2 O 100 -
[10] pentaacetic acid [20]
Sodium
Cu2O Trans +/- Diamino 7N NH3
4 ascorbate 95 -
[100] cyclohexane [20] in MeOH
[200]
a
: 4% conversion of starting material to CQ determined by 1H NMR.
Copper-catalyzed amination reactions from aryl halides using ammonium hydroxide have been
well reported in the literature, Scheme 2.2.20–22 Lang et al. reported the amination of 2-
bromopyridine without the need of a ligand or base.21 They report that amination worked best in
an aqueous or alcoholic solvent; however, they see the formation of a C-O byproduct due to
competing hydroxide attack from the solvent. When polar, aprotic solvents were used, they had
low product conversion, which was attributed to ammonia’s poor solubility in such solvents.
Similarly, Jiao et al. carried out the amination reaction on 3- and 4-bromopyridine and achieved
near quantitative yields with copper powder, exposure to air and optional use of a Lewis base.20
Yang et al. were able to successfully carry out amination of 2- and 3-bromopyridine using a ligand
81
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
Cu,
H
Br CH3NH2 N
CuO, Ligand,
Br NH3.H2O NH2
In our experimentation, the use of ammonia did not yield any desired product (Table 2.2, entries
2 and 4). The reaction is monitored by TLC for up to 24 hours, and if no product spots were
detected by TLC, the reaction was deemed unsuccessful. The use of copper powder in degassed
ethanol (EtOH) was unsuccessful, so was the use of copper(I) oxide with a base or reducing agent.
Dimethylethylenediamine (DMEDA) as a ligand and excess L-ascorbic acid as the reducing agent
for CuI results in trace amounts of the desired product (<2% conversion) and interestingly results
We then decided to use an organic azide, azidotrimethyl silane (TMSN3), to improve the solubility
of our amine source in organic solvents and increase the reaction yield. As expected, the control
experiment of 3-BrCQ in 1.5 equiv. TMSN3 (Table 2.3, entry 10), heated at 53℃ overnight, did
82
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
not yield any product. We then used CuI with an excess of potassium carbonate (K2CO3) in 1,4-
dioxane; however, no product spots were detected on TLC (entry 6). The absence of a ligand gives
a low yield of 11% (entry 5), and the yield is not significantly improved when L-proline (entry 4)
or trans +/- diamino cyclohexane (entry 7) are used as ligands. When a stoichiometric amount of
copper powder is used with excess 2-aminoethanol as a ligand, and excess sodium ascorbate (entry
8) as the reducing agent in dimethylacetamide (DMA) are used, we observe a significant increase
in yield.23 The 2-aminoethanol ligand is essential for this reaction as there is no product formation
in its absence after heating at 95℃ for 23 hours when used with stoichiometric amounts of copper
powder (entry 8). We get similar results using catalytic amounts of copper powder with DMEDA
and sodium ascorbate in anhydrous EtOH (entry 2). The use of excess ligand combined with copper
iodide gave 1.7 - fold increase in yield (entry 1). A different organic azide, tetrabutylammonium
azide (NBu4N3), was used as an amine source (entry 11); however, no product was formed from
this reaction.
83
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
We then decided to carry out the amination reaction with sodium azide as the amine source. The
control reaction does not proceed in dimethylformamide (DMF) (Table 2.4, entry 6).24,25 The use
of copper sulfate with sodium ascorbate did not yield any product (entry 4),26,27 and Cu(I) iodide
with excess potassium carbonate did not yield any product (entry 2).28 When Cu(I) iodide was used
with bidentate ligand DMEDA in a degassed solution of 70% EtOH, 30% H2O at 95℃, we saw
product formation and were able to isolate 29% of the desired product.29–31 We repeated the
experiment in anhydrous EtOH with the addition of sodium ascorbate and were able to isolate 55%
84
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
of our desired product.27,32 Goriya and Ramana reported the direct amination of aryl halides with
sodium azide under a Cu(II)-ascorbate redox system.27 They reported that arylamine formation can
occur without the presence of copper and that various concentrations of sodium ascorbate with
copper(I) sulfate (Cu2SO4), sodium carbonate (Na2CO3) and L-proline greatly increases product
formation rate and decreases reaction time. However, in our system, we have found that copper is
required for this synthesis and the combination of DMEDA and sodium ascorbate produces the
highest yield.
Reducing
Catalyst Ligand Temperature Yield
Entry agent/base Solvent
[mol%] [mol%] (oC) (%)
[mol%]
DMEDA EtOH/H2O
1 CuI [20] - 95 29
[1200] (7:3)
2 CuI [20] - K2CO3 [200] EtOH (95%) 95 -
DMEDA Sodium EtOH/H2O
3 CuI [50] 85 55
[50] Ascorbate [50] (7:3)
CuSO4.H2O L-Proline Sodium DMF/H2O -
4 85
[20] [20] Ascorbate [20] (2:1)
Sodium
CuSO4.H2O DMEDA DMF/H2O
5 Ascorbate [30] 85 -
[20] [30] (2:1)
+ Na2CO3 [20]
6 - - - DMF 100 -
Surprisingly, conditions in Table 2.4 entry 3 results in the formation of the desired 3-aminoCQ
product and the reduction of CQ as a product. We then carried out experiments in an attempt to
minimize the reductive formation of CQ. Heating the reaction in an oil bath instead of microwave-
irradiation resulted in 50% conversion of the 3-BrCQ into 3-NH2CQ and 50% to CQ at 85℃. We
then ran the reaction at 75℃ for 2 hours (Table 2.5, entry 4). However, there was little product or
byproduct formation at this temperature. The temperature was then increased to 85℃, heated for
a further 3 hours and we were able to isolate 44% of the desired product. CQ was also formed, but
85
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
it was not collected during the purification. A longer reaction time of 19 hours (entry 3) did not
improve the selectivity nor the yield of the desired product. The reaction was carried out in the
agent. We got lower yields of both 3-NH2CQ and CQ but also slightly greater selectivity for 3-
NH2CQ. When ten equiv. of TEMPO is used, we got no formation of CQ and trace amounts of 3-
NH2CQ. The greatest selectivity and yield are achieved with three equiv. NaN3, 0.5 equiv. CuI,
0.5 equiv. DMEDA, and 0.5 equiv. sodium ascorbate at 85℃ in an oil bath for 3 hours.
86
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
CuI, DMEDA,
Et2N Na ascorbate, Et2N Et2N
NH EtOH/H2O (7:3), NH NH
Br 85oC NH2
Cl N Cl N Cl N
2.2 2.10 2.11
Reaction 3-NH2CQ
Heating Sodium Conversion
Entry time NaN3 CuI DMEDA Isolated Yield
method Ascorbate (NH2:CQ)
(hours) (%)
1 Oil 3 2 0.3 0.4 0.3 - 49
2 Oil 3 2 0.3 0.4 0.3 - 32b
3 Oil 19 2 0.3 0.4 0.3 - 50
4 Oilc 5 2 0.3 0.3 0.3 - 44
5 Oil 4 2 0.3 0.3 0.3 - Traced
6 Oil 3 3 0.5 0.5 0.5 - 55
7 MWe 0.7 3 3 3 2 91 (30:70) 16
8 MWf 0.5 3 3g 3 3 81 (37:63) 19
9 MW 1 3 0.5 0.5 0.5 69 (36:64) 26
10 MW 1 3 0.5 0.5 0.5 84 (51:49) NAh
a) Reactions run at 85℃ in EtOH /H2O (7:3). b) Reaction included 2 equiv. TEMPO. c) Starting
temperature of 75℃ after 2 hours temperature increased to 85℃. d) Reaction included 10 equiv.
TEMPO, starting temperature of 75℃, after 2 hours temperature increased to 85℃. e) Heated at
100℃ for 30 minutes and then at 140℃ for 10 minutes. f) Heated at 100℃ for 30 minutes. g) 3
equiv. of copper powder used. h) Purification not carried out; product contained 5 equiv. of
TEMPO.
We then carried out this coupling reaction via microwave-assisted synthesis (MW) with reduced
reaction times. When we replicated our optimized reaction conditions in the microwave, we saw a
2-fold decrease in yield (entry 9). One might assume that it may be due to reduced reaction times,
87
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
stoichiometric amounts of our reagents and increased the temperature from 85℃ to 140℃.
However, we obtain lower yields of 3-NH2CQ and greater conversion of starting material to CQ
(entry 7 and 8). We then carried out the reaction in the presence of 5 equiv. of TEMPO and
surprisingly saw 84% conversion of the starting material with a significantly improved selectivity
for 3-NH2CQ. Hence microwave-assisted synthesis is not recommended for these Ullmann-type
CuI, DMEDA,
Et2N Na ascorbate, Et2N
NH EtOH/H2O (7:3), NH
NH2 85oC, 3h
NaN3
Cl N Cl N
0%
2.10 2.11
We were then curious to determine the stability of the Csp2-N bond. We replicated the reaction
with 3-NH2CQ as our starting material with CuI, DMEDA, sodium ascorbate and sodium azide in
degassed EtOH /H2O (7:3), which is heated at 85℃ for 3 hours. A TLC is taken, and no CQ spots
are detected, and only the 3-NH2CQ starting material is present. This indicates that there is
competition for the formation of CQ and our desired amine, Scheme 2.3.
The reductive formation of the CQ byproduct was unexpected and reduced the yield of the desired
product. We reexamined the proposed catalytic cycle for Ullmann-type coupling reactions to
determine the cause. As shown in Figure 2.3 there are two proposed pathways for this coupling
reaction, with the upper cycle being the most widely accepted.17,33,34 Copper-catalyzed Ullmann-
88
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
type coupling is believed to occur via a Cu(I) / Cu(III) catalytic cycle, which is initiated by the
coordination of the nucleophile onto the Cu(I) center. This is followed by oxidative addition of the
aryl halide to form an NR2 - Cu(III) - aryl species and the release of the halide anion. The activation
of the aryl halide is believed to be the rate-limiting step. Reductive elimination then occurs to form
the NR2 - Ar coupling product and releases Cu(I) which can then reenter the cycle. The less
probable route is the initial oxidative addition of the aryl halide to form a halide - Cu(III) - aryl
intermediate, followed by coordination of the nucleophile via the release of the halide to form an
NR2 - Cu(III) - aryl species. Reductive elimination follows to release the product and Cu(I). We
believe that both pathways contribute to the formation of our desired 3-NH2CQ product and that
the lower cycle might be the main contributor to the formation of the CQ byproduct. The formed
ligand – Cu(III) – Ar species can undergo proton-coupled electron transfer causing the release of
the reductive CQ product. It is possible that the amines present in the 3-BrCQ side-chain act as
ligands on the Cu(I) center, and if oxidative addition occurs first, then all coordination sites could
Base-H
Ar-X Oxidative
L CuI NR2 Addition
R2NH + Base X-
Reductive elimination Ar
L CuI L CuIII
NR2
Ar-NR2
Ar-X
Ar Base-HX
Oxidative L CuIII
Addition X R2NH + Base
e- + H+
Proton-coupled
Electron Transfer
L-CuI-X
Ar-H
Figure 2.3. Proposed pathways for Cu(I)/ Cu(III) Ullmann-type amination catalytic cycle.
89
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
The 2-electron catalytic cycle is the most widely accepted mechanism as the reaction is not
inhibited with radical trapping agents or radical initiators.35 We tested this with the use of TEMPO
in both traditional oil heating and with microwave-assisted synthesis. When using an oil bath with
two equiv. of TEMPO we saw a slight reduction in product and byproduct formation. When ten
equiv. of TEMPO were used, we see no formation of byproduct and little formation of the desired
product. When five equiv. of TEMPO is used under microwave-assisted synthesis, we still observe
84% conversion to products. This suggests that the radical trap has little effect on the reaction
process. It is only when a large excess is used that we see an impact on product formation.
The mentioned Cu(I) / Cu(III) cycle is not the only proposed Ullmann-type mechanism. Another
proposed mechanism is a Cu(I) / Cu(II) Single Electron Transfer (SET) mechanism with aryl
radicals that might lead to product formation.36 Through experimentation with iodoarenes, Kim
and Bunnett proposed ‘outer sphere’ electron transfer through a radical chain mechanism involving
SET as the initiation step.36 They performed experiments in the presence of tetraphenylhydrazine
(a radical trapping agent) and proposed a unimolecular radical nucleophilic substitution (SRN1), as
SET
Initator + ArX (ArX) Ar + X
Ar + NH (ArNH2)
2
90
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
A proposed mechanism that does not involve a change in oxidation state is the p-complexation of
Cu(I) with the aryl halide, Figure 2.5. This coordination of Cu(I) with the aromatic ring activates
2.4 Conclusion
The 3-substituted quinolines are surprisingly poorly explored as antimalarial drug candidates. We
functionalize this position to explore the potential advantages and disadvantages of such a
substitution. In this chapter, the reaction optimization of 3-BrCQ while using fewer reagents and
decreasing reaction times was reported. We were able to scale up the reaction from 300 mg to 1
With 3-BrCQ in hand, we were able to synthesize 3-NH2CQ via Ullmann-type coupling. We
discovered that ammonium hydroxide is not a good amine source for this coupling reaction. We
then tested azidotrimethyl silane as an amine source because of its increased solubility in organic
solvents, and it proved to be a better candidate than NH4.OH. We determined that a base is not
91
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
required for this reaction; however, the presence of the reducing agent as sodium ascorbate or L-
ascorbic acid in combination with ligand DMEDA gave the best results, although the yields were
still poor. Tetrabutylammonium azide was also tested with DMEDA and sodium ascorbate;
however, no product was formed. Lastly, sodium azide was tested, and we were able to
byproduct. Therefore, we created a 2-step synthesis to form 3-NH2CQ from CQ in 35% overall
We concluded that the formation of 3-NH2CQ and CQ occurred mainly via two competing
mechanisms. It is possible that amine formation occurs when nucleophile coordination precedes
oxidative addition and vice versa for CQ. Currently, there is no exact proven mechanism for the
Ullmann-type coupling in the literature. There may be several mechanisms through which our
byproduct is formed. As we have synthesized 3-NH2CQ, we can now derivatize the 3 position
92
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
2.5 Experimental
All amination reactions were carried out in the presence of N2 gas by standard syringe and septa
techniques. Glassware was taken directly from the oven (120oC) and allowed to cool in a desiccator
before use. Bromination reactions were carried out in 2 mL and 5 mL Biotage Microwave Vials
under air. All solvents and reagents were obtained commercially and used without further
a Bruker Maxis Impact QTOF, or a Thermo Scientific ExactivePlus Orbitrap, with results reported
as mass/charge ratios (m/z). Thin-layer chromatography (TLC) plates were visualized using ultra-
violet light, 254 nm. Flash column chromatography was carried out manually using 230 - 400 mesh
silica gel (Silicycle) using reagent grade solvents or using Biotage Isolera One system with Biotage
ZIP 30 g columns.
1 13
H and C NMR spectra were recorded at ambient temperature on Bruker AVIIIHD 400 MHz
(1H 400 MHz, 13C 100 MHz), Bruker AVIIIHD 500 MHz (1H 500 MHz, 13C 125 MHz) and Bruker
AVIIIHD 800 MHz (1H 800 MHz, 13C 201 MHz) using tetramethylsilane as the internal standard.
Chemical shifts are reported relative to the residual deuterated solvent peaks. Chemical shifts are
expressed in parts per million (ppm = δ) values and coupling constants (J) in Hertz (Hz). The terms
93
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
experiments (COSY, HSQC, HMBC) was often used to complete the assignment of 1H and 13C
signals. 1H NMR signals are described by chemical shift δ (multiplicity, J (Hz), integration). 13C
NMR signals are described by chemical shift δ and are singlets unless otherwise specified.
flask is added sodium hydroxide (1.86 g, 0.16 M) to give a pH of 11/12. At this pH all the drug
has precipitated. This mixture is then transferred into a 1 L separatory funnel, and dichloromethane
(DCM) (200 mL) is added to extract the free base (repeated three times). The combined organic
layers are washed with brine and then dried over anhydrous magnesium sulfate (MgSO4). MgSO4
is filtered off, and the solvent is removed under reduced pressure. To the flask containing the free-
base, which appears as a colourless oil, is added 15 mL of diethyl ether and is swirled. The solvent
is then removed under reduced pressure, and the flask is transferred into a vacuum oven and left
literature.38
N4-(3-bromo-7-chloroquinolin-4-yl)-N1,N1-diethylpentane-1,4-diamine
O MW, 30 min
Cl N Cl N Cl N
Monobrominated CQ Dibrominated CQ
Chloroquine free-base (100 mg, 0.31 mmol), N-Bromosuccinimide (83.5 mg, 0.47 mmol) and 0.5
mL of acetonitrile or acetic acid are placed in a 2 mL Biotage microwave vial. The mixture is
94
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
subjected to microwave irradiation at 80℃ for 30 minutes. When complete, the reaction is diluted
in ethyl acetate (EtOAc) and basified with saturated sodium hydroxide and extracted with EtOAc
(3 X 50 mL) and washed with brine. The combined organic extracts are then dried over anhydrous
MgSO4, and the solvent is removed under reduced pressure. The crude product is purified by flash
chromatography with silica gel, solvent system: 70% hexanes (Hex), 25% EtOAc, 5%
N4-(3-bromo-7-chloroquinolin-4-yl)-N1,N1-diethylpentane-1,4-diamine (2.2)
N4-(3,6-dibromo-7-chloroquinolin-4-yl)-N1,N1-diethylpentane-1,4-diamine (2.3)
Yield: 119.4 mg, 8%. 1H NMR (400 MHz, Chloroform-d) δ 8.68 (s, 1H), 8.29 (s, 1H), 8.08 (s,
1H), 4.57 (d, J = 10.1 Hz, 1H), 4.00 (dq, J = 10.0, 6.2 Hz, 1H), 2.57 (q, J = 7.2 Hz, 4H), 2.47 (d, J
= 7.0 Hz, 2H), 1.63 (dtd, J = 13.2, 6.9, 3.8 Hz, 4H), 1.29 (d, J = 6.3 Hz, 3H), 1.04 (t, J = 7.2 Hz,
13
6H). C NMR (201 MHz, Chloroform-d) δ 153.01, 148.61, 148.14, 135.46, 130.84, 128.19,
121.88, 119.74, 107.54, 55.05, 52.55, 46.92, 36.67, 23.40, 22.39, 11.26. HRMS (ESI) calcd. for
(2.4) synthesis:
Cl N Cl N
95
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
Azide (2 equiv.), copper (0.5 equiv.), ligand (0.5 equiv.) and reducing agent (0.5 equiv.) or base
(2 equiv.) are placed in a 10 mL round bottom flask fitted with a septum. A needle connected to
the vacuum/N2 manifold was inserted, the flask is evacuated and backfilled with N2 gas, this
diethylpentane-1,4-diamine (1 equiv.) dissolved in degassed EtOH/ H2O (7:3 v/v) is added to the
flask via syringe through the septum. The reaction mixture is refluxed at 85℃ under N2 for 3 hours
while stirring vigorously. After the time elapses, the reaction mixture is left to cool down to room
temperature (RT). The reaction mixture is then diluted in H2O and basified with 1 M NaOH. It is
then extracted three times with DCM or EtOAc, and the combined organic phases are washed with
brine and dried over MgSO4. The solvent is then removed by reduced pressure and purified by
prep TLC 50% Hex, 40% EtOAc, 5% methanol (MeOH), 5% NEt3 to give an orange oil 331 mg,
55%. The 1H and 13C NMR was consistent with the literature.5
96
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
2.6 References
(2) Kinawi, A.; Dietz, M.; Schmidt, R.; Löwe, W. Kurzmitteilung. Bindung von Razemischen
(3) Pou, S.; Winter, R. W.; Nilsen, A.; Kelly, J. X.; Li, Y.; Doggett, J. S.; Riscoe, E. W.;
(7), 3475–3480.
(4) Bass, G. E.; Hudson, D. R.; Parker, J. E.; Purcell, W. P. Mechanism of Antimalarial Activity
(5) Konstantinović, J.; Videnović, M.; Orsini, S.; Bogojević, K.; D’Alessandro, S.;
Scaccabarozzi, D.; Terzić Jovanović, N.; Gradoni, L.; Basilico, N.; Šolaja, B. A. Novel
(6) van Langevelde, A.; Bakker, C. N. M.; Boer, H.; Ilmer, T. A. M.; Journée-de Korver, J. A.;
(7) Edaye, S.; Tazoo, D.; Bohle, D. S.; Georges, E. 3-Iodo-4-Aminoquinoline Derivative
97
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
(8) Edaye, S.; Tazoo, D.; Bohle, D. S.; Georges, E. 3-Halo Chloroquine Derivatives Overcome
(9) Kaur, K.; Jain, M.; Reddy, R. P.; Jain, R. Quinolines and Structurally Related Heterocycles
(10) Coatney, G. R. Pitfalls in a Discovery: The Chronicle of Chloroquine *. Am. J. Trop. Med.
(11) Okombo, J.; Chibale, K. Recent Updates in the Discovery and Development of Novel
(13) Kaschula, C. H.; Egan, T. J.; Hunter, R.; Basilico, N.; Parapini, S.; Taramelli, D.; Pasini,
Role of the Group at the 7-Position. J. Med. Chem. 2002, 45 (16), 3531–3539.
(15) Egan, T. J.; Hunter, R.; Kaschula, C. H.; Marques, H. M.; Misplon, A.; Walden, J.
(16) Natarajan, J. K.; Alumasa, J. N.; Yearick, K.; Ekoue-Kovi, K. A.; Casabianca, L. B.; De
98
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
Dios, A. C.; Wolf, C.; Roepe, P. D. 4-N-, 4-S-, and 4-O-Chloroquine Analogues: Influence
of Side Chain Length and Quinolyl Nitrogen PKa on Activity vs Chloroquine Resistant
(17) Ribas, X.; Güell, I. Cu(I)/Cu(III) Catalytic Cycle Involved in Ullmann-Type Cross-
(18) Sambiagio, C.; Marsden, S. P.; Blacker, A. J.; McGowan, P. C. Copper Catalysed Ullmann
Type Chemistry: From Mechanistic Aspects to Modern Development. Chem. Soc. Rev.
(19) Terzić, N.; Konstantinović, J.; Tot, M.; Burojević, J.; Djurković-Djaković, O.; Srbljanović,
J.; Štajner, T.; Verbić, T.; Zlatović, M.; Machado, M.; Albuquerque, I. S.; Prudêncio, M.;
Sciotti, R. J.; Pecic, S.; D’Alessandro, S.; Taramelli, D.; Šolaja, B. A. Reinvestigating Old
(20) Jiao, J.; Zhang, X. R.; Chang, N. H.; Wang, J.; Wei, J. F.; Shi, X. Y.; Chen, Z. G. A Facile
(21) Lang, F.; Zewge, D.; Houpis, I. N.; Volante, R. . Amination of Aryl Halides Using Copper
(22) Yang, B.; Liao, L.; Zeng, Y.; Zhu, X.; Wan, Y. A Simple and Recyclable Copper/DTPA
Catalyst System for Amination of Aryl Halides with Aqueous Ammonia in Water. Catal.
(23) Maejima, T.; Shimoda, Y.; Nozaki, K.; Mori, S.; Sawama, Y.; Monguchi, Y.; Sajiki, H.
99
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
(25) Peterson, J. R.; Zjawiony, J. K.; Liu, S.; Hufford, C. D.; Clark, A. M.; Rogers, R. D.
(26) Liu, Y.; Xiao, Q.; Liu, Y.; Li, Z.; Qiu, Y.; Zhou, G.-B.; Yao, Z.-J.; Jiang, S. Biological
Click Linkage with Aromatic Functionalities. Eur. J. Med. Chem. 2014, 78, 248–258.
(27) Goriya, Y.; Ramana, C. V. The [Cu]-Catalyzed SNAR Reactions: Direct Amination of
Electron Deficient Aryl Halides with Sodium Azide and the Synthesis of Arylthioethers
(28) Altman, R. A.; Buchwald, S. L. Cu-Catalyzed Goldberg and Ullmann Reactions of Aryl
Halides Using Chelating N- and O-Based Ligands. Nat. Protoc. 2007, 2 (10), 2474–2479.
(29) Markiewicz, J. T.; Wiest, O.; Helquist, P. Synthesis of Primary Aryl Amines Through a
Copper-Assisted Aromatic Substitution Reaction with Sodium Azide. J. Org. Chem. 2010,
75 (14), 4887–4890.
Catalyzed Coupling of Sodium Azide with Aryl Iodides/Boronic Acids to Aryl Azides or
(31) Sklorz, J. A. W.; Hoof, S.; Rades, N.; De Rycke, N.; Könczöl, L.; Szieberth, D.; Weber, M.;
100
Chapter 2 Optimization of 3-Aminochloroquine synthesis for Chloroquine Derivatization
(33) Sung, S.; Braddock, D. C.; Armstrong, A.; Brennan, C.; Sale, D.; White, A. J. P.; Davies,
Studies on Their Role in the Modified Ullmann Amination Reaction. Chem. - A Eur. J.
