throbber
Original Article
`
`PD-1 Blockade in Tumors
`with Mismatch-Repair Deficiency
`D.T. Le, J.N. Uram, H. Wang, B.R. Bartlett, H. Kemberling, A.D. Eyring,
`A.D. Skora, B.S. Luber, N.S. Azad, D. Laheru, B. Biedrzycki, R.C. Donehower,
`A. Zaheer, G.A. Fisher, T.S. Crocenzi, J.J. Lee, S.M. Duffy, R.M. Goldberg,
`A. de la Chapelle, M. Koshiji, F. Bhaijee, T. Huebner, R.H. Hruban, L.D. Wood,
`N. Cuka, D.M. Pardoll, N. Papadopoulos, K.W. Kinzler, S. Zhou, T.C. Cornish,
`J.M. Taube, R.A. Anders, J.R. Eshleman, B. Vogelstein, and L.A. Diaz, Jr.
`
`ABSTR ACT
`
`BACKGROUND
`Somatic mutations have the potential to encode “non-self” immunogenic antigens.
`We hypothesized that tumors with a large number of somatic mutations due to
`mismatch-repair defects may be susceptible to immune checkpoint blockade.
`METHODS
`We conducted a phase 2 study to evaluate the clinical activity of pembrolizumab,
`an anti–programmed death 1 immune checkpoint inhibitor, in 41 patients with
`progressive metastatic carcinoma with or without mismatch-repair deficiency. Pem-
`brolizumab was administered intravenously at a dose of 10 mg per kilogram of body
`weight every 14 days in patients with mismatch repair–deficient colorectal cancers,
`patients with mismatch repair–proficient colorectal cancers, and patients with mis-
`match repair–deficient cancers that were not colorectal. The coprimary end points
`were the immune-related objective response rate and the 20-week immune-related
`progression-free survival rate.
`RESULTS
`The immune-related objective response rate and immune-related progression-free sur-
`vival rate were 40% (4 of 10 patients) and 78% (7 of 9 patients), respectively, for mis-
`match repair–deficient colorectal cancers and 0% (0 of 18 patients) and 11% (2 of
`18 patients) for mismatch repair–proficient colorectal cancers. The median progres-
`sion-free survival and overall survival were not reached in the cohort with mismatch
`repair–deficient colorectal cancer but were 2.2 and 5.0 months, respectively, in the
`cohort with mismatch repair–proficient colorectal cancer (hazard ratio for disease
`progression or death, 0.10 [P<0.001], and hazard ratio for death, 0.22 [P = 0.05]). Pa-
`tients with mismatch repair–deficient noncolorectal cancer had responses similar to
`those of patients with mismatch repair–deficient colorectal cancer (immune-related
`objective response rate, 71% [5 of 7 patients]; immune-related progression-free sur-
`vival rate, 67% [4 of 6 patients]). Whole-exome sequencing revealed a mean of 1782
`somatic mutations per tumor in mismatch repair–deficient tumors, as compared with
`73 in mismatch repair–proficient tumors (P = 0.007), and high somatic mutation loads
`were associated with prolonged progression-free survival (P = 0.02).
`CONCLUSIONS
`This study showed that mismatch-repair status predicted clinical benefit of immune
`checkpoint blockade with pembrolizumab. (Funded by Johns Hopkins University and
`others; ClinicalTrials.gov number, NCT01876511.)
`
`The authors’ full names, academic de-
`grees, and affiliations are listed in the Ap-
`pendix. Address reprint requests to Dr.
`Diaz at the Johns Hopkins Sidney Kim-
`mel Comprehensive Cancer Center, 1650
`Orleans St., Rm. 590, Baltimore, MD
`21287, or at ldiaz1@ jhmi . edu.
`
`This article was published on May 30, 2015,
`at NEJM.org.
`
`N Engl J Med 2015;372:2509-20.
`DOI: 10.1056/NEJMoa1500596
`Copyright © 2015 Massachusetts Medical Society.
`
`n engl j med 372;26 nejm.org
`
`june 25, 2015
`
`2509
`
`T h e ne w e ngl a nd jou r na l o f m e dicine
`
`
`
`Genome Ex. 1031
`Page 1 of 12
`
`

