Molecular Biology of Cancer Translation to the Clinic 1st Edition Raymond W. Ruddon (Eds.) 2024 scribd download
Molecular Biology of Cancer Translation to the Clinic 1st Edition Raymond W. Ruddon (Eds.) 2024 scribd download
Molecular Biology of Cancer Translation to the Clinic 1st Edition Raymond W. Ruddon (Eds.) 2024 scribd download
com
https://ebookfinal.com/download/molecular-biology-of-cancer-
translation-to-the-clinic-1st-edition-raymond-w-ruddon-eds/
OR CLICK BUTTON
DOWNLOAD EBOOK
https://ebookfinal.com/download/cancer-biology-4th-edition-raymond-w-
ruddon-m-d/
ebookfinal.com
https://ebookfinal.com/download/skin-cancer-the-biology-of-cancer-1st-
edition-po-lin-so/
ebookfinal.com
https://ebookfinal.com/download/prevention-of-cancer-the-biology-of-
cancer-1st-edition-robert-g-mckinnell/
ebookfinal.com
https://ebookfinal.com/download/international-review-of-cell-and-
molecular-biology-1st-edition-kwang-w-jeon-eds/
ebookfinal.com
International Review of Cell and Molecular Biology 280 1st
Edition Kwang W. Jeon (Eds.)
https://ebookfinal.com/download/international-review-of-cell-and-
molecular-biology-280-1st-edition-kwang-w-jeon-eds/
ebookfinal.com
https://ebookfinal.com/download/the-biology-of-cancer-1st-edition-
robert-a-weinberg/
ebookfinal.com
https://ebookfinal.com/download/international-review-of-cell-and-
molecular-biology-vol-285-1st-edition-kwang-w-jeon-ed/
ebookfinal.com
https://ebookfinal.com/download/never-fear-cancer-again-the-
revolutionary-holistic-solution-to-turn-off-cancer-cells-never-be-
raymond-francis/
ebookfinal.com
https://ebookfinal.com/download/the-biology-of-cancer-2nd-edition-
janice-ann-gabriel/
ebookfinal.com
PROGRESS IN
Molecular Biology
and Translational Science
Volume 95
This page intentionally left blank
PROGRESS IN
Volume 95
No part of this publication may be reproduced, stored in a retrieval system or transmitted in any
form or by any means electronic, mechanical, photocopying, recording or otherwise without the
prior written permission of the Publisher
Permissions may be sought directly from Elsevier’s Science & Technology Rights
Department in Oxford, UK: phone (+44) (0) 1865 843830; fax (+44) (0) 1865 853333;
email: [email protected]. Alternatively you can submit your request online by
visiting the Elsevier web site at http://elsevier.com/locate/permissions, and selecting
Obtaining permission to use Elsevier material
Notice
No responsibility is assumed by the publisher for any injury and/or damage to persons or
property as a matter of products liability, negligence or otherwise, or from any use or operation
of any methods, products, instructions or ideas contained in the material herein. Because of rapid
advances in the medical sciences, in particular, independent verification of diagnoses and drug
dosages should be made
Library of Congress Cataloging-in-Publication Data
A catalog record for this book is available from the Library of Congress
British Library Cataloguing in Publication Data
A catalogue record for this book is available from the British Library
ISBN: 978-0-12-385071-3
ISSN: 1877-1173
Contributors..................................................................................... ix
Preface............................................................................................. xi
v
vi contents
Chromosomal Aberrations in Solid Tumors . . . . . . . . . . 55
Arul M. Chinnaiyan and Nallasivam Palanisamy
I. Introduction .............................................................................. 56
II. Historical Background: Discovery of Chromosome Aberrations
in Cancer .................................................................................. 57
III. Discovery of Gene Fusions in Cancer............................................. 58
IV. New Approaches for Gene Fusion Identification .............................. 61
V. Methods for the Characterization of Chromosome Aberrations
in Solid Tumors.......................................................................... 70
VI. Next-Generation Sequencing Technology........................................ 79
VII. Structural Classification of Gene Fusions ........................................ 82
VIII. Functional Classification of Gene Fusions ....................................... 83
IX. Mechanism of the Formation of Gene Fusions in Cancer................... 84
References ................................................................................ 86
Numbers in parentheses indicate the pages on which the authors’ contributions begin.
ix
x contributors
The purpose of this volume in the Progress in Molecular Biology and Transla-
tional Science series is to explore some of the most exciting recent advances in
basic research on the molecular biology of cancer and how this knowledge leads
to advances in the diagnosis, treatment, and prevention of cancer.
The chapter topics include introduction to the molecular biology of cancer
(Ruddon), molecularly targeted approaches to the development of anticancer
drugs (Lazo), targeting chemokine ligands and their role in cancer metastasis
(Pienta), discovery of cancer cell fusion genes in solid cancers (Chinnaiyan),
role of circulating tumor cells in cancer diagnosis, disease progression and
response to therapy (Hayes), cancer stem cells (WIcha), bioinformatics and
systems biology of cancers(Omenn), progress in cancer nanotechnology
(Baker), molecular imaging (Ross), cancer epigenetics (Weber), and cancer
prevention (Brenner).
The senior investigators represented here are all University of Michigan
faculty, with the exception of John Lazo, from the University of Pittsburgh, but
even he has a U of M genealogy, having received his Ph.D. in Pharmacology
from the Ruddon lab at U of M. However, all of these scientists are leaders in
their fields of research. Thus, this is not just a parochial concatenation of
narrowly focused local research, but it is, in fact, representative of the most
advanced research in the fields discussed here.
xi
This page intentionally left blank
Introduction to the Molecular
Biology of Cancer: Translation
to the Clinic
Raymond W. Ruddon, M.D., Ph.D.
Department of Pharmacology, University of
Michigan Medical School, Ann Arbor,
Michigan, USA
Advances in molecular biology over the last several decades are being steadily
applied to our understanding of the molecular biology of cancer, and these
advances in knowledge are being translated into the clinical practice of oncology.
Many examples can be cited to demonstrate this. Some of them are listed
below. Everyone has their favorite list, of course; however, everyone would
probably agree that the items on this list should be included in any such list of
advances.1
In spite of the fact that remarkable progress has been made over the past
few decades in the understanding of the molecular, cellular, and tissue process-
es involved in precancer and during cancer progression, the development of
effective and safe modalities for prevention of cancer remains slow, inefficient,
and expensive. One of the problems is that treatment with cancer preventative
agents usually means that individuals need to take a chemopreventive agent for
many years, if not for a lifetime. This means that such treatments must be
extremely nontoxic and safe. Secondly, pharmaceutical companies are loath to
fund such clinical trials because the trials must go on for years if not decades
and involve very large numbers of individuals. All this costs a lot of money. Of
course, initially such clinical trials can be focused on high-risk individuals,
which would cut down on the scale of the trial. Also, if accurate surrogate
markers for effectiveness could be developed, the amount of time and number
of patients could be reduced significantly.20
Similarly, the application of epidemiology to cancer prevention can provide
significant information on cancer risk and the usefulness of preventive strate-
gies.21 For example, the elucidation of clues to cancer causation flows from
observed associations of population exposures to tobacco, diet, environmental
chemicals, and other exogenous factors with the development of cancer in
patients. Indeed, the one real success story for cancer prevention has come
about through smoking cessation.
Nevertheless, everyone wants the ‘‘magic pill’’ that will allow them to keep
their life styles and at the same time enable them to avoid getting cancer. As
numerous studies have shown, alterations in diet or ingestion of mega doses of
antioxidants and vitamins have so far proved to be of little avail. As better
molecular models of cancer initiation and promotion become available, the
hope is that effective chemopreventive agents may be developed. The history
of cancer prevention for the past 100 years has been recently reviewed.22
Current clinical data for ‘‘cancer risk reductive interventions’’ (also previously
known as ‘‘chemoprevention’’) are discussed by Kakarala and Brenner in
Chapter 11.
References
1. Ruddon RW. Cancer Biology. 4th ed. London: Oxford University Press; 2007. p. 1–16.
2. Fearon ER, Vogelstein BA. A genetic model for colorectal tumorigenesis. Cell 1990;61:759–67.
3. Hoeı̈jmakers JHJ. Genome maintenance mechanisms for preventing cancer. Nature
2001;411:366–74.
4. Ruddon RW. Cancer Biology. 4th ed. London: Oxford University Press; 2007. p. 321–352.
5. Ruddon RW. Cancer Biology. 4th ed. London: Oxford University Press; 2007. p. 352–367.
8 RAYMOND W. RUDDON
6. Friend SH, Bernards R, Rogelj S, Weinberg RA, Rapaport JM, Albert DM, et al. A human DNA
segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma.
