1
|
Wang L, Ren S, Lou J, Xue S, Zhou P, Zheng X, Shan F, Li X, Chen Y, Liu X. E-Urea-K-Based PSMA Imaging Served as an Alternative in Assessing Tumor Neovascularization via Targeting CD31. Mol Pharm 2025; 22:2029-2039. [PMID: 40013667 DOI: 10.1021/acs.molpharmaceut.4c01252] [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: 02/28/2025]
Abstract
To reveal the natural correlation between prostate-specific membrane antigen (PSMA) imaging and tumor neovascularization in prostate cancer and further explore E-urea-K-based PSMA-targeted (EK-PSMA) imaging as a potential indicator of tumor neovascularization, the 22Rv1 mouse models were established and underwent 99mTc-HYNIC-ALUG SPECT/CT. Pearson correlation analysis was applied to assess the relationship between tumor tracer uptake and tumor characteristics, including size, glucose metabolism, and cell phenotypes (e.g., Ki-67, VEGF, CD31, and PSMA). Then, molecular docking further identified the key factors of EK-PSMA imaging, specifically related to tumor neovascularization. Finally, animal models with positive and negative PSMA expression (22Rv1, LNCaP, U87, SAOS-2, A549, and ACHN) were subjected to antibody-targeted blockade to verify the role of these key factors in EK-PSMA imaging. The Pearson's r values of tracer uptake correlated with CD31 and tumor size were 0.82 and 0.99, respectively (P < 0.05), and the correlations of tracer uptake with SUVmax, SUVmean, Ki-67, VEGF, and PSMA expressions were 0.47, 0.20, 0.69, -0.65, and 0.20, respectively (all P > 0.05). Molecular docking confirmed the affinity of E-urea-K to PSMA (two sites, binding scores, -5.4 kcal/mol and -6.0 kcal/mol) and CD31 (one site, binding score, -5.1 kcal/mol). The blockade of the CD31 antibody partially reduced the 99mTc-HYNIC-ALUG uptake in five other types of tumors (paired t test, P = 0.0478). The Pearson's r value of CD31 staining and tracer uptake prior to the antibody blockade was 0.84 (P < 0.05). Additionally, when removing the PSMA-positive models (22Rv1 and LNCaP), the Pearson's r value of CD31 staining and tracer uptake prior to the antibody blockade was 0.99 (P < 0.05). Thus, CD31 was found to be a mutual target of EK-PSMA imaging; therefore, EK-PSMA imaging provides a viable assessment option for tumor neovascularization, especially for PSMA-negative tumors.
Collapse
Affiliation(s)
- Lan Wang
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Shengnan Ren
- Department of Nuclear Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Jingjing Lou
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Shuai Xue
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Pan Zhou
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Xiaobei Zheng
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Fengling Shan
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Xiao Li
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- Department of Radiology, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Yangchun Chen
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Xingdang Liu
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
2
|
Shiozawa Y, Parajuli KR, Pienta K, Taichman R. Role of Chemokines and Cytokines in Prostate Cancer Skeletal Metastasis. Curr Osteoporos Rep 2024; 23:3. [PMID: 39585513 DOI: 10.1007/s11914-024-00897-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/31/2024] [Indexed: 11/26/2024]
Abstract
PURPOSE OF REVIEW Once prostate cancer (PCa) bone metastases develop, the prognosis dramatically declines. The precise mechanisms regulating bone metastasis remain elusive. This review will explore recent findings related to cytokines and chemokines in the process of bone metastases. RECENT FINDINGS We discuss the role of cytokines in tumor growth, invasion, bone remodelling and angiogenesis and immune regulation in PCa skeletal metastases. Major advances in our understanding focus on immune evasion, immune checkpoint blockade, tumor-associated macrophages (TAMs), CAR-T cells, cytokine regulation of matrix metalloproteinases, cytokines including IL-10, IL-27, Interferon-γ, prostate transmembrane protein androgen induced 1 (Pmepa1), and regulation of RUNX2 transcription in supporting survival and growth of disseminated tumor cells (DTCs) and metastases development. The review highlights the complexity of cytokine actions in PCa bone metastases, suggesting potential therapeutic targets to disrupt interactions between cancer cells and their microenvironment.
