51
|
Guan Y, Ren M, Guo D, He Y. [Research Progress on Lung Cancer Screening]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:954-960. [PMID: 32819054 PMCID: PMC7679225 DOI: 10.3779/j.issn.1009-3419.2020.101.37] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
肺癌是世界上最常见的恶性肿瘤,其5年生存率仅为19.7%,严重威胁人类健康。肺癌筛查是降低肺癌死亡率的有效措施,已有的研究证明用低剂量螺旋计算机断层扫描(low-dose computed tomography, LDCT)进行筛查可降低20%的肺癌死亡,目前国际和国内均建议进行肺癌筛查。研究肺癌筛查的发展现状有助于我们明确肺癌的高危人群,探索合理的筛查方案,提高筛查的成本效益,减轻经济负担。因此本文就肺癌筛查现状、肺癌筛查的成本效益以及存在的问题综述如下。
Collapse
Affiliation(s)
- Yazhe Guan
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Meng Ren
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Dongli Guo
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Yutong He
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| |
Collapse
|
52
|
Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab. ACTA ACUST UNITED AC 2020; 1:873-881. [DOI: 10.1038/s43018-020-0096-5] [Citation(s) in RCA: 295] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 06/26/2020] [Indexed: 12/22/2022]
|
53
|
Li J, Jiang W, Wei J, Zhang J, Cai L, Luo M, Wang Z, Sun W, Wang S, Wang C, Dai C, Liu J, Wang G, Wang J, Xu Q, Deng Y. Patient specific circulating tumor DNA fingerprints to monitor treatment response across multiple tumors. J Transl Med 2020; 18:293. [PMID: 32738923 PMCID: PMC7395971 DOI: 10.1186/s12967-020-02449-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/21/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) offers a convenient way to monitor tumor progression and treatment response. Because tumor mutational profiles are highly variable from person to person, a fixed content panel may be insufficient to track treatment response in all patients. METHODS We design ctDNA fingerprint panels specific to individual patients which are based on whole exome sequencing and target to high frequency clonal population clusters in patients. We test the fingerprint panels in 313 patients who together have eight tumor types (colorectal, hepatocellular, gastric, breast, pancreatic, and esophageal carcinomas and lung cancer and cholangiocarcinoma) and exposed to multiple treatment methods (surgery, chemotherapy, radiotherapy, targeted-drug therapy, immunotherapy, and combinations of them). We also monitor drug-related mutations in the patients using a pre-designed panel with eight hotspot genes. RESULTS 291 (93.0%) designed fingerprint panels harbor less than ten previously known tumor genes. We detected 7475 ctDNA mutations in 238 (76%) patients and 6196 (96.0%) of the mutations are detected in only one test. Both the level of ctDNA content fraction (CCF) and fold change of CCF (between the definitive and proceeding tests) are highly correlated with clinical outcomes (p-values 1.36e-6 for level and 5.64e-10 for fold change, Kruskal-Wallis test). The CCFs of PD patients are an order of magnitude higher than the CCFs of SD and OR patients (median/mean 2.22%/8.96% for SD, 0.18/0.21% for PD, and 0.31/0.54% for OR; pairwise p-values 7.8e-6 for SD ~ PD, 2.7e-4 for OR ~ PD, and 7.0e-3 for SD ~ OR, Wilcoxon rank sum test). The fold change of CCF distinguishes the patient groups even better, which increases for PD, remains stable for SD, and decreases for OR patients (p-values 0.002, ~ 1, and 0.0001 respectively, Wilcoxon signed-rank test). Eleven drug-related mutations are identified from nine out of the 313 patients. CONCLUSIONS The ctDNA fingerprint method improves both specificity and sensitivity of monitoring treatment response across several tumor types. It can identify tumor relapse/recurrence potentially earlier than imaging-based diagnosis. When augmented with tumor hotspot genes, it can track acquired drug-related mutations in patients.
Collapse
Affiliation(s)
- Jiaping Li
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Wei Jiang
- Department of Radiation Oncology, Huanhu Hospital, Tianjin, China
| | - Jinwang Wei
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Jianwei Zhang
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, No. 26 Erheng Road, Tianhe District, Guangzhou, 510655, China
| | - Linbo Cai
- Department of Oncology, Guangdong 999 Brain Hospital, Guangdong, China
| | - Minjie Luo
- Department of Pediatric Neurosurgery, Zhujiang Hospital of Southern Medical University, Guangdong, China
| | - Zhan Wang
- Department of Medical Oncology, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Wending Sun
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | | | - Chen Wang
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Chun Dai
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Jun Liu
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Guan Wang
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Jiping Wang
- Division of Surgical Oncology, Brigham and Women's Hospital, Boston, MA, USA
| | - Qiang Xu
- GenomiCare Biotechnology Co. Ltd, Shanghai, China.