`

`T h e ne w e ngl a nd jou r na l o f m e dicine
`
`The programmed death 1 (PD-1) path-
`
`way is a negative feedback system that
`represses Th1 cytotoxic immune respons-
`es and that, if unregulated, can damage the
`host.1-3 It is up-regulated in many tumors and in
`their surrounding microenvironment. Blockade
`of this pathway with antibodies to PD-1 or its
`ligands has led to remarkable clinical responses
`in patients with many different types of cancer,
`including melanomas, non–small-cell lung can-
`cer, renal-cell carcinoma, bladder cancer, and
`Hodgkin’s lymphoma.4-10 The expression of PD-1
`ligands (PD-L1 or PD-L2) on the surface of tu-
`mor cells or immune cells is an important — but
`not a definitive — predictive biomarker of re-
`sponse to PD-1 blockade.4,6-8,11
`In reports of the effects of PD-1 blockade in
`human tumors, only 1 of 33 patients with
`colorectal cancer had a response to this treat-
`ment, in contrast to substantial fractions of pa-
`tients with melanomas, renal-cell cancers, and
`lung tumors who have a response.10,12 What was
`different about this single patient? We hypothe-
`sized that this patient had mismatch-repair defi-
`ciency, because mismatch-repair deficiency oc-
`curs in a small fraction of advanced colorectal
`cancers,13,14 somatic mutations found in tumors
`can be recognized by the patient’s own immune
`system,15 and mismatch repair–deficient colorec-
`tal cancers have 10 to 100 times as many so-
`matic mutations as mismatch repair–proficient
`colorectal cancers.16-18 Moreover, mismatch re-
`pair–deficient cancers contain prominent lym-
`phocyte infiltrates, a finding consistent with an
`immune response.19-22 In addition, two of the
`tumor types that were most responsive to PD-1
`blockade in a study by Topalian et al.10 had high
`numbers of somatic mutations as a result of
`exposure to cigarette smoke (lung cancers) or
`ultraviolet radiation (melanomas).23,24 Our hy-
`pothesis was correct: the tumor of the single
`patient with colorectal cancer who had a response
`to PD-1 blockade was mismatch repair–defi-
`cient.25 Therefore, we hypothesized that mis-
`match repair–deficient tumors are more re-
`sponsive to PD-1 blockade than are mismatch
`repair–proficient tumors.
`To test this hypothesis, we initiated a phase 2
`clinical trial to evaluate immune checkpoint
`blockade in patients whose tumors had or did
`not have mismatch-repair deficiency. Because
`mismatch-repair deficiency in tumors arises
`
`through two routes,26-28 we recruited patients
`with hereditary nonpolyposis colorectal cancer
`(also known as the Lynch syndrome), which re-
`sults from an inherited germline defect in one of
`four mismatch-repair genes followed by a sec-
`ond inactivating somatic change in the remain-
`ing wild-type allele. We also recruited patients
`with sporadic mismatch repair–deficient tu-
`mors, in which both alleles of a mismatch-repair
`gene are inactivated by somatic mutations or by
`epigenetic silencing.29 In either case, the neo-
`plasms that arise harbor hundreds or thousands
`of mutations.16,18
`
`Methods
`
`Patients
`Patients with treatment-refractory progressive
`metastatic cancer were recruited from three cen-
`ters for this phase 2 study (Table 1). Three co-
`horts were evaluated: cohort A included patients
`with mismatch repair–deficient colorectal ade-
`nocarcinomas, cohort B included patients with
`mismatch repair–proficient colorectal adenocar-
`cinomas, and cohort C included patients with
`mismatch repair–deficient cancers of types oth-
`er than colorectal.
`
`Study Oversight
`The protocol, available with the full text of this
`article at NEJM.org, was approved by the institu-
`tional review board at each site, and the study
`was conducted in accordance with the provisions
`of the Declaration of Helsinki and the Interna-
`tional Conference on Harmonisation Good Clin-
`ical Practice guidelines. All the patients provided
`written informed consent before study entry.
`The first author (the principal investigator) and
`the last author (the Investigational New Drug
`sponsor) were responsible for oversight of the
`study. Merck donated the study drug and re-
`viewed the final drafts of the protocol and of
`this manuscript before submission; they did not
`participate in the analysis of the data.
`
`Study Design
`This phase 2 trial was conducted with the use of
`a Green–Dahlberg two-stage design and includ-
`ed the three parallel cohorts described above.
`The study agent, pembrolizumab, was adminis-
`tered intravenously at a dose of 10 mg per kilo-
`gram of body weight every 14 days (Fig. S1 in
`
`2510
`
`n engl j med 372;26 nejm.org
`
`june 25, 2015
`
`
`
`Genome Ex. 1031
`Page 2 of 12
`
`

