throbber
|PR2017-00737
`
`Genentech 2137
`Genentech 2137 '
`Hospira v. Genentech
`Hospira v. Genentech
`IPR2017-00737
`
`

`

`The Basic Science of
`Oncology
`
`Second Edition
`
`Editors
`
`Ian F. Tannock, M.D., Ph.D.
`Richard P. Hill, Ph.D.
`
`The Ontario Cancer Institute
`and
`The University of Toronto
`Toronto, Ontario
`Canada
`
`McGraw-Hill, lnc.
`Health Professions Division
`
`San Francisco Auckland
`St. Louis
`New York
`Bogota Caracas
`Lisbon
`London Madrid
`Mexico Milan Montreal New Delhi
`Paris
`
`San Juan
`
`Singapore
`
`Sydney
`
`Tokyo
`
`Toronto
`
`

`

`
`
`THE BASIC SCIENCE OF ONCOLOGY
`
`Copyright © 1992 by McGraw-Hill, Inc. All rights reserved.
`Printed in the United States of America. Except as permitted un—
`der the United States Copyright Act of 1976, no part of this pub-
`lication may be reproduced or distributed in any form or by any
`means, or stored in a data base or retrieval system, without the
`prior written permission of the publisher.
`
`1234567890MALMAL98765432
`
`This book was set in Baskerville by Beljan, Ltd.
`The editors were jane E. Pennington and Lester A. Sheinis.
`The production supervisor was Roger Kasunic.
`The cover was designed by Carol Matsuyama.
`Malloy Lithographing, Inc., was printer and binder.
`
`ISBN 0-07-105407-3
`
`Library of Congress Cataloging-in-Publication Data
`
`The Basic science of oncology / edited by Ian F. Tannock, Richard
`P. Hill.
`p. cm.
`Includes bibliographical references and index.
`ISBN 0-07-105407-3
`1. Cancer.
`2. Oncology.
`Richard P.
`
`1. Tannock, Ian F.
`
`II. Hill,
`
`[DNLM: 1. Neoplasms. QZ 200 B3115]
`RC261.B37 1992
`616.99’ 4—dc20
`DNLM/DLC
`
`for Library of Congress
`
`92-2884CIP
`
`

`

`Contents
`AbouttheEditorsandContributors..................................................vii
`
`Preface ............................................................................ ix
`
`1. Introduction: CancerasaCellularDisease...............1
`
`Richard P. Hill and Ian F. Tarmac/c
`
`PART 1: CANCER CAUSATION
`
`2. EpidemiologyofCancer...............7
`Norman F. Boyd
`
`3. MethodsofGeneticAnalysis................................................
`Jeremy Squire and RobertA. Phillips
`
`...23
`
`4-. GeneticBasisofCancer. 4-1
`
`JeremySquireandRoberlA. Phillips
`
`SOncogenes.
`.. 61
`MarkD. Mina’eri and Anthony J Pawson
`
`6. Viruses and Cancer. .
`Sam Benchimol
`
`.
`
`.
`
`. ...................................................... 88
`
`7. Chemical Carcinogenesis......................................................102
`Michael C. Archer
`
`8/.RadiationCarcinogenesis.... 119
`[/1 A. Michael Rauth
`
`PART 2: CANCER BIOLOGY
`
`9. Properties ofMalignantCellsl39
`RonaldN. Buick and [an F. Tarmac/c
`
`10. CellProliferation.............................................................154
`
`Ian F. Tarmac/c
`11. Metasta51sl78
`Richard P. Hill
`12. TumorMarkers..................................._,................._.......196
`AaronMal/cirz
`
`13 Hormonesand Cancer. 207
`DonaldJ Sutherland and Betty G. Mobbs
`
`I
`
`(
`V
`
`1
`
`I
`
`\
`
`|
`
`|
`
`l
`.
`1
`
`I
`
`i
`
`

`

`
`
`vi
`
`CONTENTS
`
`14. ImmunologyandImmunotherapyofCancer....._._.........._......_._...._.._..
`Richard G. Miller and Ian F. Tarmock
`
`232
`
`PART 3: BIOLOGY UNDERLYING CANCER TREATMENT
`
`15. Cellular Basis of Radiotherapy .....
`Richard P. Hill
`
`. 259
`
`16. Experimental Radiotherapy............................_......_._.._..._...__._
`Richard P. Hill
`
`276
`
`17. Biological Properties of Anticancer Drugs. .
`Ian F. Tarmac/c
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`_
`
`.
`
`.
`
`_
`
`.
`
`.
`
`_
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`. ...
`
`. 302
`
`18. PharmacologyofAnticancerDrugs....._.__.............._...._._........._.....
`Charles Erlichmarz
`
`19. Experimental Chemotherapy......................_....._._.__..._._._.._.._._.
`Ian F. Tarmac/c
`
`317
`
`338
`
`20. Hyperthermia andPhotodynamic Therapy
`Richard P. Hill
`
`. 360
`
`21. Guide to Studies of Diagnostic Tests, Prognosis, and Treatment .....................
`Norman F. Boyd
`
`379
`
`Glossary.........._........_.........
`
`Index
`
`_ 395
`
`. 403
`
`