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, No. 26 Erheng Road, Tianhe District, Guangzhou, 510655, China.
| |
Collapse
|
54
|
Esagian SM, Grigoriadou GΙ, Nikas IP, Boikou V, Sadow PM, Won JK, Economopoulos KP. Comparison of liquid-based to tissue-based biopsy analysis by targeted next generation sequencing in advanced non-small cell lung cancer: a comprehensive systematic review. J Cancer Res Clin Oncol 2020; 146:2051-2066. [PMID: 32462295 PMCID: PMC7456570 DOI: 10.1007/s00432-020-03267-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE To explore whether targeted next generation sequencing (NGS) of liquid biopsy in advanced non-small cell lung cancer (NSCLC) could potentially overcome the innate problems that arise with standard tissue biopsy, like intratumoral heterogeneity and the inability to obtain adequate samples for analysis. METHODS The Scopus, Cochrane Library, and MEDLINE (via PubMed) databases were searched for studies with matched tissue and liquid biopsies from advanced NSCLC patients, analyzed with targeted NGS. The number of mutations detected in tissue biopsy only, liquid biopsy only, or both was assessed and the positive percent agreement (PPA) of the two methods was calculated for every clinically relevant gene. RESULTS A total of 644 unique relevant articles were retrieved and data were extracted from 38 studies fulfilling the inclusion criteria. The sample size was composed of 2000 mutations tested in matched tissue and liquid biopsies derived from 1141 patients. No studies analyzed circulating tumor cells. The calculated PPA rates were 53.6% (45/84) for ALK, 53.9% (14/26) for BRAF, 56.5% (13/23) for ERBB2, 67.8% (428/631) for EGFR, 64.2% (122/190) for KRAS, 58.6% (17/29) for MET, 54.6% (12/22) for RET, and 53.3% (8/15) for ROS1. We additionally recorded data for 65 genes that are not recommended by current guidelines for mutational testing. An extra category containing results of unspecified genes was added, with a PPA rate of 55.7% (122/219). CONCLUSION Despite many advantages, liquid biopsy might be unable to fully substitute its tissue counterpart in detecting clinically relevant mutations in advanced NSCLC patients. However, it may serve as a helpful tool when making therapeutic decisions. More studies are needed to evaluate its role in everyday clinical practice.
Collapse
Affiliation(s)
- Stepan M Esagian
- Oncology Working Group, Society of Junior Doctors, Athens, Greece
| | - Georgia Ι Grigoriadou
- Oncology Working Group, Society of Junior Doctors, Athens, Greece
- 1st Department of Medical Oncology, Theageneio Anticancer Hospital, Thessaloníki, Greece
| | - Ilias P Nikas
- School of Medicine, European University of Cyprus, Nicosia, Cyprus
| | - Vasileios Boikou
- Oncology Working Group, Society of Junior Doctors, Athens, Greece
- Athens University of Economics and Business, Athens, Greece
| | - Peter M Sadow
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Konstantinos P Economopoulos
- Oncology Working Group, Society of Junior Doctors, Athens, Greece.
- Department of Surgery, Duke University Medical Center, 2301 Erwin Rd, Durham, NC, 27710, USA.
| |
Collapse
|
55
|
Jia Q, Chiu L, Wu S, Bai J, Peng L, Zheng L, Zang R, Li X, Yuan B, Gao Y, Wu D, Li X, Wu L, Sun J, He J, Robinson BWS, Zhu B. Tracking Neoantigens by Personalized Circulating Tumor DNA Sequencing during Checkpoint Blockade Immunotherapy in Non-Small Cell Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903410. [PMID: 32382482 PMCID: PMC7201246 DOI: 10.1002/advs.201903410] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/24/2020] [Accepted: 03/04/2020] [Indexed: 05/10/2023]
Abstract
The evolutionary dynamics of tumor-associated neoantigens carry information about drug sensitivity and resistance to the immune checkpoint blockade (ICB). However, the spectrum of somatic mutations is highly heterogeneous among patients, making it difficult to track neoantigens by circulating tumor DNA (ctDNA) sequencing using "one size fits all" commercial gene panels. Thus, individually customized panels (ICPs) are needed to track neoantigen evolution comprehensively during ICB treatment. Dominant neoantigens are predicted from whole exome sequencing data for treatment-naïve tumor tissues. Panels targeting predicted neoantigens are used for personalized ctDNA sequencing. Analyzing ten patients with non-small cell lung cancer, ICPs are effective for tracking most predicted dominant neoantigens (80-100%) in serial peripheral blood samples, and to detect substantially more genes (18-30) than the capacity of current commercial gene panels. A more than 50% decrease in ctDNA concentration after eight weeks of ICB administration is associated with favorable progression-free survival. Furthermore, at the individual level, the magnitude of the early ctDNA response is correlated with the subsequent change in tumor burden. The application of ICP-based ctDNA sequencing is expected to improve the understanding of ICB-driven tumor evolution and to provide personalized management strategies that optimize the clinical benefits of immunotherapies.
