1
|
Ma L, Mao JH, Barcellos-Hoff MH. Systemic inflammation in response to radiation drives the genesis of an immunosuppressed tumor microenvironment. Neoplasia 2025; 64:101164. [PMID: 40184664 PMCID: PMC11999686 DOI: 10.1016/j.neo.2025.101164] [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: 12/29/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
The composition of the tumor immune microenvironment has become a major determinant of response to therapy, particularly immunotherapy. Clinically, a tumor microenvironment lacking lymphocytes, so-called "cold" tumors, are considered poor candidates for immune checkpoint inhibition. In this review, we describe the diversity of the tumor immune microenvironment in breast cancer and how radiation exposure alters carcinogenesis. We review the development and use of a radiation-genetic mammary chimera model to clarify the mechanism by which radiation acts. Using the chimera model, we demonstrate that systemic inflammation elicited by a low dose of radiation is key to the construction of an immunosuppressive tumor microenvironment, resulting in aggressive, rapidly growing tumors lacking lymphocytes. Our experimental studies inform the non-mutagenic mechanisms by which radiation affects cancer and provide insight into the genesis of cold tumors.
Collapse
Affiliation(s)
- Lin Ma
- Department of Stomatology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, School of Medicine, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94143 USA.
| |
Collapse
|
2
|
Maldonado MDM, Gracia-Hernandez M, Le LH, Iida M, Gulley JL, Donahue RN, Palena C, Schlom J, Hamilton DH. Combination of a therapeutic cancer vaccine targeting the endogenous retroviral envelope protein ERVMER34-1 with immune-oncology agents facilitates expansion of neoepitope-specific T cells and promotes tumor control. J Immunother Cancer 2025; 13:e011378. [PMID: 40360436 DOI: 10.1136/jitc-2024-011378] [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] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Endogenous retroviruses (ERVs) are remnants of retrovirus germline infections that occurred over the course of evolution and constitute between 5% and 8% of the human genome. While ERVs tend to be epigenetically silenced in normal adult human tissues, they are often overexpressed in carcinomas and may represent novel immunotherapeutic targets. This study characterizes the ERV envelope protein ERVMER34-1 as a target for a therapeutic cancer vaccine. METHODS The expression of ERVMER34-1 in multiple healthy adult and cancer tissues was assessed, as was its immunogenicity, to ascertain whether specific T cells could lyse human carcinoma cell lines expressing ERVMER34-1. Furthermore, the ability of a rationally designed ERVMER34-1-targeted therapeutic vaccine to induce tumor clearance in two murine carcinoma models expressing ERVMER34-1 was examined either as a monotherapy or in combination with anti-programmed cell death protein-1/programmed death-ligand 1 monoclonal antibody (mAb) or the interleukin-15 superagonist N-803. RESULTS The ERVMER34-1 protein was shown to be overexpressed in 232/376 of human carcinomas analyzed while being absent in most healthy adult tissues. High levels of ERVMER34-1 RNA expression associate with decreased survival in uveal melanoma, adenoid cystic, and head and neck carcinomas. ERVMER34-1-specific T cells were detected in peripheral blood mononuclear cells (PBMCs) of patients with cancer but not healthy donors following an overnight stimulation. However, reactive T cells are readily expanded from both healthy donor and patient with cancer PBMCs following a 7- day in vitro stimulation. Furthermore, ERVMER34-1-specific T cells selectively kill human carcinoma cell lines expressing ERVMER34-1. A novel, rationally designed, therapeutic cancer vaccine targeting ERVMER34-1 mediated tumor control in established syngeneic murine tumors expressing the full-length ERVMER34-1 protein. When combined with checkpoint blockade, the vaccine promoted expansion of neoepitope-reactive T cells whose function was further enhanced when combined with N-803. This expansion of neoepitope-reactive T cells was associated with tumor control. CONCLUSIONS This study reveals the potential of a vaccine that targets the retroviral envelope protein ERVMER34-1 and supports its continued development toward clinical testing as a new class of therapeutic cancer vaccine.
Collapse
Affiliation(s)
- Maria Del Mar Maldonado
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Maria Gracia-Hernandez
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Loc Huu Le
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Masafumi Iida
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - James L Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Renee N Donahue
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Claudia Palena
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Duane H Hamilton
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Wu X, Zhang J, Deng Z, Sun X, Zhang Y, Zhang C, Wang J, Yu X, Yang G. Bacteria-based biohybrids for remodeling adenosine-mediated immunosuppression to boost radiotherapy-triggered antitumor immune response. Biomaterials 2025; 316:123000. [PMID: 39674101 DOI: 10.1016/j.biomaterials.2024.123000] [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: 07/17/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
Radiotherapy (RT) can trigger immunogenic cell death (ICD) in tumor cells and release adenosine triphosphate (ATP) to activate antitumor immunity. However, the formation of immunosuppressive adenosine (ADO) mediated by ectonucleotidases including CD39 and CD73, can exacerbate the immunosuppressive effects. Herein, a radiosensitizer-based metal-organic framework (MOF) composed of bismuth (Bi) and ellagic acid (EA) was synthesized in situ on the surface of Escherichia coli Nissle 1917 (EcN) to serve as a carrier for the CD39 inhibitor sodium polyoxotungstate (POM-1). This therapeutic platform, acting as a radiosensitizer, significantly enhances cytotoxicity against tumor cells while effectively inducing ICD and releasing high concentrations of ATP. Subsequently, the released POM-1 increases the levels of pro-inflammatory extracellular ATP while preventing tumor immunosuppression caused by the accumulation of ADO. Additionally, as a natural immune adjuvant, EcN further promotes the maturation of dendritic cells (DCs) and the infiltration of cytotoxic T lymphocytes (CTLs). As a result, such treatment initiates the destruction of established tumor growth and induces strong abscopal effects, leading to a significant inhibition of tumor metastases. This strategy presents a bacterial-based biohybrid system that facilitates RT-induced ICD while simultaneously limiting the degradation of ATP into ADO, thereby achieving sustained anti-tumor immunity.
Collapse
Affiliation(s)
- Xirui Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Junjun Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou 215004, China
| | - Zheng Deng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xianglong Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yifan Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Cai Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jiadong Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xinke Yu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Guangbao Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| |
Collapse
|
4
|
Yu L, Liu J, Fan Y, Hu X, Zeng X, Luo S, Chen P. The Radiosensitizing Effect of Tumor-Derived Microparticles Co-Loaded with Sorafenib and Gold Nanoparticles on Hepatocellular Carcinoma. Int J Nanomedicine 2025; 20:5489-5508. [PMID: 40321799 PMCID: PMC12047280 DOI: 10.2147/ijn.s509936] [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: 12/05/2024] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Objective Hepatocellular carcinoma (HCC) is a highly heterogeneous tumor with features such as high recurrence, easy metastasis, and poor prognosis, posing significant challenges for clinical treatment. In this study, we introduce a novel approach for treating HCC using tumor cell-derived microparticles (MPs) co-loaded with sorafenib and gold nanoparticles (AuNP) in combination with radiotherapy. Methods MPSF@AuNP was prepared by co-incubating AuNP with sorafenib, and was evaluated using dynamic light scattering (DLS), transmission electron microscopy (TEM), ultraviolet-visible spectrophotometry (UV-Vis), inductively coupled plasma optical emission spectrometry (ICP-OES), high-performance liquid chromatography (HPLC), and SDS-PAGE electrophoresis. Subsequently, their targeting ability toward hepatocellular carcinoma cells and their combined antitumor therapeutic effects with radiotherapy were investigated through in vitro and in vivo experiments, while their in vivo safety was also assessed. Results Our results demonstrate that co-loaded microparticles (MPSF@AuNP) can effectively deliver therapeutic agents to tumor cells through homologous targeting, improving the bioavailability of therapeutic drugs and enhancing their cytotoxicity against tumor cells. Furthermore, the combination of MPSF@AuNP with radiotherapy shows a synergistic anti-tumor effect by enhancing the inhibition of tumor cell proliferation, promoting tumor cell apoptosis, remodeling the tumor microenvironment, and activating the anti-tumor immune responses. Conclusion This study offers a promising treatment approach for malignant tumors such as HCC by using MP co-loaded and delivered with anti-tumor drugs and AuNP in combination with radiotherapy.
Collapse
Affiliation(s)
- Li Yu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Department of Oncology, Jiangsu Cancer Hospital, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Jiali Liu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Yiwen Fan
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Xiao Hu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Oncology Treatment Center, People’s Hospital of Yangjiang Affiliated to Guangdong Medical University, Yangjiang, Guangdong, 529500, People’s Republic of China
| | - Xiaonan Zeng
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Shan Luo
- Tianfu Jincheng Laboratory, Chengdu, 610212, People’s Republic of China
| | - Ping Chen
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| |
Collapse
|
5
|
Wang SY, Wu JX, An X, Yuan Z, Ren YF, Yu XF, Tian XD, Wei W. Structural and temporal dynamics analysis on immune response in low-dose radiation: History, research hotspots and emerging trends. World J Radiol 2025; 17:101636. [PMID: 40309477 PMCID: PMC12038408 DOI: 10.4329/wjr.v17.i4.101636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/19/2024] [Accepted: 03/27/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Radiotherapy (RT) is a cornerstone of cancer treatment. Compared with conventional high-dose radiation, low-dose radiation (LDR) causes less damage to normal tissues while potentially modulating immune responses and inhibiting tumor growth. LDR stimulates both innate and adaptive immunity, enhancing the activity of natural killer cells, dendritic cells, and T cells. However, the mechanisms underlying the effects of LDR on the immune system remain unclear. AIM To explore the history, research hotspots, and emerging trends in immune response to LDR literature over the past two decades. METHODS Publications on immune responses to LDR were retrieved from the Web of Science Core Collection. Bibliometric tools, including CiteSpace and HistCite, were used to identify historical features, active topics, and emerging trends in this field. RESULTS Analysis of 1244 publications over the past two decades revealed a significant surge in research on immune responses to LDR, particularly in the last decade. Key journals such as INR J Radiat Biol, Cancers, and Radiat Res published pivotal studies. Citation networks identified key studies by authors like Twyman-Saint Victor C (2015) and Vanpouille-Box C (2017). Keyword analysis revealed hotspots such as ipilimumab, stereotactic body RT, and targeted therapy, possibly identifying future research directions. Temporal variations in keyword clusters and alluvial flow maps illustrate the evolution of research themes over time. CONCLUSION This bibliometric analysis provides valuable insights into the evolution of studies on responses to LDR, highlights research trends, and identifies emerging areas for further investigation.
Collapse
Affiliation(s)
- Shu-Yuan Wang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Jia-Xing Wu
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xian An
- Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Zhen Yuan
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yi-Fan Ren
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiu-Feng Yu
- Department of General Medicine, Tuberculosis Hospital of Shaanxi Province, Xi’an 710105, Shaanxi Province, China
| | - Xiao-Dong Tian
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Wei Wei
- Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
6
|
Nahm WJ, Sakunchotpanit G, Nambudiri VE. Abscopal Effects and Immunomodulation in Skin Cancer Therapy. Am J Clin Dermatol 2025:10.1007/s40257-025-00943-x. [PMID: 40180765 DOI: 10.1007/s40257-025-00943-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/05/2025]
Abstract
Radiation therapy (RT) is a crucial modality in cancer treatment, functioning through direct DNA damage and immune stimulation. However, RT's effects extend beyond targeted cells, influencing neighboring cells through the bystander effect (ByE) and distant sites via the abscopal effect (AbE). The AbE, first described by Mole in 1953, encompasses biological reactions at sites distant from the irradiation field. While RT can enhance antitumor immune responses, it may also contribute to an immunosuppressive microenvironment. To address this limitation, combining RT with immune checkpoint inhibitors (ICIs) has gained renewed interest, aiming to amplify antitumor immune responses. Evidence of AbEs has been observed in various metastatic or advanced cutaneous cancers, including melanoma, basal cell carcinoma, cutaneous lymphoma, Merkel cell carcinoma, and cutaneous squamous cell carcinoma. Clinical studies suggest combining RT with ICIs targeting CTLA-4 and PD-1/PD-L1 may enhance AbE incidence in these cancers. This review primarily explores the current understanding of AbEs in skin cancers, briefly acknowledging the ByE focusing on combining RT with immunomodulation. It focuses on proposed mechanisms, preclinical and clinical evidence, challenges in clinical translation, and future directions for harnessing AbEs in managing advanced skin malignancies. Alternative modalities for inducing abscopal-like responses are also explored. While promising, challenges remain in consistently reproducing AbEs in clinical practice, necessitating further research to optimize treatment combinations, timing, and patient selection.
Collapse
Affiliation(s)
- William J Nahm
- New York University Grossman School of Medicine, New York, NY, USA.
- Department of Dermatology, Brigham and Women's Hospital, 117 Western Avenue, Boston, MA, 02163, USA.
| | - Goranit Sakunchotpanit
- Department of Dermatology, Brigham and Women's Hospital, 117 Western Avenue, Boston, MA, 02163, USA
- Tufts University School of Medicine, Boston, MA, USA
| | - Vinod E Nambudiri
- Department of Dermatology, Brigham and Women's Hospital, 117 Western Avenue, Boston, MA, 02163, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Revelant A, Gessoni F, Montico M, Dhibi R, Brisotto G, Casarotto M, Zanchetta M, Paduano V, Sperti F, Evangelista C, Giordari F, De Re V, Trovò M, Minatel E, Mascarin M, Steffan A, Muraro E. Radical hemithorax radiotherapy induces an increase in circulating PD-1 + T lymphocytes and in the soluble levels of PD-L1 in malignant pleural mesothelioma patients: a possible synergy with PD-1/PD-L1 targeting treatment? Front Immunol 2025; 16:1534766. [PMID: 40236706 PMCID: PMC11997449 DOI: 10.3389/fimmu.2025.1534766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/17/2025] [Indexed: 04/17/2025] Open
Abstract
Malignant Pleural Mesothelioma (MPM) is an aggressive tumor associated with asbestos exposure, characterized by a poor prognosis, managed with surgery, chemotherapy and radiotherapy. Recently, immunotherapy gives a survival advantage compared to chemotherapy, but limited to the non-epithelioid histotype, the rarest type. Radical hemithorax radiotherapy (RHRT) improves the Overall Survival (OS) of MPM patients, irrespective of histotype, and is able to induce immunomodulatory effects. In this study we aim to investigate changes in circulating T lymphocytes phenotype and activity, in MPM patients undergoing RHRT, to evaluate a possible therapeutic space for immunotherapy in this setting. To assess immunomodulatory effects of RHRT we evaluate peripheral blood samples of 35 MPM patients collected before treatment, at the end of RT, and 1 month later. We first notice that higher Lymphocyte-to-Monocyte Ratio (LMR) levels, before RT, are associated with an improved OS. The immune monitoring performed by ELISA assays reveals a significant increase in the serum levels of soluble PD-L1 (sPD-L1) and IFN-γ at the end of RHRT. Furthermore, the percentage of PD-1+ cells, evaluated by flow cytometry, significantly raise after RHRT in T cells, both CD4+ and CD8+. Also the proportion of proliferative cells is significantly expanded after RHRT in all T cell subtypes. After treatment we observe a significant increase in the number of patients showing WT-1 specific CD4+ T cells, measured by intracellular staining. The TCR repertoire analysis, investigated by Next Generation Sequencing, reveals an increased number of expanded T-cell clones after RHRT, and an association between TCR clonality and the percentage of proliferating cytotoxic T lymphocytes. The comparison of TCR sequences obtained in our cohort with those described in a literature cohort of MPM patients, reveals common entries, specific for MPM-associated antigens including WT-1. In this setting, pre-treatment levels of LMR seem to have a positive prognostic role, and RHRT would appear to induce immunomodulating effects, potential biomarkers for immunotherapy eligibility: i.e. increased PD-1+ T lymphocytes, proliferating T cells, expanded T cell clones and augmented levels of sPD-L1. These data suggest the design of a prospective study evaluating a maintenance immunotherapy after RHRT in MPM, even in the epithelioid histotype.