`

`
`
`19
`
`Experimental Chemotherapy
`
`Ian F. Tannock
`
`
`
`19.1 INTRODUCTION
`19.1.1 Current Status of Chemotherapy
`19.1.2 Therapeutic Index
`19.1.3 Relationship Between Tumor Remission and Cure
`
`19.2 DRUG RESISTANCE
`19.2.1 Mechanisms of Drug Resistance
`19.2.2 Genetic Basis of Drug Resistance
`19.2.3 Resistance to Methotrexate
`19.2.4 Multiple Drug Resistance
`19.2.5 Topoisomerases and Drug Resistance
`19.2.6 Glutathione and Drug Resistance
`
`19.3.3 Directed Drug Delivery
`19.3.4 Drugs That Cause Differentiation
`19.3.5 Anti-angiogenesis
`19.3.6 Drugs for Hypoxic and Nutrient-deprived Cells
`19.3.7 High-Dose Chemotherapy with Bone-Marrow
`Transplantation
`
`19.4 TREATMENT WITH MULTIPLE AGENTS
`19.4.1 Influence on Therapeutic Index
`19.4.2 Synergy and Additivity
`19.4.3 Growth Factors and Other Modifiers of Drug Activity
`19.4.4 Radiation and Drugs
`
`19.3 EXPERIMENTAL APPROACHES TO IMPROVEMENT
`OF CHEMOTHERAPY
`19.3.1 Adjuvant Chemotherapy
`REFERENCES
`19.3.2 Antimetastatic Drugs
`
`
`19.5 SUMMARY
`
`19.1 INTRODUCTION
`
`19.1.1 Current Status of Chemotherapy
`
`In current clinical practice chemotherapy is used in the
`following ways: (a) as the major curative modality for
`a few rare types of malignancies, such as Hodgkin’s dis-
`ease and other lymphomas, acute leukemia in children,
`and testicular cancer in men; (b) as palliative treatment
`for many types of advanced cancers; (c) as adjuvant
`treatment before or after local treatment (usually sur-
`gery) for primary disease, with the aim of eradicating
`occult micrometastases; and (d) in combination with
`other modalities (usually radiotherapy) in an attempt to
`improve their therapeutic effects.
`Improvements in clinical chemotherapy have de-
`pended largely on the use of drugs in combination.
`Some drugs have been combined because there is a the—
`oretical or experimental basis for expecting synergistic
`interaction through their known mechanisms of action
`either at the molecular level or because of their comple-
`mentary effects on cell-cycle kinetics. Synergy does not
`lead, however, to therapeutic benefit unless the interac-
`tion between drugs is tumor specific (section 19.4.1).
`
`The most important factors underlying the successful
`use of drugs in combination are (a) the ability to com-
`bine drugs at close to full tolerated doses with additive
`effects against tumors and less than additive toxicities to
`normal tissues, and (b) the expectation that drug com-
`binations will include at least one drug to which the tu-
`mor is sensitive.
`
`19.1.2 Therapeutic Index
`
`All anticancer drugs have toxicity as well as antiturnOr
`effects, and toxicity to normal tissues limits the dose 0f
`drugs that can be given to patients. The relationship be-
`tween probability of a biological effect of a drug and ad-
`ministered dose is usually described by a sigmoid cuI‘Ve
`(Fig. 19.1). If the drug is to be useful, the curve describ'
`ing probability of antitumor effect (e.g., complete clin-
`ical remission) must be displaced toward lower doses as
`compared to the curve describing probability of millor
`toxicity to normal tissues (e.g. , myelosuppreSSion lead'
`ing to infection). Therapeutic index (or therapeutic ra-
`tio) may be defined from such curves as the ratio of the
`doses required to produce a given probability of tOX‘
`
`338
`
`

