throbber
Hepatotoxicity in Cancer Patients Receiving erb-38, a
`Recombinant Immunotoxin That Targets the erbB2 Receptor

`Lee H. Pai-Scherf, Jacy Villa, Deborah Pearson, et al.
`Clin Cancer Res  
`
`1999;5:2311-2315.
`




`


`
`Updated version

`
`
`
`Access the most recent version of this article at:
` http://clincancerres.aacrjournals.org/content/5/9/2311

`
`Cited Articles

`Citing articles

`
`This article cites by 14 articles, 8 of which you can access for free at:
`http://clincancerres.aacrjournals.org/content/5/9/2311.full.html#ref-list-1

`This article has been cited by 18 HighWire-hosted articles. Access the articles at:
`
`http://clincancerres.aacrjournals.org/content/5/9/2311.full.html#related-urls

`
`
`
`E-mail alerts

`Reprints and
`Subscriptions

`Permissions

`
`Sign up to receive free email-alerts
`
` related to this article or journal.
`
`To order reprints of this article or to subscribe to the journal, contact the AACR Publications
`
`.pubs@aacr.org
`Department at

`To request permission to re-use all or part of this article, contact the AACR Publications
`.
`permissions@aacr.org
`Department at

`
`
`
`Downloaded from Downloaded from
`
`
`
`clincancerres.aacrjournals.org clincancerres.aacrjournals.org
`
`
`on October 30, 2014. © 1999 American Association for Canceron October 30, 2014. © 1999 American Association for Cancer
`
`Research. Research.
`
`IMMUNOGEN 2029, pg. 1
`Phigenix v. ImmunoGen
`IPR2014-00676
`
`