Collapse
Affiliation(s)
- Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, 27157, NC, USA.
| | - Keshab Raj Parajuli
- Department of Periodontology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth Pienta
- Cancer Ecology Center, Johns Hopkins School of Medicine, The Brady Urological Institute, 600 N. Wolfe St., Marburg Building Room 113, Baltimore, 21287, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Whiting School of Engineering, Baltimore, MD, USA
| | - Russell Taichman
- Department of Periodontology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Cancer Ecology Center, Johns Hopkins School of Medicine, The Brady Urological Institute, 600 N. Wolfe St., Marburg Building Room 113, Baltimore, 21287, MD, USA.
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.
- Department of Basic & Clinical Translational Sciences, Tufts University School of Dental Medicine, Boston, MA, USA.
| |
Collapse
|
3
|
Raymakers L, Demmers TJ, Meijer GJ, Molenaar IQ, van Santvoort HC, Intven MPW, Leusen JHW, Olofsen PA, Daamen LA. The Effect of Radiation Treatment of Solid Tumors on Neutrophil Infiltration and Function: A Systematic Review. Int J Radiat Oncol Biol Phys 2024; 120:845-861. [PMID: 39009323 DOI: 10.1016/j.ijrobp.2024.07.2141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/13/2024] [Accepted: 07/02/2024] [Indexed: 07/17/2024]
Abstract
Radiation therapy (RT) initiates a local and systemic immune response which can induce antitumor immunity and improve immunotherapy efficacy. Neutrophils are among the first immune cells that infiltrate tumors after RT and are suggested to be essential for the initial antitumor immune response. However, neutrophils in tumors are associated with poor outcomes and RT-induced neutrophil infiltration could also change the composition of the tumor microenvironment (TME) in favor of tumor progression. To improve RT efficacy for patients with cancer it is important to understand the interplay between RT and neutrophils. Here, we review the literature on how RT affects the infiltration and function of neutrophils in the TME of solid tumors, using both patients studies and preclinical murine in vivo models. In general, it was found that neutrophil levels increase and reach maximal levels in the first days after RT and can remain elevated up to 3 weeks. Most studies report an immunosuppressive role of neutrophils in the TME after RT, caused by upregulated expression of neutrophil indoleamine 2,3-dioxygenase 1 and arginase 1, as well as neutrophil extracellular trap formation. RT was also associated with increased reactive oxygen species production by neutrophils, which can both improve and inhibit antitumor immunity. In addition, multiple murine models showed improved RT efficacy when depleting neutrophils, suggesting that neutrophils have a protumor phenotype after RT. We conclude that the role of neutrophils should not be overlooked when developing RT strategies and requires further investigation in specific tumor types. In addition, neutrophils can possibly be exploited to enhance RT efficacy by combining RT with neutrophil-targeting therapies.
