Abstract
Early studies shed light on the immune suppression of immune checkpoint molecules in the cancer microenvironment, with later studies applying immune checkpoint blockade (ICB) in treatment of various malignancies. Despite the encouraging efficacy of ICBs in a substantial subset of cancer patients, the treatment response varies. Gene mutations of both tumor cells and immune cells in the tumor microenvironment have recently been identified as potential predictors of the ICB response. Recent developments in gene expression profiling of tumors have allowed identification of a panel of mutated genes that may affect tumor cell response to ICB treatment. In this review, we discuss the association of the ICB response with gene expression and mutation profiles in tumor cells, which it is hoped will help to optimize the clinical application of ICBs in cancer patients.
Keywords: gene mutation, immune checkpoint blockade, tumor
Introduction
With the emergence of immune checkpoint blockades, the field of anti-cancer therapy has currently shifted its focus to antagonizing agents that target inhibitory signaling molecules on tumor and immune cells.1,2 Once the immune suppression of immune checkpoints is released by immune checkpoint blockades (ICBs), the immune system is then activated, which can be best characterized with the inflammatory response observed at tumor sites. In addition to the most intensively studied ICBs such as the antibodies targeting CTLA-4 (cytotoxic T lymphocyte-associated antigen-4),3,4 PD-1 (programmed cell death-1),5–7 and PD-L1 (PD-1 ligand),8,9 a wide range of other ICBs have also exhibited striking clinical benefits in cancer patients. An increasing number of ICBs are currently under evaluation for treatment of various advanced diseases, many of which are approved by the Food and Drug Administration each year.10–14
Despite the considerable clinical benefit of ICBs in a substantial subset of cancer patients, their efficacy varies. It is of paramount importance to identify predictive biomarkers for ICB responses. Previous reports have suggested the accumulation of gene mutations as a tumor hallmark. Although the mutational burden varies widely both within and among cancer types,15 a subset of cancer patients has demonstrated similar gene mutation profiles. It is therefore conceivable that gene alterations play a functional role in tumor response of ICBs.
Application of genomic and transcriptomic analyses can facilitate accessible gene expression profiling of tumors to identify potential prognosis predictors.16–19 Tumor-related genes described thus far are mostly involved in cytokine and chemokine signaling pathways. Many of these genes including EREG and AREG, have been reported to predict response to chemotherapy in cancer patients20,21 and their differential expression may also affect the treatment response to ICBs.22,23 Compared with the traditional quantitative immunohistochemical (IHC) method, the identification of gene expression can establish a more comprehensive response profile, in that it simultaneously evaluates substantial parameters including expression of immune checkpoint molecules and inflammatory genes. However, the gene expression assay fails to present spatial and structural details that may also serve as prognostic factors.24 The integration of multiple detecting platforms is therefore warranted to provide comprehensive data to predict ICB response.24
In this review, we briefly discuss the association of ICB response with gene expression and mutation profiles in tumor cells. With a growing body of study on the underlying mechanism of the association, we also discuss the intratumoral mutational burdens and mutational heterogeneity, which aids the stratification of ICB treatment to minimize the potential treatment resistance.
Response to ICB in the context of tumor cell gene expression
Intense investigation is currently under way into potential predictive biomarkers of the response to ICBs. Akiyama et al. established a gene panel consisting of 164 immune therapy response-associated genes from 1000 tumors based on whole-exome sequencing and gene expression profiling.25 Multiple molecular predictors have been proposed of tumor response to the ICB. High levels of PD-L1 expression, IFN-γ induced gene expression in tumor cells, and CD8+ T cell infiltrates have all been detected in patients responsive to immune checkpoint therapy.26,27 Some studies of tumor biopsies have suggested a predictive role for CD8+, CD4+, PD-1+, and PD-L1+ in tumor cells in terms of the therapeutic response to ICBs.28,29
Tumor expression of PD-L1
Based on the PD-1/PD-L1 signaling pathway, it is conceivable that anti-PD-1/anti-PD-L1 therapy is largely associated with expression of PD-L1 in tumor cells or tumor-infiltrating immune cells. Previous studies have demonstrated a correlation between tumor expression of PD-L1 and response to PD-1 inhibitors in some cancer types.5,6,27,30,31 Expression of PD-L1 on tumor cells varies from absent in Merkel cell carcinomas to 100% in liposarcomas and chondrosarcomas.32 Meanwhile, triple-negative breast cancers and colon cancers with high microsatellite instability (MSI) have significantly higher PD-L1 expression than that seen in non-triple-negative breast cancers and microsatellite stable tumors, respectively.32 In a clinical trial evaluating patients treated with the anti-PD-1 agent pembrolizumab, the higher expression of PD-L1 on tumor cells was associated with a better outcome compared with tumors with low PD-L1 expression.33 In this trial, a proportion score of ≥50% was used as the cut-off value, referred to as the PD-L1 expression on at least 50% of the tumor cells. A response rate of around 90% to nivolumab has been observed in patients with Hodgkin’s lymphoma with amplified PD-L1/PD-L2.11 Moreover, higher levels of CTLA-4 mRNA are also found to indicate better response to both anti-CTLA-4 and anti-PD-L1 therapy.22,27 The PD-L1 copy number gains in non-small-cell lung cancer (NSCLC) cells display higher expression of PD‑L1, which can potentially serve as a predictor of response to anti-PD-1/PD-L1 therapy. However, PD-L2 copy number gains fail to result in augmented PD-L2 expression.34 Kim et al. later identified nine genes related to immune checkpoints, including PD-1, PD-L1, and CTLA-4, the expression of which is positively correlated with the number of infiltrating lymphocytes in metastatic breast cancers, suggesting a potential predictive function of these gene markers.35 Taken together, this evidence suggests that expression of PD-L1/PD-L2 and CTLA-4 can be used as a target for gene expression-based immunotherapy.
