throbber

`
`
`
`
`
`-
`
`Hospira v. Genentech
`Hospira v. Genentech
`IPR2017-00737
`IPR2017—00737
`Genentech Exhibit 2038
`
`Genentech Exhibit 2038
`
`

`

`Principles and Practice
`
`Cancer Chemotherapy and Biotherapy:
`
`

`

`
`
`Cancer Chemotherapy
`and Biotherapy:
`Principles and Practice
`
`SECOND EDITION
`
`
`
`EDITED BY
`
`Bruce A. Chabner, M.D.
`Chief, Hematology/Oncology
`Clinical Director
`
`Massachusetts General Hospital Cancer Center
`Boston, Massachusetts
`
`Dan L. Longo, M.D.
`Director, Biological Response Modifiers Program
`Division of Cancer Treatment
`National Cancer Institute
`
`Frederick, Maryland
`
`
`
`
`n Lippincott - Raven
`
`
`
`Philadelphia - New York
`
`_It______
`
`

`

`
`
`Copyright © 1996 by Lippincott—Raven Publishers. All rights reserved. This book is protected by copy-
`right. No part of it may be reproduced, stored in a retrieval system, or transmitted, in any form or by
`any means—electronic, mechanical, photocopy, recording, or otherwise—without the prior written
`permission ofthe publisher, except for brief quotations embodied in critical articles and reviews. Printed
`in the United States of America. For information write Lippincott—Raven Publishers, 227 East Wash—
`ington Square, Philadelphia, PA 19106.
`
`Library of Congress Cataloging-in-Publications Data
`
`Cancer chemotherapy and biotherapy : principles and practice / edited
`by Bruce A. Chabner, Dan L. Longo -—- 2nd ed.
`p.
`cm.
`Rev. ed. of: Cancer chemotherapy.
`Includes bibliographical references and index.
`ISBN 0—397-514184 (hard : alk. paper)
`1. Cancer—Chemotherapy.
`2. Cancer—Immunotherapy.
`(Dan Louis), 1949—
`.
`II. Cancer chemotherapy.
`[DNLM: 1. Neoplasms—drug therapy.
`2. Biological Products—therapeutic use.
`3. Antineoplastic Agents—therapeutic use. 4. Chabner, Bruce. QZ 267 C21515 1996]
`RC271.CSC32219
`1996
`616.99’4061—dc20
`DNLM/DLC
`for Library of Congress
`
`I. Longo, Dan L
`
`95-38920
`CIP
`
`The material contained in this volume was submitted as previously unpublished material, except in
`the instances in which credit has been given to the source fi'om which some of the illustrative material
`was derived.
`Great care has been taken to maintain the accuracy ofthe information contained in the volume. How—
`ever, neither Lippincott—Raven Publishers nor the editors can be held responsible for errors or for any
`consequences arising from the use of the information herein.
`The authors and publisher have exerted every effort to ensure that drug selection and dosage set forth
`in this text are in accord with current recommendations and practice at the time of publication. How-
`ever, in view of ongoing research, changes in government regulations, and the constant flow of infor-
`mation relating to drug therapy and drug reactions, the reader is urged to check the package insert for
`each drug for any change in indications and dosage and for added warnings and precautions. This is
`particularly important when the recommended agent is a new or infrequently employed drug.
`Materials appearing in this book prepared by individuals as part of their official duties as US.
`Government employees are not covered by the above-mentioned copyright.
`
`987654321
`
`

