throbber
he Basic
`Science of
`
`IPR2017-01122
`
`Genentech 2137
`Genentech 2137 |
`Celltrion v. Genentech
`Celltrion v. Genentech
`IPR2017-01122
`
`

`

`The Basic Science of
`Oncology
`
`Second Edition
`
`Editors
`lan 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, Inc.
`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, orstored in a data base orretrieval system, without the
`prior written permission of the publisher.
`
`1234567890 MAL MAL98765432
`
`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.
`. cm.
`Includes bibliographical references and index.
`ISBN 0-07-105407-3
`1. Cancer.
`2. Oncology.
`Richard P.
`[DNLM: 1. Neoplasms. QZ 200 B3115]
`RC261.B37 1992
`616.99’ 4—dc20
`DNLM/DLC
`for Library of Congress
`
`92-2884CIP
`
`I. Tannock, Ian F.
`
`Il. Hill,
`
`

`

`zs
`
`BOO
`Bal!
`oeek
`
`:
`
`||
`
`)
`
`|
`
`|
`|
`|
`
`|
`
`|
`
`|
`
`Contents
`
`About the Editors and Contributors hee GaSeEe ei
`
`Preface 2.0 eee bee be eee bebe eee een bub ybbbnbbb LEE EEE bbb EER EGS ix
`
`1. ‘Introduction: Cancer as a Cellular Discasesvvssar uns suger ners eee
`Richard P. Hill and Ian F. Tannock
`
`PART 1: CANCER CAUSATION
`
`4, Epidemiology: of Canceraaniesveiaie eeee
`Norman F. Boyd
`
`3. Methods of Genetic Analysis. 00.0000 cee ence een e ee es 23
`Jeremy Squire and Robert A. Phillips
`
`4. Geneétre ‘Basis of Cancereasusianeereewse sey or ee eA
`Jeremy Squire and Robert A. Phillips
`
`5. Oncogene«2...en ee eee eee ee ee ees 61
`Mark D. Minden and Anthony J. Pawson
`
`6. Viruses and: Camcmersrccsera cc 0
`Sam Benchimol
`
`i 5Baa 88
`
`Te
`
`(Gheimical CareiieSEESIS ecco wane osi ane eecsauecsecr qm amine: cecame ese as mem ucelucerce scceuatien sameeren ns 102
`Michael C’. Archer
`a-admition Carcinogenesis nese ieee Wea OE el 1D
`ff A. Michael Rauth
`
`PART 2: CANCER BIOLOGY
`
`®.. Propertics of Malignent Odlescsesecssnasr en ea) Ra eee ee
`Ronald N. Buick and Ian F. Tannock
`
`LO),
`
`Ae Dis
`
`lan F. Tannock
`
`(EZRESPOMTetreHOM fore
`
`cies
`
`cca ceecarcasnerpunanse
`
`cau
`
`ever aesesntg ese citupse tesa epee cetera eerie erent tems
`
`iet
`
`actor
`
`aeraS eo CNET 154
`
`BVASRSUaSS eS Sea acca i Sea pe wie Aa alse tte auc La O
`Richard P. Hill
`
`U2)
`
`SEaraeas" IA earnbeaSa oe sacszer
`Aaron Malkin
`
`perce sseereeaa
`
`npenasscea
`
`gm atcse cata gap ALCS Sar
`
`ae AES TESLA SIS TT STE 196
`
`13; Horriones: and, Catacer'yaisigcicte im csce, goo boase asad wea} gumsmomsracimemsnsmepcmonsensm t@eedumym nein
`DonaldJ. Sutherland and Betty G. Mobbs
`
`smcmcancnsacmonamemeane 207
`
`
`
`

