throbber

`
`From From
`
`
`
`by guest by guest on November 17, 2014. For personal use only.on November 17, 2014. For personal use only.
`www.bloodjournal.orgwww.bloodjournal.org
`
`
`
`
`
`
`
`
`
`
`
`1999 93: 3678-3684
`
`Selective Ablation of Acute Myeloid Leukemia Using Antibody-Targeted
`Chemotherapy: A Phase I Study of an Anti-CD33 Calicheamicin
`Immunoconjugate: Presented in part at the 1997 Annual Meeting of the
`American Society of Clinical Oncology, Denver, CO; the 1997 European
`Cancer Conference, Hamburg, Germany; and the 1997 Annual Meeting of
`the American Society of Hematology, San Diego, CA.
`
`E.L. Sievers, F.R. Appelbaum, R.T. Spielberger, S.J. Forman, D. Flowers, F.O. Smith, K. Shannon-Dorcy,
`M.S. Berger and I.D. Bernstein
`
`Updated information and services can be found at:
`http://www.bloodjournal.org/content/93/11/3678.full.html
`Articles on similar topics can be found in the following Blood collections
`
` (3965 articles)Clinical Trials and Observations
`   
`
`Information about reproducing this article in parts or in its entirety may be found online at:
`http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests
`
`Information about ordering reprints may be found online at:
`http://www.bloodjournal.org/site/misc/rights.xhtml#reprints
`
`Information about subscriptions and ASH membership may be found online at:
`http://www.bloodjournal.org/site/subscriptions/index.xhtml
`
`Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society of
`Hematology, 2021 L St, NW, Suite 900, Washington DC 20036.
`Copyright 2011 by The American Society of Hematology; all rights reserved.

`
`IMMUNOGEN 2187, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`  


`

`

`From
`
`by guest
`www.bloodjournal.org
`
`
`
`
`For personal use only.on November 17, 2014.
`
`CLINICAL OBSERVATIONS, INTERVENTIONS, AND THERAPEUTIC TRIALS
`Selective Ablation of Acute Myeloid Leukemia Using Antibody-Targeted
`Chemotherapy: A Phase I Study of an Anti-CD33 Calicheamicin
`Immunoconjugate
`
`By E.L. Sievers, F.R. Appelbaum, R.T. Spielberger, S.J. Forman, D. Flowers, F.O. Smith, K. Shannon-Dorcy,
`M.S. Berger, and I.D. Bernstein
`
`Leukemic blast cells express the CD33 antigen in most
`patients with acute myeloid leukemia (AML), but this anti-
`gen is not expressed by hematopoietic stem cells. We
`conducted a study to determine whether normal hematopoi-
`esis could be restored in patients with AML by selective
`ablation of cells expressing the CD33 antigen. In a dose
`escalation study, 40 patients with relapsed or refractory
`CD331 AML were treated with an immunoconjugate (CMA-
`676) consisting of humanized anti-CD33 antibody linked to
`the potent antitumor antibiotic calicheamicin. The capacity
`of leukemic cells to efflux 3,3’-diethyloxacarbocyanine iodide
`(DiOC2) was used to estimate pretreatment functional drug
`
`PATIENTS WITH refractory or recurrent acute myeloid
`
`leukemia (AML) have a dismal prognosis. Toxic effects
`associated with additional conventional chemotherapy are often
`life-threatening, and few patients achieve a complete remission
`(CR). A therapy that more specifically targets leukemia is likely
`to be safer, and possibly more effective than current nonspecific
`chemotherapeutic agents.
`During hematopoietic development, stem cells capable of
`establishing long-term multilineage hematopoiesis give rise to
`progenitors with diminished self-renewal capacity and a greater
`degree of differentiation. During this process, hematopoietic
`cells express distinct cell surface antigens that are also ex-
`pressed by their malignant counterparts. Some of these antigens
`(eg, CD33) are present on maturing normal hematopoietic cells
`and on AML cells, but not on normal hematopoietic stem
`cells.1-4 These findings raise the possibility that an antibody
`can be used to deliver cytotoxic agents to selectively ablate
`
`From the Clinical Research Division, Fred Hutchinson Cancer
`Research Center, Seattle; the Departments of Pediatrics and Medicine,
`University of Washington, Seattle, WA; the Department of Hematology
`and Bone Marrow Transplantation, City of Hope National Medical
`Center, Duarte, CA; and Wyeth Ayerst Research, Radnor, PA.
