1
|
Yang X, Wu X, Hao X, Li T, Guo H, Yang R. Unleashing the therapeutic potential of tumor-draining lymph nodes: spotlight on bladder cancer. J Transl Med 2025; 23:489. [PMID: 40301883 PMCID: PMC12042586 DOI: 10.1186/s12967-024-05864-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/07/2024] [Indexed: 05/01/2025] Open
Abstract
Tumor-draining lymph nodes (TDLNs) are often involved during the metastasis of bladder cancer (BC), which is associated with a poor prognosis. Recent studies have shown that TDLNs are a major source of host anti-tumor immunity, which can impede tumor progression and favor tumor immunotherapy. However, during tumor progression, various tumor-derived mediators modulate the TDLN microenvironment, impairing their protective function. Ultimately, TDLNs provide the soil for the proliferation and dissemination of tumor cells. Therefore, surgical removal of TDLNs is commonly recommended in various solid tumors to prevent metastasis, but this poses significant challenges for leveraging TDLNs in immunotherapy. Additionally, lymph node dissection (LND) has not shown survival benefits in some tumors. Hence, the decision to remove TDLNs in oncological treatment needs to be reconsidered. Herein, we spotlight the TDLNs of BC and introduce how BC cells modulate stromal cells and immune cells to shape an immunosuppressive TDLN microenvironment for BC progression. We summarize the existing therapeutic strategies to reinvigorate anti-tumor immunity in TDLNs. Furthermore, we discuss whether to preserve TDLNs and the role of LND during oncological treatment.
Collapse
Affiliation(s)
- Xin Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiangyu Wu
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuyang Hao
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tianhang Li
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China.
- Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Rong Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Yang L, Wei W, Yuan X, Guo E, Peng P, Wang J, Sun W. Targeting DNA Damage Repair to Enhance Antitumor Immunity in Radiotherapy: Mechanisms and Opportunities. Int J Mol Sci 2025; 26:3743. [PMID: 40332379 PMCID: PMC12027993 DOI: 10.3390/ijms26083743] [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: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/13/2025] [Indexed: 05/08/2025] Open
Abstract
Radiotherapy is a standard cancer treatment that involves the induction of DNA damage. DNA damage repair (DDR) pathways maintain genomic integrity and make tumors resistant to radiotherapy and certain chemotherapies. In turn, DDR dysfunction results in cumulative DNA damage, leading to increased sensitivity for antitumor treatment. Moreover, radiotherapy has been shown to trigger antitumor immunity. Currently, immunotherapy has become a new and widely used standard strategy for treating a broad spectrum of tumor types. Notably, recent studies have demonstrated that DDR pathways play important roles in driving the response to immunotherapy. Herein, we review and discuss how DDR affects antitumor immunity induced by radiotherapy. Furthermore, we summarize the development of strategies for combining DDR inhibitors with radiotherapy and/or immunotherapy to enhance their efficacy against cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (L.Y.); (W.W.); (X.Y.); (E.G.); (P.P.); (J.W.)
| |
Collapse
|
3
|
Liebman LN, Shen Y, Buchwald ZS, Nepiyushchikh Z, Qi Z, García AJ, Dixon JB. Lymphatic vessel network injury reduces local tumor control despite preservation of the tumor-draining lymph node. Sci Rep 2025; 15:3485. [PMID: 39875798 PMCID: PMC11775106 DOI: 10.1038/s41598-025-85670-3] [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: 01/16/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
The lymphatic system plays complex, often contradictory, roles in many cancers, including melanoma; these roles include contributions to tumor cell metastasis and immunosuppression in the tumor microenvironment as well as generation of antitumor immunity. Advancing our understanding of lymphatic vessel involvement in regulating tumor growth and immune response may provide new therapeutic targets or treatment plans to enhance the efficacy of existing therapies. We utilized a syngeneic murine melanoma model in which we surgically disrupted the lymphatic vessel network draining from the tumor to the tumor-draining lymph node (TDLN) while leaving the TDLN intact. Although transport of lymphatic-specific molecular weight tracers to the TDLN remains present after surgery, disruption of the tumor-draining lymphatic vessels results in decreased local tumor control, as reflected in an increase in the rate of tumor growth and reduction in effector-like T cell infiltration into the tumor. Our findings suggest that preservation of the functional tumor-draining lymphatic network may be essential in promoting a robust antitumor immune response.
Collapse
Affiliation(s)
- Lauren N Liebman
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Yang Shen
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Zachary S Buchwald
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Radiation Oncology, Emory University, Atlanta, GA, USA
| | - Zhanna Nepiyushchikh
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhiming Qi
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA, 30332, USA
| | - J Brandon Dixon
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA, 30332, USA.
| |
Collapse
|
4
|
Zheng J, Zheng Z, Zhang T, Chen X, Pang Q, Wang P, Yan C, Zhang W. Optimization of radiation target volume for locally advanced esophageal cancer in the immunotherapy era. Expert Opin Biol Ther 2024; 24:1221-1232. [PMID: 39460561 DOI: 10.1080/14712598.2024.2423009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 10/28/2024]
Abstract
INTRODUCTION Locally advanced esophageal cancer (EC) has poor prognosis. Preliminary clinical studies have demonstrated the synergistic efficacy of radiotherapy combined with immunotherapy in EC. Adjusting the radiotherapy target volume to protect immune function favors immunotherapy. However, there is no clear consensus on the exact definition of the EC target volume. AREAS COVERED Preclinical studies have provided a wealth of information on immunotherapy combined with different radiotherapy modalities, and several clinical studies have evaluated the impact of immunotherapy combined with radiotherapy on locally advanced EC. Here, we illustrate the rational target volume delineation for radiotherapy in terms of patient prognosis, pattern of radiotherapy failure, treatment-related toxicities, tumor-draining lymph nodes, and systemic immunity and summarize the clinical trials of radiotherapy combined with immunotherapy in EC. EXPERT OPINION We recommend applying involved-field irradiation (IFI) instead of elective nodal irradiation (ENI) for irradiated fields when immunotherapy is combined with chemoradiotherapy (CRT) for locally advanced EC. We expect that this target design will be evaluated in clinical trials to further explore more precise diagnostic modalities, long-term toxic responses, and quality of survival, and stratification factors for personalized treatment, and to provide more treatment benefits for patients.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhunhao Zheng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tian Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xi Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qingsong Pang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
5
|
Cai C, Zhang X, Sun X, Wang H, Chen E, Chen L, Gu B, Wang J, Huang X, Lao W, Wang X, Chen M, Ding S, Du J, Song Z. Node-sparing modified short-course Radiotherapy Combined with CAPOX and Tislelizumab for locally Advanced MSS of Middle and low rectal Cancer (mRCAT): an open-label, single-arm, prospective, multicentre clinical trial. BMC Cancer 2024; 24:1247. [PMID: 39385104 PMCID: PMC11463141 DOI: 10.1186/s12885-024-12994-0] [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: 09/08/2023] [Accepted: 09/25/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy followed by total mesorectal excision is a standard treatment for locally advanced rectal cancer. Mismatch repair-deficient locally advanced rectal cancer (LARC) was highly sensitive to PD-1 blockade. However, most rectal cancers are microsatellite stable (MSS) or mismatch repair-proficient (pMMR) subtypes for which PD-1 blockade is ineffective. Radiation can trigger the activation of CD8 + T cells, further enhancing the responses of MSS/pMMR rectal cancer to PD-1 blockade. Radioimmunotherapy offers a promising therapeutic modality for rectal cancer. Progenitor T exhausted cells are abundant in tumour-draining lymph nodes and play an important role in immunotherapy. Conventional irradiation fields include the mesorectum and regional lymph nodes, which might cause considerable damage to T lymphocytes and radiation-induced fibrosis, ultimately leading to a poor response to immunotherapy and rectal fibrosis. This study investigated whether node-sparing modified short-course irradiation combined with chemotherapy and PD-1 blockade could be effective in patients with MSS/ pMMR LARC. METHODS This was a open-label, single-arm, multicentre, prospective phase II trial. 32 LARC patients with MSS/pMMR will receive node-sparing modified short-course radiotherapy (the irradiated planned target volume only included the primary tumour bed but not the tumour-draining lymph nodes, 25 Gy/5f, 5 Gy/f) followed by CAPOX and tislelizumab. CAPOX and tislelizumab will be started two days after the completion of radiotherapy: oxaliplatin 130 mg/m2 intravenous infusion, day 1; capecitabine 1000 mg/m2 oral administration, days 1-14; and tislelizumab 200 mg, intravenous infusion, day 1. There will be four 21-day cycles. TME will be performed at weeks 14-15. We will collect blood, tumour, and lymphoid specimens; perform flow cytometry and in situ multiplexed immunofluorescence detection; and analyse the changes in various lymphocyte subsets. The primary endpoint is the rate of pathological complete response. The organ preservation rate, tumour regression grade, local recurrence rate, disease-free survival, overall survival, adverse effects, and quality of life will also be analysed. DISCUSSION In our research, node-sparing modified radiotherapy combined with immunotherapy probably increased the responsiveness of immunotherapy for MSS/pMMR rectal cancer patients, reduced the occurrence of postoperative rectal fibrosis, and improved survival and quality of life. This is the first clinical trial to utilize a node-sparing radiation strategy combined with chemotherapy and PD-1 blockade in the neoadjuvant treatment of rectal cancer, which may result in a breakthrough in the treatment of MSS/pMMR rectal cancer. TRIAL REGISTRATION This study was registered at www. CLINICALTRIALS gov . TRIAL REGISTRATION NUMBER NCT05972655. Date of registration: 31 July 2023.
Collapse
Affiliation(s)
- Cheng Cai
- Department of Colorectal and Anal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Zhang
- Department of Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiaonan Sun
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huogang Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Engeng Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Benxing Gu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianping Wang
- Department of Colorectal and Anal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xuefeng Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weifeng Lao
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaowei Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shubo Ding
- Department of Radiology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jinlin Du
- Department of Colorectal and Anal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China.
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China.
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
6
|
Wang X, Zhang H, XinZhang, Liu Y. Abscopal effect: from a rare phenomenon to a new frontier in cancer therapy. Biomark Res 2024; 12:98. [PMID: 39228005 PMCID: PMC11373306 DOI: 10.1186/s40364-024-00628-3] [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: 02/25/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Radiotherapy (RT) controls local lesions, meantime it has the capability to induce systemic response to inhibit distant, metastatic, non-radiated tumors, which is referred to as the "abscopal effect". It is widely recognized that radiotherapy can stimulate systemic immune response. This provides a compelling theoretical basis for the combination of immune therapy combined with radiotherapy(iRT). Indeed, this phenomenon has also been observed in clinical treatment, bringing significant clinical benefits to patients, and a series of basic studies are underway to amplify this effect. However, the molecular mechanisms of immune response induced by RT, determination of the optimal treatment regimen for iRT, and how to amplify the abscopal effect. In order to amplify and utilize this effect in clinical management, these key issues require to be well addressed; In this review, we comprehensively summarize the growing consensus and emphasize the emerging limitations of enhancing the abscopal effect with radiotherapy or immunotherapy. Finally, we discuss the prospects and barriers to the current clinical translational applications.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Haoyu Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - XinZhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| |
Collapse
|
7
|
Iwasaki T, Yazaki Y, Myojo T, Masuko T, Nishimura T. Complete Cure of Inoperable Stage IV Locally Advanced Hypopharyngeal Squamous Cell Carcinoma by an Innovative Combination Cancer Immunotherapy Consisting of Radiation, Immune Checkpoint Inhibitors, and Dendritic Cell Vaccine. Cureus 2024; 16:e69429. [PMID: 39411614 PMCID: PMC11474011 DOI: 10.7759/cureus.69429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2024] [Indexed: 10/19/2024] Open
Abstract
We report the case of a 68-year-old man with locally advanced (LA) head and neck cancer (HNC) (LA-HNC). The patient was diagnosed with inoperable stage IVA hypopharyngeal squamous cell carcinoma with 2 cm primary and three lymph node metastatic cancers. The patient was treated with an innovative combination cancer immunotherapy (iCCI) consisting of radiotherapy, immune checkpoint inhibitors, and helper/killer-hybrid epitope long peptides (H/K-HELP)-pulsed dendritic cell vaccine. These three treatments constituting iCCI are known to show an immunomodulating effect on tumor-draining lymph nodes (TDLNs) and improve antitumor immunity in tumor microenvironments (TMEs) to reduce tumor growth. Surprisingly, the patient treated with iCCI showed a complete cure for all the cancers including primary and lymph node-metastatic cancers without standard chemotherapy. The patient is still cancer-free for almost two years. Although the destruction mechanism of cancer is not determined, we speculate this iCCI might improve the patient's antitumor immune capability around tumor sites including TDLNs and TME. Our developed iCCI will become a promising strategy to overcome inoperable cancers in the future.
