1
|
Mortaja M, Adams SR, McKay RR, Gutkind JS, Advani SJ. Spatially precise chemo-radio-immunotherapy by antibody drug conjugate directed tumor radiosensitization to potentiate immunotherapies. NPJ Precis Oncol 2025; 9:97. [PMID: 40181161 PMCID: PMC11968929 DOI: 10.1038/s41698-025-00885-x] [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: 10/01/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Concurrent chemo-radiotherapy is standard of care for locally advanced cancer patients. While radiotherapy and immuno-oncology have advanced precision oncology, chemotherapies in the chemo-radiotherapy paradigm remain non-targeted cytotoxins. Antibody drug conjugates offer an opportunity for targeted radiosensitization that stimulates immune responses while protecting normal tissues. Here, we discuss the rationale for combining antibody drug conjugates, radiotherapy and immunotherapies and opportunities for clinical translation to advance towards targeted chemo-radio-immunotherapy precision cancer care.
Collapse
Affiliation(s)
- Mahsa Mortaja
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Stephen R Adams
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rana R McKay
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- UC San Diego, Moores Cancer Center, La Jolla, CA, 92093, USA
- Department of Urology, University of California San Diego, La Jolla, CA, 92093, USA
| | - J Silvio Gutkind
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
- UC San Diego, Moores Cancer Center, La Jolla, CA, 92093, USA
| | - Sunil J Advani
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, 92093, USA.
- UC San Diego, Moores Cancer Center, La Jolla, CA, 92093, USA.
| |
Collapse
|
2
|
Bedi D, Hassan M, Yirsaw A, Vikas B, Datta P, Samuel T. The immunopeptidome of colon cancer cells treated with topoisomerase inhibiting drug reveals differential as well as common endogenous protein sampling and display of MHC I-associated peptides. Mol Cell Oncol 2025; 12:2471640. [PMID: 40051755 PMCID: PMC11881837 DOI: 10.1080/23723556.2025.2471640] [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: 01/16/2024] [Revised: 08/05/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025]
Abstract
Immunotherapy options for microsatellite stable (MSS) colorectal cancer are currently very limited. The lack of detectably unique or altered immunogens in the tumor microenvironment may be a factor. Radiation and chemotherapy may enhance immunotherapy by increasing cancer cell visibility through Major Histocompatibility Complex I (MHC I) expression. To investigate this, we treated MSS and microsatellite-instable (MSI) colon cancer cells with a topoisomerase inhibitor and analyzed MHC I-associated peptides. Treatment increased peptide numbers by 5% in RKO (MSI) cells and 83% in SW620 (MSS) cells, with 40-50% of peptides being exclusive to treatment. Additionally, clustering analysis revealed a set of peptides with uniquely conserved residues displayed only in treated MSS SW620 cells. Gene Ontology analysis of MHC I-displayed proteins revealed a treatment-induced increase in extracellular vesicle- and nuclear-derived proteins, alongside reduced cytosolic protein sampling. Overall, we present evidence for treatment-inducible differential display of peptides, some of which may affect interactions and functions of immune cells. Given the multitude of factors that modulate the effects of increased MHC I expression and associated peptides, further studies are needed to elucidate the pathophysiological implications of these changes.