`

`PD-1 Blockade in Mismatch-Repair Deficiency
`
`Table 1. Demographic and Baseline Characteristics of the Patients.*
`
`Characteristic
`Median age (range) — yr
`Sex — no. (%)
`Female
`Male
`Race — no. (%)‡
`White
`Black
`Other
`ECOG performance status — no. (%)§
`0
`1
`Cancer type — no. (%)
`Colon
`Rectal
`Ampullary or cholangiocarcinoma
`Endometrial
`Small bowel
`Gastric
`Histologic grade — no. (%)
`Well or moderately differentiated
`Poorly differentiated
`Other
`Stage IV cancer — no. (%)
`Liver metastases — no. (%)
`Median time since initial diagnosis (range) — mo
`Previous therapies — no. (%)
`1
`2
`3
`>4
`Detected germline mutation or known Lynch
`syndrome — no. (%)
`
`Yes
`No
`Unknown
`BRAF wild type — no. (%)
`Yes
`No
`Unknown
`KRAS wild type — no. (%)
`Yes
`No
`Unknown
`
`Mismatch
`Repair–Deficient
`Colorectal Cancer
`(N = 11)
`46 (24–65)
`
`Mismatch
`Repair–Proficient
`Colorectal Cancer
`(N = 21)
`61 (32–79)
`
`Mismatch
`Repair–Deficient
`Noncolorectal Cancer
`(N = 9)
`57 (34–92)
`
`5 (45)
`6 (55)
`
`8 (73)
`1 (9)
`2 (18)
`
`0
`11 (100)
`
`9 (82)
`2 (18)
`0
`0
`0
`0
`
`7 (64)
`4 (36)
`0
`11(100)
`6 (55)
`31 (6–95)
`
`0
`3 (27)
`3 (27)
`5 (45)
`
`9 (82)
`2 (18)
`0
`
`8 (73)
`0
`3 (27)
`
`6 (55)
`5 (45)
`0
`
`8 (38)
`13 (62)
`
`17 (81)
`3 (14)
`1 (5)
`
`6 (29)
`15 (71)
`
`18 (86)
`3 (14)
`NA
`NA
`NA
`NA
`
`4 (44)
`5 (56)
`
`8 (89)
`0
`1 (11)
`
`2 (22)
`7 (78)
`
`0
`0
`4 (44)
`2 (22)
`2 (22)
`1 (11)
`
`18 (86)
`3 (14)
`0
`21 (100)
`11 (52)
`58 (27–192)
`
`4 (44)
`3 (33)
`2 (22)
`9 (100)
`6 (67)
`23 (2–105)
`
`0
`4 (19)
`5 (24)
`12 (57)
`
`0
`21 (100)
`0
`
`11 (52)
`1 (5)
`9 (43)
`
`13 (62)
`8 (38)
`0
`
`1 (11)
`5 (56)
`1 (11)
`2 (22)
`
`4 (44)
`4 (44)
`1 (11)
`
`4 (44)
`0
`5 (56)
`
`4 (44)
`1 (11)
`4 (44)
`
`P Value†
`0.02
`0.72
`
`0.66
`
`0.07
`
`>0.99
`
`0.20
`
`>0.99
`>0.99
`0.07
`0.89
`
`<0.001
`
`0.64
`
`0.72
`
`* NA denotes not applicable.
`† P values are for the comparison between the cohort with mismatch repair–deficient colorectal cancer and the cohort with mismatch repair–
`proficient colorectal cancer.
`‡ Race was self-reported.
`§ Eastern Cooperative Oncology Group (ECOG) performance status is a measure of a patient’s ability to perform activities of daily living; val-
`ues range from 0 to 5, with higher scores indicating greater impairment.
`
`n engl j med 372;26 nejm.org
`
`june 25, 2015
`
`2511
`
`
`
`Genome Ex. 1031
`Page 3 of 12
`
`