Collapse
Affiliation(s)
| | | | | | - Takashi Masuko
- Pharmacy, Kindai University, Higashiosaka, JPN
- Tumor and Gene Regulation, Oncology Innovation Center, Fujita Health University, Nagoya, JPN
| | - Takashi Nishimura
- Cancer Immunotherapy, Precision Clinic Group, Tokyo, JPN
- Tumor and Gene Regulation, Oncology Innovation Center, Fujita Health University, Nagoya, JPN
| |
Collapse
|
8
|
Ma L, Lan F, Chen P, Lei L, Zou T, Fu F, Wu R, Jin J, Zhang J. Loco-regional radiotherapy in de novo metastatic nasopharyngeal carcinoma with chemotherapy and immunotherapy: A real-world retrospective study from two cancer centers. Head Neck 2024; 46:1932-1937. [PMID: 38317293 DOI: 10.1002/hed.27665] [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: 12/01/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Immunochemotherapy has become the first-line treatment for initial diagnosed metastatic nasopharyngeal carcinoma (mNPC). Loco-regional radiotherapy combined with systemic chemotherapy significantly improves the survival. However, the safety and efficacy of loco-regional radiotherapy combined with immunochemotherapy remained unknown. METHODS Patients with de novo mNPC who received immunochemotherapy followed by loco-regional radiotherapy were included from two cancer centers. Toxicity and treatment response were assessed using CTCAE 5.0 and RECIST 1.1, respectively. Overall survival (OS) and progression-free survival (PFS) were analyzed using the Kaplan-Meier method. RESULTS From 2019 to 2021, a total of 16 patients were retrospectively analyzed. The median follow-up was 28 months (range 14-47 months). No one died. One-year, 2-year, and 3-year PFS rate was 93.8%, 58.4% and 50.1%, respectively. Radiotherapy-related acute severe (grade 3 or higher) toxicity was dermatitis (1/16, 6.3%) and mucositis (2/16, 12.5%). CONCLUSIONS Loco-regional radiotherapy provided a promising efficacy with modest toxicity for patients with mNPC who received immunochemotherapy.
Collapse
Affiliation(s)
- Li Ma
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Fengming Lan
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Peng Chen
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Ling Lei
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Teng Zou
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Fangmeng Fu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Runye Wu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Jin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianghu Zhang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Takeshima T, Hirayama R, Hasegawa S. Experimental evidence that carbon-ion radiotherapy utilizes cytotoxic T lymphocyte-mediated anti-tumor immunity for shrinking tumors compared to X-ray therapy. Biochem Biophys Res Commun 2024; 718:150058. [PMID: 38729076 DOI: 10.1016/j.bbrc.2024.150058] [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: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
The therapeutic efficacy of radiotherapy (RT) is primarily driven by two factors: biophysical DNA damage in cancer cells and radiation-induced anti-tumor immunity. However, Anti-tumor immune responses between X-ray RT (XRT) and carbon-ion RT (CIRT) remain unclear. In this study, we, employed mouse models to assess the immunological contribution, especially cytotoxic T-lymphocyte (CTL)-mediated immunity, to the therapeutic effectiveness of XRT and CIRT in shrinking tumors. We irradiated mouse intradermal tumors of B16F10-ovalbumin (OVA) mouse melanoma cells and 3LL-OVA mouse lung cancer cells with carbon-ion beams or X-rays in the presence or absence of CTLs. CTL removal was performed by administration of anti-CD8 monoclonal antibody (mAb) in mice. Based on tumor growth delay, we determined the tumor growth and regression curves. The enhancement ratio (ER) of the slope of regression lines in the presence of CTLs, relative to the absence of CTLs, indicates the dependency of RT on CTLs for shrinking mouse tumors, and the biological effectiveness (RBE) of CIRT relative to XRT were calculated. Tumor growth curves revealed that the elimination of CD8+ CTLs by administrating anti-CD8 mAb accelerated tumor growth compared to the presence of CTLs in both RTs. The ERs were larger in CIRT compared to XRT in the B16F10-OVA tumor models, but not in the 3LL-OVA models, suggesting a greater contribution of CTL-mediated anti-tumor immunity to tumor reduction in CIRT compared to XRT in the B16F10-OVA tumor model. In addition, the RBE values for both models were larger in the presence of CTLs compared to models without CTLs, suggesting that CIRT may utilize CTL-mediated anti-tumor immunity more than X-ray. The findings from this study suggest that although immunological contribution to therapeutic efficacy may vary depending on the type of tumor cell, CIRT utilizes CTL-mediated immunity to a greater extent compared to XRT.
Collapse
Affiliation(s)
- Tsuguhide Takeshima
- Department of Charged Particle Therapy Research, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Ryoichi Hirayama
- Department of Charged Particle Therapy Research, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Sumitaka Hasegawa
- Department of Charged Particle Therapy Research, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan.
| |
Collapse
|
10
|
Yang X, Liao Y, Fan L, Lin B, Li J, Wu D, Liao D, Yuan L, Liu J, Gao F, Feng G, Du X. High-intensity focused ultrasound ablation combined with immunotherapy for treating liver metastases: A prospective non-randomized trial. PLoS One 2024; 19:e0306595. [PMID: 38968190 PMCID: PMC11226133 DOI: 10.1371/journal.pone.0306595] [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: 01/27/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024] Open
Abstract
PURPOSE Given the unique features of the liver, it is necessary to combine immunotherapy with other therapies to improve its efficacy in patients of advanced cancer with liver metastases (LM). High-intensity focused ultrasound (HIFU) ablation is now widely used in clinical practice and can enhanced immune benefits. The study is intended to prospectively evaluate the safety and clinical feasibility of HIFU ablation in combination with systemic immunotherapy for patients with liver metastases. METHODS The study enrolled 14 patients with LM who received ultrasound-guided HIFU ablation combined with immune checkpoint inhibitors (ICIs) such as anti-programmed cell death protein 1 (anti-PD-1 agents manufactured in China) at Mianyang Central Hospital. Patients were followed up for adverse events (AEs) during the trial, using the CommonTerminology Criteria for Adverse Events v5.0(CTCAE v5.0) as the standard. Tumour response after treatment was assessed using computerized tomography. RESULTS The 14 patients (age range, 35-84 years) underwent HIFU ablation at 19 metastatic sites and systemic immunotherapy. The mean lesion volume was 179.9 cm3 (maximum: 733.1 cm3). Median follow-up for this trial was 9 months (range: 3-21) months. The study is clinically feasible and acceptable to patients. CONCLUSION This prospective study confirmed that HIFU combined with immunotherapy is clinically feasible and safe for treating liver metastases.
Collapse
Affiliation(s)
- Xiyue Yang
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Yao Liao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Lingli Fan
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Binwei Lin
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Jie Li
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Danfeng Wu
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Dongbiao Liao
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Li Yuan
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Jihui Liu
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Feng Gao
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Gang Feng
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Xiaobo Du
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Wu Y, Yi M, Niu M, Zhou B, Mei Q, Wu K. Beyond success: unveiling the hidden potential of radiotherapy and immunotherapy in solid tumors. Cancer Commun (Lond) 2024; 44:739-760. [PMID: 38837878 PMCID: PMC11260771 DOI: 10.1002/cac2.12576] [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: 12/12/2023] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
Immunotherapy, particularly with immune checkpoint inhibitors, has significantly transformed cancer treatment. Despite its success, many patients struggle to respond adequately or sustain long-lasting clinical improvement. A growing consensus has emerged that radiotherapy (RT) enhances the response rate and overall efficacy of immunotherapy. Although combining RT and immunotherapy has been extensively investigated in preclinical models and has shown promising results, establishing itself as a dynamic and thriving area of research, clinical evidence for this combination strategy over the past five years has shown both positive and disappointing results, suggesting the need for a more nuanced understanding. This review provides a balanced and updated analysis of the combination of immunotherapy and RT. We summarized the preclinical mechanisms through which RT boosts antitumor immune responses and mainly focused on the outcomes of recently updated clinical trials, including those that may not have met expectations. We investigated the optimization of the therapeutic potential of this combined strategy, including key challenges, such as fractionation and scheduling, lymph node irradiation, and toxicity. Finally, we offered insights into the prospects and challenges associated with the clinical translation of this combination therapy, providing a realistic perspective on the current state of research and potential future directions.
Collapse
Affiliation(s)
- Yuze Wu
- Department of OncologyTongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Ming Yi
- Department of Breast SurgeryZhejiang University School of Medicine First Affiliated HospitalHangzhouZhejiangP. R. China
| | - Mengke Niu
- Department of OncologyTongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Binghan Zhou
- Department of OncologyTongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Qi Mei
- Department of OncologyTongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Kongming Wu
- Cancer CenterShanxi Bethune HospitalShanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical UniversityTaiyuanShanxiP. R. China
- Cancer CenterTongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| |
Collapse
|
12
|
Telarovic I, Yong CSM, Kurz L, Vetrugno I, Reichl S, Fernandez AS, Cheng HW, Winkler R, Guckenberger M, Kipar A, Ludewig B, Pruschy M. Delayed tumor-draining lymph node irradiation preserves the efficacy of combined radiotherapy and immune checkpoint blockade in models of metastatic disease. Nat Commun 2024; 15:5500. [PMID: 38951172 PMCID: PMC11217506 DOI: 10.1038/s41467-024-49873-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 06/21/2024] [Indexed: 07/03/2024] Open
Abstract
Cancer resistance to immune checkpoint inhibitors motivated investigations into leveraging the immunostimulatory properties of radiotherapy to overcome immune evasion and to improve treatment response. However, clinical benefits of radiotherapy-immunotherapy combinations have been modest. Routine concomitant tumor-draining lymph node irradiation (DLN IR) might be the culprit. As crucial sites for generating anti-tumor immunity, DLNs are indispensable for the in situ vaccination effect of radiotherapy. Simultaneously, DLN sparing is often not feasible due to metastatic spread. Using murine models of metastatic disease in female mice, here we demonstrate that delayed (adjuvant), but not neoadjuvant, DLN IR overcomes the detrimental effect of concomitant DLN IR on the efficacy of radio-immunotherapy. Moreover, we identify IR-induced disruption of the CCR7-CCL19/CCL21 homing axis as a key mechanism for the detrimental effect of DLN IR. Our study proposes delayed DLN IR as a strategy to maximize the efficacy of radio-immunotherapy across different tumor types and disease stages.
Collapse
Affiliation(s)
- Irma Telarovic
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Carmen S M Yong
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lisa Kurz
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Irene Vetrugno
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sabrina Reichl
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alba Sanchez Fernandez
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Rona Winkler
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anja Kipar
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Martin Pruschy
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
13
|
Kramer G, Blair T, Bambina S, Kaur AP, Alice A, Baird J, Friedman D, Dowdell AK, Tomura M, Grassberger C, Piening BD, Crittenden MR, Gough MJ. Fluorescence tracking demonstrates T cell recirculation is transiently impaired by radiation therapy to the tumor. Sci Rep 2024; 14:11909. [PMID: 38789721 PMCID: PMC11126658 DOI: 10.1038/s41598-024-62871-w] [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: 12/04/2023] [Accepted: 05/22/2024] [Indexed: 05/26/2024] Open
Abstract
T cells recirculate through tissues and lymphatic organs to scan for their cognate antigen. Radiation therapy provides site-specific cytotoxicity to kill cancer cells but also has the potential to eliminate the tumor-specific T cells in field. To dynamically study the effect of radiation on CD8 T cell recirculation, we used the Kaede mouse model to photoconvert tumor-infiltrating cells and monitor their movement out of the field of radiation. We demonstrate that radiation results in loss of CD8 T cell recirculation from the tumor to the lymph node and to distant sites. Using scRNASeq, we see decreased proliferating CD8 T cells in the tumor following radiation therapy resulting in a proportional enrichment in exhausted phenotypes. By contrast, 5 days following radiation increased recirculation of T cells from the tumor to the tumor draining lymph node corresponds with increased immunosurveillance of the treated tumor. These data demonstrate that tumor radiation therapy transiently impairs systemic T cell recirculation from the treatment site to the draining lymph node and distant untreated tumors. This may inform timing therapies to improve systemic T cell-mediated tumor immunity.