Collapse
Affiliation(s)
- Alberto Revelant
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Francesca Gessoni
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Marcella Montico
- Clinical Trial Office, Scientific Direction, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Raja Dhibi
- Immunopathology and Cancer Biomarkers Unit, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Giulia Brisotto
- Immunopathology and Cancer Biomarkers Unit, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Mariateresa Casarotto
- Immunopathology and Cancer Biomarkers Unit, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Martina Zanchetta
- Clinical Trial Office, Scientific Direction, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Veronica Paduano
- Biobank, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Filippo Sperti
- Biobank, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Chiara Evangelista
- Biobank, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Fabiana Giordari
- Immunopathology and Cancer Biomarkers Unit, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Valli De Re
- Immunopathology and Cancer Biomarkers Unit, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Marco Trovò
- Department of Radiation Oncology, Udine General Hospital, Udine, Italy
| | - Emilio Minatel
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Maurizio Mascarin
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers Unit, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Elena Muraro
- Immunopathology and Cancer Biomarkers Unit, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| |
Collapse
|
8
|
Yang Y, Yang C, Deng K, Xiao Y, Liu X, Du Z. Nucleic Acid Drugs in Radiotherapy. Chembiochem 2025; 26:e202400854. [PMID: 39903093 DOI: 10.1002/cbic.202400854] [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: 10/15/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/06/2025]
Abstract
Radiotherapy remains a cornerstone of cancer treatment, using high-energy radiation to induce DNA damage in tumor cells, leading to cell death. However, its efficacy is often hindered by challenges such as radiation resistance and side effects. As a powerful class of functional molecules, nucleic acid drugs (NADs) present a promising solution to these limitations. Engineered to target key pathways like DNA repair and tumor hypoxia, NADs can enhance radiotherapy sensitivity. NADs can also serve as delivery vehicles for radiotherapy agents such as radionuclides, improving targeting accuracy and minimizing side effects. This review explores the role of NADs in optimizing radiotherapy, highlighting their mechanisms, clinical applications, and synergies with radiotherapy, ultimately offering a promising strategy for improving patient outcomes in cancer therapy.
Collapse
Affiliation(s)
- Yuying Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Cai Yang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Kai Deng
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Yating Xiao
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, Universities and Colleges Admissions Service (UCAS), Hangzhou, 310024, China
| | - Xiangsheng Liu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Zhen Du
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
9
|
Moore J, Gkantalis J, Guix I, Chou W, Yuen K, Lazar AA, Spitzer M, Combes A, Barcellos-Hoff MH. Identification of a conserved subset of cold tumors responsive to immune checkpoint blockade. J Immunother Cancer 2025; 13:e010528. [PMID: 40050047 PMCID: PMC11887281 DOI: 10.1136/jitc-2024-010528] [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: 09/06/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The efficacy of immune checkpoint blockade (ICB) depends on restoring immune recognition of cancer cells that have evaded immune surveillance. Transforming growth factor-beta (TGFβ) is associated with immune-poor, so-called cold tumors whereas loss of its signaling promotes DNA misrepair that could stimulate immune response. METHODS We analyzed transcriptomic data from IMvigor210, The Cancer Genome Atlas, and Tumor Immune Syngeneic MOuse data sets to evaluate the predictive value of high βAlt, a score representing low expression of a signature consisting of TGFβ targets and high expression of genes involved in error-prone DNA repair. The immune context of βAlt was assessed by evaluating tumor-educated immune signatures. An ICB-resistant, high βAlt preclinical tumor model was treated with a TGFβ inhibitor, radiation, and/or ICB and assessed for immune composition and tumor control. RESULTS We found that a high βAlt score predicts ICB response yet is paradoxically associated with an immune-poor tumor microenvironmentcancer in both human and mouse tumors. We postulated that high βAlt cancers consist of cancer cells in which loss of TGFβ signaling generates a TGFβ rich, immunosuppressive tumor microenvironment. Accordingly, preclinical modeling showed that TGFβ inhibition followed by radiotherapy could convert an immune-poor, high βAlt tumor to an immune-rich, ICB-responsive tumor. Mechanistically, TGFβ inhibition increased activated natural killer (NK) cells, which were required to recruit lymphocytes to respond to ICB in irradiated tumors. NK cell activation signatures were also increased in high βAlt, cold mouse and human tumors that responded to ICB. CONCLUSIONS These studies indicate that loss of TGFβ signaling competency and gain of error-prone DNA repair identifies a subset of cold tumors that are responsive to ICB. Our mechanistic studies show that inhibiting TGFβ activity can convert a high βAlt, cold tumor into ICB-responsive tumors via NK cells. A biomarker consisting of combined TGFβ, DNA repair, and immune context signatures is a means to prospectively identify patients whose cancers may be converted from cold to hot with appropriate therapy.
Collapse
Affiliation(s)
- Jade Moore
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Jim Gkantalis
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Ines Guix
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - William Chou
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Kobe Yuen
- Oncology Biomarker Development, Genentech, South San Francisco, California, USA
| | - Ann A Lazar
- Division of Oral Epidemiology and Division of Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Matthew Spitzer
- Depts of Otolaryngology-Head and Neck Surgery and of Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
| | - Alexis Combes
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
10
|
Huang B, Liu S, Wang K, Zhao J, Li M, Wang X, Wang W, Wang X, Yu J, Meng X, Cai G. Addition of thoracic radiotherapy to a PD-L1 inhibitor plus chemotherapy regimen delays brain metastasis onset in extensive-stage small cell lung cancer patients without baseline brain metastasis. Respir Res 2025; 26:85. [PMID: 40045282 PMCID: PMC11883939 DOI: 10.1186/s12931-025-03157-1] [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: 08/11/2024] [Accepted: 02/15/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND With the application of immune checkpoint inhibitors (ICIs) and the discovery of the synergistic effect of radiotherapy and immunotherapy, the intracranial benefit of thoracic radiotherapy (TRT) is receiving signiffcant clinical attention. The purpose of this study was to analyze the cranial benefits of ICIs and TRT in patients with extensive-stage small cell lung cancer (ES-SCLC) without baseline brain metastases (BMs). MATERIALS AND METHODS From August 2019 to August 2022, data from patients diagnosed with ES-SCLC without baseline BMs were retroactively recorded. The Kaplan‒Meier method was used to calculate overall survival (OS), progression-free survival (PFS), and brain metastasis-free survival (BMFS), and the differences between the treatment groups were compared with the log-rank test. Risk factors associated with OS were analyzed via the Cox regression model. RESULTS A total of 216 patients were included, with a median follow-up of 24.73 months. Among these patients, 137 (63.4%) received first-line ICIs combined with chemotherapy (ChT), including 32 patients treated with anti-programmed death 1 antibody (αPD-1) and 105 patients treated with anti-programmed death-ligand 1 antibody (αPD-L1), and 79 patients (36.6%) received first-line ChT alone. Compared with the ChT-alone group, the ICI + ChT group demonstrated significantly improved PFS (8.07 vs. 6.87 months; p < 0.001) and OS (19.83 vs. 13.80 months; p = 0.001). The addition of ICIs to the ChT regimen did not significantly delay the onset of BMs compared to that with ChT alone (16.93 vs. 12.67 months; p = 0.379). Notably, the addition of TRT to the αPD-L1 + ChT regimen significantly prolonged BMFS compared to that without TRT (20.27 vs. 8.80 months; p = 0.045). CONCLUSION In patients with ES-SCLC without baseline BMs, first-line chemoimmunotherapy significantly improves PFS and OS. However, it does not delay intracranial metastasis. The addition of TRT to αPD-L1 + ChT therapy significant delays the development of BMs. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Baiyang Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Jinan, Shandong, 250117, China
| | - Senyuan Liu
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Kaiyue Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Jinan, Shandong, 250117, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Jiarui Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Jinan, Shandong, 250117, China
| | - Min Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Jinan, Shandong, 250117, China
| | - Xingpeng Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Jinan, Shandong, 250117, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Weiqing Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Jinan, Shandong, 250117, China
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaohan Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Jinan, Shandong, 250117, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Jinan, Shandong, 250117, China
| | - Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Jinan, Shandong, 250117, China
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Guoxin Cai
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Jinan, Shandong, 250117, China.
| |
Collapse
|
11
|
Lee D, Oh S, Lawler S, Kim Y. Bistable dynamics of TAN-NK cells in tumor growth and control of radiotherapy-induced neutropenia in lung cancer treatment. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2025; 22:744-809. [PMID: 40296792 DOI: 10.3934/mbe.2025028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Neutrophils play a crucial role in the innate immune response as a first line of defense in many diseases, including cancer. Tumor-associated neutrophils (TANs) can either promote or inhibit tumor growth in various steps of cancer progression via mutual interactions with cancer cells in a complex tumor microenvironment (TME). In this study, we developed and analyzed mathematical models to investigate the role of natural killer cells (NK cells) and the dynamic transition between N1 and N2 TAN phenotypes in killing cancer cells through key signaling networks and how adjuvant therapy with radiation can be used in combination to increase anti-tumor efficacy. We examined the complex immune-tumor dynamics among N1/N2 TANs, NK cells, and tumor cells, communicating through key extracellular mediators (Transforming growth factor (TGF-$ \beta $), Interferon gamma (IFN-$ \gamma $)) and intracellular regulation in the apoptosis signaling network. We developed several tumor prevention strategies to eradicate tumors, including combination (IFN-$ \gamma $, exogenous NK, TGF-$ \beta $ inhibitor) therapy and optimally-controlled ionizing radiation in a complex TME. Using this model, we investigated the fundamental mechanism of radiation-induced changes in the TME and the impact of internal and external immune composition on the tumor cell fate and their response to different treatment schedules.
Collapse
Affiliation(s)
- Donggu Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Republic of Korea
| | - Sunju Oh
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Sean Lawler
- Department of Pathology and Laboratory Medicine, Legorreta Brown Cancer Center, Brown University, Providence, RI 02912, USA
| | - Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul 05029, Republic of Korea
- Department of Pathology and Laboratory Medicine, Legorreta Brown Cancer Center, Brown University, Providence, RI 02912, USA
| |
Collapse
|
12
|
Nachankar A, Pelak M, Schafasand M, Martino G, Tubin S, Hug E, Carlino A, Lütgendorf-Caucig C, Stock M, Fossati P. Carbon-Ion Radiotherapy for Head and Neck Mucosal Melanoma: Preliminary Clinical Outcomes and the MedAustron Approach for Reporting RBE-Weighted Dose With 2 Models. Int J Part Ther 2025; 15:100738. [PMID: 39896178 PMCID: PMC11786705 DOI: 10.1016/j.ijpt.2025.100738] [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: 11/19/2024] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Purpose Head and neck mucosal melanomas (HNMMs) are aggressive, radiotherapy-resistant cancers. Previous JCROS studies demonstrated improved local control with carbon-ion radiotherapy (CIRT). This study evaluates early outcomes of CIRT for HNMM using the European and Japanese relative biological effectiveness (RBE)-adapted dose prescriptions. Materials and Methods Between November 2019 and April 2023, 14 HNMM patients received CIRT treatment. Postoperative CIRT for R2 resection: 9 cases; biopsies only: 5 cases. Immune checkpoint inhibitors used as primary treatment: 6 cases; salvage: 8 cases. CIRT delivered in DRBE dose of 68.8 (64.5-68.8) Gy (RBE)/16 fractions, optimized with the local effect model I (LEM-I, European) for RBE-weighted dose, recalculated using the modified-microdosimetric kinetic model (mMKM, Japanese). Results HNMM tumor and nodal stages: cT3: 2 (14%), cT4: 12 (86%), cN1: 1 (7%). The median follow-up was 22 months (range, 4-54). The 2-year local recurrence-free survival, regional recurrence-free survival, overall survival, and distant metastasis-free survival were 100%, 89% (CI, 71-100), 64% (CI, 44-95), and 43% (CI, 22-84), respectively. The median relative volumetric tumor regression at 3, 6, and 12 months post-CIRT was 40%, 63%, and 72%, respectively. CIRT-associated late toxicities were G3 mucositis: 2 (14%) and G3 anosmia: 1 (7%). The immune checkpoint inhibition-related late toxicities were G2 hypophysitis: 1 (11%) and G3 peripheral neuropathy: 1 (11%). The average attainable DRBE coverage for 95% of high-dose clinical target volume was 63.2 ± 6 Gy (RBE) (LEM-I) and 57.4 ± 5 Gy (RBE) (mMKM). The LETd distribution in high-dose clinical target volume was satisfactory, LETd50% (median) = 57.3 ± 6 keV/µm and LETd98% (near minimum) = 46.5 ± 6.1 keV/µm. Conclusion Bi-RBE model (LEM-I, mMKM) optimized CIRT protocol improved dose comparability of plans between different systems. It also improved intratumoral LETd distribution and resulted in rapid tumor regression, favorable toxicity profile, and excellent early loco-regional control. It provides a promising alternative to surgery, though distant metastasis remains the key prognostic factor.