`

`T h e ne w e ngl a nd jou r na l o f m e dicine
`
`Supplementary Appendix 1, available at NEJM.org).
`Pembrolizumab is a humanized monoclonal
`anti–PD-1 antibody of the IgG4 kappa isotype
`that blocks the interaction between PD-1 and its
`ligands, PD-L1 and PD-L2 (Fig. S1 in Supplemen-
`tary Appendix 1).
`Safety assessments were performed before
`each treatment. At the start of each treatment
`cycle, the total tumor burden was assessed by
`means of measurement of serum biomarkers.
`Radiographic assessments were performed at 12
`weeks and every 8 weeks thereafter. Further de-
`tails concerning the clinical protocol are avail-
`able at NEJM.org.
`
`Analysis of Mismatch-Repair Status
`Tumors with genetic defects in mismatch-repair
`pathways are known to harbor hundreds to
`thousands of somatic mutations, especially in
`regions of repetitive DNA known as microsatel-
`lites. The accumulation of mutations in these
`regions of the genome is termed microsatellite
`instability.26-28 Mismatch-repair status was as-
`sessed in tumors with the use of the MSI Analy-
`sis System (Promega), through the evaluation of
`selected microsatellite sequences that are par-
`ticularly prone to copying errors when mismatch
`repair is compromised.26-28 Additional details are
`provided in Supplementary Appendix 1.
`
`Genomic and Bioinformatic Analyses
`Primary tumor samples and matched normal
`peripheral-blood specimens were obtained from
`a subgroup of patients with mismatch repair–
`deficient carcinomas and a subgroup with mis-
`match repair–proficient carcinomas, for whom
`sufficient tumor tissue was available for exome
`sequencing30 and HLA haplotyping. To assess
`the potential for mutant peptide binding, so-
`matic exome data combined with each individu-
`al patient’s major histocompatibility complex
`(MHC) class I HLA haplotype were applied to an
`epitope prediction algorithm.31,32 This algorithm
`provided an estimate of the total number of
`mutation-associated neoantigens in each tumor.
`Additional details are provided in Supplementa-
`ry Appendix 1.
`
`Statistical Analysis
`The primary end points for cohorts A and B were
`the immune-related objective response rate and
`the 20-week immune-related progression-free
`
`survival rate, assessed with the use of immune-
`related response criteria.33 The primary end
`point for cohort C was the immune-related pro-
`gression-free survival rate at 20 weeks (Fig. S1 in
`Supplementary Appendix 1). Immune-related cri-
`teria (i.e., one of the types of criteria used to
`evaluate immune-based therapies) are based on
`radiographic responses, and unlike Response
`Evaluation Criteria in Solid Tumors (RECIST),
`they capture newly developed lesions detected on
`radiography in the measurement of tumor bur-
`den; these criteria are defined and compared
`with RECIST, version 1.1, in Table S1 in Supple-
`mentary Appendix 1. The response rate and 20-
`week progression-free survival rate were evalu-
`ated and reported in this study with the use of
`both RECIST, version 1.1, and immune-related
`response criteria. Progression-free survival and
`overall survival were summarized by means of
`the Kaplan–Meier method. Details of the hy-
`pothesis, the decision rules for the rejection of
`the null hypotheses, decision rules for early
`discontinuation of the study in a cohort because
`of efficacy or futility, and statistical methods are
`provided in Supplementary Appendix 1.
`
`R esults
`
`Patients
`A total of 41 consecutive patients were enrolled
`in the study and treated during the period from
`September 2013 through January 2015 (Table 1).
`Recruitment included patients in pursuit of a
`clinical trial option who were known to have
`tumors with mismatch-repair defects or who
`had tumors of unknown status who were then
`tested. One patient in the cohort with mismatch
`repair–deficient colorectal cancer was enrolled
`under an institutional review board eligibility
`waiver allowing a grade 3 bilirubin level (i.e.,
`higher than the cutoff specified in the inclusion
`criteria). A total of 32 patients with colorectal
`cancer were enrolled in cohorts A and B. All
`patients with colorectal cancer had received two
`or more previous chemotherapy regimens (a me-
`dian of four regimens), except for 1 patient with
`mismatch repair–proficient cancer who had
`received one chemotherapeutic and one (non–
`PD-1–based) immunotherapeutic regimen.
`Nine patients with mismatch repair–deficient
`solid tumors other than colorectal cancer were
`enrolled in cohort C. All patients in cohort C had
`
`2512
`
`n engl j med 372;26 nejm.org
`
`june 25, 2015
`
`
`
`Genome Ex. 1031
`Page 4 of 12
`
`

