1
|
Shi H, Yu TK, Johnson B, Selvamani SP, Zhuang L, Lee K, Klebe S, Smith S, Wong K, Chen K, Clark G, Rath EM, Pearson H, Ortega DG, Linton A, Kao S, Silveira P, Cheng YY. A combination of PD-1 and TIGIT immune checkpoint inhibitors elicits a strong anti-tumour response in mesothelioma. J Exp Clin Cancer Res 2025; 44:51. [PMID: 39939955 DOI: 10.1186/s13046-025-03314-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/02/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Finding effective and curative treatment for mesothelioma remains challenging. While the introduction of immunotherapy combinations using ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) have offered hope for some patients, a large proportion of mesothelioma cases, particularly the epithelial subtype, have minimal benefit from this. METHODS Our study was inspired by the results of the AdvanTG-105 phase I clinical trial, which showed partial response with anti-TIGIT/PD-1 treatment in two epithelioid mesothelioma patients. Here, we conducted a comprehensive in vivo experiment involving eight animal treatment groups administered with either PBS (control group), cisplatin/pemetrexed, anti-PD-1, anti-PD-1 + anti-CTLA-4, anti-TIGIT, anti-PD-1 + anti-TIGIT, anti-PD-1 + anti-CTLA-4 + anti-TIGIT, and cisplatin/pemetrexed + anti-PD-1 + anti-TIGIT. RESULTS Our results indicate that animals receiving anti-PD-1 + TIGIT exhibited a superior anti-tumour response, with 90% of the treatment group exhibiting an objective response, compared to 60%, 20% and 40% for the standard-of-care anti-PD-1 + CTLA-4, single-agent anti-PD-1 and cisplatin/pemetrexed treatment groups, respectively. Animals receiving anti-PD-1 + TIGIT displayed a significantly reduced average tumour size, with improved weight and survival rates, and fewer adverse effects than those receiving anti-PD-1 + CTLA-4 treatment. Anti-PD-1 + TIGIT-treated animals achieved complete tumour regression, with heightened effector CD8 + T cell and NK cell activity, remaining tumour-free for over 300 days without immune-related adverse events. After initial tumour elimination, anti-PD-1 + TIGIT-treated animals showed no tumour regrowth in the rechallenge experiment. CONCLUSION These findings provide rationale for the development of an anti-PD-1 + TIGIT combination immunotherapy trial for mesothelioma patients.
Collapse
Affiliation(s)
- Huaikai Shi
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia.
| | - Ta-Kun Yu
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
| | - Ben Johnson
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
| | - Sakthi Priya Selvamani
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
| | - Ling Zhuang
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
| | - Kenneth Lee
- Sydney Medical School, University of Sydney, Sydney, NSW, 2050, Australia
- SydPath, St. Vincent Health, Darlinghurst, Sydney, NSW, 2010, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, The University of Technology Sydney, Ultimo, NSW, 15 Broadway, Ultimo, NSW, 2007, Australia
| | - Sonja Klebe
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
- Pathology, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South, 5042, Australia
| | - Samuel Smith
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, NSW, 2050, Australia
| | - Kirby Wong
- Department of Radiology, Royal Prince Alfred Hospital, Sydney, 2050, Australia
| | - Kate Chen
- Dendritic Cell Research Group, ANZAC Research Institute, Sydney, NSW, 2139, Australia
| | - Georgina Clark
- Sydney Medical School, University of Sydney, Sydney, NSW, 2050, Australia
- Dendritic Cell Research Group, ANZAC Research Institute, Sydney, NSW, 2139, Australia
| | - Emma M Rath
- Victor Chang Cardiac Research Institute, 405 Liverpool St, Darlinghurst, NSW, 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Holly Pearson
- Single Cell Genomics Facility, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - David Gallego Ortega
- Single Cell Genomics Facility, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Technology Sydney, Ultimo, NSW, 2007, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW, 2033, Australia
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 380 Victoria St. Darlinghurst, Sydney, NSW, 2010, Australia
| | - Anthony Linton
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2050, Australia
| | - Steven Kao