Collapse
Affiliation(s)
- Ankita Nachankar
- ACMIT Gmbh, Wiener Neustadt, Austria
- Department of Radiation Oncology, MedAustron Ion Therapy Center, Wiener Neustadt, Austria
| | - Maciej Pelak
- University Clinic for Radiotherapy and Radio-Oncology of the Paracelsus Medical University of Salzburg, Austria
| | - Mansure Schafasand
- Department of Medical Physics, MedAustron Ion Therapy Center, Wiener Neustadt, Austria
- Division Medical Physics, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| | - Giovanna Martino
- Department of Medical Physics, MedAustron Ion Therapy Center, Wiener Neustadt, Austria
| | - Slavisa Tubin
- Department of Radiation Oncology, MedAustron Ion Therapy Center, Wiener Neustadt, Austria
| | - Eugen Hug
- Department of Radiation Oncology, MedAustron Ion Therapy Center, Wiener Neustadt, Austria
| | - Antonio Carlino
- Department of Medical Physics, MedAustron Ion Therapy Center, Wiener Neustadt, Austria
| | | | - Markus Stock
- Department of Medical Physics, MedAustron Ion Therapy Center, Wiener Neustadt, Austria
- Division Medical Physics, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| | - Piero Fossati
- Department of Radiation Oncology, MedAustron Ion Therapy Center, Wiener Neustadt, Austria
- Division Radiation Oncology, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| |
Collapse
|
13
|
Zhou L, Yu G, Shen Y, Wen R, Ding H, Zhou J, Zhu X, Hong Y, Gong H, Liu L, Wang H, Zhang H, Bai C, Hao L, Zhang W. Safety and clinical efficacy of neoadjuvant chemoradiation therapy with immunotherapy for organ preservation in ultra-low rectal cancer: preliminary results of the CHOICE-I trial: a prospective cohort study. Int J Surg 2025; 111:2487-2494. [PMID: 39764608 DOI: 10.1097/js9.0000000000002225] [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: 07/16/2024] [Accepted: 12/05/2024] [Indexed: 03/16/2025]
Abstract
OBJECTIVE To explore the safety and efficacy of neoadjuvant chemoradiotherapy (nCRT) combined with a PD-1 antibody in improving complete clinical response (cCR) and organ preservation in patients with ultra-low rectal cancer. METHODS This was a prospective phase II, single-arm, open-label trial. Patients with confirmed pMMR status T 1-3a N 0-1 M 0 retcal adenocarcinoma were included. Long-course chemoradiotherapy was delivered to a dose of 50 Gy. A PD-1 antibody was added 2 weeks after the first radiotherapy session, and two courses were administered. After chemoradiotherapy, CapeOX plus PD-1 antibody was administered to patients for two cycles. After evaluation, patients with cCR were managed with a watch-and-wait (W&W) approach. Local excision or a W&W approach was performed for patients with near complete clinical response (ncCR) as per multidisciplinary team decision. Radical surgery was recommended for poorly regressed or progressed tumors. RESULTS Twenty-five patients were enrolled, but two patients withdrew from the study. A total of 23 patients completed the entire neoadjuvant therapy. Ten and five patients achieved cCR and ncCR, respectively, and the rest had a partial clinical response. Patients with cCR were managed with W&W. Four patients with ncCR underwent local excision and were managed using W&W. Eight patients with partial clinical response underwent anus-preserving surgery. At the last follow-up, the rectum and anus preservation rates were 63.4% (14/22) and 95.5% (21/22), respectively. CONCLUSION nCRT combined with immunotherapy tended to achieve better cCR and rectum preservation rates with good tolerance in patients.
Collapse
Affiliation(s)
- Leqi Zhou
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guanyu Yu
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yuxin Shen
- Department of Radiation Oncology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Rongbo Wen
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Haibo Ding
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jidian Zhou
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaoming Zhu
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yonggang Hong
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Haifeng Gong
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lianjie Liu
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hao Wang
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Huojun Zhang
- Department of Radiation Oncology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chenguang Bai
- Department of Pathology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Liqiang Hao
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Wei Zhang
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
14
|
Moore J, Gkantalis J, Guix I, Chou W, Yuen K, Lazar AA, Spitzer M, Combes AJ, Barcellos-Hoff MH. A conserved subset of cold tumors responsive to immune checkpoint blockade. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.06.583752. [PMID: 38496519 PMCID: PMC10942434 DOI: 10.1101/2024.03.06.583752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Background The efficacy of immune checkpoint blockade (ICB) depends on restoring immune recognition of cancer cells that have evaded immune surveillance. At the time of diagnosis, patients with lymphocyte-infiltrated cancers are the most responsive to ICB, yet a considerable fraction of patients have immune-poor tumors. Methods We analyzed transcriptomic data from IMvigor210, TCGA, and TISMO datasets to evaluate the predictive value of βAlt, a score representing the negative correlation of signatures consisting of transforming growth factor beta (TGFβ) targets and genes involved in error-prone DNA repair. The immune context of βAlt was assessed by evaluating tumor-educated immune signatures. An ICB-resistant, high βAlt preclinical tumor model was treated with a TGFβ inhibitor, radiation, and/or ICB and assessed for immune composition and tumor control. Results Here, we show that high βAlt is associated with an immune-poor context yet is predictive of ICB response in both humans and mice. A high βAlt cancer in which TGFβ signaling is compromised generates a TGFβ rich, immunosuppressive tumor microenvironment. Accordingly, preclinical modeling showed that TGFβ inhibition followed by radiotherapy could convert an immune-poor, ICB-resistant tumor to an immune-rich, ICB-responsive tumor. Mechanistically, TGFβ blockade in irradiated tumors activated natural killer cells that were required to recruit lymphocytes to respond to ICB. In support of this, natural killer cell activation signatures were also increased in immune-poor mouse and human tumors that responded to ICB. Conclusions These studies suggest that loss of TGFβ competency identifies a subset of cold tumors that are candidates for ICB. Our mechanistic studies show that inhibiting TGFβ activity converts high βAlt, cold tumors into ICB-responsive tumors via NK cells. Thus, a biomarker consisting of combined TGFβ, DNA repair, and immune context signatures provides a means to prospectively identify patients whose cancers may be converted from 'cold' to 'hot,' which could be exploited for therapeutic treatment.
Collapse
Affiliation(s)
- Jade Moore
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- A member of the imCORE Network
| | - Jim Gkantalis
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Ines Guix
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - William Chou
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Kobe Yuen
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - Ann A. Lazar
- Division of Oral Epidemiology and Division of Biostatistics, University of California, San Francisco, CA, USA
| | - Mathew Spitzer
- Parker Institute for Cancer Immunotherapy, Department of Otolaryngology-Head and Neck Surgery, Department of Microbiology and Immunology, University of California, San Francisco, CA USA
- A member of the imCORE Network
| | - Alexis J. Combes
- Department of Pathology, CoLabs, University of California, San Francisco, San Francisco, CA, USA
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- A member of the imCORE Network
| |
Collapse
|
15
|
Grambozov B, Stana M, Zehentmayr F. Immediate start of durvalumab after chemoradiotherapy in unresectable non-small cell lung cancer UICC stage III: early results from the TORG1937/DATE study. J Thorac Dis 2025; 17:1-4. [PMID: 39975756 PMCID: PMC11833581 DOI: 10.21037/jtd-24-1638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/18/2024] [Indexed: 02/21/2025]
Affiliation(s)
- Brane Grambozov
- Department of Radiation Oncology, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Markus Stana
- Department of Radiation Oncology, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Franz Zehentmayr
- Department of Radiation Oncology, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
16
|
Charalampopoulou A, Barcellini A, Magro G, Bellini A, Borgna SS, Fulgini G, Ivaldi GB, Mereghetti A, Orlandi E, Pullia MG, Savazzi S, Tabarelli De Fatis P, Volpi G, Facoetti A. Advancing Radiobiology: Investigating the Effects of Photon, Proton, and Carbon-Ion Irradiation on PANC-1 Cells in 2D and 3D Tumor Models. Curr Oncol 2025; 32:49. [PMID: 39851965 PMCID: PMC11763791 DOI: 10.3390/curroncol32010049] [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: 12/01/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Introduction: Pancreatic cancer (PC) is one of the most aggressive and lethal malignancies, calling for enhanced research. Pancreatic ductal adenocarcinoma (PDAC) represents 70-80% of all cases and is known for its resistance to conventional therapies. Carbon-ion radiotherapy (CIRT) has emerged as a promising approach due to its ability to deliver highly localized doses and unique radiobiological properties compared to X-rays. In vitro radiobiology has relied on two-dimensional (2D) cell culture models so far; however, these are not sufficient to replicate the complexity of the in vivo tumor architecture. Three-dimensional (3D) models become a paradigm shift, surpassing the constraints of traditional models by accurately re-creating morphological, histological, and genetic characteristics as well as the interaction of tumour cells with the microenvironment. Materials and Methods: This study investigates the survival of pancreatic cancer cells in both 2D and spheroids, a 3D model, following photon, proton, and carbon-ion irradiation by means of clonogenic, MTT, spheroid growth, and vitality assays. Results: Our results demonstrate that carbon ions are more efficient in reducing cancer cell survival compared to photons and protons. In 2D cultures, carbon-ion irradiation reduced cell survival to approximately 15%, compared to 45% with photons and 30% with protons. In the 3D culture model, spheroid growth was similarly inhibited by carbon-ion irradiation; however, the overall survival rates were higher across all irradiation modalities compared to the 2D cultures. Carbon ions consistently showed the highest efficacy in reducing cell viability in both models. Conclusions: Our research highlights the pivotal role of 3D models in unraveling the complexities of pancreatic cancer radiobiology, offering new avenues for designing more effective and precise treatment protocols.
Collapse
Affiliation(s)
- Alexandra Charalampopoulou
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
- Hadron Academy PhD Course, School for Advanced Studies (IUSS), 27100 Pavia, Italy
| | - Amelia Barcellini
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (E.O.)
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Giuseppe Magro
- Medical Physics Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy;
| | - Anna Bellini
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
| | - Sara Sevan Borgna
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
| | - Giorgia Fulgini
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
| | - Giovanni Battista Ivaldi
- Radiation Oncology Department, Clinical Scientific Institutes Maugeri IRCCS, 27100 Pavia, Italy;
| | - Alessio Mereghetti
- Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.M.); (M.G.P.); (S.S.)
| | - Ester Orlandi
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (E.O.)
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Marco Giuseppe Pullia
- Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.M.); (M.G.P.); (S.S.)
| | - Simone Savazzi
- Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.M.); (M.G.P.); (S.S.)
| | | | - Gaia Volpi
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
- Hadron Academy PhD Course, School for Advanced Studies (IUSS), 27100 Pavia, Italy
| | - Angelica Facoetti
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
| |
Collapse
|
17
|
Xu J, Wang H, Zhang C, Jin SH, Chen X, Tan F, Frey B, Hecht M, Sun JG, Gaipl US, Ma H, Zhou JG. Efficacy of radiotherapy combined with atezolizumab or docetaxel in patients with previously treated NSCLC. iScience 2024; 27:111363. [PMID: 39640586 PMCID: PMC11617966 DOI: 10.1016/j.isci.2024.111363] [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: 08/20/2024] [Revised: 10/01/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024] Open
Abstract
Radiotherapy showed synergy with immunotherapy, yet the comparative effectiveness of combining immunotherapy (iRT) or chemotherapy (CRT) after platinum therapy failure in advanced non-small cell lung cancer (NSCLC) remains unexplored. We analyzed 163 patients (iRT: n = 120 vs. CRT: n = 43) eligible for combination radiotherapy. Before matching, median overall survival (OS) was significantly longer in iRT group (7.79 vs. 4.57 months, hazard ratio [HR]: 0.62, 95% confidence interval [CI]: 0.41-0.94, p = 0.024). After 1:2 propensity score matching (PSM) and inverse probability of treatment weighting (IPTW), iRT group showed improved OS, consistent with unmatched analysis (PSM, p = 0.033 and IPTW, p = 0.035). Exploratory analysis suggested that PD1+, central memory PD1+, and effector memory PD-L1+ CD4+ T cells were strong predictive biomarkers for iRT-treated patients (P OS = 0.025, P OS = 0.002, P OS = 0.010, respectively). Proliferative CD4+ T celllow was a prognostic (P OS = 0.008) and predictive biomarker for iRT (P OS < 0.001). Our work revealed iRT was prolonged OS in previously treated advanced NSCLC patients. Additionally, proliferative CD4+ T cell served as prognostic and predictive biomarkers.
Collapse
Affiliation(s)
- Junzhu Xu
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
| | - Haitao Wang
- Thoracic Surgery Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| | - Chi Zhang
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
| | - Su-Han Jin
- Department of Orthodontics, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaofei Chen
- Oncology Biometrics, AstraZeneca, Gaithersburg, MD 20850, USA
| | - Fangya Tan
- Harrisburg University of Science and Technology, Harrisburg, PA 17101, USA
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander- Universität Erlangen-Nurnberg, Erlangen, Germany
| | - Markus Hecht
- Department of Radiotherapy and Radiation Oncology, Saarland University Medical Center, Homburg, Germany
| | - Jian-Guo Sun
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Udo S. Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander- Universität Erlangen-Nurnberg, Erlangen, Germany
| | - Hu Ma
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
| | - Jian-Guo Zhou
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander- Universität Erlangen-Nurnberg, Erlangen, Germany
| |
Collapse
|
18
|
Georgescu A, Oved JH, Galarraga JH, Cantrell T, Mehta S, Dulmovits BM, Olson TS, Fattahi P, Wang A, Candarlioglu PL, Muvaffak A, Kim MM, Aydin SA, Seo J, Diffenderfer ES, Lynch A, Worthen GS, Huh DD. Self-organization of the hematopoietic vascular niche and emergent innate immunity on a chip. Cell Stem Cell 2024; 31:1847-1864.e6. [PMID: 39642865 DOI: 10.1016/j.stem.2024.11.003] [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: 02/01/2024] [Revised: 07/04/2024] [Accepted: 11/05/2024] [Indexed: 12/09/2024]
Abstract
Here, we present a bioengineering approach to emulate the human bone marrow in vitro. Our developmentally inspired method uses self-organization of human hematopoietic stem and progenitor cells and vascular endothelial cells cultured in a three-dimensional microphysiological system to create vascularized, perfusable tissue constructs that resemble the hematopoietic vascular niche of the human marrow. The microengineered niche is capable of multilineage hematopoiesis and can generate functionally mature human myeloid cells that can intravasate into perfused blood vessels, providing a means to model the mobilization of innate immune cells from the marrow. We demonstrate the application of this system by presenting a specialized model of ionizing radiation-induced bone marrow injury and a multiorgan model of acute innate immune responses to bacterial lung infection. Furthermore, we introduce an advanced platform that enables large-scale integration and automated experimentation of the engineered hematopoietic tissues for preclinical screening of myelotoxicity due to anti-cancer drugs.
