throbber
V O L U M E 3 1 䡠 N U M B E R 3 3 䡠 N O V E M B E R 2 0 2 0 1 3
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`O R I G I N A L R E P O R T
`
`Author affiliations appear at the end of
`this article.
`
`Published online ahead of print at
`www.jco.org on October 14, 2013.
`
`P.A. was supported by an American
`Society of Hematology (ASH) Scholar
`Award and by an American Society of
`Clinical Oncology/Conquer Cancer Foun-
`dation Career Development Award.
`D.A.R. was supported by an ASH
`Scholar Award.
`
`P.A. and A.N. contributed equally to
`this work.
`
`Authors’ disclosures of potential con-
`flicts of interest and author contribu-
`tions are found at the end of this
`article.
`
`Clinical trial information: NCT00532259.
`
`Corresponding author: Leo I. Gordon,
`MD, Northwestern University Feinberg
`School of Medicine and the Robert H.
`Lurie Comprehensive Cancer Center,
`676 North St Clair Street, Suite 850,
`Chicago, IL 60611-3008; e-mail:
`l-gordon@northwestern.edu.
`
`© 2013 by American Society of Clinical
`Oncology
`
`0732-183X/13/3133w-4199w/$20.00
`
`DOI: 10.1200/JCO.2012.48.3685
`
`Disabling Immune Tolerance by Programmed Death-1
`Blockade With Pidilizumab After Autologous
`Hematopoietic Stem-Cell Transplantation for Diffuse Large
`B-Cell Lymphoma: Results of an International Phase II Trial
`
`Philippe Armand, Arnon Nagler, Edie A. Weller, Steven M. Devine, David E. Avigan, Yi-Bin Chen,
`Mark S. Kaminski, H. Kent Holland, Jane N. Winter, James R. Mason, Joseph W. Fay, David A. Rizzieri,
`Chitra M. Hosing, Edward D. Ball, Joseph P. Uberti, Hillard M. Lazarus, Markus Y. Mapara,
`Stephanie A. Gregory, John M. Timmerman, David Andorsky, Reuven Or, Edmund K. Waller,
`Rinat Rotem-Yehudar, and Leo I. Gordon
`See accompanying article on page 4268
`
`A
`
`B
`
`S
`
`T
`
`R
`
`A
`
`C
`
`T
`
`Purpose
`The Programmed Death-1 (PD-1) immune checkpoint pathway may be usurped by tumors,
`including diffuse large B-cell lymphoma (DLBCL), to evade immune surveillance. The reconstitut-
`ing immune landscape after autologous hematopoietic stem-cell transplantation (AHSCT) may be
`particularly favorable for breaking immune tolerance through PD-1 blockade.
`Patients and Methods
`We conducted an international phase II study of pidilizumab, an anti–PD-1 monoclonal antibody, in
`patients with DLBCL undergoing AHSCT, with correlative studies of lymphocyte subsets. Patients
`received three doses of pidilizumab beginning 1 to 3 months after AHSCT.
`Results
`Sixty-six eligible patients were treated. Toxicity was mild. At 16 months after the first treatment,
`progression-free survival (PFS) was 0.72 (90% CI, 0.60 to 0.82), meeting the primary end point.
`Among the 24 high-risk patients who remained positive on positron emission tomography after
`salvage chemotherapy, the 16-month PFS was 0.70 (90% CI, 0.51 to 0.82). Among the 35 patients
`with measurable disease after AHSCT, the overall response rate after pidilizumab treatment was
`51%. Treatment was associated with increases in circulating lymphocyte subsets including
`PD-L1E–bearing lymphocytes, suggesting an on-target in vivo effect of pidilizumab.
`Conclusion
`This is the first demonstration of clinical activity of PD-1 blockade in DLBCL. Given these results,
`PD-1 blockade after AHSCT using pidilizumab may represent a promising therapeutic strategy in
`this disease.
`
`J Clin Oncol 31:4199-4206. © 2013 by American Society of Clinical Oncology
`
`INTRODUCTION
`
`PD-1 (Programmed Death-1) is a member of the B7
`receptor family. Together with its ligands (PD-L1
`and PD-L2), it functions as an important check-
`point in the regulation of immune responses.1
`Those ligands are upregulated by the inflammatory
`environment and inhibit the function of PD-1–
`bearing lymphocytes. Thus the PD-1 immune
`checkpoint pathway serves to dampen peripheral
`lymphocyte activity in the context of inflammatory
`responses. This pathway seems to be co-opted by
`many tumors, preventing effective antitumor im-
`
`munity, and therefore represents a promising ther-
`apeutic target, as demonstrated in several solid
`tumor subtypes.2-5 Pidilizumab (CureTech, Yavne,
`Israel) is an anti–PD-1 humanized immunoglobulin
`G1 monoclonal antibody with preclinical antitumor
`activity in animal models.6-8 In a phase I trial in
`patients with advanced hematologic malignancies,
`pidilizumab showed a favorable safety profile and
`early evidence of clinical activity.9
`We conducted an international phase II study
`of pidilizumab in patients with diffuse large B-cell
`lymphoma (DLBCL) and primary mediastinal
`large B-cell lymphoma (PMBCL) after autologous
`
`© 2013 by American Society of Clinical Oncology
`Downloaded from jco.ascopubs.org on August 31, 2015. For personal use only. No other uses without permission.