- Asbestos and Dust Disease Research Institute (ADDRI), 3 Hospital Road, Sydney, NSW, Concord, NSW, 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2050, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW, 2033, Australia
| | - Pablo Silveira
- Dendritic Cell Research Group, ANZAC Research Institute, Sydney, NSW, 2139, Australia
| | - Yuen Yee Cheng
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, The University of Technology Sydney, Ultimo, NSW, 15 Broadway, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
2
|
Lim ZF, Wu X, Zhu L, Albandar H, Hafez M, Zhao C, Almubarak M, Smolkin M, Zheng H, Wen S, Ma PC. Quantitative peripheral live single T-cell dynamic polyfunctionality profiling predicts lung cancer checkpoint immunotherapy treatment response and clinical outcomes. Transl Lung Cancer Res 2024; 13:3323-3343. [PMID: 39830778 PMCID: PMC11736609 DOI: 10.21037/tlcr-24-260] [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: 03/22/2024] [Accepted: 08/23/2024] [Indexed: 01/22/2025]
Abstract
Background Predictive biomarkers for immune checkpoint inhibitors (ICIs), e.g., programmed death ligand-1 (PD-L1) tumor proportional score (TPS), remain limited in clinical applications. Predictive biomarkers that require invasive tumor biopsy procedures are practically challenging especially when longitudinal follow-up is required. Clinical utility of tissue-based PD-L1 TPS also becomes diluted when ICI is combined with chemotherapies. Peripheral single T-cell dynamic polyfunctionality profiling offers the opportunity to reveal rare T-cell subpopulations that are polyfunctional and responsible for the underlying ICI treatment molecular response that bulk biological assays cannot achieve. Here, we evaluated a novel live single-cell functional liquid biopsy cytokine profiling platform, IsoLight, as a potential predictive biomarker to track ICI treatment response and clinical outcomes in non-small cell lung cancer (NSCLC). Methods Peripheral blood mononuclear cell samples of 10 healthy donors and 10 NSCLC patients undergoing ICI-based therapies were collected longitudinally pre-/post-ICI treatment after ≥2 cycles under institutional review board (IRB)-approved protocols. Cancer blood samples were collected from unresectable advanced stage (III-IV) NSCLC patients. Clinical course and treatment response and survival outcomes were extracted from electronic health records, with treatment response assessed by treating oncologists based on RECIST. CD4+ and CD8+ T-cells were enriched magnetically and analyzed on the IsoLight platform. Single T-cells were captured in microchambers on IsoCode chips for proteomic immune cytokines profiling. Functional polyfunctionality data from 55,775 single cells were analyzed with IsoSpeak software, 2D- and 3D-t-distributed stochastic neighbor embedding (t-SNE) analysis, kappa coefficient, and Kaplan-Meier survival plots. P values ≤0.05 is considered statistically significant. Results Pre-treatment baseline polyfunctionality profiles could not differentiate NSCLC patients from healthy subjects, and could not differentiate ICI responders from non-responders. We found a statistically significant difference between responders and non-responders in CD8+ T-cells' changes in overall polyfunctionality (ΔPolyFx) (P=0.01) and polyfunctional strength index (ΔPSI) (P=0.006) in our dynamic pre-/post-treatment single cell measurements, both performing better than PD-L1 TPS alone (P=0.08). In the 3D-t-SNE analysis, subpopulations of post-treatment CD8+ T-cells in ICI responders displayed distinct immune cytokine profiles from those in pre-treatment cells. CD8+ T-cells ΔPolyFx and ΔPSI scores performed better than PD-L1 TPS in ICI response correlation. Moreover, combined PD-L1 strong TPS and ΔPSI >15 scores strongly correlated with early ICI response with a robust kappa coefficient of 1.0 (P=0.003), which was previously statistically established to indicate a perfect agreement between the prediction and actual response status. Interestingly, high CD4+ T-cells ΔPSI >5 was found to correlate with a strong trend of improved progression-free survival (3.9-fold) (10.8 vs. 2.8 months; P=0.07) and overall survival (3-fold) (34.5 vs. 11.5 months; P=0.09) in ICI-treated patients. Conclusions Our study nominates single peripheral T-cell polyfunctionality dynamics analysis to be a promising liquid biopsy platform to determine potential ICI predictive biomarker in NSCLC. It warrants further studies in larger prospective cohorts to validate the clinical utilities and to further optimize cancer immunotherapy.