Collapse
Affiliation(s)
- Andrei Georgescu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Vivodyne Inc., Philadelphia, PA 19104, USA
| | - Joseph Hai Oved
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | - Samira Mehta
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brian M Dulmovits
- Blood and Marrow Transplant Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Timothy S Olson
- Blood and Marrow Transplant Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pouria Fattahi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anni Wang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Asli Muvaffak
- Complex In Vitro Models, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Michele M Kim
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sezin Aday Aydin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeongyun Seo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric S Diffenderfer
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anthony Lynch
- Complex In Vitro Models, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - G Scott Worthen
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, PA 19104, USA.
| | - Dan Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Vivodyne Inc., Philadelphia, PA 19104, USA; Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
19
|
Elguindy MM, Young JS, Ho WS, Lu RO. Co-evolution of glioma and immune microenvironment. J Immunother Cancer 2024; 12:e009175. [PMID: 39631850 PMCID: PMC11624716 DOI: 10.1136/jitc-2024-009175] [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: 04/03/2024] [Accepted: 11/03/2024] [Indexed: 12/07/2024] Open
Abstract
Glioma evolution is governed by a multitude of dynamic interactions between tumor cells and heterogenous neighboring, non-cancerous cells. This complex ecosystem, termed the tumor microenvironment (TME), includes diverse immune cell types that have gained increasing attention for their critical and paradoxical roles in tumor control and tumorigenesis. Recent work has revealed that the cellular composition and functional state of immune cells in the TME can evolve extensively depending on the tumor stage and intrinsic features of surrounding glioma cells. Concurrently, adaptations to the glioma cellular phenotype, including activation of various cellular states, occur in the context of these immune cell alterations. In this review, we summarize important features of the immune TME that play key roles during each stage of glioma progression, from initiation to immune escape, invasion and recurrence. Understanding the complex interplay between tumor and immune cells is critical for the development of effective immunotherapies for glioma treatment.
Collapse
Affiliation(s)
- Mahmoud M Elguindy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Jacob S Young
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Winson S Ho
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Rongze O Lu
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
20
|
Barcellini A, Molinelli S, Vanoli A, Vitolo V, Fossati P, Vai A, Pagani A, Inzani F, Pecorilla M, Butturini G, Klersy C, Preda L, Facoetti A, Valvo F, Orlandi E. Preoperative chemo-CIRT in Re/BRe pancreatic cancer: Insights from a multicenter prospective phase II clinical study (NCT03822936). TUMORI JOURNAL 2024; 110:470-474. [PMID: 39462835 DOI: 10.1177/03008916241291341] [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] [Indexed: 10/29/2024]
Abstract
PURPOSE There is debate about the optimal management of borderline resectable (bRe) and resectable (Re) pancreatic ductal adenocarcinoma (PDAC). Both preclinical and clinical evidence showed that carbon ion radiotherapy (CIRT) produces superior control on radioresistant histologies compared to conventional photon beam radiotherapy (RT). However, so far there is a lack of data concerning the integration of CIRT in a multimodal approach with chemotherapy and surgery for bRe/Re. METHODS We recently presented the first analysis of a multicenter prospective phase II clinical study aimed at assessing the feasibility and effectiveness of a neoadjuvant chemotherapy + short course of CIRT followed by surgery and adjuvant chemotherapy in the management of bRe/Re PDAC. The study was terminated early due to low patient enrollment.Herein, we reported a post-hoc analysis focusing on toxicity, dosimetry and translational assessment. RESULTS In our experience, CIRT can be integrated into a multimodal treatment strategy for bRe/Re PDAC, alongside chemotherapy and surgery. A case of fatal liver failure occurring three months post-surgery has been documented, likely related to the combination approach. Although the treatment plans were satisfactory according to the Local Effect Model (LEM) model, recalculations using the modified Microdosimetric Kinetic Model (mMKM) revealed suboptimal target coverage. Additionally, we observed an increased expression of PD-L1 following CIRT. CONCLUSIONS This multimodal approach was well tolerated; however, clinicians should carefully monitor for vascular disorders during follow-up and further investigate surgical techniques after CIRT. The increased PD-L1 expression supports the immunogenic effects of particle therapy and lays the groundwork for future studies. To enhance the therapeutic ratio of CIRT treatments, integrating LET-d based objectives into the plan optimization process should be considered. TRIAL REGISTRATION NUMBER ClinicalTrials.gov Identifier: NCT03822936.
Collapse
Affiliation(s)
- Amelia Barcellini
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Silvia Molinelli
- Medical Physics Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Alessandro Vanoli
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia, Italy
- Unit of Anatomic Pathology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Viviana Vitolo
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Piero Fossati
- Department of Radiation Oncology, MedAustron Ion Therapy Center, Wiener Neustadt, Austria
- Department for Basic and Translational Oncology and Haematology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Alessandro Vai
- Medical Physics Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Anna Pagani
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Frediano Inzani
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia, Italy
- Unit of Anatomic Pathology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mattia Pecorilla
- Radiology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Giovanni Butturini
- Division of Hepato-Bilio-Pancreatic Surgery, P. Pederzoli Hospital, Peschiera del Garda, Italy
| | - Catherine Klersy
- Biostatistics & Clinical Trial Center, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Lorenzo Preda
- Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
- Radiology Institute, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Angelica Facoetti
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Francesca Valvo
- Scientific Directorate, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Ester Orlandi
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
21
|
Wang M, Li S, Li R, Ning F, Tian L. Efficacy and Mechanism of Combining Radiotherapy and Immunotherapy in Stage IV Non-Small Cell Lung Cancer. Curr Treat Options Oncol 2024; 25:1605-1614. [PMID: 39625619 PMCID: PMC11638397 DOI: 10.1007/s11864-024-01260-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] [Accepted: 08/26/2024] [Indexed: 12/13/2024]
Abstract
OPINION STATEMENT Lung cancer is the leading cause of cancer-related deaths worldwide, with about 85% of patients being diagnosed as non-small cell lung cancer (NSCLC); and most presenting with stage IV disease initially. With the continuous advancement of treatment strategies of oncology, immunotherapy with/without chemo-immunotherapy has become the first-line treatment for patients with stage IV NSCLC. However, a proportion of patients still develop resistance to the treatment regimen and experience local progression, and primary lung lesion progression is the main progression pattern of stage IV NSCLC. Preclinical and clinical studies have demonstrated the potential of radiotherapy in anti-tumor treatment and suggest that administering local radiotherapy prior to cancer progression can prolong survival. Therefore, we consider whether adding local radiotherapy before the progression of a pulmonary lesion in stage IV NSCLC patients receiving chemo-immunotherapy would be beneficial. The present review aims to explore the efficacy and safety of combining radiotherapy with immunotherapy in the treatment of stage IV NSCLC, delving into the intricacies of their underlying mechanism.
Collapse
Affiliation(s)
- Mingyue Wang
- The Department of Oncology, Binzhou Medical University Hospital, Binzhou City, Shandong Province, China
| | - Shuo Li
- The Department of Oncology, Binzhou Medical University Hospital, Binzhou City, Shandong Province, China
| | - Runyu Li
- The Department of Oncology, Binzhou Medical University Hospital, Binzhou City, Shandong Province, China
| | - Fangling Ning
- The Department of Oncology, Binzhou Medical University Hospital, Binzhou City, Shandong Province, China
| | - Lijun Tian
- The Department of Oncology, Binzhou Medical University Hospital, Binzhou City, Shandong Province, China.
| |
Collapse
|
22
|
Jasim SA, Salahdin OD, Malathi H, Sharma N, Rab SO, Aminov Z, Pramanik A, Mohammed IH, Jawad MA, Gabel BC. Targeting Hepatic Cancer Stem Cells (CSCs) and Related Drug Resistance by Small Interfering RNA (siRNA). Cell Biochem Biophys 2024; 82:3031-3051. [PMID: 39060914 DOI: 10.1007/s12013-024-01423-5] [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] [Accepted: 07/10/2024] [Indexed: 07/28/2024]
Abstract
Tumor recurrence after curative therapy and hepatocellular carcinoma (HCC) cells' resistance to conventional therapies is the reasons for the worse clinical results of HCC patients. A tiny population of cancer cells with a strong potential for self-renewal, differentiation, and tumorigenesis has been identified as cancer stem cells (CSCs). The discovery of CSC surface markers and the separation of CSC subpopulations from HCC cells have been made possible by recent developments in the study of hepatic (liver) CSCs. Hepatic CSC surface markers include epithelial cell adhesion molecules (EpCAM), CD133, CD90, CD13, CD44, OV-6, ALDH, and K19. CSCs have a significant influence on the development of cancer, invasiveness, self-renewal, metastasis, and drug resistance in HCC, and thus provide a therapeutic chance to treat HCC and avoid its recurrence. Therefore, it is essential to develop treatment approaches that specifically and effectively target hepatic stem cells. Given this, one potential treatment approach is to use particular small interfering RNA (siRNA) to target CSC, disrupting their behavior and microenvironment as well as changing their epigenetic state. The characteristics of CSCs in HCC are outlined in this study, along with new treatment approaches based on siRNA that may be used to target hepatic CSCs and overcome HCC resistance to traditional therapies.
Collapse
Affiliation(s)
| | | | - H Malathi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University, Bangalore, Karnataka, India
| | - Neha Sharma
- Chandigarh Pharmacy College, Chandigarh group of Colleges, Jhanjeri, 140307, Mohali, Punjab, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Zafar Aminov
- Department of Public Health and Healthcare management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Israa Hussein Mohammed
- College of nursing, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Mohammed Abed Jawad
- Department of Medical Laboratories Technology, Al-Nisour University College, Baghdad, Iraq
| | - Benien C Gabel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq
- Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
23
|
Hamza FN, Mohammad KS. Immunotherapy in the Battle Against Bone Metastases: Mechanisms and Emerging Treatments. Pharmaceuticals (Basel) 2024; 17:1591. [PMID: 39770433 PMCID: PMC11679356 DOI: 10.3390/ph17121591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/20/2024] [Accepted: 11/23/2024] [Indexed: 01/11/2025] Open
Abstract
Bone metastases are a prevalent complication in advanced cancers, particularly in breast, prostate, and lung cancers, and are associated with severe skeletal-related events (SREs), including fractures, spinal cord compression, and debilitating pain. Conventional bone-targeted treatments like bisphosphonates and RANKL inhibitors (denosumab) reduce osteoclast-mediated bone resorption but do not directly impact tumor progression within the bone. This review focuses on examining the growing potential of immunotherapy in targeting the unique challenges posed by bone metastases. Even though immune checkpoint inhibitors (ICIs) have significantly changed cancer treatment, their impact on bone metastases appears limited because of the bone microenvironment's immunosuppressive traits, which include high levels of transforming growth factor-beta (TGFβ) and the immune-suppressing cells, such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). This review underscores the investigation of combined therapeutic approaches that might ease these difficulties, such as the synergy of immune checkpoint inhibitors with agents aimed at bones (denosumab, bisphosphonates), chemotherapy, and radiotherapy, as well as the combination of immune checkpoint inhibitors with different immunotherapeutic methods, including CAR T-cell therapy. This review provides a comprehensive analysis of preclinical studies and clinical trials that show the synergistic potential of these combination approaches, which aim to both enhance immune responses and mitigate bone destruction. By offering an in-depth exploration of how these strategies can be tailored to the bone microenvironment, this review underscores the need for personalized treatment approaches. The findings emphasize the urgent need for further research into overcoming immune evasion in bone metastases, with the goal of improving patient survival and quality of life.
Collapse
Affiliation(s)
- Fatheia N. Hamza
- Department of Biochemistry, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Khalid Said Mohammad
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
24
|
Zheng J, Zheng Z, Zhang T, Chen X, Pang Q, Wang P, Yan C, Zhang W. Optimization of radiation target volume for locally advanced esophageal cancer in the immunotherapy era. Expert Opin Biol Ther 2024; 24:1221-1232. [PMID: 39460561 DOI: 10.1080/14712598.2024.2423009] [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: 06/29/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 10/28/2024]
Abstract
INTRODUCTION Locally advanced esophageal cancer (EC) has poor prognosis. Preliminary clinical studies have demonstrated the synergistic efficacy of radiotherapy combined with immunotherapy in EC. Adjusting the radiotherapy target volume to protect immune function favors immunotherapy. However, there is no clear consensus on the exact definition of the EC target volume. AREAS COVERED Preclinical studies have provided a wealth of information on immunotherapy combined with different radiotherapy modalities, and several clinical studies have evaluated the impact of immunotherapy combined with radiotherapy on locally advanced EC. Here, we illustrate the rational target volume delineation for radiotherapy in terms of patient prognosis, pattern of radiotherapy failure, treatment-related toxicities, tumor-draining lymph nodes, and systemic immunity and summarize the clinical trials of radiotherapy combined with immunotherapy in EC. EXPERT OPINION We recommend applying involved-field irradiation (IFI) instead of elective nodal irradiation (ENI) for irradiated fields when immunotherapy is combined with chemoradiotherapy (CRT) for locally advanced EC. We expect that this target design will be evaluated in clinical trials to further explore more precise diagnostic modalities, long-term toxic responses, and quality of survival, and stratification factors for personalized treatment, and to provide more treatment benefits for patients.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhunhao Zheng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tian Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xi Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qingsong Pang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
25
|
Katoh H, Mitsuma T, Okamoto R, Naito K, Tokito T, Kikuchi M, Sangai T. Pembrolizumab with external radiation therapy effectively controlled TMB-high unresectable recurrent parathyroid cancer: a case report with review of literature. Endocr J 2024; 71:1069-1075. [PMID: 38987211 PMCID: PMC11778383 DOI: 10.1507/endocrj.ej24-0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Parathyroid cancer (PC) is extremely resistant to chemotherapy and radiotherapy (RT), but hormonally functional by producing excessive parathyroid hormone (PTH), causing remarkable hypercalcemia even in biochemical disease recurrence. Accordingly, management of hypercalcemia by calcimimetics and bisphosphonates has been main treatment for unresectable PC. Here, we report a case of unresectable tumor mutational burden (TMB)-high recurrent PC that has been effectively controlled by pembrolizumab (PEM) with RT. A 48-year-old male patient, with previous history of left single parathyroidectomy for primary hyperparathyroidism, underwent surgeries for recurrent hyperparathyroidism at 47 and 48 years of age, and was pathologically diagnosed with PC. He was referred to our hospital due to persistent hypercalcemia and elevated PTH. The recurrent tumors were identified in the superior mediastinum and radically resected, then the hyperparathyroidism was improved. A FoundationOne® CDx of the specimen called TMB-high. He demonstrated recurrent hyperparathyroidism at 49 years of age, and underwent a gross curative resection. However, hyperparathyroidism achieved only insufficient improvement, indicating biochemical residual cancer cells. PEM treatment was initiated in combination with RT to the left central-lateral neck and superior mediastinum. He successfully achieved evocalcet and zoledronate withdrawal, and the PTH level improvement was continuously observed for 8 months at present, with only grade 2 subclinical hypothyroidism. Interestingly, leukocyte fraction ratios were reversed corresponding to disease improvement. A combination of PEM and RT is a promising treatment of unresectable TMB-high PC. Recent evidence on the immunomodulatory effect of RT provides the rationale for the combination of RT and PEM.
