51
|
Kim S, Lee JW, Park YS. The Application of Next-Generation Sequencing to Define Factors Related to Oral Cancer and Discover Novel Biomarkers. Life (Basel) 2020; 10:E228. [PMID: 33023080 PMCID: PMC7599837 DOI: 10.3390/life10100228] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
Despite the introduction of next-generation sequencing in the realm of DNA sequencing technology, it is not often used in the investigation of oral squamous cell carcinoma (OSCC). Oral cancer is one of the most frequently occurring malignancies in some parts of the world and has a high mortality rate. Patients with this malignancy are likely to have a poor prognosis and may suffer from severe facial deformity or mastication problems even after successful treatment. Therefore, a thorough understanding of this malignancy is essential to prevent and treat it. This review sought to highlight the contributions of next-generation sequencing (NGS) in unveiling the genetic alterations and differential expressions of miRNAs involved in OSCC progression. By applying an appropriate eligibility criterion, we selected relevant studies for review. Frequently identified mutations in genes such as TP53, NOTCH1, and PIK3CA are discussed. The findings of existing miRNAs (e.g., miR-21) as well as novel discoveries pertaining to OSCC are also covered. Lastly, we briefly mention the latest findings in targeted gene therapy and the potential use of miRNAs as biomarkers. Our goal is to encourage researchers to further adopt NGS in their studies and give an overview of the latest findings of OSCC treatment.
Collapse
Affiliation(s)
| | | | - Young-Seok Park
- Department of Oral Anatomy and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 03968, Korea; (S.K.); (J.W.L.)
| |
Collapse
|
52
|
Ramelow J, Brooks CD, Gao L, Almiman AA, Williams TM, Villalona-Calero MA, Duan W. The oncogenic potential of a mutant TP53 gene explored in two spontaneous lung cancer mice models. BMC Cancer 2020; 20:738. [PMID: 32770960 PMCID: PMC7414707 DOI: 10.1186/s12885-020-07212-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/23/2020] [Indexed: 12/25/2022] Open
Abstract
Background Lung cancer is the number one cancer killer worldwide. A major drawback in the lung cancer treatment field is the lack of realistic mouse models that replicate the complexity of human malignancy and immune contexture within the tumor microenvironment. Such models are urgently needed. Mutations of the tumor protein p53 are among the most common alterations in human lung cancers. Methods Previously, we developed a line of lung cancer mouse model where mutant human TP53-273H is expressed in a lung specific manner in FVB/N background. To investigate whether the human TP53 mutant has a similar oncogenic potential when it is expressed in another strain of mouse, we crossed the FVB/N-SPC-TP53-273H mice to A/J strain and created A/J-SPC-TP53-273H transgenic mice. We then compared lung tumor formation between A/J-SPC-TP53-273H and FVB/N-SPC-TP53-273H. Results We found the TP53-273H mutant gene has a similar oncogenic potential in lung tumor formation in both mice strains, although A/J strain mice have been found to be a highly susceptible strain in terms of carcinogen-induced lung cancer. Both transgenic lines survived more than 18 months and developed age related lung adenocarcinomas. With micro CT imaging, we found the FVB-SPC-TP53-273H mice survived more than 8 weeks after initial detection of lung cancer, providing a sufficient window for evaluating new anti-cancer agents. Conclusions Oncogenic potential of the most common genetic mutation, TP53-273H, in human lung cancer is unique when it is expressed in different strains of mice. Our mouse models are useful tools for testing novel immune checkpoint inhibitors or other therapeutic strategies in the treatment of lung cancer.
