1
|
Deng X, Xiang K, He X, Chen S, Guo Q, Wu H, Liu X, Wen Q, Yang H. Good response of stage IV melanoma to high‑dose radiation therapy combined with immunotherapy: A case report. Oncol Lett 2024; 28:598. [PMID: 39493434 PMCID: PMC11529377 DOI: 10.3892/ol.2024.14731] [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: 01/15/2024] [Accepted: 08/22/2024] [Indexed: 11/05/2024] Open
Abstract
Patients with advanced malignant melanoma (MM) often do not receive satisfactory treatment. The present study reports the case of a 51-year-old female patient with stage IV MM of unknown primary. After undergoing immune checkpoint inhibitor therapy, the patient received multiple doses of hypofractionated radiotherapy (HFRT) for the left inguinal lymph node and single-fraction high-dose-rate brachytherapy for the left and right lung metastases. After combination treatment, the patient experienced almost complete remission of the inguinal target area, significant relief of pain and discomfort and an improved quality of life. The time of lung radiotherapy lesion control was 8 months. Meanwhile, the observed lesions (observation lesions 1, 2, 3 and 5) adjacent to the target lesion received lower doses of scattering (0.9-1.8 Gy) and the time of control for these lung observation lesions was 9 months. In addition, restarting targeted therapy after cessation of other treatments due to myelosuppression resulted in a progression-free survival time of 6 months. Nevertheless, the patient developed new metastases in the brain and abdomen. The present case report demonstrates that high-dose radiotherapy combined with immunotherapy may be effective for local lesions and that multiple doses of HFRT may be superior to single-fraction high-dose-rate brachytherapy for certain patients. Low-dose scattering also shows improvement for local lesions. Furthermore, restarting targeted therapy may be effective in the presence of target sites. Thus, the present case report provides a possible therapeutic option for the treatment of advanced melanoma.
Collapse
Affiliation(s)
- Xuemei Deng
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Kewei Xiang
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xingting He
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Shuang Chen
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Qingxi Guo
- Department Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Hong Wu
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xiaolong Liu
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Qinglian Wen
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Hongru Yang
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
2
|
Trautmann T, Yakobian N, Nguyen R. CAR T-cells for pediatric solid tumors: where to go from here? Cancer Metastasis Rev 2024; 43:1445-1461. [PMID: 39317919 DOI: 10.1007/s10555-024-10214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Despite the great success that chimeric antigen receptor (CAR) T-cells have had in patients with B-cell malignancies and multiple myeloma, they continue to have limited efficacy against most solid tumors. Especially in the pediatric population, pre- and post-treatment biopsies are rarely performed due to ethical reasons, and thus, our understanding is still very limited regarding the mechanisms in the tumor microenvironment by which tumor cells exclude effectors and attract immune-suppressive cells. Nevertheless, based on the principles that are known, current T-cell engineering has leveraged some of these processes and created more potent CAR T-cells. The recent discovery of new oncofetal antigens and progress made in CAR design have expanded the potential pool of candidate antigens for therapeutic development. The most promising approaches to enhance CAR T-cells are novel CAR gating strategies, creative ways of cytokine delivery to the TME without enhancing systemic toxicity, and hijacking the chemokine axis of tumors for migratory purposes. With these new modifications, the next step in the era of CAR T-cell development will be the clinical validation of these promising preclinical findings.
Collapse
Affiliation(s)
- Tina Trautmann
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA
| | - Natalia Yakobian
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA
| | - Rosa Nguyen
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA.
| |
Collapse
|
3
|
Mossahebi S, Molitoris JK, Poirier Y, Jatczak J, Zhang B, Mohindra P, Ferris M, Regine WF, Yi B. Clinical Implementation and Dosimetric Evaluation of a Robust Proton Lattice Planning Strategy Using Primary and Robust Complementary Beams. Int J Radiat Oncol Biol Phys 2024; 120:1149-1158. [PMID: 38936634 DOI: 10.1016/j.ijrobp.2024.06.009] [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: 10/23/2023] [Revised: 05/17/2024] [Accepted: 06/15/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE Pencil-beam scanning proton therapy has been considered a potential modality for the 3D form of spatially fractionated radiation therapy called lattice therapy. However, few practical solutions have been introduced in the clinic. Existing limitations include degradation in plan quality and robustness when using single-field versus multifield lattice plans, respectively. We propose a practical and robust proton lattice (RPL) planning method using multifield and evaluate its dosimetric characteristics compared to clinically acceptable photon lattice plans. METHODS AND MATERIALS Seven cases previously treated with photon lattice therapy were used to evaluate a novel RPL planning technique using 2-orthogonal beams: a primary beam (PB) and a robust complementary beam (RCB) that deliver 67% and 33%, respectively, of the prescribed dose to vertices inside the gross target volume (GTV). Only RCB is robustly optimized for setup and range uncertainties. The number and volume of vertices, peak-to-valley dose ratios (PVDRs), and volume of low dose to GTV of proton and photon plans were compared. The RPL technique was then used in the treatment of 2 patients and their dosimetric parameters were reported. RESULTS The RPL strategy was able to achieve the clinical planning goals. Compared to previously treated photon plans, the average number of vertices increased by 30%, the average vertex volume by 49% (18.2 ± 25.9 cc vs 12.2 ± 14.5 cc, P = .21), and higher PVDR (10.5 ± 4.8 vs 2.5 ± 0.9, P < .005) was achieved. In addition, RPL plans show more conformal dose with less low dose to GTV (V30%, 60.9% ± 7.2% vs 81.6% ± 13.9% and V10%, 88.3% ± 4.5% vs 98.6% ± 3.6% [P < .01]). The RPL plan for 2 treated patients showed PVDRs of 4.61 and 14.85 with vertices-to-GTV ratios of 1.52% and 1.30%, respectively. CONCLUSIONS A novel RPL planning strategy using a pair of orthogonal beams was developed and successfully translated to the clinic. The proposed method can generate better quality plans, a higher number of vertices, and higher PVDRs than currently used photon lattice plans.
Collapse
Affiliation(s)
- Sina Mossahebi
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; Maryland Proton Treatment Center, Baltimore, Maryland.
| | - Jason K Molitoris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; Maryland Proton Treatment Center, Baltimore, Maryland
| | - Yannick Poirier
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jenna Jatczak
- Maryland Proton Treatment Center, Baltimore, Maryland
| | - Baoshe Zhang
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; Maryland Proton Treatment Center, Baltimore, Maryland
| | - Pranshu Mohindra
- Department of Radiation Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Matthew Ferris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; Maryland Proton Treatment Center, Baltimore, Maryland
| | - William F Regine
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; Maryland Proton Treatment Center, Baltimore, Maryland
| | - ByongYong Yi
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; Maryland Proton Treatment Center, Baltimore, Maryland
| |
Collapse
|
4
|
Traveset L, Cerdán Porqueras V, Huerga Encabo H, Avalle S, Esteve-Codina A, Fornas O, Aramburu J, Lopez-Rodriguez C. NFAT5 counters long-term IFN-1 responses in hematopoietic stem cells to preserve reconstitution potential. Blood Adv 2024; 8:5510-5526. [PMID: 39208369 PMCID: PMC11538617 DOI: 10.1182/bloodadvances.2023011306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
ABSTRACT Hematopoietic stem cells (HSCs) readily recover from acute stress, but persistent stress can reduce their viability and long-term potential. Here, we show that the nuclear factor of activated T cells 5 (NFAT5), a transcription modulator of inflammatory responses, protects the HSC pool under stress. NFAT5 restrains HSC differentiation to multipotent progenitors after bone marrow transplantation and bone marrow ablation with ionizing radiation or chemotherapy. Correspondingly, NFAT5-deficient HSCs fail to support long-term reconstitution of hematopoietic progenitors and mature blood cells after serial transplant. Evidence from competitive transplant assays shows that these defects are HSC intrinsic. NFAT5-deficient HSCs exhibit enhanced expression of type 1 interferon (IFN-1) response genes after transplant, and suppressing IFN-1 receptor prevents their exacerbated differentiation and cell death after reconstitution and improves long-term regeneration potential. Blockade of IFN-1 receptor also prevented the overdifferentiation of NFAT5-deficient HSCs after bone marrow ablation. These findings show that long-term IFN-1 responses to different hematopoietic stressors drive HSCs toward more differentiated progenitors, and that NFAT5 has an HSC-intrinsic role, limiting IFN-1 responses to preserve reconstitution potential. Our identification of cell-intrinsic mechanisms that strengthen the resistance of HSCs to stress could help to devise approaches to protect long-term stemness during the treatment of hematopoietic malignancies.
Collapse
Affiliation(s)
- Laia Traveset
- Department of Medicine and Life Sciences, Immunology Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Víctor Cerdán Porqueras
- Department of Medicine and Life Sciences, Immunology Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Hector Huerga Encabo
- Department of Medicine and Life Sciences, Immunology Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Silvia Avalle
- Department of Medicine and Life Sciences, Immunology Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Anna Esteve-Codina
- Bioinformatics unit, Centro Nacional de Análisis Genómico, Centre for Genomic Regulation, Barcelona Institute for Science and Technology, Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Oscar Fornas
- Department of Medicine and Life Sciences, Flow Cytometry Unit, Universitat Pompeu Fabra, Centre for Genomic Regulation, Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Jose Aramburu
- Department of Medicine and Life Sciences, Immunology Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Cristina Lopez-Rodriguez
- Department of Medicine and Life Sciences, Immunology Unit, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
5
|
Amodio V, Vitiello PP, Bardelli A, Germano G. DNA repair-dependent immunogenic liabilities in colorectal cancer: opportunities from errors. Br J Cancer 2024; 131:1576-1590. [PMID: 39271762 DOI: 10.1038/s41416-024-02848-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the major causes of cancer death worldwide. Chemotherapy continues to serve as the primary treatment modality, while immunotherapy is largely ineffective for the majority of CRC patients. Seminal discoveries have emphasized that modifying DNA damage response (DDR) mechanisms confers both cell-autonomous and immune-related vulnerabilities across various cancers. In CRC, approximately 15% of tumours exhibit alterations in the mismatch repair (MMR) machinery, resulting in a high number of neoantigens and the activation of the type I interferon response. These factors, in conjunction with immune checkpoint blockades, collectively stimulate anticancer immunity. Furthermore, although less frequently, somatic alterations in the homologous recombination (HR) pathway are observed in CRC; these defects lead to genome instability and telomere alterations, supporting the use of poly (ADP-ribose) polymerase (PARP) inhibitors in HR-deficient CRC patients. Additionally, other DDR inhibitors, such as Ataxia Telangiectasia and Rad3-related protein (ATR) inhibitors, have shown some efficacy both in preclinical models and in the clinical setting, irrespective of MMR proficiency. The aim of this review is to elucidate how preexisting or induced vulnerabilities in DNA repair pathways represent an opportunity to increase tumour sensitivity to immune-based therapies in CRC.
Collapse
Affiliation(s)
- V Amodio
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Department of Oncology, Molecular Biotechnology Center, University of Torino, 10126, Turin, Italy
| | - P P Vitiello
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Department of Oncology, Molecular Biotechnology Center, University of Torino, 10126, Turin, Italy
| | - A Bardelli
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
- Department of Oncology, Molecular Biotechnology Center, University of Torino, 10126, Turin, Italy.
| | - G Germano
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
- Department of Medical Biotechnologies and Translational Medicine, University of Milano, 20133, Milan, Italy.
| |
Collapse
|
6
|
He J, Ren X, Zhang Q, Wang S, Li Z, Cai K, Li M, Hu Y, Ran Q, Luo Z. Nanoradiosentizers with X ray-actuatable supramolecular aptamer building units for programmable immunostimulatory T cell engagement. Biomaterials 2024; 315:122924. [PMID: 39489019 DOI: 10.1016/j.biomaterials.2024.122924] [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: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
The insufficient activation and impaired effector functions of T cells in the immunosuppressive tumor microenvironment (TME) substantially reduces the immunostimulatory effects of radiotherapy. Herein, a multifunctional nanoradiosensitizer is established by integrating molecularly engineered aptamer precursors into cisplatin-loaded liposomes for enhancing radio-immunotherapy of solid tumors. Exposure to ionizing radiation (IR) following the nanoradiosensitizer treatment would induce pronounced immunogenic death (ICD) of tumor cells through cisplatin-mediated radiosensitization while also trigger the detachment of the aptamer precursors, which further self-assemble into PD-L1/PD-1-bispecific aptamer-based T cell engagers (CA) through the bridging effect of tumor-derived ATP to direct T cell binding onto tumor cells in the post-IR TME in a spatial-temporally programmable manner. The CA-mediated post-IR tumor-T cell engagement could override the immunosuppressive barriers in TME and enhance T cell-mediated recognition and elimination of tumor cells while minimizing systemic toxicities. Overall, this work offers an innovative approach to enhance the radio-immunotherapeutic efficacy in the clinics.
Collapse
Affiliation(s)
- Jinming He
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Xijiao Ren
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Qiqi Zhang
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Shuang Wang
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Zhongjun Li
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Qian Ran
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China.
