Cap 7 - Tumor-Targeting Strategies
Cap 7 - Tumor-Targeting Strategies
7 Tumor-Targeting
Strategies
7.1 INTRODUCTION
One of the greatest challenges in the discovery and development of new therapeutic
agents and strategies for the treatment of cancer is the achievement of selectivity
between tumor cells and healthy tissues. With few exceptions, all agents in clinical
use today are associated with varying degrees of side effects (i.e., toxicities) due to
their collateral action on healthy cells and tissues. Therefore, many ingenious meth-
ods have been devised to target drugs to tumor cells or tumor masses.
One obvious way to achieve targeting is through antibodies directed towards
markers on the surface of tumor cells. This method has given rise to four strategies.
First, an antibody can be used as a single entity; this approach has been validated
through the development of such agents as trastuzumab (Herceptin™) and bevaci-
zumab (Avastin™). Second, a drug can be directly attached to a tumor-specific
antibody to guide it to tumor cells, an example of which is gemtuzumab ozogamicin
(Mylotarg™). A third antibody strategy involves the use of antibodies to guide an
attached radioactive element or ligand to the tumor. This approach was exemplified
by pemtumomab (Theragyn™), an antibody labeled with yttrium-90 (no longer in
development). A fourth strategy involves the use of antibodies to guide a pro-
drug/drug-releasing enzyme to tumor cells, such as in antibody-directed enzyme
prodrug therapy (ADEPT).
A different approach to achieving selectivity involves targeting tumor vascula-
ture. Anti-angiogenic agents work by blocking the ability of tumors to develop new
vasculature vital for their growth and survival; healthy tissues are relatively unaf-
fected by these drugs because their vasculature is already in place. This approach
has been recently validated by the success of bevacizumab (Avastin™) in the clinic.
An alternative strategy involves targeting the existing blood supply of tumors, which
can be functionally distinct from the vasculature of healthy tissue. Drugs of this
class are known as vascular disrupting agents (VDAs). In particular, because tumor
vasculature is usually less mature than that of other tissues and organs, the cells are
more prone to drugs such as combretastatin due to their lack of actin and relatively
exposed tubulin.
Other triggers, such as oxygen concentration, are also being used to selectively
release drugs at tumor sites. For example, it is well established that the centers of
most tumor masses are hypoxic compared to healthy tissue due to an abnormal
vasculature. Therefore, prodrugs that are activated under these hypoxic (i.e.,
bioreductive) conditions have been designed. This approach is exemplified by AQ4N,
157
emerge, partly due to the technical challenges involved but also because early studies
with vinblastine-antibody conjugates were disappointing. However, the clinical
potential of MAbs has been recently validated by the success of such agents as
trastuzumab (Herceptin™) and bevacizumab (Avastin™). Even so, in practice, these
MAbs tend to be most efficacious when used in combination with traditional cyto-
toxic agents. Also, for solid tumors, debulking may be initially required.
Rather than use MAbs alone, an alternative approach to enhance efficacy is to
attach a cytotoxic agent through a chemical linker. The linker can be designed to
cleave specifically at the tumor site, thus releasing the cytotoxic agent. For example,
conjugates have been reported that contain linkers designed to cleave on exposure
to the enzyme cathepsin, which is overexpressed in some tumor cell types. With this
type of construct, in which exposure to the cytoplasm within the cell is important
for drug release, it is necessary to demonstrate that, once bound to the tumor cell,
the drug-antibody conjugate is internalized. If internalization does not occur, then
the full cytotoxic effect may not be achieved, although some drug may still be
released in the vicinity of the tumor. If some release occurs externally to the cancer
cell, then the drug is free to diffuse into neighboring cells, a phenomenon known
as the bystander effect. A further consideration is that, for maximum efficacy, as
many linker-drug units as possible should be attached to a single antibody without
affecting its antigen-recognition or pharmaceutical properties, a technically chal-
lenging feat. For example, in the case of gemtuzumab ozogamicin (Mylotarg™), a
loading of calicheamycin of 4 to 6 moles per mole of antibody over 50% of the
antibody’s surface is achieved. Another approach is to ligate the MAb to a radiop-
harmaceutical to create an agent for use in radioimmunotherapy. This combines the
advantages of targeted radiation therapy and specific immunotherapy.
Furthermore, IGN311 has favorable safety and tolerability profiles and a serum half-
life of more than 20 days. Similarly, a CD4-targeted antibody called HuMax-CD4
(Genmab AS/Serono SA) is being evaluated for the treatment of T-cell lymphomas.
This antibody has been granted fast-track status by the U.S. FDA and is currently
being evaluated in a Phase III clinical trial.
hP67.6
HN
O O
O H3C CH3
O
HO
CH3 O H3C NNH S H
S N
H3C O
l O
S O H3C OCH3
N O
H O H
O OCH3 OH OH
O
OCH3 H3CH2C
H3C O
HO H3C N O
H3CO OH H3CO
O
7.2.3.1 Pemtumomab
exploited, including the folate receptor CA-125 and PEM. MAbs to PEM were
developed in the early 1980s; one of the best known is HMFG-1, the antibody that
forms the basis of pemtumomab.
Research into the tumor-associated mucin molecule led to the discovery of a
gene on chromosome 1 of the human genome, coding for the core protein of PEM.
This gene, called the MUC-1 gene, codes for the MUC-1 antigen (a 20 amino-acid
antigen) repeated in tandem, which forms the core protein of PEM. In normal
epithelial tissue, the MUC-1 gene product is fully glycosylated (covered in carbo-
hydrates) and is therefore “hidden.” However, in many tumors of epithelial origin,
including ovarian cancer, the glycosylation is incomplete, which results in exposure
of amino acid sequences of the MUC-1 gene product, which was exploited by
pemtumomab.