`Submitted August 19, 1998; accepted January 29, 1999.
`Supported by Wyeth-Ayerst Research. E.L.S. is supported by an
`American Cancer Society Clinical Oncology Career Development Award.
`I.D.B. is an American Cancer Society Clinical Research Professor.
`Presented in part at the 1997 Annual Meeting of the American
`Society of Clinical Oncology, Denver, CO; the 1997 European Cancer
`Conference, Hamburg, Germany; and the 1997 Annual Meeting of the
`American Society of Hematology, San Diego, CA.
`Address reprint requests to E.L. Sievers, MD, Fred Hutchinson
`Cancer Research Center, 1100 Fairview Ave N, D5-280, PO Box 19024,
`Seattle, WA 98109-1024; e-mail: esievers@fhcrc.org.
`The publication costs of this article were defrayed in part by page
`charge payment. This article must therefore be hereby marked ‘‘adver-
`tisement’’ in accordance with 18 U.S.C. section 1734 solely to indicate
`this fact.
`r 1999 by The American Society of Hematology.
`0006-4971/99/9311-0018$3.00/0
`
`resistance. Leukemia was eliminated from the blood and
`marrow of 8 (20%) of the 40 patients; blood counts returned
`to normal in three (8%) patients. A high rate of clinical response
`was observed in leukemias characterized by low dye efflux in
`vitro. Infusions of CMA-676 were generally well tolerated,
`and a postinfusion syndrome of fever and chills was the
`most common toxic effect. Two patients who were treated
`at the highest dose level (9 mg/m2) were neutropenic G5
`weeks after the last dose of CMA-676. These results show
`that an immunoconjugate targeted to CD33 can selectively
`ablate malignant hematopoiesis in some patients with AML.
`r 1999byTheAmericanSocietyofHematology.
`
`malignant myeloid and immature normal cells while sparing
`normal stem cells.
`The myeloid cell surface antigen CD33 is an attractive target
`for this approach, as it is expressed on AML blast cells from
`about 90% of patients.2,3 CD33 also may be expressed by most,
`if not all, of the malignant precursors in at least some cases of
`AML, as precursor cells that lack CD33 from some patients
`give rise to normal granulocyte/monocyte precursors in a
`marrow long-term culture system.5,6 The possibility that anti-
`CD33 antibody could be used to deliver a cytotoxic agent to
`malignant cells was suggested by studies showing rapid satura-
`tion of leukemic blast cells in peripheral blood and marrow after
`intravenous administration of approximately 5 mg/m2 radioio-
`dinated anti-CD33 antibodies.7,8 Further, in vitro studies showed
`rapid internalization of the antibody by the target cell.9 To test
`the concept of selective ablation of malignant hematopoiesis
`and to evaluate the safety of this approach, we treated patients
`with relapsed or refractory AML with escalating doses of
`CMA-676, antibody-targeted chemotherapy consisting of an
`engineered human anti-CD33 antibody linked with the potent
`antitumor antibiotic calicheamicin.10
`
`MATERIALS AND METHODS
`
`Patients. Patients were required to have AML that was either
`refractory to standard therapy or that had recurred after remission.
`Patients whose leukemia relapsed after a marrow or peripheral blood
`stem cell transplant were required to have had successful engraftment
`(platelets $20 3 103/µL without transfusion and an absolute neutrophil
`count [ANC] $500/µL) before relapse. Each prospective patient
`underwent a bone marrow aspirate to confirm that morphologic
`evidence of leukemia was present and to document leukemic blast cell
`surface expression of the CD33 antigen. Patients were required to have
`a Karnofsky performance status $60%, a white blood cell count #30 3
`103/µL, serum creatinine #2.0 mg/dL, and serum bilirubin #1.5 mg/dL,
`to have recovered from toxic effects of previous antineoplastic therapy,
`and to be able to give informed consent. Patients were ineligible if they
`had previously received anti-CD33 antibody treatment, were pregnant
`or nursing, had a prior malignancy, or had active symptomatic central
`nervous system leukemia or an uncontrolled life-threatening infection.
`Recombinant humanized anti-CD33-calicheamicin drug conjugate.
`CMA-676 was produced and provided for clinical study by Wyeth-
`
`3678
`
`Blood,Vol 93, No 11 (June 1), 1999: pp 3678-3684
`
`IMMUNOGEN 2187, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`From
`
`by guest
`www.bloodjournal.org
`
`
`
`
`For personal use only.on November 17, 2014.
