throbber
V O L U M E
`
`2 8
`
`䡠 N U M B E R 1 9
`
`䡠
`
`J U L Y 1
`
`2 0 1 0
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`O R I G I N A L R E P O R T
`
`From the Johns Hopkins University
`School of Medicine and the Sidney
`Kimmel Comprehensive Cancer Center,
`Baltimore, MD; Henry Ford Health
`Systems, Detroit, MI; Carolina BioOn-
`cology Institute, Huntersville, NC;
`Washington University School of Medi-
`cine Siteman Cancer Center, St Louis,
`MO; and Medarex, Bloomsbury, NJ,
`and Milpitas, CA.
`
`Submitted November 21, 2009;
`accepted March 12, 2010; published
`online ahead of print at www.jco.org on
`June 1, 2010.
`
`Supported in part by a grant from the
`Melanoma Research Alliance Founda-
`tion (L.C., D.M.P., S.L.T.).
`
`I.L. and S.L.T. contributed equally to
`this work.
`
`Presented in part at the 44th Annual
`Meeting of the American Society of
`Clinical Oncology (ASCO) May 30-June
`3, 2008, Chicago, IL, and the 45th
`Annual Meeting of ASCO, May 29-June
`2, 2009, Orlando, FL.
`
`Terms in blue are defined in the glos-
`sary, found at the end of this article
`and online at www.jco.org.
`
`Authors’ disclosures of potential con-
`flicts of interest and author contribu-
`tions are found at the end of this
`article.
`
`Clinical Trials repository link available on
`JCO.org.
`
`Corresponding author: Suzanne L.
`Topalian, MD, Department of Surgery,
`Johns Hopkins University School of
`Medicine, 1550 Orleans St, CRB 2,
`Room 508, Baltimore, MD 21231;
`e-mail: stopali1@jhmi.edu.
`
`© 2010 by American Society of Clinical
`Oncology
`
`0732-183X/10/2819-3167/$20.00
`
`DOI: 10.1200/JCO.2009.26.7609
`
`Phase I Study of Single-Agent Anti–Programmed Death-1
`(MDX-1106) in Refractory Solid Tumors: Safety, Clinical
`Activity, Pharmacodynamics, and Immunologic Correlates
`Julie R. Brahmer, Charles G. Drake, Ira Wollner, John D. Powderly, Joel Picus, William H. Sharfman,
`Elizabeth Stankevich, Alice Pons, Theresa M. Salay, Tracee L. McMiller, Marta M. Gilson, Changyu Wang,
`Mark Selby, Janis M. Taube, Robert Anders, Lieping Chen, Alan J. Korman, Drew M. Pardoll, Israel Lowy,
`and Suzanne L. Topalian
`
`A
`
`B
`
`S
`
`T
`
`R
`
`A
`
`C
`
`T
`
`Purpose
`Programmed death-1 (PD-1), an inhibitory receptor expressed on activated T cells, may suppress
`antitumor immunity. This phase I study sought to determine the safety and tolerability of anti–PD-1
`blockade in patients with treatment-refractory solid tumors and to preliminarily assess antitumor
`activity, pharmacodynamics, and immunologic correlates.
`Patients and Methods
`Thirty-nine patients with advanced metastatic melanoma, colorectal cancer (CRC), castrate-
`resistant prostate cancer, non–small-cell
`lung cancer (NSCLC), or renal cell carcinoma (RCC)
`received a single intravenous infusion of anti–PD-1 (MDX-1106) in dose-escalating six-patient
`cohorts at 0.3, 1, 3, or 10 mg/kg, followed by a 15-patient expansion cohort at 10 mg/kg. Patients
`with evidence of clinical benefit at 3 months were eligible for repeated therapy.
`
`Results
`Anti–PD-1 was well tolerated: one serious adverse event, inflammatory colitis, was observed in a
`patient with melanoma who received five doses at 1 mg/kg. One durable complete response
`(CRC) and two partial responses (PRs; melanoma, RCC) were seen. Two additional patients
`(melanoma, NSCLC) had significant lesional tumor regressions not meeting PR criteria. The serum
`half-life of anti–PD-1 was 12 to 20 days. However, pharmacodynamics indicated a sustained mean
`occupancy of ⬎ 70% of PD-1 molecules on circulating T cells ⱖ 2 months following infusion,
`regardless of dose. In nine patients examined, tumor cell surface B7-H1 expression appeared to
`correlate with the likelihood of response to treatment.
`
`Conclusion
`Blocking the PD-1 immune checkpoint with intermittent antibody dosing is well tolerated and associated
`with evidence of antitumor activity. Exploration of alternative dosing regimens and combinatorial
`therapies with vaccines, targeted therapies, and/or other checkpoint inhibitors is warranted.
`
`J Clin Oncol 28:3167-3175. © 2010 by American Society of Clinical Oncology
`
`INTRODUCTION
`
`Genetic and epigenetic aberrations occur com-
`monly in human tumors and produce altered anti-
`genic profiles that can be selectively recognized by
`the adaptive immune response.1 A dynamic inter-
`play exists between host and tumor, and the ability
`of the tumor to evade immune recognition often
`determines the clinical course of the disease.2 The
`successes of passive immunotherapies, such as
`monoclonal antibodies (mAbs) directed against
`tumor or vascular cell surface molecules or adop-
`tive transfer of tumor-specific T cells, validate the
`potential of immunotherapy to eradicate established
`
`metastatic cancers. However, active immunothera-
`peutic strategies designed to enhance endogenous
`antitumor responses, such as cancer vaccines, have
`been far less successful.
`Augmenting specific antitumor CD4⫹ and
`CD8⫹ T cell responses is a major goal of cancer
`immunotherapy. Important insights explaining the
`limitations of T cell– based cancer immunothera-
`pies have come from the discovery of inhibitory
`coreceptors and pathways termed immune check-
`points, which restrain T cell functions in normal
`physiologic settings and may be exploited by tu-
`mors.3 Preclinical cancer models demonstrate that
`inhibitory signals mediated by coreceptors on
`
`© 2010 by American Society of Clinical Oncology
`
`3167
`
`Genome Ex. 1030
`Page 1 of 9
`
`