(34) Sperotto, E.; Van Klink, G. P. M.; Van Koten, G.; De Vries, J. G. The Mechanism of the
(35) Cohen, T.; Cristea, I. Kinetics and Mechanism of the Copper(I)-Induced Homogeneous
(36) Kim, J. K.; Bunnett, J. F. Evidence for a Radical Mechanism of Aromatic “Nucleophilic”
(37) Weingarten, H. Mechanism of the Ullmann Condensation. J. Org. Chem. 1964, 29 (12),
3624–3626.
(38) Sinha, M.; Dola, V. R.; Soni, A.; Agarwal, P.; Srivastava, K.; Haq, W.; Puri, S. K.; Katti,
101
Chapter 3
102
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
3.1 Preamble
Following the optimization of 3-NH2CQ synthesis, we wanted to derivatize the 3-position further.
The installed arylamine serves as a precursor for various derivatives such as secondary amines,
benzamides, sulfonamides, triazoles, and Schiff-bases. We were interested in the synthesis of such
derivatives as possible combination therapy partners for CQ. Additionally, b-hematin may not be
their only target. 3-substituted CQs may target other stages of the parasite's life cycle. As we know,
8-AQs such as PQ are active against liver-stage hypnozoites. We discovered that 3-haloCQs when
used in combination with CQ, resensitize resistance strains of the parasite to CQ. Furthermore, the
potencies of 3-haloCQ are unaffected by VP, which suggests that they are not substrates for mutant
PfCRT. This was an exciting discovery that inspired us to create a library of novel 3-substituted
CQs. We are interested in their drug targets and their ability to resensitize resistant parasites to
CQ. Artemisinin combination therapies (ACTs) are the recommended treatments for malaria,
which are beginning to fail in Asian countries. Insights into new combination therapies, especially
3.2 Introduction
As illustrated in chapter 1, the 4-aminoquinolines are the most studied of all the classes of
antimalarial drugs. Although the quinoline structure has been extensively modified, it still has
great potential, as seen with antimalarial drug candidates FQ and AQ-13. Both antimalarial
candidates were in phase II clinical trials but have not yet advanced to phase III.
Previous results from our group included the synthesis of 3-halo CQs: 3-ClCQ, 3-BrCQ and 3-
103
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
acetonitrile at 80oC for 30 minutes, followed by CQ addition. The reaction mixture is then stirred
at 80oC for 2 hours. Yields ranged between 73% - 81%. In vitro assays were carried out and it was
determined that each derivative is equally potent against CQS and CQR P. falciparum strains;
however, they were ~8.5 - fold less potent than CQ.1 These results are in-line with previous
structure-activity relationship (SAR) reports which showed that modifications to the quinoline ring
resulted in decreased antiplasmodial activity.2,3 In CQS strain 3D7, 3-haloCQ had a 19 - fold
decrease in potency compared to CQ. The IC50 values range between 367 nM - 747 nM in CQS
3D7 strain and 623 nM – 1163 nM in CQR strain Dd2, Scheme 3.1. Of the three derivatives
synthesized, 3-ICQ was the most potent with IC50 values of 367 nM and 623 nM in 3D7 and Dd2,
respectively.4
a) O 80oC, 2h
Cl N Cl N X= Cl, Br, I
Chloroquine 3-halo chloroquine
O
N O
b)
O N O
Verapamil
Furthermore, a b-hematin inhibition assay was carried out to determine 3-ICQ's ability to inhibit
b-hematin formation, and it was found that 3-ICQ does inhibit b-hematin formation but to a lesser
extent than CQ. This is possibly due to the presence of the electron-withdrawing iodine that lowers
104
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
the pKa of the quinolinium nitrogen (Table 3.3) and may reduce the compound's affinity to
Fe(III)PIX. These results are not surprising or unexpected as substituents on the quinoline ring
affect the drug's ability to interact with heme.5 A large, electron-withdrawing group such as iodine
Verapamil (VP), Scheme 3.1, is a chemosensitizer that presumably inhibits PfCRT allowing CQ
to remain in its active site and exert its potency. When resistant parasites are treated with 3-ICQ
and VP, there was no improvement of activity, suggesting that 3-ICQ may not be a substrate for
mutant PfCRT, as its activity is independent of the presence of VP. A surprising and significant
finding of these results is that combination treatment of CQ and 3-ICQ was more potent than either
drug alone against CQR parasites. This chemosensitization effect suggests that 3-ICQ may act like
With these promising results in hand, we wanted to explore the functionality of the 3-position
further. In this chapter, we describe the synthesis of CQ derivative, their antiplasmodial activity
and their structure-activity relationships. We will also describe pKa studies carried out to determine
105
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
Previous work in our lab demonstrated the chemosensitization effect that 3-ICQ had on CQ.4 It
was evident that 3-ICQ sensitizes CQR parasites to CQ, suggesting it may not be a substrate for
mutant PfCRT. The possibility of synthesizing drugs for combination therapy was attractive as
that is the basis of ACT. We wanted to further explore modifications at the 3rd position of the
We began with the functionalization of the 3rd position of the quinoline, with the installation of an
amine functional group (discussed in chapter 2), Scheme 3.2. With the amine installed, we were
then able to carry out the final step of the synthesis. We explored three classes of derivatives:
amides, sulfonamides and Schiff-bases. The formation of amides was straightforward as benzoyl
chlorides are widely available and relatively inexpensive, Figure 3.2. Amides add rigidity to the
molecule as well as the introduction of an H-bond acceptor. The resulting derivatives were then
While designing the new CQ-derivatives, we used Lipinski's rule of 5 as a guide to possibly make
the compounds more drug-like for oral administration.6,7 The rules state that a drug should: i) have
a molar mass of less than 500 g/mol, ii) have a partition coefficient of less than 5, iii) have no more
than 5 H-bond donors and iv) have no more than 10 H-bond acceptors. As a starting point, we
began with the synthesis of benzamides and used the Craig Plot 8,9 as a guide to determine which
106
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
+s - p +s +p
-s -p -s +p
Figure 3.1. Craig Plot groups substituents with similar electronic and hydrophobic effects.
Highlighted are synthesized compounds containing substituents at various locations on the phenyl
ring.
The Craig Plot groups various substituents based on their electronic and hydrophobic properties
using benzoic acid as the reference point, the center of the graph. This helps identify simple
experimented with substitution patterns on the benzoyl ring to get a complete SAR profile.
107
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
Our collaborators, Dr. Fadi Baakdah, supervised by Dr. Elias Georges at the McGill University
Institute of Parasitology, carried out in vitro antiplasmodial assays to identify which substituents
showed the greatest potency, the results are displayed in Table 3.1.
Et2N
NH
H
N R
O
Cl N
H CN NO2 Cl F
Figure 3.2. Synthesized compounds. Colours: grey: the reference compound, blue: electron-
108
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
Table 3.1. Antiplasmodial activities of CQ derivatives in sensitive and resistant parasitic strains
The vast majority of the synthesized drug candidates show some antiplasmodial activity; however,
most are less potent than CQ in CQS 3D7 strain and CQR Dd2 strain. The benzamido-analog, 3.3,
was 2.5 - fold less potent than CQ in CQS 3D7 strains, and was 7 - fold less potent than CQ in
CQR Dd2 strains. We believe this decrease in activity is due to decreased affinity to heme, similar
to 3-ICQ, which is correlated to antiplasmodial activity.5 For the Dd2 strain, the mutations might
109
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
cause reduced accumulation of the benzamidoCQ in the DV, it was reported that accumulation is
Et2N R Et2N
NH NH H
H
N N
O
O
Cl N Cl N
1 Et2N
2
R
Et2N O O
NH R HN R
N N
H H
Cl N 3 Cl N 4
Halogen substituents such as fluoride and chloride had varying activity depending on their position
on the benzoyl ring. We discovered that the para-position gave the best activity for both halogens,
3.8 and 3.11. They were more potent than the benzamidoCQ compound; 7.2 and 5.8 - fold better
in CQS strain than benzamidoCQ for chloro- and fluoro-respectively. The activity at the meta-
position, 3.7 and 3.10, was in between that of the ortho- and para- positions. The ortho-position
gave the worst activity for both substituents, 3.6 and 3.9, with the chloro- being more potent in
CQS strain. They were more potent than benzamidoCQ in CQR strains but not as effective on CQS
Electron-withdrawing, hydrophobic nitro-substituent, 3.5, was 5.5 - fold less potent than the
reference compound in CQS strain and was 1.4 – fold more potent than the reference compound
110
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
in CQR strain. The decrease in activity in CQS strain is likely due to an increase in the size of the
moiety. Furthermore, the rigidity of this substituent might play a role in its reduced activity.
The addition of an electron-withdrawing, hydrophilic cyano group, 3.4, was 7.7 - fold weaker in
CQS strain and 1.5 - fold better in CQR than the benzamidoCQ. The decrease in activity in 3D7
strains might be due to the addition of an H-bond acceptor. The lack of flexibility at the 4th position
might reduce the compound's ability to enter the target / active site. In Dd2 strains, the activity is
essentially the same; thus, the reduced activity compared to CQ might be due to decreased
accumulation.
The electron-donating, hydrophilic amino group, 3.12, was 5 - fold more potent than
benzamidoCQ in CQS but 1.3 – fold weaker in the CQR parasite. In contrast, the methoxy, 3.13,
was 2 - fold weaker in CQS and 1.2 – fold stronger in the CQR parasite than the benzamidoCQ.
The addition of an H-bond donor NH2 group significantly improves the antiplasmodial activity in
CQS strains. This increase in activity might be due to a slight increase in accumulation as a small
percentage of the amine will be protonated at lysosomal pH. Furthermore, the ability to form strong
hydrogen bonds might further stabilize the compound.11 In contrast, the methoxy derivative's
reduced potency might result from reduced H-bonding ability and increased size of the methoxy
in CQS strains. In resistant strains, however, the H-bonding ability of the amino group is a
disadvantage, this is a trend in all our compounds. Therefore, we observe that the methoxy group,
which does not H-bond as strongly as the amine, in CQR strains, is 1.6 - fold more potent than the
amine counterpart.
111
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
3.15, have similar activity in CQS and CQR strains. They were 5 - fold less potent than
benzamidoCQ, 3.3, in CQS parasites but ~2.3 - fold more potent in resistant parasites. However,
the methylthio-substituent, 3.14, was 1.6 – fold less potent than benzamidoCQ in CQS strains but
was 5.8 - fold more potent in CQR strains. Although these substituents belong within the same
quadrant, it is important to note that there is a difference in electronegativity and steric bulk. The
methylthio-substituent is 3.4 - fold more potent than the methyl and dimethylamino-substituent in
CQS strains. The methylthio-derivative is also 3 - fold more potent than these derivatives in CQR
strains. This might be due to the favorable electronics and size of the methylthio over the simple
F
Et2N O O
Et2N F Et2N
NH NH NH
H
N N N
H H
O F
Cl N 1 Cl N 2 Cl N 3
F
O F Et2N Et2N O
Et2N NH
NH H NH
N N N F
H H
O
Cl N 4 Cl N Cl N 6
5
When looking at the possible configurations to explain differences in activity, one can see how
electronic effects and steric hindrance may impact activity (see Figure 3.3 to Figure 3.5). The
ortho-position which showed the worst antiplasmodial activity exhibits lone pair / lone pair
repulsion with the carbonyl oxygen. These forces may be unfavourable for activity. However,
112
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
these conformers also possibly exhibit intramolecular H-bonding, which could constrain the steric
effects. However, the repulsion might be stronger than H-bonding stabilization. The meta-position
contains conformers that possibly exhibit intramolecular H-bonding. This reduced repulsion might
contribute to an increase of antiplasmodial activity in both CQS and CQR strains. The para-fluoro-
substituent exhibits the least steric effects with the possible H-bond of the carbonyl and secondary
amine, further stabilizing the compound and contributing to increased activity in CQS strains.
However, it was clear that these factors do not enhance activity in CQR strains. As mentioned
Cl
Et2N O O
Et2N Cl Et2N
NH NH NH
H
N N N
H H
O Cl
Cl N 1 Cl N 2 Cl N 3
Cl
O Cl Et2N Et2N O
Et2N NH
NH H NH
N N N Cl
H H
O
Cl N 4 Cl N Cl N 6
5
In contrast, the chloro-substituent shows better antiplasmodial activity across the board in CQS
strains. A likely explanation is the reduced electronegativity of the chloride and longer C - X bond,
which minimizes the " - delocalization. Furthermore, the chloride is unable to H – bond, explaining
To summarize, in 3D7 strains, the best substituents are chloro, 3.8, fluoro, 3.11, methylthio, 3.14,
and amino 3.12. These are relatively compact or can form strong hydrogen bonds. In the case of
113
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
Dd2 strains, H-bonding substituents are disfavoured. The best substituents are chloro, 3.8, and
methylthio-substituent 3.14. These are relatively small or flexible and do not form H-bonds. The
optimal substituent positioning is the para-position. This position is void of possible steric effects,
To determine whether the synthesized derivatives were substrates for PfCRT, in vitro assays were
carried out in CQS and CQR strains in the presence of VP. VP is believed to act as a
chemosensitizer that works on resistant strains by inhibiting mutant PfCRT's ability to transport
drugs out of the DV. Assays of CQ with VP in Dd2 show a significant increase in activity as
mutant PfCRT is inhibited. When 3-ICQ is assayed with VP in 3D7 and Dd2, no significant
increase in activity is observed. This suggests that 3-ICQ is not a possible substrate for wild-type
or mutant PfCRT.4 Similarly, VP does not modulate 3.16 activity in Dd2 strains. Our next objective
when used in combination with CQ. Interestingly, VP increases the IC50 value of wild-type PfCRT,
Table 3.2. There is currently no evidence of VP interacting with wild-type PfCRT; furthermore,
there are only a few reports on the speculated normal function(s) of wild-type PfCRT.
114
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
3.3.3 Isomerization
The synthesized derivatives can adapt two main isomeric forms: the cis or trans-isomers, Figure
3.6. These isomers can be easily distinguished in an NMR spectrum as major or minor contributors.
However, we found that all of the compounds were isolated as one isomer at RT, according to the
1
H NMR. As the rotational barrier of rotamers is high, it was assumed that they were isolated as
one rotamer. The expected major isomer is the trans-amide because it exhibits the least steric
effects. This is confirmed as the NOESY NMR does not show evidence of there being the E isomer.
We do not observe the expected 2 - H / phenyl interaction associated with the E isomer.
Et2N R Et2N
NH NH
H H
N N O
O
Cl N Cl N
trans-amide cis-amide
R
115
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
O
d
N c
H
b
c
d F
a
a 3
b
O
h
N g
H
e f
e,h F
f 2
g
O
i
N j
H
F j
i 1
8.25 8.20 8.15 8.10 8.05 8.00 7.95 7.90 7.85 7.80 7.75 7.70 7.65 7.60 7.55 7.50 7.45 7.40 7.35 7.30 7.25 7.20 7.15
f1 (ppm)
For the fluoride-containing derivatives, 1H NMR analysis was carried out with H-F coupled mode.
Evident in the spectra, Figure 3.7, is the extend of fluoride coupling on the benzoyl ring. We
observe up to 4J coupling in all derivatives, as evidenced by the splitting patterns. The coupling
constant for proton i, a triplet is 6.5 Hz, while the coupling constant for proton j, a triplet is 8.4
Hz. Similarly, it is evident the extent of proton-fluorine coupling in proton b, which is a triplet of
double of doublets (tdd) where J = 1.9, 5.3 and 7.2 Hz and proton d, which is a triplet of doublets
(td) where J = 5.6 and 8 Hz. The multiplicity of proton a, is a double of double of doublets (ddd)
116
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
Et2N Cl Et2N Cl
NH NH
H
N N
S
O2
Cl N Cl N
3.17 3.18
Once we discovered that the 4-chlorobenzamidoCQ, 3.8, was the most potent compound, we
synthesized sulfonyl, 3.17, and Schiff-base, 3.18, equivalents to determine the effect of the
carbonyl group on activity, as shown in Figure 3.8. We found that the addition of the sulfone has
a negative impact on the activity of the compound. This might be due to the increased mass by 36
g/mol, which breaks the Lipinski rule that states a drug should have a molar mass less than 500
g/mol and the introduction of an additional H-bond acceptor, Figure 3.9. This further reinforces
the notion that H-bonding groups are disfavoured in resistant strains. Furthermore, the sulfone
group might sterically hinder localization in the active site. The Schiff-base was 15 - fold less
potent in the 3D7 strain and 1.3 - fold less potent in the Dd2 strain than the 4-chlorobenzamide.
The decrease in activity in the sensitive strain might indicate the need for some electron density in
the active site. Therefore, the electronegativity of the carbonyl is beneficial to activity. In Dd2, the
activity is similar, further reinforcing that H-bonding is not beneficial in mutant parasites.
117
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
Et2N Et2N R
NH NH
H O H
N N
S R S
O O O
Cl N Cl N
1 2
R
Et2N O O Et2N O
NH S NH S O
N R N
H H
Cl N Cl N
3 4
Due to the size of these compounds and the positioning of the modifications, b-hematin inhibition
assays (BHIA) were not carried out. It is known that modification on the quinoline ring reduces
the affinity of heme - CQ complex formation. These compounds are not expected to be able to
prevent b-hematin formation to any significant extent. For this reason, BHIA was not performed.
Furthermore, although the current widely accepted mechanism of action for 4-aminoquinolines is
b-hematin inhibition, it may not be the only way these compounds exert their activity.12–16
Although these derivatives may not be as potent as CQ, their activity may be linked to a different
mechanism. For this reason, the next step of this project would be to carry out assays in which
parasites are given both CQ and our derivatives to determine if they behave as 3-ICQ in a
synergistic manner.
118
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
CQ is a weak base that can accumulate within its target site via the protonation of its tertiary alkyl
nitrogen and quinoline nitrogen.17 At pH 7.4, CQ is 17% mono-protonated and still lipid-soluble
and can cross cell membranes. At lysosomal pH of 5.2, it is entirely diprotonated and no longer
membrane permeable.15 Thus, the pKas of the molecule plays a crucial role in its localization,
Table 3.3.
The presence of the electron-withdrawing halogens at the 3-position has a significant effect on the
quinolinium pKa. 3-halo CQs are 2 - 3 log units lower than CQ; therefore, they are not expected
the overall lipophilicity of the molecule, which may promote membrane permeability; however,
due to the decreased pKa, it is not expected to become acid trapped within the DV as seen with
CQ. This lack of accumulation in the target site may contribute to the observed decreased
antiplasmodial activity. Furthermore, acid trapping is not believed to be the only method used for
119
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
substituent does not lower the quinolinium pKa as much as the halogens. This log unit difference
in pKa means a greater portion of 3-NH2CQ will be protonated at physiological and lysosomal pH.
Interestingly, although the pKa is higher for 3-NH2-CQ, the IC50 values are similar to 3-ICQ in
CQS and CQR strains; this might suggest a similar mode of action for 3-substituted CQ's.
3.4 Conclusion
CQ has not been extensively modified at the 3-position in prior studies. A possible reason for this
is the reduced antiplasmodial activity resulting from the lowered pKa of the quinolinium nitrogen
parasite to CQ when 3-substituted CQs derivatives are used in combination with CQ. We were
able to further derivatize 3-substituted CQs via the installation of an amine. A library of derivatives
was successfully synthesized represented across in the Craig Plot. Derivatives contain varying
electronic and hydrophobic properties, which allow us to compare the effectiveness of substituents
and the significance of regioselectivity. We determined that the hydrophilic and electron-
withdrawing chloro-substituent was the most effective; furthermore, the para-position gives the
best activity. Although the derivatives were active towards the parasite, they were on average 8 –
fold less effective than CQ in sensitive strains and 6 – fold less effective than CQ in resistant
strains. A sulfone and Schiff-base derivative were also synthesized; however, they were less potent
than 4-chlorobenzamidCQ.
120
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
3.5 Experimental
Glassware was taken directly from the oven (120oC) and let cool in a desiccator before use. All
reactions were carried out under N2 pressure. Anhydrous DCM was obtained from MBraun MB-
SPS solvent purification system into an oven-dried flask on the day of use. All solvents and
reagents were obtained commercially and used without further purification unless noted.
Orbitrap, with results reported as mass/charge ratios (m/z). Thin-layer chromatography plates were
visualized using ultra-violet light, 254nm. Flash column chromatography was carried out manually
using 230 - 400 mesh silica gel (Silicycle) using reagent grade solvents or using Biotage Isolera
One system with Biotage ZIP 30 g columns. Cole Parmer Microcomputer pH-Vision Model
05669-20 pH Meter used with Sigma-Aldrich micro pH combination electrode, glass body. The
1 13
H and C NMR spectra were recorded at ambient temperature on Bruker AVIIIHD 400 MHz
(1H 400 MHz, 13C 100 MHz) and Bruker AVIIIHD 500 MHz (1H 500 MHz, 13C 125 MHz) using
tetramethylsilane as the internal standard. Chemical shifts are reported relative to the residual
deuterated solvent peaks. Chemical shifts are expressed in parts per million (ppm = δ) values and
coupling constants (J) in Hertz (Hz). The terms m, s, d, t and q represent multiplicities of 1H NMR
resonances: multiplet, singlet, doublet, triplet and quartet, respectively. For previously unknown
121
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
complete the assignment of 1H and 13C signals. 1H NMR signals are described by chemical shift δ
13
(multiplicity, J (Hz), integration). C NMR signals are described by chemical shift δ and are
cuvette, 1,990 µL of deionized H2O was added, and into this cuvette was added 10 µL of the 0.01
the cuvette of 2,000 µL. The pH of the solution was measured on a calibrated pH meter. The pH
should be acidic. Base (NaOH) will be added to the cuvette to increase the pH. Solutions of 10mM
and 1mM NaOH and HCl were made. 5-20 µL of NaOH was added to increase the pH by 0.2 units.
If the pH increases were too large, HCl was added to lower the pH. After each addition of NaOH,
the solution was well mixed, the pH probe was fully submerged into the cuvette, and the pH was
measured. The pH, incremental volume increase and absorbance at the lmax were recorded. NaOH
was added until a pH > 11/12 was reached or until the Absorbance versus pH graph plateaus. The
The measured absorbance was then adjusted for dilution at pHx with the following formula:
The pH and corrected absorbances were plotted on a non-linear curve fit graph on Origin Pro or
GraphPad Prism 7. The equation used to fit absorbance values to produce a pKa value is as follows:
1 1
7 = <1 − @ 7'( + < @7 !
1 + 10!"$!%& 1 + 10!"$!%& '(
122
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
Where ACQ is the absorbance of neutral CQ, ACQ+ is the absorbance of protonated CQ. Where
Parasite proliferation assays using CQ or CQ-derivatives were tested on CQS 3D7 and CQR Dd2
Plasmodium falciparum lab strains. Briefly, 0.5% ring-stage parasites were incubated in a 0.2%
hematocrit in 96 black well clear bottom plates in triplicates (Corning Inc.; ref: 3603) for 72hrs in
an incubator maintaining 37°C and atmospheric conditions of 3% O2, 5% CO2 and 92% N2. Later,
the plates were frozen at -80°C and thawed at RT. Then they were treated with a lysis-developing
solution (Tris; 20 mM pH 7.5, 5 mM EDTA; 0.008% w/v saponin, 0.08% v/v Triton X-100, 0.2
μL /ml of 10,000x SYBR® Green I Nucleic Acid gel stain dye) and kept away from light for 1
hour. Fluorescence was measured using Synergy H4 plate reader with Ex 485 nm / Em 535 nm.