Collapse
Affiliation(s)
- Julian Ramelow
- Department of Human & Molecular Genetics, Herbert Wertheim College of Medicine, The Florida International University, Miami, Florida, 33199, USA.,Biomolecular Sciences Institute, The Florida International University, Miami, Florida, 33199, USA.,Biological Sciences, College of Arts, Science and Education, The Florida International University, Miami, Florida, 33199, USA
| | - Christopher D Brooks
- Comprehensive Cancer Center at the Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Li Gao
- Department of Human & Molecular Genetics, Herbert Wertheim College of Medicine, The Florida International University, Miami, Florida, 33199, USA
| | - Abeer A Almiman
- Department of Human & Molecular Genetics, Herbert Wertheim College of Medicine, The Florida International University, Miami, Florida, 33199, USA
| | - Terence M Williams
- Comprehensive Cancer Center at the Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | | | - Wenrui Duan
- Department of Human & Molecular Genetics, Herbert Wertheim College of Medicine, The Florida International University, Miami, Florida, 33199, USA. .,Biomolecular Sciences Institute, The Florida International University, Miami, Florida, 33199, USA. .,Comprehensive Cancer Center at the Ohio State University College of Medicine, Columbus, OH, 43210, USA.
| |
Collapse
|
53
|
Zhang X, Chen J, Jin H, Zhao W, Chang Z, Wu H. Effect of erlotinib combined with cisplatin on IL-6 and IL-12 in mice with Lewis lung cancer. Oncol Lett 2020; 20:902-906. [PMID: 32566018 PMCID: PMC7285801 DOI: 10.3892/ol.2020.11632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 01/14/2020] [Indexed: 11/20/2022] Open
Abstract
Effect of erlotinib combined with cisplatin on tumor growth, interleukin-6 (IL-6) and interleukin-12 (IL-12) in mice with Lewis lung cancer (LLC) was investigated. Forty-four pure inbred SPF C57BL/6J mice were modeled for LLC and randomized into groups A, B, C and D (n=11 each group). Mice in group A were given normal saline, group B was given erlotinib, group C was given cisplatin injection and group D erlotinib combined with cisplatin. Tumor growth of the mice was observed and the tumor mass was measured. Serum IL-6 and IL-12 levels were measured by enzyme-linked immunosorbent assay (ELISA) 40 days later. At different time-points after medication, tumor volume in group D was significantly lower than that in groups A, B and C (P<0.05), and that in groups B and C was significantly lower than that in group A (P<0.05), whereas there was no significant difference between groups B and C (P>0.05). Tumor mass in groups B, C and D was significantly lower than that in group A (P<0.05), and that in group D was significantly lower than that in groups B and C (P<0.05), whereas there was no significant difference between groups B and C (P>0.05). Compared with groups B and C, mice in group D had significantly lower IL-6 level (P<0.05), but significantly higher IL-12 level (P<0.05). There was no significant difference in IL-6 and IL-12 levels between groups B and C (P>0.05). In conclusion, erlotinib combined with cisplatin can inhibit the tumor growth of mice with LLC, and inhibition of IL-6 level and upregulation of IL-12 level may be one of its therapeutic mechanisms.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Jinliang Chen
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Hongsong Jin
- Department of Oncology, The People's Hospital of Taizhou, Taizhou, Jiangsu 225300, P.R. China
| | - Wei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Zhibo Chang
- Department of Thoracic Surgery, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Huijing Wu
- Department of Medical Oncology, Radio-Chemotherapy Center, Hubei Cancer Hospital, Wuhan, Hubei 430079, P.R. China
| |
Collapse
|
54
|
Onuma AE, Zhang H, Huang H, Williams TM, Noonan A, Tsung A. Immune Checkpoint Inhibitors in Hepatocellular Cancer: Current Understanding on Mechanisms of Resistance and Biomarkers of Response to Treatment. Gene Expr 2020; 20:53-65. [PMID: 32340652 PMCID: PMC7284108 DOI: 10.3727/105221620x15880179864121] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy worldwide and a leading cause of death worldwide. Its incidence continues to increase in the US due to hepatitis C infection and nonalcoholic steatohepatitis. Liver transplantation and resection remain the best therapeutic options for cure, but these are limited by the shortage of available organs for transplantation, diagnosis at advanced stage, and underlying chronic liver disease found in most patients with HCC. Immune checkpoint inhibitors (ICIs) have been shown to be an evolving novel treatment option in certain advanced solid tumors and have been recently approved for inoperable, advanced, and metastatic HCC. Unfortunately, a large cohort of patients with HCC fail to respond to immunotherapy. In this review, we discuss the ICIs currently approved for HCC treatment and their various mechanisms of action. We will highlight current understanding of mechanism of resistance and limitations to ICIs. Finally, we will describe emerging biomarkers of response to ICIs and address future direction on overcoming resistance to immune checkpoint therapy.