`

`
`
`vi
`
`CONTENTS
`
`14,
`
`Immunology and Immunotherapy of Cancer: pura veepey epee eS eee eee
`Richard G. Miller and Ian F. Tannock
`
`ee 232
`
`PART 3: BIOLOGY UNDERLYING CANCER TREATMENT
`
`Cellular Basis of Radiotherapy.........
`Richard P. Hill
`
`. 259
`
`Experimental Radiotherapy sseeeess eae ee RS OE
`Richard P. Hill
`
`276
`
`Miblopical Properties of Agienmoer TMi 10ers caesar ee RE ET
`lan F. Tannock
`Pheomacology of Anvilcanmes Dreppececscsusesrenanocsnen eu un sv aRUaN RW CEES
`Charles Erlichman
`
`302
`
`317
`
`Eepernmential Chemiethenany sce. can anes eek eae ee RRS
`lan F. Tannock
`
`338
`
`Hyperthennuia: and Photadynamic UhetapyMaia
`Richard P. Hill
`
`Guide to Studies of Diagnostic Tests, Prognosis, and Treatment................
`Norman F. Boyd
`
`155
`
`16.
`
`ihe
`
`18.
`
`19),
`
`20.
`
`21,
`
`Glossary eee a aS
`
`|Xe
`
`403
`
`360
`
`So
`
`O90
`
`

`

`
`
`19
`
`Experimental Chemotherapy
`
`lan 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 Higli-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 currentclinical 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
`treatmentbefore 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 mechanismsof action
`either at the molecularlevel 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 tumorspecific (section 19.4.1).
`
`The most important factors underlying the successful
`use of drugs in combinationare (a) the ability to com-
`bine drugsat close to full tolerated doses with additive
`effects against tumorsandless than additivetoxicities to
`normal tissues, and (b) the expectation that drug com-
`binations will include at least one drug to which the tu-
`moris sensitive.
`
`19.1.2 Therapeutic Index
`
`All anticancer drugs havetoxicity as well as antitumor
`effects, and toxicity to normal tissues limits the dose of
`drugs that can be given to patients. Therelationship be
`tween probability ofa biological effect of a drug and ad-
`ministered doseis usually described by a sigmoid curve
`(Fig. 19.1). If the drugis to be useful, the curve describ-
`ing probability of antitumoreffect (e.g., complete clin-
`ical remission) must be displaced toward lower doses 8
`comparedto the curve describing probability of major
`toxicity to normal tissues (e.g., myelosuppression lead-
`ing to infection). Therapeutic index (or therapeutic 14”
`tio) may be defined from such curvesasthe ratio of the
`doses required to produce a given probability of tox
`
`338
`
`uffr
`
`Wie
`
`ekJui
`
`

