Purpose:

Immune checkpoint inhibitors (ICI) have revolutionized the treatment of solid tumors with dramatic and durable responses seen across multiple tumor types. However, identifying patients who will respond to these drugs remains challenging, particularly in the context of advanced and previously treated cancers.

Experimental Design:

We characterized fresh tumor biopsies from a heterogeneous pan-cancer cohort of 98 patients with metastatic predominantly pretreated disease through the Personalized OncoGenomics program at BC Cancer (Vancouver, Canada) using whole genome and transcriptome analysis (WGTA). Baseline characteristics and follow-up data were collected retrospectively.

Results:

We found that tumor mutation burden, independent of mismatch repair status, was the most predictive marker of time to progression (P = 0.007), but immune-related CD8+ T-cell and M1-M2 macrophage ratio scores were more predictive for overall survival (OS; P = 0.0014 and 0.0012, respectively). While CD274 [programmed death-ligand 1 (PD-L1)] gene expression is comparable with protein levels detected by IHC, we did not observe a clinical benefit for patients with this marker. We demonstrate that a combination of markers based on WGTA provides the best stratification of patients (P = 0.00071, OS), and also present a case study of possible acquired resistance to pembrolizumab in a patient with non–small cell lung cancer.

Conclusions:

Interpreting the tumor-immune interface to predict ICI efficacy remains challenging. WGTA allows for identification of multiple biomarkers simultaneously that in combination may help to identify responders, particularly in the context of a heterogeneous population of advanced and previously treated cancers, thus precluding tumor type–specific testing.

Translational Relevance

Clinical response to immune checkpoint inhibitors (ICI) varies significantly and the majority of studies into their effectiveness are focused on primary tumors of single histologies. This retrospective study utilizes whole genome and transcriptome analysis (WGTA) to examine a pan-cancer cohort of advanced and previously treated patients that is currently underrepresented in the field, yet encompasses a large proportion of patients with cancer routinely seen in clinics. Our results reveal that tumor mutation burden and immune expression signatures are efficient at stratifying patients in this context, but suggest that PD-L1 testing may not be the most appropriate clinical biomarker for these patients. This study also demonstrates the benefit of measuring multiple markers simultaneously, highlighting the clinical utility of WGTA in selecting patients most likely to benefit from ICIs.

Immune checkpoint inhibitors (ICI) have revolutionized the treatment of solid tumors with dramatic and durable responses in cancers with previously limited effective treatment options (1, 2). Overall response rates to ICIs are often improved compared with standard-of-care cytotoxic chemotherapies but remain relatively low in unselected populations; for example, reported overall response rates are 13.3% in head and neck squamous cell carcinomas (HNSCC) treated with nivolumab (3), and 5.3% in previously treated triple-negative breast cancers (4). Response rates in selected populations are higher, with objective response rates of 40% to pembrolizumab in mismatch repair deficient (dMMR) colorectal cancers (5) and 41% in patients with non–small cell lung cancer (NSCLC) with more than 1% tumor cell programmed death-ligand 1 (PD-L1) staining (6). dMMR, PD-L1 expression, and more recently tumor mutation burden (TMB) are clinically approved biomarkers for patient selection (7, 8). However, many patients who are negative for these markers also respond to ICI therapy.

Additional features, including tumor immunogenicity and evidence of an immune response, may be predictive of ICI efficacy. TMB and prediction of neoantigens have been associated with clinical benefit from ICIs (9–11). The tumor microenvironment, particularly the presence and spatial arrangement of T cells, has also been shown to influence response, and has been explored in various tumor types including NSCLC and melanoma (10, 12). These features have yet to be surveyed in broader advanced cancer populations.

Whole genome and transcriptome analysis (WGTA) provides the opportunity to explore a wealth of genomic and expression markers that may predict response and overall survival (OS) with ICIs. The Personalized OncoGenomics Program (POG) at BC Cancer, in Vancouver, Canada, performs WGTA on patients with advanced and metastatic disease, often after one or more lines of therapy, and therefore represents an important heterogeneous patient population in which to examine predictors of ICI response (13, 14). Here we explored proposed biomarkers of ICI sensitivity in a pan-cancer cohort of patients with advanced or metastatic disease who received ICI therapy, incorporating WGTA and clinical follow-up.

Patient enrollment

This work was approved by and conducted under the University of British Columbia BC Cancer Research Ethics Board (H12-00137, H14-00681), in compliance with the Tri-Council Policy Statement and the FDA regulations (Belmont) and the Good Clinical Practice principles (Helsinki), and approved by the institutional review board. The POG is registered under clinical trial number NCT02155621. Patients with advanced or metastatic disease gave informed written consent and were enrolled into POG as described previously (13, 14). Ninety-eight patients within the program, biopsied between April 1, 2014 and August 31, 2018, had received ICIs. A REMARK (REporting recommendations for tumour MARKer prognostic studies; ref. 15) checklist has been included for this study.

Clinical data collection and processing

Full treatment histories, response, and survival data for this cohort were collected retrospectively using the BC Cancer Pharmacy database and chart review. Follow-up was censored on March 1, 2019. Patients who received ICIs up to 14 days prior to biopsy were considered ICI treated at time of biopsy. Patients were considered to have a durable clinical benefit (DCB) if they remained on treatment for greater than 6 months without disease progression [stable disease (SD), partial (PR) or complete response (CR)]. Time to progression (TTP) was defined as the time from ICI initiation to the date of discontinuation due to progression. OS was defined as the time from ICI initiation to death.

Tissue collection and sequencing

Tumor specimens were collected using needle core biopsies, endobronchial ultrasound biopsies, or tissue resection. Solid tumor specimens were snap frozen, while liquid biopsies were spun down into a cell pellet and resuspended, followed by extraction of DNA and RNA as described in Pleasance and colleagues (14). Constitutional DNA representing normal cells was extracted from peripheral blood.

PCR-free genomic DNA libraries and poly-A selected RNA libraries were constructed using standard protocols as described previously (16, 17). Tumor genomes were sequenced to a target depth of 80× coverage and normal peripheral blood samples to 40× coverage with 125 or 150 bp reads on the Illumina HiSeq platform. Transcriptomes were sequenced targeting 150–200 million 75-bp end reads on Illumina HiSeq2500 or NextSeq500. Sequencing characteristics for each sample are reported in Supplementary Table S1.

Somatic variant detection

Sequences were aligned to human genome version hg19, and single-nucleotide variants (SNV) and insertions and deletions (indels) were called as described in Jones and colleagues (17) and Grewal and colleagues (16). Structural variants (SV) were identified followed by merging and annotating using MAVIS (v2.1.1; ref. 18), and filtered for high-quality events as described in Pleasance and colleagues (14). Variants were annotated to genes using SNPEff (v3.2) with the Ensembl database (v69; ref. 19).

Copy-number alterations were identified from read depth ratios in tumor and normal whole-genome sequencing (WGS) data using CNAseq (v0.0.6; ref. 20), and regions of loss of heterozygosity determined from allelic ratios using APOLLOH (v0.1.1; ref. 21). Estimated tumor content for each sample was obtained on the basis of the best fit of predicted allelic ratios and read depth ratios to observed data, followed by manual review, also taking into consideration the estimated tumor content from pathology review of hematoxylin and eosin–stained tumor sections.

Exomic mutation burden was computed as the total of all SNVs and indels that fall within protein coding sequence (specifically, annotated by SNPEff as HIGH, MODERATE, or LOW impact and with an entry in the “aachange” field), divided by the total coding exome size of 35,600,628 bases included in the gene annotations, based on one transcript per gene for Ensembl 69 gene models. Microsatellite instability–high (MSI-H) samples were identified as such using MSIsensor (v0.2; ref. 22).