Nature 1986;323:643–6.
7. Ma WW, Adjei AA. Novel agents on the horizon for cancer therapy. CA Cancer J Clin
2009;59:111–37.
8. DeVita Jr. VT, Chu E. A history of cancer chemotherapy. Cancer Res 2008;68:8643–53.
9. Hambley TW. Is anticancer drug development heading in the right direction? Cancer Res
2009;69:1259–62.
10. Hait WN. Targeted cancer therapeutics. Cancer Res 2009;69:1263–7.
11. Li X, Loberg R, Liao J, Ying C, Snyder LA, Pienta KJ, et al. A destructive cascade mediated by
CCL2 facilitates prostate cancer growth in bone. Cancer Res 2009;69:1685–92.
12. Maher CA, Kumar-Sinha C, Cao X, Kalyana-Sundaram S, Han B, Jing X, et al. Transcriptome
sequencing to detect gene fusions in cancer. Nature 2009;458:97–101.
13. Eliane J-P, Repollet M, Luker KE, Brown M, Rae JM, Dontu G, et al. Monitoring serial
changes in circulating human breast cancer cells in murine xenograft models. Cancer Res
2008;68:5529–32.
14. Hayes DF, Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Miller MC, et al. Circulating tumor
cells at each follow-up time point during therapy of metastatic breast cancer patients predict
progression-free and overall survival. Clin Cancer Res 2006;12:4218–24.
15. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identifica-
tion of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 2003;100:3983–8.
16. Ruddon RW. Cancer Biology. 4th ed. London: Oxford University Press; 2007. p. 376–379.
17. Nicholson JK, Lindon JC. Metabonomics. Nature 2008;455:1054–6.
18. Sreekumar A, Poisson LM, Rajendiran TM, Khan AP, Cao Q, Yu J, et al. Metabolomic profiles
delineate potential role for sarcosine in prostate cancer progression. Nature 2009;457:910–4.
19. Quintana A, Raczka E, Piehler L, Lee I, Mhyc A, Majoros I, et al. Design and function of a
dendrimer-based therapeutic nano-device targeted to tumor cells through the folate receptor.
Pharmacol Res 2002;19:1310–6.
20. Kelloff GJ, Bast Jr. RC, Coffey DS, D’Amico AV, Kerbel RS, Park JW, et al. Biomarkers,
surrogate end points, and the acceleration of drug development for cancer prevention and
treatment: an update. Clin Cancer Res 2004;10:3881–4.
21. Greenwald P, Dunn BK. Landmarks in the history of cancer epidemiology. Cancer Res
2009;69:2151–62.
22. Lippman SM, Hawk ET. Cancer prevention: from 1727 to milestones of the past 100 years.
Cancer Res 2009;69:5269–84.
Molecular Biology and
Anticancer Drug Discovery
John S. Lazo
Department of Pharmacology and Chemical
Biology, University of Pittsburgh Drug
Discovery Institute and Cancer Institute,
University of Pittsburgh, Pittsburgh,
Pennsylvania, USA
I. Introduction .................................................................................. 9
II. Phenotypic Targets .......................................................................... 12
III. Molecular Targets ........................................................................... 16
A. General Issues ........................................................................... 16
B. Drug Resistance......................................................................... 17
C. Oncogene and Nononcogene Addiction ........................................... 18
D. Synthetic Lethal ......................................................................... 20
E. Combination Chemotherapy ......................................................... 23
F. Nontraditional Targets ................................................................. 23
IV. Other Contemporary Issues in Anticancer Drug Discovery ....................... 26
V. Conclusions ................................................................................... 27
References .................................................................................... 27
I. Introduction
Contemporary cancer drug discovery largely focuses on identifying new
therapies by leveraging advances in our understanding of the molecular biology
of cancer. This has been facilitated by the enormous increase in our knowledge
concerning the molecular pathogenesis of cancer in the past two decades, which
was stimulated by the profound investment in research funding from both private
and public sources. In addition to great improvements in diagnostics and imaging,
there are now more than 200 approved drugs for cancer; hundreds are in clinical
development. Nonetheless, our ability to control, much less cure, cancer has
been disappointing. Oncology has among the worst success rates (5%) for clinical
drug development compared to all other major disease sectors.1 Drug discovery
and development in cancer remains a challenging, high-risk, inefficient, and
complex process. Why is that so?
There are several reasons for the poor success in anticancer drug discovery
and development. First, as mentioned in Chapter 1, tumors evolve through a
multistep process in which cancer cells frequently acquire a large number of low-
frequency genetic or epigenetic changes that can contribute to oncogenesis.
These mutations affect the expression or functionality of oncogenes or tumor
suppressors. The substantial number of alterations makes it challenging to distin-
guish the critical changes that are worthy of being targeted with drugs from the
nonessential ones or ‘‘passengers’’ that are a legacy of the disease process. Some
changes may have been important for the initial stages of oncogenesis but now are
completely dispensable for the oncogenic phenotype and, therefore, unworthy of
being drug targets. A second problem is that many of the oncogenes represent
overexpressed versions of normal proteins or they contain mutations that are only
subtly different from the normal gene product. Thus, selective targeting of cancer
cells may be considered problematic. Generally, drugs affect gain-of-function
abnormalities, making it at least conceptually challenging to design drugs that
will replace the loss of a tumor suppressor. Finally, the fundamental genetic
instability of cancer produces inherent plasticity in the disease that promotes
rapid drug resistance.
There is no doubt that the cancer drug discovery landscape has benefited
from developments in a host of disciplines and technologies. Advances in
organic chemistry have facilitated the complete synthesis of complex natural
products with powerful pharmacological activities.2 Combinatorial chemistry
and diversity-oriented synthesis have expanded greatly the numbers of drug-
like compounds that now can be regularly probed for anticancer activities.
Publically accessible databases of compounds and biological actions, such as
PubChem, PharmGKB, ChemSpider, and ChemExper, provide information on
tens of millions of compounds.3 Advances in automated liquid handling plat-
forms have enabled high throughput screening of hundreds of thousands of
compounds for bioactivity. Similarly, high content platforms have empowered
investigators to directly test compounds for cellular actions. Computational and
structural biology have generated tools for molecular docking and visualization
of drug or ligand interactions with putative targets.4 Innovation in drug delivery
and nanotechnology offer hope for better pharmacokinetics and drug targeting
to tumors.2 The emergence of systems biology as a discipline has created new
ANTI-CANCER DRUG DISCOVERY 11
tools to visualize signaling pathways controlling attributes that are vital for the
neoplastic phenotype. How are these developments in various disciplines
integrated into the process of discovering a new anticancer drug?
Fundamentally, there are two broad approaches used to identify new drugs:
Forward Pharmacology and Reverse Pharmacology.5 These approaches are not
unique to anticancer drug discovery, but there is some merit to the argument that
the field of oncology drug discovery has uniquely benefited from the advances of
molecular biology with the cataloging of vast numbers of oncogenes and tumor
suppressors, which has accelerated the transition from Forward Pharmacology to
Reverse Pharmacology. The oldest strategy, Forward Pharmacology, relies on
observing phenotypic changes of cells, organs, or organisms by chemical sub-
stances. Even before the acquisition of any fundamental understanding of the
regulation of physiological systems or the nature of pathological processes, one
could readily measure the inhibition of cancer cell proliferation in a culture dish.
This molecular target-unbiased approach merely focused on the desired biology
without any knowledge of how the process was regulated. There are at least six
phenotypic attributes associated with tumor cells that can be examined in culture
(Table I). Indeed, a majority of our existing clinically used anticancer drugs were
discovered using a phenotypic assay and with little knowledge of how they might
be selectively toxic to cancer cells. In contrast, Reverse Pharmacology is
practiced with the goal of finding a modulator, usually an inhibitor, of a molecular
target that is believed to be critical for the cancer phenotype.5 Clearly, molecular
biology has profoundly influenced this branch of pharmacology and anticancer
TABLE I
EXAMPLES OF PHENOTYPIC ATTRIBUTES OF CANCER CELLS AND COMPOUNDS THAT ALTER THE PROCESS
drug discovery. Molecular biology and molecular genetics have been critical in
identifying potential causal factors in neoplasia. Once identified, the molecular
target (generally an enzyme) can be produced in a recombinant form permitting
in vitro interrogation for inhibitors. This class of assays has the distinct advantage
of generally being precise, rapid, and easier to conduct than assays requiring
animal, organs, or cells. One can sometimes even determine at an atomic level
the interactions between the compound and the target, making the generation of
a chemical structure–activity relationship practical and the synthesis of more
potent analogs rational. Target specificity and modes of inhibition assays are also
conceptually easier to perform with a known target than with phenotypic assays.