However, in some reports, tumors classified as PD-L1-positive did not respond and the baseline PD-L1 status was not an independent predictor of treatment response.14,27,36,37 Such differing reports can probably be attributed to use of different tumor types and histologic categories, and heterogeneity of PD-L1 expression within the tumors themselves. Differences may also reflect the limitations of IHC staining as the detection method used for PD-L1 expression, given that IHC staining can be performed on different staining platforms with different antibodies. One such example is the comparison staining performed by Smith et al.38 They compared the performance of two PD-L1 clones (SP263 and E1L3N) and found that SP263 IHC assays had higher sensitivity and a wider dynamic range than E1L3N assays. On the other hand, Carbognin et al. found that the cut-off value for PD-L1 positive melanomas could have an impact on study conclusions.39
Despite the dramatic efficacy of PD-1/PD-L1 blockades, some patients fail to respond to the initial PD-1/PD-L1 therapy, which is known as primary resistance.7,40 Moreover, a subset of responders eventually develop acquired resistance after initial treatment. Therefore, it is of paramount importance to identify biomarkers related to ICB resistance. PD-1/PD-L1 blockade responses can be greatly impacted by biomarkers such as tumor neoantigens and MSI, which we briefly describe in the following discussion. The tumor PD-L1 expression serves as another biomarker to predict responses to PD-1/PD-L1 blockade, the clinical application of which needs in-depth consideration for tumor types.41 The tumor PD-L1 expression has been identified as an acquired resistance mechanism for tumor cells in response to immunotherapies.1,42 Importantly, it has to be addressed that one single biomarker is insufficient to predict drug resistance and combining different biomarkers is therefore a valuable option for prediction.
Tumor expression of MHC
Given that T cells recognize tumor cells through recognition of peptides that bind to the major histocompatibility complex (MHC), theoretically, decreasing expression of MHC I would promote tumor survival and reduce ICB efficacy. An early study by Restifo et al. found that melanoma patients with β2 microglobulin deficiency, which led to the incompleteness of MHC I, did not respond to T-cell-based immunotherapy.43 A growing body of literature has reinforced this finding that the loss of MHC I expression on tumor-infiltrating immune cells leads to the immune evasion of melanoma cells and their acquired resistance to anti-PD-1 agents following the initial response.44–46 On the other hand, MHC II expression on melanoma cells has also been found to predict response to anti-PD-1 treatment,47 and expression of MHC II on some melanoma cell lines can be restored with IFN-γ treatment.48
Response to ICB in the context of tumor cell gene alterations
Multiple oncogenic events have been reported to contribute to acquisition of the phenotype and malignant properties of cancer. Developments in techniques for identification of genetic abnormalities, have allowed identification of a subset of cancer patients with similar gene mutation profiles. It is therefore conceivable that gene alterations play a functional role in tumor response to ICBs. Although no clear associations have been established, this review focuses on the potential gene mutations that are predictive of therapeutic sensitivity to ICBs.
Ras and RAF gene mutation
The Ras/Raf/MEK/MAPK pathway is involved in proliferation of both normal and tumor cells, and is therefore associated with tumorigenesis and tumor progression.49 The RAS oncogene family includes KRAS, NRAS, and HRAS, which have all been characterized as potential predictors of anti-epidermal growth factor receptor (EGFR) treatment response in colorectal cancer.50 KRAS mutations occur in approximately 20% of NSCLC,51 and are associated with poor prognosis in cancer patients.52,53 Patients with KRAS mutations exhibit intrinsic resistance to anti-EGFR treatment,54 but are sensitive to MEK/ERK inhibitors such as selumetinib.55,56 Previous studies have shown that patients with KRAS mutations, especially those with concomitant TP53 mutations, had increased PD-L1 expression and remarkable clinical response to PD-1 inhibitors.57 Compared with patients with wild-type TP53, there was a significantly higher proportion of patients with mutations in TP53 who exhibited high PD-1 expression (10.9% compared with 34.2%, P = 0.023). As for NRAS mutations, patients with NRAS mutant melanoma displayed a significantly worse overall survival of 8.2 months compared with patients with wild-type BRAF/NRAS (15.1 months, P = 0.004).58 And it has been reported that patients with NRAS mutation-positive melanomas demonstrate better disease control (median progression-free survival 4.1 vs. 2.9 months, P = 0.09) when treated with CTLA-4 and PD-1 antibodies,59 and prolonged overall survival (OS, 12.1 vs. 8.03 months) when treated with ipilimumab.60 Kirchberger et al. suggested that immune checkpoint inhibition induced encouraging responses in both NRAS-mutated and NRAS wild-type melanoma patients; however, worse OS was observed in the case of NRAS mutation, but this could be overcome by additional use of MEK inhibitors.61
The predictive value of BRAF mutation in the context of treatment response to ICBs remains incompletely defined. BRAF mutation status appears to be irrelevant of disease progression or OS,62,63 but some reports have identified BRAF mutation as a negative prognostic indicator.62,64 Although immune checkpoint inhibitors have significant benefitted cancer patients in randomized clinical trials,3,4,14,65,66 the BRAF mutation status is independent of the response difference.67,68 A recent report, however, suggested that the BRAF mutation is moderately related to worse survival in melanoma patients treated with ipilimumab.69
EGFR gene mutation