For analysis of resistance alterations, frameshift and nonsense mutations were defined as truncating alterations, and missense variants as substitutions. Mutations used for this analysis were manually reviewed for validity in the Integrated Genomics Viewer (23). Biallelic loss indicated deletions predicted to affect all copies of the gene. Differences in alteration frequencies between responders (DCB) and nonresponders [no clinical benefit (NCB)] were calculated using a χ2 test.

Gene expression profiling

Reads were aligned and processed using Jaguar (v2.0.3; ref. 24) as described in Jones and colleagues (17). Gene level RPKM (reads per kilobase per million mapped reads) were calculated on the basis of Ensembl gene models (v69).

Immune cell deconvolution

Immune cell type deconvolution was performed with the CIBERSORT R package (v1.04; ref. 25), using the LM22 cell subtype signature in absolute mode, with 1,000 permutations and no quantile normalization (Supplementary Table S3). Biopsies from hematopoietic malignancies, lymphomas, and thymic cancers, and organs of the hematopoietic system such as lymph nodes, were omitted from the immune CIBERSORT analysis due to the confounding presence of many immune cells unrelated to an antitumor immune response.

M1-M2 macrophage scoring

Using the LM22 cell weight matrix from CIBERSORT, we identified the genes that most effectively discriminate between M1 and M2 macrophages. Specifically, we calculated the fold change between the weight value for M1 and for M2 for each gene in the matrix, and the fold change for M1/M2 and the other 21 cell types. We selected the 10 genes with the highest weight for M1 and M2 (and not the other cell types) that also have the largest discrimination (fold change) between M1 and M2. The M1-M2 macrophage score for each sample was derived by calculating the mean expression (RPKM) of these 10 genes (CXCL11, IDO1, CCL19, CXCL9, PLA1A, LAMP3, CCR7, APOL6, CXCL10, TNIP3). Biopsies from hematopoietic malignancies and the hematopoietic system, such as lymph nodes, were also excluded from the M1-M2 macrophage analysis.

PD-L1 IHC

PD-L1 clinical staining tests were performed with the PD-L1 IHC 22C3 pharmDx test using the Dako autostainer.

Survival and regression analyses

Kaplan–Meier survival analysis was performed for TTP and OS using the R packages survival (v2.42.3) and survminer (v0.4.2). Differences in nonparametric survival functions were assessed across groups using log-rank tests. Cox proportional hazards models were performed using the R packages survival (v2.42.3) and forest model (v0.5.0). Log-rank tests were used to calculate P values, and in the case of ties, the Efron approximation was used. Linear regression models were used to account for histology in the DCB versus NCB analyses for mutation burden.

In the case of multivariate analyses using histology, only tumor types with more than one sample were included.

Marker thresholds

Thresholds for CD274 expression (80th percentile; 4.77 RPKM), CD8+ T cell (median; 0.054), and M1-M2 macrophage score (median; 8.51 RPKM) “high” groups were determined through examination of the naïve cohort distributions (Supplementary Fig. S1A) for each marker, and ensuring that sample sizes in each group were appropriate for combining markers later on. Ten mutations/Mb was used as a threshold for TMB as this has been previously reported as an appropriate threshold for hypermutation (26). High structural variant burden was defined as ≥ 75th percentile (187.25) for the cohort.

Validation cohorts and additional markers

Data for Snyder and colleagues (27) was downloaded from Supplementary Data and https://github.com/hammerlab/multi-omic-urothelial-anti-pdl1 on April 28, 2020. High mutation burden was defined as ≥ 10/Mb. Mutation burden in this cohort was excluded from the multivariate Cox proportional hazards models as only one patient was above this threshold. Thresholds for high score were defined as: CD8 score ≥ 0.054 as the median was 0 (0.054 was the same value as the median in our cohort), M1-M2 score ≥ median (228.6), and CD274 expression ≥ 80th percentile (202.6). DCB was defined as a positive response (PR, CR, SD) without progression for at least 6 months. Data for Riaz and colleagues (28) were downloaded from the Supplementary Data and https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE91061 on April 28, 2020. High mutation burden was defined as ≥ 200 coding mutations, which corresponds to approximately 10/Mb. Clinical benefit was defined as a positive response (PR, CR, SD) only, as no progression data was available. Thresholds for high score were defined as: CD8 score ≥ median (0.075), M1-M2 score ≥ median (17.05), and CD274 expression ≥ 80th percentile (6.73). Patients with nonevaluable responses were not included in clinical benefit analyses. Raw data for Liu and colleagues (29) were downloaded from https://www.ncbi.nlm.nih.gov/projects/gap/cgi-bin/study.cgi?study_id=phs000452.v3.p1#restricted-access-section on May 25, 2020. SNV calling and gene expression profiling were performed as described above for our cohort. High mutation burden was defined as ≥ 10/Mb. Thresholds for high score were defined as: CD8 score ≥ 0.054, M1-M2 score ≥ median (8.17), and CD274 expression ≥ 80th percentile (1.51). DCB was defined as a positive response (PR, CR, SD) without progression for at least 6 months. Of these datasets, patients were only included if they were naïve for immunotherapy prior to the study and both transcriptome and mutation data were available, consistent with our study. CIBERSORT and M1-M2 scores were evaluated as described above on available expression data. Where relevant, lymph associated samples were excluded.

Cytolytic (CYT) scores were calculated using expression transcripts per million (TPM) as described in Rooney and colleagues, (30). T-cell exclusion signature (TIDE) scores and IFNγ signatures were obtained using normalized expression RPKMs with the TIDE web application http://tide.dfci.harvard.edu/ (31–33). TPMs were calculated as described in Pleasance and colleagues (14).

Data availability

Sequence data, both WGS and RNAseq, have been deposited at the European Genome–phenome Archive (EGA, http://www.ebi.ac.uk/ega/) as part of the study EGAS00001001159 with accession numbers as listed in Supplementary Table S1. Clinical data, marker status, sequencing statistics, and CIBERSORT immune scores are presented in Supplementary Tables S1–S3. Complete somatic mutation variant call format (VCF) files, RPKM expression matrices, and tables of structural variants and coding mutations described in this study are available from https://www.bcgsc.ca/downloads/immunoPOG/. Data on coding mutations, copy changes, and expression from tumor samples in the POG program are also accessible from https://www.personalizedoncogenomics.org/cbioportal/. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Advanced and pretreated tumors profiled by WGTA

We characterized tumor samples from 98 patients (Table 1; Supplementary Table S1), representing a diversity of histologies, biopsy sites, and treatment histories, who had received ICIs either following (naïve cohort, n = 82) or prior to biopsy (treated cohort, n = 16). Twenty different tumor types were profiled, with the majority of cases being lung (n = 26, 26%) and breast cancer (n = 13, 13%), or cutaneous melanoma (n = 11, 11%). The median age of advanced disease diagnosis was 57 (25–86 years) and 55% of patients were female. Biopsies were taken at a median of 10 months following advanced diagnosis (0 days–15 years), and the median follow-up time postbiopsy was 15 months (0–54 months). Patients in the naïve cohort received an ICI after the POG biopsy, typically as a third-line therapy (median, range: 1–10), and those in the treated cohort had received an ICI typically as a first-line therapy (median, range: 1–4). The naïve cohort was the primary focus of this study for examining genomic biomarkers predictive of response.

Table 1.

Cohort and treatment demographics of tumor types and drug combinations received by patients in the immunotherapy-naïve and -treated cohorts.