Collapse
Affiliation(s)
- Deepa Bedi
- Departments of Pathobiology and Biomedical Sciences, Tuskegee University, College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL, USA
| | - Mohammed Hassan
- Departments of Pathobiology and Biomedical Sciences, Tuskegee University, College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL, USA
| | - Alehegne Yirsaw
- Departments of Pathobiology and Biomedical Sciences, Tuskegee University, College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL, USA
| | - Biba Vikas
- Departments of Pathobiology and Biomedical Sciences, Tuskegee University, College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL, USA
| | - Pran Datta
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Temesgen Samuel
- Departments of Pathobiology and Biomedical Sciences, Tuskegee University, College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL, USA
| |
Collapse
|
3
|
Ampolini EA, Jimenez-Sainz J, Long DT. The Development of ATM Inhibitors in Cancer Therapy. Target Oncol 2025; 20:281-297. [PMID: 40024979 PMCID: PMC11933189 DOI: 10.1007/s11523-025-01136-6] [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: 02/18/2025] [Indexed: 03/04/2025]
Abstract
The ataxia-telangiectasia mutated (ATM) protein kinase plays a critical role in activating the cellular response to DNA double-strand breaks and promoting homology-directed repair. ATM is frequently mutated in cancer, contributing to an accumulation of DNA damage that drives genomic instability. To exploit cancer cells' inherent vulnerability to DNA damage, various small molecule inhibitors have been developed that target ATM. ATM inhibitors have shown great versatility in preclinical studies and increasing use in the clinic. Here, we review the development of ATM inhibitors and their role in cancer therapy. We describe their limitations and the advances that have led to increases in both the number and diversity of active clinical trials targeting ATM. We also discuss ATM's role in personalized medicine and the current challenges to more widespread use of ATM inhibitors in the clinic.
Collapse
Affiliation(s)
- Elizabeth A Ampolini
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Judit Jimenez-Sainz
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - David T Long
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
4
|
Jiang Q, Chen Z, Jiang J, Chen Q, Lan H, Zhu J, Mao W. The role of cGAS-STING in remodeling the tumor immune microenvironment induced by radiotherapy. Crit Rev Oncol Hematol 2025; 209:104658. [PMID: 39956501 DOI: 10.1016/j.critrevonc.2025.104658] [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: 11/04/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025] Open
Abstract
The activation of the cGAS-STING pathway occurs when tumor cell DNA is damaged by ionizing radiation. Once triggered, this pathway reshapes the tumor immune microenvironment by promoting the maturation, activation, polarization, and immune-killing capacity of immune cells, as well as by inducing the release of interferons and the expression of immune-related genes. In addition, the gut microbiota and various mechanisms of programmed cell death interact with the cGAS-STING pathway, further influencing its function in remodeling the immune microenvironment after radiotherapy. Therefore, investigating the mechanisms of the cGAS-STING pathway in reshaping the tumor immune microenvironment post-radiotherapy can not only optimize the efficacy of combined radiotherapy and immunotherapy but also provide new research directions and potential targets for cancer treatment.
Collapse
Affiliation(s)
- Qingyu Jiang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Zhejiang Chinese Medical University, Hangzhou 310053, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Zhiheng Chen
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing 31400, China
| | - Jin Jiang
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing 31400, China
| | - Qianping Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Huiyin Lan
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Ji Zhu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China.
| | - Wei Mao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China.
| |
Collapse
|
5
|
Rødland GE, Temelie M, Eek Mariampillai A, Serban AM, Edin NFJ, Malinen E, Lindbergsengen L, Gilbert A, Chevalier F, Savu DI, Syljuåsen RG. Interferon signaling is enhanced by ATR inhibition in glioblastoma cells irradiated with X-rays, protons or carbon ions. Radiother Oncol 2025; 203:110669. [PMID: 39675575 DOI: 10.1016/j.radonc.2024.110669] [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: 04/08/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND AND PURPOSE Interferon (IFN) signaling plays an important role in antitumor immune responses. Inhibitors of the DNA damage response, such as ATR inhibitors, can increase IFN signaling upon conventional radiotherapy with X-rays. However, it is not known whether such inhibitors also enhance IFN signaling after irradiation with high linear energy transfer (LET) particles. MATERIALS AND METHODS Human glioblastoma U-251 and T98G cells were irradiated with X-rays, protons (LET: 4.8 and 41.9 keV/µm) and carbon ions (LET: 28 and 73 keV/µm), with and without ATR inhibitor (VE-822) or ATM inhibitor (AZD1390). DNA damage signaling and cell cycle distribution were analyzed by immunoblotting and flow cytometry, and radiosensitivity was assessed by clonogenic survival assay. IFN-β secretion was measured by ELISA, and STAT1 activation was examined by immunoblotting. RESULTS High-LET protons and carbon ions caused stronger activation of the DNA damage response compared to low-LET protons and X-rays at similar radiation doses. G2 checkpoint arrest was abrogated by the ATR inhibitor and prolonged by the ATM inhibitor after all radiation types. The inhibitors increased radiosensitivity, as measured after X- and carbon ion irradiation. ATR inhibition increased IFN signaling following both low-LET and high-LET irradiation. ATM inhibition also increased IFN signaling, but to a lesser extent. Notably, both cell lines secreted significantly more IFN-β when the inhibitors were combined with high-LET compared to low-LET irradiation. CONCLUSION These findings indicate that DNA damage response inhibitors can enhance IFN signaling following X-, proton and carbon ion irradiation, with a strong positive dependency on LET.