EGFR mutations are often seen in patients with lung cancer, and different mutation types can potentially affect the treatment response to anti-cancer therapies.70–72 The T790M EGFR mutation has been reported to account for more than half of patients resistant to first-line EGFR tyrosine kinase inhibitors.73 Moreover, T790M mutation status is also associated with PD-L1 expression levels. Previous studies have detected a lower rate of PD-L1 positivity in patients with EGFR mutations.74 And in cancer patients with PD-L1 overexpression who are treated with pembrolizumab, those with EGFR mutations have exhibited significantly shorter OS than those with wild-type EGFR.75 In patients with EGFR mutations, increased PD-L1 expression indicates higher response rates to immuno-oncology agents.76,77 IHC results have suggested a high expression profile of PD-L1 and PD-1 in tumor specimens of patients with EGFR-mutant NSCLC, suggesting that PD-1/PD-L1 immunotherapy could potentially benefit such patients.78 Lee et al. reported that compared with chemotherapy, immune checkpoint inhibitors failed to bring survival benefits in patients with EGFR-mutant NSCLC.79 Moreover, anti-PD-1/PD-L1 therapy does not achieve a significant survival improvement in patients with EGFR mutations.80 However, compared with ICB monotherapy, the concomitant use of PD-1 and CTLA-4 inhibitors can augment the immunogenicity of EGFR-mutant tumors and increase the treatment response.81 The 2-year overall survival of advanced melanoma patients was 63.8% in a group of patients receiving nivolumab plus ipilimumab versus 53.6% in a group receiving ipilimumab monotherapy.82
Janus kinase (JAK) gene mutation
The JAK family includes four members, JAK1, JAK2, JAK3, and TYK2 (non-receptor protein-tyrosine kinase 2).83 Multiple studies have explored the role of a gene locus adjacent to JAK2, the 9p24.1, the amplicon of which is often seen in both solid tumors and hematologic malignancies.84,85 The emergence of JAK2 at the 9p24.1 gene site is crucial to PD-L1 expression, as JAK2 signaling has been found to increase PD-L1 expression.85 A study has identified the synergistic amplification of PDL1/PD-L2/JAK2 as a potential predictor for the response to immune checkpoint therapy.86 Furthermore, amplifications of the PD-L1 and JAK2 genes are mutually correlated, as observed after use of the JAK2 inhibitor TG-101348 which can cause a decrease in PD-L1 protein.87 Both somatic and germline genomic alterations of JAK3 have been reported to promote PD-L1 activation in lung cancer cells and potentially impact their response to the PD-L1 immune checkpoint therapy.88
MSI
Resulting from an intrinsic deficiency in DNA mismatch repair (MMR), MSI often occurs throughout the genome in colorectal cancers.89 Accumulation of DNA alterations is an essential step towards carcinogenesis, which reflects the somatic and germline variants involved in DNA damage and repair. Previous studies have identified use of MSI as a potential biomarker to predict sensitivity to anti-PD-1 therapy as a less than ideal treatment strategy in colorectal cancers.90
A phase II study suggested that the anti-PD-1 agent nivolumab induced encouraging anti-tumor activity in patients with MMR-deficient cancers.91 A recent study presented a case of a cancer patient with high levels of MSI who demonstrated a striking response to another PD-1 inhibitor, pembrolizumab.92 In this case, after the patient received the initial pembrolizumab treatment, her chest wall lesion was significantly softened with tumor tissue necrosis. There are also cases of other tumors, such as glioblastoma and extrahepatic cholangiocarcinoma, which lacked the expression of potential response predictors for PD-L1 inhibitors.93,94 In a phase II clinical trial,95 a more robust response to pembrolizumab was observed in patients with MMR-deficient colorectal cancer than in patients with proficient MMR, suggesting that cancers with intensive somatic mutations caused by MMR deficiency are susceptible to immune checkpoint blockade. A growing body of literature has reported similar results in an expanding tumor type profile,95–99 including the left frontal glioblastoma with DNA-repair defects in which ICB treatment results in certral nervous system (CNS) immune activation.100
Other gene mutations
Current knowledge on gene mutations as biomarkers predicting ICB response represents only the tip of the iceberg. Recent publications have highlighted the alterations of other potential predictive genes. A significant proportion of cancer patients with targetable mutations in SMO, DDR2, FGFR1, PTCH1, FGFR2, and MET have been reported to be eligible for certain checkpoint inhibitors.101 Matsuo et al. identified a subset of patients with BRCA mutant epithelial ovarian, fallopian tubal, and primary peritoneal cancers who responded to nivolumab monotherapy.102 Moreover, gene mutations of DNA polymerase epsilon (POLE) are also correlated with increased expression of certain immune checkpoint genes, indicating that cancers with POLE mutations are candidate predictors for immune checkpoint therapy.103 More recently, Miao et al. found that loss-of-function mutations of the PBRM1 gene in clear cell renal cell carcinoma can lead to remarkable responsiveness to immune checkpoint therapy.104 PTEN mutations and decreased expression of neoantigen genes have also been demonstrated as having potential predictive value for resistance to immune checkpoint therapy.105 Ongoing studies are likely to result in development of an expanding repertoire of gene mutations.
Mechanism of the correlation between ICB response and gene mutations
Tumor neoantigens and mutational load
Neoantigens are also referred to as the proteins resulting from DNA mutations within tumor cells that can be recognized as foreign proteins by the immune system.106–113 Previous reports have emphasized that tumors with higher mutational frequency generate larger numbers of neoantigens than tumors with lower mutational frequency.114 When neoantigens are released from tumor cells, cytotoxic T lymphocytes engage the tumor microenvironment, exert killer functions on the tumor cells, and induce tumor cell apoptosis, which in turn promotes release of more neoantigens and enhances the anti-tumor activities of the immune system.115 To reach maximum anti-tumor efficacy, the anti-tumor therapy needs to overcome the suppressive tumor microenvironment including the inhibitory immune checkpoint molecules. Figure 1 illustrates the interaction between tumor neoantigens, T cells, and antigen-presenting cells.
Figure 1.
Simplified overview of the interaction between tumor neoantigens, T cells, and antigen-presenting cells (APCs). MHC, class I major histocompatibility complex; TCR, T cell receptor.
