1
|
Wang C, Zhao Y, Liang W. Biomarkers to predict the benefits of immune‑checkpoint blockade‑based therapy in patients with malignant peritoneal mesothelioma (Review). Oncol Lett 2024; 28:600. [PMID: 39483967 PMCID: PMC11525615 DOI: 10.3892/ol.2024.14733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/26/2024] [Indexed: 11/03/2024] Open
Abstract
Malignant peritoneal mesothelioma (MPeM) is a type of rare and highly lethal tumor. Immune checkpoint blockade (ICB)-based therapy has shown encouraging clinical activity for MPeM. However, no definitive biomarkers have been identified for predicting which patients with MPeM will benefit from ICB-based therapy. At present, there are several novel potential biomarkers proposed for predicting the response to ICB-based therapy, and biomarkers available in MPeM cells and in the tumor microenvironment have been identified with the potential to predict the efficacy of ICB-based therapy in MPeM. According to the molecular characteristics of MPeM itself, the feasibility of biomarkers in practice, and the body of available evidence, we hypothesize that the following five types of biomarkers can be used to predict the response of ICB-based therapy in patients with MPeM: Tertiary lymphoid structures, immune checkpoints and their ligands, fusion gene neoantigen burden, BRCA1-associated protein-1 haploinsufficiency and transcriptome-based biomarkers. The present review discusses the value and limitations of each type of biomarker, and potential solutions to address the limitations are proposed. The aim of the present review is to provide a background for future studies on ICB-based therapy for MPeM.
Collapse
Affiliation(s)
- Chunhong Wang
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yan Zhao
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Wanru Liang
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
2
|
Giotti B, Dolasia K, Zhao W, Cai P, Sweeney R, Merritt E, Kiner E, Kim GS, Bhagwat A, Nguyen T, Hegde S, Fitzgerald BG, Shroff S, Dawson T, Garcia-Barros M, Abdul-Ghafar J, Chen R, Gnjatic S, Soto A, Brody R, Kim-Schulze S, Chen Z, Beaumont KG, Merad M, Flores RM, Sebra RP, Horowitz A, Marron TU, Tocheva A, Wolf A, Tsankov AM. Single-Cell View of Tumor Microenvironment Gradients in Pleural Mesothelioma. Cancer Discov 2024; 14:2262-2278. [PMID: 38959428 DOI: 10.1158/2159-8290.cd-23-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/24/2024] [Accepted: 07/02/2024] [Indexed: 07/05/2024]
Abstract
Immunotherapies have shown great promise in pleural mesothelioma (PM), yet most patients still do not achieve significant clinical response, highlighting the importance of improving the understanding of the tumor microenvironment (TME). Here, we utilized high-throughput, single-cell RNA sequencing (scRNA-seq) to de novo identify 54 expression programs and construct a comprehensive cellular catalog of the PM TME. We found four cancer-intrinsic programs associated with poor disease outcome and a novel fetal-like, endothelial cell population that likely responds to VEGF signaling and promotes angiogenesis. Across cellular compartments, we observe substantial difference in the TME associated with a cancer-intrinsic sarcomatoid signature, including enrichment in fetal-like endothelial cells, CXCL9+ macrophages, and cytotoxic, exhausted, and regulatory T cells, which we validated using imaging and bulk deconvolution analyses on independent cohorts. Finally, we show, both computationally and experimentally, that NKG2A:HLA-E interaction between NK and tumor cells represents an important new therapeutic axis in PM, especially for epithelioid cases. Significance: This manuscript presents the first single-cell RNA sequencing atlas of PM tumor microenvironment. Findings of translational relevance, validated experimentally and using independent bulk cohorts, include identification of gene programs predictive of survival, a fetal-like endothelial cell population, and NKG2A blockade as a promising new immunotherapeutic intervention in PM.
Collapse
Affiliation(s)
- Bruno Giotti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Komal Dolasia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - William Zhao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Peiwen Cai
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Robert Sweeney
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Elliot Merritt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Grace S Kim
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Atharva Bhagwat
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Thinh Nguyen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Samarth Hegde
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Bailey G Fitzgerald
- Department of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sanjana Shroff
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Travis Dawson
- The Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Monica Garcia-Barros
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jamshid Abdul-Ghafar
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rachel Chen
- The Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sacha Gnjatic
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alan Soto
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rachel Brody
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Seunghee Kim-Schulze
- The Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zhihong Chen
- The Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kristin G Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Miriam Merad
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Raja M Flores
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Robert P Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Amir Horowitz
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Thomas U Marron
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anna Tocheva
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Andrea Wolf
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexander M Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
3
|
Mitra S, Jang HJ, Kuncheria A, Kang SW, Choi JM, Shim JS, Lee C, Ranchod P, Jindra P, Ramineni M, Patel M, Ripley RT, Groth SS, Blackmon SH, Burt BM, Lee HS. Soluble mesothelin-related peptide as a prognosticator in pleural mesothelioma patients receiving checkpoint immunotherapy. J Thorac Cardiovasc Surg 2024:S0022-5223(24)00914-0. [PMID: 39395787 DOI: 10.1016/j.jtcvs.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/27/2024] [Accepted: 10/02/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Immune checkpoint therapy (ICT) has significantly impacted the treatment of malignant pleural mesothelioma (MPM). Despite some promising results from combination therapies, nearly half of MPM patients do not benefit, underscoring the urgent need for reliable predictive biomarkers. This study assesses the prognostic value of serum soluble mesothelin-related peptide (SMRP) and PD-L1 levels in MPM patients receiving ICT. METHODS We conducted a retrospective analysis of 125 MPM patients treated with ICT by measuring pre-ICT serum levels of SMRP and PD-L1. We also examined the correlation of these serum levels with tumor mRNA expressions of mesothelin and PD-L1. Both univariable and multivariable Cox regression analyses were used to determine independent prognosticators for overall survival (OS). A prospective ICT clinical trial and our historical cohort were included for validation. RESULTS Seventy-seven patients (62%) were treated with either anti-PD-(L)1 monotherapy, and the remaining 38% received combination ICT. Higher pre-ICT SMRP levels were observed in epithelioid MPM compared to nonepithelioid MPM. Serum PD-L1 levels did not differ significantly between the different histologic groups. Univariable analysis identified durable clinical benefit, development of immune-related adverse events, and SMRP levels as significantly associated with OS. Multivariable analysis confirmed SMRP as an independent prognostic factor, with lower levels (≤1.35 nmol/L) correlating with improved OS. The association of high SMRP with worse prognosis was validated in the prospective ICT clinical trial cohort and not in our historical cohort treated without ICT. CONCLUSIONS SMRP is a promising serum biomarker for predicting survival in MPM patients treated with ICT and warrants prospective investigation.
Collapse
Affiliation(s)
- Sonali Mitra
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Hee-Jin Jang
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Allen Kuncheria
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Sung Wook Kang
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Jong Min Choi
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Ji Seon Shim
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Claire Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Priyanka Ranchod
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Peter Jindra
- Immune Evaluation Laboratory, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Maheshwari Ramineni
- Department of Pathology, Baylor College of Medicine, Houston, Tex; Dan L Ducan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Tex
| | - Meera Patel
- Dan L Ducan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Tex; Division of Hemato-Oncology, Department of Medicine, Baylor College of Medicine, Houston, Tex
| | - R Taylor Ripley
- Dan L Ducan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Tex; David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Shawn S Groth
- Dan L Ducan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Tex; David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Shanda H Blackmon
- Dan L Ducan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Tex; David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Bryan M Burt
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex; Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif.
| | - Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex; Division of Hemato-Oncology, Department of Medicine, Baylor College of Medicine, Houston, Tex; David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex.
| |
Collapse
|
4
|
Liu C, Xie J, Lin B, Tian W, Wu Y, Xin S, Hong L, Li X, Liu L, Jin Y, Tang H, Deng X, Zou Y, Zheng S, Fang W, Cheng J, Dai X, Bao X, Zhao P. Pan-Cancer Single-Cell and Spatial-Resolved Profiling Reveals the Immunosuppressive Role of APOE+ Macrophages in Immune Checkpoint Inhibitor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401061. [PMID: 38569519 PMCID: PMC11186051 DOI: 10.1002/advs.202401061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/13/2024] [Indexed: 04/05/2024]
Abstract
The heterogeneity of macrophages influences the response to immune checkpoint inhibitor (ICI) therapy. However, few studies explore the impact of APOE+ macrophages on ICI therapy using single-cell RNA sequencing (scRNA-seq) and machine learning methods. The scRNA-seq and bulk RNA-seq data are Integrated to construct an M.Sig model for predicting ICI response based on the distinct molecular signatures of macrophage and machine learning algorithms. Comprehensive single-cell analysis as well as in vivo and in vitro experiments are applied to explore the potential mechanisms of the APOE+ macrophage in affecting ICI response. The M.Sig model shows clear advantages in predicting the efficacy and prognosis of ICI therapy in pan-cancer patients. The proportion of APOE+ macrophages is higher in ICI non-responders of triple-negative breast cancer compared with responders, and the interaction and longer distance between APOE+ macrophages and CD8+ exhausted T (Tex) cells affecting ICI response is confirmed by multiplex immunohistochemistry. In a 4T1 tumor-bearing mice model, the APOE inhibitor combined with ICI treatment shows the best efficacy. The M.Sig model using real-world immunotherapy data accurately predicts the ICI response of pan-cancer, which may be associated with the interaction between APOE+ macrophages and CD8+ Tex cells.