`

`L553..
`
`Cancer Chemotherapy and Mot/army, second edition,
`edited by Bruce A. Chabner and Dan L. Longo.
`Lippincott—Raven Publishers, Philadelphia ©1996
`
`
`
`_-
`
`_
`
`..
`
`Platinum Analogues
`Eddie Reed, Meenakshee Dabholkor, and Bruce A. ChObner
`
`..
`
`.
`
`
`
`
`
`The antitumor activity ofthe platinum complexes was dis-
`covered as the result ofa fortuitous observation by Rosen-
`berg et all,2 during a study of the effects of electric
`current on growing bacteria. When alternating current
`was delivered through platinum electrodes to a growing
`bacterial culture, the bacterial cells stopped dividing and
`grew into long filaments. The same result was seen when
`an attempt was made to grow the bacteria in fresh
`medium that previously had been subjected to the elec—
`tric current. Because filamentous growth was known to
`occur in bacteria subjected to alkylating agents or radia-
`tion, Rosenberg suspected that an active substance may
`have entered the medium, possibly through the release of
`soluble platinum compounds from the electrodes. It was
`found that platinum was released by electrolysis as hexa—
`chloroplatinate, which, in the presence of ammonium
`salts and light, generated the platinum complex cis-
`Pt(II)(NH3)2C12, which is now known as the anticancer
`drug cisplatin. This agent has become the primary build—
`ing block for regimens that cure patients with testicular
`carcinoma and produces high response rates in patients
`with small cell carcinoma ofthe lung, bladder cancer, and
`ovarian cancer. The key features of cisplatin and its active
`and less nephrotoxic analogue, carboplatin, are shown in
`Tables 14—1 and 14-2.
`
`CHEMISTRY
`
`have fixed bond angles and spacial configuration. The
`spacial configuration depends on whether the oxidation
`state of the platinum is +2 or +4; these oxidation states
`are usually designated as Pt(II) or Pt(IV), respectively. In
`Pt(II) complexes, the platinum atom has four bonds
`directed to the corners of a square at which the four
`ligand atoms are located (Fig. 14-1). In Pt(IV) complexes,
`there are six bonds and six ligands: four in a planar square
`configuration as in Pt(II), plus one directly above and
`one directly below the platinum atom, an octahedral con~
`figuration. Because the bonds are fixed and not readily
`exchangeable, the complexes can have distinct isomers,
`such as cis— and tram-Pt(II)(NH3)2C12 (Fig. 14-1). The
`cis isomer is the antitumor drug cisplatin. The importance
`of steric conformation is highlighted by the fact that the
`trans isomer has virtually no antitumor activity.
`Like covalent bonds in general, the bonds to the plat-
`inum atom tend to have energy barriers that limit the rate
`ofbond formation or dissociation. Some platinum bonds,
`such as those to nitrogen, are essentially irreversible under
`physiologic conditions, while others, such as the chloride-
`platinum(II) bond in cisplatin, are more labile.
`Displacement reactions can occur in Pt(II) complexes
`in which one or both ligands are displaced by a compet-
`ing nucleophile, in analogy to the reactions of alkylating
`agents. Thus the Pt(II) complexes have significant chem-
`ical and biologic similarities to alkylating agents.
`
`Covalent Bond Character
`
`Oxidation States
`
`Platinum is in the third row of transition metals in the
`periodic table and has eight electrons in the outer d shell.
`Palladium and nickel, which occupy analogous positions
`in the second and first transition series, have similar con-
`figurations of outer electrons but little or no antitumor
`activity. Because the platinum atom has a much larger
`total number of electrons, however, the orbitals of its
`- Outer electrons are more polarizable, and bonds that are
`formed from these orbitals have more covalent character.
`The covalent nature of these bonds confers two essen-
`tial properties, stereospecificity and energy barriers. Stereo-
`Schzficity means that the bonds from the platinum atom
`
`Although both Pt(II) and Pt(IV) complexes can have
`potent antitumor activities, it is unclear whether Pt(IV)
`complexes may undergo ligand displacement reactions
`and form covalent bonds under physiologic conditions.
`The activity of Pt(IV) complexes may be due to their
`reduction in vivo to the active Pt(II) state.3
`
`Displacement Reactions
`
`The biologic actions of Pt(II) complexes are due to
`displacement reactions, which cause the platinum to
`become stably bound to DNA, RNA, proteins, or other
`
`357
`
`
`
`@3315“s
`a?“
`
`\AaGiant?i
`"tauaver-.mm
`
`
`
`
`
`..-.;.a.znull-
`
`