The resulting data were analyzed using GraphPad Prism version 8.4.0 (671) to obtain the 50%
To a stirring solution of 3-NH2CQ (1 equiv.) in anhydrous DCM at 0oC is added benzoyl chloride
(1.5 equiv.) under nitrogen followed by NEt3 (3 equiv.). The solution is stirred and allowed to
warm to RT. The reaction progress is monitored by TLC. When complete, the reaction is poured
into saturated NaHCO3 solution and extracted with DCM (3 X 20 mL) and washed with brine. The
combined organic extracts are then dried over anhydrous MgSO4, and the solvent is removed under
reduced pressure. The crude product is purified by prep-TLC with silica gel, solvent system: 90%
123
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
EtOAc, 5% MeOH, 5% NEt3 or with C18 prep-TLC in 50% H2O, 50% acetonitrile, 1%
Et2N
NH
H
N
O
Cl N
N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)benzamide (3.3)
Yield: 53.3 mg, 85%. 1H NMR (800 MHz, Methanol-d4) δ 8.56 (d, J = 9.2 Hz, 2H), 8.07 (dt, J =
7.1, 1.3 Hz, 2H), 7.94 (d, J = 2.1 Hz, 1H), 7.76 (dd, J = 9.1, 2.1 Hz, 1H), 7.69 (td, J = 7.3, 1.3 Hz,
1H), 7.61 (t, J = 7.8 Hz, 2H), 4.57 (dt, J = 12.4, 7.0 Hz, 1H), 3.10 (tq, J = 12.5, 5.8, 5.1 Hz, 4H),
3.07 – 3.02 (m, 1H), 3.00 – 2.94 (m, 1H), 1.82 (dq, J = 12.1, 7.6, 6.7 Hz, 1H), 1.70 – 1.62 (m, 3H),
1.43 (d, J = 6.5 Hz, 3H), 1.20 (q, J = 6.9 Hz, 6H). 13C NMR (201 MHz, Methanol-d4) δ 169.09,
153.47, 151.04, 145.62, 139.46, 138.61, 132.71, 132.69, 128.77, 127.55, 125.29, 119.60, 117.39,
111.52, 51.57, 51.02, 33.27, 20.79, 20.20, 7.70, 7.64. HRMS (ESI) calcd. for C25H31ClN4O
Et2N CN
NH
H
N
O
Cl N
N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)-4-cyanobenzamide (3.4)
Yield: 45.6 mg, 66%, mixture of isomers. 1H NMR (500 MHz, Chloroform-d) δ 8.85 (s, 1H), 8.20
(d, J = 7.9 Hz, 2H), 7.99 (dd, J = 3.8, 2.1 Hz, 1H), 7.88 (dd, J = 9.2, 7.5 Hz, 2H), 7.80 (d, J = 8.0
Hz, 2H), 7.42 (ddd, J = 8.5, 6.1, 2.2 Hz, 1H), 4.32 (s, 1H), 3.72 (s, 1H), 2.52 (q, J = 7.1 Hz, 4H),
124
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
2.39-2.35 (m, 2H), 1.60 – 1.40 (m, 4H), 1.24 (d, J = 9.0 Hz, 3H), 0.99 (t, J = 7.2 Hz, 6H). 13C
NMR (126 MHz, Chloroform-d) δ 164.96, 150.39, 150.26, 148.53, 148.15, 145.96, 145.17,
143.09, 137.64, 135.05, 134.73, 132.72, 130.94, 129.70, 129.08, 129.05, 128.57, 127.59, 126.51,
126.47, 124.07, 123.76, 121.50, 121.26, 119.82, 119.01, 118.07, 115.85, 53.62, 53.51, 52.72,
52.60, 46.82, 46.73, 36.71, 23.78, 23.40, 22.40, 22.26, 21.69, 11.19, 10.57. HRMS (ESI) calcd.
Et2N NO2
NH
H
N
O
Cl N
N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)-4-nitrobenzamide (3.5)
Yield: 109.5 mg, 76%. 1H NMR (500 MHz, Methanol-d4) δ 8.60 (s, 1H), 8.58 (d, J = 9.3 Hz, 1H),
8.44 (d, J = 8.5 Hz, 2H), 8.29 (d, J = 8.7 Hz, 2H), 7.95 (d, J = 2.1 Hz, 1H), 7.76 (dd, J = 9.2, 1.9
Hz, 1H), 4.65 – 4.53 (m, 1H), 3.14 (q, J = 7.3 Hz, 4H), 3.10 – 3.03 (m, 2H), 1.90 – 1.81 (m, 1H),
1.77 – 1.66 (m, 3H), 1.43 (d, J = 6.5 Hz, 3H), 1.23 (t, J = 7.3 Hz, 6H). 13C NMR (126 MHz,
Methanol-d4) δ 167.24, 153.71, 150.34, 145.10, 139.63, 138.53, 138.33, 129.02, 127.57, 125.62,
123.60, 119.42, 117.04, 111.31, 51.94, 51.09, 46.94, 33.30, 20.81, 20.25, 7.62. HRMS (ESI) calcd.
125
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
Et2N Cl
NH
H
N
O
Cl N
2-chloro-N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)benzamide (3.6)
Yield: 42 mg, 59%. 1H NMR (500 MHz, Methanol-d4) δ 8.60 (s, 1H), 8.55 (d, J = 9.2 Hz, 1H),
7.95 (s, 1H), 7.79 – 7.70 (m, 2H), 7.63 – 7.48 (m, 3H), 4.73 (q, J = 6.7, 5.9 Hz, 1H), 3.14 (q, J =
7.3 Hz, 4H), 3.10 – 3.02 (m, 2H), 1.93 – 1.84 (m, 1H), 1.77 (dq, J = 12.1, 7.6, 6.0 Hz, 3H), 1.45
(d, J = 6.4 Hz, 3H), 1.22 (q, J = 7.0 Hz, 6H). 13C NMR (126 MHz, Methanol-d4) δ 170.67, 154.26,
147.24, 140.60, 136.11, 133.34, 132.14, 131.59, 130.39, 128.90, 128.66, 126.75, 121.75, 119.26,
52.65, 52.58, 34.84, 22.16, 21.95, 9.08, 8.99. HRMS (ESI) calcd. for C25H31Cl2N4O [M+H]:
Cl
Et2N
NH
H
N
O
Cl N
3-chloro-N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)benzamide (3.7)
Yield: 43.3 mg, 61%. 1H NMR (500 MHz, Chloroform-d) δ 8.87 (s, 1H), 8.10 (s, 1H), 8.01 (d, J
= 8.7 Hz, 1H), 7.91 (d, J = 9.0 Hz, 1H), 7.56 (d, J = 8.1 Hz, 1H), 7.47 (t, J = 7.8 Hz, 1H), 7.42 (d,
J = 9.6 Hz, 1H), 4.31 (d, J = 11.2 Hz, 1H), 3.73 (d, J = 13.1 Hz, 2H), 2.57 (t, J = 7.5 Hz, 5H), 2.42
(d, J = 6.7 Hz, 2H), 1.52 (dt, J = 10.6, 6.8 Hz, 1H), 1.26 (d, J = 7.3 Hz, 7H), 1.02 (t, J = 7.2 Hz,
13
7H). C NMR (126 MHz, Chloroform-d) δ 165.40, 150.70, 148.64, 145.89, 135.46, 135.18,
134.86, 132.37, 130.24, 129.15, 128.37, 126.37, 126.02, 124.30, 121.36, 119.41, 53.58, 52.48,
126
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
46.72, 36.69, 22.84, 22.52, 10.09. HRMS (ESI) calcd. for C25H31Cl2N4O [M+H]: 473.1869, found:
473.1874.
Et2N Cl
NH
H
N
O
Cl N
4-chloro-N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)benzamide (3.8)
Yield: 36.4 mg, 51%. 1H NMR (400 MHz, Chloroform-d) δ 9.35 (s, 1H), 8.83 (s, 1H), 8.11 (d, J
= 8.2 Hz, 2H), 8.04 – 7.97 (m, 1H), 7.90 (d, J = 9.0 Hz, 1H), 7.48 (dd, J = 8.4, 2.1 Hz, 2H), 7.41
(dd, J = 9.0, 2.0 Hz, 1H), 4.35 (s, 1H), 3.76 (s, 1H), 2.63 (q, J = 7.2 Hz, 4H), 2.47 (t, J = 7.4 Hz,
2H), 1.62 (d, J = 9.3 Hz, 1H), 1.58 – 1.44 (m, 3H), 1.27 (d, J = 6.4 Hz, 3H), 1.05 (t, J = 7.2 Hz,
13
6H). C NMR (126 MHz, Chloroform-d) δ 165.91, 151.05, 148.71, 146.63, 138.64, 134.81,
131.99, 129.66, 129.09, 129.04, 126.22, 124.53, 121.23, 119.38, 53.49, 52.30, 46.58, 36.58, 22.61,
22.30, 9.54. HRMS (ESI) calcd. for C25H31Cl2N4O [M+H]: 473.1869, found: 473.1872.
Et2N
NH
H
N
O F
Cl N
N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)-2-fluorobenzamide (3.9)
Yield: 45.1 mg, 66%. 1H NMR (500 MHz, Methanol-d4) δ 8.58 (d, J = 9.2 Hz, 1H), 8.56 (s, 1H),
7.99 – 7.91 (m, 2H), 7.73 (dd, J = 9.2, 1.7 Hz, 1H), 7.68 (tdd, J = 7.4, 5.1, 1.8 Hz, 1H), 7.41 (t, J
= 7.6 Hz, 1H), 7.36 (dd, J = 11.3, 8.4 Hz, 1H), 4.71 (h, J = 6.9, 6.0 Hz, 1H), 3.13 (q, J = 7.3 Hz,
4H), 3.09 – 3.00 (m, 2H), 1.87 (dt, J = 13.5, 8.2 Hz, 1H), 1.73 (dq, J = 12.4, 7.8, 6.1 Hz, 3H), 1.43
127
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
(d, J = 6.5 Hz, 3H), 1.22 (t, J = 7.3 Hz, 6H). 13C NMR (126 MHz, Methanol-d4) δ 167.59, 154.51,
147.18, 140.74, 140.26, 135.60, 135.53, 132.13, 128.89, 126.77, 126.23, 126.20, 122.98, 122.88,
19
121.20, 52.71, 52.52, 48.34, 34.71, 22.19, 21.73, 9.02. F NMR (377 MHz, Methanol-d4) δ -
115.14. HRMS (ESI) calcd. for C25H3oClFN4O [M+H]: 457.2165, found: 457.2162.
F
Et2N
NH
H
N
O
Cl N
N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)-3-fluorobenzamide (3.10)
Yield: 43.7 mg, 64%. 1H NMR (500 MHz, Chloroform-d) δ 8.89 (s, 1H), 8.75 (s, 1H), 8.00 (d, J
= 2.2 Hz, 1H), 7.88 (d, J = 9.0 Hz, 1H), 7.80 (dd, J = 28.6, 8.5 Hz, 2H), 7.50 (td, J = 8.0, 5.6 Hz,
1H), 7.42 (dd, J = 9.0, 2.1 Hz, 1H), 7.29 (dd, J = 8.0, 2.3 Hz, 1H), 4.28 (d, J = 9.5 Hz, 1H), 3.72
(s, 1H), 2.50 (q, J = 7.1 Hz, 4H), 2.40 – 2.32 (m, 2H), 1.59 – 1.42 (m, 4H), 1.22 (d, J = 6.4 Hz,
13
3H), 0.97 (t, J = 7.2 Hz, 6H). C NMR (126 MHz, Chloroform-d) δ 165.24, 164.04, 162.07,
150.30, 148.37, 145.43, 136.10, 136.04, 134.86, 130.72, 130.66, 129.14, 126.50, 123.87, 123.15,
121.43, 119.51, 119.46, 119.34, 115.25, 115.06, 53.55, 52.62, 46.77, 36.69, 23.51, 22.33, 10.82.
19
F NMR (377 MHz, Chloroform-d) δ -111.57. HRMS (ESI) calcd. for C25H31ClFN4O [M+H]:
128
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
Et2N F
NH
H
N
O
Cl N
N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)-4-fluorobenzamide (3.11)
Yield: 41 mg, 60%. 1H NMR (500 MHz, Chloroform-d) δ 8.86 (s, 1H), 8.74 (s, 1H), 8.07 (t, J =
6.7 Hz, 2H), 7.99 (d, J = 2.1 Hz, 1H), 7.88 (d, J = 9.0 Hz, 1H), 7.41 (dd, J = 9.0, 2.2 Hz, 1H), 7.18
(t, J = 8.4 Hz, 2H), 4.28 (d, J = 9.4 Hz, 1H), 3.71 (s, 1H), 2.49 (q, J = 7.2 Hz, 4H), 2.40 – 2.30 (m,
1H), 1.59 – 1.37 (m, 2H), 1.21 (d, J = 6.4 Hz, 3H), 0.97 (t, J = 7.2 Hz, 6H). 13C NMR (126 MHz,
Chloroform-d) δ 166.20, 165.35, 164.18, 150.25, 148.20, 145.37, 134.64, 130.09, 130.02, 129.78,
129.76, 128.95, 126.28, 123.79, 121.27, 119.52, 115.98, 115.81, 53.40, 52.45, 46.61, 36.50, 23.27,
19
22.19, 10.57. F NMR (377 MHz, Chloroform-d) δ -107.08. HRMS (ESI) calcd. for
Et2N NH2
NH
H
N
O
Cl N
4-amino-N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)benzamide (3.12)
Yield: 53.8 mg, 41%. 1H NMR (400 MHz, Methanol-d4) δ 8.31 (s, 1H), 8.26 (d, J = 9.1 Hz, 1H),
7.84 (d, J = 2.2 Hz, 1H), 7.83 – 7.78 (m, 2H), 7.48 (dd, J = 9.1, 2.2 Hz, 1H), 6.76 – 6.70 (m, 2H),
2.48 (q, J = 7.1 Hz, 4H), 2.39 (d, J = 8.0 Hz, 2H), 1.64 (d, J = 7.2 Hz, 1H), 1.52 – 1.38 (m, 3H),
1.26 (d, J = 6.4 Hz, 3H), 0.96 (t, J = 7.2 Hz, 6H). 13C NMR (126 MHz, Methanol-d4) δ 169.78,
154.45, 154.12, 148.95, 148.71, 136.26, 130.65, 127.88, 126.72, 125.18, 121.94, 121.23, 115.33,
129
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
114.70, 53.36, 52.16, 47.64, 36.67, 23.79, 21.90, 10.92. HRMS (ESI) calcd. for C25H33ClN5
Et2N O
NH
H
N
O
Cl N
N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)-4-methoxybenzamide
(3.13)
Yield: 52.2 mg, 75%. 1H NMR (500 MHz, Chloroform-d) δ 8.92 (s, 1H), 8.34 (s, 1H), 7.99 (d, J
= 2.2 Hz, 1H), 7.97 (d, J = 8.6 Hz, 2H), 7.87 (d, J = 9.0 Hz, 1H), 7.40 (dd, J = 9.0, 2.2 Hz, 1H),
6.99 (d, J = 8.8 Hz, 2H), 4.25 (d, J = 9.7 Hz, 1H), 3.88 (s, 3H), 3.73-3.67 (m, 1H), 2.46 (q, J = 7.1
Hz, 4H), 2.33 (t, J = 6.7 Hz, 2H), 1.62 – 1.40 (m, 4H), 1.18 (d, J = 6.3 Hz, 3H), 0.95 (t, J = 7.2
Hz, 6H). 13C NMR (126 MHz, Chloroform-d) δ 165.98, 162.96, 150.28, 148.13, 145.14, 134.61,
129.52, 129.05, 126.40, 125.98, 123.80, 121.55, 120.07, 114.20, 55.64, 53.55, 52.73, 46.81, 36.71,
23.80, 22.25, 11.22. HRMS (ESI) calcd. for C26H34ClN4O2 [M+H]: 469.2365, found: 469.2369.
Et2N SMe
NH
H
N
O
Cl N
N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)-4(methylthio)benzamide
(3.14)
Yield: 47 mg, 61%. 1H NMR (500 MHz, Chloroform-d) δ 8.80 (s, 1H), 8.17 (d, J = 8.0 Hz, 2H),
8.01 (d, J = 2.2 Hz, 1H), 7.95 (d, J = 9.0 Hz, 1H), 7.41 (dd, J = 9.0, 2.2 Hz, 1H), 7.34 (d, J = 8.6
Hz, 2H), 4.41 (d, J = 10.2 Hz, 1H), 3.80 (s, 1H), 2.77 – 2.67 (m, 4H), 2.58-2.45 (m, 5H), 1.74-1.65
130
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
(m, 1H), 1.57 – 1.44 (m, 3H), 1.33 (d, J = 6.4 Hz, 3H), 1.11 (t, J = 7.3 Hz, 6H). 13C NMR (126
MHz, Chloroform-d) δ 166.45, 151.45, 148.80, 144.30, 137.55, 134.50, 129.25, 128.81, 128.73,
125.80, 125.29, 124.96, 121.01, 119.38, 53.31, 51.95, 46.32, 36.39, 22.70, 21.15, 14.95, 8.37.
Et2N NMe2
NH
H
N
O
Cl N
N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)-4-
(dimethylamino)benzamide (3.15)
Yield: 22.3 mg, 31%. 1H NMR (800 MHz, Dimethyl sulfoxide-d6) δ 9.71 (s, 1H), 8.40 (d, J = 9.1
Hz, 1H), 8.29 (s, 1H), 7.91 (d, J = 8.9 Hz, 2H), 7.84 (d, J = 2.3 Hz, 1H), 7.50 (dd, J = 8.9, 2.3 Hz,
1H), 6.78 – 6.74 (m, 2H), 5.97 (d, J = 9.1 Hz, 1H), 3.98 (dq, J = 8.8, 6.4 Hz, 1H), 3.00 (s, 9H),
2.38 (s, 4H), 2.28 (s, 2H), 1.60 – 1.54 (m, 1H), 1.36 (ddt, J = 12.6, 9.8, 6.1 Hz, 1H), 1.29 (t, J =
7.9 Hz, 2H), 1.12 (d, J = 6.4 Hz, 3H), 0.85 (t, J = 7.1 Hz, 6H). 13C NMR (201 MHz, Dimethyl
sulfoxide-d6) δ 165.98, 154.20, 152.42, 147.67, 146.07, 133.05, 129.05, 127.50, 124.85, 124.61,
120.32, 119.96, 114.71, 110.84, 51.99, 50.41, 46.06, 39.99, 34.97, 22.61, 21.51, 7.16. HRMS
131
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
Et2N
NH
H
N
O
Cl N
N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-yl)-4-methylbenzamide
(3.16)
Yield: 38.1 mg, 59%. 1H NMR (500 MHz, Chloroform-d) δ 8.95 (s, 1H), 8.34 (s, 1H), 8.01 (d, J
= 2.2 Hz, 1H), 7.89 (dd, J = 11.0, 8.2 Hz, 3H), 7.41 (dd, J = 8.9, 2.2 Hz, 1H), 7.31 (d, J = 7.7 Hz,
2H), 4.25 (d, J = 9.8 Hz, 1H), 3.75 – 3.64 (m, 1H), 2.51 – 2.40 (m, 7H), 2.33 (t, J = 6.6 Hz, 2H),
13
1.61 – 1.41 (m, 4H), 1.19 (d, J = 6.4 Hz, 3H), 0.96 (t, J = 7.2 Hz, 6H). C NMR (126 MHz,
Chloroform-d) δ 166.42, 150.35, 148.23, 145.26, 143.02, 134.66, 130.93, 129.67, 129.09, 127.66,
126.40, 123.88, 121.54, 119.95, 53.53, 52.68, 46.80, 36.67, 23.57, 22.30, 21.69, 10.99. HRMS
solution is stirred and allowed to warm to RT. The reaction progress is monitored by TLC. After
24 hours the solvent is removed under reduced pressure. The residue is then diluted with DCM,
and the solution is poured into a flask containing saturated NaHCO3 solution and is extracted with
DCM (3 X 20 mL) and washed with brine. The combined organic extracts are then dried over
anhydrous MgSO4 and the solvent removed under reduced pressure. The crude product is purified
by prep-TLC with silica gel, solvent system: 70% Hex, 20% EtOAc, 5% MeOH, 5% NEt3 to give
a bright yellow powder. Due to the low-yielding nature of the reaction, we were unable to obtain
full NMR characterization, but preliminary data is consistent with the expected product.
132
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
Et2N Cl
NH
H
N
S
O2
Cl N
4-chloro-N-(7-chloro-4-((5-(diethylamino)pentan-2-yl)amino)quinolin-3-
yl)benzenesulfonamide (3.17)
chlorobenzaldehyde (1.5 equiv.) under nitrogen followed by NEt3 (3 equiv.). The solution is stirred
and allowed to warm to RT. The reaction progress is monitored by TLC. After 24 hours the solvent
is removed under reduced pressure. The residue is then diluted with DCM, and the solution is
poured into a flask containing saturated NaHCO3 solution and is extracted with DCM (3 X 20 mL)
and washed with brine. The combined organic extracts are then dried over anhydrous MgSO4 and
the solvent removed under reduced pressure. The crude product is purified by prep-TLC with silica
gel, solvent system: 70% Hex, 20% EtOAc, 5% MeOH, 5% NEt3 to give a bright yellow powder.
Et2N Cl
NH
N
Cl N
(E)-N4-(7-chloro-3-((4-chlorobenzylidene)amino)quinolin-4-yl)-N1,N1-diethylpentane-1,4-
diamine (3.18)
133
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
3.6 References
(1) Edaye, S.; Tazoo, D.; Bohle, D. S.; Georges, E. 3-Halo Chloroquine Derivatives Overcome
(3) Bass, G. E.; Hudson, D. R.; Parker, J. E.; Purcell, W. P. Mechanism of Antimalarial Activity
(4) Edaye, S.; Tazoo, D.; Bohle, D. S.; Georges, E. 3-Iodo-4-Aminoquinoline Derivative
(5) Kaschula, C. H.; Egan, T. J.; Hunter, R.; Basilico, N.; Parapini, S.; Taramelli, D.; Pasini,
Role of the Group at the 7-Position. J. Med. Chem. 2002, 45 (16), 3531–3539.
(6) Lipinski, C. A. Lead- and Drug-like Compounds: The Rule-of-Five Revolution. Drug
(7) Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J. Experimental and
(9) Ertl, P. Craig Plot 2.0: An Interactive Navigation in the Substituent Bioisosteric Space. J.
134
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
(10) Hawley, S. R.; Bray, P. G.; Mungthin, M.; Atkinson, J. D.; O’Neill, P. M.; Ward, S. A.
42 (3), 682–686.
(11) Madrid, P. B.; Liou, A. P.; DeRisi, J. L.; Guy, R. K. Incorporation of an Intramolecular
(12) Egan, T. J.; Marques, H. M. The Role of Haem in the Activity of Chloroquine and Related
(13) Nsumiwa, S.; Kuter, D.; Wittlin, S.; Chibale, K.; Egan, T. J. Structure–Activity
Aminoquinolines Using Experimental and Ab Initio Methods. Bioorg. Med. Chem. 2013,
21 (13), 3738–3748.
(14) Thomé, R.; Lopes, S. C. P.; Costa, F. T. M.; Verinaud, L. Chloroquine: Modes of Action of
(16) Hänscheid, T.; Egan, T. J.; Grobusch, M. P. Haemozoin: From Melatonin Pigment to Drug
Target, Diagnostic Tool, and Immune Modulator. Lancet Infect. Dis. 2007, 7 (10), 675–685.
135
Chapter 3 Synthesis of Novel Substituted Chloroquine Derivatives
136
Chapter 4
Photoaffinity label.
137
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
4.1 Preamble
In drug discovery protein:compound interactions can be detected by covalently linking the drug to
the potential active site of the protein of interest. This technique is known as photoaffinity labelling
and facilitates the determination of a compound’s mechanism of action. The formation of a label
generally involves installing a photoreactive aryl azide or an aryl/ alkyl diazirine onto the
compound. Cross-linking between the compound and protein of interest occurs following the
moiety is generally accompanied with the installation of large functional groups that may impact
the activity of the parent compound and interact with surrounding proteins. Photoaffinity labels
(PAL) can mimic the biological mechanism of the parent compound, but one cannot ignore the
influence of any additional functional group. With this in mind, we wanted to synthesize a PAL
for CQ that required the least amount of modifications to the CQ structure. With our 3-NH2CQ
precursor, we decided that it was the ideal location to install an azide PAL. We discovered that the
secondary amine at the 4-position is labile and subject to cyclization to form a triazole product
during the installation of the azide. We were able to prevent cyclization via the placement of a
methyl group to give a tertiary amine at the 4-position. All modifications resulted in the addition
of 2 small functional groups, a methyl and azide, where the methyl is unreactive. The resulting
PAL is easily activated under UV irradiation. The successful installation of a PAL with minimal
structural modifications will allow for a more precise mechanistic determination of the substrate-
binding site. The PAL is currently being used for labelling studies at the Fidock group at Columbia
University.
138
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
4.2 Introduction
Quinoline-based antimalarials have been in use for centuries. Despite the long tenure of AQs on
the commercial market, their mechanism of action is still a matter of debate.1–5 There is a consensus
that blood-stage AQs interact with heme to inhibit the parasite’s proliferation; however, it is not
believed to be their sole function. The mechanism of quinoline accumulation into its active site
and its targets are still being investigated. Thus, to determine the site and identity of the proteins
with which the antimalarial drugs interact, several PALs have been previously synthesized, Figure
4.1. These labels are useful because they form strong covalent bonds with the surrounding proteins
and allow for the isolation and identification of the ligated product.