Collapse
Affiliation(s)
- Gwen Kramer
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Tiffany Blair
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Shelly Bambina
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Aanchal Preet Kaur
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Alejandro Alice
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Jason Baird
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - David Friedman
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Alexa K Dowdell
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, 584-8540, Japan
| | - Clemens Grassberger
- Department of Radiation Oncology, University of Washington, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Brian D Piening
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA
- The Oregon Clinic, Portland, OR, 97213, USA
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA.
| |
Collapse
|
14
|
Yu B, Gao Y, Li J, Gao F, Zhang J, Li L, Feng X, Zuo D, Jin X, Chen W, Li Q. Killing two birds with one stone: Abscopal effect mechanism and its application prospect in radiotherapy. Crit Rev Oncol Hematol 2024; 196:104325. [PMID: 38462151 DOI: 10.1016/j.critrevonc.2024.104325] [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: 10/25/2023] [Revised: 02/07/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024] Open
Abstract
Abscopal effects are characterized by the emergence of neoplasms in regions unrelated to the primary radiation therapy site, displaying a gradual attenuation or regression throughout the progression of radiation therapy, which have been of interest to scientists since Mole's proposal in 1953. The incidence of abscopal effects in radiation therapy is intricately linked to the immune system, with both innate and adaptive immune responses playing crucial roles. Biological factors impacting abscopal effects ultimately exert their influence on the intricate workings of the immune system. Although abscopal effects are rarely observed in clinical cases, the underlying mechanism remains uncertain. This article examines the biological and physical factors influencing abscopal effects of radiotherapy. Through a review of preclinical and clinical studies, this article aims to offer a comprehensive understanding of abscopal effects and proposes new avenues for future research in this field. The findings presented in this article serve as a valuable reference for researchers seeking to explore this topic in greater depth.
Collapse
Affiliation(s)
- Boyi Yu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuting Gao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; College of Life Sciences, Northwest Normal University, Gansu Province, Lanzhou 730070, China
| | - Jiaxin Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feifei Gao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiahao Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; School of Public Health, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Linjing Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianglong Feng
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dashan Zuo
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
15
|
Sun L, Morikawa K, Sogo Y, Sugiura Y. MHY1485 potentiates immunogenic cell death induction and anti-cancer immunity following irradiation. JOURNAL OF RADIATION RESEARCH 2024; 65:205-214. [PMID: 38330507 PMCID: PMC10959436 DOI: 10.1093/jrr/rrad107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/17/2023] [Indexed: 02/10/2024]
Abstract
Recent in vitro experiments showed that combined treatment with MHY1485, a low-molecular-weight compound, and X-ray irradiation significantly increased apoptosis and senescence in tumor cells, which was associated with oxidative stress, endoplasmic reticulum (ER) stress and p21 stabilization, compared to radiation treatment alone. However, evidence for MHY1485 treatment-mediated suppression of tumor growth in animals is still lacking. Furthermore, it has been shown that ER stress enhances immunogenic cell death (ICD) in tumor cells, as it can exert a favorable influence on the anti-cancer immune system. In the present study, we examined whether co-treatment of MHY1485 and X-ray irradiation induces ICD and in vivo tumor growth suppression using the CT26 and Lewis lung carcinoma murine tumor cell lines. We found that MHY1485 + X-ray treatment promotes ICD more effectively than X-ray treatment alone. MHY1485 suppresses tumor growth in vivo under co-treatment with X-rays and increases INF-γ, tumor necrosis factor, interleukin-2 and interleukin-12 levels in the spleen as well as the presence of CD8+ cells in the tumor. The results suggest that MHY1485 treatment leads to the conversion of irradiated tumors into effective vaccines. Thus, MHY1485 is a promising lead compound for use in combination with radiotherapy.
Collapse
Affiliation(s)
- Lue Sun
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Kumi Morikawa
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Yu Sogo
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Yuki Sugiura
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14, Hayashi-cho, Takamatsu, Kagawa 761-0895, Japan
| |
Collapse
|
16
|
Frijlink E, Bosma DM, Busselaar J, Battaglia TW, Staal MD, Verbrugge I, Borst J. PD-1 or CTLA-4 blockade promotes CD86-driven Treg responses upon radiotherapy of lymphocyte-depleted cancer in mice. J Clin Invest 2024; 134:e171154. [PMID: 38349740 PMCID: PMC10940086 DOI: 10.1172/jci171154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 01/17/2024] [Indexed: 03/16/2024] Open
Abstract
Radiotherapy (RT) is considered immunogenic, but clinical data demonstrating RT-induced T cell priming are scarce. Here, we show in a mouse tumor model representative of human lymphocyte-depleted cancer that RT enhanced spontaneous priming of thymus-derived (FOXP3+Helios+) Tregs by the tumor. These Tregs acquired an effector phenotype, populated the tumor, and impeded tumor control by a simultaneous, RT-induced CD8+ cytotoxic T cell (CTL) response. Combination of RT with CTLA-4 or PD-1 blockade, which enables CD28 costimulation, further increased this Treg response and failed to improve tumor control. We discovered that upon RT, the CD28 ligands CD86 and CD80 differentially affected the Treg response. CD86, but not CD80, blockade prevented the effector Treg response, enriched the tumor-draining lymph node migratory conventional DCs that were positive for PD-L1 and CD80 (PD-L1+CD80+), and promoted CTL priming. Blockade of CD86 alone or in combination with PD-1 enhanced intratumoral CTL accumulation, and the combination significantly increased RT-induced tumor regression and OS. We advise that combining RT with PD-1 and/or CTLA-4 blockade may be counterproductive in lymphocyte-depleted cancers, since these interventions drive Treg responses in this context. However, combining RT with CD86 blockade may promote the control of such tumors by enabling a CTL response.
Collapse
Affiliation(s)
- Elselien Frijlink
- Division of Tumor Biology and Immunology and Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| | - Douwe M.T. Bosma
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| | - Julia Busselaar
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| | - Thomas W. Battaglia
- Division of Molecular Oncology and Immunology and Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Mo D. Staal
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| | - Inge Verbrugge
- Division of Tumor Biology and Immunology and Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jannie Borst
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
17
|
Verma N, Renauer PA, Dong C, Xin S, Lin Q, Zhang F, Glazer PM, Chen S. Genome scale CRISPR screens identify actin capping proteins as key modulators of therapeutic responses to radiation and immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.14.575614. [PMID: 38293095 PMCID: PMC10827061 DOI: 10.1101/2024.01.14.575614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Radiotherapy (RT), is a fundamental treatment for malignant tumors and is used in over half of cancer patients. As radiation can promote anti-tumor immune effects, a promising therapeutic strategy is to combine radiation with immune checkpoint inhibitors (ICIs). However, the genetic determinants that impact therapeutic response in the context of combination therapy with radiation and ICI have not been systematically investigated. To unbiasedly identify the tumor intrinsic genetic factors governing such responses, we perform a set of genome-scale CRISPR screens in melanoma cells for cancer survival in response to low-dose genotoxic radiation treatment, in the context of CD8 T cell co-culture and with anti-PD1 checkpoint blockade antibody. Two actin capping proteins, Capza3 and Capg, emerge as top hits that upon inactivation promote the survival of melanoma cells in such settings. Capza3 and Capg knockouts (KOs) in mouse and human cancer cells display persistent DNA damage due to impaired homology directed repair (HDR); along with increased radiation, chemotherapy, and DNA repair inhibitor sensitivity. However, when cancer cells with these genes inactivated were exposed to sublethal radiation, inactivation of such actin capping protein promotes activation of the STING pathway, induction of inhibitory CEACAM1 ligand expression and resistance to CD8 T cell killing. Patient cancer genomics analysis reveals an increased mutational burden in patients with inactivating mutations in CAPG and/or CAPZA3, at levels comparable to other HDR associated genes. There is also a positive correlation between CAPG expression and activation of immune related pathways and CD8 T cell tumor infiltration. Our results unveil the critical roles of actin binding proteins for efficient HDR within cancer cells and demonstrate a previously unrecognized regulatory mechanism of therapeutic response to radiation and immunotherapy.
Collapse
Affiliation(s)
- Nipun Verma
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut, USA
| | - Paul A. Renauer
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Chuanpeng Dong
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Shan Xin
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Qianqian Lin
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Feifei Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Peter M. Glazer
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut, USA
- Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Immunobiology Program, Yale University, New Haven, Connecticut, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, Connecticut, USA
- Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Biomedical Data Science, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
18
|
Swamy K. Therapeutic In Situ Cancer Vaccine Using Pulsed Stereotactic Body Radiotherapy-A Translational Model. Vaccines (Basel) 2023; 12:7. [PMID: 38276666 PMCID: PMC10819354 DOI: 10.3390/vaccines12010007] [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: 10/24/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Both radiation and cancer therapeutic vaccine research are more than 100 years old, and their potential is likely underexplored. Antiangiogenics, nanoparticle targeting, and immune modulators are some other established anticancer therapies. In the meantime, immunotherapy usage is gaining momentum in clinical applications. This article proposes the concept of a pulsed/intermittent/cyclical endothelial-sparing single-dose in situ vaccination (ISVRT) schedule distinguishable from the standard therapeutic stereotactic body radiotherapy (SBRT) and stereotactic radiosurgery (SRS) plans. This ISVRT schedule can repeatedly generate tumor-specific neoantigens and epitopes for primary and immune modulation effects, augment supplementary immune enhancement techniques, activate long-term memory cells, avoid extracellular matrix fibrosis, and essentially synchronize with the vascular normalized immunity cycle. The core mechanisms of ISVRT impacting in situ vaccination would be optimizing cascading antigenicity and adjuvanticity. The present proposed hypothesis can be validated using the algorithm presented. The indications for the proposed concept are locally progressing/metastatic cancers that have failed standard therapies. Immunotherapy/targeted therapy, chemotherapy, antiangiogenics, and vascular-lymphatic normalization are integral to such an approach.
Collapse
|
19
|
Badrigilan S, Meola A, Chang SD, Rezaeian S, Nemati H, Almasi T, Rostampour N. Stereotactic radiosurgery with immune checkpoint inhibitors for brain metastases: a meta-analysis study. Br J Neurosurg 2023; 37:1533-1543. [PMID: 34979828 DOI: 10.1080/02688697.2021.2022098] [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: 08/02/2021] [Accepted: 12/20/2021] [Indexed: 11/02/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are an emerging tool in the treatment of brain metastases (BMs), Stereotactic radiosurgery (SRS), traditionally used for BMs, elicits an immune brain response and can act synergistically with ICIs. We aim to investigate the efficacy of ICI administered with SRS and determine the impact of timing on BM response. METHODS A systematical search was performed to identify potential studies concerning BMs managed with SRS alone or with SRS + ICI with relative timing administration (ICI concurrent with SRS, ICI nonconcurrent with SRS, SRS before ICI, SRS after ICI). The overall survival (OS), 12-month OS, local progression-free survival (LPFS), 12-month local brain control (LBC), distant progression-free survival (DPFS), 12-month distant brain control (DBC), and adverse events (intracranial hemorrhage, radionecrosis) were analyzed using the random-effects model. RESULTS A total of 16 retrospective studies with 1356 BM patients were included. Compared to nonconcurrent therapy, concurrent therapy revealed a significantly longer OS (HR= 1.43; p = 0.008) and 12-months LBC (HR = 1.91; p = 0.04), a similar 12-months DBC (HR = 1.12; p = 0.547) and higher complication rate (R = 0.77; p = 0.346). Concurrent therapy leads to a significantly higher OS compared to ICI before SRS (HR = 2.55; p = 0.0003). CONCLUSION The combination of SRS with ICI improves patients' clinical and radiological outcomes. The effectiveness of the combination is subject to the identification of an optimal therapeutic window.
Collapse
Affiliation(s)
- Samireh Badrigilan
- Department of Medical Physics, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Antonio Meola
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven D Chang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Shahab Rezaeian
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hossein Nemati
- Department of Epidemiology, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Tinoosh Almasi
- Department of Medical Physics, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nima Rostampour
- Department of Medical Physics, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
20
|
SHIRATO H. Biomedical advances and future prospects of high-precision three-dimensional radiotherapy and four-dimensional radiotherapy. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2023; 99:389-426. [PMID: 37821390 PMCID: PMC10749389 DOI: 10.2183/pjab.99.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/13/2023] [Indexed: 10/13/2023]
Abstract
Biomedical advances of external-beam radiotherapy (EBRT) with improvements in physical accuracy are reviewed. High-precision (±1 mm) three-dimensional radiotherapy (3DRT) can utilize respective therapeutic open doors in the tumor control probability curve and in the normal tissue complication probability curve instead of the one single therapeutic window in two-dimensional EBRT. High-precision 3DRT achieved higher tumor control and probable survival rates for patients with small peripheral lung and liver cancers. Four-dimensional radiotherapy (4DRT), which can reduce uncertainties in 3DRT due to organ motion by real-time (every 0.1-1 s) tumor-tracking and immediate (0.1-1 s) irradiation, have achieved reduced adverse effects for prostate and pancreatic tumors near the digestive tract and with similar or better tumor control. Particle beam therapy improved tumor control and probable survival for patients with large liver tumors. The clinical outcomes of locally advanced or multiple tumors located near serial-type organs can theoretically be improved further by integrating the 4DRT concept with particle beams.