Collapse
Affiliation(s)
- Gro Elise Rødland
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Mihaela Temelie
- Department of Life and Environmental Physics, Horia Hulubei National Institute for R&D in Physics and Nuclear Engineering, 077125 Magurele, Romania
| | - Adrian Eek Mariampillai
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Ana Maria Serban
- Department of Life and Environmental Physics, Horia Hulubei National Institute for R&D in Physics and Nuclear Engineering, 077125 Magurele, Romania
| | | | - Eirik Malinen
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; Section for biophysics and medical physics, Department of Physics, University of Oslo, 0371 Oslo, Norway
| | - Lilian Lindbergsengen
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Antoine Gilbert
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France
| | - François Chevalier
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France
| | - Diana I Savu
- Department of Life and Environmental Physics, Horia Hulubei National Institute for R&D in Physics and Nuclear Engineering, 077125 Magurele, Romania.
| | - Randi G Syljuåsen
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway.
| |
Collapse
|
6
|
Ji K, Jia H, Liu Z, Yu G, Wen R, Zhang T, Peng Z, Man W, Tian Y, Wang C, Ling Q, Zhang W, Zhou L, Liu M, Zhu B. New insight in immunotherapy and combine therapy in colorectal cancer. Front Cell Dev Biol 2025; 12:1453630. [PMID: 39839672 PMCID: PMC11747282 DOI: 10.3389/fcell.2024.1453630] [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: 06/24/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025] Open
Abstract
The advent of immune checkpoint inhibitors (ICIs) in colorectal cancer (CRC) treatment marks a major breakthrough. These therapies have proven safer and more effective than traditional radiotherapy and targeted treatments. Immunotherapies like pembrolizumab, nivolumab, and ipilimumab have pioneered new treatment avenues, potentially improving patient outcomes and quality of life. Additionally, advances in immunotherapy have prompted detailed research into CRC therapies, especially those integrating ICIs with conventional treatments, providing new hope for patients and shaping future research and practice. This review delves into the mechanisms of various ICIs and evaluates their therapeutic potential when combined with radiotherapy, chemotherapy, and targeted therapies in clinical settings. It also sheds light on the current application and research involving ICIs in CRC treatment.