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
7
|
Zhang Y, Hong W, Zheng D, Li Z, Hu Y, Chen Y, Yang P, Zeng Z, Du S. Increased IFN-β indicates better survival in hepatocellular carcinoma treated with radiotherapy. Clin Exp Immunol 2024; 218:188-198. [PMID: 39185713 DOI: 10.1093/cei/uxae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 06/12/2024] [Accepted: 08/25/2024] [Indexed: 08/27/2024] Open
Abstract
Preclinical data suggest that type I interferon (IFN) responsiveness is essential for the antitumor effects of radiotherapy (RT). However, its clinical value remains unclear. This study aimed to explore this from a clinical perspective. In cohort 1, data from 152 hepatocellular carcinoma (HCC) patients who received RT were analyzed. Blood samples were taken 1 day before and 2 weeks after RT. RT was found to increase serum levels of IFN-β (a subtype of IFN-I) in HCC patients (3.42 ± 1.57 to 5.51 ± 2.11 pg/ml, P < 0.01), particularly in those with favorable responses. Higher post-RT serum IFN-β levels (≥4.77 pg/ml) were associated with better progression-free survival (HR = 0.58, P < 0.01). Cohort 2 included 46 HCC patients, including 23 who underwent preoperative RT and 23 matched control HCC who received surgical resection without RT. Formalin-fixed paraffin-embedded samples were obtained. Neoadjuvant RT significantly increased IFN-β expression in tumor tissues compared to direct surgery (8.13% ± 5.19% to 15.10% ± 5.89%, P < 0.01). Higher post-RT IFN-β (>median) indicated better disease-free survival (P = 0.049). Additionally, increased CD11c+MHCII+CD141+ antigen-presenting cell subsets and CD103+CD39+CD8+ tumor-infiltrating lymphocytes were found in the higher IFN-β group (P = 0.02, P = 0.03), which may contribute to the favorable prognosis in higher IFN-β group. Collectively, these findings suggest that IFN-β response activated by radiation may serve as a prognostic biomarker for HCC patients undergoing RT.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weifeng Hong
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Danxue Zheng
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zongjuan Li
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yong Hu
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yixing Chen
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ping Yang
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhaochong Zeng
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shisuo Du
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Wang SW, Zheng QY, Hong WF, Tang BF, Hsu SJ, Zhang Y, Zheng XB, Zeng ZC, Gao C, Ke AW, Du SS. Mechanism of immune activation mediated by genomic instability and its implication in radiotherapy combined with immune checkpoint inhibitors. Radiother Oncol 2024; 199:110424. [PMID: 38997092 DOI: 10.1016/j.radonc.2024.110424] [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: 04/07/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024]
Abstract
Various genetic and epigenetic changes associated with genomic instability (GI), including DNA damage repair defects, chromosomal instability, and mitochondrial GI, contribute to development and progression of cancer. These alterations not only result in DNA leakage into the cytoplasm, either directly or through micronuclei, but also trigger downstream inflammatory signals, such as the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. Apart from directly inducing DNA damage to eliminate cancer cells, radiotherapy (RT) exerts its antitumor effects through intracellular DNA damage sensing mechanisms, leading to the activation of downstream inflammatory signaling pathways. This not only enables local tumor control but also reshapes the immune microenvironment, triggering systemic immune responses. The combination of RT and immunotherapy has emerged as a promising approach to increase the probability of abscopal effects, where distant tumors respond to treatment due to the systemic immunomodulatory effects. This review emphasizes the importance of GI in cancer biology and elucidates the mechanisms by which RT induces GI remodeling of the immune microenvironment. By elucidating the mechanisms of GI and RT-induced immune responses, we aim to emphasize the crucial importance of this approach in modern oncology. Understanding the impact of GI on tumor biological behavior and therapeutic response, as well as the possibility of activating systemic anti-tumor immunity through RT, will pave the way for the development of new treatment strategies and improve prognosis for patients.
Collapse
Affiliation(s)
- Si-Wei Wang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China
| | - Qiu-Yi Zheng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Wei-Feng Hong
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Bu-Fu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Shu-Jung Hsu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Yang Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Xiao-Bin Zheng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Zhao-Chong Zeng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Chao Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China.
| | - Ai-Wu Ke
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China.
| | - Shi-Suo Du
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China.
| |
Collapse
|
9
|
Bugno J, Wang L, Yu X, Cao X, Wang J, Huang X, Yang K, Piffko A, Chen K, Luo SY, Naccasha E, Hou Y, Fu S, He C, Fu YX, Liang HL, Weichselbaum RR. Targeting the Dendritic Cell-Secreted Immunoregulatory Cytokine CCL22 Alleviates Radioresistance. Clin Cancer Res 2024; 30:4450-4463. [PMID: 38691100 PMCID: PMC11444901 DOI: 10.1158/1078-0432.ccr-23-3616] [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: 11/20/2023] [Revised: 03/20/2024] [Accepted: 04/29/2024] [Indexed: 05/03/2024]
Abstract
PURPOSE Radiation-mediated immune suppression limits efficacy and is a barrier in cancer therapy. Radiation induces negative regulators of tumor immunity including regulatory T cells (Treg). Mechanisms underlying Treg infiltration after radiotherapy (RT) are poorly defined. Given that conventional dendritic cells (cDC) maintain Treg, we sought to identify and target cDC signaling to block Treg infiltration after radiation. EXPERIMENTAL DESIGN Transcriptomics and high dimensional flow cytometry revealed changes in murine tumor cDC that not only mediate Treg infiltration after RT but also associate with worse survival in human cancer datasets. Antibodies perturbing a cDC-CCL22-Treg axis were tested in syngeneic murine tumors. A prototype interferon-anti-epidermal growth factor receptor fusion protein (αEGFR-IFNα) was examined to block Treg infiltration and promote a CD8+ T cell response after RT. RESULTS Radiation expands a population of mature cDC1 enriched in immunoregulatory markers that mediates Treg infiltration via the Treg-recruiting chemokine CCL22. Blocking CCL22 or Treg depletion both enhanced RT efficacy. αEGFR-IFNα blocked cDC1 CCL22 production while simultaneously inducing an antitumor CD8+ T cell response to enhance RT efficacy in multiple EGFR-expressing murine tumor models, including following systemic administration. CONCLUSIONS We identify a previously unappreciated cDC mechanism mediating Treg tumor infiltration after RT. Our findings suggest blocking the cDC1-CCL22-Treg axis augments RT efficacy. αEGFR-IFNα added to RT provided robust antitumor responses better than systemic free interferon administration and may overcome clinical limitations to interferon therapy. Our findings highlight the complex behavior of cDC after RT and provide novel therapeutic strategies for overcoming RT-driven immunosuppression to improve RT efficacy. See related commentary by Kalinski et al., p. 4260.
Collapse
Affiliation(s)
- Jason Bugno
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Xianbin Yu
- Department of Chemistry, University of Chicago, Chicago, Illinois
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois
- Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois
- Howard Hughes Medical Institute, University of Chicago, Chicago, Illinois
| | - Xuezhi Cao
- Guangzhou National Laboratory, Guangzhou, China
| | - Jiaai Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Xiaona Huang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Andras Piffko
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katherine Chen
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Stephen Y Luo
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Emile Naccasha
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Yuzhu Hou
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Sherry Fu
- UT Southwestern Medical School, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chuan He
- Department of Chemistry, University of Chicago, Chicago, Illinois
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois
- Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois
- Howard Hughes Medical Institute, University of Chicago, Chicago, Illinois
| | - Yang-Xin Fu
- Department of Basic Medical Science, Tsinghua University, Beijing, China
| | - Hua L Liang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| |
Collapse
|
10
|
Iriondo O, Mecenas D, Li Y, Chin CR, Thomas A, Moriarty A, Marker R, Wang YJ, Hendrick H, Amzaleg Y, Ortiz V, MacKay M, Dickerson A, Lee G, Harotoonian S, Benayoun BA, Smith A, Mason CE, Roussos Torres ET, Klotz R, Yu M. Hypoxic Memory Mediates Prolonged Tumor-Intrinsic Type I Interferon Suppression to Promote Breast Cancer Progression. Cancer Res 2024; 84:3141-3157. [PMID: 38990731 PMCID: PMC11444891 DOI: 10.1158/0008-5472.can-23-2028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 05/03/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
Hypoxia is a common feature of many solid tumors due to aberrant proliferation and angiogenesis that is associated with tumor progression and metastasis. Most of the well-known hypoxia effects are mediated through hypoxia-inducible factors (HIF). Identification of the long-lasting effects of hypoxia beyond the immediate HIF-induced alterations could provide a better understanding of hypoxia-driven metastasis and potential strategies to circumvent it. Here, we uncovered a hypoxia-induced mechanism that exerts a prolonged effect to promote metastasis. In breast cancer patient-derived circulating tumor cell lines and common breast cancer cell lines, hypoxia downregulated tumor-intrinsic type I IFN signaling and its downstream antigen presentation (AP) machinery in luminal breast cancer cells, via both HIF-dependent and HIF-independent mechanisms. Hypoxia induced durable IFN/AP suppression in certain cell types that was sustained after returning to normoxic conditions, presenting a "hypoxic memory" phenotype. Hypoxic memory of IFN/AP downregulation was established by specific hypoxic priming, and cells with hypoxic memory had an enhanced ability for tumorigenesis and metastasis. Overexpression of IRF3 enhanced IFN signaling and reduced tumor growth in normoxic, but not hypoxic, conditions. The histone deacetylase inhibitor entinostat upregulated IFN targets and erased the hypoxic memory. These results point to a mechanism by which hypoxia facilitates tumor progression through a long-lasting memory that provides advantages for circulating tumor cells during the metastatic cascade. Significance: Long-term cellular memory of hypoxia leads to sustained suppression of tumor-intrinsic type I IFN signaling and the antigen presentation pathway that facilitates tumorigenesis and metastasis. See related commentary by Purdy and Ford, p. 3125.
Collapse
Affiliation(s)
- Oihana Iriondo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
- Center for Cooperative Research (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Desirea Mecenas
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Yilin Li
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Christopher R Chin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Amal Thomas
- Department of Molecular and Computational Biology, USC Dana and David Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Aidan Moriarty
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rebecca Marker
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Yiru J Wang
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Haley Hendrick
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Yonatan Amzaleg
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of the University of Southern California, Los Angeles, California
| | - Veronica Ortiz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Matthew MacKay
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Amber Dickerson
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Grace Lee
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Sevana Harotoonian
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Bérénice A Benayoun
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
- Department of Molecular and Computational Biology, USC Dana and David Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California
| | - Andrew Smith
- Department of Molecular and Computational Biology, USC Dana and David Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Evanthia T Roussos Torres
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
- Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Remi Klotz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
11
|
Ho AY, Shiao S, Kobald SA, Chen J, Duda DG, Ly A, Bossuyt V, Cho HL, Arnold B, Knott S, Gupta GP, McAndrew P, Karlan S, Tighiouart M, Muzikansky A, Basho R, McArthur H. PEARL: A Phase Ib/II Biomarker Study of Adding Radiation Therapy to Pembrolizumab Before Neoadjuvant Chemotherapy in Human Epidermal Growth Factor Receptor 2-Negative Breast Cancer. J Clin Oncol 2024:JCO2400003. [PMID: 39298718 DOI: 10.1200/jco.24.00003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/11/2024] [Accepted: 07/19/2024] [Indexed: 09/22/2024] Open
Abstract
PURPOSE To assess safety and immune biomarkers after preoperative radiation therapy (RT) and anti-PD1 therapy in breast cancer. MATERIALS AND METHODS A phase I/IIb trial of pembrolizumab with RT was conducted in patients with triple-negative breast cancer (TNBC) and hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) breast cancer. All received pembrolizumab followed by a second cycle + RT (anti-PD1/RT) of 24 Gy/three daily fractions delivered to the breast tumor and then neoadjuvant chemotherapy (NAC). Blood and tumor biopsies were obtained at baseline, after anti-PD1, and after anti-PD-RT. Coprimary end points were safety and change in tumor-infiltrating lymphocytes (TILs). Secondary end points were pathologic complete response (pCR), residual cancer burden (RCB) rates, and event-free survival (EFS). RESULTS Sixty-six patients with stage I-III breast cancer (54 TNBC, 12 HR+/HER2-) were enrolled. The median follow-up was 32 months. Safety end point was met. Incidence of grade ≥3 toxicities was 41%. The pCR rate was 59.2%, 33.3%, and 54.5% for the TNBC, HR+/HER2-, and entire cohort, respectively. A total of 77.8% of TNBC and 41.6% of HR+/HER2- had a near pCR (RCB 0-1). The 3-year EFS was 80%. In the entire cohort, PD-L1 expression increased after anti-PD1 (median Combined Positive Score [CPS], 7.49-23.20; 95% CI, -41.88 to -6.30; P = .044) and anti-PD1/RT (median CPS, 7.49-23.41; 95% CI, -41.88 to -6.30; P = .009), compared with baseline. In TNBC, adding RT to anti-PD1 significantly decreased TILs (28.9%-17.1%; 95% CI, 2.46 to 21.09; P = .014). Baseline TILs correlated with PD-L1 expression and TNF-a. CONCLUSION Preoperative RT with pembrolizumab is safe and results in high pCR rates and 3-year EFS, despite the lack of pembrolizumab during NAC. PD-L1 and TILs may be predictive biomarkers for preoperative anti-PD1/RT response. Reduction in TILs after adding RT to anti-PD1 highlights the importance of treatment sequencing.