NEt2 NEt2
O HN HN
NH OMe
N3 OH N N3 N
ASA-Q 3-Azido-mepacrine
F
N3
F
F
O
HO
N F
O
I
N3 O O
N HN
N
NH HN N NH
H
S
OH O
ASA-MQ Cl N AzBCQ
When designing a PAL, one wants to mimic the biochemical binding sites of the parent compound
as closely as possible. This means that one must minimize the interactions of the label with
139
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
surrounding residues to avoid unnecessary interaction. Therefore, it is best to avoid adding reactive
functional groups to PALs. Known PALs of AQs and MP are shown in Figure 4.1 and include: N-
(l-(l-diethylamino-l-methylbutylamino)quinolin-6-yl)-4-azido-2-hydroxybenzamide (ASA-
the PALs mentioned above can cross-link to proteins of interest, their design omits certain core
functionalities of the parent compound. ASA-Q completely removes the 7-chloro group found in
CQ, which is essential for inhibiting hemozoin formation. It introduces an amide, hydroxy and
phenyl group, which can hydrogen bond and form additional p-stacking interactions. These are
unfavourable modifications as they can interact with the surrounding environment. 3-Azido-
mepacrine replaces the 7-chloro group found in MP for the photoreactive aryl azide, this is
unfavourable because, as mentioned above, the chloro group is required for hemozoin inhibition.
ASA-MQ, an MQ PAL, does a poor job of retaining the parent compound’s core functions.
Desneves et al. claim that ASA-MQ is 10-fold more potent than mefloquine and that ASA-MQ is
inactive in the CQR Kl Mef2 strain which is selected in vitro for resistance to MQ. AzBCQ is a
CQ PAL that contains a biotin tag and a perfluorophenylazido group, and the quinoline ring
remains unmodified. However, there is the addition of large groups on the side chain.
In this chapter, I describe the synthesis of the first PAL, which contains the least modifications,
where all the essential features for CQ activity remain unmodified, Figure 4.2. Photolysis studies
were carried out with the PAL to determine the optimal wavelength for its activation. Examples of
140
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
Figure 4.2. Structure of novel 3-Azidomethylchloroquine in which all important CQ features are
present.
141
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
We have synthesized a CQ PAL derivative with minimal modification of the quinoline structure.
The aryl azide is placed at the 3-position, a position that our group has previously utilized.10,11
Installation of the aryl azide can occur via Ullman coupling to form the Csp2 - N bond.12 This is
generally carried out in the presence of air, with a copper catalyst and ligand in a polar solvent,
with or without the presence of a base.13–15 In our experimentation, the azidation reaction from an
aryl bromide did not yield any trace of aryl azide. Rather an arylamine was the only product
observed. A further search of the literature shows that direct amination of aryl bromide with
sodium azide as an amine source is a common observation.16–18 Initially, Thatcher et al. suggest
this to be a result of thermal decomposition.19 Then Alami et al. proposed a mechanism of amine
formation via a nitrene intermediate, Scheme 4.1.17 However, they were unable to gather
experimental evidence of azide formation, but believe that the aryl azide is first formed and through
Cu(I) catalyzed thermo-initiation, N2 is liberated to form a nitrene, which abstracts protons and
N EtOH, 100oC N N N
Intermediate
However, Helquist et al. later disproved the notion of amine formation due to azide thermal
decomposition by heating an aryl azide in DMSO for 72 hrs and found that the azide remained
intact.16 They then heated an aryl bromide in the presence of a copper catalyst and DMEDA and
only recovered an azo compound. They determined that amine formation from an azide precursor
142
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
is dependent on the presence of a copper catalyst and excess sodium azide (see Table 2.2 – 2.4).
We did not isolate the desired azido product in our experiments, nor did we isolate any azo product;
only aryl amine was detected and isolated, Scheme 4.2. As shown in Chapter 2, regardless of the
Scheme 4.2. Ullman amination of aryl bromide in 30% aqueous solution in the presence of a ligand
With the arylamine in-hand, we continued to form the aryl azide via a route that first involves
forming a diazonium salt with sodium nitrite in the presence of sulfuric acid. Following the
diazonium’s formation, sodium azide is added to the reaction mixture to form the aryl azide.
However, the aryl azide was not isolated; rather, a cyclized triazole compound was isolated, as
depicted in Scheme 4.3. NMR and HRMS confirmed this structure. Formation of the triazole
indicates reactivity of the secondary amine at the 4-position. The reactive amine had to be
secondary amine and then deprotect after azide installation, there remains a high probability of
143
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
Cl N Cl N Cl N
0% 49%
4.3 4.3 4.4
Initially, the goal of this project was to synthesize a PAL with the least possible modifications of
the parent compound. To remain true to the objective, we opted to methylate the secondary amine.
Methylation allows for the addition of an unreactive functional group. The first attempt of
methylation involves the use of methyl iodide for direct methylation of CQ, Table 4.1.20,21
However, this method was low yielding and in some instances resulted in a dimethylated product.
Et2N Et2N
NH N
Cl N Cl N
0%
4.5 4.6
Methylating Temperature
Base Solvent Yield (%)
agent (oC)
NaH DMF MeI -20 to 0 23
NaH DMF MeI Room temp. 15
NaH DMF MeI -20 9
NaH DMF MeI -20 N.D.
NaH MeCN MeI -20 -
144
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
Varying reaction conditions did not improve product yield or selectivity, and the HRMS
continuously shows the formation of mono and dimethylated CQ. We then attempted methylating
the secondary amine via the formation of a formamide, followed by a reduction of the formamide
Cl N Cl N Cl N
4.5 4.7 4.6
Formulating
Solvent Temperature (oC) Catalyst 4.7 Yield (%)
agent
Rahman et al. reported the 60% yield of N-formamide formation from diphenylamine with formic
acid in the absence of solvent.22 Kim and Jang reported 82% formation of N-formamide from N-
methylaniline from formic acid with I2 as a catalyst,23 and Shastri et al. report the formation of N-
formamide with the use of sodium ethoxide in chloroform via the Riemer– Teimann reaction.24
However, in our experiments only starting material was present. Following these attempts, we
145
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
It was decided that methylation of the side chain followed by a nucleophilic aromatic substitution
(SNAr) reaction would be the most efficient method for the synthesis of MeCQ, Scheme 4.4.
Formylation of the primary amine with formic acid and acetic anhydride proved to be unsuccessful.
This is likely due to the insufficient formation of acetic formic anhydride before the addition of
primary amine. However, the reaction with a primary amine and formic acid produced the desired
formamide in a 57% yield.22 Reactivity is improved using ethyl formate to give 97% of a product
and easy recovery with a simple acid and base workup.25 When ethyl formate, is used the side
chain does not get protonated; as is the case with formic acid, the primary amine is more
nucleophilic resulting in our higher yield. Once the formamide is formed, the final step in the
formation of 4.10 is a reduction with lithium aluminum hydride,26 which results in a 65% yield.
Lastly, to form our desired MeCQ, we performed a condensation reaction between 4,7-
dichloroquinoline (DCQ), 4.11 and 4.10, Table 4.3. Generally, this type of condensation reaction
occurs under basic conditions. As the alkyl side chain contains a basic secondary amine, we opted
to carry out the reaction with the amine acting as the base and/or solvent.
146
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
Table 4.3. Condensation reaction between DCQ and methylated side chain under basic conditions.
Et2N
Cl N
Et2N
N
H
Cl N Cl N
4.11 4.10
4.3
Yield (%)
Base Solvent Temperature (oC) Side product % Yield of 4.3
- - 85 -
- - 120-125 6
- - 120a -
NEt3 - 125 -
NEt3 - 85a -
NEt3 EtOH 0-40 -
DBU - 80a 10
DBU - 125 82b -
DIPEA DMSO 80 -
DIPEA DMSO 120 -
DIPEA MeCN 85 -
t
BuOK DMSO 80-120 Decomp.
t
BuOK THF RT-80oC -
c
1,4-dioxane 85 8
a) Microwave-assisted reaction, b) 1-(3-((7-chloroquinolin-4-yl)amino)propyl)azepan-2-one
formed, see Scheme 4.4 b, c) Pd(OAc)2, DPEPhos, K3PO4.
There are numerous examples of thermal activation for this type of condensation reaction with
primary amines that give moderate to excellent yields.27–29 However, in our experiments, diamine
used as a base results in low yields of our product, even at elevated temperatures. The use of a
polar solvent such as ethylene glycol did not improve the yield. Therefore, it became clear that the
diamine was not nucleophilic enough to initiate the SNAr reaction; thus, we used stronger bases
147
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
Diisopropylethylamine (DIPEA) and potassium tert-butoxide (tBuOK). NEt3 did not result in
product formation, nor did DIPEA with various polar solvents. DBU gave 10% product yield at
80℃; however, when the temperature was increased to 125oC, we get 82% formation of a ring-
opened DBU adduct, as illustrated in Scheme 4.5. Such adducts of ring-opened amidine bases
have been observed in the literature, formed in the presence of an inorganic base.30,31 A recent
publication from Merck scientists details their discovery of a ring-opened DBU adduct during the
development of a hepatitis C drug. Hyde et al. hypothesized that the hydrolysis of DBU occurs in
trace amounts of H2O and high temperatures or during an aqueous workup.32 When examining the
reactivity of electrophilic reactions in the presence of DBU they proposed two pathways to the
observed ring-opened DBU adduct. In the first pathway, the amidine base is hydrolyzed generating
a primary amine, which reacts with an electrophile. In the second pathway, a cationic intermediate
is formed via a reversible interaction of an amidine and an electrophile. This then reacts with trace
amounts of H2O or an aqueous workup that results in the ring-opened product. In our experiments,
while monitoring the reaction with TLC, the product is formed before quenching the reaction; thus,
148
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
NH2
H2O Path 1 E
Hydrolysis first O
N N
E
N N
H
H2O
E
N
N
E
Path 2
a) Electrophile addition first
b)
Scheme 4.5. A) Potential pathways for DBU reaction with an electrophile. B) Crystal structure of
When potassium tert-butoxide is used, it leads to the decomposition of the reaction mixture and
149
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
Tf Et2N Et2N
Cl OH O N N
H
4.10
Cl N Cl N Cl N Cl N
4.11 4.12 4.13 4.6
Scheme 4.6. Increasing the electrophilicity of C-4 with the installation of a triflate.
We then concluded that the C-4 was not electrophilic enough; therefore, we opted for a better
leaving group than the chloride, Scheme 4.6. Triflic acid has a pKa value of -14, whereas
hydrochloric acid has a pKa of -8; therefore, we opted for the weaker conjugate base being the
triflate. Installation of the triflate involves nucleophilic substitution with acetic acid to give 7-
chloroquinolin-4-ol, 4.12. With the hydroxy in hand, we then carried out an electrophilic carbonyl
reaction to afford the triflate quinoline, 4.13.33 The final step to obtaining our desired MeCQ, 4.6,
was the condensation reaction with the methylated side chain, 4.10, Table 4.4.
Initially, the reactions were placed in an 80oC oil bath, and the reaction was monitored for one
hour. When monitoring the reaction, if no product formed after one hour, the flask was then
transferred into a 125oC oil bath and heated for a further hour before quenching. It is evident from
150
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
the results that using a weaker base as a leaving group more than doubled the product yield, thus
proving the hypothesis for a need to make C-4 more electrophilic. However, although yields were
We then decided to carry out the reaction under acidic conditions. There are examples in the
literature of this reaction being carried out in weakly acidic conditions with the use of phenol and
examples in strongly acidic conditions such as hydrochloric acid.34–37 The acid protonates the
secondary amine, which forms a quaternary ammonium cation, which would be slightly less basic
Table 4.5. Condensation reaction between DCQ 4.11, and methylated side chain 4.10 under
acidic conditions.
Et2N
Cl N
Et2N
N
Cl N H Cl N
4.10
4.11 4.6
Temperature Yield (%)
Acid Solvent Yield (%)
(oC) Side product
Phenola - 125 36b 14
Phenol - 125c 16
p-toluenesulfonic acid DMSO 125 -
Phenol is a weak acid with a pKa value of 10. It did not improve the reaction yield compared to
the use of strong bases. We observed a 7-chloro-4-phenoxyquinoline side product formation in the
reaction mixture containing phenol and NEt3. This side product has been reported by other research
151
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
groups, formed in the absence of NEt3.38 As phenol did not optimize the reaction, we decided to
use a stronger organic acid p-toluenesulfonic acid with a pKa of -3, Table 4.5. The reaction
containing DMSO did not yield any product, which might be due to hydrogen bonding between
the sulfonic acid and the sulfoxide when prevented the protonation of the amine. Pan et al. reported
similar reaction conditions between primary and secondary amines with 4-chloro-2-
cyclic secondary amine, quinoline, DMF and high temperatures give yields ranging 25 – 52%.
However, they demonstrated that they could produce good to excellent yields, 55 – 75%, under
microwave-assisted conditions and yields were improved to 81% when the solvent was removed
from the reaction.39 In our experiments, a microwave-assisted system without the use of solvent
improved the yield of the reaction to 63%. With this improved result we were satisfied with the
yield and proceeded to the final stage of the synthesis to form 3-azidomethylchloroquine (3-
N3MeCQ), 4.17.
to form the 3-NO2MeCQ, 4.15, see Scheme 4.7. Remarkably, in the case of 4.14, the optimal
152
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
reaction conditions are with the use of DIPEA in acetonitrile. This reaction occurs very quickly
and gives quantitative yields. An explanation for this observation is the proximity of the electron-
withdrawing nitro group, which greatly increases the electrophilicity of the C-4 position. The nitro
4.16. The final step is converting the amine to azide via a diazonium salt with sodium nitrite and
sulfuric acid. The diazotization reaction occurs immediately following the addition of sodium
nitrite. However, a 3-azido-7-chloroquinolin-4-ol, 4.18 side product begins to form just as rapidly;
thus, the addition of sodium azide followed by quenching the reaction with sodium carbonate must
be done within 10 minutes of starting the reaction. A proposed pathway for side product formation
is shown below, Scheme 4.8. Once the diazonium ion forms, it can form an iminium salt that
results in an electrophilic C-4. This is then subject to nucleophilic attack by H2O. The methylated
side chain is then expulsed to give 4.22. The formation of 4.18 occurs after the addition of sodium
azide. The side product is insoluble in the acidic media and is filtered out of the solution to allow
Cl N Cl N Cl N Cl N Cl N
4.19 4.20 4.21 4.22 4.18
Et2N
N
4.10 H
To reduce side product formation, we carried out the reaction in an organic solvent to minimize
H2O concentration. All reagents used have a degree of solubility in MeOH which was the chosen
solvent. The strong organic acid p-toluenesulfonic acid was chosen, and this reaction was carried
153
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
out at 0oC to RT and stirred for 18 hours. TLC showed minimal product conversion. When the
time elapsed and purification was performed only 1.4% of the product was isolated with no side
product formation. Factors affecting product formation for the new method included partial
solubility of sodium nitrite and sodium azide, therefore a lower rate of formation of the diazonium
intermediate. Another factor affecting the formation of the product is the pH of the solution. For
the nitrosonium ion to form an acidic environment is needed, which is not the case with p-
With the desired 3-N3MeCQ in hand, we have synthesized the first known quinoline-based PAL
with the least modifications to the parent compound. In doing so, we are able to retain all the
We first determined the UV-Vis spectroscopic difference between CQ, 3-NH2MeCQ and 3-
N3MeCQ. The spectroscopic profile for CQ diphosphate salt was prepared in H2O and measured
at pH 7.3. The samples for 3-NH2MeCQ and 3-N3MeCQ were prepared in MeOH. As expected,
CQ has a very different profile to its derivatives, whereas there are several similarities between 3-
NH2MeCQ and 3-N3MeCQ. As shown in Figure 4.3, there is a 6 nm bathochromic shift in the
lmax of the amine compared to the azide from 237 nm to 243 nm. Both compounds have weak
p®p* bands at 278 nm and 282 nm for the amine and azide, respectively, and weaker n®p* bands
at 357 nm and 358 nm for the amine and azide, respectively. CQ diphosphate is a colourless salt
and this is proven in the spectra where the absorbances are in the ultraviolet region. 3-NH2MeCQ
and 3-N3MeCQ have a yellow tint, this is due to the tailed absorbance at 400 nm.
154
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
3-MeN3CQ
3-MeNH2CQ
Absorbance
CQ.2H3PO4
0
200 300 400
wavelength
Figure 4.3. Overlaying spectra of 3-NH2MeCQ, 3-N3MeCQ and CQ.2H3PO4 for comparison.
We then determined the excitation wavelength for the photolabile moiety. It has been reported that
aryl azides can be irradiated at 254 nm without causing damage to the compound; therefore, we
decided to test its stability at that excitation wavelength, Figure 4.4.40,41 The azide was first
exposed to 254 nm using a UV mercury penlight in the dark, exposure lasted 1 hour, and the spectra
were taken at different time intervals. As shown in the spectra, photoexcitation/ decomposition is
rapid as the p®p* band disappears within one minute. However, photodecomposition begins to
occur within 30 minutes, with the compound having little UV-Vis signature after one hour.
155
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
1.5
1 minute
Absorbance
3 minutes
1.0 8 minutes
18 minutes
0.5 30 minutes
1 hour
0.0
200 300 400
Wavelength (nm)
With this observation, we decided that photolysis at 254 nm was too energetic and tried
photoexcitation at 365 nm. There are also many examples of azide irradiation above 300 nm which
result in insertion reactions.6,42,43 We carried out stability tests in H2O and MeOH, and the aryl
azide remains intact after longer than 18 hours with exposure to 365 nm of light, Figure 4.5.
Due to the high reactivity of the nitrene, the below spectra are the results of product formation
from the nitrene reacting with the polar solvent, Figure 4.5. These results show that the aryl azide
is indeed photolabile; however, it is also a cause of concern that the reaction with the solvent will
occur faster than insertion with a chosen compound, making detection of azide-compound
156
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
30 seconds 30 seconds
285 1 minute 1 minute
2 minutes 287 2 minutes
Absorbance
3 minutes 3 minutes
Absorbance
4 minutes 4 minutes
0.5
5 minutes 5 minutes
0.5
363
360
0.0
250 300 350 400 450 0.0
Wavelength (nm) 250 300 350 400 450
Wavelength (nm)
Figure 4.5. Photolysis of 42.97 µM N3 in H2O and 35.57 µM N3 in MeOH at 365 nm.
We then carried out the same reaction in an NMR tube with a 365 nm penlight in the dark in
deuterated MeOH.
0 minutes l g
k g e
N
i
h
e l
a f N
j
bd c
N3 k
c b
j h, i
Cl N a f h’
d
16 minutes
9.5 9.0 8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5
f1 (ppm)
157
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
0 minutes
2 minutes
6 minutes
11 minutes
16 minutes
9.2 9.1 9.0 8.9 8.8 8.7 8.6 8.5 8.4 8.3 8.2 8.1 8.0 7.9 7.8 7.7 7.6 7.5 7.4 7.3 7.2 7.1
f1 (ppm)
Changes become evident after 2 minutes of exposure. As seen in the UV spectra, the p ® p* band
disappears, in the NMR spectra we can see the aromatic protons decrease in intensity as additional
peaks arise. Additional photodecomposition occurs with longer exposure times, Figure 4.7, the
aromatic region is more affected than the aliphatic regions as one would expect. Therefore, when
carrying out photolysis experiments exposure times did not exceed 10 minutes. High-Resolution
Mass Spectroscopy was carried out for the azide samples in H2O and MeOH. We discovered the
formation of 3 consistent molecular ions with their possible structures shown below, Figure 4.8.
158
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
The formation of nitroso-methylCQ occurs in the presence of H2O and MeOH when irradiated
at 365 nm for 5 minutes, this might be a result of hydroxyl insertion. Interestingly, only in the
presence of H2O do we get the formation of a nitroso-chloroquine derivative that has lost a methyl
group. The formation of this product may also result from H2O insertion and the HRMS data
leads us to believe the shown structure is the most probable product. Not-so surprisingly, we have
the formation of an intramolecular insertion product; however, it only formed in the presence of
MeOH. Formations of such azirines are well documented for phenyl azide, which exists in
N N N
N NH N
N
N N
O O
Cl N Cl N Cl N
Formed in the presence Formed only in the Formed only in the
of water and methanol presence of water presence of methanol
We carried out experiments to determine if the expected singlet nitrene intermediate that forms
inserts into C-H, C-N or C=C bonds. To achieve this, we first used a simple tripeptide
GlyGlyGly. A UV-Vis spectrum of the azide and GlyGlyGly was taken at different
stoichiometries, Figure 4.9, and the change in the UV-Vis signature is evident at the different
concentrations. From the UV-Vis spectra it is clear that there is a change in the aromatic system
of the aryl azide when mixed with the peptide. To confirm that insertion occurred with the peptide
159
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
we carried out HRMS for the resulting product. However, there were no peaks related to an
21.49uM N3 and GlyGlyGly in H2O (1:1) 4.29uM N3 and GlyGlyGly in H2O (1:9.4)
0.08
0 secs 0 secs
245 10 secs 10 secs
20 secs 245 20 secs
0.4 30 secs 30 secs
0.06
1 minute 1 minute
2 minutes 2 minutes
285 3 minutes 3 minutes
285
Absorbance
Absorbance
4 minutes 4 minutes
0.04
5 minutes 5 minutes
0.2
0.02
358
358
0.00
0.0
250 300 350 400 450 250 300 350 400 450
Wavelength (nm) Wavelength (nm)
Figure 4.9. Photolysis of 21.49 µM 3-N3MeCQ with 22.32 µM GlyGlyGly and 4.29 µM 3-
We then decided to carry out UV-Vis and NMR studies to trap the nitrene with strong electron
acceptors. First, we used N-phenylmaleimide with 3-N3MeCQ in the dark exposed to 365 nm of
light, Figure 4.10. At higher concentrations, the absorbance associated with N-Phenylmaleimide
begin to obscure that of the quinoline; however, it seems to follow a similar pattern to that of the
control experiment. The HRMS did not detect any molecular ions associated with a possible
insertion product.
160
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
19.56uM N3 and N-Phenylmaleimide in MeOH (1:1) 3.56uM N3 and N-Phenylmaleimide in MeOH (1:10.6)
0.8 218
220 0 secs 0 secs
1.0
10 secs 10 secs
20 secs 20 secs
30 secs 30 secs
0.6
1 minute 1 minute
2 minutes 2 minutes
3 minutes 3 minutes
Absorbance
Absorbance
4 minutes 4 minutes
0.4
286 5 minutes 5 minutes
0.5
286
0.2
354
0.0 0.0
250 300 350 400 450 250 300 350 400 450
Wavelength (nm) Wavelength (nm)
Figure 4.10. Photolysis of 19.56 µM 3-N3MeCQ with 18.77 µM maleimide, 7.11 µM 3-N3MeCQ
with 33.37 µM maleimide and 3.56 µM 3-N3MeCQ and 37.54 µM maleimide at 365 nm excitation
over 5 minutes.
We then carried out the same experiment in an NMR tube to detect product formation at higher
concentrations of both reagents. As shown in Figure 4.11, doublets appear at 6.53 ppm and 6.29
ppm. To further investigate N-phenylmaleimide, was irradiated alone in MeOH for a total of nine
minutes, and we saw the appearance of the vinyl protons indicating independent activation of the
161
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
3-N3MeCQ
3-N3MeCQ + N-Phenylmaleimide
365 nm for 6 minutes
6.53 4
6.29
9.5 9.0 8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5 0.0
f1 (ppm)
Figure 4.11. Photolysis of 26.7mM 3-N3MeCQ and 26.6mM maleimide at 365 nm in d4-MeOD.
Hott and Heusinger (1977) investigated the photolysis of maleimides in polar solutions using
electron spin resonance.47 They note the detection of radical anions of maleimides in alcohol
solutions. They determined that radical anions are the primary ion species. Kiselev et al. have
reported the slow hydrolysis of N-phenylmaleimide to the acid in the presence of H2O.48 It is
possible that the radical anion formation speeds up the hydrolysis of the maleimide, resulting in
162
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
No light
O
N
2
O
8.0 7.9 7.8 7.7 7.6 7.5 7.4 7.3 7.2 7.1 7.0 6.9 6.8 6.7 6.6 6.5 6.4 6.3 6.2 6.1 6.0
f1 (ppm)
We then decided to use the stronger electron acceptor tetracyanoethylene (TCNE). Murata et al.
reported the photochemical reaction of mesityl azide with TCNE with 360 nm ± 15 nm light
produced two adducts formed via singlet nitrene trapping.49 Irradiation of our aryl azide and TCNE
for 9 minutes shows the formation of a minor product (23% conversion); however, the HRMS of
the mixture did not detect the formation of the expected photolysis product but rather corresponds
163
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
CN
N NC N
N CN N
H
N N CN
CN
Cl N Cl N
Expected product Possible product
a) 23% conversion
9.3 9.2 9.1 9.0 8.9 8.8 8.7 8.6 8.5 8.4 8.3 8.2 8.1 8.0 7.9 7.8 7.7 7.6 7.5 7.4 7.3 7.2 7.1 7.0
b) f1 (ppm)
Figure 4.13. a) Desired product v possible observed photolysis product, b) NMR of photolysis
product formation.