`
`SELECTIVE ABLATION OF AML USING ANTIBODY-TARGETED CHEMOTHERAPY
`
`3679
`
`Ayerst Research (Radnor, PA) under an Investigational New Drug
`Application. Celltech Therapeutics, Ltd (Slough, UK) transformed the
`murine-derived IgG1 monoclonal antibody (p67.6)8 into an engineered
`human IgG4 antibody (hP67.6), which was then conjugated to the
`enediyne calicheamicin10 using a Nac-gamma linker molecule (Hamann
`et al, manuscript in preparation). Calicheamicin binds in a sequence-
`specific manner to the minor groove of DNA and induces double-strand
`DNA breaks that ultimately induce cell death.10 Specific cytotoxicity of
`CMA-676 against CD331 leukemia cells was documented in colony-
`forming assays in vitro, against leukemic cell lines in vitro, and in vivo
`in nude mice (data not shown).
`In this open, single-arm, phase I dose escalation
`Protocol design.
`study, CMA-676 was administered to patients with relapsed or refrac-
`tory CD331 AML at two study sites. Informed consent was obtained
`from all patients in accordance with the institutional review boards of
`the participating institutions. In a single-dose tolerance study with
`CMA-676 in chimpanzees, an intravenous dose of 0.5 mg/m2 (twice the
`initial clinical dose of 0.25 mg/m2) was well
`tolerated, with no
`compound-related clinical signs of clinical pathology changes evident
`throughout a 15-day postinfusion period (data not shown). In previous
`clinical studies in patients using radiolabeled mP67.6 antibody,8 dose-
`limiting toxicity was not observed at doses up to and including 17
`mg/m2 antibody trace-labeled with 131I. However, saturation of CD33
`binding sites was achieved at a dose level of approximately 5 mg/m2.
`Hence, at
`least
`three patients were treated at each dose level of
`CMA-676 with up to three infusions of 0.25, 0.5, 1, 2, 4, 5, 6, and 9 mg
`protein per square meter of body surface area. If one or two of the first
`three of these patients experienced a grade III toxic effect (see below),
`three additional patients were to be treated at the same dose. The
`maximum tolerated dose was defined as one dose level below the dose
`that was associated with unacceptable CMA-676–related toxicity.
`Dose-limiting toxicity was defined as severe marrow hypoplasia of
`more than 6 weeks duration after a single dose of CMA-676, one grade
`IV study drug-related toxic effect or two grade IV toxic conditions of
`ambiguous relationship to CMA-676.
`CMA-676 was administered as a single 2-hour intravenous infusion.
`All cytokines or chemotherapeutic agents were withdrawn before
`treatment. Patients without leukemic progression and drug-related,
`nonhematologic toxic effects judged to be grade III or less could receive
`one or two subsequent cycles of CMA-676 with at least 14 days
`between cycles. Patients were treated with acetaminophen 650 mg
`orally and diphenhydramine 25 to 50 mg intravenously 15 to 30 minutes
`before infusion of CMA-676. Patients who achieved a CR and
`subsequently had a relapse of their leukemia could receive retreatment
`at the dosage level being evaluated at the time of retreatment.
`Patient evaluation. Patients were examined for acute toxic effects
`in accordance with the World Health Organization toxic effect grades.
`Peripheral blood specimens were obtained to study the pharmacokinet-
`ics of CMA-676, to evaluate for evidence of CMA-676 binding to
`CD33-positive cells, and to measure the effects of CMA-676 on
`hematologic variables and serum chemistry parameters. A complete
`blood count was obtained before the initial dose and each day for 2
`weeks thereafter, or until recovery of granulocyte and platelet levels to
`the prestudy levels. Hepatic and renal function were measured before
`initial dose administration, three times per week thereafter, and on day
`28 after the last dose of CMA-676.
`Bone marrow aspirates were obtained before initial dose administra-
`tion with CMA-676 on days 1, 7, and 14 of each treatment cycle and on
`days 1, 7, 14, and 28 of the last treatment cycle. All marrow specimens
`were examined by light microscopy to estimate cellularity and to detect
`any residual leukemic blast cells. Patients with progressive disease were
`removed from the study. Disappearance of leukemia was defined as the
`absence of peripheral blasts and the presence of #5% blast cells in the
`bone marrow by light microscopic evaluation. CR was defined as
`
`disappearance of leukemia in addition to an ANC .1,500/µL and a
`platelet count .100 3 103/µL, without transfusions.