Collapse
Affiliation(s)
- Zuan-Fu Lim
- Cancer Cell Biology Program, West Virginia University School of Medicine, West Virginia University, Morgantown, WV, USA
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Penn State College of Medicine, Penn State University, Hershey, PA, USA
| | - Xiaoliang Wu
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Penn State College of Medicine, Penn State University, Hershey, PA, USA
| | - Lin Zhu
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Penn State College of Medicine, Penn State University, Hershey, PA, USA
| | - Heidar Albandar
- WVU Cancer Institute, Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Maria Hafez
- WVU Cancer Institute, Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Chenchen Zhao
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Penn State College of Medicine, Penn State University, Hershey, PA, USA
| | - Mohammed Almubarak
- WVU Cancer Institute, Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV, USA
- Division of Hematology & Oncology, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV, USA
| | - Matthew Smolkin
- Department of Pathology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Hong Zheng
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Penn State College of Medicine, Penn State University, Hershey, PA, USA
| | - Sijin Wen
- Department of Biostatistics, School of Public Health, West Virginia University, Morgantown, WV, USA
| | - Patrick C. Ma
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Penn State College of Medicine, Penn State University, Hershey, PA, USA
| |
Collapse
|
3
|
Deboever N, Zhou N, McGrail DJ, Tomczak K, Oliva JL, Feldman HA, Parra E, Zhang J, Lee PP, Antonoff MB, Hofstetter WL, Mehran RJ, Rajaram R, Rice DC, Roth JA, Swisher SS, Vaporciyan AA, Altan M, Weissferdt A, Tsao AS, Haymaker CL, Sepesi B. Radiographic response to neoadjuvant therapy in pleural mesothelioma should serve as a guide for patient selection for cytoreductive operations. Front Oncol 2023; 13:1216999. [PMID: 37637041 PMCID: PMC10455934 DOI: 10.3389/fonc.2023.1216999] [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: 05/10/2023] [Accepted: 07/10/2023] [Indexed: 08/29/2023] Open
Abstract
Background Malignant pleural mesothelioma (MPM) is associated with poor prognosis despite advances in multimodal therapeutic strategies. While patients with resectable disease may benefit from added survival with oncologic resection, patient selection for mesothelioma operations often relies on both objective and subjective evaluation metrics. We sought to evaluate factors associated with improved overall survival (OS) in patients with mesothelioma who underwent macroscopic complete resection (MCR). Methods Patients with MPM who received neoadjuvant therapy and underwent MCR were identified in a prospectively maintained departmental database. Clinicopathologic, blood-based, and radiographic variables were collected and included in a Cox regression analysis (CRA). Response to neoadjuvant therapy was characterized by a change in tumor thickness from pretherapy to preoperative scans using the modified RECIST criteria. Results In this study, 99 patients met the inclusion criteria. The median age of the included patients was 64.7 years, who were predominantly men, had smoking and asbestos exposure, and who received neoadjuvant therapy. The median change in tumor thickness following neoadjuvant therapy was -16.5% (interquartile range of -49.7% to +14.2%). CRA demonstrated reduced OS associated with non-epithelioid histology [hazard ratio (HR): 3.06, 95% confidence interval (CI): 1.62-5.78, p < 0.001] and a response to neoadjuvant therapy inferior to the median (HR: 2.70, CI: 1.55-4.72, p < 0.001). Patients who responded poorly (below median) to neoadjuvant therapy had lower median survival (15.8 months compared to 38.2 months, p < 0.001). Conclusion Poor response to neoadjuvant therapy in patients with MPM is associated with poor outcomes even following maximum surgical cytoreduction and should warrant a patient-centered discussion regarding goals of care and may therefore help guide further therapeutic decisions.
