1
|
Kret ZS, Sweder RJ, Pollock R, Tinoco G. Potential Mechanisms for Immunotherapy Resistance in Adult Soft-Tissue Sarcoma. Target Oncol 2025:10.1007/s11523-025-01145-5. [PMID: 40289241 DOI: 10.1007/s11523-025-01145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2025] [Indexed: 04/30/2025]
Abstract
Soft-tissue sarcomas represent a diverse group of rare malignancies originating from mesenchymal tissue, accounting for less than 1% of adult cancers in the USA. With over 13,000 new cases and around 5350 deaths annually, patients with metastatic soft-tissue sarcomas face limited therapeutic options and an estimated median overall survival of 18 months. While immunotherapy has demonstrated effectiveness in several cancers, its application in soft-tissue sarcomas remains challenging owing to the tumors' largely "cold" immunological environment, characterized by low levels of tumor-infiltrating lymphocytes and a lack of soft-tissue sarcoma-specific biomarkers. This review examines potential mechanisms underlying immunotherapy resistance in soft-tissue sarcomas, including the complex interplay between innate and adaptive immunity, the tumor microenvironment, and the role of immune-related genes. Despite preliminary findings suggesting correlations between immune profiles and histological subtypes, consistent biomarkers for predicting immunotherapeutic responses across soft-tissue sarcoma types are absent. Emerging strategies focus on converting "cold" tumors to "hot" tumors, enhancing their susceptibility to immunologic activation. While research is ongoing, personalized treatment approaches may offer hope for overcoming the inherent heterogeneity and resistance seen in soft-tissue sarcomas, ultimately aiming to improve outcomes for affected patients.
Collapse
Affiliation(s)
- Zaina S Kret
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ryan J Sweder
- The Ohio State University College of Arts and Sciences and College of Medicine, Columbus, OH, USA
| | - Raphael Pollock
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Gabriel Tinoco
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 1800 Cannon Drive, 1240 Lincoln Tower, Columbus, OH, 43210, USA.
| |
Collapse
|
2
|
Gao T, He X, Wang J, Liu J, Hu X, Bai C, Yin S, Shi Y, Wang Y, Tan Z, Cao F, Li S, Shi YJ, Xue R, Li J, He Y, Li J, Lu H, Zhang H, Zhang L, Fang Z, Wang X, Liu M, Fu W, Tang L, Ye B, Fan Z, Xi JJ. Self-assembled patient-derived tumor-like cell clusters for personalized drug testing in diverse sarcomas. Cell Rep Med 2025; 6:101990. [PMID: 40054460 PMCID: PMC11970405 DOI: 10.1016/j.xcrm.2025.101990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/30/2024] [Accepted: 02/04/2025] [Indexed: 03/21/2025]
Abstract
Several patient-derived tumor models have emerged recently. However, soft tissue sarcomas (STSs) present a challenge in developing preclinical drug-testing models due to their non-epithelial and complex nature. Here, we report a model termed patient-derived tumor-like cell clusters (PTCs) derived from STS patients. PTCs result from the self-assembly and proliferation of mesenchymal stem cells (MSCs), epithelial cells, and immune cells, faithfully recapitulating the morphology and function of the original tumors. Through standardized culture and drug-response assessment protocols, PTCs facilitate personalized drug testing, evaluating hundreds of therapies within two weeks. Notably, PTCs exhibit 100% accuracy in distinguishing between complete or partial response and disease progression. We demonstrate the utility of PTCs in guiding chemotherapy selection for a patient with relapse and metastases following conventional therapy, who exhibited a positive response after non-conventional therapy identified through PTC. These findings underscore the potential of PTCs for prospective use in clinical decision-making regarding therapy selection.
Collapse
Affiliation(s)
- Tian Gao
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xinyu He
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Junyi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jiayong Liu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiongbing Hu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Chujie Bai
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shenyi Yin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; GeneX Health Co., Ltd., Beijing 100195, China
| | - Yunfei Shi
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yanmin Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Zhichao Tan
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fang Cao
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shu Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yan-Jie Shi
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ruifeng Xue
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Juan Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Yang He
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jiaxin Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China
| | - Huinan Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China
| | - Hanshuo Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; GeneX Health Co., Ltd., Beijing 100195, China
| | - Lu Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhiwei Fang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xinyu Wang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Mengmeng Liu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wenjun Fu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lei Tang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Buqing Ye
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Zhengfu Fan
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Jianzhong Jeff Xi
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China.
| |
Collapse
|
3
|
Blomain ES, Soudi S, Wang Z, Somani A, Subramanian A, Nouth SCL, Oladipo E, New C, Kenney DE, Nemat-Gorgani N, Kindler T, Avedian RS, Steffner RJ, Mohler DG, Hiniker SM, Chin AL, Kalbasi A, Binkley MS, Fried M, Gaida MM, van de Rijn M, Moding EJ. Evolutionary Pressures Shape Undifferentiated Pleomorphic Sarcoma Development and Radiotherapy Response. Cancer Res 2025; 85:1162-1174. [PMID: 39808162 DOI: 10.1158/0008-5472.can-24-3281] [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: 09/10/2024] [Revised: 10/19/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
Radiotherapy is an integral component in the treatment of many types of cancer, with approximately half of patients with cancer receiving radiotherapy. Systemic therapy applies pressure that can select for resistant tumor subpopulations, underscoring the importance of understanding how radiation impacts tumor evolution to improve treatment outcomes. We integrated temporal genomic profiling of 120 spatially distinct tumor regions from 20 patients with undifferentiated pleomorphic sarcomas (UPS), longitudinal circulating tumor DNA analysis, and evolutionary biology computational pipelines to study UPS evolution during tumorigenesis and in response to radiotherapy. Most unirradiated UPSs displayed initial linear evolution, followed by subsequent branching evolution with distinct mutational processes during early and late development. Metrics of genetic divergence between regions provided evidence of strong selection pressures during UPS development that further increased during radiotherapy. Subclone abundance changed after radiotherapy with subclone contraction tied to alterations in calcium signaling, and inhibiting calcium transporters radiosensitized sarcoma cells. Finally, circulating tumor DNA analysis accurately measured subclone abundance and enabled noninvasive monitoring of subclonal changes. These results demonstrate that radiation exerts selective pressures on UPSs and suggest that targeting radioresistant subclonal populations could improve outcomes after radiotherapy. Significance: Radiotherapy mediates tumor evolution by leading to the expansion of resistant subclonal cancer cell populations, indicating that developing approaches to target resistant subclones will be crucial to improve radiotherapy response.
Collapse
Affiliation(s)
- Erik S Blomain
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Shaghayegh Soudi
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Ziwei Wang
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Anish Somani
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Ajay Subramanian
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Serey C L Nouth
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Eniola Oladipo
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Christin New
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Deborah E Kenney
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Neda Nemat-Gorgani
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Thomas Kindler
- Department of Internal Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- TRON, Translational Oncology at the University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Raffi S Avedian
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Robert J Steffner
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - David G Mohler
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Susan M Hiniker
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Alexander L Chin
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Anusha Kalbasi
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Michael S Binkley
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Marius Fried
- Department of Internal Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias M Gaida
- TRON, Translational Oncology at the University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Pathology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matt van de Rijn
- Department of Pathology, Stanford University, Stanford, California
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, California
- Stanford Cancer Institute, Stanford University, Stanford, California
| |
Collapse
|
4
|
Pasquali S, Moura DS, Danks MR, Manasterski PJ, Zaffaroni N, Stacchiotti S, Mondaza-Hernandez JL, Kerrison WGJ, Martin-Broto J, Huang PH, Brunton VG. Preclinical models of soft tissue sarcomas - generation and applications to enhance translational research. Crit Rev Oncol Hematol 2025; 207:104621. [PMID: 39824369 DOI: 10.1016/j.critrevonc.2025.104621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025] Open
Abstract
Soft tissue sarcomas (STS) represent a large group of rare and ultra-rare tumors distinguished by unique morphological, molecular and clinical features. Patients with such rare cancers are generally underrepresented in clinical trials which has limited the introduction of new treatment options and subsequent improvement of patient outcomes. Preclinical models of STS that recapitulate the human disease can aid progress in identifying new effective treatments. However, due to the rarity of these tumors there are limited STS models available. Here we review the existing preclinical models of STS, including patient-derived cell lines and organoids, patient-derived xenografts and genetically engineered mouse models. We discuss the advantages and disadvantages of the different models and describe to what extent they have aided clinical translation. Finally, we consider what can be done in the future to enhance their predictivity in the preclinical setting.
Collapse
Affiliation(s)
- Sandro Pasquali
- Molecular Pharmacology, Department of Experimental Oncology, Fondazione IRCCS Instituto Nazionale dei Tumori di Milano, via G. Amadeo 42, Milano 20133, Italy
| | - David S Moura
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain; Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain; University Hospital General of Villalba, Madrid, Spain
| | - Molly R Danks
- Edinburgh Cancer Research, CRUK Scotland Centre, University of Edinburgh, Crewe Road South, Edinburgh EH4 2RX, UK
| | - Piotr J Manasterski
- Edinburgh Cancer Research, CRUK Scotland Centre, University of Edinburgh, Crewe Road South, Edinburgh EH4 2RX, UK
| | - Nadia Zaffaroni
- Molecular Pharmacology, Department of Experimental Oncology, Fondazione IRCCS Instituto Nazionale dei Tumori di Milano, via G. Amadeo 42, Milano 20133, Italy
| | - Silvia Stacchiotti
- Molecular Pharmacology, Department of Experimental Oncology, Fondazione IRCCS Instituto Nazionale dei Tumori di Milano, via G. Amadeo 42, Milano 20133, Italy
| | - Jose L Mondaza-Hernandez
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain; Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain; University Hospital General of Villalba, Madrid, Spain
| | - William G J Kerrison
- Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road Sutton, London, SM2 5NG, UK
| | - Javier Martin-Broto
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain; Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain; University Hospital General of Villalba, Madrid, Spain
| | - Paul H Huang
- Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road Sutton, London, SM2 5NG, UK
| | - Valerie G Brunton
- Edinburgh Cancer Research, CRUK Scotland Centre, University of Edinburgh, Crewe Road South, Edinburgh EH4 2RX, UK.
| |
Collapse
|
5
|
Ni H, Reitman ZJ, Zou W, Akhtar MN, Paul R, Huang M, Zhang D, Zheng H, Zhang R, Ma R, Ngo G, Zhang L, Diffenderfer ES, Motlagh SAO, Kim MM, Minn AJ, Dorsey JF, Foster JB, Metz J, Koumenis C, Kirsch DG, Gong Y, Fan Y. FLASH radiation reprograms lipid metabolism and macrophage immunity and sensitizes medulloblastoma to CAR-T cell therapy. NATURE CANCER 2025; 6:460-473. [PMID: 39910249 DOI: 10.1038/s43018-025-00905-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/07/2025] [Indexed: 02/07/2025]
Abstract
FLASH radiotherapy holds promise for treating solid tumors given the potential lower toxicity in normal tissues but its therapeutic effects on tumor immunity remain largely unknown. Using a genetically engineered mouse model of medulloblastoma, we show that FLASH radiation stimulates proinflammatory polarization in tumor macrophages. Single-cell transcriptome analysis shows that FLASH proton beam radiation skews macrophages toward proinflammatory phenotypes and increases T cell infiltration. Furthermore, FLASH radiation reduces peroxisome proliferator-activated receptor-γ (PPARγ) and arginase 1 expression and inhibits immunosuppressive macrophage polarization under stimulus-inducible conditions. Mechanistically, FLASH radiation abrogates lipid oxidase expression and oxidized low-density lipid generation to reduce PPARγ activity, while standard radiation induces reactive oxygen species-dependent PPARγ activation in macrophages. Notably, FLASH radiotherapy improves infiltration and activation of chimeric antigen receptor (CAR) T cells and sensitizes medulloblastoma to GD2 CAR-T cell therapy. Thus, FLASH radiotherapy reprograms macrophage lipid metabolism to reverse tumor immunosuppression. Combination FLASH-CAR radioimmunotherapy may offer exciting opportunities for solid tumor treatment.
Collapse
Affiliation(s)
- Haiwei Ni
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Zachary J Reitman
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Wei Zou
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Md Naushad Akhtar
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ritama Paul
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Menggui Huang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Hao Zheng
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruitao Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruiying Ma
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Gina Ngo
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric S Diffenderfer
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Michele M Kim
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Andy J Minn
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
- Mark Foundation Center for Immunotherapy, Immune Signaling, and Radiation, University of Pennsylvania, Philadelphia, PA, USA
| | - Jay F Dorsey
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica B Foster
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - James Metz
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
- Mark Foundation Center for Immunotherapy, Immune Signaling, and Radiation, University of Pennsylvania, Philadelphia, PA, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA.
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA.
- Mark Foundation Center for Immunotherapy, Immune Signaling, and Radiation, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Gao M, Zhang W, Li X, Li S, Wang W, Han P. LCAT in Cancer Biology: Embracing Epigenetic Regulation, Immune Interactions, and Therapeutic Implications. Int J Mol Sci 2025; 26:1453. [PMID: 40003919 PMCID: PMC11855027 DOI: 10.3390/ijms26041453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Lecithin cholesterol acyltransferase (LCAT) is a crucial enzyme in high-density lipoprotein (HDL) metabolism that is often dysregulated in cancers, affecting tumor growth and therapy response. We extensively studied LCAT expression in various malignancies, linking it to clinical outcomes and genetic/epigenetic alterations. We analyzed LCAT expression in multiple cancers and used the Cox regression model to correlate it with patient survival metrics, including overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI). We also examined the copy number variations (CNVs), single-nucleotide variations (SNVs), DNA methylation, and N6-methyladenosine (m6A) modifications of LCAT and their connections to tumor immune responses and drug sensitivity. LCAT expression varies among cancers and correlates with patient outcomes. Low expression is linked to poor prognosis in low-grade glioma (LGG) and liver hepatocellular carcinoma (LIHC), while high expression is associated with better outcomes in adrenocortical carcinoma (ACC) and colon adenocarcinoma (COAD). In kidney renal papillary cell carcinoma (KIRP) and uterine corpus endometrial carcinoma (UCEC), LCAT CNV and methylation levels are prognostic markers. LCAT interacts with m6A modifiers and immune molecules, suggesting a role in immune evasion and as a biomarker for immunotherapy response. LCAT expression correlates with chemotherapeutic drug IC50 values, indicating potential for predicting treatment response. In ACC and COAD, LCAT may promote tumor growth, while in LGG and LIHC, it may inhibit progression. LCAT expression and activity regulation could be a new cancer therapy target. As a key molecule linking lipid metabolism, immune modulation, and tumor progression, the potential of LCAT in cancer therapy is significant. Our findings provide new insights into the role of LCAT in cancer biology and support the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Manzhi Gao
- Department of Aerospace Hygiene, School of Aerospace Medicine, Air Force Medical University, Xi’an 710032, China; (M.G.); (W.Z.); (X.L.); (S.L.)
- Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi’an 710032, China
| | - Wentian Zhang
- Department of Aerospace Hygiene, School of Aerospace Medicine, Air Force Medical University, Xi’an 710032, China; (M.G.); (W.Z.); (X.L.); (S.L.)
- Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi’an 710032, China
| | - Xinxin Li
- Department of Aerospace Hygiene, School of Aerospace Medicine, Air Force Medical University, Xi’an 710032, China; (M.G.); (W.Z.); (X.L.); (S.L.)
- Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi’an 710032, China
| | - Sumin Li
- Department of Aerospace Hygiene, School of Aerospace Medicine, Air Force Medical University, Xi’an 710032, China; (M.G.); (W.Z.); (X.L.); (S.L.)
- Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi’an 710032, China
| | - Wenlan Wang
- Department of Aerospace Hygiene, School of Aerospace Medicine, Air Force Medical University, Xi’an 710032, China; (M.G.); (W.Z.); (X.L.); (S.L.)
- Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi’an 710032, China
| | - Peijun Han
- Department of Aerospace Hygiene, School of Aerospace Medicine, Air Force Medical University, Xi’an 710032, China; (M.G.); (W.Z.); (X.L.); (S.L.)
- Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi’an 710032, China
| |
Collapse
|
7
|
Sheng H, Luo Y, Zhong L, Wang Z, Sun Z, Gao X, He X, Zhu Z, Wu D, Sun J, Cao C. Antiangiogenic Treatment Facilitates the Abscopal Effect of Radiation Therapy Combined With Anti-PD-1 in Hepatocellular Carcinoma. Int J Radiat Oncol Biol Phys 2025; 121:534-546. [PMID: 39299549 DOI: 10.1016/j.ijrobp.2024.09.024] [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: 04/17/2024] [Revised: 09/02/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE Metastasis is one of the most important factors contributing to poor prognosis in hepatocellular carcinoma. Radiation therapy (RT), along with its induced abscopal effect, is a promising treatment for metastatic patients. However, the incidence of abscopal effect in clinical practice is rare, even when RT is combined with immune checkpoint inhibitors (ICIs). In this study, we aim to investigate the role of antiangiogenic treatment on the abscopal effect induced by RT + ICIs. METHODS AND MATERIALS Bilateral subcutaneous and orthotopic Hepa1-6 and Hep53.4 models were established and treated with different combination treatments. We evaluated changes in the immune microenvironment and vascular normalization by flow cytometry, T cell receptor sequencing, chemotactic gene array, enzyme linked immunosorbent assays, and immunofluorescence. RESULTS Our studies showed that antiangiogenic treatment with RT + ICIs increased the antitumor response of the unirradiated lesions. Mechanistically, the blockade of vascular endothelial receptor 2 (anti-VEGFR2) increased the activation and maturation of dendritic cells and promoted the production of CD8+ T cells in irradiated tumors. These CD8+ T cells were attracted by anti-VEGFR2-induced CCL5 secretion from M1 macrophages in unirradiated tumors. Besides that, anti-VEGFR2 enhanced the function of CD8+ T cells by reducing myeloid-derived suppressor cells and regulatory T cells. CONCLUSIONS This study demonstrated that the combination of antiangiogenic treatment with RT and ICIs enhanced the abscopal effects. The application of triple therapy and its induced abscopal effect may offer a novel therapeutic approach for hepatocellular carcinoma, particularly for cases with multiple metastatic lesions.
Collapse
Affiliation(s)
- Hailong Sheng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yongyi Luo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liting Zhong
- Department of Oncology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Oncology, Ganzhou People's Hospital, Ganzhou, China
| | - Zhiyi Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhichao Sun
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinna Gao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinrong He
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenru Zhu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dehua Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jingyuan Sun
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Chuanhui Cao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Zhang J, Peng Q, Fan J, Liu F, Chen H, Bi X, Yuan S, Jiang W, Pan T, Li K, Tan S, Chen P. Single-cell and spatial transcriptomics reveal SPP1-CD44 signaling drives primary resistance to immune checkpoint inhibitors in RCC. J Transl Med 2024; 22:1157. [PMID: 39736762 DOI: 10.1186/s12967-024-06018-5] [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: 06/10/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are a cornerstone therapy for advanced renal cell carcinoma (RCC). However, significant rates of primary resistance hinder their efficacy, and the underlying mechanisms remain poorly understood. This study aims to unravel the tumor-immune interactions and signaling pathways driving primary resistance to ICIs in RCC. METHODS We integrated single-cell RNA sequencing, spatial transcriptomics, and clinical sample analysis to investigate the tumor microenvironment and intercellular signaling. Advanced computational methods, including cell-cell communication networks, pseudotime trajectories, and gene set enrichment analysis (GSEA), were employed to uncover the underlying resistance mechanisms. RESULTS Compared to the sensitive group, the primary resistance group exhibited a significant increase in SPP1-CD44 signaling-mediated interactions between tumor cells and immune cells. These interactions disrupted antigen presentation in immune effector cells and suppressed key chemokine and cytokine pathways, thereby impairing effective immune responses. In contrast, the sensitive group showed more active antigen presentation and cytokine signaling, which facilitated stronger immune responses. Furthermore, the interaction between SPP1-secreting tumor cells and CD44-expressing exhausted CD8 + T cells activated the MAPK signaling pathway within CD8 + Tex cells, exacerbating T cell exhaustion and driving the development of ICI resistance in RCC. CONCLUSION Our findings reveal a potential mechanism by which SPP1-CD44 signaling mediates tumor-immune cell interactions leading to ICI resistance, providing a theoretical basis for targeting and disrupting this signaling to overcome primary resistance in RCC.
Collapse
Affiliation(s)
- Junfeng Zhang
- Department of Urology, Xinjiang Medical University Affiliated Cancer Hospital, Urumqi, China
| | - Qingyan Peng
- School of Nursing, Xinjiang Medical University, Urumqi, China
| | - Jin Fan
- Department of Oncology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, China
| | - Fuzhong Liu
- Cancer Institute, Xinjiang Medical University Affiliated Cancer Hospital, Urumqi, China
| | - Hongbo Chen
- Department of Urology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, China
| | - Xing Bi
- Department of Urology, Xinjiang Medical University Affiliated Cancer Hospital, Urumqi, China
| | - Shuai Yuan
- Department of Urology, Xinjiang Medical University Affiliated Cancer Hospital, Urumqi, China
| | - Wei Jiang
- Department of Urology, Xinjiang Medical University Affiliated Cancer Hospital, Urumqi, China
| | - Ting Pan
- Department of Urology, Xinjiang Medical University Affiliated Cancer Hospital, Urumqi, China
| | - Kailing Li
- Department of Urology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, China
| | - Sihai Tan
- Department of Pediatric, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, China.
| | - Peng Chen
- Department of Urology, Xinjiang Medical University Affiliated Cancer Hospital, Urumqi, China.
| |
Collapse
|
9
|
Gu Y, Feng Z, Xu X, Jin L. Identification of a novel immune-related gene signature by single-cell and bulk sequencing for the prediction of the immune landscape and prognosis of breast cancer. Cancer Cell Int 2024; 24:393. [PMID: 39627792 PMCID: PMC11613745 DOI: 10.1186/s12935-024-03589-7] [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: 07/24/2024] [Accepted: 11/26/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND As a common cause of cancer-related deaths in women, BRCA (breast cancer) shows complexity and requires precise biomarkers and treatment methods. This study delves into the molecular makeup of BRCA, focusing on immune profiles, molecular subtypes, gene expression and single-cell analysis. METHODS XCell was used to assess immune infiltration based on TCGA (the Cancer Genome Atlas) data and the clustering analysis was made. Differentially expressed genes were examined in distinct clusters, and the WGCNA (weighted correlation network analysis) was made to establish co-expression networks. The prognostic models were developed by Cox and LASSO-Cox regression. The clustering analysis, GSEA (Gene set enrichment analysis), GSVA (gene set variation analysis) and communication analysis of the single-cell dataset GSE161529 were performed to investigate the functional relevance. Real-time polymerase chain reaction (RT-PCR) was employed for evaluating gene expression. RESULTS The results revealed significant differences in immune cell infiltration between two clusters (C1 and C2). C2 had poorer survival outcomes, which was associated with higher expression of immune checkpoints PD1 and PD-L1. The gene modules identified via WGCNA were correlated with the immune-based subtypes. Then, a prognostic model comprising seven genes (ACSL1, ABCB5, XG, ADH4, OPN4, NPR3, NLGN1) was used to divide patients into high- and low-risk subgroups. The high-risk group had worse prognosis and higher scores of TIDE (Tumor Immune Dysfunction and Exclusion). The single-cell analysis depicted the immune landscape. Macrophages and endothelial cells exhibited higher AUCell scores. In cellular communication analysis, notably significant ligand-receptor interactions of HLA-DRA-> CD4 and TNFSF13B-> HLA-DPB1 were observed. The proportion of endothelial cells was correlated with risk scores. Finally, RT-PCR results illustrated the expression of seven genes in BRCA specimens. CONCLUSION The integrative analysis provides new insights into molecular complexities of BRCA. Immune profiles and gene signatures hold potential for improving stratification of BRCA patients and guiding the development of personalized immunotherapy strategies.
Collapse
Affiliation(s)
- Yanlin Gu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Soochow University, Jiangsu, China
| | - Zhengyang Feng
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Jiangsu, China
| | - Xiaoyan Xu
- Department of Operating Room, Traditional Chinese Medicine Hospital of Kunshan, Jiangsu, China
| | - Liyan Jin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Soochow University, Jiangsu, China.
| |
Collapse
|
10
|
Huo J, Wang Z, Zhao W, Chen M, Li H, He F, Tian X, Ma Y, Husanova F, Ma L, Ni Y, Ding H, Li W, Xu H. Investigating intra-tumoural heterogeneity and microenvironment diversity in primary cardiac angiosarcoma through single-cell RNA sequencing. Clin Transl Med 2024; 14:e70113. [PMID: 39658531 PMCID: PMC11631565 DOI: 10.1002/ctm2.70113] [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: 06/08/2024] [Revised: 11/04/2024] [Accepted: 11/16/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Primary cardiac angiosarcoma (PCAS) is a rare and aggressive heart tumour with limited treatment options and a poor prognosis. Understanding cellular heterogeneity and tumour microenvironment (TME) is crucial for the development of effective therapies. Here, we investigated the intratumoural heterogeneity and TME diversity of PCAS using single-cell RNA sequencing (scRNA-seq). METHODS We performed scRNA-seq analysis on tumour samples from four patients with PCAS, supplemented with multicolour immunohistochemistry for identification. We used scRNA-seq data from five normal cardiac tissue samples downloaded from public databases for comparative analyses. Bioinformatic analyses, including Cell Ranger, Seurat, Monocle2, hdWGCNA, SCENIC and NicheNet, were utilized to identify distinct cell populations, transcriptional patterns, and co-regulating gene modules. RESULTS Our analysis revealed significant intratumoural heterogeneity in PCAS driven by diverse biological processes such as protein synthesis, degradation, and RIG-I signalling inhibition. The SCENIC analysis identified three primary transcription factors' clusters (CEBPB, MYC and TAL1). T-cell subset analysis showed exhausted antigen-specific T-cells, complicating the efficacy of immune checkpoint blockade. Furthermore, we observed suppressive macrophages (SPP1+ and OLR1+) and reduced mitochondrial gene MT-RNR2 (MTRNR2L12) expression in TME-infiltrating cells, indicating impaired mitochondrial function. CONCLUSION This study elucidates the complex cellular landscape and immune microenvironment of PCAS, highlighting potential molecular targets for the development of novel therapies. These findings underscore the importance of a multifaceted therapeutic approach for addressing the challenges posed by PCAS's heterogeneity and immune evasion. KEY POINTS Insights into the heterogeneity and transcriptional patterns of sarcoma cells may explain the challenges in treating primary cardiac angiosarcoma (PCAS) using the current therapeutic modalities. Characterization of the immune microenvironment revealed significant immunosuppression mediated by specific myeloid cell populations (SPP1+ and OLR1+ macrophages). Identification of mitochondrial dysfunction in immune cells within the PCAS microenvironment, particularly the notable downregulation of the MTRNR2L12 protein, suggests a new avenue for therapeutic targeting.
Collapse
Affiliation(s)
- Jingyuan Huo
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Zhen Wang
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Wenting Zhao
- Department of CardiologySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Miao Chen
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Haoyang Li
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Fengpu He
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Xiao Tian
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Yaqi Ma
- Department of PathologySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Firyuza Husanova
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Liang Ma
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Yiming Ni
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Hongda Ding
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Weidong Li
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Hongfei Xu
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| |
Collapse
|
11
|
Mowery YM, Ballman KV, Hong AM, Schuetze SM, Wagner AJ, Monga V, Heise RS, Attia S, Choy E, Burgess MA, Bae S, Pryor DI, Van Tine BA, Tinoco G, Chmielowski B, Freeman C, Gronchi A, Meyer CF, Dickson MA, Hartner L, Davis LE, Powers BC, Moding EJ, Weinhold KJ, van de Rijn M, Brigman BE, Riedel RF, Kirsch DG. Safety and efficacy of pembrolizumab, radiation therapy, and surgery versus radiation therapy and surgery for stage III soft tissue sarcoma of the extremity (SU2C-SARC032): an open-label, randomised clinical trial. Lancet 2024; 404:2053-2064. [PMID: 39547252 PMCID: PMC11842127 DOI: 10.1016/s0140-6736(24)01812-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/05/2024] [Accepted: 08/28/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Approximately half of patients with localised, high-risk soft tissue sarcoma of the extremity develop metastases. We aimed to assess whether the addition of pembrolizumab to preoperative radiotherapy and surgery would improve disease-free survival. METHODS We completed an open-label, randomised clinical trial in patients with grade 2 or 3, stage III undifferentiated pleomorphic sarcoma or dedifferentiated or pleomorphic liposarcoma of the extremity and limb girdle. Patients were enrolled at 20 academic institutions in Australia, Canada, Italy, and the USA. Patients were randomly assigned to preoperative radiotherapy then surgery (control group) or preoperative pembrolizumab with radiotherapy (initiated 1-14 days after the first dose of pembrolizumab) then surgery and postoperative pembrolizumab (experimental group). Pembrolizumab (200 mg intravenously every 3 weeks) was administered as three neoadjuvant cycles (before, during, and after radiotherapy) and 14 or less adjuvant cycles. Primary endpoint was disease-free survival. This study is registered with ClincialTrials.gov (NCT03092323). FINDINGS Between Nov 18, 2017, and Nov 14, 2023, 143 participants were randomly assigned to treatment. A modified intention-to-treat analysis of 127 patients with median follow-up of 43 months showed that the experimental group (n=64) had significantly longer disease-free survival than the control group (n=63; log-rank one-sided p=0·035; hazard ratio [HR] 0·61; 90% CI 0·39-0·96). The 2-year disease-free survival increased by 15% with addition of pembrolizumab: 52% (90% CI 42-64) and 67% (90% CI 58-78) for the control and experimental groups, respectively. Disease-free survival was similarly improved with pembrolizumab for the intention-to-treat patient population (HR 0·61 [90% CI 0·39-0·95]). Grade 3 or higher adverse events occurred more frequently in the experimental group (56%) than the control group (31%). INTERPRETATION Addition of pembrolizumab to preoperative radiotherapy and surgery improves disease-free survival for patients with stage III undifferentiated pleomorphic sarcoma and pleomorphic or dedifferentiated liposarcoma of the extremity, which establishes a promising new treatment option for these patients. FUNDING Stand Up to Cancer and Merck Sharp & Dohme.