`Copyright © 2013 American Society of Clinical Oncology. All rights reserved.
`
`4199
`
`1 of 8
`
`BI Exhibit 1012
`
`

`

`Armand et al
`
`Table 1. Baseline Patient Characteristics (eligible patients)
`
`Table 1. Baseline Patient Characteristics (eligible patients) (continued)
`
`Variable
`
`Total No.ⴱ
`
`%
`
`Variable
`
`Total No.ⴱ
`
`Response to salvage therapy by PET
`Negative
`Positive
`PET not done
`Status before pidilizumab treatment#
`By CT imaging
`CR
`Not in CR
`By PET imaging
`Negative
`Positive
`PET not done/missing
`
`31
`24
`11
`
`31
`35
`
`45
`9
`12
`
`%
`
`47
`36
`17
`
`47
`53
`
`68
`18
`14
`
`57
`19-80
`
`25
`8-186
`
`No. of patients
`Age, years
`Median
`Range
`Race
`Asian
`Black
`White
`Hispanic
`Country
`Chile
`India
`Israel
`United States
`Sex
`Male
`Female
`Disease
`De novo DLBCL
`PMBCL
`Transformed indolent B-NHL
`IPI score at diagnosis
`0-1
`2
`3
`4-5
`Unknown
`Response to first-line therapy
`Complete remission
`Partial remission
`Stable or progressive disease
`Unknown
`Time from diagnosis to AHSCT, months
`Median
`Range
`Characteristics at relapse
`Stage†
`I
`II
`III
`IV
`Bulky‡
`Extranodal involvement§
`Marrow involvement储
`IPI¶
`0-1
`2
`3
`4-5
`No. of prior treatments
`1
`2
`3
`4
`Rituximab use
`With first-line therapy
`With salvage therapy
`With conditioning
`Radiation after transplantation
`(continued in next column)
`
`66
`
`7
`3
`52
`4
`
`1
`3
`7
`55
`
`43
`23
`
`49
`4
`13
`
`15
`11
`7
`7
`26
`
`45
`15
`5
`1
`
`6
`8
`9
`18
`18
`18
`29
`
`13
`7
`8
`2
`
`3
`47
`13
`3
`
`56
`54
`10
`5
`
`11
`5
`79
`6
`
`1
`4
`11
`83
`
`65
`35
`
`74
`6
`20
`
`23
`17
`11
`11
`39
`
`68
`23
`8
`1
`
`9
`12
`14
`27
`27
`27
`44
`
`20
`11
`12
`3
`
`5
`71
`20
`5
`
`85
`82
`15
`8
`
`Abbreviations: AHSCT, autologous hematopoietic stem-cell transplantation;
`B-NHL, B-cell non-Hodgkin lymphoma; CR, complete remission; CT, com-
`puted tomography; DLBCL, diffuse large B-cell lymphoma; IPI, International
`Prognostic Index; PET, positron emission tomography; PMBCL, primary
`mediastinal B-cell lymphoma; PR, partial remission; SD, stable disease.
`ⴱPercentages may not add to 100 because of rounding. Denominator used
`was all patients, including those with missing data.
`†Data missing on 25 patients.
`‡Data missing on nine patients.
`§Data missing on 10 patients.
`储Marrow biopsy was not performed on three patients; results were deter-
`mined at time of diagnosis for primary refractory patients (SD⫹PD) or at last
`relapse before transplant for the others.
`¶Data missing on 36 patients.
`#CT was required per protocol and used for eligibility determination; PET
`was obtained at the discretion of the treating clinician and not used for
`eligibility determination.
`
`hematopoietic stem-cell transplantation (AHSCT). PD-L1 is ex-
`pressed on suppressor immune cells in the tumor microenvironment
`and in at least a subset of DLBCL and PMBCL tumors,10-13 where it
`may alter the composition and function of tumor-infiltrating lym-
`phocytes,14 and therefore represents a valid therapeutic target.11,12
`Moreover, the post-AHSCT setting may be a particularly fertile con-
`text for PD-1 blockade. This is a state of low-volume residual disease,
`during which there is a remodeling of the immune system. Indeed, the
`majority of the circulating leukocytes in the first few months after
`AHSCT are natural killer cells, CD45RO⫹ memory/effector cells, and
`monocytes, which comprise pidilizumab’s target populations and
`whose presence in DLBCL tumors has been associated with a favorable
`prognosis.15-17 Therefore, PD-1 blockade early after AHSCT for pa-
`tients with DLBCL may prevent a tumor-dependent, PD-1 driven
`exhaustion of antitumor lymphocytes, leading to eradication of resid-
`ual disease and improvement in progression-free survival (PFS).