`Laboratory investigations. Total hP67.6 antibody concentrations in
`plasma samples were determined using an enzyme-linked immunosor-
`bent assay (ELISA). Formation of antigen-antibody CMA-676 bound to
`peripheral blood mononuclear cells was detected by flow microfluorim-
`etry. Cells were incubated with biotinylated goat monoclonal anti-
`human IgG4, followed by avidin-fluorescein isothiocyanate. Cells
`incubated with avidin-fluorescein isothiocyanate alone comprised the
`negative control. The saturation percentage was defined as 100 times the
`ratio of the fluorescence intensity of patient mononuclear cells (minus
`the negative control) over the maximum fluorescence intensity (minus
`the negative control). Maximum achievable saturation was determined
`by incubating patient mononuclear cells from the same time point with
`saturating amounts of CMA-676 in vitro before the addition of the
`anti-human IgG4 antibody. The efflux of 3,3’-diethyloxacarbo-
`cyanine iodide (DiOC2) from CD33-positive blast cells was measured
`as an indication of functional drug efflux.11 Serum samples obtained
`from each patient before CMA-676 administration, on day 7 after
`initial doses, and on days 7, 14, 21, and 28 after administration of the
`final dose of CMA-676 were analyzed for anti-hP67.6 (humanized
`mouse antibody) or anti-calicheamicin/linker immune response by
`ELISA.
`
`RESULTS
`
`Patient characteristics. A total of 40 patients with relapsed
`or refractory AML were enrolled. Patient characteristics are
`shown in Table 1. Three to eight patients were treated at each of
`eight dose levels of CMA-676: 0.25, 0.5, 1, 2, 4, 5, 6, and 9 mg
`protein per square meter of body surface area.
`Nonhematologic toxicity. The most frequently reported
`drug-related adverse events are summarized in Table 2. The
`most commonly observed nonhematologic side effect, a syn-
`drome of fever and chills, occurred in 32 of 40 (80%) patients
`beginning 2 to 4 hours after the start of the 2-hour intravenous
`infusion. The syndrome was limited to grade I to II except in
`three instances. Grade III fever and chills occurred in two
`patients after receiving CMA-676 at 6 and 9 mg/m2, respec-
`tively. The third patient (COH-008) who had asymptomatic
`
`Table 1. Patient Characteristics (N 5 40)
`
`Age: median (range)
`Sex
`Stage of disease
`First relapse
`$Second relapse
`Prior stem cell transplant
`Allogeneic
`Autologous
`None
`Cytogenetic abnormality
`t(8;21)
`inv(16)
`t(4;11)
`t(6;11)
`t(9;11)
`Complex abnormality
`None detected
`Unknown
`Prior myelodysplastic phase
`Present
`Absent
`
`54 (24-73)
`21M:19F
`
`20
`20
`
`14
`4
`22
`
`1
`2
`1
`1
`2
`22
`10
`1
`
`9
`31
`
`IMMUNOGEN 2187, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`From
`
`by guest
`www.bloodjournal.org
`
`
`
`
`For personal use only.on November 17, 2014.
`
`3680
`
`SIEVERS ET AL
`
`Table 2. Most Frequently Reported Drug-Related Adverse Events (No. of Patients)
`
`Event*
`Grade 0-I fever/chills
`Grade II fever/chills
`Grade III† fever/chills
`Grade IV fever/chills
`Grade III hepatic enzyme
`elevation (AST or ALT)
`Grade IV hepatic enzyme
`elevation (AST or ALT)
`
`0.25
`
`(n 5 4)
`4
`—
`—
`—
`
`—
`
`—
`
`0.5
`
`(n 5 3)
`2
`1
`—
`—
`
`1
`
`—
`
`1
`
`(n 5 4)
`1
`3
`—
`—
`
`1
`
`—
`
`CMA-676 Dose (mg/m2)
`
`2
`
`(n 5 3)
`1
`2
`—
`—
`
`—
`
`—
`
`4
`
`(n 5 6)
`4
`2
`—
`—
`
`—
`
`—
`
`5
`
`(n 5 6)
`1
`5
`—
`—
`
`4
`
`—
`
`6
`
`(n 5 8)
`3
`3
`1‡
`1§
`
`1
`
`1\
`
`9
`
`(n 5 7)
`3
`3
`1
`—
`
`1
`
`—
`
`Abbreviations: AST, alanine serum transaminase; ALT, alanine aminotransferase.