Collapse
Affiliation(s)
- Hiroshi Katoh
- Department of Breast and Thyroid Surgery, Kitasato University Hospital, Kanagawa 252-0374, Japan
| | - Tomoya Mitsuma
- Department of Breast and Thyroid Surgery, Kitasato University Hospital, Kanagawa 252-0374, Japan
| | - Riku Okamoto
- Department of Breast and Thyroid Surgery, Kitasato University Hospital, Kanagawa 252-0374, Japan
| | - Kanako Naito
- Department of Breast and Thyroid Surgery, Kitasato University Hospital, Kanagawa 252-0374, Japan
| | - Takaaki Tokito
- Department of Breast and Thyroid Surgery, Kitasato University Hospital, Kanagawa 252-0374, Japan
| | - Mariko Kikuchi
- Department of Breast and Thyroid Surgery, Kitasato University Hospital, Kanagawa 252-0374, Japan
| | - Takafumi Sangai
- Department of Breast and Thyroid Surgery, Kitasato University Hospital, Kanagawa 252-0374, Japan
| |
Collapse
|
26
|
Li S, Chen K, Sun Z, Chen M, Pi W, Zhou S, Yang H. Radiation drives tertiary lymphoid structures to reshape TME for synergized antitumour immunity. Expert Rev Mol Med 2024; 26:e30. [PMID: 39438247 PMCID: PMC11505612 DOI: 10.1017/erm.2024.27] [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: 09/07/2023] [Revised: 04/24/2024] [Accepted: 07/15/2024] [Indexed: 10/25/2024]
Abstract
Radiotherapy (RT) plays a key role in the tumour microenvironment (TME), impacting the immune response via cellular and humoral immunity. RT can induce local immunity to modify the TME. It can stimulate dendritic cell maturation and T-cell infiltration. Moreover, B cells, macrophages and other immune cells may also be affected. Tertiary lymphoid structure (TLS) is a unique structure within the TME and a class of aggregates containing T cells, B cells and other immune cells. The maturation of TLS is determined by the presence of mature dendritic cells, the density of TLS is determined by the number of immune cells. TLS maturation and density both affect the antitumour immune response in the TME. This review summarized the recent research on the impact and the role of RT on TLS, including the changes of TLS components and formation conditions and the mechanism of how RT affects TLS and transforms the TME. RT may promote TLS maturation and density to modify the TME regarding enhanced antitumour immunity.
Collapse
Affiliation(s)
- Shuling Li
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Kuifei Chen
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Zhenwei Sun
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Meng Chen
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Wenhu Pi
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Suna Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Haihua Yang
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| |
Collapse
|
27
|
Jansen CS, Pagadala MS, Cardenas MA, Prabhu RS, Goyal S, Zhou C, Chappa P, Vo BT, Ye C, Hopkins B, Zhong J, Klie A, Daniels T, Admassu M, Green I, Pfister NT, Neill SG, Switchenko JM, Prokhnevska N, Hoang KB, Torres MA, Logan S, Olson JJ, Nduom EK, Del Balzo L, Patel K, Burri SH, Asher AL, Wilkinson S, Lake R, Kesarwala AH, Higgins KA, Patel P, Dhere V, Sowalsky AG, Carter H, Khan MK, Kissick H, Buchwald ZS. Pre-operative stereotactic radiosurgery and peri-operative dexamethasone for resectable brain metastases: a two-arm pilot study evaluating clinical outcomes and immunological correlates. Nat Commun 2024; 15:8854. [PMID: 39402027 PMCID: PMC11473782 DOI: 10.1038/s41467-024-53034-6] [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: 07/01/2024] [Accepted: 09/29/2024] [Indexed: 10/17/2024] Open
Abstract
Enhancing the efficacy of immunotherapy in brain metastases (BrM) requires an improved understanding of the immune composition of BrM and how this is affected by radiation and dexamethasone. Our two-arm pilot study (NCT04895592) allocated 26 patients with BrM to either low (Arm A) or high (Arm B) dose peri-operative dexamethasone followed by pre-operative stereotactic radiosurgery (pSRS) and resection (n= 13 per arm). The primary endpoint, a safety analysis at 4 months, was met. The secondary clinical endpoints of overall survival, distant brain failure, leptomeningeal disease and local recurrence at 12-months were 66%, 37.3%, 6%, and 0% respectively and were not significantly different between arms (p= 0.7739, p= 0.3884, p= 0.3469). Immunological data from two large retrospective BrM datasets and confirmed by correlates from both arms of this pSRS prospective trial revealed that BrM CD8 T cells were composed of predominantly PD1+ TCF1+ stem-like and PD1+ TCF1-TIM3+ effector-like cells. Clustering of TCF1+ CD8 T cells with antigen presenting cells in immune niches was prognostic for local control, even without pSRS. Following pSRS, CD8 T cell and immune niche density were transiently reduced compared to untreated BrM, followed by a rebound 6+ days post pSRS with an increased frequency of TCF1- effector-like cells. In sum, pSRS is safe and therapeutically beneficial, and these data provide a framework for how pSRS may be leveraged to maximize intracranial CD8 T cell responses.
Collapse
Affiliation(s)
| | - Meghana S Pagadala
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | | | - Roshan S Prabhu
- Southeast Radiation Oncology Group, Levine Cancer Institute, Atrium Health, Charlotte, USA
| | - Subir Goyal
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, USA
| | - Chengjing Zhou
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - Prasanthi Chappa
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - BaoHan T Vo
- Department of Urology, Emory University, Atlanta, USA
| | - Chengyu Ye
- Department of Urology, Emory University, Atlanta, USA
| | - Benjamin Hopkins
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - Jim Zhong
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - Adam Klie
- Biomedical Sciences Program, University of California San Diego, La Jolla, USA
| | - Taylor Daniels
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - Maedot Admassu
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - India Green
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - Neil T Pfister
- Department of Radiation Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | | | - Jeffrey M Switchenko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, USA
| | | | - Kimberly B Hoang
- Winship Cancer Institute, Emory University, Atlanta, USA
- Department of Neurosurgery, Emory University, Atlanta, USA
| | - Mylin A Torres
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - Suzanna Logan
- Department of Pathology, Nationwide Children's Hospital, Columbus, USA
| | - Jeffrey J Olson
- Winship Cancer Institute, Emory University, Atlanta, USA
- Department of Neurosurgery, Emory University, Atlanta, USA
| | - Edjah K Nduom
- Winship Cancer Institute, Emory University, Atlanta, USA
- Department of Neurosurgery, Emory University, Atlanta, USA
| | | | | | - Stuart H Burri
- Southeast Radiation Oncology Group, Levine Cancer Institute, Atrium Health, Charlotte, USA
| | | | - Scott Wilkinson
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, USA
| | - Ross Lake
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, USA
| | - Aparna H Kesarwala
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - Kristin A Higgins
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - Pretesh Patel
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - Vishal Dhere
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - Adam G Sowalsky
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, USA
| | - Hannah Carter
- Department of Medicine, Division of Medical Genetics, University of California San Diego, La Jolla, USA
| | - Mohammad K Khan
- Department of Radiation Oncology, Emory University, Atlanta, USA
- Winship Cancer Institute, Emory University, Atlanta, USA
| | - Haydn Kissick
- Department of Urology, Emory University, Atlanta, USA.
- Department of Microbiology and Immunology, Emory University, Atlanta, USA.
| | - Zachary S Buchwald
- Department of Radiation Oncology, Emory University, Atlanta, USA.
- Winship Cancer Institute, Emory University, Atlanta, USA.
| |
Collapse
|
28
|
Yang G, Koom WS, Lee BM, Isozaki T, Shinoto M, Yamada S, Seong J. Reduced Risk of Severe Radiation-Induced Lymphopenia in Carbon Ion Radiation Therapy for Locally Advanced Pancreatic Cancer: A Comparative Analysis of Carbon Versus Photon Therapy. Int J Radiat Oncol Biol Phys 2024; 120:544-554. [PMID: 38713122 DOI: 10.1016/j.ijrobp.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/15/2024] [Accepted: 04/03/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE Radiation-induced lymphopenia (RIL) is associated with poor prognosis in patients with locally advanced pancreatic cancers. However, there are no reports comparing the effects of carbon ion radiation therapy (CIRT) and photon beam radiation therapy (RT) on the development of RIL. Differences in RIL after CIRT or photon beam RT and predictive factors for RIL in patients with locally advanced pancreatic cancer were investigated. MATERIALS AND METHODS This retrospective study cohort included 834 patients who received concurrent chemoradiotherapy (CCRT) in 2 separate institutions: 337 and 497 in the CIRT and photon beam RT groups, respectively. Severe RIL was defined as an absolute lymphocyte count (ALC) <0.5 × 109 cells/L. A 1:1 propensity score-matching analysis was performed between the CIRT and photon beam RT groups. Patients were categorized into 3 groups according to the development of recovery from severe RIL: no severe RIL (Group A), recovery from severe RIL (Group B), and no recovery from severe RIL (Group C). Logistic regression analysis was performed to identify the predictive value of severe RIL. The prognostic factors of overall survival (OS) were determined using Cox regression analysis. RESULTS After propensity score matching, the baseline ALC and planning target volume of the CIRT and photon beam RT groups were comparable. During CCRT, the ALC of the entire cohort decreased and was significantly lower in the photon beam RT group than in the CIRT group (P < .001). Multivariate logistic regression analysis showed that CIRT reduced severe RIL more than photon beam RT. After adjusting for other factors, the RT modality and RIL were significantly associated with OS. Photon beam RT showed a significantly worse OS than CIRT, and Group C showed a significantly worse OS than Group A. CONCLUSIONS CIRT seems to reduce the development of severe RIL. The RT modality and development/recovery from severe RIL were associated with OS in patients who received CCRT for locally advanced pancreatic cancer. The reduction of severe RIL through optimized RT may be essential for improving treatment outcomes.
Collapse
Affiliation(s)
- Gowoon Yang
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byung Min Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tetsuro Isozaki
- QST Hospital, National Institutes for Quantum Science and Technology, Inageku, Chiba, Japan
| | - Makoto Shinoto
- QST Hospital, National Institutes for Quantum Science and Technology, Inageku, Chiba, Japan
| | - Shigeru Yamada
- QST Hospital, National Institutes for Quantum Science and Technology, Inageku, Chiba, Japan.
| | - Jinsil Seong
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Bo Y, Wang H. Biomaterial-Based In Situ Cancer Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2210452. [PMID: 36649567 PMCID: PMC10408245 DOI: 10.1002/adma.202210452] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/06/2023] [Indexed: 06/17/2023]
Abstract
Cancer immunotherapies have reshaped the paradigm for cancer treatment over the past decade. Among them, therapeutic cancer vaccines that aim to modulate antigen-presenting cells and subsequent T cell priming processes are among the first FDA-approved cancer immunotherapies. However, despite showing benign safety profiles and the capability to generate antigen-specific humoral and cellular responses, cancer vaccines have been limited by the modest therapeutic efficacy, especially for immunologically cold solid tumors. One key challenge lies in the identification of tumor-specific antigens, which involves a costly and lengthy process of tumor cell isolation, DNA/RNA extraction, sequencing, mutation analysis, epitope prediction, peptide synthesis, and antigen screening. To address these issues, in situ cancer vaccines have been actively pursued to generate endogenous antigens directly from tumors and utilize the generated tumor antigens to elicit potent cytotoxic T lymphocyte (CTL) response. Biomaterials-based in situ cancer vaccines, in particular, have achieved significant progress by taking advantage of biomaterials that can synergize antigens and adjuvants, troubleshoot delivery issues, home, and manipulate immune cells in situ. This review will provide an overview of biomaterials-based in situ cancer vaccines, either living or artificial materials, under development or in the clinic, and discuss the design criteria for in situ cancer vaccines.
Collapse
Affiliation(s)
- Yang Bo
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Cancer Center at Illinois (CCIL), Urbana, IL, 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
30
|
Holtermann A, Gislon M, Angele M, Subklewe M, von Bergwelt-Baildon M, Lauber K, Kobold S. Prospects of Synergy: Local Interventions and CAR T Cell Therapy in Solid Tumors. BioDrugs 2024; 38:611-637. [PMID: 39080180 PMCID: PMC11358237 DOI: 10.1007/s40259-024-00669-y] [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] [Accepted: 06/17/2024] [Indexed: 08/30/2024]
Abstract
Chimeric antigen receptor T cell therapy has been established in the treatment of various B cell malignancies. However, translating this therapeutic effect to treat solid tumors has been challenging because of their inter-tumoral as well as intratumoral heterogeneity and immunosuppressive microenvironment. Local interventions, such as surgery, radiotherapy, local ablation, and locoregional drug delivery, can enhance chimeric antigen receptor T cell therapy in solid tumors by improving tumor infiltration and reducing systemic toxicities. Additionally, ablation and radiotherapy have proven to (re-)activate systemic immune responses via abscopal effects and reprogram the tumor microenvironment on a physical, cellular, and chemical level. This review highlights the potential synergy of the combined approaches to overcome barriers of chimeric antigen receptor T cell therapy and summarizes recent studies that may pave the way for new treatment regimens.
Collapse
Affiliation(s)
- Anne Holtermann
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Mila Gislon
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
| | - Martin Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München-German Research Center for Environmental Health Neuherberg, Munich, Germany.
| |
Collapse
|
31
|
Kluge A, Baum RP, Bitterlich N, Kulkarni HR, Schorr-Neufing U, van Echteld CJA. Immune Response to Molecular Radiotherapy with 177Lu-DOTATOC: Predictive Value of Blood Cell Counts for Therapy Outcome. Cancer Biother Radiopharm 2024; 39:541-550. [PMID: 38905126 DOI: 10.1089/cbr.2024.0031] [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] [Indexed: 06/23/2024] Open
Abstract
Purpose: In a prior, retrospective study, 76% of patients with advanced neuroendocrine tumors undergoing 177Lu-DOTATOC molecular radiotherapy (MRT) showed their best response within 8 months from the first MRT cycle. In 24% of patients, latency was much greater up to >22 months after the first cycle, and long after near-complete decay of 177Lu from the last cycle. An immune response induced by MRT seems a likely explanation. As a crude measure of immunocompetence, the authors investigated whether blood cell counts (BCCs) may have predictive value for MRT outcome with 177Lu-DOTATOC. Methods: 56 Patients with neuroendocrine tumors (NET) were administered 177Lu-DOTATOC (mean 2.1 cycles; range 1-4) with median radioactivity of 7.0 GBq/cycle at 3-month intervals. Patients' BCCs were evaluated for four responder categories: CR, PR, SD, and PD (RECIST 1.1). Furthermore, baseline BCCs were correlated with progression-free survival (PFS). Finally, BCCs of patients with (PMT+) and without prior medical therapy (PMT-) were compared. Results: Significant differences between responder categories were found for baseline hemoglobin (Hb), erythrocytes, neutrophils, lymphocytes, neutrophil/lymphocyte ratio (NLR), platelet/lymphocyte ratio (PLR), and LEHN-score, integrating lymphocyte, erythrocyte, and neutrophil counts, and Hb level, but not for leukocytes and platelets. LEHN-score yielded an almost complete separation between CR and PD groups. In analogy, PFS times showed significant correlations with baseline Hb, erythrocytes, neutrophils, lymphocytes, NLR, PLR, and LEHN-score, the LEHN-score showing the strongest correlation, but not with leukocytes and platelets. For PMT- patients, median PFS was 34.5 months, compared with 20.8 months in PMT+ patients, with corresponding baseline lymphocyte (32.1 ± 9.6% vs. 24.5 ± 11.6%, p = 0.028) and neutrophil (54.9 ± 11.6% vs. 63.5 ± 13.7%, p = 0.039) counts. Conclusion: These findings emphasize the significance of an immune response to MRT for obtaining optimal therapy efficacy and support concepts to enhance the immune response of less immunocompetent patients before MRT. It seems advisable to avoid prior or concomitant immunosuppressant medical therapy.