`
`PATIENTS AND METHODS
`
`Patients
`Patients 18 years and older could be consented for this study if they
`planned or had undergone AHSCT for DLBCL, PMBCL, or transformed
`indolent B-cell non-Hodgkin lymphoma. Only patients with chemotherapy-
`sensitive disease (at least partial remission18 after salvage therapy by computed
`tomography [CT] scans) were eligible. Confirmatory screening was performed
`between 30 and 90 days after AHSCT. To enroll onto the study and receive
`treatment, patients had to have CT scans before first drug administration
`showing no evidence of progressive disease (PD) from pretransplant assess-
`ment, as well as normal hematologic, renal, hepatic, and cardiac function.
`Patients with type 1 diabetes, immune deficiency, active autoimmune disease,
`
`4200
`
`JOURNAL OF CLINICAL ONCOLOGY
`© 2013 by American Society of Clinical Oncology
`Downloaded from jco.ascopubs.org on August 31, 2015. For personal use only. No other uses without permission.
`Copyright © 2013 American Society of Clinical Oncology. All rights reserved.
`
`2 of 8
`
`BI Exhibit 1012
`
`

`

`PD-1 Blockade for DLBCL After AHSCT
`
`Table 2. Adverse Events
`
`Severity Grade
`
`All Grades
`
`1
`
`2
`
`3
`
`4
`
`5
`
`Event
`
`No. of
`Patients
`
`No. of
`Events
`
`No. of
`Patients
`
`No. of
`Events
`
`No. of
`Patients
`
`No. of
`Events
`
`No. of
`Patients
`
`No. of
`Events
`
`No. of
`Patients
`
`No. of
`Events
`
`No. of
`Patients
`
`No. of
`Events
`
`69
`19
`18
`14
`12
`12
`10
`9
`9
`
`613
`25
`21
`15
`19
`14
`15
`12
`12
`
`65
`3
`16
`9
`10
`11
`5
`8
`6
`
`392
`3
`19
`10
`14
`13
`6
`10
`8
`
`49
`7
`2
`5
`4
`1
`—
`2
`3
`
`145
`7
`2
`5
`5
`1
`
`2
`3
`
`Any AE
`Neutropenia
`Fatigue
`Upper respiratory tract infection
`Diarrhea
`Cough
`Thrombocytopenia
`Hyperglycemia
`Leukopenia
`Anemia
`Pyrexia
`Renal failure
`Vomiting
`Lymphopenia
`Cardiac arrest
`Duodenal ulcer
`GI hemorrhage
`General physical health decline
`Pain
`Clostridium difficile colitis
`Herpes zoster
`Lobar pneumonia
`Urinary tract infection
`Vascular injury
`Accident
`Fall
`Pelvic fracture
`Head injury
`Facial bone fracture
`aPTT prolonged
`Hypophosphatemia
`Bone pain
`Myositis
`Rhabdomyolysis
`Myelodysplastic syndrome
`Glioma
`Intracranial hemorrhage
`Subarachnoid hemorrhage
`Headache
`Tachypnea
`COPD
`ARDS
`Pneumothorax
`Hyperhidrosis
`Cholecystectomy
`DVT
`Hypertension
`Hypotension
`
`1
`
`1ⴱ
`—
`—
`—
`—
`—
`—
`—
`—
`
`1
`
`1
`
`9
`5
`—
`—
`—
`—
`2
`—
`—
`
`1
`
`1
`
`1†
`
`1
`1
`
`1
`
`14
`5
`
`4
`
`1
`
`1
`
`1
`
`1
`1
`
`1
`
`30
`9
`—
`—
`—
`—
`4
`—
`1
`3
`2
`2
`1
`1
`1
`1
`1
`1
`1
`1
`1
`1
`1
`1
`
`1
`1
`
`1
`1
`1
`1
`1
`1
`
`1
`
`1
`1
`1
`1
`
`1
`1
`1
`1
`1
`
`60
`10
`
`5
`
`1
`3
`2
`2
`2
`1
`1
`1
`1
`1
`1
`1
`1
`1
`1
`1
`
`1
`1
`
`1
`1
`1
`1
`1
`1
`
`1
`
`1
`1
`1
`1
`
`1
`1
`1
`1
`1
`
`NOTE. Data are shown as number of patients with a given AE and number of events. Only AEs representing ⱖ 2% of total events are shown for grade 1 and
`2 events.
`Abbreviations: AE, adverse event; aPTT, activated partial thromboplastin time; ARDS, acute respiratory distress syndrome; COPD, chronic obstructive pulmonary
`disorder; DVT; deep venous thrombosis.
`ⴱOne patient developed fatal disseminated zoster infection during the follow-up period.
`†One patient with pre-existing leukopenia and thrombocytopenia developed myelodysplasia 13 months after the last dose of pidilizumab. This was considered
`unrelated to study drug.