Collapse
Affiliation(s)
- Chuan Liu
- Department of Medical OncologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310003China
| | - Jindong Xie
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Bo Lin
- College of Computer Science and TechnologyZhejiang UniversityHangzhou310053China
- Innovation Centre for InformationBinjiang Institute of Zhejiang UniversityHangzhou310053China
| | - Weihong Tian
- Changzhou Third People's HospitalChangzhou Medical CenterNanjing Medical UniversityChangzhou213000China
| | - Yifan Wu
- School of softwareZhejiang UniversityNingbo315100China
| | - Shan Xin
- Department of GeneticsYale School of medicineNew HavenCT06510USA
| | - Libing Hong
- Department of Medical OncologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310003China
| | - Xin Li
- Department Chronic Inflammation and CancerGerman Cancer Research Center (DKFZ)69120HeidelbergGermany
| | - Lulu Liu
- Department of Medical OncologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310003China
| | - Yuzhi Jin
- Department of Medical OncologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310003China
| | - Hailin Tang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Xinpei Deng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Yutian Zou
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Shaoquan Zheng
- Breast Disease CenterThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510060China
| | - Weijia Fang
- Department of Medical OncologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310003China
| | - Jinlin Cheng
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhou310003China
| | - Xiaomeng Dai
- Department of Medical OncologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310003China
| | - Xuanwen Bao
- Department of Medical OncologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310003China
| | - Peng Zhao
- Department of Medical OncologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310003China
| |
Collapse
|
5
|
Dey S, Devender M, Rani S, Pandey RK. Recent advances in CAR T-cell engineering using synthetic biology: Paving the way for next-generation cancer treatment. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:91-156. [PMID: 38762281 DOI: 10.1016/bs.apcsb.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
This book chapter highlights a comprehensive exploration of the transformative innovations in the field of cancer immunotherapy. CAR (Chimeric Antigen Receptor) T-cell therapy represents a groundbreaking approach to treat cancer by reprogramming a patient immune cells to recognize and destroy cancer cells. This chapter underscores the critical role of synthetic biology in enhancing the safety and effectiveness of CAR T-cell therapies. It begins by emphasizing the growing importance of personalized medicine in cancer treatment, emphasizing the shift from one-size-fits-all approaches to patient-specific solutions. Synthetic biology, a multidisciplinary field, has been instrumental in customizing CAR T-cell therapies, allowing for fine-tuned precision and minimizing unwanted side effects. The chapter highlights recent advances in gene editing, synthetic gene circuits, and molecular engineering, showcasing how these technologies are optimizing CAR T-cell function. In summary, this book chapter sheds light on the remarkable progress made in the development of CAR T-cell therapies using synthetic biology, providing hope for cancer patients and hinting at a future where highly personalized and effective cancer treatments are the norm.
Collapse
Affiliation(s)
- Sangita Dey
- CSO Department, Cellworks Research India Pvt Ltd, Bengaluru, Karnataka, India
| | - Moodu Devender
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Swati Rani
- ICAR, National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, India
| | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden.
| |
Collapse
|
6
|
Lin Y, Burt BM, Lee HS, Nguyen TT, Jang HJ, Lee C, Hong W, Ripley RT, Amos CI, Cheng C. Clonal gene signatures predict prognosis in mesothelioma and lung adenocarcinoma. NPJ Precis Oncol 2024; 8:47. [PMID: 38396241 PMCID: PMC10891127 DOI: 10.1038/s41698-024-00531-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare but lethal pleural cancer with high intratumor heterogeneity (ITH). A recent study in lung adenocarcinoma has developed a clonal gene signature (ORACLE) from multiregional transcriptomic data and demonstrated high prognostic values and reproducibility. However, such a strategy has not been tested in other types of cancer with high ITH. We aimed to identify biomarkers from multi-regional data to prognostically stratify MPM patients. We generated a multiregional RNA-seq dataset for 78 tumor samples obtained from 26 MPM patients, each with one sample collected from a superior, lateral, and inferior region of the tumor. By integrating this dataset with the Cancer Genome Atlas MPM RNA-seq data, we selected 29 prognostic genes displaying high variability across different tumors but low ITH, which named PRACME (Prognostic Risk Associated Clonal Mesothelioma Expression). We evaluated PRACME in two independent MPM datasets and demonstrated its prognostic values. Patients with high signature scores are associated with poor prognosis after adjusting established clinical factors. Interestingly, the PRACME and the ORACLE signatures defined respectively from MPM and lung adenocarcinoma cross-predict prognosis between the two cancer types. Further investigation indicated that the cross-prediction ability might be explained by the high similarity between the two cancer types in their genomic regions with copy number variation, which host many clonal genes. Overall, our clonal signature PRACME provided prognostic stratification in MPM and this study emphasized the importance of multi-regional transcriptomic data for prognostic stratification based on clonal genes.
Collapse
Affiliation(s)
- Yupei Lin
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, 77030, USA
- The Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Bryan M Burt
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Hyun-Sung Lee
- The Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Thinh T Nguyen
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hee-Jin Jang
- The Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Claire Lee
- The Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wei Hong
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, 77030, USA
- The Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Robert Taylor Ripley
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
- Mesothelioma Treatment Center, Baylor St. Luke's Medical Center, Houston, TX, 77030, USA
| | - Christopher I Amos
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, 77030, USA.
- The Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, 77030, USA.
- The Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
7
|
Gao Z, Azar J, Zhu H, Williams-Perez S, Kang SW, Marginean C, Rubinstein MP, Makawita S, Lee HS, Camp ER. Translational and oncologic significance of tertiary lymphoid structures in pancreatic adenocarcinoma. Front Immunol 2024; 15:1324093. [PMID: 38361928 PMCID: PMC10867206 DOI: 10.3389/fimmu.2024.1324093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/11/2024] [Indexed: 02/17/2024] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is an aggressive tumor with poor survival and limited treatment options. PDAC resistance to immunotherapeutic strategies is multifactorial, but partially owed to an immunosuppressive tumor immune microenvironment (TiME). However, the PDAC TiME is heterogeneous and harbors favorable tumor-infiltrating lymphocyte (TIL) populations. Tertiary lymphoid structures (TLS) are organized aggregates of immune cells that develop within non-lymphoid tissue under chronic inflammation in multiple contexts, including cancers. Our current understanding of their role within the PDAC TiME remains limited; TLS are complex structures with multiple anatomic features such as location, density, and maturity that may impact clinical outcomes such as survival and therapy response in PDAC. Similarly, our understanding of methods to manipulate TLS is an actively developing field of research. TLS may function as anti-tumoral immune niches that can be leveraged as a therapeutic strategy to potentiate both existing chemotherapeutic regimens and potentiate future immune-based therapeutic strategies to improve patient outcomes. This review seeks to cover anatomy, relevant features, immune effects, translational significance, and future directions of understanding TLS within the context of PDAC.
Collapse
Affiliation(s)
- Zachary Gao
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Joseph Azar
- The Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Huili Zhu
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Sophia Williams-Perez
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Sung Wook Kang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Celia Marginean
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Mark P. Rubinstein
- The Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Shalini Makawita
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Hyun-Sung Lee
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - E. Ramsay Camp
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX, United States
| |
Collapse
|
8
|
Wang XY, Xu YM, Lau ATY. Proteogenomics in Cancer: Then and Now. J Proteome Res 2023; 22:3103-3122. [PMID: 37725793 DOI: 10.1021/acs.jproteome.3c00196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
For years, the paths of sequencing technologies and mass spectrometry have occurred in isolation, with each developing its own unique culture and expertise. These two technologies are crucial for inspecting complementary aspects of the molecular phenotype across the central dogma. Integrative multiomics strives to bridge the analysis gap among different fields to complete more comprehensive mechanisms of life events and diseases. Proteogenomics is one integrated multiomics field. Here in this review, we mainly summarize and discuss three aspects: workflow of proteogenomics, proteogenomics applications in cancer research, and the SWOT (Strengths, Weaknesses, Opportunities, Threats) analysis of proteogenomics in cancer research. In conclusion, proteogenomics has a promising future as it clarifies the functional consequences of many unannotated genomic abnormalities or noncanonical variants and identifies driver genes and novel therapeutic targets across cancers, which would substantially accelerate the development of precision oncology.
Collapse
Affiliation(s)
- Xiu-Yun Wang
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| |
Collapse
|
9
|
Cedres S, Serna G, Gonzalez-Medina A, Valdivia A, Assaf-Pastrana JD, Iranzo P, Callejo A, Pardo N, Navarro A, Martinez-Marti A, Priano I, Fasani R, Guardia X, Gonzalo J, Carbonell C, Frigola J, Amat R, Navarro V, Dienstmann R, Vivancos A, Nuciforo P, Felip E. Expression of TILs and Patterns of Gene Expression from Paired Samples of Malignant Pleural Mesothelioma (MPM) Patients. Cancers (Basel) 2023; 15:3611. [PMID: 37509274 PMCID: PMC10377125 DOI: 10.3390/cancers15143611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/31/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
MPM is an aggressive disease with an immunosuppressive tumor microenvironment, and interest in exploring immunotherapy in this disease has been increasing. In the first line of treatment, the combination of nivolumab and ipilimumab demonstrated an improvement in survival over chemotherapy. The presence of TILs has been recognized as a marker of antitumor immune response to chemotherapy in solid tumors. The aim of our study is to identify the effect of treatment on immune cells and the immune gene profile in MPM. We investigated the changes in expression of TILs in 10 human MPM paired tumor tissues using immunohistochemistry and gene expression analysis from paired untreated and treated samples. In this small series, we demonstrated that during the evolution of disease without any treatment there was an increase in the inflammatory component in tumor samples. After systemic treatment there was a decrease in the number of TILs. We observed that after systemic treatment or disease progression immune gene signatures were suppressed. Our integrated analysis of paired samples with immune profile and genomic changes on MPM suggested that during the evolution of the disease the immune system tends to switch, turning off with treatment.
Collapse
Affiliation(s)
- Susana Cedres
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Garazi Serna
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | | | - Augusto Valdivia
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Juan David Assaf-Pastrana
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Patricia Iranzo
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Ana Callejo
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Nuria Pardo
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Alejandro Navarro
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Alex Martinez-Marti
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Ilaria Priano
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Roberta Fasani
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Xavier Guardia
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Javier Gonzalo
- Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Caterina Carbonell
- Clinical Research Department, Vall d'Hebron Institute of Oncology (VHIO), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Joan Frigola
- Clinical Research Department, Vall d'Hebron Institute of Oncology (VHIO), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Ramon Amat
- Clinical Research Department, Vall d'Hebron Institute of Oncology (VHIO), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Victor Navarro
- Oncology Data Science Group, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Rodrigo Dienstmann
- Oncology Data Science Group, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Lab, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Enriqueta Felip
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| |
Collapse
|
10
|
Farahmand P, Gyuraszova K, Rooney C, Raffo-Iraolagoitia XL, Jayasekera G, Hedley A, Johnson E, Chernova T, Malviya G, Hall H, Monteverde T, Blyth K, Duffin R, Carlin LM, Lewis D, Le Quesne J, MacFarlane M, Murphy DJ. Asbestos accelerates disease onset in a genetic model of malignant pleural mesothelioma. FRONTIERS IN TOXICOLOGY 2023; 5:1200650. [PMID: 37441092 PMCID: PMC10333928 DOI: 10.3389/ftox.2023.1200650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Hypothesis: Asbestos-driven inflammation contributes to malignant pleural mesothelioma beyond the acquisition of rate-limiting mutations. Methods: Genetically modified conditional allelic mice that were previously shown to develop mesothelioma in the absence of exposure to asbestos were induced with lentiviral vector expressing Cre recombinase with and without intrapleural injection of amosite asbestos and monitored until symptoms required euthanasia. Resulting tumours were examined histologically and by immunohistochemistry for expression of lineage markers and immune cell infiltration. Results: Injection of asbestos dramatically accelerated disease onset and end-stage tumour burden. Tumours developed in the presence of asbestos showed increased macrophage infiltration. Pharmacological suppression of macrophages in mice with established tumours failed to extend survival or to enhance response to chemotherapy. Conclusion: Asbestos-driven inflammation contributes to the severity of mesothelioma beyond the acquisition of rate-limiting mutations, however, targeted suppression of macrophages in established epithelioid mesothelioma showed no therapeutic benefit.