Collapse
Affiliation(s)
- Amblessed E. Onuma
- *Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Hongji Zhang
- *Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- †Department of Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Hai Huang
- *Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Terence M. Williams
- ‡Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Anne Noonan
- §Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Allan Tsung
- *Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
55
|
Xu LB, Zhao ZG, Xu SF, Zhang XX, Liu T, Jing CY, Zhang SG, Yu SJ. The landscape of gene mutations and clinical significance of tumor mutation burden in patients with soft tissue sarcoma who underwent surgical resection and received conventional adjuvant therapy. Int J Biol Markers 2020; 35:14-22. [PMID: 32520634 DOI: 10.1177/1724600820925095] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the landscape of gene mutations and the clinical significance of tumor mutation burden (TMB) in patients with soft tissue sarcoma who underwent surgical resection and received conventional adjuvant therapy. METHODS A total of 68 patients with soft tissue sarcoma were included. Postoperative tumor tissue specimens from the patients were collected for DNA extraction. Targeted next-generation sequencing of cancer-relevant genes was performed for the detection of gene mutations and the analysis of TMB. Univariate analysis between TMB status and prognosis was carried out using the Kaplan-Meier survival analysis, and multivariate analysis was adjusted by the Cox regression model. RESULTS No specific genetic mutations associated with soft tissue sarcoma were found. The mutation frequency of TP53, PIK3C2G, NCOR1, and KRAS of the 68 patients with soft tissue sarcoma were observed in 19 cases (27.94%), 15 cases (22.06%), 14 cases (20.59%), and 14 cases (20.59%), respectively. With regard to the analysis of TMB, the overall TMB of the 68 patients with soft tissue sarcoma was relatively low (median: 2.05 per Mb (range: 0∼15.5 per Mb)). Subsequently, TMB status was divided into TMB-Low and TMB-Middle according to the median TMB. Patients with TMB-Low and TMB-Middle were 37 cases (54.41%) and 31 cases (45.59%), respectively. Overall survival analysis indicated that the median overall survival of patients with TMB-Low and TMB-Middle was not reached, and 4.5 years, respectively (P=0.015). CONCLUSION This study characterizes the genetic background of patients with STS soft tissue sarcoma. The TMB was of clinical significance for patients with soft tissue sarcoma who underwent surgical resection and received conventional adjuvant therapy.
