301
|
Tonse R, Rubens M, Appel H, Tom MC, Hall MD, Odia Y, McDermott MW, Ahluwalia MS, Mehta MP, Kotecha R. Systematic review and meta-analysis of PD-L1 expression discordance between primary tumor and lung cancer brain metastasis. Neurooncol Adv 2021; 3:vdab166. [PMID: 34988451 PMCID: PMC8704382 DOI: 10.1093/noajnl/vdab166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Novel immunotherapeutic strategies targeting the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis are often administered when metastatic tumors show PD-L1 positivity, even in the setting of lung cancer brain metastasis (LCBM). However, biological differences exist between primary tumors and metastatic sites. The objective of this study was to analyze rates of PD-L1 receptor discordance between primary tumors and LCBM. Methods A systematic review of studies of biopsied or resected LCBM evaluating PD-L1 discordance published in the Medline database was performed using PRISMA guidelines. Weighted random effects models were used to calculate pooled estimates. Results Six full-text articles (n = 230 patients) with a median of 32 patients in each study (range: 24–73) reported PD-L1 receptor expression analyses of both primary lung tumors and brain metastases and met inclusion criteria. The pooled estimate for tumor cell (TC) PD-L1 receptor discordance between primary tumors and LCBM was 19% (95% confidence interval [CI]: 10–27%). For PD-L1 receptor expression in tumor-infiltrating lymphocytes (TIL), the weighted pooled estimate for discordance was 21% (95% CI: 8–44%). For primary versus LCBM, the positive rates by expression levels of <1%, 1–50%, and >50% were 52% (95% CI: 30–73%) versus 56% (95% CI: 34–76%), 30% (95% CI: 22–40%) versus 20% (95% CI: 10–35%), and 15% (95% CI: 6–36%) versus 22% (95% CI: 15–31%) (P = .425), respectively. Conclusions PD-L1 discordance occurs in ~20% of LCBM, with the greatest discordance in the 1–50% expression category. Although controversial, confirming discordance might be important for selection of immune checkpoint inhibitor therapy and in the analysis of patterns of failure after treatment.
Collapse
Affiliation(s)
- Raees Tonse
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Muni Rubens
- Office of Clinical Research, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Haley Appel
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Martin C Tom
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Matthew D Hall
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Yazmin Odia
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
- Division of Neuro-Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Michael W McDermott
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
- Department of Neurosurgery, Miami Neuroscience Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Manmeet S Ahluwalia
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| |
Collapse
|
302
|
The Landscape of Immunotherapy in Advanced NSCLC: Driving Beyond PD-1/PD-L1 Inhibitors (CTLA-4, LAG3, IDO, OX40, TIGIT, Vaccines). Curr Oncol Rep 2021; 23:126. [PMID: 34453261 PMCID: PMC8397682 DOI: 10.1007/s11912-021-01124-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW In this review, we analyzed the current landscape of non-PD-(L)1 targeting immunotherapy. RECENT FINDINGS The advent of immunotherapy has completely changed the standard approach toward advanced NSCLC. Inhibitors of the PD-1/PD-L1 axis have quickly taken place as first-line treatment for NSCLC patients without targetable "driver" mutations. However, a non-negligible portion of patients derive modest benefit from immune-checkpoint inhibitors, and valid second-line alternatives are lacking, pushing researchers to analyze other molecules and pathways as potentially viable targets in the struggle against NSCLC. Starting from the better characterized CTLA-4 inhibitors, we then critically collected the actual knowledge on NSCLC vaccines as well as on other emerging molecules, many of them in their early phase of testing, to provide to the reader a comprehensive overview of the state of the art of immunotherapy in NSCLC beyond PD-1/PD-L1 inhibitors.
Collapse
|
303
|
Pepe F, Pisapia P, Gristina V, Rocco D, Micheli M, Micheli P, Iaccarino A, Tufano R, Gragnano G, Russo G, De Luca C, Sgariglia R, Nacchio M, Girolami I, Eccher A, Russo A, Troncone G, Malapelle U. Tumor mutational burden on cytological samples: A pilot study. Cancer Cytopathol 2020; 129:460-467. [PMID: 33378102 DOI: 10.1002/cncy.22400] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Immune-checkpoint inhibitors (ICIs) represent an important treatment option for patients who have advanced stage non-small cell lung cancer (NSCLC). Currently, evaluation of the expression level of programmed death-ligand 1 (PD-L1) has proven highly successful as a positive predictive biomarker for ICIs. In addition to PD-L1, other promising predictive biomarkers are emerging, including high tumor mutational burden (TMB-H). However, measuring TMB-H remains challenging for several reasons, among which is the difficulty in obtaining adequate tissue material from NSCLC patients. There are no data in the current literature regarding the possibility of adopting cell blocks (CBs) for TMB evaluation; therefore, our goal was to evaluate the feasibility of analyzing TMB on CBs. METHODS For evaluation of differences in run metric parameters, 8 pairs of histological and CB samples from patients with NSCLC were analyzed using the Oncomine Tumor Mutational Load Assay on Ion Torrent S5 GS next-generation sequencing (NGS) platform. RESULTS Most CBs (6/8, 75.0%) were successfully analyzed by adopting the broad NGS panel approach. CBs provided results similar to those obtained on histological matched specimens in terms of median total reads (7207048.80 vs 7558817.80), median mapped reads (7075753.83 vs 7513822.00), median read lengths (115.50 vs. 113.00), median percentage of reads on-target (97.49% vs. 98.45%), median average reads per amplicon (454.67 vs 476.14), and median uniformity of amplicon coverage (83.52% vs 84.13%). CONCLUSION In this pilot study, we demonstrated the technical feasibility of assessing TMB on CBs.
Collapse
Affiliation(s)
- Francesco Pepe
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Valerio Gristina
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Danilo Rocco
- Department of Oncology, A.O.R.N. Azienda Ospedaliera dei Colli, Naples, Italy
| | | | - Pietro Micheli
- Department of Pathology, A.O.R.N. Azienda Ospedaliera dei Colli, Naples, Italy
| | - Antonino Iaccarino
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | | | - Gianluca Gragnano
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Caterina De Luca
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Roberta Sgariglia
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Mariantonia Nacchio
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Ilaria Girolami
- Division of Pathology, Central Hospital Bolzano, Bolzano, Italy
| | - Albino Eccher
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| |
Collapse
|
304
|
Si H, Kuziora M, Quinn KJ, Helman E, Ye J, Liu F, Scheuring U, Peters S, Rizvi NA, Brohawn PZ, Ranade K, Higgs BW, Banks KC, Chand VK, Raja R. A Blood-based Assay for Assessment of Tumor Mutational Burden in First-line Metastatic NSCLC Treatment: Results from the MYSTIC Study. Clin Cancer Res 2020; 27:1631-1640. [PMID: 33355200 DOI: 10.1158/1078-0432.ccr-20-3771] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/07/2020] [Accepted: 12/17/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Tumor mutational burden (TMB) has been shown to be predictive of survival benefit in patients with non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitors. Measuring TMB in the blood (bTMB) using circulating cell-free tumor DNA (ctDNA) offers practical advantages compared with TMB measurement in tissue (tTMB); however, there is a need for validated assays and identification of optimal cutoffs. We describe the analytic validation of a new bTMB algorithm and its clinical utility using data from the phase III MYSTIC trial. PATIENTS AND METHODS The dataset used for the clinical validation was from MYSTIC, which evaluated first-line durvalumab (anti-PD-L1 antibody) ± tremelimumab (anticytotoxic T-lymphocyte-associated antigen-4 antibody) or chemotherapy for metastatic NSCLC. bTMB and tTMB were evaluated using the GuardantOMNI and FoundationOne CDx assays, respectively. A Cox proportional hazards model and minimal P value cross-validation approach were used to identify the optimal bTMB cutoff. RESULTS In MYSTIC, somatic mutations could be detected in ctDNA extracted from plasma samples in a majority of patients, allowing subsequent calculation of bTMB. The success rate for obtaining valid TMB scores was higher for bTMB (809/1,001; 81%) than for tTMB (460/735; 63%). Minimal P value cross-validation analysis confirmed the selection of bTMB ≥20 mutations per megabase (mut/Mb) as the optimal cutoff for clinical benefit with durvalumab + tremelimumab. CONCLUSIONS Our study demonstrates the feasibility, accuracy, and reproducibility of the GuardantOMNI ctDNA platform for quantifying bTMB from plasma samples. Using the new bTMB algorithm and an optimal bTMB cutoff of ≥20 mut/Mb, high bTMB was predictive of clinical benefit with durvalumab + tremelimumab versus chemotherapy.
Collapse
Affiliation(s)
- Han Si
- AstraZeneca, Gaithersburg, Maryland
| | | | | | | | - Jiabu Ye
- AstraZeneca, Gaithersburg, Maryland
| | - Feng Liu
- AstraZeneca, Gaithersburg, Maryland
| | | | - Solange Peters
- Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
305
|
Li Y, Zhao R, Cheng K, Zhang K, Wang Y, Zhang Y, Li Y, Liu G, Xu J, Xu J, Anderson GJ, Shi J, Ren L, Zhao X, Nie G. Bacterial Outer Membrane Vesicles Presenting Programmed Death 1 for Improved Cancer Immunotherapy via Immune Activation and Checkpoint Inhibition. ACS NANO 2020; 14:16698-16711. [PMID: 33232124 DOI: 10.1021/acsnano.0c03776] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Natural, extracellular membrane vesicles secreted by Gram-negative bacteria, outer membrane vesicles (OMVs), contain numerous pathogen-associated molecular patterns which can activate systemic immune responses. Previous studies have shown that OMVs induce strong IFN-γ- and T cell-mediated anti-tumor effects in mice. However, IFN-γ is known to upregulate immunosuppressive factors in the tumor microenvironment, especially the immune checkpoint programmed death 1 ligand 1 (PD-L1), which may hamper T cell function and limit immunotherapeutic effectiveness. Here, we report the development of genetically engineered OMVs whose surface has been modified by insertion of the ectodomain of programmed death 1 (PD1). This genetic modification does not affect the ability of OMVs to trigger immune activation. More importantly, the engineered OMV-PD1 can bind to PD-L1 on the tumor cell surface and facilitate its internalization and reduction, thereby protecting T cells from the PD1/PD-L1 immune inhibitory axis. Through the combined effects of immune activation and checkpoint suppression, the engineered OMVs drive the accumulation of effector T cells in the tumor, which, in turn, leads to a greater impairment of tumor growth, compared with not only native OMVs but also the commonly used PD-L1 antibody. In conclusion, this work demonstrates the potential of bioengineered OMVs as effective immunotherapeutic agents that can comprehensively regulate the tumor immune microenvironment to effect markedly increased anti-tumor efficacy.
Collapse
Affiliation(s)
- Yao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, College of Materials, Xiamen University, Xiamen, Fujian 361005, China
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, College of Materials, Xiamen University, Xiamen, Fujian 361005, China
| | - Kaiyue Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yazhou Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yujing Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaqi Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gregory J Anderson
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Ren
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, College of Materials, Xiamen University, Xiamen, Fujian 361005, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
306
|
Lagos GG, Izar B, Rizvi NA. Beyond Tumor PD-L1: Emerging Genomic Biomarkers for Checkpoint Inhibitor Immunotherapy. Am Soc Clin Oncol Educ Book 2020; 40:1-11. [PMID: 32315237 DOI: 10.1200/edbk_289967] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the success of immune checkpoint blockade as a strategy for activating an antitumor immune response and promoting cancer regression, only a subset of patients have durable clinical benefit. Efforts are ongoing to identify robust biomarkers that can effectively predict treatment response to immune checkpoint inhibitors (ICIs). Although PD-L1 expression is useful for stratifying patients, it is an imperfect tool. Comprehensive next-generation sequencing platforms that are readily used in clinical practice to identify a tumor's potentially actionable genetic alterations also reveal tumor genomic features, including tumor mutation burden (TMB), that may impact the response to ICIs. High TMB enhances tumor immunogenicity through increased numbers of tumor neoantigens that may promote an immune response. Defective DNA repair, leading to microsatellite instability, is an endogenous mechanism for increased tumor TMB that augments response to anti-PD-1 blockade. Alternatively, DNA damage from exogenous factors is responsible for high TMB seen in melanoma, lung cancer, and urothelial carcinoma, among tumor subtypes with higher response rates to ICIs. In this review, we summarize data supporting the use of TMB as a biomarker as well as its known limitations. We also highlight specific tumor suppressor genes and oncogenes that are under investigation as biomarkers for ICI response and resistance. Efforts are ongoing to delineate which genomic tumor characteristics can eventually be utilized in clinical practice to ascertain the benefit of ICIs for an individual patient.