Collapse
Affiliation(s)
- Léon Raymakers
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Radiation Oncology, University Medical Center Utrecht, UMC Utrecht Cancer Center, Utrecht, The Netherlands
| | - Thijs J Demmers
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gert J Meijer
- Department of Radiation Oncology, University Medical Center Utrecht, UMC Utrecht Cancer Center, Utrecht, The Netherlands
| | - I Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital Nieuwegein, Utrecht University, Utrecht, The Netherlands
| | - Hjalmar C van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital Nieuwegein, Utrecht University, Utrecht, The Netherlands
| | - Martijn P W Intven
- Department of Radiation Oncology, University Medical Center Utrecht, UMC Utrecht Cancer Center, Utrecht, The Netherlands
| | - Jeanette H W Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Patricia A Olofsen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lois A Daamen
- Department of Radiation Oncology, University Medical Center Utrecht, UMC Utrecht Cancer Center, Utrecht, The Netherlands; Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital Nieuwegein, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
4
|
Lynch C, Pitroda SP, Weichselbaum RR. Radiotherapy, immunity, and immune checkpoint inhibitors. Lancet Oncol 2024; 25:e352-e362. [PMID: 39089313 DOI: 10.1016/s1470-2045(24)00075-5] [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: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/29/2024] [Indexed: 08/03/2024]
Abstract
Radiotherapy exerts immunostimulatory and immunosuppressive effects, both locally, within the irradiated tumour microenvironment, and systemically, outside the radiation field. Inspired by preclinical data that showed synergy between radiotherapy and immune checkpoint inhibitors, multiple clinical trials were initiated with the hypothesis that combined treatment with radiotherapy and immune checkpoint inhibitors could stimulate a robust systemic immune response and improve clinical outcomes. However, despite early optimism, radioimmunotherapy trials in the curative and metastatic settings have met with little success. In this Review, we summarise the immunostimulatory effects of radiotherapy that provided the theoretical basis for trials of combination radiotherapy and immune checkpoint inhibitors. We also discuss findings from clinical trials incorporating radiotherapy and immune checkpoint inhibitors and examine the success of these trials in the context of the immunosuppressive effects of radiotherapy. We conclude by highlighting targets for relieving radiotherapy-induced immunosuppression with the goal of enhancing the combined effects of radiotherapy and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Connor Lynch
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Sean P Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
5
|
Hou Y, Yang K, Wang L, Wang J, Huang X, Piffko A, Luo SZ, Yu X, Rao E, Martinez C, Bugno J, Mack M, Vokes EE, Pitroda SP, Chmura SJ, Weichselbaum RR, Liang HL. Radiotherapy Enhances Metastasis Through Immune Suppression by Inducing PD-L1 and MDSC in Distal Sites. Clin Cancer Res 2024; 30:1945-1958. [PMID: 38427437 PMCID: PMC11062826 DOI: 10.1158/1078-0432.ccr-23-3206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/22/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
PURPOSE Radiotherapy (RT) is a widely employed anticancer treatment. Emerging evidence suggests that RT can elicit both tumor-inhibiting and tumor-promoting immune effects. The purpose of this study is to investigate immune suppressive factors of radiotherapy. EXPERIMENTAL DESIGN We used a heterologous two-tumor model in which adaptive concomitant immunity was eliminated. RESULTS Through analysis of PD-L1 expression and myeloid-derived suppressor cells (MDSC) frequencies using patient peripheral blood mononuclear cells and murine two-tumor and metastasis models, we report that local irradiation can induce a systemic increase in MDSC, as well as PD-L1 expression on dendritic cells and myeloid cells, and thereby increase the potential for metastatic dissemination in distal, nonirradiated tissue. In a mouse model using two distinct tumors, we found that PD-L1 induction by ionizing radiation was dependent on elevated chemokine CXCL10 signaling. Inhibiting PD-L1 or MDSC can potentially abrogate RT-induced metastasis and improve clinical outcomes for patients receiving RT. CONCLUSIONS Blockade of PD-L1/CXCL10 axis or MDSC infiltration during irradiation can enhance abscopal tumor control and reduce metastasis.
Collapse
Affiliation(s)
- Yuzhu Hou
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University; Xi’an, ShaanXi 710061, China
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Jiaai Wang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Xiaona Huang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Andras Piffko
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Sean Z. Luo
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: Biomedical Engineering program, Northwestern University; Evanston, IL 60201, USA
| | - Xinshuang Yu
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: Department of Oncology, First Affiliated Hospital of Shandong, First Medical University and Shandong Provincial Qianfoshan Hospital; Jinan, Shandong 250014, China
| | - Enyu Rao
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: Cancer Institute, Xuzhou Medical University; Xuzhou, Jiangsu 221004, China
| | - Carlos Martinez