For molecular targeted anticancer drugs, it is desirable that most, if not all, vital
cell types in the human body not depend on the target for survival. Otherwise,
the drug is likely to have a narrow therapeutic window owing to the requirement
for the target in normal cells. Although currently there is enormous enthusiasm
for target-driven anticancer drug discovery, phenotypic screening approaches
are regaining popularity in part because they can be designed to ensure com-
pound entry into cells and stability, the failure of which can lead to the demise of
compounds identified by more reductionist assays.
from primary tumors. After all, most tumors isolated from patients do not yield
cells that can grow productively in culture. Moreover, cell lines adapt distinc-
tive phenotypes after multiple passages in different laboratories. Thus, a HeLa
cell that has been extensively passaged in one laboratory is not necessarily the
same as that studied in another laboratory. Nonetheless, the US National
Cancer Institute has established a large publicly available annotated database,
which can be mined with programs, such as the COMPARE algorithm
(see http://dtp.nci.nih.gov/docs/compare/compare.html), to examine the profile
of drug sensitivity to the panel. Studies using this algorithm have clustered
compounds with common mechanisms of action, identified compounds with
new mechanisms of action, and exposed agents that are substrates for multiple
drug resistance.13 The landmark work of the National Cancer Institute has also
spawned other tumor cell line panels in an effort to develop an ideal tumor cell
screening panel.13
All cell proliferation or cell death assays depend on the assay protocol but
most importantly on the endpoint that is being measured. Some assays simply
measure the total number of cells on the plate after a fixed time point. Others
determine the number of viable cells using, for example, Trypan blue exclusion
or Alamar blue reduction. In most cases, however, there is little distinction
between cell growth inhibition and cell death. There is a growing interest
in developing assays that will differentiate between apoptosis, necrosis, necrop-
tosis, anoikis, autophagy, and other processes associated with cell death.
Obviously, it is desirable to establish preferential toxicity against cancer cells
versus normal cells, but the question of selecting the appropriate normal cell
type for the comparison often is difficult to answer.
Phenotypic cytotoxicity assays clearly are only biological models: the com-
position of the culture medium, the presence of fetal bovine serum, and the
absence of other stromal cells make the environment extremely artificial.
Additionally, the nonphysiological oxygen concentrations and the lack of a
normal extracellular matrix and stromal cells prevent a precise recapitulation
of the environment in which tumor cells find themselves in vivo. The impor-
tance of the tumor microenvironment in response to therapy is well estab-
lished.13 Studies with three-dimensional cultures, tumor spheroid systems, and
mixed stromal cell substrates may provide improved platforms for future drug
testing systems.
Although drugs identified with phenotypic assays or Forward Pharma-
cology are generally regarded in the current parlance as ‘‘cytotoxics’’ and
not targeted, all probably function by influencing a molecular target. Potent
inhibitors of dihydrofolate reductase, topoisomerase I, topoisomerase II, ribo-
nucleotide reductase, microtubule stabilizers, and tubulin disruptors were first
identified as inhibitors of cancer cell proliferation and only later were their
mechanisms of action, namely molecular targets, determined (often with the
14 JOHN S. LAZO
dramatically improved the prognosis for patients with this disease. Exposure of
APL cells in culture to ATRA leads to differentiation into mature neutrophils,
most likely due to the induction of CCAAT/enhancer binding proteins.12 There
is preclinical evidence that methyltransferase inhibitors, such as azacytidine,
and the histone deactylase inhibitor sodium phenylbutrate can also induce
differentiation in APL.17 In addition, arsenic trioxide has been documented
to induce differentiation in APL cells, leading to clinical remissions, although
an incontrovertible mechanism of action is lacking.
In contrast to differentiation, tumor cell senescence is the loss of replicative
ability that is accompanied by cellular enlargement, flattened morphology with
an increased cytoplasmic area, increased granularity, extensive cytoplasmic
vacuoles, and multinucleation. In addition, senescent cells have changes in
gene expression that is largely independent of cell type, including an increase in
the expression senescence-associated b-galactosidase, cyclin D, and cyclin-
dependent kinase inhibitors, p21 and p16. Senescent cells also have a reduction
in the tumor suppressor protein Rb. A surprisingly large number of clinically
used, mechanistically distinct anticancer agents, including DNA alkylators,
topoisomerase poisons, antimetabolites, and microtubule stabilizers, as well
as irradiation, have been reported to induce senescence.10 Typically, senes-
cence is seen with cultured human tumor cells at low drug concentrations,
while apoptosis is observed with higher drug concentrations, presumably
because of the greater cell damage. For example, treatment of human hepato-
ma cells with a low concentration of the indirect DNA-damaging agent doxo-
rubicin (< 100 nM) causes senescence, whereas higher concentrations (20 mM)
produces frank apoptosis. It should be emphasized, however, that the impor-
tance of the phenomenon of senescence for the clinical activity of any drug
remains to be determined.
Interest also exists in identifying drug-like compounds that can disrupt the
essential dialogue between tumor cells and stromal cells, but currently, there
are no clinically used agents that faithfully act in this manner. As will be
mentioned in Chapter 6, there is considerable enthusiasm for identifying
drugs that selectively kill cancer stem cells. Unfortunately, useful in vitro assays
for identifying agents that specifically kill epithelial cancer stem cells have not
been available because of the paucity of these cancer stem cells within tumor
cell populations and their instability in culture. The recent development of an
innovative assay by Gupta et al.11 may have overcome this barrier, however.
Using this assay, Gupta et al.11 discovered several compounds, including sali-
nomycin, with selective toxicity for breast cancer stem cells. Treatment of mice
with salinomycin inhibited mammary tumor growth in vivo and induced
increased epithelial differentiation of tumor cells. Thus, it may be possible to
target what is thought to be a critical cancer cell population normally resistant
to existing therapies.
16 JOHN S. LAZO
drug resistance has enabled the successful design and development of second
generation anticancer drugs that retain efficacy in tumor cells with acquired or
innate resistance to the first generation agents. For example, soratinib is
effective against tumor cells resistant to imatinib. Premetrexate inhibits multi-
ple folate-related targets, some of which are involved in resistance to metho-
trexate. Many of the second generation compounds now have embedded in
them multiple targets, some of which are considered resistance mechanisms or
participants in the oncogenic phenotype.
TABLE II
EXAMPLES OF ONCOGENE AND NONONCOGENE ADDICTION TARGETS AND AGENTS
Target Agent
Oncogene targets
BCR-Abl, c-Kit Imatinib, Dasatinib
Retinoic acid receptor, Retinoic X receptor ATRA (Tretinoin)
ERBB2 Trastuzumab
Epidermal growth factor receptor Erlotinib, Gefitinib
MDM2 Nutlin-3
BCL-2 Oblimersen, ABT-737
PI3K GDC-0941, BEZ235
Nononcogene targets
mTOR Temsirolimus, Everolimus
Topoisomerase I Topotecan, Irinotecan
Dihydrofolate reductase Methotrexate, Pemetrexed
Vascular endothelial growth factor Bevacizumab
PARP1 AZD2281, ABT-888, AG014699
Heat shock protein-90 Geldanamycin, Alvespimycin
Mitotic Spindles Vinblastine, Vincristine, Paclitaxel, Epothilone B
Chk1 PF-00477736
TABLE III
GENETIC SYNTHETIC LETHALITY
Deletion/Mutation Viable
Deletion/Mutation þ Viable
Deletion/Mutation þ Viable
Deletion/Mutation þ þ Death
TABLE IV
MIXED DRUG AND GENETIC SYNTHETIC LETHALITY
Treatment/Deletion/Mutation Viable
Treatment/Deletion/Mutation þ Viable
Treatment/Deletion/Mutation þ Viable
Treatment/Deletion/Mutation þ þ Death
that a large fraction of the remaining genes can collaborate in death process-
es.28,29 Many of the targets for existing cytotoxic agents are essential gene
products such as tubulin or topoisomerase. First principles would teach us
that they are not attractive because they are essential for both normal and
malignant cells; this is why existing compounds have such a narrow therapeutic
index. If the synthetic lethal concept from yeast genetic screens could be
extended to human tumor cells, which have preexisting mutations, deletions,
and epigenetic abnormalities, it might be possible to identify innovative drug
targets. It might even be possible that some of our current cytotoxic drugs are
efficacious because they inadvertently exploit synthetic lethality. In this model,
one of the gene deletions or mutations is replaced with a drug (Table IV).