Collapse
Affiliation(s)
- Yvonne M Mowery
- Department of Radiation Oncology, UPMC Hillman Cancer Center-University of Pittsburgh, Pittsburgh, PA, USA; Department of Radiation Oncology, Duke University, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA
| | - Karla V Ballman
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Angela M Hong
- Department of Radiation Oncology, Chris O'Brien Lifehouse-Royal Prince Alfred Hospital, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Scott M Schuetze
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Andrew J Wagner
- Sarcoma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Varun Monga
- Division of Hematology and Oncology, Department of Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA; Division of Hematology and Oncology, Department of Medicine, UCSF Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Rachel S Heise
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Steven Attia
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Edwin Choy
- Division of Hematology Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Melissa A Burgess
- Division of Hematology and Oncology, Department of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Susie Bae
- Department of Medical Oncology, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - David I Pryor
- Department of Radiation Oncology, Princess Alexandra Hospital and Queensland University of Technology, Brisbane, QLD, Australia
| | - Brian A Van Tine
- Division of Oncology, Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Gabriel Tinoco
- Division of Medical Oncology, Department of Medicine, The Ohio State University, Arthur G James Comprehensive Cancer Center, Columbus, OH, USA
| | - Bartosz Chmielowski
- Division of Hematology and Medical Oncology, Department of Medicine, Jonsson Comprehensive Cancer Center, The University of California Los Angeles, Los Angeles, CA, USA
| | - Carolyn Freeman
- Department of Radiation Oncology, McGill University Health Centre, Montreal, QC, Canada
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Christian F Meyer
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Mark A Dickson
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lee Hartner
- Division of Hematology and Medical Oncology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Lara E Davis
- Division of Hematology and Medical Oncology, Department of Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Benjamin C Powers
- Division of Medical Oncology, Department of Internal Medicine, The University of Kansas Cancer Center, Kansas City, KS, USA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Palo Alto, CA, USA
| | - Kent J Weinhold
- Department of Surgery, Duke University, Durham, NC, USA; Department of Immunology, Duke University, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA
| | - Matt van de Rijn
- Department of Pathology, Stanford University, Palo Alto, CA, USA
| | - Brian E Brigman
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA
| | - Richard F Riedel
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
12
|
Li AY, Bu J, Xiao HN, Zhao ZY, Zhang JL, Yu B, Li H, Li JP, Xiao T. Two-step consensus clustering approach to immune cell infiltration: An integrated exploration and validation of prognostic and immune implications in sarcomas. Heliyon 2024; 10:e38253. [PMID: 39492897 PMCID: PMC11531637 DOI: 10.1016/j.heliyon.2024.e38253] [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: 03/29/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 11/05/2024] Open
Abstract
To conduct a comprehensive investigation of the sarcoma immune cell infiltration (ImmCI) patterns and tumoral microenvironment (TME). We utilized transcriptomic, clinical, and mutation data of sarcoma patients (training cohort) obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) server. Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) and Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) algorithms were applied to decipher the immune cell infiltration landscape and TME profiles of sarcomas. An unsupervised clustering method was utilized for classifying ImmCI clusters (initial clustering) and ImmCI-based differentially expressed gene-driven clusters (secondary clustering). Mortality rates and immune checkpoint gene levels was analyzed among the identified clusters. We calculated the ImmCI score through principal component analysis. The tumor immune dysfunction evaluation (TIDE) score was also employed to quantify immunotherapy efficacy between two ImmCI score groups. We further validated the biomarkers for ImmCI and gene-driven clusters via experimental verification and the accuracy of the ImmCI score in predicting survival outcomes and immunotherapy efficacy by external validation cohorts (testing cohort). We demonstrated that ImmCI cluster A and gene-driven cluster A, were beneficial prognostic biomarkers and indicators of immune checkpoint blockade response in sarcomas via in-silico and laboratory experiments. Additionally, the ImmCI score exhibited independent prognostic significance and was predictive of immunotherapy response. Our research underscores the clinical significance of ImmCI scores in identifying sarcoma patients likely to respond to immunotherapy.
Collapse
Affiliation(s)
- Ao-Yu Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jie Bu
- Department of Orthopedics, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hui-Ni Xiao
- Department of Gastroenterology, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Zi-Yue Zhao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jia-Lin Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Bin Yu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Hui Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jin-Ping Li
- Department of Orthopedics, Changsha Central Hospital, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Tao Xiao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| |
Collapse
|
13
|
Prasanna PGS, Ahmed MM, Hong JA, Coleman CN. Best practices and novel approaches for the preclinical development of drug-radiotherapy combinations for cancer treatment. Lancet Oncol 2024; 25:e501-e511. [PMID: 39362261 DOI: 10.1016/s1470-2045(24)00199-2] [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: 03/01/2024] [Revised: 03/31/2024] [Accepted: 04/04/2024] [Indexed: 10/05/2024]
Abstract
Drug-radiation combination therapy is a practical approach to improving clinical outcomes for many tumours. Unfortunately, most clinical combination studies combine drugs with radiotherapy empirically and do not exploit mechanistic synergy in cell death and the interconnectivity of molecular pathways of tumours or rationale for selecting the dose, fractionation, and schedule, which can result in suboptimal efficacy and exacerbation of toxic effects. However, opportunities exist to generate compelling preclinical evidence for combination therapies from fit-for-purpose translational studies for simulating the intended clinical study use scenarios with standardised preclinical assays and algorithms to evaluate complex molecular interactions and analysis of synergy before clinical research. Here, we analyse and discuss the core issues in the translation of preclinical data to enhance the relevance of preclinical assays, in vitro clonogenic survival along with apoptosis, in vivo tumour regression and growth delay assays, and toxicology of organs at risk without creating barriers to innovation and provide a synopsis of emerging areas in preclinical radiobiology.
Collapse
Affiliation(s)
- Pataje G S Prasanna
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Mansoor M Ahmed
- Division of Radiation Biology and Molecular Therapeutics, Department of Radiation Oncology, Albert Einstein College of Medicine, New York, NY, USA
| | - Julie A Hong
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - C Norman Coleman
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
14
|
Luthria K, Shah P, Caldwell B, Melms JC, Abuzaid S, Jakubikova V, Brodtman DZ, Bose S, Amin AD, Ho P, Biermann J, Tagore S, Ingham M, Schwartz GK, Izar B. Single-Cell Profiling of Sarcomas from Archival Tissue Reveals Programs Associated with Resistance to Immune Checkpoint Blockade. Clin Cancer Res 2024; 30:4530-4541. [PMID: 39083415 PMCID: PMC11443197 DOI: 10.1158/1078-0432.ccr-23-2976] [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: 09/28/2023] [Revised: 05/25/2024] [Accepted: 07/29/2024] [Indexed: 08/02/2024]
Abstract
PURPOSE Sarcoma encompasses a diverse group of cancers that are typically resistant to current therapies, including immune checkpoint blockade (ICB), and underlying mechanisms are poorly understood. The contexture of sarcomas limits generation of high-quality data using cutting-edge molecular profiling methods, such as single-cell RNA-sequencing, thus hampering progress in understanding these understudied cancers. EXPERIMENTAL DESIGN Here, we demonstrate feasibility of producing multimodal single-cell genomics and whole-genome sequencing data from frozen tissues, profiling 75,716 cell transcriptomes of five undifferentiated pleomorphic sarcoma and three intimal sarcoma samples, including paired specimens from two patients treated with ICB. RESULTS We find that genomic diversity decreases in patients with response to ICB, and, in unbiased analyses, identify cancer cell programs associated with therapy resistance. Although interactions of tumor-infiltrating T lymphocytes within the tumor ecosystem increase in ICB responders, clonal expansion of CD8+ T cells alone was insufficient to predict drug responses. CONCLUSIONS This study provides a framework for studying rare tumors and identifies salient and treatment-associated cancer cell intrinsic and tumor microenvironmental features in sarcomas.
Collapse
Affiliation(s)
- Karan Luthria
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - Parin Shah
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Division of Hematology/Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Blake Caldwell
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - Johannes C Melms
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Division of Hematology/Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York
| | - Sinan Abuzaid
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Division of Hematology/Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Viktoria Jakubikova
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Division of Hematology/Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - D Zack Brodtman
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Division of Hematology/Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Sminu Bose
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Division of Hematology/Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Amit Dipak Amin
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Division of Hematology/Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York
| | - Patricia Ho
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York
| | - Jana Biermann
- Division of Hematology/Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Somnath Tagore
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York
| | - Matthew Ingham
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Division of Hematology/Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Gary K Schwartz
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Division of Hematology/Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Benjamin Izar
- Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
- Division of Hematology/Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
15
|
Jagodinsky JC, Vera JM, Jin WJ, Shea AG, Clark PA, Sriramaneni RN, Havighurst TC, Chakravarthy I, Allawi RH, Kim K, Harari PM, Sondel PM, Newton MA, Crittenden MR, Gough MJ, Miller JR, Ong IM, Morris ZS. Intratumoral radiation dose heterogeneity augments antitumor immunity in mice and primes responses to checkpoint blockade. Sci Transl Med 2024; 16:eadk0642. [PMID: 39292804 PMCID: PMC11522033 DOI: 10.1126/scitranslmed.adk0642] [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: 07/31/2023] [Revised: 04/03/2024] [Accepted: 08/08/2024] [Indexed: 09/20/2024]
Abstract
Radiation therapy (RT) activates multiple immunologic effects in the tumor microenvironment (TME), with diverse dose-response relationships observed. We hypothesized that, in contrast with homogeneous RT, a heterogeneous RT dose would simultaneously optimize activation of multiple immunogenic effects in a single TME, resulting in a more effective antitumor immune response. Using high-dose-rate brachytherapy, we treated mice bearing syngeneic tumors with a single fraction of heterogeneous RT at a dose ranging from 2 to 30 gray. When combined with dual immune checkpoint inhibition in murine models, heterogeneous RT generated more potent antitumor responses in distant, nonirradiated tumors compared with any homogeneous dose. The antitumor effect after heterogeneous RT required CD4 and CD8 T cells and low-dose RT to a portion of the tumor. At the 3-day post-RT time point, dose heterogeneity imprinted the targeted TME with spatial differences in immune-related gene expression, antigen presentation, and susceptibility of tumor cells to immune-mediated destruction. At a later 10-day post-RT time point, high-, moderate-, or low-RT-dose regions demonstrated distinct infiltrating immune cell populations. This was associated with an increase in the expression of effector-associated cytokines in circulating CD8 T cells. Consistent with enhanced adaptive immune priming, heterogeneous RT promoted clonal expansion of effector CD8 T cells. These findings illuminate the breadth of dose-dependent effects of RT on the TME and the capacity of heterogeneous RT to promote antitumor immunity when combined with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Justin C. Jagodinsky
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Jessica M. Vera
- Department of Statistics and Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
- Sage Bionetworks, 2901 Third Ave. Suite 330, Seattle, WA 98121, USA
| | - Won Jong Jin
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Amanda G. Shea
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Paul A. Clark
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Raghava N. Sriramaneni
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Thomas C. Havighurst
- Department of Statistics and Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Ishan Chakravarthy
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Raad H. Allawi
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - KyungMann Kim
- Department of Statistics and Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Paul M. Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Paul M. Sondel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Michael A. Newton
- Department of Statistics and Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Marka R. Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR 97213, USA
- Oregon Clinic, Portland, OR 97232, USA
| | - Michael J. Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR 97213, USA
| | - Jessica R. Miller
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Irene M. Ong
- Department of Statistics and Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
16
|
Su C, Kent CL, Pierpoint M, Floyd W, Luo L, Williams NT, Ma Y, Peng B, Lazarides AL, Subramanian A, Himes JE, Perez VM, Hernansaiz-Ballesteros RD, Roche KE, Modliszewski JL, Selitsky SR, Shinohara ML, Wisdom AJ, Moding EJ, Mowery YM, Kirsch DG. Enhancing radiotherapy response via intratumoral injection of a TLR9 agonist in autochthonous murine sarcomas. JCI Insight 2024; 9:e178767. [PMID: 39133651 PMCID: PMC11383182 DOI: 10.1172/jci.insight.178767] [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: 01/03/2024] [Accepted: 06/11/2024] [Indexed: 08/21/2024] Open
Abstract
Radiation therapy (RT) is frequently used to treat cancers, including soft-tissue sarcomas. Prior studies established that the toll-like receptor 9 (TLR9) agonist cytosine-phosphate-guanine oligodeoxynucleotide (CpG) enhances the response to RT in transplanted tumors, but the mechanisms of this enhancement remain unclear. Here, we used CRISPR/Cas9 and the chemical carcinogen 3-methylcholanthrene (MCA) to generate autochthonous soft-tissue sarcomas with high tumor mutation burden. Treatment with a single fraction of 20 Gy RT and 2 doses of CpG significantly enhanced tumor response, which was abrogated by genetic or immunodepletion of CD8+ T cells. To characterize the immune response to CpG+RT, we performed bulk RNA-Seq, single-cell RNA-Seq, and mass cytometry. Sarcomas treated with 20 Gy and CpG demonstrated increased CD8 T cells expressing markers associated with activation and proliferation, such as Granzyme B, Ki-67, and IFN-γ. CpG+RT also upregulated antigen presentation pathways on myeloid cells. Furthermore, in sarcomas treated with CpG+RT, TCR clonality analysis suggests an increase in clonal T cell dominance. Collectively, these findings demonstrate that CpG+RT significantly delays tumor growth in a CD8 T cell-dependent manner. These results provide a strong rationale for clinical trials evaluating CpG or other TLR9 agonists with RT in patients with soft-tissue sarcoma.
Collapse
Affiliation(s)
- Chang Su
- Department of Pharmacology and Cancer Biology and
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Collin L Kent
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Matthew Pierpoint
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Warren Floyd
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Nerissa T Williams
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Brian Peng
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Alexander L Lazarides
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Ajay Subramanian
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Jonathon E Himes
- Department of Pharmacology and Cancer Biology and
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | - Kimberly E Roche
- Tempus AI Inc., Durham, North Carolina, USA
- QuantBio LLC, Durham, North Carolina, USA
| | - Jennifer L Modliszewski
- QuantBio LLC, Durham, North Carolina, USA
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Sara R Selitsky
- Tempus AI Inc., Durham, North Carolina, USA
- QuantBio LLC, Durham, North Carolina, USA
| | - Mari L Shinohara
- Department of Integrative Immunology
- Department of Molecular Genetics and Microbiology, and
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Amy J Wisdom
- Harvard Radiation Oncology Program, Boston, Massachusetts, USA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
- Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Yvonne M Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David G Kirsch
- Department of Pharmacology and Cancer Biology and
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Radiation Oncology and
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Rickard AG, Mowery YM, Bassil A, Rouse DC, Williams NT, Charity T, Belloni R, Crouch B, Ramanujam N, Stevenson D, Castillo R, Blocker S, Epel B, Kotecha M, Palmer GM. Evaluating Tumor Hypoxia Radiosensitization Via Electron Paramagnetic Resonance Oxygen Imaging (EPROI). Mol Imaging Biol 2024; 26:435-447. [PMID: 37721686 DOI: 10.1007/s11307-023-01855-0] [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: 05/03/2023] [Revised: 07/30/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023]
Abstract
PURPOSE Tumor hypoxia contributes to aggressive phenotypes and diminished therapeutic responses to radiation therapy (RT) with hypoxic tissue being 3-fold less radiosensitive than normoxic tissue. A major challenge in implementing hypoxic radiosensitizers is the lack of a high-resolution imaging modality that directly quantifies tissue-oxygen. The electron paramagnetic resonance oxygen-imager (EPROI) was used to quantify tumor oxygenation in two murine tumor models: E0771 syngeneic transplant breast cancers and primary p53/MCA soft tissue sarcomas, with the latter autochthonous model better recapitulating the tumor microenvironment in human malignancies. We hypothesized that tumor hypoxia differs between these models. We also aimed to quantify the absolute change in tumor hypoxia induced by the mitochondrial inhibitor papaverine (PPV) and its effect on RT response. PROCEDURES Tumor oxygenation was characterized in E0771 and primary p53/MCA sarcomas via EPROI, with the former model also being quantified indirectly via diffuse reflectance spectroscopy (DRS). After confirming PPV's effect on hypoxic fraction (via EPROI), we compared the effect of 0 versus 2 mg/kg PPV prior to 20 Gy on tumor growth delay and survival. RESULTS Hypoxic sarcomas were more radioresistant than normoxic sarcomas (p=0.0057, 2-way ANOVA), and high baseline hypoxic fraction was a significant (p=0.0063, Cox Regression Model) hazard in survivability regardless of treatment. Pre-treatment with PPV before RT did not radiosensitize tumors in the sarcoma or E0771 model. In the sarcoma model, EPROI successfully identified baseline hypoxic tumors. DRS quantification of total hemoglobin, saturated hemoglobin, changes in mitochondrial potential and glucose uptake showed no significant difference in E0771 tumors pre- and post-PPV. CONCLUSION EPROI provides 3D high-resolution pO2 quantification; EPR is better suited than DRS to characterize tumor hypoxia. PPV did not radiosensitize E0771 tumors nor p53/MCA sarcomas, which may be related to the complex pattern of vasculature in each tumor. Additionally, understanding model-dependent tumor hypoxia will provide a much-needed foundation for future therapeutic studies with hypoxic radiosensitizers.