`*Grades are defined according to modified World Health Organization toxicity grading scale.
`†Modified to include fever ,40°C with asymptomatic hypotension.
`‡This patient (FH-007) is represented in the table twice. He experienced grade II fever/chills after his initial treatment at 1 mg/m2. He developed
`shortness of breath in association with retreatment with CMA-676 after his leukemia relapsed, presumably caused by an immune reaction to
`CMA-676.
`§This patient (COH-008) who had asymptomatic hypotension and was receiving low-dose dopamine before CMA-676 administration, had a
`temperature .40°C and reversible symptomatic hypotension 5 hours after the initiation of the CMA-676 infusion at 6 mg/m2.
`\This patient (FH-020) with documented concurrent cholelithiasis had a pretreatment AST of 13 U/L that increased to 304 U/L 4 days after
`receiving the first dose of CMA-676, and rapidly declined to the normal range within a week. Immediately before receipt of the second dose of
`CMA-676, her AST increased to 1,804 U/L and again rapidly returned to the normal range. Elevated enzymes were thought to be primarily due to
`the patient’s cholelithiasis.
`
`hypotension and was receiving low-dose dopamine before
`CMA-676 administration, had a temperature .40°C and revers-
`ible symptomatic hypotension 5 hours after the initiation of the
`CMA-676 infusion at 6 mg/m2. Another patient (FH-007)
`developed transient shortness of breath in association with
`retreatment with CMA-676 after his leukemia relapsed, presum-
`ably caused by an immune reaction to CMA-676 (see below).
`Nausea and fatigue were among other less frequent toxic effects
`thought possibly related to CMA-676.
`Reversible, possibly drug-related hepatic transaminase eleva-
`tions from 5 to 10 times the normal range were observed in eight
`patients (Table 2). One additional patient with documented
`concurrent cholelithiasis had a pretreatment alanine serum
`transaminase (AST) of 13 U/L that increased to 304 U/L 4 days
`after receiving the first dose of CMA-676, and rapidly declined
`to the normal range within a week. Immediately before receipt
`of the second dose of CMA-676, her AST increased to 1,804
`U/L and again rapidly returned to the normal range. In this
`patient, elevated enzymes were thought to be primarily due to
`the patient’s cholelithiasis. No patient had a drug-related
`bilirubin elevation of greater than five times the normal range.
`No significant study drug-related central nervous system,
`cardiac, or renal toxic effects were observed. With regard to
`nonhematologic toxic effects, a maximum tolerated dose was not
`reached, and dose-limiting toxicity was not observed.
`Hematologic toxicity. Because most patients had concur-
`rent neutropenia and thrombocytopenia caused by active AML
`at the time of enrollment, whether hematologic toxic effects
`were directly attributable to CMA-676 was often impossible to
`ascertain. However, among nine patients with neutrophil counts
`.1,500 cells/µL before treatment, eight had severe neutropenia
`(,200 cells/µL) within 14 days of CMA-676 infusion regard-
`less of dose level. Figure 1 shows the decline and recovery of
`the neutrophil count from a representative patient during the 2
`weeks after each infusion. Although dose-limiting toxicity was
`
`not formally observed, two of seven evaluable patients had
`prolonged drug-related neutropenia after treatment at a dose
`level of 9 mg/m2. The first patient (FH-023) required 38 days to
`recover to an ANC .500 cells/µL after the third dose of
`CMA-676, and the second patient (COH-015) did not achieve
`neutrophil recovery and died of sepsis 50 days after receiving
`the second dose (Table 3).
`Elimination of blast cells from the peripheral blood and bone
`marrow. Blast cells were identified on a peripheral blood
`smear in 31 (78%) patients before infusion of CMA-676. One
`week after treatment, 22 (71%) of these patients had fewer blast
`cells and in six (19%) of these patients, blast cells had
`completely disappeared from the peripheral blood smears. The
`proportion of patients who experienced reductions in peripheral
`blast cell counts was highest among those who received higher
`
`Fig 1. Relationship between hematologic parameters and time for
`a representative patient (FH-012) who received CMA-676 at 4 mg/m2
`per dose. Arrows denote infusions of CMA-676. All counts refer to
`peripheral blood counts.