`

`358
`
`CANCER CHEMOTHERAPY AND BIOTHERAPY
`
`
`
`Table 14-]. Key Features of Cisplatin
`
`
`
`Metabolism:
`
`Pharmacokinetics:
`
`Elimination:
`
`
`
`ligands such as sulfur. This type of mechanism could
`
`Dose: 20mg/m2/day times 5 days every 3—4 weeks
`Mechanism of action: Covalently binds to DNA bases and disrupts DNA funcu'on; In some settings, toxicity may be related to_drug
`binding to proteins.
`Inactivated intracellularly and in the blood stream by sulfhydryl groups. Drug covalently binds to glutathione,
`metallothioneins, and sulfhydryls on proteins.
`After IV bolus,
`‘1/2a =
`tl/ZB = approx 60 min
`t1fl'y = approx 24 h
`After “high dose,” two: = 30 min
`Approximately 25% of an administered dose is excreted from the body during the first 24 hours. Of that
`portion eliminated, excretion is: renal >90% and bile <10%.
`Systemic thiosulfates (IV thiosulfate, possibly WR—2721, possibly diethyldithiocarbamate) may theoretically
`inactivate drug systemically.
`Renal insufliciency with Mg2+ wasting
`Nausea and vomiting
`Peripheral neuropathy
`. Auditory impairment
`Myelosuppression (thrombocytopenia primarily; WBC suppression seen with carboplatin)
`. Visual impairment (rare)
`. Hypersensitivity (rare)
`. Seizures (rare)
`Use with caution in the presence of other nephrotoxic drugs (aminoglycosides, etc)
`. Monitor electrolytes, Mg“, and Ca2+
`Maintain high urine flow during therapy
`Aggressive premedicau'on with antiemetics is usually helpful (ondansetron or metoclopromide i decadron)
`Full dose can be administered if 24-hour creatinine clearance is >60 ml/h and patient can tolerate vigor-
`ous hydration. If 24-hour creatinine clearance is less than 60 ml/h, carboplatin (also given with vigorous
`hydration) should be considered.
`
`Drug interactions:
`
`Toxicity:
`
`Precautions:
`
`
`
`mesaer°°\I°~sm>s~.~re
`
`alkylating agents such as nitrogen mustards. Ligand
`displacement reactions in platinum complexes occur with
`retention of the spatial configuration of the bond angles,
`in contrast to alkylating agents, where configuration typ-
`ically becomes inverted.
`The stability of the binding of different ligands to
`Pt(II) varies greatly. Although the binding to sulfur or
`nitrogen sites is essentially irreversible under physiologic
`conditions, the stability of binding to halogens is much
`lower, in the order I‘ > Br‘ > Cl‘, and the stability of
`binding to H20 is weaker still.
`The selectivity among different sites of binding, how-
`ever, may be determined more by the relative reaction
`rates than by the equilibrium binding stabilities. The bind-
`ing to sulfhydryl groups generally is very rapid as well as
`very strong. The binding to amines, however, is relatively
`slow, even though it is strong. Binding may, ofcourse, be
`hindered by steric factors. Another important determinant
`of displacement reactions is the rate of dissociation.
`The dissociation of sulfur or nitrogen ligands is gener-
`ally negligible under physiologic conditions (except pos-
`sibly as noted below). The aquo (H20) ligand dissociates
`rapidly, whereas chloride dissociates more slowly. The
`dissociation rate also can be influenced by the identity of
`the other ligands in the complex, especially by the ligand
`located trans to the dissociating ligand. This “kinetic
`trans effect” is exerted most strongly by highly polarizable
`
`critical biomolecules. All the antitumor active complexes
`are bifunctional in that they can form, by successive dis—
`placement reactions, two stable bonds under physiologic
`conditions so as to produce a covalent cross-link between
`two nucleophilic atoms of a macromolecule. In this
`respect, the Pt(II) complexes are similar to bifunctional
`
`
`
`'—
`Table 14-2. Key Features of Corboplafin
`
`sees.
`
`i
`
`3‘82
`ism
`efi4n
`
`_sun-rdf
`
`
`
`Pharmacokinetics:
`
`365 mg/m2 every 3—4 weeks
`Dose:
`Mechanism of action: Covalent binding to DNA
`Metabolism:
`Drug is primarily excreted. Only
`a small fraction is metabolized
`After IV bolus,
`t1not = 12—24 min
`rl/ZB = 1.3—1.7 h
`‘1/2'Y =
`h
`Approximately 90% excreted in
`the urine in 24 h
`
`Elimination:
`
`Toxicity:
`
`Drug interactions:
`
`None described in detail; see
`cisplan'n
`1. Myelosuppression, especially
`thrombocytopenia
`2. Nausea and vomiting
`3. Nephrotoxicity at high doses
`and in patients with prior renal
`dysfunction
`1. Reduce dose in proportion to
`Precautions:
`reductions in creatinine clearance
`
`
`
`
`