Collapse
Affiliation(s)
- Ashlyn G Rickard
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Yvonne M Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA.
- Department of Head and Neck Surgery & Communication Sciences, Duke University Medical Center, Durham, NC, USA.
| | - Alex Bassil
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Douglas C Rouse
- Division of Laboratory Animal Resources, Duke University School of Medicine, Durham, NC, USA
| | - Nerissa T Williams
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Theresa Charity
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Rafaela Belloni
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Brian Crouch
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nimmi Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Rico Castillo
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Stephanie Blocker
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Boris Epel
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
- O2M Technologies LLC, Chicago, IL, USA
| | | | - Gregory M Palmer
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
18
|
Patel R, Cooper DE, Kadakia KT, Allen A, Duan L, Luo L, Williams NT, Liu X, Locasale JW, Kirsch DG. Targeting glutamine metabolism improves sarcoma response to radiation therapy in vivo. Commun Biol 2024; 7:608. [PMID: 38769385 PMCID: PMC11106276 DOI: 10.1038/s42003-024-06262-x] [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: 07/17/2023] [Accepted: 04/29/2024] [Indexed: 05/22/2024] Open
Abstract
Diverse tumor metabolic phenotypes are influenced by the environment and genetic lesions. Whether these phenotypes extend to rhabdomyosarcoma (RMS) and how they might be leveraged to design new therapeutic approaches remains an open question. Thus, we utilized a Pax7Cre-ER-T2/+; NrasLSL-G12D/+; p53fl/fl (P7NP) murine model of sarcoma with mutations that most frequently occur in human embryonal RMS. To study metabolism, we infuse 13C-labeled glucose or glutamine into mice with sarcomas and show that sarcomas consume more glucose and glutamine than healthy muscle tissue. However, we reveal a marked shift from glucose consumption to glutamine metabolism after radiation therapy (RT). In addition, we show that inhibiting glutamine, either through genetic deletion of glutaminase (Gls1) or through pharmacological inhibition of glutaminase, leads to significant radiosensitization in vivo. This causes a significant increase in overall survival for mice with Gls1-deficient compared to Gls1-proficient sarcomas. Finally, Gls1-deficient sarcomas post-RT elevate levels of proteins involved in natural killer cell and interferon alpha/gamma responses, suggesting a possible role of innate immunity in the radiosensitization of Gls1-deficient sarcomas. Thus, our results indicate that glutamine contributes to radiation response in a mouse model of RMS.
Collapse
Affiliation(s)
- Rutulkumar Patel
- Department of Radiation Oncology, Baylor College of Medicine, 7200 Cambridge St, Houston, TX, 77030, USA
| | - Daniel E Cooper
- Department of Radiation Oncology, Duke University, Box 3085, Duke Cancer Center, Medicine Circle, Durham, NC, 27710, USA
| | - Kushal T Kadakia
- Department of Radiation Oncology, Duke University, Box 3085, Duke Cancer Center, Medicine Circle, Durham, NC, 27710, USA
| | - Annamarie Allen
- Department of Pharmacology and Cancer Biology, Duke University, Box 3813, 308 Research Drive, Durham, NC, 27710, USA
| | - Likun Duan
- Department of Pharmacology and Cancer Biology, Duke University, Box 3813, 308 Research Drive, Durham, NC, 27710, USA
- Department of Molecular and Structural Biochemistry, NC State University, Box 7622, 128 Polk Hall, Raleigh, NC, 27695, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University, Box 3085, Duke Cancer Center, Medicine Circle, Durham, NC, 27710, USA
| | - Nerissa T Williams
- Department of Radiation Oncology, Duke University, Box 3085, Duke Cancer Center, Medicine Circle, Durham, NC, 27710, USA
| | - Xiaojing Liu
- Department of Molecular and Structural Biochemistry, NC State University, Box 7622, 128 Polk Hall, Raleigh, NC, 27695, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Box 3813, 308 Research Drive, Durham, NC, 27710, USA
- Department of Molecular and Structural Biochemistry, NC State University, Box 7622, 128 Polk Hall, Raleigh, NC, 27695, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University, Box 3085, Duke Cancer Center, Medicine Circle, Durham, NC, 27710, USA.
- Department of Pharmacology and Cancer Biology, Duke University, Box 3813, 308 Research Drive, Durham, NC, 27710, USA.
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, M5G 2M9, Canada.
- Department of Radiation Oncology, University of Toronto, 149 College Street, Suite 504, Toronto, ON, M5T 1P5, Canada.
- Department of Medical Biophysics, University of Toronto, 101 College Street, Room 15-701, Toronto, ON, M5G 1L7, Canada.
| |
Collapse
|
19
|
Lu Y, Chen D, Wang B, Chai W, Yan M, Chen Y, Zhan Y, Yang R, Zhou E, Dai S, Li Y, Dong R, Zheng B. Single-cell landscape of undifferentiated pleomorphic sarcoma. Oncogene 2024; 43:1353-1368. [PMID: 38459120 DOI: 10.1038/s41388-024-03001-8] [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: 08/24/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/10/2024]
Abstract
Undifferentiated pleomorphic sarcoma (UPS) is a highly aggressive malignant soft tissue tumor with a poor prognosis; however, the identity and heterogeneity of tumor populations remain elusive. Here, eight major cell clusters were identified through the RNA sequencing of 79,569 individual cells of UPS. UPS originates from mesenchymal stem cells (MSCs) and features undifferentiated subclusters. UPS subclusters were predicted to exist in two bulk RNA datasets, and had a prognostic value in The Cancer Genome Atlas (TCGA) dataset. The functional heterogeneity of malignant UPS cells and the immune microenvironment were characterized. Additionally, the fused cells were innovatively detected by expressing both monocyte/macrophage markers and other subcluster-associated genes. Based on the ligand-receptor interaction analysis, cellular interactions with epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) were abundant. Furthermore, 73% of patients with UPS (48/66) showed positive EGFR expression, which was associated with a poor prognosis. EGFR blockade with cetuximab inhibited tumor growth in a patient-derived xenograft model. Our transcriptomic studies delineate the landscape of UPS intratumor heterogeneity and serve as a foundational resource for further discovery and therapeutic exploration.
Collapse
Affiliation(s)
- Yifei Lu
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Deqian Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Bingnan Wang
- Department of Musculoskeletal Oncology, Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wenjun Chai
- Department of Animal Experimental Center, Fudan University Shanghai Cancer Center, Shanghai, 201102, China
| | - Mingxia Yan
- Department of Animal Experimental Center, Fudan University Shanghai Cancer Center, Shanghai, 201102, China
| | - Yong Chen
- Department of Musculoskeletal Oncology, Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yong Zhan
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Ran Yang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Enqing Zhou
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Shuyang Dai
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Yi Li
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China.
| | - Biqiang Zheng
- Department of Musculoskeletal Oncology, Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
20
|
Abedini-Nassab R, Taheri F, Emamgholizadeh A, Naderi-Manesh H. Single-Cell RNA Sequencing in Organ and Cell Transplantation. BIOSENSORS 2024; 14:189. [PMID: 38667182 PMCID: PMC11048310 DOI: 10.3390/bios14040189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024]
Abstract
Single-cell RNA sequencing is a high-throughput novel method that provides transcriptional profiling of individual cells within biological samples. This method typically uses microfluidics systems to uncover the complex intercellular communication networks and biological pathways buried within highly heterogeneous cell populations in tissues. One important application of this technology sits in the fields of organ and stem cell transplantation, where complications such as graft rejection and other post-transplantation life-threatening issues may occur. In this review, we first focus on research in which single-cell RNA sequencing is used to study the transcriptional profile of transplanted tissues. This technology enables the analysis of the donor and recipient cells and identifies cell types and states associated with transplant complications and pathologies. We also review the use of single-cell RNA sequencing in stem cell implantation. This method enables studying the heterogeneity of normal and pathological stem cells and the heterogeneity in cell populations. With their remarkably rapid pace, the single-cell RNA sequencing methodologies will potentially result in breakthroughs in clinical transplantation in the coming years.
Collapse
Affiliation(s)
- Roozbeh Abedini-Nassab
- Faculty of Mechanical Engineering, Tarbiat Modares University, Tehran P.O. Box 1411944961, Iran
| | - Fatemeh Taheri
- Biomedical Engineering Department, University of Neyshabur, Neyshabur P.O. Box 9319774446, Iran
| | - Ali Emamgholizadeh
- Faculty of Mechanical Engineering, Tarbiat Modares University, Tehran P.O. Box 1411944961, Iran
| | - Hossein Naderi-Manesh
- Department of Nanobiotechnology, Faculty of Bioscience, Tarbiat Modares University, Tehran P.O. Box 1411944961, Iran;
- Department of Biophysics, Faculty of Bioscience, Tarbiat Modares University, Tehran P.O. Box 1411944961, Iran
| |
Collapse
|
21
|
Su C, Kim SK, Wang CX, Kirsch DG, Monjazeb AM. Radiotherapy Combined with Intralesional Immunostimulatory Agents for Soft Tissue Sarcomas. Semin Radiat Oncol 2024; 34:243-257. [PMID: 38508788 PMCID: PMC11216412 DOI: 10.1016/j.semradonc.2024.01.001] [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] [Indexed: 03/22/2024]
Abstract
Immunotherapy has shifted the treatment paradigm for many types of cancer. Unfortunately, the most commonly used immunotherapies, such as immune checkpoint inhibitors (ICI), have yielded limited benefit for most types of soft tissue sarcoma (STS). Radiotherapy (RT) is a mainstay of sarcoma therapy and can induce immune modulatory effects. Combining immunotherapy and RT in STS may be a promising strategy to improve sarcoma response to RT and increase the efficacy of immunotherapy. Most combination strategies have employed immunotherapies, such as ICI, that derepress immune suppressive networks. These have yielded only modest results, possibly due to the limited immune stimulatory effects of RT. Combining RT with immune stimulatory agents has yielded promising preclinical and clinical results but can be limited by the toxic nature of systemic administration of immune stimulants. Using intralesional immune stimulants may generate stronger RT immune modulation and less systemic toxicity, which may be a feasible strategy in accessible tumors such as STS. In this review, we summarize the immune modulatory effects of RT, the mechanism of action of various immune stimulants, including toll-like receptor agonists, and data for combinatorial strategies utilizing these agents.
Collapse
Affiliation(s)
- Chang Su
- Department of Radiation Oncology, Duke University, Durham, NC
| | - Soo Kyoung Kim
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, UC Davis Health, Davis, CA
| | - Charles X Wang
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, UC Davis Health, Davis, CA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University, Durham, NC; Department of Radiation Oncology, Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Arta M Monjazeb
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, UC Davis Health, Davis, CA.
| |
Collapse
|
22
|
Hendrickson PG, Oristian KM, Browne MR, Luo L, Ma Y, Cardona DM, Nash JO, Ballester PL, Davidson S, Shlien A, Linardic CM, Kirsch DG. Spontaneous expression of the CIC::DUX4 fusion oncoprotein from a conditional allele potently drives sarcoma formation in genetically engineered mice. Oncogene 2024; 43:1223-1230. [PMID: 38413794 PMCID: PMC11027086 DOI: 10.1038/s41388-024-02984-8] [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: 10/24/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 02/29/2024]
Abstract
CIC::DUX4 sarcoma (CDS) is a rare but highly aggressive undifferentiated small round cell sarcoma driven by a fusion between the tumor suppressor Capicua (CIC) and DUX4. Currently, there are no effective treatments and efforts to identify and translate better therapies are limited by the scarcity of patient tumor samples and cell lines. To address this limitation, we generated three genetically engineered mouse models of CDS (Ch7CDS, Ai9CDS, and TOPCDS). Remarkably, chimeric mice from all three conditional models developed spontaneous soft tissue tumors and disseminated disease in the absence of Cre-recombinase. The penetrance of spontaneous (Cre-independent) tumor formation was complete irrespective of bi-allelic Cic function and the distance between adjacent loxP sites. Characterization of soft tissue and presumed metastatic tumors showed that they consistently expressed the CIC::DUX4 fusion protein and many downstream markers of the disease credentialing the models as CDS. In addition, tumor-derived cell lines were generated and ChIP-seq was preformed to map fusion-gene specific binding using an N-terminal HA epitope tag. These datasets, along with paired H3K27ac ChIP-sequencing maps, validate CIC::DUX4 as a neomorphic transcriptional activator. Moreover, they are consistent with a model where ETS family transcription factors are cooperative and redundant drivers of the core regulatory circuitry in CDS.
Collapse
Affiliation(s)
- Peter G Hendrickson
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | | | - MaKenna R Browne
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
- Developmental and Stem Cell Biology Program, Duke University Medical Center, Durham, NC, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Diana M Cardona
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Joshua O Nash
- Program in Genetics and Genome Biology, The Hospital for Sick Children (SickKids), University of Toronto, Toronto, ON, Canada
- Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Pedro L Ballester
- Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Scott Davidson
- Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Adam Shlien
- Program in Genetics and Genome Biology, The Hospital for Sick Children (SickKids), University of Toronto, Toronto, ON, Canada
- Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Corinne M Linardic
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA.
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA.
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
23
|
Wisdom AJ, Barker CA, Chang JY, Demaria S, Formenti S, Grassberger C, Gregucci F, Hoppe BS, Kirsch DG, Marciscano AE, Mayadev J, Mouw KW, Palta M, Wu CC, Jabbour SK, Schoenfeld JD. The Next Chapter in Immunotherapy and Radiation Combination Therapy: Cancer-Specific Perspectives. Int J Radiat Oncol Biol Phys 2024; 118:1404-1421. [PMID: 38184173 DOI: 10.1016/j.ijrobp.2023.12.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/20/2023] [Accepted: 12/30/2023] [Indexed: 01/08/2024]
Abstract
Immunotherapeutic agents have revolutionized cancer treatment over the past decade. However, most patients fail to respond to immunotherapy alone. A growing body of preclinical studies highlights the potential for synergy between radiation therapy and immunotherapy, but the outcomes of clinical studies have been mixed. This review summarizes the current state of immunotherapy and radiation combination therapy across cancers, highlighting existing challenges and promising areas for future investigation.
Collapse
Affiliation(s)
- Amy J Wisdom
- Harvard Radiation Oncology Program, Boston, Massachusetts
| | - Christopher A Barker
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joe Y Chang
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Clemens Grassberger
- Department of Radiation Oncology, University of Washington, Fred Hutch Cancer Center, Seattle, Washington
| | - Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Bradford S Hoppe
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida
| | - David G Kirsch
- Department of Radiation Oncology, University of Toronto, Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ariel E Marciscano
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jyoti Mayadev
- Department of Radiation Oncology, UC San Diego School of Medicine, San Diego, California
| | - Kent W Mouw
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Manisha Palta
- Department of Radiation Oncology, Duke Cancer Center, Durham, North Carolina
| | - Cheng-Chia Wu
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
| | - Jonathan D Schoenfeld
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
24
|
Ewongwo A, Hui C, Moding EJ. Opportunity in Complexity: Harnessing Molecular Biomarkers and Liquid Biopsies for Personalized Sarcoma Care. Semin Radiat Oncol 2024; 34:195-206. [PMID: 38508784 DOI: 10.1016/j.semradonc.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Due to their rarity and complexity, sarcomas represent a substantial therapeutic challenge. However, the incredible diversity within and across sarcoma subtypes presents an opportunity for personalized care to maximize efficacy and limit toxicity. A deeper understanding of the molecular alterations that drive sarcoma development and treatment response has paved the way for molecular biomarkers to shape sarcoma treatment. Genetic, transcriptomic, and protein biomarkers have become critical tools for diagnosis, prognostication, and treatment selection in patients with sarcomas. In the future, emerging biomarkers like circulating tumor DNA analysis offer the potential to improve early detection, monitoring response to treatment, and identifying mechanisms of resistance to personalize sarcoma treatment. Here, we review the current state of molecular biomarkers for sarcomas and highlight opportunities and challenges for the implementation of new technologies in the future.