During development, the fact that ovarian cancer is normally confined to the
peritoneal cavity was also exploited, and so the radiolabeled antibody was admin-
istered intraperitoneally. This strategy, known as regional immunotherapy, results
in increased localization of the antibody compared to that achievable when given
systemically, thus increasing the radiation dose to the tumor. Initially, iodine-131
was used as the isotope because it had been successfully used itself in the treatment
of other tumor types (e.g., thyroid cancer). However, the major disadvantage of
iodine-131 was that, being a γ-emitter, patients had to be isolated and kept in the
hospital for at least 5 days. Exposure of hospital staff to the radiation was also a
potential hazard. To overcome these disadvantages, the pure β-emitting isotope
yttrium-90 was used instead. This isotope had already been used on its own to treat
liver cancer (hepatoma) and raised none of the same concerns regarding exposure,
isolation, or long hospitalization. However, in order to use high amounts of this
radioactive metal, new chemical chelates had to be developed that could bind
yttrium to the antibody in an almost nonreversible manner.
The initial clinical trials of the yttrium-90 labeled MAb (pemtumomab) gave
promising results. Patients underwent surgery followed by chemotherapy and then
waited for some time before receiving pemtumomab. Those who received the agent
4 to 6 weeks after their chemotherapy, and at a time when they had no evidence of
disease, survived longer than patients who only received surgery and chemotherapy.
Five-year survival data on patients in a Phase II trial surpassed any previously
reported therapy for the treatment of ovarian cancer, which led to a multinational,
multicentered Phase III clinical trial to evaluate pemtumomab in a much wider setting
using a greater number of patients. Unfortunately, the results of this trial, published
in 2004, suggested that the benefits to patients were not significant enough to warrant
continued development.
by Folkman in the early 1970s. It is now recognized that for any tumor to grow
beyond a volume of 1 to 2 mm3, a so-called “angiogenic switch” must be present,
prompting the formation of new vasculature (i.e., neovascularization) (Figure 7.1
and Scheme 7.1). Since Folkman’s original observations, key molecules in the
angiogenesis process have gradually been identified, such as VEGF and its receptors,
culminating in the recent clinical proof of the concept of targeting VEGF in colorectal
cancer with the humanized MAb bevacizumab (Avastin™). Many small-molecule
inhibitors of VEGF receptors are also now in clinical development (e.g., SU11248
and PTK787/ZK22854).
An alternative but complementary strategy to anti-angiogenic agents is that of
targeting established tumor vasculature (Scheme 7.1). This approach was first sug-
gested as long ago as the 1920s, but research in this area was given impetus by
Denekamp in the early 1980s and has led to a class of drugs now known as Vascular
Disruptive Agents (VDAs), whose role is to cause a rapid and selective shutdown
of existing tumor blood vessels. The first known agent of this type was combretastatin
(and its derivatives), which was followed by related prodrug forms, such as CA4P
and Oxi4503. 5,6-Dimethylxanthenone-4-acetic acid (DMXAA), an agent with a
similar effect on existing tumor vasculature, is presently in late-stage clinical trials.
One attraction for clinicians is that agents such as combretastatin and DMXAA
cause a rapid and selective vascular shutdown in tumors in a matter of minutes to
hours, and so can be given in intermittent doses to potentiate the activity of conven-
tional chemotherapeutic agents (see below). Many other potential drugs of this type
are at the discovery and development stage; examples include ZD6126 and
AVE8062A.
FIGURE 7.1 Photomicrograph showing the process of angiogenesis. The image shows the
formation of numerous new blood vessels in the area of a metastasis on the surface of a
human lung.
DMXAA
Bavacizumab
AVE8062A
ZD6126
SUII248 PTK787/ZK22854
Small solid tumors with new blood Larger solid tumors with
vessels. Anti-angiogenic agents established blood vessels.
have major effect on tumor VDAs have a major effect on the
periphery inhibiting endothelial central part of the tumor causing
proliferation and migration. vessel occlusion and necrosis.
SCHEME 7.1 Mechanism of action of anti-angiogenic agents and VDAs. (Adapted with
permission from Kelland, L.R., Curr. Cancer Ther. Rev., 1:1-9, 2005).
7.3.1.1 Bevacizumab
7.3.2.1 Combretastatins
(a) (b)
FIGURE 7.2 (a) African bush willow tree; (b) mohair tapestry produced in the Zulu village
of Howick in Kwa-Zulu (Natal, South Africa) showing the gently sweeping elegance of the
bush willow along the Mooi River in Northern Natal.
STRUCTURE 7.2 Structures of the combretastatins A-1 and A-4, and related prodrugs CA4P
and Oxi4503.
rotation about the olefinic bridge is crucial for biological activity, as is the distance
between the two rings.
The interesting feature of the combretastatins is their selectivity for tumor
vasculature rather than healthy tissue. Combretastatin A-1(CA-1) is known to bind
to tubulin with a higher affinity for β-tubulin than colchicine at or near colchicine-
binding sites, thus destabilizing the tubulin cytoskeleton. When this tubulin structure
is disrupted, the endothelial cells change from a flat streamlined profile to a round
swollen shape because the internal tubulin skeleton is no longer able to mechanically
support their elongated shape. Due to this shape change, the cells effectively plug
the capillaries, thus restricting tumor blood flow. In addition, a process of apoptosis
is initiated in the cells of tumor blood vessels. This appears to first kill cancer cells
in the core of the tumor but with the apoptotic effect then radiating out, thus starving
the tumor of nutrients which, in turn, leads to exposure of the basement membrane,
hemorrhage, and coagulation (i.e., tumor necrosis). The selectivity arises because,
compared to mature endothelial cells that line blood vessels in normal tissues, tumor
blood vessels are usually newly formed and immature, and only vessels of this type
can be affected by combretastatin. Actin, a protein not present in immature endo-
thelial cells, protects the tubulin in mature endothelial cells from the effects of
combretastatin, and hence their shape is maintained. Interestingly, actin does not
appear for a number of days after the formation of new endothelial cells, and so this
provides a window of opportunity to achieve selectivity toxicity toward the tumor
microvasculature.