`

`Platinum Analogues
`
`Figure 14-]. Cisplatin, the inactive isomer transplatin, and
`other platinum analogues of clinical interest. While most
`compounds have platinum in the (II) valence state, note the
`platinum(lV) derivatives ormaplatin and tetraplatin; also
`note the several analogues that contain a stabilizing cyclo-
`hexane group attached to the platinum molecule.
`
`CI
`
`NH3
`
`CISPLATIN fl
`
`Cl
`
`NH3
`
`
`
`marized in Fig. 14—2. Although it is possible that a chlo—
`ride ligand in cisplatin might in some cases be displaced
`directly in a reaction with a macromolecule, it is generally
`agreed that the more usual path is via an initial aquation
`reaction in which a chloride is replaced by a water mole-
`cule. The aquation reaction is driven by the high concen—
`tration of water and low concentration of chloride in the
`
`tissues. The aquated platinum complex can then react
`rapidly with a variety of strong binding sites.
`The equilibrium reaction in water (Fig. 14-2) may be
`of pharmacologic importance, not only because of the
`high reactivity of the aquated species but also because of
`the different ionic states that may affect the ability of
`particular species to penetrate through lipid membranes.
`The uncharged species,
`including the dichloro- and
`chlorohydroxy complexes, would be expected to pene-
`trate membranes most easily; these species are shown with
`a zero charge exponent in Fig. 14-2.
`The equilibrium and rate constants of the aquation
`reactions (reactions 1 and 2 in Fig. 14-2) are listed in
`Table 14-3. The values listed indicate that for 4.4 mM
`
`cisplatin in water at 37°C, the first aquation occurs with
`a half-time of 2.5 hours and would eventually reach an
`equilibrium ratio of 1:1 for the dichloro:chloroaquo
`species. Because of the slowness of the reaction and the
`presence of reactive constituents other than water, it is
`unlikely that this equilibrium would be approached in
`biologic systems.
`In blood plasma, the high chloride concentration of
`approximately 100 mM would keep cisplatin predomi-
`nantly in the uncharged and relatively unreactive dichloro
`form. This form may react to some degree with sulfhydryl
`groups of plasma proteins. The fi'ee dichloro form could
`enter cells by passive diffusion. In the cytoplasm, the
`relatively low chloride concentration of approximately
`4 mM would favor the aquation reaction, which would
`yield highly reactive species whose ionic charge may
`retard exit from the cell.
`
`Reactions with DNA
`
`Cisplatin has been noted to bind to all DNA bases, but
`in intact DNA there seems to be preferential binding to
`the N—7 positions of guanine and adenine?»5 This may be
`due to the high nucleophilicity of the imidazole ring,
`particularly at the N-7 position.” Cisplatin binds to RNA
`more extensively than to DNA and to DNA more than to
`protein when binding is assessed as moles of drug per
`gram of macromolecule.8
`In the reaction of cisplatin with DNA or other macro-
`molecules, the two chloride ligands can (usually after aqua-
`tion) react with two diEerent sites to produce cross—links.
`The bond distance between the chloride leaving groups of
`cisplatin is fixed at approximately 3 A5 as compared with
`the corresponding bond distance for alkylating agents,
`which can range between 7 and 10 A. A more complete
`description of various aspects of the chemistry of cisplatin
`can be found in several excellent recent reviews.4’9’1°
`
`“JV‘V
`
`G
`
`1
`
`tmflflfifl
`.1.“l»1
`
`
`
`333,};“iii
`
`t
`
`'33.
`is;‘ 74:1
`
`
`
`TRANSPLATINUM
`
`CI
`
`NH3
`
`flNH3
`
`01
`
`CI
`
`NH2\ | / c:
`/Pt\
`1
`C]
`
`Cl
`
`~~NH2
`

`
`""3
`
`o \
`
`‘\3 \
`
`TETRAPLATIN
`
`CARBOPLATIN
`
`PLATINUM-DACH
`
`ORMAPLATIN
`
`OXALIPLATIN
`
`o‘c\
`
`CI
`
`a
`
`NH3
`
`NH2 H
`
`Cl
`
`NH2
`H
`
`NH2
`
`ell
`CE :Pt<
`
`NH;
`
`l
`c:
`
`cu
`
`c:
`
`o
`NH2\ /0 —c/
`/P1\
`l
`° _ C§O
`
`""2
`
`CH3
`
`
`
`
`NHz\m/°
`
`NH2/
`
`\0
`
`0
`
`j
`
`I- 73
`
`c 9
`
`Pcrhaps allow some platinum ligands to be dislodged
`fl0m tight binding sites_under physiologic conditions
`and may account for the biologic difl‘erences of various
`Platinum complexes.
`
`Reactions in Water and Biologic Fluids
`
`The pharmacologic behavior of cisplatin is in part
`determined by its reactions in water, which are sum—
`
`
`
`

