scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer

TL;DR: Treatment efficacy was associated with a higher number of mutations in the tumors, and a tumor-specific T cell response paralleled tumor regression in one patient, suggesting that the genomic landscape of lung cancers shapes response to anti–PD-1 therapy.
Abstract: Immune checkpoint inhibitors, which unleash a patient’s own T cells to kill tumors, are revolutionizing cancer treatment. To unravel the genomic determinants of response to this therapy, we used whole-exome sequencing of non–small cell lung cancers treated with pembrolizumab, an antibody targeting programmed cell death-1 (PD-1). In two independent cohorts, higher nonsynonymous mutation burden in tumors was associated with improved objective response, durable clinical benefit, and progression-free survival. Efficacy also correlated with the molecular smoking signature, higher neoantigen burden, and DNA repair pathway mutations; each factor was also associated with mutation burden. In one responder, neoantigen-specific CD8+ T cell responses paralleled tumor regression, suggesting that anti–PD-1 therapy enhances neoantigen-specific T cell reactivity. Our results suggest that the genomic landscape of lung cancers shapes response to anti–PD-1 therapy.
Citations
More filters
Journal ArticleDOI
TL;DR: Nivolumab was associated with even greater efficacy than docetaxel across all end points in subgroups defined according to prespecified levels of tumor-membrane expression (≥1, ≥5%, and ≥10%) of the PD-1 ligand.
Abstract: BackgroundNivolumab, a fully human IgG4 programmed death 1 (PD-1) immune-checkpoint–inhibitor antibody, disrupts PD-1–mediated signaling and may restore antitumor immunity. MethodsIn this randomized, open-label, international phase 3 study, we assigned patients with nonsquamous non–small-cell lung cancer (NSCLC) that had progressed during or after platinum-based doublet chemotherapy to receive nivolumab at a dose of 3 mg per kilogram of body weight every 2 weeks or docetaxel at a dose of 75 mg per square meter of body-surface area every 3 weeks. The primary end point was overall survival. ResultsOverall survival was longer with nivolumab than with docetaxel. The median overall survival was 12.2 months (95% confidence interval [CI], 9.7 to 15.0) among 292 patients in the nivolumab group and 9.4 months (95% CI, 8.1 to 10.7) among 290 patients in the docetaxel group (hazard ratio for death, 0.73; 96% CI, 0.59 to 0.89; P=0.002). At 1 year, the overall survival rate was 51% (95% CI, 45 to 56) with nivolumab ve...

7,474 citations

Journal ArticleDOI
TL;DR: Among patients with advanced, previously treated squamous-cell NSCLC, overall survival, response rate, and progression-free survival were significantly better with nivolumab than with docetaxel, regardless of PD-L1 expression level.
Abstract: BackgroundPatients with advanced squamous-cell non–small-cell lung cancer (NSCLC) who have disease progression during or after first-line chemotherapy have limited treatment options. This randomized, open-label, international, phase 3 study evaluated the efficacy and safety of nivolumab, a fully human IgG4 programmed death 1 (PD-1) immune-checkpoint–inhibitor antibody, as compared with docetaxel in this patient population. MethodsWe randomly assigned 272 patients to receive nivolumab, at a dose of 3 mg per kilogram of body weight every 2 weeks, or docetaxel, at a dose of 75 mg per square meter of body-surface area every 3 weeks. The primary end point was overall survival. ResultsThe median overall survival was 9.2 months (95% confidence interval [CI], 7.3 to 13.3) with nivolumab versus 6.0 months (95% CI, 5.1 to 7.3) with docetaxel. The risk of death was 41% lower with nivolumab than with docetaxel (hazard ratio, 0.59; 95% CI, 0.44 to 0.79; P<0.001). At 1 year, the overall survival rate was 42% (95% CI, 3...

6,869 citations

Journal ArticleDOI
TL;DR: This study showed that mismatch-repair status predicted clinical benefit of immune checkpoint blockade with pembrolizumab, and high somatic mutation loads were associated with prolonged progression-free survival.
Abstract: BackgroundSomatic 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. MethodsWe 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. Pembrolizumab 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 mismatch 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. ResultsThe immune-related objective response rate and immune-related progression-free survival ...

6,835 citations

Journal ArticleDOI
TL;DR: This review summarizes the clinical efficacy, perspectives, and future challenges of using PD-1/PD-L1-directed antibodies in the treatment of breast cancer.
Abstract: Immune checkpoint inhibition represents a major recent breakthrough in the treatment of malignant diseases including breast cancer. Blocking the programmed death receptor-1 (PD-1) and its ligand, PD-L1, has shown impressive antitumor activity and may lead to durable long-term disease control, especially in the triple-negative subtypes of breast cancer (TNBC). Although immune checkpoint blockade is generally well tolerated, specific immune-related adverse events (irAEs) may occur. This review summarizes the clinical efficacy, perspectives, and future challenges of using PD-1/PD-L1-directed antibodies in the treatment of breast cancer.