`

`EXPERIMENTAL CHEMOTHERAPY
`
`339
`
`
` a) Anti-tumor,
`
`effect b) Normal tissue
`
`| |
`
`1.0
`
`"
`
`a “surgically confirmed complete remission” may be
`compatible with the presence of a large numberof tu-
`morcells. Tumor cure requires eradication of all tumor
`cells that have the capacity for tumor regeneration. The
`2
`toxicity
`
`proportion of such stem cells amongthose of the tumor
`@ 05
`population is unknown(see chapter9, section 9.2), but
`=
`Therapeutic index
`
`a
`|
`_
`TD-05
`clinical and even surgically confirmed complete remis-
`||=©9)-Ep-50
`sions are compatible with the presence ofa substantial
`
`residual population of tumorstem cells. Attainment of
`f
`complete remission is but a small step toward tumor
`cure.
`ED-50 TD-05 TD-50
`Dose of Drug
`
`For many drugstherelationship between cell survival
`and doseis close to exponential, so that a constantfrac-
`tion ofthe cells (rather than a constant number)is killed
`by a given doseof drug (chapter 17, section 17.2.4).
`Drugsare usually given in sequential courses, with dos-
`age and schedule limited by normal-tissue tolerance.
`Somerepopulation of tumorcells may take place be-
`tween courses, so that the numberof tumorcells in a
`drug-sensitive tumor may change with time during a
`course of chemotherapy, asillustrated in Figure 19.2.
`In this example, each course ofdrug kills 90% ofthe tu-
`morcells, and starting from a large (100 g) tumor, com-
`plete clinical remission is achieved after three courses.
`Note thata furthersix to ten courses (depending on the
`prevalence of tumorstem 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
`
`Figure 19.1. Schematic relationships between dose of a drug
`and a, the probability of a given measure of antitumoreffect,
`andb, 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-tissue damage and
`antitumoreffects. However,if the endpoint for toxicity is se-
`vere (e.g., sepsis due to bone-marrow suppression), it would
`be more appropriate to define the therapeutic index at a
`lowerprobability of toxicity (e.g., TD-05/ED-50).
`
`icity and antitumor-effect. Therapeutic index in Fig-
`ure 19.1 might be represented by the ratio of the 5%
`level of probability of severe toxicity (sometimesreferred
`to as Toxic Dose-05 or TD-05), and the 50% probabil-
`ity of antitumoreffect(i.e., effective dose 50 or ED-50).
`Anystated levels of probability might be used, and the
`appropriate endpoints of tumor response andtoxicity
`will depend onthelimiting toxicity of the drug,the in-
`tent of treatment(i.e., cure versus palliation), and on
`whethertreatmentis 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
`effects in man.
`Improvementin the therapeutic index is the goal of
`experimental chemotherapy. The concept emphasizes
`that any modification in treatment that leads to in-
`creased killing of tumorcells in tissue culture or animals
`must be assessedforits effects on critical normal tissues’
`prior to therapeutictrials.
`
`19.1.3 Relationship Between Tumor
`Remission and Cure
`
`For most solid tumors the limit of clinical and/orradio-
`logic detection is about 1 g of tissue (~10° cells). If
`therapy can reduce the numberof malignantcells below
`this limit of detection, the patient will be described as
`being in complete clinical remission. Surgical biopsy of
`sites that were known to be previously involved with tu-
`mor maylowerthelimit of detection, but a pathologist
`is unlikely to detect sporadic tumorcells presentat a fre-
`quencyofless than 1 in 1000 normal cells, so that even
`
`
`
`Numberoftumorcells
`
`
` Limitofclinical detection —
`
`
`“Complete remission’
`
`
`
`3Tumorweight(g/
`
`lo
`
`10° Treatments t
`
`
`}
`Response
`
`t
`
`}
`t
`Drug resistance
`
`
`
`
`r
`
`Tumor,
`cur
`
`Time (months)
`
`Figure 19.2.Illustration of the relationship between tumor
`remission and cure. In this hypothetical example, treatment
`of a human tumorstarts whenit has 10"cells (~100 g), and
`each treatment, given at monthlyintervals, kills 90% of the
`cells present. This course of therapy leads to complete disap-
`pearanceofclinical tumor. Drugresistance then develops, and
`the tumor grows despite continued treatment. Note that
`despite the attainment of a complete clinical response there
`are always at least 10° 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 andleads to relapse, as shown
`in Figure 19.2.
`
`19.2 DRUG RESISTANCE
`
`19.2.1 Mechanisms of Drug Resistance
`
`Manytypesof cancer that occur commonly in man (e.g.,
`colon cancer, lung cancer other than small-cell type)
`show only infrequent responsesto treatment with anti-
`cancer drugs. This resistance to chemotherapy may be
`influenced by such factorsas the proliferative state of
`the cells (chapter 17, section 17.2) and vascular access
`and penetration ofdrugs into tissue (section 19.3.6), but
`the most importantfactor is the intrinsic resistance of the
`tumorcells to available anticancer drugs. Other human
`tumors (e.g., breast cancer or small-cell cancer of the
`lung) often respondto initial treatment, but acquired re-
`sistance to further therapy usually prevents drugtreat-
`ment from being curative. Thusintrinsic and acquired
`drug resistance are the major factorsthat 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, leadingto cross-linkages
`and breaks in DNAstrands(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 compoundssuch as glutathione
`(section 19.2.6), increased removal of drug adducts
`from DNA,andincreased 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
`compoundssuch as doxorubicin and etoposide, due to
`common mechanisms of stimulated drug efflux from
`cells (section 19.2.4), or to decreasedactivity of the en-
`zyme topoisomerase II that allows conformational
`changes in DNA(section 19.2.5).
`‘A number of mechanisms mayalso lead to resistance
`to antimetabolite drugs. These mechanisms include im-
`paired drug transport into cells, overproductionor 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,
`
`Ao
`
`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
`
`Cisplatin, doxorubicin, etoposide
`melphalan, 6-mercaptopurine,
`methotrexate, nitrogen mustard,
`vinblastine, vincristine
`Methotrexate
`
`Increase in levels of
`“target” enzyme
`Alterations in “target”
`enzyme
`Decrease in drug
`activation
`
`5-Fluorouracil, 6-mercaptopurine,
`methotrexate, 6-thioguanine
`Cytosine arabinoside, doxorubicin,
`5-fluorouracil, 6-mercaptopurine,
`6-thioguanine
`Bleomycin, cytosine arabinoside,
`6-mercaptopurine
`Cytosine arabinoside
`
`Increase in drug
`degradation
`Alternative bio-
`chemical pathways
`Inactivation of active
`metabolites by binding
`to sulfhydryl
`compounds
`Amsacrine, doxorubicin, etoposide
`Decrease activity of
`topoisomerase IT
`
`Alkylating agents, cisplatin,
`doxorubicin
`
`and the varied mechanismsofresistance 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
`
`Thefollowing evidence suggests that manytypes of drug
`resistance are genetic in origin.
`
`1. Characteristics of drug-resistantcells (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 thatis consistent with knownrates of ge-
`netic mutation.
`!
`3. Generation of drug-resistantcells is increased by €%-
`posure to compounds(e.g., ethyl methane sulfonate)
`that induce mutation. This property has been used
`to generate andselect a large number of drug-resis-
`tant mutants that have been usedto study drug-resis-
`tant phenotypes (Fig. 19.3).
`4. Altered gene products have beenidentified in some
`drug-resistantcells, and someof the genes have bee®
`cloned and sequenced.
`
`