Collapse
Affiliation(s)
- Hiroki SHIRATO
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
21
|
Xin Z, Liu Q, Ai D, Chen K, Mariamidze E, Sumon MA, Devnani B, Pihlak R, Zhu H, Zhao K. Radiotherapy for Advanced Esophageal Cancer: from Palliation to Curation. Curr Treat Options Oncol 2023; 24:1568-1579. [PMID: 37812321 DOI: 10.1007/s11864-023-01134-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2023] [Indexed: 10/10/2023]
Abstract
OPINION STATEMENT Esophageal cancer is a global health problem, which is 7th most common and 6th most deadly cancer. It has been the era of immuno-oncology for esophageal cancer management. Radiation therapy has been one of the key local therapeutic approaches for esophageal cancer treatment, while its role in advanced disease is challenging and debatable. There have been emerging clinical and translational studies of radiation therapy in recurrent or metastatic esophageal cancer. Immunotherapy has been established the standard care of 1st and 2nd line systemic therapies of advanced esophageal cancer, and the development of tumor immunity has opened a new chapter for the esophageal cancer radiation therapy. The current review will summarize the classic radiation therapy research in advanced esophageal cancer, as well as the most recent key findings. The subtitles will cover palliative radiotherapy for dysphagia, re-radiation for recurrent disease, oligo-focal disease management and stereotactic radiation therapy, and radiotherapy with immunotherapy. Radiotherapy plays vital role in multidisciplinary management of advanced EC. External or intratumoral irradiation has been used for palliation of dysphagia and improving QOL in esophageal cancer patients traditionally, while recent clinical and technical advance enables radiotherapy to be considered in recurrent or metastatic disease for curation attention. Novel clinical and translational investigation is opening a new chapter of radiotherapy with immunotherapy for benefiting advanced EC patients.
Collapse
Affiliation(s)
- Zhuocheng Xin
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, 270 DongAn Road, Shanghai, 200032, China
| | - Qi Liu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, 270 DongAn Road, Shanghai, 200032, China
| | - Dashan Ai
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, 270 DongAn Road, Shanghai, 200032, China
| | - Ke Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Endoscopy, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Elene Mariamidze
- Oncology and Hematology Department, Research Institute of Clinical Medicine After Academician F. Todua, Tbilisi, Georgia
| | - Mostafa Aziz Sumon
- Department of Radiation Oncology, Kurmitola General Hospital, Dhaka, Bangladesh
| | - Bharti Devnani
- Department of Radiotherapy and Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Rille Pihlak
- Medical Oncology Department, St Bartholomew's Hospital, London, UK
| | - Hongcheng Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China.
- Shanghai Key Laboratory of Radiation Oncology, 270 DongAn Road, Shanghai, 200032, China.
| | - Kuaile Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China.
- Shanghai Key Laboratory of Radiation Oncology, 270 DongAn Road, Shanghai, 200032, China.
| |
Collapse
|
22
|
Masuda K, Nagai Y, Amari H, Tahara H, Maeda Y, Shiihara J, Ohta H, Hiruta M, Yamaguchi Y. Hyperprogressive disease in lung metastases without target lesion progression after durvalumab consolidation therapy: A case report. Thorac Cancer 2023; 14:3161-3165. [PMID: 37699791 PMCID: PMC10626253 DOI: 10.1111/1759-7714.15104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Hyperprogressive disease (HPD) is a novel progressive pattern that occurs after immune checkpoint inhibitor (ICI) administration. Here, a 74-year-old woman who had undergone right lower lobectomy for lung cancer received curative chemoradiotherapy followed by consolidation therapy with durvalumab for metastatic recurrence confined to the mediastinal lymph nodes. Three weeks later, multiple randomly distributed nodular shadows appeared on chest CT, and thoracoscopic lung biopsy led to the diagnosis of multiple pulmonary metastases. HPD may be suspected when multiple metastases appear in new organs early after the administration of ICIs. This phenomenon may occur not only with ICI monotherapy but also with the administration of ICIs after chemoradiotherapy. Therefore, patients who have received radiation therapy should also be given similar attention early after the administration of ICIs.
Collapse
Affiliation(s)
- Kosuke Masuda
- Division of Respiratory Medicine, Clinical Department of Internal MedicineJichi Medical University Saitama Medical CenterSaitamaJapan
| | - Yoshiaki Nagai
- Division of Respiratory Medicine, Clinical Department of Internal MedicineJichi Medical University Saitama Medical CenterSaitamaJapan
| | - Hikari Amari
- Division of Respiratory Medicine, Clinical Department of Internal MedicineJichi Medical University Saitama Medical CenterSaitamaJapan
| | - Hiroki Tahara
- Division of Respiratory Medicine, Clinical Department of Internal MedicineJichi Medical University Saitama Medical CenterSaitamaJapan
| | - Yuki Maeda
- Division of Respiratory Medicine, Clinical Department of Internal MedicineJichi Medical University Saitama Medical CenterSaitamaJapan
| | - Jun Shiihara
- Division of Respiratory Medicine, Clinical Department of Internal MedicineJichi Medical University Saitama Medical CenterSaitamaJapan
| | - Hiromitsu Ohta
- Division of Respiratory Medicine, Clinical Department of Internal MedicineJichi Medical University Saitama Medical CenterSaitamaJapan
| | - Masahiro Hiruta
- Department of PathologyJichi Medical University Saitama Medical CenterSaitamaJapan
| | - Yasuhiro Yamaguchi
- Division of Respiratory Medicine, Clinical Department of Internal MedicineJichi Medical University Saitama Medical CenterSaitamaJapan
| |
Collapse
|
23
|
Jalalpour Choupanan M, Shahbazi S, Reiisi S. Naringenin in combination with quercetin/fisetin shows synergistic anti-proliferative and migration reduction effects in breast cancer cell lines. Mol Biol Rep 2023; 50:7489-7500. [PMID: 37480513 DOI: 10.1007/s11033-023-08664-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/03/2023] [Indexed: 07/24/2023]
Abstract
INTRODUCTION & AIM Breast cancer is one of the most common cancers with a high mortality rate among women worldwide. Quercetin/fisetin and naringenin, three well-known flavonoids, have been used to fight against various cancers. The aim of the present study was to investigate the possible synergism of quercetin/fisetin with naringenin on MCF7 and MDA-MB-231 breast cancer cell lines. METHODS In this study, cultured MCF7 and MDA-MB-231 cells were treated with different concentrations of quercetin/fisetin individually and in combination with naringenin. MTT assay and scratch assay was employed to determine cell viability and migration respectively. Real-time PCR was used to study the expression level of apoptosis genes and miR-1275 (tumor suppressor miRNA) and mir-27a-3p (oncogenic miRNA). RESULTS A synergism effect of quercetin/fisetin and naringenin (CI < 1) was observed for both cell lines. Combination therapies were significantly more effective in cell growth reduction, migration suppression and apoptosis induction than single therapies. Gene expression analysis revealed the upregulation of miR-1275 and downregulation miR-27a-3p. CONCLUSION Our results indicate that quercetin/fisetin enhances the anti-proliferative and anti-migratory activities in combination with naringenin in MCF7 and MDA-MB-231 human breast cancer cell lines. Therefore, the combination of Que/Fis and Nar can be proposed as a promising therapeutic strategy for further investigations.
Collapse
Affiliation(s)
| | - Shahrzad Shahbazi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Somayeh Reiisi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran.
| |
Collapse
|
24
|
Sharon S, Daher-Ghanem N, Zaid D, Gough MJ, Kravchenko-Balasha N. The immunogenic radiation and new players in immunotherapy and targeted therapy for head and neck cancer. FRONTIERS IN ORAL HEALTH 2023; 4:1180869. [PMID: 37496754 PMCID: PMC10366623 DOI: 10.3389/froh.2023.1180869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023] Open
Abstract
Although treatment modalities for head and neck cancer have evolved considerably over the past decades, survival rates have plateaued. The treatment options remained limited to definitive surgery, surgery followed by fractionated radiotherapy with optional chemotherapy, and a definitive combination of fractionated radiotherapy and chemotherapy. Lately, immunotherapy has been introduced as the fourth modality of treatment, mainly administered as a single checkpoint inhibitor for recurrent or metastatic disease. While other regimens and combinations of immunotherapy and targeted therapy are being tested in clinical trials, adapting the appropriate regimens to patients and predicting their outcomes have yet to reach the clinical setting. Radiotherapy is mainly regarded as a means to target cancer cells while minimizing the unwanted peripheral effect. Radiotherapy regimens and fractionation are designed to serve this purpose, while the systemic effect of radiation on the immune response is rarely considered a factor while designing treatment. To bridge this gap, this review will highlight the effect of radiotherapy on the tumor microenvironment locally, and the immune response systemically. We will review the methodology to identify potential targets for therapy in the tumor microenvironment and the scientific basis for combining targeted therapy and radiotherapy. We will describe a current experience in preclinical models to test these combinations and propose how challenges in this realm may be faced. We will review new players in targeted therapy and their utilization to drive immunogenic response against head and neck cancer. We will outline the factors contributing to head and neck cancer heterogeneity and their effect on the response to radiotherapy. We will review in-silico methods to decipher intertumoral and intratumoral heterogeneity and how these algorithms can predict treatment outcomes. We propose that (a) the sequence of surgery, radiotherapy, chemotherapy, and targeted therapy should be designed not only to annul cancer directly, but to prime the immune response. (b) Fractionation of radiotherapy and the extent of the irradiated field should facilitate systemic immunity to develop. (c) New players in targeted therapy should be evaluated in translational studies toward clinical trials. (d) Head and neck cancer treatment should be personalized according to patients and tumor-specific factors.
Collapse
Affiliation(s)
- Shay Sharon
- Department of Oral and Maxillofacial Surgery, Hadassah Medical Center, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Oral and Maxillofacial Surgery, Boston University and Boston Medical Center, Boston, MA, United States
| | - Narmeen Daher-Ghanem
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Deema Zaid
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael J. Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
25
|
Wang L, Yang Z, Guo F, Chen Y, Wei J, Dai X, Zhang X. Research progress of biomarkers in the prediction of anti-PD-1/PD-L1 immunotherapeutic efficiency in lung cancer. Front Immunol 2023; 14:1227797. [PMID: 37465684 PMCID: PMC10351040 DOI: 10.3389/fimmu.2023.1227797] [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: 05/23/2023] [Accepted: 06/13/2023] [Indexed: 07/20/2023] Open
Abstract
Currently, anti-PD-1/PD-L1 immunotherapy using immune checkpoint inhibitors is widely used in the treatment of multiple cancer types including lung cancer, which is a leading cause of cancer death in the world. However, only a limited proportion of lung cancer patients will benefit from anti-PD-1/PD-L1 therapy. Therefore, it is of importance to predict the response to immunotherapy for the precision treatment of patients. Although the expression of PD-L1 and tumor mutation burden (TMB) are commonly used to predict the clinical response of anti-PD-1/PD-L1 therapy, other factors such as tumor-specific genes, dMMR/MSI, and gut microbiome are also promising predictors for immunotherapy in lung cancer. Furthermore, invasive peripheral blood biomarkers including blood DNA-related biomarkers (e.g., ctDNA and bTMB), blood cell-related biomarkers (e.g., immune cells and TCR), and other blood-related biomarkers (e.g., soluble PD-L1 and cytokines) were utilized to predict the immunotherapeutic response. In this review, the current achievements of anti-PD-1/PD-L1 therapy and the potential biomarkers for the prediction of anti-PD-1/PD-L1 immunotherapy in lung cancer treatment were summarized and discussed.