In addition to these fundamental differences between immature and mature
endothelial cells, characteristics of the tumor microcirculation, such as high inter-
stitial fluid pressure, procoagulant status, vessel tortuosity, and heterogeneous blood
flow distribution, assist the selectivity process. Reduced blood flow results in sig-
nificant hypoxia within the tumor, leading to cell death and regression. The study
of these agents in the clinic has been facilitated by the development of techniques
using positron emission tomography (PET) and magnetic resonance imaging (MRI)
that allow tumor blood flow to be measured in patients during treatment.
It is noteworthy that neither of the two side effects commonly associated with
cytotoxic agents (i.e., leukopenia and alopecia) occur during or after treatment with
combretastatin. However, one of the most serious side effects is tumor pain and, at
high doses (more than 90 mg/m2), some patients show signs of lung and heart
toxicity. Other more minor adverse effects observed in clinical trials include faint
flush, transient blood pressure changes, abdominal pain, nausea, vomiting, fever,
light-headedness, headache, diarrhea, bradycardia, and tachycardia. Finally, on an
electrocardiogram, nonspecific ST-T wave changes have been observed that peak
between 3 and 5 hours after infusion.
Despite extensive clinical evaluation of the original combretastatin compounds,
efficacy proved disappointing. The poor aqueous solubility of these molecules was
thought to be partly responsible for their poor performance in the clinic, and so a
number of phosphate prodrugs have prepared, such as Oxi4503 and CA4P (see
Section 7.3.2.2). These prodrugs are presently being evaluated in the clinic. Other
types of tubulin depolymerizing agents are also in clinical development (e.g., ZD6126
and AVE8062), along with DMXAA, which has a different mechanism of action.
Interest in this nontubulin class of VDAs began in the mid-1980s, with the seren-
dipitous discovery of the antitumor properties of flavone-8-acetic acid (FAA), a
molecule originally synthesized as a nonsteroidal anti-inflammatory agent by Lyon-
naise Industrielle Pharmaceutique (Structure 7.3). FAA was found to be active (even
curative) against a wide variety of preclinical solid tumor models, and its mechanism
of action appeared to be related to the production of tumor necrosis factor (TNF).
Initial Phase I studies using an ester prodrug (LM985) of FAA showed that the dose-
limiting toxicity was acute, reversible hypotension occurring during drug infusion
at doses of 1500 mg/m2. Later Phase I studies using FAA itself (LM975) showed
that hypotension was again dose-limiting but at the significantly higher dose of 10
g/m2. However, no evidence of efficacy was apparent from any of these trials,
including a Phase II study in NSCLC. A Phase I trial of FAA in combination with
recombinant interleukin-2 gave one complete and two partial responses, but severe
hypotension (grade 3 or 4) was observed after the third dose of FAA, with no sign
of increased tumor necrosis in several tumor biopsies. This led to the clinical
abandonment of FAA on the grounds that it did not elicit similar antivascular effects
in humans as those seen in many murine tumor models.
A synthetic program to identify a more potent inducer of TNF-α was initiated
by Denny, Baguley, and colleagues at the University of Auckland, New Zealand,
O
O
O
H3C O
CH2COOH
CH3 CH2COOH
Flavone-8-acetic acid (FAA) DMXAA
Blood capillaries
Invasive zone
Zone of cell
stasis low in oxygen
Necrotic center
Proliferating zone with well-
of dead or
developed capillary network
dying cells
and high oxygen level
SCHEME 7.2 Section through a solid tumor showing the necrotic center and the development
of a network of small blood vessels.
Pro-drug
Cellular enzyme
Prodrug
Low oxygen intermediate High oxygen Trigger Effector
(tumors) (normal tissues)
Activated Restored Superoxide
drug prodrug radical Link broken
This part on activation Effective
May be toxic Potentially
reduced (reduction) drug
directly or release damaging but cell
by cells
an ‘effector’ has defenses
A B
SCHEME 7.3 (a) Schematic diagram showing the bioreductive prodrug activation pathway;
(b) design of bioreductive prodrug.
in the activating process is oxygen-reactive. In this case, the relatively higher oxygen
level in healthy cells and tissues protects the prodrug from reduction by reacting
with an intermediate (usually a short-lived “free radical”), thus restoring the drug
to its original (prodrug) form.
A number of examples of bioreductive drugs exist, some of which are either in
clinical use or are currently undergoing preclinical or clinical studies. The first group
consists of molecules containing quinone moieties; best-known members include
mitomycin C (which contains an indolequinone nucleus) and the experimental agent
EO9. The mechanism of action of mitomycin is described in detail in Chapter 3.
The second group of bioreductive agents includes molecules based on aromatic N-
oxides (e.g., tirapazamine) and aliphatic N-oxides (e.g., AQ4N). The latter is pres-
ently being evaluated in the clinic, and preliminary pharmacodynamic and pharmaco-
kinetic results convincingly demonstrate that it is working through a bioreductive
mechanism (see Section 7.3.3.1). Another group comprises molecules containing
nitroheterocycles and includes experimental agents such as CB-1954, SN-23862,
NITP, SR4554, and pimonidazole, the latter of which is also being evaluated in the
clinic for use as a hypoxia marker.
7.3.3.1 AQ4N
Apart from the natural product mitomycin C, AQ4N is the first example of a
bioreductive agent to provide encouraging results in the clinic (see Structure 7.4).