`

`
`
`EXPERIMENTAL CHEMOTHERAPY
`EXPERIMENTAL CHEMOTHERAPY
`
`341
`
`———_—
`
`Mutagen LD MutagenDD DP
`
`—_— —
`
`Chinese Hamster
`Ovary Cells
`
`Culture in low
`concentration of
`drug
`
`Culture surviving
`cells in intermediate
`concentration of
`drug
`
`Culture surviving
`cells in high concentration
`of drug
`
`Setect clones
`
`|
`
`Selectclones
`
`|
`
`|
`
`
`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 chapter3, section 3.3.9.
`
`Atleast 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 usedto 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 exposingcells to graded increases in the
`concentration ofthe 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 mdr genes that
`encode P-glycoprotein (section 19.2.4).
`The frequency of occurrence of drug-resistantcells 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-
`notypesare in the range 10-’-10~°. 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 bacteriophageresistance
`
`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.
`Manyanticancer 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 developmentofresistance 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 maystimulate the develop-
`ment of drug resistance.
`Second, thereis evidence that different populations of
`cells may interact to influence the drugsensitivities of
`each other (e.g., Heppner, 1984). This influence may
`involve sharing of drug metabolites or increased activa-
`tion of a drug by oneofthecell populations. Cell fusion,
`the transfer of DNA between cells, or the uptake of
`DNAfrom dead cells are also potential mechanismsthat
`could lead to horizontal transmission of drug resistance
`between mammalian cells.
`Generation of drug-resistant mutants amongcells in
`human tumors has implications for planning optimal
`chemotherapy. Goldie and Coldman (1984) have dem-
`onstrated that the probability of there being at least one
`drug-resistantcell 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-
`morsizes (~6 doublings) with the critical size depend-
`ing on the rate of mutation to drug resistance. This
`effect, and others, imply a greater chanceofcure if ther-
`apy is begun early, when only microscopicfoci of tumor
`
`
`
`