`

`EXPERIMENTAL CHEMOTHERAPY
`
`339
`

`3
`
`o g
`
`S g
`
`b) Normal tissue
`toxicity
`
`
`
`
`Therapeuticindex
`
`l
`ea) TD-05
`l
`ED-50
`
`I
`I
`I
`ED-50 TD-05 TD-5o
`Dose of Drug
`
`| I
`
`1.0
`
`g
`
`it? o 5
`E
`”L
`

`
`Figure 191. Schematic relationships between dose of a drug
`and a the probability of a given measure of antitumor effect,
`and b, the probability of a given measure of normal—-tissue tox-
`icity. The therapeutic index might be defined as the ratio of
`doses to give 50% probabilities of normal-t-issue damage and
`antitumor effects. However, if the endpoint for toxicity is se-
`vere (eg., sepsis due to bone-marrow suppression), it would
`be more appropriate to define the therapeutic index at a
`lower probability of toxicity (e.g, TD-05/ED-5-0.)
`
`icity and antitumor “effect Therapeutic indexIn Fig-
`ure 19.1 might be represented by the ratio of the 5%
`level of probability of severe toxicity (sometimes referred
`to as Toxic Dose-05 or TD——05), and the 50% probabil-
`ity of antitumor effect (i. e. ,effective dose 50 or ED-~50).
`Any stated levels of probability might be used, and the
`appropriate endpoints of tumor response and toxicity
`will depend on the limiting toxicity of the drug, the1n-
`tent of treatment (i e., cure versus palliation), and on
`whether treatment is given to a patient or an experimen-
`tal animal However, dose—response curves similar to
`those of Figure 19.1 have been defined rarely for drug
`effectsin man.
`
`Improvement1n the therapeutic indexis the goal of
`experimental chemotherapy The concept emphasizes
`that any modification1n treatment that leads to in-
`creased killing of tumor cells1n tissue culture or animals
`must be assessed forIts effects on critical normal tissues'
`prior to therapeutic trials.
`
`a “surgically confirmed complete remission” may be
`compatible with the presence of a large number of tu-
`mor cells. Tumor cure requires eradication of all tumor
`cells that have the capacity for tumor regeneration. The
`proportion of such stem cells among those of the tumor
`population18 unknown (see chapter 9, section 9. 2), but
`clinical and even surgically confirmed complete remis-
`sions are compatible with the presence of a substantial
`residual population of tumor stem cells. Attainment of
`complete remission is but a small step toward tumor
`cure.
`
`For many drugs the relationship between cell survival
`and dose is close to exponential, so that a constantfrac-
`tion of the cells (rather than a constant number) is killed
`by a given dose of drug (chapter 17, section 17.2.4).
`Drugs are usually given in sequential courses, with dos-
`age and schedule limited by normal-tissue tolerance.
`Some repopulation of tumor cells may take place be-
`tween courses, so that the number of tumor cells in a
`
`drug-sensitive tumor may change with time during a
`course of chemotherapy, as illustrated in Figure 19.2.
`In this example, each course of drug kills 90% of the tu-
`mor cells, and starting from a large (100 g) tumor, com-
`plete clinical remission is achieved after three courses.
`Note that a further six to ten courses (depending on the
`prevalence of tumor stem cells) would be required to
`achieve cure. Realization of the need to continue aggres-
`sive treatment during complete remission, demonstrated
`originally by the experimental work of Skipper and his
`
`I2
`
`lc
`
`l l l I l I l
`
`'0
`
`I09
`
`
`
`2 3.
`l0 §1:.hi
`I0
`1?,
`we 5
`I. g
`'0 ,3
`
`"Complete remission"
`
`f
`
`l
`Response
`
`l
`i
`Drug resistance
`
`a
`g '0 Treatmenisl
`
`E
`3IO
`
`
`
`Time (months)
`
`"Tumor.
`CUIL
`
`19.1.3 Relationship Between Tumor
`Remission and Cure
`
`l
`
`For most solid tumors the limit of clinical and/or radio-
`logic detectionis about 1 g of tissue (~109 cells). If
`therapy can reduce the number of malignant cells below
`this limit of detection, the patient will be described as
`beingin complete clinical remission. Surgical biopsy of
`sites that were known to be previously involved with tu-
`mor may lower the limit of detection, but a pathologist
`is unlikely to detect sporadic tumor cells present at a fre-
`quency of less than 1in 1000 normal cells, so that even
`
`figure 19.2. Illustration of the relationship between tumor
`remission and cure. In this hypothetical example, treatment
`of a human tumor starts when it has 1011 cells (~100 g), and
`each treatment, given at monthly intervals, kills 90% of the
`cells present. This course of therapy leads to complete disap-
`pearance of clinical tumor. Drug resistance then develops, and
`the tumor grows despite continued treatment. Note that
`despite the attainment of a complete clinical response there
`are always at least 108 viable cells present, and that the
`reduction'In cell numbersIs small compared to that required
`for cure
`
`
`
`