CharacteristicNaïve (n = 82)Treated (n = 16)
Median age of advanced disease diagnosis (range) 56 (25–86) 63 (37–76) 
Sex, n (%) 
 Female 48 (59) 6 (38) 
 Male 24 (41) 10 (63) 
Tumor type, n (%) 
 Lung (LUNG) 25 (30) 1 (6) 
 Breast (BRCA) 12 (15) 1 (6) 
 Skin cutaneous melanoma (SKCM) 7 (9) 4 (25) 
 Colorectal (COLO) 6 (7) 0 (0) 
 Sarcoma (SARC) 5 (6) 0 (0) 
 Head and neck squamous cell carcinoma (HNSC) 4 (5) 1 (6) 
 Cholangiocarcinoma (CHOL) 3 (4) 0 (0) 
 Uterine corpus endometrial carcinoma (UCEC) 3 (4) 0 (0) 
 Apocrine eccrine carcinoma/sweat gland carcinoma (AECA) 2 (2) 0 (0) 
 Kidney (KDNY) 2 (2) 0 (0) 
 Pancreatic (PANC) 2 (2) 0 (0) 
 Stomach adenocarcinoma (STAD) 2 (2) 0 (0) 
 Uveal melanoma (UVM) 2 (2) 7 (44) 
 Adenoid cystic carcinoma (ACC) 2 (2) 0 (0) 
 Oesophageal (ESCA) 1 (1) 0 (0) 
 Germ cell (GCT) 1 (1) 0 (0) 
 Lymphoma (LYMP) 1 (1) 0 (0) 
 Ovarian (OV) 1 (1) 1 (6) 
 Thymic (THYM) 1 (1) 0 (0) 
 Cervical squamous cell carcinoma (CERV) 0 (0) 1 (6) 
ICI received, n (%) 
 PD-1 45 (55) 8 (50) 
 PD-L1 10 (12) 2 (13) 
 PD-1/IDO1 8 (10) 0 (0) 
 PD-L1/CTLA-4 8 (10) 0 (0) 
 NKG2A 2 (2) 0 (0) 
 CTLA-4 1 (1) 3 (19) 
 PD-L1/OX40 1 (1) 2 (13) 
 PD-1/CTLA-4 1 (1) 1 (6) 
 OX40 1 (1) 0 (0) 
 PD-1/Chemo 1 (1) 0 (0) 
 PD-1/LAG-3 1 (1) 0 (0) 
 PD-L1/CTLA-4/Chemo 1 (1) 0 (0) 
 PD-L1/HER2 1 (1) 0 (0) 
 PD-L1/SMAC 1 (1) 0 (0) 
Lines of systemic treatment prior to ICI, n (%) 
 0 14 (17) 10 (63) 
 1 17 (21) 3 (19) 
 2 20 (24) 1 (6) 
 3 14 (17) 2 (13) 
 4 9 (11) 0 (0) 
 5 or more 8 (10) 0 (0) 
CharacteristicNaïve (n = 82)Treated (n = 16)
Median age of advanced disease diagnosis (range) 56 (25–86) 63 (37–76) 
Sex, n (%) 
 Female 48 (59) 6 (38) 
 Male 24 (41) 10 (63) 
Tumor type, n (%) 
 Lung (LUNG) 25 (30) 1 (6) 
 Breast (BRCA) 12 (15) 1 (6) 
 Skin cutaneous melanoma (SKCM) 7 (9) 4 (25) 
 Colorectal (COLO) 6 (7) 0 (0) 
 Sarcoma (SARC) 5 (6) 0 (0) 
 Head and neck squamous cell carcinoma (HNSC) 4 (5) 1 (6) 
 Cholangiocarcinoma (CHOL) 3 (4) 0 (0) 
 Uterine corpus endometrial carcinoma (UCEC) 3 (4) 0 (0) 
 Apocrine eccrine carcinoma/sweat gland carcinoma (AECA) 2 (2) 0 (0) 
 Kidney (KDNY) 2 (2) 0 (0) 
 Pancreatic (PANC) 2 (2) 0 (0) 
 Stomach adenocarcinoma (STAD) 2 (2) 0 (0) 
 Uveal melanoma (UVM) 2 (2) 7 (44) 
 Adenoid cystic carcinoma (ACC) 2 (2) 0 (0) 
 Oesophageal (ESCA) 1 (1) 0 (0) 
 Germ cell (GCT) 1 (1) 0 (0) 
 Lymphoma (LYMP) 1 (1) 0 (0) 
 Ovarian (OV) 1 (1) 1 (6) 
 Thymic (THYM) 1 (1) 0 (0) 
 Cervical squamous cell carcinoma (CERV) 0 (0) 1 (6) 
ICI received, n (%) 
 PD-1 45 (55) 8 (50) 
 PD-L1 10 (12) 2 (13) 
 PD-1/IDO1 8 (10) 0 (0) 
 PD-L1/CTLA-4 8 (10) 0 (0) 
 NKG2A 2 (2) 0 (0) 
 CTLA-4 1 (1) 3 (19) 
 PD-L1/OX40 1 (1) 2 (13) 
 PD-1/CTLA-4 1 (1) 1 (6) 
 OX40 1 (1) 0 (0) 
 PD-1/Chemo 1 (1) 0 (0) 
 PD-1/LAG-3 1 (1) 0 (0) 
 PD-L1/CTLA-4/Chemo 1 (1) 0 (0) 
 PD-L1/HER2 1 (1) 0 (0) 
 PD-L1/SMAC 1 (1) 0 (0) 
Lines of systemic treatment prior to ICI, n (%) 
 0 14 (17) 10 (63) 
 1 17 (21) 3 (19) 
 2 20 (24) 1 (6) 
 3 14 (17) 2 (13) 
 4 9 (11) 0 (0) 
 5 or more 8 (10) 0 (0) 

High TMB is associated with a longer duration on treatment

Tumors with a high TMB and dMMR have been associated with a good response to ICIs in previously untreated tumors of a single type (5, 34). We explored whether TMB was predictive in our naïve cohort comprised of mixed tumor types and prior exposure to other cancer therapies. We calculated TMB as mutations per Mb both within coding regions (exomic TMB; Fig. 1A) and across the entire genome (genomic TMB). Measuring TMB in coding regions could reflect mutations with direct potential to elicit an immune response, and is more consistent with previous studies that use whole-exome sequencing or panels such as FoundationOne CDx (35, 36). However, we observed that the two approaches produced strongly correlated TMB values (Fig. 1B; r = 0.95, P < 2.2 × 10−16, Spearman). Exomic TMB ranged from 0.70 to 85 mutations/Mb (median: 4.6) in the naïve cohort, with cutaneous melanomas and lung cancers having some of the highest TMBs (Fig. 1A). Only three samples in this cohort were dMMR, one colorectal and two lung cancers, all of which had TMB > 30 mutations/Mb.

Figure 1.

Mutation burden can predict response to ICIs. A, Distribution of TMB (exomic mutations/Mb) by tumor type. Median values are indicated by horizontal red lines. Solid diamonds indicate dMMR. B, Spearman correlation between exomic and genomic TMB across the cohort. Exomic (C) and genomic (E) TMB in patients with a DCB versus patients with NCB. P values were calculated by a two-sided Wilcoxon rank-sum test. D, TTP and OS for patients with high exomic TMB (≥10 mutations/Mb, red) versus low TMB (blue), and (F) for patients with a high genomic TMB (≥10 mutations/Mb, red) versus low TMB (blue). P values calculated by a two-sided log-rank test. All data are from the ICI-naïve cohort. Tumor types as described in Table 1.

Figure 1.

Mutation burden can predict response to ICIs. A, Distribution of TMB (exomic mutations/Mb) by tumor type. Median values are indicated by horizontal red lines. Solid diamonds indicate dMMR. B, Spearman correlation between exomic and genomic TMB across the cohort. Exomic (C) and genomic (E) TMB in patients with a DCB versus patients with NCB. P values were calculated by a two-sided Wilcoxon rank-sum test. D, TTP and OS for patients with high exomic TMB (≥10 mutations/Mb, red) versus low TMB (blue), and (F) for patients with a high genomic TMB (≥10 mutations/Mb, red) versus low TMB (blue). P values calculated by a two-sided log-rank test. All data are from the ICI-naïve cohort. Tumor types as described in Table 1.