Collapse
|
307
|
Ferrara R, Imbimbo M, Malouf R, Paget-Bailly S, Calais F, Marchal C, Westeel V. Single or combined immune checkpoint inhibitors compared to first-line platinum-based chemotherapy with or without bevacizumab for people with advanced non-small cell lung cancer. Cochrane Database Syst Rev 2020; 12:CD013257. [PMID: 33316104 PMCID: PMC8094159 DOI: 10.1002/14651858.cd013257.pub2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 axis have changed the first-line treatment of people with advanced non-small cell lung cancer (NSCLC). Single-agent pembrolizumab (a PD-1 inhibitor) is currently the standard of care as monotherapy in patients with PD-L1 expression ≥ 50%, either alone or in combination with chemotherapy when PD-L1 expression is less than 50%. Atezolizumab (PD-L1 inhibitor) has also been approved in combination with chemotherapy and bevacizumab (an anti-angiogenic antibody) in first-line NSCLC regardless of PD-L1 expression. The combination of first-line PD-1/PD-L1 inhibitors with anti-CTLA-4 antibodies has also been shown to improve survival compared to platinum-based chemotherapy in advanced NSCLC, particularly in people with high tumour mutational burden (TMB). The association of ipilimumab (an anti CTLA4) and nivolumab (PD-1 inhibitor) has been approved by the US Food and Drug Administration (FDA) in all patients with PD-L1 expression ≥1%. Although these antibodies are currently used in clinical practice, some questions remain unanswered, such as the best-treatment strategy, the role of different biomarkers for treatment selection and the effectiveness of immunotherapy according to specific clinical characteristics. OBJECTIVES Primary objective: to determine the effectiveness and safety of first-line immune checkpoint inhibitors (ICIs), as monotherapy or in combination, compared to platinum-based chemotherapy, with or without bevacizumab for people with advanced NSCLC, according to the level of PD-L1 expression. SECONDARY OBJECTIVE to maintain the currency of evidence using a living systematic review approach. SEARCH METHODS We performed an electronic search of the main databases (Cochrane Lung Cancer Group Trial Register, Cochrane Central Register of Controlled Trials, MEDLINE, Embase) from inception until 21 October 2020 and conferences meetings from 2015 onwards. SELECTION CRITERIA We included randomised controlled trials (RCTs) reporting on the efficacy or safety of first-line ICI treatment for adults with advanced NSCLC who had not previously received any anticancer treatment. We included trials comparing single- or double-ICI treatment to standard first-line therapy (platinum-based chemotherapy +/- bevacizumab). All data come from 'international multicentre studies involving adults, age 18 or over, with histologically-confirmed stage IV NSCLC who had not received any previous systemic anti-cancer treatment for advanced disease. DATA COLLECTION AND ANALYSIS Three review authors independently assessed the search results and a fourth review author resolved any disagreements. Primary outcomes were overall survival (OS) and progression-free survival (PFS); secondary outcomes were overall objective response rate (ORR) by RECIST v 1.1, grade 3 to 5 treatment-related adverse events (AEs) (CTCAE v 5.0) and health-related quality of life (HRQoL). We performed meta-analyses where appropriate using the random-effects model for hazard ratios (HRs) or risk ratios (RRs), with 95% confidence intervals (95% CIs), and used the I² statistic to investigate heterogeneity. MAIN RESULTS Main results We identified 15 trials for inclusion, seven completed and eight ongoing trials. We obtained data for 5893 participants from seven trials comparing first-line single- (six trials) or double- (two trials) agent ICI with platinum-based chemotherapy, one trial comparing both first-line single- and double-agent ICsI with platinum-based chemotherapy. All trials were at low risk of selection and detection bias, some were classified at high risk of performance, attrition or other source of bias. The overall certainty of evidence according to GRADE ranged from moderate-to-low because of risk of bias, inconsistency, or imprecision. The majority of the included trials reported their outcomes by PD-L1 expressions, with PD-L1 ≥ 50 being considered the most clinically useful cut-off level for decision makers. Also, iIn order to avoid overlaps between various PDL-1 expressions we prioritised the review outcomes according to PD-L1 ≥ 50. Single-agent ICI In the PD-L1 expression ≥ 50% group single-agent ICI probably improved OS compared to platinum-based chemotherapy (hazard ratio (HR) 0.68, 95% confidence interval (CI) 0.60 to 0.76, 6 RCTs, 2111 participants, moderate-certainty evidence). In this group, single-agent ICI also may improve PFS (HR: 0.68, 95% CI 0.52 to 0.88, 5 RCTs, 1886 participants, low-certainty evidence) and ORR (risk ratio (RR):1.40, 95% CI 1.12 to 1.75, 4 RCTs, 1672 participants, low-certainty evidence). HRQoL data were available for only one study including only people with PD-L1 expression ≥ 50%, which suggested that single-agent ICI may improve HRQoL at 15 weeks compared to platinum-based chemotherapy (RR: 1.51, 95% CI 1.08 to 2.10, 1 RCT, 297 participants, low-certainty evidence). In the included studies, treatment-related AEs were not reported according to PD-L1 expression levels. Grade 3-4 AEs may be less frequent with single-agent ICI compared to platinum-based chemotherapy (RR: 0.41, 95% CI 0.33 to 0.50, I² = 62%, 5 RCTs, 3346 participants, low-certainty evidence). More information about efficacy of single-agent ICI compared to platinum-based chemotherapy according to the level of PD-L1 expression and to TMB status or specific clinical characteristics is available in the full text. Double-agent ICI Double-ICI treatment probably prolonged OS compared to platinum-based chemotherapy in people with PD-L1 expression ≥50% (HR: 0.72, 95% CI 0.59 to 0.89 2 RCTs, 612 participants, moderate-certainty evidence). Trials did not report data on HRQoL, PFS and ORR according to PD-L1 groups. Treatment related AEs were not reported according to PD-L1 expression levels. The frequency of grade 3-4 AEs may not differ between double-ICI treatment and platinum-based chemotherapy (RR: 0.78, 95% CI 0.55 to 1.09, I² = 81%, 2 RCTs, 1869 participants, low-certainty evidence). More information about efficacy of double-agent ICI according to the level of PD-L1 expression and to TMB status is available in the full text. AUTHORS' CONCLUSIONS Authors' conclusions The evidence in this review suggests that single-agent ICI in people with NSCLC and PD-L1 ≥50% probably leads to a higher overall survival rate and may lead to a higher progression-free survival and overall response rate when compared to platinum-based chemotherapy and may also lead to a lower rate of adverse events and higher HRQoL. Combined ICI in people with NSCLC and PD-L1 ≥50% also probably leads to a higher overall survival rate when compared to platinum-based chemotherapy, but its effect on progression-free survival, overall response rate and HRQoL is unknown due to a lack of data. The rate of adverse events may not differ between groups.
Collapse
Affiliation(s)
- Roberto Ferrara
- Thoracic Oncology Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, Milano, Italy
| | - Martina Imbimbo
- Department of Oncology, Lausanne University Hospital, and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Reem Malouf
- National Perinatal Epidemiology Unit (NPEU), University of Oxford, Oxford, UK
| | - Sophie Paget-Bailly
- Methodological and Quality of Life in Oncology Unit, University Hospital of Besançon, Besançon, France
- Université de Franche-Comté, INSERM, EFS BFC, UMR 1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France, Besançon, France
| | - François Calais
- Bibliothèque universitaire de Santé, Université de Franche-Comté, Besançon, France
| | | | - Virginie Westeel
- Methodological and Quality of Life in Oncology Unit, University Hospital of Besançon, Besançon, France
- Department of Thoracic Oncology, University Hospital of Besançon, Besançon, France
- Université Bourgogne Franche-Comté, INSERM, EFS BFC, UMR 1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| |
Collapse
|
308
|
Pang Z, Chen X, Wang Y, Wang Y, Yan T, Wan J, Wang K, Du J. Long non-coding RNA C5orf64 is a potential indicator for tumor microenvironment and mutation pattern remodeling in lung adenocarcinoma. Genomics 2020; 113:291-304. [PMID: 33309768 DOI: 10.1016/j.ygeno.2020.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/06/2020] [Accepted: 12/06/2020] [Indexed: 12/22/2022]
Abstract
Understanding the synergistic and antagonistic effects of tumor microenvironment (TME) and tumor mutation pattern on lung adenocarcinoma (LUAD) is urgently needed. Herein, we applied ESTIMATE and CIBERSORT methods to calculate the ratio of immune and stromal components and TIICs proportion of LUAD samples from TCGA database. Immune-related genes were analyzed by Lasso regression analysis and used for ceRNA network construction. A 14-lncRNA immune-related signature was developed, among which C5orf64 was found to be positively correlated with abundances of M2 macrophages, monocytes, eosinophils and neutrophils, but negatively correlated with Tregs and plasma cells. PD-1, PD-L1 and CTLA-4 were demonstrated to be high expressed in high-level C5orf64 groups. However, C5orf64 had a negative correlation with TP53 mutation frequency. A novel model was built based on age, tumor stage and immune-related lncRNA signature. To conclude, lncRNA C5orf64 had potential to be an indicator for TME modulation and tumor mutation pattern remodeling in LUAD.
Collapse
Affiliation(s)
- Zhaofei Pang
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Xiaowei Chen
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Yu Wang
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China; Department of Respiratory Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Yadong Wang
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Tao Yan
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Jun Wan
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Kai Wang
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Jiajun Du
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China; Department of Thoracic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China; Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
309
|
Sha D, Jin Z, Budczies J, Kluck K, Stenzinger A, Sinicrope FA. Tumor Mutational Burden as a Predictive Biomarker in Solid Tumors. Cancer Discov 2020; 10:1808-1825. [PMID: 33139244 PMCID: PMC7710563 DOI: 10.1158/2159-8290.cd-20-0522] [Citation(s) in RCA: 409] [Impact Index Per Article: 102.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/03/2020] [Accepted: 09/09/2020] [Indexed: 11/16/2022]
Abstract
Tumor mutational burden (TMB), defined as the number of somatic mutations per megabase of interrogated genomic sequence, varies across malignancies. Panel sequencing-based estimates of TMB have largely replaced whole-exome sequencing-derived TMB in the clinic. Retrospective evidence suggests that TMB can predict the efficacy of immune checkpoint inhibitors, and data from KEYNOTE-158 led to the recent FDA approval of pembrolizumab for the TMB-high tumor subgroup. Unmet needs include prospective validation of TMB cutoffs in relationship to tumor type and patient outcomes. Furthermore, standardization and harmonization of TMB measurement across test platforms are important to the successful implementation of TMB in clinical practice. SIGNIFICANCE: Evaluation of TMB as a predictive biomarker creates the need to harmonize panel-based TMB estimation and standardize its reporting. TMB can improve the predictive accuracy for immunotherapy outcomes, and has the potential to expand the candidate pool of patients for treatment with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Dan Sha
- Departments of Medicine and Gastrointestinal Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Zhaohui Jin
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Jan Budczies
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg Partner Site, Heidelberg, Germany
| | - Klaus Kluck
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg Partner Site, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg Partner Site, Heidelberg, Germany
| | - Frank A Sinicrope
- Departments of Medicine and Gastrointestinal Research Unit, Mayo Clinic, Rochester, Minnesota.
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
- Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| |
Collapse
|
310
|
Hu-Lieskovan S, Bhaumik S, Dhodapkar K, Grivel JCJB, Gupta S, Hanks BA, Janetzki S, Kleen TO, Koguchi Y, Lund AW, Maccalli C, Mahnke YD, Novosiadly RD, Selvan SR, Sims T, Zhao Y, Maecker HT. SITC cancer immunotherapy resource document: a compass in the land of biomarker discovery. J Immunother Cancer 2020; 8:e000705. [PMID: 33268350 PMCID: PMC7713206 DOI: 10.1136/jitc-2020-000705] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2020] [Indexed: 02/07/2023] Open
Abstract
Since the publication of the Society for Immunotherapy of Cancer's (SITC) original cancer immunotherapy biomarkers resource document, there have been remarkable breakthroughs in cancer immunotherapy, in particular the development and approval of immune checkpoint inhibitors, engineered cellular therapies, and tumor vaccines to unleash antitumor immune activity. The most notable feature of these breakthroughs is the achievement of durable clinical responses in some patients, enabling long-term survival. These durable responses have been noted in tumor types that were not previously considered immunotherapy-sensitive, suggesting that all patients with cancer may have the potential to benefit from immunotherapy. However, a persistent challenge in the field is the fact that only a minority of patients respond to immunotherapy, especially those therapies that rely on endogenous immune activation such as checkpoint inhibitors and vaccination due to the complex and heterogeneous immune escape mechanisms which can develop in each patient. Therefore, the development of robust biomarkers for each immunotherapy strategy, enabling rational patient selection and the design of precise combination therapies, is key for the continued success and improvement of immunotherapy. In this document, we summarize and update established biomarkers, guidelines, and regulatory considerations for clinical immune biomarker development, discuss well-known and novel technologies for biomarker discovery and validation, and provide tools and resources that can be used by the biomarker research community to facilitate the continued development of immuno-oncology and aid in the goal of durable responses in all patients.
Collapse
Affiliation(s)
- Siwen Hu-Lieskovan
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Kavita Dhodapkar
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | | | - Sumati Gupta
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Brent A Hanks
- Duke University Medical Center, Durham, North Carolina, USA
| | | | | | - Yoshinobu Koguchi
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Amanda W Lund
- Oregon Health and Science University, Portland, Oregon, USA
| | | | | | | | | | - Tasha Sims
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | | |
Collapse
|
311
|
Wojas-Krawczyk K, Kubiatowski T. Imperfect Predictors for Lung Cancer Immunotherapy-A Field for Further Research. Front Oncol 2020; 10:568174. [PMID: 33330041 PMCID: PMC7734866 DOI: 10.3389/fonc.2020.568174] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 10/26/2020] [Indexed: 12/16/2022] Open
Abstract
The armamentarium for lung cancer immunotherapy has been strengthened using two groups of monoclonal antibodies: 1) anti-PD-1 antibodies, including pembrolizumab and nivolumab, which block the programmed death 1 receptor on the lymphocyte surface, resulting in increasing activity of these cells, and 2) anti-PD-L1 antibodies, including atezolizumab, durvalumab, and avelumab, which block the ligand for the PD-1 molecule on tumor cells and on tumor-infiltrating immune cells. The effectiveness of both groups of antibodies has been proven in many clinical trials, which translates into positive immunotherapeutic registrations for cancer patients. Regarding the predictive factor, PD-L1 expression on cancer cells is the only biomarker validated in prospective clinical trials used for qualification to immunotherapy in advanced non-small cell lung cancer (NSCLC) patients. However, it is not an ideal one. Unfortunately, no clinical benefits could be noted in patients with high PD-L1 expression on tumor cells against the effectiveness of immunotherapy that may be observed in patients without PD-L1 expression. Furthermore, the mechanism of antitumor immune response is extremely complex, multistage, and depends on many factors. Cancer cells could be recognized by the immune system, provided tumor-specific antigen presentation, and these arise as a result of somatic mutations in tumor cells. Based on novel immunotherapy registration, high tumor mutation burden (TMB) has become an important predictive factor. The intensity of lymphocyte infiltration in tumor tissue may be another predictive factor. The effectiveness of anti-PD-L1 immunotherapy is observed in patients with high expression of genes associated with the effector function of T lymphocytes (i.e., their ability to produce IFN-gamma). This does not end the list of potential factors that become useful in qualification of cancer patients for immunotherapy. There remains a need to search for new and perfect predictive factors for immunotherapy.