`
`IMMUNOGEN 2187, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`From
`
`by guest
`www.bloodjournal.org
`
`
`
`
`For personal use only.on November 17, 2014.
`
`SELECTIVE ABLATION OF AML USING ANTIBODY-TARGETED CHEMOTHERAPY
`
`3681
`
`Table 3. Clinical Responses and Recovery of Hematologic Function in Patients Who Received Treatment With CMA-676
`
`Time in Days From Last Dose to:
`
`Clinical
`Response
`
`Patient
`No.
`
`Age
`(yr)
`
`Cytogenetic
`Abnormality
`
`Dose
`Level
`(mg/m2)
`
`Peripheral
`Blasts
`(per µL)
`
`Percentage
`of Marrow
`Blasts
`
`ANC
`.1,500/
`µL
`
`Platelet
`Count
`.100 3 103/µL
`
`50
`83
`
`60
`
`88
`67
`8
`
`40
`23
`95
`
`12
`22,377
`
`748
`
`1,912
`3,560
`0
`
`2,445
`0
`300
`
`1
`4
`
`9
`
`5 6 9 9 9 6
`
`CR*
`
`FH-007
`FH-012†
`
`43 t(6;11)
`29 t(6;9)
`
`FH-023†
`
`34 None
`
`Elimination of
`blast cells from
`peripheral
`blood and
`marrow‡
`
`FH-013
`FH-019§
`FH-022
`
`FH-024
`COH-015§
`FH-007\
`
`74
`53
`65
`
`52
`34
`43
`
`None
`t(8;21)
`None
`
`t(1;7); t(12;17)
`Trisomy 11; del(15)
`t(6;11)
`
`4
`20
`
`42
`
`7
`35
`
`160
`
`Relapse
`
`132
`188
`
`.623
`
`Not attained
`Not attained
`18
`
`Not attained
`Not attained
`Not attained
`
`70
`Not applicable
`40
`
`20
`Not attained
`8
`
`Not evaluable¶
`Not attained
`Not attained
`
`14
`Not applicable
`56
`
`Site of
`Recurrence
`
`Bone; marrow
`Testes; central
`nervous system
`None
`
`Marrow
`Not applicable
`Marrow; central
`nervous system
`Marrow
`Not applicable
`Marrow
`
`*Patient achieved transfusion independence with ANC .1,500/µL, platelet count .100 3 103/µL, and ,5% blasts in the bone marrow aspirate.
`†Patient received donor lymphocyte infusion after attaining CR (FH-012) or before achieving platelet count .100 3 103/µL (FH-023).
`‡Patient had ,5% blasts in the bone marrow aspirate with various degrees of hematopoietic recovery.
`§Patients died of infection 12 (FH-019) and 50 (COH-015) days after receiving a third dose of CMA-676.
`\Patient attained CR after receiving CMA-676 at 1 mg/m2 and then was retreated with CMA-676 at a dose of 6 mg/m2.
`¶Patient received a bone marrow transplant shortly after leukemia disappeared from the bone marrow and eventually recovered a normal
`platelet count.
`
`doses of CMA-676. Reduced numbers of peripheral blast cells
`were observed in 10 of 11 patients treated with either 6 or 9
`mg/m2 of CMA-676 compared with 12 of 20 patients treated at
`dose levels from 0.25 to 5 mg/m2.
`Morphologic evidence of leukemia within the bone marrow
`(.5% blast cells) was seen in all patients before treatment.
`After treatment with one to three doses of the drug conjugate, 8
`(20%) of 40 patients had ,5% leukemic blast cells on
`morphologic examination of bone marrow aspirate and biopsy
`specimens (Table 3). In one of these patients (FH-007), blast
`cells were also eliminated from the bone marrow on a second
`occasion after retreatment with CMA-676 (see below).
`Recovery of normal blood counts. Recovery of blood
`neutrophil counts to greater than 1,500 cells/µL was observed in
`five of eight patients who had ,5% leukemic blasts by
`morphologic examination of bone marrow aspirate and biopsy
`specimens (Table 3). In addition, three of these five patients
`achieved normal platelet counts. The first patient, FH-007,
`achieved a complete hematologic and cytogenetic remission 7
`days after receiving the third dose of CMA-676 at 1 mg/m2.