Collapse
Affiliation(s)
- Kai Ji
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hang Jia
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zixuan Liu
- 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
| | - Rongbo Wen
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tianshuai Zhang
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhiying Peng
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Wenjiang Man
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yucheng Tian
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Can Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qianlong Ling
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Wei Zhang
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Leqi Zhou
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Mulin Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Bing Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
7
|
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
|
8
|
Mahin J, Xu X, Li L, Zhang C. cGAS/STING in skin melanoma: from molecular mechanisms to therapeutics. Cell Commun Signal 2024; 22:553. [PMID: 39558334 PMCID: PMC11571982 DOI: 10.1186/s12964-024-01860-y] [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: 08/07/2024] [Accepted: 09/29/2024] [Indexed: 11/20/2024] Open
Abstract
Melanoma, recognized as the most aggressive type of skin cancer, has experienced a notable increase in cases, especially within populations with fair skin. This highly aggressive cancer is largely driven by UV radiation exposure, resulting in the uncontrolled growth and malignant transformation of melanocytes. The cGAS-STING pathway, an immune signaling mechanism responsible for detecting double-stranded DNA in the cytoplasm, is essential for mediating the immune response against melanoma. This pathway serves a dual purpose: it enhances antitumor immunity by activating immune cells, but it can also promote tumor growth when chronically activated by creating an immunosuppressive environment. This review comprehensively examines the multifaceted implication of the cGAS-STING pathway in melanoma pathogenesis and treatment. We explore its molecular mechanisms, including epigenetic regulation, interaction with signaling pathways such as AR signaling, and modulation by various cellular effectors like TG2 and activin-A. The therapeutic potential of modulating the cGAS-STING pathway is highlighted, with promising results from STING agonists, combination therapies with immune checkpoint inhibitors, and novel drug delivery systems, including nanoparticles and synthetic drugs. Our findings underscore the importance of the cGAS-STING pathway in melanoma, presenting it as a critical target for enhancing anti-tumor immunity. By leveraging this pathway, future therapeutic strategies can potentially convert 'cold' tumors into 'hot' tumors, making them more susceptible to immune responses.
Collapse
Affiliation(s)
- Jafaridarabjerdi Mahin
- Department of Dermatology, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Xuezhu Xu
- Department of Dermatology, The Second Hospital of Dalian Medical University, Dalian, 116023, China.
| | - Ling Li
- Department of Dermatology, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Cong Zhang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, 116044, China
| |
Collapse
|
9
|
Du S, Liang Q, Shi J. Progress of ATM inhibitors: Opportunities and challenges. Eur J Med Chem 2024; 277:116781. [PMID: 39173286 DOI: 10.1016/j.ejmech.2024.116781] [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: 06/22/2024] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
Ataxia-telangiectasia mutated (ATM) was first discovered in patients with AT (ataxia telangiectasia), which is characteristic with cerebellar degeneration, immunodeficiency, being susceptible to malignant tumors and sensitive to radiation. ATM kinase could detect DNA double-strand breaks and play a vital role in the DNA damage response. Inhibiting the function of ATM could sensitize tumor cells to both ionizing radiation (IR) and chemotherapy, as well as improve the chemoresistance and radioresistance observed in some patients. As such, ATM is a novel and important target for the cancer therapy. We reviewed ATM inhibitors reported in the last two decades, focusing on their development process, structure-activity relationships, inhibitory efficacy, pharmacokinetics and pharmacodynamics characteristics in the preclinical and clinical studies. We summarized the clinical value of ATM inhibitors in tumors and some neurodegenerative diseases, as well as the main challenges to the development of the drugs, providing directions and references for the future development of ATM inhibitors.
Collapse
Affiliation(s)
- Shan Du
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Qi Liang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
10
|
Qian C, Li X, Zhang J, Wang Y. Small Molecular Inhibitors That Target ATM for Drug Discovery: Current Research and Potential Prospective. J Med Chem 2024; 67:14742-14767. [PMID: 39149790 DOI: 10.1021/acs.jmedchem.4c01064] [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: 08/17/2024]
Abstract
The protein kinase ataxia telangiectasia mutated (ATM) is a constituent of the phosphatidylinositol 3-kinase-related kinase (PIKK) family, exerting a pivotal influence on diverse cellular processes, notably the signaling of double-strand DNA breaks (DSB) and stress response. The dysregulation of ATM is implicated in the pathogenesis of cancer and other diseases such as neurodegeneration. Hence, ATM is deemed a promising candidate for potential therapeutic interventions across a spectrum of diseases. Presently, while ATM small molecule inhibitors are not commercially available, various selective inhibitors have progressed to the clinical research phase. Specifically, AZD1390, WSD0628, SYH2051, and ZN-B-2262 are under investigation in clinical studies pertaining to glioblastoma multiforme and advanced solid tumors, respectively. In this Perspective, we encapsulate the structure, biological functions, and disease relevance of ATM. Subsequently, we concentrate on the design concepts and structure-activity relationships (SAR) of ATM inhibitors, delineating potential avenues for the development of more efficacious ATM-targeted inhibitors.