To understand the key to the success of ICBs, it is important yet challenging to identify antigens uniquely expressed on tumor cells and not on normal tissues. One approach is application of tumor mutational burden to represent mutated peptides on T cells that exist only on tumor cells and not the normal genome.114 Correlations of durable responses to ICBs with mutational load and neoantigens have long been confirmed,106,114 with some evidence suggesting that the mutational load may be more accurate than PD-L1 IHC in predicting the presence of clinical variables such as the number of tumor infiltrating lymphocytes.106 Sequencing of tumor genomes can be used to predict the clinical benefit of ICBs because cancer patients with higher mutational loads have demonstrated improved OS after receiving ICBs.116
Previous studies on cancer-related gene profiles have demonstrated a significantly higher tumor mutation burden in immunotherapy responders compared with non-responders.9,117 On the other hand, Hellmann et al. used whole-exome sequencing to evaluate the therapeutic efficacy of PD-1 plus CTLA-4 blockade in NSCLC patients, and found that patients with higher tumor mutation had improved objective response and progression-free survival.118 It has been reported that melanoma cells are prone to damage by ultraviolet radiation, which causes an accumulating number of mutations in the melanocyte genome, leading to a higher level of mutational load.119–121 In this subset of melanoma patients, higher mutational load is associated with better response to anti-CTLA4 therapy.22 However, in one subtype of melanoma, the desmoplastic melanoma, no significant difference was detected between mutational loads in PD-1 blockade responders and non-responders.122
Although a higher mutational load is reported to be correlated with a durable clinical response to CTLA-4 blockade, tumor mutational load alone is not sufficient to indicate the clinical benefit, because not all tumors with high mutational loads respond to immune checkpoint therapy.117 Furthermore, the mutation burden varies dramatically within tumors and among different tumor types, reflecting vast differences in the DNA-damage-repairing abilities of different tumors.15
Intratumoral mutational heterogeneity
In addition to high levels of neoantigens, therapeutic sensitivity to ICBs is also associated with low intratumoral heterogeneity.113 Tumors with DNA damage response-deficiency exhibit random gene mutations, leading to profoundly high intratumoral heterogeneity.123 High levels of mutational heterogeneity potentially enhance the interaction between tumor antigens and MHC molecules, and consequently induce the antigen presentation to T cells. Moreover, heterogeneous gene mutations increase the chance of tumor cells being identified by T cells by widening the T-cell killing repertoire, including recognition of hidden neoantigens by ICBs.124 Safonov et al. recently suggested that the mutational heterogeneity is negatively associated with some immune metagenes in breast cancers, and that mutational heterogeneity leads to minimal immune cell infiltration, pointing to an escape mechanism of tumor cell from immune surveillance.125 Therefore, more immunotherapy strategies are warranted to activate the anti-cancer immune activities against a genetically diverse neoplastic population.
Perspectives
An accumulation of new literature in recent years has reported therapeutic resistance to ICBs in cancer patients, which is one of the main obstacles in application of ICBs. Several mechanisms for such resistance have been proposed including PTEN loss which reduces the intratumoral infiltration and tumor killing of T cells, leading to resistance to anti-PD-1 or anti-CTLA-4 treatment in this subset of patients.126 Immune resistance has also been detected in melanoma patients with the loss of β2-microglobulin.43 Using whole genome shotgun (WGS) technology, patients failing to respond to anti-PD-1 therapy display increased expression of genes involved in the carcinogenesis process such as epithelial mesenchymal transition.127 All these efforts provide new insights into the mechanism of drug resistance, suggesting that the gene mutation may be an attractive biomarker for predicting therapeutic efficacy.
Another challenge is how we broaden the utility of gene mutations in immune checkpoint therapy. One avenue is to identify more potential mutated genes that would have an impact on treatment response. One such strategy, genomic analysis, has been refined in recent decades and introduces new opportunities for cancer treatment. Genomic analysis technology, such as next-generation sequencing, can help to identify expressed mutations of tumors and predicts candidate peptides that may still remain after ICB treatment.128 Another potential option is improvement of the cancer vaccine concept. Cancer vaccines involve injection of cancer-specific elements into patients to induce anti-tumor immune responses. To achieve an adequate and durable T cell response, multiple vaccinations and incorporation of MHC class II peptides are required.129,130 Study on these subjects will undoubtedly provide new perspectives for improvement of the ICB treatment response.
Funding
This work is supported by the National Natural Science Foundation of China (No. 81602492) and the National Key Research and Development Program of China (No. 2016YFA0201402). This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
Conflict of interest statement
None declared.
References
- 1. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 2015;27:450–61. 10.1016/j.ccell.2015.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature 2011;480:480–9. 10.1038/nature10673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711–23. 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011;364:2517–26. 10.1056/NEJMoa1104621. [DOI] [PubMed] [Google Scholar]
- 5. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443–54. 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Brahmer JR, Drake CG, Wollner I, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 2010;28:3167–75. 10.1200/JCO.2009.26.7609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Hamid O, Robert C, Daud A, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 2013;369:134–44. 10.1056/NEJMoa1305133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012;366:2455–65. 10.1056/NEJMoa1200694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 2016;387:1909–20. 10.1016/s0140-6736(16)00561-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015;372:320–30. 10.1056/NEJMoa1412082. [DOI] [PubMed] [Google Scholar]