Collapse
Affiliation(s)
- Alice Y Ho
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC
| | - Stephen Shiao
- Department of Radiation Oncology, Cedars Sinai Medical Center, Los Angeles, CA
| | | | | | - Dan G Duda
- Massachusetts General Hospital, Boston, MA
| | - Amy Ly
- Massachusetts General Hospital, Boston, MA
| | | | | | | | | | | | - Philomena McAndrew
- Department of Radiation Oncology, Cedars Sinai Medical Center, Los Angeles, CA
| | - Scott Karlan
- Department of Radiation Oncology, Cedars Sinai Medical Center, Los Angeles, CA
| | - Mourad Tighiouart
- Department of Radiation Oncology, Cedars Sinai Medical Center, Los Angeles, CA
| | | | - Reva Basho
- Ellison Institute of Technology, Los Angeles, CA
| | | |
Collapse
|
12
|
Berzaghi R, Gundersen K, Dille Pedersen B, Utne A, Yang N, Hellevik T, Martinez-Zubiaurre I. Immunological signatures from irradiated cancer-associated fibroblasts. Front Immunol 2024; 15:1433237. [PMID: 39308864 PMCID: PMC11412886 DOI: 10.3389/fimmu.2024.1433237] [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: 05/15/2024] [Accepted: 08/14/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Cancer-associated fibroblasts (CAFs) are abundant and influential elements of the tumor microenvironment (TME), giving support to tumor development in multiple ways. Among other mechanisms, CAFs are important regulators of immunological processes occurring in tumors. However, CAF-mediated tumor immunomodulation in the context of radiotherapy remains poorly understood. In this study, we explore effects of radiation on CAF-derived immunoregulatory signals to the TME. Methods Primary CAF cultures were established from freshly collected human NSCLC lung tumors. CAFs were exposed to single-high or fractionated radiation regimens (1x18Gy or 3x6Gy), and the expression of different immunoregulatory cell-associated and secreted signaling molecules was analyzed 48h and 6 days after initiation of treatment. Analyses included quantitative measurements of released damage-associated molecular patterns (DAMPs), interferon (IFN) type I responses, expression of immune regulatory receptors, and secretion of soluble cytokines, chemokines, and growth factors. CAFs are able to survive ablative radiation regimens, however they enter into a stage of premature cell senescence. Results Our data show that CAFs avoid apoptosis and do not contribute by release of DAMPs or IFN-I secretion to radiation-mediated tumor immunoregulation. Furthermore, the secretion of relevant immunoregulatory cytokines and growth factors including TGF-β, IL-6, IL-10, TNFα, IL-1β, VEGF, CXCL12, and CXCL10 remain comparable between non-irradiated and radiation-induced senescent CAFs. Importantly, radiation exposure modifies the cell surface expression of some key immunoregulatory receptors, including upregulation of CD73 and CD276. Discussion Our data suggest that CAFs do not participate in the release of danger signals or IFN-I secretion following radiotherapy. The immune phenotype of CAFs and radiation-induced senescent CAFs is similar, however, the observed elevation of some cell surface immunological receptors on irradiated CAFs could contribute to the establishment of an enhanced immunosuppressive TME after radiotherapy.
Collapse
Affiliation(s)
- Rodrigo Berzaghi
- Department of Clinical Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Kristian Gundersen
- Department of Clinical Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Brede Dille Pedersen
- Department of Radiation Oncology, University Hospital of North Norway, Tromsø, Norway
| | - Amalie Utne
- Department of Radiation Oncology, University Hospital of North Norway, Tromsø, Norway
| | - Nannan Yang
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Turid Hellevik
- Department of Radiation Oncology, University Hospital of North Norway, Tromsø, Norway
| | - Inigo Martinez-Zubiaurre
- Department of Clinical Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
13
|
Yan W, Ren Z, Chen X, Zhang R, Lv J, Verma V, Wu M, Chen D, Yu J. Potential Role of Lymphocyte CD44 in Determining Treatment Selection Between Stereotactic Body Radiation Therapy and Surgery for Early-Stage Non-Small Cell Lung Cancer. Int J Radiat Oncol Biol Phys 2024; 120:89-101. [PMID: 38447611 DOI: 10.1016/j.ijrobp.2024.02.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 02/03/2024] [Accepted: 02/12/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE Stereotactic body radiation therapy (SBRT) versus surgery for operable early-stage non-small cell lung cancer (ES-NSCLC) remains highly debated. Herein, we used spatial proteomics to identify whether any molecular biomarker(s) associate with the efficacy of either modality, in efforts to optimize treatment selection between surgery and SBRT for this population. METHODS AND MATERIALS We evaluated biopsy tissue samples from 44 patients with ES-NSCLC treated with first-line SBRT (cohort 1) by GeoMx Digital Spatial Profiling (DSP) with a panel of 70 proteins in 5 spatial molecular compartments: tumor (panCK+), leukocyte (CD45+), lymphocyte (CD3+), macrophage (CD68+), and stroma (α-SMA+). To validate the findings in cohort 1, biopsy samples from 52 patients with ES-NSCLC who received SBRT (cohort 2) and 62 patients with ES-NSCLC who underwent surgery (cohort 3) were collected and analyzed by multiplex immunofluorescence (mIF). RESULTS In cohort 1, higher CD44 expression in the lymphocyte compartment was associated with poorer recurrence-free survival (RFS) (DSP: P < .001; mIF: P < .001) and higher recurrence rate (DSP: P = .001; mIF: P = .004). mIF data from cohort 2 validated these findings (P < .05 for all). From cohort 3, higher lymphocyte CD44 predicted higher RFS after surgery (P = .003). Intermodality comparisons demonstrated that SBRT was associated with significantly higher RFS over surgery in CD44-low patients (P < .001), but surgery was superior to SBRT in CD44-high cases (P = .016). CONCLUSIONS Lymphocyte CD44 may not only be a predictor of SBRT efficacy in this population but also an important biomarker (pending validation by large prospective data) that could better sharpen selection for SBRT versus surgery in ES-NSCLC.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/radiotherapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/surgery
- Radiosurgery
- Hyaluronan Receptors/analysis
- Hyaluronan Receptors/metabolism
- Lung Neoplasms/radiotherapy
- Lung Neoplasms/pathology
- Lung Neoplasms/surgery
- Male
- Female
- Aged
- Lymphocytes
- Middle Aged
- Antigens, Differentiation, Myelomonocytic/analysis
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, CD/analysis
- Leukocyte Common Antigens/analysis
- Leukocyte Common Antigens/metabolism
- Aged, 80 and over
- CD3 Complex/analysis
- CD3 Complex/metabolism
- Neoplasm Staging
- Biomarkers, Tumor/analysis
- Patient Selection
- CD68 Molecule
Collapse
Affiliation(s)
- Weiwei Yan
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ziyuan Ren
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xi Chen
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ran Zhang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Juncai Lv
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Vivek Verma
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Meng Wu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Dawei Chen
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Jinming Yu
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
14
|
Shaitelman SF, Le-Petross H, Raso MG, Swanson DM, Schalck AP, Contreras A, Yang F, Muruganandham M, Zhao GZ, Sawakuchi GO, Kim LH, Batra H, Smith BD, Stauder MC, Woodward WA, Reddy JP, Litton JK, Thompson A, Bedrosian I, Mittendorf EA. PRECISE: Preoperative Radiation Therapy to Elicit Critical Immune Stimulating Effects - A Phase 2 Clinical Trial. Int J Radiat Oncol Biol Phys 2024:S0360-3016(24)03231-0. [PMID: 39147206 DOI: 10.1016/j.ijrobp.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/23/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE Radiation therapy is an underinvestigated tool for priming the immune system in intact human breast cancers. We sought here to investigate if a preoperative radiation therapy boost delivered was associated with a significant change in tumor-infiltrating lymphocytes (TILs) in the tumor in estrogen receptor positive, HER2Neu nonamplified breast cancers. METHODS AND MATERIALS A total of 20 patients were enrolled in a phase 2 clinical trial and received either 7.5 Gy × 1 fraction or 2 Gy × 5 fractions, completed 6 to 8 days before surgery. Percent stromal TILs were evaluated on hematoxylin and eosin-stained samples. Short-term safety was assessed based on time to surgery, toxicities, and cosmesis up to 6 months after boost. RESULTS Stromal TIL increased 6 to 8 days after completion of boost radiation therapy (median 3.0 [IQR, 1.0-6.5]) before radiation therapy versus median 5.0 (IQR, 1.5-8.0) after radiation therapy, P = .0037. Zero grade ≥3 toxicities up to 6 months after boost were experienced. In all, 94% (16/17) patients with 6-month follow-up cosmetic assessment after breast conservation had good-excellent cosmesis by physician assessment. CONCLUSION In this phase 2 trial, preoperative radiation therapy boost resulted in a short-term increase in stromal TIL with minimal toxicities. Preoperative breast radiation therapy appears to be safe and may be a feasible means for priming the tumor microenvironment.
Collapse
Affiliation(s)
- Simona F Shaitelman
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Huong Le-Petross
- Department of Breast Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria G Raso
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David M Swanson
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aislyn P Schalck
- Department of Genomics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alejandro Contreras
- Department of Anatomical Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fei Yang
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Manickam Muruganandham
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - George Z Zhao
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel O Sawakuchi
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Leonard H Kim
- Department of Radiation Oncology, MD Anderson Cancer Center at Cooper, Camden, New Jersey
| | - Harsh Batra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Benjamin D Smith
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael C Stauder
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wendy A Woodward
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jay P Reddy
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer K Litton
- Department of Clinical Research, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alastair Thompson
- Section of Breast Surgery, Division of Surgical Oncology, Baylor College of Medicine, Houston, Texas
| | - Isabelle Bedrosian
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth A Mittendorf
- Division of Breast Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Gellert J, Agardy DA, Kumar S, Kourtesakis A, Boschert T, Jähne K, Breckwoldt MO, Bunse L, Wick W, Davies MA, Platten M, Bunse T. Tumoral Interferon Beta Induces an Immune-Stimulatory Phenotype in Tumor-Associated Macrophages in Melanoma Brain Metastases. CANCER RESEARCH COMMUNICATIONS 2024; 4:2189-2202. [PMID: 39056192 PMCID: PMC11337092 DOI: 10.1158/2767-9764.crc-24-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/14/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
Type I interferons (IFN) are immune-stimulatory cytokines involved in antiviral and antitumor immune responses. They enhance the efficacy of immunogenic anticancer therapies such as radiotherapy by activating both innate and adaptive immune cells. Macrophages are one of the most abundant innate immune cells in the immune microenvironment of melanoma brain metastases (MBM) and can exert potent immune-suppressive functions. Here, we investigate the potential of tumoral type I IFNs to repolarize tumor-associated macrophages (TAM) in two murine MBM models and assess the effects of radiotherapy-induced type I IFN on TAMs in a transcriptomic MBM patient dataset. In mice, we describe a proinflammatory M1-like TAM phenotype induced by tumoral IFNβ and identify a myeloid type I IFN-response signature associated with a high M1/M2-like TAM ratio. Following irradiation, patients with MBM displaying a myeloid type I IFN-response signature showed increased overall survival, providing evidence that tumoral IFNβ supports an effective antitumor immune response by re-educating immune-regulatory TAM. These findings uncover type I IFN-inducing therapies as a potential macrophage-targeting therapeutic approach and provide a rationale for combining radiotherapy with concomitant immunotherapy to improve treatment response in patients with MBM. SIGNIFICANCE Our study shows that re-education of tumor-associated macrophages by tumoral IFNβ translates into improved clinical outcome in patients with melanoma brain metastases, providing pathomechanistic insights into synergistic type I interferon-inducing therapies with immunotherapies and warranting investigation of IFNβ as a predictive biomarker for combined radioimmunotherapy.
Collapse
Affiliation(s)
- Julia Gellert
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, Heidelberg, Germany.
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience (MCTN), Heidelberg University, Mannheim, Germany.
| | - Dennis A. Agardy
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, Heidelberg, Germany.
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience (MCTN), Heidelberg University, Mannheim, Germany.
- Faculty of Bioscience, Heidelberg University, Heidelberg, Germany.
| | - Swaminathan Kumar
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Alexandros Kourtesakis
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany.
- Faculty of Bioscience, Heidelberg University, Heidelberg, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit (CCU) Neurooncology, Heidelberg, Germany.
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.
| | - Tamara Boschert
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, Heidelberg, Germany.
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany.
- Helmholtz Institute for Translational Oncology Mainz (HI-TRON Mainz)—A Helmholtz Institute of the DKFZ, Mainz, Germany.
| | - Kristine Jähne
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, Heidelberg, Germany.
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience (MCTN), Heidelberg University, Mannheim, Germany.
| | - Michael O. Breckwoldt
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, Heidelberg, Germany.
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany.
| | - Lukas Bunse
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, Heidelberg, Germany.
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience (MCTN), Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
- Hertie Network of Excellence in Clinical Neuroscience, Frankfurt, Germany.
| | - Wolfgang Wick
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit (CCU) Neurooncology, Heidelberg, Germany.
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.
| | - Michael A. Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Michael Platten
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, Heidelberg, Germany.
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience (MCTN), Heidelberg University, Mannheim, Germany.
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.
- Helmholtz Institute for Translational Oncology Mainz (HI-TRON Mainz)—A Helmholtz Institute of the DKFZ, Mainz, Germany.
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany.
| | - Theresa Bunse
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, Heidelberg, Germany.