`

`PD-1 Blockade in Mismatch-Repair Deficiency
`
`Table 2. Objective Responses According to RECIST Criteria.
`
`Type of Response
`
`Complete response — no. (%)
`
`Partial response — no. (%)
`
`Stable disease at week 12 — no. (%)
`
`Progressive disease — no. (%)
`
`Could not be evaluated — no. (%)‡
`
`Objective response rate (95% CI) — %
`
`Disease control rate (95% CI) — %§
`
`Median duration of response — wk
`
`Median time to response (range) — wk
`
`Mismatch
`Repair–Deficient
`Colorectal Cancer
` (N = 10)
`
`Mismatch
`Repair–Proficient
`Colorectal Cancer
`(N = 18)
`
`Mismatch
`Repair–Deficient
`Noncolorectal Cancer
`(N = 7)
`
`0
`
`4 (40)
`
`5 (50)
`
`1 (10)
`
`0
`
`40 (12–74)
`
`90 (55–100)
`
`Not reached
`
`28 (13–35)
`
`0
`
`0
`
`2 (11)
`
`11 (61)
`
`5 (28)
`
`0 (0–19)
`
`11 (1–35)
`
`NA¶
`
`NA¶
`
`1 (14)*
`
`4 (57)†
`
`0
`
`2 (29)
`
`0
`
`71 (29–96)
`
`71 (29–96)
`
`Not reached
`
`12 (10–13)
`
`* The patient had a partial response at 12 weeks, which then became a complete response at 20 weeks.
`† One patient had a partial response at 12 weeks.
`‡ Patients could not be evaluated if they did not undergo a scan at 12 weeks because of clinical progression.
`§ The rate of disease control was defined as the percentage of patients who had a complete response, partial response,
`or stable disease for 12 weeks or more.
`¶ The median time to response was not applicable (NA) because no responses were observed among patients with mis-
`match repair–proficient colorectal cancer.
`
`received one or more previous therapeutic regi-
`mens (a median of two regimens).
`
`Primary End Point
`The immune-related objective response rate in
`cohort A was 40% (4 of 10 patients; 95% confi-
`dence interval [CI], 12 to 74), and the immune-
`related progression-free survival rate at 20 weeks
`was 78% (7 of 9 patients; 95% CI, 40 to 97)
`(Table S2 in Supplementary Appendix 1); the
`corresponding rates in cohort C were 71% (5 of
`7 patients; 95% CI, 29 to 96) and 67% (4 of 6
`patients; 95% CI, 22 to 96). In cohort B, which
`included patients with mismatch repair–profi-
`cient colorectal cancers, the immune-related
`objective response rate was 0% (95% CI, 0 to 20),
`and the immune-related progression-free sur-
`vival rate at 20 weeks was 11% (2 of 18 patients;
`95% CI, 1 to 35). Both cohorts with mismatch
`repair–deficient cancers (cohorts A and C)
`reached the prespecified point at which the pro-
`tocol indicated that the study reached its pri-
`mary efficacy end point when 4 patients were
`free from disease progression at 20 weeks and
`objective responses on the basis of immune-re-
`lated response criteria were observed in 4 pa-
`tients (Table S2 and the Methods section in
`Supplementary Appendix 1).
`The median follow-up was 36 weeks (range, 5
`
`to 55) for patients with mismatch repair–deficient
`colorectal cancer (cohort A), 20 weeks (range, 4 to
`52) for patients with mismatch repair–proficient
`colorectal cancer (cohort B), and 21 weeks (range,
`0.1 to 49) for patients with mismatch repair–
`deficient noncolorectal cancer (cohort C). All pa-
`tients for whom the 20-week immune-related
`progression-free survival rate could be evaluated
`were followed for at least 20 weeks.
`
`Radiographic Evaluation
`Of the 10 patients with mismatch repair–defi-
`cient colorectal cancer (cohort A) who could be
`evaluated for RECIST, 4 (40%; 95% CI, 12 to 74)
`had objective responses according to these crite-
`ria (Table 2 and Fig. 1, and Fig. S2 in Supple-
`mentary Appendix 1). Patients were considered
`not to have been evaluated unless they under-
`went a radiographic scan at 12 weeks. The rate
`of disease control, which was defined as the
`percentage of patients who had an objective re-
`sponse or whose disease was stable, was 90% in
`cohort A (9 of 10 patients; 95% CI, 55 to 100).
`Of the 7 patients in cohort C who could be
`evaluated, 5 (71%; 95% CI, 29 to 96) had objec-
`tive responses as defined by RECIST (Table 2
`and Fig. 1, and Fig. S2 in Supplementary Appen-
`dix 1), and the rate of disease control was 71%
`(5 of 7 patients; 95% CI, 29 to 96).
`
`n engl j med 372;26 nejm.org
`
`june 25, 2015
`
`2513
`
`
`
`Genome Ex. 1031
`Page 5 of 12
`
`