`

`
`
`340
`
`THE BASIC SCIENCE OF ONCOLOGY
`
`led to success in the treatment of
`colleagues (1964),
`acute lymphoblastic leukemia in children, and subse-
`quently to cures in other tumors such as lymphomas.
`Unfortunately, for most solid tumors, a drug-resistant
`subpopulation emerges and leads to relapse, as shown
`in Figure 19.2.
`
`19.2 DRUG RESISTANCE
`
`19.2.1 Mechanisms of Drug Resistance
`
`Many types of cancer that occur commonly in man (e. g. ,
`colon cancer, lung cancer other than small-cell type)
`show only infrequent responses to treatment with anti-
`cancer drugs. This resistance to chemotherapy may be
`influenced by such factors as the proliferative state of
`the cells (chapter 17, section 17.2) and vascular access
`and penetration of drugs into tissue (section 19.3.6), but
`the most important factor is the intrinsic resistance of the
`tumor cells to available anticancer drugs. Other human
`tumors (e.g., breast cancer or small-cell cancer of the
`lung) often respond to initial treatment, but acquired re-
`sistance to further therapy usually prevents drug treat-
`ment from being curative. Thus intrinsic and acquired
`drug resistance are the major factors that limit the suc-
`cessful use of chemotherapy. A wide range of metabolic
`or structural properties of cells may lead to drug re-
`sistance, and some of the mechanisms underlying in-
`trinsic or acquired drug resistance are summarized in
`Table 19.1.
`
`Alkylating agents and cisplatin cause cellular dam—
`age by binding with DNA, leading to cross-linkages
`and breaks in DNA strands (chapter 18, section 18.3).
`Cells may be resistant to these drugs through a num-
`ber of mechanisms, including decreased cellular up-
`take, reduced drug activation, binding of alkylating
`species by sulfhydryl compounds such as glutathione
`(section 19.2.6), increased removal of drug adducts
`from DNA, and increased repair of DNA (see chap-
`ter 4, section 4.4 and chapter 17, section 17.2). Cross-
`resistance to several chemically unrelated drugs has
`been observed for naturally occurring and semisynthetic
`compounds such as doxorubicin and etoposide, due to
`common mechanisms of stimulated drug efflux from
`cells (section 19.2.4), or to decreased activity of the en-
`zyme topoisomerase II that allows conformational
`changes in DNA (section 19.2.5).
`‘A number of mechanisms may also lead to resistance
`to antimetabolite drugs. These mechanisms include im-
`paired drug transport into cells, overproduction or re-
`duced affinity of the molecular target, stimulation of
`alternative biochemical pathways, and impaired activa-
`tion or increased catabolism of the drug. The most
`widely studied of the antimetabolites is methotrexate,
`
`Table 19.1. Probable Mechanisms Associated with
`Resistance to Some Commonly Used
`Anticancer Drugs
`
`
`
`Mechanism Drugs
`
`Increase in proficiency Alkylating agents, cisplatin
`of repair of DNA
`
`Decrease in cellular
`uptake or increase in
`efflux of drugs
`
`Increase in levels of
`“target” enzyme
`
`Cisplatin, doxorubicin, etoposide
`melphalan, 6-mercaptopurine,
`methotrexate, nitrogen mustard,
`vinblastine, vincristine
`Methotrexate
`
`Alterations in “target”
`enzyme
`
`5-F1uorouracil, 6-mercaptopurine,
`methotrexate, 6—thioguanine
`
`Decrease in drug
`activation
`
`Increase in drug
`degradation
`
`Alternative bio-
`chemical pathways
`
`Inactivation of active
`metabolites by binding
`to sulfhydryl
`compounds
`
`Cytosine arabinoside, doxorubicin,
`5-fluorouracil, 6—rnercaptopurine,
`6-thioguanine
`
`Bleomycin, cytosine arabinoside,
`6-mercaptopurine
`
`Cytosine arabinoside
`
`Alkylating agents, cisplatin,
`doxorubicin
`
`Decrease activity of
`topoisomerase II
`
`Amsacrine, doxorubicin, etoposide
`
`
`and the varied mechanisms of resistance to this agent
`will be described as an example for other antimetabo-
`lites in section 19.2.3.
`
`19.2.2 Genetic Basis of Drug Resistance
`
`The following evidence suggests that many types of drug
`resistance are genetic in origin.
`
`1. Characteristics of drug-resistant cells (i.e. , their phe-
`notypes) are often stably inherited in the absence of
`the selecting drug.
`2. Drug-resistant cells are spontaneously generated
`with a rate that is consistent with known rates of ge'
`netic mutation.
`.
`
`3. Generation of drug-resistant cells is increased by ex‘
`posure to compounds (e.g. , ethyl methane sulfonate)
`that induce mutation. This property has been used
`to generate and select a large number of drug-resis‘
`tant mutants that have been used to study drug—resis—
`tant phenotypes (Fig. 19.3).
`4. Altered gene products have been identified in so!ne
`drug-resistant cells, and some of the genes have been
`cloned and sequenced.
`
`l'
`t
`
`1
`
`
`
`u.
`
`