`

`Vol. 5, 2311–2315, September 1999
`
`Clinical Cancer Research
`
`2311
`
`Advances in Brief
`
`Hepatotoxicity in Cancer Patients Receiving erb-38, a Recombinant
`Immunotoxin That Targets the erbB2 Receptor
`
`Lee H. Pai-Scherf, Jacy Villa, Deborah Pearson,
`Thelma Watson, Edison Liu,
`Mark C. Willingham, and Ira Pastan1
`Laboratory of Molecular Biology, Division of Basic Sciences
`[L. H. P-S., I. P.], and Laboratory of Molecular Signaling and
`Oncogenesis [J. V., E. L.] and Medicine Branch [D. P., T. W.],
`Division of Clinical Sciences National Cancer Institute, NIH,
`Bethesda, Maryland 20892; and Department of Pathology, Wake
`Forest University, Winston-Salem, North Carolina 27157 [M. C. W.]
`
`Abstract
`To exploit overexpression of erbB2 in human cancers,
`we constructed a single-chain immunotoxin (erb-38) that
`contains the Fv portion of monoclonal antibody e23 fused to
`a truncated form of Pseudomonas exotoxin A. In a Phase I
`study, five breast cancer patients and one esophageal cancer
`patient received three doses of erb-38 at 1.0 and 2.0 mg/kg.
`Hepatotoxicity was observed in all patients. Immunohisto-
`chemistry showed the presence of erbB2 on hepatocytes
`explaining the liver toxicity of erb-38. We suggest that tar-
`geting of tumors with antibodies to erbB2 armed with ra-
`dioisotopes or other toxic agents may result in unexpected
`organ toxicities due to erbB2 on normal cells.
`
`Introduction
`erbB2/HER2/neu encodes an Mr 185,000 cell membrane
`glycoprotein with tyrosine kinase activity. Overexpression of
`the erbB2 protein and its inherent tyrosine kinase activity causes
`loss of growth control and has an important role in the devel-
`opment of breast cancer and several other human cancers (1–3).
`erbB2 has been reported previously to be minimally expressed
`in normal adult tissues (4). For this reason, erbB2 is an attractive
`target for antibody-directed therapies. To exploit the overex-
`pression of erbB2 in many human cancers (breast, stomach,
`lung, and ovary), we made a single-chain immunotoxin erb-38
`using MAb2 e23 (5), which reacts with erbB2.
`erb-38 is a Mr 63,000 recombinant protein composed of the
`Fv portion of MAb e23 fused to PE38, a truncated form of PE
`with a molecular weight of 38,000. MAb e23 was selected from
`
`Received 2/18/99; revised 5/14/99; accepted 5/26/99.
`The costs of publication of this article were defrayed in part by the
`payment of page charges. This article must therefore be hereby marked
`advertisement in accordance with 18 U.S.C. Section 1734 solely to
`indicate this fact.
`1 To whom requests for reprints should be addressed, at Laboratory of
`Molecular Biology, National Cancer Institute, NIH, 37 Convent Drive,
`MSC 4255, Bethesda, MD 20892-4255. Phone: (301) 496-4797; Fax:
`(301) 402-1344; E-mail: pasta@helix.nih.gov.
`2 The abbreviations used are: MAb, monoclonal antibody; PE, Pseudo-
`monas exotoxin A; AST, aspartate aminotransferase; ALT, alanine
`aminotransferase; NCI, National Cancer Institute; MTD, maximum tol-
`erated dose.
`
`a panel of antibodies to erbB2 because it produced very active
`conventional immunotoxins when it was chemically linked to
`PE (6). PE is a Mr 66,000 protein secreted by Pseudomonas
`aeruginosa. PE is composed of three major structural domains
`(7): an NH2-terminal cell binding domain (domain Ia), a central
`translocation domain (domain II), and a COOH-terminal cata-
`lytically active domain (domain III). Domain III catalyzes the
`ADP ribosylation and inactivation of elongation factor 2, which
`inhibits protein synthesis and leads to cell death. Deletion of
`domain Ia of PE (amino acids 1–252) and part of domain Ib
`produces a Mr 38,000 protein (PE38) that has low cellular and
`animal cytotoxicity because it cannot bind to PE-specific cellu-
`lar receptors. The gene encoding PE38 was fused to a gene
`encoding the Fv portion of MAb e23 to form erb-38
`[e23(dsFv)PE38]. In this recombinant immunotoxin,
`the Fv
`fragment contains a disulfide bond connecting the light and
`heavy chains of MAb e23 (8). The IC50 of erb-38 is 0.2– 4 ng/ml
`on the various tumor cells lines that express the erbB2 antigen.
`erb-38 is capable of causing complete remission in nude mice
`bearing epidermoid carcinoma (A431) and breast cancer
`(MCF-7; Ref. 9). Preclinical toxicity experiments indicated that
`the LD50 of erb-38 in mice was 450 mg/kg every other day times
`three doses. Death was caused by acute hepatic necrosis. He-
`patic and renal injury was observed in cynomolgus monkeys at
`three doses of $500 mg/kg.3
`On the basis of the biological activity and safety testing
`results, we concluded that erb-38 had excellent antitumor activ-
`ity and acceptable animal toxicities. We initiated a Phase I study
`of erb-38 in patients with advanced carcinoma who had failed
`standard therapy.
`
`Patients and Methods
`Eligibility. Adult patients with metastatic carcinoma
`known to express erbB2 were eligible for this study. Tumors were
`considered positive if they expressed erbB2 on $30% of the tumor
`cells, as determined by immunohistochemistry on formaldehyde-
`fixed, paraffin-embedded tumor blocks. Tumor histology was con-
`firmed by a NIH pathologist. Other eligibility criteria included:
`advanced unresectable disease, failed conventional therapy, Eastern
`Cooperative Oncology Group performance status of 0 or 1, AST
`and ALT levels of ,1.5 times the upper limits of normal, total
`bilirubin levels within normal limits, absolute granulocyte counts of
`.2,000/mm3, and platelet counts of .100,000/mm3. Patients with
`positive hepatitis B antigen, history of any other prior liver disease
`(e.g., alcohol liver disease), central nervous system metastasis, or
`seizure disorders were excluded. Patients with positive antibodies
`to erb-38 were excluded. The clinical protocol and the consent form
`were approved by the Institutional Review Board of the NCI. Prior
`to therapy, patients underwent complete physical examination,
`
`3 Unpublished data.
`
`
`
`clincancerres.aacrjournals.org Downloaded from
`
`on October 30, 2014. © 1999 American Association for Cancer
`Research.
`
`IMMUNOGEN 2029, pg. 2
`Phigenix v. ImmunoGen
`IPR2014-00676
`
`