`

`T h e ne w e ngl a nd jou r na l o f m e dicine
`
`3 of 11 patients (27%) with tumors associated
`with the Lynch syndrome had a response (Table
`S3 in Supplementary Appendix 2) (P = 0.009). No
`other baseline characteristics had a significant
`association with objective responses.
`Among the 18 patients with mismatch re-
`pair–proficient colorectal cancers in cohort B,
`no objective responses as defined by RECIST
`were observed (Table 2 and Fig. 1, and Fig. S2 in
`Supplementary Appendix 1). In this group, the
`rate of disease control was 11% (2 of 18 patients;
`95% CI, 1 to 35).
`All the patients who had a response as de-
`fined by RECIST (Table 2) also had a response
`according to immune-related response criteria
`(Table S2 in Supplementary Appendix 1).
`
`Survival
`In the cohort of patients with mismatch repair–
`deficient colorectal cancer (cohort A), the medi-
`an progression-free survival and median overall
`survival were not reached (Fig. 2). In contrast,
`among the patients with mismatch repair–profi-
`cient cancers (cohort B), the median progres-
`sion-free survival was only 2.2 months (95% CI,
`1.4 to 2.8), and the median overall survival was
`5.0 months (95% CI, 3.0 to not estimable). In
`cohort C (patients with mismatch repair–defi-
`cient noncolorectal cancer), the median progres-
`sion-free survival was 5.4 months (95% CI, 3 to
`not estimable), and the median overall survival
`was not reached. A post hoc comparison of the
`cohorts with mismatch repair–deficient and
`mismatch repair–proficient colorectal cancers
`showed hazard ratios for disease progression or
`death (0.10; 95% CI, 0.03 to 0.37; P<0.001) and
`for death (0.22; 95% CI, 0.05 to 1.00; P = 0.05)
`that favored patients with mismatch repair–defi-
`cient colorectal cancer (Fig. 2).
`To evaluate whether the difference in survival
`might be due to prognostic differences, we mea-
`sured the time since the diagnosis of metastatic
`disease and the clinical performance of the
`regimen that patients had received before enroll-
`ment. We found that there was no significant
`difference between patients with mismatch re-
`pair–deficient colorectal cancer and patients
`with mismatch repair–proficient colorectal can-
`cer with respect to the duration of metastatic
`disease (P = 0.77 by the log-rank test) or the
`median progression-free survival while receiving
`their previous regimens (P = 0.60 by the log-rank
`
`Mismatch repair–proficient colorectal cancer
`Mismatch repair–deficient colorectal cancer
`Mismatch repair–deficient noncolorectal cancer
`
`0% (no change)
`
`100
`
`200
`
`300
`
`400
`
`Days
`
`A BiochemicalResponse
`200
`
`100
`
`0
`
`(%)
`
`ChangeinTumorMarkerLevel
`
`−100
`
`0
`
`B RadiographicResponse
`100
`
`Mismatch repair–proficient colorectal cancer
`Mismatch repair–deficient colorectal cancer
`Mismatch repair–deficient noncolorectal cancer
`
`20% increase (progressive disease)
`
`30% decrease (partial response)
`
`50
`
`0
`
`−50
`
`−100
`
`ofLongestDiameters(%)
`
`ChangefromBaselineintheSum
`
`Figure 1. Clinical Responses to Pembrolizumab Treatment.
`The biochemical responses to pembrolizumab treatment are shown in Pan-
`el A. Serum levels of protein biomarkers were measured at the start of each
`treatment cycle, and the values represent percentage changes from base-
`line. Each line represents one patient; patients were included if their base-
`line tumor marker values were higher than the upper limit of normal. CA-
`125 was used as the biomarker for one patient with endometrial cancer,
`CA19-9 was used for one patient with cholangiocarcinoma and one patient
`with ampullary cancer, and carcinoembryonic antigen (CEA) was used for
`all other patients. Radiographic responses to treatment with pembrolizum-
`ab, evaluated on the basis of Response Evaluation Criteria in Solid Tumors
`(RECIST), are shown in Panel B. Tumor responses were measured at regu-
`lar intervals, and the values shown are the largest percentage change in the
`sum of longest diameters from the baseline measurements of each mea-
`surable tumor. Each bar represents one patient.
`
`Patients in cohort C had faster responses
`than did patients in cohort A (median time to
`response according to RECIST, 12 weeks vs. 28
`weeks; P = 0.03). Furthermore, all 6 patients
`(100%) with mismatch repair–deficient tumors
`that were not associated with the Lynch syn-
`drome had an objective response, whereas only
`
`2514
`
`n engl j med 372;26 nejm.org
`
`june 25, 2015
`
`
`
`Genome Ex. 1031
`Page 6 of 12
`
`