Collapse
Affiliation(s)
- Pooyeh Farahmand
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Claire Rooney
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- Department of Respiratory Medicine, Royal Infirmary, Glasgow, United Kingdom
| | | | - Geeshath Jayasekera
- Glasgow Pleural Disease Unit, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Ann Hedley
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Emma Johnson
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Tatyana Chernova
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Gaurav Malviya
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Holly Hall
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Tiziana Monteverde
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Kevin Blyth
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Glasgow Pleural Disease Unit, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Rodger Duffin
- Centre for Inflammation Research, Edinburgh, United Kingdom
| | - Leo M. Carlin
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - David Lewis
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - John Le Quesne
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Department of Histopathology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Marion MacFarlane
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Daniel J. Murphy
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| |
Collapse
|
11
|
Al Khatib MHDO, Pinton G, Moro L, Porta C. Benefits and Challenges of Inhibiting EZH2 in Malignant Pleural Mesothelioma. Cancers (Basel) 2023; 15:1537. [PMID: 36900330 PMCID: PMC10000483 DOI: 10.3390/cancers15051537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/25/2023] [Accepted: 02/26/2023] [Indexed: 03/04/2023] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive thoracic cancer that is mainly associated with prior exposure to asbestos fibers. Despite being a rare cancer, its global rate is increasing and the prognosis remains extremely poor. Over the last two decades, despite the constant research of new therapeutic options, the combination chemotherapy with cisplatin and pemetrexed has remained the only first-line therapy for MPM. The recent approval of immune checkpoint blockade (ICB)-based immunotherapy has opened new promising avenues of research. However, MPM is still a fatal cancer with no effective treatments. Enhancer of zeste homolog 2 (EZH2) is a histone methyl transferase that exerts pro-oncogenic and immunomodulatory activities in a variety of tumors. Accordingly, a growing number of studies indicate that EZH2 is also an oncogenic driver in MPM, but its effects on tumor microenvironments are still largely unexplored. This review describes the state-of-the-art of EZH2 in MPM biology and discusses its potential use both as a diagnostic and therapeutic target. We highlight current gaps of knowledge, the filling of which will likely favor the entry of EZH2 inhibitors within the treatment options for MPM patients.
Collapse
Affiliation(s)
- MHD Ouis Al Khatib
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
- Center for Translational Research on Autoimmune & Allergic Diseases (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
| | - Giulia Pinton
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
| | - Laura Moro
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
| | - Chiara Porta
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
- Center for Translational Research on Autoimmune & Allergic Diseases (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
| |
Collapse
|
12
|
Lee HS, Jang HJ, Ramineni M, Wang DY, Ramos D, Choi JM, Splawn T, Espinoza M, Almarez M, Hosey L, Jo E, Hilsenbeck S, Amos CI, Ripley RT, Burt BM. A Phase II Window of Opportunity Study of Neoadjuvant PD-L1 versus PD-L1 plus CTLA-4 Blockade for Patients with Malignant Pleural Mesothelioma. Clin Cancer Res 2023; 29:548-559. [PMID: 36469573 PMCID: PMC9898180 DOI: 10.1158/1078-0432.ccr-22-2566] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/13/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE We report the results of a phase II, randomized, window-of-opportunity trial of neoadjuvant durvalumab versus durvalumab plus tremelimumab followed by surgery in patients with resectable malignant pleural mesothelioma (MPM; NCT02592551). PATIENTS AND METHODS The primary objective was alteration of the intratumoral CD8/regulatory T cell (Treg) ratio after combination immune checkpoint blockade (ICB) therapy. Secondary and exploratory objectives included other changes in the tumor microenvironment, survival, safety, tumor pathologic response (PR), and systemic immune responses. RESULTS Nine patients received monotherapy and 11 received combination therapy. Seventeen of the 20 patients (85%) receiving ICB underwent planned thoracotomy. Both ICB regimens induced CD8 T-cell infiltration into MPM tumors but did not alter CD8/Treg ratios. At 34.1 months follow-up, patients receiving combination ICB had longer median overall survival (not reached) compared with those receiving monotherapy (14.0 months). Grade ≥3 immunotoxicity occurred in 8% of patients in the monotherapy group and 27% of patients in the combination group. Tumor PR occurred in 6 of 17 patients receiving ICB and thoracotomy (35.3%), among which major PR (>90% tumor regression) occurred in 2 (11.8%). Single-cell profiling of tumor, blood, and bone marrow revealed that combination ICB remodeled the immune contexture of MPM tumors; mobilized CD57+ effector memory T cells from the bone marrow to the circulation; and increased the formation of tertiary lymphoid structures in MPM tumors that were rich in CD57+ T cells. CONCLUSIONS These data indicate that neoadjuvant durvalumab plus tremelimumab orchestrates de novo systemic immune responses that extend to the tumor microenvironment and correlate with favorable clinical outcomes.
Collapse
Affiliation(s)
- Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hee-Jin Jang
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maheshwari Ramineni
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Daniel Y. Wang
- Section of Hematology and Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daniela Ramos
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jong Min Choi
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Taylor Splawn
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Monica Espinoza
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michelle Almarez
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Leandria Hosey
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eunji Jo
- Advanced Technology Cores, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Susan Hilsenbeck
- Advanced Technology Cores, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Christopher I. Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX
| | - R. Taylor Ripley
- David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bryan M. Burt
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
13
|
Jang HJ, Truong CY, Lo EM, Holmes HM, Ramos D, Ramineni M, Lee JS, Wang DY, Pietropaolo M, Ripley RT, Burt BM, Lee HS. Inhibition of Cyclin Dependent Kinase 4/6 Overcomes Primary Resistance to Programmed Cell Death 1 Blockade in Malignant Mesothelioma. Ann Thorac Surg 2022; 114:1842-1852. [PMID: 34592265 PMCID: PMC8957629 DOI: 10.1016/j.athoracsur.2021.08.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/20/2021] [Accepted: 08/30/2021] [Indexed: 11/01/2022]
Abstract
BACKGROUND Despite the profound number of malignant pleural mesothelioma (MPM) patients now treated with programmed cell death 1 (PD-1) blockade, insight into the underpinnings of rational therapeutic strategies to treat resistance to checkpoint immunotherapy remains unrealized. Our objective was to develop a novel therapeutic approach to overcome primary resistance to PD-1 blockade in MPM. METHODS We generated a transcriptome signature of resistance to PD-1 blockade in MPM patients treated with nivolumab (4 responders and 4 nonresponders). We used The Cancer Genome Atlas MPM cohort (n = 73) to determine what genomic alterations were associated with the resistance signature. We tested whether regulation of identified molecules could overcome resistance to PD-1 blockade in an immunocompetent mouse malignant mesothelioma model. RESULTS Immunogenomic analysis by applying our anti-PD-1 resistance signature to The Cancer Genome Atlas cohort revealed that deletion of cyclin dependent kinase inhibitor 2A (CDKN2A) was highly associated with primary resistance to PD-1 blockade. Under the hypothesis that resistance to PD-1 blockade can be overcome by cyclin dependent kinase 4/6 (CDK4/6) inhibition, we tested whether CDK4/6 inhibitors could overcome resistance to PD-1 blockade in subcutaneous tumors derived from Cdkn2a-/- AB1 malignant mesothelioma cells, which were resistant to PD-1 blockade. The combination of daily oral administration of CDK4/6 inhibitors (abemaciclib or palbociclib) and intraperitoneal anti-PD-1 treatment markedly suppressed tumor growth compared with anti-PD-1 or CDK4/6 inhibitor alone. CONCLUSIONS We identified a therapeutic target, CDK4/6, to overcome primary resistance to PD-1 blockade through comprehensive immunogenomic approaches. These data provide a rationale for undertaking clinical trials of CDK4/6 inhibitors in more than 40% of patients with MPM who demonstrate loss of CDKN2A.
Collapse
Affiliation(s)
- Hee-Jin Jang
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Cynthia Y Truong
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Eric M Lo
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Hudson M Holmes
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Daniela Ramos
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | | | - Ju-Seog Lee
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Y Wang
- Department of Hematology and Oncology, Baylor College of Medicine, Houston, Texas
| | - Massimo Pietropaolo
- Division of Endocrinology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - R Taylor Ripley
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Bryan M Burt
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Hyun-Sung Lee
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
14
|
Nguyen TT, Lee HS, Burt BM, Amos CI, Cheng C. A combination of intrinsic and extrinsic features improves prognostic prediction in malignant pleural mesothelioma. Br J Cancer 2022; 127:1691-1700. [PMID: 35999269 PMCID: PMC9596423 DOI: 10.1038/s41416-022-01950-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Malignant pleural mesothelioma (MPM) is a lung pleural cancer with very poor disease outcome. With limited curative MPM treatment available, it is vital to study prognostic biomarkers to categorise different patient risk groups. METHODS We defined gene signatures to separately characterise intrinsic and extrinsic features, and investigated their interactions in MPM tumour samples. Specifically, we calculated gene signature scores to capture the downstream pathways of major mutated driver genes (BAP1, NF2, SETD2 and TP53) as tumour-intrinsic features. Similarly, we inferred the infiltration levels for major immune cells in the tumour microenvironment to characterise tumour-extrinsic features. Lastly, we integrated these features with clinical factors to predict prognosis in MPM. RESULTS The gene signature scores were more prognostic than the corresponding genomic mutations, mRNA and protein expression. High immune infiltration levels were associated with prolonged survival. The integrative model indicated that tumour features provided independent prognostic values than clinical factors and were complementary with each other in survival prediction. CONCLUSIONS By using an integrative model that combines intrinsic and extrinsic features, we can more correctly predict the clinical outcomes of patients with MPM.