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience (MCTN), Heidelberg University, Mannheim, Germany.
| |
Collapse
|
16
|
Passelli K, Repáraz D, Kinj R, Herrera FG. Strategies for overcoming tumour resistance to immunotherapy: harnessing the power of radiation therapy. Br J Radiol 2024; 97:1378-1390. [PMID: 38833685 PMCID: PMC11256940 DOI: 10.1093/bjr/tqae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 06/06/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized cancer treatment; yet their efficacy remains variable across patients. This review delves into the intricate interplay of tumour characteristics contributing to resistance against ICI therapy and suggests that combining with radiotherapy holds promise. Radiation, known for its ability to trigger immunogenic cell death and foster an in situ vaccination effect, may counteract these resistance mechanisms, enhancing ICI response and patient outcomes. However, particularly when delivered at high-dose, it may trigger immunosuppressive mechanism and consequent side-effects. Notably, low-dose radiotherapy (LDRT), with its capacity for tumour reprogramming and reduced side effects, offers the potential for widespread application. Preclinical and clinical studies have shown encouraging results in this regard.
Collapse
Affiliation(s)
- Katiuska Passelli
- Centre Hospitalier Universitaire Vaudoise, Service of Radiation Oncology, Department of Oncology, University of Lausanne, AGORA Center for Cancer Research, Swiss Cancer Center Leman, 1012-Lausanne, Switzerland
| | - David Repáraz
- Centre Hospitalier Universitaire Vaudoise, Service of Radiation Oncology, Department of Oncology, University of Lausanne, AGORA Center for Cancer Research, Swiss Cancer Center Leman, 1012-Lausanne, Switzerland
| | - Remy Kinj
- Centre Hospitalier Universitaire Vaudoise, Service of Radiation Oncology, Department of Oncology, University of Lausanne, 1012-Lausanne, Switzerland
| | - Fernanda G Herrera
- Centre Hospitalier Universitaire Vaudois, Service of Radiation Oncology and Service of Immuno-oncology, Department of Oncology, University of Lausanne, Ludwig Institute for Cancer Research, Agora Center for Cancer Research, Swiss Cancer Center Leman, 1012-Lausanne, Switzerland
| |
Collapse
|
17
|
Bliley R, Avant A, Medina TM, Lanning RM. Radiation and Melanoma: Where Are We Now? Curr Oncol Rep 2024; 26:904-914. [PMID: 38822928 DOI: 10.1007/s11912-024-01557-y] [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: 05/21/2024] [Indexed: 06/03/2024]
Abstract
PURPOSE OF REVIEW This review summarizes the current role of radiotherapy for the treatment of cutaneous melanoma in the definitive, adjuvant, and palliative settings, and combinations with immunotherapy and targeted therapies. RECENT FINDINGS Definitive radiotherapy may be considered for lentigo maligna if surgery would be disfiguring. High risk, resected melanoma may be treated with adjuvant radiotherapy, but the role is poorly defined since the advent of effective systemic therapies. For patients with metastatic disease, immunotherapy and targeted therapies can be delivered safely in tandem with radiotherapy to improve outcomes. Radiotherapy and modern systemic therapies act in concert to improve outcomes, especially in the metastatic setting. Further prospective data is needed to guide the use of definitive radiotherapy for lentigo maligna and adjuvant radiotherapy for high-risk melanoma in the immunotherapy era. Current evidence does not support an abscopal response or at least identify the conditions necessary to reliably produce one with combinations of radiation and immunotherapy.
Collapse
Affiliation(s)
- Roy Bliley
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Adam Avant
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Theresa M Medina
- Department of Medical Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ryan M Lanning
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
18
|
Shen Y, Zhao X, Chen L, Wang X, Wang D, Zhang H, Zheng Z, Huang W, Zheng C, Chen Y, Chen C, Chen Q. A modified HSV-1 oncolytic virus reconciles antiviral and antitumor immunity via promoting IFNβ expression and inhibiting PKR. Int J Biol Macromol 2024; 274:133297. [PMID: 38925170 DOI: 10.1016/j.ijbiomac.2024.133297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/17/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024]
Abstract
Type I interferon (IFN-I) is a potent immune modulator intricately involved in regulating tumor immunity. Meanwhile, the integrity of the IFN-I signaling pathway is essential for radiotherapy, chemotherapy, targeted therapy, and immunotherapy. However, the clinical application of IFN-I remains challenging due to its non-specific cytotoxicity and limited half-life. To overcome these limitations, we developed a gene delivery platform, CRISPR-V, enabling the rapid creation of novel HSV-1 oncolytic viruses. Utilizing this platform, we created an oncolytic virus, OVH-IFNβ, in which the IFNβ gene was incorporated into the HSV-1 genome. However, exogenous IFNβ expression significantly inhibited OVH-IFNβ replication. Through transcriptome data analyses, we identified several ISG genes inhibiting OVH-IFNβ replication. By gene knockout and functional studies of the downstream effectors, we confirmed the prominent antiviral activities of protein kinase R (PKR). To balance the antitumor and antiviral immunity of IFNβ, we developed a novel HSV-1 oncolytic virus, OVH-IFNβ-iPKR, which can express IFNβ while inhibiting PKR, leading to a potent antitumor immunity while reducing the antiviral capacity of IFNβ. OVH-IFNβ-iPKR shows a strong ability to induce immunogenic cell death and activate tumor-specific CD8+ T cells, leading to de novo immune responses and providing a novel strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- Yangkun Shen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Xiangqian Zhao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Lizhu Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xin Wang
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Dawei Wang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Hucheng Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Zuda Zheng
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Weiwei Huang
- Department of Medical Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Yu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| | - Chuanben Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China.
| |
Collapse
|
19
|
Amit U, Uslu U, Verginadis II, Kim MM, Motlagh SAO, Diffenderfer ES, Assenmacher CA, Bicher S, Atoche SJ, Ben-Josef E, Young RM, June CH, Koumenis C. Proton radiation boosts the efficacy of mesothelin-targeting chimeric antigen receptor T cell therapy in pancreatic cancer. Proc Natl Acad Sci U S A 2024; 121:e2403002121. [PMID: 39047033 PMCID: PMC11294999 DOI: 10.1073/pnas.2403002121] [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: 02/12/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents a challenge in oncology, with limited treatment options for advanced-stage patients. Chimeric antigen receptor T cell (CAR T) therapy targeting mesothelin (MSLN) shows promise, but challenges such as the hostile immunosuppressive tumor microenvironment (TME) hinder its efficacy. This study explores the synergistic potential of combining proton radiation therapy (RT) with MSLN-targeting CAR T therapy in a syngeneic PDAC model. Proton RT significantly increased MSLN expression in tumor cells and caused a significant increase in CAR T cell infiltration into tumors. The combination therapy reshaped the immunosuppressive TME, promoting antitumorigenic M1 polarized macrophages and reducing myeloid-derived suppressor cells (MDSC). In a flank PDAC model, the combination therapy demonstrated superior attenuation of tumor growth and improved survival compared to individual treatments alone. In an orthotopic PDAC model treated with image-guided proton RT, tumor growth was significantly reduced in the combination group compared to the RT treatment alone. Further, the combination therapy induced an abscopal effect in a dual-flank tumor model, with increased serum interferon-γ levels and enhanced proliferation of extratumoral CAR T cells. In conclusion, combining proton RT with MSLN-targeting CAR T therapy proves effective in modulating the TME, enhancing CAR T cell trafficking, and exerting systemic antitumor effects. Thus, this combinatorial approach could present a promising strategy for improving outcomes in unresectable PDAC.
Collapse
Affiliation(s)
- Uri Amit
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Radiation Oncology, Tel Aviv Medical Center, Tel Aviv64239, Israel
| | - Ugur Uslu
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Ioannis I. Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Michele M. Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Seyyedeh Azar Oliaei Motlagh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Eric S. Diffenderfer
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, Comparative Pathology Core, University of Pennsylvania, Philadelphia, PA19104
| | - Sandra Bicher
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Sebastian J. Atoche
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Edgar Ben-Josef
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Regina M. Young
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Carl H. June
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
20
|
Choi S, Bae HG, Jo DG, Kim WY. The Role of IRF9 Upregulation in Modulating Sensitivity to Olaparib and Platinum-Based Chemotherapies in Breast Cancer. Genes (Basel) 2024; 15:959. [PMID: 39062738 PMCID: PMC11276373 DOI: 10.3390/genes15070959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/19/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024] Open
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors are targeted therapies that accumulate DNA damage by interfering with DNA repair mechanisms and are approved for treating several cancers with BRCA1/2 mutations. In this study, we utilized CRISPR-dCas9 interference screening to identify genes regulating sensitivity to PARP inhibitors in breast cancer cell lines. Our findings indicated that the interferon (IFN) signaling gene IRF9 was critically involved in modulating sensitivity to these inhibitors. We revealed that the loss of IRF9 leads to increased resistance to the PARP inhibitor in MDA-MB-468 cells, and a similar desensitization was observed in another breast cancer cell line, MDA-MB-231. Further analysis indicated that while the basal expression of IRF9 did not correlate with the response to the PARP inhibitor olaparib, its transcriptional induction was significantly associated with increased sensitivity to the DNA-damaging agent cisplatin in the NCI-60 cell line panel. This finding suggests a mechanistic link between IRF9 induction and cellular responses to DNA damage. Additionally, data from the METABRIC patient tissue study revealed a complex network of IFN-responsive gene expressions postchemotherapy, with seven upregulated genes, including IRF9, and three downregulated genes. These findings underscore the intricate role of IFN signaling in the cellular response to chemotherapy. Collectively, our CRISPR screening data and subsequent bioinformatic analyses suggest that IRF9 is a novel biomarker for sensitivity to DNA-damaging agents, such as olaparib and platinum-based chemotherapeutic agents. Our findings for IRF9 not only enhance our understanding of the genetic basis of drug sensitivity, but also elucidate the role of IRF9 as a critical effector within IFN signaling pathways, potentially influencing the association between the host immune system and chemotherapeutic efficacy.
Collapse
Affiliation(s)
- SeokGyeong Choi
- College of Pharmacy, Sookmyung Women’s University, Seoul 04310, Republic of Korea;
| | - Han-Gyu Bae
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; (H.-G.B.); (D.-G.J.)
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; (H.-G.B.); (D.-G.J.)
| | - Woo-Young Kim
- College of Pharmacy, Sookmyung Women’s University, Seoul 04310, Republic of Korea;
- Muscle Physiome Research Center, Sookmyung Women’s University, Seoul 04310, Republic of Korea
- Drug Information Research Institute, Sookmyung Women’s University, Seoul 04310, Republic of Korea
| |
Collapse
|
21
|
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
|
22
|
Takashima ME, Berg TJ, Morris ZS. The Effects of Radiation Dose Heterogeneity on the Tumor Microenvironment and Anti-Tumor Immunity. Semin Radiat Oncol 2024; 34:262-271. [PMID: 38880534 DOI: 10.1016/j.semradonc.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Radiotherapy elicits dose- and lineage-dependent effects on immune cell survival, migration, activation, and proliferation in targeted tumor microenvironments. Radiation also stimulates phenotypic changes that modulate the immune susceptibility of tumor cells. This has raised interest in using radiotherapy to promote greater response to immunotherapies. To clarify the potential of such combinations, it is critical to understand how best to administer radiation therapy to achieve activation of desired immunologic mechanisms. In considering the multifaceted process of priming and propagating anti-tumor immune response, radiation dose heterogeneity emerges as a potential means for simultaneously engaging diverse dose-dependent effects in a single tumor environment. Recent work in spatially fractionated external beam radiation therapy demonstrates the expansive immune responses achievable when a range of high to low dose radiation is delivered in a tumor. Brachytherapy and radiopharmaceutical therapies deliver inherently heterogeneous distributions of radiation that may contribute to immunogenicity. This review evaluates the interplay of radiation dose and anti-tumor immune response and explores emerging methodological approaches for investigating the effects of heterogeneous dose distribution on immune responses.
Collapse
Affiliation(s)
- Maya E Takashima
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Tracy J Berg
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| |
Collapse
|
23
|
Wang L, Lynch C, Pitroda SP, Piffkó A, Yang K, Huser AK, Liang HL, Weichselbaum RR. Radiotherapy and immunology. J Exp Med 2024; 221:e20232101. [PMID: 38771260 PMCID: PMC11110906 DOI: 10.1084/jem.20232101] [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: 02/29/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 05/22/2024] Open
Abstract
The majority of cancer patients receive radiotherapy during the course of treatment, delivered with curative intent for local tumor control or as part of a multimodality regimen aimed at eliminating distant metastasis. A major focus of research has been DNA damage; however, in the past two decades, emphasis has shifted to the important role the immune system plays in radiotherapy-induced anti-tumor effects. Radiotherapy reprograms the tumor microenvironment, triggering DNA and RNA sensing cascades that activate innate immunity and ultimately enhance adaptive immunity. In opposition, radiotherapy also induces suppression of anti-tumor immunity, including recruitment of regulatory T cells, myeloid-derived suppressor cells, and suppressive macrophages. The balance of pro- and anti-tumor immunity is regulated in part by radiotherapy-induced chemokines and cytokines. Microbiota can also influence radiotherapy outcomes and is under clinical investigation. Blockade of the PD-1/PD-L1 axis and CTLA-4 has been extensively investigated in combination with radiotherapy; we include a review of clinical trials involving inhibition of these immune checkpoints and radiotherapy.