Collapse
Affiliation(s)
- Agnes Ewongwo
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Caressa Hui
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, CA.; Stanford Cancer Institute, Stanford University, Stanford, CA..
| |
Collapse
|
25
|
Zhang QS, Hayes JP, Gondi V, Pollack SM. Immunotherapy and Radiotherapy Combinations for Sarcoma. Semin Radiat Oncol 2024; 34:229-242. [PMID: 38508787 DOI: 10.1016/j.semradonc.2023.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Sarcomas are a heterogeneous group of bone and soft tissue tumors. Survival outcomes for advanced (unresectable or metastatic) disease remain poor, so therapeutic improvements are needed. Radiotherapy plays an integral role in the neoadjuvant and adjuvant treatment of localized disease as well as in the treatment of metastatic disease. Combining radiotherapy with immunotherapy to potentiate immunotherapy has been used in a variety of cancers other than sarcoma, and there is opportunity to further investigate combining immunotherapy with radiotherapy to try to improve outcomes in sarcoma. In this review, we describe the diversity of the tumor immune microenvironments for sarcomas and describe the immunomodulatory effects of radiotherapy. We discuss studies on the timing of radiotherapy relative to immunotherapy and studies on the radiotherapy dose and fractionation regimen to be used in combination with immunotherapy. We describe the impact of radiotherapy on the tumor immune microenvironment. We review completed and ongoing clinical trials combining radiotherapy with immunotherapy for sarcoma and propose future directions for studies combining immunotherapy with radiotherapy in the treatment of sarcoma.
Collapse
Affiliation(s)
- Qian S Zhang
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - John P Hayes
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Vinai Gondi
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Seth M Pollack
- Division of Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL..
| |
Collapse
|
26
|
Odhiambo DA, Pittman AN, Rickard AG, Castillo RJ, Bassil AM, Chen J, Ravotti ML, Xu ES, Himes JE, Daniel AR, Watts TL, Williams NT, Luo L, Kirsch DG, Mowery YM. Preclinical Evaluation of the ATR Inhibitor BAY 1895344 as a Radiosensitizer for Head and Neck Squamous Cell Carcinoma. Int J Radiat Oncol Biol Phys 2024; 118:1315-1327. [PMID: 38104870 PMCID: PMC11294978 DOI: 10.1016/j.ijrobp.2023.12.012] [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: 08/16/2023] [Revised: 11/17/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
PURPOSE Despite aggressive multimodal treatment that typically includes definitive or adjuvant radiation therapy (RT), locoregional recurrence rates approach 50% for patients with locally advanced human papillomavirus (HPV)-negative head and neck squamous cell carcinoma (HNSCC). Thus, more effective therapeutics are needed to improve patient outcomes. We evaluated the radiosensitizing effects of ataxia telangiectasia and RAD3-related (ATR) inhibitor (ATRi) BAY 1895344 in preclinical models of HNSCC. METHODS AND MATERIALS Murine and human HPV-negative HNSCC cells (MOC2, MOC1, JHU-012) were treated with vehicle or ATRi with or without 4 Gy. Checkpoint kinase 1 phosphorylation and DNA damage (γH2AX) were evaluated by Western blot, and ATRi half-maximal inhibitory concentration was determined by MTT assay for HNSCC cells and immortalized murine oral keratinocytes. In vitro radiosensitization was tested by clonogenic assay. Cell cycle distribution and mitotic catastrophe were evaluated by flow cytometry. Mitotic aberrations were quantified by fluorescent microscopy. Tumor growth delay and survival were assessed in mice bearing MOC2 or JHU-012 transplant tumors treated with vehicle, ATRi, RT (10 Gy × 1 or 8 Gy × 3), or combined ATRi + RT. RESULTS ATRi caused dose-dependent reduction in checkpoint kinase 1 phosphorylation at 1 hour post-RT (4 Gy) and dose-dependent increase in γH2AX at 18 hours post-RT. Addition of RT to ATRi led to decreased BAY 1895344 half-maximal inhibitory concentration in HNSCC cell lines but not in normal tissue surrogate immortalized murine oral keratinocytes. Clonogenic assays demonstrated radiosensitization in the HNSCC cell lines. ATRi abrogated the RT-induced G2/M checkpoint, leading to mitosis with unrepaired DNA damage and increased mitotic aberrations (multinucleated cells, micronuclei, nuclear buds, nucleoplasmic bridges). ATRi and RT significantly delayed tumor growth in MOC2 and JHU-012 in vivo models, with improved overall survival in the MOC2 model. CONCLUSIONS These findings demonstrated that BAY 1895344 increased in vitro and in vivo radiosensitivity in HPV-negative HNSCC preclinical models, suggesting therapeutic potential warranting evaluation in clinical trials for patients with locally advanced or recurrent HPV-negative HNSCC.
Collapse
Affiliation(s)
| | | | - Ashlyn G Rickard
- Dept. of Radiation Oncology, UPMC Hillman Cancer Center/University of Pittsburgh
| | | | | | - Joshua Chen
- College of Arts and Sciences, Duke University
| | - Madison L Ravotti
- Dept. of Radiation Oncology, UPMC Hillman Cancer Center/University of Pittsburgh
| | - Eric S Xu
- Dept. of Radiation Oncology, Duke University
| | | | | | - Tammara L Watts
- Dept. of Head and Neck Surgery & Communication Sciences, Duke University
| | | | - Lixia Luo
- Dept. of Radiation Oncology, Duke University
| | - David G Kirsch
- Dept. of Radiation Oncology, Duke University
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network
- Dept. of Radiation Oncology and Dept. of Medical Biophysics, University of Toronto
| | - Yvonne M Mowery
- Dept. of Radiation Oncology, Duke University
- Dept. of Radiation Oncology, UPMC Hillman Cancer Center/University of Pittsburgh
- Dept. of Head and Neck Surgery & Communication Sciences, Duke University
| |
Collapse
|
27
|
Pang X, Gao S, Liu T, Xu FX, Fan C, Zhang JF, Jiang H. Identification of STAT3 as a biomarker for cellular senescence in liver fibrosis: A bioinformatics and experimental validation study. Genomics 2024; 116:110800. [PMID: 38286349 DOI: 10.1016/j.ygeno.2024.110800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND Cellular senescence is associated with a dysregulated inflammatory response, which is an important driver of the development of liver fibrosis (LF). This study aimed to investigate the effect of cellular senescence on LF and identify potential key biomarkers through bioinformatics analysis combined with validation experiments in vivo and in vitro. METHODS The Gene Expression Omnibus (GEO) database and GeneCards database were used to download the LF dataset and the aging-related gene set, respectively. Functional enrichment analysis of differential genes was then performed using GO and KEGG. Hub genes were further screened using Cytoscape's cytoHubba. Diagnostic values for hub genes were evaluated with a receiver operating characteristic (ROC) curve. Next, CIBERSORTx was used to estimate immune cell types and ratios. Finally, in vivo and in vitro experiments validated the results of the bioinformatics analysis. Moreover, molecular docking was used to simulate drug-gene interactions. RESULTS A total of 44 aging-related differentially expressed genes (AgDEGs) were identified, and enrichment analysis showed that these genes were mainly enriched in inflammatory and immune responses. PPI network analysis identified 6 hub AgDEGs (STAT3, TNF, MMP9, CD44, TGFB1, and TIMP1), and ROC analysis showed that they all have good diagnostic value. Immune infiltration suggested that hub AgDEGs were significantly associated with M1 macrophages or other immune cells. Notably, STAT3 was positively correlated with α-SMA, COL1A1, IL-6 and IL-1β, and was mainly expressed in hepatocytes (HCs). Validation experiments showed that STAT3 expression was upregulated and cellular senescence was increased in LF mice. A co-culture system of HCs and hepatic stellate cells (HSCs) further revealed that inhibiting STAT3 reduced HCs senescence and suppressed HSCs activation. In addition, molecular docking revealed that STAT3 was a potential drug therapy target. CONCLUSIONS STAT3 may be involved in HCs senescence and promote HSCs activation, which in turn leads to the development of LF. Our findings suggest that STAT3 could be a potential biomarker for LF.
Collapse
Affiliation(s)
- Xue Pang
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, Anhui, China
| | - Shang Gao
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, Anhui, China
| | - Tao Liu
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, Anhui, China
| | - Feng Xia Xu
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, Anhui, China
| | - Chang Fan
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Jia Fu Zhang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Hui Jiang
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, Anhui, China.
| |
Collapse
|
28
|
Mortezaee K. WNT/β-catenin regulatory roles on PD-(L)1 and immunotherapy responses. Clin Exp Med 2024; 24:15. [PMID: 38280119 PMCID: PMC10822012 DOI: 10.1007/s10238-023-01274-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/29/2023] [Indexed: 01/29/2024]
Abstract
Dysregulation of WNT/β-catenin is a hallmark of many cancer types and a key mediator of metastasis in solid tumors. Overactive β-catenin signaling hampers dendritic cell (DC) recruitment, promotes CD8+ T cell exclusion and increases the population of regulatory T cells (Tregs). The activity of WNT/β-catenin also induces the expression of programmed death-ligand 1 (PD-L1) on tumor cells and promotes programmed death-1 (PD-1) upregulation. Increased activity of WNT/β-catenin signaling after anti-PD-1 therapy is indicative of a possible implication of this signaling in bypassing immune checkpoint inhibitor (ICI) therapy. This review is aimed at giving a comprehensive overview of the WNT/β-catenin regulatory roles on PD-1/PD-L1 axis in tumor immune ecosystem, discussing about key mechanistic events contributed to the WNT/β-catenin-mediated bypass of ICI therapy, and representing inhibitors of this signaling as promising combinatory regimen to go with anti-PD-(L)1 in cancer immunotherapy. Ideas presented in this review imply the synergistic efficacy of such combination therapy in rendering durable anti-tumor immunity.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
29
|
Ha CT, Tageldein MM, Harding SM. The entanglement of DNA damage and pattern recognition receptor signaling. DNA Repair (Amst) 2024; 133:103595. [PMID: 37988925 DOI: 10.1016/j.dnarep.2023.103595] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/05/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
Cells are under constant pressure to suppress DNA damage originating from both exogenous and endogenous sources. Cellular responses to DNA damage help to prevent mutagenesis and cell death that arises when DNA damage is either left unrepaired or repaired inaccurately. During the "acute phase" of DNA damage signaling, lesions are recognized, processed, and repaired to restore the primary DNA sequence whilst cell cycle checkpoints delay mitotic progression, cell death and the propagation of errors to daughter cells. Increasingly, there is recognition of a "chronic phase" of DNA damage signaling, exemplified by the secretion of dozens of cytokines days after the inciting damage event. In this review, we focus on the cellular origin of these chronic responses, the molecular pathways that control them and the increasing appreciation for the interconnection between acute and chronic DNA damage responses.
Collapse
Affiliation(s)
- Cindy T Ha
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Maha M Tageldein
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Shane M Harding
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada; Departments of Radiation Oncology and Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
30
|
Lukas L, Zhang H, Cheng K, Epstein A. Immune Priming with Spatially Fractionated Radiation Therapy. Curr Oncol Rep 2023; 25:1483-1496. [PMID: 37979032 PMCID: PMC10728252 DOI: 10.1007/s11912-023-01473-7] [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] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE OF REVIEW This review aims to summarize the current preclinical and clinical evidence of nontargeted immune effects of spatially fractionated radiation therapy (SFRT). We then highlight strategies to augment the immunomodulatory potential of SFRT in combination with immunotherapy (IT). RECENT FINDINGS The response of cancer to IT is limited by primary and acquired immune resistance, and strategies are needed to prime the immune system to increase the efficacy of IT. Radiation therapy can induce immunologic effects and can potentially be used to synergize the effects of IT, although the optimal combination of radiation and IT is largely unknown. SFRT is a novel radiation technique that limits ablative doses to tumor subvolumes, and this highly heterogeneous dose deposition may increase the immune-rich infiltrate within the targeted tumor with enhanced antigen presentation and activated T cells in nonirradiated tumors. The understanding of nontargeted effects of SFRT can contribute to future translational strategies to combine SFRT and IT. Integration of SFRT and IT is an innovative approach to address immune resistance to IT with the overall goal of improving the therapeutic ratio of radiation therapy and increasing the efficacy of IT.
Collapse
Affiliation(s)
- Lauren Lukas
- Department of Radiation Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Hualin Zhang
- Department of Radiation Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Karen Cheng
- Department of Radiation Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Alan Epstein
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
31
|
Hendrickson PG, Oristian KM, Browne MR, Luo L, Ma Y, Cardona DM, Linardic CM, Kirsch DG. Expression of the CIC-DUX4 fusion oncoprotein mimics human CIC-rearranged sarcoma in genetically engineered mouse models. RESEARCH SQUARE 2023:rs.3.rs-3487637. [PMID: 37961185 PMCID: PMC10635354 DOI: 10.21203/rs.3.rs-3487637/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
CIC-DUX4 sarcoma (CDS) is a rare but highly aggressive undifferentiated small round cell sarcoma driven by a fusion between the tumor suppressor Capicua (CIC) and DUX4. Currently, there are no effective treatments and efforts to identify and translate better therapies are limited by the scarcity of patient tumor samples and cell lines. To address this limitation, we generated three genetically engineered mouse models of CDS (Ch7CDS, Ai9CDS, and TOPCDS). Remarkably, chimeric mice from all three conditional models developed spontaneous tumors and widespread metastasis in the absence of Cre-recombinase. The penetrance of spontaneous (Cre-independent) tumor formation was complete irrespective of bi-allelic CIC function and the distance between loxP sites. Characterization of primary and metastatic mouse tumors showed that they consistently expressed the CIC-DUX4 fusion protein as well as other downstream markers of the disease credentialing these models as CDS. In addition, tumor-derived cell lines were generated and ChIP-seq was preformed to map fusion-gene specific binding using an N-terminal HA epitope tag. These datasets, along with paired H3K27ac ChIP-seq maps, validate CIC-DUX4 as a neomorphic transcriptional activator. Moreover, they are consistent with a model where ETS family transcription factors are cooperative and redundant drivers of the core regulatory circuitry in CDS.
Collapse
Affiliation(s)
- Peter G. Hendrickson
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | | | - MaKenna R. Browne
- Developmental and Stem Cell Biology Program, Duke University Medical Center, Durham, NC, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Dianna M. Cardona
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Corinne M. Linardic
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - David G. Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
Hendrickson PG, Oristian KM, Browne MR, Lou L, Ma Y, Cardona DM, Linardic CM, Kirsch DG. Expression of the CIC-DUX4 fusion oncoprotein mimics human CIC-rearranged sarcoma in genetically engineered mouse models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559519. [PMID: 37808628 PMCID: PMC10557731 DOI: 10.1101/2023.09.26.559519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
CIC-DUX4 sarcoma (CDS) is a rare but highly aggressive undifferentiated small round cell sarcoma driven by a fusion between the tumor suppressor Capicua (CIC) and DUX4. Currently, there are no effective treatments and efforts to identify and translate better therapies are limited by the scarcity of tissues and patients. To address this limitation, we generated three genetically engineered mouse models of CDS (Ch7CDS, Ai9CDS, and TOPCDS). Remarkably, chimeric mice from all three conditional models developed spontaneous tumors and widespread metastasis in the absence of Cre-recombinase. The penetrance of spontaneous (Cre-independent) tumor formation was complete irrespective of bi-allelic CIC function and loxP site proximity. Characterization of primary and metastatic mouse tumors showed that they consistently expressed the CIC-DUX4 fusion protein as well as other downstream markers of the disease credentialing these models as CDS. In addition, tumor-derived cell lines were generated and ChIP-seq was preformed to map fusion-gene specific binding using an N-terminal HA epitope tag. These datasets, along with paired H3K27ac ChIP-seq maps, validate CIC-DUX4 as a neomorphic transcriptional activator. Moreover, they are consistent with a model where ETS family transcription factors are cooperative and redundant drivers of the core regulatory circuitry in CDS.