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: University of Illinois at Chicago, Chicago, IL, 60607 USA
| | - Jason Bugno
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- The Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, IL 600637, USA
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, 93042 Regensburg, Germany
| | - Everett E. Vokes
- Department of Medicine, University of Chicago, Chicago, IL, 60637 USA
| | - Sean P. Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Steven J. Chmura
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
| | - Ralph R. Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Hua Laura Liang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| |
Collapse
|
6
|
Kim H, Lee E, Cho H, Kim E, Jang WI, Yang K, Lee YJ, Kim TJ, Kim MS. Five-Day Spacing of Two Fractionated Ablative Radiotherapies Enhances Antitumor Immunity. Int J Radiat Oncol Biol Phys 2024; 118:498-511. [PMID: 37717785 DOI: 10.1016/j.ijrobp.2023.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/10/2023] [Accepted: 09/09/2023] [Indexed: 09/19/2023]
Abstract
PURPOSE This study aimed to enhance tumor control and abscopal effects by applying diverse stereotactic ablative radiation therapy (SABR) schedules. METHODS AND MATERIALS FSaII, CT-26, and 4T1 cells were used for tumor growth delay and lung metastases analysis after 1- or 5-day intervals radiation therapy (RT) with 40, 20, and 20 Gy, respectively. Immunodeficient BALB/c-nude, immunocompetent C3H, and BALB/c mouse models were used. For immune monitoring, FSaII tumors were analyzed using flow cytometry, immunofluorescence staining, and real-time quantitative reverse transcription polymerase chain reaction. The spleens were used for the ELISpot assay and flow cytometry to determine effector CD8 T cells. For abscopal effect analysis in CT-26 tumors, the volume of the nonirradiated secondary tumors was measured after primary tumors were irradiated with 1-day or 5-day intervals. RESULTS Contrary to the high-dose 1-day interval RT, the 5-day interval RT significantly delayed tumor growth in immunocompetent mice, which was not observed in immunodeficient mice. In addition, the 5-day interval RT significantly reduced the number of lung metastases in FSaII and CT-26 tumors. Five-day spacing was more effective than 1-day interval in enhancing the antitumor immunity via increasing the secretion of tumor-specific IFN-γ, activating the CD8 T cells, and suppressing the monocytic myeloid-derived suppressor cells. The 5-day spacing inhibited nonirradiated secondary tumor growth more effectively than did the 1-day interval. CONCLUSIONS Compared with the 1-day interval RT, the 5-day interval RT scheme demonstrated enhanced antitumor immunity of CD8 T cells associated with inhibition of myeloid-derived suppressor cells. Enhancing antitumor immunity leads to significant improvements in both primary tumor control and the abscopal effect.
Collapse
Affiliation(s)
| | - Eunju Lee
- Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea; Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul, Korea
| | - Haeun Cho
- Departments of Radiation Oncology and; Department of Radiological & Medico-Oncological Science, University of Science and Technology, Daejeon, Korea
| | - Eunji Kim
- Departments of Radiation Oncology and
| | | | | | - Yoon-Jin Lee
- Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Tae-Jin Kim
- Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea.
| | - Mi-Sook Kim
- Departments of Radiation Oncology and; Department of Radiological & Medico-Oncological Science, University of Science and Technology, Daejeon, Korea.
| |
Collapse
|
7
|
McMahon RA, D'Souza C, Neeson PJ, Siva S. Innate immunity: Looking beyond T-cells in radiation and immunotherapy combinations. Neoplasia 2023; 46:100940. [PMID: 37913654 PMCID: PMC10637988 DOI: 10.1016/j.neo.2023.100940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
Radiation therapy is an established and effective anti-cancer treatment modality. Extensive pre-clinical experimentation has demonstrated that the pro-inflammatory properties of irradiation may be synergistic with checkpoint immunotherapy. Radiation induces double-stranded DNA breaks (dsDNA). Sensing of the dsDNA activates the cGAS/STING pathway, producing Type 1 interferons essential to recruiting antigen-presenting cells (APCs). Radiation promotes cytotoxic CD8 T-cell recruitment by releasing tumour-associated antigens captured and cross-presented by surveying antigen-presenting cells. Radiation-induced vascular normalisation may further promote T-cell trafficking and drug delivery. Radiation is also immunosuppressive. Recruitment of regulatory T cells (Tregs) and innate cells such as myeloid-derived suppressive cells (m-MDSCs) all counteract the immunostimulatory properties of radiation. Many innate immune cell types operate at the interface of the adaptive immune response. Innate immune cells, such as m-MDSCs, can exert their immunosuppressive effects by expressing immune checkpoints such as PD-L1, further highlighting the potential of combined radiation and checkpoint immunotherapy. Several early-phase clinical studies investigating the combination of radiation and immunotherapy have been disappointing. A greater appreciation of radiotherapy's impact on the innate immune system is essential to optimise radioimmunotherapy combinations. This review will summarise the impact of radiotherapy on crucial cells of the innate immune system and vital immunosuppressive cytokines.