Collapse
Affiliation(s)
- Qingzhu Jia
- Institute of CancerXinqiao HospitalThe Army Medical UniversityXinqiao Main StreetChongqing400037China
- Chongqing Key Laboratory of ImmunotherapyXinqiao Main StreetChongqing400037China
| | - Luting Chiu
- Berry Oncology CorporationNO. 4 Science Park Road, Changping DistrictBeijing102206China
| | - Shuangxiu Wu
- Berry Oncology CorporationNO. 4 Science Park Road, Changping DistrictBeijing102206China
| | - Jian Bai
- Berry Oncology CorporationNO. 4 Science Park Road, Changping DistrictBeijing102206China
| | - Lina Peng
- Institute of CancerXinqiao HospitalThe Army Medical UniversityXinqiao Main StreetChongqing400037China
- Chongqing Key Laboratory of ImmunotherapyXinqiao Main StreetChongqing400037China
| | - Linpeng Zheng
- Institute of CancerXinqiao HospitalThe Army Medical UniversityXinqiao Main StreetChongqing400037China
- Chongqing Key Laboratory of ImmunotherapyXinqiao Main StreetChongqing400037China
| | - Rui Zang
- Institute of CancerXinqiao HospitalThe Army Medical UniversityXinqiao Main StreetChongqing400037China
| | - Xueqin Li
- Institute of CancerXinqiao HospitalThe Army Medical UniversityXinqiao Main StreetChongqing400037China
| | - Bibo Yuan
- Institute of CancerXinqiao HospitalThe Army Medical UniversityXinqiao Main StreetChongqing400037China
| | - Yixing Gao
- Institute of CancerXinqiao HospitalThe Army Medical UniversityXinqiao Main StreetChongqing400037China
- Chongqing Key Laboratory of ImmunotherapyXinqiao Main StreetChongqing400037China
| | - Dingyong Wu
- Institute of CancerXinqiao HospitalThe Army Medical UniversityXinqiao Main StreetChongqing400037China
| | - Xiaohong Li
- Institute of CancerXinqiao HospitalThe Army Medical UniversityXinqiao Main StreetChongqing400037China
| | - Lin Wu
- Berry Oncology CorporationNO. 4 Science Park Road, Changping DistrictBeijing102206China
| | - Jianguo Sun
- Institute of CancerXinqiao HospitalThe Army Medical UniversityXinqiao Main StreetChongqing400037China
- Chongqing Key Laboratory of ImmunotherapyXinqiao Main StreetChongqing400037China
| | - Ji He
- GeneCast Biotechnology Co., Ltd.35 Huayuan North Road, HealthWork Suite 901Beijing100191China
| | - Bruce W. S. Robinson
- School of Medicine and PharmacologyUniversity of Western Australia35 Stirling HighwayPerthWestern Australia6009Australia
- National Centre for Asbestos Related DiseasesGPO Box U1908PerthWestern Australia6845Australia
| | - Bo Zhu
- Institute of CancerXinqiao HospitalThe Army Medical UniversityXinqiao Main StreetChongqing400037China
- Chongqing Key Laboratory of ImmunotherapyXinqiao Main StreetChongqing400037China
| |
Collapse
|
56
|
de Vos L, Jung M, Koerber RM, Bawden EG, Holderried TAW, Dietrich J, Bootz F, Brossart P, Kristiansen G, Dietrich D. Treatment Response Monitoring in Patients with Advanced Malignancies Using Cell-Free SHOX2 and SEPT9 DNA Methylation in Blood: An Observational Prospective Study. J Mol Diagn 2020; 22:920-933. [PMID: 32361006 DOI: 10.1016/j.jmoldx.2020.04.205] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 11/14/2019] [Accepted: 04/04/2020] [Indexed: 12/30/2022] Open
Abstract
Patients with incurable cancer usually receive palliative treatment with significant toxicity and limited efficacy. Methylation analysis of circulating cell-free DNA (ccfDNA) in blood from cancer patients represents a promising approach for minimally invasive, real-time monitoring of treatment response. Short stature homeobox 2 (SHOX2) and septin 9 (SEPT9) methylation was analyzed in N = 8865 malignant and N = 746 normal adjacent tissues across 33 different malignancies from The Cancer Genome Atlas. Furthermore, we performed quantitative SHOX2 and SEPT9 ccfDNA methylation analysis in plasma obtained before and consecutively during treatment from prospectively enrolled N = 115 patients with various advanced cancers. SHOX2 and/or SEPT9 hypermethylation in malignant tissues is present in various carcinomas, sarcoma, melanoma, brain tumors, mesothelioma, and hematopoietic malignancies. Among the prospectively enrolled cancer patients, 61% (70/115) of patients had a baseline-positive blood cumulative ccfDNA methylation score (CMS) and were eligible for response monitoring. Dynamic changes of CMS during treatment were strongly associated with treatment response. A CMS increase indicated response up to 80 days before conventional monitoring. SHOX2 and SEPT9 ccfDNA methylation represents a pan-cancer biomarker and has the potential to be a powerful tool for monitoring treatment response in patients with solid tumors and lymphomas. The early identification of nonresponders might allow for a timely change of treatment regimen.
Collapse
Affiliation(s)
- Luka de Vos
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Maria Jung
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Ruth-Miriam Koerber
- Department of Oncology, Hematology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Emma G Bawden
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Tobias A W Holderried
- Department of Oncology, Hematology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Jörn Dietrich
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Friedrich Bootz
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Peter Brossart
- Department of Oncology, Hematology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | | | - Dimo Dietrich
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
57
|
Rossi G, Russo A, Tagliamento M, Tuzi A, Nigro O, Vallome G, Sini C, Grassi M, Dal Bello MG, Coco S, Longo L, Zullo L, Tanda ET, Dellepiane C, Pronzato P, Genova C. Precision Medicine for NSCLC in the Era of Immunotherapy: New Biomarkers to Select the Most Suitable Treatment or the Most Suitable Patient. Cancers (Basel) 2020; 12:E1125. [PMID: 32365882 PMCID: PMC7281184 DOI: 10.3390/cancers12051125] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
In recent years, the evolution of treatments has made it possible to significantly improve the outcomes of patients with non-small cell lung cancer (NSCLC). In particular, while molecular targeted therapies are effective in specific patient sub-groups, immune checkpoint inhibitors (ICIs) have greatly influenced the outcomes of a large proportion of NSCLC patients. While nivolumab activity was initially assessed irrespective of predictive biomarkers, subsequent pivotal studies involving other PD-1/PD-L1 inhibitors in pre-treated advanced NSCLC (atezolizumab within the OAK study and pembrolizumab in the Keynote 010 study) reported the first correlations between clinical outcomes and PD-L1 expression. However, PD-L1 could not be sufficient on its own to select patients who may benefit from immunotherapy. Many studies have tried to discover more precise markers that are derived from tumor tissue or from peripheral blood. This review aims to analyze any characteristics of the immunogram that could be used as a predictive biomarker for response to ICIs. Furthermore, we describe the most important genetic alteration that might predict the activity of immunotherapy.