Collapse
Affiliation(s)
- Chunlin Qian
- Department of Respiratory and Critical Care Medicine and Targeted Tracer Research and Development Laboratory and Institute of Respiratory Health and State Key Laboratory of Respiratory Health and Multimorbidity and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
| | - Xiaoxue Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
| | - Jifa Zhang
- Department of Respiratory and Critical Care Medicine and Targeted Tracer Research and Development Laboratory and Institute of Respiratory Health and State Key Laboratory of Respiratory Health and Multimorbidity and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine and Targeted Tracer Research and Development Laboratory and Institute of Respiratory Health and State Key Laboratory of Respiratory Health and Multimorbidity and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan China
| |
Collapse
|
11
|
Majumder B, Nataraj NB, Maitreyi L, Datta S. Mismatch repair-proficient tumor footprints in the sands of immune desert: mechanistic constraints and precision platforms. Front Immunol 2024; 15:1414376. [PMID: 39100682 PMCID: PMC11294168 DOI: 10.3389/fimmu.2024.1414376] [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: 04/08/2024] [Accepted: 06/17/2024] [Indexed: 08/06/2024] Open
Abstract
Mismatch repair proficient (MMRp) tumors of colorectal origin are one of the prevalent yet unpredictable clinical challenges. Despite earnest efforts, optimal treatment modalities have yet to emerge for this class. The poor prognosis and limited actionability of MMRp are ascribed to a low neoantigen burden and a desert-like microenvironment. This review focuses on the critical roadblocks orchestrated by an immune evasive mechanistic milieu in the context of MMRp. The low density of effector immune cells, their weak spatiotemporal underpinnings, and the high-handedness of the IL-17-TGF-β signaling are intertwined and present formidable challenges for the existing therapies. Microbiome niche decorated by Fusobacterium nucleatum alters the metabolic program to maintain an immunosuppressive state. We also highlight the evolving strategies to repolarize and reinvigorate this microenvironment. Reconstruction of anti-tumor chemokine signaling, rational drug combinations eliciting T cell activation, and reprograming the maladapted microbiome are exciting developments in this direction. Alternative vulnerability of other DNA damage repair pathways is gaining momentum. Integration of liquid biopsy and ex vivo functional platforms provide precision oncology insights. We illustrated the perspectives and changing landscape of MMRp-CRC. The emerging opportunities discussed in this review can turn the tide in favor of fighting the treatment dilemma for this elusive cancer.