Collapse
Affiliation(s)
- Thinh T Nguyen
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hyun-Sung Lee
- Division of General Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Bryan M Burt
- Division of General Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chao Cheng
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Yang K, Yang T, Yang T, Yuan Y, Li F. Unraveling tumor microenvironment heterogeneity in malignant pleural mesothelioma identifies biologically distinct immune subtypes enabling prognosis determination. Front Oncol 2022; 12:995651. [PMID: 36237331 PMCID: PMC9552848 DOI: 10.3389/fonc.2022.995651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/14/2022] [Indexed: 11/30/2022] Open
Abstract
Background Malignant pleural mesothelioma (MPM) is a rare and intractable disease exhibiting a remarkable intratumoral heterogeneity and dismal prognosis. Although immunotherapy has reshaped the therapeutic strategies for MPM, patients react with discrepant responsiveness. Methods Herein, we recruited 333 MPM patients from 5 various cohorts and developed an in-silico classification system using unsupervised Non-negative Matrix Factorization and Nearest Template Prediction algorithms. The genomic alterations, immune signatures, and patient outcomes were systemically analyzed across the external TCGA-MESO samples. Machine learning-based integrated methodology was applied to identify a gene classifier for clinical application. Results The gene expression profiling-based classification algorithm identified immune-related subtypes for MPMs. In comparison with the non-immune subtype, we validated the existence of abundant immunocytes in the immune subtype. Immune-suppressed MPMs were enriched with stroma fraction, myeloid components, and immunosuppressive tumor-associated macrophages (TAMs) as well exhibited increased TGF-β signature that informs worse clinical outcomes and reduced efficacy of anti-PD-1 treatment. The immune-activated MPMs harbored the highest lymphocyte infiltration, growing TCR and BCR diversity, and presented the pan-cancer immune phenotype of IFN-γ dominant, which confers these tumors with better drug response when undergoing immune checkpoint inhibitor (ICI) treatment. Genetically, BAP1 mutation was most commonly found in patients of immune-activated MPMs and was associated with a favorable outcome in a subtype-specific pattern. Finally, a robust 12-gene classifier was generated to classify MPMs with high accuracy, holding promise value in predicting patient survival. Conclusions We demonstrate that the novel classification system can be exploited to guide the identification of diverse immune subtypes, providing critical biological insights into the mechanisms driving tumor heterogeneity and responsible for cancer-related patient prognoses.
Collapse
Affiliation(s)
- Kaidi Yang
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army General Hospital, Sanya, China
- *Correspondence: Kaidi Yang, ; Fang Li,
| | - Tongxin Yang
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army General Hospital, Sanya, China
| | - Tao Yang
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army General Hospital, Sanya, China
| | - Ye Yuan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fang Li
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army General Hospital, Sanya, China
- *Correspondence: Kaidi Yang, ; Fang Li,
| |
Collapse
|
16
|
Zhu J, Chang R, Wei B, Fu Y, Chen X, Liu H, Zhou W. Photothermal Nano-Vaccine Promoting Antigen Presentation and Dendritic Cells Infiltration for Enhanced Immunotherapy of Melanoma via Transdermal Microneedles Delivery. Research (Wash D C) 2022; 2022:9816272. [PMID: 36157510 PMCID: PMC9484834 DOI: 10.34133/2022/9816272] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/09/2022] [Indexed: 11/06/2022] Open
Abstract
Immunotherapy has demonstrated the potential to cure melanoma, while the current response rate is still unsatisfactory in clinics. Extensive evidence indicates the correlation between the efficacy and pre-existing T-cell in tumors, whereas the baseline T-cell infiltration is lacking in low-response melanoma patients. Herein, we demonstrated the critical contribution of dendritic cells (DCs) on melanoma survival and baseline T-cell level, as well as the efficacy of immunotherapy. Capitalized on this fact, we developed a photothermal nano-vaccine to simultaneously promote tumor antigens presentation and DCs infiltration for enhanced immunotherapy. The nano-vaccine was composed of polyserotonin (PST) core and tannic acid (TA)/Mn2+ coordination-based metal-organic-framework (MOF) shell for β-catenin silencing DNAzyme loading, which was further integrated into dissolving microneedles to allow noninvasive and transdermal administration at melanoma skin. The nano-vaccine could rapidly penetrate skin upon microneedles insertion and exert a synergistically amplified photothermal effect to induce immunogenic cell death (ICD). The MOF shell then dissociated and released Mn2+ as a cofactor to self-activate DNAzyme for β-catenin suppression, which in turn caused a persistent CCL4 excretion to promote the infiltration of DCs into the tumor. Meanwhile, the liberated PST core could effectively capture and facilitate tumor antigens presentation to DCs. As a result, potent antitumor efficacies were achieved for both primary and distal tumors without any extra treatment, indicating the great promise of such a nano-vaccine for on-demand personalized immunotherapy of melanoma.
Collapse
Affiliation(s)
- Jiaojiao Zhu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Ruimin Chang
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Benliang Wei
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Yao Fu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Hong Liu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
17
|
Zemek RM, Chin WL, Fear VS, Wylie B, Casey TH, Forbes C, Tilsed CM, Boon L, Guo BB, Bosco A, Forrest ARR, Millward MJ, Nowak AK, Lake RA, Lassmann T, Joost Lesterhuis W. Temporally restricted activation of IFNβ signaling underlies response to immune checkpoint therapy in mice. Nat Commun 2022; 13:4895. [PMID: 35986006 PMCID: PMC9390963 DOI: 10.1038/s41467-022-32567-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 08/06/2022] [Indexed: 02/08/2023] Open
Abstract
The biological determinants of the response to immune checkpoint blockade (ICB) in cancer remain incompletely understood. Little is known about dynamic biological events that underpin therapeutic efficacy due to the inability to frequently sample tumours in patients. Here, we map the transcriptional profiles of 144 responding and non-responding tumours within two mouse models at four time points during ICB. We find that responding tumours display on/fast-off kinetics of type-I-interferon (IFN) signaling. Phenocopying of this kinetics using time-dependent sequential dosing of recombinant IFNs and neutralizing antibodies markedly improves ICB efficacy, but only when IFNβ is targeted, not IFNα. We identify Ly6C+/CD11b+ inflammatory monocytes as the primary source of IFNβ and find that active type-I-IFN signaling in tumour-infiltrating inflammatory monocytes is associated with T cell expansion in patients treated with ICB. Together, our results suggest that on/fast-off modulation of IFNβ signaling is critical to the therapeutic response to ICB, which can be exploited to drive clinical outcomes towards response. Immune checkpoint blockade (ICB) is partially successful as a cancer therapy. Here using mouse models, the authors transcriptionally monitor responding and non-responding tumours showing that responding tumours were associated with transient IFN-β signalling which could promote the anti-tumour response.
Collapse
|
18
|
Jang HJ, Lee HS, Yu W, Ramineni M, Truong CY, Ramos D, Splawn T, Choi JM, Jung SY, Lee JS, Wang DY, Sederstrom JM, Pietropaolo M, Kheradmand F, Amos CI, Wheeler TM, Ripley RT, Burt BM. Therapeutic Targeting of Macrophage Plasticity Remodels the Tumor-Immune Microenvironment. Cancer Res 2022; 82:2593-2609. [PMID: 35709756 PMCID: PMC9296613 DOI: 10.1158/0008-5472.can-21-3506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/31/2022] [Accepted: 05/04/2022] [Indexed: 01/21/2023]
Abstract
SIGNIFICANCE Comprehensive single-cell proteomics analyses of lung adenocarcinoma progression reveal the role of tumor-associated macrophages in resistance to PD-1 blockade therapy. See related commentary by Lee et al., p. 2515.
Collapse
Affiliation(s)
- Hee-Jin Jang
- Systems Onco-Immunology Laboratory, Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA.,Hee-Jin Jang and Hyun-Sung Lee have equally contributed as first authors
| | - Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA.,Hee-Jin Jang and Hyun-Sung Lee have equally contributed as first authors.,Hyun-Sung Lee and Bryan M. Burt have equally contributed as corresponding authors
| | - Wendong Yu
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maheshwari Ramineni
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cynthia Y. Truong
- Systems Onco-Immunology Laboratory, Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daniela Ramos
- Systems Onco-Immunology Laboratory, Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Taylor Splawn
- Systems Onco-Immunology Laboratory, Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jong Min Choi
- Systems Onco-Immunology Laboratory, Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sung Yun Jung
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ju-Seog Lee
- Department of Systems Biology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Daniel Y. Wang
- Division of Hemato-Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joel M. Sederstrom
- Advanced Technology Cores, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Massimo Pietropaolo
- Division of Endocrinology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Farrah Kheradmand
- Division of Pulmonology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs, Houston, TX, United States
| | - Christopher I. Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX
| | - Thomas M. Wheeler
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - R. Taylor Ripley
- Systems Onco-Immunology Laboratory, Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bryan M. Burt
- Systems Onco-Immunology Laboratory, Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA.,Hyun-Sung Lee and Bryan M. Burt have equally contributed as corresponding authors
| |
Collapse
|
19
|
Sun B, Dong Y, Xu J, Wang Z. Current status and progress in immunotherapy for malignant pleural mesothelioma. Chronic Dis Transl Med 2022; 8:91-99. [PMID: 35774429 PMCID: PMC9215716 DOI: 10.1002/cdt3.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/09/2021] [Indexed: 11/09/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare and aggressive malignant disease. Currently, the platinum doublet of pemetrexed and cisplatin is the standard first-line treatment for unresectable MPM. However, recent promising results of immunotherapy have markedly changed the landscape of MPM treatment. Further, the ongoing innovative therapeutic strategies are expected to expand the range of treatment options; however, several questions remain unanswered. First, establishing predictive biomarkers with high potency is urgently needed to optimize the patient selection process. Second, further exploration of the combination algorithm is expected to unveil more effective and safe regimens. Moreover, other dilemmas, such as the resistance mechanism of immunotherapy and the role of immunotherapy in perioperative settings, still warrant further exploration.
Collapse
Affiliation(s)
- Boyang Sun
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100021China
| | - Yiting Dong
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100021China
| | - Jiachen Xu
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100021China
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100021China
| |
Collapse
|
20
|
Shah R, Klotz LV, Glade J. Current Management and Future Perspective in Pleural Mesothelioma. Cancers (Basel) 2022; 14:1044. [PMID: 35205798 PMCID: PMC8869935 DOI: 10.3390/cancers14041044] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 11/16/2022] Open
Abstract
Pleural mesothelioma is an aggressive malignancy arising from pleural mesothelial cell lining, predominantly associated with prior exposure to asbestos. The ban on asbestos use has led to its lower incidence in many countries, but globally the disease burden is expected to rise. Therefore, well-planned research is needed to develop more effective, tolerable and affordable drugs. The development of novel treatment has been too slow, with only two regimens of systemic therapy with robust phase 3 data approved formally to date. The treatment scenario for resectable disease remains controversial. However, recent developments in the understanding of disease and clinical trials have been encouraging, and may add better treatment options in the coming years. In this review, we discuss the current treatment options for pleural mesothelioma and shed light on some recent studies and ongoing trials.