Collapse
Affiliation(s)
- Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Connor Lynch
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
- Ludwig Center for Metastasis Research, 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
| | - András Piffkó
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Amy K. Huser
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Hua Laura Liang
- 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
|
24
|
Zemek RM, Anagnostou V, Pires da Silva I, Long GV, Lesterhuis WJ. Exploiting temporal aspects of cancer immunotherapy. Nat Rev Cancer 2024; 24:480-497. [PMID: 38886574 DOI: 10.1038/s41568-024-00699-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2024] [Indexed: 06/20/2024]
Abstract
Many mechanisms underlying an effective immunotherapy-induced antitumour response are transient and critically time dependent. This is equally true for several immunological events in the tumour microenvironment induced by other cancer treatments. Immune checkpoint therapy (ICT) has proven to be very effective in the treatment of some cancers, but unfortunately, with many cancer types, most patients do not experience a benefit. To improve outcomes, a multitude of clinical trials are testing combinations of ICT with various other treatment modalities. Ideally, those combination treatments should take time-dependent immunological events into account. Recent studies have started to map the dynamic cellular and molecular changes that occur during treatment with ICT, in the tumour and systemically. Here, we overlay the dynamic ICT response with the therapeutic response following surgery, radiotherapy, chemotherapy and targeted therapies. We propose that by combining treatments in a time-conscious manner, we may optimally exploit the interactions between the individual therapies.
Collapse
Affiliation(s)
- Rachael M Zemek
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Valsamo Anagnostou
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Inês Pires da Silva
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine & Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Crown Princess Mary Cancer Centre Westmead, Blacktown Hospital, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine & Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Willem Joost Lesterhuis
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia.
| |
Collapse
|
25
|
Mitrea DA, Froicu EM, Prenen H, Gambacorta MA, Span PN, Poortmans P. Combining immunotherapy and radiation therapy in gastrointestinal cancers: A review. Crit Rev Oncol Hematol 2024; 199:104381. [PMID: 38735504 DOI: 10.1016/j.critrevonc.2024.104381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024] Open
Abstract
INTRODUCTION AND PURPOSE With a significant global impact, treatment of gastrointestinal (GI) cancers still presents with challenges, despite current multimodality approaches in advanced stages. Clinical trials are expanding for checkpoint inhibition (ICI) combined with radiation therapy (RT). This review intends to offer a comprehensive image of the current data regarding the effectiveness of this association, and to reflect on possible directions to further optimize the results. RESULTS Several early phase studies demonstrated encouraging potential. However, translating preclinical outcomes to clinical settings proves challenging, especially in immunologically "cold" environments. GI cancers exhibit heterogeneity, requiring tailored approaches based on disease stage and patient characteristics. Current results, though promising, lack the power of evidence to influence the general practice. CONCLUSIONS Finding biomarkers for identifying or converting resistant cancers is essential for maximizing responses, moreover in this context strategic RT parameters need to be carefully considered. Our review emphasizes the significance of having a thorough grasp of how immunology, tumour biology, and treatment settings interact in order to propose novel research avenues and efficient GI cancer therapy.
Collapse
Affiliation(s)
- Diana A Mitrea
- Department of Radiation Oncology, Centre Antoine-Lacassagne, 33 Av. de Valombrose, Nice 06100, France.
| | - Eliza M Froicu
- Department of Medical Oncology, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Hans Prenen
- Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Maria A Gambacorta
- Department of Radiation Oncology Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Paul N Span
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Philip Poortmans
- Department of Radiation Oncology, Iridium Netwerk, Wilrijk-Antwerp, Belgium; University of Antwerp, Faculty of Medicine and Health Sciences, Wilrijk-Antwerp, Belgium
| |
Collapse
|
26
|
Akhavan D, Subham S, Jeppson JD, Aguilar B, Wong RA, Hibbard JC, Hui S, Wong JYC, Forman SJ, Alizadeh D, Brown CE. Evaluation of the Immunomodulatory Effects of Radiation for Chimeric Antigen Receptor T Cell Therapy in Glioblastoma Multiforme. Cells 2024; 13:1075. [PMID: 38994929 PMCID: PMC11240512 DOI: 10.3390/cells13131075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/05/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
Standard-of-care treatment for Glioblastoma Multiforme (GBM) is comprised of surgery and adjuvant chemoradiation. Chimeric Antigen Receptor (CAR) T cell therapy has demonstrated disease-modifying activity in GBM and holds great promise. Radiation, a standard-of-care treatment for GBM, has well-known immunomodulatory properties and may overcome the immunosuppressive tumor microenvironment (TME); however, radiation dose optimization and integration with CAR T cell therapy is not well defined. Murine immunocompetent models of GBM were treated with titrated doses of stereotactic radiosurgery (SRS) of 5, 10, and 20 Gray (Gy), and the TME was analyzed using Nanostring. A conditioning dose of 10 Gy was determined based on tumor growth kinetics and gene expression changes in the TME. We demonstrate that a conditioning dose of 10 Gy activates innate and adaptive immune cells in the TME. Mice treated with 10 Gy in combination with mCAR T cells demonstrated enhanced antitumor activity and superior memory responses to rechallenge with IL13Rα2-positive tumors. Furthermore, 10 Gy plus mCAR T cells also protected against IL13Rα2-negative tumors through a mechanism that was, in part, c-GAS-STING pathway-dependent. Together, these findings support combination conditioning with low-dose 10 Gy radiation in combination with mCAR T cells as a therapeutic strategy for GBM.
Collapse
Affiliation(s)
- David Akhavan
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, KS 66160, USA
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Cancer Center, Kansas City, KS 66160, USA
- Department of Cancer Biology, University of Kansas Cancer Center, Kansas City, KS 66160, USA
- Bioengineering Program, University of Kansas, Lawrence, KS 66045, USA
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Siddharth Subham
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, KS 66160, USA
- Department of Cancer Biology, University of Kansas Cancer Center, Kansas City, KS 66160, USA
- Bioengineering Program, University of Kansas, Lawrence, KS 66045, USA
| | - John D Jeppson
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, KS 66160, USA
| | - Brenda Aguilar
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Robyn A Wong
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jonathan C Hibbard
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Susanta Hui
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jeffrey Y C Wong
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Stephen J Forman
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Darya Alizadeh
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Christine E Brown
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
27
|
Jin Y, Jiang J, Mao W, Bai M, Chen Q, Zhu J. Treatment strategies and molecular mechanism of radiotherapy combined with immunotherapy in colorectal cancer. Cancer Lett 2024; 591:216858. [PMID: 38621460 DOI: 10.1016/j.canlet.2024.216858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024]
Abstract
Radiotherapy (RT) remodels the tumor immune microenvironment (TIME) and modulates the immune response to indirectly destroy tumor cells, in addition to directly killing tumor cells. RT combined with immunotherapy may significantly enhance the efficacy of RT in colorectal cancer by modulating the microenvironment. However, the molecular mechanisms by which RT acts as an immunomodulator to modulate the immune microenvironment remain unclear. Further, the optimal modalities of RT combined with immunotherapy for the treatment of colorectal cancer, such as the time point of combining RT and immunization, the fractionation pattern and dosage of radiotherapy, and other methods to improve the efficacy, are also being explored parallelly. To address these aspects, in this review, we summarized the mechanisms by which RT modulates TIME and concluded the progress of RT combined with immunization in preclinical and clinical trials. Finally, we discussed heavy ion radiation therapy and the efficacy of prediction markers and other immune combination therapies. Overall, combining RT with immunotherapy to enhance antitumor effects will have a significant clinical implication and will help to facilitate individualized treatment modalities.
Collapse
Affiliation(s)
- Yuzhao Jin
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China; Wenzhou Medical University, Wenzhou, 325000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China
| | - Jin Jiang
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, 31400, China
| | - Wei Mao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China
| | - Minghua Bai
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China
| | - Qianping Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 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, 310000, China; Wenzhou Medical University, Wenzhou, 325000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China.
| |
Collapse
|
28
|
Hodson D, Mistry H, Yates J, Guzzetti S, Davies M, Aarons L, Ogungbenro K. Hierarchical cluster analysis and nonlinear mixed-effects modelling for candidate biomarker detection in preclinical models of cancer. Eur J Pharm Sci 2024; 197:106774. [PMID: 38641123 DOI: 10.1016/j.ejps.2024.106774] [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: 08/18/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Preclinical models of cancer can be of translational benefit when assessing how different biomarkers are regulated in response to particular treatments. Detection of molecular biomarkers in preclinical models of cancer is difficult due inter-animal variability in responses, combined with limited accessibility of longitudinal data. METHODS Nonlinear mixed-effects modelling (NLME) was used to analyse tumour growth data based on expected tumour growth rates observed 7 days after initial doses (DD7) of Radiotherapy (RT) and Combination of RT with DNA Damage Response Inhibitors (DDRi). Cox regression was performed to confirm an association between DD7 and survival. Hierarchical Cluster Analysis (HCA) was then used to identify candidate biomarkers impacting responses to RT and RT/DDRi and these were validated using NLME. RESULTS Cox regression confirmed significant associations between DD7 and survival. HCA of RT treated samples, combined with NLME confirmed significant associations between DD7 and Cluster specific CD8+ Ki67 MFI, as well as DD7 and cluster specific Natural Killer cell density in RT treated mice. CONCLUSION Application of NLME, as well as HCA of candidate biomarkers may provide additional avenues to assess the effect of RT in MC38 syngeneic tumour models. Additional studies would need to be conducted to confirm association between DD7 and biomarkers in RT/DDRi treated mice.
Collapse
Affiliation(s)
- David Hodson
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Stopford Building, University of Manchester, Manchester M13 9PT, UK
| | - Hitesh Mistry
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Stopford Building, University of Manchester, Manchester M13 9PT, UK
| | - James Yates
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Sofia Guzzetti
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Michael Davies
- DMPK, Research and Early Development, Neuroscience R&D, AstraZeneca, Cambridge, UK
| | - Leon Aarons
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Stopford Building, University of Manchester, Manchester M13 9PT, UK
| | - Kayode Ogungbenro
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Stopford Building, University of Manchester, Manchester M13 9PT, UK.
| |
Collapse
|
29
|
Ednacot EMQ, Nabhani A, Dinh DM, Morehouse BR. Pharmacological potential of cyclic nucleotide signaling in immunity. Pharmacol Ther 2024; 258:108653. [PMID: 38679204 DOI: 10.1016/j.pharmthera.2024.108653] [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: 01/07/2024] [Revised: 03/16/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Cyclic nucleotides are important signaling molecules that play many critical physiological roles including controlling cell fate and development, regulation of metabolic processes, and responding to changes in the environment. Cyclic nucleotides are also pivotal regulators in immune signaling, orchestrating intricate processes that maintain homeostasis and defend against pathogenic threats. This review provides a comprehensive examination of the pharmacological potential of cyclic nucleotide signaling pathways within the realm of immunity. Beginning with an overview of the fundamental roles of cAMP and cGMP as ubiquitous second messengers, this review delves into the complexities of their involvement in immune responses. Special attention is given to the challenges associated with modulating these signaling pathways for therapeutic purposes, emphasizing the necessity for achieving cell-type specificity to avert unintended consequences. A major focus of the review is on the recent paradigm-shifting discoveries regarding specialized cyclic nucleotide signals in the innate immune system, notably the cGAS-STING pathway. The significance of cyclic dinucleotides, exemplified by 2'3'-cGAMP, in controlling immune responses against pathogens and cancer, is explored. The evolutionarily conserved nature of cyclic dinucleotides as antiviral agents, spanning across diverse organisms, underscores their potential as targets for innovative immunotherapies. Findings from the last several years have revealed a striking diversity of novel bacterial cyclic nucleotide second messengers which are involved in antiviral responses. Knowledge of the existence and precise identity of these molecules coupled with accurate descriptions of their associated immune defense pathways will be essential to the future development of novel antibacterial therapeutic strategies. The insights presented herein may help researchers navigate the evolving landscape of immunopharmacology as it pertains to cyclic nucleotides and point toward new avenues or lines of thinking about development of therapeutics against the pathways they regulate.
Collapse
Affiliation(s)
- Eirene Marie Q Ednacot
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Ali Nabhani
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - David M Dinh
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Benjamin R Morehouse
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92697, USA; Center for Virus Research, University of California Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
30
|
Zhou Z, Huang S, Fan F, Xu Y, Moore C, Li S, Han C. The multiple faces of cGAS-STING in antitumor immunity: prospects and challenges. MEDICAL REVIEW (2021) 2024; 4:173-191. [PMID: 38919400 PMCID: PMC11195429 DOI: 10.1515/mr-2023-0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/28/2024] [Indexed: 06/27/2024]
Abstract
As a key sensor of double-stranded DNA (dsDNA), cyclic GMP-AMP synthase (cGAS) detects cytosolic dsDNA and initiates the synthesis of 2'3' cyclic GMP-AMP (cGAMP) that activates the stimulator of interferon genes (STING). This finally promotes the production of type I interferons (IFN-I) that is crucial for bridging innate and adaptive immunity. Recent evidence show that several antitumor therapies, including radiotherapy (RT), chemotherapy, targeted therapies and immunotherapies, activate the cGAS-STING pathway to provoke the antitumor immunity. In the last decade, the development of STING agonists has been a major focus in both basic research and the pharmaceutical industry. However, up to now, none of STING agonists have been approved for clinical use. Considering the broad expression of STING in whole body and the direct lethal effect of STING agonists on immune cells in the draining lymph node (dLN), research on the optimal way to activate STING in tumor microenvironment (TME) appears to be a promising direction. Moreover, besides enhancing IFN-I signaling, the cGAS-STING pathway also plays roles in senescence, autophagy, apoptosis, mitotic arrest, and DNA repair, contributing to tumor development and metastasis. In this review, we summarize the recent advances on cGAS-STING pathway's response to antitumor therapies and the strategies involving this pathway for tumor treatment.