Collapse
Affiliation(s)
- Luyao Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Zongxing Yang
- Department of Clinical Laboratory, First Hospital of Jilin University, Changchun, China
| | - Fucheng Guo
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Yurong Chen
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Jiarui Wei
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Elbers JBW, Gunsch PA, Debets R, Keereweer S, van Meerten E, Zindler J, van Norden Y, Hoogeman MS, Verduijn GM, Kroesen M, Nout RA. HYpofractionated, dose-redistributed RAdiotherapy with protons and photons to combat radiation-induced immunosuppression in head and neck squamous cell carcinoma: study protocol of the phase I HYDRA trial. BMC Cancer 2023; 23:541. [PMID: 37312053 DOI: 10.1186/s12885-023-11031-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Radiotherapy (RT) is the standard of care for most advanced head and neck squamous cell carcinoma (HNSCC) and results in an unfavorable 5-year overall survival of 40%. Despite strong biological rationale, combining RT with immune checkpoint inhibitors does not result in a survival benefit. Our hypothesis is that the combination of these individually effective treatments fails because of radiation-induced immunosuppression and lymphodepletion. By integrating modern radiobiology and innovative radiotherapy concepts, the patient's immune system could be maximally retained by (1) increasing the dose per fraction so that the total dose and number of fractions can be reduced (HYpofractionation), (2) redistributing the radiation dose towards a higher peak dose within the tumor center and a lowered elective lymphatic field dose (Dose-redistribution), and (3) using RAdiotherapy with protons instead of photons (HYDRA). METHODS The primary aim of this multicenter study is to determine the safety of HYDRA proton- and photon radiotherapy by conducting two parallel phase I trials. Both HYDRA arms are randomized with the standard of care for longitudinal immune profiling. There will be a specific focus on actionable immune targets and their temporal patterns that can be tested in future hypofractionated immunoradiotherapy trials. The HYDRA dose prescriptions (in 20 fractions) are 40 Gy elective dose and 55 Gy simultaneous integrated boost on the clinical target volume with a 59 Gy focal boost on the tumor center. A total of 100 patients (25 per treatment group) will be recruited, and the final analysis will be performed one year after the last patient has been included. DISCUSSION In the context of HNSCC, hypofractionation has historically only been reserved for small tumors out of fear for late normal tissue toxicity. To date, hypofractionated radiotherapy may also be safe for larger tumors, as both the radiation dose and volume can be reduced by the combination of advanced imaging for better target definition, novel accelerated repopulation models and high-precision radiation treatment planning and dose delivery. HYDRA's expected immune-sparing effect may lead to improved outcomes by allowing for future effective combination treatment with immunotherapy. TRIAL REGISTRATION The trial is registered at ClinicalTrials.gov; NCT05364411 (registered on May 6th, 2022).
Collapse
Affiliation(s)
- Joris B W Elbers
- Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
- Department of Radiotherapy, HollandPTC, Delft, The Netherlands.
| | - Pascal A Gunsch
- Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Reno Debets
- Department of Medical Oncology, Laboratory of Tumor Immunology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Stijn Keereweer
- Department of Otorhinolaryngology head and neck surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Esther van Meerten
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jaap Zindler
- Department of Radiotherapy, HollandPTC, Delft, The Netherlands
- Department of Radiotherapy, Haaglanden Medical Center, Den Haag, The Netherlands
| | - Yvette van Norden
- Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Mischa S Hoogeman
- Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Radiotherapy, HollandPTC, Delft, The Netherlands
| | - Gerda M Verduijn
- Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Michiel Kroesen
- Department of Radiotherapy, HollandPTC, Delft, The Netherlands
| | - Remi A Nout
- Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
27
|
Chi A, Nguyen NP. Mechanistic rationales for combining immunotherapy with radiotherapy. Front Immunol 2023; 14:1125905. [PMID: 37377970 PMCID: PMC10291094 DOI: 10.3389/fimmu.2023.1125905] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Immunotherapy consisted mainly of immune checkpoint inhibitors (ICIs) has led to significantly improved antitumor response. However, such response has been observed only in tumors possessing an overall responsive tumor immune micro-environment (TIME), in which the presence of functional tumor-infiltrating lymphocytes (TILs) is critical. Various mechanisms of immune escape from immunosurveillance exist, leading to different TIME phenotypes in correlation with primary or acquired resistance to ICIs. Radiotherapy has been shown to induce antitumor immunity not only in the irradiated primary tumor, but also at unirradiated distant sites of metastases. Such antitumor immunity is mainly elicited by radiation's stimulatory effects on antigenicity and adjuvanticity. Furthermore, it may be significantly augmented when irradiation is combined with immunotherapy, such as ICIs. Therefore, radiotherapy represents one potential therapeutic strategy to restore anti-tumor immunity in tumors presenting with an unresponsive TIME. In this review, the generation of anti-tumor immunity, its impairment, radiation's immunogenic properties, and the antitumor effects of combining radiation with immunotherapy will be comprehensively discussed.
Collapse
Affiliation(s)
- Alexander Chi
- Department of Radiation Oncology, Capital Medical University Xuanwu Hospital, Beijing, China
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Nam Phong Nguyen
- Department of Radiation Oncology, Howard University, Washington, DC, United States
| |
Collapse
|
28
|
Hannon G, Lesch ML, Gerber SA. Harnessing the Immunological Effects of Radiation to Improve Immunotherapies in Cancer. Int J Mol Sci 2023; 24:7359. [PMID: 37108522 PMCID: PMC10138513 DOI: 10.3390/ijms24087359] [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: 03/22/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Ionizing radiation (IR) is used to treat 50% of cancers. While the cytotoxic effects related to DNA damage with IR have been known since the early 20th century, the role of the immune system in the treatment response is still yet to be fully determined. IR can induce immunogenic cell death (ICD), which activates innate and adaptive immunity against the cancer. It has also been widely reported that an intact immune system is essential to IR efficacy. However, this response is typically transient, and wound healing processes also become upregulated, dampening early immunological efforts to overcome the disease. This immune suppression involves many complex cellular and molecular mechanisms that ultimately result in the generation of radioresistance in many cases. Understanding the mechanisms behind these responses is challenging as the effects are extensive and often occur simultaneously within the tumor. Here, we describe the effects of IR on the immune landscape of tumors. ICD, along with myeloid and lymphoid responses to IR, are discussed, with the hope of shedding light on the complex immune stimulatory and immunosuppressive responses involved with this cornerstone cancer treatment. Leveraging these immunological effects can provide a platform for improving immunotherapy efficacy in the future.
Collapse
Affiliation(s)
- Gary Hannon
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Maggie L. Lesch
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Scott A. Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
29
|
Nelson BE, Adashek JJ, Lin SH, Subbiah V. On target methods to induce abscopal phenomenon for Off-Target effects: From happenstance to happenings. Cancer Med 2023; 12:6451-6465. [PMID: 36411943 PMCID: PMC10067075 DOI: 10.1002/cam4.5454] [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: 09/16/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
Although the "abscopal phenomenon" has been described several decades ago, this phenomenon lately has been obtaining momentous traction with the dawn of immune-based therapies. There has been increased cross talk among radiation oncologists, oncologists and immunologists and consequently a surge in the number of prospective clinical trials. This must be coupled with translation work from these clinical trials to aid in eventual identification of patients who may benefit. Abscopal effects may be induced by local and systemic methods, conventional radiotherapy, particle radiation, radionucleotide methods, cryoablation and brachytherapy. These approaches have all been reported to be stimulate abscopal effect. Immune induction by immune checkpoint therapy, immune adjuvants, cellular therapy including CAR and NK cell therapies may generate systemic abscopal response. With increasing recognition of this effect, there remains a lot of work to explore the modalities of inducing abscopal responses and ultimate prediction or prognostication on stratifying who may benefit. Ultimately, there is an urgent need for prospective studies and data to tease apart which one of these modalities can be applied to the appropriate candidate, to the appropriate cancer at the appropriate setting. This review seeks to elucidate readers on the different modalities of radiation, systemic therapies and other techniques rarely explored to potentiate the abscopal effect from a mere coincidence to a finite occurrence.
Collapse
Affiliation(s)
- Blessie Elizabeth Nelson
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Jacob J. Adashek
- Department of OncologyThe Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins HospitalBaltimoreMarylandUSA
| | - Steven H. Lin
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Vivek Subbiah
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
30
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
31
|
Chen Z, Nonaka H, Onishi H, Nakatani E, Oguri M, Saito M, Aoki S, Marino K, Komiyama T, Kuriyama K, Araya M, Tominaga L, Saito R, Maehata Y, Shinohara R. Impact of Systemic Autoimmune Diseases on Treatment Outcomes and Radiation Toxicities in Patients with Stage I Non-Small Cell Lung Cancer Receiving Stereotactic Body Radiation Therapy: A Matched Case-Control Analysis. Cancers (Basel) 2022; 14:cancers14235915. [PMID: 36497397 PMCID: PMC9740448 DOI: 10.3390/cancers14235915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
We aimed to evaluate the impact of systemic autoimmune diseases (SADs) on treatment outcomes and radiation toxicities following stereotactic body radiation therapy (SBRT) for stage I non-small cell lung cancer (NSCLC). We queried an institution-based database on patients with SADs treated with SBRT for lung cancer between 2001 and 2016 (SAD group). Each patient was matched to three controls without SADs. The primary outcomes of interest were the overall survival (OS) and local control rate (LCR). The secondary outcomes were radiation toxicities of grades ≥2 (≥G2). Twelve patients with SADs were matched to 36 controls. The median follow-up duration was 3.6 years. There was a significant intergroup difference in the OS (hazard ratio [HR]: 4.11, 95% confidence incidence [CI]: 1.82−9.27, p < 0.001) and LCR (HR: 15.97, 95% CI: 2.89−88.29, p < 0.001). However, there were no significant intergroup differences in the odds of acute (odds ratio [OR]: 0.38, 95% CI: 0.02−8.91, p = 0.550) and late (OR: 2.20, 95% CI: 0.32−15.10, p = 0.422) ≥G2 radiation pneumonitis. No other ≥G2 toxicities were identified. In conclusion, although radiation toxicities are not enhanced by SADs, SADs are risk factors of poor prognosis following SBRT for stage I NSCLC.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Radiology, Shizuoka General Hospital, Shizuoka 420-8527, Japan
- Department of Radiology, School of Medicine, University of Yamanashi, Chuo 409-3898, Japan
- Correspondence: ; Tel.: +81-54-247-6111
| | - Hotaka Nonaka
- Department of Radiology, Fuji City General Hospital, Fuji 417-8567, Japan
| | - Hiroshi Onishi
- Department of Radiology, School of Medicine, University of Yamanashi, Chuo 409-3898, Japan
| | - Eiji Nakatani
- Graduate School of Public Health (Medical Statistics), Shizuoka Graduate University of Public Health, Shizuoka 420-0881, Japan
| | - Mitsuhiko Oguri
- Department of Radiology, Shizuoka General Hospital, Shizuoka 420-8527, Japan
| | - Masahide Saito
- Department of Radiology, School of Medicine, University of Yamanashi, Chuo 409-3898, Japan
| | - Shinichi Aoki
- Department of Radiology, School of Medicine, University of Yamanashi, Chuo 409-3898, Japan
| | - Kan Marino
- Department of Radiology, School of Medicine, University of Yamanashi, Chuo 409-3898, Japan
| | - Takafumi Komiyama
- Department of Radiology, School of Medicine, University of Yamanashi, Chuo 409-3898, Japan
| | - Kengo Kuriyama
- Department of Radiology, Shizuoka General Hospital, Shizuoka 420-8527, Japan
| | - Masayuki Araya
- Proton Therapy Center, Aizawa Hospital, Matsumoto 390-8510, Japan
| | - Licht Tominaga
- Department of Radiology, Toranomon Hospital, Tokyo 105-8470, Japan
| | - Ryo Saito
- Department of Radiology, Shimada Municipal Hospital, Shimada 427-8502, Japan
| | - Yoshiyasu Maehata
- Department of Radiology, Yamanashi Prefectural Hospital, Kofu 400-8506, Japan
| | - Ryoji Shinohara
- Department of Health Sciences, Basic Science for Clinical Medicine, University of Yamanashi, Chuo 409-3898, Japan
| |
Collapse
|
32
|
Turchan WT, Pitroda SP, Weichselbaum RR. Beyond the Visible Spectrum: Considering the Oligometastatic Hypothesis in the Light of a New Era. Int J Radiat Oncol Biol Phys 2022; 114:581-586. [DOI: 10.1016/j.ijrobp.2022.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/10/2022] [Accepted: 04/15/2022] [Indexed: 11/16/2022]
|
33
|
Centrosome de-clustering of cancer cells induces cGAS-STING-mediated innate immunity of tumor-associated tumor cells in response to irradiation. Biochem Biophys Res Commun 2022; 636:24-30. [DOI: 10.1016/j.bbrc.2022.10.092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 11/24/2022]
|
34
|
Alvi MA, Asher AL, Michalopoulos GD, Grills IS, Warnick RE, McInerney J, Chiang VL, Attia A, Timmerman R, Chang E, Kavanagh BD, Andrews DW, Walter K, Bydon M, Sheehan JP. Factors associated with progression and mortality among patients undergoing stereotactic radiosurgery for intracranial metastasis: results from a national real-world registry. J Neurosurg 2022; 137:985-998. [PMID: 35171833 DOI: 10.3171/2021.10.jns211410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 10/14/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Stereotactic radiosurgery (SRS) has been increasingly employed in recent years to treat intracranial metastatic lesions. However, there is still a need for optimization of treatment paradigms to provide better local control and prevent progressive intracranial disease. In the current study, the authors utilized a national collaborative registry to investigate the outcomes of patients with intracranial metastatic disease who underwent SRS and to determine factors associated with lesion treatment response, overall progression, and mortality. METHODS The NeuroPoint Alliance SRS registry was queried for all patients with intracranial metastatic lesions undergoing single- or multifraction SRS at participating institutions between 2016 and 2020. The main outcomes of interest included lesion response (lesion-level analysis), progression using Response Assessment for Neuro-Oncology criteria, and mortality (patient-level analysis). Kaplan-Meier analysis was used to report time to progression and overall survival, and multivariable Cox proportional hazards analysis was used to investigate factors associated with lesion response, progression, and mortality. RESULTS A total of 501 patients (1447 intracranial metastatic lesions) who underwent SRS and had available follow-up were included in the current analyses. The most common primary tumor was lung cancer (49.5%, n = 248), followed by breast (15.4%, n = 77) and melanoma (12.2%, n = 61). Most patients had a single lesion (44.9%, n = 225), 29.3% (n = 147) had 2 or 3 lesions, and 25.7% (n = 129) had > 3 lesions. The mean sum of baseline measurements of the lesions according to Response Evaluation Criteria in Solid Tumors (RECIST) was 35.54 mm (SD 25.94). At follow-up, 671 lesions (46.4%) had a complete response, 631 (43.6%) had a partial response (≥ 30% decrease in longest diameter) or were stable (< 30% decrease but < 20% increase), and 145 (10%) showed progression (> 20% increase in longest diameter). On multivariable Cox proportional hazards analysis, melanoma-associated lesions (HR 0.48, 95% CI 0.34-0.67; p < 0.001) and larger lesion size (HR 0.94, 95% CI 0.93-0.96; p < 0.001) showed lower odds of lesion regression, while a higher biologically effective dose was associated with higher odds (HR 1.001, 95% CI 1.0001-1.00023; p < 0.001). A total of 237 patients (47.3%) had overall progression (local failure or intracranial progressive disease), with a median time to progression of 10.03 months after the index SRS. Factors found to be associated with increased hazards of progression included male sex (HR 1.48, 95% CI 1.108-1.99; p = 0.008), while administration of immunotherapy (before or after SRS) was found to be associated with lower hazards of overall progression (HR 0.62, 95% CI 0.460-0.85; p = 0.003). A total of 121 patients (23.95%) died during the follow-up period, with a median survival of 19.4 months from the time of initial SRS. A higher recursive partitioning analysis score (HR 21.3485, 95% CI 1.53202-3.6285; p < 0.001) was found to be associated with higher hazards of mortality, while single-fraction treatment compared with hypofractionated treatment (HR 0.082, 95% CI 0.011-0.61; p = 0.015), administration of immunotherapy (HR 0.385, 95% CI 0.233-0.64; p < 0.001), and presence of single compared with > 3 lesions (HR 0.427, 95% CI 0.187-0.98; p = 0.044) were found to be associated with lower risk of mortality. CONCLUSIONS The comparability of results between this study and those of previously published clinical trials affirms the value of multicenter databases with real-world data collected without predetermined research purpose.