`

`EXPERIMENTAL CHEMOTHERAPY
`EXPERIMENTAL CHEMOTHERAPY
`
`341
`
`MutagenQ Mutagen*9
`
`Mutagen
`
`Chinese Hamster
`Ovary Cells
`
`cultuye in low
`concentration of
`drug
`
`Culture surviving
`cells in intermediate
`concentration of
`drug
`
`Culture surviving
`cells'In high concentration
`of drug
`
`Select clones
`
`1
`
`Select clones
`
`l
`
`1
`
`
`
`Cells with increased levels of drug resistance
`
`figure 19.3. General method used for stepwise selection of drug-resistance mutant
`cells.
`
`
`
`
`
`5. Some drug-resistant phenotypes have been trans-
`ferred to drug-sensitive cells by transfer of genes,
`using techniques described in chapter 3, section 3.3.9.
`
`At least two types of genetic lesion may convey drug
`resistance: point mutation and gene amplification. Drug
`resistance due to point mutation may be expected to oc-
`cur in a single step that is not critically dependent on
`the concentration of drug used to select for resistance,
`and the degree of resistance should be quite stable. One
`of many well-characterized examples is mutation in the
`gene that codes for the enzyme HGPRT (hypoxanthine
`guanine phosphoribosyl transferase), which is neces-
`sary for activation of the drug 6-thioguanine. In con-
`trast to point mutation, gene amplification occurs in a
`stepwise manner, and drug-resistant cells are selected
`more rapidly by exposing cells to graded increases in the
`concentration of the selecting drug. The best-character-
`ized examples (Schimke, 1984; Bradley et al. , 1988) are
`amplification of the gene encoding dihydrofolate reduc-
`tase (DHFR), the target enzyme of methotrexate (sec-
`tion 19.2.3), and amplification of the Mir genes that
`encode P-glycoprotein (section 19.2.4).
`The frequency of occurrence of drug-resistant cells in
`a cell culture or tumor population may be estimated by
`plating the cells after exposure to the drug at a pharma-
`cologically achievable concentration and counting the
`resultant colonies. Typical spontaneous frequencies for
`the incidence of a variety of stable drug-resistant phe-
`notypes are in the range 10—7—10‘5. These frequencies
`are consistent with origin by spontaneous mutation, and
`definitive evidence for this origin has been obtained for
`a few drugs by performing a fluctuation test—a test de-
`vised by Luria and Delbruck (1943) to distinguish be-
`tween (a) mutation and selection and (b) induction by
`a drug, as the major cause of bacteriophage resistance
`
`in bacteria. The fluctuation test has been performed
`using only a few drugs (e.g., ouabain, L-asparaginase,
`and hydroxyurea), but has established mutation and se-
`lection as one major mechanism for generation of drug-
`resistant mutants.
`
`At least two other mechanisms might contribute to
`the generation of genetically based drug resistance.
`Many anticancer drugs (especially alkylating agents) are
`themselves mutagenic: treatment with such drugs may
`be expected to increase the rate of generation of mutant
`cells and to accelerate the development of resistance to
`themselves and to other drugs. Also, drugs that cause
`cell-cycle-progression delay during DNA synthesis may
`allow an increased rate of gene amplification. Thus,
`treatment with some drugs may stimulate the develop-
`ment of drug resistance.
`Second, there is evidence that different populations of
`cells may interact to influence the drug sensitivities of
`each other (e.g. , Heppner, 1984). This influence may
`involve sharing of drug metabolites or increased activa-
`tion of a drug by one of the cell populations. Cell fusion,
`the transfer of DNA between cells, or the uptake of
`DNA from dead cells are also potential mechanisms that
`could lead to horizontal transmission of drug resistance
`between mammalian cells.
`
`Generation of drug-resistant mutants among cells in
`human tumors has implications for planning optimal
`chemotherapy. Goldie and Goldman (1984) have dem-
`onstrated that the probability of there being at least one
`drug-resistant cell in a tumor population is dependent
`on tumor size (Fig. 19.4). This probability increases
`from near zero to near unity over a small range of tu-
`mor sizes (~6 doublings) with the critical size depend-
`ing on the rate of mutation to drug resistance. This
`effect, and others, imply a greater chance of cure if ther-
`apy is begun early, when only microscopic foci of tumor
`
`
`
`