Collapse
Affiliation(s)
- Zheqi Zhou
- Peking University International Cancer Institute, Peking University Cancer Hospital and Institute, Health Science Center, Peking University, Beijing, China
| | - Sanling Huang
- Peking University International Cancer Institute, Peking University Cancer Hospital and Institute, Health Science Center, Peking University, Beijing, China
| | - Fangying Fan
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China
| | - Yan Xu
- Peking University International Cancer Institute, Peking University Cancer Hospital and Institute, Health Science Center, Peking University, Beijing, China
| | - Casey Moore
- Departments of Immunology, Pathology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Sirui Li
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chuanhui Han
- Peking University International Cancer Institute, Peking University Cancer Hospital and Institute, Health Science Center, Peking University, Beijing, China
| |
Collapse
|
31
|
Zenga J, Awan MJ, Frei A, Massey B, Bruening J, Shukla M, Sharma GP, Shreenivas A, Wong SJ, Zimmermann MT, Mathison AJ, Himburg HA. Type I interferon signaling promotes radioresistance in head and neck cancer. Transl Cancer Res 2024; 13:2535-2543. [PMID: 38881922 PMCID: PMC11170510 DOI: 10.21037/tcr-23-2104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/08/2024] [Indexed: 06/18/2024]
Abstract
Despite the promise of concurrent radiotherapy (RT) and immunotherapy in head and neck cancer (HNC), multiple randomized trials of this combination have had disappointing results. To evaluate potential immunologic mechanisms of RT resistance, we compared pre-treatment HNCs that developed RT resistance to a matched cohort that achieved curative status. Gene set enrichment analysis demonstrated that a pre-treatment pro-immunogenic tumor microenvironment (TME), including type II interferon [interferon gamma (IFNγ)] and tumor necrosis factor alpha (TNFα) signaling, predicted cure while type I interferon [interferon alpha (IFNα)] enrichment was associated with an immunosuppressive TME found in tumors that went on to recur. We then used immune deconvolution of RNA sequencing datasets to evaluate immunologic cell subset enrichment. This identified M2 macrophage signaling associated with type I IFN pathway expression in RT-recurrent disease. To further dissect mechanism, we then evaluated differential gene expression between pre-treatment and RT-resistant HNCs from sampled from the same patients at the same anatomical location in the oral cavity. Here, recurrent samples exhibited upregulation of type I IFN-stimulated genes (ISGs) including members of the IFN-induced protein with tetratricopeptide repeats (IFIT) and IFN-induced transmembrane (IFITM) gene families. While several ISGs were upregulated in each recurrent cancer, IFIT2 was significantly upregulated in all recurrent tumors when compared with the matched pre-RT specimens. Based on these observations, we hypothesized sustained type I IFN signaling through ISGs, such as IFIT2, may suppress the intra-tumoral immune response thereby promoting radiation resistance.
Collapse
Affiliation(s)
- Joseph Zenga
- Department of Otolaryngology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Musaddiq J Awan
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Anne Frei
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Becky Massey
- Department of Otolaryngology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jennifer Bruening
- Department of Otolaryngology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Monica Shukla
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Guru Prasad Sharma
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Aditya Shreenivas
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Stuart J Wong
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael T Zimmermann
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Angela J Mathison
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Heather A Himburg
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
32
|
Peng BL, Ran T, Chen X, Ding JC, Wang ZR, Li WJ, Liu W. A CARM1 Inhibitor Potently Suppresses Breast Cancer Both In Vitro and In Vivo. J Med Chem 2024; 67:7921-7934. [PMID: 38713486 PMCID: PMC11129188 DOI: 10.1021/acs.jmedchem.3c02315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/22/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024]
Abstract
CARM1, belonging to the protein arginine methyltransferase (PRMT) family, is intricately associated with the progression of cancer and is viewed as a promising target for both cancer diagnosis and therapy. However, the number of specific and potent CARM1 inhibitors is limited. We herein discovered a CARM1 inhibitor, iCARM1, that showed better specificity and activity toward CARM1 compared to the known CARM1 inhibitors, EZM2302 and TP-064. Similar to CARM1 knockdown, iCARM1 suppressed the expression of oncogenic estrogen/ERα-target genes, whereas activated type I interferon (IFN) and IFN-induced genes (ISGs) in breast cancer cells. Consequently, iCARM1 potently suppressed breast cancer cell growth both in vitro and in vivo. The combination of iCARM1 with either endocrine therapy drugs or etoposide demonstrated synergistic effects in inhibiting the growth of breast tumors. In summary, targeting CARM1 by iCARM1 effectively suppresses breast tumor growth, offering a promising therapeutic approach for managing breast cancers in clinical settings.
Collapse
Affiliation(s)
- Bing-ling Peng
- Fujian
Provincial Key Laboratory of Innovative Drug Target Research, School
of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
- State
Key Laboratory of Cellular Stress Biology, School of Pharmaceutical
Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
- Xiang
An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty
of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
| | - Ting Ran
- Bioland
Laboratory (Guangzhou Regenerative Medicine and Health - Guangdong
Laboratory), KaiYuan
Road, Guangzhou, Guangdong 510530, China
| | - Xue Chen
- Fujian
Provincial Key Laboratory of Innovative Drug Target Research, School
of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
- State
Key Laboratory of Cellular Stress Biology, School of Pharmaceutical
Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
- Xiang
An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty
of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
| | - Jian-cheng Ding
- Fujian
Provincial Key Laboratory of Innovative Drug Target Research, School
of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
- State
Key Laboratory of Cellular Stress Biology, School of Pharmaceutical
Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
- Xiang
An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty
of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
| | - Zi-rui Wang
- Fujian
Provincial Key Laboratory of Innovative Drug Target Research, School
of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
- State
Key Laboratory of Cellular Stress Biology, School of Pharmaceutical
Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
| | - Wen-juan Li
- Fujian
Provincial Key Laboratory of Innovative Drug Target Research, School
of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
- State
Key Laboratory of Cellular Stress Biology, School of Pharmaceutical
Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
- Xiang
An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty
of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
| | - Wen Liu
- Fujian
Provincial Key Laboratory of Innovative Drug Target Research, School
of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
- State
Key Laboratory of Cellular Stress Biology, School of Pharmaceutical
Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
- Xiang
An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty
of Medicine and Life Sciences, Xiamen University, Xiang’an South Road, Xiamen, Fujian 361102, China
| |
Collapse
|
33
|
Arimoto KI, Miyauchi S, Liu M, Zhang DE. Emerging role of immunogenic cell death in cancer immunotherapy. Front Immunol 2024; 15:1390263. [PMID: 38799433 PMCID: PMC11116615 DOI: 10.3389/fimmu.2024.1390263] [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: 02/23/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Cancer immunotherapy, such as immune checkpoint blockade (ICB), has emerged as a groundbreaking approach for effective cancer treatment. Despite its considerable potential, clinical studies have indicated that the current response rate to cancer immunotherapy is suboptimal, primarily attributed to low immunogenicity in certain types of malignant tumors. Immunogenic cell death (ICD) represents a form of regulated cell death (RCD) capable of enhancing tumor immunogenicity and activating tumor-specific innate and adaptive immune responses in immunocompetent hosts. Therefore, gaining a deeper understanding of ICD and its evolution is crucial for developing more effective cancer therapeutic strategies. This review focuses exclusively on both historical and recent discoveries related to ICD modes and their mechanistic insights, particularly within the context of cancer immunotherapy. Our recent findings are also highlighted, revealing a mode of ICD induction facilitated by atypical interferon (IFN)-stimulated genes (ISGs), including polo-like kinase 2 (PLK2), during hyperactive type I IFN signaling. The review concludes by discussing the therapeutic potential of ICD, with special attention to its relevance in both preclinical and clinical settings within the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Kei-ichiro Arimoto
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Sayuri Miyauchi
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Mengdan Liu
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
- School of Biological Sciences, University of California San Diego, La Jolla, CA, United States
| | - Dong-Er Zhang
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
- School of Biological Sciences, University of California San Diego, La Jolla, CA, United States
- Department of Pathology, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
34
|
Felchle H, Gissibl J, Lansink Rotgerink L, Nefzger SM, Walther CN, Timnik VR, Combs SE, Fischer JC. Influence of intestinal microbial metabolites on the abscopal effect after radiation therapy combined with immune checkpoint inhibitors. Clin Transl Radiat Oncol 2024; 46:100758. [PMID: 38500667 PMCID: PMC10945164 DOI: 10.1016/j.ctro.2024.100758] [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: 09/04/2023] [Revised: 02/16/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024] Open
Abstract
Background Most clinical studies failed to elicit a strong antitumor immune response and subsequent systemic tumor regression after radiation therapy (RT), even in combination with the immune checkpoint inhibitors (ICI) anti-CTLA4 or anti-PD1. Mechanistically, type I interferon (IFN-I) activation is essential for the development of such abscopal effects (AE); however, mechanisms driving or limiting IFN-I activation are ill defined. Groundbreaking discoveries have shown that antibiotics (ABx) can affect oncological outcomes and that microbiota-derived metabolites can modulate systemic antitumor immunity. Recent studies have demonstrated that the bacterial metabolites desaminotyrosine (DAT) and indole-3-carboxaldehyde (ICA) can enhance IFN-I activation in models of inflammatory diseases. Materials and Methods The subcutaneous bilateral MC38 tumor model is a widely used experimental tool to study the AE in mice. We applied it to explore the influence of broad-spectrum ABx, DAT and ICA on the AE after radioimmunotherapy (RIT). We performed 1x8 Gy of the primary tumor ± anti-CTLA4 or anti-PD1, and ± daily oral application of ABx or metabolites. Result Combinatory ABx had neither a significant effect on tumor growth of the irradiated tumor nor on tumor progression of the abscopal tumor after RIT with anti-CTLA4. Furthermore, DAT and ICA did not significantly impact on the AE after RIT with anti-CTLA4 or anti-PD1. Surprisingly, ICA even appears to reduce outcomes after RIT with anti-CTLA4. Conclusion We did not find a significant impact of combinatory ABx on the AE. Experimental application of the IFN-I-inducing metabolites DAT or ICA did not boost the AE after combined RIT. Additional studies are important to further investigate whether the intestinal microbiota or specific microbiota-derived metabolites modulate the AE.
Collapse
Affiliation(s)
- Hannah Felchle
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Julia Gissibl
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Laura Lansink Rotgerink
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Sophie M. Nefzger
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Caroline N. Walther
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Vincent R. Timnik
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Stephanie E. Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz Zentrum München, Institute of Radiation Medicine, 85764 Neuherberg, Germany
| | - Julius C. Fischer
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
35
|
Bower JE, Ganz PA, Irwin MR, Crespi CM, Petersen L, Asher A, Hurvitz SA, Cole SW. Type I interferons, inflammation, and fatigue in a longitudinal RNA study of women with breast cancer. Brain Behav Immun 2024; 118:312-317. [PMID: 38325563 PMCID: PMC11095951 DOI: 10.1016/j.bbi.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/12/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Fatigue is a common side effect of cancer and its treatment and is thought to be driven in part by activation of the proinflammatory cytokine network. However, the cellular and molecular underpinnings of cancer-related fatigue (CRF) have not been determined, nor have immune pathways beyond inflammation been carefully investigated. The goal of this study was to examine the association between CRF and activation of canonical proinflammatory gene regulation pathways and Type I interferon (IFN) signaling pathways in breast cancer patients during and after treatment. METHODS Women diagnosed with early-stage breast cancer (n = 181) completed assessments before and after treatment with radiation and/or chemotherapy and at 6, 12, and 18-month post-treatment follow-ups. Assessments included self-reported fatigue (Multidimensional Fatigue Symptom Inventory - Short Form) and expression of pre-specified sets of Type I IFN and pro-inflammatory immune response genes determined from mRNA sequencing of PBMCs. Mixed effect linear models examined changes in fatigue and immune gene expression over time and tested the hypothesis that fatigue would be associated with increased expression of Type I IFN and inflammatory response genes. RESULTS There were significant changes in fatigue and immune gene expression across the assessment period; all measures increased from pre- to post-treatment but showed diverging patterns over the follow-up, with declines in fatigue and persistent elevations in Type I IFN and proinflammatory gene expression. In mixed effect linear models, expression of Type I IFN response genes was elevated in association with fatigue across the assessment period, from pre-treatment to 18-month follow-up. In contrast, pro-inflammatory gene expression was associated with fatigue only at 6, 12, and 18-month follow-ups. Analyses controlling for changes in leukocyte subsets continued to show a significant association between fatigue and Type I IFN gene expression but reduced the time-dependent association with pro-inflammatory gene expression to non-significant. CONCLUSIONS Results revealed unexpected complexity in the immune underpinnings of CRF and identify a novel role for IFN signaling as a robust contributor to this symptom before, during, and after treatment. Pro-inflammatory gene expression emerged as a predictor of fatigue later in the cancer trajectory, and that effect was primarily accounted for by a concurrent increase in monocyte prevalence.