`

`360
`
`CANCER CHEMOTHERAPY AND BIOTHERAPY
`
`
`7
`
`Figure 14-2. Aquation and hydrolysis equilibria of cisplatin (pKa values are from ref. 31 3). Note that reactions 3 and 6 are favored
`at physiologic pH and yield products that have a neutral charge and that theoretically could readily cross cell membranes. The rate
`constants tor the initial aquation reactions (1 and 2) are listed in Table 14-3.
`
`
`o
`
`+
`
`H3N\ ,0
`pl
`H3N/
`‘cn
`dichtoro
`
`,ow2
`1
`. H3N\
`‘—
`\CI
`HaN/
`chbtoaqm
`
`P!
`
`3
`
`pK.~7.4
`
`+ +
`
`,OH2
`.__2_.. H3N\
`w—-
`’Pt\
`H3N
`OH2
`
`
`
`
`
`+
`H
`“3” \Pt ’0“ —7-8 “WM/ONR ’NH3
`I \0/ \NH
`dimer
`
`+ +
`
`are consistent with earlier reports from Lippard’s group.
`Using an analysis based on gel electrophoretic mobility,
`reactivity with chemical probes, and molecular mechan-
`ics, Sip et al.13 have demonstrated that a centrally placed
`interstrand cross—link in a 22-mer DNA duplex bends the
`double helix by approximately 55 degrees toward the
`major groove. This results in exposing the cytosine
`residues but does not result in local denaturation within
`the flanking sequences.
`A similarly placed intrastrand d(GpG) adduct bends
`DNA to a lesser extent.14 The frequency of specific inter-
`and intrastrand adducts found in mammalian cells after
`exposure to platinum derivatives is shown in Table 14-4.
`
`
`
`Table 14-4. Types oi DNA Lesions Caused by
`
`Cis latin and Carbo lat'n
`P
`P
`l
`
`PERCENT
`OF TOTAL
`DNA
`DAMAGE
`
`DNA LESION
`
`N7— d(GpG)-intrastrand
`adduct
`
`N7- d(ApG)—intrastrand
`adduct
`
`N7- d(GpoG)—intrastrand
`adduct
`
`~60%
`
`~ 30%
`
`~10%
`
`
`
`N7- d(X)-d(X)-interstrand
`cross-link
`
`~ < 1%
`
`Important in
`gene-specific
`
`repair studies
`
`COMMENT
`
`Possibly lethal
`to cells
`
`Possibly lethal
`to cells
`
`Potential lethality
`unclear
`
`_‘l
`
`
`
`Using x-ray crystallographic techniques, Sherman
`et a1.11 studied the crystal and molecular structure of the
`intrastrand deG diammine-platinum adduct, which
`comprises approximately 60% of total platinum binding
`to DNA following in Vitro or in vivo drug exposures.
`These studies show that when cisplatin is covalently
`bound to the d(GpG) dinucleotide, the conformation of
`the dinucleotides largely depends on the stereochemical
`requirements for maintaining the planar platinum coor-
`dination relationship. Further, it appears that the forma—
`tion of an intramolecuiar hydrogen bond between a
`proton of the amine ligand and an oxygen atom of the
`terminal 51-phosphate contributes to the stabilization of
`the platinum dinucleotide complex.
`Kozelka and Chottard12 studied cisplatin binding to
`defined double-stranded decanucleotides and showed
`that cisplatin causes kinking of the helix by approximately
`60 degrees. The bend occurs toward the major groove
`and is associated with limited unwinding. These findings
`
`
`
`Table “'3' EqUilibfiUm and Rate Constants For
`Aquation oi Cisplafinalz—sis
`
`EQUILIBRIUM
`REACTION CONSTANT, (mM)
`(REFERTO
`FIG. 14-2)
`
`AT 25°C AT 37°C AT 25°C AT 37°C
`
`RATE OF FORWARD
`REACTION(t1/2, H)
`
`
` 1
`
`2
`
`3.6
`0.11
`
`4.4
`0.19
`
`7.7
`5.8
`
`2.5 —i
`
`