Collapse
Affiliation(s)
- Giovanni Rossi
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| | | | - Marco Tagliamento
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Alessandro Tuzi
- UO Oncologia, ASST Sette Laghi, 21100 Varese, Italy; (A.T.); (O.N.)
| | - Olga Nigro
- UO Oncologia, ASST Sette Laghi, 21100 Varese, Italy; (A.T.); (O.N.)
| | - Giacomo Vallome
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Claudio Sini
- Oncologia Medica e CPDO, ASSL di Olbia-ATS Sardegna, 07026 Olbia, Italy;
| | - Massimiliano Grassi
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Maria Giovanna Dal Bello
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Simona Coco
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Luca Longo
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Lodovica Zullo
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Enrica Teresa Tanda
- Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Chiara Dellepiane
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Paolo Pronzato
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Carlo Genova
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| |
Collapse
|
58
|
Chang S, Hur JY, Choi YL, Lee CH, Kim WS. Current status and future perspectives of liquid biopsy in non-small cell lung cancer. J Pathol Transl Med 2020; 54:204-212. [PMID: 32460474 PMCID: PMC7253954 DOI: 10.4132/jptm.2020.02.27] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/27/2020] [Indexed: 12/17/2022] Open
Abstract
With advances in target therapy, molecular analysis of tumors is routinely required for treatment decisions in patients with advanced non-small cell lung cancer (NSCLC). Liquid biopsy refers to the sampling and analysis of circulating cell-free tumor DNA (ctDNA) in various body fluids, primarily blood. Because the technique is minimally invasive, liquid biopsies are the future in cancer management. Epidermal growth factor receptor (EGFR) ctDNA tests have been performed in routine clinical practice in advanced NSCLC patients to guide tyrosine kinase inhibitor treatment. In the near future, liquid biopsy will be a crucial prognostic, predictive, and diagnostic method in NSCLC. Here we present the current status and future perspectives of liquid biopsy in NSCLC.
Collapse
Affiliation(s)
- Sunhee Chang
- Department of Pathology, Inje University Ilsan Paik Hospital, Goyang, Korea
| | - Jae Young Hur
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chang Hun Lee
- Department of Pathology, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea
| | - Wan Seop Kim
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
59
|
Filipovic A, Miller G, Bolen J. Progress Toward Identifying Exact Proxies for Predicting Response to Immunotherapies. Front Cell Dev Biol 2020; 8:155. [PMID: 32258034 PMCID: PMC7092703 DOI: 10.3389/fcell.2020.00155] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Clinical value and utility of checkpoint inhibitors, a drug class targeting adaptive immune suppression pathways (PD-1, PDL-1, and CTLA-4), is growing rapidly and maintains status of a landmark achievement in oncology. Their efficacy has transformed life expectancy in multiple deadly cancer types (melanoma, lung cancer, renal/urothelial carcinoma, certain colorectal cancers, lymphomas, etc.). Despite significant clinical development efforts, therapeutic indication of approved checkpoint inhibitors are not as wide as the oncology community and patients would like them to be, potentially bringing into question their universal efficacy across tumor histologies. With the main goal of expanding immunotherapy applications, identifying of biomarkers to accurately predict therapeutic response and treatment related side-effects are a paramount need in the field. Specificities surrounding checkpoint inhibitors in clinic, such as unexpected tumor response patterns (pseudo- and hyper-progression), late responders, as well as specific immune mediated toxicities, complicate the management of patients. They stem from the complexities and dynamics of the tumor/host immune interactions, as well as baseline tumor biology. Search for clinically effective biomarkers therefore calls for a holistic approach, rather than implementation of a single analyte. The goal is to achieve dynamic and comprehensive acquisition, analyses and interpretation of immunological and biologic information about the tumor and the immune system, and to compute these parameters into an actionable, maximally predictive value at the individual patient level. Limitation delaying swift incorporation of validated immuno-oncology biomarkers span from standardized biospecimens acquisition and processing, selection of proficient biomarker discovery and validation methods, to establishing multidisciplinary consortiums and data sharing platforms. Multi-disciplinary efforts have already yielded some approved (PDL-1 and MSI-status) and other advanced tests (TMB, neoantigen pattern, and TIL infiltration rate). Importantly, clinical trial taskforces now recognize the imperative of the biomarker-driven trial design and execution, to enable translating biomarker discoveries into the clinical setting. This will ensure we utilize the “conspiracy” between the peripheral and intra-tumoral dynamic markers in shaping responses to checkpoint blockade, for the ultimate patient benefit.