Collapse
Affiliation(s)
- Kamila Wojas-Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Tomasz Kubiatowski
- Department of Clinical Oncology, Saint John of Dukla Oncology Centre of the Lublin Region, Lublin, Poland
| |
Collapse
|
312
|
Khasraw M, Walsh KM, Heimberger AB, Ashley DM. What is the Burden of Proof for Tumor Mutational Burden in gliomas? Neuro Oncol 2020; 23:noaa256. [PMID: 33252666 PMCID: PMC7849945 DOI: 10.1093/neuonc/noaa256] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Indexed: 12/17/2022] Open
Abstract
The treatment of patients with a variety of solid tumors has benefitted from immune checkpoint inhibition targeting the anti-programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) axis. The US Food and Drug Administration (FDA) granted accelerated approval of PD-1 inhibitor pembrolizumab for the treatment of adult and pediatric patients with TMB-high (TMB-H), solid tumors that have progressed following prior treatment and who have no other treatment options, including the extension to tumors of the Central Nervous System (CNS). In general, pan-cancer approvals are viewed positively to empower patients and clinicians. There are subsets (eg, BRAF, NTRK) for which this pathway for approval is appropriate. However, the pan-cancer FDA approval of pembrolizumab raises several concerns regarding the generalizability of the evidence to other tumor types, including managing patients with gliomas and other CNS tumors. The cut off for TMB-H is not well defined. There are intrinsic immunological differences between gliomas and other cancers types, including the immunosuppressive glioma microenvironment, the tumor's effects on systemic immune function, and the transformation of the T cell populations to an exhausted phenotype in glioma. Here we address the caveats with pan-cancer approvals concerning gliomas, complexities of the unique CNS immune environment, and discuss potential predictive biomarkers, including TMB, and explain why the recent approval should be applied with caution in CNS tumors.
Collapse
Affiliation(s)
- Mustafa Khasraw
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Kyle M Walsh
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David M Ashley
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
313
|
Dagogo-Jack I, Mino-Kenudson M. Reply to the Letter to the Editor From Zhou et al. J Thorac Oncol 2020; 15:e136-e137. [PMID: 32718538 DOI: 10.1016/j.jtho.2020.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/01/2020] [Indexed: 11/16/2022]
Affiliation(s)
- Ibiayi Dagogo-Jack
- Massachusetts General Hospital Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
314
|
Sutherland KD, Vissers JHA. Balancing the Count: Harmonizing Panel-Based Tumor Mutational Burden Assessment. J Thorac Oncol 2020; 15:1106-1109. [PMID: 32593443 DOI: 10.1016/j.jtho.2020.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 11/19/2022]
Affiliation(s)
- Kate D Sutherland
- Australian Cancer Research Foundation Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.
| | - Joseph H A Vissers
- Centre for Cancer Research and Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia; Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
315
|
Abstract
PURPOSE OF REVIEW Positive results from recent immunotherapy trials of non-small cell lung cancer (NSCLC) have coincided with a greater appreciation for the impact of radiation therapy (RT) on tumor immunity. Here, we summarize key clinical findings and ongoing efforts to combine immunotherapy and RT for the treatment of NSCLC. RECENT FINDINGS The role of immunotherapy for NSCLC has expanded significantly following the pivotal approvals of nivolumab and pembrolizumab for metastatic NSCLC, maintenance durvalumab in unresectable stage III NSCLC, and atezolizumab for metastatic NSCLC. Several small early-phase trials have demonstrated the ability of RT to elicit clinically significant tumor immunity. These positive findings support current trial efforts combining RT with immunotherapy for NSCLC. Recently initiated trials of RT and immunotherapy hold significant promise in expanding the therapeutic options for NSCLC. Optimization of therapy will require careful patient selection to yield meaningful improvements in clinical outcomes.
Collapse
|
316
|
Kachroo S, Shao C, Desai K, He J, Jin F, Sen S. Association of clinico-genomic characteristics with tumor mutational burden in small cell lung cancer patients. Future Oncol 2020; 17:423-433. [PMID: 33198513 DOI: 10.2217/fon-2020-0728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: We evaluated the relationship between clinical and genomic characteristics and tumor mutational burden (TMB) in small cell lung cancer. Materials & methods: In a retrospective analysis of small cell lung cancer patients aged ≥18, we assessed treatment patterns and survival in relation to TMB; the association of clinical and genomic characteristics with TMB was determined by multivariate regression. High TMB (TMB-H) was defined as ≥10 mutations/megabase. Results: Among 186 patients, treatment patterns and overall survival were similar for TMB-H and non-TMB-H patients. TMB was determined for 179 patients, 41.9% of whom were TMB-H. Short variants of LRP1B, FAT3, MLL3, MED12 and NOTCH3 were significantly associated with TMB-H (p ≤ 0.01). Conclusion: Neither treatment patterns nor survival differed by TMB status.
Collapse
Affiliation(s)
| | | | | | - Jinghua He
- Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - Fan Jin
- Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | | |
Collapse
|
317
|
Lang GT, Jiang YZ, Shi JX, Yang F, Li XG, Pei YC, Zhang CH, Ma D, Xiao Y, Hu PC, Wang H, Yang YS, Guo LW, Lu XX, Xue MZ, Wang P, Cao AY, Ling H, Wang ZH, Yu KD, Di GH, Li DQ, Wang YJ, Yu Y, Shi LM, Hu X, Huang W, Shao ZM. Characterization of the genomic landscape and actionable mutations in Chinese breast cancers by clinical sequencing. Nat Commun 2020; 11:5679. [PMID: 33173047 PMCID: PMC7656255 DOI: 10.1038/s41467-020-19342-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 10/07/2020] [Indexed: 12/31/2022] Open
Abstract
The remarkable advances in next-generation sequencing technology have enabled the wide usage of sequencing as a clinical tool. To promote the advance of precision oncology for breast cancer in China, here we report a large-scale prospective clinical sequencing program using the Fudan-BC panel, and comprehensively analyze the clinical and genomic characteristics of Chinese breast cancer. The mutational landscape of 1,134 breast cancers reveals that the most significant differences between Chinese and Western patients occurred in the hormone receptor positive, human epidermal growth factor receptor 2 negative breast cancer subtype. Mutations in p53 and Hippo signaling pathways are more prevalent, and 2 mutually exclusive and 9 co-occurring patterns exist among 9 oncogenic pathways in our cohort. Further preclinical investigation partially suggests that NF2 loss-of-function mutations can be sensitive to a Hippo-targeted strategy. We establish a public database (Fudan Portal) and a precision medicine knowledge base for data exchange and interpretation. Collectively, our study presents a leading approach to Chinese precision oncology treatment and reveals potentially actionable mutations in breast cancer. Chinese breast cancer patients have not been well represented in clinical sequencing studies. Here the authors analyse the mutational landscape of 1,134 Chinese breast cancer patients, finding actionable targets and a higher prevalence of p53 and Hippo pathway mutations compared to Western cohorts.
Collapse
Affiliation(s)
- Guan-Tian Lang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong'an Road, 200032, Shanghai, P.R. China
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China
| | - Jin-Xiu Shi
- Department of Genetics, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Academy of Science and Technology (SAST), 250 Bibo Road, 201203, Shanghai, P.R. China
| | - Fan Yang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong'an Road, 200032, Shanghai, P.R. China
| | - Xiao-Guang Li
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China
| | - Yu-Chen Pei
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China.,Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, 688 Hongqu Road, 201315, Shanghai, P.R. China
| | - Chen-Hui Zhang
- Department of Genetics, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Academy of Science and Technology (SAST), 250 Bibo Road, 201203, Shanghai, P.R. China
| | - Ding Ma
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China
| | - Yi Xiao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China
| | - Peng-Chen Hu
- Department of Genetics, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Academy of Science and Technology (SAST), 250 Bibo Road, 201203, Shanghai, P.R. China
| | - Hai Wang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong'an Road, 200032, Shanghai, P.R. China
| | - Yun-Song Yang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong'an Road, 200032, Shanghai, P.R. China
| | - Lin-Wei Guo
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong'an Road, 200032, Shanghai, P.R. China
| | - Xun-Xi Lu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong'an Road, 200032, Shanghai, P.R. China
| | - Meng-Zhu Xue
- SARI Center for Stem Cell and Nanomedicine, Shanghai Advanced Research Institute, Chinese Academy of Sciences, 201210, Shanghai, P.R. China
| | - Peng Wang
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, 200031, Shanghai, P.R. China
| | - A-Yong Cao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China
| | - Hong Ling
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China
| | - Zhong-Hua Wang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China
| | - Gen-Hong Di
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China
| | - Da-Qiang Li
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China
| | - Yun-Jin Wang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China.,Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, 688 Hongqu Road, 201315, Shanghai, P.R. China
| | - Ying Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Fudan University, 2005 Songhu Road, 200438, Shanghai, P.R. China
| | - Le-Ming Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Fudan University, 2005 Songhu Road, 200438, Shanghai, P.R. China
| | - Xin Hu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China. .,Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, 688 Hongqu Road, 201315, Shanghai, P.R. China.
| | - Wei Huang
- Department of Genetics, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Academy of Science and Technology (SAST), 250 Bibo Road, 201203, Shanghai, P.R. China. .,Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, 688 Hongqu Road, 201315, Shanghai, P.R. China.
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, 270 Dong'an Road, 200032, Shanghai, P.R. China. .,Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, 688 Hongqu Road, 201315, Shanghai, P.R. China.
| |
Collapse
|
318
|
Kim B, Kang SY, Kim KM. DNA-protein biomarkers for immunotherapy in the era of precision oncology. J Pathol Transl Med 2020; 55:26-32. [PMID: 33153244 PMCID: PMC7829578 DOI: 10.4132/jptm.2020.09.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/23/2020] [Indexed: 12/02/2022] Open
Abstract
The use of biomarkers to guide patient and therapy selection has gained much attention to increase the scope and complexity of targeted therapy options and immunotherapy. Clinical trials provide a basis for discovery of biomarkers, which can then aid in development of new drugs. To that end, samples from cancer patients, including DNA, RNA, protein, and the metabolome isolated from cancer tissues and blood or urine, are analyzed in various ways to identify relevant biomarkers. In conjunction with nucleotide-based, high-throughput, next-generation sequencing techniques, therapy-guided biomarker assays relying on protein-based immunohistochemistry play a pivotal role in cancer care. In this review, we discuss the current knowledge regarding DNA and protein biomarkers for cancer immunotherapy.
Collapse
Affiliation(s)
- Binnari Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Pathology, Ulsan University Hospital, Ulsan, Korea
| | - So Young Kang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Center of Clinical Genomics, Samsung Medical Center, Seoul, Korea
| |
Collapse
|
319
|
Dammeijer F, van Gulijk M, Mulder EE, Lukkes M, Klaase L, van den Bosch T, van Nimwegen M, Lau SP, Latupeirissa K, Schetters S, van Kooyk Y, Boon L, Moyaart A, Mueller YM, Katsikis PD, Eggermont AM, Vroman H, Stadhouders R, Hendriks RW, Thüsen JVD, Grünhagen DJ, Verhoef C, van Hall T, Aerts JG. The PD-1/PD-L1-Checkpoint Restrains T cell Immunity in Tumor-Draining Lymph Nodes. Cancer Cell 2020; 38:685-700.e8. [PMID: 33007259 DOI: 10.1016/j.ccell.2020.09.001] [Citation(s) in RCA: 332] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/28/2020] [Accepted: 08/31/2020] [Indexed: 12/31/2022]
Abstract
PD-1/PD-L1-checkpoint blockade therapy is generally thought to relieve tumor cell-mediated suppression in the tumor microenvironment but PD-L1 is also expressed on non-tumor macrophages and conventional dendritic cells (cDCs). Here we show in mouse tumor models that tumor-draining lymph nodes (TDLNs) are enriched for tumor-specific PD-1+ T cells which closely associate with PD-L1+ cDCs. TDLN-targeted PD-L1-blockade induces enhanced anti-tumor T cell immunity by seeding the tumor site with progenitor-exhausted T cells, resulting in improved tumor control. Moreover, we show that abundant PD-1/PD-L1-interactions in TDLNs of nonmetastatic melanoma patients, but not those in corresponding tumors, associate with early distant disease recurrence. These findings point at a critical role for PD-L1 expression in TDLNs in governing systemic anti-tumor immunity, identifying high-risk patient groups amendable to adjuvant PD-1/PD-L1-blockade therapy.
Collapse
Affiliation(s)
- Floris Dammeijer
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands; Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Mandy van Gulijk
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands; Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Evalyn E Mulder
- Department of Surgical Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Melanie Lukkes
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Larissa Klaase
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Menno van Nimwegen
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Sai Ping Lau
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Surgical Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Kitty Latupeirissa
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Sjoerd Schetters
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Louis Boon
- Polpharma Biologics, Utrecht, the Netherlands
| | - Antien Moyaart
- Department of Pathology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Yvonne M Mueller
- Department of Immunology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Peter D Katsikis
- Department of Immunology, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Heleen Vroman
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands; Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Cell Biology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jan von der Thüsen
- Department of Pathology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Dirk J Grünhagen
- Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Surgical Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Cornelis Verhoef
- Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Surgical Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands.
| | - Joachim G Aerts
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands; Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
320
|
Chen S, Huang Z, Jia W, Tao H, Zhang S, Ma J, Liu Z, Wang J, Wang L, Cui P, Zhang Z, Huang D, Wu Z, Zheng X, Hu Y. Association of the Pretreatment Lung Immune Prognostic Index with Survival Outcomes in Advanced Hepatocellular Carcinoma Patients Treated with PD-1 Inhibitors. J Hepatocell Carcinoma 2020; 7:289-299. [PMID: 33173757 PMCID: PMC7646485 DOI: 10.2147/jhc.s277453] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/10/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose At present, there are no validated biomarkers that can predict whether patients with advanced hepatocellular carcinoma (aHCC) are likely to benefit from anti-PD-1 therapy. We aimed to determine whether lung immune prognostic index (LIPI) is associated with outcomes in patients with aHCC treated with PD-1 inhibitors. Patients and Methods Patients undergoing initial treatment with PD-1 inhibitors for aHCC at a single center from January 1, 2015 to August 31, 2019 were included. The patients were stratified according to pretreatment LIPI based on a derived neutrophils/(leukocytes minus neutrophils) ratio (dNLR) ≥ 3 and a lactate dehydrogenase (LDH) level ≥ the upper limit of normal (ULN). Kaplan–Meier analysis and the Log rank test were used to calculate and compare survival between good LIPI and intermediate/poor LIPI scores. The prognostic values of LIPI for survival and disease control rate were evaluated using Cox proportional hazard and logistic regression models, respectively. Results Of the 108 study patients, 53 (49%) had a good LIPI (dNLR < 3 and LDH normal) and 55 (51%) had intermediate/poor LIPI (dNLR ≥ 3 or/and LDH ≥ ULN). With a median follow-up of 12.4 months, intermediate/poor LIPI was independently associated with shorter overall survival (OS) (hazard ratio [HR] 4.00; 95% CI, 2.00–8.03) and progression-free survival (PFS) (HR 2.65; 95% CI, 1.61–4.37). The median OS for good and intermediate/poor LIPI was not reached and was 13.7 (95% CI, 8.2–19.1) months, respectively, and the median PFS was 10.9 (95% CI, 8.9–12.9) and 4.0 (95% CI, 2.2–5.8) months (both P < 0.001), respectively. Conclusion Pretreatment LIPI combined with a dNLR ≥ 3 and/or LDH ≥ ULN is associated with poor outcomes in patients with aHCC treated with PD-1 inhibitors. Further validation in large, prospective studies are warranted.