- 11. Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med 2015;372:311–9. 10.1056/NEJMoa1411087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Ferris RL, Blumenschein G Jr., Fayette J, et al. Nivolumab vs investigator’s choice in recurrent or metastatic squamous cell carcinoma of the head and neck: 2-year long-term survival update of CheckMate 141 with analyses by tumor PD-L1 expression. Oral Oncol 2018;81:45–51. 10.1016/j.oraloncology.2018.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Reck M, Rodríguez-Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016;375:1823–33. 10.1056/NEJMoa1606774. [DOI] [PubMed] [Google Scholar]
- 14. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 2015;373:23–34. 10.1056/NEJMoa1504030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Lawrence MS, Stojanov P, Mermel CH, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 2014;505:495–501. 10.1038/nature12912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Dave SS, Wright G, Tan B, et al. Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells. N Engl J Med 2004;351:2159–69. 10.1056/NEJMoa041869. [DOI] [PubMed] [Google Scholar]
- 17. Kratz JR, He J, Van Den Eeden SK, et al. A practical molecular assay to predict survival in resected non-squamous, non-small-cell lung cancer: development and international validation studies. Lancet 2012;379:823–32. 10.1016/S0140-6736(11)61941-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med 2004;351:2817–26. 10.1056/NEJMoa041588. [DOI] [PubMed] [Google Scholar]
- 19. van ‘t Veer LJ, Dai H, van de Vijver MJ, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 2002;415:530–6. 10.1038/415530a. [DOI] [PubMed] [Google Scholar]
- 20. Baker JB, Dutta D, Watson D, et al. Tumour gene expression predicts response to cetuximab in patients with KRAS wild-type metastatic colorectal cancer. Br J Cancer 2011;104:488–95. 10.1038/sj.bjc.6606054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Kim HK, Choi IJ, Kim CG, et al. Three-gene predictor of clinical outcome for gastric cancer patients treated with chemotherapy. Pharmacogenomics J 2012;12:119–27. 10.1038/tpj.2010.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Van Allen EM, Miao D, Schilling B, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 2015;350:207–11. 10.1126/science.aad0095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Ji RR, Chasalow HD, Wang L, et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol Immunother 2012;61:1019–31. 10.1007/s00262-011-1172-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Galon J, Mlecnik B, Bindea G, et al. Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J Pathol 2014;232:199–209. 10.1002/path.4287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Akiyama Y, Kondou R, Iizuka A, et al. Immune response-associated gene analysis of 1,000 cancer patients using whole-exome sequencing and gene expression profiling-Project HOPE. Biomed Res 2016;37:233–42. 10.2220/biomedres.37.233. [DOI] [PubMed] [Google Scholar]
- 26. Hegde PS, Karanikas V, Evers S. The where, the when, and the how of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition. Clin Cancer Res 2016;22:1865–74. 10.1158/1078-0432.CCR-15-1507. [DOI] [PubMed] [Google Scholar]
- 27. Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014;515:563–7. 10.1038/nature14011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Taube JM, Klein A, Brahmer JR, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 2014;20:5064–74. 10.1158/1078-0432.CCR-13-3271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Tumeh PC, Harview CL, Yearly JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568–71. 10.1038/nature13954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Powles T, Eder JP, Fine GD, et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature 2014;515:558–62. 10.1038/nature13904. [DOI] [PubMed] [Google Scholar]
- 31. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015;372:2018–28. 10.1056/NEJMoa1501824. [DOI] [PubMed] [Google Scholar]
- 32. Gatalica Z, Snyder C, Maney T, et al. Programmed cell death 1 (pd-1) and its ligand (PD-L1) in common cancers and their correlation with molecular cancer type. Cancer Epidemiol Biomarkers Prev 2014;23:2965–70. 10.1158/1055-9965.EPI-14-0654. [DOI] [PubMed] [Google Scholar]
- 33. Garon EB, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015;372:2018–28. 10.1056/NEJMoa1501824. [DOI] [PubMed] [Google Scholar]
- 34. Inoue Y, Yoshimura K, Mori K, et al. Clinical significance of PD-L1 and PD-L2 copy number gains in non-small-cell lung cancer. Oncotarget 2016;7:32113–28. 10.18632/oncotarget.8528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Kim JY, Lee E, Park K, et al. Immune signature of metastatic breast cancer: identifying predictive markers of immunotherapy response. Oncotarget 2017;8:47400–11. 10.18632/oncotarget.17653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther 2015;14:847–56. 10.1158/1535-7163.MCT-14-0983. [DOI] [PubMed] [Google Scholar]
- 37. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med 2015;373:123–35. 10.1056/NEJMoa1504627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Smith J, Robida MD, Acosta K, et al. Quantitative and qualitative characterization of two PD-L1 clones: SP263 and E1L3N. Diagn Pathol 2016;11:44. 10.1186/s13000-016-0494-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Carbognin L, Pilotto S, Milella M, et al. Differential activity of nivolumab, pembrolizumab and MPDL3280A according to the tumor expression of programmed death-ligand-1 (PD-L1): sensitivity analysis of trials in melanoma, lung and genitourinary cancers. PLoS One 2015;10:e0130142. 10.1371/journal.pone.0130142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Sharma P, Hu-Lieskovan S, Wargo JA, et al. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017;168:707–23. 10.1016/j.cell.2017.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Chen Q, Li T, Yue W. Drug response to PD-1/PD-L1 blockade: based on biomarkers. Onco Targets Ther 2018;11:4673–83. 10.2147/OTT.S168313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Taube JM, Anders RA, Young GD, et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med 2012;4:127ra137. 