5,777 citations

Journal ArticleDOI
TL;DR: Pembrolizumab had an acceptable side-effect profile and showed antitumor activity in patients with advanced non-small-cell lung cancer and PD-L1 expression in at least 50% of tumor cells correlated with improved efficacy of pembrolIZumab.
Abstract: BackgroundWe assessed the efficacy and safety of programmed cell death 1 (PD-1) inhibition with pembrolizumab in patients with advanced non–small-cell lung cancer enrolled in a phase 1 study. We also sought to define and validate an expression level of the PD-1 ligand 1 (PD-L1) that is associated with the likelihood of clinical benefit. MethodsWe assigned 495 patients receiving pembrolizumab (at a dose of either 2 mg or 10 mg per kilogram of body weight every 3 weeks or 10 mg per kilogram every 2 weeks) to either a training group (182 patients) or a validation group (313 patients). We assessed PD-L1 expression in tumor samples using immunohistochemical analysis, with results reported as the percentage of neoplastic cells with staining for membranous PD-L1 (proportion score). Response was assessed every 9 weeks by central review. ResultsCommon side effects that were attributed to pembrolizumab were fatigue, pruritus, and decreased appetite, with no clear difference according to dose or schedule. Among all ...

4,834 citations

References
More filters
Journal ArticleDOI
25 Mar 2011-Science
TL;DR: A unifying conceptual framework called “cancer immunoediting,” which integrates the immune system’s dual host-protective and tumor-promoting roles is discussed.
Abstract: Understanding how the immune system affects cancer development and progression has been one of the most challenging questions in immunology. Research over the past two decades has helped explain why the answer to this question has evaded us for so long. We now appreciate that the immune system plays a dual role in cancer: It can not only suppress tumor growth by destroying cancer cells or inhibiting their outgrowth but also promote tumor progression either by selecting for tumor cells that are more fit to survive in an immunocompetent host or by establishing conditions within the tumor microenvironment that facilitate tumor outgrowth. Here, we discuss a unifying conceptual framework called "cancer immunoediting," which integrates the immune system's dual host-protective and tumor-promoting roles.

5,026 citations

Journal ArticleDOI
Michael S. Lawrence1, Petar Stojanov1, Petar Stojanov2, Paz Polak3, Paz Polak1, Paz Polak2, Gregory V. Kryukov3, Gregory V. Kryukov1, Gregory V. Kryukov2, Kristian Cibulskis1, Andrey Sivachenko1, Scott L. Carter1, Chip Stewart1, Craig H. Mermel2, Craig H. Mermel1, Steven A. Roberts4, Adam Kiezun1, Peter S. Hammerman2, Peter S. Hammerman1, Aaron McKenna5, Aaron McKenna1, Yotam Drier, Lihua Zou1, Alex H. Ramos1, Trevor J. Pugh1, Trevor J. Pugh2, Nicolas Stransky1, Elena Helman1, Elena Helman6, Jaegil Kim1, Carrie Sougnez1, Lauren Ambrogio1, Elizabeth Nickerson1, Erica Shefler1, Maria L. Cortes1, Daniel Auclair1, Gordon Saksena1, Douglas Voet1, Michael S. Noble1, Daniel DiCara1, Pei Lin1, Lee Lichtenstein1, David I. Heiman1, Timothy Fennell1, Marcin Imielinski1, Marcin Imielinski2, Bryan Hernandez1, Eran Hodis2, Eran Hodis1, Sylvan C. Baca1, Sylvan C. Baca2, Austin M. Dulak2, Austin M. Dulak1, Jens G. Lohr1, Jens G. Lohr2, Dan A. Landau2, Dan A. Landau7, Dan A. Landau1, Catherine J. Wu2, Jorge Melendez-Zajgla, Alfredo Hidalgo-Miranda, Amnon Koren2, Amnon Koren1, Steven A. McCarroll2, Steven A. McCarroll1, Jaume Mora8, Ryan S. Lee9, Ryan S. Lee2, Brian D. Crompton2, Brian D. Crompton9, Robert C. Onofrio1, Melissa Parkin1, Wendy Winckler1, Kristin G. Ardlie1, Stacey Gabriel1, Charles W. M. Roberts9, Charles W. M. Roberts2, Jaclyn A. Biegel10, Kimberly Stegmaier2, Kimberly Stegmaier1, Kimberly Stegmaier9, Adam J. Bass1, Adam J. Bass2, Levi A. Garraway1, Levi A. Garraway2, Matthew Meyerson2, Matthew Meyerson1, Todd R. Golub, Dmitry A. Gordenin4, Shamil R. Sunyaev2, Shamil R. Sunyaev3, Shamil R. Sunyaev1, Eric S. Lander1, Eric S. Lander2, Eric S. Lander6, Gad Getz1, Gad Getz2 
11 Jul 2013-Nature
TL;DR: A fundamental problem with cancer genome studies is described: as the sample size increases, the list of putatively significant genes produced by current analytical methods burgeons into the hundreds and the list includes many implausible genes, suggesting extensive false-positive findings that overshadow true driver events.
Abstract: Major international projects are underway that are aimed at creating a comprehensive catalogue of all the genes responsible for the initiation and progression of cancer. These studies involve the sequencing of matched tumour-normal samples followed by mathematical analysis to identify those genes in which mutations occur more frequently than expected by random chance. Here we describe a fundamental problem with cancer genome studies: as the sample size increases, the list of putatively significant genes produced by current analytical methods burgeons into the hundreds. The list includes many implausible genes (such as those encoding olfactory receptors and the muscle protein titin), suggesting extensive false-positive findings that overshadow true driver events. We show that this problem stems largely from mutational heterogeneity and provide a novel analytical methodology, MutSigCV, for resolving the problem. We apply MutSigCV to exome sequences from 3,083 tumour-normal pairs and discover extraordinary variation in mutation frequency and spectrum within cancer types, which sheds light on mutational processes and disease aetiology, and in mutation frequency across the genome, which is strongly correlated with DNA replication timing and also with transcriptional activity. By incorporating mutational heterogeneity into the analyses, MutSigCV is able to eliminate most of the apparent artefactual findings and enable the identification of genes truly associated with cancer.