The exact mass and chemical formula obtained from HRMS ([M+H+] m/z: 374.21) led us to the
proposed structure, Figure 4.13. This product was not isolated and longer excitation times leads
164
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
4.4 Conclusion
We have synthesized the first photolabile CQ derivative with minimal modifications of the parent
compound. The modification involved installing a methyl group on the 4-AQ to prevent
cyclization to a triazole derivative. In order to install the amine we first methylated the side chain
and then carried out a condensation reaction with 4,7-dichloro-3-nitroquinoline. Once the desired
azide was obtained we carried out UV-Vis stability tests to determine the best-suited
photoexcitation energy. We found that irradiation at 254 nm was too energetic and led to the
decomposition of the aromatic system and 365 nm of light was the most suitable as it activated the
azide, causing a release of nitrogen gas to form a new compound. The reactive nitrene intermediate
species forms an adduct with the surrounding solvent which was detected by HRMS. We then
carried out photolysis studies with a GlyGlyGly, N-phenylmaleimide and tetracyanoethylene and
discovered that the electron acceptor and the solvent determine the conditions for insertion. We
could not detect the expected insertion product with TCNE in which the nitrene intermediate is
inserted into the C-N triple bond; however, we detected a possible HCN insertion product. The
PAL is currently being used for photoaffinity labelling on PfCRT with the Fidock group at
Columbia University.
165
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
4.5 Experimental
Glassware was taken directly from the oven (120oC) and let cool in a desiccator before use. NMR
experiments were carried out in New Era NMR routine grade tubes 5 mm X 8”. All solvents and
reagents were obtained commercially and used without further purification unless noted.
UV irradiation was carried out using Analytik Jena UVP Pen-Ray UV mercury lamp 254 nm and
365 nm. High-Resolution Mass Spectroscopy (HRMS) was obtained by positive/negative ESI, or
Orbitrap, with results reported as mass/charge ratios (m/z). Thin-layer chromatography plates were
visualized using ultra-violet light, 254nm. Flash column chromatography was carried out manually
using 230 - 400 mesh silica gel (Silicycle) using reagent grade solvents or using Biotage Isolera
One system with Biotage ZIP 30 g columns. Cole Parmer Microcomputer pH-Vision Model
05669-20 pH Meter used with Sigma-Aldrich micro pH combination electrode and glass body.
1 13
H and C NMR spectra were recorded at ambient temperature on Bruker AVIIIHD 400 MHz
(1H 400 MHz, 13C 100 MHz) and Bruker AVIIIHD 500 MHz (1H 500 MHz, 13C 125 MHz) using
tetramethylsilane as the internal standard. Chemical shifts are reported relative to the residual
deuterated solvent peaks. Chemical shifts are expressed in parts per million (ppm = δ) values and
coupling constants (J) in Hertz (Hz). The terms m, s, d, t and q represent multiplicities of 1H NMR
resonances: multiplet, singlet, doublet, triplet and quartet, respectively. For previously unknown
complete the assignment of 1H and 13C signals. 1H NMR signals are described by chemical shift δ
166
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
13
(multiplicity, J (Hz), integration). C NMR signals are described by chemical shift δ and are
N
N N
N
2H3PO4
Cl N
4-(7-chloro-1H-[1,2,3]triazolo[4,5-c]quinolin-1-yl)-N,N-diethylpentan-1-amine diphosphate
(4.4)
mmol) in of 3.2 M sulfuric acid (98%), is cooled to 0oC and a 1 mL solution of 2.5 M sodium
nitrite is added dropwise. The reaction mixture is stirred at this temperature for 15 minutes. After
15 minutes, 1 mL of 4.5 M sodium azide is added dropwise to the reaction mixture at 0oC. Addition
of azide results in visible N2 gas evolution. The reaction mixture is left to stir for a further 1 hour
and the mixture is allowed to warm from 0oC to RT. Once complete, the reaction mixture is treated
with saturated Na2CO3 solution and the compound is extracted with DCM (3 X 10 mL). The
combined organic layers are rinsed with brine and dried over anhydrous MgSO4. The solvent is
removed under reduced vacuum to reveal a bright orange oil. The oil was purified by flash
chromatography on silica gel with the solvent system: 55% Hex, 40% DCM, 5% NEt3 to give a
The free-base is then converted to a phosphate salt with phosphoric acid as follows: To a
amine (24 mg, 0.069 mmol) in 1 mL of MeOH is added, with cooling, a phosphoric acid solution
167
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
made from 0.14 g of 85% phosphoric acid and 3 mL of MeOH. Isopropyl alcohol is then added to
separate the salt from the solution as an oil. The oil is triturated with ether and stirred until it
solidifies. The mixture was then filtered, the diphosphate washed with ether and quickly placed in
Yield: 26.3 mg, 70%. 1H NMR (400 MHz, Deuterium Oxide-d2) δ 9.26 (s, 1H), 8.26 (d, J = 9.0
Hz, 1H), 7.78 (d, J = 2.1 Hz, 1H), 7.69 (dd, J = 8.9, 2.2 Hz, 1H), 5.43 (p, J = 6.6 Hz, 1H), 3.19 –
3.09 (m, 6H), 2.47 – 2.34 (m, 1H), 2.28 – 2.15 (m, 1H), 1.81 (d, J = 6.6 Hz, 3H), 1.79 – 1.63 (m,
2H), 1.22 – 1.18 (m, 6H). 13C NMR (126 MHz, Deuterium Oxide-d2) δ 144.70, 142.82, 139.06,
135.94, 133.40, 129.23, 126.81, 123.73, 113.20, 57.99, 50.95, 47.23, 32.16, 19.96, 19.89, 8.07 (d,
J = 4.6 Hz). 31P NMR (162 MHz, Deuterium Oxide-d2) δ 0.03. HRMS (ESI) calcd. for C18H25ClN5
N
N
Cl N
N4-(7-chloroquinolin-4-yl)-N1,N1-diethyl-N4-methylpentane-1,4-diamine (4.6)
1,4-diamine (652.6 mg, 3.8 mmol, 1.5 equiv.) are placed in a microwave flask with p-
toluenesulfonic acid (816 mg, 4.3 mmol, 1.7 equiv.). This mixture was heated at 120oC for 1.5
hours. Once complete, the reaction is let cool to RT. The mixture is poured into ice H2O, and then
1 M NaOH is added. This is extracted with EtOAc (3 X 100 mL), washed with brine, and the
organic layers are then dried over anhydrous MgSO4. The solvent is removed under reduced
vacuum to reveal an orange oil. The oil was purified by flash chromatography on silica gel with
168
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
the solvent system: 50% Hex, 50% to 100% EtOAc and 5 - 10% NEt3 to give an orange oil 534.5
Yield: 534.5mg, 63%. 1H NMR (500 MHz, Chloroform-d) δ 8.60 (d, J = 5.2 Hz, 1H), 7.99 (d, J
= 2.2 Hz, 1H), 7.88 (d, J = 9.0 Hz, 1H), 7.35 (dd, J = 9.0, 2.2 Hz, 1H), 6.75 (d, J = 5.2 Hz, 1H),
3.85 (h, J = 6.8 Hz, 1H), 2.85 (s, 3H), 2.43 (qd, J = 7.1, 1.6 Hz, 4H), 2.34 – 2.29 (m, 2H), 1.76 –
1.65 (m, 1H), 1.56 – 1.47 (m, 1H), 1.46 – 1.31 (m, 0H), 1.25 (d, J = 6.5 Hz, 3H), 0.94 (t, J = 7.1
Hz, 6H). 13C NMR (126 MHz, Chloroform-d) δ 157.55, 151.53, 150.78, 134.64, 128.90, 125.98,
125.22, 121.86, 109.02, 58.80, 52.87, 46.91, 32.49, 32.35, 24.64, 17.10, 11.70. HRMS (APCI)
O
N
N H
H
N-(5-(diethylamino)pentan-2-yl)formamide (4.9)
equiv.) is heated at 70°C for 18 hours in a round-bottom flask with molecular sieves. After
completion the mixture was cooled to RT and EtOAc was added. The resulting solution was
washed with 1 M HCl and extracted with EtOAc (3 X 150 mL). The organic layer was then basified
with 1 M NaOH and extracted with EtOAc (3X 200 mL) then rinsed with brine and dried over
anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure to get N-(5-
Yield: 5.689 g, 97%. Major trans rotamer: 1H NMR (500 MHz, Chloroform-d) δ 8.10 (s, 1H),
6.55 (s, 1H), 4.08 – 3.97 (m, 1H), 2.53 (dd, J = 7.2, 4.7 Hz, 4H), 2.45 – 2.35 (m, 2H), 1.50 (dd, J
169
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
= 6.2, 3.8 Hz, 4H), 1.15 (d, J = 6.6 Hz, 3H), 1.04 – 1.00 (m, 6H). 13C NMR (126 MHz, Chloroform-
d) δ 160.68, 53.02, 46.77, 44.01, 34.80, 23.14, 20.84, 11.42. Minor cis rotamer: 1H NMR (500
MHz, Chloroform-d) δ 8.07 (d, J = 12.0 Hz, 1H), 5.93 (s, 1H), 3.49 (s, 1H), 2.53 (dd, J = 7.2, 4.7
Hz, 4H), 2.45 – 2.35 (m, 2H), 1.50 (dd, J = 6.2, 3.8 Hz, 4H), 1.21 (d, J = 6.6 Hz, 3H), 0.99 (s,
13
6H). C NMR (126 MHz, Chloroform-d) δ 163.78, 52.65, 48.37, 46.86, 36.00, 23.58, 22.79,
11.62. HRMS (APCI) calcd. for C10H23ON2 [M+H]: 187.18049, found: 187.18033.
N
N
H
N1,N1-diethyl-N4-methylpentane-1,4-diamine (4.10)
mmol), which is dissolved and degassed in anhydrous THF, is added portion-wise under stirring
and cooling with an ice bath. The mixture is let stir at 0oC for 30 minutes, and then the reaction
mixture is refluxed for 24 hours under nitrogen. After completion 1 M NaOH is added dropwise
to hydrolyze any remaining LiAlH4. All precipitates are filtered off and washed with THF. The
filtrate is then placed in a separatory funnel and the organic layer separates. The organic layer is
then acidified with 1 M HCl and extracted with DCM (3 X 200 mL). The combined organic layers
are then basified with sat. NaOH and extracted with DCM (3 X 300 mL). The organic layers are
then rinsed with brine and dried over anhydrous MgSO4 and the solvent is removed under reduced
Yield: 2.685 g, 65% yield. 1H NMR (500 MHz, Chloroform-d) δ 2.52 (q, J = 7.2 Hz, 4H), 2.40
(d, J = 6.5 Hz, 5H), 1.51 – 1.42 (m, 3H), 1.34 – 1.23 (m, 1H), 1.05 (d, J = 6.3 Hz, 3H), 1.01 (t, J
170
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
= 7.2 Hz, 6H). 13C NMR (126 MHz, Chloroform-d) δ 55.07, 53.32, 46.87, 34.91, 33.79, 23.68,
19.79, 11. HRMS (APCI) calcd. for C10H25N2 [M+H]: 173.20123, found: 173.20097.
N NH
Cl N
1-(3-((7-chloroquinolin-4-yl)amino)propyl)azepan-2-one
mg, 0.58 mmol) and DBU (0.43 mL, 2.9 mmol) are placed in a round bottom flask and heated to
125oC. The mixture was let stir for 18 hours. The brown mixture is then cooled to RT and washed
with sat. NaOH and extracted with EtOAc (3 X 100 mL). The combined organic layers are then
rinsed with H2O and dried over anhydrous MgSO4 and the solvent was removed under reduced
pressure. The oil was purified by flash chromatography on silica gel with the solvent system: 85%,
EtOAc, 10% MeOH and 5% NEt3 to give a white solid, 173.8 mg, 82% yield.
Yield: 173.8 mg, 82%.1H NMR (500 MHz, Chloroform-d) δ 8.46 (d, J = 5.5 Hz, 1H), 8.00 (d, J
= 9.0 Hz, 1H), 7.95 (d, J = 2.2 Hz, 1H), 7.39 (dd, J = 9.0, 2.2 Hz, 1H), 7.02 (s, 1H), 6.39 (d, J =
5.6 Hz, 1H), 3.53 – 3.47 (m, 2H), 3.41 – 3.33 (m, 4H), 2.64 – 2.58 (m, 2H), 1.85 – 1.70 (m, 6H),
1.67 (p, J = 5.6 Hz, 2H). 13C NMR (126 MHz, Chloroform-d) δ 177.42, 151.23, 150.33, 148.73,
135.16, 127.91, 125.47, 122.25, 117.65, 98.09, 49.88, 45.11, 38.47, 37.17, 29.93, 28.44, 25.63,
23.46. HRMS (ESI) calcd. for C18H23ClN3O [M+H]: 332.1524, found: 332.1520.
171
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
N
N
NO2
Cl N
N4-(7-chloro-3-nitroquinolin-4-yl)-N1,N1-diethyl-N4-methylpentane-1,4-diamine (4.15)
diisopropylethylamine (0.54 mL, 1.5 equiv.) which is stirred until it formed a homogenous
solution. This mixture is let stir at RT for up to 10 minutes. The reaction mixture is then transferred
to an 85oC oil bath and let stir for 30 minutes. Once complete the reaction mixture is evaporated
to dryness under reduced pressure. The residue is diluted with EtOAc and H2O and basified with
1 M NaOH. The resulting solution is extracted with EtOAc (3 X 100 mL). The combined organic
layers were then rinsed with brine and dried over anhydrous MgSO4. The solvent is removed under
reduced pressure. The crude product is purified by flash silica column chromatography with 85%
Hex, 10% EtOAc, 5% NEt3 to reveal a bright orange solid, 719.7mg, 92%.
Yield: 719.7 mg, 92%. 1H NMR (500 MHz, Chloroform-d) δ 8.93 (s, 1H), 8.06 (d, J = 2.2 Hz,
1H), 8.02 (d, J = 9.2 Hz, 1H), 7.50 (dd, J = 9.1, 2.2 Hz, 1H), 4.03 – 3.95 (m, 1H), 2.85 (s, 3H),
2.51 (d, J = 7.3 Hz, 4H), 2.42 (d, J = 7.7 Hz, 2H), 1.89 – 1.81 (m, 1H), 1.65 (dq, J = 13.5, 8.0 Hz,
1H), 1.47 (q, J = 7.8 Hz, 2H), 1.43 (d, J = 6.6 Hz, 3H), 1.00 (t, J = 7.1 Hz, 6H). 13C NMR (126
MHz, Chloroform-d) δ 151.13, 150.70, 147.90, 137.63, 137.04, 129.68, 127.55, 127.18, 123.82,
60.99, 52.76, 46.94, 33.91, 32.73, 24.46, 18.19, 11.42. HRMS (APCI) calcd. for C19H27ClN4O2
172
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
N
N
NH2
Cl N
7-chloro-N4-(5-(diethylamino)pentan-2-yl)-N4-methylquinoline-3,4-diamine (4.16)
mmol) and stannous chloride (1.49 g, 5 equiv.) are placed in a round bottom flask. EtOH is added
and the mixture was let stir at 80oC for 2 hours. Once complete the mixture is let cool to RT. Once
cool, the solvent is removed by reduced pressure. The residue is diluted with EtOAc and basified
with sat. NaOH and extracted with EtOAc (3 X 50 mL). The combined organic layers are washed
with brine and dried over anhydrous MgSO4. The solvent is removed under reduced pressure to
reveal a dark orange oil. This was purified by column chromatography with the solvent system:
EtOAc (100→90%)/ MeOH (0→10%)/ NEt3 (5%) to give an orange oil, 313 mg, 68% yield.
Yield: 313 mg, 68%. 1H NMR (400 MHz, Dimethyl sulfoxide-d6) δ 9.40 (s, 1H), 8.62 (s, 1H),
7.97 (d, J = 9.2 Hz, 1H), 7.92 (d, J = 2.2 Hz, 1H), 7.53 (dd, J = 9.2, 2.2 Hz, 1H), 3.56 – 3.47 (m,
1H), 3.10 – 2.99 (m, 3H), 2.93 (s, 5H), 1.66 – 1.48 (m, 4H), 1.18 (d, J = 6.5 Hz, 3H), 1.12 (t, J =
13
7.2 Hz, 6H). C NMR (101 MHz, Dimethyl sulfoxide-d6) δ 140.47, 139.38, 138.79, 138.67,
130.61, 127.23, 126.25, 125.55, 124.01, 55.24, 50.92, 46.45, 46.30, 35.02, 31.32, 20.25, 17.77,
8.55 (d, J = 1.7 Hz). HRMS (ESI) calcd. for C19H30ClN4O2 [M+H-]: 349.2153, found: 349.2143.
173
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
N
N
N3
Cl N
N4-(3-azido-7-chloroquinolin-4-yl)-N1,N1-diethyl-N4-methylpentane-1,4-diamine (4.17)
is dissolved in a solution of concentrated sulfuric acid 98% and H2O (3.2 M). The resultant solution
is cooled to 0°C; to this, a sodium nitrite solution in H2O (2.5 M) is added dropwise with stirring.
The solution was stirred at this temperature for 5 min. Then a solution of sodium azide in H2O (4.5
M) is added to it with vigorous stirring. The mixture was stirred for 5 minutes at 0°C. The reaction
mixture is then quenched with sat. NaCO3 and extracted with EtOAc (3 X 25 mL). The combined
extracts are rinsed with brine and dried over anhydrous MgSO4 and solvent is removed under
reduced pressure to reveal a dark orange oil. This oil is purified by column chromatography with
the solvent system: 65% EtOAc, 35% Hex, 5% NEt3 to reveal a dark orange oil, 101 mg, 28%.
Yield: 101 mg, 28%. 1H NMR (500 MHz, Chloroform-d) δ 8.67 (s, 1H), 8.00 (d, J = 2.2 Hz, 1H),
7.98 (d, J = 9.0 Hz, 1H), 7.42 (dd, J = 9.0, 2.2 Hz, 1H), 3.49 (q, J = 6.4 Hz, 1H), 2.49 (q, J = 7.2
Hz, 4H), 2.38 (t, J = 7.3 Hz, 2H), 1.69 – 1.61 (m, 1H), 1.53 – 1.43 (m, 3H), 1.21 (d, J = 6.5 Hz,
13
3H), 0.98 (t, J = 7.1 Hz, 6H). C NMR (126 MHz, Chloroform-d) δ 147.93, 146.13, 145.08,
134.30, 128.91, 128.14, 127.48, 125.96, 125.91, 58.61, 53.02, 46.94, 35.52, 32.74, 24.11, 17.90,
11.54. IR (cm-1) 2966.6, 2933.2, 2110.6, 1559.3, 1451.4, 1381.8, 1319.9, 1073.4, 915.8. HRMS
174
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
OH
N3
Cl N
3-azido-7-chloroquinolin-4-ol (4.18)
See procedure for 4.17 and 4.18 was a precipitate that was filtered, washed with H2O and dried
under house vacuum overnight to give a brown solid, 81.1 mg, 39%.
Yield: 81.1 mg, 39%. 1H NMR (500 MHz, Dimethyl sulfoxide-d6) δ 12.23 (d, J = 6.2 Hz, 1H),
8.11 (d, J = 8.7 Hz, 1H), 7.95 (d, J = 6.3 Hz, 1H), 7.63 (d, J = 2.0 Hz, 1H), 7.38 (dd, J = 8.7, 2.0
Hz, 1H). 13C NMR (126 MHz, Dimethyl sulfoxide-d6) δ 171.72, 139.35, 136.45, 130.19, 127.04,
124.10, 122.37, 120.30, 117.71. IR (cm-1) 3075.6, 2124.7, 1627.9, 1558.2, 1512.3, 1460.4, 1350.8,
1188.7, 1075.2, 812.8. HRMS (ESI) calcd for C9H5ClN4O [M+Na+]: 243.0044, found: 243.0043.
one are grown from DCM and Hex. A large 0.1 mm x 0.1 mm x 0.06 mm prism is mounted on a
glass fibre with epoxy resin. Single-crystal X-ray diffraction is then measured at RT with a
monochromated MoKα radiation (λ = 0.71073 Å). The crystal crystallizes in the monoclinic space
group P21/c with the unit cell parameters a = 10.65(2); b = 16.68(3); c = 10.68(2) Å and α = γ =
integration of the intensity reflections and scaling and SADABS for absorption correction. A
combination of intrinsic phasing and direct methods were used for solving the structure which
were subsequently anisotropically refined after all non-hydrogen atoms were located by difference
Fourier maps and final solution refinements are solved by full-matrix least-squares method on F2
175
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
of all data, by using SHELXTL software. The hydrogen atoms are placed in calculated positions.
2.9 mg of 3-N3MeCQ is dissolved in 1 mL of MeOH and the resulting mixture is diluted with 29
deionized H2O to make a 669.6 µM stock solution. Then 6.5 mg of N-Phenylmaleimide was
General procedure
UV-Vis Spectrophotometer for the area 190 nm – 1100 nm. To a different 4 mL quartz cuvette is
added 333 µL of the 257.8 µM stock solution and 1,667 µL of deionized H2O to make a 42.97 µM
solution. A stir bar is placed in the cuvette and the cuvette is capped and placed in a cuvette holder
fitted with a 365 nm penlight and the setup is then covered with aluminum foil. The control
spectrum is measured with the light off. The experiment is then started in kinetics mode for a
spectrum to be taken every 10 seconds for 300 seconds whilst the light is on and the solution is
stirring. When the experiment is finished the resulting solution is transferred into a 2mL Eppendorf
176
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
To a different 4 mL quartz cuvette the stock 3-N3MeCQ solution is diluted with deionized H2O or
MeOH to make the desired concentrated solution. This is thoroughly mixed using a micropipette
to make a homogeneous solution. A stir bar is placed in the cuvette and the cuvette is capped and
placed in a cuvette holder fitted with a 365 nm penlight and the setup is then covered with
aluminum foil. The control spectrum is measured with the light off. The experiment is then started
in kinetics mode for a spectrum to be taken every 10 seconds for 300 seconds whilst the light is on
and the solution is stirring. When the experiment is finished the resulting solution is transferred
Insertion experiments
5,000 µL of the 257.8 µM 3-N3MeCQ stock solution is further diluted into 30 mL of deionized
H2O to make a 42.97 µM solution, and 1,000 µL of this solution is then transferred into a 4 mL
quartz cuvette. 2,000 µL of the 669.6 µM GlyGlyGly stock solution is further diluted into 30 mL
of deionized H2O to make a 44.64 µM and 1,000 µL of this solution is added into the above 4 mL
cuvette to give final concentrations of 21.49 µM and 22.32 µM of 3-N3MeCQ and GlyGlyGly
200 µL of the freshly made 42.97 µM 3-N3MeCQ solution is transferred into a 4 mL quartz cuvette.
To this is added 1,800 µL of the freshly made 44.64 µM GlyGlyGly solution to give final
concentrations of 4.29 µM and 40.18 µM of 3-N3MeCQ and GlyGlyGly, respectively. The kinetics
177
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
5,000 µL of the 213.4 µM 3-N3MeCQ stock solution is further diluted into 30 mL of MeOH to
make a 35.57 µM solution and 1,100 µL of this solution is then transferred into a 4 mL quartz
cuvette. 1,000 µL of the 1,251.2 µM N-Phenylmaleimide stock solution is further diluted into 30
mL of MeOH to make a 41.71 µM solution and 900 µL of this solution is added into the above 4
400 µL of the freshly made 35.57 µM 3-N3MeCQ solution is transferred into a 4 mL quartz cuvette.
To this is added 1,600 µL of the freshly made 41.71 µM N-Phenylmaleimide solution to give final
200 µL of the freshly made 35.57 µM 3-N3MeCQ solution is transferred into a 4 mL quartz cuvette.
To this is added 1,800 µL of the freshly made 41.71 µM N-Phenylmaleimide solution to give final
178
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
General procedure
In a 0.5-dram vial is added 3-N3MeCQ, the electron acceptor and 500 µL of deuterated MeOH.
This is thoroughly mixed to make a homogenous solution. This mixture is then transferred into an
8” NMR tube fitted with a stir bar. A 365 nm penlight is positioned 5 cm from the NMR tube. In
a dark fume hood with the setup covered in aluminum foil, the solution is let stir exposed to 365
nm for 6 to 9 minutes. Once the time elapses the stir bar is removed and a 1H NMR spectrum is
measured.
5 mg of 3-N3MeCQ and 2.3 mg of N-Phenylmaleimide were used and irradiated for 6 minutes.
N-Phenylmaleimide
5 mg of 3-N3MeCQ and 2.3 mg of Tetracyanoethylene were used and irradiated for 9 minutes.
