1
|
Kim Y, Jeon SH, Kim S, Kang MH, Han MG, Lee SY, Kim IA. In vitro-irradiated cancer vaccine enhances anti-tumor efficacy of radiotherapy and PD-L1 blockade in a syngeneic murine breast cancer model. Radiother Oncol 2024; 200:110480. [PMID: 39159681 DOI: 10.1016/j.radonc.2024.110480] [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: 03/18/2024] [Revised: 07/18/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND AND PURPOSE Local radiotherapy (RT) exerts immunostimulatory effects by inducing immunogenic cell death. However, it remains unknown whether in vitro-irradiated tumor cells can elicit anti-tumor responses and enhance the efficacy of local RT and immune checkpoint inhibitors when injected in vivo. METHODS AND MATERIALS We tested the "in vitro-irradiated cancer vaccine (ICV)", wherein tumor cells killed by varying doses of irradiation and their supernatants are intravenously injected. We examined the efficacy of combining local RT (24 Gy in three fractions), PD-L1 blockade, and the ICV in a murine breast cancer model. The immune cell profiles were analyzed via flow cytometry and immunohistochemistry. The cytokine levels were measured by multiplex immunoassays. RESULTS The ICV significantly increased the effector memory phenotype and interferon-γ production capacity in splenic CD8+ T cells. The in vitro-irradiated products contained immune response-related molecules. When combined with local RT and PD-L1 blockade, the ICV significantly delayed the growth of irradiated and non-irradiated tumors. The triple combination therapy increased the proportions of CD8+ T cells and effector memory CD8+ T cells while decreasing the proportion of CTLA-4+ exhausted CD8+ T cells within tumor microenvironment. Additionally, plasma level of interferon-γ and proliferation of effector T cells in the spleen and tumor-draining lymph nodes were significantly increased by the triple combination therapy. CONCLUSIONS The ICV enhanced the therapeutic efficacy of local RT and PD-L1 blockade by augmenting anti-tumor immune responses. Our findings suggest a therapeutic potential of in vitro-irradiation products of tumor cells.
Collapse
Affiliation(s)
- Yoomin Kim
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
| | - Seung Hyuck Jeon
- Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Seongmin Kim
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Integrated Major in Innovative Medical Science, Seoul National University, Seoul, Republic of Korea
| | - Mi Hyun Kang
- Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Min Guk Han
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Se Yup Lee
- Korea Nuclear Engineering Co., Ltd, Seoul, Republic of Korea
| | - In Ah Kim
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Integrated Major in Innovative Medical Science, Seoul National University, Seoul, Republic of Korea; Department of Radiation Oncology, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Lopez-Bujanda ZA, Hadavi SH, Ruiz De Porras V, Martínez-Balibrea E, Dallos MC. Chemotactic signaling pathways in prostate cancer: Implications in the tumor microenvironment and as potential therapeutic targets. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 388:162-205. [PMID: 39260936 DOI: 10.1016/bs.ircmb.2024.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Prostate cancer (PCa) stands as a significant global health concern, ranking among the leading causes of cancer deaths in men. While there are several treatment modalities for localized PCa, metastatic castration-resistant PCa (mCRPC) remains incurable. Despite therapeutic advancements showing promise in mCRPC, their impact on overall survival has been limited. This chapter explores the process by which tumors form, reviews our current understanding of PCa progression to mCRPC, and addresses the challenges of boosting anti-tumor immune responses in these tumors. It specifically discusses how chemotactic signaling affects the tumor microenvironment and its role in immune evasion and cancer progression. The chapter further examines the rationale of directly or indirectly targeting these pathways as adjuvant therapies for mCRPC, highlighting recent pre-clinical and clinical studies currently underway. The discussion emphasizes the potential of targeting specific chemokines and chemokine receptors as combination therapies with mainstream treatments for PCa and mCRPC to maximize long-term survival for this deadly disease.
Collapse
Affiliation(s)
- Zoila A Lopez-Bujanda
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, United States.
| | - Shawn H Hadavi
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Vicenç Ruiz De Porras
- Badalona Applied Research Group of Oncology (B-ARGO), Catalan Institute of Oncology, Badalona, BCN, Spain; CARE program, Germans Trias i Pujol Research Institute (IGTP), Badalona, BCN, Spain
| | - Eva Martínez-Balibrea
- CARE program, Germans Trias i Pujol Research Institute (IGTP), Badalona, BCN, Spain; ProCURE Program, Catalan Institute of Oncology, Badalona, BCN, Spain
| | - Matthew C Dallos
- Memorial Solid Tumor Group, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
3
|
Song JY, Han MG, Kim Y, Kim MJ, Kang MH, Jeon SH, Kim IA. Combination of local radiotherapy and anti-glucocorticoid-induced tumor necrosis factor receptor (GITR) therapy augments PD-L1 blockade-mediated anti-tumor effects in murine breast cancer model. Radiother Oncol 2024; 190:109981. [PMID: 37925106 DOI: 10.1016/j.radonc.2023.109981] [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: 05/03/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/06/2023]
Abstract
PURPOSE In this study, we investigated whether local radiotherapy (RT) and an anti-glucocorticoid-induced tumor necrosis factor receptor (GITR) agonist could increase the efficacy of PD-L1 blockade. METHODS AND MATERIALS We analyzed a breast cancer dataset from the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) to determine the role of GITR in breast cancer. We used the 4T1 murine TNBC model (primary and secondary tumors) to investigate the efficacy of PD-L1 blockade, local RT, anti-GITR agonist, and their combinations. We assessed tumor growth by tumor volume measurements, in vivo bioluminescence imaging, and metastatic lung nodule counts to evaluate the effects of these treatments. Flow cytometry and immunohistochemistry determined the proportions and phenotypes of CD8+ T-cells and regulatory T-cells (Tregs) in the tumors and spleen. Plasma cytokine levels were measured by enzyme-linked immunosorbent assay. RESULTS In the METABRIC cohort, patients with high expression of TNFRSF18, which encodes GITR, had significantly better survival than those with low expression. Adding local RT or anti-GITR agonist to PD-L1 blockade did not significantly augment efficacy compared to PD-L1 blockade alone; however, adding both to PD-L1 blockade significantly reduced tumor growth and lung metastasis. The benefits of the triple combination were accompanied by increased CD8+ T-cells and decreased Tregs in the tumor microenvironment and spleen. CONCLUSIONS The combination of local RT and an anti-GITR agonist significantly enhanced the anti-tumor immune responses induced by PD-L1 blockade. These results provide the preclinical rationale for the combination of therapy.