One attractive example is the sensitivity of cancers carrying BRCA1 mutations,
which is responsible for homologous DNA recombination repair. These tumors
are exceptionally sensitive to DNA-damaging and cross-linking agents such as
temozolomide and cisplatin.6 These tumors are heavily reliant on orthogonal
forms of DNA repair, such base-excision repair that is mediated by poly-ADP-
ribose polumerase (PARP1). PARP1 facilitates repair of single-strand breaks.
In nonmalignant cells, endogenous DNA damage generated by PARP1 inhibi-
tion by agents, such as ABT-888, is well tolerated because of functional
compensation from homologous recombination-mediated repairs. This is an
22 JOHN S. LAZO
TABLE V
DRUG–DRUG SYNTHETIC LETHALITY
Treatment Viable
Treatment þ Viable
Treatment þ Viable
Treatment þ þ Death
E. Combination Chemotherapy
Almost all regimens for the treatment of cancer rely on drug combinations.
They have been created almost exclusively by empirically amalgamating drugs
with different mechanisms of action, untoward effects, and mechanisms of
resistance. Molecular biology has provided new information about the inter-
connections of the anticancer targets that could provide some theoretical
guidance for the formulation of drug combinations. Nonetheless, even with
the introduction of targeted therapies where there is a quantifiable pharmaco-
logical effect, it is most common to develop drug combinations in the clinic by
escalating the individual agents to the maximum tolerated dose until the
aggregate effect of toxicity is considered to be excessive. This approach pre-
sumes, perhaps incorrectly, that the maximum tolerated dose is the maximum
effective dose. There is no incontrovertible clinical evidence to support this
notion. It has been extremely challenging to provide any in vitro guidance for
this important question, especially as we see the use of combinations containing
multiple agents. We know each of the individual drugs is likely to exert multiple
pharmacological effects at different concentrations and each has a unique
pharmacokinetic profile that alters the plasma and tumor concentration over
time. Thus, it is important to address this issue, because there is compelling
evidence that the anticancer effect of drug combinations can be profoundly
dependent on the ratio of the individual drugs.33
F. Nontraditional Targets
As mentioned above, the primary targets for almost all current drugs are the
catalytic site of enzymes or ligand binding sites. Many enzymes, however, depend
on allosteric regulation to be fully active, and there is a growing interest in
disrupting this process. We know that the intracellular spatial, kinetic, and
substrate specificity regulation of enzymes can be controlled by multiple
protein–protein interactions (PPIs). These are widely believed to control all
major cellular functions, including the maintenance of DNA topology, DNA
replication, mRNA transcription, protein translation and its proper folding, the
assembly and maintenance of morphological structures, and the regulation of
cellular metabolism and signaling pathways. The protein interactions comprise
identical or homotypic binding and nonidentical or heterotypic binding between
two or more polypeptides; they can be characterized thermodynamically and
kinetically as anything from high affinity stable contacts to low affinity transient
interactions. Some are critical for functionality and others are gratuitous.
It is easy to imagine how the critical PPIs might provide valuable potential
molecular targets that would be mechanistically distinct from the common active
sites or ligand binding drug discovery targets. While PPIs might superficially
seem attractive, until recently it was widely believed that PPI surfaces were not
24 JOHN S. LAZO
druggable because they were large and amorphous.34 Refined structural eluci-
dation of many protein–protein complexes indicate that the protein-binding
interfaces can be dissected into discrete patches with critical residues, termed
‘‘hot spots,’’ that are vital for binding.34 Biochemical PPI assays exploiting
methods, such as co-precipitation, co-purification, affinity chromatography, ul-
tracentrifugation, nuclear magnetic resonance, surface plasmon resonance, mass
spectrometry, and isothermal titration calorimetry, have been developed.34,35
Additionally, higher throughput biochemical PPI screening assay formats have
been developed, including capture ELISAs, cell surface binding, fluorescence
polarization, time-resolved fluorescence, ligand-induced changes in thermal
stability, bead-based technologies, such as AlphaScreen and Luminex, and reso-
nance energy transfer assays.34,35 Cell-based PPIs assays exploiting two-hybrid
transcriptional reporter systems have also been developed in mammalian cells.
Several imaging-based high content screening assay formats have also been
employed to study PPIs, such as the co-localization of fluorescently labeled
protein partners, fluorescent resonance energy transfer measurements between
PPI partners bearing donor and acceptor fluorescently labeled protein, protein
fragment complementation assays, and positional biosensors that measure the
PPI-induced redistribution of fluorescently labeled protein partners.
Perhaps one of the most successful discovery examples of PPI inhibitors
has been with p53 and MDM2. The p53 tumor suppressor is a transcriptional
activator that regulates the expression of target genes involved in processes that
serve to restrict the initiation, progression, or survival of cancer cells. p53
controls cell cycle arrest, DNA damage repair, apoptosis, senescence, metasta-
sis, and angiogenesis. In more than half of all human cancers, p53 is inactivated
by single-point missense mutation in the DNA-binding domain, resulting in
deficient regulation of p53 target genes. In a significant proportion of the
remaining cancers where wild-type p53 is functional, MDM2 is overexpressed
and blocks the tumor suppressor activity of p53. In this case, MDM2 binds to
the N-terminal transactivation domain of p53, thereby inhibiting activation of
p53 target genes. Because MDM2 contains a E3-ubiquitin-ligase activity, it also
tags p53 for degradation by the proteasome.36 The structure of the protein–
protein binding interfaces between p53 and MDM2 has been comprehensively
mapped and characterized; there are three amino acid residues in the p53 N-
terminus (Phe19, Trp23, and Leu26) that bind to a small hydrophobic pocket
on the surface of MDM2. This protein–protein binding interface was targeted
using a combination of structure-based drug design and an SPR assay that
measured the disruption of the p53-MDM2 binding.37 A cis-imidazoline com-
pound, called nutlin, was identified as a small molecule that occupied the
hydrophobic pocket on the surface of MDM2 disrupting the p53-MDM2
PPI. Nutlin caused stabilization of p53 in cells and suppressed tumor growth
in xenograft models.37 The initial success of nutlin has spawned several other
ANTI-CANCER DRUG DISCOVERY 25
groups to investigate and identify small molecules that focus on disrupting the
p53–MDM2 interaction. Benzodiazepene analogs such as TDP665759 have
been shown to disrupt p53–MDM2 interactions in vitro with low micromolar
concentrations.36 They have also been found to suppress the growth of p53
wild-type tumor cells in culture. Treatment of normal mice with TDP665759
produces an increase in the p53-dependent transcript, cyclin-dependent kinase
inhibitor p21 in liver. It is therapeutically interesting that TDP665759 had a
synergistic growth-inhibitory effect with doxorubicin both in culture and in
xenografts. Potent spirooxindoles such as MI-215 have been synthesized that
inhibit the p53–MDM2 interaction in vitro in multiple tumor cell lines. MI-219
displays good pharmacokinetic and bioavailability properties in mice and
induces tumor regression without obvious reported untoward effects.
There is a vast array of potential PPI targets. It seems likely that the initial
success with p53-MDM2 PPI inhibitors will stimulate the field. There are
already efforts to generate PPI inhibitors for MYC-MAX38 and BCL2-BAX39
heterodimers. One would expect that some of these will enter advance preclin-
ical development in the near future. In addition, others are attempting to
produce inhibitors of protein–DNA interactions using analogs of natural anti-
biotics like netropsin and distamycin.40 These compounds might be able to
regulate transcription in tumor cells but the preclinical development of these
has been quite slow probably due to poor uptake of the compounds and the
lack of the requisite cancer selectivity.
Additional strategies are being pursued to reactivate p53 tumor suppressor
activity as potential cancer therapeutics. These include searching for com-
pounds that inhibit MDM2 ubiquitin E3 ligase activity or restore the thermal
stability and DNA-binding activity of p53 DNA-binding mutants. The core
domain of wild-type p53 is somewhat unstable, with a melting temperature of
44 C and a short half-life of nine minutes. Some of the known p53 mutations
add to the thermal instability of the protein, leading to the loss of DNA binding
and the p53 response. Small molecules that could stabilize p53 in its active
biological conformation and, thus, restore its DNA-binding functionality, could
potentially rescue wild-type p53 function. Proof-of-principle studies were
provided with antibodies that bind the C-terminus of p53 and with synthetic
peptides derived from the C-terminal domain.41 The antibodies and peptides
showed a stimulatory effect on the DNA-binding ability of p53. Because these
antibodies and peptides lack ideal pharmacological attributes, drug discovery
campaigns have been conducted to identify small molecules that would emu-
late the antibodies and peptides.