`

`Inwith:jgig-gum\aw"
`atiiiHRH
`
`—«‘hé‘!ktnhaltai‘5‘:
`E:-.‘.sWfififiéflmmaa
`
`i-
`
`5%.;
`its;
`
`''Jfi'glé
`'5
`
`Platinum Analogues
`
`METHODS OF MEASUREMENT
`
`Shortly after the observation that cisplatin reacts with
`DNA, methods began to be developed to measure the
`binding of drug to DNA and the production of particu-
`lar types of cisplatin-DNA lesions. There are now several
`methods available for measuring cisplatin-DNA adducts.
`The standard for the quantitation of elemental plat-
`inum is atomic absorption spectrometry (AAS).1° AAS
`utilizes high temperature to atomize molecules within a
`graphite chamber. The chamber is monitored by an opti-
`cal system that reads the absorbance of the atomic cloud
`generated. One metal can be distinguished from another
`by its absorbance at specific wavelengths. Platinum is
`measured at a wavelength of 265.9 nm. Recently, AAS
`instruments have been equipped with optional Zeeman
`background correction for nonspecific absorption.15
`Using Zeeman correction, one can now obtain highly
`precise and sensitive measurements of platinum bound to
`DNA after therapeutic levels of drug exposure.16
`An alternative method to assess cisplatin—induced
`DNA lesions in cells treated at pharmacologically relevant
`dosage is DNA alkaline elution.17 In this method, cellular
`DNA is prelabeled with a radioisotope such as [3H]
`thymidine. After exposure to drug, the cells are deposited
`on a filter surface and lysed with a detergent solution. An
`alkaline solution (pH 12) is then added. The high pH
`disrupts the hydrogen-bonded structure that holds the
`DNA strands together. The alkaline solution is pumped
`slowly through the filter, and the rate of elution of the
`single-stranded DNA is determined. Using appropriate
`modifications of this technique, it is possible to quantify
`DNA interstrand cross-links, DNA-protein cross—links,
`and DNA strand breaks, all ofwhich modulate the rate of
`DNA elution from the filter. The method detects inter-
`
`strand cross—links, which retard elution of DNA, as well
`as DNA strand breaks, which accelerate elution. The
`technique does not detect DNA intrastrand cross-links or
`DNA-drug monoadducts, which can be measured by
`HPLC methodsmil"
`
`A number of ELISA assays can assess drug binding to
`DNA in various settings. The first such assay was devel-
`oped by Poirier and colleagues”23 and has been used to
`Study tissues from human cancer patients as well as tissues
`from experimental animals. It utilizes polyclon‘al antisera
`Clicited in rabbits against an antigen ofintact calf thymus
`DNA modified with cisplatin. Although Poirier’s method
`quantitates an antigen that correlates well with cisplatin
`biologic efiect, it measures only a fiacfion ofthe covalent
`Platinum-DNA complexes measured by AAS.24 Subse-
`quent ELISA assays developed by Fichtinger-Schepman
`and colleagues“ use polyclonal antisera elicited against
`purified,
`isolated cisplatin-DNA adducts, and those
`developed by Lippard and colleagues26 utilize murine
`monoclonal antibodies elicited against cisplatin-modified
`iIltact calfthymus DNA. Fichtinger—Schepman’s assay can
`quantitate cisplatin-DNA lesions only after the DNA has
`
`been degraded and adducts have been purified by HPLC.
`Fichtinger-Schepman et al.27728 performed limited studies
`with material from human cancer patients, and it is
`unclear whether the need to subject isolated DNA to en-
`zymatic degradation and subsequent HPLC purification
`limits the application of this assay to the study of small
`numbers of samples. However, this assay correlates well
`with AAS measurements of plau'num binding to DNA.
`The determination of the nature of the DNA lesions
`
`formed by cisplatin after defined drug exposures was
`made possible by HPLC analyses of cisplatin-modified
`DNA after that DNA was degraded by enzymatic or
`chemical hydrolysis”,19 Generally, HPLC methods can
`readily assess the relative percentages ofspecific platinum-
`DNA lesions and have been used to measure the relative
`
`proportions of the deG and dApG diammineplatinum
`adducts after cisplatin exposures. However, these meth-
`ods do not determine the absolute amount of drug
`bound to DNA.
`
`Terheggen and colleagues”31 have developed an
`immunocytochemical technique that uses microdensi-
`tometry to assess relative cisplatin binding to DNA in
`rodent and in human tissues. Their assay utilizes a poly-
`clonal antiserum elicited by the method of Poirier. A
`“double peroxidase” approach is used to visualize the
`binding sites of the antiserum on the DNA. With this
`method, as few as several hundred cells are required to
`obtain an assessment ofthe relative level of cisplatin bind-
`ing to DNA, a threshold that is 5 to 6 logs less than the
`number of cells needed in the assays of Poitier and
`Fichtinger-Schepman. It is currently not clear if this assay
`correlates with AAS determinations. Others have adopted
`these methods to measure gene-specific repair.32
`Sancar and colleagues“,34 used the uvrABC excision
`nuclease complex from Escherichia 5011'. Such a method
`may prove useful in determining the persistence of cis-
`platin-DNA adducts in viable tissues of human cancer
`patients months after drug exposure.35
`Taq polymerase, used in PCR reactions to amplify
`DNA, is inhibited by cisplatin DNA adducts and can de-
`tect as few as one adduct per 2000 base pairs?”8 This
`sensitivity may not be adequate to measure adducts in
`clinical
`specimens,
`which
`reach
`levels
`of
`1 adduct in 10,000 to 100,000 base pairs.
`
`MECHANISM OF ACTION
`
`Most data are consistent with the hypothesis that the
`major cytotoxic target of cisplatin is DNA. The types of
`DNA lesions primarily responsible for the cytotoxicity
`and antitumor activity, however, are not clearly estab-
`lished. Analogues of cisplatin that are tumoricidal in
`experimental systems have chemical leaving groups in the
`cis configuration and have effects on DNA that are in
`most respects similar to those of the parent compound.
`
`
`
`