Close modal

Patients that demonstrated a DCB (see Materials and Methods) did not have a significantly higher TMB overall than those with NCB (Fig. 1C; median 12.7 vs. 4.3, P = 0.16 and Fig. 1E; median 18.4 vs. 5.8, P = 0.053), though there was a significant difference after correction for histology [P = 0.035 (exome) and P = 0.029 (genome)]. A high TMB (≥ 10 mutations/Mb; Materials and Methods; Supplementary Fig. S1A) was strongly predictive of longer TTP for both exome (Fig. 1D; median 179 days vs. 79, P = 0.007) and genome-derived counts (Fig. 1F; median 156 days vs. 79.5, P = 0.0027). As TMB can vary substantially by tumor type, we additionally noted that this observation remained true after accounting for histology [exome; HR = 0.36 (0.16–0.83), P = 0.02, genome; HR = 0.34 (0.15–0.74), P = 0.007]. TMB was less predictive of OS, and using the same TMB threshold, exome-derived counts (Fig. 1D; median 442 days vs. 240, P = 0.039) were more effective than genomic (Fig. 1F; median 396 vs. 265, P = 0.17). This also remained constant after correction for tumor type [exome; HR = 0.41 (0.18–0.92), P = 0.03, genome; HR = 0.54 (0.26–1.12), P = 0.1]. Overall, there was a stronger stratification of patients based on TMB for TTP than OS, which is likely influenced by higher TMBs being a poor prognostic marker (37, 38).

The burden of SVs in the genome was also evaluated as a potential prognostic marker for ICIs, but we could not identify a consistent trend with response (Supplementary Figs. S1B and S1C). Despite some difference in OS between patients with high (≥75th percentile) and low SV burden, we did not see the same trend with TTP on ICI therapy, and we did not observe a difference in SV burden between patients with and without clinical benefit. This may indicate some OS benefit associated with higher structural variant burden that is independent of treatment with ICIs.

Immune cell expression profiles predict response to ICIs

The composition of the tumor microenvironment, in particular, the presence and spatial arrangement of immune cells, has been reported to influence patient response to ICIs (12). As these investigations have generally been conducted using IHC, we sought to investigate the predictive power of immune cell gene expression profiles derived from WGTA (Materials and Methods). We observed a trend of higher T-cell expression scores in samples biopsied from lymph nodes and in lymphoid cancers (Supplementary Fig. S1D, P = 0.063, Wilcoxon rank sum). As presence of immune cells in these tissues was expected, and may not specifically indicate detection of an antitumor immune response, we excluded these (n = 17 naïve samples of mixed histologies; Supplementary Table S1) from immune cell expression analyses. Immune cell expression profiles varied by tumor type (Fig. 2A), with highest variability found in CD8+ T cell, M0 macrophage, and neutrophil scores [standard deviation (StdDev) 0.19, 0.16, and 0.16, respectively]. Cholangiocarcinomas displayed the highest overall levels of immune cell expression, which may be in part a result of the lower tumor content of these sample biopsies (Supplementary Fig. S1E). Cholangiocarcinoma, skin cutaneous melanomas (SKCM), and HNSCCs displayed the highest levels of expression for CD8+ T cells.

Figure 2.

Immune expression profiles can predict response to ICIs. A, Mean CIBERSORT immune cell scores for samples stratified by tumor type. Only tumor types with at least three samples are shown. B, CD8+ T-cell scores in patients with a DCB versus patients with NCB. C, TTP and OS for patients with high CD8+ scores (≥ median, red) versus low scores (blue). D, M1-M2 macrophage scores (Materials and Methods) in patients with a DCB versus patients with NCB. E, TTP and OS for patients with high M1-M2 macrophage scores (≥ median, red) versus low scores (blue). F, Distribution of CD274 (PD-L1) gene expression by tumor type. Median values are indicated by horizontal red lines. G,CD274 expression in patients with a DCB versus patients with NCB. H, TTP and OS for patients with high CD274 expression (≥ 80th percentile, red) versus low expression (blue). P values for B, D, and G calculated by two-sided Wilcoxon rank-sum tests, and those for C, E, and H by two-sided log-rank tests. All data are from the ICI-naïve cohort, excluding samples from lymph nodes or lymphoid cancers (see Materials and Methods). Tumor types as described in Table 1.

Figure 2.

Immune expression profiles can predict response to ICIs. A, Mean CIBERSORT immune cell scores for samples stratified by tumor type. Only tumor types with at least three samples are shown. B, CD8+ T-cell scores in patients with a DCB versus patients with NCB. C, TTP and OS for patients with high CD8+ scores (≥ median, red) versus low scores (blue). D, M1-M2 macrophage scores (Materials and Methods) in patients with a DCB versus patients with NCB. E, TTP and OS for patients with high M1-M2 macrophage scores (≥ median, red) versus low scores (blue). F, Distribution of CD274 (PD-L1) gene expression by tumor type. Median values are indicated by horizontal red lines. G,CD274 expression in patients with a DCB versus patients with NCB. H, TTP and OS for patients with high CD274 expression (≥ 80th percentile, red) versus low expression (blue). P values for B, D, and G calculated by two-sided Wilcoxon rank-sum tests, and those for C, E, and H by two-sided log-rank tests. All data are from the ICI-naïve cohort, excluding samples from lymph nodes or lymphoid cancers (see Materials and Methods). Tumor types as described in Table 1.

Close modal

We also observed that the median CD8+ T-cell score in responders (DCB) was over double that of nonresponders, though the overall difference was not significant (Fig. 2B; median 0.13 vs. 0.05, P = 0.11). Higher CD8+ T-cell scores (≥ median; Supplementary Fig. S1A) were predictive of a longer TTP (Fig. 2C; 102 days vs. 70, P = 0.0059) and OS (Fig. 2C; 370 days vs. 160, P = 0.0014). In contrast to TMB, high CD8+ scores had a more profound influence on OS than TTP, highlighting the underlying positive prognostic value of CD8+ T cells (39). Evaluating expression signatures from other immune cells revealed that only CD8+ T-cell scores were predictive for OS (P = 0.03, Holm–Bonferroni correction), consistent with the central role of CD8+ T cells in eliminating tumor cells (40).

M1 macrophages are known to have antitumor characteristics (41). Conversely, M2 macrophages are thought to have opposing effects and repolarization of these to an M1 phenotype may improve therapy response (41, 42). As individual M1 and M2 macrophage expression scores were not predictive, and the ratio of M1/M2 macrophages has been described as having prognostic value in cancer (43), we developed a score for M1-M2 macrophages expression (Materials and Methods), and investigated its utility in predicting response to ICIs. We discovered that the difference in M1-M2 score between patients with a DCB and NCB was more pronounced than for the CD8+ T-cell score (Fig. 2D; median 21.2 vs. 7.6, P = 0.018). Similar to the CD8+ score, a high M1-M2 macrophage score (≥ median; Supplementary Fig. S1A) was associated with longer TTP (Fig. 2E; median 102 vs. 65, P = 0.041) and OS (median 373 days vs. 146, P = 0.0012).

In addition to CD8+ T-cell scores, and M1-M2 macrophage expression, we were also able to validate previously published immunologic biomarkers of response in our cohort (Supplementary Fig. S2A—S2L) including a CYT score (30) and IFNγ expression signature (31) which both correlated well with our CD8+ score (R = 0.8 and 0.74, respectively, Spearman). The previously published TIDE (33) had an inverse relationship (R = −0.5, Spearman) with our CD8+ score, and was not predictive in our pan-cancer cohort, unlike in the melanoma tumors in which this was originally developed.