One compound, CP31398, was identified using an in vitro assay for confor-
mational refolding of mutant p53 to a wild-type shape.41 CP31398 induced the
expression of both reporter and endogenous p53 target genes. More recent
results suggest that CP31398 did not act as a PPI inhibitor but rather
26 JOHN S. LAZO
intercalated with DNA, altering and destabilizing the p53-DNA core domain
complex. CP31398 also decreased sequence-specific DNA binding of wild-type
p53 and the His273 mutant p53. It also produced cellular toxicity that was
independent of mutant p53. Therefore, there continues to be a challenge with
respect to reactivating mutant p53 with small molecules. If such compounds
were discovered, they will probably need to target a large fraction of the known
mutation observed in p53 to be clinically interesting.
are several practical and philosophical reasons for this reluctance. One concern
with the genetically engineered mouse models is that the resulting tumors are
thought to be genetically too homogenous. While this uniformity may concep-
tually be beneficial for preclinical evaluation of drugs, some believe that the
tumors lack the complex genetic heterogeneity seen in human tumors, which
may be an important factor determining the responsiveness to therapy. Using
genetically engineered mouse models of cancer required the establishment of a
substantial infrastructure and commitment of resources. In general, there is the
need to use a larger number of mice than with xenograft models to generate the
appropriate preclinical dataset. Recent studies with conditional mutant KRAS-
driven non-small-cell lung cancer and pancreatic adenocarcinoma models illus-
trate the power and perhaps the superiority of genetically engineered mouse
models.48 The investigators examined standard-of-care agents both alone and in
combination with clinically used agents that block the Epidermal Growth Factor
receptor and the Vascular Endothelial Growth Factor using noninvasive imaging
methods and conventional survival endpoints. They found a concordance be-
tween the preclinical results and existing clinical data leading them to conclude
that the genetically engineered mouse platform model human disease well and
that they had predictive value for anti-cancer drug development.
V. Conclusions
Molecular biology has irreversibly altered the manner in which new anti-
cancer drugs are discovered. It has provided numerous validated molecular
targets that have been the subject of high throughput drug screens. It has also
provided reagents to conduct imaging-based cellular screens and to generate
new animal models of cancer. Although the inherent complexities of human
cancer remain, the new knowledge obtained about the processes permitting
the development of cancer should enable the creation of more efficacious drugs
for the treatment of hematological and solid tumors.
References
1. Kamb A, Wee S, Lengauer C. Why is cancer drug discovery so difficult? Nat Rev Drug Discov
2007;6:115–20.
2. Pors K, Goldberg FW, Leamon CP, Rigby AC, Snyder SA, Falconer RA. The changing
landscape of cancer drug discovery: a challenge to the medicinal chemist of tomorrow. Drug
Discov Today 2009;14:1045–50.
3. Lazo JS, Brady LS, Dingledine RJ. Building a pharmacological lexicon: small molecule discov-
ery in academia. Mol Pharmacol 2007;72:1–7.
28 JOHN S. LAZO
4. Bakan A, Lazo JS, Wipf P, Brummond KM, Bahar I. Toward a molecular understanding of the
interaction of dual specificity phosphatases with substrates: insights from structure-based
modeling and high throughput screening. Curr Med Chem 2008;15:2536–44.
5. Lazo JS. Rear-view mirrors and crystal balls: a brief reflection on drug discovery. Mol Interv
2008;8:60–3.
6. Luo J, Solimini NL, Elledge SJ. Principles of cancer therapy: oncogene and non-oncogene
addiction. Cell 2009;136:823–37.
7. Lapenna S, Giordano A. Cell cycle kinases as therapeutic targets for cancer. Nat Rev Drug
Discov 2009;8:547–66.
8. Kerbel RS. Antiangiogenic therapy: a universal chemosensitization strategy for cancer? Science
2006;312:1171–5.
9. Bottone Jr. FG, Moon Y, Kim JS, Alston-Mills B, Ishibashi M, et al. The anti-invasive activity of
cyclooxygenase inhibitors is regulated by the transcription factor ATF3 (activating transcription
factor 3). Mol Cancer Ther 2005;4:693–703.
10. Schmitt CA. Cellular senescence and cancer treatment. Biochim Biophys Acta
2007;1775:5–20.
11. Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg RA, et al. Identification of
selective inhibitors of cancer stem cells by high-throughput screening. Cell 2009;138:645–59.
12. Truong BT, Lee YJ, Lodie TA, Park DJ, Perrotti D, Watanabe N, et al. CCAAT/enhancer
binding proteins repress the leukemic phenotype of acute myeloid leukemia. Blood
2003;101:1141–8.
13. Sharma SV, Haber DA, Settleman J. Cell line-based platforms to evaluate the therapeutic
efficacy of candidate anticancer agents. Nat Rev Cancer 2010;10:241–53.
14. Borrell B. How accurate are cancer cell lines? Nature 2010;463:245.
15. Achiwa H, Lazo JS. PRL-1 tyrosine phosphatase regulates c-Src levels, adherence, and invasion
in human lung cancer cells. Cancer Res 2007;67:643–50.
16. Lazo JS, Ruddon RW. Neurite extensions and malignancy of murine neuroblastoma cells after
treatment with prostaglandin E1 and papaverine. J Natl Cancer Inst 1977;59:137–43.
17. Spira AJ, Carducci MA. Differentiation therapy. Curr Opin Pharmacol 2003;3:338–43.
18. Tierno MB, Kitchens CA, Petrik B, Graham TH, Wipf P, Xu F, et al. Microtubule binding and
disruption and induction of premature senescence by Disorazole C1. J Pharmacol Exp Ther
2009;328:715–22.
19. Vogt A, Cholewinski A, Shen S, Nelson S, Lazo JS, Tsang M, et al. Automated image-based
phenotypic analysis in zebrafish embryos. Dev Dynamics 2009;238:656–63.
20. MacKeigan JP, Murphy LO, Blenis J. Sensitized RNAi screen of human kinases and phospha-
tases identified new regulators of apoptosis and chemoresistance. Nat Cell Biol 2005;7:591–600.
21. Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitors transactivate RAF
dimers and ERK signalling in cells with wild-type BRAF. Nature 2010;464:427–31.
22. Payne DJ, Gwynn MN, Holmes DJ, Pompliano DL. Drugs for bad bugs: confronting the
challenges of antibacterial discovery. Nat Rev Drug Discov 2007;6:29–40.
23. Weinstein IB. Cancer. Addition to oncogenes-the Achilles heal of cancer. Science
2002;297:63–4.
24. Weinstein IB, Joe A. Oncogene addiction. Cancer Res 2008;68:3077–80.
25. Bertotti A, Burbridge MF, Gastaldi S, Galimi F, Torti D, Medico E, et al. Only a subset of met-
activated pathways are required to sustain oncogene addiction. Sci Signal 2009;2:er11.
26. Felsher DW, Bishop JM. Transient excess of MYC activity can elicit genomic instability and
tumorigenesis. Proc Natl Acad Sci USA 1999;96:3940–4.
27. Ventura A, Kirsch DG, McLaughlin ME, Tuveson DA, Grimm J, Lintault L, et al. Restoration
of p53 function leads to tumor regression in vivo. Nature 2007;445:661–5.
ANTI-CANCER DRUG DISCOVERY 29
28. Tong AH, Evangelista M, Parsons AB, Xu H, Bader GD, Pagé N, et al. Systematic genetic
analysis with ordered arrays of yeast deletion mutants. Science 2001;294:2364–8.
29. Kaelin J, Kaelin WG. The concept of synthetic lethality in the context of anticancer therapy. Nat
Rev Cancer 2005;5:689–98.
30. Stockwell BR, Haggarty SJ, Schreiber SL. High-throughput screening of small molecules in
miniaturized mammalian cell-based assays involving post-translational modifications. Chem
Biol 1999;6:71–83.
31. Torrance CJ, Agrawal V, Vogelstein B, Kinzler KW. Use of isogenic human cancer cells for high-
throughput screening and drug discovery. Nat Biotech 2001;19:940–5.
32. Dolma S, Lessnick SL, Hahn WC, Stockwell BR. Identification of genotype-selective anti-
tumor agents using synthetic lethal chemical screening in engineered human tumor cells.
Cancer Cell 2003;3:285–96.
33. Tardi PG, Dos Santos N, Harasym TO, Johnstone SA, Zisman N, Tsang AW, et al. Drug ratio-
dependent antitumor activity of irinotecan and cisplatin combinations in vitro and in vivo. Mol
Cancer Ther 2009;8:2266–75.
34. Colas P. High-throughput screening assays to discover small-molecule inhibitors of protein
interactions. Curr Drug Discov Technol 2008;5:190–9.