Collapse
Affiliation(s)
- Nathaniel Deboever
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nicolas Zhou
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Daniel J. McGrail
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katarzyna Tomczak
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jacqueline L. Oliva
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hope A. Feldman
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Edwin Parra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Percy P. Lee
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mara B. Antonoff
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Wayne L. Hofstetter
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Reza J. Mehran
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ravi Rajaram
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - David C. Rice
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jack A. Roth
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stephen S. Swisher
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ara A. Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mehmet Altan
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Annikka Weissferdt
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anne S. Tsao
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cara L. Haymaker
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
4
|
Babar Q, Saeed A, Tabish TA, Sarwar M, Thorat ND. Targeting the tumor microenvironment: Potential strategy for cancer therapeutics. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166746. [PMID: 37160171 DOI: 10.1016/j.bbadis.2023.166746] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023]
Abstract
Cellular and stromal components including tumor cells, immune cells, mesenchymal cells, cancer-linked fibroblasts, and extracellular matrix, constituent tumor microenvironment (TME). TME plays a crucial role in reprogramming tumor initiation, uncontrolled proliferation, invasion and metastasis as well as response to therapeutic modalities. In recent years targeting the TME has developed as a potential strategy for treatment of cancer because of its life-threatening functions in restricting tumor development and modulating responses to standard-of-care medicines. Cold atmospheric plasma, oncolytic viral therapy, bacterial therapy, nano-vaccine, and repurposed pharmaceuticals with combination therapy, antiangiogenic drugs, and immunotherapies are among the most effective therapies directed by TME that have either been clinically authorized or are currently being studied. This article discusses above-mentioned therapies in light of targeting TME. We also cover problems related to the TME-targeted therapies, as well as future insights and practical uses in this rapidly growing field.
Collapse
Affiliation(s)
- Quratulain Babar
- Department of Biochemistry Government College University, Faisalabad, Pakistan
| | - Ayesha Saeed
- Department of Biochemistry Government College University, Faisalabad, Pakistan
| | - Tanveer A Tabish
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Mohsin Sarwar
- Department of Biochemistry University of Management and Technology, Lahore, Pakistan
| | - Nanasaheb D Thorat
- Department of Physics, Bernal Institute, Castletroy, Limerick V94T9PX, Ireland; Nuffield Department of Women's and Reproductive Health, John Radcliffe Hospital, Medical Sciences Division, University of Oxford, Oxford OX3 9DU, United Kingdom; Limerick Digital Cancer Research Centre (LDCRC) University of Limerick, Castletroy, Limerick V94T9PX, Ireland.
| |
Collapse
|
5
|
Perrino M, De Vincenzo F, Cordua N, Borea F, Aliprandi M, Santoro A, Zucali PA. Immunotherapy with immune checkpoint inhibitors and predictive biomarkers in malignant mesothelioma: Work still in progress. Front Immunol 2023; 14:1121557. [PMID: 36776840 PMCID: PMC9911663 DOI: 10.3389/fimmu.2023.1121557] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
Malignant mesothelioma (MM) is a rare and aggressive neoplasm, usually associated with a poor prognosis (5 years survival rate <10%). For unresectable disease, platinum and pemetrexed chemotherapy has been the only standard of care in first line for more than two decades, while no standard treatments have been approved in subsequent lines. Recently, immunotherapy has revolutionized the therapeutic landscape of MM. In fact, the combination of ipilimumab plus nivolumab has been approved in first line setting. Moreover, immune checkpoint inhibitors (ICIs) showed promising results also in second-third line setting after platinum-based chemotherapy. Unfortunately, approximately 20% of patients are primary refractory to ICIs and there is an urgent need for reliable biomarkers to improve patient's selection. Several biological and molecular features have been studied for this goal. In particular, histological subtype (recognized as prognostic factor for MM and predictive factor for chemotherapy response), programmed death ligand 1 (PD-L1) expression, and tumor mutational burden (widely hypothesized as predictive biomarkers for ICIs in several solid tumors) have been evaluated, but with unconclusive results. On the other hand, the deep analysis of tumor infiltrating microenvironment and the improvement in genomic profiling techniques has led to a better knowledge of several mechanisms underlying the MM biology and a greater or poorer immune activation. Consequentially, several potential biomarkers predictive of response to immunotherapy in patients with MM have been identified, also if all these elements need to be further investigated and prospectively validated. In this paper, the main evidences about clinical efficacy of ICIs in MM and the literature data about the most promising predictive biomarkers to immunotherapy are reviewed.