`

`
`
`342
`
`THE BASIC SCIENCE OF ONCOLOGY
`
`oON Probabilityofatleastone
`
`drug-resisantcell oa
`
`fo
`‘waa rate
`
`=10°6
`
`——®
`
`Oo
`
`3
`
`°@
`
`°a
`
`2°B
`
`ae=:
`
`i
`
`10*
`
`107
`Numberofcells in tumor
`
`10°
`
`10°
`
`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-® (open symbols) and 107+
`(closed symbols) percell per generation. Note that this prob-
`ability increases from low to high values overa relatively short
`period in thelife history of the tumorand 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 modelalso predicts a better therapeutic effect
`when two equally effective and non-cross-resistant
`drugs are alternated, rather than given sequentially,
`since this minimizes the emergenceofcell populations
`that are resistant to both drugs.
`Although drug-resistant phenotypes in many types of
`cell have been shownto 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 byselection in high concen-
`trations of drug (Fig. 19.3) may predisposeto selection
`of cells with genetically based drug resistance. Exposure
`ofcells 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 somecells in the population may occur spon-
`taneously, without prior drug exposure (Cillo et al.,
`1989). Mechanisms underlying drugresistance that is
`unstable may includethe transient amplification ofgenes
`or changesin patterns of DNA methylation or of other
`factors that influence gene expression (sometimesre-
`ferred to as “epigenetic”). Such changes in drug sensi-
`tivity might be expected to occurin vivo,since thecells
`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 tumorin vivo, but no
`significant resistance was observed whencells derived
`from the tumors were exposed to the same drugsin cul-
`ture (Teicheret al., 1990). It was proposed that changes
`in the tumorcells led to changes in drugdistribution in
`
`
`
`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-dependentactive transport
`system. Drug-resistant cells may arise that have im-
`paired transport of methotrexate into thecell, probably
`due to point mutation (e.g., Flintoff et al., 1976).
`Transport-deficient cells also show a decrease in poly-
`glutamation of intracellular methotrexate (Frei etal.,
`1984); primary defects in polyglutamationorin intra-
`cellular binding could lead to decreased uptake of meth-
`otrexate in the drug-resistant mutants.
`A second type of mutation maylead to production of
`variant forms of DHFR,the target enzyme for metho-
`trexate. Variant enzymes have been foundto retain ad-
`equate function for reduction of their normal substrate
`(dihydrofolate), but to have a greatly decreasedaffinity
`for methotrexate (e.g., Goldie et al., 1980).
`The most common mechanism leading to methotrex-
`ate resistancein cell lines and experimental tumors that
`are exposed to increasing concentrations of the drugis
`overproduction of DHFR from amplified genes. This
`process has been extensively characterized by Schimke
`and hiscolleagues (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
`
`Seee
`
`if
`
`Decreased formation
`of polyglutamates
`Decreased transport
`
`i
`Variant forms o!
`of MTX into cell
`
`“oH FR with low
`
`affinity for MTX
`
`
`
`|
`Methotrexate
`Polyglutomate
`
`
`(MTXKw ) formation
`
`
`Decreased binding
`of MTX to its target
`enzyme OHFR
`
`
`
`Artplitied genes
`for DHFR on
`chromosomes or
`double minutes
`(leads to large
`increase in OHFR
`copies)
`
`Figure 19.5. Probable mechanisms underlying cellular resis-
`tance to methotrexate.
`
`