Collapse
Affiliation(s)
- Peter G. Hendrickson
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | | | - MaKenna R. Browne
- Developmental and Stem Cell Biology Program, Duke University Medical Center, Durham, NC, USA
| | - Lixia Lou
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Dianna M. Cardona
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Corinne M. Linardic
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - David G. Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
33
|
Kohlmeyer JL, Lingo JJ, Kaemmer CA, Scherer A, Warrier A, Voigt E, Garay JAR, McGivney GR, Brockman QR, Tang A, Calizo A, Pollard K, Zhang X, Hirbe AC, Pratilas CA, Leidinger M, Breheny P, Chimenti MS, Sieren JC, Monga V, Tanas MR, Meyerholz DK, Darbro BW, Dodd RD, Quelle DE. CDK4/6-MEK Inhibition in MPNSTs Causes Plasma Cell Infiltration, Sensitization to PD-L1 Blockade, and Tumor Regression. Clin Cancer Res 2023; 29:3484-3497. [PMID: 37410426 PMCID: PMC10528807 DOI: 10.1158/1078-0432.ccr-23-0749] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/22/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023]
Abstract
PURPOSE Malignant peripheral nerve sheath tumors (MPNST) are lethal, Ras-driven sarcomas that lack effective therapies. We investigated effects of targeting cyclin-dependent kinases 4 and 6 (CDK4/6), MEK, and/or programmed death-ligand 1 (PD-L1) in preclinical MPNST models. EXPERIMENTAL DESIGN Patient-matched MPNSTs and precursor lesions were examined by FISH, RNA sequencing, IHC, and Connectivity-Map analyses. Antitumor activity of CDK4/6 and MEK inhibitors was measured in MPNST cell lines, patient-derived xenografts (PDX), and de novo mouse MPNSTs, with the latter used to determine anti-PD-L1 response. RESULTS Patient tumor analyses identified CDK4/6 and MEK as actionable targets for MPNST therapy. Low-dose combinations of CDK4/6 and MEK inhibitors synergistically reactivated the retinoblastoma (RB1) tumor suppressor, induced cell death, and decreased clonogenic survival of MPNST cells. In immune-deficient mice, dual CDK4/6-MEK inhibition slowed tumor growth in 4 of 5 MPNST PDXs. In immunocompetent mice, combination therapy of de novo MPNSTs caused tumor regression, delayed resistant tumor outgrowth, and improved survival relative to monotherapies. Drug-sensitive tumors that regressed contained plasma cells and increased cytotoxic T cells, whereas drug-resistant tumors adopted an immunosuppressive microenvironment with elevated MHC II-low macrophages and increased tumor cell PD-L1 expression. Excitingly, CDK4/6-MEK inhibition sensitized MPNSTs to anti-PD-L1 immune checkpoint blockade (ICB) with some mice showing complete tumor regression. CONCLUSIONS CDK4/6-MEK inhibition induces a novel plasma cell-associated immune response and extended antitumor activity in MPNSTs, which dramatically enhances anti-PD-L1 therapy. These preclinical findings provide strong rationale for clinical translation of CDK4/6-MEK-ICB targeted therapies in MPNST as they may yield sustained antitumor responses and improved patient outcomes.
Collapse
Affiliation(s)
- Jordan L Kohlmeyer
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Joshua J Lingo
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Courtney A Kaemmer
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Amanda Scherer
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Akshaya Warrier
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Ellen Voigt
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | | | - Gavin R McGivney
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
| | - Qierra R Brockman
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Amy Tang
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Center, Eastern Virginia Medical School, Norfolk, Virginia
| | - Ana Calizo
- Department of Oncology, Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Kai Pollard
- Department of Oncology, Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Xiaochun Zhang
- Division of Medical Oncology, Washington University, St. Louis, Missouri
| | - Angela C Hirbe
- Division of Medical Oncology, Washington University, St. Louis, Missouri
| | - Christine A Pratilas
- Department of Oncology, Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Mariah Leidinger
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Patrick Breheny
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa
| | - Michael S Chimenti
- Iowa Institute of Human Genetics, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Jessica C. Sieren
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Radiation, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Varun Monga
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Munir R Tanas
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - David K Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Benjamin W Darbro
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Rebecca D Dodd
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Dawn E Quelle
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
34
|
Schmidt DR, Gramatikov IMT, Sheen A, Williams CL, Hurwitz M, Dodge LE, Holupka E, Kiger WS, Cornwall-Brady MR, Huang W, Mak HH, Cormier KS, Condon C, Dane Wittrup K, Yilmaz ÖH, Stevenson MA, Down JD, Floyd SR, Roper J, Vander Heiden MG. Ablative radiotherapy improves survival but does not cure autochthonous mouse models of prostate and colorectal cancer. COMMUNICATIONS MEDICINE 2023; 3:108. [PMID: 37558833 PMCID: PMC10412558 DOI: 10.1038/s43856-023-00336-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 07/24/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Genetically engineered mouse models (GEMMs) of cancer are powerful tools to study mechanisms of disease progression and therapy response, yet little is known about how these models respond to multimodality therapy used in patients. Radiation therapy (RT) is frequently used to treat localized cancers with curative intent, delay progression of oligometastases, and palliate symptoms of metastatic disease. METHODS Here we report the development, testing, and validation of a platform to immobilize and target tumors in mice with stereotactic ablative RT (SART). Xenograft and autochthonous tumor models were treated with hypofractionated ablative doses of radiotherapy. RESULTS We demonstrate that hypofractionated regimens used in clinical practice can be effectively delivered in mouse models. SART alters tumor stroma and the immune environment, improves survival in GEMMs of primary prostate and colorectal cancer, and synergizes with androgen deprivation in prostate cancer. Complete pathologic responses were achieved in xenograft models, but not in GEMMs. CONCLUSIONS While SART is capable of fully ablating xenografts, it is unable to completely eradicate disease in GEMMs, arguing that resistance to potentially curative therapy can be modeled in GEMMs.
Collapse
Affiliation(s)
- Daniel R Schmidt
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Iva Monique T Gramatikov
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Allison Sheen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christopher L Williams
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Boston, MA, USA
| | - Martina Hurwitz
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Laura E Dodge
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Edward Holupka
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - W S Kiger
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Milton R Cornwall-Brady
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Wei Huang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Howard H Mak
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kathleen S Cormier
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charlene Condon
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - K Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ömer H Yilmaz
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, USA
| | - Mary Ann Stevenson
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Julian D Down
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Scott R Floyd
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Jatin Roper
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Division of Gastroenterology, and Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
35
|
Monti S, Truppa ME, Albanese S, Mancini M. Radiomics and Radiogenomics in Preclinical Imaging on Murine Models: A Narrative Review. J Pers Med 2023; 13:1204. [PMID: 37623455 PMCID: PMC10455673 DOI: 10.3390/jpm13081204] [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: 06/09/2023] [Revised: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023] Open
Abstract
Over the past decade, medical imaging technologies have become increasingly significant in both clinical and preclinical research, leading to a better understanding of disease processes and the development of new diagnostic and theranostic methods. Radiomic and radiogenomic approaches have furthered this progress by exploring the relationship between imaging characteristics, genomic information, and outcomes that qualitative interpretations may have overlooked, offering valuable insights for personalized medicine. Preclinical research allows for a controlled environment where various aspects of a pathology can be replicated in animal models, providing radiomic and radiogenomic approaches with the unique opportunity to investigate the causal connection between imaging and molecular factors. The aim of this review is to present the current state of the art in the application of radiomics and radiogenomics on murine models. This review will provide a brief description of relevant articles found in the literature with a discussion on the implications and potential translational relevance of these findings.
Collapse
Affiliation(s)
| | | | - Sandra Albanese
- National Research Council, Institute of Biostructures and Bioimaging, 80145 Naples, Italy; (S.M.); (M.E.T.); (M.M.)
| | | |
Collapse
|
36
|
Vonderhaar EP, Dwinell MB, Craig BT. Targeted immune activation in pediatric solid tumors: opportunities to complement local control approaches. Front Immunol 2023; 14:1202169. [PMID: 37426669 PMCID: PMC10325564 DOI: 10.3389/fimmu.2023.1202169] [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: 04/07/2023] [Accepted: 05/31/2023] [Indexed: 07/11/2023] Open
Abstract
Surgery or radiation therapy is nearly universally applied for pediatric solid tumors. In many cases, in diverse tumor types, distant metastatic disease is present and evades surgery or radiation. The systemic host response to these local control modalities may lead to a suppression of antitumor immunity, with potential negative impact on the clinical outcomes for patients in this scenario. Emerging evidence suggests that the perioperative immune responses to surgery or radiation can be modulated therapeutically to preserve anti-tumor immunity, with the added benefit of preventing these local control approaches from serving as pro-tumorigenic stimuli. To realize the potential benefit of therapeutic modulation of the systemic response to surgery or radiation on distant disease that evades these modalities, a detailed knowledge of the tumor-specific immunology as well as the immune responses to surgery and radiation is imperative. In this Review we highlight the current understanding of the tumor immune microenvironment for the most common peripheral pediatric solid tumors, the immune responses to surgery and radiation, and current evidence that supports the potential use of immune activating agents in the perioperative window. Finally, we define existing knowledge gaps that limit the current translational potential of modulating perioperative immunity to achieve effective anti-tumor outcomes.
Collapse
Affiliation(s)
- Emily P. Vonderhaar
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Center for Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michael B. Dwinell
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Center for Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Brian T. Craig
- Center for Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
37
|
Wang J, Ge H, Tian Z. Immunotherapy Plus Radiotherapy for the Treatment of Sarcomas: Is There a Potential for Synergism? Onco Targets Ther 2023; 16:385-397. [PMID: 37313391 PMCID: PMC10258041 DOI: 10.2147/ott.s410693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Soft tissue sarcoma (STS) is a highly heterogeneous malignant tumor derived from mesenchymal tissue. Advanced STS has a poor response to the current anti-cancer therapeutic options, with a median overall survival of less than two years. Thus, new and more effective treatment methods for STS are needed. Increasing evidence has shown that immunotherapy and radiotherapy have synergistic therapeutic effects against malignant tumors. In addition, immunoradiotherapy has yielded positive results in clinical trials for various cancers. In this review, we discuss the synergistic mechanism of immunoradiotherapy in cancer treatment and the application of this combined regimen for the treatment of several cancers. In addition, we summarize the existing evidence on the use of immunoradiotherapy for the treatment of STS and the relevant clinical trials that are currently ongoing. Furthermore, we identify challenges in the use of immunoradiotherapy for the treatment of sarcomas and propose methods and precautions for overcoming these challenges. Lastly, we propose clinical research strategies and future research directions to help in the research and treatment of STS.
Collapse
Affiliation(s)
- Jiaqiang Wang
- Department of Bone and Soft Tissue, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan Province, 450008, People’s Republic of China
| | - Hong Ge
- Department of Radiotherapy, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan Province, 450008, People’s Republic of China
| | - Zhichao Tian
- Department of Bone and Soft Tissue, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan Province, 450008, People’s Republic of China
| |
Collapse
|
38
|
Mortaezaee K, Majidpoor J. Mechanisms of CD8 + T cell exclusion and dysfunction in cancer resistance to anti-PD-(L)1. Biomed Pharmacother 2023; 163:114824. [PMID: 37141735 DOI: 10.1016/j.biopha.2023.114824] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/26/2023] [Accepted: 04/30/2023] [Indexed: 05/06/2023] Open
Abstract
CD8+ T cells are the front-line defensive cells against cancer. Reduced infiltration and effector function of CD8+ T cells occurs in cancer and is contributed to defective immunity and immunotherapy resistance. Exclusion and exhaustion of CD8+ T cells are the two key factors associated with reduced durability of immune checkpoint inhibitor (ICI) therapy. Initially activated T cells upon exposure to chronic antigen stimulation or immunosuppressive tumor microenvironment (TME) acquire a hyporesponsive state that progressively lose their effector function. Thus, a key strategy in cancer immunotherapy is to look for factors contributed to defective CD8+ T cell infiltration and function. Targeting such factors can define a promising supplementary approach in patients receiving anti-programmed death-1 receptor (PD-1)/anti-programmed death-ligand 1 (PD-L1) therapy. Recently, bispecific antibodies are developed against PD-(L)1 and a dominant factor within TME, representing higher safety profile and exerting more desired outcomes. The focus of this review is to discuss about promoters of deficient infiltration and effector function of CD8+ T cells and their addressing in cancer ICI therapy.
Collapse
Affiliation(s)
- Keywan Mortaezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
39
|
Su C, Himes JE, Kirsch DG. Relationship between the tumor microenvironment and the efficacy of the combination of radiotherapy and immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:201-232. [PMID: 37438018 DOI: 10.1016/bs.ircmb.2023.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Activating and recruiting the immune system is critical for successful cancer treatment. Since the discovery of immune checkpoint inhibitors, immunotherapy has become the standard of care for many types of cancers. However, many patients fail to respond to immunotherapy. Further research is needed to understand the mechanisms of resistance and adjuvant therapies that can help sensitize patients to immunotherapies. Here, we will discuss how radiotherapy can change the tumor microenvironment and work synergistically with immunotherapy. We will examine different pre-clinical models focusing on their limitations and their unique advantages in studying the efficacy of treatments and the tumor microenvironment. We will also describe emerging findings from clinical trials testing the combination of immunotherapy and radiotherapy.
Collapse
Affiliation(s)
- Chang Su
- Molecular Cancer Biology Program and Medical Scientist Training Program, Duke University School of Medicine, Durham, NC, United States
| | - Jonathon E Himes
- Molecular Cancer Biology Program and Medical Scientist Training Program, Duke University School of Medicine, Durham, NC, United States
| | - David G Kirsch
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, United States; Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC, United States.
| |
Collapse
|
40
|
Bernhardt SM, Mitchell E, Stamnes S, Hoffmann RJ, Calhoun A, Klug A, Russell TD, Pennock ND, Walker JM, Schedin P. Isogenic Mammary Models of Intraductal Carcinoma Reveal Progression to Invasiveness in the Absence of a Non-Obligatory In Situ Stage. Cancers (Basel) 2023; 15:2257. [PMID: 37190184 PMCID: PMC10136757 DOI: 10.3390/cancers15082257] [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: 03/13/2023] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
In breast cancer, progression to invasive ductal carcinoma (IDC) involves interactions between immune, myoepithelial, and tumor cells. Development of IDC can proceed through ductal carcinoma in situ (DCIS), a non-obligate, non-invasive stage, or IDC can develop without evidence of DCIS and these cases associate with poorer prognosis. Tractable, immune-competent mouse models are needed to help delineate distinct mechanisms of local tumor cell invasion and prognostic implications. To address these gaps, we delivered murine mammary carcinoma cell lines directly into the main mammary lactiferous duct of immune-competent mice. Using two strains of immune-competent mice (BALB/c, C57BL/6), one immune-compromised (severe combined immunodeficiency; SCID) C57BL/6 strain, and six different murine mammary cancer cell lines (D2.OR, D2A1, 4T1, EMT6, EO771, Py230), we found early loss of ductal myoepithelial cell differentiation markers p63, α-smooth muscle actin, and calponin, and rapid formation of IDC in the absence of DCIS. Rapid IDC formation also occurred in the absence of adaptive immunity. Combined, these studies demonstrate that loss of myoepithelial barrier function does not require an intact immune system, and suggest that these isogenic murine models may prove a useful tool to study IDC in the absence of a non-obligatory DCIS stage-an under-investigated subset of poor prognostic human breast cancer.