Collapse
Affiliation(s)
- R A McMahon
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.
| | - C D'Souza
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia; Cancer Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - P J Neeson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia; Cancer Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - S Siva
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Chang CH, Chen CJ, Yu CF, Tsai HY, Chen FH, Chiang CS. Targeting M-MDSCs enhances the therapeutic effect of BNCT in the 4-NQO-induced murine head and neck squamous cell carcinoma model. Front Oncol 2023; 13:1263873. [PMID: 37886177 PMCID: PMC10598372 DOI: 10.3389/fonc.2023.1263873] [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: 07/20/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023] Open
Abstract
Purpose Malignant head and neck squamous cell carcinoma (HNSCC) is characterized by a poor prognosis and resistance to conventional radiotherapy. Infiltrating myeloid-derived suppressive cells (MDSCs) is prominent in HNSCC and is linked to immune suppression and tumor aggressiveness. This study aimed to investigate the impact of boron neutron capture therapy (BNCT) on the MDSCs in the tumor microenvironment and peripheral blood and to explore the potential for MDSCs depletion combined with BNCT to reactivate antitumor immunity. Methods and materials Carcinogen, 4-NQO, -induced oral tumors were irradiated with a total physical dose of 2 Gy BNCT in Tsing Hua Open Reactor (THOR). Flow cytometry and immunohistochemistry accessed the dynamics of peripheral MDSCs and infiltrated MDSCs within the tumor microenvironment. Mice were injected with an inhibitor of CSF-1 receptor (CSF-1R), PLX3397, to determine whether modulating M-MDSCs could affect mice survival after BNCT. Results Peripheral CD11b+Ly6ChighLy6G- monocytic-MDSCs (M-MDSCs), but not CD11b+Ly6CloLy6Ghigh polymorphonuclear-MDSCs (PMN-MDSCs), increased as tumor progression. After BNCT treatment, there were temporarily decreased and persistent increases of M-MDSCs thereafter, either in peripheral blood or in tumors. The administration of PLX-3397 hindered BNCT-caused M-MDSCs infiltration, prolonged mice survival, and activated tumor immunity by decreasing tumor-associated macrophages (TAMs) and increasing CD8+ T cells. Conclusion M-MDSCs were recruited into 4-NQO-induced tumors after BNCT, and their number was also increased in peripheral blood. Assessment of M-MDSCs levels in peripheral blood could be an index to determine the optimal intervention window. Their temporal alteration suggests an association with tumor recurrence after BNCT, making M-MDSCs a potential intervention target. Our preliminary results showed that PLX-3397 had strong M-MDSCs, TAMs, and TIL (tumor-infiltrating lymphocyte) modulating effects that could synergize tumor control when combined with BNCT.