10.1126/scitranslmed.3003689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Restifo NP, Marincola FM, Kawakami Y, et al. Loss of functional beta 2-microglobulin in metastatic melanomas from five patients receiving immunotherapy. J Natl Cancer Inst 1996;88:100–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016;375:819–29. 10.1056/NEJMoa1604958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Khong HT, Wang QJ, Rosenberg SA. Identification of multiple antigens recognized by tumor-infiltrating lymphocytes from a single patient: tumor escape by antigen loss and loss of MHC expression. J Immunother 2004;27:184–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Bradley SD, Chen Z, Melendez B, et al. Brafv600e co-opts a conserved mhc class I internalization pathway to diminish antigen presentation and CD8+ t-cell recognition of melanoma. Cancer Immunol Res 2015;3:602–9. 10.1158/2326-6066.CIR-15-0030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Johnson DB, Estrada MV, Salgado R, et al. Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat Commun 2016;7:10582. 10.1038/ncomms10582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Mendez R, Aptsiauri N, Del Campo A, et al. HLA and melanoma: multiple alterations in HLA class I and II expression in human melanoma cell lines from ESTDAB cell bank. Cancer Immunol Immunother 2009;58:1507–15. 10.1007/s00262-009-0701-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Cobb MH, Goldsmith EJ. How MAP kinases are regulated. J Biol Chem 1995;270:14843–46. [DOI] [PubMed] [Google Scholar]
- 50. Allegra CJ, Rumble RB, Hamilton SR, et al. Extended ras gene mutation testing in metastatic colorectal carcinoma to predict response to anti-epidermal growth factor receptor monoclonal antibody therapy: american society of clinical oncology provisional clinical opinion update 2015. J Clin Oncol 2016;34:179–85. 10.1200/JCO.2015.63.9674. [DOI] [PubMed] [Google Scholar]
- 51. Ding L, Getz G, Wheeler DA, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature 2008;455:1069–75. 10.1038/nature07423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Huncharek M, Muscat J, Geschwind JF. K-ras oncogene mutation as a prognostic marker in non-small cell lung cancer: a combined analysis of 881 cases. Carcinogenesis 1999;20:1507–10. [DOI] [PubMed] [Google Scholar]
- 53. Pao W, Wang TY, Riely GJ, et al. KRAS mutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib. PLoS Med 2005;2:e17. 10.1371/journal.pmed.0020017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Linardou H, Dahabreh IJ, Kanaloupiti D, et al. Assessment of somatic k-RAS mutations as a mechanism associated with resistance to EGFR-targeted agents: a systematic review and meta-analysis of studies in advanced non-small-cell lung cancer and metastatic colorectal cancer. Lancet Oncol 2008;9:962–72. 10.1016/S1470-2045(08)70206-7. [DOI] [PubMed] [Google Scholar]
- 55. Solit DB, Garraway LA, Pratilas CA, et al. BRAF mutation predicts sensitivity to MEK inhibition. Nature 2006;439:358–62. 10.1038/nature04304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Garon EB, Finn RS, Hosmer W, et al. Identification of common predictive markers of in vitro response to the Mek inhibitor selumetinib (AZD6244; ARRY-142886) in human breast cancer and non-small cell lung cancer cell lines. Mol Cancer Ther 2010;9:1985–94. 10.1158/1535-7163.MCT-10-0037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Dong ZY, Zhong WZ, Zhang XC, et al. Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma. Clin Cancer Res 2017;23:3012–24. 10.1158/1078-0432.ccr-16-2554. [DOI] [PubMed] [Google Scholar]
- 58. Jakob JA, Bassett RL Jr., Ng CS, et al. NRAS mutation status is an independent prognostic factor in metastatic melanoma. Cancer 2012;118:4014–23. 10.1002/cncr.26724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Johnson DB, Lovly CM, Flavin M, et al. Impact of NRAS mutations for patients with advanced melanoma treated with immune therapies. Cancer Immunol Res 2015;3:288–95. 10.1158/2326-6066.CIR-14-0207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Mangana J, Cheng PF, Schindler K, et al. Analysis of BRAF and nras mutation status in advanced melanoma patients treated with anti-CTLA-4 antibodies: association with overall survival? PLoS One 2015;10:e0139438. 10.1371/journal.pone.0139438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Kirchberger MC, Ugurel S, Mangana J, et al. MEK inhibition may increase survival of NRAS-mutated melanoma patients treated with checkpoint blockade: results of a retrospective multicentre analysis of 364 patients. Eur J Cancer 2018;98:10–6. 10.1016/j.ejca.2018.04.010. [DOI] [PubMed] [Google Scholar]
- 62. Long GV, Menzies AM, Nagrial AM, et al. Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol 2011;29:1239–46. 10.1200/JCO.2010.32.4327. [DOI] [PubMed] [Google Scholar]
- 63. Carlino MS, Haydu LE, Kakavand H, et al. Correlation of BRAF and NRAS mutation status with outcome, site of distant metastasis and response to chemotherapy in metastatic melanoma. Br J Cancer 2014;111:292–9. 10.1038/bjc.2014.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. von Moos R, Seifert B, Simcock M, et al. First-line temozolomide combined with bevacizumab in metastatic melanoma: a multicentre phase II trial (SAKK 50/07). Ann Oncol 2012;23:531–6. 10.1093/annonc/mdr126. [DOI] [PubMed] [Google Scholar]
- 65. Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med 2015;372:2006–17. 10.1056/NEJMoa1414428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Weber JS, D’Angelo SP, Minor D, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol 2015;16:375–84. 10.1016/s1470-2045(15)70076-8. [DOI] [PubMed] [Google Scholar]
- 67. Larkin J, Lao CD, Urba WJ, et al. Efficacy and safety of nivolumab in patients with BRAF V600 mutant and BRAF wild-type advanced melanoma: a pooled analysis of 4 clinical trials. JAMA Oncol 2015;1:433–40. 10.1001/jamaoncol.2015.1184. [DOI] [PubMed] [Google Scholar]
- 68. Shahabi V, Whitney G, Hamid O, et al. Assessment of association between BRAF-V600E mutation status in melanomas and clinical response to ipilimumab. Cancer Immunol Immunother 2012;61:733–7. 10.1007/s00262-012-1227-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Rossfeld K, Hade EM, Gangi A, et al. Metastatic melanoma patients’ sensitivity to ipilimumab cannot be predicted by tumor characteristics. Int J Surg Oncol 2017;2:e43. 10.1097/ij9.0000000000000043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Yang JC, Wu YL, Schuler M, et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol 2015;16:141–51. 10.1016/S1470-2045(14)71173-8. [DOI] [PubMed] [Google Scholar]