Collapse
Affiliation(s)
- Shixue Chen
- Department of Graduate Administration, Chinese PLA General Hospital, Beijing, People's Republic of China.,Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Ziwei Huang
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China.,School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Wangping Jia
- Department of Graduate Administration, Chinese PLA General Hospital, Beijing, People's Republic of China.,Institute of Geriatrics, Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatrics Diseases, Second Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Haitao Tao
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Sujie Zhang
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Junxun Ma
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhefeng Liu
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jinliang Wang
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lijie Wang
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Pengfei Cui
- Department of Graduate Administration, Chinese PLA General Hospital, Beijing, People's Republic of China.,Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhibo Zhang
- Department of Graduate Administration, Chinese PLA General Hospital, Beijing, People's Republic of China.,Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Di Huang
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China.,School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Zhaozhen Wu
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China.,School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Xuan Zheng
- Department of Graduate Administration, Chinese PLA General Hospital, Beijing, People's Republic of China.,Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yi Hu
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
321
|
Hissong E, Baek I, Costa V, Beneck D, Saxena A, Solomon JP, Song W. Identification of a Microsatellite Stable, EGFR-Mutant Lung Adenocarcinoma Developing in a Patient With Lynch Syndrome. JCO Precis Oncol 2020; 4:818-822. [PMID: 35050755 DOI: 10.1200/po.20.00074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Erika Hissong
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY
| | - Inji Baek
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY
| | - Victoria Costa
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY
| | - Debra Beneck
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY
| | - Ashish Saxena
- Department of Medicine New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY
| | - James P Solomon
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY
| | - Wei Song
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY
| |
Collapse
|
322
|
Halbert B, Einstein DJ. Hot or Not: Tumor Mutational Burden (TMB) as a Biomarker of Immunotherapy Response in Genitourinary Cancers. Urology 2020; 147:119-126. [PMID: 33137348 DOI: 10.1016/j.urology.2020.10.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/31/2022]
Abstract
Pembrolizumab was recently approved for treatment of cancers with high tumor mutational burden (TMB). We conduct a focused literature review of TMB as a predictive biomarker. TMB quantifies the sum of nonsynonymous coding mutations (typically single nucleotide substitutions and short insertion-deletions) per megabase of sequenced DNA. As a proxy for expression of immunogenic neoantigens, TMB may be an effective predictive biomarker for response to immune checkpoint inhibitors. However, like other biomarkers in this setting, TMB has many limitations; the effect of this FDA approval in the current management of genitourinary cancers is likely limited to select situations.
Collapse
Affiliation(s)
- Brian Halbert
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA
| | - David J Einstein
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA.
| |
Collapse
|
323
|
Montesion M, Murugesan K, Jin DX, Sharaf R, Sanchez N, Guria A, Minker M, Li G, Fisher V, Sokol ES, Pavlick DC, Moore JA, Braly A, Singal G, Fabrizio D, Comment LA, Rizvi NA, Alexander BM, Frampton GM, Hegde PS, Albacker LA. Somatic HLA Class I Loss Is a Widespread Mechanism of Immune Evasion Which Refines the Use of Tumor Mutational Burden as a Biomarker of Checkpoint Inhibitor Response. Cancer Discov 2020; 11:282-292. [PMID: 33127846 DOI: 10.1158/2159-8290.cd-20-0672] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/14/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022]
Abstract
Neoantigen presentation arises as a result of tumor-specific mutations and is a critical component of immune surveillance that can be abrogated by somatic LOH of the human leukocyte antigen class I (HLA-I) locus. To understand the role of HLA-I LOH in oncogenesis and treatment, we utilized a pan-cancer genomic dataset of 83,644 patient samples, a small subset of which had treatment outcomes with immune checkpoint inhibitors (ICI). HLA-I LOH was common (17%) and unexpectedly had a nonlinear relationship with tumor mutational burden (TMB). HLA-I LOH was frequent at intermediate TMB, yet prevalence decreased above 30 mutations/megabase, suggesting highly mutated tumors require alternate immune evasion mechanisms. In ICI-treated patients with nonsquamous non-small cell lung cancer, HLA-I LOH was a significant negative predictor of overall survival. Survival prediction improved when combined with TMB, suggesting TMB with HLA-I LOH may better identify patients likely to benefit from ICIs. SIGNIFICANCE: This work shows the pan-cancer landscape of HLA-I LOH, revealing an unexpected "Goldilocks" relationship between HLA-I LOH and TMB, and demonstrates HLA-I LOH as a significant negative predictor of outcomes after ICI treatment. These data informed a combined predictor of outcomes after ICI and have implications for tumor vaccine development.This article is highlighted in the In This Issue feature, p. 211.
Collapse
Affiliation(s)
| | | | - Dexter X Jin
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | - Radwa Sharaf
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | - Nora Sanchez
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | - Ameet Guria
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | - Max Minker
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | - Gerald Li
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | | | | | | | - Jay A Moore
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | - Alan Braly
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | | | | | | | - Naiyer A Rizvi
- Columbia University Irving Medical Center, New York, New York
| | | | | | | | | |
Collapse
|
324
|
Medjebar S, Truntzer C, Perrichet A, Limagne E, Fumet JD, Richard C, Elkrief A, Routy B, Rébé C, Ghiringhelli F. Angiotensin-converting enzyme (ACE) inhibitor prescription affects non-small-cell lung cancer (NSCLC) patients response to PD-1/PD-L1 immune checkpoint blockers. Oncoimmunology 2020; 9:1836766. [PMID: 33178495 PMCID: PMC7595630 DOI: 10.1080/2162402x.2020.1836766] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 10/06/2020] [Accepted: 10/10/2020] [Indexed: 12/15/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitors are frequently used to treat hypertension and congestive heart failure. Preclinical data show that ACE plays a role on both innate and adaptive immune responses. Since interactions between ACE inhibitors and immune checkpoint inhibitors (ICIs) have not been reported, the aim of this study is to investigate the influence of ACE inhibitors on non-small cell lung cancer (NSCLC) patients treated with programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) inhibitors. We conducted a retrospective cohort analysis of NSCLC patients treated with PD-1/PD-L1 inhibitors. Clinical and co-medication data as well as tumor biopsies were collected. Groups were defined according to patients' co-medications at the time of ICI initiation. Among the 178 patients included, 22 (13.1%) received ACE inhibitors. While baseline characteristics were similar in both groups, ACE inhibitors group had a shorter median PFS (Progression-Free Survival) compared to the control group: 1.97 vs. 2.56 months, p = .01 (HR = 1.8 CI95% 1.1-2.8). Using CIBERSORT, RNA sequencing suggested that tumors from the ACE inhibitors group had less M1 macrophages, activated mast cells, NK cells and memory activated T cells, thus suggesting an immunosuppressed state. In vitro, the ACE inhibitor, captopril, induced M2 marker at the cell surface of monocytes engaged into M1 differentiation. Thus, ACE inhibitors prescription concomitant to PD-1/PD-L1 inhibitors treatment seems to be associated with impaired outcome and with a tumor immunosuppressed state in patients with advanced NSCLC. These results should be validated in larger prospective cohorts.
Collapse
Affiliation(s)
- Soleine Medjebar
- Department of Medical Oncology, GF Leclerc Centre, Dijon, France
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
| | - Caroline Truntzer
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
- Genetic and Immunology Medical Institute (GIMI), Dijon, France
- INSERM UMR1231, Dijon, France
| | - Anaïs Perrichet
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
- INSERM UMR1231, Dijon, France
| | - Emeric Limagne
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
- INSERM UMR1231, Dijon, France
| | - Jean-David Fumet
- Department of Medical Oncology, GF Leclerc Centre, Dijon, France
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
| | - Corentin Richard
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
| | - Arielle Elkrief
- Research Centre for the University of Montréal (CRCHUM), Montréal. Hematology-Oncology Division, Department of Medicine, University of Montreal Healthcare Centre (CHUM), Montreal, Canada
| | - Bertrand Routy
- Research Centre for the University of Montréal (CRCHUM), Montréal. Hematology-Oncology Division, Department of Medicine, University of Montreal Healthcare Centre (CHUM), Montreal, Canada
| | - Cédric Rébé
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
- INSERM UMR1231, Dijon, France
| | - François Ghiringhelli
- Department of Medical Oncology, GF Leclerc Centre, Dijon, France
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
- Genetic and Immunology Medical Institute (GIMI), Dijon, France
- INSERM UMR1231, Dijon, France
| |
Collapse
|
325
|
Gobbini E, Swalduz A, Giaj Levra M, Ortiz-Cuaran S, Toffart AC, Pérol M, Moro-Sibilot D, Saintigny P. Implementing ctDNA Analysis in the Clinic: Challenges and Opportunities in Non-Small Cell Lung Cancer. Cancers (Basel) 2020; 12:E3112. [PMID: 33114393 PMCID: PMC7693855 DOI: 10.3390/cancers12113112] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor genomic profiling has a dramatic impact on the selection of targeted treatment and for the identification of resistance mechanisms at the time of progression. Solid tissue biopsies are sometimes challenging, and liquid biopsies are used as a non-invasive alternative when tissue is limiting. The clinical relevance of tumor genotyping through analysis of ctDNA is now widely recognized at all steps of the clinical evaluation process in metastatic non-small cell lung cancer (NSCLC) patients. ctDNA analysis through liquid biopsy has recently gained increasing attention as well in the management of early and locally advanced, not oncogene-addicted, NSCLC. Its potential applications in early disease detection and the response evaluation to radical treatments are promising. The aim of this review is to summarize the landscape of liquid biopsies in clinical practice and also to provide an overview of the potential perspectives of development focusing on early detection and screening, the assessment of minimal residual disease, and its potential role in predicting response to immunotherapy. In addition to available studies demonstrating the clinical relevance of liquid biopsies, there is a need for standardization and well-designed clinical trials to demonstrate its clinical utility.
Collapse
Affiliation(s)
- Elisa Gobbini
- Thoracic Oncology Unit, CHU Grenoble-Alpes, 38700 Grenoble, France or (E.G.); (M.G.L.); (A.-C.T.); (D.M.-S.)
- Univ Lyon, Université Claude Bernard Lyon, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69373 Lyon, France; (S.O.-C.)
| | - Aurélie Swalduz
- Department of Medical Oncology, Centre Léon Bérard, 69373 Lyon, France; (A.S.); (M.P.)
| | - Matteo Giaj Levra
- Thoracic Oncology Unit, CHU Grenoble-Alpes, 38700 Grenoble, France or (E.G.); (M.G.L.); (A.-C.T.); (D.M.-S.)
| | - Sandra Ortiz-Cuaran
- Univ Lyon, Université Claude Bernard Lyon, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69373 Lyon, France; (S.O.-C.)
| | - Anne-Claire Toffart
- Thoracic Oncology Unit, CHU Grenoble-Alpes, 38700 Grenoble, France or (E.G.); (M.G.L.); (A.-C.T.); (D.M.-S.)
| | - Maurice Pérol
- Department of Medical Oncology, Centre Léon Bérard, 69373 Lyon, France; (A.S.); (M.P.)
| | - Denis Moro-Sibilot
- Thoracic Oncology Unit, CHU Grenoble-Alpes, 38700 Grenoble, France or (E.G.); (M.G.L.); (A.-C.T.); (D.M.-S.)
| | - Pierre Saintigny
- Univ Lyon, Université Claude Bernard Lyon, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69373 Lyon, France; (S.O.-C.)
- Department of Medical Oncology, Centre Léon Bérard, 69373 Lyon, France; (A.S.); (M.P.)
| |
Collapse
|
326
|
Hallqvist A, Rohlin A, Raghavan S. Immune checkpoint blockade and biomarkers of clinical response in non-small cell lung cancer. Scand J Immunol 2020; 92:e12980. [PMID: 33015859 PMCID: PMC7757202 DOI: 10.1111/sji.12980] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 12/26/2022]
Abstract
Immunotherapy with PD‐1 and PD‐L1 inhibitors has revolutionized the treatment for patients with NSCLC the last years with increased overall survival and in particular increased number of long‐time survivors in patients with metastatic disease. It is now a treatment of choice for patients with distant metastases (stage IV) and in conjunction with chemoradiotherapy for patients with limited spread confined to the chest (stage III). PD‐1 inhibition has been proven to be superior to standard chemotherapy, both as a single treatment and when combined with either chemotherapy or CTLA‐4 inhibition. Despite the success of immunotherapy, the majority of patients do not respond or relapse within a short time frame. Biomarkers that would help to properly select patients with a high likelihood of clinical response to PD‐1 and PD‐L1 inhibitors are scarce and far from optimal, and only one (PD‐L1 expression) has reached clinical practice. Thus for immunotherapy to be effective, the discovery and validation of additional biomarkers is critical for patient selection and prediction of clinical response. In this mini‐review, we give an overview of current clinical management of NSCLC including treatment landscape with regard to immunotherapy, as well as discuss the current genetic and immune cell biomarker studies and their potential for introduction into clinical practice.