Collapse
Affiliation(s)
| | - George Miller
- New York University School of Medicine, New York, NY, United States
| | | |
Collapse
|
60
|
Kiriu T, Yamamoto M, Nagano T, Hazama D, Sekiya R, Katsurada M, Katsurada N, Tachihara M, Kobayashi K, Nishimura Y. Pseudo-Progression and the Neutrophil-to-Lymphocyte Ratio in Non-Small Cell Lung Cancer Treated with Immune Checkpoint Inhibitors: A Case-Control Study. Onco Targets Ther 2019; 12:10559-10568. [PMID: 31819535 PMCID: PMC6897378 DOI: 10.2147/ott.s228138] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/10/2019] [Indexed: 12/21/2022] Open
Abstract
Purpose Pseudo-progression (PsPD) is a rare phenomenon observed in <5% of cases of non-small cell lung cancer (NSCLC). This event is challenging for both clinicians and patients. Viable biomarkers to distinguish between PsPD and true progressive disease (TPD) are lacking. The aim of our study was to determine the correlation between PsPD and the neutrophil-to-lymphocyte ratio (NLR) in patients with NSCLC treated with immune checkpoint inhibitors (ICIs). Patients and methods We retrospectively reviewed the clinical records of NSCLC patients treated with ICI monotherapy from December 2015 to October 2018 at Kobe University Hospital, Japan. Twenty-five patients were determined to have either PsPD (n =4) or TPD (n =21). We focused on longitudinal radiological images and NLRs. Results Here, we report four patients with PsPD. The pre- and post-treatment NLRs were significantly lower in patients with PsPD than in patients with TPD (p = 0.019 and p = 0.007, respectively). The receiver operating characteristic curve according to the pre- and post-treatment NLR showed areas under the curve of 0.82 and 0.94, respectively. The optimal cut-off values for pre- and post-treatment NLR were 4.1 and 3.2, respectively. The pre- and post-treatment NLRs were useful in distinguishing between PsPD and TPD. Both a pre-treatment NLR <4.1 and a post-treatment NLR <3.2 were significantly associated with longer overall survival compared to a pre-treatment NLR ≥4.1 (p < 0.001) and post-treatment NLR ≥3.2 (p = 0.004), respectively. Conclusion The NLR could be a viable clue for distinguishing between PsPD and TPD. Patients with a high post-treatment NLR in this study all had TPD, suggesting that these subjects should be considered for an early transition to the next drug treatment regimen.
Collapse
Affiliation(s)
- Tatsunori Kiriu
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Masatsugu Yamamoto
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Daisuke Hazama
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Reina Sekiya
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Masahiro Katsurada
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Naoko Katsurada
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Motoko Tachihara
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Kazuyuki Kobayashi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| |
Collapse
|
61
|
Costantini A, Takam Kamga P, Dumenil C, Chinet T, Emile JF, Giroux Leprieur E. Plasma Biomarkers and Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer: New Tools for Better Patient Selection? Cancers (Basel) 2019; 11:cancers11091269. [PMID: 31470546 PMCID: PMC6769436 DOI: 10.3390/cancers11091269] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment landscape for patients with non-small cell lung cancer (NSCLC). Although some patients can experience important response rates and improved survival, many others do not benefit from ICIs developing hyper-progressive disease or immune-related adverse events. This underlines the need to select biomarkers for ICIs use in order to better select patients. There is currently no universally validated robust biomarker for daily use of ICIs. Programmed death-ligand 1 (PD-L1) or tumor mutational burden (TMB) are sometimes used but still have several limitations. Plasma biomarkers are a promising approach in ICI treatment. This review will describe the development of novel plasma biomarkers such as soluble proteins, circulating tumor DNA (ctDNA), blood TMB, and blood microbiome in NSCLC patients treated with ICIs and their potential use in predicting response and toxicity.
Collapse
Affiliation(s)
- Adrien Costantini
- Department of Respiratory Diseases and Thoracic Oncology, APHP-Hôpital Ambroise Paré, 92100 Boulogne-Billancourt, France
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France
| | - Paul Takam Kamga
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France
| | - Coraline Dumenil
- Department of Respiratory Diseases and Thoracic Oncology, APHP-Hôpital Ambroise Paré, 92100 Boulogne-Billancourt, France
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France
| | - Thierry Chinet
- Department of Respiratory Diseases and Thoracic Oncology, APHP-Hôpital Ambroise Paré, 92100 Boulogne-Billancourt, France
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France
| | - Jean-François Emile
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France
- Department of Pathology, APHP-Hôpital Ambroise Pare, 92100 Boulogne-Billancourt, France
| | - Etienne Giroux Leprieur
- Department of Respiratory Diseases and Thoracic Oncology, APHP-Hôpital Ambroise Paré, 92100 Boulogne-Billancourt, France.