- 71. Shim HS, Lee DH, Park EJ, et al. Histopathologic characteristics of lung adenocarcinomas with epidermal growth factor receptor mutations in the International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society lung adenocarcinoma classification. Arch Pathol Lab Med 2011;135:1329–34. 10.5858/arpa.2010-0493-OA. [DOI] [PubMed] [Google Scholar]
- 72. Shigematsu H, Lin L, Takahashi T, et al. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst 2005;97:339–46. 10.1093/jnci/dji055. [DOI] [PubMed] [Google Scholar]
- 73. Kobayashi S, Boggon TJ, Dayaram T, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med 2005;352:786–92. 10.1056/NEJMoa044238. [DOI] [PubMed] [Google Scholar]
- 74. Haratani K, Hayashi H, Tanaka T, et al. Tumor immune microenvironment and nivolumab efficacy in EGFR mutation-positive non-small-cell lung cancer based on T790M status after disease progression during EGFR-TKI treatment. Ann Oncol 2017;28:1532–9. 10.1093/annonc/mdx183. [DOI] [PubMed] [Google Scholar]
- 75. Ramalingam S, Hui R, Gandhi L, et al. P2.39: Long-term OS for patients with advanced NSCLC enrolled in the keynote-001 study of pembrolizumab: track: immunotherapy. J Thorac Oncol 2016;11:S241–2. 10.1016/j.jtho.2016.08.110. [DOI] [Google Scholar]
- 76. Akbay EA, Koyama S, Carretero J, et al. Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov 2013;3:1355–63. 10.1158/2159-8290.CD-13-0310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Azuma K, Ota K, Kawahara A, et al. Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Ann Oncol 2014;25:1935–40. 10.1093/annonc/mdu242. [DOI] [PubMed] [Google Scholar]
- 78. Lin C, Chen X, Li M, et al. Programmed death-ligand 1 expression predicts tyrosine kinase inhibitor response and better prognosis in a cohort of patients with epidermal growth factor receptor mutation-positive lung adenocarcinoma. Clin Lung Cancer 2015;16:e25–35. 10.1016/j.cllc.2015.02.002. [DOI] [PubMed] [Google Scholar]
- 79. Lee CK, Man J, Lord S, et al. Checkpoint inhibitors in metastatic egfr-mutated non-small cell lung cancer—a meta-analysis. J Thorac Oncol 2017;12:403–7. 10.1016/j.jtho.2016.10.007. [DOI] [PubMed] [Google Scholar]
- 80. Jiang Q, Xie M, He M, et al. Anti-PD-1/PD-L1 antibodies versus docetaxel in patients with previously treated non-small-cell lung cancer. Oncotarget 2018;9:7672–83. 10.18632/oncotarget.23584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Hellmann MD, Gettinger SN, Goldman JW, et al. Oral01.03: checkmate 012: safety and efficacy of first-line nivolumab and ipilimumab in advanced NSCLC: topic: medical oncology. J Thorac Oncol 2016;11:S250–1. 10.1016/j.jtho.2016.09.008. [DOI] [Google Scholar]
- 82. Hodi FS, Chesney J, Pavlick AC, et al. Combined nivolumab and ipilimumab versus ipilimumab alone in patients with advanced melanoma: 2-year overall survival outcomes in a multicentre, randomised, controlled, phase 2 trial. Lancet Oncol 2016;17:1558–68. 10.1016/S1470-2045(16)30366-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Verstovsek S. Janus-activated kinase 2 inhibitors: a new era of targeted therapies providing significant clinical benefit for Philadelphia chromosome-negative myeloproliferative neoplasms. J Clin Oncol 2011;29:781–3. 10.1200/JCO.2010.33.4508. [DOI] [PubMed] [Google Scholar]
- 84. Cancer Genome Atlas Research Network . Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014;513:202–9. 10.1038/nature13480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Green MR, Monti S, Rodig SJ, et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood 2010;116:3268–77. 10.1182/blood-2010-05-282780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Gupta S, Vanderbilt CM, Cotzia P, et al. JAK2, PD-L1, and PD-L2 (9p24.1) amplification in metastatic mucosal and cutaneous melanomas with durable response to immunotherapy. Hum Pathol 2018. 10.1016/j.humpath.2018.08.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Ikeda S, Okamoto T, Okano S, et al. PD-L1 is upregulated by simultaneous amplification of the PD-L1 and JAK2 genes in non-small cell lung cancer. J Thorac Oncol 2016;11:62–71. 10.1016/j.jtho.2015.09.010. [DOI] [PubMed] [Google Scholar]
- 88. Van Allen EM, Golay HG, Liu Y, et al. Long-term benefit of PD-L1 blockade in lung cancer associated with jak3 activation. Cancer Immunol Res 2015;3:855–63. 10.1158/2326-6066.cir-15-0024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Sinicrope FA, Foster NR, Thibodeau SN, et al. DNA mismatch repair status and colon cancer recurrence and survival in clinical trials of 5-fluorouracil-based adjuvant therapy. J Natl Cancer Inst 2011;103:863–75. 10.1093/jnci/djr153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Dudley JC, Lin MT, Le DT, et al. Microsatellite instability as a biomarker for PD-1 blockade. Clin Cancer Res 2016;22:813–20. 10.1158/1078-0432.CCR-15-1678. [DOI] [PubMed] [Google Scholar]
- 91. Overman MJ, McDermott R, Leach JL, et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol 2017;18:1182–91. 10.1016/s1470-2045(17)30422-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Greene C, Nakakura EK, Ko AH. Gastrocutaneous fistula in a patient with locally recurrent MSI-high colorectal cancer: local complications arising from therapeutic response to immune checkpoint blockade. Anticancer Res 2017;37:3679–84. 10.21873/anticanres.11739. [DOI] [PubMed] [Google Scholar]
- 93. Czink E, Kloor M, Goeppert B, et al. Successful immune checkpoint blockade in a patient with advanced stage microsatellite-unstable biliary tract cancer. Cold Spring Harb Mol Case Stud 2017;3. 10.1101/mcs.a001974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. AlHarbi M, Ali Mobark N, AlMubarak L, et al. Durable response to nivolumab in a pediatric patient with refractory glioblastoma and constitutional biallelic mismatch repair deficiency. Oncologist 2018;23:1401–6. 10.1634/theoncologist.2018-0163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:2509–20. 10.1056/NEJMoa1500596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409–13. 10.1126/science.aan6733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Waddell N, Pajic M, Patch AM, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015;518:495–501. 10.1038/nature14169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Diaz LA, et al. Programmed death-1 blockade in mismatch repair deficient cancer independent of tumor histology. J Clin Oncol 2016;34:3003–3. 10.1200/JCO.2016.34.15_suppl.3003. [DOI] [Google Scholar]