Collapse
Affiliation(s)
- Andreas Hallqvist
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna Rohlin
- Laboratory Medicine, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Unit of Genetic Analysis and Bioinformatics, Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sukanya Raghavan
- Department of Microbiology and Immunology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
327
|
Li M, Huang L, Ren X, Liu L, Shi Q, Liu L, Wang X, Tian Y, Yu L, Mi F. The incidence risk of programmed cell death-1/programmed cell death ligand 1 inhibitor-related alopecia for cancer patients: A systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e22555. [PMID: 33080690 PMCID: PMC7571948 DOI: 10.1097/md.0000000000022555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE To evaluate the incidence risk of programmed cell death-1/programmed cell death ligand 1 (PD-1/PD-L1) inhibitor-related alopecia for cancer patients, the meta-analysis was put into practice. METHOD The meta-analysis was designed and put into practice according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. RESULTS After rigorous screening and verification, 22 clinical trials involving PD-1/PD-L1 inhibitors were collected for the final comprehensive analysis. The incidence risk of alopecia for all-grade in the PD-1/PD-L1 group was significantly lower than that in the control chemotherapy group (odds ratio [OR] = 0.01, 95% confidence interval [CI]: [0.01, 0.04], I = 86%, Z = 8.73 [P < .00001]). Similar to the above, the incidence risk of alopecia for grade 3-5 related to PD-1/PD-L1 was obvious lower than the control group (OR = 0.17, 95% CI:[0.05, 0.55], I = 0%, Z = 2.97 [P = .003]). When 7 clinical trials (PD-1/PD-L1 + Chemotherapy vs Chemotherapy) were taken to evaluate the risk of alopecia for all-grade and grade 3-5, no statistically significant results were found. CONCLUSION The incidence risk of alopecia caused by PD-1/PD-L1 is significantly lower than chemotherapy, and there is no statistical significant evidence that PD-1/PD-L1 combined with chemotherapy would increase the incidence risk of alopecia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lili Yu
- Department of Radiotherapy Oncology
| | - Fuli Mi
- Gastrointestinal Endoscopy Center, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
328
|
Li S, Zhang C, Pang G, Wang P. Emerging Blood-Based Biomarkers for Predicting Response to Checkpoint Immunotherapy in Non-Small-Cell Lung Cancer. Front Immunol 2020; 11:603157. [PMID: 33178229 PMCID: PMC7596386 DOI: 10.3389/fimmu.2020.603157] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have brought impressive clinical benefits in a variety of malignancies over the past years, which dramatically revolutionized the cancer treatment paradigm. Monotherapy or in combination with chemotherapy of ICIs targeting programmed death 1/programmed death ligand 1 (PD-L1) has emerged as an alternative treatment for patients with advanced non-small-cell lung cancer (NSCLC). However, constrained by primary or acquired resistance, most patients obtain limited benefits from ICIs and occasionally suffer from severe immune-related adverse events. Moreover, owing to the complexity of the tumor microenvironment and the technical limitations, clinical application of PD-L1 and tumor mutation burden as biomarkers shows many deficiencies. Thus, additional predictive biomarkers are required to further advance the precision of proper patient selection, avoiding the exposure of potential non-responders to unnecessary immunotoxicity. Nowadays, an increasing number of investigations are focusing on peripheral blood as a noninvasive alternative to tissue biopsy in predicting and monitoring treatment outcomes. Herein, we summarize the emerging blood-based biomarkers that could predict the clinical response to checkpoint immunotherapy, specifically in patients with NSCLC.
Collapse
Affiliation(s)
- Shumin Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Chengyan Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Guanchao Pang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Pingli Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
329
|
Sesma A, Pardo J, Cruellas M, Gálvez EM, Gascón M, Isla D, Martínez-Lostao L, Ocáriz M, Paño JR, Quílez E, Ramírez A, Torres-Ramón I, Yubero A, Zapata M, Lastra R. From Tumor Mutational Burden to Blood T Cell Receptor: Looking for the Best Predictive Biomarker in Lung Cancer Treated with Immunotherapy. Cancers (Basel) 2020; 12:E2974. [PMID: 33066479 PMCID: PMC7602200 DOI: 10.3390/cancers12102974] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/30/2020] [Accepted: 10/12/2020] [Indexed: 12/20/2022] Open
Abstract
Despite therapeutic advances, lung cancer (LC) is one of the leading causes of cancer morbidity and mortality worldwide. Recently, the treatment of advanced LC has experienced important changes in survival benefit due to immune checkpoint inhibitors (ICIs). However, overall response rates (ORR) remain low in unselected patients and a large proportion of patients undergo disease progression in the first weeks of treatment. Therefore, there is a need of biomarkers to identify patients who will benefit from ICIs. The programmed cell death ligand 1 (PD-L1) expression has been the first biomarker developed. However, its use as a robust predictive biomarker has been limited due to the variability of techniques used, with different antibodies and thresholds. In this context, tumor mutational burden (TMB) has emerged as an additional powerful biomarker based on the observation of successful response to ICIs in solid tumors with high TMB. TMB can be defined as the total number of nonsynonymous mutations per DNA megabases being a mechanism generating neoantigens conditioning the tumor immunogenicity and response to ICIs. However, the latest data provide conflicting results regarding its role as a biomarker. Moreover, considering the results of the recent data, the use of peripheral blood T cell receptor (TCR) repertoire could be a new predictive biomarker. This review summarises recent findings describing the clinical utility of TMB and TCRβ (TCRB) and concludes that immune, neontigen, and checkpoint targeted variables are required in combination for accurately identifying patients who most likely will benefit of ICIs.
Collapse
Affiliation(s)
- Andrea Sesma
- Medical Oncology Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain; (M.C.); (M.G.); (D.I.); (M.O.); (E.Q.); (I.T.-R.); (A.Y.); (M.Z.); (R.L.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
| | - Julián Pardo
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
- ARAID Foundation (IIS Aragón), 50009 Zaragoza, Spain
- Microbiology, Preventive Medicine and Public Health Department, Medicine, University of Zaragoza, 50009 Zaragoza, Spain
- Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine Network (CIBER-BBN), 28029 Madrid, Spain
| | - Mara Cruellas
- Medical Oncology Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain; (M.C.); (M.G.); (D.I.); (M.O.); (E.Q.); (I.T.-R.); (A.Y.); (M.Z.); (R.L.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
| | - Eva M. Gálvez
- Instituto de Carboquímica (ICB-CSIC), Miguel Luesma 4, 50018 Zaragoza, Spain;
| | - Marta Gascón
- Medical Oncology Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain; (M.C.); (M.G.); (D.I.); (M.O.); (E.Q.); (I.T.-R.); (A.Y.); (M.Z.); (R.L.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
| | - Dolores Isla
- Medical Oncology Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain; (M.C.); (M.G.); (D.I.); (M.O.); (E.Q.); (I.T.-R.); (A.Y.); (M.Z.); (R.L.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
| | - Luis Martínez-Lostao
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
- Immunology Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain
- Department of Microbiology, Pediatrics, Radiology and Public Health, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Nanoscience Institute, 50018 Zaragoza, Spain
- Aragon Materials Science Institute, 50009 Zaragoza, Spain
| | - Maitane Ocáriz
- Medical Oncology Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain; (M.C.); (M.G.); (D.I.); (M.O.); (E.Q.); (I.T.-R.); (A.Y.); (M.Z.); (R.L.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
| | - José Ramón Paño
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
- Infectious Disease Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain
| | - Elisa Quílez
- Medical Oncology Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain; (M.C.); (M.G.); (D.I.); (M.O.); (E.Q.); (I.T.-R.); (A.Y.); (M.Z.); (R.L.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
| | - Ariel Ramírez
- Nanotoxicology and Immunotoxicology Unit (IIS Aragón), 50009 Zaragoza, Spain;
| | - Irene Torres-Ramón
- Medical Oncology Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain; (M.C.); (M.G.); (D.I.); (M.O.); (E.Q.); (I.T.-R.); (A.Y.); (M.Z.); (R.L.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
| | - Alfonso Yubero
- Medical Oncology Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain; (M.C.); (M.G.); (D.I.); (M.O.); (E.Q.); (I.T.-R.); (A.Y.); (M.Z.); (R.L.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
| | - María Zapata
- Medical Oncology Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain; (M.C.); (M.G.); (D.I.); (M.O.); (E.Q.); (I.T.-R.); (A.Y.); (M.Z.); (R.L.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
| | - Rodrigo Lastra
- Medical Oncology Department, University Hospital Lozano Blesa, 50009 Zaragoza, Spain; (M.C.); (M.G.); (D.I.); (M.O.); (E.Q.); (I.T.-R.); (A.Y.); (M.Z.); (R.L.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (J.P.); (L.M.-L.); (J.R.P.)
| |
Collapse
|
330
|
Abstract
Immunotherapy has transformed the treatment of many tumors. Robust data demonstrating improved overall survival and progression-free survival in patients treated with monoclonal antibodies have established immune checkpoint inhibitors as standard of care in stages III and IV non-small cell lung cancer. Nivolumab is effective in previously treated patients with metastatic non-small cell lung cancer. Pembrolizumab and atezolizumab are approved as monotherapy and in combination with other therapies. Ongoing trials investigate the potential role of immunotherapy in earlier disease settings. Identifying predictive biomarkers of response will further amplify the impact of immune checkpoint inhibitors in the treatment of non-small cell lung cancer.
Collapse
Affiliation(s)
- Melinda L Hsu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, 1650 Orleans Street CRB1 186, Baltimore, MD 21287, USA.
| | - Jarushka Naidoo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, 401 North Broadway, Baltimore, MD 21287, USA. https://twitter.com/DrJNaidoo
| |
Collapse
|
331
|
Huang Z, Su W, Lu T, Wang Y, Dong Y, Qin Y, Liu D, Sun L, Jiao W. First-Line Immune-Checkpoint Inhibitors in Non-Small Cell Lung Cancer: Current Landscape and Future Progress. Front Pharmacol 2020; 11:578091. [PMID: 33117170 PMCID: PMC7577011 DOI: 10.3389/fphar.2020.578091] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is one of the most common cancers and the leading cause of cancer-related deaths worldwide. Most of these patients with non-small cell lung cancer (NSCLC) present with the advanced stage of the disease at the time of diagnosis, and thus decrease the 5-year survival rate to about 5%. Immune checkpoint inhibitors (ICIs) can act on the inhibitory pathway of cancer immune response, thereby restoring and maintaining anti-tumor immunity. There are already ICIs targeting different pathways, including the programmed cell death 1 (PD-1), programmed cell death ligand 1 (PD-L1), and cytotoxic T lymphocyte antigen 4 (CTLA-4) pathway. Since March 2015, the US Food and Drug Administration (FDA) approved nivolumab (anti-PD-1 antibody) as the second-line option for treatment of patients with advanced squamous NSCLC. Additionally, a series of inhibitors related to PD-1/PD-L1 immune-checkpoints have helped in the immunotherapy of NSCLC patients, and modified the original treatment model. However, controversies remain regarding the use of ICIs in a subgroup with targeted oncogene mutations is a problem that we need to solve. On the other hand, there are continuous efforts to find biomarkers that effectively predict the response of ICIs to screen suitable populations. In this review, we have reviewed the history of the continuous developments in cancer immunotherapy, summarized the mechanism of action of the immune-checkpoint pathways. Finally, based on the results of the first-line recent trials, we propose a potential first-line immunotherapeutic strategy for the treatment of the patients with NSCLC.
Collapse
Affiliation(s)
- Zhangfeng Huang
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenhao Su
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tong Lu
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuanyong Wang
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanting Dong
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yi Qin
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dahai Liu
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lili Sun
- Department of Ultrasound, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Wenjie Jiao
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
332
|
Yang Z, Wei S, Deng Y, Wang Z, Liu L. Clinical significance of tumour mutation burden in immunotherapy across multiple cancer types: an individual meta-analysis. Jpn J Clin Oncol 2020; 50:1023-1031. [PMID: 32542383 DOI: 10.1093/jjco/hyaa076] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 05/07/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Biomarkers for stratifying patients that could benefit from immune checkpoint inhibitors are necessary. Tumour mutation burden has recently become a promising biomarker in cancer, but the associations between tumour mutation burden and outcomes of immune checkpoint inhibitors treatment were not well-documented in present studies. METHODS We searched PubMed, Web of Science and EMBASE databases up to 1 October 2019. Studies evaluated the association between tumour mutation burden and clinical outcomes were included. Hazard ratios and odds ratios were applied to estimate the association of tumour mutation burden score with overall survival, progression-free survival and response rate, respectively. The best cut-off value was chosen by best discriminated overall survival using Contal and O'Quigley method. RESULTS Twenty-two studies involving 6171 patients in diverse cancers were included. The individual participant data meta-analysis demonstrated that high tumour mutation burden was associated with better overall survival (HR = 0.57, 95% CI = 0.50-0.64) and progression-free survival (HR = 0.50, 95% CI = 0.40-0.63) and higher response rate. The best cut-off values in each cancer type were 17.7/MB in non-small cell lung cancer, 7.9/MB in bladder cancer, 6.1/MB in melanoma, 12.3/MB in colorectal cancer, 6.9/MB in esophagogastric cancer, 10.5/MB in head and neck cancer. The pooled meta-analysis showed the prognosis value was robust and the sensitivity, specificity and area under the receiver operating characteristic curves in predicting response rates were 0.63, 0.71 and 0.73, respectively. CONCLUSIONS The present meta-analysis indicates tumour mutation burden is a promising predictor of immune checkpoint inhibitors therapy but the cut-off value differs in different cancers.
Collapse
Affiliation(s)
- Zhenyu Yang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China.,Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Shiyou Wei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China.,Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Yulan Deng
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China.,Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Zihuai Wang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China.,Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Lunxu Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China.,Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| |
Collapse
|
333
|
Pender A, Titmuss E, Pleasance ED, Fan KY, Pearson H, Brown SD, Grisdale CJ, Topham JT, Shen Y, Bonakdar M, Taylor GA, Williamson LM, Mungall KL, Chuah E, Mungall AJ, Moore RA, Lavoie JM, Yip S, Lim H, Renouf DJ, Sun S, Holt RA, Jones SJM, Marra MA, Laskin J. Genome and Transcriptome Biomarkers of Response to Immune Checkpoint Inhibitors in Advanced Solid Tumors. Clin Cancer Res 2020; 27:202-212. [PMID: 33020056 DOI: 10.1158/1078-0432.ccr-20-1163] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/06/2020] [Accepted: 09/30/2020] [Indexed: 11/16/2022]
Abstract
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.