`

`Brahmer et al
`
`tumor-specific T cells impede antitumor immunity and suggest that
`blockade of such interactions can release the brakes on immune re-
`sponsiveness leading to tumor elimination. The most extensively
`studied inhibitory T cell coreceptor, CTLA-4 (CD152), has been
`evaluated in patients with advanced cancers. As originally pre-
`dicted by murine models, anti–CTLA-4 therapy in humans resulted
`in objective tumor regressions including durable complete re-
`sponses (CRs) in some patients. However, as anticipated from the
`uncontrolled lymphoproliferation observed in CTLA-4 null mice,4
`anti–CTLA-4 therapy was associated with a significant frequency of
`serious immunologic adverse events (AEs).5 Thus, investigators have
`searched for new checkpoint blocking agents with more favorable
`therapeutic profiles.
`Programmed death-1 (PD-1, CD279) is an inhibitory coreceptor
`expressed on antigen-activated and exhausted T and B cells.6 It bears
`homology to CTLA-4 but provides distinct immune-inhibitory sig-
`nals. In contrast to early lethality in CTLA-4 knockout mice, PD-1
`knockouts demonstrate modest late-onset strain- and organ-specific
`autoimmunity.7,8 There are two known ligands for PD1: B7-H1/PD-L1
`(hereafter B7-H1), the predominant mediator of PD-1–dependent
`immunosuppression, and B7-DC/PD-L2. In murine tumor models,
`B7-H1 expression confers immune resistance, and interrupting PD-1:
`B7-H1 interactions has antitumor effects.9-11 B7-H1 is highly upregu-
`lated in many murine and human tumors (either in tumor cells or
`nontransformed cells in the tumor microenvironment such as
`antigen-presenting cells),12 and its expression is associated with poor
`outcome for patients with certain epithelial cancers.13,14 These find-
`ings have focused attention on PD-1:B7-H1 blockade as a strategy for
`cancer immunotherapy. On the basis of these considerations, we ini-
`tiated a phase I clinical trial of PD-1 blockade with the fully human
`mAb MDX-1106 in 39 patients with advanced treatment-refractory
`solid tumors. We report here the safety, antitumor activity, pharma-
`codynamics, and correlative in vitro results from this trial.
`
`PATIENTS AND METHODS
`
`MDX-1106
`MDX-1106 (BMS-936558/ONO-4538) is a genetically engineered, fully
`human immunoglobulin G4 (IgG4) mAb specific for human PD-1 (Appendix
`Fig A1A, online only). Mice transgenic for human Ig loci were immunized with
`Chinese hamster ovary cell PD-1 transfectants and a PD-1/human IgG1 Fc
`fusion protein. MDX-1106 contains an engineered hinge region mutation
`(S228P) designed to prevent exchange of IgG4 molecules; the IgG4 isotype
`minimizes cellular and complement-mediated cytolytic functions. MDX-1106
`⫽ 2.6 nmol/L by Scatchard analysis to
`binds PD-1 with high affinity (KD
`polyclonally activated human T cells), blocks its interactions with both B7-H1
`and B7-DC (Appendix Fig A1B), and enhances tumor antigen-specific T cell
`proliferation and secretion of cytokines in vitro.15
`
`Patients
`Eligible patients had treatment-refractory metastatic melanoma,
`castrate-resistant prostate cancer, renal cell carcinoma (RCC), non–small-cell
`lung cancer (NSCLC), or colorectal cancer (CRC), and had no cancer therapy
`for at least 4 weeks before enrollment. Patients were ⱖ 18 years old with a life
`expectancy of ⱖ 12 weeks, an Eastern Cooperative Oncology Group (ECOG)
`performance status of 0 or 1, adequate organ function, and no ongoing sys-
`temic infections or history of autoimmune disease. Concurrent antineoplastic
`therapies, systemic steroids, and prior treatment with anti–CTLA-4 were not
`permitted. Patients with treated stable brain metastases were eligible.
`
`Study Design and Procedures
`This multi-institutional, first in-human, open-label, phase I, dose-
`escalation study was approved by local institutional review boards. All partic-
`ipating patients signed informed consent. The primary objectives were to