Collapse
|
12
|
Kerr CP, Sheehan-Klenk J, Grudzinski JJ, Adam DP, Nguyen TPT, Ferreira CA, Bates AM, Jin WJ, Kwon O, Olson AP, Lin W, Hyun M, Jagodinsky JC, Powers M, Sriramaneni RN, Clark PA, Shea AG, Rojas HC, Choi C, Massey CF, Zangl LM, Pinchuk AN, Aluicio-Sarduy E, Kim K, Engle JW, Hernandez R, Bednarz BP, Weichert JP, Morris ZS. Effects of clinically relevant radionuclides on the activation of a type I interferon response by radiopharmaceuticals in syngeneic murine tumor models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602990. [PMID: 39071353 PMCID: PMC11275738 DOI: 10.1101/2024.07.10.602990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Radiopharmaceutical therapies (RPT) activate a type I interferon (IFN1) response in tumor cells. We hypothesized that the timing and amplitude of this response varies by isotope. We compared equal doses delivered by 90 Y, 177 Lu, and 225 Ac in vitro as unbound radionuclides and in vivo when chelated to NM600, a tumor-selective alkylphosphocholine. Response in murine MOC2 head and neck carcinoma and B78 melanoma was evaluated by qPCR and flow cytometry. Therapeutic response to 225 Ac-NM600+anti-CTLA4+anti-PD-L1 immune checkpoint inhibition (ICI) was evaluated in wild-type and stimulator of interferon genes knockout (STING KO) B78. The timing and magnitude of IFN1 response correlated with radionuclide half-life and linear energy transfer. CD8 + /Treg ratios increased in tumors 7 days after 90 Y- and 177 Lu-NM600 and day 21 after 225 Ac-NM600. 225 Ac-NM600+ICI improved survival in mice with WT but not with STING KO tumors, relative to monotherapies. Immunomodulatory effects of RPT vary with radioisotope and promote STING-dependent enhanced response to ICIs in murine models. Teaser This study describes the time course and nature of tumor immunomodulation by radiopharmaceuticals with differing physical properties.
Collapse
|
13
|
Lu C, Gao Z, Wu D, Zheng J, Hu C, Huang D, He C, Liu Y, Lin C, Peng T, Dou Y, Zhang Y, Sun F, Jiang W, Yin G, Han R, He Y. Understanding the dynamics of TKI-induced changes in the tumor immune microenvironment for improved therapeutic effect. J Immunother Cancer 2024; 12:e009165. [PMID: 38908857 PMCID: PMC11328648 DOI: 10.1136/jitc-2024-009165] [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: 06/08/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND The dynamic interplay between tyrosine kinase inhibitors (TKIs) and the tumor immune microenvironment (TME) plays a crucial role in the therapeutic trajectory of non-small cell lung cancer (NSCLC). Understanding the functional dynamics and resistance mechanisms of TKIs is essential for advancing the treatment of NSCLC. METHODS This study assessed the effects of short-term and long-term TKI treatments on the TME in NSCLC, particularly targeting epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) mutations. We analyzed changes in immune cell composition, cytokine profiles, and key proteins involved in immune evasion, such as laminin subunit γ-2 (LAMC2). We also explored the use of aspirin as an adjunct therapy to modulate the TME and counteract TKI resistance. RESULTS Short-term TKI treatment enhanced T cell-mediated tumor clearance, reduced immunosuppressive M2 macrophage infiltration, and downregulated LAMC2 expression. Conversely, long-term TKI treatment fostered an immunosuppressive TME, contributing to drug resistance and promoting immune escape. Differential responses were observed among various oncogenic mutations, with ALK-targeted therapies eliciting a stronger antitumor immune response compared with EGFR-targeted therapies. Notably, we found that aspirin has potential in overcoming TKI resistance by modulating the TME and enhancing T cell-mediated tumor clearance. CONCLUSIONS These findings offer new insights into the dynamics of TKI-induced changes in the TME, improving our understanding of NSCLC challenges. The study underscores the critical role of the TME in TKI resistance and suggests that adjunct therapies, like aspirin, may provide new strategies to enhance TKI efficacy and overcome resistance.