`

`2312 Phase I Study of Anti-erbB2 Immunotoxin
`
`Table 1 Patient characteristics
`
`Diagnosis
`Age/sex
`Breast cancer
`49/F
`Breast cancer
`36/F
`58/M Esophageal cancer
`35/F
`Breast cancer
`57/F
`Breast cancer
`
`erbB2
`expression
`(%)
`80
`80
`80
`80
`100
`
`Patient
`no.
`1
`2
`3
`4
`5
`
`6
`
`Sites of metastasis
`Prior therapy
`XRTa, Doxo, cyclophosphamide, paclitaxel
`Lung, pleura, bone, skin, LN
`Pleura, pericardium, skin, LN
`Surgery, XRT, CMF, Doxo, 5-FU, taxotere, navalbine
`Liver, lung, LN
`XRT, cisplatin, paclitaxel
`Surgery, CAF, Taxol, Ctx, etoposide/melphalan (PBSC) Lung, skin, LN
`Surgery, XRT, CAF, TAM, arimidex, magace,
`LN
`cyclophosphamide/novantrone/5-FU
`Cyclophosphamide/thiotepa/carboplatin (PBSC)
`Surgery, XRT, CAF, paclitaxel,
`Etoposide/melphalan (PBSC)
`a XRT, radiation therapy; Doxo, doxorubicin; CMF, cyclophosphamide, methotrexate and 5-FU; 5-FU, 5-fluorouracil; CAF, cyclophosphamide,
`Doxo, and 5-FU; Ctx, cyclophosphamide; PBSC, peripheral blood stem cell support; TAM, tamoxifen; LN, lymph node.
`
`40/F
`
`Breast cancer
`
`100
`
`Lung, bone, skin
`
`Table 2 Dosing and drug-related toxicity
`
`Patient
`no.
`1
`2
`3
`4
`5
`6
`
`Dose
`(mg/kg)
`2.0
`2.0
`2.0
`2.0
`1.0
`1.0
`
`AST/ALT
`2
`3
`2
`3
`3
`3
`
`Toxicity grade
`
`Nausea/vomiting
`
`Skin rash
`
`2
`
`1
`
`3
`
`complete blood count, determination of chemistry profile, urinaly-
`sis, chest X-ray, electrocardiogram, tumor staging work-up, and
`tumor measurement. Informed consent was obtained from all pa-
`tients.
`erb-38.
`erb-38 (NSC 683039) was produced and placed
`in vials by NCI-Frederick Cancer Research & Development
`Center. erb-38 was supplied through the Cancer Therapy Eval-
`uation Program, NCI, under Investigational New Drug No.
`7373. erb-38 in phosphate buffer solution was supplied as a
`sterile solution at 0.5 mg/ml in 2-ml vials containing 1.0 mg of
`erb-38. Test and treatment doses were diluted in 0.9% normal
`saline and 0.2% albumin.
`Trial Design.
`erb-38 was administered by i.v. infusion
`over 30 min on days 1, 3, and 5. All patients received a 10-mg test
`dose on day 1 given as a bolus over 2 min. The second and third
`doses were delayed or withheld if any measure of toxicity was not
`less than grade II on the scheduled day of administration. A cycle
`could be repeated every 28 days, provided that the patient had not
`developed neutralizing antibodies against erb-38 and had no dis-
`ease progression. Patients were to be entered in groups of three.
`Dose escalation was to based on the modified Fibonacci series.
`While on study, patients were followed with complete
`blood count, blood chemistry and urine. Tumor imaging was
`repeated 1 month after therapy. Toxicity and response to treat-
`ment were graded by the NCI Common Toxicity Criteria. Lim-
`iting toxicity was defined as any grade III or grade IV toxicity.
`Pharmacokinetics. Blood samples were collected at the