`
`www.jco.org
`
`© 2013 by American Society of Clinical Oncology
`Downloaded from jco.ascopubs.org on August 31, 2015. For personal use only. No other uses without permission.
`Copyright © 2013 American Society of Clinical Oncology. All rights reserved.
`
`4201
`
`3 of 8
`
`BI Exhibit 1012
`
`

`

`Armand et al
`
`CNS involvement by lymphoma, active infection, other serious illness, con-
`current investigational treatment, or performance status more than 1 were
`excluded, as were pregnant or nursing patients.
`Patients were recruited at 30 centers in the United States, Israel, Chile,
`and India. All patients provided written informed consent. The study was
`approved by the offices for human research studies at the participating insti-
`tution and conducted in accordance with the principles of the Declaration of
`Helsinki. The study was supported by CureTech, and the data were analyzed by
`three of the authors (P.A., E.A.W., and L.I.G.) and by CureTech.
`Treatment and Monitoring
`Patients received treatment with pidilizumab administered intrave-
`nously at a dose of 1.5 mg/kg every 42 days for three cycles, beginning 30 to 90
`days from AHSCT. Premedication consisted of acetaminophen or ibuprofen,
`as well as diphenhydramine or promethazine. Patients were restaged with CT
`scans (with or without positron emission tomography [PET] scans, at the
`discretion of the treating clinicians) at confirmatory screening, then before the
`second and third cycles, and at 30, 44, and 69 weeks from the first day of
`treatment. Treatment was stopped if there was evidence of PD based on
`standard criteria.18 Patients were observed until 16 months from first pidili-
`zumab treatment, which corresponded to approximately 18 months from
`AHSCT. For patients with measurable disease at post-AHSCT screening, re-
`sponse to pidilizumab treatment was assessed18 according to the restaging
`schedule described previously, using the post-AHSCT measurements as the
`pretreatment baseline. Toxicity was graded using National Cancer Institute
`Common Terminology Criteria of Adverse Events v3.0.
`Correlative Studies
`Blood samples collected from all treatment sites from patients treated at
`least once with pidilizumab were analyzed by flow cytometry at two central
`laboratories of Esoterix LabCorp Services (Austin, TX) using study-specific
`validated methodologies. Forty-one prospectively specified leukocyte subsets
`based on cluster of differentiation marker expression were evaluated for abso-
`lute (per microliter) and relative numbers, as well as molecules of equivalent
`soluble fluorochrome (MESF). Validation studies for marker stability and
`inter- and intra-assay precision were conducted before initiating the tests in
`this study.
`Statistical Considerations
`The primary end point of this study was the 16-month progression-free
`proportion from the time of first pidilizumab administration among all eligi-
`ble patients who received at least one dose of pidilizumab. Secondary end
`points included safety and toxicity, PFS, and overall survival (OS); immuno-
`genicity of pidilizumab; and immune subset analyses. OS was defined as the
`time from first treatment to death, and PFS as the time to death, relapse, or
`
`progression. OS and PFS were calculated using the Kaplan-Meier method. On
`the basis of data available at the time of study design, the 18-month PFS after
`transplantation for chemosensitive patients was estimated to be approximately
`60% to 65%.19,20 This time corresponds to approximately 16 months from the
`planned start of pidilizumab on this trial. Because the number of patients
`enrolled was anticipated to range from 64 to 80 patients, an observed 16-
`month PFS from start of pidilizumab of at least 69% was considered to warrant
`further study. This design had at least 87% probability of concluding the
`treatment promising if the true 16-month PFS was 75% and less than 10%
`probability if the true 16-month PFS was 60% (given the exact binomial
`distribution applied to all possible sample sizes between 64 and 80 patients).
`Patients were eligible if they met all eligibility criteria and received at least one
`dose of pidilizumab.
`For exploratory measurements of changes in immune subsets, we com-
`pared absolute numbers of prespecified circulating lymphocytes of a given
`immunophenotype before the first treatment and at 24 hours, 6, 12, and 16
`weeks afterwards. We also measured MESF to assess for change in surface
`expression of selected markers. Pre- and post-treatment values were compared
`using paired Wilcoxon signed rank testing for individual time points, adjusted
`for multiple comparisons, as well as repeated measures analysis using a log10
`transformation (with SAS proc mixed). All P values are two-tailed, using a
`threshold for statistical significance of .05 except as noted. The data were
`analyzed using SAS version 9.2 (SAS Institute, Cary, NC).