CD274 gene expression is poorly associated with response and survival

Expression of measured by IHC, is used as a clinical biomarker to select patients that may benefit from ICIs (8), but there is evidence that PD-L1–low or –negative patients can also respond to therapy (44, 45). We therefore sought to investigate the utility of RNA expression of the PD-L1 gene (CD274) as a biomarker of response to ICIs in this cohort. Expression of CD274 was highest in a lymphoma and a thymic carcinoma (Fig. 2F), consistent with its natural expression in the immune cells composing these tumors. Some LUNG, cholangiocarcinoma, and SKCM also exhibited high expression of CD274. Comparison with PD-L1 IHC staining available for a subset (n = 11) of NSCLCs showed that samples with the highest PD-L1 staining in tumor cells (>50%) consistently had the highest levels of CD274 expression (Supplementary Fig. S1F; P = 0.047, Kruskal–Wallis), suggesting that CD274 expression may be a useful surrogate for IHC staining. Furthermore, consistent with a recent study examining PD-L1 status (46), we observed that CD274 expression was not correlated with TMB (R = 0.051, P = 0.65, Spearman).

Median CD274 expression was not significantly higher in responders (Fig. 2G; median 8.9 RPKM vs. 1.8, P = 0.12), though the data may suggest a trend. In addition, we noted that many responders had little to no CD274 expression, and that nonresponders often also had high expression. Interestingly, the highest expressing responders encompassed a variety of tumor types (one melanoma, one thymic, one colorectal, one NSCLC, and one sarcomatoid carcinoma of the lung) and not just NSCLCs for which the biomarker is currently validated. Patients with high CD274 expression (≥80th percentile, 4.8 RPKM) had a longer TTP (median 104 days vs. 79, P = 0.032), but had no OS benefit (Fig. 2H; median, 340 days vs. 271, P = 0.87). This suggests that, while high CD274 expression can be observed across tumor types, it may not be the most informative biomarker for response in a pan-cancer setting.

Combination biomarkers improve prediction of ICI response

Clinically approved biomarkers for patient selection for ICI therapy, such as PD-L1 positivity or dMMR status, are currently considered in isolation (47). We investigated the potential for improved predictive value of combinations of biomarkers. Using TMB, CD8+ T-cell score, M1-M2 score, and CD274 expression, we performed multivariate Cox proportional hazards models, also incorporating tumor type to control for any underlying variation in response (see Materials and Methods). High CD8+ T-cell scores (≥ median; HR = 0.41, P = 0.03) and TMB (≥10 mutations/Mb; HR = 0.36, P = 0.05) were most predictive of TTP (Fig. 3A). M1-M2 scores were most beneficial for OS (≥ median; HR = 0.26, P = 9.4 × 10−04) and a high TMB or CD8+ T-cell score also provided prognostic benefit (HR = 0.36, P = 0.03 and HR = 0.46, P = 0.06, respectively). Of the 33 samples with high CD8+ T-cell score, 75% also showed a high M1-M2 score; 12.5% of samples were high for only one of these. Despite this overlap, these results indicate that both biomarkers contribute to patient response. The presence of high CD274 expression did not provide any additional benefit in TTP [≥ 80th percentile; HR = 0.84 (0.32–2.22), P = 0.72] or OS [HR = 0.93 (0.35–2.47), P = 0.89], indicating that any benefit from CD274 expression can be explained in our cohort by positivity for another marker. We additionally examined the markers as continuous covariates but did not find the results to be as robust (overall model TTP; P = 0.04, OS; P = 0.6). It is possible that the effect of these markers on response and prognosis is only observed at higher levels, consistent with high TMB thresholds described previously (35, 36) and is not as discriminating at lower values.

Figure 3.

Combination biomarkers. Cox proportional hazards model for TTP (A) and OS (B), taking high CD8+ T-cell score, high mutation burden, high M1-M2 score, high CD274 expression, and tumor type into account. Patients were split into binary high and low groups for each marker based on the thresholds defined in the Materials and Methods. Only tumor types with at least two samples were included in the models. Overall model significance was determined using a log-rank test. C, The number of positive biomarkers in patients with a DCB versus patients with NCB. D, TTP and OS stratified by the number of positive markers in each sample. The P value for C was calculated by a two-sided Wilcoxon rank-sum test, and global P values for D by log-rank tests. All data are from the naïve cohort, excluding samples from lymph nodes or lymphoid cancers (see Methods). Tumor types as described in Table 1.

Figure 3.

Combination biomarkers. Cox proportional hazards model for TTP (A) and OS (B), taking high CD8+ T-cell score, high mutation burden, high M1-M2 score, high CD274 expression, and tumor type into account. Patients were split into binary high and low groups for each marker based on the thresholds defined in the Materials and Methods. Only tumor types with at least two samples were included in the models. Overall model significance was determined using a log-rank test. C, The number of positive biomarkers in patients with a DCB versus patients with NCB. D, TTP and OS stratified by the number of positive markers in each sample. The P value for C was calculated by a two-sided Wilcoxon rank-sum test, and global P values for D by log-rank tests. All data are from the naïve cohort, excluding samples from lymph nodes or lymphoid cancers (see Methods). Tumor types as described in Table 1.

Close modal

Samples with a DCB had more positive markers than those with NCB (Fig. 3C; median 2 vs. 1, P = 0.0028), excluding CD274 due to its lack of predictive value. Interestingly, all patients with a DCB were positive for at least two markers, except for one patient with stomach adenocarcinoma whose biopsy sample was negative for all markers that we evaluated. Samples with more markers had significantly improved TTP [Fig. 3D; HR = 0.62 (0.47–0.81), P = 0.0006] and OS [Fig. 3D; HR = 0.56 (0.42–0.75), P = 0.0001].

In addition to our pan-cancer cohort, we also found that there may be predictive value in using the combination of TMB, CD8+ T-cell score, and M1-M2 score in independent, previously published cohorts from single tumor types (Supplementary Fig. S3; refs. 27, 28). While the number of samples in these cohorts is smaller, precluding statistical significance, we still observe a trend in the same direction toward the individual predictive value of CD8 score, TMB, and M1-M2 macrophage score, and that responders trend in the direction of having more than one marker. In another published melanoma cohort by Liu and colleagues (29), the authors report no trend with respect to TMB or T-cell score; our reanalysis of this melanoma cohort confirmed their findings, and additionally showed no trend with M1-M2 macrophage score. The authors attributed the lack of observed association partly to confounding rare tumor subtypes. This exception to the consistent trends found in other tumor datasets underscores the complexity of evaluating biomarkers in heterogeneous cohorts, and suggests that some rare tumor subtypes may be more amenable to alternative biomarkers, the elucidation of which will require further detailed study.

Landscape of alterations in ICI resistance genes

We examined alterations in genes in antigen presentation and JAK/STAT signaling pathways, as these have been reported as ICI resistance mechanisms (48, 49). Considering all types of somatic alterations in these genes, we observed no difference in the proportion of altered samples between DCB and NCB groups (Fig. 4A; 24% vs. 31%, P = 0.78, χ2 test). Notably, however, all samples with biallelic alterations (events which affect all copies of the gene due to copy loss or loss of heterozygosity) were in the nonresponder group. This observation could not reach statistical significance (0% vs. 12%, P = 0.28) due to the small number of samples with these events (n = 8). In the 16 patients who progressed on ICIs before WGTA, biallelic losses were only present in patients who had previously exhibited a DCB, but the small number of patients (n = 8) prevented a statistical interpretation of this observation (Fig. 4B).

Figure 4.

Potential markers of resistance to ICIs. Genomic alterations in ICI-naïve (A) and ICI-treated (B) patients in genes previously associated with resistance to ICIs. Patients are ordered by clinical benefit (DCB), and time to progression on ICIs. Biallelic mutations are indicated by black circles inside the tile. Biallelic loss refers to a genomic deletion predicted to affect all copies of the gene. The event track highlights if patients have had a progression or death event. Those without an event had no progression events at the time of censoring. C, An example of a patient with advanced lung cancer who had a mixed response to pembrolizumab following treatment for 104 days; the original biopsied site (liver) responded, yet progression was seen at the posttreatment biopsy site (supraclavicular node). The second biopsy was taken 135 days after treatment initiation which revealed biallelic frameshift mutations in both JAK1 and HLA-A, neither of which were found in the liver biopsy before treatment.

Figure 4.