Collapse
Affiliation(s)
- Mohammed Ali Alvi
- 1Mayo Clinic Neuro-Informatics Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota
- 2Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Anthony L Asher
- 3Neuroscience Institute, Carolinas Healthcare System and Carolina Neurosurgery & Spine Associates, Charlotte, North Carolina
| | - Giorgos D Michalopoulos
- 1Mayo Clinic Neuro-Informatics Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota
- 2Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Inga S Grills
- 4Department of Neurological Surgery, Beaumont Health System, Royal Oak, Michigan
| | - Ronald E Warnick
- 5Department of Neurosurgery, The Jewish Hospital, Cincinnati, Ohio
| | - James McInerney
- 6Department of Neurosurgery, Penn State Health, Hershey, Pennsylvania
| | - Veronica L Chiang
- 7Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Albert Attia
- 8Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Robert Timmerman
- 9Department of Neurological Surgery, UT Southwestern Medical Center, Dallas, Texas
| | - Eric Chang
- 10Department of Radiation Oncology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Brian D Kavanagh
- 11Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - David W Andrews
- 12Department of Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Kevin Walter
- 13Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York; and
| | - Mohamad Bydon
- 1Mayo Clinic Neuro-Informatics Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota
- 2Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Jason P Sheehan
- 14Department of Neurological Surgery, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|
35
|
Liu Z, Yu Z, Chen D, Verma V, Yuan C, Wang M, Wang F, Fan Q, Wang X, Li Y, Ma Y, Wu M, Yu J. Pivotal roles of tumor-draining lymph nodes in the abscopal effects from combined immunotherapy and radiotherapy. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 42:971-986. [PMID: 35962977 PMCID: PMC9558691 DOI: 10.1002/cac2.12348] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/28/2022] [Accepted: 07/27/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Currently, due to synergy enhancement of anti-tumor effects and potent stimulation of abscopal effects, combination therapy with irradiation and programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) immune checkpoint inhibition (immuno-radiotherapy, iRT) has revolutionized the therapeutic guidelines. It has been demonstrated that tumor-draining lymph nodes (TDLN) are essential for effective antitumor immunity induced by radiotherapy, immunotherapy, or iRT. Given that the function of TDLN in iRT remains unclear, this study aimed to investigate the function and mechanism of TDLN in iRT-induced abscopal effects. METHODS The function of TDLN was evaluated using unilateral or bilateral MC38 and B16F10 subcutaneous tumor models with or without indicated TDLN. The flow cytometry, multiple immunofluorescence analysis, and NanoString analysis were utilized to detect the composition and function of the immune cells in the primary and abscopal tumor microenvironment. Additionally, we tempted to interrogate the possible mechanisms via RNA-sequencing of tumor-infiltrating lymphocytes and TDLN. RESULTS TDLN deficiency impaired the control of tumor growth by monotherapy. Bilateral TDLN removal rather than unilateral TDLN removal substantially curtailed iRT-stimulated anti-tumor and abscopal effects. Furthermore, in the absence of TDLN, the infiltration of CD45+ and CD8+ T cells was substantially reduced in both primary and abscopal tumors, and the anti-tumor function of CD8+ T cells was attenuated as well. Additionally, the polarization of tumor-associated macrophages in primary and abscopal tumors were found to be dependent on intact bilateral TDLN. RNA-sequencing data indicated that impaired infiltration and anti-tumor effects of immune cells partially attributed to the altered secretion of components from the tumor microenvironment. CONCLUSIONS TDLN play a critical role in iRT by promoting the infiltration of CD8+ T cells and maintaining the M1/M2 macrophage ratio.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. 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, 250117, P. R. China.,Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Zhiyong Yu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Dawei Chen
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. 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, 250117, P. R. China
| | - Vivek Verma
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, United States
| | - Chenxi Yuan
- 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, 250117, P. R. China
| | - Minglei Wang
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. 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, 250117, P. R. China
| | - Fei Wang
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. 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, 250117, P. R. China
| | - Qing Fan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Xingwu Wang
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. 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, 250117, P. R. China
| | - Yang Li
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. 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, 250117, P. R. China
| | - Yuequn Ma
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. 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, 250117, P. R. China
| | - Meng Wu
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. 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, 250117, P. R. China
| | - Jinming Yu
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. 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, 250117, P. R. China.,Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| |
Collapse
|
36
|
Misaki S, Murata S, Shimoji M, Iwai T, Sihombing AM, Aoki K, Takahashi Y, Watanabe Y. Enhancement of antitumor immune response by radiation therapy combined with dual immune checkpoint inhibitor in a metastatic model of HER2-positive murine tumor. Jpn J Radiol 2022; 40:1307-1315. [PMID: 35763240 DOI: 10.1007/s11604-022-01303-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 06/01/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Treatments for metastatic human epidermal growth factor receptor 2 (HER2)-positive tumors are improving but remain inadequate. We investigated activating antitumor immune response by combining radiation therapy with immune checkpoint inhibitors using mouse tumors overexpressing HER2, a pivotal driver oncogenic antigen, to develop new immunotherapies for metastatic HER2-positive tumors. MATERIALS AND METHODS NT2.5 cells were inoculated into the two mammary fat pads of FVB/N mice, which were divided into four groups: no treatment (Non), anti-PD-1 and anti-CTLA4 antibodies (P1C4), irradiation of the large tumor (Rad), and combination (R + P1C4) groups. Tumor growth, immunostaining of tumor-infiltrating lymphocytes, and the proportion of HER2-tumor antigen-specific CD8-positive T cells in the spleen and tumor-infiltrating lymphocytes were analyzed. RESULTS In the Rad group, unirradiated and irradiated tumors shrank after treatment. Besides the directly irradiated tumors, the unirradiated tumors in the R + P1C4 group shrank the most. In the unirradiated tumors, CD8-positive T cells and FOXP3-positive T cells accumulated significantly more in the R + P1C4 group than in the P1C4 and the Rad groups (all p < 0.001). CD4-positive helper T cells accumulated significantly more in the R + P1C4 group than in the Rad group (p < 0.05), but this was not significantly different from the P1C4 group. HER2-specific CD8-positive T cells in the spleen and tumor-infiltrating lymphocytes were significantly increased in the R + P1C4 group compared to the P1C4 and Rad groups (all p < 0.0001). CONCLUSION Irradiation of HER2-positive tumors induced an antitumor immune effect against the unirradiated tumor, which was enhanced by the combined use of immune checkpoint inhibitors and was mediated by enhanced recruitment of HER2-tumor antigen-specific cytotoxic T lymphocytes at the tumor site in an HER2-positive mouse tumor model. Harnessing the distant antitumor immune response induced by the combination of radiation therapy and immune checkpoint inhibitors could be a promising treatment strategy for metastatic HER2-positive tumors.
Collapse
Affiliation(s)
- Sayaka Misaki
- Department of Radiology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan.
| | - Satoshi Murata
- Cancer Center, Shiga University of Medical Science Hospital, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
- Department of Surgery, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Miyuki Shimoji
- Department of Surgery, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Takayasu Iwai
- Department of Radiology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Andreas Michael Sihombing
- Department of Surgery, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Ken Aoki
- Department of Radiology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Yutaka Takahashi
- Department of Medical Physics and Engineering, Osaka University Graduate School of Medicine, 1-7, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yoshiyuki Watanabe
- Department of Radiology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
37
|
Zhao LP, Hu JH, Hu D, Wang HJ, Huang CG, Luo RH, Zhou ZH, Huang XY, Xie T, Lou JS. Hyperprogression, a challenge of PD-1/PD-L1 inhibitors treatments: potential mechanisms and coping strategies. Biomed Pharmacother 2022; 150:112949. [PMID: 35447545 DOI: 10.1016/j.biopha.2022.112949] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/01/2022] [Accepted: 04/08/2022] [Indexed: 11/29/2022] Open
Abstract
Immunotherapy is now a mainstay in cancer treatments. Programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) immune checkpoint inhibitor (ICI) therapies have opened up a new venue of advanced cancer immunotherapy. However, hyperprogressive disease (HPD) induced by PD-1/PD-L1 inhibitors caused a significant decrease in the overall survival (OS) of the patients, which compromise the efficacy of PD-1/PD-L1 inhibitors. Therefore, HPD has become an urgent issue to be addressed in the clinical uses of PD-1/PD-L1 inhibitors. The mechanisms of HPD remain unclear, and possible predictive factors of HPD are not well understood. In this review, we summarized the potential mechanisms of HPD and coping strategies that can effectively reduce the occurrence and development of HPD.
Collapse
Affiliation(s)
- Li-Ping Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jun-Hu Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Die Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Hao-Jie Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Chang-Gang Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ru-Hua Luo
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Zhao-Huang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xin-Yun Huang
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA.
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Jian-Shu Lou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| |
Collapse
|
38
|
Tagliaferri L, Lancellotta V, Fionda B, Mangoni M, Casà C, Di Stefani A, Pagliara MM, D’Aviero A, Schinzari G, Chiesa S, Mazzarella C, Manfrida S, Colloca GF, Marazzi F, Morganti AG, Blasi MA, Peris K, Tortora G, Valentini V. Immunotherapy and radiotherapy in melanoma: a multidisciplinary comprehensive review. Hum Vaccin Immunother 2022; 18:1903827. [PMID: 33847208 PMCID: PMC9122308 DOI: 10.1080/21645515.2021.1903827] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/16/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Melanoma is an extremely aggressive tumor and is considered to be an extremely immunogenic tumor because compared to other cancers it usually presents a well-expressed lymphoid infiltration. The aim of this paper is to perform a multidisciplinary comprehensive review of the evidence available about the combination of radiotherapy and immunotherapy for melanoma. Radiation, in fact, can increase tumor antigens visibility and promote priming of T cells but can also exert immunosuppressive action on tumor microenvironment. Combining radiotherapy with immunotherapy provides an opportunity to increase immunostimulatory potential of radiation. We therefore provide the latest clinical evidence about radiobiological rationale, radiotherapy techniques, timing, and role both in advanced and systemic disease (with a special focus on ocular melanoma and brain, liver, and bone metastases) with a particular attention also in geriatric patients. The combination of immunotherapy and radiotherapy seems to be a safe therapeutic option, supported by a clear biological rationale, even though the available data confirm that radiotherapy is employed more for metastatic than for non-metastatic disease. Such a combination shows promising results in terms of survival outcomes; however, further studies, hopefully prospective, are needed to confirm such evidence.