It is a water-soluble anthracene derivative that, when administered intravenously,
targets the hypoxic compartments of tumors. Although AQ4N enters all cells freely,
in theory it is only converted to a charged (at physiological pH) tertiary amine species
in hypoxic tumor cells. This amine species becomes trapped in the hypoxic cells,
killing them through DNA intercalation and inhibition of topoisomerase II. Clini-
cians are generally enthusiastic about this mechanism of action because treating the
hypoxic fractions of tumors has the potential to significantly enhance selective
toxicity and thus improve therapeutic index.
AQ4N itself is a bis(N-oxide) which, because it is uncharged, crosses cell
membranes with ease. However, in hypoxic tumor cells, it is selectively and irre-
versibly reduced to AQ4, the charged quaternary amino form, where it remains
CH3 CH3
O−
N NH
OH O HN + CH3 OH O HN + CH3
Bioreductive
Conditions
CH3 CH3
OH O HN O− OH O HN
N NH
+ CH + CH
3 3
AQ4N AQ4
(N-Oxides are not charged overall and (After reduction, tertiary amines cause
allow molecule to cross cell membrane) molecule to become “trapped” in cell
due to charge)
STRUCTURE 7.4 Structures of AQ4N and its biologically active metabolite (AQ4).
localized. The bioreduction process is enhanced in the absence of oxygen but inhib-
ited strongly in its presence. It is thought that when radiotherapy or chemotherapy
kills oxygenated cells on the outside of a tumor, the more-central quiescent AQ4-
containing cells become reoxygenated. When these cells resume replication, they
are rapidly killed by the antitopoisomerase II activity of AQ4. As demonstrated in
a recent Phase I clinical trial, systemic toxicity is thus reduced to a very low level
(i.e., the agent has a high maximum tolerated dose) because the prodrug form
(AQ4N) is relatively nontoxic and is only transformed into the active agent (i.e.,
AQ4) when it is inside hypoxic tumor cells.
Phase I clinical trials involving patients with non-Hodgkin’s lymphoma and
advanced solid tumors are presently underway. AQ4N is also being evaluated in the
treatment of esophageal cancer in combination with radiation therapy. During these
trials, it has been possible to confirm by studying biopsy material that the drug is
converted into its tertiary amine form (AQ4) in hypoxic cells. One curious but
nonproblematic side effect observed with AQ4N in these clinical trials was that the
mucous membranes of patients acquired a blue coloration, as did body fluids such
as urine and saliva. Interestingly, this effect was welcomed by many patients, who
felt that the unusually visible sign of the drug in their bodies might be associated
with a beneficial effect on their tumors.
7.4.1 ADEPT
Bagshawe and co-workers first introduced ADEPT in the early 1990s. In this two-
or three-component approach, an antitumor antibody linked to an enzyme is first
used to target the enzyme to tumor cells. A second antibody selective for the
antibody-enzyme conjugate may then be introduced to clear the conjugate from
general circulation, thus reducing systemic toxicity. A prodrug form of a cytotoxic
agent is then administered, which is transformed into the active agent by the anti-
body-bound enzyme selectively at the tumor site (Scheme 7.4 and Scheme 7.5).
Collateral damage to healthy tissue is theoretically avoided because the enzyme
chosen has no human equivalent (e.g., a bacterial enzyme). Variations of this
approach (GDEPT and VDEPT) were later established in which the activating
enzyme is introduced through organ-specific gene therapy or other means rather than
through an antibody-enzyme conjugate.
The ADEPT system originally proposed by Bagshawe utilizes the bacterial
enzyme carboxypeptidase G2 (CPG2), which is a metalloenzyme isolated from a
Pseudomonas species. It has no mammalian counterpart and catalyzes the conversion
of both reduced and nonreduced folates into pteroates and L-glutamic acid. It is also
known that CPG2 is capable of cleaving the amidic bond of methotrexate and folic
acid at the benzoylglutamate linkage. Thus, a conjugate of CPG2 with an appropriate
monoclonal antibody was designed for the selective activation of chemically
“masked” nitrogen mustards. The original experimental mustard prodrugs were
bifunctional alkylating agents incorporating either chlorine- or sulfonate-leaving
groups in which the activating effect of the ionizable carboxyl group had been
inhibited by protection with a glutamic acid moiety connected through an amide
linkage that formed the substrate for CPG2 (Scheme 7.4).
SCHEME 7.4 Examples of ADEPT prodrugs and their conversion to active agents. The
original experimental mustard prodrugs were bifunctional alkylating agents incorporating
either chlorine or sulfonate leaving groups (1-3).
Cancer Cell
Ab CPG2
O
H
HO N O
HO
O l
HO O N
l
N
l
ADEPT Prodrug ZD2767P l
Active Agent:
Di-iodophenyl mustard
SCHEME 7.5 The principle of ADEPT, showing the conversion of the di-iodomustard pro-
drug ZD2767P, presently in Phase I clinical trials, into the active di-iodophenyl mustard at
the tumor site by the Ab-CPG2 conjugate.
COMET assay, and the extent of genomic damage at any time can be defined for a
population of tumor or normal cells. Experiments with the ZD2767/ADEPT system
in tissue culture showed that cross-links were produced in colon carcinoma cells in
a dose-dependent fashion that related to the degree of cell kill. However, the cross-
links were repaired within 24 hours in a sufficient proportion of cells, suggesting a
high probability of tumor regrowth in the clinic. This finding was supported by
similar findings in nude mice bearing xenografts of human colon carcinoma. There-
fore, a third clinical trial utilizing ZD2767 focused on studying DNA cross-links.