`

`
`
`342
`
`THE BASIC SCIENCE OF ONCOLOGY
`
`0
`
`D
`/.’. —/'__.r0
`0/
`
`/.
`Mutation rate
`=l0‘“
`
`0
`
`Mutation rate
`= IO'6
`
`o
`
`.0ca
`
`9or
`
`.0a
`
`9N Probabilityofat/605/one
`
`drug-resisan/cell Ei-
`
`|
`
`no“
`:02
`Number of cells in tumor
`
`no“
`
`lo“
`
`Figure 19.4. Probability that there will be at least one drug-
`resistant cell in a tumor containing varying numbers of cells,
`based on rates of mutation of 10—6 (open symbols) and 10“4
`(closed symbols) per cell per generation. Note that this prob-
`ability increases from low to high values over a relatively short
`period in the life history of the tumor and that drug-resistant
`cells are likely to be established prior to clinical detection.
`(Adapted from Goldie and Coldman, 1984.)
`
`cells are present (see section 19.3.1). The Goldie—
`Coldman model also predicts a better therapeutic effect
`when two equally effective and non—cross-resistant
`drugs are alternated, rather than given sequentially,
`since this minimizes the emergence of cell populations
`that are resistant to both drugs.
`Although drug-resistant phenotypes in many types of
`cell have been shown to be due to mutation or amplifi-
`cation of genes, this could be due in part to methods
`used for selection of drug-resistant cells. Exposure of
`cells to mutagens, followed by selection in high concen-
`trations of drug (Fig. 19.3) may predispose to selection
`of cells with genetically based drug resistance. Exposure
`of cells to lower concentrations of drugs, without prior
`exposure to mutagens, often leads to cells that show
`transient resistance to drugs; indeed, transient resis-
`tance of some cells in the population may occur spon-
`taneously, without prior drug exposure (Cillo et al.,
`1989). Mechanisms underlying drug resistance that is
`unstable may include the transient amplification of genes
`or changes in patterns of DNA methylation or of other
`factors that influence gene expression (sometimes re-
`ferred to as “epigenetic”). Such changes in drug sensi-
`tivity might be expected to occur in vivo, since the cells
`are exposed to relatively low concentrations of drugs
`during cancer treatment. In one study, repeated treat-
`ment of tumor-bearing mice with alkylating agents led
`to high levels of resistance of the tumor in vivo, but no
`significant resistance was observed when cells derived
`from the tumors were exposed to the same drugs in cul-
`ture (Teicher et al., 1990). It was proposed that changes
`in the tumor cells led to changes in drug distribution in
`
`‘Qif‘l
`w" M
`
`vivo. Clinically important drug resistance is probably
`due to both genetic and nongenetic mechanisms.
`
`19.2.3 Resistance to Methotrexate
`
`Resistance to methotrexate may occur by several mech-
`anisms (Bertino et al., 1981; Fig. 19.5). Methotrexate
`is transported across cell membranes both by passive
`diffusion and by an energy-dependent active transport
`system. Drug-resistant cells may arise that have im-
`paired transport of methotrexate into the cell, probably
`due to point mutation (e.g. , Flintoff et a1. , 1976).
`Transport-deficient cells also show a decrease in poly-
`glutamation of intracellular methotrexate (Frei et al.,
`1984-); primary defects in polyglutamation or in intra-
`cellular binding could lead to decreased uptake of meth-
`otrexate in the drug-resistant mutants.
`A second type of mutation may lead to production of
`variant forms of DHFR, the target enzyme for metho-
`trexate. Variant enzymes have been found to retain ad-
`equate function for reduction of their normal substrate
`(dihydrofolate), but to have a greatly decreased affinity
`for methotrexate (e.g., Goldie et al., 1980).
`The most common mechanism leading to methotrex-
`ate resistance in cell lines and experimental tumors that
`are exposed to increasing concentrations of the drug is
`overproduction of DHFR from amplified genes. This
`process has been extensively characterized by Schimkc
`and his colleagues (e.g., Schimke, 1984-) and their ma-
`jor findings are:
`
`1. Selection for methotrexate resistance in mammalian
`
`cells by stepwise increase in drug concentration in
`
`Decreased formation
`of polyglutamaies
`
`Decreased transport
`'
`
`VarIant forms 0
`of MTX into cell
`
`\DH FR with low
`affinity for MTX
`
`
`|
`
`Methotrexate
`Polygluiamate
`Iormation
`(MTX)
`
`f
`
`Decreased binding
`of MTX to its turn:
`enzyme DHFR
`
`
`1/ Am/plified genes
`
`for DHFR on
`chromosomes or
`double minutes
`(leads to large
`increase in DHFR
`copies)
`
`Figure 19.5. Probable mechanisms underlying cellular resis-
`tance to methotrexate.
`
`