Collapse
Affiliation(s)
- Sarah M. Bernhardt
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Elizabeth Mitchell
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Stephanie Stamnes
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Reuben J. Hoffmann
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andrea Calhoun
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alex Klug
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Tanya D. Russell
- Center for Advancing Professional Excellence, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nathan D. Pennock
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Joshua M. Walker
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Pepper Schedin
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, CO 80045, USA
| |
Collapse
|
41
|
Chow J, Khan A, Gaudieri M, Wasik BJ, Conway A, Soh KT, Repasky EA, Schwaab T, Wallace PK, Abrams SI, Singh AK, Muhitch JB. Tumor and immune remodeling following radiotherapy in human renal cell carcinoma. J Immunother Cancer 2023; 11:e006392. [PMID: 37080610 PMCID: PMC10124322 DOI: 10.1136/jitc-2022-006392] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND Studies evaluating peripheral patient samples show radiation can modulate immune responses, yet the biological changes in human tumors particularly at the cellular level remain largely unknown. Here, we address how radiation treatment shapes the immune compartment and interactions with cancer cells within renal cell carcinoma (RCC) patient tumors. METHODS To identify how radiation shaped the immune compartment and potential immune interactions with tumor cells we evaluated RCC tumors from patients treated only with nephrectomy or with radiation followed by nephrectomy. Spectral flow cytometry using a 35-marker panel was performed on cell suspensions to evaluate protein expression within immune subsets. To reveal how radiation alters programming of immune populations and interactions with tumor cells, we examined transcriptional changes by single-cell RNA sequencing (scRNAseq). RESULTS Spectral flow cytometry analysis revealed increased levels of early-activated as well as effector programmed cell death protein-1 (PD-1)+ CD8 T-cell subsets within irradiated tumors. Following quality control, scRNAseq of tumor samples from nephrectomy-only or radiation followed by nephrectomy-treated patients generated an atlas containing 34,626 total cells. Transcriptional analysis revealed increased transition from stem-like T-cell populations to effector T cells in irradiated tumors. Interferon (IFN) pathways, that are central to radiation-induced immunogenicity, were enriched in irradiated lymphoid, myeloid, and cancer cell populations. Focused cancer cell analysis showed enhanced antigen presentation and increased predicted TRAIL-mediated and IFN-mediated interactions between tumor cells and the same effector T-cell subsets increased by radiation. TRAIL and IFN pathways enriched in irradiated tumors were associated with survival in patients treated with immunotherapy. CONCLUSIONS These findings identify the source of IFN enrichment within irradiated RCC and reveal heightened levels of PD-1+ CD8+ T-cell subsets and increased probability of interactions with tumor cells following standalone radiation treatment. This study provides a window into the irradiated tumor-immune microenvironment of patients and rationale for treatment combinations.
Collapse
Affiliation(s)
- Jacky Chow
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Adil Khan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Madeline Gaudieri
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Brianna J Wasik
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Alexis Conway
- Department of Flow and Image Cytometry, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Kah Teong Soh
- Department of Flow and Image Cytometry, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Thomas Schwaab
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Paul K Wallace
- Department of Flow and Image Cytometry, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Anurag K Singh
- Department of Radiation Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Jason B Muhitch
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| |
Collapse
|
42
|
Verhaegen F, Butterworth KT, Chalmers AJ, Coppes RP, de Ruysscher D, Dobiasch S, Fenwick JD, Granton PV, Heijmans SHJ, Hill MA, Koumenis C, Lauber K, Marples B, Parodi K, Persoon LCGG, Staut N, Subiel A, Vaes RDW, van Hoof S, Verginadis IL, Wilkens JJ, Williams KJ, Wilson GD, Dubois LJ. Roadmap for precision preclinical x-ray radiation studies. Phys Med Biol 2023; 68:06RM01. [PMID: 36584393 DOI: 10.1088/1361-6560/acaf45] [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: 05/12/2022] [Accepted: 12/30/2022] [Indexed: 12/31/2022]
Abstract
This Roadmap paper covers the field of precision preclinical x-ray radiation studies in animal models. It is mostly focused on models for cancer and normal tissue response to radiation, but also discusses other disease models. The recent technological evolution in imaging, irradiation, dosimetry and monitoring that have empowered these kinds of studies is discussed, and many developments in the near future are outlined. Finally, clinical translation and reverse translation are discussed.
Collapse
Affiliation(s)
- Frank Verhaegen
- MAASTRO Clinic, Radiotherapy Division, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
- SmART Scientific Solutions BV, Maastricht, The Netherlands
| | - Karl T Butterworth
- Patrick G. Johnston, Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Anthony J Chalmers
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Rob P Coppes
- Departments of Biomedical Sciences of Cells & Systems, Section Molecular Cell Biology and Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Dirk de Ruysscher
- MAASTRO Clinic, Radiotherapy Division, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sophie Dobiasch
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine and Klinikum rechts der Isar, Germany
- Department of Medical Physics, Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Germany
| | - John D Fenwick
- Department of Medical Physics & Biomedical Engineering University College LondonMalet Place Engineering Building, London WC1E 6BT, United Kingdom
| | | | | | - Mark A Hill
- MRC Oxford Institute for Radiation Oncology, University of Oxford, ORCRB Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU München, Munich, Germany
- German Cancer Consortium (DKTK), Partner site Munich, Germany
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester, NY, United States of America
| | - Katia Parodi
- German Cancer Consortium (DKTK), Partner site Munich, Germany
- Department of Medical Physics, Faculty of Physics, Ludwig-Maximilians-Universität München, Garching b. Munich, Germany
| | | | - Nick Staut
- SmART Scientific Solutions BV, Maastricht, The Netherlands
| | - Anna Subiel
- National Physical Laboratory, Medical Radiation Science Hampton Road, Teddington, Middlesex, TW11 0LW, United Kingdom
| | - Rianne D W Vaes
- MAASTRO Clinic, Radiotherapy Division, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | - Ioannis L Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jan J Wilkens
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine and Klinikum rechts der Isar, Germany
- Physics Department, Technical University of Munich (TUM), Germany
| | - Kaye J Williams
- Division of Pharmacy and Optometry, University of Manchester, Manchester, United Kingdom
| | - George D Wilson
- Department of Radiation Oncology, Beaumont Health, MI, United States of America
- Henry Ford Health, Detroit, MI, United States of America
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
43
|
Peehl DM, Badea CT, Chenevert TL, Daldrup-Link HE, Ding L, Dobrolecki LE, Houghton AM, Kinahan PE, Kurhanewicz J, Lewis MT, Li S, Luker GD, Ma CX, Manning HC, Mowery YM, O’Dwyer PJ, Pautler RG, Rosen MA, Roudi R, Ross BD, Shoghi KI, Sriram R, Talpaz M, Wahl RL, Zhou R. Animal Models and Their Role in Imaging-Assisted Co-Clinical Trials. Tomography 2023; 9:657-680. [PMID: 36961012 PMCID: PMC10037611 DOI: 10.3390/tomography9020053] [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: 01/26/2023] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
The availability of high-fidelity animal models for oncology research has grown enormously in recent years, enabling preclinical studies relevant to prevention, diagnosis, and treatment of cancer to be undertaken. This has led to increased opportunities to conduct co-clinical trials, which are studies on patients that are carried out parallel to or sequentially with animal models of cancer that mirror the biology of the patients' tumors. Patient-derived xenografts (PDX) and genetically engineered mouse models (GEMM) are considered to be the models that best represent human disease and have high translational value. Notably, one element of co-clinical trials that still needs significant optimization is quantitative imaging. The National Cancer Institute has organized a Co-Clinical Imaging Resource Program (CIRP) network to establish best practices for co-clinical imaging and to optimize translational quantitative imaging methodologies. This overview describes the ten co-clinical trials of investigators from eleven institutions who are currently supported by the CIRP initiative and are members of the Animal Models and Co-clinical Trials (AMCT) Working Group. Each team describes their corresponding clinical trial, type of cancer targeted, rationale for choice of animal models, therapy, and imaging modalities. The strengths and weaknesses of the co-clinical trial design and the challenges encountered are considered. The rich research resources generated by the members of the AMCT Working Group will benefit the broad research community and improve the quality and translational impact of imaging in co-clinical trials.
Collapse
Affiliation(s)
- Donna M. Peehl
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA; (J.K.); (R.S.)
| | - Cristian T. Badea
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA;
| | - Thomas L. Chenevert
- Department of Radiology and the Center for Molecular Imaging, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; (T.L.C.); (G.D.L.); (B.D.R.)
| | - Heike E. Daldrup-Link
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305, USA; (H.E.D.-L.); (R.R.)
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (L.D.); (S.L.); (C.X.M.)
| | - Lacey E. Dobrolecki
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA;
| | | | - Paul E. Kinahan
- Department of Radiology, University of Washington, Seattle, WA 98105, USA;
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA; (J.K.); (R.S.)
| | - Michael T. Lewis
- Departments of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Shunqiang Li
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (L.D.); (S.L.); (C.X.M.)
| | - Gary D. Luker
- Department of Radiology and the Center for Molecular Imaging, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; (T.L.C.); (G.D.L.); (B.D.R.)
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Cynthia X. Ma
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (L.D.); (S.L.); (C.X.M.)
| | - H. Charles Manning
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Yvonne M. Mowery
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708, USA;
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine, Durham, NC 27708, USA
| | - Peter J. O’Dwyer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.J.O.); (M.A.R.); (R.Z.)
| | - Robia G. Pautler
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Mark A. Rosen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.J.O.); (M.A.R.); (R.Z.)
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raheleh Roudi
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305, USA; (H.E.D.-L.); (R.R.)
| | - Brian D. Ross
- Department of Radiology and the Center for Molecular Imaging, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; (T.L.C.); (G.D.L.); (B.D.R.)
- Department of Biological Chemistry, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Kooresh I. Shoghi
- Mallinckrodt Institute of Radiology (MIR), Washington University School of Medicine, St. Louis, MO 63110, USA; (K.I.S.); (R.L.W.)
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA; (J.K.); (R.S.)
| | - Moshe Talpaz
- Division of Hematology/Oncology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA;
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Richard L. Wahl
- Mallinckrodt Institute of Radiology (MIR), Washington University School of Medicine, St. Louis, MO 63110, USA; (K.I.S.); (R.L.W.)
| | - Rong Zhou
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.J.O.); (M.A.R.); (R.Z.)
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
44
|
A New Signature of Sarcoma Based on the Tumor Microenvironment Benefits Prognostic Prediction. Int J Mol Sci 2023; 24:ijms24032961. [PMID: 36769292 PMCID: PMC9918054 DOI: 10.3390/ijms24032961] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/13/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Sarcomas are a group of malignant tumors derived from mesenchymal tissues that display complex and variable pathological types. The impact of the immune properties of the tumor microenvironment (TME) on the prognosis, treatment, and management of sarcomas has attracted attention, requiring the exploration of sensitive and accurate signatures. In this study, The Cancer Genome Atlas (TCGA) database was searched to screen for an RNA sequencing dataset, retrieving 263 sarcoma and 2 normal samples with survival data. Genes associated with immune regulation in sarcomas were retrieved from the Tumor Immune Estimation Resource database to estimate tumor purity and immune cell infiltration levels. The samples were then divided into the immune-high and immune-low groups. Then, we screened for differentially expressed genes (DEGs) between the two groups. The intersection between immune-related genes and DEGs was then determined. Univariate Cox and least absolute shrinkage and selection operator analyses were used to select ideal genes for prognostic prediction and subsequent construction of a risk signature. A survival analysis was performed to reveal the dissimilarity in survival between the high- and low-score groups. Finally, a nomogram was generated to verify the accuracy and reliability of the signature. Through Estimation of STromal and Immune cells in MAlignant Tumour tissues using Expression (ESTIMATE) analysis, high ESTIMATE, and low tumor purity were significantly associated with a favorable prognosis. Moreover, a total of 5259 DEGs were retrieved, the majority of which were downregulated. In total, 590 immune-associated genes overlapped with the DEGs, among which nine hub genes were identified. Finally, two candidate genes, ACVR2B and NFYA, were identified, based on which a risk signature was constructed. The risk signature constructed in this study is accurate and reliable for the prognostic prediction and phenotyping of sarcomas.
Collapse
|
45
|
Marritt KL, Hildebrand KM, Hildebrand KN, Singla AK, Zemp FJ, Mahoney DJ, Jirik FR, Monument MJ. Intratumoral STING activation causes durable immunogenic tumor eradication in the KP soft tissue sarcoma model. Front Immunol 2023; 13:1087991. [PMID: 36700206 PMCID: PMC9868147 DOI: 10.3389/fimmu.2022.1087991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction Soft tissue sarcomas (STS) are highly metastatic, connective-tissue lineage solid cancers. Immunologically, sarcomas are frequently characterized by a paucity of tumor infiltrating lymphocytes and an immune suppressive microenvironment. Activation of the STING pathway can induce potent immune-driven anti-tumor responses within immunogenic solid tumors; however, this strategy has not been evaluated in immunologically cold sarcomas. Herein, we assessed the therapeutic response of intratumoral STING activation in an immunologically cold murine model of undifferentiated pleomorphic sarcoma (UPS). Materials and Results A single intratumoral injection of the murine STING agonist, DMXAA resulted in durable cure in up to 60% of UPS-bearing mice. In mice with synchronous lung metastases, STING activation within hindlimb tumors resulted in 50% cure in both anatomic sites. Surviving mice all rejected UPS re-challenge in the hindlimb and lung. Therapeutic efficacy of STING was inhibited by lymphocyte deficiency but unaffected by macrophage deficiency. Immune phenotyping demonstrated enrichment of lymphocytic responses in tumors at multiple timepoints following treatment. Immune checkpoint blockade enhanced survival following STING activation. Discussion These data suggest intratumoral activation of the STING pathway elicits local and systemic anti-tumor immune responses in a lymphocyte poor sarcoma model and deserves further evaluation as an adjunctive local and systemic treatment for sarcomas.
Collapse
Affiliation(s)
- Kayla L. Marritt
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Karys M. Hildebrand
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kurt N. Hildebrand
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Arvind K. Singla
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Franz J. Zemp
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Douglas J. Mahoney
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Frank R. Jirik
- McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Michael J. Monument
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,*Correspondence: Michael J. Monument,
| |
Collapse
|
46
|
Patel R, Mowery YM, Qi Y, Bassil AM, Holbrook M, Xu ES, Hong CS, Himes JE, Williams NT, Everitt J, Ma Y, Luo L, Selitsky SR, Modliszewski JL, Gao J, Jung SH, Kirsch DG, Badea CT. Neoadjuvant Radiation Therapy and Surgery Improves Metastasis-Free Survival over Surgery Alone in a Primary Mouse Model of Soft Tissue Sarcoma. Mol Cancer Ther 2023; 22:112-122. [PMID: 36162051 PMCID: PMC9812921 DOI: 10.1158/1535-7163.mct-21-0991] [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: 12/09/2021] [Revised: 06/28/2022] [Accepted: 09/20/2022] [Indexed: 02/03/2023]
Abstract
This study aims to investigate whether adding neoadjuvant radiotherapy (RT), anti-programmed cell death protein-1 (PD-1) antibody (anti-PD-1), or RT + anti-PD-1 to surgical resection improves disease-free survival for mice with soft tissue sarcomas (STS). We generated a high mutational load primary mouse model of STS by intramuscular injection of adenovirus expressing Cas9 and guide RNA targeting Trp53 and intramuscular injection of 3-methylcholanthrene (MCA) into the gastrocnemius muscle of wild-type mice (p53/MCA model). We randomized tumor-bearing mice to receive isotype control or anti-PD-1 antibody with or without radiotherapy (20 Gy), followed by hind limb amputation. We used micro-CT to detect lung metastases with high spatial resolution, which was confirmed by histology. We investigated whether sarcoma metastasis was regulated by immunosurveillance by lymphocytes or tumor cell-intrinsic mechanisms. Compared with surgery with isotype control antibody, the combination of anti-PD-1, radiotherapy, and surgery improved local recurrence-free survival (P = 0.035) and disease-free survival (P = 0.005), but not metastasis-free survival. Mice treated with radiotherapy, but not anti-PD-1, showed significantly improved local recurrence-free survival and metastasis-free survival over surgery alone (P = 0.043 and P = 0.007, respectively). The overall metastasis rate was low (∼12%) in the p53/MCA sarcoma model, which limited the power to detect further improvement in metastasis-free survival with addition of anti-PD-1 therapy. Tail vein injections of sarcoma cells into immunocompetent mice suggested that impaired metastasis was due to inability of sarcoma cells to grow in the lungs rather than a consequence of immunosurveillance. In conclusion, neoadjuvant radiotherapy improves metastasis-free survival after surgery in a primary model of STS.