Collapse
Affiliation(s)
- Andreas Kluge
- ABX-CRO Advanced Pharmaceutical Services, Dresden, Germany
| | - Richard P Baum
- Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka, Bad Berka, Germany
- CURANOSTICUM Wiesbaden-Frankfurt-Advanced Theranostics Center for Radiomolecular Precision Oncology, HELIOS DKD Klinik, Wiesbaden, Germany
| | | | - Harshad R Kulkarni
- Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka, Bad Berka, Germany
- BAMF Health, Grand Rapids, Michigan, USA
| | | | - Cees J A van Echteld
- ABX-CRO Advanced Pharmaceutical Services, Dresden, Germany
- Helacor Consultancy, Hillegom, The Netherlands
| |
Collapse
|
32
|
Ma Z, Liu Y, Bao Y, Liu Q, Yang X, Men Y, Wang J, Deng L, Zhai Y, Hu C, Bi N, Wang L, Hui Z. Higher immune cell radiation dose is correlated with poor tumor control and survival in patients with non-small cell lung cancer receiving postoperative radiotherapy. BMC Cancer 2024; 24:968. [PMID: 39112971 PMCID: PMC11308139 DOI: 10.1186/s12885-024-12699-4] [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: 11/13/2023] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
INTRODUCTION The estimated dose of radiation to immune cells (EDRIC) has been shown to correlate with the overall survival (OS) of patients who receive definitive thoracic radiotherapy. However, the planning target volume (PTV) may be a confounding factor. We assessed the prognostic value of EDRIC for non-small cell lung cancer (NSCLC) in patients who underwent postoperative radiotherapy (PORT) with homogeneous PTV. METHODS Patients with NSCLC who underwent PORT between 2004 and 2019 were included. EDRIC was computed as a function of the number of radiation fractions and mean doses to the lungs, heart, and remaining body. The correlations between EDRIC and OS, disease-free survival (DFS), locoregional-free survival (LRFS), and distant metastasis-free survival (DMFS) were analyzed using univariate and multivariate Cox models. Kaplan-Meier analysis was performed to assess the survival difference between low- and high-EDRIC groups. RESULTS In total, 345 patients were analyzed. The mean EDRIC was 6.26 Gy. Multivariate analysis showed that higher EDRIC was associated with worse outcomes in terms of OS (hazard ratio [HR] 1.207, P = .007), DFS (HR 1.129, P = .015), LRFS (HR 1.211, P = .002), and DMFS (HR 1.131, P = .057). In the low- and high-EDRIC groups, the 3-year OS was 81.2% and 74.0%, DFS 39.8% and 35.0%, LRFS 70.4% and 60.5%, and DMFS 73.9% and 63.1%, respectively. CONCLUSIONS EDRIC is an independent prognostic factor for survival in patients with NSCLC undergoing PORT. Higher doses of radiation to the immune system are associated with tumor progression and poor survival. Organs at risk for the immune system should be considered during radiotherapy planning.
Collapse
Affiliation(s)
- Zeliang Ma
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunsong Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongxing Bao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Men
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianyang Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Deng
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yirui Zhai
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Hu
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Luhua Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhouguang Hui
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
33
|
Xu N, Wang J, Liu L, Gong C. Injectable hydrogel-based drug delivery systems for enhancing the efficacy of radiation therapy: A review of recent advances. CHINESE CHEM LETT 2024; 35:109225. [DOI: 10.1016/j.cclet.2023.109225] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
34
|
Al-Eryani K, Epstein JB, Monreal AV, Villa A. Orofacial complications from immune checkpoint inhibitors: A retrospective analysis from two academic medical centers. Head Neck 2024; 46:1865-1872. [PMID: 38258988 DOI: 10.1002/hed.27646] [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: 11/04/2023] [Revised: 12/31/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are FDA-approved for various cancers, yet their orofacial immune-related adverse events (irAEs) remain poorly understood. Our two-center retrospective study aims to better understand the prevalence and nature of these orofacial irAEs. METHODS We retrospectively collected demographics, ICI details, and onset of orofacial irAEs in ICI-treated patients at University of California San Francisco and City of Hope (2013-2021). Orofacial irAEs were identified by ICD-10 codes and data categorized as dry mouth/xerostomia, oral mucosal lesions, and orofacial neuropathies. Patients with pre-existing orofacial conditions resembling the reported irAEs were excluded. RESULTS Among 3768 ICI-treated patients, 408 (10.8%) developed 467 orofacial irAEs: oral mucosal diseases (41.4%), dry mouth/xerostomia (41.0%), and orofacial neuropathies (17.6%). Notably, head and neck cancers had the highest incidence of orofacial irAEs. CONCLUSIONS Orofacial irAEs are relatively common in patients receiving ICIs, necessitating careful monitoring and management of these complications during and after the treatment.
Collapse
Affiliation(s)
- Kamal Al-Eryani
- Department of Orofacial Sciences, University of California, San Francisco, California, USA
| | - Joel B Epstein
- Dental Oncology Services, City of Hope Comprehensive Cancer Center, Duarte, California, USA
- Cedars Sinai Health System, Los Angeles, California, USA
| | - Anette Vistoso Monreal
- Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Alessandro Villa
- Department of Orofacial Sciences, University of California, San Francisco, California, USA
- Oral Medicine, Oral Oncology and Dentistry, Miami Cancer Institute, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| |
Collapse
|
35
|
Zhu J, Li M, Zhang Y, Lv Z, Zhao Z, Guo Y, Chen Y, Ren X, Cheng X, Shi H. S-Sulfenylation Driven Antigen Capture Boosted by Radiation for Enhanced Cancer Immunotherapy. ACS NANO 2024. [PMID: 39066710 DOI: 10.1021/acsnano.4c02206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Radiotherapy (RT)-induced in situ vaccination greatly promotes the development of personalized cancer vaccines owing to the massive release of antigens initiated by tumor-localized RT eliciting the tumor-specific immune response. However, its broad application in cancer treatment is seriously impeded by poor antigen cross-presentation, low response rate, and short duration of efficacy. Herein, the tumor-antigen-capturing nanosystem dAuNPs@CpG consisting of gold nanoparticles, 3,5-cyclohexanedione (CHD), and immunoadjuvant CpG were fabricated to enhance RT-induced vaccination. Taking advantage of the specific covalent binding between CHD and sulfenic acids of antigen proteins, we show that this nanoplatform has an unexpected potential to capture the sulfenylated tumor-derived protein antigens (TDPAs) induced by RT to in situ generate a vaccination effect, achieving significant growth suppression of both primary and distant tumors in combination with PD-1 blockade. We thus believe that our work presents a powerful and effective means to improve the synergistic tumor radioimmunotherapy.
Collapse
Affiliation(s)
- Jinfeng Zhu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Roma 00133, Italy
| | - Miao Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Yuqi Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhengzhong Lv
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhongsheng Zhao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Yirui Guo
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Yan Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Xingxiang Ren
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Xiaju Cheng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Haibin Shi
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| |
Collapse
|
36
|
Takashima ME, Berg TJ, Morris ZS. The Effects of Radiation Dose Heterogeneity on the Tumor Microenvironment and Anti-Tumor Immunity. Semin Radiat Oncol 2024; 34:262-271. [PMID: 38880534 DOI: 10.1016/j.semradonc.2024.04.004] [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: 06/18/2024]
Abstract
Radiotherapy elicits dose- and lineage-dependent effects on immune cell survival, migration, activation, and proliferation in targeted tumor microenvironments. Radiation also stimulates phenotypic changes that modulate the immune susceptibility of tumor cells. This has raised interest in using radiotherapy to promote greater response to immunotherapies. To clarify the potential of such combinations, it is critical to understand how best to administer radiation therapy to achieve activation of desired immunologic mechanisms. In considering the multifaceted process of priming and propagating anti-tumor immune response, radiation dose heterogeneity emerges as a potential means for simultaneously engaging diverse dose-dependent effects in a single tumor environment. Recent work in spatially fractionated external beam radiation therapy demonstrates the expansive immune responses achievable when a range of high to low dose radiation is delivered in a tumor. Brachytherapy and radiopharmaceutical therapies deliver inherently heterogeneous distributions of radiation that may contribute to immunogenicity. This review evaluates the interplay of radiation dose and anti-tumor immune response and explores emerging methodological approaches for investigating the effects of heterogeneous dose distribution on immune responses.
Collapse
Affiliation(s)
- Maya E Takashima
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Tracy J Berg
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| |
Collapse
|
37
|
Dionisi F, Landoni V, Widesott L, Nardangeli A, Fracchiolla F, Siniscalchi B, Soriani A, Turkaj A, Righetto R, Amelio D, Farace P, Goanta L, Trianni A, Lorentini S, Cianchetti M, Sanguineti G. Dosimetric and NTCP advantages of robust proton therapy over robust VMAT for Stage III NSCLC in the immunotherapy era. Phys Med 2024; 123:103410. [PMID: 38878630 DOI: 10.1016/j.ejmp.2024.103410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/29/2024] [Accepted: 06/10/2024] [Indexed: 07/13/2024] Open
Abstract
AIMS To assess the robustness and to define the dosimetric and NTCP advantages of pencil-beam-scanning proton therapy (PBSPT) compared with VMAT for unresectable Stage III non-small lung cancer (NSCLC) in the immunotherapy era. MATERIAL AND METHODS 10 patients were re-planned with VMAT and PBSPT using: 1) ITV-based robust optimization with 0.5 cm setup uncertainties and (for PBSPT) 3.5 % range uncertainties on free-breathing CT 2) CTV-based RO including all 4DCTs anatomies. Target coverage (TC), organs at risk dose and TC robustness (TCR), set at V95%, were compared. The NTCP risk for radiation pneumonitis (RP), 24-month mortality (24MM), G2 + acute esophageal toxicity (ET), the dose to the immune system (EDIC) and the left anterior descending (LAD) coronary artery V15 < 10 % were registered. Wilcoxon test was used. RESULTS Both PBSPT methods improved TC and TCR (p < 0.01). The mean lung dose and lung V20 were lower with PBSPT (p < 0.01). Median mean heart dose reduction with PBSPT was 8 Gy (p < 0.001). PT lowered median LAD V15 (p = 0.004). ΔNTCP > 5 % with PBSPT was observed for two patients for RP and for five patients for 24 MM. ΔNTCP for ≥ G2 ET was not in favor of PBSPT for all patients. PBSPT halved median EDIC (4.9/5.1 Gy for ITV/CTV-based VMAT vs 2.3 Gy for both ITV/CTV-based PBSPT, p < 0.01). CONCLUSIONS PBSPT is a robust approach with significant dosimetric and NTCP advantages over VMAT; the EDIC reduction could allow for a better integration with immunotherapy. A clinical benefit for a subset of NSCLC patients is expected.
Collapse
Affiliation(s)
- F Dionisi
- Department of Research and Advanced Technology, Radiotherapy Unit, IRCCS Regina Elena National Cancer Institute-Rome, Italy.
| | - V Landoni
- Laboratory of Medical Physics and Expert Systems, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - L Widesott
- Medical Physics Department, APSS, Trento, Italy
| | - A Nardangeli
- Department of Research and Advanced Technology, Radiotherapy Unit, IRCCS Regina Elena National Cancer Institute-Rome, Italy
| | | | | | - A Soriani
- Laboratory of Medical Physics and Expert Systems, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - A Turkaj
- Proton Therapy Unit, APSS, Trento, Italy
| | - R Righetto
- Medical Physics Department, APSS, Trento, Italy
| | - D Amelio
- Proton Therapy Unit, APSS, Trento, Italy
| | - P Farace
- Medical Physics Department, APSS, Trento, Italy
| | - L Goanta
- Department of Research and Advanced Technology, Radiotherapy Unit, IRCCS Regina Elena National Cancer Institute-Rome, Italy
| | - A Trianni
- Medical Physics Department, APSS, Trento, Italy
| | - S Lorentini
- Medical Physics Department, APSS, Trento, Italy
| | | | - G Sanguineti
- Department of Research and Advanced Technology, Radiotherapy Unit, IRCCS Regina Elena National Cancer Institute-Rome, Italy
| |
Collapse
|
38
|
Li J, Hu YT, Liu CC, Wang LH, Ju HX, Huang XF, Chi P, Du JL, Wang JP, Xiao Y, Lin GL, Zhang W, Zhao H, Liu M, Song YM, Xu D, Wang JW, Sun LF, Xie HT, Cao HF, Xiao Q, Wang J, Wu QB, Li DC, Dai S, Jiang WZ, Shen L, Yuan Y, Wang ZQ, Ding KF. Primary Surgery Followed by Selective Chemoradiotherapy Versus Preoperative Chemoradiotherapy Followed by Surgery for Locally Advanced Rectal Cancer: A Randomized Clinical Trial. Int J Radiat Oncol Biol Phys 2024; 119:884-895. [PMID: 38185388 DOI: 10.1016/j.ijrobp.2023.12.027] [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: 06/13/2023] [Revised: 11/27/2023] [Accepted: 12/19/2023] [Indexed: 01/09/2024]
Abstract
PURPOSE The aim of this work was to determine whether locally advanced rectal cancer (LARC) with negative mesorectal fascia (MRF) predicted by magnetic resonance imaging (MRI) can be excluded from preoperative radiation therapy treatment. METHODS AND MATERIALS This multicenter, open-label, non-inferiority, randomized clinical trial enrolled patients with LARC within 6 to 12 cm from the anal verge and with negative MRI-predicted MRF. Participants were randomized to the intervention group (primary surgery, in which the patients with positive pathologic [CRM] circumferential margins were subjected to chemoradiotherapy [CRT] and those with negative CRM underwent adjuvant chemotherapy according to pathologic staging) or the control group (preoperative CRT, in which all patients underwent subsequent surgery and adjuvant chemotherapy). The primary endpoint was 3-year disease-free survival (DFS). RESULTS A total of 275 patients were randomly assigned to the intervention (n = 140) and control (n = 135) groups, in which 33.57% and 28.15% patients were at clinical T4 stage and 85.92% and 80.45% patients were at "bad" or "ugly" risk in the intervention and control groups, respectively. There were 2 patients (1.52%) and 1 patient (0.77%) with positive CRM in the intervention and control groups, respectively (P > .05). The non-adherence rates for the intervention and control groups were 3.6% and 23.7%, respectively. After a median follow-up of 34.6 months (IQR, 18.2-45.7), 43 patients had positive events (28 patients and 15 patients in the intervention and control groups, respectively). There were 6 patients (4.4%) with local recurrence in the intervention group and none in the control group, which led to the termination of the trial. The 3-year DFS rate was 81.82% in the intervention group (95% CI, 78.18%-85.46%) and 85.37% in the control group (95% CI, 81.75%-88.99%), with a difference of -3.55% (95% CI, -3.71% to -3.39%; hazard ratio, 1.76; 95% CI, 0.94-3.30). In the per-protocol data set, the difference between 3-year DFS rates was -5.44% (95% CI, -5.63% to -5.25%; hazard ratio, 2.02; 95% CI, 1.01-4.06). CONCLUSIONS Based on the outcomes of this trial, in patients with LARC and MRI-negative MRF, primary surgery could negatively influence their DFS rates. Therefore, primary surgery was an inferior strategy compared with preoperative CRT followed by surgery and cannot be recommended for patients with LARC.