Collapse
Affiliation(s)
- Li-Bin Xu
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhen-Guo Zhao
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Song-Feng Xu
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Xin Zhang
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Liu
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chang-You Jing
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Guang Zhang
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sheng-Ji Yu
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
56
|
Zhao YC, Tang D, Yang S, Liu H, Luo S, Stinchcombe TE, Glass C, Su L, Shen S, Christiani DC, Wei Q. Novel Variants of ELP2 and PIAS1 in the Interferon Gamma Signaling Pathway Are Associated with Non-Small Cell Lung Cancer Survival. Cancer Epidemiol Biomarkers Prev 2020; 29:1679-1688. [PMID: 32493705 DOI: 10.1158/1055-9965.epi-19-1450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/12/2020] [Accepted: 05/29/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND IFNγ is a pleiotropic cytokine that plays critical immunomodulatory roles in intercellular communication in innate and adaptive immune responses. Despite recognition of IFNγ signaling effects on host defense against viral infection and its utility in immunotherapy and tumor progression, the roles of genetic variants of the IFNγ signaling pathway genes in survival of patients with cancer remain unknown. METHODS We used a discovery genotyping dataset from the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial (n = 1,185) and a replication genotyping dataset from the Harvard Lung Cancer Susceptibility Study (n = 984) to evaluate associations between 14,553 genetic variants in 150 IFNγ pathway genes and survival of non-small cell lung cancer (NSCLC). RESULTS The combined analysis identified two independent potentially functional SNPs, ELP2 rs7242481G>A and PIAS1 rs1049493T>C, to be significantly associated with NSCLC survival, with a combined HR of 0.85 (95% confidence interval, 0.78-0.92; P < 0.0001) and 0.87 (0.81-0.93; P < 0.0001), respectively. Expression quantitative trait loci analyses showed that the survival-associated ELP2 rs7242481A allele was significantly associated with increased mRNA expression levels of elongator acetyltransferase complex subunit 2 (ELP2) in 373 lymphoblastoid cell lines and 369 whole-blood samples. The PIAS1 rs1049493C allele was significantly associated with decreased mRNA expression levels of PIAS1 in 383 normal lung tissues and 369 whole-blood samples. CONCLUSIONS Genetic variants of IFNγ signaling genes are potential prognostic markers for NSCLC survival, likely through modulating the expression of key genes involved in host immune response. IMPACT Once validated, these variants could be useful predictors of NSCLC survival.
Collapse
Affiliation(s)
- Yu Chen Zhao
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.,Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Dongfang Tang
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.,Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Sen Yang
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.,Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.,Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | - Thomas E Stinchcombe
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.,Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Carolyn Glass
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University School of Medicine, Durham, North Carolina
| | - Li Su
- Department of Environmental Health and Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Sipeng Shen
- Department of Environmental Health and Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - David C Christiani
- Department of Environmental Health and Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts.,Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina. .,Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina.,Department of Medicine, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
57
|
Pestinger V, Smith M, Sillo T, Findlay JM, Laes JF, Martin G, Middleton G, Taniere P, Beggs AD. Use of an Integrated Pan-Cancer Oncology Enrichment Next-Generation Sequencing Assay to Measure Tumour Mutational Burden and Detect Clinically Actionable Variants. Mol Diagn Ther 2020; 24:339-349. [PMID: 32306292 PMCID: PMC7264086 DOI: 10.1007/s40291-020-00462-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The identification of tumour mutational burden (TMB) as a biomarker of response to programmed cell death protein 1 (PD-1) immunotherapy has necessitated the development of genomic assays to measure this. We carried out comprehensive molecular profiling of cancers using the Illumina TruSight Oncology 500 (TSO500) panel and compared these to whole-genome sequencing (WGS). METHODS Cancer samples derived from formalin-fixed material were profiled on the TSO500 panel, sequenced on an Illumina NextSeq 500 instrument and processed through the TSO500 Docker pipeline. Either FASTQ files (PierianDx) or vcf files (OncoKDM) were processed to understand clinical actionability. RESULTS In total, 108 samples (a mixture of colorectal, lung, oesophageal and control samples) were processed via the DNA panel. There was good correlation between TMB, single-nucleotide variants (SNVs), indels and copy-number variations as predicted by TSO500 and WGS (R2 > 0.9) and good reproducibility, with less than 5% variability between repeated controls. For the RNA panel, 13 samples were processed, with all known fusions observed via orthogonal techniques. For clinical actionability, 72 tier 1 variants and 297 tier 2 variants were detected, with clinical trials identified for all patients. CONCLUSIONS The TSO500 assay accurately measures TMB, microsatellite instability, SNVs, indels, copy-number/structural variation and gene fusions when compared to WGS and orthogonal technologies. Coupled with a clinical annotation pipeline, this provides a powerful methodology for identification of clinically actionable variants.