Collapse
Affiliation(s)
- Luca Tagliaferri
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Valentina Lancellotta
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Bruno Fionda
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Monica Mangoni
- Sezione di Radioterapia Oncologica, Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università di Firenze, Florence, Italy
| | - Calogero Casà
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Alessandro Di Stefani
- UOC Dermatologia, Dipartimento di Scienze mediche e chirurgiche, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Monica Maria Pagliara
- UOC Oncologia Oculare, Dipartimento di Scienze dell'Invecchiamento, neurologiche ortopediche e della testa collo, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Andrea D’Aviero
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Giovanni Schinzari
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Silvia Chiesa
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Ciro Mazzarella
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Stefania Manfrida
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Giuseppe Ferdinando Colloca
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Fabio Marazzi
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Alessio Giuseppe Morganti
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale Settore Scientifico Disciplinare, Università di Bologna, Bologna, Italy
| | - Maria Antonietta Blasi
- UOC Oncologia Oculare, Dipartimento di Scienze dell'Invecchiamento, neurologiche ortopediche e della testa collo, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Ketty Peris
- UOC Dermatologia, Dipartimento di Scienze mediche e chirurgiche, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Giampaolo Tortora
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Vincenzo Valentini
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
39
|
Baxevanis CN, Gritzapis AD, Voutsas IF, Batsaki P, Goulielmaki M, Adamaki M, Zoumpourlis V, Fortis SP. T-Cell Repertoire in Tumor Radiation: The Emerging Frontier as a Radiotherapy Biomarker. Cancers (Basel) 2022; 14:cancers14112674. [PMID: 35681654 PMCID: PMC9179913 DOI: 10.3390/cancers14112674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Radiotherapy constitutes an essential component of the treatment for malignant disease. Besides its direct effect on cancer cells, namely, DNA damage and cell death, ionizing irradiation also mediates indirect antitumor effects that are mostly mediated by the immune system. Investigations into the processes underlying the interaction between radiotherapy and the immune system have uncovered mechanisms that can be exploited to promote the antitumor efficacy of radiotherapy both locally in the irradiated primary tumor and also at distant lesions in non-irradiated tumors. Because of its capacity to stimulate antitumor immunity, radiotherapy is also applied in combination with immune-checkpoint-inhibition-based immunotherapy. This review discusses the important pathways that govern the synergistic interactions between ionizing radiation and antitumor immune reactivity. Unravelling these involved mechanisms is mandatory for the successful application of anticancer radiotherapy and immunotherapy. We also place emphasis on the need for biomarkers that will aid in the selection of patients most likely to benefit from such combined treatments. Abstract Radiotherapy (RT) is a therapeutic modality that aims to eliminate malignant cells through the induction of DNA damage in the irradiated tumor site. In addition to its cytotoxic properties, RT also induces mechanisms that result in the promotion of antitumor immunity both locally within the irradiation field but also at distant tumor lesions, a phenomenon that is known as the “abscopal” effect. Because the immune system is capable of sensing the effects of RT, several treatment protocols have been assessing the synergistic role of radiotherapy combined with immunotherapy, collectively referred to as radioimmunotherapy. Herein, we discuss mechanistic insights underlying RT-based immunomodulation, which also enhance our understanding of how RT regulates antitumor T-cell-mediated immunity. Such knowledge is essential for the discovery of predictive biomarkers and for the improvement of clinical trials investigating the efficacy of radio-immunotherapeutic modalities in cancer patients.
Collapse
Affiliation(s)
- Constantin N. Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Angelos D. Gritzapis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Ioannis F. Voutsas
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Panagiota Batsaki
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Maria Goulielmaki
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Maria Adamaki
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece; (M.A.); (V.Z.)
| | - Vassilios Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece; (M.A.); (V.Z.)
| | - Sotirios P. Fortis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
- Correspondence: ; Tel.: +30-2106409462
| |
Collapse
|
40
|
Turchan WT, Pitroda SP, Weichselbaum RR. Combined radio-immunotherapy: An opportunity to increase the therapeutic ratio of oligometastasis-directed radiotherapy. Neoplasia 2022; 27:100782. [PMID: 35303578 PMCID: PMC8931441 DOI: 10.1016/j.neo.2022.100782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 11/24/2022]
Abstract
The utility of radiotherapy as a means of palliating symptoms due to metastatic cancer is well-accepted. A growing body of literature suggests that radiotherapy may play a role beyond palliation in some patients with low-burden metastatic disease. Recent data suggest that oligometastasis-directed radiotherapy may improve progression-free and even overall survival in select patients. Immunotherapy also has a growing role in the management of patients with metastatic cancer and, like radiotherapy, appears to be most effective in the setting of low-volume disease. Thus, the addition of immunotherapy may be a feasible means of increasing the therapeutic ratio of metastasis-directed radiotherapy, particularly among patients with oligometastatic cancer.
Collapse
Affiliation(s)
- William Tyler Turchan
- University of Chicago, Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, 5758 S Maryland Ave, Chicago, IL 60637, United States
| | - Sean P Pitroda
- University of Chicago, Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, 5758 S Maryland Ave, Chicago, IL 60637, United States
| | - Ralph R Weichselbaum
- University of Chicago, Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, 5758 S Maryland Ave, Chicago, IL 60637, United States.
| |
Collapse
|
41
|
Tumor draining lymph nodes, immune response, and radiotherapy: Towards a revisal of therapeutic principles. Biochim Biophys Acta Rev Cancer 2022; 1877:188704. [DOI: 10.1016/j.bbcan.2022.188704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/06/2022] [Accepted: 02/21/2022] [Indexed: 12/20/2022]
|
42
|
Askar MA, El-Nashar HA, Al-Azzawi MA, Rahman SSA, Elshawi OE. Synergistic Effect of Quercetin Magnetite Nanoparticles and Targeted Radiotherapy in Treatment of Breast Cancer. BREAST CANCER: BASIC AND CLINICAL RESEARCH 2022; 16:11782234221086728. [PMID: 35359610 PMCID: PMC8961357 DOI: 10.1177/11782234221086728] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/17/2022] [Indexed: 01/16/2023] Open
Abstract
Quercetin is a potent cancer therapeutic agent present in fruits and vegetables. The pharmaceutical uses of quercetin are limited due to many problems associated with low solubility, bioavailability, permeability, and instability. In addition, the high doses of quercetin show toxic effects in clinical and experimental studies. Therefore, a new strategy is warranted to overcome these problems without the use of toxic doses. The iron oxide nanoparticles can be used as a drug delivery system. This study aimed to prepare quercetin-conjugated magnetite nanoparticles (QMNPs) using biological simple nanoprecipitation and mediated by fungus Aspergillus oryzae. Also, we initiated in vitro and in vivo studies to determine whether QMNPs might sensitize breast cancer to radiotherapy treatment. The structural, morphological, and magnetic properties of the prepared nanoparticles were studied. The results indicated that QMNPs were spherical in shape and 40 nm in diameter. The in vitro studies showed that the incubation of MCF-7, HePG-2, and A459 cancer cells with QMNPs for 24 h effectively inhibited the growth of cancer cell lines in a concentration-dependent manner with IC50 values of 11, 77.5, and104 nmol/mL, respectively. The combination of QMNPs with irradiation (IR) potently blocked MCF-7 cancer cell proliferation and showed significant changes in the morphology of these cells as observed by bright-field inverted light microscopy. Focusing on the long-term toxicity of QMNPs (20 ml/kg), the assessment of hematological, hepatic, and renal markers indicated no toxic effect. Besides, QMNPs inhibited tumor growth and potently enhanced the lateral radiotherapy treatment in N-methyl-N-nitrosourea (MNU)-induced breast cancer in female white albino rats. These anticancer and radiosensitizing activities were ascribed to cytotoxicity, cell cycle arrest, immunomodulation, and efficiency through induction of apoptosis. In a conclusion, these observations suggest that the QMNPs combined with LRT could act as a potential targeted therapy in breast cancer.
Collapse
Affiliation(s)
- Mostafa A Askar
- Department of Radiation Biology, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Heba As El-Nashar
- Department of Pharmacognosy and Center of Drug Discovery Research and Development, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mahmood A Al-Azzawi
- Department of Pathological Analysis Techniques, College of Medical & Health Technologies, Ahl Al Bayt University, Karbala, Iraq
| | - Sahar S Abdel Rahman
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Omama E Elshawi
- Department of Health and Radiation Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
43
|
Friedrich T, Scholz M, Durante M. A predictive biophysical model of the combined action of radiotherapy and immunotherapy in cancer. Int J Radiat Oncol Biol Phys 2022; 113:872-884. [DOI: 10.1016/j.ijrobp.2022.03.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/24/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
|
44
|
Meningeal lymphatics regulate radiotherapy efficacy through modulating anti-tumor immunity. Cell Res 2022; 32:543-554. [PMID: 35301438 PMCID: PMC9159979 DOI: 10.1038/s41422-022-00639-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 02/11/2022] [Indexed: 12/15/2022] Open
Abstract
As a first-line treatment, radiotherapy (RT) is known to modulate the immune microenvironment of glioma, but it is unknown whether the meningeal lymphatic vessel (MLV)-cervical lymph node (CLN) network regulates the process or influences RT efficacy. Here, we show that the MLV-CLN network contributes to RT efficacy in brain tumors and mediates the RT-modulated anti-tumor immunity that is enhanced by vascular endothelial growth factor C (VEGF-C). Meningeal lymphatic dysfunction impaired tumor-derived dendritic cell (DC) trafficking and CD8+ T cell activation after RT, whereas tumors overexpressing VEGF-C with meningeal lymphatic expansion were highly sensitive to RT. Mechanistically, VEGF-C-driven modulation of RT-triggered anti-tumor immunity was attributed to C-C Motif Chemokine Ligand 21 (CCL21)-dependent DC trafficking and CD8+ T cell activation. Notably, delivery of VEGF-C mRNA significantly enhanced RT efficacy and anti-tumor immunity in brain tumors. These findings suggest an essential role of the MLV-CLN network in RT-triggered anti-tumor immunity, and highlight the potential of VEGF-C mRNA for brain tumor therapy.
Collapse
|
45
|
Hannan R, Mohamad O, Diaz de Leon A, Manna S, Pop LM, Zhang Z, Mannala S, Christie A, Christley S, Monson N, Ishihara D, Hsu EJ, Ahn C, Kapur P, Chen M, Arriaga Y, Courtney K, Cantarel B, Wakeland EK, Fu YX, Pedrosa I, Cowell L, Wang T, Margulis V, Choy H, Timmerman RD, Brugarolas J. Outcome and Immune Correlates of a Phase II Trial of High-Dose Interleukin-2 and Stereotactic Ablative Radiotherapy for Metastatic Renal Cell Carcinoma. Clin Cancer Res 2021; 27:6716-6725. [PMID: 34551906 PMCID: PMC9924935 DOI: 10.1158/1078-0432.ccr-21-2083] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/13/2021] [Accepted: 09/20/2021] [Indexed: 01/04/2023]
Abstract
PURPOSE This phase II clinical trial evaluated whether the addition of stereotactic ablative radiotherapy (SAbR), which may promote tumor antigen presentation, improves the overall response rate (ORR) to high-dose IL2 (HD IL2) in metastatic renal cell carcinoma (mRCC). PATIENTS AND METHODS Patients with pathologic evidence of clear cell renal cell carcinoma (RCC) and radiographic evidence of metastasis were enrolled in this single-arm trial and were treated with SAbR, followed by HD IL2. ORR was assessed based on nonirradiated metastases. Secondary endpoints included overall survival (OS), progression-free survival (PFS), toxicity, and treatment-related tumor-specific immune response. Correlative studies involved whole-exome and transcriptome sequencing, T-cell receptor sequencing, cytokine analysis, and mass cytometry on patient samples. RESULTS Thirty ethnically diverse mRCC patients were enrolled. A median of two metastases were treated with SAbR. Among 25 patients evaluable by RECIST v1.1, ORR was 16% with 8% complete responses. Median OS was 37 months. Treatment-related adverse events (AE) included 22 grade ≥3 events that were not dissimilar from HD IL2 alone. There were no grade 5 AEs. A correlation was observed between SAbR to lung metastases and improved PFS (P = 0.0165). Clinical benefit correlated with frameshift mutational load, mast cell tumor infiltration, decreased circulating tumor-associated T-cell clones, and T-cell clonal expansion. Higher regulatory/CD8+ T-cell ratios at baseline in the tumor and periphery correlated with no clinical benefit. CONCLUSIONS Adding SAbR did not improve the response rate to HD IL2 in patients with mRCC in this study. Tissue analyses suggest a possible correlation between frameshift mutation load as well as tumor immune infiltrates and clinical outcomes.