Collapse
Affiliation(s)
- Rajiv Shah
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital, 69126 Heidelberg, Germany
| | - Laura V. Klotz
- Department of Thoracic Surgery, Thoraxklinik, Heidelberg University Hospital, 69126 Heidelberg, Germany;
| | - Julia Glade
- Institute for Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| |
Collapse
|
21
|
Lapidot M, Saladi SV, Salgia R, Sattler M. Novel Therapeutic Targets and Immune Dysfunction in Malignant Pleural Mesothelioma. Front Pharmacol 2022; 12:806570. [PMID: 35069219 PMCID: PMC8776703 DOI: 10.3389/fphar.2021.806570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
Advances in the treatment of malignant pleural mesothelioma (MPM) have been disappointing, despite the apparent need for new therapeutic options for this rare and devastating cancer. Drug resistance is common and surgical intervention has brought benefits only to a subset of patients. MPM is a heterogenous disease with a surprisingly low mutation rate and recent sequencing efforts have confirmed alterations in a limited number of tumor suppressors that do not provide apparent insights into the molecular mechanisms that drive this malignancy. There is increasing evidence that epigenetic regulation leads to immune evasion and transformation in MPM. Further, the low efficacy of immune checkpoint inhibitors is consistent with a suppression of genes involved in the anti-tumor immune response. We review three promising emerging therapeutic targets (STAT3, KDM4A, heparanase) and highlight their potential effects on the immune response.
Collapse
Affiliation(s)
- Moshe Lapidot
- Department of Thoracic Surgery, Galilee Medical Center, Nahariya, Israel
| | - Srinivas Vinod Saladi
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States
- Broad Institute of Harvard and MIT, Cambridge, MA, United States
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA, United States
| | - Martin Sattler
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| |
Collapse
|
22
|
Duraiswamy J, Turrini R, Minasyan A, Barras D, Crespo I, Grimm AJ, Casado J, Genolet R, Benedetti F, Wicky A, Ioannidou K, Castro W, Neal C, Moriot A, Renaud-Tissot S, Anstett V, Fahr N, Tanyi JL, Eiva MA, Jacobson CA, Montone KT, Westergaard MCW, Svane IM, Kandalaft LE, Delorenzi M, Sorger PK, Färkkilä A, Michielin O, Zoete V, Carmona SJ, Foukas PG, Powell DJ, Rusakiewicz S, Doucey MA, Dangaj Laniti D, Coukos G. Myeloid antigen-presenting cell niches sustain antitumor T cells and license PD-1 blockade via CD28 costimulation. Cancer Cell 2021; 39:1623-1642.e20. [PMID: 34739845 PMCID: PMC8861565 DOI: 10.1016/j.ccell.2021.10.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 07/06/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022]
Abstract
The mechanisms regulating exhaustion of tumor-infiltrating lymphocytes (TIL) and responsiveness to PD-1 blockade remain partly unknown. In human ovarian cancer, we show that tumor-specific CD8+ TIL accumulate in tumor islets, where they engage antigen and upregulate PD-1, which restrains their functions. Intraepithelial PD-1+CD8+ TIL can be, however, polyfunctional. PD-1+ TIL indeed exhibit a continuum of exhaustion states, with variable levels of CD28 costimulation, which is provided by antigen-presenting cells (APC) in intraepithelial tumor myeloid niches. CD28 costimulation is associated with improved effector fitness of exhausted CD8+ TIL and is required for their activation upon PD-1 blockade, which also requires tumor myeloid APC. Exhausted TIL lacking proper CD28 costimulation in situ fail to respond to PD-1 blockade, and their response may be rescued by local CTLA-4 blockade and tumor APC stimulation via CD40L.
Collapse
Affiliation(s)
- Jaikumar Duraiswamy
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Riccardo Turrini
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Aspram Minasyan
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - David Barras
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; Bioinformatics Core Facility, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Isaac Crespo
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Alizée J Grimm
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Julia Casado
- Research Program of Systems Oncology, University of Helsinki, 00014 Helsinki, Finland
| | - Raphael Genolet
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Fabrizio Benedetti
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Alexandre Wicky
- Center for Precision Oncology, Department of Oncology, CHUV, 1011 Lausanne, Switzerland
| | - Kalliopi Ioannidou
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Wilson Castro
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Christopher Neal
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Amandine Moriot
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Stéphanie Renaud-Tissot
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; Center of Experimental Therapeutics, Department of Oncology, CHUV, 1011 Lausanne, Switzerland
| | - Victor Anstett
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Noémie Fahr
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Janos L Tanyi
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Monika A Eiva
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Connor A Jacobson
- Harvard Ludwig Center, Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Kathleen T Montone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Lana E Kandalaft
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; Center of Experimental Therapeutics, Department of Oncology, CHUV, 1011 Lausanne, Switzerland
| | - Mauro Delorenzi
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland; Department of Oncology, UNIL, 1011 Lausanne, Switzerland
| | - Peter K Sorger
- Harvard Ludwig Center, Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Anniina Färkkilä
- Research Program of Systems Oncology, University of Helsinki, 00014 Helsinki, Finland; Department of Obstetrics and Gynecology, Helsinki University Hospital, 00014 Helsinki, Finland
| | - Olivier Michielin
- Center for Precision Oncology, Department of Oncology, CHUV, 1011 Lausanne, Switzerland
| | - Vincent Zoete
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Santiago J Carmona
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Periklis G Foukas
- 2nd Department of Pathology, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Daniel J Powell
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sylvie Rusakiewicz
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; Center of Experimental Therapeutics, Department of Oncology, CHUV, 1011 Lausanne, Switzerland
| | - Marie-Agnès Doucey
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Denarda Dangaj Laniti
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - George Coukos
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland.
| |
Collapse
|
23
|
Cersosimo F, Barbarino M, Lonardi S, Vermi W, Giordano A, Bellan C, Giurisato E. Mesothelioma Malignancy and the Microenvironment: Molecular Mechanisms. Cancers (Basel) 2021; 13:cancers13225664. [PMID: 34830817 PMCID: PMC8616064 DOI: 10.3390/cancers13225664] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022] Open
Abstract
Several studies have reported that cellular and soluble components of the tumor microenvironment (TME) play a key role in cancer-initiation and progression. Considering the relevance and the complexity of TME in cancer biology, recent research has focused on the investigation of the TME content, in terms of players and informational exchange. Understanding the crosstalk between tumor and non-tumor cells is crucial to design more beneficial anti-cancer therapeutic strategies. Malignant pleural mesothelioma (MPM) is a complex and heterogenous tumor mainly caused by asbestos exposure with few treatment options and low life expectancy after standard therapy. MPM leukocyte infiltration is rich in macrophages. Given the failure of macrophages to eliminate asbestos fibers, these immune cells accumulate in pleural cavity leading to the establishment of a unique inflammatory environment and to the malignant transformation of mesothelial cells. In this inflammatory landscape, stromal and immune cells play a driven role to support tumor development and progression via a bidirectional communication with tumor cells. Characterization of the MPM microenvironment (MPM-ME) may be useful to understand the complexity of mesothelioma biology, such as to identify new molecular druggable targets, with the aim to improve the outcome of the disease. In this review, we summarize the known evidence about the MPM-ME network, including its prognostic and therapeutic relevance.
Collapse
Affiliation(s)
- Francesca Cersosimo
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
| | - Marcella Barbarino
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (M.B.); (A.G.); (C.B.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy; (S.L.); (W.V.)
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy; (S.L.); (W.V.)
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Antonio Giordano
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (M.B.); (A.G.); (C.B.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Cristiana Bellan
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (M.B.); (A.G.); (C.B.)
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
- Correspondence: ; Tel.: +39-057-723-2125
| |
Collapse
|
24
|
Huang R, Zheng Z, Liu S, Yan P, Song D, Yin H, Hu P, Zhu X, Chang Z, Liu Y, Zhuang J, Meng T, Huang Z, Zhang J. Identification of prognostic and bone metastasis-related alternative splicing signatures in mesothelioma. Cancer Med 2021; 10:4478-4492. [PMID: 34041868 PMCID: PMC8267146 DOI: 10.1002/cam4.3977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 01/18/2023] Open
Abstract
Mesothelioma (MESO) is an infrequent tumor derived from mesothelial cells of pleura, peritoneum, pericardium, and tunica vaginalis testis. Despite advancement in technologies and better understanding of tumor progression mechanism, the prognosis of MESO remains poor. The role of alternative splicing events (ASEs) in the oncogenesis, tumor metastasis and drug resistance has been widely discussed in multiple cancers. But the prognosis and potential therapeutic value of ASEs in MESO were not clearly studied by now. We constructed a prognostic model using RNA sequencing data and matched ASE data of MESO patients obtained from the TCGA and TCGASpliceSeq database. A total of 3,993 ASEs were identified associated with overall survival using Cox regression analysis. Eight of them were finally figured out to institute the model by lasso regression analysis. The risk score of the model can predict the prognosis independently. Among the identified 390 splicing factors (SF), HSPA1A and DDX3Y was significantly associated with 43 OS-SEs. Among these OS-SEs, SNX5-58744-AT (p = 0.048) and SNX5-58745-AT (p = 0.048) were significantly associated with bone metastasis. Co-expression analysis of signal pathways and SNX5-58744-AT, SNX5-58745-AT was also depicted using GSVA. Finally, we proposed that splicing factor (SF) HSPA1A could regulate SNX5-58744-AT (R = -0.414) and SNX5-58745-AT (R = 0.414) through the pathway "Class I MHC mediated antigen processing and presentation" (R = 0.400). In this way, tumorigenesis and bone metastasis of MESO were controlled.
Collapse
Affiliation(s)
- Runzhi Huang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China.,Division of Spine, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Tongji University School of Medicine, Shanghai, China
| | - Zixuan Zheng
- Tongji University School of Medicine, Shanghai, China
| | - Sijia Liu
- Tongji University School of Medicine, Shanghai, China
| | - Penghui Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Huabin Yin
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Peng Hu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaolong Zhu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengyan Chang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yihan Liu
- Tongji University School of Medicine, Shanghai, China
| | - Juanwei Zhuang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tong Meng
- Division of Spine, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zongqiang Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Zhang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China.,Division of Spine, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Napoli F, Listì A, Zambelli V, Witel G, Bironzo P, Papotti M, Volante M, Scagliotti G, Righi L. Pathological Characterization of Tumor Immune Microenvironment (TIME) in Malignant Pleural Mesothelioma. Cancers (Basel) 2021; 13:2564. [PMID: 34073720 PMCID: PMC8197227 DOI: 10.3390/cancers13112564] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/08/2023] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare and highly aggressive disease that arises from pleural mesothelial cells, characterized by a median survival of approximately 13-15 months after diagnosis. The primary cause of this disease is asbestos exposure and the main issues associated with it are late diagnosis and lack of effective therapies. Asbestos-induced cellular damage is associated with the generation of an inflammatory microenvironment that influences and supports tumor growth, possibly in association with patients' genetic predisposition and tumor genomic profile. The chronic inflammatory response to asbestos fibers leads to a unique tumor immune microenvironment (TIME) composed of a heterogeneous mixture of stromal, endothelial, and immune cells, and relative composition and interaction among them is suggested to bear prognostic and therapeutic implications. TIME in MPM is known to be constituted by immunosuppressive cells, such as type 2 tumor-associated macrophages and T regulatory lymphocytes, plus the expression of several immunosuppressive factors, such as tumor-associated PD-L1. Several studies in recent years have contributed to achieve a greater understanding of the pathogenetic mechanisms in tumor development and pathobiology of TIME, that opens the way to new therapeutic strategies. The study of TIME is fundamental in identifying appropriate prognostic and predictive tissue biomarkers. In the present review, we summarize the current knowledge about the pathological characterization of TIME in MPM.