Collapse
Affiliation(s)
- Valerie Pestinger
- Surgical Research Laboratory, Institute of Cancer and Genomic Sciences, University of Birmingham, Vincent Drive, Birmingham, B15 2TT, UK
| | | | - Toju Sillo
- Surgical Research Laboratory, Institute of Cancer and Genomic Sciences, University of Birmingham, Vincent Drive, Birmingham, B15 2TT, UK
| | | | | | | | - Gary Middleton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | | - Andrew D Beggs
- Surgical Research Laboratory, Institute of Cancer and Genomic Sciences, University of Birmingham, Vincent Drive, Birmingham, B15 2TT, UK.
- Queen Elizabeth Hospital Birmingham, Birmingham, UK.
| |
Collapse
|
58
|
Loupakis F, Depetris I, Biason P, Intini R, Prete AA, Leone F, Lombardi P, Filippi R, Spallanzani A, Cascinu S, Bonetti LR, Maddalena G, Valeri N, Sottoriva A, Zapata L, Salmaso R, Munari G, Rugge M, Dei Tos AP, Golovato J, Sanborn JZ, Nguyen A, Schirripa M, Zagonel V, Lonardi S, Fassan M. Prediction of Benefit from Checkpoint Inhibitors in Mismatch Repair Deficient Metastatic Colorectal Cancer: Role of Tumor Infiltrating Lymphocytes. Oncologist 2020; 25:481-487. [PMID: 31967692 PMCID: PMC7288636 DOI: 10.1634/theoncologist.2019-0611] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/27/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Immunotherapy with immune checkpoint inhibitors (ICIs) is highly effective in microsatellite instability-high (MSI-H) metastatic colorectal cancer (mCRC); however, specific predictive biomarkers are lacking. PATIENTS AND METHODS Data and samples from 85 patients with MSI-H mCRC treated with ICIs were gathered. Tumor infiltrating lymphocytes (TILs) and tumor mutational burden (TMB) were analyzed in an exploratory cohort of "super" responders and "clearly" refractory patients; TILs were then evaluated in the whole cohort of patients. Primary objectives were the correlation between the number of TILs and TMB and their role as biomarkers of ICI efficacy. Main endpoints included response rate (RR), progression-free survival (PFS), and overall survival (OS). RESULTS In the exploratory cohort, an increasing number of TILs correlated to higher TMB (Pearson's test, p = .0429). In the whole cohort, median number of TILs was 3.6 in responders compared with 1.8 in nonresponders (Mann-Whitney test, p = .0448). RR was 70.6% in patients with high number of TILs (TILs-H) compared with 42.9% in patients with low number of TILs (odds ratio = 3.20, p = .0291). Survival outcomes differed significantly in favor of TILs-H (PFS: hazard ratio [HR] = 0.42, p = .0278; OS: HR = 0.41, p = .0463). CONCLUSION A significant correlation between higher TMB and increased number of TILs was shown. A significantly higher activity and better PFS and OS with ICI in MSI-H mCRC were reported in cases with high number of TILs, thus supporting further studies of TIL count as predictive biomarker of ICI efficacy. IMPLICATIONS FOR PRACTICE Microsatellite instability is the result of mismatch repair protein deficiency, caused by germline mutations or somatic modifications in mismatch repair genes. In metastatic colorectal cancer (mCRC), immunotherapy (with immune checkpoint inhibitors [ICIs]) demonstrated remarkable clinical benefit in microsatellite instability-high (MSI-H) patients. ICI primary resistance has been observed in approximately 25% of patients with MSI-H mCRC, underlining the need for predictive biomarkers. In this study, tumor mutational burden (TMB) and tumor infiltrating lymphocyte (TIL) analyses were performed in an exploratory cohort of patients with MSI-H mCRC treated with ICIs, demonstrating a significant correlation between higher TMB and increased number of TILs. Results also demonstrated a significant correlation between high number of TILs and clinical responses and survival benefit in a large data set of patients with MSI-H mCRC treated with ICI. TMB and TILs could represent predictive biomarkers of ICI efficacy in MSI-H mCRC and should be incorporated in future trials testing checkpoint inhibitors in colorectal cancer.