Collapse
Affiliation(s)
- Chun-Hsiang Chang
- Department of Biomedical Engineering and Environment Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chi-Jui Chen
- Department of Biomedical Engineering and Environment Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Fang Yu
- Institute for Radiological Research, Chang Gung University, Taoyuan, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan, Taiwan
| | - Hui-Yu Tsai
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Fang-Hsin Chen
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environment Sciences, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
9
|
Wang L, Dou X, Chen S, Yu X, Huang X, Zhang L, Chen Y, Wang J, Yang K, Bugno J, Pitroda S, Ding X, Piffko A, Si W, Chen C, Jiang H, Zhou B, Chmura SJ, Luo C, Liang HL, He C, Weichselbaum RR. YTHDF2 inhibition potentiates radiotherapy antitumor efficacy. Cancer Cell 2023; 41:1294-1308.e8. [PMID: 37236197 PMCID: PMC10524856 DOI: 10.1016/j.ccell.2023.04.019] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/23/2022] [Accepted: 04/28/2023] [Indexed: 05/28/2023]
Abstract
RNA N6-methyladenosine (m6A) modification is implicated in cancer progression. However, the impact of m6A on the antitumor effects of radiotherapy and the related mechanisms are unknown. Here we show that ionizing radiation (IR) induces immunosuppressive myeloid-derived suppressor cell (MDSC) expansion and YTHDF2 expression in both murine models and humans. Following IR, loss of Ythdf2 in myeloid cells augments antitumor immunity and overcomes tumor radioresistance by altering MDSC differentiation and inhibiting MDSC infiltration and suppressive function. The remodeling of the landscape of MDSC populations by local IR is reversed by Ythdf2 deficiency. IR-induced YTHDF2 expression relies on NF-κB signaling; YTHDF2 in turn leads to NF-κB activation by directly binding and degrading transcripts encoding negative regulators of NF-κB signaling, resulting in an IR-YTHDF2-NF-κB circuit. Pharmacological inhibition of YTHDF2 overcomes MDSC-induced immunosuppression and improves combined IR and/or anti-PD-L1 treatment. Thus, YTHDF2 is a promising target to improve radiotherapy (RT) and RT/immunotherapy combinations.
Collapse
Affiliation(s)
- Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Xiaoyang Dou
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, University of Chicago, Chicago, IL 60637, USA
| | - Shijie Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xianbin Yu
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, University of Chicago, Chicago, IL 60637, USA
| | - Xiaona Huang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Linda Zhang
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, University of Chicago, Chicago, IL 60637, USA
| | - Yantao Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jiaai Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Jason Bugno
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA; The Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, IL 600637, USA
| | - Sean Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Xingchen Ding
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Andras Piffko
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA; Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Wei Si
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Chao Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hualiang Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Bing Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Steven J Chmura
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Cheng Luo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China.
| | - Hua Laura Liang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA.
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, University of Chicago, Chicago, IL 60637, USA; Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA.
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
10
|
Sadhukhan P, Seiwert TY. The role of macrophages in the tumor microenvironment and tumor metabolism. Semin Immunopathol 2023; 45:187-201. [PMID: 37002376 DOI: 10.1007/s00281-023-00988-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/08/2023] [Indexed: 04/03/2023]
Abstract
The complexity and plasticity of the tumor microenvironment (TME) make it difficult to fully understand the intratumoral regulation of different cell types and their activities. Macrophages play a crucial role in the signaling dynamics of the TME. Among the different subtypes of macrophages, tumor-associated macrophages (TAMs) are often associated with poor prognosis, although some subtypes of TAMs can at the same time improve treatment responsiveness and lead to favorable clinical outcomes. TAMs are key regulators of cancer cell proliferation, metastasis, angiogenesis, extracellular matrix remodeling, tumor metabolism, and importantly immunosuppression in the TME by modulating various chemokines, cytokines, and growth factors. TAMs have been identified as a key contributor to resistance to chemotherapy and cancer immunotherapy. In this review article, we aim to discuss the mechanisms by which TAMs regulate innate and adaptive immune signaling in the TME and summarize recent preclinical research on the development of therapeutics targeting TAMs and tumor metabolism.
Collapse
Affiliation(s)
- Pritam Sadhukhan
- Johns Hopkins University, Skip Viragh Outpatient Cancer Building, Baltimore, MD, 21287, USA
| | - Tanguy Y Seiwert
- Johns Hopkins University, Skip Viragh Outpatient Cancer Building, Baltimore, MD, 21287, USA.