`

`EXPERIMENTAL CHEMOTHERAPY
`
`343
`
`Table 19.2. Drugs Associated with Cross-Resistance and
`Increased Expression of P-Glycoprotein
`
`Actinomycin D
`Colchicine
`Daunorubicin
`Doxorubicin
`
`Etoposide (VP-16)
`Mitoxantrone
`Puromycin
`Vinblastine
`
`Vincristine
`(Adriamycin)
`
`Epirubicin Vindesine
`
`that are selected for resistance to an individual drug of
`the group are cross-resistant to other members of the
`group but usually show the highest degree of resistance
`to the drug that was used for selection. Thus, this mech-
`anism of drug resistance shows some degree of specific-
`ity. Multidrug—resistant cells do not usually show major
`cross-resistance to antimetabolites or to alkylating
`agents, and increased sensitivity has been reported to a
`few drugs, including steroid hormones.
`When membrane proteins from multidrug—resistant
`cells are separated by electrophoresis, they are found
`to contain a glycoprotein whose molecular weight is
`about 170 kDa, which has been termed 'P-glycoprotein
`(Fig. 19.6).
`A direct correlation has been found between the con-
`
`tent of P-glycoprotein in the cell membrane and the de-
`gree of resistance to the selecting drug. Drug resistance
`can be transferred to drug-sensitive cells by transfection
`
`<8
`’0
`3' silage"
`v a? a? 5
`
`Origin...
`
`hi] P—glycoprotein
`'
`
`”Huh
`
`'50"
`IOOL
`
`50:
`
`l
`
`-
`
`
`
`%
`\.
`
`5‘
`
`k

`
`the medium may lead to as many as 100—1000 cop-
`ies of the DHFR gene, and to high levels of drug
`resmtance.
`
`2. High levels of resistance to methotrexate cannot be
`obtained in a single-step selection process. Gene am-
`plification occurs in small steps.
`3. Resistance occurs from overproduction of the normal
`enzyme (although overproduction of variant forms of
`DHFR has also been observed).
`4-. Drug resistance and amplified genes may be either
`stable or unstable when cells are passaged in the ab-
`sence of the drug. Stable amplification is usually as-
`sociated with chromosomal location of the genes,
`seen as homogeneously staining regions in stained
`chromosome preparations (chapter 4-, section 4.3.1)
`Unstable amplification is usually associated with lo-
`cation of the genes in extrachromosomal chromatin
`structures known as double minutes. Both locations
`
`may be evident during selection for drug resistance.
`5. Gene amplification appears to take place by multiple
`replication ofthe DHFR gene (and flanking sequences)
`during the S phase of the cell cycle. Interruption
`of DNA synthesis in synchronized cells by drugs
`such as hydroxyurea, or by transient exposure to
`hypoxic conditions (Rice et al. , 1986), leads to an in-
`crease in the rate of gene amplification. Other anti-
`cancer drugs may increase resistance to methotrexate
`by gene amplification.
`6. Gene amplification and overproduction of DHFR
`have been observed in cells from human tumors
`treated with methotrexate.
`
`Although gene amplification has been studied most
`extensively in relation to methotrexate, there is increas-
`ing evidence for the importance of this mechanism in
`determining resistance to several other drugs, including
`5-fluorouracil and the multiple drug-resistance pheno-
`type described below.
`
`19.2.4 Multiple Drug Resistance
`
`Several investigators have observed that cells which are
`selected for resistance to one of a group of drugs (Ta-
`ble 19.2) may show cross-resistance to each of the other
`drugs in the group (for reviews, see Bradley et al. , 1988;
`Endicott and Ling, 1989). Thus, Chinese hamster ovary
`(CHO) cells selected for resistance to colchicine after
`prior treatment with a mutagen were found subse-
`quently to be cross—resistant to several anticancer drugs,
`including anthracyclines (e.g., doxorubicin) and vinca
`alkaloids (e.g. , Vinblastine), and to other drugs such as
`puromycin, an inhibitor of protein synthesis. Many of
`these drugs have quite different chemical structures, but
`most of them are derived from natural products. Cells
`
`IOIV
`
`Front.
`
`figure 19.6. Increased levels of P—glycoprotein in increasingly
`drug-resistant mutant Chinese hamster ovary cells derived
`from wild type (Aux B1 cells) by stepwise selection (as indi-
`cated in Fig. 19.3). Membrane components were separated by
`SDS gel electrophoresis, transferred to nitrocellulose paper,
`and stained using a radiolabeled hetero-antiserum. (Reprinted
`with permission from Ling, ”Genetic basis of drug resistance
`in mammalian cells.” In: Bruchovsky, Goldie (eds). Drugs and
`Hormone Resistance in Neoplasia. Copyright 1982, CRC Press,
`Boca Raton, FL.)
`
`
`
`