35. Pagliaro L, Felding J, Audouze K, Nielsen SJ, Terry RB, Krog-Jensen C, et al. Emerging classes
of protein–protein interaction inhibitors and new tools for their development. Curr Opin Chem
Biol 2004;8:442–9.
36. Brown CJ, Lain S, Verma CS, Fersht AR, Lane DP. Awakening guardian angels: drugging the
p53 pathway. Nat Rev Cancer 2009;9:862–73.
37. Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z, et al. In vivo activation of the
p53 pathway by small-molecule antagonists of MDM2. Science 2004;303:844–8.
38. Yin XY, Giap C, Lazo JS, Prochownik EV. Low molecular weight inhibitors of Myc-Max
interaction and function. Oncogene 2003;22:6151–9.
39. Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Augeri DJ, Belli BA, et al. An
inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 2005;435:677–81.
40. Reddy BS, Sharma SK, Lown JW. Recent developments in sequence selective minor groove
DNA effectors. Curr Med Chem 2001;8:475–508.
41. Foster BA, Coffey HA, Morin MJ, Rastinejad F. Pharmacological rescue of mutant p53
conformation and function. Science 1999;286:2507–10.
42. Kelland LR. Of mice and men: values and liabilities of the athymic nude mouse model in
anticancer drug development. Eur J Cancer 2004;40:827–36.
43. Klein S, Levitzki A. Targeted cancer therapy: promise and reality. Adv Cancer Res
2007;97:295–319.
44. Suggitt M, Bibby MC. 50 years of preclinical anticancer drug screening: empirical to target-
driven approaches. Clin Cancer Res 2005;11:971–81.
45. Hall LA, Krauthauser CM, Wexler RS, Hollingshead MG, Slee AM, Kerr JS. The hollow fiber
assay: continued characterization with novel approaches. Anticancer Res 2000;20:903–11.
46. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, et al. Inhibition
of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic
cancer. Science 2009;324:1457–61.
47. Olive KP, Tuveson DA. The use of targeted mouse models for preclinical testing of novel cancer
therapeutics. Clin Cancer Res 2006;12:5277–87.
48. Singh M, Lima A, Molina R, Hamilton P, Clermont AC, Devasthali V, et al. Assessing
therapeutic responses in Kras mutant cancers using genetically engineered mouse models.
Nat Biotech 2010;28:585–93.
This page intentionally left blank
Targeting Chemokine
(C-C motif) Ligand 2 (CCL2)
as an Example of Translation
of Cancer Molecular Biology
to the Clinic
Jian Zhang,*,{ Lalit Patel,*,{
and Kenneth J. Pienta*,{
*Department of Medicine, Michigan Center
for Translational Pathology and the
University of Michigan Comprehensive
Cancer Center, University of Michigan,
Ann Arbor, Michigan, USA
{
Department of Urology, Michigan Center
for Translational Pathology and the
University of Michigan Comprehensive
Cancer Center, University of Michigan,
Ann Arbor, Michigan, USA
Chemokines are a family of small and secreted proteins that play pleiotropic
roles in inflammation-related pathological diseases, including cancer. Among
the identified 50 human chemokines, chemokine (C-C motif) ligand 2 (CCL2)
is of particular importance in cancer development since it serves as one of the
key mediators of interactions between tumor and host cells. CCL2 is produced
by cancer cells and multiple different host cells within the tumor microenvi-
ronment. CCL2 mediates tumorigenesis in many different cancer types. For
example, CCL2 has been reported to promote prostate cancer cell prolifera-
tion, migration, invasion, and survival, via binding to its functional receptor
CCR2. Furthermore, CCL2 induces the recruitment of macrophages and
induces angiogenesis and matrix remodeling. Targeting CCL2 has been
demonstrated as an effective therapeutic approach in preclinical prostate
cancer models, and currently, neutralizing monoclonal antibody against
CCL2 has entered into clinical trials in prostate cancer. In this chapter,
targeting CCL2 in prostate cancer will be used as an example to show
translation of laboratory findings from cancer molecular biology to the clinic.
I. Biology of CCL2
A. CCL2 Basics
Chemokines, a family of chemoattractant cytokines, are classified into four
sub-families as CXC, CC, CX3C, and C based upon the number and location of
the cysteine residues at the N-terminus of the protein. Chemokine (C-C motif)
ligand 2 (CCL2), also known as monocyte chemotactic protein-1 (MCP-1), is a
small, secreted protein that belongs to the CC chemokine family. CCL2 was
purified and cloned in 1989 from human gliomas and myelomonocytic cells by
two independent research groups based on its ability to chemoattract mono-
cytes.1,2 Subsequent to its cloning, it was confirmed that this protein was also
identical to the product of the human JE gene. The JE gene, originally identi-
fied in mouse fibroblasts, is a platelet-derived growth factor (PDGF)-inducible
gene. Since then, CCL2 has been shown to display chemoattractic activity for
not only monocytes, but also memory T cells, natural killer (NK) cells, and
perhaps dendritic cells, resulting in recruiting of these cells to sites of tissue
injury and inflammatory responses.3,4 The human CCL2 cDNA encodes a 99
amino acid residue precursor protein with a hydrophobic signal peptide of 23
amino acids and a mature peptide of 76 amino acids.5,6 The CCL2 gene is
located on the chromosome 17 where many of the genes of the CC chemokine
family are located. The mouse or rat CCL2 gene has about 75% homology to
humans. CCL2 functions through binding to a functional chemokine receptor
CCR2, although it also binds to CCR4.7 The roles of CCL2 have been impli-
cated in the pathogenesis of various diseases that associate with monocyte
infiltration, for example, rheumatoid arthritis, atherosclerosis, and multiple
types of cancer [see review in Ref. 8].
TARGETING CHEMOKINE (C-C MOTIF) LIGAND 2 (CCL2) 33
B. CCL2 Expression
CCL2 is expressed in a wide array of tissues. It can be produced by multiple
cell types, including fibroblasts, macrophages, lymphocytes, astrocytes, mast
cells, endothelial cells, and osteoblasts.1,9–14 In addition, CCL2 can also be
produced by a variety of human and murine malignant cells [see review in Refs.
15–17]. In prostate cancer, determined by immunohistochemical staining on a
human tissue microarray, CCL2 positive staining was located mostly in epithe-
lial and fibromuscular stromal cells18; however, CCL2 positive staining was also
observed in the extracellular areas surrounding neoplastic glands and epithelial
cells,18 suggesting both autocrine and paracrine production of CCL2 in the
tumor microenvironment. In at least one report, CCL2 expression levels
positively correlated with Gleason score (a measure of tumor aggressiveness)
and pathologic stages.18–20 CCL2 production has been determined by enzyme-
linked immunosorbent assay (ELISA) in conditioned media collected from
prostate cancer cell lines and compared to primary prostate epithelial cells.18
Prostate cancer cells produce higher amounts of CCL2 as compared to non-
malignant prostate epithelial cells.18 Higher production of CCL2 at the sites of
bone metastases was demonstrated by a clinically related report in which
Loberg et al. collected tumor bone metastatic and normal bone specimens
from vertebral lesions in three patients with prostate cancer.21 Total protein
lysates were isolated and analyzed by cytokine array. Elevated CCL2 produc-
tion was identified in the tumor-bone microenvironment compared to normal
bone microenvironment, suggesting that CCL2 plays a critical role in prostate
tumorigenesis in bone metastases.21 In vitro, it has been further reported that
human osteoblasts and bone marrow endothelial cells produced higher amount
of CCL2 compared to prostate epithelial cells.13 CCL2 can also be produced by
osteoclasts.22–25 Its production can be induced by Receptor Activator of NF-kB
ligand (RANKL) and tumor necrosis factor alpha (TNFa).22–25 To date, it
remains unclear which cell type(s) play a major role in the production of
CCL2 in the tumor microenvironment.
C. CCL2 Functions
CCL2 functions as a chemoattractant through binding to its receptor on
monocytes, macrophages, and lymphocytes [see review in Ref. 26]. The exis-
tence of CCL2/CCR2 axis has been validated using CCR2 knockout animals.27
In acute inflammatory response, CCL2 has been shown to actively recruit
monocytes to the site of inflammation.28,29 CCL2 also plays important roles
in T-cell immunity, and CCL2 expression is associated with Th2 response.30–32
For example, CCL2 is overproduced in an animal model of Th2 immune-
mediated asthma.33 CCL2 is a potent factor in the polarization of Th0 cells
toward a Th2 phenotype.34 It has been demonstrated that CCL2 induced
34 ZHANG ET AL.
pulmonary disease [see review in Ref. 56]. In a prostate cancer study, a CCR2
antagonist has been shown to diminish the prostate cancer cell proliferation and
invasion in vitro.18
Recently, CCR2 was reported as a key factor in balancing the bone
remodeling process.57 It was shown that CCR2 knockout mice had high bone
mass and stability (biomechanical properties by compression) due to a decrease
in number, size, and function of osteoclasts.57 RANK expression is diminished
in CCR2 knockout mice, and CCL2 enhances RANK expression via NFkB and
ERK1/2 pathways57; therefore, CCR2 could become a therapeutic target in
postmenopausal bone loss.