`characterize the safety and tolerability of a single dose of MDX-1106 in patients
`with selected malignancies and to determine the maximum-tolerated dose
`(MTD) and pharmacokinetics. Secondary objectives included assessing anti-
`tumor activity, pharmacodynamics, and immunologic end points. Sequential
`cohorts of six patients received a 60-minute intravenous infusion of MDX-
`1106 at 0.3, 1, 3, or 10 mg/kg and were evaluated for toxicities on a weekly basis
`for 8 weeks. Dose-limiting toxicity (DLT) was defined as a treatment-related
`grade ⱖ 3 AE or laboratory abnormality occurring ⱕ 28 days postdose. The
`MTD was the highest dose at which no more than one of six patients experi-
`enced a DLT. Fifteen additional patients were planned to be enrolled at the
`MTD or the highest planned dose (10 mg/kg) to confirm safety.
`Patients were restaged radiographically (Response Evaluation Criteria in
`Solid Tumors [RECIST] 1.0) at 8 and 12 weeks. Patients with progressive
`disease were taken off study. Those with stable disease or evidence of lesional
`tumor regression, no AE grade ⱖ 3, and no evidence of human antihuman Ab
`at a 1:10 serum dilution received additional doses of MDX-1106 at weeks 12
`and 16 and were then observed for 3 months and restaged. Those with contin-
`ued clinical benefit could receive two more doses, spaced by 4 weeks. Each
`re-treatment phase was 16 weeks. Patients with objective partial responses
`(PRs) or CRs were observed, with optional re-treatment on progression.
`Responses are reported as of January 2010.
`
`Pharmacokinetics
`Serum samples were collected serially before and up to 85 days after the
`first dose of MDX-1106. MDX-1106 serum concentrations were determined
`with a quantitative enzyme-linked immunosorbent assay (ELISA) capable of
`detecting ⱖ 1.2 ␮g/mL, using 96-well plates coated with chimeric PD-1/
`human IgG1 Fc protein (R&D Systems, Minneapolis, MN).
`
`Tumor Biopsies
`Sections of formalin-fixed, paraffin-embedded tumor specimens ar-
`chived before protocol entry or core-needle or excisional biopsies obtained
`immediately pre- and post-therapy were subjected to hematoxylin and
`eosin staining and immunohistochemistry (IHC) to detect lymphoid infil-
`trates (anti-CD3, anti-CD4, and anti-CD8) and B7-H1 expression (murine
`antihuB7-H1, clone 5H1; previously described13). Pigmented melanoma sam-
`ples were bleached before staining and visualized with a red chromogen.
`Tumors were considered B7-H1–positive if ⱖ 5% of tumor cells showed
`membranous staining with 5H1.
`
`Immunologic Assessments
`Delayed-type hypersensitivity reactions to Candida albicans and tetanus
`toxoid were assessed along with viral antigen recall reactions (details are in-
`cluded in the Appendix, online only). Peripheral blood lymphocyte (PBL)
`phenotypes were also assessed. Serially collected blood was analyzed for the
`presence and activation status of various lymphocyte subsets, as detailed in
`the Appendix.
`
`PD-1 Receptor Occupancy (pharmacodynamics)
`MDX-1106 binding to PD-1 molecules on circulating CD3⫹ PBLs was
`investigated with flow cytometric analysis of serially collected blood samples
`(see PBL phenotyping schedule in the Appendix). Peripheral blood mononu-
`clear cells were preincubated (30 minutes at 4°C) with a saturating concentra-
`tion (20 ␮g/mL) of either unlabeled huIgG4 (isotype control) or MDX-1106,
`washed extensively, and then costained with anti-CD3 fluorescein isothiocya-
`nate and murine antihuIgG4 biotin (Invitrogen, Carlsbad, CA) plus
`streptavidin-phycoerythrin. PD-1 occupancy by infused MDX-1106 was esti-
`mated as the ratio of the percent of CD3⫹ cells stained with antihuIgG4 after in
`vitro saturation with isotype control Ab (indicating in vivo binding) to that
`observed after MDX-1106 saturation (indicating total available binding sites).
`
`3168
`
`© 2010 by American Society of Clinical Oncology
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`Genome Ex. 1030
`Page 2 of 9
`
`