`
`RESULTS
`
`Patients
`Patients could sign consent before or after AHSCT, but were
`required to pass confirmatory screening to be enrolled onto the study
`and treated. Among the 97 patients who gave consent, 25 were screen
`failures at confirmatory screening (including six with PD, five on
`concurrent disallowed treatment, four who withdrew consent, two
`with CNS disease, and two with infection). Therefore, 72 patients
`received at least one dose of pidilizumab (treated subset) at a median
`of 2.6 months after AHSCT (range, 1.1 to 4.1 months). Sixty patients
`(83%) completed all three cycles. Ten patients withdrew from the
`study before the 16-month follow-up visit for reasons other than
`death or progression: loss to follow-up (n ⫽ 2), investigator decision
`(n ⫽ 3), withdrawal of consent (n ⫽ 3), protocol violation (n ⫽ 1),
`and adverse event (AE; n ⫽ 1). On final review, six patients were
`
`OS
`PFS
`
`4
`8
`12
`Time Since First Treatment (months)
`
`16
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`Survival (%)
`
`B
`
`OS
`PFS
`
`4
`8
`12
`Time Since First Treatment (months)
`
`16
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`A
`
`Survival (%)
`
`Fig 1. Progression-free survival (PFS) and overall survival (OS) after pidilizumab treatment. (A) PFS and OS of all eligible patients. (B) PFS and OS of the 24 eligible
`patients who remained positive on positron emission tomography after salvage therapy.
`
`4202
`
`JOURNAL OF CLINICAL ONCOLOGY
`© 2013 by American Society of Clinical Oncology
`Downloaded from jco.ascopubs.org on August 31, 2015. For personal use only. No other uses without permission.
`Copyright © 2013 American Society of Clinical Oncology. All rights reserved.
`
`4 of 8
`
`BI Exhibit 1012
`
`

`

`PD-1 Blockade for DLBCL After AHSCT
`
`Total CD14+
`CD14+ PD-L1+
`CD14+ PD-L2+
`Total CD4+ CD25+
`CD4+ CD25+ PD-L1+
`
`24 hours
`
`6 weeks
`
`12 weeks
`Time
`
`16 weeks
`
`CD4+ CD62L+ CD127+
`CD4+ CD62L- CD127+
`CD8+ CD62L+ CD127+
`CD8+ CD62L- CD127+
`
`24 hours
`
`6 weeks
`
`12 weeks
`Time
`
`16 weeks
`
`PD-L1 on CD14+
`PD-L2 on CD14+
`PD-L1 on CD4+ CD25+
`CD127 on CD4+ CD62L+
`CD127 on CD4+ CD62L-
`CD127 on CD8+ CD62L+
`CD127 on CD8+ CD62L-
`
`24 hours
`
`6 weeks
`
`12 weeks
`Time
`
`16 weeks
`
`200
`180
`160
`140
`120
`100
`80
`60
`40
`20
`0
`
`350
`
`300
`
`250
`
`200
`
`150
`
`100
`
`50
`
`0
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`-20
`
`A
`
`Mean Change From Baseline
`
`No. of Cells/μL (%)
`
`B
`
`Mean Change From Baseline
`
`No. of Cells/μL (%)
`
`C
`
`Mean Change From Baseline
`
`No. of Cells/μL (%)
`
`Fig 2. Changes in absolute number of circulating lymphocyte subsets and
`surface marker expression after pidilizumab administration. (A) Changes in
`circulating number of PD-L1 (B7-H1) and PD-L2 (B7-DC) –positive monocytes
`and T cells.
`(B) Changes in circulating number of peripheral and central
`memory CD8 T cells. (C) Changes in expression of selected surface markers
`on monocytes and T cells.
`
`pidilizumab resulted in a significant increase in the absolute number
`of PD-L1–bearing activated helper T cells (CD4⫹ CD25⫹ PD-L1⫹),
`apparent 24 hours after first treatment and sustained until at least 16
`weeks (Fig 2A). There also seemed to be some changes in PD-1 ligand-
`bearing monocytes (CD14⫹ PD-L1⫹ and CD14⫹ PD-L2⫹ cells). For
`the latter subsets, the mean percentage increase was larger than the
`
`determined ineligible (four because of refractoriness to salvage ther-
`apy, one for PD on the first day of treatment, and the other for
`disallowed concomitant treatments). The baseline characteristics of
`the 66 eligible patients are shown in Table 1. At the time of post-
`AHSCT restaging, 47% of patients were in complete remission (CR)
`by CT. Fifty-five patients had a PET scan after salvage; 31 (47%) were
`in PET-CR. Fifty-four patients had a post-AHSCT PET scan; 45 (68%)
`were in PET-CR at that time.
`
`Safety and Toxicity
`Among all 72 treated patients, a total of 613 AEs occurred in 69
`(96%) of patients (Table 2), among which 135 were considered related
`to treatment. The most frequently reported grade 3 to 4 AEs were
`neutropenia (19% of patients) and thrombocytopenia (8%). All pa-
`tients with grade 4 neutropenia responded to growth factor treatment
`and remained asymptomatic. One patient died of disseminated herpes
`zoster 10 months after the third dose of pidilizumab, which was
`considered unrelated to study treatment. Twenty-three patients
`(32%) experienced at least one serious AE, and three patients (4%)
`experienced a related serious AE. There was no evidence of significant
`autoimmune toxicity, no infusion reactions, and no treatment-
`related mortality.