`

`362
`
`CANCER CHEMOTHERAPY AND BIOTHERAPY
`
`The cytotoxicity ofcisplau'n against cells in culture cor-
`relates closely with total platinum binding to DNA)",39 to
`interstrand cross-links,17’4° and to the formation of intra-
`strand bidentate N-7 adducts at d(GpG) and d(ApGYML42
`(Fig. 14-3). The relative percentages of these lesions have
`been studied after treatment of isolated DNA with cis-
`platin,“"19 as well as after treatment ofmalignant and non-
`malignant rodent cells”,43 and after treatment of human
`cancer patients.“44 HPLC analysis of degraded DNA
`indicates that the d(GpG) lesion makes up 60% to 70% of
`total platinum binding to DNA, the d(ApG) lesion is 20%
`to 30% of total platinum binding, and the remaining 10%
`is a mixture of various intrastrand d(GpoG) adducts, in—
`terstrand cross—links, and monoadducts.“"19’45 Analogous
`studies have been carried out in which intact cells were
`treated with cisplatin. In L1210 murine leukemia cells43
`and in Chinese hamster ovary cells,25 the relative percent-
`ages of adducts measured by HPLC analysis approximate
`the following:
`interstrand cross-links,
`less
`than 1%;
`d(GpG) intrastrand adducts, 60%; d(ApG) adducts, 30%.
`Composing the remaining 10% are a variety of DNA
`lesions, including DNA—protein cross-links, DNA mono-
`adducts, and d(GpoG) and d(ApoG) intrastrand
`adducts. In human leukocyte DNA after a Cisplatin intra-
`venous infusion?!"44 the relative percentages of the
`Cisplatin-DNA lesions formed in human leukocyte DNA
`were the same as those reported for L1210 cells, Chinese
`hamster ovary cells, and salmon sperm DNA.
`
`Several recent reports have focused on the inhibi-
`tion of DNA replication and specific enzymatic activities
`by different Cisplatin adducts. Pinto and Lippard45 com-
`pared the ability of Cisplatin and transplatinum to inhibit
`DNA synthesis using a single-stranded DNA template and
`DNA polymerase I. Cisplatin was much more effective at
`inhibiting DNA replication than was the trams-Ptal) iso-
`mer in this system. A d(GpG) adduct was very effective in
`inhibiting replication of DNA polymerase I.
`Intrastrand d(GpG)-Cisplatin adducts cause a marked
`distortion of DNA conformation. Lippard and col-
`leagues‘“48 have described two relevant alterations. In
`one study, it was shown that the d( GpG)-cisplatin lesion
`disrupts the local structure of the DNA, allowing the
`cytosine bases that normally are paired to guanines to
`become partially or totally unpaired, as indicated by their
`becoming accessible to binding by an antiserum that
`recognizes unpaired DNA bases.47 This disruption of
`structure requires bidentate binding by platinum because
`it did not occur in DNA treated with a monodentate plat-
`inum analogue. In another set of studies, Lippard’s group
`has shown that the intrastrand adduct introduces a sharp
`bend in the helix axis of the DNA and that this is a con-
`sequence of the geometry of the cis—Pt(II) complex
`bound to two adjacent guanine-N-7 positions.48 Lippard
`suggested that these alterations in local DNA structure
`that are caused by a Cisplatin intrastrand adduct may be
`responsible for the cytotoxic effect.49
`
`
`
`
`
`PM“
`t, “w
`M;
`Film1 : 5M"'ttlrr
`must?
`
`“nma
`
`
`
`-Nsa-d’l£451
`
`Figure 14-3. Biiunctional adducts of Cisplatin with DNA. Lesions indicated in panels A, B, and C represent different intrastrand
`adducts, which together account For >90% of total platinum binding to DNA. The lesion indicated in panel D is the interstrand cross-
`link measured by alkaline elution and accounts for less than 5% of total platinum binding to DNA. See text For discussion.
`
`d(GpG) Adduct
`
`d(ApG) Adduct
`
`H3N
`
`NH3
`\ /
`Pt
`
`/\
`5'— A—G _3'
`O
`O
`
`3'— T—C —5’
`B
`
`lnterstrand Crosslink
`
`H3N
`
`NH3
`
`d(GpoG) Adduct
`
`NH3
`
`HEN
`\W/’
`5'— G{x>e— 3'
`o
`o
`.
`
`3,—C—X—C—5'
`C
`
`
`
`
`