Collapse
Affiliation(s)
- Alexandra Pender
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Emma Titmuss
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Erin D Pleasance
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Kevin Y Fan
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hillary Pearson
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Scott D Brown
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Cameron J Grisdale
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | | | - Yaoqing Shen
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Melika Bonakdar
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Gregory A Taylor
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Laura M Williamson
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Karen L Mungall
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Eric Chuah
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Jean-Michel Lavoie
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Howard Lim
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Daniel J Renouf
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
- Pancreas Centre BC, Vancouver, British Columbia, Canada
| | - Sophie Sun
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Robert A Holt
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Janessa Laskin
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada.
| |
Collapse
|
334
|
Shao C, Li G, Huang L, Pruitt S, Castellanos E, Frampton G, Carson KR, Snow T, Singal G, Fabrizio D, Alexander BM, Jin F, Zhou W. Prevalence of High Tumor Mutational Burden and Association With Survival in Patients With Less Common Solid Tumors. JAMA Netw Open 2020; 3:e2025109. [PMID: 33119110 PMCID: PMC7596577 DOI: 10.1001/jamanetworkopen.2020.25109] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
IMPORTANCE Tumor mutational burden (TMB) is a potential biomarker associated with response to immune checkpoint inhibitor therapies. The prognostic value associated with TMB in the absence of immunotherapy is uncertain. OBJECTIVE To assess the prevalence of high TMB (TMB-H) and its association with overall survival (OS) among patients not treated with immunotherapy with the same 10 tumor types from the KEYNOTE-158 study. DESIGN, SETTING, AND PARTICIPANTS This retrospective cohort study evaluated the prognostic value of TMB-H, assessed by Foundation Medicine (FMI) and defined as at least 10 mutations/megabase (mut/Mb) in the absence of immunotherapy. Data were sourced from the deidentified Flatiron Health-FMI clinicogenomic database collected up to July 31, 2018. Eligible patients were aged 18 years or older with any of the following solid cancer types: anal, biliary, endometrial, cervical, vulvar, small cell lung, thyroid, salivary gland, mesothelioma, or neuroendocrine tumor. Patients with microsatellite instability-high tumors were excluded from primary analysis. For OS analysis, patients were excluded if immunotherapy started on the FMI report date or earlier or if patients died before January 1, 2012, and patients were censored if immunotherapy was started later than the FMI report date. Data were analyzed from November 2018 to February 2019. MAIN OUTCOMES AND MEASURES Overall survival was analyzed using the Kaplan-Meier method and Cox proportional hazards model, adjusting for age, sex, cancer types, practice type, and albumin level. RESULTS Of 2589 eligible patients, 1671 (64.5%) were women, and the mean (SD) age was 63.7 (11.7) years. Median (interquartile range) TMB was 2.6 (1.7-6.1) mut/Mb, and 332 patients (12.8%) had TMB-H (≥10 mut/Mb). Prevalence of TMB-H was highest among patients with small cell lung cancer (40.0%; 95% CI, 34.7%-45.6%) and neuroendocrine tumor (29.3%; 95% CI, 22.8%-36.6%) and lowest was among patients with mesothelioma (1.2%; 95% CI, 0.3%-4.4%) and thyroid cancer (2.7%; 95% CI, 1.2%-5.7%). Adjusted hazard ratio for OS of patients not treated with immunotherapy with TMB-H vs those without TMB-H was 0.94 (95% CI, 0.77-1.13). Comparable results were observed when including patients with high microsatellite instability tumors and calculating OS from first observed antineoplastic treatment date. CONCLUSIONS AND RELEVANCE These findings suggest that prevalence of TMB-H varies widely depending on tumor type and TMB-H does not appear to be a factor associated with OS among patients across these cancer types treated in the absence of immunotherapy.
Collapse
Affiliation(s)
| | - Gerald Li
- Foundation Medicine, Cambridge, Massachusetts
| | | | | | | | | | | | | | | | | | | | - Fan Jin
- Merck and Co, Kenilworth, New Jersey
| | - Wei Zhou
- Merck and Co, Kenilworth, New Jersey
| |
Collapse
|
335
|
Imyanitov EN, Ivantsov AO, Tsimafeyeu IV. Harmonization of Molecular Testing for Non-Small Cell Lung Cancer: Emphasis on PD-L1. Front Oncol 2020; 10:549198. [PMID: 33102215 PMCID: PMC7554524 DOI: 10.3389/fonc.2020.549198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/14/2020] [Indexed: 12/23/2022] Open
Abstract
Comprehensive molecular testing plays a critical role in the choice of treatment for non-small lung cell cancer (NSCLC). The analysis of druggable alterations in EGFR, BRAF, MET, KRAS, ALK, ROS1, RET and NTRK1/2/3 genes is more or less standardized and can be achieved using a single diagnostic platform, e.g., next generation sequencing (NGS) or polymerase chain reaction (PCR). In contrast to above targets, PD-L1 testing requires the use of immunohistochemistry (IHC). There are multiple PD-L1 IHC assays, which utilize distinct antibodies and detection systems. These PD-L1 tests are tailored to distinct drugs, often rely on different thresholds and scoring guidelines, and are characterized by incomplete inter-laboratory and inter-observer reproducibility. Several studies evaluated the performance of PD-L1 RNA expression tests, as PCR-based RNA analysis is compatible with other NSCLC molecular testing platforms, can be performed in a semi-automated manner, and has a potential for proper standardization. These investigations revealed a correlation between PD-L1 protein and RNA expression; however, there were NSCLCs demonstrating decent amounts of PD-L1 transcript in the absence of PD-L1 IHC staining. Clinical studies are required to evaluate, which of the two PD-L1 testing approaches, i.e., RNA or protein expression measurement, has a better predictive value.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St. Petersburg, Russia.,Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, Saint Petersburg, Russia
| | - Alexandr O Ivantsov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St. Petersburg, Russia.,Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, Saint Petersburg, Russia
| | | |
Collapse
|
336
|
Morphological, immune and genetic features in biopsy sample associated with the efficacy of pembrolizumab in patients with non-squamous non-small cell lung cancer. J Cancer Res Clin Oncol 2020; 147:1227-1237. [PMID: 32997195 DOI: 10.1007/s00432-020-03413-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/24/2020] [Indexed: 10/23/2022]
Abstract
INTRODUCTION The usefulness of the histopathology of biopsy samples for predicting the efficacy of immunotherapy in non-squamous, non-small cell lung cancer (NSq NSCLC) patients remains unclear. METHODS We retrospectively investigated the associations between the histopathological features in biopsy samples and survival outcomes in advanced NSq NSCLC patients receiving pembrolizumab. NSq NSCLC was classified histopathologically as morphological adenocarcinoma or non-small cell carcinoma (NSCC: absence of definitive features of either adenocarcinoma or a squamous morphology). We investigated the association between the tumor morphological features and immune/genetic features by examining the tumor PD-L1 expression and tumor mutation burden (TMB). RESULTS Among 33 advanced NSq NSCLC patients with tumor PD-L1 scores ≥ 50% receiving pembrolizumab as first-line therapy, a biopsy diagnosis of NSCC was associated with a significantly longer progression-free survival [median 16.8 vs. 2.3 months; hazard ratio (HR) 0.26; 95% CI 0.10-0.62, P = 0.01] and overall survival (median NR vs. 10.1 months; HR 0.35; 0.12-0.97, P = 0.04) as compared to that of morphological adenocarcinoma. In an analysis of 367 biopsy samples, the NSCC group showed a higher percentage of samples with PD-L1 scores ≥ 50% than the morphological adenocarcinoma group (35% vs. 10%). The NSCC group (n = 8) also showed a significantly higher TMB than the morphological adenocarcinoma group (n = 7) (median 236 vs. 25 mutations/whole exome, P = 0.01). CONCLUSION Absence of definitive morphological features in a biopsy sample could be a useful predictor of the efficacy of pembrolizumab in NSq NSCLC patients with tumor PD-L1 scores ≥ 50%, as these tumors are likely to show high tumor PD-L1 expression and high TMB.
Collapse
|
337
|
Xiang L, Fu X, Wang X, Li W, Zheng X, Nan K, Tian T. A Potential Biomarker of Combination of Tumor Mutation Burden and Copy Number Alteration for Efficacy of Immunotherapy in KRAS-Mutant Advanced Lung Adenocarcinoma. Front Oncol 2020; 10:559896. [PMID: 33072585 PMCID: PMC7541961 DOI: 10.3389/fonc.2020.559896] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/03/2020] [Indexed: 01/16/2023] Open
Abstract
Objectives The Kirsten Rat Sarcoma (KRAS) mutation is the commonest oncogenic drive mutation in lung adenocarcinoma (LUAD) and immunotherapy may be quite promising for KRAS-mutant LUAD. While the effects of tumor mutation burden (TMB) and copy number alteration (CNA) are poorly understood in this illness, our study aimed to explore the roles TMB and CNA play in the prediction of response to immune checkpoint inhibitor (ICI) therapy in advanced KRAS-mutant LUAD. Methods Mutation and clinical data were downloaded from cBioPortal. We evaluated KRAS mutation status and divided patients into different subgroups based on TMB and CNA cutoffs to investigate the predictive value of these biomarkers on ICI response. Results KRAS mutation with concurrent TP53 or STK11 mutations had higher TMB and CNA compared to KRAS mutation alone. The KRAS G12C and G > T mutation subgroups, with TP53 or STK11 co-mutation, also had higher TMB and CNA. We found that TMB and CNA were independently associated with progression-free survival (PFS) and durable clinical benefits (DCB); TMB was positively correlated with PFS (P = 0.0074) and DCB (P = 0.0008) while low CNA was associated with prolonged PFS (P = 0.0060) and DCB (P = 0.0018). However, TMB alone did not distinguish benefits among KRAS-mutant patients. Notably, when combining TMB and CNA, low TMB and high CNA revealed worse outcomes of ICI therapy (mPFS: 2.20m, P = 0.0023; proportion of DCB: 24%, P = 0.0001). Conclusion The combination of TMB and CNA provides more sensible and accurate prediction of ICI response than individual factors in KRAS-mutant LUAD. Moreover, low TMB and high CNA can be utilized as a potential biomarker to predict adverse outcome in KRAS-mutant LUAD.
Collapse
Affiliation(s)
- Luochengling Xiang
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao Fu
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao Wang
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wenyuan Li
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoqiang Zheng
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kejun Nan
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Oncology Hospital, Xi'an International Medical Center Hospital, Xi'an, China
| | - Tao Tian
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
338
|
中国临床肿瘤学会非小细胞肺癌专家委员会. [Chinese Expert Consensus on Next Generation Sequencing Diagnosis
for Non-small Cell Lung Cancer (2020 Edition)]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:741-761. [PMID: 32957170 PMCID: PMC7519957 DOI: 10.3779/j.issn.1009-3419.2020.101.45] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
339
|
Li G, Wang G, Guo Y, Li S, Zhang Y, Li J, Peng B. Development of a novel prognostic score combining clinicopathologic variables, gene expression, and mutation profiles for lung adenocarcinoma. World J Surg Oncol 2020; 18:249. [PMID: 32950055 PMCID: PMC7502202 DOI: 10.1186/s12957-020-02025-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Integrating phenotypic and genotypic information to improve prognostic prediction is under active investigation for lung adenocarcinoma (LUAD). In this study, we developed a new prognostic model for event-free survival (EFS) and recurrence-free survival (RFS) based on the combination of clinicopathologic variables, gene expression, and mutation data. METHODS We enrolled a total of 408 patients from the Cancer Genome Atlas Lung Adenocarcinoma (TCGA-LUAD) project for the study. We pre-selected gene expression or mutation features and constructed 14 different input feature sets for predictive model development. We assessed model performance with multiple evaluation metrics including the distribution of C-index on testing dataset, risk score significance, and time-dependent AUC under competing risks scenario. We stratified patients into higher- and lower-risk subgroups by the final risk score and further investigated underlying immune phenotyping variations associated with the differential risk. RESULTS The model integrating all three types of data achieved the best prediction performance. The resultant risk score provided a higher-resolution risk stratification than other models within pathologically defined subgroups. The score could account for extra EFS-related variations that were not captured by clinicopathologic scores. Being validated for RFS prediction under a competing risks modeling framework, the score achieved a significantly higher time-dependent AUC as compared to that of the conventional clinicopathologic variables-based model (0.772 vs. 0.646, p value < 0.001). The higher-risk patients were characterized with transcriptional aberrations of multiple immune-related genes, and a significant depletion of mast cells and natural killer cells. CONCLUSIONS We developed a novel prognostic risk score with improved prediction accuracy, using clinicopathologic variables, gene expression and mutation profiles as input, for LUAD. Such score was a significant predictor of both EFS and RFS. TRIAL REGISTRATION This study was based on public open data from TCGA and hence the study objects were retrospectively registered.
Collapse
Affiliation(s)
- Guofeng Li
- Department of Thoracic Surgery, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Luohu District, Shenzhen, 518020, China
| | - Guangsuo Wang
- Department of Thoracic Surgery, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Luohu District, Shenzhen, 518020, China
| | - Yanhua Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Yangpu District, Shanghai, 200433, China
| | - Shixuan Li
- Department of Thoracic Surgery, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Luohu District, Shenzhen, 518020, China
| | - Youlong Zhang
- Department of Biostatistics, HuaJia Biomedical Intelligence, Shenzhen Overseas Chinese High-Tech Venture Park, Nanshan District, Shenzhen, 518057, China
| | - Jialu Li
- Department of Biostatistics, HuaJia Biomedical Intelligence, Shenzhen Overseas Chinese High-Tech Venture Park, Nanshan District, Shenzhen, 518057, China.
| | - Bin Peng
- Department of Thoracic Surgery, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Luohu District, Shenzhen, 518020, China.
| |
Collapse
|
340
|
Schardt J. [The use of immune checkpoint inhibitors in routine oncology]. Z Rheumatol 2020; 79:809-817. [PMID: 32936368 PMCID: PMC7653782 DOI: 10.1007/s00393-020-00876-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2020] [Indexed: 11/12/2022]
Abstract
Hintergrund Die Einführung von Immuncheckpoint-Inhibitoren (ICI) hat die Behandlungskonzepte der Onkologie für eine Vielzahl von unterschiedlichen Krebsarten maßgeblich verändert. Dabei werden in der klinischen Routine v. a. humanisierte Antikörper gegen Immuncheckpoints wie „cytotoxic T‑lymphocyte associated protein 4“ (CTLA-4) oder „programmed cell death 1/programmed cell death ligand 1“ (PD1/PD-L1) eingesetzt. Fragestellung Übersicht zur Therapielandschaft mit Immuncheckpoint-Inhibitoren bei mehrheitlich soliden Tumoren in der Onkologie. Material und Methoden Darstellung und Diskussion aktueller Studienresultate, Einbezug aktueller Behandlungsempfehlungen und Zulassungsindikationen. Ergebnisse Sieben verschiedene Immuncheckpoint-Inhibitoren werden in der Onkologie therapeutisch eingesetzt: ein Anti-CTLA-4-Antikörper, 3 Anti-PD1-Antikörper und 3 Anti-PD-L1-Antiköper. FDA-Zulassung auf dem US-Markt für 17 verschiedene Tumorentitäten und einer agnostischen Indikation (Tumoren mit defizienter Mismatch-repair-Maschinerie/hohe Mikrosatelliteninstabilität). Langzeitremissionen sind in ca. zwei Drittel der Patienten mit Tumoransprechen möglich. Schlussfolgerungen Nutzen der Immuncheckpoint-Inhibitoren nur für einen Teil der behandelten Patienten. Primäre und sekundäre Resistenzmechanismen erst in Anfängen verstanden. Kombinationstherapien der Immuncheckpoint-Inhibitoren mit z. B. Chemotherapie, neuen Immuncheckpoint-Inhibitoren (z. B. Anti-LAG3-Antikörper) oder gezielten Therapien (z. B. CDK4/6, PARP-Inhibitoren) zur Verbesserung der Wirksamkeit werden in klinischen Studien untersucht. Verlässliche, prädiktive Marker sind dringend erforderlich.