`
`Clinical Outcome
`Among the 66 eligible patients, 18 experienced disease progres-
`sion or died before the 16-month time point. The 16-month PFS from
`first treatment (the primary end point) was 0.72 (90% CI, 0.60 to 0.82;
`Fig 1A). The study therefore met its primary end point. Nine patients
`died during the study period between 2.3 and 15.3 months; the cause
`of death was lymphoma in eight patients and disseminated herpes
`zoster in one patient. The 16-month OS for eligible patients was 0.85
`(90% CI, 0.74 to 0.92; Fig 1A). Among the 24 patients who remained
`PET-positive at the conclusion of salvage therapy, 16-month PFS was
`0.70 (90% CI, 0.51 to 0.82; Fig 1B). Among the 31 PET-negative
`patients, 16-month PFS was 0.72 (90% CI, 0.56 to 0.84); among the 11
`patients who did not have a postsalvage PET scan, 16-month PFS was
`0.72 (90% CI, 0.42 to 0.88). No significant difference was detected in
`the PFS or OS between patients when stratified by disease status
`assessed by CT scans after AHSCT, age, time to first relapse, time from
`diagnosis to AHSCT, or salvage regimen; however, there was limited
`power for those comparisons. We also performed an intent-to-treat
`analysis for the 72 treated patients. The PFS at 16 months from first
`pidilizumab treatment in this cohort was 0.68 (90% CI, 0.59 to 0.77),
`and OS was 0.84 (90% CI, 0.77 to 0.91).
`Among the 35 eligible patients with measurable disease at
`screening post-AHSCT and before the first dose of pidilizumab, 12
`(34%) achieved a CR by CT criteria after pidilizumab treatment,
`and six (17%) achieved a partial remission (PR), for an overall
`response rate of 51%. In addition, 13 patients (37%) had stable
`disease, whereas four (11%) had PD. The median time to docu-
`mented response was 30 weeks (range, 6 to 69 weeks). Among the
`nine patients who had residual disease after AHSCT and a positive
`PET scan, the overall response rate was 33%, and an additional
`44% had stable disease.
`
`Immune Subset Analyses
`Figure 2 and Table 3 detail changes in selected lymphocyte sub-
`sets and marker expression among eligible patients. Treatment with
`
`www.jco.org
`
`© 2013 by American Society of Clinical Oncology
`Downloaded from jco.ascopubs.org on August 31, 2015. For personal use only. No other uses without permission.
`Copyright © 2013 American Society of Clinical Oncology. All rights reserved.
`
`4203
`
`5 of 8
`
`BI Exhibit 1012
`
`

`

`Armand et al
`
`Table 3. Changes in Selected Lymphocyte Subsets and Marker Expression
`
`24 Hours
`
`6 Weeks
`
`12 Weeks
`
`16 Weeks
`
`Variable
`
`Adjusted
`P
`
`Mean
`Change
`
`Median
`Change
`
`P
`
`Mean
`Change
`
`Median
`Change
`
`P
`
`Mean
`Change
`
`Median
`Change
`
`P
`
`Mean
`Change
`
`Median
`Change
`
`P
`
`.2
`.3
`.3
`< .0001
`.0004
`.0025
`.07
`.003
`.0002
`
`Lymphocyte population
`CD14⫹
`CD14⫹ PD-L1⫹
`CD14⫹ PD-L2⫹
`CD4⫹CD25⫹
`CD4⫹CD25⫹ PD-L1⫹
`CD4⫹CD62L⫹ CD127⫹
`CD4⫹CD62L– CD127⫹
`CD8⫹CD62L⫹ CD127⫹
`CD8⫹CD62L– CD127⫹
`Marker expression
`PD-L1 on CD14⫹
`.6
`PD-L2 on CD14⫹
`.2
`PD-L1 on CD4⫹ CD25⫹
`.04
`CD127 on CD4⫹CD62L⫹
`.0001
`CD127 on CD4⫹CD62L–
`.0001
`CD127 on CD8⫹CD62L⫹ < .0001
`CD127 on CD8⫹CD62L–
`.7
`
`⫹0.00
`⫹6.9
`⫺9.65
`⫹47.6
`⫺8.82
`⫹22.6
`⫹7.77
`⫹7.7
`⫹80.4 ⫹29.17
`⫹72.6
`⫹9.97
`⫹12.8
`⫹6.83
`⫹34.6 ⫹25.93 < .0001
`⫹27.9 ⫹11.04
`.03
`
`.015
`.041
`.003
`.02
`
`⫹0.0
`⫹14.4
`⫹53.3 ⫹10.0
`⫹61.4 ⫹17.3
`⫹5.9
`⫹0.0
`⫹87.1
`⫹0.0
`⫹134.6
`-5.5
`⫹18.1
`⫹6.5
`⫹29.4
`⫹9.7
`⫹55.3
`⫹2.3
`
`⫺5.3
`⫹9.3
`⫹1.0 ⫺20.2
`⫺6.7
`⫹60.5
`⫹70.7 ⫺31.3
`⫹75.6 ⫹12.1
`⫹86.7
`⫺1.6
`.09
`⫹50.7 ⫹30.0
`⫹16.7
`⫺2.6
`.0001
`⫹119.9 ⫹50.0 < .0001
`.013 ⫹137.4 ⫹16.7
`⫹346.7 ⫹24.2
`⫹192.8
`⫺2.9
`.0005
`⫹32.8 ⫹19.8
`⫹23.6
`⫹6.3
`.002
`⫹119.8 ⫹37.1
`⫹52.2
`⫹9.0
`.0002
`⫹88.8 ⫹54.9 < .0001 ⫹108.1 ⫹22.5
`.001
`
`.02
`.08
`.01
`
`.006
`
`⫹0.0
`⫹8.8
`⫺1.0
`⫹0.6
`⫹5.0
`⫹6.9
`⫹15.9 ⫹15.9
`⫹18.9
`⫹9.3
`⫹8.8
`⫹6.2
`⫹1.8
`⫺2.9
`
`⫹6.5
`⫹0.3
`⫹6.5
`⫹14.0
`⫹20.8
`⫹29.4
`⫹0.7
`
`⫹2.75
`⫺1.14
`⫹4.58
`⫹3.69
`⫹6.39
`⫹7.33
`⫹0.04
`
`.05
`.05
`.036
`
`⫹1.0
`⫹11.4
`⫺5.1
`⫺0.5
`⫹8.4
`⫹7.3
`⫹27.9 ⫹21.4
`⫹40.7 ⫹20.6
`⫹68.0 ⫹24.4
`⫹1.0
`⫺0.9
`
`.09
`
`⫹4.6
`⫹14.1
`⫺6.8
`⫺2.2
`⫹18.2 ⫹15.1
`⫹39.1 ⫹28.5 < .0001
`⫹58.2 ⫹33.0 < .0001
`⫹81.6 ⫹37.0 < .0001
`⫹3.6
`⫹1.5
`
`.003
`.001
`.0007
`
`.0007
`.004
`
`NOTE. Changes are reported compared with baseline values as both the mean and median percentage change in the absolute number of the selected subset or