`

`EXPERIMENTAL CHEMOTHERAPY
`
`343
`
`the medium may lead to as many as 100-1000 cop-
`ies of the DHFR gene, and to high levels of drug
`resistance.
`2. Highlevels 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
`DHFRhasalso been observed).
`4, Drugresistance and amplified genes may be either
`stable or unstable whencells 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
`chromosomepreparations (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 duringselection for drug resistance.
`5, Gene amplification appears to take place by multiple
`replication ofthe DHFR gene(andflanking 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 mayincrease resistance to methotrexate
`by gene amplification.
`6. Gene amplification and overproduction of DHFR
`have been observed in cells from human tumors
`treated with methotrexate.
`
`Table 19.2. Drugs Associated with Cross-Resistance and
`Increased Expression of P-Glycoprotein
`
`Etoposide (VP-16)
`Actinomycin D
`Mitoxantrone
`Colchicine
`Puromycin
`Daunorubicin
`Vinblastine
`Doxorubicin
`Vincristine
`(Adriamycin)
`
`Epirubicin Vindesine
`
`that are selected for resistance to an individual drug of
`the group are cross-resistant to other membersofthe
`group but usually show the highest degree ofresistance
`to the drug that was used for selection. Thus, this mech-
`anism of drug resistance shows some degreeofspecific-
`ity. Multidrug-resistant cells do not usually show major
`cross-resistance to antimetabolites or to alkylating
`agents, and increasedsensitivity has been reported to a
`few drugs, including steroid hormones.
`When membraneproteins 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 membraneandthe de-
`gree ofresistance to the selecting drug. Drugresistance
`can be transferred to drug-sensitive cells by transfection
`
`Sf
`4
`x ate&
`eT eeég
`
`Although gene amplification has been studied most
`extensively in relation to methotrexate, there is increas-
`ing evidence for the importance of this mechanism in
`determiningresistance to several other drugs, including
`5-fluorouracil and the multiple drug-resistance pheno-
`50
`> > aatee
`
`type described below.
`*&ee
`
`ee | P-glycoprotein
`<
`
`Origin
`pa
`100
`
`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
`
`>
`=
`
`L
`
`10
`
`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-
`catedin Fig. 19.3). Membrane components were separated by
`SDSgel electrophoresis, transferred to nitrocellulose paper,
`and stained using a radiolabeled hetero-antiserum.(Reprinted
`with permission from Ling, “Genetic basis of drug resistance
`in mammaliancells.” In: Bruchovsky, Goldie (eds.). Drugs and
`HormoneResistance 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
`(knownas mdr, or multidrug resistance genes) that en-
`code P-glycoprotein, demonstrating a cause-and-effect
`relationship between the presence of P-glycoprotein and
`drugresistance. P-glycoprotein is present at low concen-
`tration in the cell membraneof drug-sensitivecells, 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 DHFRgene, which leadsto resistance to
`methotrexate (section 19.2.3). In some multidrug resis-
`tant cells, there is increased transcription of mdr mRNA,
`without amplification of mdr genes.
`The mechanism underlyingthis type of multidrugre-
`sistance is reduced accumulation of drugsinside thecell.
`Studies using isotopically labeled drug (Fig. 19.7), or
`fluorescent compoundssuch 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 somecells that P-glycoprotein may act as a
`permeability barrier to drug uptake, but its major func-
`tion appearsto be as an energy-dependentefflux pump
`which extrudesa 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 mdr
`genes, which have been cloned and sequenced from hu-
`man, hamster and murinecells (Gottesman and Pastan,
`1988; Raymond and Gros, 1989; Endicott and Ling,
`1989). Two homologous linked genes (mdr1 and mdr2
`have been located to chromosome7 in humancells, but
`only the protein product of mdr1 is associated with drug
`
`resistance. Three mdr genes have been identified in
`mouse and hamstercells, 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 homologoushalves, suggesting
`that they were produced originally by duplication of a
`’ smaller gene. Small variations in sequenceleadto pref-
`erential resistance to different drugs (Safaet al., 1990).
`Sequencingofthe geneshas allowed determination ofthe
`amino acid composition of the correspondingproteins,
`andthe location ofhydrophilic and lipophilic components
`has allowed modeling ofthe orientation of P-glycopro-
`tein in the cell membrane(Fig. 19.8). This modelsug-
`gests that there are 12 transmembrane domains, with 2
`internal ATP-bindingsites. P-glycoprotein hassignifi-
`cant homology with a number of ATP-bindingtransport
`proteins found in bacteria, suggesting a general role for
`this type of protein in the energy-dependentefflux of a
`variety of agents from cells.
`Use of monoclonal antibodies against P-glycoprotein,
`or of specific genetic probes, that can bind to mdr
`mRNAin 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; andatlow levels in most
`other tissues (Fojo et al., 1987). Immunohistochemical
`techniques have shownthat P-glycoproteinis localized
`to the surfaces o

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