Collapse
Affiliation(s)
- Julienne E Bower
- UCLA Department of Psychology, United States; UCLA Department of Psychiatry and Biobehavioral Sciences, United States; Cousins Center for Psychoneuroimmunology, United States; Jonsson Comprehensive Cancer Center, United States.
| | - Patricia A Ganz
- Jonsson Comprehensive Cancer Center, United States; UCLA Schools of Medicine and Public Health, United States
| | - Michael R Irwin
- UCLA Department of Psychiatry and Biobehavioral Sciences, United States; Cousins Center for Psychoneuroimmunology, United States
| | - Catherine M Crespi
- Jonsson Comprehensive Cancer Center, United States; UCLA Department of Biostatistics, United States
| | | | - Arash Asher
- Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, United States
| | - Sara A Hurvitz
- University of Washington Department of Medicine, United States; Fred Hutchinson Cancer Center, United States
| | - Steve W Cole
- UCLA Department of Psychiatry and Biobehavioral Sciences, United States; Cousins Center for Psychoneuroimmunology, United States; UCLA Department of Medicine, United States
| |
Collapse
|
36
|
Bergerud KMB, Berkseth M, Pardoll DM, Ganguly S, Kleinberg LR, Lawrence J, Odde DJ, Largaespada DA, Terezakis SA, Sloan L. Radiation Therapy and Myeloid-Derived Suppressor Cells: Breaking Down Their Cancerous Partnership. Int J Radiat Oncol Biol Phys 2024; 119:42-55. [PMID: 38042450 PMCID: PMC11082936 DOI: 10.1016/j.ijrobp.2023.11.050] [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: 05/16/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
Radiation therapy (RT) has been a primary treatment modality in cancer for decades. Increasing evidence suggests that RT can induce an immunosuppressive shift via upregulation of cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs). MDSCs inhibit antitumor immunity through potent immunosuppressive mechanisms and have the potential to be crucial tools for cancer prognosis and treatment. MDSCs interact with many different pathways, desensitizing tumor tissue and interacting with tumor cells to promote therapeutic resistance. Vascular damage induced by RT triggers an inflammatory signaling cascade and potentiates hypoxia in the tumor microenvironment (TME). RT can also drastically modify cytokine and chemokine signaling in the TME to promote the accumulation of MDSCs. RT activation of the cGAS-STING cytosolic DNA sensing pathway recruits MDSCs through a CCR2-mediated mechanism, inhibiting the production of type 1 interferons and hampering antitumor activity and immune surveillance in the TME. The upregulation of hypoxia-inducible factor-1 and vascular endothelial growth factor mobilizes MDSCs to the TME. After recruitment, MDSCs promote immunosuppression by releasing reactive oxygen species and upregulating nitric oxide production through inducible nitric oxide synthase expression to inhibit cytotoxic activity. Overexpression of arginase-1 on subsets of MDSCs degrades L-arginine and downregulates CD3ζ, inhibiting T-cell receptor reactivity. This review explains how radiation promotes tumor resistance through activation of immunosuppressive MDSCs in the TME and discusses current research targeting MDSCs, which could serve as a promising clinical treatment strategy in the future.
Collapse
Affiliation(s)
| | - Matthew Berkseth
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sudipto Ganguly
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lawrence R Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, Minnesota
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - David A Largaespada
- Departments of Pediatrics and Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota
| | | | - Lindsey Sloan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
37
|
Tong J, Song J, Zhang W, Zhai J, Guan Q, Wang H, Liu G, Zheng C. When DNA-damage responses meet innate and adaptive immunity. Cell Mol Life Sci 2024; 81:185. [PMID: 38630271 PMCID: PMC11023972 DOI: 10.1007/s00018-024-05214-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024]
Abstract
When cells proliferate, stress on DNA replication or exposure to endogenous or external insults frequently results in DNA damage. DNA-Damage Response (DDR) networks are complex signaling pathways used by multicellular organisms to prevent DNA damage. Depending on the type of broken DNA, the various pathways, Base-Excision Repair (BER), Nucleotide Excision Repair (NER), Mismatch Repair (MMR), Homologous Recombination (HR), Non-Homologous End-Joining (NHEJ), Interstrand Crosslink (ICL) repair, and other direct repair pathways, can be activated separately or in combination to repair DNA damage. To preserve homeostasis, innate and adaptive immune responses are effective defenses against endogenous mutation or invasion by external pathogens. It is interesting to note that new research keeps showing how closely DDR components and the immune system are related. DDR and immunological response are linked by immune effectors such as the cyclic GMP-AMP synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway. These effectors act as sensors of DNA damage-caused immune response. Furthermore, DDR components themselves function in immune responses to trigger the generation of inflammatory cytokines in a cascade or even trigger programmed cell death. Defective DDR components are known to disrupt genomic stability and compromise immunological responses, aggravating immune imbalance and leading to serious diseases such as cancer and autoimmune disorders. This study examines the most recent developments in the interaction between DDR elements and immunological responses. The DDR network's immune modulators' dual roles may offer new perspectives on treating infectious disorders linked to DNA damage, including cancer, and on the development of target immunotherapy.
Collapse
Affiliation(s)
- Jie Tong
- College of Life Science, Hebei University, Baoding, 071002, China
- Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Jiangwei Song
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100089, China
| | - Wuchao Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, 071000, China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, 028000, China
| | - Qingli Guan
- The Affiliated Hospital of Chinese PLA 80th Group Army, Weifang, 261000, China
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Gentao Liu
- Department of Oncology, Tenth People's Hospital Affiliated to Tongji University & Cancer Center, Tongji University School of Medicine, Shanghai, 20000, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
38
|
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
|
39
|
Schmid M, Fischer P, Engl M, Widder J, Kerschbaum-Gruber S, Slade D. The interplay between autophagy and cGAS-STING signaling and its implications for cancer. Front Immunol 2024; 15:1356369. [PMID: 38660307 PMCID: PMC11039819 DOI: 10.3389/fimmu.2024.1356369] [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: 12/15/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Autophagy is an intracellular process that targets various cargos for degradation, including members of the cGAS-STING signaling cascade. cGAS-STING senses cytosolic double-stranded DNA and triggers an innate immune response through type I interferons. Emerging evidence suggests that autophagy plays a crucial role in regulating and fine-tuning cGAS-STING signaling. Reciprocally, cGAS-STING pathway members can actively induce canonical as well as various non-canonical forms of autophagy, establishing a regulatory network of feedback mechanisms that alter both the cGAS-STING and the autophagic pathway. The crosstalk between autophagy and the cGAS-STING pathway impacts a wide variety of cellular processes such as protection against pathogenic infections as well as signaling in neurodegenerative disease, autoinflammatory disease and cancer. Here we provide a comprehensive overview of the mechanisms involved in autophagy and cGAS-STING signaling, with a specific focus on the interactions between the two pathways and their importance for cancer.
Collapse
Affiliation(s)
- Maximilian Schmid
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- MedAustron Ion Therapy Center, Wiener Neustadt, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Labs, Vienna Biocenter, Vienna, Austria
| | - Patrick Fischer
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- MedAustron Ion Therapy Center, Wiener Neustadt, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Labs, Vienna Biocenter, Vienna, Austria
| | - Magdalena Engl
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Labs, Vienna Biocenter, Vienna, Austria
- Vienna Biocenter PhD Program, a Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Joachim Widder
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Sylvia Kerschbaum-Gruber
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- MedAustron Ion Therapy Center, Wiener Neustadt, Austria
| | - Dea Slade
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- MedAustron Ion Therapy Center, Wiener Neustadt, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Labs, Vienna Biocenter, Vienna, Austria
| |
Collapse
|
40
|
Yu T, Jiang W, Wang Y, Zhou Y, Jiao J, Wu M. Chimeric antigen receptor T cells in the treatment of osteosarcoma (Review). Int J Oncol 2024; 64:40. [PMID: 38390935 PMCID: PMC10919759 DOI: 10.3892/ijo.2024.5628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
Osteosarcoma (OS) is a frequently occurring primary bone tumor, mostly affecting children, adolescents and young adults. Before 1970, surgical resection was the main treatment method for OS, but the clinical results were not promising. Subsequently, the advent of chemotherapy has improved the prognosis of patients with OS. However, there is still a high incidence of metastasis or recurrence, and chemotherapy has several side effects, thus making the 5‑year survival rate markedly low. Recently, chimeric antigen receptor T (CAR‑T) cell therapy represents an alternative immunotherapy approach with significant potential for hematologic malignancies. Nevertheless, the application of CAR‑T cells in the treatment of OS faces numerous challenges. The present review focused on the advances in the development of CAR‑T cells to improve their clinical efficacy, and discussed ways to overcome the difficulties faced by CAR T‑cell therapy for OS.
Collapse
Affiliation(s)
- Tong Yu
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Weibo Jiang
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yang Wang
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Ying Zhou
- Department of Operating Room, The Third Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, P.R. China
| | - Jianhang Jiao
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Minfei Wu
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
41
|
Su C, Kim SK, Wang CX, Kirsch DG, Monjazeb AM. Radiotherapy Combined with Intralesional Immunostimulatory Agents for Soft Tissue Sarcomas. Semin Radiat Oncol 2024; 34:243-257. [PMID: 38508788 PMCID: PMC11216412 DOI: 10.1016/j.semradonc.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Immunotherapy has shifted the treatment paradigm for many types of cancer. Unfortunately, the most commonly used immunotherapies, such as immune checkpoint inhibitors (ICI), have yielded limited benefit for most types of soft tissue sarcoma (STS). Radiotherapy (RT) is a mainstay of sarcoma therapy and can induce immune modulatory effects. Combining immunotherapy and RT in STS may be a promising strategy to improve sarcoma response to RT and increase the efficacy of immunotherapy. Most combination strategies have employed immunotherapies, such as ICI, that derepress immune suppressive networks. These have yielded only modest results, possibly due to the limited immune stimulatory effects of RT. Combining RT with immune stimulatory agents has yielded promising preclinical and clinical results but can be limited by the toxic nature of systemic administration of immune stimulants. Using intralesional immune stimulants may generate stronger RT immune modulation and less systemic toxicity, which may be a feasible strategy in accessible tumors such as STS. In this review, we summarize the immune modulatory effects of RT, the mechanism of action of various immune stimulants, including toll-like receptor agonists, and data for combinatorial strategies utilizing these agents.
Collapse
Affiliation(s)
- Chang Su
- Department of Radiation Oncology, Duke University, Durham, NC
| | - Soo Kyoung Kim
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, UC Davis Health, Davis, CA
| | - Charles X Wang
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, UC Davis Health, Davis, CA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University, Durham, NC; Department of Radiation Oncology, Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Arta M Monjazeb
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, UC Davis Health, Davis, CA.
| |
Collapse
|
42
|
Li G, Zhao X, Zheng Z, Zhang H, Wu Y, Shen Y, Chen Q. cGAS-STING pathway mediates activation of dendritic cell sensing of immunogenic tumors. Cell Mol Life Sci 2024; 81:149. [PMID: 38512518 PMCID: PMC10957617 DOI: 10.1007/s00018-024-05191-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/09/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
Type I interferons (IFN-I) play pivotal roles in tumor therapy for three decades, underscoring the critical importance of maintaining the integrity of the IFN-1 signaling pathway in radiotherapy, chemotherapy, targeted therapy, and immunotherapy. However, the specific mechanism by which IFN-I contributes to these therapies, particularly in terms of activating dendritic cells (DCs), remains unclear. Based on recent studies, aberrant DNA in the cytoplasm activates the cyclic GMP-AMP synthase (cGAS)- stimulator of interferon genes (STING) signaling pathway, which in turn produces IFN-I, which is essential for antiviral and anticancer immunity. Notably, STING can also enhance anticancer immunity by promoting autophagy, inflammation, and glycolysis in an IFN-I-independent manner. These research advancements contribute to our comprehension of the distinctions between IFN-I drugs and STING agonists in the context of oncology therapy and shed light on the challenges involved in developing STING agonist drugs. Thus, we aimed to summarize the novel mechanisms underlying cGAS-STING-IFN-I signal activation in DC-mediated antigen presentation and its role in the cancer immune cycle in this review.
Collapse
Affiliation(s)
- Guohao Li
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Xiangqian Zhao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Zuda Zheng
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Hucheng Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Yundi Wu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Yangkun Shen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China.
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China.
| |
Collapse
|
43
|
Mecca M, Picerno S, Cortellino S. The Killer's Web: Interconnection between Inflammation, Epigenetics and Nutrition in Cancer. Int J Mol Sci 2024; 25:2750. [PMID: 38473997 DOI: 10.3390/ijms25052750] [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: 12/20/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Inflammation is a key contributor to both the initiation and progression of tumors, and it can be triggered by genetic instability within tumors, as well as by lifestyle and dietary factors. The inflammatory response plays a critical role in the genetic and epigenetic reprogramming of tumor cells, as well as in the cells that comprise the tumor microenvironment. Cells in the microenvironment acquire a phenotype that promotes immune evasion, progression, and metastasis. We will review the mechanisms and pathways involved in the interaction between tumors, inflammation, and nutrition, the limitations of current therapies, and discuss potential future therapeutic approaches.