Potential markers of resistance to ICIs. Genomic alterations in ICI-naïve (A) and ICI-treated (B) patients in genes previously associated with resistance to ICIs. Patients are ordered by clinical benefit (DCB), and time to progression on ICIs. Biallelic mutations are indicated by black circles inside the tile. Biallelic loss refers to a genomic deletion predicted to affect all copies of the gene. The event track highlights if patients have had a progression or death event. Those without an event had no progression events at the time of censoring. C, An example of a patient with advanced lung cancer who had a mixed response to pembrolizumab following treatment for 104 days; the original biopsied site (liver) responded, yet progression was seen at the posttreatment biopsy site (supraclavicular node). The second biopsy was taken 135 days after treatment initiation which revealed biallelic frameshift mutations in both JAK1 and HLA-A, neither of which were found in the liver biopsy before treatment.

Close modal

One patient with NSCLC is particularly remarkable; she presented (ages 50) with stage IV squamous cell lung cancer with bilateral lung masses, mediastinal adenopathy, and liver metastases. She had a 10-pack-year smoking history but had quit 20 years prior to this diagnosis. This was her fourth cancer diagnosed within 10 years, having had surgery for a T2 breast cancer, T3N1 colorectal cancer, and a stage IA endometrial adenocarcinoma. Next-generation sequencing of germline DNA was uninformative; however, subsequent clinical testing identified constitutive MLH1 promoter hypermethylation in the patient. For her NSCLC she received palliative radiation to her lung mass and initiated first-line pembrolizumab 4 weeks later. The patient underwent a biopsy for WGTA both before (liver biopsy) and after (supraclavicular node) pembrolizumab. On the basis of WGTA this patient was a strong candidate for receiving ICIs (dMMR, high TMB, CD8+ T cell and M1-M2 scores, and high CD274 expression; Fig. 4C). Although her liver and lung nodules did respond to pembrolizumab, she had progression in her supraclavicular fossa and bone after 104 days on therapy, therefore, she did not have a DCB. The second biopsy of the growing supraclavicular mass was taken 135 days after treatment initiation which revealed biallelic frameshift mutations in both JAK1 and HLA-A, neither of which were found in the other biopsy (liver) before treatment (Fig. 4C). This may represent an acquired resistance mechanism to ICIs, or presence of a resistant subclone which was selected for by ICI treatment, which highlights the importance of profiling metastatic lesions.

As the complexity of cancer care increases with technological advances and emergence of new therapeutic options, identification of the optimal patient population for any given intervention is crucial. Current molecular testing to align patients to immunotherapies are usually panel- or IHC-based approaches focused on individual biomarkers, for example, MMR status and PD-L1 expression. However, it is becoming clear that individual biomarkers are neither necessary nor sufficient to select patients most likely to benefit. WGTA has the advantage of producing a wealth of information that would otherwise require multiple independent tests, which is particularly valuable given our findings that multiple markers are predictive of ICI response. Our accrual of WGTA data for a pan-cancer cohort of ICI-treated patients will permit examination of more unusual responders, and can be used retrospectively (see Data Availability) to identify and evaluate new markers of response and resistance.

Our results lend support to development of TMB as a tumor agnostic clinical biomarker, confirming previous findings (11, 34), and the recent tumor-agnostic FDA approval of pembrolizumab in patients with a TMB >10 mutations/Mb using the FoundationOne CDx assay (7). Our results also demonstrate a strong relationship between exome- and genome-derived TMB counts with little difference in predictive value, providing further support for the applicability of WGTA in clinical testing.

Our findings emphasize the fundamental predictive value of immune-related markers for ICI response. Consistent observations across tumor types, cohorts, and different measures of T-cell activity [CD8 score, IFNγ score (31), and cytolytic score (30)] suggest a clear role for expression-based signatures of T-cell activity in a pan-cancer context. Our macrophage score, reflecting the expression of M1 and M2 macrophage markers, has not been previously described and evaluation both in our cohort and in published datasets suggests this may warrant further investigation as another independently predictive expression-based immune-related marker of response.

In contrast, we found that CD274 expression was not predictive in a tumor agnostic context, despite good concordance with PD-L1 IHC, a clinical biomarker evaluated in specific tumor types including NSCLCs. Even within this population, there are many PD-L1 diagnostic tests available with varying thresholds paired with specific drugs (50), suggesting that the clinical benefit from PD-L1 may be tumor or drug specific.

ICI resistance is reported across multiple cancer types with patient-based and in vivo studies suggesting roles for antigen presentation and JAK/STAT signaling pathways (48, 49, 51). Loss-of-function mutations in JAK1 similar to those found here have been previously associated with resistance to pembrolizumab in patients with melanoma (51), supporting similar mechanisms of resistance across tumor types. Our finding that biallelic mutations and copy losses in these pathways were limited to patients with NCB, or patients post-ICI treatment, suggests that complete loss of gene function may be key to resistance. This has parallels to the patterns of alterations of tumor suppressor genes during cancer development (52). We did not observe recurrent alterations, suggesting that resistance may be driven by loss of a pathway, rather than a single gene. Future work in larger patient cohorts will expand on the clinical relevance of these and other potential resistance alterations.

In summary, we have presented a unique WTGA dataset for pan-cancer ICI-treated tumors, and demonstrated that a combination of biomarkers including high TMB, CD8+ expression, and M1-M2 macrophage scores can predict both TTP and OS on ICIs. These data support a potential move away from PD-L1 testing as the clinical biomarker of choice for immunotherapy, and point toward the use of a multifaceted assessment of many tumor features, as is afforded by WGTA. Challenges in measuring and standardizing biomarkers for clinical use across tumor types, as illustrated by work to standardize TMB measures across panel platforms (53), demonstrate the importance of exploration in pan-cancer cohorts for hypothesis generation, and follow-up with prospective clinical trials. A pan-cancer approach based on WGTA data and integrating multiple biomarkers based on the data presented here has been implemented in a phase II clinical trial at BC Cancer (Vancouver, Canada), CAPTIV-8, which uses WGTA to select patients for treatment with the PD-L1 inhibitor atezolizumab (NCT04273061). This illustrates the potential of integrative genomics approaches to identify patients best suited for immunotherapies in current medical practice.

A. Pender reports other from Roche Canada during the conduct of the study; A. Pender also reports other from Guardant Health and BMS outside the submitted work. C.J. Grisdale reports grants from NIH during the conduct of the study. R.A. Moore reports grants from Genome Canada, TFRI, and BC Cancer Foundation during the conduct of the study. J.-M. Lavoie reports personal fees from Hoffmann-La Roche Limited outside the submitted work. S. Yip reports receiving (advisory boards) travel allowances from Amgen, AstraZeneca, Bayer, Norvatis, Pfizer, and Roche, none of which are associated with the submitted work. H. Lim reports other from Ipsen, Merck, BMS, Taiho, Eisai, and Roche outside the submitted work. D.J. Renouf reports grants and personal fees from Roche, as well as personal fees from Bayer, Celgene, Servier, Ipsen, Taiho, and AstraZeneca outside the submitted work. S.J.M. Jones reports grants from BC Cancer Foundation, Terry Fox Research Institute, Canada Foundation for Innovation, Canada Research Chairs, and BC Knowledge Development Fund during the conduct of the study. J. Laskin reports grants from BC Cancer Foundation and Roche Canada during the conduct of the study; grants from Roche Canada; and personal fees from Roche Canada, Pfizer, Eli Lilly, and AstraZeneca outside the submitted work. No disclosures were reported by the other authors.