Collapse
Affiliation(s)
- Julian Schardt
- Universitätsklinik für Medizinische Onkologie, Inselspital, Freiburgstr. 41G, 3010, Bern, Schweiz.
| |
Collapse
|
341
|
Champiat S, Tselikas L, Farhane S, Raoult T, Texier M, Lanoy E, Massard C, Robert C, Ammari S, De Baère T, Marabelle A. Intratumoral Immunotherapy: From Trial Design to Clinical Practice. Clin Cancer Res 2020; 27:665-679. [PMID: 32943460 DOI: 10.1158/1078-0432.ccr-20-0473] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/10/2020] [Accepted: 09/14/2020] [Indexed: 11/16/2022]
Abstract
Systemic immunotherapies such as immune checkpoint blockade targeted at PD(L)1 and CTLA4 have demonstrated their ability to provide durable tumor responses and long-term overall survival benefits for some patients in several solid tumor types. However, a majority of patients remain resistant to these treatments and a significant proportion of them develop severe autoimmune and inflammatory adverse events. Preclinical studies have demonstrated that intratumoral injections of immunostimulatory products (oncolytics, pattern recognition receptor agonists,…) that are able to trigger type I IFN release and enhance tumor antigen presentation on immune cells could generate a strong antitumor immunity and overcome the resistance to systemic immune checkpoint blockade therapies. The intratumoral immunotherapy strategies that are currently in clinical development offer a unique therapeutic and exploratory setting to better understand the immune contexture across tumor lesions of patients with metastatic cancer. Also these local therapeutic products could turn cold tumors into hot and improve the response rates to cancer immunotherapies while diminishing their systemic exposure and toxicities. Intratumoral immunotherapies could prime or boost the immunity against tumors and therefore radically change the combinatorial therapeutic strategies currently pursued for metastatic and local cancers to improve their long-term survival. We aimed to review and discuss the scientific rationale for intratumoral immunotherapy, the challenges raised by this strategy in terms of drug development within clinical trials and the current state-of-the-art regarding the clinical practice of this innovative approach.
Collapse
Affiliation(s)
- Stéphane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France.,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France
| | - Lambros Tselikas
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France.,Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Siham Farhane
- Gustave Roussy Immunotherapy Program (GRIP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thibault Raoult
- Service de Promotion des Etudes Cliniques (SPEC), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Matthieu Texier
- Service de Biostatistiques et d'Epidémiologie (SBE), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Emilie Lanoy
- Service de Biostatistiques et d'Epidémiologie (SBE), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Christophe Massard
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Caroline Robert
- Département de Médecine Oncologique (DMO), Gustave Roussy, Université Paris Saclay, Villejuif, France.,Université Paris Saclay, Saint-Aubin, France
| | - Samy Ammari
- Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thierry De Baère
- Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France.,Université Paris Saclay, Saint-Aubin, France
| | - Aurélien Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France. .,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France.,Gustave Roussy Immunotherapy Program (GRIP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| |
Collapse
|
342
|
Marabelle A, Fakih M, Lopez J, Shah M, Shapira-Frommer R, Nakagawa K, Chung HC, Kindler HL, Lopez-Martin JA, Miller WH, Italiano A, Kao S, Piha-Paul SA, Delord JP, McWilliams RR, Fabrizio DA, Aurora-Garg D, Xu L, Jin F, Norwood K, Bang YJ. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol 2020; 21:1353-1365. [PMID: 32919526 DOI: 10.1016/s1470-2045(20)30445-9] [Citation(s) in RCA: 1351] [Impact Index Per Article: 337.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Tumour mutational burden (TMB) has been retrospectively correlated with response to immune checkpoint blockade. We prospectively explored the association of high tissue TMB (tTMB-high) with outcomes in ten tumour-type-specific cohorts from the phase 2 KEYNOTE-158 study, which assessed the anti-PD-1 monoclonal antibody pembrolizumab in patients with selected, previously treated, advanced solid tumours. METHODS In the multi-cohort, open-label, non-randomised, phase 2 KEYNOTE-158 study, patients were enrolled from 81 academic facilities and community-based institutions across 21 countries in Africa, the Americas, Asia, and Europe. Eligible patients were aged 18 years or older, had a histologically or cytologically confirmed advanced (ie, unresectable or metastatic, or both) incurable solid tumour (eligible tumour types were anal, biliary, cervical, endometrial, mesothelioma, neuroendocrine, salivary, small-cell lung, thyroid, and vulvar), progression on or intolerance to one or more lines of standard therapy, had measurable disease per Response Evaluation Criteria in Solid Tumors (RECIST; version 1.1) assessed by independent central radiological review, Eastern Cooperative Oncology Group performance status of 0 or 1, life expectancy of at least 3 months, adequate organ function, and a tumour sample for biomarker analysis. Participants were given pembrolizumab 200 mg intravenously every 3 weeks for up to 35 cycles. Tissue TMB (tTMB) was assessed in formalin-fixed paraffin-embedded tumour samples using the FoundationOne CDx assay (Foundation Medicine, Cambridge, MA, USA). The prespecified definition of tTMB-high status was at least 10 mutations per megabase. The primary endpoint was the proportion of patients with an objective response (complete or partial response) as per Response Evaluation Criteria in Solid Tumours (version 1.1) by independent central review. This prespecified analysis assessed the association between antitumour activity and tTMB in treated patients with evaluable tTMB data. Efficacy was assessed in all participants who received at least one dose of pembrolizumab, had evaluable tTMB data, and were enrolled at least 26 weeks before data cutoff (June 27, 2019), and safety was assessed in all participants who received at least one dose of pembrolizumab and had tTMB-high status. KEYNOTE-158 is registered at ClinicalTrials.gov, NCT02628067, and is ongoing. FINDINGS Between Jan 15, 2016, and June 25, 2019, 1073 patients were enrolled. 1066 participants were treated as of data cutoff (June 27, 2019), of whom 805 (76%) were evaluable for TMB, and 105 (13%) of 805 had tTMB-high status and were assessed for safety. 1050 (98%) of 1066 patients enrolled by at least 26 weeks before data cutoff, of whom 790 (75%) were evaluable for TMB and included in efficacy analyses. 102 (13%) of these 790 patients had tTMB-high status (≥10 mutations per megabase), and 688 (87%) patients had non-tTMB-high status (<10 mutations per megabase). Median study follow-up was 37·1 months (IQR 35·0-38·3). Objective responses were observed in 30 (29%; 95% CI 21-39) of 102 patients in the tTMB-high group and 43 (6%; 5-8) of 688 in the non-tTMB-high group. 11 (10%) of 105 patients had treatment-related serious adverse events. 16 (15%) participants had a grade 3-5 treatment-related adverse event, of which colitis was the only such adverse event that occurred in more than one patient (n=2). One patient had fatal pneumonia that was assessed by the investigator to be treatment related. INTERPRETATION tTMB-high status identifies a subgroup of patients who could have a robust tumour response to pembrolizumab monotherapy. tTMB could be a novel and useful predictive biomarker for response to pembrolizumab monotherapy in patients with previously treated recurrent or metastatic advanced solid tumours. FUNDING Merck Sharp & Dohme Corp, a subsidiary of Merck & Co, Inc.
Collapse
Affiliation(s)
- Aurélien Marabelle
- Gustave Roussy, INSERM U1015, Université Paris-Saclay, Villejuif, France.
| | - Marwan Fakih
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Juanita Lopez
- The Royal Marsden Foundation Trust and the Institute of Cancer Research, London, UK
| | - Manisha Shah
- Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | | | | | - Hyun Cheol Chung
- Yonsei Cancer Center and Yonsei University College of Medicine, Seoul, South Korea
| | | | | | - Wilson H Miller
- Jewish General Hospital and McGill University, Montréal, QC, Canada
| | | | - Steven Kao
- Chris O'Brien Lifehouse, Sydney, NSW, Australia
| | | | | | | | | | | | - Lei Xu
- Merck & Co, Kenilworth, NJ, USA
| | - Fan Jin
- Merck & Co, Kenilworth, NJ, USA
| | | | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
343
|
Clinical biomarkers directing the management of patients with colon and lung cancer (beyond oncogene-addicted NSCLC). FORUM OF CLINICAL ONCOLOGY 2020. [DOI: 10.2478/fco-2019-0014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Abstract
Treatment personalisation plays a key role in the current management of patients with cancer. Several biomarkers have shown clinical utility and may guide therapeutic decisions. Amongst patients with lung cancer, the level of expression of programmed death ligand 1 (PD-L1) has both prognostic and predictive values in terms of response to the inhibition of programmed cell death protein 1 (PD-1). Depending on the clinical setting, the expression of PD-L1 ≥1% or ≥50% has been associated with improved outcomes amongst patients receiving pembrolizumab. Regarding patients with colorectal carcinoma, mutations in the KRAS oncogene predict the responsiveness to the inhibition of epidermal growth factor receptor (EGFR). Only patients with wild-type KRAS tumours derive benefit from cetuximab and panitumumab in terms of response and survival. In conclusion, future research should aim in the optimisation of the use of biomarker in the clinical practice in order to provide the optimal drug combination to each individual patient.
Collapse
|
344
|
Hu H, She L, Liao M, Shi Y, Yao L, Ding D, Zhu Y, Zeng S, Carbone DP, Huang J. Cost-Effectiveness Analysis of Nivolumab Plus Ipilimumab vs. Chemotherapy as First-Line Therapy in Advanced Non-Small Cell Lung Cancer. Front Oncol 2020; 10:1649. [PMID: 33014826 PMCID: PMC7507990 DOI: 10.3389/fonc.2020.01649] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/27/2020] [Indexed: 01/10/2023] Open
Abstract
Background: The CheckMate 227 trial has indicated that nivolumab plus ipilimumab compared with chemotherapy significantly increases long-term survival in the first-line setting of advanced non-small-cell lung cancer (NSCLC). Methods: A Markov model was built to estimate the cost and effectiveness of nivolumab plus ipilimumab vs. chemotherapy as the first-line therapy in patients with advanced NSCLC based on outcomes data from the CheckMate 227 trial. We calculated the cost and health outcomes at a willingness-to-pay (WTP) threshold of $150,000 per quality adjusted life year (QALY) in populations with different programmed death ligand 1 (PD-L1) expression levels (≥50, ≥1, and <1%) or a high tumor mutational burden (TMB) (≥10 mutations per megabase). Sensitivity analysis were used to test the model stability. Results: The outcomes showed that the incremental costs and QALYs by using nivolumab plus ipilimumab were $124180.76 and 1.16, $70951.42 and 0.53, $144093.63 and 0.83 for the advanced NSCLC patients with a PD-L1 expression ≥50%, ≥1%, and <1%, which led to an incremental cost-effective ratio (ICER) of $107403.72, $133732.20, and $172589.15 per QALY, respectively. For patients with a high TMB, nivolumab plus ipilimumab contributed an extra 2.04 QALYs at a cost of $69182.50 per QALY. Conclusion: Nivolumab plus ipilimumab as first-line therapy makes a better cost-effective strategy than chemotherapy in advanced NSCLC patients with PD-L1 expression levels ≥50% and ≥1% or a high TMB, at a willingness-to-pay threshold of $150,000 per QALY, but not in the patients with a PD-L1 expression <1%.
Collapse
Affiliation(s)
- Huabin Hu
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Longjiang She
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Mengting Liao
- Xiangya Hospital, Central South University, Changsha, China
| | - Yin Shi
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Linli Yao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Dong Ding
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Youwen Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - David P Carbone
- Barbara J. Bonner Chair in Lung Cancer Research, James Thoracic Center, James Cancer Center, The Ohio State University Medical Center, Columbus, OH, United States
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
345
|
Alborelli I, Bratic Hench I, Chijioke O, Prince SS, Bubendorf L, Leuenberger LP, Tolnay M, Leonards K, Quagliata L, Jermann P, Matter MS. Robust assessment of tumor mutational burden in cytological specimens from lung cancer patients. Lung Cancer 2020; 149:84-89. [PMID: 32980613 DOI: 10.1016/j.lungcan.2020.08.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/23/2020] [Accepted: 08/31/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Tumor mutational burden (TMB) has emerged as a promising predictive biomarker for immune checkpoint inhibitor therapy. While the feasibility of TMB analysis on formalin-fixed paraffin-embedded (FFPE) samples has been thoroughly evaluated, only limited analyses have been performed on cytological samples, and no dedicated study has investigated concordance of TMB between different sample types. Here, we assessed TMB on matched histological and cytological samples from lung cancer patients and evaluated the accuracy of TMB estimation in these sample types. MATERIALS AND METHODS We analyzed mutations and resulting TMB in FFPE samples and matched ethanol-fixed cytological smears (n = 12 matched pairs) by using a targeted next-generation sequencing assay (Oncomine™ Tumor Mutational Load). Two different variant allele frequency (VAF) thresholds were used to estimate TMB (VAF = 5% or 10%). RESULTS At 5% VAF threshold, 73% (107/147) of mutations were concordantly detected in matched histological and cytological samples. Discordant variants were mainly unique to FFPE samples (34/40 discordant variants) and mostly C:G > T:A transitions with low allelic frequency, likely indicating formalin fixation artifacts. Increasing the VAF threshold to 10% clearly increased the number of concordantly detected mutations in matched histological and cytological samples to 96% (100/106 mutations), and drastically reduced the number of FFPE-only mutations (from 34 to 4 mutations). In contrast, cytological samples showed consistent mutation count and TMB values at both VAF thresholds. Using FFPE samples, 2 out of 12 patients were classified as TMB-high at VAF cutoff of 5% but TMB-low at 10%, whereas cytological specimens allowed consistent patient classification independently from VAF cutoff. CONCLUSION Our results show that cytological smears provide more consistent TMB values due to high DNA quality and lack of formalin-fixation induced artifacts. Therefore, cytological samples should be the preferred sample type for robust TMB estimation.