Tumor biopsies and peripheral blood lymphocytes were obtained on a small number
of patients for COMET assay. One patient with evidence of tumor response had
extensive cross-linking in tumor cells but no increase over controls in lymphocytes.
However, a patient with no evidence of response had no significant cross-links in
either his or her tumor or lymphocyte cells. The fact that tumor progression followed
in the patient with evidence of response is consistent with human tumor xenograft
model findings of repair of the cross-links induced by ZD2767. Therefore, research
is currently underway to develop novel nonmustard prodrugs suitable for ADEPT
that produce DNA adducts resistant to repair.
but the activated agent must be able diffuse back across the cellular membrane in
order to elicit a BE. This is not a requirement when the enzyme is expressed on the
outer cell surface because it can activate the prodrug extracellularly. Although the
latter mechanism can enhance a BE, the disadvantage is that activated drug can
potentially leak back into the general circulation and lead to toxic effects, thus
lowering the therapeutic index.
The prodrugs used for GDEPT and VDEPT should ideally possess such char-
acteristics as limited cytotoxicity until activated, good chemical stability under
physiological conditions, a favorable ADME profile, the ability to kill both prolif-
erative and quiescent cells after activation, and the capacity to induce a BE. Also,
for intracellular activation, the prodrugs should have sufficient lipophilicity to pen-
etrate cell membranes or otherwise utilize an active transport mechanism. Many
prodrugs in current use, such as the nucleoside analogs (e.g., 5-FC, CP, and CMDA)
and the alkylating agent CB-1954, penetrate cells by passive diffusion.
The precise mechanism of activation of prodrugs is also important. The prodrugs
used in GDEPT are commonly based on antimetabolites that require cycling cells
(e.g., in S phase) for cytotoxicity and are not active in quiescent cells. Therefore, it
has been suggested that alkylating prodrugs may have an advantage over, for exam-
ple, purine nucleosides or 5-FC in that they are cytotoxic to noncycling as well as
proliferative cells. In non-self-immolative prodrugs, the active agent is formed
directly following a one-step process. Most prodrugs used in clinical trials to date
have been based on licensed anticancer drugs whose characteristics are already well
known. Examples of activation mechanisms exploited include scission reactions
(e.g., CPG2, CE, CPA, PNP, PGA, MDAE, TP, β-GAL, β-L, and β-Glu), reductions
(e.g., nitroreductase [NR] and DT-diaphorase), phosphorylation (e.g., HSV-TK,
VZV-TK, and dCK), hydroxylation (e.g., CYP4B1), functional group substitution
(e.g., NH2 to OH; CD), deoxyribosylation (e.g., XGPRT), or oxidation (e.g., DAAO).
A challenge related to the choice of prodrug is that the enzymes used in GDEPT
impose rigid structural requirements on the prodrug substrates, which limits the
choice of anticancer drugs employed One approach to this problem is to design a
self-immolative prodrug that, when activated, forms an unstable intermediate that
extrudes the active agent through a series of subsequent degradative steps. For self-
immolative prodrugs, the initial activation step is generally enzymatic in nature and
distinct from the extrusion step, which is usually chemical and relies on spontaneous
fragmentation. Examples of self-immolative mechanisms include 1,4- or 1,6-elimi-
nations or cyclizations. A major advantage of this approach is that the structure of
the active drug is independent of the substrate requirements of the enzyme being
used in the system. Therefore, a broad range of drugs of various structural classes
can be converted to self-immolative prodrugs.
It is also an advantage if the expressed enzyme can activate the prodrug directly
without multiple catalytic steps because the requisite host endogenous tumor
enzymes can become defective or deficient in some tumor cells. For example,
alkylating agent prodrugs such as CP and CB-1954 (5-[aziridin-1-yl]-2,4-dini-
trobenzamide) have an activation cascade that depends on endogenous enzymes. In
the latter case, nitroreductase (NR) activates CB-1954 to the 5-aziridinyl-4-hydrox-
ylamino-2-nitrobenzamide intermediate, which is followed by a further enzymatic
N N N
CONH2 2
CONH2 CONH2
CB-1954
SCHEME 7.6 Activation of prodrug CB-1954 by rat DT diaphorase [DT (rat)] or E. coli NR
(NR [E. coli]).
step that converts it to a powerful electrophile that alkylates DNA. An optimal half-
life for the active agent produced is also critical; it should be short enough to prevent
drug leakage from the tumor site but long enough to allow a local BE.
A recent GDEPT clinical trial involving NR to activate the prodrug CB-1954
used a replication-deficient adenovirus vector expressing NR from the CMV pro-
moter (CTL102). Patients with operable primary or metastatic liver tumors were
treated with increasing doses of CTL102 given as a single intratumor injection prior
to radical surgery. This trial demonstrated the tolerability of the vector and the ability
of CTL102 to induce a dose-dependent NR expression in the tumor. At the highest
dose of 5 × 1011 virus particles, NR expression occurred in up to 50% of the tumor
cells and was detectable in more than 50% of the specimen slides.
A detailed knowledge of the relevant enzymology is crucial when developing
X-DEPT approaches. For example, it is known that reduction of the prodrug CB-
1954 by rat DT-diaphorase results in conversion of the 4-nitro group to a hydroxyl-
amine moiety, leading to an agent capable of cross-linking DNA. Human DT-
diaphorase can also perform this reduction but at a slower rate, which explains why
degrees of activation are reported to be up to 10,000-fold in rodent cell lines, whereas
an activation of only 670-fold is observed in human cell lines. This finding probably
explains, in part, why CB-1954 is devoid of antitumor activity in humans. Interest-
ingly, Escherichia coli NR is able to reduce both the 2- and 4-nitro groups efficiently
and thus provides a good choice of enzyme for GDEPT studies (Scheme 7.6).