A. Pender: Conceptualization, data curation, supervision, writing-original draft, writing-review and editing. E. Titmuss: Conceptualization, data curation, formal analysis, validation, investigation, visualization, methodology, writing-original draft, writing-review and editing. E.D. Pleasance: Conceptualization, formal analysis, supervision, investigation, methodology, writing-original draft, writing-review and editing. K.Y. Fan: Data curation, formal analysis, writing-original draft, writing-review and editing. H. Pearson: Conceptualization, data curation. S.D. Brown: Investigation, writing-review and editing. C.J. Grisdale: Data curation, writing-review and editing. J.T. Topham: Investigation, writing-review and editing. Y. Shen: Resources, writing-review and editing. M. Bonakdar: Resources, writing-review and editing. G.A. Taylor: Resources, writing-review and editing. L.M. Williamson: Resources, supervision, writing-review and editing. K.L. Mungall: Resources, data curation, methodology, writing-review and editing. E. Chuah: Resources, methodology, writing-review and editing. A.J. Mungall: Resources, methodology, writing-review and editing. R.A. Moore: Resources, methodology, writing-review and editing. J.-M. Lavoie: Data curation, writing-review and editing. S. Yip: Writing-review and editing. H. Lim: Writing-review and editing. D.J. Renouf: Writing-review and editing. S. Sun: Writing-review and editing. R. Holt: Conceptualization, writing-review and editing. S.J.M. Jones: Supervision, funding acquisition, writing-review and editing. M.A. Marra: Supervision, funding acquisition, writing-review and editing. J. Laskin: Conceptualization, supervision, funding acquisition, writing-original draft, writing-review and editing.