`following times for pharmacokinetics: pretreatment; 2, 15, 30,
`45, 60, and 90 min; and 2, 4, 8, 12, 24, and 48 h. After the
`second and third doses of erb-38, samples were collected at 2
`min, 30 min, and 4 h. The concentration of erb-38 was deter-
`
`mined by incubating various dilutions of serum with N87 cells,
`a gastric cancer cell line known to express erbB2, and measuring
`its ability to inhibit protein synthesis. A standard curve was used
`to determine the amount of erb-38 in each sample. Data were
`weighted inversely, and the fit program used was RSTRIP
`(MicroMath Scientific Inc., Salt Lake City, UT).
`Immunogenicity of erb-38. Antibodies against erb-38
`were assessed pretreatment, on day 21, and then bimonthly
`thereafter by a serum neutralization assay: erb-38 was added to
`serum samples at 0.0 (control), 0.1, 0.5, and 1.0 mg/ml and
`incubated at 37°C for 15 min. The activity of erb-38 was
`assayed by incubating the samples with MCF-7 cells and meas-
`uring its ability to inhibit protein synthesis. A standard curve
`with erb-38 was used to determine IC50. A serum sample was
`considered negative for antibodies against erb-38 when the
`cytotoxic activity of erb-38 was not neutralized when incubated
`with sera at concentrations of .0.1 mg/ml erb-38.
`Immunohistochemistry of Normal Liver. Fresh normal
`human liver samples were frozen by immersion in liquid nitro-
`gen. Cryostat sections (;6 mm) were thaw-mounted on slides
`and fixed in 3.7% formaldehyde in PBS for 10 min at 23°C,
`followed by washing in PBS. Sections were not treated with
`H2O2 or any organic solvents. After blocking in 1% BSA in PBS
`for 10 min, sections were incubated in mouse monoclonal anti-
`erbB2 (CB11; Novocastra Laboratories, Novocastra antibody,
`Vector Laboratories, Burlingame, CA) at 10 mg/ml in BSA-PBS
`for 30 min at 23°C. Alternatively, sections were incubated in
`affinity-purified rabbit anti-erbB2 (code A 0485; Dako Corpo-
`ration, Carpinteria, CA) at 7 mg/ml in BSA-PBS. After further
`washing in PBS, sections were incubated, as appropriate, with
`either goat antimouse IgG or goat antirabbit IgG conjugated
`with horseradish peroxidase (Jackson ImmunoResearch) at 25
`mg/ml in BSA-PBS for 30 min. Peroxidase was detected using
`diaminobenzidine (1 ng/ml) with 0.01% H2O2 in PBS for 10
`min. Controls included deletion of the first antibody step. Sec-
`tions were mounted using dehydration and Permount without
`further counterstains and viewed using bright-field microscopy.
`
`Results
`Six patients were enrolled in this trial. Patient characteris-
`tics are shown in Table 1. The initial dose level was 2 mg/kg
`times three, which was 1/250 of the MTD in monkeys and 1/225
`
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`on October 30, 2014. © 1999 American Association for Cancer
`Research.
`
`IMMUNOGEN 2029, pg. 3
`Phigenix v. ImmunoGen
`IPR2014-00676
`
`