Another recent example of GDEPT is a clinical trial involving prostate cancer,
in which researchers are using CTL102 delivered via multiple intraprostatic injec-
tions in combination with intravenous CB-1954. These trials have demonstrated the
safety of the treatment and have provided preliminary evidence of biological activity.
7.4.3 PDEPT
PDEPT is an experimental two-phase antitumor strategy that employs a combination
of polymer-prodrug and polymer-enzyme conjugates to generate a cytotoxic species
selectively within the tumor mass. The treatment is based on the principle that
polymeric materials are known to accumulate in tumors due to their poor vasculature
(the so-called enhanced permeation and retention (EPR) effect).
family, and the second challenge is to design prodrugs that are not toxic to normal
tissues but that can be converted to cytotoxic species by the relevant enzyme.
In terms of the choice of enzyme, recent attention has focused on the cytochrome
P450 family which, in addition to endogenous substances, metabolizes many drugs
and other xenobiotics (i.e., foreign) substances. The P450s are mixed function
oxidases displaying selective aromatic hydroxylase activity that can be exploited by
medicinal chemists to activate prodrugs. One particular family member, CYP1B1,
is thought to be overexpressed in a wide variety of human tumors but not in normal
tissues, and so is presently being studied more than any other family members in
the context of drug activation.
It is worth noting that subtle genetic modifications (i.e., base pair changes) in
the gene coding for the P450 drug metabolizing enzymes are often used to explain
differences between the response of individuals to cancer chemotherapy agents.
Thus, it is thought that relatively modest genetic changes can have an impact on the
pharmacodynamic and pharmacokinetic characteristics of any anticancer drug and
may have an especially significant effect on those agents with narrow therapeutic
indices. The detection of variations in the P450 genes is therefore growing in
importance as a means to select the most beneficial anticancer drugs for an individual,
and the most appropriate doses and dose schedules to use. This approach has led to
the identification of many different types of P450 enzymes, such as those in the
CYP2C and CYP3A families, as well as CYP1B1 and CYP2D6. In particular, studies
on the CYP2C and CYP3A families have shown that polymorphisms (i.e., subtle
changes of base-pair sequence) in individual genes can lead to significant changes
in the cytotoxic potential of certain drugs. In addition, the etiology of a range of
human cancers is now thought to be related to polymorphisms in the CYP1B1 gene.
Prodrugs based on this approach are still at the discovery stage and so are only
just beginning to reach the clinical evaluation. A number of biotech companies have
been set up to exploit this new therapeutic strategy, and so the structures of novel
prodrugs are not always freely available. However, prodrugs based on the duocar-
mycin DNA minor-groove alkylating agents are in development that require hydrox-
ylation in order to become DNA reactive. Similarly, Phortress, a novel agent that
has just reached Phase I trials, is activated by the CYP family of enzymes to give
an intermediate that spontaneously fragments into DNA-damaging species.
The major criticism of this therapeutic approach is that, given the heterogeneity
of tumors, it is highly unlikely that all tumor cells in a tumor mass will robustly
overexpress the chosen enzyme and will therefore succumb to the drug released.
However, this problem may be somewhat alleviated by a bystander effect. A second
criticism is that it is unlikely that the chosen enzyme would not be expressed
elsewhere in the body. Therefore, taken together, these points suggest that the
therapeutic index of such a therapy is likely to be narrower in practice than expected.
In preclinical studies, the presence of NRH has been shown to provide a very
large (i.e., 100- to 3000-fold) increase in the cytotoxicity of CB1954 in both non-
transfected human tumor cell lines and NQO2-transfected rodent cells. Other
reduced pyridinium compounds have been shown to serve as cosubstrates for NQO2,
and Knox and co-workers have identified key SAR features through a medicinal
chemistry program. One derivative in particular, 1-carbamoylmethyl-3-carbamoyl-
1,4-dihydropyridine (Structure 7.6), has been identified as a good cosubstrate for
NQO2 but with several key advantages over NRH, such as greater stability and
cellular penetration properties, along with the ability to potentiate the cytotoxicity
of CB1954.
CH2 CONH2
CONH2
7.5.3 ASPARAGINASE
Asparaginase (Cristanaspase™; Erwinase™) therapy takes advantage of the fact that
certain types of tumor cells (in particular lymphoblastic leukemia cells) are unable
to synthesize their own asparagine. Systemic administration of the enzyme aspara-
ginase causes a significant reduction in systemic concentrations of asparagine, thus
starving the cancer cells of this nutrient and leading to cell death while healthy cells
continue to synthesize their own. This process is described in detail in Chapter 8
(Biological Agents).
OCH3
O
O
CH3
CH3
NaO2C(CH2)2 N CH3 H3C N O H3C N (CH2)2CO2Na
NH HN H NH HN H H NH HN
CO(CH3)2
H3C O C CH3
N N H2 N
CH3 CH3
H
R= HO CH and/or C CH3 n = 0-6
H
CH3
The cytotoxicity and consequent antitumor effect of porfimer sodium are light
and oxygen dependent. After intravenous injection, the agent clears from most tissues
in 40 to 72 hours. However, organs of the reticuloendothelial system (including the
liver and spleen), the skin and, most importantly, tumor tissue, retain the agent for
longer periods of time. Therefore, selectivity occurs through a combination of
preferential uptake and retention of the agent by the tumor and accurate delivery of
the laser light, which induces a photochemical rather than a thermal effect.
At the molecular level, propagation of radical reactions, initiated after porfimer
sodium absorbs light to form singlet oxygen, is thought to be responsible for the
cellular damage caused by this agent. Hydroxyl and superoxide radicals, lethal to
cells, are thought to form during subsequent radical reactions. Ischemic necrosis
secondary to vascular occlusion, thought to be partly mediated through thromboxane
A2 release, can also cause tumor shrinkage. From a patient-care perspective, it is
important to note that the necrotic reaction and associated inflammatory responses
may develop over several days.