`

`Platinum Analogues
`
`When cisplatin binds to cellular DNA and/or RNA,
`such lesions may inhibit the fiinction of a number of
`enzymatic processes important to the cell. Such functions
`include inhibition of SI nuclease (an endonuclease spe-
`cific for single—stranded DNA)“; inhibition of restriction
`enzymes“; inhibition of DNA polymerase 1”; inhibition
`of RNA polymerase“; and inhibition of RNA transla-
`tion“,55 Synthesis of new DNA or RNA can proceed
`across cisplatin intrastrand cross—links, as mediated by
`DNA and RNA polymerases, respectively,“ although
`only 10% of adducts are bypassed. The d(GpG) lesion is
`the most inhibitory. In E. coli, the intrastrand d(ApG)
`adduct is five times more mutagenic than d(GpG).57 In
`this system the d(ApG) may not be an effective blocking
`lesion for either DNA or RNA synthesis.53 Guanine-rich
`sequences appear to be preferred targets for cisplatin
`attack and mutation.58 Interestingly, the type and extent
`of helical unwinding and bending are similar for the two
`intrastrand adducts.59
`The tmm—Pt(II) isomer of cisplatin is capable of bind-
`ing and cross-linking DNA but has much lower cytotox—
`icity and no antitumor activity. Many studies have inves-
`tigated the origin of the biologic difference between the
`cis— and tmm—Pt(II) isomers to elucidate the mechanism
`of action of cisplatin. Although both isomers produce in-
`terstrand cross-links in purified DNA, only the cis isomer
`(cisplatin) produces substantial interstrand cross-linking
`in mammalian cells.”40 The trans isomer, however,
`produces more DNA-protein cross-links than does cis-
`platin. This argues against DNA-protein cross-links as the
`primary cytotoxic lesions; however, this conclusion is not
`definitive because of possible differences between the cis
`and trans isomers in regard to the nature of the proteins
`bound and the locations of their binding in the genome.
`Ciccarelli et al.‘50 compared the actions of cisplatin and
`its trans isomer in SV40-infected green monkey CV—l
`cells. In this system, the trans isomer was much less
`potent than cisplatin in generating Pt adducts of SV40
`DNA. At equal extracellular concentrations, much less
`trans than cis isomer was bound to the viral DNA isolated
`
`from treated cells. The inhibition of SV40 DNA replica-
`tion, when gauged on the basis of amount of Pt bound to
`the DNA, was reported to be nearly equal for the two iso-
`mers. These data were interpreted as indicating a rapid
`removal of bound tram-Pt from the DNA oftreated cells,
`in contrast to little or no such repair in the case of
`cisplatin. Roberts and Friedlos,61 however, were not able
`to confirm the proposed rapid repair of tram-Pt DNA
`adducts and attributed the apparent
`reduction in
`I)t/DNA ratio not to a removal of Pt but to continued
`DNA synthesis in the trans—Pt treated cells.
`The reason for the high cytotoxicity of cisplatin and
`10W cytotoxicity of trans-PtaI) thus is still unclear. It may
`be that the interstrand cross-links, even though they con-
`stitute only a small fraction of cisplatin DNA adducts, are
`the major cytotoxic lesions because the trans isomer pro-
`duces virtually no interstrand cross-links in mammalian
`Cells. A second possibility is that the d(GpG) or d(ApG)
`
`intrastrand cross-links produced by cisplatin are much
`more cytotoxic than are the d(GpoG) intrastrand cross-
`links produced by the trans isomer. Tram-Ptal), because
`of its particular geometry, cannot form intrastrand cross-
`links between adjacent bases, but it can form intrastrand
`cross—links in which one base is skipped, as in d(GpoG).
`A third possibility is that cisplatin produces an especially
`cytotoxic type of DNA-protein cross—link different fi'om
`the trans isomer.
`
`Because cisplatin could react avidly with many accessi-
`ble sulfur and nitrogen sites on a variety of proteins, it is
`difficult to evaluate the possible importance of non-DNA
`targets in the mechanism of antitumor action or mecha-
`nism of toxicity. Platinum complexes bind extensively to
`sulfliydryl groups of plasma proteins, and it has been hy-
`pothesized that ligand exchange reactions with sulfhydryl
`groups of critical enzymes may be responsible for to

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