`

`
`
`344
`
`THE BASIC SCIENCE OF ONCOLOGY
`
`of DNA from drug-resistant cells or with cloned genes
`(known as mdr, or multidrug resistance genes) that en-
`code P-glycoprotein, demonstrating a cause-and-effect
`relationship between the presence of P-glycoprotein and
`drug resistance. P-glycoprotein is present at low concen-
`tration in the cell membrane of drug-sensitive cells, and
`drug resistance is most often due to amplification of mdr
`genes with a consequent increase in the production of
`P-glycoprotein. This effect is analogous to the amplifi-
`cation of the DHFR gene, which leads to resistance to
`methotrexate (section 19.2.3). In some multidrug resis-
`tant cells, there is increased transcription of Mr mRNA,
`without amplification of mdr genes.
`The mechanism underlying this type of multidrug re-
`sistance is reduced accumulation of drugs inside the cell.
`Studies using isotopically labeled drug (Fig. 19.7), or
`fluorescent compounds such as doxorubicin, have dem-
`onstrated a decrease in the net rate of uptake, and more
`rapid efflux, of drug from drug-resistant cells. There is
`evidence in some cells that P-glycoprotein may act as a
`permeability barrier to drug uptake, but its major func-
`tion appears to be as an energy-dependent efflux pump
`which extrudes a variety of unrelated compounds from
`cells (Bradley et al., 1988).
`Genetic analysis has demonstrated that P-glycopro-
`teins are produced by a small family of homologous ma’r
`genes, which have been cloned and sequenced from hu-
`man, hamster and murine cells (Gottesman and Pastan,
`1988; Raymond and Gros, 1989; Endicott and Ling,
`1989). Two homologous linked genes (ma’rl and W112
`have been located to chromosome 7 in human cells, but
`only the protein product of ma'rl is associated with drug
`
`resistance. Three ma’r genes have been identified in
`mouse and hamster cells, two of which appear to encode
`a functional P—glycoprotein. The sequences of the hu-
`man and rodent genes show about 80% identity, and
`each gene contains two homologous halves, suggesting
`that they were produced originally by duplication of a
`‘ smaller gene. Small variations in sequence lead to pref-
`erential resistance to different drugs (Safa et a1. , 1990).
`Sequencing of the genes has allowed determination of the
`amino acid composition of the corresponding proteins,
`and the location ofhydrophilic and lipophilic components
`has allowed modeling of the orientation of P-glycopro-
`tein in the cell membrane (Fig. 19.8). This model sug-
`gests that there are 12 transmembrane domains, with 2
`internal ATP-binding sites. P-glycoprotein has signifi-
`cant homology with a number of ATP—binding transport
`proteins found in bacteria, suggesting a general role for
`this type of protein in the energy-dependent efflux of a
`variety of agents from cells.
`Use of monoclonal antibodies against P-glycoprotein,
`or of specific genetic probes, that can bind to mdr
`mRNA in northern blots (chapter 3, section 3.3.4), has
`allowed quantitation of P—glycoprotein, or its mRNA,
`in cells from a variety of tumors and normal tissues.
`P-glycoprotein is present at high levels in the normal hu—
`man kidney and adrenal gland; at intermediate levels in
`lung, liver, colon, and rectum; and at low levels in most
`other ti

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket