throbber
”Want-m,q
`zsmrmsm
`
` 2
`
`Hepatotoxic Effects of Oncotherapeutic
`and Immunosuppressive Agents
`
`and chemical
`Chemotherapy for neoplastic disease
`imimmosuppression involve the use of agents that inter—
`fere selectively with metabolic pathways or have other
`cytotoxic effects; several of these agents are hepatotoxic
`(1—8). Other agents in the group appear to spare the liver
`or to produce hepatic injury rarely and as the result of
`host idiosyncrasy (5). Although abnormal values for bio-
`chemical tests are common, clinically significant hepatic
`injury is relatively uncommon. It seems almost paradoxi-
`cal that some agents that are potent cell poisons, and that
`are metabolized by the hepatocyte, produce little or no
`hepatic damage.
`Several factors appear to account for the sparing of the
`hepatocyte by agents so lethal
`to the neoplastic cell.
`These agents exert their toxic effects preferentially on
`rapidly proliferating tissues, such as those of the bone
`marrow, gastrointestinal
`tract,
`and neoplasms
`(3).
`Accordingly, the slowly multiplying hepatic tissue should
`be relatively unsusceptible (3). Furthermore, the broad
`range of metabolic activity of hepatocytes may provide
`opportunity for balance between production and disposi—
`tion of toxic metabolites; whereas transformation of the
`drug in the neoplastic cell (e.g., by action ofphosphami-
`dase or cyclophosphamide) may convert it to a cytolytic
`alkylating agent (5). Additionally, administration of anti~
`cancer drugs may lead to amplification of the multidrug-
`resistance gene, which encodes an organic ion transporter
`(p-glycoprotein). Conceivably, this may add protection by
`enhanced excretion of potentially hepatotoxic drugs (8).
`Nevertheless, considering the role of alkylation and ary-
`lation of cell macromolecules of the hepatocyte in the
`production of hepatic injury by known hepatotoxins
`(Chapter 5), the oligotoxicity for the liver of alkylating
`antineoplastic agents is somewhat surprising.
`Also relevant to the hepatotoxic effects of anticancer
`drugs is the difficulty of identifying any hepatic injury
`they may cause. Toxic liver damage resulting from onco—
`
`therapy must be distinguished from the effects on the
`liver of the neoplasm, coincidental viral hepatitis, other
`infections, other drugs, and total parenteral nutrition.
`The number of candidate oncotherapeutic agents stud-
`ied in experimental animals and tested in patients is far
`too great, and the data regarding the possible adverse
`effects of many of them on the liver are too scant, to per-
`mit systematic analysis of the hepatotoxicity of cancer
`chemotherapy. Nevertheless, sufficient
`information is
`available to define the effects of some agents on the liver
`(Table 23.1) and to discern some general principles
`regarding the relation between the type of agent and the
`potential for producing hepatic injury.
`
`FORMS OF HEPATIC INJURY
`
`For the most part, the forms of acute (Table 23.2) and
`chronic (Table 23.3) hepatic injury produced by oncother-
`apeutic agents are similar to those produced by other
`agents, Several forms of the injury, however, are particuo
`larly prominent. Steatosis is more frequently noted in the
`hepatic injury of several anticancer agents than in that pro-
`duced by most other drugs. Steatocirrhosis seen with metl -
`otrexate (MTX) therapy but is not seen with most other
`medicinal hepatic injury. Venn-occlusive disease (VOD),
`the dramatic lesion characteristically resulting from pyrro—
`lizidine alkaloids, is produced by several oncotherapeutic
`and immunosuppressive agents, singly and in various com—
`binations (4—6,8—l l), and not by other medicinals (Table
`23.4). Another unique drug-induced lesion is the scleros-
`ing cholangitis that is seen as a complication of “pump”
`infiision of floxuridine into the hepatic artery for treatment
`of metastatic hepatic carcinoma (12—21) (Table 23.5).
`
`Acute Injury
`
`The acute hepatic lesions of cancer chemotherapy are
`mainly hepatocellular. A few anticancer drugs are intrin-
`
`673
`
`IMMUNOGEN 2041, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`s‘l._
`,_
`
`y.
`
`_
`
`IMMUNOGEN 2041, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`+
`
`+++++|
`
`I
`
`l++ml+++
`
`+ l
`
`1++il
`
`|l+|1
`
`+l
`
`|+
`
`+
`
`+
`
`674 / HEPATOTOXICITY
`
`
`
`TABLE 23.1. Some hepatic lesions produced by agents used in cancer therapy
`VOD
`Peliosis
`Cholestasis
`Fat
`Necrosis
`Biochemica pattern of injury
`Drug
`______________________________________._.___——————————-——-—-—
`AT
`Aclarubicin
`Cholestatic
`Aminoglutethamide
`Amsacrine
`Hepatoce u ar/cholestatic
`Cholestat'c
`Anabolic steroida
`Hepatoce u ar
`Asparaginase
`Hepatocellular
`Azacytidine
`Hepatoce u ar
`Azatepa
`Hepatocel ular/cholestatic
`Azathioprine
`Hepatoce u ar
`Bacille Calmette-Guérinc
`Hepatoce ular”
`Bleomycin
`Cholestatic
`Busulfand
`Hepatoce u ar/cholestatic
`Carboplatin
`Carmustine
`Hepatocel ular
`Hepatoce u ar
`Chlorambucil
`Hepatocel ular
`Chloropurine
`Chlorozocin
`Hepatocellu ar/cholestatic
`Hepatoce u at
`Cisplatin
`None
`Cladribine
`Cholestatic
`Cyclosporine
`Hepatoce u ar
`Cyclophosphamide
`Hepatocel ular
`Cyproterone
`Hepatocellu ar
`Cytarabine
`Dacarbazine
`Hepatoce u ar
`Hepatocellular
`Dactinomycin (actinomycin D)9
`Daunorubicin
`Hepatoce ulare
`Dichloromethotrexate
`Hepatocellu ar
`Hepatoce lular
`Diethylstilbestrol
`Cholestatic
`Dimethylbusulfan
`Doxorubicin
`Hepatoce ular
`Estramustine
`Hepatocel|u|ar
`Cholestat'c
`Estrogens (Steroids)
`Hepatocellular
`Etoposide
`Floxuridine’
`Hepatoce ular/chotestatic
`AT
`Fluorouracil
`Flutamide
`Hepatoce Iular/cholestatic
`Frentizole
`Hepatoce ular
`AT
`Homoharringtonine
`Hepatoce ular
`Hydrazines
`Cholestatic
`Hydroxyprogesterone
`Hepatocellular
`Hydroxyurea
`AT
`Idarubicin
`AT
`lfosfamide
`Indicine-N-oxide
`Hepatocellular
`AT
`Interferons
`Cholestatic
`Interleukin—29
`Cholestatic
`Interleukin—6
`None
`Lomustine
`AT
`Maytansine
`None
`Mechlorethamine
`Cholestatic
`Medroxyprogesterone
`AT
`Melphalan
`Hepatocellular/cholestatic
`Mercaptopurine
`Methotrexate
`Hepatocel|u|ar
`Hepatocellular
`Mithramycin
`Hepatocellular
`Mitomycin
`AT
`Mitotane
`AT
`Mitoxantrone
`HepatoceHular
`Monomethylformamide
`Ni|utamide
`Hepatocellular
`Pentostatin
`Hepatocellular
`Procarbazine
`Hepatocellulari
`Hepatocellular
`Puromycin
`Hepatocellular
`Pseudisocytidine
`AT
`Semustine
`Hepatoceltular
`treptozocin
`_s_______________________________..____———————————-———
`
`(|+|!
`
`|+++
`
`+I
`
`|l++I
`
`++
`+t
`
`I |
`
`++++++
`
`IMMUNOGEN 2041, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`+l
`
`I++|
`
`+ +
`
`+
`
`|I++|
`
`|++|
`
`l+
`
`IMMUNOGEN 2041, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`—
`—
`—
`+
`—
`~
`
`—
`+
`+
`+
`,
`—
`
`TABLE 23.1. Continued.W
`Drug
`Biochemical pattern of injury
`Fat
`Necrosis
`Cholestasis
`VOD
`Peliosis
`Tacrolimus
`Hepatocellular
`Tamoxifen
`Cholestatic/hepatocelluIar
`Teniposide
`Hepatocellular
`Thioguanine
`Hepatocellular/cholestatic
`Thiotepa
`Hepatocellular
`Trimetrexate
`None
`Triethylenemelamlne
`None
`Uracil mustard
`AT
`-
`+
`—
`+
`-
`Urethane
`Hepatccellular
`
`Vinca alkaloids _WHepatocellular — +517 _ +5
`
`
`
`
`
`AT, increased aminotransferase levels but no jaundice; +, present ;~, absent; i, uncertain.
`alncriminated in hepatic adenoma and carcinoma.
`”Hepatic injury is minor component of adverse effects of drug.
`CGranuloma.
`dAlso incriminated, with other agents, in production of nodular regenerative hyperplasia,
`EOnly when given with other agents or radiotherapy.
`’Characteristic injury is sclerosing cholangitis when drug is administered by pump into hepatic artery,
`9Pattern of injury is consistently cholestatic. Histological changes show more variation.
`hin large parenteral doses.
`’ Rare
`
`+
`_
`+
`_
`_
`-
`
`_
`_
`+
`_
`L
`-
`
`+
`_
`_
`_
`_
`-
`
`TABLE 23.2. Acute hepatic injury associated with agents used in cancer therapy.W
`
`Cytotoxic
`
`Necrosis
`Dose—related, relatively frequent
`
`Dose-unrelated, relatively rare
`
`Steatosis
`Steatosis plus necrosis
`Cholestatic
`
`Mixed cholestatic—cytotoxic
`aIn high dose.
`
`Agents
`
`Mithramycin, N—methyltormamide, mercaptopurine, nitrosoureas, streptozocin,
`methotrexatea
`
`Chlorambucii, cyclophosphamide, cytarabine, cytoproterone, etoposide, flutamide,
`frentlzole, thioguanine
`Dactinomycin, asparaginase, bieomycin, methotrexate, mitomycin,a puromycin
`Asparaginase, cisplatin, methotrexate,a thiotepa
`Anabolic steroids, estrogenic steroids, aminogutethamide, azathioprine, amsacrine,
`busultan, 4,4’—diaminodiphenylamine, medroxyprogesterone, tamoxifen
`Chlorozocin, mercaptopurine, thioguanine, azathlopurine
`
`TABLE 23.3. Chronic hepatic lesions associated with agents used in cancer therapyW
`Type of injury AgentsW
`
`Cytotoxic injury
`Chronic hepatitis
`Steatosis
`Phospholipidosis
`Mallory bodies (alcoholic hyaline)
`Cirrhosis
`Steatocirrhosis
`Biliary cirrhosis
`Congestive cirrhosis
`Cholestatic lesions
`Chronic intrahepatic cholestasis
`Sclerosing cholangitis
`Vascular lesions
`Peliosis hepatis
`
`Azathioprine, doxorubicin
`Daotinomycin, asparaginase, glucocortisteroids, methotrexate, mitomycin C, puromycin
`amphophilic compounds
`Diethylstilbestrol, tamoxifen
`
`Methotrexate
`Azathioprine, floxuridine
`Drugs that lead to VOD or hepatic thrombosis
`
`Azathioprine
`Floxuridine
`
`Anabolic steroids, azathioprine, medroxyprogesterone, hydroxyurea, tamoxifen, OCs,
`DES, thioguanine
`Combination therapy (Table 23.18)
`(Table 23.4)
`Many drugs
`
`Hepatic vein thrombosis
`Veno—occlusive disease
`Granulomas
`Neoplasms
`Adenomas
`Carcinoma, hepatocellular
`Angiosarcoma
`Non-cirrhotic portal hypertension
`Nodular regenerative hyperplasia,
`Combination therapy (Table 28.18)
`hepatoportal sclerosis
`DES, diethylstilbestroi; OCs, oral contraceptives; VOD, veno-occlusive disease.
`
`Anabolic steroids, OCs
`Azathioprine, anabolic steroids, chlorambucil, OCs, DES, methotrexate
`Anabolic, OCs, DES
`
`IMMUNOGEN 2041, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`IMMUNOGEN 2041, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`676 / HEPATOTOXICITY
`
`TABLE 23.4. Oncotherapeutic and immunisuppressive
`______________________.__._————-—-—————-—-——-
`agents that can lead to veno-occlusive diseasea
`
`wA
`
`zathioprine
`Busultan
`Carmustine
`Cisplatin
`Cyclophosphamimide
`Cytarabine
`
`Dacarbazine
`Dactinomycin
`Daunorubicin
`Dimethylbusufan
`Doxorubicin
`Floxuridine
`Indicine-N-oxide
`Mechlorethamine
`B-Mercaptopurine
`Mitomycin
`Thioguanine
`Urethane
`Vincristine
`X-lrradiation
`Combinations
`__________._____—_.———————————————-———-
`
`aAlone or in combination with other agents or x—irradiation.
`
`sic hepatotoxins that produce necrosis as a dose—depen-
`dent effect. Most produce necrosis relatively rarely as a
`dose-independent idiosyncratic reaction (6). Acute chole-
`stasis is an uncommon form of anticancer drug injury,
`although busulfan (23,24), azathioprine (25,26), amino-
`glutethamide (27,28), amsacrine (6), tamoxifen (29,30),
`medroxyprogesterone (8), and the obsolete agent, 4,4’-
`diaminodiphenylamine (6) can cause cholestatic injury.
`
`Chronic Injury
`
`Cytotoxic Lesions
`
`Steatosis is a frequent chronic cytotoxic lesion resulting
`from several anticancer drugs, especially MTX (6) (Table
`23.3). Chronic hepatitis resembling the autoimmune type,
`a lesion and syndrome that can be produced by several
`
`Imuran
`Myleran
`BCNN. BlCNU
`Platinol
`Cytoxan
`Cyosine arabinoside, ARA-C,
`Cytosar-U
`DTIC
`Actinomycin, Cosmegen
`Cerubidine
`——
`Adriamycin
`FUDR
`——
`Mustargen
`Purinethanol
`Mitocin. Mutamycin
`TG
`Ethyl carbamate
`Oncovin
`
`TABLE 23.5. Published reports of sclerosing cholangitis occurring as a complication of pump infusion of
`floxuridine into the hepatic artery
`
`Authors
`
`No. cases
`
`incidence of lesion (°/o)
`
`Year
`
`Ref no.
`
`16
`1986
`40
`20
`Anderson et al.
`13
`1985
`33a
`2
`Bolton et al.
`14
`1985
`7
`6
`Botet et al.
`18
`1986
`100”
`8
`Doria et al.
`19
`1987
`100
`20
`Hermann et al.
`12
`1986
`56
`31
`Hohn et al.C
`15
`1985
`17
`8
`Kemeny et al.
`20
`1989
`—
`1
`Ludwig et al.
`
`
`
`
`17 100 1986Shea et alf 17W—
`aReflects authors‘ estimate that “one-third" of treated patients develop the lesion.
`thetlects all of patients studied.
`CThe two reports are from the same institution and figures may overlap.
`
`is not readily ascribable to any of the cancer
`drugs,
`chemotherapy agents. However, I have seen the lesion as
`an apparent reaction to azathioprine and in published (31)
`photomicrographs of doxorubicin-induced injury.
`Phospholipidosis, a lesion characterized by phospho-
`lipid—engorged lysosomes and produced by a number of
`amphiphilic drugs (32), has not been recorded as a lesion
`of oncotherapeutic agents other than the investigational
`antileukemic drug, AC3579 (33). The Mallory body, a
`lesion seen in alcoholics and in several other clinical con-
`ditions (Chapter 6), is not seen with any of the usual anti-
`cancer agents. It has been produced in experimental ani—
`mals (34) by administration of diethylstilbestrol and in a
`patient with prostatic carcinoma (35) treated with that
`drug. Tamoxifen also has been implicated in the develop—
`ment of Mallory bodies (35a,35b). Several types of cir-
`rhosis may occur after anticancer therapy (Table 23.3).
`
`Cholestatic Injury
`
`Chronic intrahepatic cholestasis may follow the acute
`cholestasis induced by azathioprine (Fig. 23.1). Refer-
`ence has been made to the striking sclerosing cholangitis
`(Fig. 23.2) produced by hepatic artery infusion of flex-
`uridine (12—21) in the treatment of metastatic hepatic car—
`cinoma (Table 23.5).
`
`Vhscular Lesions
`
`Peliosis Hepatitis. This lesion has been associated in
`the past with terminal
`tuberculosis or carcinomatosis
`(36), more recently with Cl7—alkylated steroids, and,
`rarely, with oral contraceptives (Chapter 4). Peliosis hep-
`atis also has been seen in patients (8) taking tamoxifen
`(37), hydroxyurea (4) thioguanine, medroxyprogesterone
`acid (8), or azathioprine (4) (Table 23.3).
`Hepatic Vein Thrombosis. The drugs most associated
`with hepatic vein thrombosis are the oral contraceptives
`(38). Cancer chemotherapy is .rarely the cause of the
`injury, but the lesion has been reported as a complication
`of chemotherapy for Hodgkin’s disease (39) and adminis-
`
`tration of dacarbazine (DTIC) (40).
`
`IMMUNOGEN 2041, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`IMMUNOGEN 2041, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`23: ONCOTHERAPEUTIC AND IMMUNOSUPPRESSIVE AGENTS / 677
`
`
`
`FIG 23.1. Liver biopsy from a patient who developed chronic cholestasis (vanishing bile duct syndrome)
`after taking azathioprine for many months. Jaundice had been present for 11 months at that time of the
`biopsy. A: Note cholestasis with huge bile casts in canaliculus. B: Cholate stasis (arrow) reflected by
`foamy striated cells. C: Portal areas (P) showing fibrosis and lack of bile ducts.
`
`Veno-Occlusive Disease. This lesion leads to morpho-
`logic changes and a syndrome resembling that of hepatic
`vein thrombosis, namely the Budd-Chiari syndrome (41).
`In VOD, however, the lesion is fibrotic and involves par-
`tial or complete obliteration of the lumina of the efferent
`venules rather than thrombotic occlusion of the hepatic
`veins (41). The lesion is typical of pyrrolizidine alkaloid
`toxicity, which is the most common cause of the entity
`worldwide (9,41,42). In this country the most common
`causes are anticancer and immunosuppressive drugs
`(22,43—49) and radiation injury (50) of the liver. It is rel-
`atively common in patients receiving chemotherapy or
`
`immunosuppressive therapy, particularly in recipients of
`bone marrow transplants (ll). VOD develops in 10% to
`60% of patients during the first few weeks after bone
`marrow transplantation (4,6,9—1 1,43—49). Superb exposi-
`tion of the VOD that is associated with bone marrow
`transplantation is continued in the writing of Jones et al.
`(9), McDonald et al. (10), and Bearmen (1 1). It is char-
`acterized by partial or complete fibrotic occlusion of ter—
`minal hepatic venules (central veins) with congestion and
`necrosis of the perivenous area (zone 3). The diagnosis
`should be suspected when abdominal
`swelling and
`increased aminotransferase levels develop during or after
`
`
`
`FIG. 23.2. A: Retrograde cholangiogram showing sclerosing cholangitis in a patient after 6 months of
`pump infusion therapy with floxuridine. (Courtesy of Dr. Paul Shorb, George Washington University Med
`ical Center; from ref. 5, with permission.) B: Section from explant liver of patient who underwent trans-
`plantation for sclerosing cholangitis. Note damaged bile ducts and striking periductal sclerosis.
`
`IMMUNOGEN 2041, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`IMMUNOGEN 2041, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`678 / HEPATOTOXlCITY
`
`chemotherapy. As a therapeutic misadventure, VOD is a
`lesion virtually specifically related to anticancer and
`immunosuppressive drugs and irradiation (Table 23.4).
`No other therapeutic agents lead to it, although several
`herbal preparation taken as alternative medications can
`(Chapter 24).
`Portal ijerz‘enslon. Nodular regenerative hyperpla-
`sia and hepatoportal sclerosis, which are lesions that can
`lead to portal hypertension, have been described in recip—
`ients of chemotherapy involving several anticancer drugs
`in combination (51~57). Hepatoportal sclerosis has been
`produced by organic arsenicals and by azathioprine, and
`nodular regenerative hyperplasia has been attributed to
`azathiopiine, dactinomycin, busulfan plus thioguanine,
`and other protocols employed in preparation for bone
`marrow transplantation (9—1 1,54—57).
`
`Granulomas
`
`Many drugs produce granulomatous lesions (58,59), but
`the anticancer drugs show no special tendency to do so.
`
`Neoplasms
`
`(60), azathioprine (61), MTX (62),
`Chlorambucil
`androgens (63), and diethylstilbestrol
`(64) have been
`implicated in the development of hepatic carcinoma.
`Androgens, estrogens, and procarbazine also have been
`linked to angiosarcoma (65—67).
`
`MECHANISMS OF INJURY
`
`Like other drugs, anticancer agents produce injury as
`intrinsic hepatotoxins or as idiosyncratic reactions. The
`antimetabolites, some selective enzyme poisons, and
`some antibiotics tend to be intrinsic hepatotoxins that
`produce dose—dependent liver damage. Urethane and the
`nitrosourea analogues, which become alkylating agents
`only after biotransformation, also appear to act as hepa-
`totoxins. However, the directly alkylating oncotherapeu-
`tic agents such as nitrogen mustards generally do not
`produce hepatic injury, or do so only as very rare idio—
`syncratic reactions. Hypersensitivity appears to be a less
`common form of idiosyncratic reaction to anticancer
`agents than metabolic idiosyncrasy, as judged from co]-
`lateral clinical features (6).
`The remainder of this chapter describes hepatic injury
`produced by individual drugs.
`
`ALKYLATING AGENTS
`
`Five groups of alkylating agents (Table 23.6) have been
`used for oncotherapy. The nitrogen mustards (Fig. 23.3)
`show little hepatotoxic potential. Mechlorethamine and
`melphalan seem free of responsibility for hepatic injury,
`and the ethylenimine derivatives seem hardly ever to have
`
`caused liver damage. Cholestatic jaundice has been
`attributed to an alkylsulfonate (23,24), but rarely. More
`convincing evidence for some hepatotoxic potential
`applies to the nitrosoureas (68), streptozocin (4), and a
`triazine (4).
`
`Nitrogen Mustards
`
`I?”
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Mechlorellzamine (HNg). This alkylating agent (Mus-
`targen, Merck & Co, West Point, PA), the first to be used,
`has been found to produce little or no hepatic injury. One
`report described focal hepatic necrosis in three patients
`and worsening of jaundice in two others among 50
`patients receiving the drug (69). in a prospective study,
`my group found no hepatic injury (70).
`Melphalari (MLD). This drug (Alkeran, Glaxo Well-
`come, NC) has not been incriminated in significant
`hepatic injury to my knowledge. Large doses have been
`accompanied by mildly abnormal aminotransferase levels
`(7).
`(CLB). This agent (Leukeran, Glaxo
`Clllormnbucil
`Wellcome, NC) has been implicated in several reports of
`hepatic injury (71,72). The cases are few, but the form of
`injury appears to have been uniformly hepatocellular.
`Massive necrosis can occur (4). The possible hepatocar-
`cinogenic role of the agent has been reported (60).
`Cyclop/zosplzamicle (CTX). This drug (Cytoxan, Bris-
`tol-Myers Squibb Oncology/1mmLmology, Princeton, NJ)
`seems to have led to a few instances of hepatic injury.
`Several reports have drawn attention to mild hepatic
`injury in humans and in experimental animals. Instances
`of marked elevation of aspartate aminotransferase (AST),
`of massive necrosis, and of VOD have been reported
`(73—75). This drug also has been incriminated, both in
`concert with busulfan and other drugs (76—80) and alone
`(81,82), in the production ofVOD.
`[fro/amide.
`I fosfamide (lfex, Bristol—Mvers Squibb
`Oncology/1771mmology, Princeton, NJ), a derivative, has
`seemed largely clear of hepatotoxic potential. Two cases
`of severe Cholestatic injury have been described in
`patients receiving both ifosfamide and etoposide (83).
`Uracil Mustard. This agent was incriminated in sev-
`eral instances of mild hepatic injury, but apparently none
`lately (6).
`
`
`
`
`
`
`
`
`
`
`
`Ethylenimine Derivatives
`
`Triethylenemelamine (TEM) seems not to have caused
`hepatic injury. Triethylenethiophosphoramide, or thiotepa,
`has been incriminated in a case of liver failure (84), and a
`derivative, azatepa, has been implicated in severe hepatic
`injury resembling phosphorus poisoning (85) (Fig. 23.4).
`
`Alkylsulfonatcs
`
`Busulfan (Myleran, Glaxo Wellcome, NC) (Fig. 23.5),
`an agent long in use in the treatment of granulocytic-
`
`
`
`IMMUNOGEN 2041, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`IMMUNOGEN 2041, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`23: ONCOTHERAPEUTIC AND IMMUNOSUPPRESSIVE AGENTS / 679
`
`TABLE 23.6. Adverse effects on liver of alky/ating anticancer drugsW
`_._______..__—-————-———————-———————————————-—————————
`Drug
`Hepatic injury
`Type
`Mechanism
`Nitrogen mustards
`Mechlorethamine (HNz Mustargen)
`Melphaian (MLP Alkeran)
`Cyclophosphamide (CTX Cytoxan)
`Chlorambucil (Leukeran)
`Uracil Mustard (Ura-M)
`Itasfamide (lfex)
`Ethylenimine
`Triethylenemelamine (TEM)
`Thiotepa (Thioplex)
`Azatepa
`Alkylsulfonates
`Busulfan (Myleran)="
`Dimethylbusulian
`Nitrosoureas
`Carmustine (BCNU)
`Lomustine (CCNU)
`Semustine (Methyl—CCNU)
`Estramustine (Emcyt)
`Streptozocin (Zanosar)
`Chlorozocin
`Triazines
`TOX, Idio
`VOD
`Yes
`Dacarbazine (DTlC)
`
`Procarbazine (Matulane) IdioWVR ' H-Cell
`
`
`
`VR, very rare; H—Ceil, Hepatocellular; Chol, Cholestasis; Necr, necrosis, AT, levels of aminotrans-
`ferases increased, but overt liver disease very rare; Idio, idiosyncrasy;TOX, intrinsic toxicity;VOD, veno—
`occlusive disease.
`ain concert with other treatment has led to VOD, portal hypertension due to hepatoportal sclerosis or
`nodular regenerative hyperplasia (see Table 23.18).
`
`——
`—
`H-Cell/Necr
`H—Cell/Necr
`H<Cell/Mild
`
`-
`—
`Idio
`Idio
`Idio
`
`——
`ldio
`ldio
`
`ldio
`ldio
`
`TOX
`TOX
`TOX
`——
`TQX
`TOX
`
`None
`None
`VR
`VFt
`VFi
`
`None
`VR
`VR
`
`VR
`VR
`
`Yes
`Yes
`Yes
`Yes
`Yes
`Yes
`
`—
`H-Cell
`H-Cell
`
`Chol
`Chol
`
`H-Ceii
`AT
`AT
`H-Celi
`H—Cell
`H-Cell/Chol
`
`chN:
`
`CH -CH ~Cl
`2
`2
`CHz-CHZ-Ci
`
`.CHz-CHz-Cl
`0 M
`GEOCW‘CHz-Cl
`
`0
`
`'t‘H
`
`it:
`
`o
`
`,CHz—CHz-CI
`N‘
`
`CHz-CHz-Cl
`
`Mechlorelhamine
`
`Cyclophosphamide
`
`Uracil Mustard
`
`,CHz-CH2~CI
`HOOC-GH-CH2‘©‘N‘
`CHz'CHz'CI
`NH;
`
`0. (,0
`F’s
`<
`_
`_
`LAO“. CH2 CH2 (3'
`CHZ-CHa-Ci
`
`,CHfCH >Cl
`COOH—CHg—CHg—CH2~@~N\
`2
`CH -CH -Ci
`2
`2
`
`Melphalan
`
`iiosfamide
`
`Chlorambucil
`
`FIG. 23.3. Derivatives of nitrogen mustard.
`
`N
`N
`,CTz
`(I3H2\
`CH! YNY ‘CHQ
`NON
`Y
`N
`CH{—‘CH2
`
`CH2WCH2
`CHA
`9‘
`,CH2
`N—P—N\
`i
`l
`CH{
`'3'
`CH2
`
`,CH2
`9
`(3 WP N]
`l
`\il/CZHS
`CH2
`N——N
`
`2
`
`Triethylenemelamine (TEM)
`
`Thiolepa
`
`Azalepa
`
`FIG. 23.4. Derivatives of ethyleneimine.
`
`CHS‘SOZ’O-‘CHZ-CHg-CHz-CHa—O-SOQ-CH3
`FIG. 23.5. Busultan
`
`IMMUNOGEN 2041, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`IMMUNOGEN 2041, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`680 / HEPATOTOXICITY
`
`H
`
`eHon
`0H
`
`OH
`
`O
`0
`CI-CHQ-CHz-tf-(E‘NH‘CHZ'CHZ'CI C‘_CH2.CH2.N_E_NH_<:>
`NO
`NO
`
`OH
`
`no
`OH
`H NH~§-N~CH3
`o
`
`Carmustine (BCNU)
`
`9
`4. _ -
`_
`.
`CH
`9' CHECK)“ c “HO 3
`NO
`
`Semustlne
`
`Lornusline (CCNU)
`
`Streptozolncin
`
`H
`
`CHEOH
`O H
`
`OH?
`H
`OH NO.
`on 0H
`OH
`H NH-(E—N-CHg-CHz-Cl
`(CICHZCH2)2N-COO @.
`O
`Ch’orozotocin
`Estramustine
`
`logues of folic acid, pyrimidines, and purines (Table 23.7).
`
`FIG. 23.6. Nitrosourea derivatives.
`
`(methyl-CCNU) have led to elevated aminotransferase
`concentrations (9).
`Streptozocin. This antibiotic (Zanosar, Pharmacia &
`Upjohn, Bridgewater, NJ), a nitrosourea—related com-
`pound, can produce hepatocellular
`injury including
`necrosis (4). Abnormal aminotransferase levels have been
`reported in 15% (3) and 67% (9) of recipients of the drug;
`but the abnormalities are of little clinical moment (3).
`Chlorozocin has been reported to cause cholestatic injury
`accompanied by hepatocellular damage (7,8,9). The
`injury appears to be dose related (9).
`Triazenes. Despite the antimetabolite structure of
`dacarbizine (DTIC, Bayer Pharmaceutical Products Inc,
`Liberty Corner, NJ) (Fig. 23.7), its antitumor effect is
`mediated apparently by metabolic activation in the liver
`to an alkylating agent (7). The hepatic injury attributed to
`this drug may also result from production of the alkylat-
`ing metabolite in the liver. The most important lesion is
`VOD, of which a number of fatal cases have been
`reported (4,89,11,41). Some of these cases developed
`during the administration of DTIC concurrently with
`other agents; others followed the use of DTIC alone.
`Hepatic vein thrombosis also has been recorded in recip-
`ients of DTIC (39,40).
`Procarbazine. This drug (Matulane, Roche Pharma-
`ceuticals, Basel, Switzerland) appears to produce little
`hepatic injury in most recipients, although jaundice,
`apparently hepatocellular, has been noted in about 1%
`(1,9). The extrahepatic threat, however, outweighs in
`importance the hepatotoxicity of this agent (9).
`
`_
`
`ANTIMETABOLITES
`
`Antimetabolites employed in oncotherapy include ana-
`
`leukemia, polycythemia vera, and primary thrombocyto-
`sis and in the preparation for bone marrow transplanta—
`tion, has been implicated in several cases of cholestatic
`jaundice (23,24) and in a case of porphyria cutanea tarda
`(86).
`In concert with thioguanine,
`it also has been
`incriminated in the production of nodular regenerative
`hyperplasia and portal hypertension (54—57). Also, in
`combination with cyclophosphamide, busulfan has been
`incriminated in the production of VOD (11,87).
`Dimethylbusulfan, a related agent, also has been asso-
`ciated with VOD (9,49).
`
`Nitrosoureas
`
`These compounds apparently act as alkylating agents
`through the liberation of active products (7) (Fig. 23.6).
`The parent compound, carmustine (BCNU), has pro—
`duced hepatic injury in experimental animals and hepatic
`dysfunction in up to 25% of patients taking therapeutic
`doses (68,88—90). The hepatic injury, manifested by
`increased AST activity and jaundice as well as necrosis,
`appears to be reversible. Kupffer cell injury is presumed
`to account for abnormal hepatic scans when radioactive
`colloid is used for scintigraphy (68). In animals, BCNU
`has been found to lead to pericholangitis and necrosis of
`bile ducts
`(6). Lomusrine
`(CCNU)
`and semustine
`
`CHZ—u—u—CHQ
`H H
`
`HCI
`
`o
`
`CH
`
`tort—NH—b'
`CH2
`
`o.
`
`.
`C‘NH
`
`2
`,CH3
`N=N—N‘
`CH3
`
`N
`
`f
`HN—-
`
`Dacarbazine
`
`Procarbazine
`
`FlG. 23.7. Triazines
`
`IMMUNOGEN 2041, pg. 8
`Phigenix v. Immunogen
`|PR2014—00676
`
`IMMUNOGEN 2041, pg. 8
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`23: ONCOTHERAPEUTIC AND IMMUNOSUPPRESSIVE AGENTS / 681
`
`Fat, cirrhosisa
`Fat, necrosis
`Hepatoceliular
`
`Hepatocellular/cholestatic
`Hepatocellular/cholestatic
`Hepatoceiluiar/VOD
`H-Ceil, Chol, VOD, peiiosis
`Fat, necrosis, cirrhosis
`Hepatoceilular
`None reported
`None reported
`
`TABLE 23.7. Hepatotoxic effects of anticancer and immunosuppress/ve drugs: ant/metabolites
`Mechanism
`Hepatic injury
`Drugs
`______________________________.__.——————————————-——-—-—-
`Antitolates
`Intrinsic toxicity
`Methotrexate (Mexate)
`intrinsic toxicity
`Dichloromethotexate
`Intrinsic toxicity
`083717”
`Antipurines
`Mercaptopurine (Purinethol)
`Chloropurine
`Thioguanine (TG, Lanvis)
`Azathioprine (lmuran)
`Frentizoie
`Pentostatin (Nipent)
`Cladribine (Leustatin)
`Fludarabine (Fludara)
`Antipyrimidines
`Hepatocellularc
`5-Fiuorouracilc (Adrucii)
`Cholestatic,d VOD
`Floxuridine (FUDFI)
`Fat, Necrosis
`5-Azacytidine
`Hepatocellular
`6-Azacytidine
`Hepatocellular, VOD
`Cytarabine (ara-C)
`Hepatoceliular
`Pseudoisocytidine
`H-cell, hepatocellular, presumably necrosis; Chol, cholestasis; VOD, veno-occlusive disease.
`aCan cause necrosis in large parenteral doses.
`”08371 7 is a quinazoline thymidylate synthase inhibitor; it is Ni0-propargyl-5,8-dideazoiolic acid.
`COnly with parenteral administration.
`dCholestasis due to scierosing cholangitis when drug administered by pump—perfusion into hepatic
`artery.
`
`intrinsic toxicity
`intrinsic toxicity
`intrinsic toxicity idiosyncrasy
`intrinsic toxicity idiosyncrasy
`intrinsic toxicity
`—
`—
`——
`
`intrinsic toxicity
`Intrinsic toxicity
`intrinsic toxicity
`intrinsic toxicity
`Intrinsic toxicity idiosyncrasy
`intrinsic toxicity
`
`Each category contains agents that can cause hepatic
`injury (6). L-Asparaginase is an enzyme discussed with
`the natural products. Although not an antimetabolite in the
`usual sense, it behaves like one, because it leads to selec-
`tive deprivation of an amino acid (6).
`
`plastic disease, where usually very large doses are given
`intermittently,
`the hepatic injury has been acute bio-
`chemical abnormality or clinical disease of no lasting
`importance. In rheumatoid arthritis, hepatic injury has
`been relatively minor, consisting of biochemical abnor—
`malities and fibrosis.
`
`Methotrexate
`
`This folate antagonist (Fig. 23.8) has been used in th'e
`treatment of leukemia and other neoplastic disease for
`many years. Its mode of action appears to be inhibition of
`dihydrofolate reductase and a consequent inhibition of
`synthesis of purines and pyrimidines (7). A voluminous
`literature details its hepatotoxic effects (l~9,88’187).
`Within a few years after the introduction of MTX into
`clinical use, hints that it might be hepatotoxic appeared.
`At first in children with leukemia (9le94), and later in
`patients with psoriasis (95’135), evidence of the hepato-
`toxic potential of MTX accumulated. Treatment of neo-
`plastic disease including leukemia (9i—94,l42—i45),
`choriocarcinoma (146), localized tumors, and other male
`ignant states (147—153) and of uveitis (145) also led to
`hepatic injury. In more recent years, decade, widespread
`use of MTX for the treatment of rheumatoid arthritis has
`led to limited evidence of hepatic injury (154—167).
`Adverse effects of the drug relate to the clinical set—
`ting. In psoriasis, the significant hepatic injury attrib—
`uted to MTX has been steatosis and cirrhosis. In neo~
`
`Hepatic Injury in the Treatment of Psoriasis
`Most of the information on the hepatotoxic effects of
`this anticancer agent has been acquired in the course of
`its use in psoriasis. During three decades of that use,
`many recipients of MTX were found to have fatty liver,
`and some had fibrosis or cirrhosis (6,96—136). Figures
`for cirrhosis ranged from 4% to more than 25%; for
`fibrosis, from 8% to 30%; and for cirrhosis plus fibrosis,
`about 50% in some reports (Table 23.8). Most of the
`reports, however, did not distinguish between preexisting
`liver abnormalities and the effects of MTX, because in
`most instances information on pretreatment status of the
`liver was not available (134). Only with reports of hepatic
`
`H2N
`
`N
`N
`Y
`CH3
`0
`COOH
`,
`. /
`.
`.
`NO 0 H r'r-@~'c c'H
`N
`e
`N
`CHa-CHZ-COOH
`NH2
`H
`H
`
`FIG. 23.8. Structure of methotrexate.
`
`IMMUNOGEN 2041, pg. 9
`Phigenix v. Immunogen
`IPR2014-00676
`
`IMMUNOGEN 2041, pg. 9
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Year
`1970
`1971
`1971
`1972
`
`1972
`
`1973
`1973
`1973
`1974
`1974
`1977
`1980
`
`injury from MTX treatment, confirmed by comparison of
`pretreatment and posttreatment biopsies, was the phe-
`nomenon clearly established and the issue put in perspec—
`tive (134,136). The suspicion that some of the abnormal—
`ities attributed to MTX were preexisting and apparently
`associated with the psoriasis was confirmed (134). Stea-
`tosis, inflammation, and necrosis are common in the liv-
`ers of untreated patients with psoriasis. Fibrosis and cir-
`rhosis, however, are not common (129), and the reported
`incidence of these lesions in patients treated with MTX,
`although wide—ranging (Table 23.8), demonstrates an
`adverse effect of the drug. Perhaps the most important
`figures are those of Zacchariae et al. (129), who found
`that psoriatic patients who had not received MTX had a
`0.6% prevalence of cirrhosis, whereas those treated with
`the drug for 5 years had a 25% incidence.
`There is now a consensus that MTX can be hepato—
`toxic, that attention to factors modifying the hepatotoxic
`potential can decrease the likelihood of significant hep—
`atic injury, and that appropriate monitoring can keep it
`contained (134,136). However, there is also the view that
`the hepatotoxic threat of MTX, although real, has been
`
`682 / HEPATOTOXlClTY
`TABLE 23.8. Incidence of hepatic fibrosis cirrhosis or both after methotrexate treatment for psoriasis
`(no "baseline" biopsies obtained)
`°/o
` incidence ( )
`
`Fibrosis
`

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