`marker. Unadjusted P values are based on Wilcoxon signed rank testing; only P values ⬍ .1 are reported. Bolded P values indicate significance after adjustment for
`multiple comparisons (P ⬍ .0125). Adjusted P values test the significance of changes over time using repeated measurement analysis (see Patients and Methods).
`
`median increase at all time points (Table 3), suggesting that pidili-
`zumab induced large increases in those cells that were early and
`sustained, but restricted to a subset of patients. MESF analysis
`excluded the possibility that these changes could be solely ex-
`plained by upregulation of surface markers. There were also signif-
`icant increases in the absolute number of circulating CD8⫹
`peripheral (CD62L-CD127⫹) and central (CD62L⫹CD127⫹)
`memory T cells, as well as in CD4⫹ central memory T cells (Fig 2B).
`Finally, we found an increase in the cell surface expression of the
`interleukin 7␣receptor (CD127) on peripheral and central mem-
`ory CD4⫹ and CD8⫹ T cells (Fig 2C).
`
`DISCUSSION
`
`Monoclonal antibody therapy as a means of targeting immune
`checkpoints has emerged as a viable and effective antitumor
`strategy.3-5,9,21,22 Hematologic malignancies may be particularly at-
`tractive targets for this type of treatment, as patients with even ad-
`vanced myeloid or lymphoid tumors can be cured by adoptive
`immunotherapy delivered in the context of allogeneic HSCT. This
`raises the possibility that patients’ own immune systems can be har-
`nessed to eradicate those diseases, if the mechanisms that lead to
`immune tolerance of the tumor can be safely disabled. In the present
`trial, we show that the anti–PD-1 monoclonal antibody pidilizumab
`can be safely given to patients with DLBCL after AHSCT. The lack of
`significant autoimmune toxicity in our trial stands in contrast to the
`clinical experience so far with cytotoxic T-lymphocyte antigen-4
`blockade.23 Treatment was associated with an apparent CR rate of
`34% and overall response rate of 51% among patients with measur-
`able disease after transplant. This suggests direct antitumor activity,
`although given this study’s design, we cannot rule out the possibility
`
`that the residual radiologic abnormalities in some cases reflected
`treated disease or inflammation rather than the presence of viable
`lymphoma. Moreover, with a 16-month PFS of 0.72, the study met its
`prespecified primary end point. These results may compare favorably
`with those of two recent multicenter randomized clinical trials in this
`population, although our exclusion of patients who experienced re-
`lapse early after AHSCT precludes a direct comparison.24,25 In the
`rituximab era, as the prognosis of patients with relapsed or refractory
`disease is worse,24 new therapies are needed to increase the efficacy of
`salvage and to increase the success rate of AHSCT in patients without
`a PET-CR at transplantation26-28 who have a poorer outcome. We
`recently reported the outcomes of 105 patients with DLBCL who
`underwent transplantation in the last decade at our own institutions.29
`Among this cohort, we examined (on an institutional review board–
`approved study) the outcome of the 46 patients who were chemosen-
`sitive but PET positive after salvage and who would have otherwise
`met the eligibility criteria for the present study, including no progres-
`sion or relapse within 2 months of AHSCT. In this group, the 18-
`month post-AHSCT PFS was 0.52 (90% CI, 0.39 to 0.63). In the
`present study, the PFS was 0.72 among PET-positive patients treated
`with pidilizumab. This compares favorably with our historical experi-
`ence and with other published cohorts.26-28,30-32 Although not all
`patients on the present study had a postsalvage PET (which was not
`mandated per protocol), the absence of an apparent difference in
`outcome between those who did and those who did not argues against
`a strong selection bias, again with the caveat of the small numbers
`involved. We emphasize that the study was not powered for a com-
`parison among PET subgroups or with historical controls. Nonethe-
`less, our findings support the hypothesis that PD-1 blockade may be a
`viable therapeutic strategy in the high-risk subset of patients with
`residual disease and may overcome the negative prognostic value of a
`
`4204
`
`JOURNAL OF CLINICAL ONCOLOGY
`© 2013 by American Society of Clinical Oncology
`Downloaded from jco.ascopubs.org on August 31, 2015. For personal use only. No other uses without permission.