`

`Single-Agent Anti–PD-1 Therapy for Refractory Solid Tumors
`
`Table 1. Patient Characteristics
`
`Table 2. Treatment Characteristics and Clinical Response to Therapy
`
`Characteristic
`
`No. of Patients
`
`Sex
`Male
`Female
`Age, years
`Median
`Range
`Tumor histology
`Colorectal cancer
`Melanoma
`Prostate cancer
`NSCLC
`Renal cell carcinoma
`ECOG PS
`0
`1
`Prior therapies
`Median
`Range
`Chemotherapyⴱ
`Radiation therapy
`Surgery
`Immunotherapy
`Biologics†
`Hormonal therapy
`
`62
`42-84
`
`4
`1-13
`
`22
`17
`
`14
`10
`8
`6
`1
`
`13
`26
`
`36
`13
`39
`14
`26
`8
`
`%
`
`56.4
`43.6
`
`35.9
`25.6
`20.5
`15.4
`2.6
`
`33.3
`66.7
`
`92.3
`33.3
`100
`35.9
`66.7
`20.5
`
`Abbreviations: NSCLC, non–small-cell lung cancer; ECOG, Eastern Coopera-
`tive Oncology Group; PS, performance status.
`ⴱIncludes molecularly targeted therapies.
`†Includes monoclonal antibody therapies.
`
`RESULTS
`
`Patients and Treatments
`Thirty-nine patients with advanced metastatic NSCLC, mela-
`noma, castrate-resistant prostate cancer, RCC, or CRC received
`MDX-1106 in four escalating dose cohorts of 0.3 to 10 mg/kg and an
`expansion cohort at 10 mg/kg, from October 2006 through June 2009
`(Tables 1 and 2). Their median age was 62 years. All had progressive
`treatment-refractory disease, and they had undergone a median of
`four prior therapies.
`
`Treatment-Related Toxicities
`MDX-1106 was well-tolerated: no DLTs were observed after one
`dose, and an MTD was not defined in this study. Grade ⱖ 2 adverse
`clinical and laboratory events are summarized in Appendix Table A1
`(online only). Most frequent were decreased CD4⫹ lymphocyte
`counts (14 patients, 35.9%), lymphopenia (10 patients, 25.6%), fa-
`tigue and musculoskeletal events (six patients each, 15.4%). No pa-
`tient developed human antihuman Ab, even after multiple doses.
`Immune-related AEs (irAEs) were of special interest because of
`the presumed mechanism of action of anti–PD-1 and prior experience
`with anti–CTLA-4.5 No grade ⱖ 3 irAE occurred in the 28-day period
`following the first dose of anti–PD-1. One patient with metastatic
`ocular melanoma developed grade 3 inflammatory colitis following
`five doses (1 mg/kg) administered over 8 months (Appendix Fig A2,
`online only), which responded to steroids and infliximab. One patient
`
`Dose
`(mg/kg)
`
`No. of
`Patients
`
`0.3
`1
`3
`10
`
`Total
`
`6
`6
`6
`21
`
`39
`
`Total No. of Doses
`
`1
`
`6
`3
`3
`15
`
`27
`
`2
`
`0
`1
`0
`1
`
`2
`
`3
`
`0
`1
`2
`4
`
`7
`
`5
`
`0
`1
`1
`0
`
`2
`
`11
`
`0
`0
`0
`1
`
`1
`
`Best Response
`(duration in months)ⴱ
`
`N/A
`1 MXR (1)
`1 CR (21⫹)†
`2 PR (3⫹, 16⫹)‡§
`1 MXR (1)
`1 CR, 2 PR, 2 MXR
`
`Abbreviations: N/A, not applicable; MXR, mixed response defined as regres-
`sion in some lesions but concomitant progression in others; CR, complete
`response; PR, partial response.
`ⴱCR and PR by Response Evaluation Criteria in Solid Tumors 1.0 criteria.
`†This patient with stage IV colorectal cancer had previously shown progres-
`sive disease after receiving chemotherapy regimens including bevacizumab
`and cetuximab.
`‡PR duration of 3⫹ months was preceded by an MXR in this patient with
`melanoma lasting 20 months. Previous therapies that were ineffective in-
`cluded high-dose interleukin-2 and temozolomide.
`§PR duration of 16⫹ months was preceded by an MXR in this patient with renal
`cell carcinoma lasting 4 months. Previous therapies that were ineffective included
`sunitinib, sorafenib, and an experimental histone deacetylase inhibitor.
`
`(10 mg/kg) experienced grade 2 hypothyroidism requiring hormone
`replacement. Two patients (at 3 and 10 mg/kg) developed grade 2
`polyarticular arthropathies requiring oral steroids and were not fur-
`ther treated; in retrospect, both had potentially contributory predis-
`posing factors, one with a history of Lyme arthritis and polymyalgia
`rheumatica, the other with a preexisting antinuclear antibody titer
`⬎ 1:1000.
`
`Antitumor Activity
`One patient with CRC (3 mg/kg) achieved a CR, and two patients
`with RCC (10 mg/kg) and melanoma (10 mg/kg) experienced PRs to
`therapy. A 67-year-old male with CRC metastatic to intra-abdominal
`lymph nodes received five doses of MDX-1106 and experienced a CR
`persisting 21⫹ months. A 72-year-old male with multiorgan meta-
`static RCC had a mixed response after one dose of MDX-1106, with
`progression in a pancreatic metastasis but regression in other sites; this
`evolved to an overall PR after two additional doses, lasting 16⫹
`months without further therapy (Fig 1A). A 51-year-old female with
`melanoma metastatic to multiple lymph nodes and liver sites initially
`experienced a mixed response, with regression at all sites except an
`enlarging subpectoral lymph node, and achieved a PR after receiving
`11 doses of MDX-1106 over 24 months (Fig 1B). Two additional
`patients had significant lesional or mixed tumor regressions (defined
`as regression in individual lesions with concomitant progression at
`other sites), including one with NSCLC (1 mg/kg) and another with
`melanoma (10 mg/kg). In total, 12 patients with stable disease or
`lesional tumor regressions at the first disease assessment received
`multiple doses of MDX-1106 (Table 2).
`
`Tumor Biopsies
`B7-H1 expression on tumors may affect the ability to respond to
`PD-1 blockade. To explore this, tumor biopsies from nine patients
`undergoing treatment with MDX-1106 were analyzed for B7-H1 ex-
`pression with IHC (Appendix Table A2, online only). In two cases,
`both pre- and post-treatment samples were available; in the others,
`only pretreatment (six cases) or post-treatment (one case) samples
`
`www.jco.org
`
`© 2010 by American Society of Clinical Oncology
`
`3169
`
`Genome Ex. 1030
`Page 3 of 9
`
`