Side effects of porfimer sodium include severe photosensitivity, with sunscreens
offering no protection. Although some individuals are photosensitive to porfimer
sodium for 90 days or more after administration due to residual drug in all areas of
the skin, in general, patients are advised to avoid bright indoor light or direct sunlight
for at least 30 days after treatment. Therefore, careful monitoring of treated patients
is required, and it is important to expose small test areas of skin to sunlight imme-
diately after treatment to assess the degree of photosensitivity likely to occur. Con-
stipation can also be a problematic but treatable side effect.
7.6.2 TEMOPORFIN
Temoporfin (Foscan™) is a second-generation photosensitizing agent for PDT intro-
duced in the mid- to late-1990s (Structure 7.9). The main advantage of temoporfin
over porfimer sodium is that it is a discrete chemical moiety rather than a polymeric
mixture. It is licensed for the PDT of advanced head and neck cancer.
Temoporfin is administered by intravenous injection over at least 6 minutes. Side
effects include photosensitivity (as with porfimer sodium, sunscreens offer no pro-
tection), and so exposure of eyes and skin to bright indoor light or direct sunlight
should be avoided for at least 15 days after treatment. Other side effects include
constipation, local hemorrhage, facial pain and edema, dysphagia, and possible
scarring near the treatment site. Temoporfin is contraindicated in patients with
porphyria or other diseases exacerbated by light. In addition, ophthalmic slit-lamp
examination cannot be carried out for 30 days after administration.
OH
HO
N HN
NH N
OH
HO
thermalized at depth in the tissue and are captured by the 10B atoms, with the resultant
reaction producing α-particles (4He) and lithium-7 (7Li) ions that destroy the tumor
tissue:
These particles are energetic but have a limited range of less than 9 μm in tissue,
thereby preferentially targeting the radiation to the tumor while sparing surrounding
tissues. They also have high linear energy transfer (LET) characteristics compared
to X-rays and gamma rays and, as a consequence, the level of tissue oxygenation
is less important, which is an additional advantage if the tumors contain hypoxic
cells.
A major advantage of BNCT over conventional radiotherapy is that it enables
relatively large volumes of normal tissue to be irradiated with a reduced risk of
adverse effects. This is because the presence of a neutron capture agent ensures that
the radiation dose to the tumor is significantly higher than that to the surrounding
normal tissue, despite the fact that the delivery of low energy neutrons cannot be
focused. However, this advantage is dependent upon successful selective uptake of
the neutron capture agent by the tumor. A major disadvantage is the lack of avail-
ability of expensive nuclear reactor or linear accelerator facilities (required to pro-
duce neutron beams) in most hospitals, and a paucity of effective BNCT delivery
agents.
Experimental and clinical studies relating to the use of BNCT have largely, but
not exclusively, focused on brain tumors— glioblastomas, in particular. High-grade
gliomas are frequently inoperable and, without treatment, can prove fatal within a
few months of diagnosis, with death being due to extensive proximal disease and
local metastatic spread. Gliomas rarely metastasize via the blood and are only
occasionally dispersed along cerebrospinal fluid pathways. Therefore, a treatment
capable of controlling the primary disease and local metastatic spread could poten-
tially result in patient cures. Unfortunately, high-grade gliomas do not respond well
to conventional radiation or traditional chemotherapeutic agents. Enhanced radio-
therapy methods, including hypoxic cell sensitizers and fast neutrons, have also
proved ineffective. Therefore, BNCT has the potential to address this challenging
clinical problem provided that suitable 10B delivery agents can be found, which is
an active area of research.
A number of boron delivery agents have either been studied or are in develop-
ment. For example, borocaptate sodium (Na2B12H11SH) has been shown to be effec-
tive in limited clinical trials in Japan (but in fewer than 150 patients). In these trials,
patients were irradiated with thermal neutrons after the administration of BSH and,
in those with relatively small superficial (less than 6-cm deep), high-grade brain
tumors, 5-year survival rates approached 60%. This compared well with a 5-year
survival rate of less than 3% in patients with comparable tumors treated with
conventional radiotherapy. However, the use of thermal neutrons with limited pen-
etration characteristics in tissue was a major limitation of this Japanese study.
More recently, much effort has focused on the development of more-deeply
penetrating epithermal neutron beams. At present, nuclear reactor based epithermal
beam facilities are available at the Brookhaven National Laboratory (BNL) in the
U.S., at the European Joint Research Center (Petten) in the Netherlands, and at the
University of Helsinki in Finland. In addition, the development of a new British
epithermal beam facility has been funded at the University of Birmingham. This
facility is unique in that it is based on a linear accelerator.
OH
B
HO NH2
OH
Clinical BNCT studies using an epithermal beam have been undertaken at BNL
using the delivery agent p-boronophenylalanine (BPA) dissolved in a fructose solu-
tion and administered intravenously (Structure 7.10). Preliminary results have been
encouraging in terms of tumor responses observed, and no evidence of radioinduced
brain necrosis has been observed.
BNCT also has the potential to treat tumors other than those of the central
nervous system. For example, clinical trials in Japan involving the treatment of
malignant melanoma using BPA as the neutron capture agent have provided encour-
aging results. The development of improved neutron capture agents could potentially
micelles have a spherical, core-shell structure, with the hydrophobic block forming
the core of the micelle and the hydrophilic block or (blocks) forming the shell. They
have promising properties as drug carriers both in terms of their size and architecture.
The hydrophobic drug molecules partition inside the micelles, which are 15 to 35 nm
in diameter. It is thought that these structures enter cells by phagocytosis or endo-
cytosis. The drug is thus delivered inside the cells by local delivery or by fusion of
the particle with the cell membrane, which destabilizes the micelle.