Collapse
Affiliation(s)
- Conghua Lu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Ziyuan Gao
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Di Wu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Jie Zheng
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Chen Hu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Daijuan Huang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Chao He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Yihui Liu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Caiyu Lin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Tao Peng
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Yuanyao Dou
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Yimin Zhang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Fenfen Sun
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Weiling Jiang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Guoqing Yin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Rui Han
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
- School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
14
|
Colangelo NW, Gerber NK, Vatner RE, Cooper BT. Harnessing the cGAS-STING pathway to potentiate radiation therapy: current approaches and future directions. Front Pharmacol 2024; 15:1383000. [PMID: 38659582 PMCID: PMC11039815 DOI: 10.3389/fphar.2024.1383000] [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: 02/06/2024] [Accepted: 03/15/2024] [Indexed: 04/26/2024] Open
Abstract
In this review, we cover the current understanding of how radiation therapy, which uses ionizing radiation to kill cancer cells, mediates an anti-tumor immune response through the cGAS-STING pathway, and how STING agonists might potentiate this. We examine how cGAS-STING signaling mediates the release of inflammatory cytokines in response to nuclear and mitochondrial DNA entering the cytoplasm. The significance of this in the context of cancer is explored, such as in response to cell-damaging therapies and genomic instability. The contribution of the immune and non-immune cells in the tumor microenvironment is considered. This review also discusses the burgeoning understanding of STING signaling that is independent of inflammatory cytokine release and the various mechanisms by which cancer cells can evade STING signaling. We review the available data on how ionizing radiation stimulates cGAS-STING signaling as well as how STING agonists may potentiate the anti-tumor immune response induced by ionizing radiation. There is also discussion of how novel radiation modalities may affect cGAS-STING signaling. We conclude with a discussion of ongoing and planned clinical trials combining radiation therapy with STING agonists, and provide insights to consider when planning future clinical trials combining these treatments.
Collapse
Affiliation(s)
- Nicholas W. Colangelo
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, NY, United States
| | - Naamit K. Gerber
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, NY, United States
| | - Ralph E. Vatner
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Benjamin T. Cooper
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
15
|
Koukourakis IM, Xanthopoulou E, Koukourakis MI, Tiniakos D, Kouloulias V, Zygogianni A. IFN-Type-I Response and Systemic Immunity in Rectal Adenocarcinoma Patients Treated with Conventional or Hypofractionated Neoadjuvant Radiotherapy. Biomolecules 2024; 14:448. [PMID: 38672465 PMCID: PMC11048635 DOI: 10.3390/biom14040448] [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: 02/28/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
The IFN-type-I pathway is involved in radiotherapy (RT)-mediated immune responses. Large RT fractions have been suggested to potently induce this pathway. Neoadjuvant hypofractionated short-course (scRT) and conventional long-course (lcRT) RT applied for the treatment of locally advanced rectal adenocarcinoma patients provides a unique model to address the immuno-stimulatory properties of RT on a systemic level. We prospectively analyzed the IFNβ plasma levels and lymphocyte counts (LCs) of rectal adenocarcinoma patients before and after treatment with scRT (n = 22) and lcRT (n = 40). Flow cytometry was conducted to assess the effects on lymphocytic subpopulations in a subset of 20 patients. A statistically significant increase in the post-RT IFNβ plasma levels was noted in patients undergoing scRT (p = 0.004). Improved pathological tumor regression was associated with elevated post-RT IFNβ levels (p = 0.003). Although all patients experienced substantial lymphopenia after treatment, the post-RT LC of patients treated with scRT were significantly higher compared to lcRT (p = 0.001). Patients undergoing scRT displayed significantly lower percentages of regulatory CD4+/CD25+ T-cells after therapy (p = 0.02). scRT enables effective stimulation of the IFN-type-I pathway on a systemic level and confers decreased lymphocytic cytotoxicity and limited regulatory T-cell activation compared to lcRT, supporting its increasing role in immuno-RT trials.
Collapse
Affiliation(s)
- Ioannis M. Koukourakis
- Radiation Oncology Unit, 1st Department of Radiology, School of Medicine, Aretaieion Hospital, National and Kapodistrian University of Athens (NKUOA), 11528 Athens, Greece;
| | - Erasmia Xanthopoulou
- Department of Radiotherapy/Oncology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.X.); (M.I.K.)
| | - Michael I. Koukourakis
- Department of Radiotherapy/Oncology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.X.); (M.I.K.)