Collapse
Affiliation(s)
- Jun Yeong Song
- Department of Radiation Oncology, Seoul National University School of Medicine, Republic of Korea.
| | - Min Guk Han
- Medical Science Research Institute, Seoul National University Bundang Hospital, Republic of Korea
| | - Yoomin Kim
- Department of Tumor Biology, Graduate School of Medicine & Cancer Research Institute, Seoul National University, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Republic of Korea
| | - Min Ji Kim
- Department of Tumor Biology, Graduate School of Medicine & Cancer Research Institute, Seoul National University, Republic of Korea
| | - Mi Hyun Kang
- Medical Science Research Institute, Seoul National University Bundang Hospital, Republic of Korea
| | - Seung Hyuck Jeon
- Department of Radiation Oncology, Seoul National University Bundang Hospital, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Republic of Korea
| | - In Ah Kim
- Department of Radiation Oncology, Seoul National University School of Medicine, Republic of Korea; Department of Radiation Oncology, Seoul National University Bundang Hospital, Republic of Korea; Department of Tumor Biology, Graduate School of Medicine & Cancer Research Institute, Seoul National University, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Republic of Korea.
| |
Collapse
|
4
|
Chen L, Shi V, Wang S, Freeman R, Ruiz F, Jayachandran K, Zhang J, Cosper P, Sun L, Luke CJ, Spina C, Grigsby PW, Schwarz JK, Markovina S. SCCA1/SERPINB3 promotes suppressive immune environment via STAT-dependent chemokine production, blunting the therapy-induced T cell responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526675. [PMID: 36778224 PMCID: PMC9915608 DOI: 10.1101/2023.02.01.526675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Radiotherapy is a commonly used cancer treatment; however, patients with high serum squamous cell carcinoma antigen (SCCA1/SERPINB3) are associated with resistance and poor prognosis. Despite being a strong clinical biomarker, the modulation of SERPINB3 in tumor immunity is poorly understood. We investigated the microenvironment of SERPINB3 high tumors through RNAseq of primary cervix tumors and found that SERPINB3 was positively correlated with CXCL1/8, S100A8/A9 and myeloid cell infiltration. Induction of SERPINB3 in vitro resulted in increased CXCL1/8 and S100A8/A9 production, and supernatants from SERPINB3-expressing cultures attracted monocytes and MDSCs. In murine tumors, the orthologue mSerpinB3a promoted MDSC, TAM, and M2 macrophage infiltration contributing to an immunosuppressive phenotype, which was further augmented upon radiation. Radiation-enhanced T cell response was muted in SERPINB3 tumors, whereas Treg expansion was observed. A STAT-dependent mechanism was implicated, whereby inhibiting STAT signaling with ruxolitinib abrogated suppressive chemokine production. Patients with elevated pre-treatment serum SCCA and high pSTAT3 had increased intratumoral CD11b+ myeloid cell compared to patients with low SCCA and pSTAT3 cohort that had overall improved cancer specific survival after radiotherapy. These findings provide a preclinical rationale for targeting STAT signaling in tumors with high SERPINB3 to counteract the immunosuppressive microenvironment and improve response to radiation.
Collapse
|
5
|
Predicting tumour radiosensitivity to deliver precision radiotherapy. Nat Rev Clin Oncol 2023; 20:83-98. [PMID: 36477705 DOI: 10.1038/s41571-022-00709-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2022] [Indexed: 12/13/2022]
Abstract
Owing to advances in radiotherapy, the physical properties of radiation can be optimized to enable individualized treatment; however, optimization is rarely based on biological properties and, therefore, treatments are generally planned with the assumption that all tumours respond similarly to radiation. Radiation affects multiple cellular pathways, including DNA damage, hypoxia, proliferation, stem cell phenotype and immune response. In this Review, we summarize the effect of these pathways on tumour responses to radiotherapy and the current state of research on genomic classifiers designed to exploit these variations to inform treatment decisions. We also discuss whether advances in genomics have generated evidence that could be practice changing and whether advances in genomics are now ready to be used to guide the delivery of radiotherapy alone or in combination.
Collapse
|
6
|
Abstract
Radiation therapy continues to break down technological barriers to deliver ionizing radiation with exceptional anatomical precision. However, some tumor types and subtypes exhibit intrinsic biological resistance to radiotherapy, which can result in unsuccessful tumor eradication or symptom palliation. Radiation resistance can result from alterations in diverse genetic, epigenetic, and metabolic pathways. Therapeutic targeting of these tumor-specific alterations may provide tumor-selective radiosensitization with relative sparing of adjacent normal tissues. This issue of Seminars in Radiation Oncology presents a series of articles that describe recent progress towards genomically-directed radiosensitization.
Collapse
Affiliation(s)
- Gaorav P Gupta
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC.
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale University, New Haven, CT.
| |
Collapse
|