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
11
|
HIF-1α Inhibition Improves Anti-Tumor Immunity and Promotes the Efficacy of Stereotactic Ablative Radiotherapy (SABR). Cancers (Basel) 2022; 14:cancers14133273. [PMID: 35805044 PMCID: PMC9265101 DOI: 10.3390/cancers14133273] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Stereotactic ablative radiotherapy (SABR), which irradiates tumors with high-dose radiation per fraction, promotes anti-tumor immunity by stimulating various immune processes. SABR also induces vascular damage and obstructs blood flow, thereby increasing tumor hypoxia and upregulation of hypoxia-inducible factors HIF-1α and HIF-2α, master transcription factors for the cellular response to hypoxia. HIF-1α and HIF-2α are key players in the upregulation of immune suppression in hypoxia. Therefore, the radiation-induced increase in anti-tumor immunity is masked by the HIF-mediated immune suppression. Pre-clinical experiments show that inhibition of HIF-1α effectively prevents immune suppression and improves anti-tumor immunity. A combination of HIF-1α inhibitors with immunotherapy with checkpoint blocking antibodies may represent a novel approach to boost anti-tumor immunity and enhance the efficacy of SABR. Abstract High-dose hypofractionated radiation such as SABR (stereotactic ablative radiotherapy) evokes an anti-tumor immune response by promoting a series of immune-stimulating processes, including the release of tumor-specific antigens from damaged tumor cells and the final effector phase of immune-mediated lysis of target tumor cells. High-dose hypofractionated radiation also causes vascular damage in tumors, thereby increasing tumor hypoxia and upregulation of hypoxia-inducible factors HIF-1α and HIF-2α, the master transcription factors for the cellular response to hypoxia. HIF-1α and HIF-2α are critical factors in the upregulation of immune suppression and are the master regulators of immune evasion of tumors. Consequently, SABR-induced increase in anti-tumor immunity is counterbalanced by the increase in immune suppression mediated by HIFα. Inhibition of HIF-1α with small molecules such as metformin downregulates immunosuppressive pathways, including the expression of immune checkpoints, and it improves or restores the anti-tumor immunity stimulated by irradiation. Combinations of HIFα inhibitors, particularly HIF-1α inhibitors, with immune checkpoint blocking antibodies may represent a novel approach to boost the overall anti-tumor immune profile in patients and thus enhance outcomes after SABR.
Collapse
|
12
|
Jiménez-Cortegana C, Galassi C, Klapp V, Gabrilovich DI, Galluzzi L. Myeloid-Derived Suppressor Cells and Radiotherapy. Cancer Immunol Res 2022; 10:545-557. [DOI: 10.1158/2326-6066.cir-21-1105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/21/2022] [Accepted: 02/22/2022] [Indexed: 11/16/2022]
Abstract
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of pathologically activated, mostly immature, myeloid cells that exert robust immunosuppressive functions. MDSCs expand during oncogenesis and have been linked to accelerated disease progression and resistance to treatment in both preclinical tumor models and patients with cancer. Thus, MDSCs stand out as promising targets for the development of novel immunotherapeutic regimens with superior efficacy. Here, we summarize accumulating preclinical and clinical evidence indicating that MDSCs also hamper the efficacy of radiotherapy (RT), as we critically discuss the potential of MDSC-targeting strategies as tools to achieve superior immunotherapeutic tumor control by RT in the clinic.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville, Spain
| | - Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
| | - Vanessa Klapp
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
| | | | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Sandra and Edward Meyer Cancer Center, New York, New York
- Caryl and Israel Englander Institute for Precision Medicine, New York, New York
| |
Collapse
|
13
|
Yu CF, Chang CH, Wang CC, Hong JH, Chiang CS, Chen FH. Local Interleukin-12 Treatment Enhances the Efficacy of Radiation Therapy by Overcoming Radiation-Induced Immune Suppression. Int J Mol Sci 2021; 22:ijms221810053. [PMID: 34576217 PMCID: PMC8468040 DOI: 10.3390/ijms221810053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 01/31/2023] Open
Abstract
Radiation therapy (RT) recruits myeloid cells, leading to an immunosuppressive microenvironment that impedes its efficacy against tumors. Combination of immunotherapy with RT is a potential approach to reversing the immunosuppressive condition and enhancing tumor control after RT. This study aimed to assess the effects of local interleukin-12 (IL-12) therapy on improving the efficacy of RT in a murine prostate cancer model. Combined treatment effectively shrunk the radioresistant tumors by inducing a T helper-1 immune response and influx of CD8+ T cells. It also delayed the radiation-induced vascular damage accompanied by increased α-smooth muscle actin-positive pericyte coverage and blood perfusion. Moreover, RT significantly reduced the IL-12-induced levels of alanine aminotransferase in blood. However, it did not further improve the IL-12-induced anti-tumor effect on distant tumors. Upregulated expression of T-cell exhaustion-associated genes was found in tumors treated with IL-12 only and combined treatment, suggesting that T-cell exhaustion is potentially correlated with tumor relapse in combined treatment. In conclusion, this study illustrated that combination of radiation and local IL-12 therapy enhanced the host immune response and promoted vascular maturation and function. Furthermore, combination treatment was associated with less systemic toxicity than IL-12 alone, providing a potential option for tumor therapy in clinical settings.