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France.
| |
Collapse
|
62
|
Alama A, Coco S, Genova C, Rossi G, Fontana V, Tagliamento M, Giovanna Dal Bello M, Rosa A, Boccardo S, Rijavec E, Biello F, Longo L, Cavalieri Z, Bruzzo C, Grossi F. Prognostic Relevance of Circulating Tumor Cells and Circulating Cell-Free DNA Association in Metastatic Non-Small Cell Lung Cancer Treated with Nivolumab. J Clin Med 2019; 8:jcm8071011. [PMID: 31295929 PMCID: PMC6679117 DOI: 10.3390/jcm8071011] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/18/2022] Open
Abstract
The treatment of advanced non-small cell lung cancer (NSCLC) has been revolutionized by immune checkpoint inhibitors (ICIs). The identification of prognostic and predictive factors in ICIs-treated patients is presently challenging. Circulating tumor cells (CTCs) and cell-free DNA (cfDNA) were evaluated in 89 previously treated NSCLC patients receiving nivolumab. Blood samples were collected before therapy and at the first and second radiological response assessments. CTCs were isolated by a filtration-based method. cfDNA was extracted from plasma and estimated by quantitative PCR. Patients with baseline CTC number and cfDNA below their median values (2 and 836.5 ng from 3 mL of blood and plasma, respectively) survived significantly longer than those with higher values (p = 0.05 and p = 0.04, respectively). The two biomarkers were then used separately and jointly as time-dependent covariates in a regression model confirming their prognostic role. Additionally, a four-fold risk of death for the subgroup presenting both circulating biomarkers above the median values was observed (p < 0.001). No significant differences were found between circulating biomarkers and best response. However, progressing patients with concomitant lower CTCs and cfDNA performed clinically well (p = 0.007), suggesting that jointed CTCs and cfDNA might help discriminate a low-risk population which might benefit from continuing ICIs beyond progression.
Collapse
Affiliation(s)
- Angela Alama
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy.
| | - Simona Coco
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Carlo Genova
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Giovanni Rossi
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Vincenzo Fontana
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Marco Tagliamento
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Maria Giovanna Dal Bello
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Alessandra Rosa
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Simona Boccardo
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Erika Rijavec
- Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza, 28, 20122 Milan, Italy
| | - Federica Biello
- AOU Maggiore della Carità, Corso Mazzini 18, 28100 Novara, Italy
| | - Luca Longo
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Zita Cavalieri
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Cristina Bruzzo
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Francesco Grossi
- Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza, 28, 20122 Milan, Italy
| |
Collapse
|
63
|
Cabel L, Proudhon C, Romano E, Girard N, Lantz O, Stern MH, Pierga JY, Bidard FC. Clinical potential of circulating tumour DNA in patients receiving anticancer immunotherapy. Nat Rev Clin Oncol 2019; 15:639-650. [PMID: 30050094 DOI: 10.1038/s41571-018-0074-3] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Considerable interest surrounds the use of immune-checkpoint inhibitors in patients with solid tumours following the demonstration of the impressive clinical efficacy of anti-programmed cell death protein 1 and anti-programmed cell death 1 ligand 1 antibodies in several tumour types. However, the emergence of unexpected tumour response patterns, such as pseudoprogression or hyperprogression, might complicate the management of patients receiving these agents. Analysis of circulating tumour DNA (ctDNA) has been shown to have prognostic value by enabling the detection of residual proliferating disease in the adjuvant setting and estimation of tumour burden in the metastatic setting, which are key stratification biomarkers for use of immune-checkpoint inhibition (ICI). Furthermore, examinations of ctDNA for genetic predictors of responsiveness to immunotherapy, such as mutations, tumour mutational load, and microsatellite instability provide a noninvasive surrogate for tumour biopsy sampling. Proof-of-concept reports have also demonstrated that quantitative changes in ctDNA levels early in the course of disease are a promising tool for the assessment of responsiveness to ICI that might complement standard imaging approaches. Other applications of this technology are also currently under investigation, such as early detection of resistance to immunotherapy and characterization of mechanisms of resistance. The aim of this Review is to summarize available data on the application of ctDNA in patients receiving immunotherapy and to discuss the most promising future directions.
Collapse
Affiliation(s)
- Luc Cabel
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France.,Versailles Saint Quentin en Yvelines University, Paris Saclay University, Saint Cloud, Paris, France.,Circulating Tumor Biomarkers Laboratory, Institut Curie, PSL Research University, Paris, France
| | - Charlotte Proudhon
- Circulating Tumor Biomarkers Laboratory, Institut Curie, PSL Research University, Paris, France
| | - Emanuela Romano
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France.,INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Nicolas Girard
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France
| | - Olivier Lantz
- INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Marc-Henri Stern
- INSERM U830, Institut Curie, PSL Research University, Paris, France
| | - Jean-Yves Pierga
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France.,Paris Descartes University, Paris, France
| | - François-Clément Bidard
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France. .,Versailles Saint Quentin en Yvelines University, Paris Saclay University, Saint Cloud, Paris, France. .,Circulating Tumor Biomarkers Laboratory, Institut Curie, PSL Research University, Paris, France.
| |
Collapse
|
64
|
Filipska M, Pedraz-Valdunciel C, Chaib I, Rosell R. Biological therapies in lung cancer treatment: using our immune system as an ally to defeat the malignancy. Expert Opin Biol Ther 2019; 19:457-467. [PMID: 30763126 DOI: 10.1080/14712598.2019.1582635] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Biological therapies, with immunotherapy leading the field, have arisen as one of the quickest expanding areas of research for cancer treatment in the last few years. The clear benefits for patients are undeniable, satisfying the long-awaited necessity of a target-specific therapy. However, its full potential remains still unexploited due to a lack of response in a majority of patients and pending reliable biomarkers. AREAS COVERED This review provides a summarizing view of the current biological therapies for lung cancer, focusing on immunotherapy - including immune checkpoint inhibitors, adoptive cell therapy and vaccines available in clinical/pre-clinical settings or currently in development. A thorough analysis of the technical and functional differences among all therapies is provided, along with a critical discussion of prospective treatments and potential biomarkers. EXPERT OPINION The use of immunotherapy in the treatment of cancer has provided clear benefits for patients. Still, exploitation of the full potential of immune checkpoint inhibitors alone or in combination, or adoptive cell therapies is hampered by, amongst other reasons, the lack of reliable biomarkers and possible adverse immune effects. We postulate that the development of liquid biopsy-based diagnostics will help to overcome these limitations in the near future.