2. CCR2 REGULATION
Little is known about the regulation of the CCR2 gene in normal or
cancerous tissues. It is downregulated as monocytes move down the macro-
phage differentiation pathway while other related chemokine receptors are
not.78 IFNgþ M-CSF or PMA þ ionomycin downregulate CCR2 expression in
monocytes, and this can be replicated with a 1220/þ115 hCCR2 promoter-
pGL3 luciferase reporter.78 Peroxisome proliferator-activated receptor-gamma
(PPAR-g) ligands (i.e., Rosiglitazone) also downregulate CCR2 in circulating
monocytes, while cholesterol slightly upregulated CCR2.79 While proinflam-
matory cytokines rapidly reduce CCR2 expression in monocytes, they upregu-
late CCR2 expression in the brain.80 Constitutive tissue-specific expression of
CCR2 in THP-1 monocyte cells has been shown to be dependent upon a 31-bp
region ( 89 to 59) adjacent to the TATA box that contains an Oct-1 binding
site and a pair of tandem C/EBP binding sites located in the 50 UTR (þ 50 to
þ 77 bp).81 In addition to the Oct-1 and C/EBP binding sites that function in
monocyte CCR2 expression, the hCCR2 50 flank and UTR contains an array of
possible binding sites for PPAR/RXR, SREBP, GATA, STAT, NFAT, and AP-1.
It remains unknown whether these sequences are sufficient for positive
TARGETING CHEMOKINE (C-C MOTIF) LIGAND 2 (CCL2) 37
B. Angiogenesis
Chemokines play an important role in the maintenance of hematopoietic
homeostasis, regulation of cell proliferation, tissue morphogenesis, and angio-
genesis.93 In human breast cancer, it was reported that CCL2 levels in the
excised breast cancer tissue were correlated significantly with the levels of
angiogenic factors, including vascular endothelial growth factor (VEGF), thy-
midine phosphorylase, TNFa, and IL-8.94 Transfection of colon cancer cells
with the CCL2 gene induces angiogenesis in a murine model.95 It has been
demonstrated that both CCL2 and VEGF expression positively correlates with
TAM infiltration and angiogenesis in breast cancer.96 In prostate cancer, it has
been reported that CCL2 induces tumor cells to produce the pro-angiogenic
factor VEGF-A, which indirectly induces sprout formation in human bone
marrow endothelial cells.71 In vivo, it has been shown that administration of
neutralizing antibody against CCL2 significantly reduces tumor blood vessel
density and decreases the prostate cancer tumor burden (Fig. 1)71,97; therefore,
CCL2 is a key mediator of tumor angiogenesis.
Other documents randomly have
different content
mandó que se volviesen á sus pueblos, y luego les mandó salir de
Méjico.
Dejemos á los mensajeros, que luego salieron, y los mejicanos
por tres partes con la mayor furia que hasta allí habiamos visto, y se
vienen á nosotros, y en todos tres reales nos dieron muy recia
guerra; y puesto que les heriamos y matábamos muchos dellos,
paréceme que deseaban morir peleando, y entónces cuando más
recios andaban con nosotros pié con pié peleando, nos decian:
—«Tenitoz Rey Castilla, Tenitoz Ajaca;» que quiere decir en su
lengua: «¿Qué dirá el Rey de Castilla? ¿Qué dirá ahora?»
Y con estas palabras tirar vara y piedra y flecha, que cubrian el
suelo y calzada.
Dejemos esto, que ya les íbamos ganando gran parte de la
ciudad, y en ellos sentiamos que, puesto que peleaban muy como
varones, no se remudaban ya tantos escuadrones como solian, ni
abrian zanjas ni calzadas; mas otra cosa tenian muy cierta, que al
tiempo que nos retraiamos nos venian siguiendo hasta nos echar
mano; y tambien se nos habia acabado ya la pólvora en todos tres
reales, y en aquel instante habia venido á la Villa-Rica un navío que
era de una armada de un licenciado Lúcas Vazquez de Aillon, que se
perdió y desbarató en las islas de la Florida, y el navío aportó á
aquel puerto, como dicho tengo, y venian en él ciertos soldados y
pólvora y ballestas y otras cosas; y el teniente que estaba en la Villa-
Rica, que se decia Rodrigo Rangel, que tenia en guarda á Narvaez,
envió luego á Cortés pólvora y ballestas y soldados.
Y volvamos á nuestra conquista, por abreviar: que mandó y
acordó Cortés con todos los demás capitanes y soldados que les
entrásemos todo cuanto pudiésemos hasta llegalles al Tatelulco, que
es la plaza mayor, adonde estaban sus altos cues y adoratorios; y
Cortés por su parte y Sandoval por la suya, y nosotros por la
nuestra, les íbamos ganando puentes y albarradas, y Cortés les
entró hasta una plazuela donde tenian otros adoratorios.
En aquellos cues estaban unas vigas, y en ellas muchas cabezas
de nuestros soldados que habian muerto y desbaratado en las
batallas pasadas, y tenian los cabellos y barbas muy crecidas, más
que cuando eran vivos, y no lo habia yo creido si no lo viera desde
tres dias, que como fuimos ganando por nuestra parte dos aberturas
y puentes, tuvimos lugar de las ver, é yo conocia tres soldados mis
compañeros; y cuando las vimos de aquella manera se nos saltaron
las lágrimas de los ojos; y en aquella sazon se quedaron allí donde
estaban, más desde á doce dias se quitaron, y las pusimos aquellas
y otras cabezas que tenian ofrecidas á otros ídolos, y las enterramos
en una iglesia que se dice ahora los Mártires, que nosotros hicimos.
Dejemos desto y digamos cómo fuimos batallando por la parte de
Pedro de Albarado y llegamos al Tatelulco, y habia tantos mejicanos
en guarda de sus ídolos y altos cues, y tenian tantas albarradas, que
estuvimos bien dos horas que no se lo pudimos tomar; y cómo
podian ya correr caballos, puesto que les hirieron á los más; mas
nos ayudaron muy bien y alancearon muchos mejicanos; y como
habia tantos contrarios en tres partes, fuimos las tres capitanías á
batallar con ellos; y á la una capitanía, que era de un Gutierre de
Badajoz, mandó Pedro de Albarado que subiese en el alto cu de
Huichilóbos, y peleó muy bien con los contrarios y muchos papas
que en las casas de los adoratorios estaban, y de tal manera le
daban guerra los contrarios, que le hacian venir las gradas abajo; y
luego Pedro de Albarado nos mandó que le fuésemos á socorrer y
dejásemos el combate en que estábamos; é yendo que íbamos, nos
siguieron los escuadrones con quien peleábamos, y todavía les
subiamos sus gradas arriba.
Aquí habia bien que decir en qué trabajo nos vimos los unos y los
otros en ganalles aquellas fortalezas, que ya he dicho otras veces
que eran muy altas; y en aquellas batallas nos tornaron á herir á
todos muy malamente, y todavía les pusimos fuego á los ídolos, y
levantamos nuestras banderas, y estuvimos batallando en lo llano,
despues de le haber puesto fuego, hasta la noche, que no nos
podiamos valer de tanto guerrero.
Dejemos de hablar en ello, y digamos que como Cortés y sus
capitanes vieron en aquella sazon desde sus barrios y calles en sus
partes léjos del alto cu, y las llamaradas en que el cu mayor ardia, y
nuestras banderas encima, se holgó mucho, y se quisieran hallar en
él; mas no podian, porque habian un cuarto de legua de la una parte
á la otra, y tenian muchas puentes y aberturas de agua por ganar, y
por donde andaba le daban recia guerra, y no podian entrar tan
presto como quisieran en el cuerpo de la ciudad; mas dende á
cuatro dias se juntó con nosotros, así Cortés como Sandoval, é
podiamos ir desde un real á otro por las calles y casas derrocadas y
puentes y albarradas deshechas y aberturas de agua todo ciego; y
en este instante se iban retrayendo Guatemuz con todos sus
guerreros en una parte de la ciudad dentro de la laguna, porque las
casas y palacios en que vivia ya estaban por el suelo; y con todo
esto, no dejaban cada dia de salir á nos dar guerra, y al tiempo de
retraer nos iban siguiendo muy mejor que de ántes; é viendo esto
Cortés, que se pasaban muchos dias, y no venian de paz ni tal
pensamiento tenian, acordó con todos nuestros capitanes que les
echásemos celadas.