179
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
4.6 References
(3) Sullivan, D. J. Quinolines Block Every Step of Malaria Heme Crystal Growth. Proc. Natl.
(4) Kapishnikov, S.; Staalsø, T.; Yang, Y.; Lee, J.; Pérez-Berná, A. J.; Pereiro, E.; Yang, Y.;
Werner, S.; Guttmann, P.; Leiserowitz, L.; Als-Nielsen, J. Mode of Action of Quinoline
(5) Bray, P. G.; Park, B.; Asadollaly, E.; Biagini, G.; Jeyadevan, J.; Berry, N.; Ward, S.; O’
(6) Foley, M.; Deady, L. W.; Ng, K.; Cowman, A. F.; Tilley, L. Photoaffinity Labeling of
6955–6961.
(7) Mueller, D. M.; Hudson, R. A.; Lee, C.-P. Azide Photoaffinity Analogs for Acridine Dye
(8) Desneves, J.; Thorn, G.; Berman, A.; Galatis, D.; La Greca, N.; Sinding, J.; Foley, M.;
180
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
(9) Lekostaj, J. K.; Natarajan, J. K.; Paguio, M. F.; Wolf, C.; Roepe, P. D. Photoaffinity
(10) Edaye, S.; Tazoo, D.; Bohle, D. S.; Georges, E. 3-Iodo-4-Aminoquinoline Derivative
(11) Edaye, S.; Tazoo, D.; Bohle, D. S.; Georges, E. 3-Halo Chloroquine Derivatives Overcome
(12) Ribas, X.; Güell, I. Cu(I)/Cu(III) Catalytic Cycle Involved in Ullmann-Type Cross-
(13) Andersen, J.; Bolvig, S.; Liang, X. Efficient One-Pot Synthesis of 1-Aryl 1,2,3-Triazoles
from Aryl Halides and Terminal Alkynes in the Presence of Sodium Azide. Synlett 2005,
Catalyzed Coupling of Sodium Azide with Aryl Iodides/Boronic Acids to Aryl Azides or
(15) Zhu, W.; Ma, D. Synthesis of Aryl Azides and Vinyl Azides via Proline-Promoted CuI-
(16) Markiewicz, J. T.; Wiest, O.; Helquist, P. Synthesis of Primary Aryl Amines Through a
Copper-Assisted Aromatic Substitution Reaction with Sodium Azide. J. Org. Chem. 2010,
75 (14), 4887–4890.
181
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
(18) Goriya, Y.; Ramana, C. V. The [Cu]-Catalyzed SNAR Reactions: Direct Amination of
Electron Deficient Aryl Halides with Sodium Azide and the Synthesis of Arylthioethers
(19) Qin, Z.; Kastrati, I.; Chandrasena, R. E. P.; Liu, H.; Yao, P.; Petukhov, P. A.; Bolton, J. L.;
Oxidative Activity and Receptor Affinity. J. Med. Chem. 2007, 50 (11), 2682–2692.
(20) Gehringer, M.; Forster, M.; Pfaffenrot, E.; Bauer, S. M.; Laufer, S. A. Novel Hinge-Binding
(21) Kaila, N.; Green, N.; Li, H.-Q.; Hu, Y.; Janz, K.; Gavrin, L. K.; Thomason, J.; Tam, S.;
Powell, D.; Cuozzo, J.; Hall, J. P.; Telliez, J.-B.; Hsu, S.; Nickerson-Nutter, C.; Wang, Q.;
6442.
(22) Rahman, M.; Kundu, D.; Hajra, A.; Majee, A. Formylation without Catalyst and Solvent at
(23) Kim, J.-G.; Jang, D. Facile and Highly Efficient N-Formylation of Amines Using a Catalytic
Amount of Iodine under Solvent-Free Conditions. Synlett 2010, 2010 (14), 2093–2096.
(24) Shastri, L. A.; Shastri, S. L.; Bathula, C. D.; Basanagouda, M.; Kulkarni, M. V. Mild,
Simple, and Efficient Method for N -Formylation of Secondary Amines via Reimer–
182
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
(25) Collet, J. W.; Ackermans, K.; Lambregts, J.; Maes, B. U. W.; Orru, R. V. A.; Ruijter, E.
(26) Pajkert, R.; Böttcher, T.; Ponomarenko, M.; Bremer, M.; Röschenthaler, G.-V. Synthesis
(27) Sánchez-Martín, R.; Campos, J. M.; Conejo-García, A.; Cruz-López, O.; Báñez-Coronel,
M.; Rodríguez-González, A.; Gallo, M. A.; Lacal, J. C.; Espinosa, A. Symmetrical Bis-
Activity against the HT-29 Cell Line. J. Med. Chem. 2005, 48 (9), 3354–3363.
(28) Rodrigues, T.; Guedes, R. C.; dos Santos, D. J. V. A.; Carrasco, M.; Gut, J.; Rosenthal, P.
J.; Moreira, R.; Lopes, F. Design, Synthesis and Structure–Activity Relationships of (1H-
(13), 3476–3480.
(29) Natarajan, J. K.; Alumasa, J. N.; Yearick, K.; Ekoue-Kovi, K. A.; Casabianca, L. B.; De
Dios, A. C.; Wolf, C.; Roepe, P. D. 4-N-, 4-S-, and 4-O-Chloroquine Analogues: Influence
of Side Chain Length and Quinolyl Nitrogen PKa on Activity vs Chloroquine Resistant
(31) Kraft, A. Branched Non-Covalent Complexes between Carboxylic Acids and Two
183
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
(32) Hyde, A. M.; Calabria, R.; Arvary, R.; Wang, X.; Klapars, A. Investigating the
Related Unsaturated Nitrogenous Bases. Org. Process Res. Dev. 2019, 23 (9), 1860–1871.
(33) Sather, A. C.; Lee, H. G.; De La Rosa, V. Y.; Yang, Y.; Müller, P.; Buchwald, S. L. A
Fluorination of Aryl Triflates and Bromides. J. Am. Chem. Soc. 2015, 137 (41), 13433–
13438.
(34) Sinha, M.; Dola, V. R.; Soni, A.; Agarwal, P.; Srivastava, K.; Haq, W.; Puri, S. K.; Katti,
(35) Johnson, W. S.; Buell, B. G. A New Synthesis of Chloroquine. J. Am. Chem. Soc. 1952, 74
(18), 4513–4516.
(36) Gorlitzer, K.; Kramer, C.; Meyer, H.; Walter, R. D.; Jomaa, H.; Wiesner, J. Pyrido[3,2-
b]Indol-4-Yl-Amine – Synthese Und Prufung Auf Wirksamkeit Gegen Malaria. Die Pharm.
(37) Liu, Y.; Zhang, Z.; Wu, A.; Yang, X.; Zhu, Y.; Zhao, N. A Novel Process for Antimalarial
Drug Pyronaridine Tetraphosphate. Org. Process Res. Dev. 2014, 18 (2), 349–353.
(38) Elslager, E. F.; Gold, E. H.; Tendick, F. H.; Werbel, L. M.; Worth, D. F. Amodiaquine N-
6–12.
(39) Wang, X.; Pan, W.; Cai, Y.; Xie, X.; Huang, C.; Li, J.; Chen, W.; He, M. Microwave-
184
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
(40) Bayley, H.; Staros, J. V. Photoaffinity Labeling and Related Techniques. In Azides and
(41) Vezmar, M.; Georges, E. Direct Binding of Chloroquine to the Multidrug Resistance Protein
(42) Baruah, H.; Puthenveetil, S.; Choi, Y.-A.; Shah, S.; Ting, A. Y. An Engineered Aryl Azide
(43) Berman, A.; Shearing, L. N.; Ng, K. F.; Jinsart, W.; Foley, M.; Tilley, L. Photoaffinity
(44) Wang, J.; Burdzinski, G.; Platz, M. S. Ultrafast Time-Resolved Studies of the
(45) Vyas, S.; Winter, A. H.; Hadad, C. M. Theory and Computation in the Study of Nitrenes
and Their Excited-State Photoprecursors. In Nitrenes and Nitrenium Ions; John Wiley &
(46) Bräse, S.; Banert, K. Organic Azides; Brse, S., Banert, K., Eds.; John Wiley & Sons, Ltd:
(47) Zott, H.; Heusinger, H. Photolysis of Maleic Anhydride and Maleimides in Solution Studied
(48) Kiselev, V. D.; Kashaeva, H. A.; Potapova, L. N.; Kornilov, D. A.; Latypova, L. I.;
2205.
185
Chapter 4 Synthesis and Reactivity of a Compact Chloroquine Photoaffinity label.
(49) Murata, S.; Abe, S.; Tomioka, H. Photochemical Reactions of Mesityl Azide with
186
Chapter 5
Transporter (PfCRT)
187
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
5.1 Preamble
In the previous chapter, the synthesis and utilization of our photoaffinity label was described. This
label is expected to cross-link to PfCRT. PfCRT contains several point mutations that have been
directly linked to CQ resistance. There is much debate about this protein’s function, whether it acts
between different classes of antimalarials, and the mechanism of substrate recognition is also
poorly understood.2 As our photoaffinity label contains minor modifications compared to CQ, this
possibly avoids unnecessary interactions with surrounding proteins. PfCRT has been isolated, and
its cryo-electron microscopy structure was determined for the first time.3 Therefore, before
and binding affinity of PfCRT with CQ and heme. In collaboration with the Fidock group who
provided PfCRT isoform, we carried out fluorescence and UV-Vis studies. These studies were
carried out in buffers of pH 7.0 – 8.0, which is within the range of cytosolic pH. The challenge
with carrying out this assay is that PfCRT is a transmembrane protein with one side exposed to pH
5.2 and the other at pH 7.4. Therefore we chose to model the cytosolic pH.
5.2 Introduction
CQ, first synthesized in 1934, revolutionized antimalarial therapy. It was viewed as the drug that
would eradicate malaria. Unfortunately, in the early 1960s, resistance to CQ was discovered.4 A
tremendous amount of effort has been made in determining the antimalarial mechanism of CQ and
the origin of its resistance.5–9 The structure of hemozoin which is a biomineralization byproduct
synthesized by the parasite to detoxify free heme in the DV was solved in 2000.10 A lysine to
188
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
threonine mutation at position 76 (K76T) on the PfCRT is a biomarker for CQ resistance that was
also discovered in 2000.6 PfCRT has been sequenced, and there has been much speculation on the
mechanism in which the protein recognizes antimalarial drugs. Moreover, for the first time, a
PfCRT isoform of CQ-resistant 7G8 parasites was isolated using single-particle cryo-electron
The isolated PfCRT isoform is a 424 amino acid protein with a molecular weight of 48 kDa. It is
a monomeric transmembrane protein that consists of ten transmembrane (TM) helices and two
juxtamembrane helices. One is exposed to the parasite cytosol and the other to the DV (Figure
5.1). The transmembrane domains form 5 helical pairs, and TM1-4 and 6-9 form a central cavity
of around 3,300 Å3. The K76T mutation is located within the lining of the cavity. The cavity has
a net negative charge influenced by three aspartate residues, suggesting positively charged
a) b)
Figure 5.1. Structure of PfCRT 7G8 isoform. A) Topology showing inverted antiparallel TM
helices. B) Surface representation of the electrostatic potential of the central cavity with red and
blue indicating negative and positive residues, respectively. Figures adapted from Kim et al.3
189
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
techniques used to determine interactions between CQ and heme with PfCRT are described.
Including two control substrates: empty nanodisc (1D1), which has the same lipid membrane used
for PfCRT filled with lipids and unrelated GtrB from an enterobacteria phage enclosed in a
different nanodisc (1E3D1). This ensemble will be referred to as membrane proteins and lipid
190
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
In collaboration with Prof. David Fidock’s lab, we were generously gifted PfCRT 7G8 isoform,
nanodisc and GtrB. It was first believed that the cavity with a volume of 3,300 Å3 was large enough
to transport out the CQ - heme complex that forms within the DV. CQ and hemin have approximate
volumes of 396 Å3 and 702 Å3, respectively (as estimated from crystallographic results). We began
with binding studies of the individual drugs with the ensembles to determine possible recognition
of CQ or heme with the transporter protein. To determine the interactions between CQ, heme and
proposed that CQ is transported out of the DV by PfCRT, which will entail recognition, binding
and transport.2,6 The observed PfCRT fluorescence primarily originates from tryptophan (Trp) 280
and 316, with the empty nanodisc contains 2 Trp residues and GtrB 3 Trp residues. As Trp
emission is highly sensitive to its microenvironment, changes in fluorescence are used to determine
protein-drug interactions.12 PfCRT’s two Trp residues are located in the juxtamembrane, two and
TM eight, exposed to the DV, Figure 5.1. Fluorescence quenching and shifts in the emission
191
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
Fluorescence Intensity
Fluorescence Intensity
2eq 2eq
1.5×107
3eq 3eq
6×106 4eq
4eq
1.0×107
4×106
2×106 5.0×106
0 0.0
350 400 450 500 350 400 450
Wavelength (nm) Wavelength (nm)
3.16uM GtrB 5.92uM Nanodisc
4×107 1×107
GtrB Nanodisc
1eq 1eq
Fluorescence Intensity
Fluorescence Intensity
2eq 2eq
3×107
3eq 3eq
4eq 4eq
2×107 5×106
1×107
0 0
350 400 450 350 400 450 500
Wavelength (nm) Wavelength (nm)
solution as CQH2+). As shown in Figure 5.2, increasing amounts of CQ, up to 4 equivalents, causes
a slight decrease in emission energy, and this decrease is more pronounced in empty nanodisc.
Furthermore, we observe the following shifts in the fluorescence maxima: PfCRT pH 8 10 nm red
shift, GtrB 5 nm red shift and nanodisc 5 nm red shift. The fluorescence quenching constant was
192
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
H)
= 1 + I* J) [L] = 1 + B+, [L]
H
where F0 and F are the fluorescence intensities of the protein in the absence and presence of the
lifetime, KSV is the Stern-Volmer quenching constant and [Q] is the concentration of the quencher.
Table 5.1 shows the KSV values of our membrane proteins and lipid membrane ensemble
(MPLME) and CQ. The higher the quenching constant, the stronger the association. The data
shows that nanodisc has the highest quenching constant. In general, a linear plot suggests the
occurrence of either static or dynamic quenching, Figure 5.3. Static quenching involves the
formation of nonfluorescent complexes between the fluorophore and the quencher that occur in
the ground state. Dynamic quenching, which occurs in the excited state, involves the diffusion of
the fluorophore during the lifetime of the excited state. The fluorophore then returns to the ground
state upon contact without emission of a photon. Both types of quenching mechanisms occur only
through space contact between the fluorophore and quencher and do not result in a permanent
193
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
1.15
1.2
F0/F (345nm)
F0/F (335nm)
1.10
1.1
1.05
1.0
1.00
0.95 0.9
0.0 5.0×10-6 1.0×10-5 1.5×10-5 2.0×10-5 0.0 5.0×10-6 1.0×10-5 1.5×10-5
[CQ] [CQ]
3.16uM GtrB 5.92uM Nanodisc
1.3 2.0
1.8
1.2
F0/F (340nm)
F0/F (340nm)
1.6
1.1
1.4
1.0
1.2
0.9 1.0
0 2×10-6 4×10-6 6×10-6 8×10-6 1×10-5 0.0 5.0×10-6 1.0×10-5 1.5×10-5 2.0×10-5
[CQ] [CQ]
Figure 5.3. Stern-Volmer plots for PfCRT, GtrB and nanodisc quenched with up to 4 equivalents
A factor affecting the specific quenching mechanism is the accessibility of the quencher to the
fluorophore. The fraction of accessible fluorophores can be measured using the modified Stern-
H) 1 1
= +
H) − H NB& [L] N
Where F0 and F are the fluorescence intensities in the presence and absence of the quencher, α is
the fraction of accessible fluorophores, Ka is the effective quenching constant and [Q] is the
194
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
15
40
F0/F0-F
F0/F0-F
10
20
5
0 0
0 2×105 4×105 6×105 8×105 1×106 0 500000 1×106 2×106
1/[CQ] 1/[CQ]
3.16uM GtrB 5.92uM Nanodisc
40 8
30 6
F0/F0-F
F0/F0-F
20 4
10 2
0 0
0.0 5.0×105 1.0×106 1.5×106 2.0×106 0 2×105 4×105 6×105 8×105 1×106
1/[CQ] 1/[CQ]
Figure 5.4. Modified Stern-Volmer plots for PfCRT, GtrB and nanodisc quenched with up to 4
The modified Stern-Volmer plots, shown in Figure 5.4, give an N value of 35% or less. In general,
N values less than 100% are a result of fractional accessibility, which is due to the existence of at
least two fluorophore populations with one that is accessible and the other is buried or
inaccessible.13 Each of these fluorophores would have varying KSV values and are differently
accessible to the quencher. The Ka value, in Table 5.2, is the effective quenching constant of
accessible fluorophores, this is in accordance with the KSV values in Table 5.1. CQ in nanodisc is
exposed to the highest fraction of accessible fluorophores as it has the highest KSV value. CQ has
195
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
Interestingly, there appears to be a concentration dependant response from the Stern-Volmer plot
for PfCRT pH 7 and CQ, Figure 5.5. . The data points of 0 - 1 equiv. give KSV of 1.69 × 10-4 M-
1
and 1 - 4 equiv. of 0.19 × 10-4 M-1. This may be due to conformational change following binding
Similarly, for PfCRT pH 8 and CQ, data points of 0 – 2 equiv. give KSV of 1.5 × 10-4 M-1; however,
196
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
1.15
1.10
F0/F (345nm)
F0/F (345nm)
1.10
1.05
1.05
1.00
1.00
0.95 0.95
0 2×10-6 4×10-6 6×10-6 0.0 5.0×10-6 1.0×10-5 1.5×10-5 2.0×10-5
[CQ] [CQ]
1.2 1.2
F0/F (335nm)
F0/F (335nm)
1.1 1.1
1.0 1.0
0.9 0.9
0 2×10-6 4×10-6 6×10-6 8×10-6 1×10-5 6.0×10-6 8.0×10-6 1.0×10-5 1.2×10-5 1.4×10-5
[CQ] [CQ]
To determine the number of binding sites for CQ present on the MPLME, we use the following
equation (3)14:
H) − H
log = log B- + ) log [L]
H
where F0 and F are fluorescent intensities in the presence and absence of a quencher, Kb is the
binding constant, n is the number of binding sites and [Q] is the quencher concentration.
197
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
Table 5.3. Binding constant and number of binding sites for CQ with MPLME.
PROTEIN logKb n
PfCRT pH 7 0.06 ± 0.83 0.27 ± 0.16
PfCRT pH 8 0.01 ± 0.87 0.18 ± 0.17
GtrB 1.89 ± 0.58 0.54 ± 0.11
Nanodisc 3.05 ± 0.37 0.67 ± 0.07
Stronger binding interactions give higher logKb values, and the values in Table 5.3. are in
accordance with previous KSV and Ka values. Nanodisc displays the strongest binding interactions
with CQ in the ensemble, although with an n value of 0.67, we cannot conclusively say that
nanodisc contains a binding site for CQ, Figure 5.6. PfCRT at both pH’s form weak associations
with CQ and does not have a specific CQ binding site. The control, GtrB, forms stronger
associations with CQ than PfCRT; however, it does not contain a binding site for CQ.
198
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
-1.0
-0.5
Log(F0-F)/F
Log(F0-F)/F
-1.2
-1.4 -1.0
-1.6
-1.5
-1.8
-2.0 -2.0
-6.5 -6.0 -5.5 -5.0 -4.5 -6.5 -6.0 -5.5 -5.0 -4.5
Log[CQ] Log[CQ]
3.16uM GtrB 5.92uM Nanodisc
0.0 0.0
-0.5 -0.2
Log(F0-F)/F
-1.5 -0.6
-2.0 -0.8
-7.0 -6.5 -6.0 -5.5 -5.0 -4.5 -6.0 -5.5 -5.0 -4.5
Log[CQ] Log[CQ]
Figure 5.6. Binding plot to determine the number of CQ binding sites and binding constant.
monoprotonated and 17% diprotonated. Thus, the results showing a lower binding affinity for CQ
5.215, this is the optimal condition for PfCRT activity, therefore it is not surprising that we do not
this supports the belief that PfCRT is responsible for CQ efflux from the DV, and not its influx as
199
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
Heme can form reversible non-covalent binding interactions with proteins and these interactions
cause spectral changes that can be measured by UV-Vis and Fluorescence spectroscopy.13 High-
spin Fe3+ porphyrins are fluorescence quenchers due to their low-lying excited states and
paramagnetism allowing for multiple relaxation pathways.17 Therefore, any observed fluorescence
quenching between heme and proteins results from non-covalent complex formation.
5.0×106
0 0.0
350 400 450 350 400 450
Wavelength (nm) Wavelength (nm)
Fluorescence Intensity
2eq 2eq
3×107 3eq 3eq
4eq 4eq
2×107 5×106
1×107
0 0
350 400 450 350 400 450
Wavelength (nm) Wavelength (nm)
Figure 5.7. Effect of heme on fluorescence spectrum of PfCRT at pH 7 and 8, GtrB and nanodisc.
As shown in Figure 5.7 in PfCRT and nanodisc, increasing amounts of hemin cause a decrease in
fluorescence intensity. However, in the case of GtrB a decrease in fluorescence is only observed
200
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
when at least 4 equiv. of hemin is added. To determine the fluorescence quenching constant for
The KSV values, in Table 5.4, show a strong quenching constant of heme in PfCRT at pH 8 and to
a lesser extent in nanodisc as can be seen in Figure 5.8. we get a negative slope when heme is
added to GtrB, this is attributed to the inaccessibility of heme to GtrB fluorophores. In this
experiment, we can see the effect of pH and protein conformation with heme association. There is
an order of magnitude difference in the Stern-Volmer constant with the pH is lowered by one unit.
201
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
F0/F (340nm)
F0/F (340nm)
2.0
1.5
1
1.0 0
0.0 5.0×10-6 1.0×10-5 1.5×10-5 2.0×10-5 0.0 5.0×10-6 1.0×10-5 1.5×10-5
[Heme] [Heme]
1.1
F0/F (340nm)
F0/F (345nm)
2
1.0
1
0.9
0.8 0
0 2×10-6 4×10-6 6×10-6 8×10-6 0.0 5.0×10-6 1.0×10-5 1.5×10-5 2.0×10-5
[Heme] [Heme]
Figure 5.8. Stern-Volmer plot for PfCRT, GtrB and nanodisc quenched with up to 4 equivalents
To determine the fraction of accessible fluorophores, we used the modified Stern-Volmer equation
(2), the results are displayed in Table 5.5 and Figure 5.9. The fraction of accessible fluorophores
is greater for PfCRT at pH 8 than pH 7. Whereas in the case of GtrB there are no accessible
fluorophores for heme to interact with. This is in agreement with the null KSV value in Table 5.4.
202
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
Lastly, to determine whether the proteins contain binding sites for heme, we used equation (3), the
plots are shown in Figure 5.10 and values are shown in Figure 5.6.
6
4
F0/F0-F
F0/F0-F
2
2
0 0
0 2×105 4×105 6×105 8×105 1×106 0.0 5.0×105 1.0×106 1.5×106
1/[Heme] 1/[Heme]
50 3
F0/F0-F
F0/F0-F
0 2
-50 1
-100 0
0.0 5.0×105 1.0×106 1.5×106 2.0×106 0 2×105 4×105 6×105 8×105 1×106
1/[Heme] 1/[Heme]
Figure 5.9. Modified Stern-Volmer plots for PfCRT, GtrB and nanodisc quenched with up to 4
GtrB does not contain any binding sites for heme, and we get a negative slope and intercept values
for the Stern-Volmer quenching constants; thus, we can say that heme is not a substrate for GtrB.
203
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
PfCRT at pH 8 contains one binding site for heme, this is in accordance with the 90% accessibility
Table 5.6. Binding constants and number of binding sites for heme with MPLME.
PROTEIN logKb n
PfCRT pH 7 2.08 ± 0.39 0.43 ± 0.08
PfCRT pH 8 5.34 ± 0.25 1.02 ± 0.05
GtrB - -
Nanodisc 2.81 ± 0.26 0.54 ± 0.05
The difference of one pH unit has a 1,000 - fold impact as at pH 7 PfCRT does not contain any
binding sites for heme. Consequently, heme has access to a lesser percentage of the fluorophore
resulting in a low quenching constant. Finally, nanodisc does not contain any binding sites for
heme, which is also in accordance with previous results. The same nanodisc is incorporated onto
PfCRT to keep it membrane-bound; thus, these results indicate that at pH 8 heme is truly binding
to the protein not the membrane and that there is a significant conformational change to the protein
204
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
0.0
Log(F0-F)/F
Log(F0-F)/F
0.0
-0.2
-0.4
-0.5
-0.6
-0.8 -1.0
-6.5 -6.0 -5.5 -5.0 -4.5 -6.5 -6.0 -5.5 -5.0 -4.5
Log[Heme] Log[Heme]
0.2
-1.2
Log(F0-F)/F
Log(F0-F)/F
0.0
-1.4
-0.2
-1.6
-0.4
-1.8 -0.6
-7.0 -6.5 -6.0 -5.5 -5.0 -6.5 -6.0 -5.5 -5.0 -4.5
Log[Heme] Log[Heme]
Figure 5.10. Binding plot to determine the number of heme binding sites and binding constant.