`

`A
`
`B
`
`Brahmer et al
`
`01/15/08
`
`04/22/08
`
`07/22/08
`
`03/25/08
`
`04/14/09
`
`Pre-Rx
`
`12 wk post Dose 1
`
`8 wk post Dose 3
`
`Pre-Rx
`
`4 wk post Dose 1
`
`4 wk post Dose 3
`
`Fig 1. Objective tumor responses in patients with metastatic renal cell carcinoma (RCC) and melanoma after repeated dosing with anti–programmed death-1
`monoclonal antibody (MDX-1106) at 10 mg/kg. (A) Patient 4033 with RCC experienced a partial response (PR) after receiving three doses of MDX-1106. Regression
`of metastases in mediastinal lymph nodes and bone (scapula) demonstrated on contrast-enhanced computed tomography scans are representative of lesions at other
`sites including lung, muscle, pancreas, and pericolic lymph node. Date of first treatment was January 29, 2008. (B) Patient 3019 experienced a PR after receiving 11
`doses of MDX-1106. Serial core-needle biopsies of a regressing axillary lymph node metastasis were stained with anti-CD8, revealing a moderate post-treatment
`infiltrate. Infiltration of CD4⫹ cells was not observed (not shown). 20⫻ objective. Rx, treatment; wk, week.
`
`were accessible. Tumor cell staining for B7-H1 expression fell into
`three patterns: negative, intracytoplasmic, or membranous (cell sur-
`face). Among nine patients studied, four exhibited membranous
`B7-H1 staining (Fig 2): three of these patients experienced tumor
`regressions following MDX-1106 therapy; the fourth was treated in
`the 0.3-mg/kg cohort where no responses were observed. Con-
`versely, among five patients whose tumors failed to express B7-H1
`at the cell surface, there was no evidence of clinical response.
`B7-H1 staining patterns were consistent in five patients from
`
`whom multiple biopsies were available (AppendixTable A2). In
`this small sample size, the correlation between membranous
`B7-H1 expression on tumor cells and the likelihood of tumor
`regression following PD-1 blockade suggested potential signifi-
`cance (two-sided P ⫽ .0476; Fisher’s exact test).
`Melanoma patient 3019, who experienced a PR to anti–PD-1
`therapy, underwent pre- and post-treatment biopsies of an axillary
`lymph node metastasis for characterization of intratumoral lymphoid
`infiltrates by IHC. Whereas the pretreatment biopsy contained only
`
`3170
`
`© 2010 by American Society of Clinical Oncology
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`Genome Ex. 1030
`Page 4 of 9
`
`