- 99. Kelderman S, Schumacher TN, Kvistborg P. Mismatch repair-deficient cancers are targets for anti-PD-1 therapy. Cancer Cell 2015;28:11–3. 10.1016/j.ccell.2015.06.012. [DOI] [PubMed] [Google Scholar]
- 100. Johanns TM, Miller CA, Dorward IG, et al. Immunogenomics of hypermutated glioblastoma: a patient with germline POLE deficiency treated with checkpoint blockade immunotherapy. Cancer Discov 2016;6:1230–6. 10.1158/2159-8290.cd-16-0575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Colli LM, Machiela MJ, Zhang H, et al. Landscape of combination immunotherapy and targeted therapy to improve cancer management. Cancer Res 2017;77:3666–71. 10.1158/0008-5472.can-16-3338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Matsuo K, Spragg SE, Ciccone MA, et al. Nivolumab use for BRCA gene mutation carriers with recurrent epithelial ovarian cancer: a case series. Gynecol Oncol Rep 2018;25:98–101. 10.1016/j.gore.2018.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Mehnert JM, Panda A, Zhong H, et al. Immune activation and response to pembrolizumab in POLE-mutant endometrial cancer. J Clin Invest 2016;126:2334–40. 10.1172/jci84940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Miao D, Margolis CA, Gao W, et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science 2018;359:801–6. 10.1126/science.aan5951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. George S, Miao D, Demetri GD, et al. Loss of PTEN is associated with resistance to anti-PD-1 checkpoint blockade therapy in metastatic uterine leiomyosarcoma. Immunity 2017;46:197–204. 10.1016/j.immuni.2017.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Rizvi NA, Hellmann MD, Snyder A, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124–8. 10.1126/science.aaa1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Cohen CJ, Gartner JJ, Horovitz-Fried M, et al. Isolation of neoantigen-specific T cells from tumor and peripheral lymphocytes. J Clin Invest 2015;125:3981–91. 10.1172/JCI82416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Lu YC, Yao X, Crystal JS, et al. Efficient identification of mutated cancer antigens recognized by T cells associated with durable tumor regressions. Clin Cancer Res 2014;20:3401–10. 10.1158/1078-0432.CCR-14-0433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Pasetto A, Gros A, Robbins PF, et al. Tumor- and neoantigen-reactive T-cell receptors can be identified based on their frequency in fresh tumor. Cancer Immunol Res 2016;4:734–43. 10.1158/2326-6066.CIR-16-0001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Robbins PF, Lu YC, El-Gamil M, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 2013;19:747–52. 10.1038/nm.3161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Kvistborg P, Philips D, Kelderman S, et al. Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med 2014;6:254ra128. 10.1126/scitranslmed.3008918. [DOI] [PubMed] [Google Scholar]
- 112. van Rooij N, van Buuren MM, Philips D, et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol 2013;31:e439–442. 10.1200/JCO.2012.47.7521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. McGranahan N, Furness AJ, Rosenthal R, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016;351:1463–9. 10.1126/science.aaf1490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015;348:69–74. 10.1126/science.aaa4971. [DOI] [PubMed] [Google Scholar]
- 115. Lake RA, Robinson BW. Immunotherapy and chemotherapy—a practical partnership. Nat Rev Cancer 2005;5:397–405. 10.1038/nrc1613. [DOI] [PubMed] [Google Scholar]
- 116. Yaghmour G, Pandey M, Ireland C, et al. Role of genomic instability in immunotherapy with checkpoint inhibitors. Anticancer Res 2016;36:4033–8. [PubMed] [Google Scholar]
- 117. Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371:2189–99. 10.1056/NEJMoa1406498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Hellmann MD, Nathanson T, Rizvi H, et al. Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell 2018;33:843–852.e844. 10.1016/j.ccell.2018.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Pleasance ED, Cheetham RK, Stephens PJ, et al. A comprehensive catalogue of somatic mutations from a human cancer genome. Nature 2010;463:191–6. 10.1038/nature08658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Shain AH, Yeh I, Kovalyshyn I, et al. The genetic evolution of melanoma from precursor lesions. N Engl J Med 2015;373:1926–36. 10.1056/NEJMoa1502583. [DOI] [PubMed] [Google Scholar]
- 121. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature 2013;500:415–21. 10.1038/nature12477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Eroglu Z, Zaretsky JM, Hu-Lieskovan S, et al. High response rate to PD-1 blockade in desmoplastic melanomas. Nature 2018;553:347–50. 10.1038/nature25187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Burrell RA, McGranahan N, Bartek J, et al. The causes and consequences of genetic heterogeneity in cancer evolution. Nature 2013;501:338–45. 10.1038/nature12625. [DOI] [PubMed] [Google Scholar]
- 124. Novellino L, Castelli C, Parmiani G. A listing of human tumor antigens recognized by T cells: March 2004 update. Cancer Immunol Immunother 2005;54:187–207. 10.1007/s00262-004-0560-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Safonov A, Jiang T, Bianchini G, et al. Immune gene expression is associated with genomic aberrations in breast cancer. Cancer Res 2017;77:3317–24. 10.1158/0008-5472.CAN-16-3478. [DOI] [PubMed] [Google Scholar]
- 126. Peng W, Chen JQ, Liu C, et al. Loss of pten promotes resistance to T cell-mediated immunotherapy. Cancer Discov 2016;6:202–16. 10.1158/2159-8290.CD-15-0283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Hugo W, Zaretsky JM, Sun L, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 2016;165:35–44. 10.1016/j.cell.2016.02.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Bernicker E. Next-generation sequencing and immunotherapy biomarkers: a medical oncology perspective. Arch Pathol Lab Med 2016;140:245–8. 10.5858/arpa.2015-0287-SA. [DOI] [PubMed] [Google Scholar]
- 129. Kreiter S, Vormehr M, van de Roemer N, et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 2015;520:692–6. 10.1038/nature14426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Castle JC, Kreiter S, Diekmann J, et al. Exploiting the mutanome for tumor vaccination. Cancer Res 2012;72:1081–91. 10.1158/0008-5472.CAN-11-3722. [DOI] [PubMed] [Google Scholar]