As mentioned above, it is important to remember that heme is possibly removed out of the DV by
PfCRT at pH 5.2. Secondly, the two PfCRT Trp residues are located on the DV portion of the
membrane; thus, at physiological pH results may differ. It may be the case that PfCRT indeed
contains at least one binding site for CQ and heme at physiological pH. Nonetheless, we were able
The PfCRT central cavity has a volume of approximately 3,300 Å3, heme and CQ molecules have
approximate volumes of 702 Å3 and 396 Å3, respectively. Furthermore, the cavity has a max
diameter of 25 Å, with the max diameters for heme and CQ being approximately 14 Å and 8 Å,
respectively. Hence, it is possible that the resistance protein is capable of effluxing both heme and
205
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
The heme Soret, Q-bands and charge transfer bands can be identified by UV-Vis spectroscopy and
are used to determine changes to heme oxidation state and spin state. We can use the observed
changes of these bands to determine whether heme is involved in any binding interactions,
All heme solutions were made in a 40% DMSO/ aqueous mixture, in this solution, heme is in its
monomeric form.18 As shown in Figure 5.11, for GtrB, there is no observed shift of the Soret band
when two molar equivalents of heme are added. Heme added to the empty nanodisc causes a small
2 nm hypochromic shift (blue shift) from 412 nm to 410 nm and a greater 6 nm blue shift for the
Q band. When heme is added to PfCRT, we observe the greatest change, an 8 nm blue shift, from
403 nm to 395 nm with no change to the Q band. Changes to the Soret peak which are associated
with " → "* transition suggesting the occurrence of heme binding interactions.19
206
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
2.0
2.82µM PfCRT pH 8 2.16µM GtrB
0.6
PfCRT 0.4eq
0.4eq 0.8eq
1.5 0.8eq 1.2eq
0.4
Absorbance
Absorbance
1.2eq 1.6eq
1.6eq 2eq
1.0 2eq
0.2 !max 413→413
!max 403→395
0.5
0.0
0.0
300 400 500 600 300 400 500 600
Wavelength (nm) Wavelength (nm)
6µM Nanodisc
0.8
Nanodisc
0.4eq
0.6 0.8eq
Absorbance
1.2eq
1.6eq
0.4 2eq
!max 412→410
0.2
0.0
300 400 500 600
Wavelength (nm)
The control protein GtrB does not bind with heme as there is no measured shift in the Soret peak.
This was confirmed with fluorescence assays which showed that GtrB has no binding sites for
heme (Table 5.6). In the case of lipid nanodisc, we observe a small 2 nm shift when two molar
equivalents of heme are added, which we attribute to weak associations of heme with the
membrane. Fluorescence studies showed that heme has a logKb value of 2.81, which is stronger
than with GtrB but less than with PfCRT at pH 8. When heme is added to PfCRT at pH 8 we see
the greatest shift of 8 nm, indicating greater binding interactions between PfCRT - Heme than with
nanodisc and GtrB. Furthermore, these interactions are independent of the nanodisc, which gave a
207
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
2 nm shift. These results are in accordance with fluorescent studies, which show that PfCRT at pH
8 contains one binding site for heme; furthermore, heme has access to 90% of its Trp fluorophores.
5.4 Conclusion
CQ resistance was first discovered in the 1960s, and there has been sustained research into
determining the mechanism(s) of resistance and how to reverse and/or prevent it. A central protein
involved in CQ resistance has for the first time been isolated, this breakthrough will allow for a
better understanding of the resistance mechanism. In collaboration with the Fidock group, we
worked with samples of the PfCRT 7G8 isoform and were able to carry out fluorescence and UV-
Vis studies to determine binding of CQ and heme to the protein (an empty nanodisc membrane in
which PfCRT is embedded in and an unrelated control protein GtrB) at cytosolic pH.
Fluorescence studies with GtrB show that it does not bind CQ or heme. We also determined that
CQ and heme have little to no access to fluorophores located on the protein. Empty nanodisc does
not bind CQ or heme, indicating that these drugs do not get embedded within these membranes.
Additionally, heme has access to a greater fraction of fluorophores than CQ, which highlights the
different interactions between the drugs and the protein. Lastly, PfCRT interactions are pH-
dependent. At pH 7 PfCRT does not bind CQ or heme, this is in accordance with previous reports
that state PfCRT effluxes CQ from the DV into the cytosol, it does not transport CQ into the DV.
We also carried out UV-Vis experiments of heme with GtrB, nanodisc and PfCRT at pH 8. We
used the change in the Soret peak as an indication of non-covalent binding interactions. There were
208
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
no observed changes in the Soret peak with GtrB and a 2 nm blue shift with nanodisc. We observed
an 8 nm blue shift of the Soret peak in PfCRT. This 8 nm shift indicates significant interactions
The results of our fluorescence and UV-Vis studies demonstrate that PfCRT at pH 8, contains one
binding site for heme and at lower pH does not bind heme. At pH 7 and 8, PfCRT does not form
significant binding interactions with CQ. This supports the belief that PfCRT transports CQ from
the DV into the cytosol. Future work would involve performing fluorescence assays at vacuolar
209
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
5.5 Experimental
All solvents and reagents were obtained commercially and used without further purification unless
noted. The UV-Vis spectra were measured on Agilent 8453. The fluorescence measurements are
performed on a Molecular Devices SpectraMax i3x, equipped with a black bottom 96 well plate
The PfCRT 7G8 isoform, nanodisc (MSP1D1 + POPG) and GtrB (encapsulated in MSP1E3D1)
gifted from Prof. David Fidock’s group at Columbia University. PfCRT at pH 7, nanodisc and
GtrB were provided in a buffer solution containing 20 mM HEPES and 150 mM NaCl. PfCRT at
pH 8 was provided in a buffer solution containing 20 mM TRIS pH 8 and 150 mM NaCl. CQ and
All solutions are freshly prepared and deionized H2O is used in the preparation of all buffer
solutions. All data analysis is performed using Prism 7.0a (GraphPad Software). Cole Parmer
combination electrode, glass body, which is calibrated with standard buffer solutions before use.
The emission spectra are then measured at 25.0oC and are recorded in a wavelength range of 305-
500 nm for nanodisc and GtrB following excitation at 280 nm. The emission spectra for PfCRT
210
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
are recorded with a wavelength range of 315-500 nm following excitation at 290 nm. The
excitation and emission bandwidths are 5 nm. The initial volume of all proteins in the 96 well
PfCRT pH 7 and nanodisc experiments: 39 μL of hemin stock solution is diluted into 1 mL 40%
DMSO, 60% buffer mixture to make a 39 μM 40% hemin solution. 39 μL of CQ stock is then
solution.
PfCRT pH 8 experiments: 66.2 μL of hemin stock solution is diluted into 2 mL 40% DMSO, 60%
buffer mixture to make a 33.1 μM 40% hemin solution. 82.7 μL of CQ stock is then diluted into 5
mL of buffer to make a 33.1 μM solution. PfCRT is used undiluted as a 4.98 μM TRIS solution.
GtrB experiments: 21.1 μL of hemin stock solution is diluted into 1 mL 40% DMSO, 60% buffer
mixture to make a 21.1 μM 40% hemin solution. 52.8 μL of CQ stock is then diluted into 5 mL of
buffer to make a 21.1 μM solution. GtrB is used undiluted as a 3.16 μM HEPES solution.
Once prepared the solutions are kept on ice in a polystyrene cooler box. CQ or hemin is then
titrated in 3 μL increments which correspond to 0.2 molar equivalents addition. Once four equiv.
of the drug are added subsequent additions are in 15 μL increments until 7 molar equivalents are
added. The equilibrium time between each addition is 10 minutes. During titration the solution is
stirred and kept at a constant temperature of 25oC. The total volume of drug added is 150 μL.
211
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
At RT, the spectrophotometer is equipped with a 1 cm quartz cuvette of 4 mL capacity. All spectra
are recorded against the appropriate blank buffer solution in the range of 260-800 nm.
a 6 μM solution. 400 μL of hemin stock solution is diluted into 1 mL 40% DMSO, 60% buffer
PfCRT: 100 μL of 14.1 μM PfCRT is diluted to 500 μL TRIS pH 8 buffer to make a 2.82 μM
solution. 188 μL of hemin stock solution is diluted into 1 mL 40% DMSO, 60% buffer mixture to
GtrB: 340 μL of 3.18 μM GtrB is diluted into 500 μL HEPES pH 7.5 buffer to make a 2.16 μM
solution. 144 μL of hemin stock solution is diluted into 1 mL 40% DMSO, 60% buffer mixture to
Heme is titrated in 3 μL increments which correspond to 0.4 molar equivalents addition. Heme is
added until 2 molar equiv. are reached, with an equilibrium time of 10 minutes between additions.
212
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
5.6 References
(1) Sanchez, C. P.; Stein, W. D.; Lanzer, M. Is PfCRT a Channel or a Carrier? Two Competing
(2) Johnson, D. J.; Fidock, D. A.; Mungthin, M.; Lakshmanan, V.; Sidhu, A. B. S.; Bray, P. G.;
Ward, S. A. Evidence for a Central Role for PfCRT in Conferring Plasmodium Falciparum
(3) Kim, J.; Tan, Y. Z.; Wicht, K. J.; Erramilli, S. K.; Dhingra, S. K.; Okombo, J.; Vendome,
J.; Hagenah, L. M.; Giacometti, S. I.; Warren, A. L.; Nosol, K.; Roepe, P. D.; Potter, C. S.;
Carragher, B.; Kossiakoff, A. A.; Quick, M.; Fidock, D. A.; Mancia, F. Structure and Drug
Resistance of the Plasmodium Falciparum Transporter PfCRT. Nature 2019, 576 (7786),
315–320.
(4) Ecker, A.; Lehane, A. M.; Clain, J.; Fidock, D. A. PfCRT and Its Role in Antimalarial Drug
(5) Chinappi, M.; Via, A.; Marcatili, P.; Tramontano, A. On the Mechanism of Chloroquine
(6) David A. Fidock; Nomura, T.; Talley, A. K.; Cooper, R. A.; Dzekunov, S. M.; Ferdig, M.
T.; Ursos, L. M. B.; Sidhu, A. bir S.; Naudé, B.; Deitsch, K. W.; Su, X.; Wootton, J. C.;
Transmembrane Protein PfCRT and Evidence for Their Role in Chloroquine Resistance.
(7) Djimdé, A.; Doumbo, O. K.; Cortese, J. F.; Kayentao, K.; Doumbo, S.; Diourté, Y.;
Coulibaly, D.; Dicko, A.; Su, X.; Nomura, T.; Fidock, D. A.; Wellems, T. E.; Plowe, C. V.
213
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
(8) Price, R. N.; Cassar, C.; Brockman, A.; Duraisingh, M.; Van Vugt, M.; White, N. J.; Nosten,
(9) Duraisingh, M. T.; Cowman, A. F. Contribution of the Pfmdr1 Gene to Antimalarial Drug-
(10) Pagola, S.; Stephens, P. W.; Bohle, D. S.; Kosar, A. D.; Madsen, S. K. The Structure of
(12) Marković, O. S.; Cvijetić, I. N.; Zlatović, M. V.; Opsenica, I. M.; Konstantinović, J. M.;
Terzić Jovanović, N. V.; Šolaja, B. A.; Verbić, T. Human Serum Albumin Binding of
Certain Antimalarials. Spectrochim. Acta - Part A Mol. Biomol. Spectrosc. 2018, 192, 128–
139.
(13) Makarska-Bialokoz, M. Interactions of Hemin with Bovine Serum Albumin and Human
(14) Tunç, S.; Duman, O.; Bozoǧlan, B. K. Studies on the Interactions of Chloroquine
Diphosphate and Phenelzine Sulfate Drugs with Human Serum Albumin and Human
214
Chapter 5 The Determination of Heme and Chloroquine interactions with PfCRT
1013.
(17) Dodd, E. L.; Bohle, D. S. Orienting the Heterocyclic Periphery: A Structural Model for
(18) Egan, T. J.; Mavuso, W. W.; Ross, D. C.; Marques, H. M. Thermodynamic Factors
(19) Dayer, M.; Moosavi-Movahedi, A.; Dayer, M. Band Assignment in Hemoglobin Porphyrin
Ring Spectrum: Using Four-Orbital Model of Gouterman. Protein Pept. Lett. 2010, 17 (4),
473–479.
215
Chapter 6
216
Chapter 6 Conclusions and Future Work
6.1 Conclusion
When I started my Ph.D., the main objective was to synthesize a chloroquine photoaffinity label
with the simple addition of an azide at the 3-position. In pursuit of the azide, the discovery of
undesired products led to the creation of projects that make the body of my thesis. This is a
testament to not dismissing side/byproduct formation. As chemists, we must truly get in the flask
to understand why and what reactions are occurring to the best of our ability. In that sense, is there
In my first chapter, I described the optimization of 3-NH2CQ synthesis. This project was born out
of the formation of the 3-NH2CQ rather than 3-N3CQ. To optimize amine formation, large amounts
of the precursor 3-BrCQ were required. However, the known synthetic route for 3-BrCQ at the
time did not allow large-scale synthesis. Therefore, 3-BrCQ synthesis was optimized using
microwave-assisted synthesis and excess amounts of a brominating agent. We were able to scale
the reaction to gram scale and obtain a 64% yield. With large amounts of my precursor, we were
able to carry out a series of experiments with varying copper catalysts, ligands, solvents, reducing
agents, and nitrogen sources. A moderate yield of 55% with the optimized reaction conditions was
reached.
The formation of 3-NH2CQ opened up the possibility of derivatization, which paved the way for
chapter 2. A quick search of the literature for 4-aminoquinolines reveals compounds that only
contain a 7-chloro substituent on the quinoline ring. Fortunately, previous work done in our lab
showed that 3-ICQ, when used in combination with CQ, resensitizes the parasite to CQ. Therefore,
217
Chapter 6 Conclusions and Future Work
derivatives. These derivatives would then be tested for any synergistic effects they may have in
combination therapy. Derivatives were synthesized with varying electronic and hydrophobic
effects, with guidance from the Craig Plot and consideration of Lipinski’s Rule of 5. Various
benzamides were synthesized, and we determined that para - chloro substituent showed the best
antiplasmodial activity. In CQS 3D7 strains, it was 3 – fold more effective, and in CQR Dd2 strains
it was 1.8 – fold less effective than CQ. The equivalent para – chloro sulfonamide and Schiff base
The precursor for the desired photoaffinity label was 3-NH2CQ. However, as explained in chapter
4, we were unable to synthesize the photoaffinity label with only one modification at the 3-
position. Doing so leads to the creation of a triazole product. To prevent cyclization, we blocked
the reactivity of the 4-amine by installing a methyl to form a tertiary amine. A methyl was chosen
due to its small size and lack of reactivity. This minor modification then allowed the synthesis of
the desired photoaffinity label. Preliminary experiments to test the reactivity of the aryl azide were
carried out.
The final chapter is a result of a collaboration that arose after an oral conference presentation. Prof.
Fidock was interested in the potential of the photoaffinity label and disclosed that they had, for the
first time, resolved the cryo-EM structure of Plasmodium falciparum Chloroquine Resistant
Transporter (PfCRT) isoform. We obtained samples of the isoform and were able to carry out
binding studies. We determined that there was no evidence of CQ binding to PfCRT at pH 7 and
218
Chapter 6 Conclusions and Future Work
Regarding the 3-substituted CQ library, future steps involve isobolographic analysis to determine
whether interactions between the derivatives and CQ are synergistic, additive, or antagonistic.
Results from such experiments can give more detailed S.A.R. data about the derivatives and their
targets. The 3-NH2CQ can be further derivatized to expand the library and synthesize different
families of antimalarial compounds. As the synthetic route for 3-N3CQ has been developed. It can
be used as a precursor to a family of triazole derivatives for click chemistry, thus further expanding
The photoaffinity label has been synthesized and can be irradiated with UV light of 365 nm. The
synthetic route developed can be used for various 4-aminoquinolines, thus allowing adequate
labelling of many antimalarial drug candidates. More studies need to be carried out to determine
the best conditions required for C-H, C-N, and C-C insertion and the optimal stoichiometry that
will produce the highest conversion and yield. Future work involves using the label with proteins
of interest to determine the insertion of the PAL. Such studies will have a significant impact on
our understanding of CQ’s biological pathway. Furthermore, once successfully incorporated into
the target protein, one can carry out protein digestion for mass spectroscopy. This will allow for
We have demonstrated that PfCRT binds heme at pH 8 and does not bind CQ at pH 7 and 8. The
next area of interest is determining the binding mechanism (static or dynamic) by carrying out
between the quencher and the fluorophore in the ground state. Dynamic quenching is a result of
219
Chapter 6 Conclusions and Future Work
interactions between the quencher and fluorophore in the excited state. Quenching emission is
the mechanism of interaction between the quencher and fluorophore. Lastly, it would be of great
interest to determine the binding behaviour of PfCRT towards CQ and heme at lower pH such as
pH 5.2.
220
Appendix
221
Appendix
Chapter 3
pH:
CQ titration 5.08
CQ pKa
7.58
4.87 7.71
1.5
5.55 7.92
5.84 8.11 0.8
Absorbance
1.0
6.03 8.49
6.1 8.65
A342
0.5 6.24 8.88 0.6
6.57 9.06
0.0 6.7 9.21
250 300 350 400 6.96 9.54
Wavelength (nm)
7.13
0.4
9.78
7.34 10 6 8 10
pH
4.27 6.43
1.0
A342
4.47 6.57
4.81 6.77 0.4
0.5
5 6.94
5.11 7.12
0.0 5.29 7.39
250 300 350 400
Wavelength (nm) 5.45 7.88 0.2
3 4 5 6 7 8
pH
222
Appendix
pH:
3.42 5.69
3-BrCQ pKa
3-BrCQ titration
3.57 5.85
1.5 3.77 6
4.13 6.12
0.6
Absorbance
A342
4.62 6.48 0.4
0.5
4.81 6.61
4.98 6.72
0.0
250 300 350 400
5.17 6.89 0.2
Wavelength (nm) 5.26 7.08
5.41 7.58
4 6 8
5.53 8.14
pH
pH:
3-ICQ pKa
3.39 5.8
3-ICQ titration
3.53 5.94
3.73 6.09 0.5
1.5
3.97 6.23
Absorbance
4.42 6.34
1.0 0.4
4.77 6.44 A342
4.83 6.59
0.5
4.95 6.69
0.3
5.02 6.92
0.0 5.21 7.14
250 300 350 400
5.43 7.52 0.2
Wavelength (nm)
5.61 8.04 4 6 8
pH
223
Appendix
pH:
3.58 5.89
3-NH2CQ pKa
3-NH2CQ titration
3.61 6.04 0.40
1.5 3.7 6.14
4
3.9 6.3 0.35
Absorbance
A342
4.42 6.92
0.5
4.72 7.05 0.25
4.81 7.36
0.0 0.20
250 300 350 400 450 4.94 7.65
Wavelength (nm) 5.19 7.91
0.15
5.37 8.24 4 6 8
5.57 8.47
pH
5.7
224
Appendix
Chapter 5
!!
= 1 + &" '! [)] = 1 + +#$ [)]
!
!! 1 1
= +
!! − ! -+% [)] -
225
Appendix
!! − !
log = log +& + 2 log [)]
!
!!
= 1 + &" '! [)] = 1 + +#$ [)]
!
226
Appendix
!! 1 1
= +
!! − ! -+% [)] -
!! − !
log = log +& + 2 log [)]
!