`After 140 days, local leukemia recurrences in his femurs and
`iliac wing were identified by magnetic resonance imaging and
`confirmed by surgical biopsy. He was treated with conventional
`chemotherapy and local radiation and achieved another CR. He
`experienced relapse again 293 days after completing the first
`course of CMA-676, was treated with two additional doses of
`CMA-676 at 6 mg/m2, and his leukemia disappeared again for
`56 days. His leukemic cells expressed CD33 at the time of each
`of his relapses.
`The second patient, FH-012, achieved a complete hemato-
`logic and cytogenetic remission 35 days after the third dose of 4
`mg/m2. In an attempt to consolidate his remission, he was given
`an infusion of lymphocytes from his bone marrow donor 40
`days after infusion of the third dose of CMA-676. He remained
`
`in CR for 214 days after treatment until testicular and central
`nervous system relapses occurred.
`The third patient, FH-023, received three doses of CMA-676
`at 9 mg/m2, and his ANC recovered to .1,500/µL 42 days after
`he received the third dose of CMA-676. His platelet count
`reached a peak of 36 3 103/µL 23 days after he received the
`third dose of CMA-676, but the patient subsequently became
`platelet
`transfusion-dependent for approximately 3 weeks.
`Evidence of antibody on the patient’s platelet surfaces was
`documented. Megakaryocytes were present and platelet matura-
`tion appeared normal on examination of the marrow aspirate. In
`an attempt
`to consolidate his remission, he was given an
`infusion of lymphocytes from his bone marrow donor 54 days
`after receiving the third dose, and his platelet count eventually
`rose to .100 3 103/µL 106 days later. At the time of this report,
`he remained in continuous cytogenetic and hematologic remis-
`sion .623 days after receiving the third dose of CMA-676.
`Pharmacokinetics and saturation of peripheral CD33 sites.
`Detectable plasma levels of CMA-676 were identified in all
`patients immediately after intravenous administration and the
`half-life of CMA-676 was estimated to be 38 6 21 hours for
`patients receiving the 9 mg/m2 dose. Figure 2 shows the
`relationship between the total number of cell surface CD33 sites
`and CD33 sites bound with CMA-676 in peripheral blood cells
`for a characteristic patient over time. Thirty minutes after
`infusion, CD33 sites were almost completely saturated with
`CMA-676. Twenty-four hours after administration, fewer CD33
`sites and bound drug conjugate were evident, suggesting
`internalization of the CD33-immunoconjugate complex. In the
`patients who received the maximum dose (9 mg/m2), a median
`of 92.2% (range, 79.2% to 100%) of CD33 binding sites on
`peripheral blood blast-sized cells were bound by the drug
`conjugate 30 minutes after infusion of a dose. Peak CD33
`saturation levels in relation to treatment response are shown in
`
`IMMUNOGEN 2187, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`From
`
`by guest
`www.bloodjournal.org
`
`
`
`
`For personal use only.on November 17, 2014.
`
`3682
`
`SIEVERS ET AL
`
`relationship between measured leukemic blast cell drug efflux
`levels and saturation of CD33 sites with response. Elimination
`of leukemia appeared to be correlated with a low capacity by
`leukemic blast cells to extrude DiOC2. For example, of the 30
`patients evaluated with the assay in whom doses of CMA-676
`saturated .75% of available CD33 sites on peripheral blood
`blast cells, 8 of 17 patients with leukemic blast cells that showed
`#40 channel numbers of DiOC2 efflux had ,5% blasts in the
`bone marrow after treatment. In contrast, none of the 13 patients
`with leukemic blast cells expressing .40 channel numbers of
`DiOC2 efflux entered remission.
`Immune responses. No humoral responses to anti-hP67.6
`antibody were detected. A humoral response to the calicheamicin-
`linker complex was documented in one patient after receiving a
`third dose of CMA-676 and in a second patient during
`retreatment with CMA-676.
`
`DISCUSSION
`
`This study describes the use of an antibody-drug conjugate
`capable of targeting and safely eliminating target leukemic cells
`in vivo. Disappearance of AML cells from the bone marrow and
`peripheral blood of eight patients with complete restoration of
`normal hematopoiesis in three patients was observed after
`treatment with antibody-targeted chemotherapy. These results
`show that eradication of CD331 AML cells can allow restora-
`tion of normal hematopoiesis by remaining CD332 precursors.