4,411 citations

Journal ArticleDOI
27 Nov 2014-Nature
TL;DR: Evaluated data suggest that MPDL3280A is most effective in patients in which pre-existing immunity is suppressed by PD-L1, and is re-invigorated on antibody treatment, as well as across multiple cancer types.
Abstract: The development of human cancer is a multistep process characterized by the accumulation of genetic and epigenetic alterations that drive or reflect tumour progression. These changes distinguish cancer cells from their normal counterparts, allowing tumours to be recognized as foreign by the immune system. However, tumours are rarely rejected spontaneously, reflecting their ability to maintain an immunosuppressive microenvironment. Programmed death-ligand 1 (PD-L1; also called B7-H1 or CD274), which is expressed on many cancer and immune cells, plays an important part in blocking the 'cancer immunity cycle' by binding programmed death-1 (PD-1) and B7.1 (CD80), both of which are negative regulators of T-lymphocyte activation. Binding of PD-L1 to its receptors suppresses T-cell migration, proliferation and secretion of cytotoxic mediators, and restricts tumour cell killing. The PD-L1-PD-1 axis protects the host from overactive T-effector cells not only in cancer but also during microbial infections. Blocking PD-L1 should therefore enhance anticancer immunity, but little is known about predictive factors of efficacy. This study was designed to evaluate the safety, activity and biomarkers of PD-L1 inhibition using the engineered humanized antibody MPDL3280A. Here we show that across multiple cancer types, responses (as evaluated by Response Evaluation Criteria in Solid Tumours, version 1.1) were observed in patients with tumours expressing high levels of PD-L1, especially when PD-L1 was expressed by tumour-infiltrating immune cells. Furthermore, responses were associated with T-helper type 1 (TH1) gene expression, CTLA4 expression and the absence of fractalkine (CX3CL1) in baseline tumour specimens. Together, these data suggest that MPDL3280A is most effective in patients in which pre-existing immunity is suppressed by PD-L1, and is re-invigorated on antibody treatment.