Collapse
Affiliation(s)
- Francesca Napoli
- Department of Oncology, University of Turin, 10043 Orbassano, Italy; (F.N.); (V.Z.); (P.B.); (M.P.); (M.V.); (G.S.)
| | - Angela Listì
- Thoracic Oncology Unit, San Luigi Hospital, 10043 Orbassano, Italy;
| | - Vanessa Zambelli
- Department of Oncology, University of Turin, 10043 Orbassano, Italy; (F.N.); (V.Z.); (P.B.); (M.P.); (M.V.); (G.S.)
| | - Gianluca Witel
- Department of Medical Sciences, University of Turin, City of Health and Science, 10126 Torino, Italy;
| | - Paolo Bironzo
- Department of Oncology, University of Turin, 10043 Orbassano, Italy; (F.N.); (V.Z.); (P.B.); (M.P.); (M.V.); (G.S.)
- Thoracic Oncology Unit, San Luigi Hospital, 10043 Orbassano, Italy;
| | - Mauro Papotti
- Department of Oncology, University of Turin, 10043 Orbassano, Italy; (F.N.); (V.Z.); (P.B.); (M.P.); (M.V.); (G.S.)
- Pathology Unit, City of Health and Science, 10126 Torino, Italy
| | - Marco Volante
- Department of Oncology, University of Turin, 10043 Orbassano, Italy; (F.N.); (V.Z.); (P.B.); (M.P.); (M.V.); (G.S.)
| | - Giorgio Scagliotti
- Department of Oncology, University of Turin, 10043 Orbassano, Italy; (F.N.); (V.Z.); (P.B.); (M.P.); (M.V.); (G.S.)
- Thoracic Oncology Unit, San Luigi Hospital, 10043 Orbassano, Italy;
| | - Luisella Righi
- Department of Oncology, University of Turin, 10043 Orbassano, Italy; (F.N.); (V.Z.); (P.B.); (M.P.); (M.V.); (G.S.)
| |
Collapse
|
26
|
Principe N, Kidman J, Lake RA, Lesterhuis WJ, Nowak AK, McDonnell AM, Chee J. Malignant Pleural Effusions-A Window Into Local Anti-Tumor T Cell Immunity? Front Oncol 2021; 11:672747. [PMID: 33987104 PMCID: PMC8111299 DOI: 10.3389/fonc.2021.672747] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/07/2021] [Indexed: 01/01/2023] Open
Abstract
The success of immunotherapy that targets inhibitory T cell receptors for the treatment of multiple cancers has seen the anti-tumor immune response re-emerge as a promising biomarker of response to therapy. Longitudinal characterization of T cells in the tumor microenvironment (TME) helps us understand how to promote effective anti-tumor immunity. However, serial analyses at the tumor site are rarely feasible in clinical practice. Malignant pleural effusions (MPE) associated with thoracic cancers are an abnormal accumulation of fluid in the pleural space that is routinely drained for patient symptom control. This fluid contains tumor cells and immune cells, including lymphocytes, macrophages and dendritic cells, providing a window into the local tumor microenvironment. Recurrent MPE is common, and provides an opportunity for longitudinal analysis of the tumor site in a clinical setting. Here, we review the phenotype of MPE-derived T cells, comparing them to tumor and blood T cells. We discuss the benefits and limitations of their use as potential dynamic biomarkers of response to therapy.
Collapse
Affiliation(s)
- Nicola Principe
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - Joel Kidman
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - Richard A. Lake
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - Willem Joost Lesterhuis
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- Telethon Kids Institute, Perth, WA, Australia
| | - Anna K. Nowak
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, University of Western Australia, Nedlands, WA, Australia
- School of Medicine, University of Western Australia, Crawley, WA, Australia
| | | | - Jonathan Chee
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
27
|
Alay A, Cordero D, Hijazo-Pechero S, Aliagas E, Lopez-Doriga A, Marín R, Palmero R, Llatjós R, Escobar I, Ramos R, Padrones S, Moreno V, Nadal E, Solé X. Integrative transcriptome analysis of malignant pleural mesothelioma reveals a clinically relevant immune-based classification. J Immunother Cancer 2021; 9:jitc-2020-001601. [PMID: 33632900 PMCID: PMC7908918 DOI: 10.1136/jitc-2020-001601] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2021] [Indexed: 12/24/2022] Open
Abstract
Background Malignant pleural mesothelioma (MPM) is a rare and aggressive neoplasia affecting the lung mesothelium. Immune checkpoint inhibitors (ICI) in MPM have not been extremely successful, likely due to poor identification of suitable candidate patients for the therapy. We aimed to identify cellular immune fractions associated with clinical outcome and classify patients with MPM based on their immune contexture. For each defined group, we sought for molecular specificities that could help further define our MPM classification at the genomic and transcriptomic level, as well as identify differential therapeutic strategies based on transcriptional signatures predictive of drug response. Methods The abundance of 20 immune cell fractions in 516 MPM samples from 7 gene expression datasets was inferred using gene set variation analysis. Identification of clinically relevant fractions was performed with Cox proportional-hazards models adjusted for age, stage, sex, and tumor histology. Immune-based groups were defined based on the identified fractions. Results T-helper 2 (TH2) and cytotoxic T (TC) cells were found to be consistently associated with overall survival. Three immune clusters (IG) were subsequently defined based on TH2 and TC immune infiltration levels: IG1 (54.5%) was characterized by high TH2 and low TC levels, IG2 (37%) had either low or high levels of both fractions, and IG3 (8.5%) was defined by low TH2 and high TC levels. IG1 and IG3 groups were associated with worse and better overall survival, respectively. While no differential genomic alterations were identified among immune groups, at the transcriptional level, IG1 samples showed upregulation of proliferation signatures, while IG3 samples presented upregulation of immune and inflammation-related pathways. Finally, the integration of gene expression with functional signatures of drug response showed that IG3 patients might be more likely to respond to ICI. Conclusions This study identifies a novel immune-based signature with potential clinical relevance based on TH2 and TC levels, unveiling a fraction of patients with MPM with better prognosis and who might benefit from immune-based therapies. Molecular specificities of the different groups might be used to tailor specific potential therapies in the future.
Collapse
Affiliation(s)
- Ania Alay
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology (ICO)-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - David Cordero
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology (ICO)-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Biomedical Research Centre Network for Epidemiology & Public Health (CIBERESP), Madrid, Comunidad de Madrid, Spain
| | - Sara Hijazo-Pechero
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology (ICO)-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Elisabet Aliagas
- Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Adriana Lopez-Doriga
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology (ICO)-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Biomedical Research Centre Network for Epidemiology & Public Health (CIBERESP), Madrid, Comunidad de Madrid, Spain
| | - Raúl Marín
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology (ICO)-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ramón Palmero
- Department of Medical Oncology, Catalan Institute of Oncology (ICO)-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Roger Llatjós
- Department of Pathology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ignacio Escobar
- Department of Thoracic Surgery, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ricard Ramos
- Department of Thoracic Surgery, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Susana Padrones
- Department of Respiratory Medicine, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Víctor Moreno
- Biomedical Research Centre Network for Epidemiology & Public Health (CIBERESP), Madrid, Comunidad de Madrid, Spain.,Oncology Data Analytics Program, Catalan Institute of Oncology (ICO)-Bellvitge Biomedical Research Institute (IDIBELL)-University of Barcelona (UB), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ernest Nadal
- Department of Medical Oncology, Catalan Institute of Oncology (ICO)-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Xavier Solé
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology (ICO)-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain .,Biomedical Research Centre Network for Epidemiology & Public Health (CIBERESP), Madrid, Comunidad de Madrid, Spain
| |
Collapse
|
28
|
Lee HS, Hamaji M, Palivela N, Jang HJ, Splawn T, Ramos D, Lee AK, Raghuram AC, Ramineni M, Amos CI, Ripley RT, Burt BM. Prognostic Role of Programmed Cell Death 1 Ligand 1 in Resectable Pleural Mesothelioma. Ann Thorac Surg 2020; 112:1575-1583. [PMID: 33248997 DOI: 10.1016/j.athoracsur.2020.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/08/2020] [Accepted: 10/25/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND The prognostic role of programmed cell death 1 ligand 1 (PD-L1) in malignant pleural mesothelioma (MPM) is incompletely understood. Our objectives were to evaluate the evidence for tumor PD-L1 as a prognostic biomarker in MPM through meta-analysis and to determine whether tumor PD-L1 expression is associated with survival in MPM patients undergoing macroscopic complete resection. METHODS Meta-analysis was performed to determine the association of PD-L1 with overall survival in MPM (n = 1655) from 14 studies containing overall survival and tumor PD-L1 expression. Univariable and multivariable analyses tested the relationship of tumor PD-L1 with overall survival and recurrence-free survival in an institutional cohort of MPM patients treated by macroscopic complete resection (n = 75). To validate the association of PD-L1 with overall survival, we utilized two independent MPM cohorts (n = 284). RESULTS Meta-analysis demonstrated that high tumor PD-L1 expression was associated with poor overall survival. Among 75 patients undergoing macroscopic complete resection, 49 tumors (65%) expressed PD-L1 (1% or more), and high PD-L1 (50% or greater) was more commonly expressed on nonepithelial (29%) compared with epithelial tumors (14%). High tumor PD-L1 expression was independently associated with poor overall survival (P < .001, hazard ratio 5.67) and recurrence-free survival (P = .003, hazard ratio 3.28). The association of PD-L1 overexpression with unfavorable survival was more significant in epithelial MPMs than nonepithelial MPMs. These findings were validated in RNA sequencing analyses in two independent cohorts. Exploratory transcriptome analysis revealed that MPM tumors with PD-L1 overexpression displayed coexpression of other immune regulatory molecules, programmed cell death 1 ligand 2 and T-cell immunoglobulin mucin receptor 3. CONCLUSIONS Tumor PD-L1 expression is a prognostic biomarker in patients undergoing surgical resection for MPM and may be useful in perioperative decision making.