There is now significant ongoing research into new types of nanoparticles and
the nanoencapsulation of drugs for targeted delivery to tumors of various organs
both as a single therapy and in combination with other treatments, such as radio-
therapy. In particular, there is growing interest in attaching entities such as antibody
fragments to the surface of drug-containing nanoparticles in order to guide them
specifically to a tumor, where they can release the active agent. Nanoparticles are
also being used in gene therapy strategies as previously described. Finally, nano-
technology-based diagnostics can be combined with therapeutics, which is likely to
be important in the future for the growing emphasis on the personalized management
of cancer.
diblock copolymers, mixtures of these, and mixtures of such polymers with PEGy-
lated diacylphospholipids.
Ultrasound has been used extensively for both medical diagnostics and physical
therapy, and is widely regarded to be safe when exposed to healthy tissues. Advan-
tages of ultrasound include the fact that it is noninvasive, and that its energy can be
controlled and focused easily with a capability of penetrating deep into tissues to
predetermined depths. In particular, large amounts of ultrasonic energy can be
accurately deposited deep into tumors. The depth of penetration and the shape of
the energy deposition pattern may be controlled by varying the ultrasound frequency
and the type and shape of the transducer. Optimal power densities at the target site
may be obtained by adjusting the output power of the ultrasonic transducer.
Two possible mechanisms for the ultrasound enhancement of chemotherapy have
been proposed. The first relates to enhanced drug release from micelles (as previously
described). The other is associated with the apparent enhanced uptake of the micellar-
encapsulated drug by cells. For example, there are literature reports of ultrasound-
induced hypersensitization of anthracycline-sensitive cell lines, which would appear
to support the enhanced drug-uptake mechanism. However, other experiments on
the effect of low-frequency ultrasound pulse duration on drug uptake suggest that
both mechanisms may work in concert.
Both low-frequency and high-frequency ultrasound have proved effective in
triggering drug release from micelles. Low-frequency ultrasound is more effective
but does not allow sharp focusing. In contrast, high-frequency ultrasound allows
sharp focusing but does not penetrate as deeply into the interior of the body. There-
fore, optimal design of ultrasound treatment will depend on tumor size and location.
For example, for tumors 2 cm in diameter or larger, application of 100 kHz (or a
lower frequency) ultrasound is feasible, with optimal power densities achieved by
controlling output energy. For smaller tumors that are not so deep, a higher-frequency
ultrasound can be used because it provides sharper focusing. In addition, power
densities produced at the focal site by existing hyperthermia devices appear sufficient
to cause drug release from micelles. Importantly, in vivo experiments have demon-
strated improved survival rates for ovarian carcinoma–bearing mice treated with 3
mg/kg of doxorubicin in PLURONIC micelles delivered intraperitoneally in com-
bination with ultrasound (1 MHz, 30 s, 1.2 W/cm2) applied 1 hour after injection
of the drug. In the same experiment, no effect was observed in untreated control
mice or mice treated with 3 mg/kg of doxorubicin administered intraperitoneally in
a physiological solution.
The current findings suggest that ultrasound targeting is a promising new area
of research, particularly as high-frequency ultrasound is widely used in clinical
practice for imaging purposes (though at much lower power densities than used in
these experiments). The ideal scenario would be to ultimately combine imaging and
therapeutic ultrasound transducer arrays in one instrument that could be used initially
to image the tumor but, followed by automatic focusing of the ultrasound beam for
therapeutic purposes. Finally, the combination of micellar drug delivery carriers and
ultrasound is especially promising for treating MDR-positive tumors that do not
respond to conventional treatment regimens.
FURTHER READING
Algire, G.H., and Chalkley, H.W. “Vascular Reactions of Normal and Malignant Tissue In
Vivo,” J. Nat. Cancer Inst., 6:73-85, 1945.
Denekamp, J. “Endothelial Cell Proliferation as a Novel Approach to Targeting Tumour
Therapy,” Br. J. Cancer, 45:136-139, 1982.
Folkman, J. “Tumor Angiogenesis: Therapeutic Implications,” NEJM, 285:1182-86, 1971.
Folkman, J. “Angiogenesis in Cancer, Vascular, Rheumatoid, and Other Disease,” Nat. Med.,
1:27-31, 1996.
Hahnfeldt, P., et al. “Tumor Development under Angiogenic Signaling: A Dynamical Theory
of Tumor Growth, Treatment Response, and Postvascular Dormancy,” Cancer Res.,
59:4770-75, 1999.
Hua, J., et al. “The Role of New Agents in the Treatment of Colorectal Cancer,” Oncology,
66(1):1-17, 2004.
Hurwitz, H., et al. “Bevacizumab Plus Irinotecan, Fluorouracil, and Leucovorin for Metastatic
Colorectal Cancer,” NEJM, 350:2335-42, 2004.
Presta, L.G., et al. “Humanization of an Anti-Vascular Endothelial Growth Factor Monoclonal
Antibody for the Therapy of Solid Tumors and Other Disorders,” Cancer Res.,
57:4593-99, 1997.
Saaristo, A., et al. “Mechanisms of Angiogenesis and Their Use in the Inhibition of Tumor
Growth and Metastasis,” Oncogene, 19:6122–29, 2000.
Salgaller, M.L. “Technology Evaluation: Bevacizumab, Genentech/Roche,” Curr. Opinion
Mol. Ther., 5(6):657-67, 2003.
Sheng, Y., et al. “Combretastatin Family Member OXI4503 Induces Tumor Vascular Collapse
through the Induction of Endothelial Apoptosis,” Int. J. Cancer, 111:604-10, 2004.