`

`Single-Agent Anti–PD-1 Therapy for Refractory Solid Tumors
`
`Isotype Control
`
`Anti-B7-H1
`
`A
`
`B
`
`Fig 2. Membranous pattern of B7-H1 ex-
`pression demonstrated on (A) renal cell car-
`cinoma cells in a tumor thrombus from
`patient 4033, and (B) melanoma cells in an
`axillary lymph node metastasis from patient
`3019. Both patients experienced partial re-
`sponses after anti–programmed death-1
`monoclonal antibody (MDX-1106) therapy.
`40⫻ objective.
`
`sparse lymphoid cells, subsequent tumor regression was accompanied
`by a moderate infiltration of CD8⫹, but not CD4⫹, T cells (Fig 1B).
`
`Reactivity to Recall Antigens and PBL Phenotypes
`No significant effects of MDX-1106 therapy on delayed-type
`hypersensitivity responses against C albicans or tetanus toxoid or
`antiviral recall responses by interferon gamma ELISpot analysis were
`observed in 15 and six patients examined, respectively. The effects of a
`single 10-mg/kg dose of MDX-1106 on PBL numbers, subset profiles,
`and activation status were analyzed in 17 patients (Fig 3). Twenty-four
`hours postdose, total lymphocyte as well as CD3, CD4, and CD8
`numbers declined and then rebounded from days 2 through 29 and
`declined again from days 29 through 85. These trends were not ob-
`served for CD19 (B lymphocyte) or CD56 (natural killer) cells (not
`shown), suggesting a selective effect on T cells. Percentages of CD3,
`CD4, and CD8 cells declined steadily over the 85-day observation
`period, with a reciprocal increase in CD19 and CD56 cell percentages
`(Appendix Fig A3A, online only). No significant changes were ob-
`served in expression of the T cell activation markers CD25, CD45RO,
`or HLA-DR by CD4⫹ or CD8⫹ PBLs (Appendix Fig A3B).
`
`PD-1 Receptor Occupancy (pharmacodynamics)
`PD-1 has predominant cell surface expression, unlike CTLA-4,
`which is displayed only transiently at the T cell surface. Thus, it was
`possible to develop flow cytometric methods to evaluate the pharma-
`
`codynamics of infused MDX-1106, estimating PD-1 occupancy on
`circulating T cells over time. Standard pharmacokinetic measure-
`ments of MDX-1106 serum concentrations yielded an approximate
`serum half-life (t1/2) of 12 days (0.3-, 1-, or 3-mg/kg dose) to 20 days
`(10 mg/kg), with maximum concentration (Cmax) and AUC directly
`related to dose. However, pharmacokinetics and pharmacodynamics
`were unexpectedly discordant in 15 patients studied (Fig 4). PD-1
`occupancy appeared to be dose-independent, with a mean peak occu-
`pancy of 85% (range, 70% to 97%) and a mean plateau occupancy of
`72% (range, 59% to 81%) observed at 4 to 24 hours and ⱖ 57 days,
`respectively, after one infusion (Fig 4A). These data are consistent with
`the high affinity of MDX-1106 for PD-1—in vitro, 0.04 ␮g/mL MDX-
`1106 is sufficient to occupy ⬎ 70% PD-1 molecules on T cells (not
`shown)—suggesting that even when serum levels are undetectable
`(⬍ 1.2 ␮g/mL), sufficient concentrations persist to maintain plateau
`PD-1 occupancy. Occupancy eventually decayed after 85 days (Fig 4B,
`top and middle panels). In patients receiving repeated infusions of
`MDX-1106 at 10 mg/kg, troughs and peaks of PD-1 occupancy
`around each dose were observed (Fig 4B, middle and bottom panels),
`although 100% occupancy was not achieved. In vitro experiments
`indicate that PD-1 occupancy analyses of cryopreserved PBLs may
`underestimate occupancy on fresh PBLs (not shown). It is unknown
`whether these findings in circulating lymphocytes reflect PD-1 occu-
`pancy on lymphocytes in the tumor, secondary lymphoid organs,
`and/or tissues.
`
`www.jco.org
`
`© 2010 by American Society of Clinical Oncology
`
`3171
`
`Genome Ex. 1030
`Page 5 of 9
`
`