Collapse
Affiliation(s)
- Martyna Filipska
- a Cancer Biology and Precision Medicine , Institute for Health Science Research Germans Trias i Pujol (IGTP) , Badalona , Spain
| | - Carlos Pedraz-Valdunciel
- a Cancer Biology and Precision Medicine , Institute for Health Science Research Germans Trias i Pujol (IGTP) , Badalona , Spain
| | - Imane Chaib
- a Cancer Biology and Precision Medicine , Institute for Health Science Research Germans Trias i Pujol (IGTP) , Badalona , Spain
| | - Rafael Rosell
- a Cancer Biology and Precision Medicine , Institute for Health Science Research Germans Trias i Pujol (IGTP) , Badalona , Spain.,b Pangaea Oncology , Laboratory of Molecular Biology , Barcelona, Spain.,c Institute of Oncology Rosell (IOR) , Quiron-Dexeus University Institute , Barcelona , Spain.,d Catalan Institute of Oncology (ICO) , Hospital Germans Trias i Pujol , Badalona , Spain
| |
Collapse
|
65
|
Herbreteau G, Vallée A, Charpentier S, Normanno N, Hofman P, Denis MG. Circulating free tumor DNA in non-small cell lung cancer (NSCLC): clinical application and future perspectives. J Thorac Dis 2019; 11:S113-S126. [PMID: 30775034 DOI: 10.21037/jtd.2018.12.18] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Major advances in the treatment of non-small cell lung cancer (NSCLC) patients have been obtained during the last decade. Molecular testing of tumor samples is therefore mandatory in routine clinical practice. Tumor DNA is also present as cell-free molecules in blood, which is therefore a very useful and convenient source of tumor DNA. In this review, we discuss pre-analytical and analytical aspects of circulating tumor DNA (ctDNA) analysis. We also describe the use of ctDNA analysis in routine clinical practice, and discuss the potential use of ctDNA monitoring both to identify minimal residual disease and as a potential tool to early identify patients' response to treatment.
Collapse
Affiliation(s)
- Guillaume Herbreteau
- Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, F-44093 Nantes Cedex, France
| | - Audrey Vallée
- Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, F-44093 Nantes Cedex, France
| | - Sandrine Charpentier
- Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, F-44093 Nantes Cedex, France
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori, IRCCS, "Fondazione G. Pascale", Naples, Italy
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Inserm U1081/CNRS 7284, Université Côte d'Azur, CHU Nice and FHU OncoAge, and Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice, France
| | - Marc G Denis
- Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, F-44093 Nantes Cedex, France
| |
Collapse
|
66
|
Blons H, Garinet S, Laurent-Puig P, Oudart JB. Molecular markers and prediction of response to immunotherapy in non-small cell lung cancer, an update. J Thorac Dis 2019; 11:S25-S36. [PMID: 30775025 DOI: 10.21037/jtd.2018.12.48] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Immunotherapy represents one of the most promising therapeutic approaches in lung cancer, however 50% of lung cancer patients will not respond to this treatment, while others will have transitory or durable responses. Because side effects may be life threatening and treatment costs remain very high, the identification of predictive markers is mandatory and actually extensively studied. Factors that determine response to immune checkpoint inhibitors (ICI) are numerous including tumor microenvironment, immune tumor infiltrates, expression of immune checkpoint proteins (PD-1/PD-L1), gene expression signatures and molecular tumor profiles. Based on high impact factor publications and recent literature this review focuses on the potential predictive value of tumor molecular alterations and tumor mutation burden as predictive markers of response or resistance to ICI. We also discuss the role of circulating tumor DNA (ctDNA) to monitor ICI responses and propose an algorithm that integrates molecular markers upcoming recommendations for first line treatment.