`

`PD-1 Blockade in Mismatch-Repair Deficiency
`
`A Progression-freeSurvivalinCohortswithColorectalCancer
`1.0
`
`P<0.001 by log-rank test
`
`B OverallSurvivalinCohortswithColorectalCancer
`1.0
`
`P=0.03 by log-rank test
`
`Mismatch repair–deficient
`
`Mismatch repair–proficient
`
`12
`
`15
`
`0 0
`
`1 1
`
`9
`
`5 1
`
`Months
`
`6
`
`7 5
`
`0
`
`3
`
`11
`
`21
`
`9
`
`12
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`ProbabilityofOverallSurvival
`
`No.atRisk
`Mismatch repair–
`deficient
`Mismatch repair–
`proficient
`
`Mismatch repair–deficient
`
`Mismatch repair–proficient
`
`12
`
`15
`
`0 0
`
`0 0
`
`9
`
`2 0
`
`Months
`
`6
`
`6 1
`
`3
`
`8 2
`
`0
`
`11
`
`21
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`Survival
`
`ProbabilityofProgression-free
`
`No.atRisk
`Mismatch repair–
`deficient
`Mismatch repair–
`proficient
`
`C Progression-freeSurvivalinCohortwithMismatchRepair–Deficient
`NoncolorectalCancer
`1.0
`
`D OverallSurvivalinCohortwithMismatchRepair–Deficient
`NoncolorectalCancer
`1.0
`
`0
`
`9
`
`3
`
`6
`
`6
`
`2
`
`Months
`
`9
`
`1
`
`12
`
`0
`
`15
`
`0
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`ProbabilityofOverallSurvival
`
`0
`
`9
`
`3
`
`5
`
`6
`
`1
`
`Months
`
`9
`
`0
`
`12
`
`0
`
`15
`
`0
`
`No.atRisk
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`Survival
`
`ProbabilityofProgression-free
`
`No.atRisk
`
`Figure 2. Clinical Benefit of Pembrolizumab Treatment According to Mismatch-Repair Status.
`Kaplan–Meier curves are shown for progression-free survival in the cohorts with colorectal cancer (Panel A), overall survival in the co-
`horts with colorectal cancer (Panel B), progression-free survival among patients with mismatch repair–deficient noncolorectal cancers
`(Panel C), and overall survival among patients with mismatch repair–deficient noncolorectal cancers (Panel D). In both cohorts with
`mismatch repair–deficient tumors, median overall survival was not reached. Patients in the cohort with mismatch repair–proficient can-
`cers had a median progression-free survival of 2.2 months (95% CI, 1.4 to 2.8) and a median overall survival of 5.0 months (95% CI, 3.0
`to not estimable). Patients with mismatch repair–deficient noncolorectal cancers had a median progression-free survival of 5.4 months
`(95% CI, 3 to not estimable).
`
`test) (Fig. S3 in Supplementary Appendix 1). We
`also performed an additional multivariate analy-
`sis of progression-free and overall survival to
`examine the difference in outcomes between
`mismatch repair–deficient colorectal cancer and
`mismatch repair–proficient colorectal cancer,
`adjusting for elapsed time since the initial diag-
`nosis. The magnitude of the hazard ratios for
`disease progression or death (hazard ratio, 0.04;
`95% CI 0.01 to 0.21; P<0.001) and for death
`
`(hazard ratio, 0.18; 95% CI, 0.03 to 1.01; P = 0.05),
`representing the differing effects of pembroli-
`zumab between mismatch repair–deficient tu-
`mors and mismatch repair–proficient tumors,
`was maintained after adjustment for this poten-
`tial difference.
`
`Safety Assessment
`Adverse events occurring in more than 5% of
`patients are listed in Table 3. Events of clinical
`
`n engl j med 372;26 nejm.org
`
`june 25, 2015
`
`2515
`
`
`
`Genome Ex. 1031
`Page 7 of 12
`
`