`For some patients, we hypothesize that the CD332 precursors
`are predominantly or completely nonmalignant. This is based
`on findings that, in some cases of AML, the clonal abnormality
`originated in either a committed progenitor or an early multipo-
`tent cell whose proliferative expression is mainly restricted to
`the granulocyte/monocyte lineage.12 Because selection of CD332
`precursors from some of these leukemias allowed normal
`hematopoietic growth in culture,
`the malignant clone may
`involve few, if any, CD332 precursors.5,6
`In other cases of AML, particularly in older patients, the
`clonal abnormality has been found in both the erythroid and
`myeloid lineages, showing malignant involvement of multipo-
`tent precursors.12 In addition, clonal karyotypic abnormalities
`have been found in primitive precursor cells from some AML
`patients.13,14 It is conceivable that normal hematopoiesis could
`also be restored in these patients even if a substantial portion of
`CD332 precursors were malignant because normal precursors
`express at least a short-term proliferative advantage. While this
`explanation appears inconsistent with the observed leukemic
`growth in a immunodeficient mouse model after infusion of
`isolated primitive (CD341 CD382) precursors from human
`leukemia specimens,15 it is consistent with the report that a
`patient undergoing allogeneic transplantation who was inadver-
`tently given an infusion of donor AML cells initially recovered
`with normal donor hematopoiesis.16
`Each patient whose blood counts returned completely to
`normal had experienced relapse after an allogeneic bone
`marrow transplant (data not shown). Therefore, it is possible
`that a graft-versus-leukemia effect eliminated the remaining
`CD332 leukemic cells after elimination of the bulk of AML
`cells. However,
`in a recently initiated phase II study of
`CMA-676, normal hematopoiesis was restored in some patients
`
`Fig 2. Relationship between total number and CMA-676–bound
`CD33 sites on peripheral blood cells in a characteristic patient over
`time. The solid line represents the number of CD33 sites available for
`binding to CMA-676 as estimated by the maximal fluorescence
`intensity obtained by incubating an aliquot of cells in vitro with
`excess CMA-676. The dashed line represents the fluorescence inten-
`sity of bound CMA-676 to cell surfaces. Near-complete saturation is
`seen 30 minutes after the start of the infusion.
`
`Fig 3. These results show that saturation of sites is not, by itself,
`sufficient to insure treatment response.
`Response correlation with drug efflux. We evaluated the
`efflux of DiOC2 from pretreatment leukemic blast cells as a
`measure of functional drug resistance. Figure 3 shows the
`
`Fig 3. Relation of leukemic blast cell dye efflux and maximum
`CMA-676 saturation of CD33 sites on peripheral blood blast-sized
`cells with treatment response (N 5 36). (m) Denotes patients who had
`F5% leukemic blasts by morphologic examination of bone marrow
`aspirate and biopsy specimens after treatment. (d) Denotes patients
`whose leukemia did not disappear. Peripheral blood samples from
`four patients were unavailable for analysis. The efflux from the
`dominant population is represented in the five instances in which
`efflux profiles were bimodal. *Leukemic cell specimens from patients
`FH-023 and FH-024 were 2 and 3 days old, respectively, at the time of
`efflux measurement. Because blast-sized cells from each showed
`uncharacteristically low DiOC2 loading, obtained efflux values may
`underestimate true efflux.
`
`IMMUNOGEN 2187, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`From
`
`by guest
`www.bloodjournal.org
`
`
`
`
`For personal use only.on November 17, 2014.
`
`SELECTIVE ABLATION OF AML USING ANTIBODY-TARGETED CHEMOTHERAPY
`
`3683
`
`who had not received a transplant.17 Molecular remissions have
`been observed in a portion of patients with acute promyelocytic
`leukemia who received unconjugated anti-CD33 antibody
`(HuM195).18 Although an AML patient with 8% blasts in his
`bone marrow achieved a CR with HuM195 given at a supersatu-
`rating dose, complete clinical responses have not been reported
`in patients with large tumor burdens.19 Hence, the observed
`elimination of leukemia after treatment with CMA-676 was not
`likely due to antibody-mediated effects alone.
`Myelosuppression was the most clinically significant adverse
`event. The severe marrow hypoplasia observed in two patients
`who received 9 mg/m2 is consistent with depletion of CD331
`hematopoietic progenitor cells and with the time required for
`CD332 cells to restore hematopoietic function.20 To prevent
`prolo

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