Collapse
Affiliation(s)
- Hyun-Sung Lee
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Masatsugu Hamaji
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Nihanth Palivela
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Hee-Jin Jang
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Taylor Splawn
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Daniela Ramos
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Alice K Lee
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas; Department of Kinesiology, Rice University, Houston, Texas
| | - Anjali C Raghuram
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | | | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, Texas
| | - R Taylor Ripley
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Bryan M Burt
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
29
|
Abstract
Malignant mesothelioma (MM) is uncommon, but very aggressive tumor arising from the mesothelial cells of pleura, pericardium, peritoneum, and tunica vaginalis. Despite multimodality treatments 5-year survival is only 5% after the diagnosis. Histology and TNM staging system are still the best prognostic factors. Furthermore, histologic subtype of MM determines the clinical management of the patients. According to the 2015 WHO classification, MM is divided into diffuse, localized and well differentiated papillary mesothelioma. Major histologic subtypes of diffuse MM, namely epithelioid, biphasic and sarcomatoid, have different prognosis. However, in the last decade it has become evident that more detailed subclassification and histologic/cytological characterization of MM have prognostic and perhaps predictive implications. In this review, major histologic subtypes and cytological features of MM are presented and their relation with prognosis and predictive biomarkers is discussed.
Collapse
Affiliation(s)
- Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Izidor Kern
- Cytology and Pathology Laboratory, University Clinic of Respiratory and Allergic Diseases, Golnik, Slovenia
| |
Collapse
|
30
|
Zemek RM, Fear VS, Forbes C, de Jong E, Casey TH, Boon L, Lassmann T, Bosco A, Millward MJ, Nowak AK, Lake RA, Lesterhuis WJ. Bilateral murine tumor models for characterizing the response to immune checkpoint blockade. Nat Protoc 2020; 15:1628-1648. [PMID: 32238953 DOI: 10.1038/s41596-020-0299-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/16/2020] [Indexed: 02/01/2023]
Abstract
The therapeutic response to immune checkpoint blockade (ICB) is highly variable, not only between different cancers but also between patients with the same cancer type. The biological mechanisms underlying these differences in response are incompletely understood. Identifying correlates in patient tumor samples is challenging because of genetic and environmental variability. Murine studies usually compare different tumor models or treatments, introducing potential confounding variables. This protocol describes bilateral murine tumor models, derived from syngeneic cancer cell lines, that display a symmetrical yet dichotomous response to ICB. These models enable detailed analysis of whole tumors in a highly homogeneous background, combined with knowledge of the therapeutic outcome within a few weeks, and could potentially be used for mechanistic studies using other (immuno-)therapies. We discuss key considerations and describe how to use two cell lines as fully optimized models. We discuss experimental details, including proper inoculation technique to achieve symmetry and one-sided surgical tumor removal, which takes only 5 min per mouse. Furthermore, we outline the preparation of bulk tissue or single-cell suspensions for downstream analyses such as bulk RNA-seq, immunohistochemistry, single-cell RNA-seq and flow cytometry.
Collapse
Affiliation(s)
- Rachael M Zemek
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia. .,National Centre for Asbestos Related Diseases, Nedlands, Western Australia, Australia. .,Telethon Kids Institute, University of Western Australia, West Perth, Western Australia, Australia.
| | - Vanessa S Fear
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia.,National Centre for Asbestos Related Diseases, Nedlands, Western Australia, Australia.,Telethon Kids Institute, University of Western Australia, West Perth, Western Australia, Australia
| | - Cath Forbes
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia.,National Centre for Asbestos Related Diseases, Nedlands, Western Australia, Australia.,Telethon Kids Institute, University of Western Australia, West Perth, Western Australia, Australia
| | - Emma de Jong
- Telethon Kids Institute, University of Western Australia, West Perth, Western Australia, Australia
| | - Thomas H Casey
- National Centre for Asbestos Related Diseases, Nedlands, Western Australia, Australia
| | | | - Timo Lassmann
- Telethon Kids Institute, University of Western Australia, West Perth, Western Australia, Australia
| | - Anthony Bosco
- Telethon Kids Institute, University of Western Australia, West Perth, Western Australia, Australia
| | - Michael J Millward
- National Centre for Asbestos Related Diseases, Nedlands, Western Australia, Australia.,Medical School, University of Western Australia, Crawley, Western Australia, Australia.,Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Anna K Nowak
- National Centre for Asbestos Related Diseases, Nedlands, Western Australia, Australia.,Medical School, University of Western Australia, Crawley, Western Australia, Australia.,Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Richard A Lake
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia.,National Centre for Asbestos Related Diseases, Nedlands, Western Australia, Australia
| | - W Joost Lesterhuis
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia. .,National Centre for Asbestos Related Diseases, Nedlands, Western Australia, Australia. .,Telethon Kids Institute, University of Western Australia, West Perth, Western Australia, Australia.
| |
Collapse
|
31
|
Expression of V-set immunoregulatory receptor in malignant mesothelioma. Mod Pathol 2020; 33:263-270. [PMID: 31363159 DOI: 10.1038/s41379-019-0328-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/20/2019] [Accepted: 06/21/2019] [Indexed: 11/09/2022]
Abstract
Malignant mesothelioma is a highly lethal cancer. V-set immunoregulatory receptor (VSIR, also known as V-domain Ig suppressor T-cell activation, VISTA), a negative immune checkpoint regulator, was reported to be expressed in malignant mesothelioma; however, its detailed expression pattern and clinicopathological significance have not been elucidated. We examined the expression of VSIR and CD274 and CD8+ tumor-infiltrating lymphocytes in a total of 124 samples from 66 patients with malignant mesothelioma and analyzed the clinicopathological characteristics and their relationship with the immunohistochemical findings. A total of 553 non-small cell lung carcinomas were also evaluated for VSIR expression. VSIR expression was higher in epithelioid type mesothelioma (p < 0.001), whereas CD274 expression was higher in sarcomatoid type (p < 0.001). CD8+ tumor-infiltrating lymphocytes were more abundant in sarcomatoid mesotheliomas (p < 0.001), VSIR-low tumors (p = 0.045), and CD274-high tumors (p < 0.001). VSIR and CD274 were differentially expressed in each histological component of the biphasic type. VSIR expression was associated with favorable survival (p = 0.008). Two patients with VSIR-high tumors had received pembrolizumab; however, they showed progressive disease. No VSIR expression was observed in tumor cells of non-small cell lung carcinomas. In conclusion, VSIR expression may define a unique class of mesothelioma, characterized by predominantly epithelioid type and favorable prognosis. VSIR expression may be used as an immunohistochemical diagnostic marker for epithelioid mesothelioma. CD274 expression was associated with sarcomatoid mesothelioma and high infiltration of CD8+ lymphocytes. Because VSIR is a negative immune regulator and expressed in malignant mesothelioma, further study is warranted to investigate the therapeutic significance of VSIR blockade in this deadly cancer.
Collapse
|
32
|
de Perrot M, Wu L, Cabanero M, Perentes JY, McKee TD, Donahoe L, Bradbury P, Kohno M, Chan ML, Murakami J, Keshavjee S, Tsao MS, Cho BCJ. Prognostic influence of tumor microenvironment after hypofractionated radiation and surgery for mesothelioma. J Thorac Cardiovasc Surg 2019; 159:2082-2091.e1. [PMID: 31866087 DOI: 10.1016/j.jtcvs.2019.10.122] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/20/2019] [Accepted: 10/21/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Cytotoxic CD8+ tumor infiltrating lymphocytes (TILs) can contribute to the benefit of hypofractionated radiation, but programmed cell death pathways (programmed cell death 1 and programmed cell death ligand 1 [PD-1/PD-L1]) may provide a mechanism of tumor immune escape. We therefore reviewed the influence of PD-1/PD-L1 and CD8+ TILs on survival after accelerated hypofractionated hemithoracic radiation followed by extrapleural pneumonectomy for malignant pleural mesothelioma (MPM). METHODS Sixty-nine consecutive patients undergoing the protocol of Surgery for Mesothelioma after Radiation Therapy (SMART) between November 2008 and February 2016 were analyzed for the presence of PD-L1 on tumor cells, PD-1 on inflammatory cells, and CD8+ TILs. Comparison was made with a cohort of patients undergoing extrapleural pneumonectomy after induction chemotherapy (n = 14) and no induction (n = 2) between March 2005 and October 2008. PD-L1 expression on tumor cells ≥1% was considered positive. CD8+ TILs and PD-1 expression were scored as a percentage of positive cells. RESULTS PD-L1 was negative in 75% of MPM after completion of SMART. CD8+ TILs ranged between 0.24% and 8.47% (median 2%). CD8+ TILs ≥2% was associated with significantly better survival in epithelioid MPM (median survival 3.7 years vs 2.3 years in CD8+ TILs <2%; P = .02). PD-L1 positivity was associated with worse survival in biphasic MPM (median survival, 0.4 years vs 1.5 years in biphasic PD-L1 negative tumors; P = .07) after SMART. Multivariate analysis demonstrated that epithelioid MPM, nodal disease, and CD8+ TILs were independent predictors of survival after SMART. CONCLUSIONS The influence of tumor microenvironment on survival differs between epithelioid and nonepithelioid MPM. CD8+ TILs is an independent factor associated with better survival in epithelioid MPM treated with SMART.
Collapse
Affiliation(s)
- Marc de Perrot
- Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada; Latner Thoracic Surgery Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada.
| | - Licun Wu
- Latner Thoracic Surgery Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Michael Cabanero
- Department of Pathology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - J Yannis Perentes
- Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Trevor D McKee
- STTARR Program, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Laura Donahoe
- Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Penelope Bradbury
- Division of Medical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mikihiro Kohno
- Latner Thoracic Surgery Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mei-Lin Chan
- Latner Thoracic Surgery Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Junichi Murakami
- Latner Thoracic Surgery Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada; Latner Thoracic Surgery Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Ming-Sound Tsao
- Department of Pathology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - B C John Cho
- Department of Radiation Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Chen-Yoshikawa TF. Commentary: A journey of a thousand miles begins with a single step. J Thorac Cardiovasc Surg 2019; 159:2092-2093. [PMID: 31627947 DOI: 10.1016/j.jtcvs.2019.09.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 11/19/2022]
|
34
|
Hinz TK, Heasley LE. Translating mesothelioma molecular genomics and dependencies into precision oncology-based therapies. Semin Cancer Biol 2019; 61:11-22. [PMID: 31546009 DOI: 10.1016/j.semcancer.2019.09.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 12/28/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a rare, yet lethal asbestos-induced cancer and despite marked efforts to reduce occupational exposure, the incidence has not yet significantly declined. Since 2003, combined treatment with a platinum-based agent and pemetrexed has been the first-line therapy and no effective or approved second-line treatments have emerged. The seemingly slow advance in developing new MPM treatments does not appear to be related to a low level of clinical and pre-clinical research activity. Rather, we suggest that a key hurdle in successfully translating basic discovery into novel MPM therapeutics is the underlying assumption that as a rare cancer, it will also be molecularly and genetically homogeneous. In fact, lung adenocarcinoma and melanoma only benefitted from precision oncology upon full appreciation of the high degree of molecular heterogeneity inherent in these cancers, especially regarding the diversity of oncogenic drivers. Herein, we consider the recent explosion of molecular and genetic information that has become available regarding MPM and suggest ways in which the unfolding landscape may guide identification of novel therapeutic vulnerabilities within subsets of MPM that can be targeted in a manner consistent with the tenets of precision oncology.