227
Appendix
NMR Spectra
228
9.5
1H
Et2N
9.0
Appendix
Cl
Br
1.00 8.68
8.5
1.09 8.29
2.3
1.00 8.08
8.0
NH
Br
7.5
NMR (400 MHz, Chloroform-d)
7.0
6.5
6.0
4.58
4.56
5.5
4.04
4.02
4.01
5.0
4.00
4.00
1.07 3.99
f1 (ppm)
4.5
3.98
3.96
EtOAc
1.23 2.60
4.0
2.58
2.56
2.54
3.5
2.49
2.48
2.46
3.0
1.68
4.53 1.68
2.5 1.67
2.23
1.66
1.65
EtOAc
1.65
2.0
1.63
1.63
4.69
1.5
1.62
3.53 1.62
6.64 1.61
EtOAc
1.0
1.60
1.30
1.29
0.5
1.06
1.04
1.02
0.0
229
Appendix
153.01
148.61
148.14
135.46
130.84
128.19
121.88
119.74
107.54
55.05
52.55
46.92
36.67
23.40
22.39
11.26
13
C NMR (201 MHz, Chloroform-d)
210 200 190 180 170 160 150 140 130 120 110 100 90 80 70 60 50 40 30 20 10 0 -10
f1 (ppm)
230
8.56
8.55
Et2N
8.08
9.0
1H
Cl
Appendix
8.08
8.07
8.07
8.5
8.07
8.07
8.0
7.94
NH
7.94
3.3
N
H
7.76
7.5
7.76
O
7.75
7.75
7.0
7.70
7.70
7.69
NMR (800 MHz, Methanol-d4)
6.5
7.69
7.69
7.68
6.0
7.62
7.62
7.61
5.5
7.60
7.60
4.57
5.0
4.57
3.10
3.09
f1 (ppm)
4.5
3.08
3.07
3.05
4.0
3.05
3.04
3.04
3.5
2.99
2.98
2.98
3.0
2.97
1.83
2.5 1.82
1.82
1.81
2.0
1.66
1.66
1.66
1.5
1.65
1.65
1.64
1.0
1.43
1.43
1.21
0.5
1.21
1.20
1.19
0.0
231
13
210
Appendix
200
190
180
169.09
170
160
153.47
C NMR (201 MHz, Methanol-d4)
151.04
145.62
150
139.46
138.61
140
132.71
132.69
130
128.77
127.55
125.29
120
119.60
117.39
110
111.52
100
f1 (ppm)
80
70
60 90
51.57
50
51.02
40
33.27
30
20.79
20
20.20
7.70
10
7.64
0
-10
232
9.5
Et2N
0.32 8.92
9.0
1H
Cl
Appendix
1.00 8.85
8.20
8.5
8.19
2.03 8.00
7.99
N
1.59
NH
8.0
7.99
2.35
7.98
N
H
3.4
2.25
7.89
O
7.5
1.57
7.88
0.77 7.88
7.87
7.0
7.86
7.81
CN
7.80
6.5
NMR (500 MHz, Chloroform-d)
7.43
7.43
7.42
6.0
7.42
7.41
5.5
7.40
7.40
7.32
5.0
7.30
4.5
4.31
f1 (ppm)
1.56 4.27
4.24
4.0
1.66 3.72
3.5
2.54
2.53
2.52
3.0
2.50
4.49 2.48
3.81 2.48
2.5
2.18 2.47
2.45
1.10
2.44
2.0
2.39
7.33 2.38
2.36
1.5
3.38
2.36
1.25
2.34
6.14
1.0
2.33
2.92 1.53
1.53
0.5
1.51
1.50
1.48
0.0
1.47
1.45
233
13
230
Appendix
220
210
200
190
180
164.96
C NMR (126 MHz, Chloroform-d)
150.39
170
150.26
148.53
160
148.15
145.96
150
143.09
137.64
135.05
140
134.73
132.72
130
130.94
129.70
129.08
129.05
f1 (ppm)
128.57
120 110
127.59
126.51
100
126.47
124.07
123.76
90
121.26
119.01
80
118.07
115.85
70
53.62
53.51
52.72
60
52.60
46.82
50
46.78
46.73
40
36.71
30
29.85
23.78
23.40
20
22.40
22.26
10
21.69
11.19
0
10.57
234
8.60
8.59
Et2N
9.0
8.57
Cl
1H
Appendix
8.45
0.75 8.43
8.5
1.20 8.30
1.95 8.29
N
2.05 8.28
NH
8.0
1.00 7.96
7.95
N
H
0.99
3.5 7.77
O
7.5
7.77
7.75
7.75
7.0
NO2
6.5
4.60
4.59
4.58
6.0
4.57
4.57
4.56
5.5
4.55
3.17
3.15
5.0
3.14
3.12
1.02
f1 (ppm)
3.09
4.5
3.07
3.07
3.06
4.0
3.06
3.04
3.04
3.5
1.89
4.06 1.88
2.08 1.86
3.0
1.85
1.84
2.5 1.83
1.81
1.74
2.0
1.73
1.14 1.72
3.09 1.71
1.5
3.00 1.71
6.16 1.70
1.69
1.0
1.67
1.44
1.43
0.5
1.25
1.23
1.22
0.0
235
13
230
220
Appendix
210
200
190
180
170
C NMR (126 MHz, Methanol-d4)
167.24
160
153.71
150.34
145.10
150
139.63
138.53
140
138.33
129.02
130
127.57
125.62
123.60
119.42
117.04
f1 (ppm)
111.31
33.30
30
20.81
20
20.25
10
7.62
0
-10
236
9.5
8.56
8.54
Et2N
1H
7.95
9.0
Cl
Appendix
7.78
7.77
7.76
8.5
7.76
7.74
N
NH
7.72
8.0
3.6
7.61
N
H
7.61
Cl
7.5
7.60
7.59
7.59
7.0
7.58
7.57
NMR (500 MHz, Methanol-d4)
7.57
6.5
7.56
7.55
7.54
6.0
7.53
7.52
7.52
5.5
7.51
7.50
4.75
5.0
4.74
4.72
4.71
f1 (ppm)
4.5
3.16
3.16
3.14
4.0
3.13
3.11
3.11
3.5
3.09
3.09
3.07
3.0
1.89
1.88
2.5 1.86
1.80
1.79
2.0
1.78
1.77
1.77
1.5
1.76
1.74
1.46
1.0
1.45
1.44
1.24
0.5
1.22
1.21
1.19
0.0
237
13
230
220
Appendix
210
200
190
180
170.67
C NMR (126 MHz, Methanol-d4)
170
154.26
147.24
160
140.60
136.11
150
133.34
132.14
140
131.59
130.39
130
128.90
128.66
126.75
121.75
119.26
f1 (ppm)
120 110 100
90
80
70
60 52.65
52.58
50
40
34.84
30
22.16
21.95
20
9.08
10
8.99
0
-10
238
9.0
Et2N
1.00 8.87
1H
Cl
Appendix
8.10
8.02
8.5
8.00
0.98 7.91
2.17
N
7.90
8.0
NH
1.39 7.57
7.55
3.7
1.10
N
H
1.26
O 7.48
7.5
1.51 7.47
7.45
Cl
7.43
7.0
7.41
NMR (500 MHz, Chloroform-d)
5.0 6.5
6.0
5.5
4.5
f1 (ppm)
4.32
1.04
4.30
4.0
3.78
1.12 3.75
3.72
3.5
3.0
2.60
2.59
4.94 2.57
2.5 3.05 2.56
2.43
2.42
2.0
1.55
1.54
1.52
1.5
4.88 1.52
2.73 1.51
7.33 1.49
1.0
1.27
1.26
1.04
0.5
1.02
1.01
0.99
0.0
239
13
230
220
Appendix
210
200
190
180
170
C NMR (126 MHz, Chloroform-d)
165.40
160
150.70
150
148.64
145.89
135.46
140
135.18
134.86
130
132.37
130.24
129.15
128.37
126.37
f1 (ppm)
126.02
124.30
90
80
70
60
53.58
52.48
50
46.72
40
36.69
30
22.84
22.52
20
10.09
10
0
-10
240
1H
Et2N
10.0
Cl
Appendix
9.5
0.93 9.35
8.83
9.0
8.12
NH
1.00
3.8
N 8.09
H
8.01
8.5
O 8.01
1.98 8.00
1.03 7.91
8.0
1.06 7.89
7.50
Cl
2.04
NMR (400 MHz, Chloroform-d)
7.5
7.49
1.09
7.47
7.47
7.0
7.42
7.42
6.5
7.40
7.39
5.5 6.0
5.0
f1 (ppm)
4.5
1.05 4.35
4.0
1.11 3.76
3.5
2.66
2.64
2.62
3.0
2.60
4.12 2.49
2.47
2.5
2.28
2.45
2.43
2.0
1.10 1.53
1.52
1.5
3.03
3.74 1.52
1.51
6.08
1.0
1.50
1.28
1.26
0.5
1.07
1.05
1.03
0.0
241
13
230
220
Appendix
210
200
190
180
170
C NMR (126 MHz, Chloroform-d)
165.91
151.05
160
148.71
146.63
150
138.64
134.81
131.99
140
129.66
129.09
130
129.04
126.22
124.53
121.23
119.38
f1 (ppm)
120 110 100
90
80
70
60
53.49
50 52.30
46.58
40
36.58
30
22.61
22.30
20
10
9.54
0
242
-10
9.5
Et2N
9.0
1H
Cl
8.59
Appendix
1.04 8.57
8.56
8.5
1.03
7.95
7.95
N
NH
1.89 7.94
8.0
1.00 7.94
3.9
N
H
0.97 7.94
O
7.74
7.5
0.96
7.74
1.00
F
7.73
7.72
7.0
7.70
7.69
NMR (500 MHz, Methanol-d4)
7.68
6.5
7.68
7.67
7.42
6.0
7.41
7.39
5.5
7.37
7.36
7.35
5.0
7.34
1.07 4.72
4.71
f1 (ppm)
4.5
4.70
3.15
3.14
4.0
3.12
3.11
3.09
3.5
3.08
4.07 3.07
1.92 3.06
3.0
3.06
3.04
3.03
2.5
1.88
1.86
1.84
2.0
1.05 1.75
3.10 1.74
1.73
1.5
3.04
1.73
6.02
1.72
1.43
1.0
1.42
1.23
1.22
0.5
1.21
0.0
243
Appendix
167.60
167.58
154.51
147.18
140.74
140.26
135.60
135.53
132.14
132.12
128.89
126.77
126.23
126.20
122.98
122.88
121.20
52.71
52.52
48.34
34.71
22.19
21.73
9.02
13
C NMR (126 MHz, Methanol-d4)
Trifluoroacetic
Trifluoroacetic
acid
acid
230 220 210 200 190 180 170 160 150 140 130 120 110 100 90 80 70 60 50 40 30 20 10 0 -10
f1 (ppm)
244
Appendix
-115.14
-76.94
Trifluoroacetic
acid
19
F NMR (377 MHz, Methanol-d4)
10 0 -10 -20 -30 -40 -50 -60 -70 -80 -90 -100 -110 -120 -130 -140 -150 -160 -170 -180 -190 -200 -210
f1 (ppm)
245
9.5
Et2N
1H
9.0
Cl
1.00 8.89
Appendix
0.84 8.00
8.00
8.5
7.89
7.87
N
1.08
7.84
NH
8.0
1.17
7.82
N
H
2.12
3.10
7.78
O
1.10 7.76
7.5
1.18 7.51
F
0.95 7.50
7.49
7.0
7.49
7.47
NMR (500 MHz, Chloroform-d)
6.5
7.43
7.42
7.41
6.0
7.41
7.30
7.30
5.5
7.30
7.29
7.28
5.0
f1 (ppm)
4.5
4.29
1.04
4.27
4.0
1.12 3.72
3.5
2.52
2.51
3.0
2.49
2.48
4.25 2.37
2.5 2.16 2.36
2.36
2.35
2.0
1.54
1.54
1.53
1.5
4.28
1.52
3.02
1.23
6.24 1.22
1.0
0.99
0.97
0.96
0.5
0.0
246
13
230
Appendix
220
210
200
190
180
C NMR (126 MHz, Chloroform-d)
170
165.24
164.04
162.07
160
150.30
150
148.37
145.42
136.10
140
136.04
134.86
130
130.72
130.66
129.14
126.50
f1 (ppm)
123.87
120 110
123.16
123.14
100
121.44
119.51
90
119.46
119.34
115.25
80
115.06
70
60
53.55
52.62
50
46.77
40
36.69
30
23.51
22.33
20
10.82
10
0
247
Appendix
-111.57
19
F NMR (377 MHz, Chloroform-d)
10 0 -10 -20 -30 -40 -50 -60 -70 -80 -90 -100 -110 -120 -130 -140 -150 -160 -170 -180 -190 -200 -210
f1 (ppm)
248
9.5
8.86
Et2N
9.0
1H
Cl
8.74
Appendix
1.00
8.08
0.82
8.07
8.5
8.06
7.99
2.19
N
7.98
NH
8.0
1.09 7.88
N
H
1.22 7.87
3.11
O
7.42
7.5
1.14 7.41
2.25 7.40
7.40
7.0
7.20
F
7.18
NMR (500 MHz, Chloroform-d)
7.16
5.0 6.5
6.0
5.5
f1 (ppm)
4.5
4.29
1.12
4.28
4.0
1.20 3.71
3.5
2.51
2.50
3.0
2.48
2.47
4.37 2.37
2.5 2.35
2.43
2.34
2.33
2.0
1.54
1.53
1.5
4.78
1.52
3.35
1.51
6.66
1.0
1.48
1.48
1.22
0.5
1.21
0.98
0.97
0.0
0.95
249
13
230
220
Appendix
210
200
190
180
166.20
170
C NMR (126 MHz, Chloroform-d)
165.35
164.18
160
150.25
150
148.20
145.37
134.64
140
130.09
130.02
130
129.78
129.76
128.95
126.28
123.79
f1 (ppm)
121.27
119.52
90
80
70
60
53.40
50 52.45
46.61
40
36.50
30
23.27
22.19
20
10.57
10
0
-10
250
Appendix
-107.08
19
F NMR (377 MHz, Chloroform-d)
10 0 -10 -20 -30 -40 -50 -60 -70 -80 -90 -100 -110 -120 -130 -140 -150 -160 -170 -180 -190 -200 -210
f1 (ppm) 251
1.00 8.31
8.27
1.15
Et2N
8.25
1H
Cl
8.0
7.84
Appendix
0.88 7.83
2.02 7.82
1.19 7.80
7.5
7.49
N
NH
7.49
7.47
3.12
N
H
7.0
7.46
O
2.19 6.74
6.72
6.5
NMR (400 MHz, Methanol-d4)
NH2
4.5 6.0
5.5
5.0
4.13
4.11
1.62 4.10
f1 (ppm)
4.0
4.09
4.07
3.5
3.0
2.51
2.49
3.66 2.47
2.5
2.45
2.02
2.40
2.38
2.0
1.65
1.63
1.47
1.41
1.45
1.5
3.24 1.44
3.30 1.42
6.15 1.26
1.0
1.25
0.98
0.96
0.5
0.95
252
0.0
13
220
210
Appendix
200
190
180
169.78
170
C NMR (126 MHz, Methanol-d4)
160
154.45
154.12
148.95
150
148.71
136.26
140
130.65
127.88
130
126.72
125.18
121.94
120
121.23
115.33
114.69
110 100
f1 (ppm)
80
70
60 90
53.36
52.16
50
40 47.64
36.67
30
23.79
21.90
20
10.92
10
0
-10
253
9.5
Et2N
8.92
9.0
1H
Cl
Appendix
1.00
8.34
8.00
7.99
8.5
0.94
7.98
1.05
N
7.96
NH
1.97
8.0
7.88
1.13
N
H
7.86
3.13
7.41
O
7.5
1.09 7.41
7.40
7.39
2.25
7.0
6.99
6.98
OMe
5.0 6.5
6.0
5.5
f1 (ppm)
4.5
4.26
1.05 4.24
3.88
4.0
3.23
1.22 3.73
3.70
3.68
3.5
2.48
2.47
3.0
2.45
2.44
2.5 4.60 2.34
2.33 2.33
2.32
2.30
2.0
1.53
1.51
1.5
4.68
1.50
3.31
1.49
6.80
1.0
1.49
1.47
1.19
0.5
1.18
0.97
0.95
254
0.0
0.94
13
210
Appendix
200
190
180
170
165.98
162.96
C NMR (126 MHz, Chloroform-d)
160
150.28
150
148.13
145.14
134.61
140
129.52
129.05
130
126.40
125.98
123.80
120
121.55
120.07
110
114.20
f1 (ppm)
90
80
70
60100 55.64
53.55
50
52.73
46.81
40
36.71
30
23.80
22.25
20
11.22
10
0
255
9.5
Et2N
Cl
1H
9.0
Appendix
8.80
1.00
8.18
8.17
8.5
8.02
1.94
N
8.01
NH
1.02 7.95
8.0
1.17 7.94
N
H
3.14 7.42
O
7.41
7.5
1.15
2.24 7.40
7.40
7.35
7.0
7.33
SMe
6.5
6.0
4.42
4.40
3.84
5.5
3.80
3.77
2.73
5.0
2.72
2.58
2.54
4.5
f1 (ppm)
1.19
2.54
2.53
2.52
4.0
1.26 2.52
2.51
3.5
2.50
2.48
2.46
3.0
1.71
4.16 1.66
1.65
6.77
2.5 1.55
1.53
1.53
2.0
1.52
2.33 1.50
3.44 1.50
1.5
3.56 1.49
1.48
6.36
1.47
1.0
1.46
1.34
1.32
0.5
1.13
1.11
0.0
1.10
256
13
230
220
Appendix
210
200
190
180
170
C NMR (126 MHz, Chloroform-d)
166.45
151.45
160
148.80
144.30
137.55
150
134.50
129.25
140
128.81
128.73
130
125.80
125.29
124.96
121.01
119.38
f1 (ppm)
120 110 100
90
80
70
60
53.31
51.95
50
46.32
40
36.39
30
22.70
20
21.15
14.95
10
8.37
0
-10
257
10.5
Et2N
10.0
1H
Cl
Appendix
1.06 9.71
9.5
9.0
8.40
N
NH
8.39
8.29
8.5
N
H
3.15
1.15 7.92
O
1.00 7.91
7.85
8.0
2.11
7.84
0.98
7.50
1.07
7.5
7.50
7.49
NMe2
7.49
NMR (800 MHz, Dimethyl sulfoxide-d6)
7.0
6.77
2.34
6.76
6.5
5.98
1.14
6.0
5.97
5.5
3.99
f1 (ppm)
5.0
3.98
3.98
3.97
4.5
3.02
3.02
1.41
4.0
3.01
3.01
3.01
3.5
3.00
2.99
6.84 2.98
3.0
2.5 2.98
4.21 2.41
2.28 2.41
2.38
2.0
1.58
1.57
1.93 1.57
1.5
2.02 1.57
3.20 1.36
1.0
6.45 1.35
1.13
1.12
0.5
0.86
0.85
0.84
0.0
258
13
210
Appendix
200
190
180
170
165.98
160
154.20
152.42
150
147.67
146.07
133.05
140
C NMR (201 MHz, Dimethyl sulfoxide-d6)
129.05
127.50
130
124.85
124.61
120
120.32
119.96
114.71
110
110.84
100
f1 (ppm)
80
70
60 90
51.99
50
50.41
40 46.06
39.99
34.97
30
22.61
21.51
20
10
7.16
0
-10
259
9.5
8.95
Et2N
9.0
1H
Cl
1.00 8.95
Appendix
8.34
8.01
8.5
8.00
0.93 7.91
N
1.13 7.89
NH
8.0
3.40 7.89
N
H
7.87
3.16
O
7.42
7.5
1.20
7.42
2.39 7.41
7.40
7.0
7.32
7.31
NMR (500 MHz, Chloroform-d)
5.0 6.5
6.0
5.5
f1 (ppm)
4.5
4.26
1.18
4.24
4.0
1.31 2.49
2.47
3.5
2.46
2.44
2.35
3.0
2.33
2.32
8.24 2.32
2.5 1.53
2.64
1.51
1.51
2.0
1.50
1.49
1.49
1.5
4.90
1.48
3.60
1.48
6.98
1.0
1.47
1.47
1.20
0.5
1.18
0.97
0.96
0.0
0.94
260
13
230
220
Appendix
210
200
190
180
170
C NMR (126 MHz, Chloroform-d)
166.42
150.35
160
148.23
145.26
150
143.02
134.66
130.93
140
129.67
129.09
130
127.66
126.40
123.87
121.53
119.95
f1 (ppm)
120 110 100
90
77.41
77.37
80
77.16
77.05
70
76.90
60
53.53
52.67
50
46.80
40
36.67
30
23.57
22.30
22.28
20
21.69
10.99
10
0
-10
261
Appendix
N
N N
N
2H3PO4
Cl N
4.4
1H NMR (400 MHz, Deuterium Oxide-d2)
262
Appendix
13
C NMR (126 MHz, Deuterium Oxide-d2)
263
Appendix
31
P NMR (162 MHz, Deuterium Oxide-d2)
264
8.61
1H
8.60
7.99
9.0
Appendix
7.98
N
7.89
Cl
1.00
7.87
8.5
7.36
7.35
4.6
0.94
8.0
7.34
N
N
1.05 7.34
6.75
7.5
6.74
1.02
2.45
2.45
7.0
2.44
NMR (500 MHz, Chloroform-d)
1.05 2.44
2.42
6.5
2.42
2.41
2.41
6.0
2.33
2.33
2.32
5.5
2.32
2.32
2.31
5.0
2.30
2.30
1.72
4.5
f1 (ppm)
1.71
1.70
1.69
4.0
1.08 1.53
1.52
3.5
1.52
1.51
1.50
3.0
1.42
3.09
1.41
1.40
2.5
4.14 1.40
2.06 1.40
1.40
2.0
1.39
1.14 1.38
1.12 1.38
1.5
1.37
2.14
1.37
3.15
1.36
1.0
6.14
1.35
1.26
1.24
0.5
0.96
0.94
0.93
0.0
265
Appendix
157.56
151.53
150.78
134.64
128.90
125.99
125.22
121.86
109.02
58.79
52.87
46.90
32.48
32.34
24.63
17.09
11.69
13
C NMR (126 MHz, Chloroform-d)
210 200 190 180 170 160 150 140 130 120 110 100 90 80 70 60 50 40 30 20 10 0
f1 (ppm)
266
9.5
1H
Appendix
9.0
N
8.5
8.10
4.9
1.00
8.08
H
N
0.40
8.0
8.06
O
7.5
NMR (500 MHz, Chloroform-d)
7.0
1.06 6.55
6.5
6.0
0.23 5.93
5.0 5.5
f1 (ppm)
4.5
4.04
4.03
4.02
1.21
4.0
4.02
4.01
0.29
3.5
2.54
2.53
2.52
3.0
2.51
2.42
2.5 6.50 2.42
3.26 2.42
1.51
1.51
2.0
1.50
1.49
6.54
1.49
1.5
0.89
1.21
3.93
1.20
8.98 1.16
1.0
2.28 1.15
1.03
1.02
0.5
1.00
1.00
267
0.0
0.99
Appendix
163.78
160.68
53.02
52.65
48.37
46.86
46.77
44.01
36.00
34.80
23.58
23.14
22.79
20.84
11.62
11.42
13
C NMR (126 MHz, Chloroform-d)
210 200 190 180 170 160 150 140 130 120 110 100 90 80 70 60 50 40 30 20 10 0
f1 (ppm)
268
9.5
1H
Appendix
9.0
N
8.5
4.10
8.0
H
N
7.5
7.0
NMR (500 MHz, Chloroform-d)
6.5
6.0
5.5
2.55
2.53
5.0
2.52
2.50
f1 (ppm)
2.43
4.5
2.41
2.40
1.49
4.0
1.48
1.48
3.5
1.48
1.48
1.47
3.0
1.47
1.46
5.13 1.46
2.5
1.45
5.18
1.45
1.45
2.0
1.31
1.31
1.31
3.00
1.5
1.30
1.04
1.30
3.04
1.29
1.0
6.54 1.29
1.06
1.04
0.5
1.03
1.01
1.00
0.0
269
Appendix
55.07
53.32
46.87
34.91
33.79
23.68
19.79
11.70
13
C NMR (126 MHz, Chloroform-d)
210 200 190 180 170 160 150 140 130 120 110 100 90 80 70 60 50 40 30 20 10 0
f1 (ppm)
270
9.5
1H
9.0
Appendix
Cl
8.47
N 8.45
8.5
1.00
8.01
1.00 7.99
8.0
7.96
1.00
N 7.95
NH
7.40
7.5
1.01 7.40
7.38
0.97 7.38
7.0
NMR (500 MHz, Chloroform-d)
7.02
6.39
6.5
1.03
6.38
5.0 6.0
5.5
f1 (ppm)
4.5
3.51
4.0
3.50
3.49
2.12 3.39
3.5
4.15 3.38
3.37
3.36
3.0
2.62
2.61
2.14
2.60
2.5 2.60
1.80
1.80
2.0
6.46 1.79
2.31 1.79
1.79
1.5
1.78
1.77
1.0
1.74
1.73
1.72
0.5
1.68
1.67
0.0
271
Appendix
13
C NMR (126 MHz, Chloroform-d)
272
1H
9.0
8.93
N
1.00
Appendix
Cl
8.5
8.06
8.06
1.00
4.15
8.03
N
N
8.0
1.08
8.01
7.51
1.08 7.50
7.5
NO2
7.49
7.49
7.0
NMR (500 MHz, Chloroform-d)
5.0 6.5
6.0
5.5
4.01
4.00
4.5
f1 (ppm)
4.00
4.00
1.20
4.0
3.99
3.99
3.98
3.5
3.97
2.85
2.52
3.0
2.50
3.07
2.45
4.10
2.45
2.5 2.15 2.43
2.41
2.0
1.36
1.86
1.40
1.84
2.06 1.67
1.5
3.24 1.65
1.48
6.42 1.46
1.0
1.45
1.43
0.5
1.42
1.01
1.00
0.0
0.98
).
273
Appendix
151.14
150.71
147.91
137.64
137.05
129.68
127.56
127.19
123.83
60.99
52.76
46.94
33.91
32.72
24.45
18.18
11.42
13
C NMR (126 MHz, Chloroform-d)
210 200 190 180 170 160 150 140 130 120 110 100 90 80 70 60 50 40 30 20 10 0
f1 (ppm)
274
10.0
Et2N
9.5
0.91 9.40
Cl
Appendix
9.0
1.00 8.62
8.5
N
N
4.16
7.98
7.96
7.93
8.0
1.05
NH2
7.92
1.05
7.55
1.07
7.5
7.54
7.52
6
7.52
1H NMR (400 MHz, Dimethyl sulfoxide-d )
5.0 7.0
6.5
6.0
5.5
f1 (ppm)
4.04.5
3.55
3.53
1.78 3.52
3.5
3.51
4.05 3.05
3.0
5.13 3.04
3.03
2.5 2.96
2.95
2.93
2.0
1.59
1.58
4.27 1.56
1.5
1.56
3.31
1.55
6.21
1.54
1.0
1.52
1.19
1.17
0.5
1.14
1.12
0.0
1.10
275
Appendix
140.47
139.38
138.79
138.67
130.61
127.23
126.25
125.55
124.01
55.24
50.92
46.45
46.30
35.02
31.32
20.25
17.77
8.56
8.54
13
C NMR (101 MHz, Dimethyl sulfoxide-d6)
Trifluoroacetic Trifluoroacetic
acid acid
210 200 190 180 170 160 150 140 130 120 110 100 90 80 70 60 50 40 30 20 10 0 -10
f1 (ppm)
276
9.5
1H
N
9.0
Appendix
Cl
1.00 8.67
8.5
8.00
4.17
8.00
N
N
1.03
8.0
7.98
1.34
7.97
N3
7.43
7.5
1.21 7.42
7.41
7.41
NMR (500 MHz, Chloroform-d)
5.0 7.0
6.5
6.0
5.5
3.51
3.50
3.48
f1 (ppm)
4.5
3.47
2.93
2.51
4.0
2.50
2.49
1.31 2.47
3.5
2.40
2.38
2.37
3.0
4.12
1.68
1.67
4.48
2.5 1.66
2.05 1.65
1.65
2.0
1.65
1.63
1.37 1.49
1.5
3.30 1.48
3.31 1.48
6.65 1.47
1.0
1.47
1.21
1.20
0.5
1.00
0.98
0.97
0.0
277
Appendix
147.93
146.13
145.08
134.30
128.91
128.14
127.48
125.96
125.91
58.61
53.02
46.94
35.52
32.74
24.11
17.90
11.54
13
C NMR (126 MHz, Chloroform-d)
230 220 210 200 190 180 170 160 150 140 130 120 110 100 90 80 70 60 50 40 30 20 10 0
f1 (ppm)
278
Appendix
279
Appendix
12.23
12.22
8.12
8.10
7.95
7.94
7.63
7.62
7.40
7.39
7.38
7.37
OH
N3
Cl N
4.18
1H NMR (500 MHz, Dimethyl sulfoxide-d6)
1.06
1.03
1.00
1.00
1.05
12.5 12.0 11.5 11.0 10.5 10.0 9.5 9.0 8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5 0.0
f1 (ppm)
280
Appendix
171.72
139.35
136.45
130.19
127.04
124.10
122.37
120.30
117.71
13
C NMR (126 MHz, Dimethyl sulfoxide-d6)
210 200 190 180 170 160 150 140 130 120 110 100 90 80 70 60 50 40 30 20 10 0 -10
f1 (ppm)
281
Appendix
282
283