Collapse
Affiliation(s)
- Ching-Fang Yu
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital Linkou Branch, Chang Gung University, Taoyuan 33302, Taiwan; (C.-F.Y.); (C.-C.W.); (J.-H.H.)
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan 33382, Taiwan
| | - Chun-Hsiang Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Chun-Chieh Wang
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital Linkou Branch, Chang Gung University, Taoyuan 33302, Taiwan; (C.-F.Y.); (C.-C.W.); (J.-H.H.)
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan 33382, Taiwan
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ji-Hong Hong
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital Linkou Branch, Chang Gung University, Taoyuan 33302, Taiwan; (C.-F.Y.); (C.-C.W.); (J.-H.H.)
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan 33382, Taiwan
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan;
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu 30013, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence: (C.-S.C.); (F.-H.C.); Tel.: +886-3-5733168 (C.-S.C.); +886-3-2118800 (ext. 3838) (F.-H.C.)
| | - Fang-Hsin Chen
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital Linkou Branch, Chang Gung University, Taoyuan 33302, Taiwan; (C.-F.Y.); (C.-C.W.); (J.-H.H.)
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan 33382, Taiwan
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 33302, Taiwan
- Correspondence: (C.-S.C.); (F.-H.C.); Tel.: +886-3-5733168 (C.-S.C.); +886-3-2118800 (ext. 3838) (F.-H.C.)
| |
Collapse
|
14
|
Lee YH, Yu CF, Yang YC, Hong JH, Chiang CS. Ablative Radiotherapy Reprograms the Tumor Microenvironment of a Pancreatic Tumor in Favoring the Immune Checkpoint Blockade Therapy. Int J Mol Sci 2021; 22:2091. [PMID: 33669885 PMCID: PMC7923299 DOI: 10.3390/ijms22042091] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 12/25/2022] Open
Abstract
The low overall survival rate of patients with pancreatic cancer has driven research to seek a new therapeutic protocol. Radiotherapy (RT) is frequently an option in the neoadjuvant or palliative settings for pancreatic cancer treatment. This study explored the effect of RT protocols on the tumor microenvironment (TME) and their consequent impact on anti-programmed cell death ligand-1 (PD-L1) therapy. Using a murine orthotopic pancreatic tumor model, UN-KC-6141, RT-disturbed TME was examined by immunohistochemical staining. The results showed that ablative RT is more effective than fractionated RT at recruiting T cells. On the other hand, fractionated RT induces more myeloid-derived suppressor cell infiltration than ablative RT. The RT-disturbed TME presents a higher perfusion rate per vessel. The increase in vessel perfusion is associated with a higher amount of anti-PD-L1 antibody being delivered to the tumor. Animal survival is increased by anti-PD-L1 therapy after ablative RT, with 67% of treated animals surviving more than 30 days after tumor inoculation compared to a median survival time of 16.5 days for the control group. Splenocytes isolated from surviving animals were specifically cytotoxic for UN-KC-6141 cells. We conclude that the ablative RT-induced TME is more suited than conventional RT-induced TME to combination therapy with immune checkpoint blockade.
Collapse
Affiliation(s)
- Yu-Hung Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Ching-Fang Yu
- Radiation Biology Research Center, Institute for Radiologic Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan 333323, Taiwan; (C.-F.Y.); (J.-H.H.)
| | - Ying-Chieh Yang
- Radiation Oncology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu City 300195, Taiwan;
| | - Ji-Hong Hong
- Radiation Biology Research Center, Institute for Radiologic Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan 333323, Taiwan; (C.-F.Y.); (J.-H.H.)
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan 333423, Taiwan
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 333323, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan;
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu 30013 Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|