Y fué desta manera: que de todos tres reales se juntaron hasta
treinta de á caballo y cien soldados los más sueltos y guerreros que
conocia Cortés, y envió á llamar de todos tres reales mil tlascaltecas,
y nos metimos en unas casas grandes que habian sido de un señor
de Méjico, y esto fué muy de mañana, y Cortés iba entrando con los
demás de á caballo que le quedaban, y sus soldados y ballesteros y
escopeteros por las calles y calzadas como solia; y ya llegaba Cortés
á una abertura y puente de agua, y entónces estaban peleando con
los escuadrones de mejicanos que para ello estaban aparejados, y
muchos más que Guatemuz enviaba para guardar la puente; y como
Cortés vió que habia gran número de contrarios, hizo que se retraia
y mandaba echar los amigos fuera de la calzada, porque creyesen
que de hecho se iban retrayendo; y le iban siguiendo al principio
poco á poco, y cuando vieron que de hecho hacia que iba huyendo,
van tras él todos los poderes que en aquella calzada le daban
guerra; y como Cortés vió que habia pasado algo adelante de las
casas á donde estaba la celada, tiraron dos tiros juntos, que era
señal de cuándo habiamos de salir de la celada, y salen los de á
caballo primero, y salimos todos los soldados y dimos en ellos á
placer; pues luego volvió Cortés con los suyos y nuestros amigos los
tlascaltecas, é hicieron gran matanza.
Por manera que se hirieron y mataron muchos, y desde allí
adelante no nos seguian al tiempo del retraer; y tambien en el real
de Pedro de Albarado les echó una celada, mas no tan buena como
esta; y en aquel dia no me hallé yo en nuestro real con Pedro de
Albarado por causa que Cortés me mandó que para la celada
quedase con él.
Dejemos desto, y digamos cómo estábamos ya en el Tatelulco, y
Cortés nos mandó que pasásemos todas las capitanías á estar con
él, é que allí velásemos, por causa que veniamos más de media
legua desde el real á batallar con los mejicanos; y estuvimos allí tres
dias sin hacer cosa que de contar sea, porque nos mandó que no les
entrásemos más en la ciudad ni les derrocásemos más casas, porque
les queria tornar á requerir con las paces; y en aquellos dias que allí
estuvimos en el Tatelulco envió Cortés á Guatemuz rogándole que se
diese y no hubiese miedo, y con grandes ofrecimientos que le
prometia que su persona seria muy acatada y honrada dél, y que
mandaria á Méjico y á todas sus tierras y ciudades como solia; y les
envió bastimentos y regalos, que eran tortillas y gallinas y cerezas y
tunas y caza, é que no tenian otra cosa; y el Guatemuz entró en
consejo con sus capitanes, y lo que le aconsejaron fué, que dijese
que queria paz, é que aguardarian tres dias, é que al cabo de los
tres dias se verian el Guatemuz y Cortés, y se darian los conciertos
de las paces; y en aquellos tres dias tenian tiempo de aderezar
puentes y abrir calzadas y adobar piedra y vara y flecha y hacer
albarradas; y envió Guatemuz cuatro mejicanos principales con
aquella respuesta; é creiamos que eran verdaderas las paces, y
Cortés les mandó dar muy bien de comer y beber, y les tornó á
enviar á Guatemuz, y con ellos les envió más refresco como de
ántes; y el Guatemuz tornó á enviar á Cortés otros mensajeros, y
con ellos dos mantas ricas, y dijeron que Guatemuz vernia para
cuando estaba acordado; y por no gastar más razones sobre el caso,
él nunca quiso venir, porque le aconsejaron que no creyese á Cortés,
y poniéndole por delante el fin de su tio el gran Montezuma y sus
parientes y la destruccion de todo el linaje noble de los mejicanos, é
que dijese que estaba malo, é que saliesen todos de guerra, é que
placeria á sus dioses, que les darian vitoria contra nosotros, pues
tantas veces se la habia prometido.
Pues como estábamos aguardando al Guatemuz y no venia, vimos
luego la burla que de nosotros hacia; y en aquel instante salian
tantos batallones de mejicanos con sus divisas, y dan á Cortés tanta
guerra, que no se podia valer; y otro tanto fué por nuestra parte de
nuestro real; pues en el de Sandoval lo mismo; y era de tal manera,
que parecia que entónces comenzaban de nuevo á batallar; y como
estábamos algo descuidados, creyendo que estaban ya de paz,
hirieron á muchos de nuestros soldados, y tres fueron heridos muy
malamente, y el uno dellos murió, y mataron dos caballos y hirieron
otros más; é ellos no se fueron mucho alabando, que muy bien lo
pagaron; y como esto vido Cortés, mandó que luego les tornásemos
á dar guerra y les entrásemos en su ciudad á la parte donde se
habian recogido; y cómo vieron que les íbamos ganando toda la
ciudad, envió Guatemuz á decir á Cortés que queria hablar con él
desde una gran abertura de agua, y habia de ser Cortés de la una
parte y el Guatemuz de la otra, y señalaron el tiempo para otro dia
de mañana; y fué Cortés para hablar con él, y no quiso Guatemuz
venir al puesto, sino envió á muchos principales, los cuales dijeron
que su señor Guatemuz no osaba venir por temor que cuando
estuviese hablando le tirarian escopetas y ballestas y le matarian; y
entónces Cortés les prometió con juramento que no les enojaria en
cosa ninguna, y no aprovechó, que no le creyeron.
En aquella sazon dos principales de los que hablaban con Cortés
sacaron de un fardalejo que traian tortillas é una pierna de gallina y
cerezas, y sentáronse muy de espacio á comer, porque Cortés los
viese y entendiese que no tenian hambre; y desde allí le envió á
decir á Guatemuz, que pues no queria venir, que no le daba nada y
que presto les entraria en todas sus casas, y veria si tenia maíz,
cuanto más gallinas; y desta manera se estuvieron otros cuatro ó
cinco dias que no les dábamos guerra; y en este instante se salian
de noche muchos pobres indios que no tenian qué comer, y se
venian al real de Cortés y al nuestro, como aburridos de hambre; y
cuando aquello vió Cortés, mandó que en bueno ni en malo no les
diésemos guerra, é que quizá se les mudaria la voluntad para venir
de paz, y no venian; y en el real de Cortés estaba un soldado que
decia el mismo que él habia estado en Italia en compañía del Gran
Capitan, y se halló en la chirinola de Garayana y en otras grandes
batallas, y decia muchas cosas de ingenios de la guerra, é que haria
un trabuco en el Tatelulco, con que en dos dias que con él tirase á la
parte y casas de la ciudad adonde el Guatemuz se habia retraido,
que las haria que luego se diesen de paz; y tantas cosas dijo á
Cortés sobre ello, que luego puso en obra hacer el trabuco, y
trajeron piedra, cal y madera de la manera que él la demandó, y
carpinteros y clavazon, y todo lo perteneciente para hacer el
trabuco, é hicieron dos hondas de recias sogas, y trujeron grandes
piedras, y mayores que botijas de arroba; é ya que estaba armado el
trabuco segun y de la manera que el soldado dió la órden, y dijo que
estaba bueno para tirar, y pusieron en la honda una piedra hechiza,
lo que con ella se hizo es, que no pasó adelante del trabuco, porque
fué por alto y luego cayó allí donde estaba armado; y desque
aquello vió Cortés hubo mucho enojo del soldado que le dió la órden
para que lo hiciese, y tenia pesar en sí mismo, porque él creido tenia
que no era para en la guerra ni para en cosa de afrenta, y no era
más de hablar, que se habia hallado de la manera que he dicho; y
segun el mismo soldado decia, que se decia Fulano de Sotelo,
natural de Sevilla, y luego Cortés mandó deshacer el trabuco.
Dejemos desto, y digamos que como vió que el trabuco era cosa
de burla, acordó que con todos doce bergantines fuese en ellos
Gonzalo de Sandoval por capitan general y entrase en el rincon de la
ciudad adonde se habia retraido Guatemuz, el cual estaba en parte
que no podian entrar en sus palacios y casas sino por el agua, y
luego Sandoval apercibió á todos los capitanes de los bergantines; y
lo que hizo diré adelante cómo y de qué manera pasó.
CAPÍTULO CLVI.
CÓMO SE PRENDIÓ GUATEMUZ.