`

`Clinical Cancer Research
`
`2313
`
`Fig. 1 Time course of AST
`(E) and ALT (F) of patient 2,
`who received 2.0 mg/kg (A) and
`patient 5, who received 1.0
`mg/kg erb-38 (B).
`
`of the LD50 in mice. This conservative starting dose was chosen
`due to the fact that the toxicity of immunotoxins in humans has
`thus far been found to be unpredictable. Furthermore, 2.0 mg/kg
`times three has been shown to be safe with two other recombi-
`nant immunotoxins made with PE (LMB-7 and LMB-2).3
`Toxicity. Transient elevation of AST and ALT was ob-
`served in all patients. At 2.0 mg/kg, two patients had grade II
`and two patients had grade III elevations of AST and ALT. The
`toxicity observed in the first cohort prompted us to amend the
`protocol to decrease the dose by 50%. Two patients were treated
`at the 1.0 mg/kg dose level. Both patients had a grade 3 trans-
`aminase elevation after one cycle of erb-38 (Table 2).
`Increases in AST and ALT usually occurred within 24 h
`after the first or second dose of erb-38, and peak levels were
`reached 24 – 48 h after the last dose. Two patients required a
`50% dose reduction for the third dose (patients 2 and 6), and in
`two patients, the third dose was held (patients 4 and 5). Hepatic
`transaminases returned to baseline levels 14 –21 days following
`therapy. Fig. 1 shows the time course of AST and ALT of
`patient 2, who received 2.0 mg/kg erb-38, and patient 5, who
`received 1.0 mg/kg erb-38. There was no evidence of liver
`enlargement or right upper quadrant tenderness on exam. Two
`patients had symptoms of nausea and vomiting during therapy,
`with unclear relation to the hepatic injury. No significant drug-
`related changes in bilirubin, alkaline phosphatase, serum albu-
`min, or prothrombin time were observed. Other erb-38-related
`toxicities include a skin rash in one patient 4 days after erb-38
`and nausea and vomiting in two patients.
`As expected in a group of patients with advanced carci-
`noma, several untoward events occurred while the patients were
`on study: pain (n 5 3), malignant pleural effusion (n 5 2),
`malignant pericardial effusion (n 5 1), catheter infection (n 5
`2), venous thrombosis (n 5 1), and proteinuria (n 5 1). One
`patient (patient 6) died of acute respiratory failure in her home-
`town 10 days after discharge, due to pulmonary emboli. An
`autopsy was not performed. These events were attributed to the
`patient’s underlying disease and not related to erb-38.
`Localization of erbB2 in Normal Liver. Although prior
`literature had suggested that erbB2 was not expressed in normal
`liver (4), we used a sensitive immunohistochemical method with
`antibodies to erbB2 that were not available until recently. Rap-
`
`idly frozen normal human liver samples were sectioned with a
`cryostat and postfixed using formaldehyde but were not exposed
`to H2O2 or any organic solvents. Mouse monoclonal CB11 and
`rabbit polyclonal A0485 were used for the detection of erbB2
`using affinity-purified antiglobulins labeled with horseradish
`peroxidase. When viewed without counterstains, the localization
`of erbB2 to the sinusoidal surface of normal hepatocytes was
`clearly visible using either primary antibody (CB11 and AD485;
`Fig. 2). The same result was observed from samples of normal
`liver derived from at least three different patients. Similar results
`were also obtained with antibody E23 (data not shown).
`Pharmacokinetics. Blood samples were collected for
`pharmacokinetics in all patients. erb-38 is cleared monoexpo-
`nentially from the circulation, with a T1/2 that varies from 2.4 to
`10.3 (mean, 3.6 h). A high degree of variation was observed
`from patient to patient. The peak concentration achieved at the
`end of infusion ranged from ,20 ng/ml (patient 1) to 105 ng/ml
`(patient 3). At 1.0 mg/kg, the peak plasma levels were 38.1 and
`39.4 ng/ml. The volumes of distribution of central compartment
`and area under the concentration versus time curve were found
`to be 28.0 6 8.6 ml/kg and 248.7 6 139.6 ng/mlzh, respectively.
`Although full pharmacokinetic analysis was not possible due to
`limited sampling after second and third doses, clearance rates
`appeared to be similar to that after the first dose.
`Immunogenicity. Patients had no neutralizing antibodies
`against erb-38 prior to therapy, as required by protocol. At the
`end of one cycle, only one patient (patient 3) developed neu-
`tralizing antibodies (.1.0 mg/ml) against erb-38 14 days after
`treatment. Retreatment was not possible in this patient, due to
`disease progression.
`Response. No objective responses were observed in the
`trial. One patient had stable disease for 3 months, four patients
`progressed after one cycle of therapy, and one patient was not
`evaluable for response. Patient 2 had improvement of chest wall
`pain after erb-38 that lasted for 3 weeks. Fentanyl transdermal
`patch requirement decreased from 225 to 25 mg/h in this patient.
`
`Discussion
`We have shown that, when a recombinant immunotoxin,
`erb-38, which targets erbB2, is given to cancer patients, it causes
`hepatic injury, manifested by transient elevation of transami-
`
`Downloaded from
`
`on October 30, 2014. © 1999 American Association for Cancerclincancerres.aacrjournals.org
`
`
`Research.
`
`IMMUNOGEN 2029, pg. 4
`Phigenix v. ImmunoGen
`IPR2014-00676
`
`