`Copyright © 2013 American Society of Clinical Oncology. All rights reserved.
`
`6 of 8
`
`BI Exhibit 1012
`
`

`

`PD-1 Blockade for DLBCL After AHSCT
`
`pretransplant positive PET scan. This hypothesis should now be tested
`in a randomized clinical trial.
`The increase in the number of some PD-1 ligand-bearing lym-
`phocyte subsets within 24 hours of drug infusion is consistent with an
`on-target effect of pidilizumab. Indeed, the interaction of PD-1 with
`either of its cognate ligands seems to induce cell death and signal
`suppression in lymphocytes and monocytes.33 The increase in circu-
`lating levels of cells expressing PD-1 ligands may therefore reflect the
`reversal of PD-1’s inhibition of cell survival or proliferation, although
`the rapidity of some of the changes occurring after treatment suggests
`that blockade of apoptosis is not the main or only mechanism at play
`and that mobilization of those cells from their reservoirs may be
`important. Also, our results suggest significant variability in the
`changes among different patients, implying additional complexity in
`the immune effects of PD-1 blockade in the setting of a reconstituting
`immune system. We also found an increase in effector and peripheral
`memory cell subsets, consistent with in vitro data that pidilizumab
`enhances the survival of human CD4⫹CD45RO⫹ cells (CureTech,
`unpublished data) within 72 hours of treatment. The increase in the
`cell surface expression of the interleukin 7␣receptor CD127, pivotal
`for the maturation and survival of memory T cells, suggests that
`pidilizumab induces molecular events associated with the fate of
`specific memory T-cell subsets. These analyses are only explor-
`atory, reported for hypothesis-generating purposes, and should be
`prospectively validated.
`Because the expression of PD-L1 on DLBCL cells may be re-
`stricted to a subset of tumors,10 it may be that future selection of
`patients for PD-1 blockade on the basis of ligand expression in the
`tumor or microenvironment could lead to a greater clinical benefit in
`the appropriate patient subgroups; this could not be ascertained on
`this study because we did not have access to tumor material for most
`patients. Furthermore, the ability to evaluate for on-target effects that
`predict outcome could provide a method to adapt post-AHSCT ther-
`apy in those patients. Although the answers to those questions must
`await future prospective trials, the present study represents the first
`efficacy trial of immune checkpoint blockade in hematologic malig-
`nancies. Our rapidly expanding scientific knowledge in this area, cou-
`pled with the availability of a growing number of monoclonal
`antibodies targeting those pathways, will doubtlessly lead into broader
`investigations of this strategy in other settings.
`
`AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS
`OF INTEREST
`
`Although all authors completed the disclosure declaration, the following
`author(s) and/or an author’s immediate family member(s) indicated a
`financial or other interest that is relevant to the subject matter under
`consideration in this article. Certain relationships marked with a “U” are
`those for which no compensation was received; those relationships marked
`with a “C” were compensated. For a detailed description of the disclosure
`categories, or for more information about ASCO’s conflict of interest policy,
`please refer to the Author Disclosure Declaration and the Disclosures of
`Potential Conflicts of Interest section in Information for Contributors.
`Employment or Leadership Position: Rinat Rotem-Yehudar, Cure Tech
`(C) Consultant or Advisory Role: Philippe Armand, Merck (C); Arnon
`Nagler, CureTech (C); Edie A. Weller, CureTech (C); Yi-Bin Chen,
`Otsuka (C), Genzyme (C); David A. Rizzieri, Teva (C), Celgene (C),
`Celgene (C), Novartis (C), Spectrum (C); Leo I.

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