Collapse
Affiliation(s)
- Matteo Perrino
- Department of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy
| | - Fabio De Vincenzo
- Department of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy
| | - Nadia Cordua
- Department of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy
| | - Federica Borea
- Department of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Marta Aliprandi
- Department of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Armando Santoro
- Department of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Paolo Andrea Zucali
- Department of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy,Department of Biomedical Sciences, Humanitas University, Milan, Italy,*Correspondence: Paolo Andrea Zucali,
| |
Collapse
|
6
|
Song Y, Baxter SS, Dai L, Sanders C, Burkett S, Baugher RN, Mellott SD, Young TB, Lawhorn HE, Difilippantonio S, Karim B, Kadariya Y, Pinto LA, Testa JR, Shoemaker RH. Mesothelioma Mouse Models with Mixed Genomic States of Chromosome and Microsatellite Instability. Cancers (Basel) 2022; 14:3108. [PMID: 35804881 PMCID: PMC9264972 DOI: 10.3390/cancers14133108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/10/2022] [Accepted: 06/21/2022] [Indexed: 12/10/2022] Open
Abstract
Malignant mesothelioma (MMe) is a rare malignancy originating from the linings of the pleural, peritoneal and pericardial cavities. The best-defined risk factor is exposure to carcinogenic mineral fibers (e.g., asbestos). Genomic studies have revealed that the most frequent genetic lesions in human MMe are mutations in tumor suppressor genes. Several genetically engineered mouse models have been generated by introducing the same genetic lesions found in human MMe. However, most of these models require specialized breeding facilities and long-term exposure of mice to asbestos for MMe development. Thus, an alternative model with high tumor penetrance without asbestos is urgently needed. We characterized an orthotopic model using MMe cells derived from Cdkn2a+/-;Nf2+/- mice chronically injected with asbestos. These MMe cells were tumorigenic upon intraperitoneal injection. Moreover, MMe cells showed mixed chromosome and microsatellite instability, supporting the notion that genomic instability is relevant in MMe pathogenesis. In addition, microsatellite markers were detectable in the plasma of tumor-bearing mice, indicating a potential use for early cancer detection and monitoring the effects of interventions. This orthotopic model with rapid development of MMe without asbestos exposure represents genomic instability and specific molecular targets for therapeutic or preventive interventions to enable preclinical proof of concept for the intervention in an immunocompetent setting.
Collapse
Affiliation(s)
- Yurong Song
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (S.S.B.); (L.D.); (L.A.P.)
| | - Shaneen S. Baxter
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (S.S.B.); (L.D.); (L.A.P.)
| | - Lisheng Dai
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (S.S.B.); (L.D.); (L.A.P.)
| | - Chelsea Sanders
- Animal Research Technical Support of Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (C.S.); (S.D.)
| | - Sandra Burkett
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD 21702, USA;
| | - Ryan N. Baugher
- CLIA Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (R.N.B.); (S.D.M.); (T.B.Y.); (H.E.L.)
| | - Stephanie D. Mellott
- CLIA Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (R.N.B.); (S.D.M.); (T.B.Y.); (H.E.L.)
| | - Todd B. Young
- CLIA Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (R.N.B.); (S.D.M.); (T.B.Y.); (H.E.L.)
| | - Heidi E. Lawhorn
- CLIA Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (R.N.B.); (S.D.M.); (T.B.Y.); (H.E.L.)
| | - Simone Difilippantonio
- Animal Research Technical Support of Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (C.S.); (S.D.)
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA;
| | - Yuwaraj Kadariya
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (Y.K.); (J.R.T.)
| | - Ligia A. Pinto
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (S.S.B.); (L.D.); (L.A.P.)
| | - Joseph R. Testa
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (Y.K.); (J.R.T.)
| | - Robert H. Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD 20892, USA;
| |
Collapse
|