`

`2314 Phase I Study of Anti-erbB2 Immunotoxin
`
`Fig. 2 Localization of erbB2
`to the surface of normal human
`hepatocytes. Frozen sections of
`normal human liver were incu-
`bated with either a mouse MAb
`to erbB2 (CB11; B) or a rabbit
`polyclonal affinity-purified an-
`ti-erbB2 (A0485; D). Corre-
`sponding controls with deletion
`of the first step but with the
`second step included are shown
`for antimouse IgG (A) or anti-
`rabbit IgG (C). The characteris-
`tic pattern, especially at inter-
`cellular
`junctions,
`of
`the
`sinusoidal surface of hepato-
`cytes is evident (arrows) with
`both antibodies
`(B and D).
`These sections were not coun-
`terstained, and the nuclear lo-
`calization seen in D represents a
`nonspecific pattern seen only
`with the rabbit anti-erbB2 anti-
`(Magnification, 3375;
`body.
`scale bar, 50 mm).
`
`nases. The dose-limiting toxicities of erb-38 occurred at 1.0 and
`2.0 mg/kg when it was given i.v. every other day times three.
`These doses are significantly below the doses necessary to show
`antitumor effects in animals. Although the MTD was not deter-
`mined, based on our preclinical experience with this agent,
`effective serum levels cannot be achieved at doses ,1.0 mg/kg
`(6). In addition, we found that the erb-38 was cleared monoex-
`ponentially from the circulation, with a T1/2 that varied from 2.4
`to 10.3 h. As compared with other biological agents of this
`nature, a great deal of variation was observed from patient to
`patient. At 2.0 mg/kg, the maximum concentration varied from
`,20 to 105 ng/ml. No clear correlation can be made between the
`pharmacokinetic findings and the tumor burden, toxicity, or
`expression of erb-38 on tumor cells (all patients had tumors that
`expressed erbB2 on $80% of the cells).
`The toxicity of erb-38 is most likely due to the presence of
`erbB2 on hepatocytes, not detected by immunohistochemical
`staining in earlier publications (4). We have now repeated these
`assays using newly available antibody to erbB2 (affinity-puri-
`fied rabbit anti-erbB2, code A 0485; Dako Corp.) and have
`shown here that erbB2 is clearly present on the surface of
`normal hepatocytes. The expression of erbB2 is known to be
`very high in ;30% of breast cancers, often due to gene ampli-
`
`fication. Despite the fact that there is a very large difference in
`the amount of erbB2 on the surface of cancer cells relative to the
`small amount present on liver cells, liver toxicity was the first
`biological effect seen in this study. Several factors contribute to
`this finding. One is that hepatocytes are more rapidly exposed to
`agents injected into the circulation than tumor cells. There is a
`barrier to proteins entering tumors because tumors do not have
`lymphatics and there is no connective flow. As a consequence,
`mixing within tumors is solely by diffusion and, therefore, very
`slow. In addition, tumors are often poorly vascularized (10).
`There are several ways in which antibodies are being used
`as antitumor agents. Antibodies have been used by themselves
`to produce antitumor activity, taking advantage of their ability to
`carry out complement-mediated cell killing or ADCC (antibody
`dependent cell- and complement-mediated cytotoxicity) or to
`induce apoptosis directly. One or more of these effects explains
`how Rituximab, which binds to CD20, causes regression of
`lymphomas (11). Recently, clinical data concerning the safety
`and efficacy of a humanized antibody to erbB2 termed Hercep-
`tin were presented (12, 13). The antibody alone has been found
`to produce objective responses in breast cancer and when com-
`bined with chemotherapy results in an increased response rate.
`It is likely that the antitumor activity of the antibody in this
`
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`on October 30, 2014. © 1999 American Association for Cancer
`Research.
`
`IMMUNOGEN 2029, pg. 5
`Phigenix v. ImmunoGen
`IPR2014-00676
`
`