`

`T h e ne w e ngl a nd jou r na l o f m e dicine
`
`interest included rash or pruritus (24%); thyroid-
`itis, hypothyroidism, or hypophysitis (10%); and
`asymptomatic pancreatitis (15%). Although the
`numbers were small, thyroid-function abnor-
`malities were limited to the cohorts with mis-
`match repair–deficient cancer (Table 3).
`
`Tumor Markers
`In the two cohorts with colorectal cancer, it was
`possible to evaluate levels of carcinoembryonic
`antigen (CEA) before enrollment; in 29 of 32
`patients, these levels were above the upper limit
`of normal (3 mg per deciliter). Substantial de-
`creases in CEA level occurred in 7 of the 10 pa-
`tients with mismatch repair–deficient colorectal
`cancer and in none of the 19 patients with mis-
`match repair–proficient colorectal cancer in whom
`CEA could be evaluated (Fig. 1, and Fig. S4 in
`Supplementary Appendix 1). Among patients
`with mismatch repair–deficient noncolorectal
`cancer, levels of tumor markers (CEA, CA19-9,
`or CA-125) were elevated above the upper limit
`of normal in 4 patients. Declines in CA19-9 or
`CA-125 of more than 70% occurred in 3 of these
`4 patients. Tumor marker kinetics in all three
`cohorts are shown in Figure 1. The degree of
`CEA decline after one dose (between day 14 and
`day 28) of pembrolizumab was predictive of
`both progression-free survival (P = 0.01) and
`overall survival (P = 0.02). The CEA response oc-
`curred well in advance of radiographic confirma-
`tion of disease control (range, 10 to 35 weeks). In
`contrast, patients who had disease progression
`had rapid biomarker elevation within 30 days
`after the initiation of therapy. Thus, changes in
`CEA levels significantly preceded and correlated
`with ultimate radiographic changes.
`
`Genomic Analysis
`The analysis of whole-exome sequences showed
`a mean of 1782 somatic mutations per tumor in
`patients with mismatch repair–deficient cancer
`(nine patients), as compared with 73 mutations
`per tumor in patients with mismatch repair–
`proficient cancer (six patients) (P = 0.007 by
`nonparametric Wilcoxon test) (Fig. S5 in Sup-
`plementary Appendix 1 and Table S3 in Supple-
`mentary Appendix 2). Most of these mutations
`(63%) are predicted to alter amino acids.
`These mutations were then assessed for their
`immunogenic potential in the context of each
`patient’s MHC haplotype. We identified a mean
`of 578 potential mutation-associated neoanti-
`
`gens from the tumors of patients with mismatch
`repair–deficient cancers; 21 such neoantigens
`were identified in tumors from patients with
`mismatch repair–proficient cancers (Table S3 in
`Supplementary Appendix 2). The percentage of
`potential mutation-associated neoantigens among
`all somatic mutations was similar in the two
`cohorts (a mean of 32% in patients with mis-
`match repair–deficient cancer and 29% in pa-
`tients with mismatch repair–proficient cancer).
`High numbers of somatic mutations and poten-
`tial mutation-associated neoantigens were asso-
`ciated with longer progression-free survival and
`with a trend toward objective response (Fig. S5
`and Table S4 in Supplementary Appendix 1).
`
`Immunohistochemical Analysis
`The expression of CD8 and PD-L1 was evaluated
`within the tumor and at the invasive fronts of
`the tumor in an immunohistochemical analysis
`in the 30 cases in which tumor tissue was avail-
`able (Fig. S6 in Supplementary Appendix 1). Tu-
`mors from patients in cohorts A and C con-
`tained a greater density of CD8-positive lymphoid
`cells than did tumors from patients in cohort B
`(P = 0.10) (Fig. S7 in Supplementary Appendix 1),
`and CD8 labeling was associated with a trend
`toward objective response and stable disease
`(Fig. S8 and Table S5 in Supplementary Appen-
`dix 1). This CD8-positive lymphoid infiltrate was
`especially prominent at the invasive fronts of the
`tumors (P = 0.04) (Fig. S7 in Supplementary Ap-
`pendix 1). Membranous PD-L1 expression occurred
`only in patients with mismatch repair–deficient
`cancer and was prominent on tumor-infiltrating
`lymphocytes and tumor-associated macrophages
`located at the invasive fronts of the tumor
`(P = 0.04) (Fig. S7 in Supplementary Appendix 1).
`The expression of CD8 and PD-L1 was not sig-
`nificantly associated with progression-free sur-
`vival or overall survival (Table S5 in Supplemen-
`tary Appendix 1).
`
`Discussion
`
`The data from this small phase 2 trial of pem-
`brolizumab for the treatment of tumors with
`and tumors without mismatch-repair deficiency
`support the hypothesis that mismatch repair–
`deficient tumors are more responsive to PD-1
`blockade than are mismatch repair–proficient
`tumors. Mismatch-repair deficiency occurs in
`many cancers, including those of the colorec-
`
`2516
`
`n engl j med 372;26 nejm.org
`
`june 25, 2015
`
`
`
`Genome Ex. 1031
`Page 8 of 12
`
`

`

`PD-1 Blockade in Mismatch-Repair Deficiency
`
`tum, uterus, stomach, biliary tract, pancreas,
`ovary, prostate, and small intestine.18,34-42 It is
`possible that patients with mismatch repair–
`deficient tumors of these types may also bene-
`fit from anti–PD-1 therapy, as may patients
`whose tumors contain other DNA repair defi-
`ciencies, such as those with mutations in POLD,
`POLE, or MYH.18,43,44
`The hypothesis that mismatch repair–defi-
`cient tumors stimulate the immune system is
`not a new idea45; it has been supported by obser-
`vations of the dense immune infiltration and
`Th1-associated cytokine-rich environment in
`mismatch repair–deficient tumors.19-22,46 A recent
`study refined these classic observations by show-
`ing that the mismatch repair–deficient tumor
`microenvironment strongly expressed several
`immune checkpoint ligands, including PD-1, PD-
`L1, CTLA-4, LAG-3, and IDO, which indicates
`that their active immune microenvironment is
`counterbalanced by immune inhibitory signals
`that resist tumor elimination.47 The most likely
`explanation for both the old and new findings
`was that the immune infiltrate associated with
`mismatch repair–deficient carcinomas was di-
`rected at neoantigens. The correlation of a
`higher mutational load and a higher rate of re-
`sponse to anti–CTLA-4 in melanoma41 and anti–
`PD-1 in lung cancer48 provides further support
`for the idea that mutation-associated neoantigen
`recognition is an important compo

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