Collapse
Affiliation(s)
- Rutulkumar Patel
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Yvonne M. Mowery
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
- Department of Head and Neck Surgery & Communication Sciences, Duke University Medical Center, Durham, NC 27710
| | - Yi Qi
- Department of Radiology, Duke University Medical Center, Durham, NC 27710
| | - Alex M. Bassil
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Matt Holbrook
- Department of Radiology, Duke University Medical Center, Durham, NC 27710
| | - Eric S. Xu
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Cierra S. Hong
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Jonathon E. Himes
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Nerissa T. Williams
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Jeffrey Everitt
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710
| | - Yan Ma
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | | | | | - Junheng Gao
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC
| | - Sin-Ho Jung
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC
| | - David G. Kirsch
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710
| | - Cristian T. Badea
- Department of Radiology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
47
|
Boumelha J, de Carné Trécesson S, Law EK, Romero-Clavijo P, Coelho MA, Ng K, Mugarza E, Moore C, Rana S, Caswell DR, Murillo M, Hancock DC, Argyris PP, Brown WL, Durfee C, Larson LK, Vogel RI, Suárez-Bonnet A, Priestnall SL, East P, Ross SJ, Kassiotis G, Molina-Arcas M, Swanton C, Harris R, Downward J. An Immunogenic Model of KRAS-Mutant Lung Cancer Enables Evaluation of Targeted Therapy and Immunotherapy Combinations. Cancer Res 2022; 82:3435-3448. [PMID: 35930804 PMCID: PMC7613674 DOI: 10.1158/0008-5472.can-22-0325] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/01/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022]
Abstract
Mutations in oncogenes such as KRAS and EGFR cause a high proportion of lung cancers. Drugs targeting these proteins cause tumor regression but ultimately fail to elicit cures. As a result, there is an intense interest in how to best combine targeted therapies with other treatments, such as immunotherapies. However, preclinical systems for studying the interaction of lung tumors with the host immune system are inadequate, in part due to the low tumor mutational burden in genetically engineered mouse models. Here we set out to develop mouse models of mutant KRAS-driven lung cancer with an elevated tumor mutational burden by expressing the human DNA cytosine deaminase, APOBEC3B, to mimic the mutational signature seen in human lung cancer. This failed to substantially increase clonal tumor mutational burden and autochthonous tumors remained refractory to immunotherapy. However, establishing clonal cell lines from these tumors enabled the generation of an immunogenic syngeneic transplantation model of KRAS-mutant lung adenocarcinoma that was sensitive to immunotherapy. Unexpectedly, antitumor immune responses were not directed against neoantigens but instead targeted derepressed endogenous retroviral antigens. The ability of KRASG12C inhibitors to cause regression of KRASG12C -expressing tumors was markedly potentiated by the adaptive immune system, highlighting the importance of using immunocompetent models for evaluating targeted therapies. Overall, this model provides a unique opportunity for the study of combinations of targeted and immunotherapies in immune-hot lung cancer. SIGNIFICANCE This study develops a mouse model of immunogenic KRAS-mutant lung cancer to facilitate the investigation of optimal combinations of targeted therapies with immunotherapies.
Collapse
Affiliation(s)
| | | | - Emily K. Law
- Department of Biochemistry, Molecular Biology and Biophysics,
University of Minnesota, Minneapolis, MN, USA, 55455
- Howard Hughes Medical Institute, University of Minnesota,
Minneapolis, MN, USA, 55455
| | | | | | - Kevin Ng
- Retroviral Immunology Laboratory
| | | | | | - Sareena Rana
- Oncogene Biology Laboratory
- Lung Cancer Group, Division of Molecular Pathology, Institute of
Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | | | - Miguel Murillo
- Oncogene Biology Laboratory
- Lung Cancer Group, Division of Molecular Pathology, Institute of
Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | | | - Prokopios P. Argyris
- Department of Biochemistry, Molecular Biology and Biophysics,
University of Minnesota, Minneapolis, MN, USA, 55455
- Division of Oral and Maxillofacial Pathology, School of Dentistry,
University of Minnesota, Minneapolis, MN, USA, 55455
| | - William L. Brown
- Department of Biochemistry, Molecular Biology and Biophysics,
University of Minnesota, Minneapolis, MN, USA, 55455
- Institute for Molecular Virology, University of Minnesota,
Minneapolis, MN, USA, 55455
| | - Cameron Durfee
- Department of Biochemistry, Molecular Biology and Biophysics,
University of Minnesota, Minneapolis, MN, USA, 55455
- Institute for Molecular Virology, University of Minnesota,
Minneapolis, MN, USA, 55455
- Department of Biochemistry and Structural Biology, University of
Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Lindsay K. Larson
- Department of Biochemistry, Molecular Biology and Biophysics,
University of Minnesota, Minneapolis, MN, USA, 55455
- Institute for Molecular Virology, University of Minnesota,
Minneapolis, MN, USA, 55455
| | - Rachel I. Vogel
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN,
USA, 55455
- Department of Obstetrics, Gynecology, and Women’s Health,
University of Minnesota, Minneapolis, MN, USA, 55455
| | - Alejandro Suárez-Bonnet
- Experimental Histopathology, Francis Crick Institute, 1 Midland
Road, London NW1 1AT, UK
- Department of Pathobiology and Population Sciences, Royal Veterinary
College, Hatfield, AL9 7TA, UK
| | - Simon L. Priestnall
- Experimental Histopathology, Francis Crick Institute, 1 Midland
Road, London NW1 1AT, UK
- Department of Pathobiology and Population Sciences, Royal Veterinary
College, Hatfield, AL9 7TA, UK
| | | | | | | | | | | | - Reuben Harris
- Department of Biochemistry, Molecular Biology and Biophysics,
University of Minnesota, Minneapolis, MN, USA, 55455
- Howard Hughes Medical Institute, University of Minnesota,
Minneapolis, MN, USA, 55455
- Department of Biochemistry and Structural Biology, University of
Texas Health San Antonio, San Antonio, TX 78229, USA
- Howard Hughes Medical Institute, University of Texas Health San
Antonio, San Antonio, TX 78229, USA
| | - Julian Downward
- Oncogene Biology Laboratory
- Lung Cancer Group, Division of Molecular Pathology, Institute of
Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| |
Collapse
|
48
|
Borchmann S, Selenz C, Lohmann M, Ludwig H, Gassa A, Brägelmann J, Lohneis P, Meder L, Mattlener J, Breid S, Nill M, Fassunke J, Wisdom AJ, Compes A, Gathof B, Alakus H, Kirsch D, Hekmat K, Büttner R, Reinhardt HC, Hallek M, Ullrich RT. Tripartite antigen-agnostic combination immunotherapy cures established poorly immunogenic tumors. J Immunother Cancer 2022; 10:e004781. [PMID: 36223955 PMCID: PMC9562723 DOI: 10.1136/jitc-2022-004781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Single-agent immunotherapy has shown remarkable efficacy in selected cancer entities and individual patients. However, most patients fail to respond. This is likely due to diverse immunosuppressive mechanisms acting in a concerted way to suppress the host anti-tumor immune response. Combination immunotherapy approaches that are effective in such poorly immunogenic tumors mostly rely on precise knowledge of antigenic determinants on tumor cells. Creating an antigen-agnostic combination immunotherapy that is effective in poorly immunogenic tumors for which an antigenic determinant is not known is a major challenge. METHODS We use multiple cell line and poorly immunogenic syngeneic, autochthonous, and autologous mouse models to evaluate the efficacy of a novel combination immunotherapy named tripartite immunotherapy (TRI-IT). To elucidate TRI-ITs mechanism of action we use immune cell depletions and comprehensive tumor and immune infiltrate characterization by flow cytometry, RNA sequencing and diverse functional assays. RESULTS We show that combined adoptive cellular therapy (ACT) with lymphokine-activated killer cells, cytokine-induced killer cells, Vγ9Vδ2-T-cells (γδ-T-cells) and T-cells enriched for tumor recognition (CTLs) display synergistic antitumor effects, which are further enhanced by cotreatment with anti-PD1 antibodies. Most strikingly, the full TRI-IT protocol, a combination of this ACT with anti-PD1 antibodies, local immunotherapy of agonists against toll-like receptor 3, 7 and 9 and pre-ACT lymphodepletion, eradicates and induces durable anti-tumor immunity in a variety of poorly immunogenic syngeneic, autochthonous, as well as autologous humanized patient-derived models. Mechanistically, we show that TRI-IT coactivates adaptive cellular and humoral, as well as innate antitumor immune responses to mediate its antitumor effect without inducing off-target toxicity. CONCLUSIONS Overall, TRI-IT is a novel, highly effective, antigen-agnostic, non-toxic combination immunotherapy. In this study, comprehensive insights into its preclinical efficacy, even in poorly immunogenic tumors, and mode of action are given, so that translation into clinical trials is the next step.
Collapse
Affiliation(s)
- Sven Borchmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Carolin Selenz
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Mia Lohmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Hanna Ludwig
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Asmae Gassa
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Johannes Brägelmann
- Mildred Scheel School of Oncology, University Hospital Cologne, Medical Faculty, Cologne, Germany
| | - Philipp Lohneis
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Lydia Meder
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Julia Mattlener
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Sara Breid
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Marieke Nill
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Jana Fassunke
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Amy J Wisdom
- Department of Radiation Oncology and Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Anik Compes
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Birgit Gathof
- Institute of Transfusion Medicine, University of Cologne, Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - David Kirsch
- Department of Radiation Oncology and Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Khosro Hekmat
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | | | - H Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen,University Duisburg-Essen, German Cancer Consortium (DKTK partner site Essen), Essen, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Roland T Ullrich
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
49
|
Jagodinsky JC, Bates AM, Clark PA, Sriramaneni RN, Havighurst TC, Chakravarty I, Nystuen EJ, Kim K, Sondel PM, Jin WJ, Morris ZS. Local TLR4 stimulation augments in situ vaccination induced via local radiation and anti-CTLA-4 checkpoint blockade through induction of CD8 T-cell independent Th1 polarization. J Immunother Cancer 2022; 10:e005103. [PMID: 36192087 PMCID: PMC9535200 DOI: 10.1136/jitc-2022-005103] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Radiation therapy (RT) has been demonstrated to generate an in situ vaccination (ISV) effect in murine models and in patients with cancer; however, this has not routinely translated into enhanced clinical response to immune checkpoint inhibition (ICI). We investigated whether the commonly used vaccine adjuvant, monophosphoryl lipid A (MPL) could augment the ISV regimen consisting of combination RT and ICI. MATERIALS/METHODS We used syngeneic murine models of melanoma (B78) and prostate cancer (Myc-CaP). Tumor-bearing mice received either RT (12 Gy, day 1), RT+anti-CTLA-4 (C4, day 3, 6, 9), MPL (20 µg IT injection days 5, 7, 9), RT+C4+MPL, or PBS control. To evaluate the effect of MPL on the irradiated tumor microenvironment, primary tumor with tumor draining lymph nodes were harvested for immune cell infiltration analysis and cytokine profiling, and serum was collected for analysis of antitumor antibody populations. RESULTS Combination RT+C4+MPL significantly reduced tumor growth, increased survival and complete response rate compared with RT+C4 in both B78 and Myc-CaP models. MPL favorably reprogrammed the irradiated tumor-immune microenvironment toward M1 macrophage and Th1 TBET+CD4+ T cell polarization. Furthermore, MPL significantly increased intratumoral expression of several Th1-associated and M1-associated proinflammatory cytokines. In co-culture models, MPL-stimulated macrophages directly activated CD8 T cells and polarized CD4 cells toward Th1 phenotype. MPL treatment significantly increased production of Th1-associated, IgG2c antitumor antibodies, which were required for and predictive of antitumor response to RT+C4+MPL, and enabled macrophage-mediated antibody-dependent direct tumor cell killing by MPL-stimulated macrophages. Macrophage-mediated tumor cell killing was dependent on FcγR expression. In metastatic models, RT and MPL generated a systemic antitumor immune response that augmented response to ICIs. This was dependent on macrophages and CD4+ but not CD8+T cells. CONCLUSIONS We report the potential for MPL to augment the ISV effect of combination RT+C4 through FcγR, macrophage, and TBET+CD4+ Th1 cell dependent mechanisms. To our knowledge, this is the first report describing generation of a CD8+ T cell-independent, Th1 polarized, systemic antitumor immune response with subsequent generation of immunologic memory. These findings support the potential for vaccine adjuvants to enhance the efficacy of in situ tumor vaccine approaches.
Collapse
Affiliation(s)
- Justin C Jagodinsky
- Department of Human Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Amber M Bates
- Department of Human Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Paul A Clark
- Department of Human Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Raghava N Sriramaneni
- Department of Human Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Thomas C Havighurst
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Ishan Chakravarty
- Department of Human Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Erin J Nystuen
- Department of Human Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Paul M Sondel
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Won Jong Jin
- Department of Human Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
50
|
Blair TC, Bambina S, Kramer GF, Dowdell AK, Alice AF, Baird JR, Lund AW, Piening BD, Crittenden MR, Gough MJ. Fluorescent tracking identifies key migratory dendritic cells in the lymph node after radiotherapy. Life Sci Alliance 2022; 5:e202101337. [PMID: 35487695 PMCID: PMC9058260 DOI: 10.26508/lsa.202101337] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022] Open
Abstract
Radiation therapy generates extensive cancer cell death capable of promoting tumor-specific immunity. Within the tumor, conventional dendritic cells (cDCs) are known to carry tumor-associated antigens to the draining lymph node (TdLN) where they initiate T-cell priming. How radiation influences cDC migration is poorly understood. Here, we show that immunological efficacy of radiation therapy is dependent on cDC migration in radioimmunogenic tumors. Using photoconvertible mice, we demonstrate that radiation impairs cDC migration to the TdLN in poorly radioimmunogenic tumors. Comparative transcriptional analysis revealed that cDCs in radioimmunogenic tumors express genes associated with activation of endogenous adjuvant signaling pathways when compared with poorly radioimmunogenic tumors. Moreover, an exogenous adjuvant combined with radiation increased the number of migrating cDCs in these poorly radioimmunogenic tumors. Taken together, our data demonstrate that cDC migration play a critical role in the response to radiation therapy.
Collapse
Affiliation(s)
- Tiffany C Blair
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Shelly Bambina
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Gwen F Kramer
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Alexa K Dowdell
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Alejandro F Alice
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Jason R Baird
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Amanda W Lund
- Ronald O Perelman Department of Dermatology, Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Brian D Piening
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Marka R Crittenden
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
- The Oregon Clinic, Portland, OR, USA
| | - Michael J Gough
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| |
Collapse
|