`

`Clinical Cancer Research
`
`2315
`
`setting is dependent on genetic perturbations that alter the con-
`figuration of downstream signaling events, and, thereby, the
`responsiveness of the cancer cells to Herceptin. Thus, the mech-
`anism of killing depends on a genetic abnormality present in the
`cancer cells. In contrast, if the antibody is used to deliver a
`cytotoxic agent, such as a bacterial toxin or radioisotope, the
`death of the target cell will be principally dependent on the
`amount of agent delivered to the cell. In the case of erb-38, it
`appears that liver cells are the initial site of drug action. It has
`also been found in the Herceptin trial that there is evidence of
`cardiotoxicity, perhaps due to the presence of small amounts of
`erbB2 on myocytes or endocardial cells. Unfortunately, the
`information concerning the cardiotoxicity of Herceptin was not
`yet reported at the time of this trial. We did not include a
`throughout cardiac evaluation with echocardiogram or MUGA
`(Multigated Radionuclide Angiography) scan in the initial
`work-up. However, none of the patients had clinical evidence of
`myocardial dysfunction or arrhythmia before or after therapy
`with erb-38 based on their history, physical exam, chest X-ray,
`and electrocardiogram. It is quite possible that, because the
`concentrations of erb-38 given were low, overt cardiotoxicity
`was detected in this small trial. This might have been observed
`if higher doses of erb-38 had been administered.
`The new findings of reactivity with anti-erbB2 antibodies
`on hepatocytes are, for the most part, the result of the availabil-
`ity of the affinity-purified rabbit anti-erbB2. With this antibody,
`which has been shown to be 2– 4-fold more sensitive in detect-
`ing erbB2 in paraffin sections of breast tumors, we were able to
`evaluate carefully the conditions used for histochemistry to
`optimize localization of erbB2 in breast cancer specimens. With
`these new conditions, which include the use of frozen sections
`of freshly frozen tissues, formaldehyde postsectioning fixation,
`the deletion of treatments with any peroxide or organic solvents,
`and the deletion of counterstains, the localization of erbB2 was
`optimal in tumor specimens as well as in normal human liver.
`These results allowed us to go back and test previously available
`monoclonal antibodies, such as CB11 and E23, both of which
`also showed specific surface labeling at a low level in hepato-
`cytes. As shown in Fig. 2, this labeling was homogeneous,
`surface related, and absent in parallel controls for all of the
`antibodies, implying that it was truly specific for erbB2.
`Two other recombinant immunotoxins developed by our
`group have recently been evaluated in Phase 1 trials. Both can
`be given at much higher doses than erb-38. One of these is
`LMB-7 [B3(Fv)PE38], which targets a carbohydrate antigen
`present in colon, breast, and other epithelial cancers (14). The
`MTD of LMB-7 was found to be 24 mg/kg when given every
`other day times three. The dose-limiting toxicities were nausea,
`vomiting and diarrhea, secondary to the targeting of the carbo-
`hydrate antigen present on the gastric mucosa.4 LMB-2 [anti-
`Tac(Fv)PE38] is a recombinant immunotoxin that targets CD25
`(15), the a-subunit of the interleukin 2 receptor. In patients with
`
`leukemia and lymphoma, the MTD was 40 mg/kg, at which
`several clinical responses have been observed.5
`We conclude that the toxicity observed with erb-38 is most
`likely due to the presence of erbB2 on hepatocytes. The target-
`ing of tumors with antibodies to erbB2 that are armed with
`radioisotopes and other toxic agents may result in unexpected
`organ toxicities due to erbB2 expression on normal tissues.
`
`References
`1. Slamon, D., Godolphim, W., Jones, L. A., Holt J. A., Wong, S. B.,