Collapse
Affiliation(s)
- Ilaria Alborelli
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland.
| | - Ivana Bratic Hench
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Obinna Chijioke
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Spasenija Savic Prince
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Lukas Bubendorf
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Laura P Leuenberger
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Markus Tolnay
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Katharina Leonards
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | | | - Philip Jermann
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Matthias S Matter
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| |
Collapse
|
346
|
Ferrara R, Naigeon M, Auclin E, Duchemann B, Cassard L, Jouniaux JM, Boselli L, Grivel J, Desnoyer A, Mezquita L, Texier M, Caramella C, Hendriks L, Planchard D, Remon J, Sangaletti S, Proto C, Garassino MC, Soria JC, Marabelle A, Voisin AL, Farhane S, Besse B, Chaput N. Circulating T-cell Immunosenescence in Patients with Advanced Non-small Cell Lung Cancer Treated with Single-agent PD-1/PD-L1 Inhibitors or Platinum-based Chemotherapy. Clin Cancer Res 2020; 27:492-503. [PMID: 32887723 DOI: 10.1158/1078-0432.ccr-20-1420] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/15/2020] [Accepted: 08/31/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE CD28, CD57, and KLRG1 have been previously identified as markers of T-cell immunosenescence. The impact of immunosenescence on anti-PD(L)-1 (ICI) or platinum-based chemotherapy (PCT) in patients with advanced non-small cell lung cancer (aNSCLC) is unknown. EXPERIMENTAL DESIGN The percentage of CD28-, CD57+, KLRG1+ among CD8+ T cells [senescent immune phenotype (SIP)] was assessed by flow cytometry on blood from patients with aNSCLC before single-agent ICI (discovery cohort). A SIP cut-off was identified by log-rank maximization method and patients with aNSCLC treated with ICI (validation cohort) or PCT were classified accordingly. Proliferation and functional properties of SIP+ CD8+ T cells were assessed in vitro. RESULTS In the ICI discovery cohort (N = 37), SIP cut-off was 39.5%, 27% of patients were SIP+. In the ICI validation cohort (N = 46), SIP+ status was found in 28% of patients and significantly correlated with worse objective response rate (ORR; 0% vs. 30%, P = 0.04), median progression-free survival (PFS) [1.8 (95% confidence interval (CI), 1.3-NR) vs. 6.4 (95% CI, 2-19) months, P = 0.009] and median overall survival, OS [2.8 (95% CI, 2.0-NR) vs. 20.8 (95% CI, 6.0-NR) months, P = 0.02]. SIP+ status was significantly associated with circulating specific immunephenotypes, in vitro lower CD8+ T cells proliferation, lower IL2 and higher TNFα and IFNγ production. In the ICI-pooled population (N = 83), SIP+ status did not correlate with any clinical characteristics and it was associated with significantly worse ORR, PFS, and OS. In PCT cohort (N = 61), 11% of patients were SIP+. SIP status did not correlate with outcomes upon PCT. CONCLUSIONS Circulating T-cell immunosenescence is observed in up to 28% of patients with aNSCLC and correlates with lack of benefit from ICI but not from PCT.See related commentary by Salas-Benito et al., p. 374.
Collapse
Affiliation(s)
- Roberto Ferrara
- Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Villejuif, France.,Department of Cancer Medicine, Gustave Roussy, Villejuif, France.,Department of Medical Oncology, Thoracic Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.,Department of Research, Molecular Immunology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Marie Naigeon
- Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Villejuif, France.,Faculty of Medicine, University Paris-Saclay, Orsay, France
| | - Edouard Auclin
- Department of Hepato-Gastroenterology and Gastrointestinal Oncology, Sorbonne Paris-Cité, Paris Descartes University, Hôpital Européen Georges Pompidou, Paris, France
| | - Boris Duchemann
- Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Villejuif, France
| | - Lydie Cassard
- Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Villejuif, France
| | - Jean-Mehdi Jouniaux
- Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Villejuif, France
| | - Lisa Boselli
- Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Villejuif, France
| | - Jonathan Grivel
- Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Villejuif, France
| | - Aude Desnoyer
- Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Villejuif, France
| | - Laura Mezquita
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Matthieu Texier
- Biostatistics and Epidemiology Department, Gustave Roussy, Villejuif, France
| | | | - Lizza Hendriks
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France.,Department of Pulmonary Diseases GROW - School for oncology and developmental biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - David Planchard
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Jordi Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Sabina Sangaletti
- Department of Research, Molecular Immunology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Claudia Proto
- Department of Medical Oncology, Thoracic Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Marina C Garassino
- Department of Medical Oncology, Thoracic Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | | | - Aurelien Marabelle
- Departement d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Anne-Laure Voisin
- Gustave Roussy, Université Paris-Saclay, Unité de Pharmacovigilance, Villejuif, France
| | - Siham Farhane
- Gustave Roussy, Université Paris-Saclay, Unité de Pharmacovigilance, Villejuif, France
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France. .,Faculty of Medicine, University Paris-Saclay, Orsay, France
| | - Nathalie Chaput
- Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Villejuif, France. .,Faculté de Pharmacie, University Paris-Saclay, Chatenay-Malabry, France
| |
Collapse
|
347
|
Burdett N, Desai J. New biomarkers for checkpoint inhibitor therapy. ESMO Open 2020; 5:e000597. [PMID: 32933940 PMCID: PMC7493090 DOI: 10.1136/esmoopen-2019-000597] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/20/2019] [Accepted: 11/27/2019] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitor blockade has vastly changed treatment paradigms and improved outcomes of many solid organ malignancies. The achievements of the last decade have transformed the outcomes of several tumour types, most notably metastatic melanoma. There are, however, still large numbers of patients who receive checkpoint inhibitor therapy and do not respond. In addition to potential lack of efficacy, checkpoint inhibitors also come with a unique and sometimes devastating side-effect profile. There exists a strong need for biomarkers to accurately predict response, improve treatment selection and avoid exposing patients to toxicity where there is minimal likelihood of response. There is a wide range of methodologies investigating predictive biomarkers in this space; in this review, we address the major putative biomarkers of interest. These include conventional serum tests such as lymphocyte indices and lactate dehydrogenase, and more novel research markers such as interleukin-6 and T receptor clonality. We discuss tumorous factors that may be of interest in certain tumour types, and finally gene expression profiling. Significant research continues into many of these potential predictive biomarkers in response to the emergent need to better select patients who will benefit from treatment.
Collapse
Affiliation(s)
- Nikki Burdett
- Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jayesh Desai
- Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
348
|
Inoue Y, Yoshimura K, Nishimoto K, Inui N, Karayama M, Yasui H, Hozumi H, Suzuki Y, Furuhashi K, Fujisawa T, Enomoto N, Nakamura Y, Asada K, Uto T, Fujii M, Matsui T, Matsuura S, Hashimoto D, Toyoshima M, Kusagaya H, Matsuda H, Inami N, Kaida Y, Niwa M, Ito Y, Sugimura H, Suda T. Evaluation of Programmed Death Ligand 1 (PD-L1) Gene Amplification and Response to Nivolumab Monotherapy in Non-small Cell Lung Cancer. JAMA Netw Open 2020; 3:e2011818. [PMID: 32955570 PMCID: PMC7506518 DOI: 10.1001/jamanetworkopen.2020.11818] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IMPORTANCE Robust predictors for response to anti-programmed death 1 and its ligand (PD-1/PD-L1) immunotherapy in non-small cell lung cancer (NSCLC) are not fully characterized. OBJECTIVE To evaluate whether PD-L1 (CD274) copy number gains (CNGs), comprising amplification and polysomy, in pretreatment specimens assessed by fluorescence in situ hybridization are associated with response to nivolumab monotherapy in NSCLC. DESIGN, SETTING, AND PARTICIPANTS This multicenter cohort study enrolled 200 patients, of whom 194 had assessable tumors, with advanced or recurrent NSCLC who were treated with nivolumab after progression following prior treatment at 14 institutions in Japan between July 2016 and December 2018. Median (interquartile range) duration of follow-up was 12.6 (5.6-20.4) months. Data were analyzed from December 2019 to February 2020. EXPOSURES Sequential nivolumab was given on day 1 of a 14-day cycle. Response was assessed every 4 cycles using Response Evaluation Criteria in Solid Tumors version 1.1. MAIN OUTCOMES AND MEASURES Overall response rate (ORR) according to the PD-L1 copy number status. Additional end points were progression-free survival, overall survival, and PD-L1 tumor proportion score (TPS) assessed by immunohistochemistry based on PD-L1 copy number status. RESULTS A total of 6 of the 200 patients were excluded because of poor-quality tumor specimens for the biomarker study, resulting in 194 assessable patients. Of these, 155 (79.9%) were men, with a median (range) age of 69 (43-83) years. PD-L1 CNGs were identified in 32 patients (16.5%), including 5 (2.6%) with amplification and 27 (13.9%) with polysomy. The ORR among patients with and without PD-L1 CNGs was 28.1% (95% CI, 13.7%-46.7%) and 17.9% (95% CI, 12.3%-24.7%), respectively. Although patients with PD-L1 polysomy did not demonstrate improved ORR (18.5% [95% CI, 6.3%-38.1%]) compared with those without PD-L1 CNGs, 4 of 5 patients (80.0% [95% CI, 28.4%-99.5%]) with PD-L1 amplification showed response, among whom median duration of response was not reached. Patients with PD-L1 amplification showed excellent survival outcomes for progression-free and overall survival. Overall, 3 PD-L1-amplified tumors (60.0%) showed PD-L1 TPS of at least 80%, but 2 (40.0%) had PD-L1 TPS of 15% or less. CONCLUSIONS AND RELEVANCE In this study, tumor PD-L1 amplification but not polysomy was associated with response to nivolumab monotherapy among patients with NSCLC. External validation with a larger sample size is warranted.
Collapse
Affiliation(s)
- Yusuke Inoue
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Katsuhiro Yoshimura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Koji Nishimoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Clinical Oncology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideki Yasui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuhiro Asada
- Department of Respiratory Medicine, Shizuoka General Hospital, Shizuoka, Japan
| | - Tomohiro Uto
- Department of Respiratory Medicine, Iwata City Hospital, Iwata, Japan
| | - Masato Fujii
- Department of Respiratory Medicine, Shizuoka City Shizuoka Hospital, Shizuoka, Japan
| | - Takashi Matsui
- Department of Respiratory Medicine, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Shun Matsuura
- Department of Respiratory Medicine, Fujieda Municipal General Hospital, Fujieda, Japan
| | - Dai Hashimoto
- Department of Pulmonary Medicine, Seirei Hamamatsu General Hospital, Hamamatsu, Japan
| | - Mikio Toyoshima
- Department of Respiratory Medicine, Hamamatsu Rosai Hospital, Hamamatsu, Japan
| | - Hideki Kusagaya
- Department of Respiratory Medicine, Shizuoka Saiseikai General Hospital, Shizuoka, Japan
| | - Hiroyuki Matsuda
- Department of Respiratory Medicine, Japanese Red Cross Shizuoka Hospital, Shizuoka, Japan
| | - Nao Inami
- Department of Respiratory Medicine, Shizuoka City Shimizu Hospital, Shizuoka, Japan
| | - Yusuke Kaida
- Department of Respiratory Medicine, Ensyu Hospital, Hamamatsu, Japan
| | - Mitsuru Niwa
- Department of Respiratory Medicine, Hamamatsu Medical Center, Hamamatsu, Japan
| | - Yasuhiro Ito
- Department of Respiratory Medicine, Tenryu Hospital, National Hospital Organization, Hamamatsu, Japan
| | - Haruhiko Sugimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
349
|
Jia XH, xu H, Geng LY, Jiao M, Wang WJ, Jiang LL, Guo H. Efficacy and safety of neoadjuvant immunotherapy in resectable nonsmall cell lung cancer: A meta-analysis. Lung Cancer 2020; 147:143-153. [DOI: 10.1016/j.lungcan.2020.07.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/22/2020] [Accepted: 07/01/2020] [Indexed: 01/17/2023]
|
350
|
Li R, Han D, Shi J, Han Y, Tan P, Zhang R, Li J. Choosing tumor mutational burden wisely for immunotherapy: A hard road to explore. Biochim Biophys Acta Rev Cancer 2020; 1874:188420. [PMID: 32828886 DOI: 10.1016/j.bbcan.2020.188420] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 12/28/2022]
Abstract
Immunotherapy has revolutionized the treatment of cancer due to its remarkable efficacy and extensive survival benefit in multiple tumor types. However, predictive biomarkers are required to identify patients who are likely to respond to immunotherapy. Recently, tumor mutational burden (TMB) has been shown to be associated with clinical outcomes in diverse cancers, such as melanoma, non-small-cell lung cancer and colorectal cancer. Several studies have demonstrated that high TMB can effectively predict the objective response rate and progression-free survival, but the ability of TMB to predict overall survival is limited. Thus, the clinical utility of TMB as a predictive and prognostic biomarker in immunotherapy is currently controversial. Importantly, multiple factors can affect the accurate assessment of TMB and further interfere with its prediction of clinical outcomes. These factors include preanalytical factors such as sample status, analytical factors such as differences in platforms and methods for determining TMB and variability of cutoff values, and postanalytical factors such as inconsistent interpretation and reporting of results. In addition, the optimal definition and quantification of TMB are unclear and require harmonization and standardization for reliable clinical application. This review elaborates on the factors affecting TMB status in primary tumors, summarizes the clinical utility of TMB as a biomarker in immunotherapy, and evaluates the impact of each analysis stage on the accurate estimation of TMB, especially its quantification, aiming to facilitate TMB assessment in routine clinical settings.
Collapse
Affiliation(s)
- Rui Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, PR China
| | - Dongsheng Han
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, PR China
| | - Jiping Shi
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Peking University Fifth School of Clinical Medicine, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, PR China
| | - YanXi Han
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, PR China
| | - Ping Tan
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, PR China
| | - Rui Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, PR China.
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, PR China.
| |
Collapse
|