Collapse
Affiliation(s)
- Fotios Loupakis
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PaduaItaly
| | - Ilaria Depetris
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PaduaItaly
| | - Paola Biason
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PaduaItaly
| | - Rossana Intini
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PaduaItaly
| | - Alessandra Anna Prete
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PaduaItaly
| | - Francesco Leone
- Medical Oncology, ASL BiellaBiellaItaly
- Medical Oncology, Candiolo Cancer Institute, Fondazione Piemonte per l'Oncologia, IRCCSCandioloItaly
| | - Pasquale Lombardi
- Medical Oncology, Candiolo Cancer Institute, Fondazione Piemonte per l'Oncologia, IRCCSCandioloItaly
- Department of Oncology, University of TurinTurinItaly
| | - Roberto Filippi
- Medical Oncology, Candiolo Cancer Institute, Fondazione Piemonte per l'Oncologia, IRCCSCandioloItaly
- Department of Oncology, University of TurinTurinItaly
| | - Andrea Spallanzani
- Department of Oncology and Haematology, University Hospital of Modena and Reggio EmiliaModenaItaly
| | - Stefano Cascinu
- Department of Oncology and Haematology, University Hospital of Modena and Reggio EmiliaModenaItaly
| | | | - Giulia Maddalena
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PaduaItaly
| | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer ResearchLondonUnited Kingdom
- Department of Medicine, The Royal Marsden National Health Service (NHS) TrustLondonUnited Kingdom
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer ResearchLondonUnited Kingdom
| | - Luis Zapata
- Centre for Evolution and Cancer, The Institute of Cancer ResearchLondonUnited Kingdom
| | - Roberta Salmaso
- Surgical Pathology and Cytopathology Unit, Department of Medicine (DIMED), University of Padua, Padua University HospitalPaduaItaly
| | - Giada Munari
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PaduaItaly
| | - Massimo Rugge
- Surgical Pathology and Cytopathology Unit, Department of Medicine (DIMED), University of Padua, Padua University HospitalPaduaItaly
| | - Angelo Paolo Dei Tos
- Department of Pathology and Molecular Genetics, Treviso General HospitalTrevisoItaly
| | | | | | | | - Marta Schirripa
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PaduaItaly
| | - Vittorina Zagonel
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PaduaItaly
| | - Sara Lonardi
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PaduaItaly
| | - Matteo Fassan
- Surgical Pathology and Cytopathology Unit, Department of Medicine (DIMED), University of Padua, Padua University HospitalPaduaItaly
| |
Collapse
|
59
|
Oligoprogressive Non-Small-Cell Lung Cancer under Treatment with PD-(L)1 Inhibitors. Cancers (Basel) 2020; 12:cancers12041046. [PMID: 32340408 PMCID: PMC7226015 DOI: 10.3390/cancers12041046] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 11/23/2022] Open
Abstract
Oligoprogression (OPD) of non-small-cell lung cancer (NSCLC) occurs in approximately half of patients under targeted compounds (TKI) and facilitates use of regional therapies that can prolong survival. In order to characterize OPD in immunotherapy (IO)-treated NSCLC, we analyzed the failure pattern under PD-1/PD-L1 inhibitors (n = 297) or chemoimmunotherapy (n = 75). Under IO monotherapy, OPD was more frequent (20% vs. 10%, p < 0.05), occurred later (median 11 vs. 5 months, p < 0.01), affected fewer sites (mean 1.1 vs. 1.5, p < 0.05), and involved fewer lesions (1.4 vs. 2.3, p < 0.05) in the first compared to later lines. Lymph nodes (42%, mainly mediastinal) and the brain (39%) were mostly affected, followed by the lung (24%) and other organs. Compared to multifocal progression, OPD occurred later (11 vs. 4 months, p < 0.001) and was associated with longer survival (26 vs. 13 months, p < 0.001) and higher tumor PD-L1 expression (p < 0.001). Chemoimmunotherapy showed a similar incidence of OPD as IO monotherapy (13% vs. 11% at 2 years). Local treatments were applied regularly for brain but only in 50% for extracranial lesions. Thus, NSCLC oligoprogression is less common under IO than under TKI, but also favorable. Since its frequency drops later in the disease, regular restaging and multidisciplinary evaluation are essential in order to exploit the full therapeutic potential.