`

`Brahmer et al
`
`treatment will be necessary to make direct comparisons with anti–
`CTLA-4.
`While efficacy was not a primary end point of this trial, we found
`evidence of antitumor activity, with one CR (ongoing at 21 months)
`and two PRs (ongoing at 3 and 16 months) in patients with CRC,
`melanoma, and RCC; significant lesional regressions were seen in two
`additional patients with melanoma and NSCLC. The observed clinical
`activity of MDX-1106 is most likely exerted through immunologic
`mechanisms rather than direct tumoricidal effects, since nonhe-
`matologic tumors do not express PD-1. Importantly, tumor re-
`gressions were seen in patients with colon and lung cancer,
`suggesting that the capacity of anti–PD-1 to enhance antitumor
`immunity extends beyond the classically immunogenic tumor types
`of melanoma and RCC.
`Two possible scenarios for T cell inhibition via PD-1:B7-H1
`ligation have been proposed in the context of cancer.16,17 First, lym-
`phocytes may encounter B7-H1 expressed on activated antigen-
`presenting cells during priming or reactivation in lymphoid organs. In
`addition, tumor-specific effector lymphocytes may be inhibited on
`contact with B7-H1–positive tumor cells. In the latter case, direct
`tumor assessment for B7-H1 expression may provide a powerful pre-
`dictive biomarker for responsiveness to anti–PD-1. Analysis of serial
`tumor biopsies may provide additional insights into specific antitu-
`mor mechanisms resulting from PD-1 blockade. A regressing lesion in
`one melanoma patient on this study revealed a significantly increased
`and selective CD8⫹ T cell infiltrate post-therapy. While this kind of
`analysis needs to be performed on much larger numbers of patients, in
`light of the acute decline in CD3⫹ T cell numbers observed post-
`treatment, it is interesting to speculate that anti–PD-1 may cause
`redistribution of lymphocyte subsets from the blood into tumor and
`tissue sites.
`There is a single previous report18 of cancer therapy with a differ-
`ent anti–PD-1 antibody, CT-011, in 17 patients with hematologic
`malignancies. One CR was reported in a non-Hodgkin’s lymphoma
`patient. Because the CT-011 trial evaluated patients with a completely
`distinct and nonoverlapping set of cancers relative to this study, it is
`difficult to compare the two antibodies at this time.
`The promising safety profile and evidence of durable clinical
`activity in this phase I trial is encouraging, and early results from a
`follow-up trial (NCT00730639) appear to confirm the tolerability and
`activity of anti–PD-1 in patients for whom multiple prior therapies
`failed. However, it is important to consider factors that might
`improve the response rate. Despite the consistently high level of
`PD-1 occupancy observed on PBLs and the fact that T cells continu-
`ously redistribute between blood, lymph, and tissue, repeated dosing
`of MDX-1106 at shorter intervals might increase occupancy and tissue
`penetration and should be explored. Patient selection is an important
`issue, since anti–PD-1 may have suboptimal effects in patients with
`compromised immune systems. This notion is supported by preclin-
`ical models demonstrating that anti–PD-1 is most effective against
`immunogenic tumors in intact mice. Another important factor in
`determining outcomes may be intratumoral expression of B7-H1,
`either by tumor cells or by nontransformed cells in the tumor micro-
`environment. As suggested by our preliminary biopsy analysis, tumor
`cell surface expression of B7-H1 may be a predictor of responsiveness
`to PD-1 blockade. Finally, on the basis of murine models showing that
`anti–PD-1 is highly synergistic in combination with tumor vaccines,19
`we anticipate that more effective uses of this agent for cancer therapy
`
`1,500
`
`(17)(15)
`
`(14)
`
`1,000
`
`(16)
`
`(12)
`
`(13)
`
`Lymphocytes
`CD3
`CD4
`CD8
`
`(10)
`
`500
`
`0
`
`1
`
`2
`
`20
`40
`Time (days)
`
`60
`
`80
`
`100
`
`1,500
`
`2,000
`
`1,500
`
`1,000
`
`500
`
`0
`
`1
`
`2
`
`Time (days)
`
`A
`
`Lymphocytes (per mm3)
`
`B
`
`Lymphocytes (per mm3)
`
`Fig 3. Effects of a single dose of anti–programmed death-1 monoclonal
`antibody (MDX-1106; 10 mg/kg) on circulating lymphocyte numbers. (A) Twenty-
`four hours postdose, a decline in total lymphocyte as well as CD3, CD4, and CD8
`numbers was observed (two-sided P ⫽ .004, .002, ⬍ .001, and .01 respectively;
`Wilcoxon signed rank test). These parameters followed similar trends, rebound-
`ing from days 2 through 29 (two-sided P ⬍ .001; two-sided P ⫽ .01 for CD8), and
`declining again from days 29 through 85 (two-sided P ⬍ .001; mixed model test
`for trend with knots [changes in slopes] and repeated measures). Means ⫾
`standard error of mean are shown; numbers of patients studied at each time
`point are indicated in parentheses.
`(B) Paired analysis of total
`lymphocyte
`numbers in 16 patients, comparing immediate pretreatment samples (day 1) with
`24-hour post-treatment samples (day 2). A significant decline at day 2 was
`observed (two-sided P ⫽ .004; Wilcoxon signed rank test). Dotted line indicates
`the lower limit of normal lymphocyte counts.
`
`DISCUSSION
`
`We report results from a dose-escalation trial of a fully human anti–
`PD-1 mAb, MDX-1106, in 39 patients with refractory metastatic can-
`cers. MDX-1106 was well tolerated to the maximum planned dose of
`10 mg/kg. While both anti–PD-1 and anti–CTLA-4 therapies are
`associated with irAEs, as predicted by preclinical models and consis-
`tent with the physiologic roles of these molecules, these toxicities
`appear to be less frequent and milder in patients receiving anti–PD-1.
`Among 39 patients treated with MDX-1106, a single attributable seri-
`ous AE, inflammatory colitis, occurred. Low-grade irAEs occurred in
`two patients with polyarticular arthropathies requiring oral steroids
`and one patient with hypothyroidism requiring hormone replace-
`ment. However, expanded experience with multidose anti–PD-1
`
`3172
`
`© 2010 by American Society of Clinical Oncology
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`Genome Ex. 1030
`Page 6 of 9
`
`