`Keith, D. E., Levin, W. J., Stuart, S. G., Udove, J., Ullrich, A., and Press,
`M. F. Studies of the HER2/neu protooncogene in human breast and
`ovarian cancer. Science (Washington DC), 244: 707–712, 1989.
`2. Schneider, P. M., Hung, M. C., Chiocca, S. M., Manning, J., Zhao,
`X. Y., Fang, K., et al. Differential expression of the erbB2 gene in
`human small cell and non-small cell lung cancer. Cancer Res., 49:
`4968 – 4971, 1989.
`3. Park, J., Rhim, J. S., Park, S. C., Kimm, S. W., and Kraus, M. H.
`Amplification, overexpression and rearrangement of the erbB-2 pro-
`tooncogene in primary human stomach carcinomas. Cancer Res., 49:
`6605– 6609, 1989.
`4. Press, M. F., Cordon-Cardo, C., and Slamon, D. J. Expression of the
`HER-2/neu proto-oncogene in normal human adult and fetal tissues.
`Oncogene, 5: 953–962, 1990.
`5. Kasprzyk, P. G., Song, S. V., DiFiore, P. P., and King, C. R. Therapy
`of an animal model of human gastric cancer using a combination of
`anti-erbB2 monoclonal antibodies. Cancer Res., 52: 2771–2776, 1992.
`6. Reiter, Y., Brinkmann, U., Jung, S-H., Pastan, I., and Lee, B. Im-
`proved binding and antitumor activity of a recombinant anti-erbB2
`immunotoxin by disulfide Stabilization of the Fv fragment. J. Biol.
`Chem., 269: 18327–18331, 1994.
`7. Allured, V. S., Collier, R. J., Carroll, S. F., and McKay, D. B.
`Structure of exotoxin A of Pseudomonas aeruginosa at 3.0 angstrom
`resolution. Proc. Natl. Acad. Sci. USA, 83: 1320 –1324, 1986.
`8. Reiter, Y., Brinkmann, U., Kreitman, R., Jung, S-H., Lee, B., and
`Pastan, I. Stabilization of the Fv fragments in recombinant immunotox-
`ins by disulfide bonds engineered into conserved framework regions.
`Biochemistry, 33: 5451–5459, 1994.
`9. Reiter, Y., and Pastan, I. Antibody engineering of recombinant Fv
`immunotoxins for improved targeting of cancer: disulfide-stabilized Fv
`immunotoxins. Clin. Cancer Res., 2: 245–252, 1998.
`10. Jain, R. K. Delivery of novel therapeutic agents in tumors: physi-
`ological Barriers and strategies. J. Natl. Cancer Inst., 8: 570 –576, 1989.
`11. Maloney, D. G., Grillo-Lopes, A. J., Bodkin, D, White, C. A., Liles,
`T. M., and Royston, I. IDEC-C2B8 (Rituximab) Anti-CD20 monoclonal
`antibody therapy in patients with relapsed low-grade non-Hodgkin’s
`lymphoma. Blood, 90: 2188 –2195, 1997.
`12. Cobleigh, M. A., Vogel, C. L., Tripathy, N. J., Tripathy, D., Robert,
`N. J., Scholl, S., Fehrenbacher, L., Paton, V., Shak, S., Lieberman, G.,
`and Slamon, D. Efficacy and safety of Herceptin (humanized anti-Her2
`antibody) as a single agent in 222 humans with Her2 overexpression
`who relapsed following chemotherapy for metastatic breast cancer.
`Proc. Am. Soc. Clin. Oncol., Abstract 376, 97a, 1998.
`13. Slamon, D., Leyland-Jones, B., Shak, S., Paton, V., Bajamonde, A.,
`Fleming, T., Eiermann, W., Wolter, J., Baselga, J., and Norton, L.
`Addition of Herceptin (humanized anti-Her2 antibody) to first line
`chemotherapy for HER2 overexpressing metastatic breast cancer
`(HEr21/MBC) markedly increases anticancer activity: a randomized,
`multinational controlled Phase III trial. Proc. Am. Soc. Clin. Oncol.,
`Abstract 397, 98a, 1998.
`14. Pai, L. H., and Pastan, I. Immunotoxin therapy for cancer. J. Am.
`Med. Assoc., 269: 78 – 81, 1993.
`15. Kreitman, R. J., and Pastan, I. Targeting Pseudomonas exotoxin to
`hematologic malignancies. Semin. Cancer Biol., 6: 297–306, 1995.
`
`4 L. H. Pai-Scherf and I. Pastan, manuscript in preparation.
`
`5 R. J. Kreitman, T. Waldmann, and I. Pastan, manuscript in preparation.
`
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`on October 30, 2014. © 1999 American Association for Cancer
`Research.
`
`IMMUNOGEN 2029, pg. 6
`Phigenix v. ImmunoGen
`IPR2014-00676
`
`

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