Collapse
|
60
|
|
61
|
Muñoz-Hidalgo L, San-Miguel T, Megías J, Monleón D, Navarro L, Roldán P, Cerdá-Nicolás M, López-Ginés C. Somatic copy number alterations are associated with EGFR amplification and shortened survival in patients with primary glioblastoma. Neoplasia 2019; 22:10-21. [PMID: 31751860 PMCID: PMC6864306 DOI: 10.1016/j.neo.2019.09.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/16/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary tumor of the central nervous system. With no effective therapy, the prognosis for patients is terrible poor. It is highly heterogeneous and EGFR amplification is its most frequent molecular alteration. In this light, we aimed to examine the genetic heterogeneity of GBM and to correlate it with the clinical characteristics of the patients. For that purpose, we analyzed the status of EGFR and the somatic copy number alterations (CNAs) of a set of tumor suppressor genes and oncogenes. Thus, we found GBMs with high level of EGFR amplification, low level and with no EGFR amplification. Highly amplified tumors showed histological features of aggressiveness. Interestingly, accumulation of CNAs, as a measure of tumor mutational burden, was frequent and significantly associated to shortened survival. EGFR-amplified GBMs displayed both a higher number of concrete CNAs and a higher global tumor mutational burden than their no EGFR-amplified counterparts. In addition to genetic changes previously described in GBM, we found PARK2 and LARGE1 CNAs associated to EGFR amplification. The set of genes analyzed allowed us to explore relevant signaling pathways on GBM. Both PARK2 and LARGE1 are related to receptor tyrosine kinase/PI3K/PTEN/AKT/mTOR-signaling pathway. Finally, we found an association between the molecular pathways altered, EGFR amplification and a poor outcome. Our results underline the potential interest of categorizing GBM according to their EGFR amplification level and the usefulness of assessing the tumor mutational burden. These approaches would open new knowledge possibilities related to GBM biology and therapy.
Collapse
Affiliation(s)
| | - Teresa San-Miguel
- INCLIVA Research Institute, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain; Department of Pathology, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain.
| | - Javier Megías
- INCLIVA Research Institute, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain; Department of Pathology, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Daniel Monleón
- INCLIVA Research Institute, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain; Department of Pathology, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Lara Navarro
- Consortium Hospital General Universitario de Valencia, Av. Tres cruces, 2, 46014 Valencia, Spain
| | - Pedro Roldán
- Department of Neurosurgery, Hospital Clínico Universitario de Valencia, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain
| | - Miguel Cerdá-Nicolás
- INCLIVA Research Institute, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain; Department of Pathology, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Concha López-Ginés
- INCLIVA Research Institute, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain; Department of Pathology, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| |
Collapse
|
62
|
Rebuzzi SE, Leonetti A, Tiseo M, Facchinetti F. Advances in the prediction of long-term effectiveness of immune checkpoint blockers for non-small-cell lung cancer. Immunotherapy 2019; 11:993-1003. [DOI: 10.2217/imt-2019-0107] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Sara Elena Rebuzzi
- Medical Oncology Unit, University Hospital of Parma, 43122 Parma, Italy
- Medical Oncology Unit 1, Ospedale Policlinico San Martino of Genova, 16132 Genova, Italy
| | | | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, 43122 Parma, Italy
- Department of Medicine & Surgery, University of Parma, 43122 Parma, Italy
| | - Francesco Facchinetti
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris Saclay, 94800 Villejuif, France
| |
Collapse
|