`

`Single-Agent Anti–PD-1 Therapy for Refractory Solid Tumors
`
`100
`
`B
`
`100
`
`0.3 mg/kg
`
`[MDX-1106] (ug/mL)
`PD-1 occupancy (%)
`
`300
`
`200
`
`100
`
`[MDX-1106] (ug/mL)
`
`A
`
`Pt. 4025
`
`100
`
`200
`300
`Time (days)
`
`400
`
`500
`
`600
`
`Pt. 4033
`
`400
`
`500
`
`600
`
`80
`
`60
`
`40
`
`20
`
`0
`
`100
`
`80
`
`60
`
`40
`
`20
`
`PD-1 Occupancy (%)
`
`PD-1 Occupancy (%)
`
`PD-1 Occupancy (%)
`
`80
`
`60
`
`40
`
`20
`
`0
`
`100
`
`PD-1 Occupancy (%)
`
`80
`
`60
`
`40
`
`20
`
`0
`
`0
`
`20
`
`40
`60
`Time (days)
`
`80
`
`1 mg/kg
`
`[MDX-1106] (ug/mL)
`PD-1 occupancy (%)
`
`300
`
`200
`
`100
`
`0
`
`[MDX-1106] (ug/mL)
`
`20
`
`40
`60
`Time (days)
`
`80
`
`0
`
`100
`
`200
`300
`Time (days)
`
`Pt. 3019
`
`100
`
`200
`300
`Time (days)
`
`400
`
`500
`
`600
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`PD-1 Occupancy (%)
`
`100
`
`PD-1 Occupancy (%)
`
`80
`
`60
`
`40
`
`20
`
`0
`
`100
`
`PD-1 Occupancy (%)
`
`80
`
`60
`
`40
`
`20
`
`0
`
`3 mg/kg
`
`[MDX-1106] (ug/mL)
`PD-1 occupancy (%)
`
`300
`
`200
`
`100
`
`[MDX-1106] (ug/mL)
`
`0
`
`20
`
`40
`60
`Time (days)
`
`80
`
`10 mg/kg
`(n = 10)
`
`[MDX-1106] (ug/mL)
`PD-1 occupancy (%)
`
`300
`
`200
`
`100
`
`[MDX-1106] (ug/mL)
`
`0
`
`20
`
`40
`60
`Time (days)
`
`80
`
`Fig 4. Pharmacodynamics of anti–programmed death-1 (PD-1) monoclonal antibody (MDX-1106). (A) PD-1 occupancy on circulating CD3⫹ T cells after one infusion
`of MDX-1106 is shown for single patients (Pts.) each receiving 0.3, 1, or 3 mg/kg, and for 10 patients receiving 10 mg/kg (mean ⫾ standard error of mean; solid squares).
`Serum concentrations of MDX-1106 at the same time points are indicated (open diamonds). (B) Long-term PD-1 occupancy analysis in patients receiving one (top panel)
`or multiple doses (middle and bottom panels) of MDX-1106 at 10 mg/kg. All patients received infusions at day 1; additional infusions are indicated by arrows. Results
`in (B) middle and bottom panels are representative of five patients receiving multiple doses.
`
`www.jco.org
`
`© 2010 by American Society of Clinical Oncology
`
`3173
`
`Genome Ex. 1030
`Page 7 of 9
`
`

`

`Brahmer et al
`
`will involve combinatorial therapies with other agents that boost en-
`dogenous antitumor immunity. Treatment regimens combining
`MDX-1106 with vaccines, molecularly targeted therapies, or other
`immunomodulators are already under evaluation in the laboratory
`for near-term clinical development.
`
`AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS
`OF INTEREST
`
`Although all authors completed the disclosure declaration, the following
`author(s) indicated a financial or other interest that is relevant to the subject
`m

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