Collapse
Affiliation(s)
- Raquibul Hannan
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas.
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Osama Mohamad
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Alberto Diaz de Leon
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Subrata Manna
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Laurentiu M Pop
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ze Zhang
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Samantha Mannala
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alana Christie
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Scott Christley
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nancy Monson
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Dan Ishihara
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Eric J Hsu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chul Ahn
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Payal Kapur
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Mingyi Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yull Arriaga
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kevin Courtney
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Brandi Cantarel
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Edward K Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ivan Pedrosa
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Lindsay Cowell
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tao Wang
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Vitaly Margulis
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hak Choy
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Robert D Timmerman
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - James Brugarolas
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
46
|
Sun C, Zhang M, Qiao Q, Wang Y. Integrating Intrinsic Radiosensitivity and Immune Status for Predicting Benefits of Radiotherapy in Head and Neck Squamous Cell Carcinoma. Med Sci Monit 2021; 27:e932126. [PMID: 34670999 PMCID: PMC8547194 DOI: 10.12659/msm.932126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Background HNSCC (head and neck squamous cell carcinoma) is a heterogeneous disease for which radiotherapy is a main treatment. As intrinsic radiosensitivity and immune status affect the initial and effective stage of the radiation-induced cancer immunity cycle, respectively, it is important to consider both of them when we select patients who can benefit from radiotherapy. Material/Methods Our study included all HNSCC patients with complete survival and radiotherapy information in TCGA database. Patients were divided into RS (radiosensitive), RR (radioresistant), immune, and non-immune groups according to their RSI (radiosensitivity index) and immune score calculated by the ESTIMATE algorithm. Survival analysis was performed to compare OS (overall survival) between patients receiving and not receiving radiotherapy. GO and KEGG pathway enrichment analysis was performed for functional analysis. Univariate Cox and ridge regression analysis were performed to construct a predictive gene signature based on the combined stratification. Results Only patients in the RS-immune group could benefit from radiotherapy, and the survival analysis results remained consistent after we performed propensity score matching between patients receiving and not receiving radiotherapy. The differentially expressed genes between the RS-immune and non-RS-immune groups were mainly enriched in pathways related to immune process. The 3-gene signature we built exhibited predictive value in training and validation cohorts when treated as a binary or continuous variable. Conclusions The combined stratification of intrinsic radiosensitivity and immune status was superior to considering intrinsic radiosensitivity or immune status alone and could be used in preclinical evaluation to select patients or to decide whether radiotherapy sensitizers and immunotherapy should be used at the same time.
Collapse
Affiliation(s)
- Chuyao Sun
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland).,Department of Radiation Oncology, The Fourth Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Miao Zhang
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Qiao Qiao
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yanli Wang
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
47
|
Oba T, Kajihara R, Yokoi T, Repasky EA, Ito F. Neoadjuvant in situ immunomodulation enhances systemic antitumor immunity against highly metastatic tumors. Cancer Res 2021; 81:6183-6195. [PMID: 34666993 DOI: 10.1158/0008-5472.can-21-0939] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/21/2021] [Accepted: 10/14/2021] [Indexed: 11/16/2022]
Abstract
Neoadjuvant immunotherapy, given before surgical resection, is a promising approach to develop systemic antitumor immunity for the treatment of high-risk resectable disease. Here, using syngeneic and orthotopic mouse models of triple-negative breast cancer, we have tested the hypothesis that generation of tumor-specific T-cell responses by induction and activation of tumor-residing Batf3-dependent conventional type 1 dendritic cells (cDC1) before resection improves control of distant metastatic disease and survival. Mice bearing highly metastatic orthotopic tumors were treated with a combinatorial in situ immunomodulation (ISIM) regimen comprised of intratumoral administration of Flt3L, local radiotherapy, and in situ TLR3/CD40 stimulations, followed by surgical resection. Neoadjuvant ISIM generated tumor-specific CD8+ T cells that infiltrated into distant non-irradiated metastatic sites, which delayed the progression of lung metastases and improved survival after the resection of primary tumors. The efficacy of neoadjuvant ISIM was dependent on de novo adaptive T-cell immunity elicited by Batf3-dependent DCs and was enhanced by increasing dose and fractionation of radiotherapy, and early surgical resection after the completion of neoadjuvant ISIM. Importantly, neoadjuvant ISIM synergized with PD-L1 blockade to improve control of distant metastases and prolong survival, while removal of tumor-draining lymph nodes abrogated the antimetastatic efficacy of neoadjuvant ISIM. Our findings illustrate the therapeutic potential of neoadjuvant multimodal intralesional therapy for the treatment of resectable tumors with high risk of relapse.
Collapse
Affiliation(s)
- Takaaki Oba
- Division of Breast and Endocrine Surgery, Department of Surgery (II), Shinshu University School of Medicine
| | - Ryutaro Kajihara
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center
| | - Toshihiro Yokoi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center
| | | | - Fumito Ito
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center
| |
Collapse
|
48
|
Boustani J, Lecoester B, Baude J, Latour C, Adotevi O, Mirjolet C, Truc G. Anti-PD-1/Anti-PD-L1 Drugs and Radiation Therapy: Combinations and Optimization Strategies. Cancers (Basel) 2021; 13:cancers13194893. [PMID: 34638376 PMCID: PMC8508444 DOI: 10.3390/cancers13194893] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Although immune checkpoint blockade has yielded unprecedented and durable responses in cancer patients, the efficacy of this treatment remains limited. Radiation therapy can induce immunogenic cell death that contributes to the local efficacy of irradiation. However, radiation-induced systemic responses are scarce. Studies combining radiation with checkpoint inhibitors suggest a synergistic potential of this strategy. In this review, we focused on parameters that can be optimized to enhance the anti-tumor immune response that results from this association, in order to achieve data on dose, fractionation, target volume, lymph nodes sparing, radiation particles, and other immunomodulatory agents. These factors should be considered in future trials for better clinical outcomes. To this end, we discussed the main preclinical and clinical data available to optimize the efficacy of the treatment combination. Abstract Immune checkpoint inhibitors have been associated with long-term complete responses leading to improved overall survival in several cancer types. However, these novel immunotherapies are only effective in a small proportion of patients, and therapeutic resistance represents a major limitation in clinical practice. As with chemotherapy, there is substantial evidence that radiation therapy promotes anti-tumor immune responses that can enhance systemic responses to immune checkpoint inhibitors. In this review, we discuss the main preclinical and clinical evidence on strategies that can lead to an enhanced response to PD-1/PD-L1 blockade in combination with radiation therapy. We focused on central issues in optimizing radiation therapy, such as the optimal dose and fractionation for improving the therapeutic ratio, as well as the impact on immune and clinical responses of dose rate, target volume, lymph nodes irradiation, and type of radiation particle. We explored the addition of a third immunomodulatory agent to the combination such as other checkpoint inhibitors, chemotherapy, and treatment targeting the tumor microenvironment components. The strategies described in this review provide a lead for future clinical trials.
Collapse
Affiliation(s)
- Jihane Boustani
- Department of Radiation Oncology, Centre Georges François Leclerc, UNICANCER, 21079 Dijon, France; (J.B.); (J.B.); (C.L.); (G.T.)
- Department of Radiation Oncology, University Hospital of Besançon, 25000 Besançon, France
- INSERM, EFS BFC, UMR1098, RIGHT, Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, 25000 Besançon, France; (B.L.); (O.A.)
| | - Benoît Lecoester
- INSERM, EFS BFC, UMR1098, RIGHT, Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, 25000 Besançon, France; (B.L.); (O.A.)
| | - Jérémy Baude
- Department of Radiation Oncology, Centre Georges François Leclerc, UNICANCER, 21079 Dijon, France; (J.B.); (J.B.); (C.L.); (G.T.)
| | - Charlène Latour
- Department of Radiation Oncology, Centre Georges François Leclerc, UNICANCER, 21079 Dijon, France; (J.B.); (J.B.); (C.L.); (G.T.)
- INSERM UMR 1231, Cadir Team, 21000 Dijon, France
| | - Olivier Adotevi
- INSERM, EFS BFC, UMR1098, RIGHT, Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, 25000 Besançon, France; (B.L.); (O.A.)
- Department of Medical Oncology, University Hospital of Besançon, 25000 Besançon, France
| | - Céline Mirjolet
- Department of Radiation Oncology, Centre Georges François Leclerc, UNICANCER, 21079 Dijon, France; (J.B.); (J.B.); (C.L.); (G.T.)
- INSERM UMR 1231, Cadir Team, 21000 Dijon, France
- Correspondence:
| | - Gilles Truc
- Department of Radiation Oncology, Centre Georges François Leclerc, UNICANCER, 21079 Dijon, France; (J.B.); (J.B.); (C.L.); (G.T.)
| |
Collapse
|
49
|
Chua KLM, Chu PL, Tng DJH, Soo KC, Chua MLK. Repurposing Proton Beam Therapy through Novel Insights into Tumour Radioresistance. Clin Oncol (R Coll Radiol) 2021; 33:e469-e481. [PMID: 34509347 DOI: 10.1016/j.clon.2021.08.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/02/2021] [Accepted: 08/25/2021] [Indexed: 12/11/2022]
Abstract
Despite improvements in radiotherapy, radioresistance remains an important clinical challenge. Radioresistance can be mediated through enhanced DNA damage response mechanisms within the tumour or through selective pressures exerted by the tumour microenvironment (TME). The effects of the TME have in recent times gained increased attention, in part due to the success of immune modulating strategies, but also through improved understanding of the downstream effects of hypoxia and dysregulated wound healing processes on mediating radioresistance. Although we have a better appreciation of these molecular mechanisms, efforts to address them through novel combination approaches have been scarce, owing to limitations of photon therapy and concerns over toxicity. At the same time, proton beam therapy (PBT) represents an advancement in radiotherapy technologies. However, early clinical results have been mixed and the clinical strategies around optimal use and patient selection for PBT remain unclear. Here we highlight the role that PBT can play in addressing radioresistance, through better patient selection, and by providing an improved toxicity profile for integration with novel agents. We will also describe the developments around FLASH PBT. Through close examination of its normal tissue-sparing effects, we will highlight how FLASH PBT can facilitate combination strategies to tackle radioresistance by further improving toxicity profiles and by directly mediating the mechanisms of radioresistance.
Collapse
Affiliation(s)
- K L M Chua
- Oncology Academic Clinical Programme, Duke-NUS Medical School, Singapore; Division of Radiation Oncology, National Cancer Centre Singapore, Singapore
| | - P L Chu
- Oncology Academic Clinical Programme, Duke-NUS Medical School, Singapore
| | - D J H Tng
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore
| | - K C Soo
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore; Division of Surgical Oncology, National Cancer Centre Singapore, Singapore
| | - M L K Chua
- Oncology Academic Clinical Programme, Duke-NUS Medical School, Singapore; Division of Radiation Oncology, National Cancer Centre Singapore, Singapore; Division of Medical Sciences, National Cancer Centre Singapore, Singapore.
| |
Collapse
|
50
|
He K, Barsoumian HB, Sezen D, Puebla-Osorio N, Hsu EY, Verma V, Abana CO, Chen D, Patel RR, Gu M, Cortez MA, Welsh JW. Pulsed Radiation Therapy to Improve Systemic Control of Metastatic Cancer. Front Oncol 2021; 11:737425. [PMID: 34497773 PMCID: PMC8419338 DOI: 10.3389/fonc.2021.737425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/06/2021] [Indexed: 12/25/2022] Open
Abstract
Radiation therapy (RT) is emerging as an interventional modality in the cancer-immunity cycle, augmenting the activation of an adaptive immune response against tumors. RT, particularly in combination with immunotherapy, can enhance immune memory effects and shape the tumor-directed T-cell populations. However, a single cycle of RT delivered to a limited number of polymetastatic lesions is rarely sufficient to achieve systemic control. We hypothesize that several rounds of RT, akin to several rounds of immunotherapeutic drugs, is likely to provide greater clinical benefit to patients with metastatic disease. We propose that the repeated exposure to tumor antigens released by “pulsed-RT” (i.e., treating 2-4 tumor lesions with 3 irradiation cycles given one month apart) may amplify the adaptive immune response by expanding the tumor-specific T-cell receptor repertoire, the production of high-affinity tumor antibodies, and the generation of memory lymphocytes and thereby improve immune control of systemic disease.
Collapse
Affiliation(s)
- Kewen He
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Duygu Sezen
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Radiation Oncology, Koç University School of Medicine, Istanbul, Turkey
| | - Nahum Puebla-Osorio
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ethan Y Hsu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vivek Verma
- Department of Radiation Oncology, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Chike O Abana
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dawei Chen
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | | | - Meidi Gu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|