Collapse
Affiliation(s)
- Jun Li
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China
| | - Ye-Ting Hu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China
| | - Cheng-Cheng Liu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China
| | - Liu-Hong Wang
- Department of Radiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Xing Ju
- Department of Colorectal Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Xue-Feng Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Pan Chi
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jin-Lin Du
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jian-Ping Wang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Yi Xiao
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Guo-Le Lin
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Zhang
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hong Zhao
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming Liu
- Department of General Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong-Mao Song
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China
| | - Dong Xu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China
| | - Jian-Wei Wang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China
| | - Li-Feng Sun
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China
| | - Hai-Ting Xie
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China
| | - Hong-Feng Cao
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China
| | - Qian Xiao
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China
| | - Jian Wang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China
| | - Qing-Bin Wu
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - De-Chuan Li
- Department of Colorectal Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Sheng Dai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei-Zhong Jiang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Li Shen
- Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China; Department of Radiation Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Yuan
- Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China; Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zi-Qiang Wang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
| | - Ke-Feng Ding
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Zhejiang, China.
| |
Collapse
|
39
|
Seyedin SN, Harada G, Garemanian E, Rafizadeh D, Kaakour D, Dwabe S, Daneshvar M, Mar N. Use of Radiation Therapy for Ataxia-Telangiectasia Mutated (ATM)-Mutation Metastatic Renal Cell Carcinoma: A Case Report. Cureus 2024; 16:e64781. [PMID: 39156348 PMCID: PMC11329860 DOI: 10.7759/cureus.64781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 08/20/2024] Open
Abstract
Papillary renal cell carcinoma (pRCC) is a rare kidney cancer with limited treatment options and poor outcomes when metastatic. We present a case of a 42-year-old male with metastatic pRCC harboring a somatic ataxia-telangiectasia mutated (ATM) mutation who was treated at our institution. After progression of disease (POD) on ipilimumab/nivolumab, followed by POD on cabozantinib, the patient was treated with radiation therapy to metastatic cervical lymphadenopathy to 60 Gy in 15 fractions as well as retroperitoneal lymphadenopathy to 36 Gy in 9 fractions, which was curtailed due to intolerance. This was followed by sequential systemic therapy with a poly (ADP-ribose) polymerase (PARP) inhibitor and pembrolizumab, which was also discontinued due to adverse effects. Despite not receiving any treatment for 10 months, his disease remains stable. We believe that the prolonged progression-free survival of this patient with ATM-mutation metastatic pRCC is likely due to the enhanced sensitivity of the tumor to radiation therapy due to ATM loss.
Collapse
Affiliation(s)
- Steven N Seyedin
- Radiation Oncology, University of California San Francisco Medical Center, San Francisco, USA
| | - Garrett Harada
- Radiation Oncology, University of California Irvine Medical Center, Orange, USA
| | | | - Desiree Rafizadeh
- Radiation Oncology, University of California Irvine School of Medicine, Irvine, USA
| | - Dalia Kaakour
- Hematology and Oncology, University of California Irvine Medical Center, Orange, USA
| | - Sami Dwabe
- Hematology and Oncology, University of California Irvine Medical Center, Orange, USA
| | - Michael Daneshvar
- Urology, University of California Irvine Medical Center, Orange, USA
| | - Nataliya Mar
- Hematology and Oncology, University of California Irvine Medical Center, Orange, USA
| |
Collapse
|
40
|
Gatfield ER, Tadross J, Ince W. Immune checkpoint inhibitor use in head and neck squamous cell carcinoma: the current landscape and future perspectives. Future Oncol 2024; 20:1695-1711. [PMID: 38889284 PMCID: PMC11485897 DOI: 10.1080/14796694.2024.2362612] [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: 02/27/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
Immune checkpoint inhibitors are licensed for use in patients with unresectable, recurrent or metastatic head and neck squamous cell carcinoma. Multiple published and ongoing trials are assessing efficacy in the curative management of patients in the concomitant, neoadjuvant and/or adjuvant settings, as well as part of multimodality treatment in patients with metastatic disease. This review evaluates the evidence for use of immune checkpoint inhibitors in all stages of head and neck squamous cell carcinoma and considers future approaches.
Collapse
Affiliation(s)
- Elinor R Gatfield
- Oncology Centre, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - John Tadross
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
- Cambridge Genomics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ,UK
| | - William Ince
- Oncology Centre, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| |
Collapse
|
41
|
Patel JS, McCall NS, Thomas M, Zhou J, Higgins KA, Bradley JD, Tian S, McDonald MW, Kesarwala AH, Stokes WA. Immune System Dose With Proton Versus Photon Radiotherapy for Treatment of Locally Advanced NSCLC. Int J Part Ther 2024; 12:100016. [PMID: 38832321 PMCID: PMC11145531 DOI: 10.1016/j.ijpt.2024.100016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/25/2024] [Accepted: 02/12/2024] [Indexed: 06/05/2024] Open
Abstract
Purpose Emerging data have illuminated the impact of effective radiation dose to immune cells (EDIC) on outcomes in patients with locally advanced, unresectable non-small cell lung cancer (NSCLC) treated with intensity-modulated radiotherapy (IMRT). Hypothesizing that intensity-modulated proton therapy (IMPT) may reduce EDIC versus IMRT, we conducted a dosimetric analysis of patients treated at our institution. Materials and Methods Data were retrospectively collected for 12 patients with locally advanced, unresectable NSCLC diagnosed between 2019 and 2021 who had physician-approved IMRT and IMPT plans. Data to calculate EDIC from both Jin et al (PMID: 34944813) and Ladbury et al's (PMID: 31175902) models were abstracted. Paired t tests were utilized to compare the difference in mean EDIC between IMPT and IMRT plans. Results IMPT decreased EDIC for 11 of 12 patients (91.7%). The mean EDIC per the Jin model was significantly lower with IMPT than IMRT (3.04 GyE vs 4.99 Gy, P < .001). Similarly, the mean EDIC per the Ladbury model was significantly lower with IMPT than IMRT (4.50 GyE vs 7.60 Gy, P < .002). Modeled 2-year overall survival was significantly longer with IMPT than IMRT (median 71% vs 63%; P = .03). Conclusion IMPT offers a statistically significant reduction in EDIC compared to IMRT. Given the emergence of EDIC as a modifiable prognostic factor in treatment planning, our dosimetric study highlights a potential role for IMPT to address an unmet need in improving oncologic outcomes in patients with locoregionally advanced NSCLC.
Collapse
Affiliation(s)
- Jimmy S. Patel
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Neal S. McCall
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Matthew Thomas
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jun Zhou
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kristin A. Higgins
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jeffrey D. Bradley
- Department of Radiation Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sibo Tian
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mark W. McDonald
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Aparna H. Kesarwala
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - William A. Stokes
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
42
|
Ying X, You G, Shao R. The analysis of the efficacy and safety of stereotactic body radiotherapy with sequential immune checkpoint inhibitors in the management of oligoprogressive advanced non-small cell lung cancer. Transl Cancer Res 2024; 13:2408-2418. [PMID: 38881915 PMCID: PMC11170538 DOI: 10.21037/tcr-23-2232] [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: 12/04/2023] [Accepted: 03/31/2024] [Indexed: 06/18/2024]
Abstract
Background No standardized treatment strategy exists for managing oligoprogression during maintenance therapy in driver-negative advanced non-small cell lung cancer (NSCLC). Similarly, a uniform response to oligoprogression during maintenance therapy using immune checkpoint inhibitors (ICIs) has not been established. Consequently, our investigation focused on assessing the efficacy and safety of employing stereotactic total body radiotherapy in conjunction with ICIs to address oligoprogression in advanced NSCLC. Methods We conducted a retrospective analysis of patients diagnosed with driver-negative advanced NSCLC who received stereotactic body radiotherapy (SBRT) in combination with ICIs to manage oligoprogressive lesions within the period from October 2018 to October 2023 at our institution. Oligoprogression, defined as progression occurring in three or fewer disease sites, was the focus of our investigation. Our assessment encompassed various parameters including the local control rate (LCR), progression-free survival post-oligoprogression (PFS-P), overall survival post-oligoprogression (OS-P), progression-free survival (PFS), overall survival (OS), and the safety profile associated with SBRT followed by sequential ICIs after oligoprogression. Results A total of 15 patients were enrolled in this study, all at stage IV, with 12 (80%) receiving a diagnosis of adenocarcinoma. Before oligoprogression, 11 (73.3%) patients had undergone immunotherapy. Following the treatment of oligoprogressed lung cancer with SBRT sequential ICIs, the median PFS-P and OS-P were 8 months (95% CI: 2.7-13.3) and 12 months (95% CI: 7.3-16.7), respectively. Additionally, the median PFS and OS were 26 months (95% CI: 8.0-44.0) and 30 months (not reached), respectively. The median local control (LC) of 15 oligoprogressed lesions was 13 months (95% CI: 5.3-20.2), with a 1-year LCR of 77.9%. Notably, patients with a performance status (PS) score of less than 2 demonstrated a more favorable survival benefit. Conclusions Stereotactic systemic radiation therapy, combined with sequential ICIs, enhances both LC and survival in advanced NSCLC characterized by oligoprogression and negative driver gene mutations. This approach also exhibits the potential to postpone the transition between systemic chemotherapy regimens. Manageable adverse reactions were observed, with the absence of grade 4 reactions.
Collapse
Affiliation(s)
- Xixi Ying
- Department of Radiology and Oncology, Taizhou Cancer Hospital, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Intervention and Big Data Artificial Intelligence, Taizhou, China
| | - Guangxian You
- Department of Radiology and Oncology, Taizhou Cancer Hospital, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Intervention and Big Data Artificial Intelligence, Taizhou, China
| | - Rongjun Shao
- Department of Radiology and Oncology, Taizhou Cancer Hospital, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Intervention and Big Data Artificial Intelligence, Taizhou, China
| |
Collapse
|
43
|
Tam A, Ladbury C, Kassardjian A, Modi B, McGee H, Melstrom L, Margolin K, Xing Y, Amini A. Combined Regional Approach of Talimogene laherparepvec and Radiotherapy in the Treatment of Advanced Melanoma. Cancers (Basel) 2024; 16:1951. [PMID: 38893072 PMCID: PMC11171111 DOI: 10.3390/cancers16111951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Talimogene laherparepvec (TVEC) is a genetically modified oncolytic herpes simplex virus (HSV-1) that is used for the intralesional treatment of advanced or metastatic melanoma. Given that TVEC produces the granulocyte-macrophage colony-stimulating factor (GM-CSF), recent reports have suggested that radiation treatment (RT) given in conjunction with TVEC may provide synergistic immune activation at the site, and possibly systemically. However, studies on combining RT with TVEC remain limited. We conducted a retrospective review of melanoma patients from a single cancer center who received TVEC and RT in the same region of the body and compared them to patients who received TVEC with RT at another site (other than the site of TVEC injection). Between January 2015 and September 2022, we identified twenty patients who were treated with TVEC and RT; fourteen patients received TVEC and RT in the same region, and six had treatments in separate regions. Regions were determined at the time of analysis and were based on anatomic sites (such as arm, leg, torso, etc.). Kaplan-Meier analysis of progression-free survival (PFS), analyses of time to distant metastasis (DM), overall survival (OS), and locoregional control (LRC), and the corresponding log-rank test were performed. With a median follow-up of 10.5 months [mos] (range 1.0-58.7 mos), we found an improvement in PFS with TVEC and RT in the same region compared to different regions, which were 6.4 mos (95% CI, 2.4-NR mos) and 2.8 mos (95% CI, 0.7-4.4 mos), respectively; p = 0.005. There was also a significant improvement in DM when TVEC and RT were used in the same region compared to different regions: 13.8 mos (95% CI, 4.6-NR mos) and 2.8 mos (95% CI, 0.7-4.4 mos), respectively (p = 0.001). However, we found no difference in overall survival (OS) between patients who had TVEC and RT in the same region (19.0 mos, 95% confidence interval [CI], 4.1-not reached [NR] mos) and those who received treatments in different regions (18.5 mos, 95% CI, 1.0-NR mos); p = 0.366. There was no statistically significant improvement in locoregional control (LRC) in patients who had TVEC and RT in the same region was 26.0 mos (95% CI, 6.4-26.0 mos) compared to patients who received TVEC and RT in different regions (4.4 mos) (95% CI, 0.7-NR mos) (p = 0.115). No grade 3 or higher toxicities were documented in either group. Overall, there were improvements in PFS and DM when TVEC and RT were delivered to the same region of the body compared to when they were used in different regions. However, we did not find a significant difference in locoregional recurrence or OS. Future studies are needed to assess the sequence and timing of combining RT and TVEC to potentially enhance the immune response both locally and distantly.