The POG program, and subsequent studies, would not be possible without the participation of our patients and families, and support from the POG team. This work was supported by the British Columbia Cancer Foundation, Genome British Columbia (B20POG), Genome Canada and Genome BC (202SEQ, 212SEQ, 12002), Canada Foundation for Innovation (20070, 30981, 30198, 33408, 35444), and the BC Knowledge Development Fund.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Motzer
RJ
,
Tannir
NM
,
McDermott
DF
,
Arén Frontera
O
,
Melichar
B
,
Choueiri
TK
, et al
Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma
.
N Engl J Med
2018
;
378
:
1277
90
.
2.
Wolchok
JD
,
Kluger
H
,
Callahan
MK
,
Postow
MA
,
Rizvi
NA
,
Lesokhin
AM
, et al
Nivolumab plus ipilimumab in advanced melanoma
.
N Engl J Med
2013
;
369
:
122
33
.
3.
Addeo
R
,
Ghiani
M
,
Merlino
F
,
Ricciardiello
F
,
Caraglia
M
. 
CheckMate 141 trial: all that glitters is not gold
.
Expert Opin Biol Ther
2019
;
19
:
169
171
.
4.
Adams
S
,
Schmid
P
,
Rugo
HS
,
Winer
EP
,
Loirat
D
,
Awada
A
, et al
Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase II KEYNOTE-086 study
.
Ann Oncol
2019
;
30
:
397
404
.
5.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
6.
Ready
N
,
Hellmann
MD
,
Awad
MM
,
Otterson
GA
,
Gutierrez
M
,
Gainor
JF
, et al
First-line nivolumab plus ipilimumab in advanced non–small-cell lung cancer (checkmate 568): outcomes by programmed death ligand 1 and tumor mutational burden as biomarkers
.
J Clin Oncol
2019
;
37
:
992
1000
.
7.
U.S. Food and Drug Administration
. 
FDA approves pembrolizumab for adults and children with TMB-H solid tumors
; 
2020
.
Available from
: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-pembrolizumab-adults- and-children-tmb-h-solid-tumors.
8.
Planchard
D
,
Popat
S
,
Kerr
K
,
Novello
S
,
Smit
EF
,
Faivre-Finn
C
, et al
Metastatic non-small cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up.
Ann Oncol
2018
;
29
:
iv192
iv237
.
9.
Anagnostou
V
,
Niknafs
N
,
Marrone
K
,
Bruhm
DC
,
White
JR
,
Naidoo
J
, et al
Multimodal genomic features predict outcome of immune checkpoint blockade in non-small-cell lung cancer
.
Nat Cancer
2020
;
1
:
99
111
.
10.
Cristescu
R
,
Mogg
R
,
Ayers
M
,
Albright
A
,
Murphy
E
,
Yearley
J
, et al
Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy
.
Science
2018
;
362
:
eaar3593
.
11.
Rizvi
NA
,
Hellmann
MD
,
Snyder
A
,
Kvistborg
P
,
Makarov
V
,
Havel
JJ
, et al
Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer
.
Science
2015
;
348
:
124
8
.
12.
Uryvaev
A
,
Passhak
M
,
Hershkovits
D
,
Sabo
E
,
Bar-Sela
G
. 
The role of tumor-infiltrating lymphocytes (TILs) as a predictive biomarker of response to anti-PD1 therapy in patients with metastatic non-small cell lung cancer or metastatic melanoma
.
Med Oncol
2018
;
35
:
25
.
13.
Laskin
J
,
Jones
S
,
Aparicio
S
,
Chia
S
,
Ch'ng
C
,
Deyell
R
, et al
Lessons learned from the application of whole-genome analysis to the treatment of patients with advanced cancers
.
Cold Spring Harb Mol Case Stud
2015
;
1
:
a000570
.
14.
Pleasance
E
,
Titmuss
E
,
Williamson
L
,
Kwan
H
,
Culibrk
L
,
Zhao
EY
, et al
Pan-cancer analysis of advanced patient tumors reveals interactions between therapy and genomic landscapes
.
Nature Cancer
2020
;
1
:
452
68
.
15.
McShane
LM
,
Sauerbrei
W
,
Taube
SE
,
Gion
M
,
Clark
GM
. 
REporting recommendations for tumour MARKer prognostic studies (REMARK)
.
Br J Cancer
2005
;
93
:
387
91
.
16.
Grewal
JK
,
Eirew
P
,
Jones
M
,
Chiu
K
,
Tessier-Cloutier
B
,
Karnezis
AN
, et al
Detection and genomic characterization of a mammary-like adenocarcinoma
.
Cold Spring Harb Mol Case Stud
2017
;
3
:
a002170
.
17.
Jones
MR
,
Lim
H
,
Shen
Y
,
Pleasance
E
,
Ch'ng
C
,
Reisle
C
, et al
Successful targeting of the NRG1 pathway indicates novel treatment strategy for metastatic cancer
.
Ann Oncol
2017
;
28
:
3092
7
.
18.
Reisle
C
,
Mungall
KL
,
Choo
C
,
Paulino
D
,
Bleile
DW
,
Muhammadzadeh
A
, et al
MAVIS: merging, annotation, validation, and illustration of structural variants
.
Bioinformatics
2019
;
35
:
515
7
.
19.
Flicek
P
,
Amode
MR
,
Barrell
D
,
Beal
K
,
Billis
K
,
Brent
S
, et al
Ensembl 2014
.
Nucleic Acids Res
2014
;
42
:
D749
755
.
20.
Jones
SJ
,
Laskin
J
,
Li
YY
,
Griffith
OL
,
An
J
,
Bilenky
M
, et al
Evolution of an adenocarcinoma in response to selection by targeted kinase inhibitors
.
Genome Biol
2010
;
11
:
R82
.
21.
Ha
G
,
Roth
A
,
Lai
D
,
Bashashati
A
,
Ding
J
,
Goya
R
, et al
Integrative analysis of genome-wide loss of heterozygosity and monoallelic expression at nucleotide resolution reveals disrupted pathways in triple-negative breast cancer
.
Genome Res
2012
;
22
:
1995
2007
.
22.
Niu
B
,
Ye
K
,
Zhang
Q
,
Lu
C
,
Xie
M
,
McLellan
MD
, et al
MSIsensor: microsatellite instability detection using paired tumor-normal sequence data
.
Bioinformatics
2014
;
30
:
1015
6
.
23.
Robinson
JT
,
Thorvaldsdóttir
H
,
Wenger
AM
,
Zehir
A
,
Mesirov
JP
. 
Variant review with the integrative genomics viewer
.
Cancer Res
2017
;
77
:
e31
e4
.
24.
Butterfield
YS
,
Kreitzman
M
,
Thiessen
N
,
Corbett
RD
,
Li
Y
,
Pang
J
, et al
JAGuaR: junction alignments to genome for RNA-seq reads
.
PLoS One
2014
;
9
:
e102398
.
25.
Newman
AM
,
Liu
CL
,
Green
MR
,
Gentles
AJ
,
Feng
W
,
Xu
Y
, et al
Robust enumeration of cell subsets from tissue expression profiles
.
Nat Methods
2015
;
12
:
453
7
.
26.
Campbell
BB
,
Light
N
,
Fabrizio
D
,
Zatzman
M
,
Fuligni
F
,
de Borja
R
, et al
Comprehensive analysis of hypermutation in human cancer
.
Cell
2017
;
171
:
1042
56
.
27.
Snyder
A
,
Nathanson
T
,
Funt
SA
,
Ahuja
A
,
Buros Novik
J
,
Hellmann
MD
, et al
Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: an exploratory multi-omic analysis
.
PLoS Med
2017
;
14
:
e1002309
.
28.
Riaz
N
,
Havel
JJ
,
Makarov
V
,
Desrichard
A
,
Urba
WJ
,
Sims
JS
, et al
Tumor and microenvironment evolution during immunotherapy with nivolumab
.
Cell
2017
;
171
:
934
49
.
29.
Liu
D
,
Schilling
B
,
Liu
D
,
Sucker
A
,
Livingstone
E
,
Jerby-Arnon
L
, et al
Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma
.
Nat Med
2019
;
25
:
1916
27
.
30.
Rooney
MS
,
Shukla
SA
,
Wu
CJ
,
Getz
G
,
Hacohen
N
. 
Molecular and genetic properties of tumors associated with local immune cytolytic activity
.
Cell
2015
;
160
:
48
61
.
31.
Ayers
M
,
Lunceford
J
,
Nebozhyn
M
,
Murphy
E
,
Loboda
A
,
Kaufman
DR
, et al
IFN-γ–related mRNA profile predicts clinical response to PD-1 blockade
.
J Clin Invest
2017
;
127
:
2930
40
.
32.
Fu
J
,
Li
K
,
Zhang
W
,
Wan
C
,
Zhang
J
,
Jiang
P
, et al
Large-scale public data reuse to model immunotherapy response and resistance
.
Genome Med
2020
;
12
:
21
.
33.
Jiang
P
,
Gu
S
,
Pan
D
,
Fu
J
,
Sahu
A
,
Hu
X
, et al
Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response
.
Nat Med
2018
;
24
:
1550
8
.
34.
Greillier
L
,
Tomasini
P
,
Barlesi
F
. 
The clinical utility of tumor mutational burden in non-small cell lung cancer
.
Transl Lung Cancer Res
2018
;
7
:
639
46
.
35.
Fancello
L
,
Gandini
S
,
Pelicci
PG
,
Mazzarella
L
. 
Tumor mutational burden quantification from targeted gene panels: major advancements and challenges
.
J Immunother Cancer
2019
;
7
:
183
.
36.
Klempner
SJ
,
Fabrizio
D
,
Bane
S
,
Reinhart
M
,
Peoples
T
,
Ali
SM
, et al
Tumor mutational burden as a predictive biomarker for response to immune checkpoint inhibitors: a review of current evidence
.
Oncologist
2020
;
25
:
e147
e59
.
37.
Haricharan
S
,
Bainbridge
MN
,
Scheet
P
,
Brown
PH
. 
Somatic mutation load of estrogen receptor-positive breast tumors predicts overall survival: an analysis of genome sequence data
.
Breast Cancer Res Treat
2014
;
146
:
211
20
.
38.
Miller
A
,
Asmann
Y
,
Cattaneo
L
,
Braggio
E
,
Keats
J
,
Auclair
D
, et al
High somatic mutation and neoantigen burden are correlated with decreased progression-free survival in multiple myeloma
.
Blood Cancer J
2017
;
7
:
e612
.
39.
Ali
HR
,
Provenzano
E
,
Dawson
S-J
,
Blows
FM
,
Liu
B
,
Shah
M
, et al
Association between CD8+ T-cell infiltration and breast cancer survival in 12 439 patients
.
Ann Oncol
2014
;
25
:
1536
43
.
40.
Zhang
N
,
Bevan
MJ
. 
CD8+ T cells: foot soldiers of the immune system
.
Immunity
2011
;
35
:
161
8
.
41.
Brown
JM
,
Recht
L
,
Strober
S
. 
The promise of targeting macrophages in cancer therapy
.
Clin Cancer Res
2017
;
23
:
3241
50
.
42.
Kulkarni
A
,
Chandrasekar
V
,
Natarajan
SK
,
Ramesh
A
,
Pandey
P
,
Nirgud
J
, et al
A designer self-assembled supramolecule amplifies macrophage immune responses against aggressive cancer
.
Nat Biomed Eng
2018
;
2
:
589
99
.
43.
Jayasingam
SD
,
Citartan
M
,
Thang
TH
,
Mat Zin
AA
,
Ang
KC
,
Ch'ng
ES
. 
Evaluating the polarization of tumor-associated macrophages into M1 and M2 phenotypes in human cancer tissue: technicalities and challenges in routine clinical practice
.
Front Oncol
2020
;
9
:
1512
.
44.
Gandhi
L
,
Rodríguez-Abreu
D
,
Gadgeel
S
,
Esteban
E
,
Felip
E
,
De Angelis
F
, et al
Pembrolizumab plus chemotherapy in metastatic non–small-cell lung cancer
.
N Engl J Med
2018
;
378
;
2078
92
.
45.
Shen
X
,
Zhao
B
. 
Efficacy of PD-1 or PD-L1 inhibitors and PD-L1 expression status in cancer: meta-analysis
.
BMJ
2018
;
362
:
k3529
.
46.
Hellmann
MD
,
Nathanson
T
,
Rizvi
H
,
Creelan
BC
,
Sanchez-Vega
F
,
Ahuja
A
, et al
Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer
.
Cancer Cell
2018
;
33
:
843
52
.
47.
Hellmann
MD
,
Ciuleanu
T-E
,
Pluzanski
A
,
Lee
JS
,
Otterson
GA
,
Audigier-Valette
C
, et al
Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden
.
N Engl J Med
2018
;
378
:
2093
104
.
48.
Gettinger
S
,
Choi
J
,
Hastings
K
,
Truini
A
,
Datar
I
,
Sowell
R
, et al
Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer
.
Cancer Discov
2017
;
7
:
1420
35
.
49.
Jenkins
RW
,
Barbie
DA
,
Flaherty
KT
. 
Mechanisms of resistance to immune checkpoint inhibitors
.
Br J Cancer
2018
;
118
:
9
16
.
50.
Ionescu
DN
,
Downes
MR
,
Christofides
A
,
Tsao
MS
. 
Harmonization of PD-L1 testing in oncology: a Canadian pathology perspective
.
Curr Oncol
2018
;
25
:
e209
e16
.
51.
Zaretsky
JM
,
Garcia-Diaz
A
,
Shin
DS
,
Escuin-Ordinas
H
,
Hugo
W
,
Hu-Lieskovan
S
, et al
Mutations associated with acquired resistance to PD-1 blockade in melanoma
.
N Engl J Med
2016
;
375
:
819
29
.
52.
Bailey
MH
,
Tokheim
C
,
Porta-Pardo
E
,
Sengupta
S
,
Bertrand
D
,
Weerasinghe
A
, et al
Comprehensive characterization of cancer driver genes and mutations
.
Cell
2018
;
174
:
1034
5
.
53.
Merino
DM
,
McShane
LM
,
Fabrizio
D
,
Funari
V
,
Chen
S-J
,
White
JR
, et al
Establishing guidelines to harmonize tumor mutational burden (TMB): in silico assessment of variation in TMB quantification across diagnostic platforms: phase I of the Friends of Cancer Research TMB Harmonization Project
.
J Immunother Cancer
2020
;
8
:
e000147
.