Collapse
Affiliation(s)
- Hélène Blons
- INSERM UMR-S1147, Paris Sorbonne Cite University, Paris, France.,Department of Biochemistry, Unit of Pharmacogenetics and Molecular Oncology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Simon Garinet
- INSERM UMR-S1147, Paris Sorbonne Cite University, Paris, France.,Department of Biochemistry, Unit of Pharmacogenetics and Molecular Oncology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Pierre Laurent-Puig
- INSERM UMR-S1147, Paris Sorbonne Cite University, Paris, France.,Department of Biochemistry, Unit of Pharmacogenetics and Molecular Oncology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Baptiste Oudart
- Department of Biochemistry, Unit of Pharmacogenetics and Molecular Oncology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
67
|
Seijo LM, Peled N, Ajona D, Boeri M, Field JK, Sozzi G, Pio R, Zulueta JJ, Spira A, Massion PP, Mazzone PJ, Montuenga LM. Biomarkers in Lung Cancer Screening: Achievements, Promises, and Challenges. J Thorac Oncol 2018; 14:343-357. [PMID: 30529598 DOI: 10.1016/j.jtho.2018.11.023] [Citation(s) in RCA: 341] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/20/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
The present review is an update of the research and development efforts regarding the use of molecular biomarkers in the lung cancer screening setting. The two main unmet clinical needs, namely, the refinement of risk to improve the selection of individuals undergoing screening and the characterization of undetermined nodules found during the computed tomography-based screening process are the object of the biomarkers described in the present review. We first propose some principles to optimize lung cancer biomarker discovery projects. Then, we summarize the discovery and developmental status of currently promising molecular candidates, such as autoantibodies, complement fragments, microRNAs, circulating tumor DNA, DNA methylation, blood protein profiling, or RNA airway or nasal signatures. We also mention other emerging biomarkers or new technologies to follow, such as exhaled breath biomarkers, metabolomics, sputum cell imaging, genetic predisposition studies, and the integration of next-generation sequencing into study of circulating DNA. We also underline the importance of integrating different molecular technologies together with imaging, radiomics, and artificial intelligence. We list a number of completed, ongoing, or planned trials to show the clinical utility of molecular biomarkers. Finally, we comment on future research challenges in the field of biomarkers in the context of lung cancer screening and propose a design of a trial to test the clinical utility of one or several candidate biomarkers.
Collapse
Affiliation(s)
- Luis M Seijo
- Clinica Universidad de Navarra, Madrid, Spain; CIBERES, Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Madrid, Spain
| | - Nir Peled
- Oncology Division, The Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka Medical Center and Ben-Gurion University, Beer-Sheva, Israel
| | - Daniel Ajona
- Solid Tumors Program, Centro de Investigación Médica Aplicada, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Mattia Boeri
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - John K Field
- The Roy Castle Lung Cancer Research Programme, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Gabriella Sozzi
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ruben Pio
- Solid Tumors Program, Centro de Investigación Médica Aplicada, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Javier J Zulueta
- Department of Pulmonology, Clinica Universidad de Navarra, Pamplona, Spain; Visiongate Inc., Phoenix, Arizona
| | - Avrum Spira
- Boston University School of Medicine, Boston, Massachusetts
| | | | | | - Luis M Montuenga
- Solid Tumors Program, Centro de Investigación Médica Aplicada, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.
| |
Collapse
|
68
|
Costantini A, Julie C, Dumenil C, Hélias-Rodzewicz Z, Tisserand J, Dumoulin J, Giraud V, Labrune S, Chinet T, Emile JF, Giroux Leprieur E. Predictive role of plasmatic biomarkers in advanced non-small cell lung cancer treated by nivolumab. Oncoimmunology 2018; 7:e1452581. [PMID: 30221046 PMCID: PMC6136870 DOI: 10.1080/2162402x.2018.1452581] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/07/2018] [Accepted: 03/09/2018] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors, as nivolumab, are used in advanced non-small cell lung cancer (NSCLC). However, no associated biomarker is validated in clinical practice with this drug. We investigated herein immune-related blood markers in patients with advanced NSCLC treated with nivolumab. Plasma of 43 consecutive patients were prospectively collected at time of the diagnosis of cancer, at the initiation of nivolumab and at the first tumour evaluation (2 months). Concentrations of PD-L1 (sPD-L1), soluble PD-L2 (sPD-L2), Interleukine-2 (sIl-2), Interferon-gamma (sIFN-γ), and Granzyme B (sGranB) were quantified by ELISA. Cell free RNA was quantified by Reverse Transcriptase -PCR), and plasmatic microRNAs (miRNAs) were evaluated by targeted sequencing. Expression of PD-L1 on tumour biopsies was performed by immunohistochemistry using E13LN. High sPD-L1 at 2 months and increase of sPD-L1 concentrations were associated with poor response and absence of clinical benefit (nivolumab treatment less than 6 months). The variation of sPD-L1 concentrations were confirmed by RNA quantification. sPD-L1 concentrations were not correlated with PD-L1 expression on corresponding tumour samples. Low sGranB at nivolumab initiation was also associated with poor response. High sPD-L1 and low sGranB were associated with poor progression-free survival (PFS) and overall survival (OS). Low sPD-L2, low sIl-2 and high sIFN-γ were associated with grade 3-4 toxicities. Finally, miRNA screening showed that patients with clinical benefit (n = 9) had down-expression of miRNA-320b and -375 compared to patients with early progression at 2 months (n = 9). In conclusion, our results highlight the interest of circulating biomarkers in patients treated with nivolumab.
Collapse
Affiliation(s)
- Adrien Costantini
- EA4340, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Catherine Julie
- EA4340, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
- Department of Pathology, APHP – Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Coraline Dumenil
- EA4340, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Zofia Hélias-Rodzewicz
- EA4340, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
- Department of Pathology, APHP – Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Julie Tisserand
- EA4340, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
- Department of Pathology, APHP – Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Jennifer Dumoulin
- EA4340, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Violaine Giraud
- EA4340, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Sylvie Labrune
- EA4340, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Thierry Chinet
- EA4340, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Jean-François Emile
- EA4340, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
- Department of Pathology, APHP – Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Etienne Giroux Leprieur
- EA4340, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Ambroise Pare Hospital, Boulogne-Billancourt, France
| |
Collapse
|