Collapse
Affiliation(s)
- Andrew Tam
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.)
| | - Colton Ladbury
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.)
| | - Ari Kassardjian
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.)
| | - Badri Modi
- Department of Dermatology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA
| | - Heather McGee
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.)
| | - Laleh Melstrom
- Department of Surgery, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA
| | - Kim Margolin
- St. John’s Cancer Institute, 2121 Santa Monica Blvd., Santa Monica, CA 90404, USA
| | - Yan Xing
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010, USA
| | - Arya Amini
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.)
| |
Collapse
|
44
|
Liu YL, Weigelt B. A tale of two pathways: Review of immune checkpoint inhibitors in DNA mismatch repair-deficient and microsatellite instability-high endometrial cancers. Cancer 2024; 130:1733-1746. [PMID: 38422006 PMCID: PMC11058027 DOI: 10.1002/cncr.35267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
The DNA mismatch repair (MMR) pathway is critical for correcting DNA mismatches generated during DNA replication. MMR-deficiency (MMR-D) leads to microsatellite instability (MSI) associated with an increased mutation rate, driving cancer development. This is particularly relevant in endometrial cancer (EC) as 25%-30% of tumors are of MMR-D/MSI-high (MSI-H) phenotype. Comprehensive assessment using immunohistochemistry (IHC) and sequencing-based techniques are necessary to fully evaluate ECs given the importance of molecular subtyping in staging and prognosis. This also influences treatment selection as clinical trials have demonstrated survival benefits for immune checkpoint inhibitors (ICIs) alone and in combination with chemotherapy for MMR-D/MSI-H EC patients in various treatment settings. As a portion of MMR-D/MSI-H ECs are driven by Lynch syndrome, an inherited cancer predisposition syndrome that is also associated with colorectal cancer, this molecular subtype also prompts germline assessment that can affect at-risk family members. Additionally, heterogeneity in the tumor immune microenvironment and tumor mutation burden (TMB) have been described by MMR mechanism, meaning MLH1 promoter hypermethylation versus germline/somatic MMR gene mutation, and this may affect response to ICI therapies. Variations by ancestry in prevalence and mechanism of MMR-D/MSI-H tumors have also been reported and may influence health disparities given observed differences in tumors of Black compared to White patients which may affect ICI eligibility. These observations highlight the need for additional prospective studies to evaluate the nuances regarding MMR-D heterogeneity as well as markers of resistance to inform future trials of combination therapies to further improve outcomes for patients with EC.
Collapse
Affiliation(s)
- Ying L Liu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
45
|
He M, Pan Y, You C, Gao H. CircRNAs in cancer therapy tolerance. Clin Chim Acta 2024; 558:119684. [PMID: 38649011 DOI: 10.1016/j.cca.2024.119684] [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: 03/20/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
The rapidly expanding field of circular RNA (circ-RNA) research has opened new avenues in cancer diagnostics and treatment, highlighting the role of serum circRNAs as potential biomarkers for assessing tumor therapy resistance. This review comprehensively compiles existing knowledge regarding the biogenesis, function, and clinical relevance of circRNAs, emphasizing their stability, abundance, and cell type-specific expression profiles, which make them ideal candidates for noninvasive early biomarkers in cancer treatment. We explored the roles of circRNAs in oncogenesis and tumor progression and their complex interactions with patient responses to various cancer treatments, such as chemotherapy, radiotherapy, targeted therapy, and immunotherapy. Through the analysis of data from recent studies and clinical trials, we underscore the prognostic significance of serum circRNAs in predicting therapeutic outcomes, their involvement in resistance mechanisms, and their capacity to inform personalized treatment approaches. Additionally, this review addresses the obstacles inherent in circRNA research, including the need for standardized protocols for circRNA extraction and quantification and the elucidation of the clinical significance of circRNAs. Furthermore, our investigation extends to future prospects, including embedding circRNA profiling into regular clinical workflows and pioneering circRNA-based therapeutic approaches. We underscore the transformative potential of serum circRNAs in enhancing cancer diagnosis, improving the accuracy of therapy tolerance predictions, and ultimately fostering the advent of precision oncology.
Collapse
Affiliation(s)
- Miao He
- The Second Hospital of Lanzhou University, Laboratory Medicine Center, Lanzhou 730030, PR China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, PR China
| | - Yunyan Pan
- The Second Hospital of Lanzhou University, Laboratory Medicine Center, Lanzhou 730030, PR China
| | - Chongge You
- The Second Hospital of Lanzhou University, Laboratory Medicine Center, Lanzhou 730030, PR China.
| | - Hongwei Gao
- The Second Hospital of Lanzhou University, Laboratory Medicine Center, Lanzhou 730030, PR China.
| |
Collapse
|
46
|
Ahn CR, Ha IJ, Kim JE, Ahn KS, Park J, Baek SH. Inhibiting AGS Cancer Cell Proliferation through the Combined Application of Aucklandiae Radix and Hyperthermia: Investigating the Roles of Heat Shock Proteins and Reactive Oxygen Species. Antioxidants (Basel) 2024; 13:564. [PMID: 38790669 PMCID: PMC11118127 DOI: 10.3390/antiox13050564] [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: 03/10/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer is a major global health concern. To address this, the combination of traditional medicine and newly appreciated therapeutic modalities has been gaining considerable attention. This study explores the combined effects of Aucklandiae Radix (AR) and 43 °C hyperthermia (HT) on human gastric adenocarcinoma (AGS) cell proliferation and apoptosis. We investigated the synergistic effects of AR and HT on cell viability, apoptosis, cell cycle progression, and reactive oxygen species (ROS)-dependent mechanisms. Our findings suggest that the combined treatment led to a notable decrease in AGS cell viability and increased apoptosis. Furthermore, cell cycle arrest at the G2/M phase contributed to the inhibition of cancer cell proliferation. Notably, the roles of heat shock proteins (HSPs) were highlighted, particularly in the context of ROS regulation and the induction of apoptosis. Overexpression of HSPs was observed in cells subjected to HT, whereas their levels were markedly reduced following AR treatment. The suppression of HSPs and the subsequent increase in ROS levels appeared to contribute to the activation of apoptosis, suggesting a potential role for HSPs in the combined therapy's anti-cancer mechanisms. These findings provide valuable insights into the potential of integrating AR and HT in cancer and HSPs.
Collapse
Affiliation(s)
- Chae Ryeong Ahn
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea
| | - In Jin Ha
- Korean Medicine Clinical Trial Center (K-CTC), Korean Medicine Hospital, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jai-Eun Kim
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Soeul 02447, Republic of Korea
| | - Jinbong Park
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Soeul 02447, Republic of Korea
| | - Seung Ho Baek
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea
| |
Collapse
|
47
|
Zabransky DJ, Yarchoan M, Ho WJ. Reply: The Hepa 1-6 may not be suitable for use in hepatocellular carcinoma models to explore responses to drug therapy. Hepatology 2024; 79:E125-E126. [PMID: 38147327 DOI: 10.1097/hep.0000000000000738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/27/2023]
Affiliation(s)
- Daniel J Zabransky
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark Yarchoan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Won Jin Ho
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Flow/Mass Cytometry Facility, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
48
|
Kim DW, Chul Cho B, Pachipala K, Kim SW, Wang CL, Chang GC, Ahn MJ, Alvarez R, Chiu CH, Trigo J, Estival A, Karam SD, O'Brien C, Gowda H, Jiang H, Bauman JE. Durvalumab in combination with chemoradiotherapy for patients with unresectable stage III non-small-cell lung cancer: Results from the phase 1 CLOVER study. Lung Cancer 2024; 190:107530. [PMID: 38471416 DOI: 10.1016/j.lungcan.2024.107530] [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: 12/22/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/14/2024]
Abstract
INTRODUCTION For patients with unresectable, stage III non-small-cell lung cancer (NSCLC), current standard of care is concurrent chemoradiotherapy (cCRT) followed by consolidation durvalumab. However, earlier initiation of durvalumab simultaneously with cCRT may increase antitumor activity relative to initiation after cCRT. The phase 1 CLOVER study (NCT03509012) evaluated durvalumab combined with cCRT in patients with advanced solid tumors; we report findings from the NSCLC cohort. METHODS CLOVER comprised a dose-limiting toxicity (DLT) assessment part, followed by an expansion part. In the NSCLC cohort, patients with previously untreated, unresectable, stage III NSCLC were enrolled in three treatment arms: durvalumab every 4 weeks (Q4W) + cisplatin + etoposide + radiotherapy (Arm 1); durvalumab Q4W + carboplatin + paclitaxel + radiotherapy (Arm 2); or durvalumab Q4W + carboplatin or cisplatin + pemetrexed + radiotherapy (non-squamous histology only; Arm 3). Patients received durvalumab until disease progression or unacceptable toxicity. The primary endpoint was safety and tolerability. RESULTS Sixty-four patients were enrolled: 21, 22, and 21 in Arms 1, 2, and 3, respectively. One patient in Arm 1 had DLT (grade 3 aspartate aminotransferase increase and grade 4 alanine aminotransferase increase); no DLTs were observed in Arms 2 or 3. Grade 3/4 adverse events occurred in 76.6 % of patients overall; the most common were neutropenia (51.6 %), leukopenia (20.3 %), and anemia (17.2 %). In a post-hoc analysis, 7.8 % of patients had grade 3 pneumonitis/radiation pneumonitis (grouped term) events. Overall, the objective response rate was 60.9 % (95 % confidence interval [CI], 47.9-72.9); median duration of response was 15.8 months (95 % CI, 9.0-not estimable [NE]). Median progression-free survival was 13.4 months (95 % CI, 8.8-20.1) and median overall survival was not reached (95 % CI, 21.9-NE). CONCLUSION Durvalumab in combination with cCRT was well tolerated, with a manageable safety profile and showed encouraging antitumor activity in patients with unresectable, stage III NSCLC.
Collapse
Affiliation(s)
- Dong-Wan Kim
- Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Republic of Korea.
| | - Byoung Chul Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | - Sang-We Kim
- Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | - Gee-Chen Chang
- School of Medicine and Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan; Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Myung-Ju Ahn
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Rosa Alvarez
- Hospital General Universitario Gregorio Marañón, Instituto de Investigacion Sanitaria Gregorio Marañón, Madrid, Spain
| | | | - José Trigo
- UGC Intercentros Oncología Hospital Regional y Virgen de la Victoria, Málaga, Spain
| | - Anna Estival
- Hospital Germans Trias i Pujol, Barcelona, Spain
| | - Sana D Karam
- University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | | | | | | | | |
Collapse
|
49
|
Rai P, Mahajan A, Shukla S, Agarwal U. Double Whammy: Abscopal Effect and Pseudoprogression in a Case of Non-small Cell Lung Carcinoma With Brain Metastases. Cureus 2024; 16:e59099. [PMID: 38803768 PMCID: PMC11128365 DOI: 10.7759/cureus.59099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Abscopal effect and pseudoprogression are terms used in modern oncological imaging. Abscopal effect refers to the elicitation of tumor response away from the site of primary disease. Pseudoprogression is the increase in size or enhancement of the treated tumor or the appearance of new lesions that remain stable or show subsequent decrease without any change in therapy. Both of these are known to be associated with radiation therapy. We present a case of adenocarcinoma of the lung, which developed both these phenomena throughout the course of their therapy. Out-of-target responses secondary to radiotherapy have been discussed extensively in the literature and may pave the way for future oncological management as the targeted therapies become more specific. At the same time, atypical, however not uncommon, phenomena such as pseudoprogression should always be kept in the back of a clinician's mind as further course of clinical management may change.
Collapse
Affiliation(s)
- Pranjal Rai
- Radiodiagnosis, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, IND
| | - Abhishek Mahajan
- Imaging Department, The Clatterbridge Cancer Centre National Health Service (NHS), Liverpool, GBR
| | - Shreya Shukla
- Radiodiagnosis, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, IND
| | - Ujjwal Agarwal
- Radiodiagnosis, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, IND
| |
Collapse
|
50
|
Verma S, Young S, Boldt G, Blanchette P, Lock M, Helou J, Raphael J. Immunotherapy and Radiation Therapy Sequencing in Breast Cancer: A Systematic Review. Int J Radiat Oncol Biol Phys 2024; 118:1422-1434. [PMID: 38195030 DOI: 10.1016/j.ijrobp.2024.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/01/2024] [Indexed: 01/11/2024]
Abstract
PURPOSE In the past decade, immune checkpoint inhibitors (ICIs) have emerged as a treatment option for metastatic breast cancer (BC). More recently, ICIs have been approved in the perioperative setting. This has led to clinical scenarios where radiation therapy (RT) is given concurrently with ICIs. On the other hand, moderate and ultrahypofractionated schedules of RT are being widely adopted in the adjuvant setting, in addition to an increased use of metastasis-directed therapy. Furthermore, RT can modulate the tumor microenvironment and induce a systemic response at nonirradiated sites, an "abscopal effect." The amplification of antitumor immune response is used as the rationale behind the concomitant use of ICIs and RT. To date, there is a lack of literature on the optimal sequence, timing, dose/fractionation schema, and treated RT volumes with ICIs in patients with BC, especially in the era of ultrahypofractionation. METHODS AND MATERIALS We conducted a systematic review to delineate the reported treatment details, safety, and efficacy of combining ICI and RT in patients with BC. PubMed, Embase, and Cochrane CENTRAL were searched between 2014 and 2023. Data were extracted to assess the details of ICIs/RT delivery, safety, and efficacy. RESULTS Of the 12 eligible studies, 9 involved patients with metastatic BC. Most studies were phase 1/2, had a small sample size (range, 8-28), and were heterogenous in patient population and reported outcomes. The combination was reported to be safe. We identified 1 study in the perioperative setting, which did a posthoc analysis of safety/efficacy of ICIs in the adjuvant setting with receipt and pattern of RT. CONCLUSIONS In conclusion, there are limited data on the dose, timing, fractionation, and volumes of RT in both the adjuvant and metastatic setting in BC. Ongoing/future trials should collect and report such data on RT details, whenever RT is used in combination with ICIs.
Collapse
Affiliation(s)
- Saurav Verma
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; London Regional Cancer Program at London Health Sciences Centre, London, Ontario, Canada
| | - Sympascho Young
- London Regional Cancer Program at London Health Sciences Centre, London, Ontario, Canada; Division of Radiation Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Gabriel Boldt
- London Regional Cancer Program at London Health Sciences Centre, London, Ontario, Canada
| | - Phillip Blanchette
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; London Regional Cancer Program at London Health Sciences Centre, London, Ontario, Canada
| | - Michael Lock
- London Regional Cancer Program at London Health Sciences Centre, London, Ontario, Canada; Division of Radiation Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Joelle Helou
- London Regional Cancer Program at London Health Sciences Centre, London, Ontario, Canada; Division of Radiation Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Jacques Raphael
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; London Regional Cancer Program at London Health Sciences Centre, London, Ontario, Canada.
| |
Collapse
|