Collapse
Affiliation(s)
- Marisabel Mecca
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, PZ, Italy
| | - Simona Picerno
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, PZ, Italy
| | - Salvatore Cortellino
- Laboratory of Preclinical and Translational Research, Responsible Research Hospital, 86100 Campobasso, CB, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, 80138 Naples, NA, Italy
- S.H.R.O. Italia Foundation ETS, 10060 Candiolo, TO, Italy
| |
Collapse
|
44
|
Gunturu DR, Hassan M, Bedi D, Datta P, Manne U, Samuel T. Unlocking the Potential of Therapy-Induced Cytokine Responses: Illuminating New Pathways in Cancer Precision Medicine. Curr Oncol 2024; 31:1195-1206. [PMID: 38534922 PMCID: PMC10968790 DOI: 10.3390/curroncol31030089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 05/26/2024] Open
Abstract
Precision cancer medicine primarily aims to identify individual patient genomic variations and exploit vulnerabilities in cancer cells to select suitable patients for specific drugs. These genomic features are commonly determined by gene sequencing prior to therapy, to identify individuals who would be most responsive. This precision approach in cancer therapeutics remains a powerful tool that benefits a smaller pool of patients, sparing others from unnecessary treatments. A limitation of this approach is that proteins, not genes, are the ultimate effectors of biological functions, and therefore the targets of therapeutics. An additional dimension in precision medicine that considers an individual's cytokine response to cancer therapeutics is proposed. Cytokine responses to therapy are multifactorial and vary among individuals. Thus, precision is dictated by the nature and magnitude of cytokine responses in the tumor microenvironment exposed to therapy. This review highlights cytokine responses as modules for precision medicine in cancer therapy, including potential challenges. For solid tumors, both detectability of cytokines in tissue fluids and their being amenable to routine sensitive analyses could address the difficulty of specimen collection for diagnosis and monitoring. Therefore, in precision cancer medicine, cytokines offer rational targets that can be utilized to enhance the efficacy of cancer therapy.
Collapse
Affiliation(s)
- Dilip R. Gunturu
- Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA;
| | - Mohammed Hassan
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA (T.S.)
| | - Deepa Bedi
- Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA;
| | - Pran Datta
- School of Medicine-Medicine-Hematology & Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Temesgen Samuel
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA (T.S.)
| |
Collapse
|
45
|
Zhang Q, Jiang L, Wang W, Huber AK, Valvo VM, Jungles KM, Holcomb EA, Pearson AN, The S, Wang Z, Parsels LA, Parsels JD, Wahl DR, Rao A, Sahai V, Lawrence TS, Green MD, Morgan MA. Potentiating the radiation-induced type I interferon antitumoral immune response by ATM inhibition in pancreatic cancer. JCI Insight 2024; 9:e168824. [PMID: 38376927 PMCID: PMC11063931 DOI: 10.1172/jci.insight.168824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/14/2024] [Indexed: 02/21/2024] Open
Abstract
Radiotherapy induces a type I interferon-mediated (T1IFN-mediated) antitumoral immune response that we hypothesized could be potentiated by a first-in-class ataxia telangiectasia mutated (ATM) inhibitor, leading to enhanced innate immune signaling, T1IFN expression, and sensitization to immunotherapy in pancreatic cancer. We evaluated the effects of AZD1390 or a structurally related compound, AZD0156, on innate immune signaling and found that both inhibitors enhanced radiation-induced T1IFN expression via the POLIII/RIG-I/MAVS pathway. In immunocompetent syngeneic mouse models of pancreatic cancer, ATM inhibitor enhanced radiation-induced antitumoral immune responses and sensitized tumors to anti-PD-L1, producing immunogenic memory and durable tumor control. Therapeutic responses were associated with increased intratumoral CD8+ T cell frequency and effector function. Tumor control was dependent on CD8+ T cells, as therapeutic efficacy was blunted in CD8+ T cell-depleted mice. Adaptive immune responses to combination therapy provided systemic control of contralateral tumors outside of the radiation field. Taken together, we show that a clinical candidate ATM inhibitor enhances radiation-induced T1IFN, leading to both innate and subsequent adaptive antitumoral immune responses and sensitization of otherwise resistant pancreatic cancer to immunotherapy.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Radiation Oncology and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | | | - Kassidy M. Jungles
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | - Stephanie The
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | | | | | | | - Daniel R. Wahl
- Department of Radiation Oncology and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Arvind Rao
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Vaibhav Sahai
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Division of Hematology and Oncology, Department of Internal Medicine, and
| | - Theodore S. Lawrence
- Department of Radiation Oncology and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael D. Green
- Department of Radiation Oncology and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Meredith A. Morgan
- Department of Radiation Oncology and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
46
|
Pannunzio S, Di Bello A, Occhipinti D, Scala A, Messina G, Valente G, Quirino M, Di Salvatore M, Tortora G, Cassano A. Multimodality treatment in recurrent/metastatic squamous cell carcinoma of head and neck: current therapy, challenges, and future perspectives. Front Oncol 2024; 13:1288695. [PMID: 38239635 PMCID: PMC10794486 DOI: 10.3389/fonc.2023.1288695] [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: 09/04/2023] [Accepted: 12/05/2023] [Indexed: 01/22/2024] Open
Abstract
Squamous cell carcinoma of the head and neck is a complex group of diseases that presents a challenge to the clinician. The prognosis in the recurrent/metastatic disease is particularly dismal, with a median survival of approximately 12 months. Recently, the personalized and multimodal approach has increased prognosis by integrating locoregional strategies (salvage surgery and stereotactic radiotherapy) and systemic treatments (chemotherapy, immunotherapy, and target therapy). Malnutrition is a significant clinical problem that interferes with dose intensity, and thus, feeding supplementation is critical not only to increase the quality of life but also to improve overall survival. With this review, we want to emphasize the importance of the multidisciplinary approach, quality of life, and nutritional supportive care and to integrate the latest updates of predictive biomarkers for immunotherapy and future therapeutic strategies.
Collapse
Affiliation(s)
- Sergio Pannunzio
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Armando Di Bello
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Denis Occhipinti
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Alessandro Scala
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Gloria Messina
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Giustina Valente
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Michela Quirino
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Mariantonietta Di Salvatore
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Giampaolo Tortora
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Alessandra Cassano
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| |
Collapse
|
47
|
Lote H, Chau I. Immunotherapy in Gastrointestinal Cancers. Cancer Treat Res 2024; 192:277-303. [PMID: 39212926 DOI: 10.1007/978-3-031-61238-1_14] [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: 09/04/2024]
Abstract
Immunotherapy has revolutionised cancer treatment over the past decade. Long-term durable responses can be achieved in some cancer patient populations that were previously facing terminal disease. In this chapter, we summarise current phase 3 clinical trial evidence for the use of immunotherapy in gastrointestinal cancers (oesophageal squamous cell carcinoma, oesophago-gastric adenocarcinoma, pancreatic cancer, biliary cancer, hepatocellular carcinoma, colorectal cancer, and squamous cell cancer of the anus). We discuss meaningful biomarkers used in clinical trials to select patients most likely to benefit from immunotherapy, such as mismatch-repair deficiency (MMRd)/microsatellite instability (MSI) and programmed-death-ligand-1 (PD-L1) immunohistochemistry (IHC) expression. Clinical questions are arising regarding the role of immunotherapy in the adjuvant/perioperative setting, optimal timing of surgery in patients who respond to immunotherapy, and toxicities specific to patients with gastrointestinal malignancies. We outline the current landscape and future horizon of immunotherapy in gastrointestinal cancers, such as strategies to increase effectiveness of checkpoint blockade through combinations with other checkpoint inhibitors, cytotoxic chemotherapy, targeted agents, radiotherapy, CAR-T therapy, and cancer vaccines.
Collapse
Affiliation(s)
- Hazel Lote
- The Royal Marsden Hospital NHS Foundation Trust, London and Sutton, UK
- Institute of Cancer Research, Sutton, UK
| | - Ian Chau
- The Royal Marsden Hospital NHS Foundation Trust, London and Sutton, UK.
| |
Collapse
|
48
|
Viswanath D, Park J, Misra R, Pizzuti VJ, Shin SH, Doh J, Won YY. Nanotechnology-enhanced radiotherapy and the abscopal effect: Current status and challenges of nanomaterial-based radio-immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1924. [PMID: 37632203 DOI: 10.1002/wnan.1924] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/03/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
Rare but consistent reports of abscopal remission in patients challenge the notion that radiotherapy (RT) is a local treatment; radiation-induced cancer cell death can trigger activation and recruitment of dendritic cells to the primary tumor site, which subsequently initiates systemic immune responses against metastatic lesions. Although this abscopal effect was initially considered an anomaly, combining RT with immune checkpoint inhibitor therapies has been shown to greatly improve the incidence of abscopal responses via modulation of the immunosuppressive tumor microenvironment. Preclinical studies have demonstrated that nanomaterials can further improve the reliability and potency of the abscopal effect for various different types of cancer by (1) altering the cell death process to be more immunogenic, (2) facilitating the capture and transfer of tumor antigens from the site of cancer cell death to antigen-presenting cells, and (3) co-delivering immune checkpoint inhibitors along with radio-enhancing agents. Several unanswered questions remain concerning the exact mechanisms of action for nanomaterial-enhanced RT and for its combination with immune checkpoint inhibition and other immunostimulatory treatments in clinically relevant settings. The purpose of this article is to summarize key recent developments in this field and also highlight knowledge gaps that exist in this field. An improved mechanistic understanding will be critical for clinical translation of nanomaterials for advanced radio-immunotherapy. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Dhushyanth Viswanath
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Jeehun Park
- SOFT Foundry Institute, Seoul National University, Seoul, Republic of Korea
| | - Rahul Misra
- Analytical Sciences, Sanofi, Toronto, Ontario, Canada
| | - Vincenzo J Pizzuti
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sung-Ho Shin
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Junsang Doh
- SOFT Foundry Institute, Seoul National University, Seoul, Republic of Korea
- Department of Materials Science and Engineering, Institute of Engineering Research, BioMAX, Seoul National University, Seoul, Republic of Korea
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana, USA
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
49
|
Galassi C, Klapp V, Yamazaki T, Galluzzi L. Molecular determinants of immunogenic cell death elicited by radiation therapy. Immunol Rev 2024; 321:20-32. [PMID: 37679959 PMCID: PMC11075037 DOI: 10.1111/imr.13271] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Cancer cells undergoing immunogenic cell death (ICD) can initiate adaptive immune responses against dead cell-associated antigens, provided that (1) said antigens are not perfectly covered by central tolerance (antigenicity), (2) cell death occurs along with the emission of immunostimulatory cytokines and damage-associated molecular patterns (DAMPs) that actively engage immune effector mechanisms (adjuvanticity), and (3) the microenvironment of dying cells is permissive for the initiation of adaptive immunity. Finally, ICD-driven immune responses can only operate and exert cytotoxic effector functions if the microenvironment of target cancer cells enables immune cell infiltration and activity. Multiple forms of radiation, including non-ionizing (ultraviolet) and ionizing radiation, elicit bona fide ICD as they increase both the antigenicity and adjuvanticity of dying cancer cells. Here, we review the molecular determinants of ICD as elicited by radiation as we critically discuss strategies to reinforce the immunogenicity of cancer cells succumbing to clinically available radiation strategies.
Collapse
Affiliation(s)
- Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Vanessa Klapp
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| |
Collapse
|
50
|
Lu Q, Yan W, Zhu A, Tubin S, Mourad WF, Yang J. Combining spatially fractionated radiation therapy (SFRT) and immunotherapy opens new rays of hope for enhancing therapeutic ratio. Clin Transl Radiat Oncol 2024; 44:100691. [PMID: 38033759 PMCID: PMC10684810 DOI: 10.1016/j.ctro.2023.100691] [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: 07/12/2023] [Revised: 10/04/2023] [Accepted: 10/15/2023] [Indexed: 12/02/2023] Open
Abstract
Spatially Fractionated Radiation Therapy (SFRT) is a form of radiotherapy that delivers a single large dose of radiation within the target volume in a heterogeneous pattern with regions of peak dosage and regions of under dosage. SFRT types can be defined by how the heterogeneous pattern of radiation is obtained. Immune checkpoint inhibitors (ICIs) have been approved for various malignant tumors and are widely used to treat patients with metastatic cancer. The efficacy of ICI monotherapy is limited due to the "cold" tumor microenvironment. Fractionated radiotherapy can achieve higher doses per fraction to the target tumor, and induce immune activation (immodulate tumor immunogenicity and microenvironment). Therefore, coupling ICI therapy and fractionated radiation therapy could significantly improve the outcome of metastatic cancer. This review focuses on both preclinical and clinical studies that use a combination of radiotherapy and ICI therapy in cancer.
Collapse
Affiliation(s)
- Qiuxia Lu
- Foshan Fosun Chancheng Hospital, P.R. China
- Junxin Precision Oncology Group, P.R. China
| | - Weisi Yan
- Baptist Health System, Lexington, KY, United States
- Junxin Precision Oncology Group, P.R. China
| | - Alan Zhu
- Mayo Clinic Alix School of Medicine, Scottsdale, AZ, United States
| | - Slavisa Tubin
- Albert Einstein Collage of Medicine New York, Center for Ion Therapy, Medaustron, Austria
| | - Waleed F. Mourad
- Department of Radiation Medicine Markey Cancer Center, University of Kentucky - College of Medicine, United States
| | - Jun Yang
- Foshan Fosun Chancheng Hospital, P.R. China
- Junxin Precision Oncology Group, P.R. China
| |
Collapse
|