| | - Dina Tiniakos
- Department of Pathology, Aretaieion University Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Vassilis Kouloulias
- Radiotherapy Unit, 2nd Department of Radiology, Attikon Hospital, School of Medicine, Rimini 1, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Anna Zygogianni
- Radiation Oncology Unit, 1st Department of Radiology, School of Medicine, Aretaieion Hospital, National and Kapodistrian University of Athens (NKUOA), 11528 Athens, Greece;
| |
Collapse
|
16
|
Shea AG, Idrissou MB, Torres AI, Chen T, Hernandez R, Morris ZS, Sodji QH. Immunological effects of radiopharmaceutical therapy. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1331364. [PMID: 39355211 PMCID: PMC11440989 DOI: 10.3389/fnume.2024.1331364] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/14/2024] [Indexed: 10/03/2024]
Abstract
Radiation therapy (RT) is a pillar of cancer therapy used by more than half of all cancer patients. Clinically, RT is mostly delivered as external beam radiation therapy (EBRT). However, the scope of EBRT is limited in the metastatic setting, where all sites of disease need to be irradiated. Such a limitation is attributed to radiation-induced toxicities, for example on bone marrow and hematologic toxicities, resulting from a large EBRT field. Radiopharmaceutical therapy (RPT) has emerged as an alternative to EBRT for the irradiation of all sites of metastatic disease. While RPT can reduce tumor burden, it can also impact the immune system and anti-tumor immunity. Understanding these effects is crucial for predicting and managing treatment-related hematological toxicities and optimizing their integration with other therapeutic modalities, such as immunotherapies. Here, we review the immunomodulatory effects of α- and β-particle emitter-based RPT on various immune cell lines, such as CD8+ and CD4+ T cells, natural killer (NK) cells, and regulatory T (Treg) cells. We briefly discuss Auger electron-emitter (AEE)-based RPT, and finally, we highlight the combination of RPT with immune checkpoint inhibitors, which may offer potential therapeutic synergies for patients with metastatic cancers.
Collapse
Affiliation(s)
- Amanda G. Shea
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Malick Bio Idrissou
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Ana Isabel Torres
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Tessa Chen
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Reiner Hernandez
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Quaovi H. Sodji
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
17
|
Wang X, Wang Y, Zhang Y, Shi H, Liu K, Wang F, Wang Y, Chen H, Shi Y, Wang R. Immune modulatory roles of radioimmunotherapy: biological principles and clinical prospects. Front Immunol 2024; 15:1357101. [PMID: 38449871 PMCID: PMC10915027 DOI: 10.3389/fimmu.2024.1357101] [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: 12/17/2023] [Accepted: 01/31/2024] [Indexed: 03/08/2024] Open
Abstract
Radiation therapy (RT) not only can directly kill tumor cells by causing DNA double-strand break, but also exerts anti-tumor effects through modulating local and systemic immune responses. The immunomodulatory effects of RT are generally considered as a double-edged sword. On the one hand, RT effectively enhances the immunogenicity of tumor cells, triggers type I interferon response, induces immunogenic cell death to activate immune cell function, increases the release of proinflammatory factors, and reshapes the tumor immune microenvironment, thereby positively promoting anti-tumor immune responses. On the other hand, RT stimulates tumor cells to express immunosuppressive cytokines, upregulates the function of inhibitory immune cells, leads to lymphocytopenia and depletion of immune effector cells, and thus negatively suppresses immune responses. Nonetheless, it is notable that RT has promising abscopal effects and may achieve potent synergistic effects, especially when combined with immunotherapy in the daily clinical practice. This systematic review will provide a comprehensive profile of the latest research progress with respect to the immunomodulatory effects of RT, as well as the abscopal effect of radioimmunotherapy combinations, from the perspective of biological basis and clinical practice.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yu 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
| | - Yonggang Zhang
- Department of Head and Neck Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Hongyun Shi
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Kuan Liu
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Fang Wang
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yue Wang
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Huijing Chen
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yan Shi
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Ruiyao Wang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| |
Collapse
|