Collapse
Affiliation(s)
- Trista K Hinz
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Lynn E Heasley
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States.
| |
Collapse
|
35
|
Abstract
INTRODUCTION Combination chemotherapy is currently standard care for advanced mesothelioma. Checkpoint blockade is a promising new treatment. AREAS COVERED This review covers clinical use and biomarkers of checkpoint blockade. Medline search used keywords 'mesothelioma' combined with 'checkpoint blockade' OR 'PD-L1' OR 'PD1' OR 'anti-CTLA4'; the search terms AND 'clinical trial' or AND 'biomarker*' were added. Handsearching covered abstracts from relevant meetings from 2016 to 2018 and reference lists. Data informed a narrative review. EXPERT OPINION Single agent anti-CTLA4 blockade is inactive in mesothelioma. Single agent PD-1 blockade as second or subsequent treatment gives 20-29% partial responses; no randomized comparisons against placebo or chemotherapy are available. Biomarkers of response have been difficult to identify. There is no consensus as to whether tumor PD-L1 expression predicts outcomes. Combination checkpoint inhibitors (CTLA4 and PD1 blockade) provide a small incremental increase in response rates and progression-free survival. Chemoimmunotherapy is the next frontier.
Collapse
Affiliation(s)
- Anna K Nowak
- a National Centre for Asbestos Related Diseases , University of Western Australia , Crawley , Australia
- b Institute for Respiratory Health, University of Western Australia , Nedlands , Australia
- c Medical School , University of Western Australia , Crawley , Australia
| | - Alison McDonnell
- a National Centre for Asbestos Related Diseases , University of Western Australia , Crawley , Australia
- b Institute for Respiratory Health, University of Western Australia , Nedlands , Australia
- c Medical School , University of Western Australia , Crawley , Australia
| | - Alistair Cook
- a National Centre for Asbestos Related Diseases , University of Western Australia , Crawley , Australia
- b Institute for Respiratory Health, University of Western Australia , Nedlands , Australia
- c Medical School , University of Western Australia , Crawley , Australia
| |
Collapse
|
36
|
Shrestha R, Nabavi N, Lin YY, Mo F, Anderson S, Volik S, Adomat HH, Lin D, Xue H, Dong X, Shukin R, Bell RH, McConeghy B, Haegert A, Brahmbhatt S, Li E, Oo HZ, Hurtado-Coll A, Fazli L, Zhou J, McConnell Y, McCart A, Lowy A, Morin GB, Chen T, Daugaard M, Sahinalp SC, Hach F, Le Bihan S, Gleave ME, Wang Y, Churg A, Collins CC. BAP1 haploinsufficiency predicts a distinct immunogenic class of malignant peritoneal mesothelioma. Genome Med 2019; 11:8. [PMID: 30777124 PMCID: PMC6378747 DOI: 10.1186/s13073-019-0620-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 02/07/2019] [Indexed: 02/06/2023] Open
Abstract
Background Malignant peritoneal mesothelioma (PeM) is a rare and fatal cancer that originates from the peritoneal lining of the abdomen. Standard treatment of PeM is limited to cytoreductive surgery and/or chemotherapy, and no effective targeted therapies for PeM exist. Some immune checkpoint inhibitor studies of mesothelioma have found positivity to be associated with a worse prognosis. Methods To search for novel therapeutic targets for PeM, we performed a comprehensive integrative multi-omics analysis of the genome, transcriptome, and proteome of 19 treatment-naïve PeM, and in particular, we examined BAP1 mutation and copy number status and its relationship to immune checkpoint inhibitor activation. Results We found that PeM could be divided into tumors with an inflammatory tumor microenvironment and those without and that this distinction correlated with haploinsufficiency of BAP1. To further investigate the role of BAP1, we used our recently developed cancer driver gene prioritization algorithm, HIT’nDRIVE, and observed that PeM with BAP1 haploinsufficiency form a distinct molecular subtype characterized by distinct gene expression patterns of chromatin remodeling, DNA repair pathways, and immune checkpoint receptor activation. We demonstrate that this subtype is correlated with an inflammatory tumor microenvironment and thus is a candidate for immune checkpoint blockade therapies. Conclusions Our findings reveal BAP1 to be a potential, easily trackable prognostic and predictive biomarker for PeM immunotherapy that refines PeM disease classification. BAP1 stratification may improve drug response rates in ongoing phases I and II clinical trials exploring the use of immune checkpoint blockade therapies in PeM in which BAP1 status is not considered. This integrated molecular characterization provides a comprehensive foundation for improved management of a subset of PeM patients. Electronic supplementary material The online version of this article (10.1186/s13073-019-0620-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raunak Shrestha
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Bioinformatics Training Program, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Noushin Nabavi
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Yen-Yi Lin
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Fan Mo
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,International Precision Medicine Research Centre, Zhejiang-California International Nanosystems Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China.,Neoantigen Therapeutics, Inc., Hangzhou, 310051, Zhejiang, China
| | - Shawn Anderson
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Stanislav Volik
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Hans H Adomat
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Dong Lin
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Hui Xue
- BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Xin Dong
- BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Robert Shukin
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Robert H Bell
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Brian McConeghy
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Anne Haegert
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Sonal Brahmbhatt
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Estelle Li
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Htoo Zarni Oo
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | | | - Ladan Fazli
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Joshua Zhou
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Yarrow McConnell
- Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Andrea McCart
- Mount Sinai Hospital, 600 University Ave, Toronto, ON, M5G 1X5, Canada
| | - Andrew Lowy
- Moores Cancer Center, 3855 Health Sciences Dr, La Jolla, CA, 92093, USA
| | - Gregg B Morin
- BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Tianhui Chen
- Zhejiang Academy of Medical Sciences, Tianmushan Road 182, Hangzhou, 310013, China
| | - Mads Daugaard
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - S Cenk Sahinalp
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,School of Informatics and Computing, Indiana University, Bloomington, IN, 47408, USA
| | - Faraz Hach
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Stephane Le Bihan
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Martin E Gleave
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.,BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Andrew Churg
- Department of Pathology, Vancouver General Hospital, Vancouver, BC, V5Z 1M9, Canada.
| | - Colin C Collins
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada. .,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
37
|
Mistry AM, Greenplate AR, Ihrie RA, Irish JM. Beyond the message: advantages of snapshot proteomics with single-cell mass cytometry in solid tumors. FEBS J 2019; 286:1523-1539. [PMID: 30549207 DOI: 10.1111/febs.14730] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/17/2018] [Accepted: 12/12/2018] [Indexed: 12/19/2022]
Abstract
Single-cell technologies that can quantify features of individual cells within a tumor are critical for treatment strategies aiming to target cancer cells while sparing or activating beneficial cells. Given that key players in protein networks are often the primary targets of precision oncology strategies, it is imperative to transcend the nucleic acid message and read cellular actions in human solid tumors. Here, we review the advantages of multiplex, single-cell mass cytometry in tissue and solid tumor investigations. Mass cytometry can quantitatively probe nearly any cellular feature or target. In discussing the ability of mass cytometry to reveal and characterize a broad spectrum of cell types, identify rare cells, and study functional behavior through protein signaling networks in millions of individual cells from a tumor, this review surveys publications of scientific advances in solid tumor biology made with the aid of mass cytometry. Advances discussed include functional identification of rare tumor and tumor-infiltrating immune cells and dissection of cellular mechanisms of immunotherapy in solid tumors and the periphery. The review concludes by highlighting ways to incorporate single-cell mass cytometry in solid tumor precision oncology efforts and rapidly developing cytometry techniques for quantifying cell location and sequenced nucleic acids.
Collapse
Affiliation(s)
- Akshitkumar M Mistry
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allison R Greenplate
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rebecca A Ihrie
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
38
|
Ye L, Ma S, Robinson BW, Creaney J. Immunotherapy strategies for mesothelioma - the role of tumor specific neoantigens in a new era of precision medicine. Expert Rev Respir Med 2018; 13:181-192. [PMID: 30596292 DOI: 10.1080/17476348.2019.1563488] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Immunotherapy has long been considered a potential therapy for malignant mesothelioma and is currently being pursued as such. Some of the early phase clinical trials involving immunomodulators have demonstrated encouraging results and numerous clinical trials are underway to further investigate this treatment approach in various treatment settings and larger patient cohorts. Areas covered: This review summarizes the current and emerging clinical evidence for checkpoint blockade and other immunotherapeutic strategies in mesothelioma. The mesothelioma tumor immune microenvironment and mutational landscape are also discussed, including their impact on treatment strategies. We also provide an evaluation of the current evidence for neoantigen targeted personalized immunotherapy. Expert opinion: Immune checkpoint inhibitors work by unleashing the host immune response against probable neoantigens. Despite impressive activity in a small subset of patients and the potential for prolonged responses, most patients experience treatment failure. Neoantigen vaccines provide a potential complementary therapeutic strategy by increasing the immunogenic antigen load, which can lead to an increased tumor specific immune response. Further research is needed explore this treatment option in mesothelioma and technological advances are required to translate this concept into clinical practice.
Collapse
Affiliation(s)
- Linda Ye
- a Department of Medical Oncology , Sir Charles Gairdner Hospital , Nedlands , Australia
| | - Shaokang Ma
- b National Centre for Asbestos Related Disease , University of Western Australia , Nedlands , Australia
| | - Bruce W Robinson
- b National Centre for Asbestos Related Disease , University of Western Australia , Nedlands , Australia.,c Department of Respiratory Medicine , Sir Charles Gairdner Hospital , Nedlands , Australia
| | - Jenette Creaney
- b National Centre for Asbestos Related Disease , University of Western Australia , Nedlands , Australia.,c Department of Respiratory Medicine , Sir Charles Gairdner Hospital , Nedlands , Australia.,d Institute of Respiratory Health , University of Western Australia , Nedlands , Australia
| |
Collapse
|