4,227 citations

Journal ArticleDOI
01 Jan 2014-Nature
TL;DR: In this paper, the authors report molecular profiling of 230 resected lung adnocarcinomas using messenger RNA, microRNA and DNA sequencing integrated with copy number, methylation and proteomic analyses.
Abstract: Adenocarcinoma of the lung is the leading cause of cancer death worldwide. Here we report molecular profiling of 230 resected lung adenocarcinomas using messenger RNA, microRNA and DNA sequencing integrated with copy number, methylation and proteomic analyses. High rates of somatic mutation were seen (mean 8.9 mutations per megabase). Eighteen genes were statistically significantly mutated, including RIT1 activating mutations and newly described loss-of-function MGA mutations which are mutually exclusive with focal MYC amplification. EGFR mutations were more frequent in female patients, whereas mutations in RBM10 were more common in males. Aberrations in NF1, MET, ERBB2 and RIT1 occurred in 13% of cases and were enriched in samples otherwise lacking an activated oncogene, suggesting a driver role for these events in certain tumours. DNA and mRNA sequence from the same tumour highlighted splicing alterations driven by somatic genomic changes, including exon 14 skipping in MET mRNA in 4% of cases. MAPK and PI(3)K pathway activity, when measured at the protein level, was explained by known mutations in only a fraction of cases, suggesting additional, unexplained mechanisms of pathway activation. These data establish a foundation for classification and further investigations of lung adenocarcinoma molecular pathogenesis.

4,104 citations

Journal ArticleDOI
TL;DR: An analysis tool for the detection of somatic mutations and copy number alterations in exome data from tumor-normal pairs is presented and new light is shed on the landscape of genetic alterations in ovarian cancer.
Abstract: Exome sequencing of tumor samples and matched normal controls has the potential to rapidly identify protein-altering mutations across hundreds of patients, potentially enabling the discovery of recurrent events driving tumor development and growth (International Cancer Genome Consortium 2010; Stratton 2011). Yet the analysis of such data presents significant challenges. Sequencing coverage is nonuniform across targeted regions and from one sample to the next (Ng et al. 2009; Bainbridge et al. 2010; Teer et al. 2010). Many regions achieve high read depth (more than 100×), which can confound variant callers and depth-based filters if not properly addressed (Ku et al. 2011). Repetitive and paralogous sequences can give rise to numerous false positives. The detection of somatic mutations in tumor genomes is even more challenging. The genomes of primary tumors are genetically heterogeneous (Ding et al. 2010), with frequent rearrangements (Campbell et al. 2008) and copy number alterations (CNAs) (Beroukhim et al. 2010). Further, somatic mutations are relatively rare compared with germline variation, often representing <0.1% of variants in a tumor genome (Ley et al. 2008; Mardis et al. 2009). Simply subtracting variants in the matched normal from variants in the tumor (Wei et al. 2011) is poorly suited for the analysis of exome sequence data, because it fails to account for regions that were undersampled in the normal. Accurate mutation detection requires a direct, simultaneous comparison of tumor–normal pairs at every position in the exome, but few algorithms to do so have been described. Numerous algorithms have been developed to assess genome-wide copy number using whole-genome sequencing (WGS) data. Most of these approaches (Campbell et al. 2008; Alkan et al. 2009; Chiang et al. 2009; Yoon et al. 2009; Abyzov et al. 2011) would be confounded by exome data sets, because of the biases introduced by hybridization and the sparse and uneven coverages throughout the genome. However, when both DNA samples in a tumor–normal pair were captured and sequenced under identical hybridization conditions, we reasoned that it might be possible to detect somatic CNAs (SCNAs) as deviations from the log-ratio of sequence coverage depth within a tumor–normal pair, and then quantify the deviations statistically. Such an approach would provide a gene-centric view of copy number in a tumor sample, though it would be limited to the ∼1% of the genome captured by current exome platforms. Previously, we published VarScan (Koboldt et al. 2009), an algorithm for variant detection in next-generation sequencing data. We have since released a new tool, VarScan 2 (http://varscan.sourceforge.net), with several improvements, including the ability to identify somatic mutation, loss of heterozygosity (LOH), and CNA events in tumor–normal pairs. VarScan 2 analyzes sequence data from a tumor sample and its corresponding normal sample simultaneously, applying heuristic methods and a statistical test to detect variants—single nucleotide variants (SNVs) and insertions/deletions (indels)—and classify them by somatic status. By direct comparison of normalized sequence depth, our method also detects SCNAs in the tumor genome. Here, we utilize VarScan 2 for the analysis of exome sequence data from 151 patients with high-grade serous ovarian adenocarcinoma (HGS-OVCa) that were initially characterized within the Cancer Genome Atlas (TCGA) project (Cancer Genome Atlas Research Network 2011). We present a robust pipeline for the detection of both germline (inherited) and somatic (acquired) mutations by exome sequencing and describe filtering approaches for detecting variants with high sensitivity and specificity. To evaluate the performance of our SCNA detection algorithm, we compare our results to copy number data from high-density SNP array and WGS approaches. Our results demonstrate the accuracy of VarScan 2 for somatic mutation and CNA detection and enable a new survey of the genetic landscape in ovarian carcinoma.

4,096 citations

Related Papers (5)