1
|
Ma F, Miao X, Lin Y, Luo X, Chen T, Ni Z, Wang X. Microwave ablation combined with α-PD-L1 enhances abscopal effect and promotes CTL activation and intratumoral homing by a cytokine network involving IFN-γ and CXCL9. Int Immunopharmacol 2025; 153:114498. [PMID: 40101420 DOI: 10.1016/j.intimp.2025.114498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/26/2025] [Accepted: 03/13/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND AND PURPOSE Microwave ablation (MWA) has demonstrated promising potential in instigating an anti-tumor immune response, potentially resulting in the regression of distant tumors. Nevertheless, the observed abscopal effect remains modest, and its underlying mechanism remains poorly elucidated. This study aims to systematically examine the utilization of α-PD-L1 to enhance the abscopal effect induced by MWA and to comprehensively investigate the associated mechanisms. METHODS The Lewis lung cancer model was employed to investigate the abscopal effect of MWA and α-PD-L1 co-treatment. The maturation status of bone marrow-derived dendritic cells (BMDCs) was observed after stimulating with the fragments of tumor cells harvested by microwave treatment. Flow cytometry analysis was employed to scrutinize the ratio of T cells and dendritic cells, along with the quantification of IFN-γ and GzmB expression levels. The expression of CXCL9 or PD-L1 was detected by immunofluorescence. RESULTS Local MWA treatment on one site of tumor did not yield a significant reduction in the volume of distant tumors. However, when combined with intraperitoneal α-PD-L1 injection, a notable abscopal effect was induced, resulting in decreased distant tumor volume and prolonged survival in mice. Flow cytometry analysis revealed an increase in infiltrating and activated CD8+ T cells in distant tumors, with up-regulation of IFN-γ and GzmB expression. Notably, the expression of CXCR3 was increased in CD8+ T cells. The mRNA and protein levels of CXCL9 were elevated within distant tumors, concomitant with increased infiltration of macrophages. Moreover, dendritic cells (DCs) in tumor-draining lymph nodes (TDLNs) exhibited enhanced maturation. Additionally, in vitro experiments showed enhanced maturation of BMDCs following stimulation with fragments of Lewis cells harvested via microwave treatment. CONCLUSIONS MWA exhibited the potential to stimulate systemic immune responses, it did not lead to a significant abscopal effect. However, the combination of MWA with α-PD-L1 treatment effectively induced the occurrence of the abscopal effect. This phenomenon appears to be linked to the activation of cytotoxic T lymphocytes (CTLs), phagocytosis by macrophages, and the maturation of DCs, facilitated by a cytokine network involving IFN-γ and CXCL9.
Collapse
Affiliation(s)
- Fuqi Ma
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Xiayi Miao
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Yuhua Lin
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Xuming Luo
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Teng Chen
- Department Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China.
| | - Zhenhua Ni
- Experimental Medicine Centre, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China.
| | - Xiongbiao Wang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| |
Collapse
|
2
|
Rafiq Z, Kang M, Barsoumian HB, Manzar GS, Hu Y, Leuschner C, Huang A, Masrorpour F, Lu W, Puebla-Osorio N, Welsh JW. Enhancing immunotherapy efficacy with synergistic low-dose radiation in metastatic melanoma: current insights and prospects. J Exp Clin Cancer Res 2025; 44:31. [PMID: 39881333 PMCID: PMC11781074 DOI: 10.1186/s13046-025-03281-2] [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: 09/25/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
Recent advances in oncology research have highlighted the promising synergy between low-dose radiation therapy (LDRT) and immunotherapies, with growing evidence highlighting the unique benefits of the combination. LDRT has emerged as a potent tool for stimulating the immune system, triggering systemic antitumor effects by remodeling the tumor microenvironment. Notably, LDRT demonstrates remarkable efficacy even in challenging metastatic sites such as the liver (uveal) and brain (cutaneous), particularly in advanced melanoma stages. The increasing interest in utilizing LDRT for secondary metastatic sites of uveal, mucosal, or cutaneous melanomas underscores its potential efficacy in combination with various immunotherapies. This comprehensive review traverses the journey from laboratory research to clinical applications, elucidating LDRT's immunomodulatory role on the tumor immune microenvironment (TIME) and systemic immune responses. We meticulously examine the preclinical evidence and ongoing clinical trials, throwing light on the promising prospects of LDRT as a complementary therapy in melanoma treatment. Furthermore, we explore the challenges associated with LDRT's integration into combination therapies, addressing crucial factors such as optimal dosage, fractionation, treatment frequency, and synergy with other pharmacological agents. Considering its low toxicity profile, LDRT presents a compelling case for application across multiple lesions, augmenting the antitumor immune response in poly-metastatic disease scenarios. The convergence of LDRT with other disciplines holds immense potential for developing novel radiotherapy-combined modalities, paving the way for more effective and personalized treatment strategies in melanoma and beyond. Moreover, the dose-related toxicities of immunotherapies may be reduced by synergistic amplification of antitumor efficacy with LDRT.
Collapse
Affiliation(s)
- Zahid Rafiq
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79968, USA
| | - Mingyo Kang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gohar S Manzar
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yun Hu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Carola Leuschner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ailing Huang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Fatemeh Masrorpour
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Weiqin Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79968, USA
| | - Nahum Puebla-Osorio
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
3
|
Eapen RS, Williams SG, Macdonald S, Keam SP, Lawrentschuk N, Au L, Hofman MS, Murphy DG, Neeson PJ. Neoadjuvant lutetium PSMA, the TIME and immune response in high-risk localized prostate cancer. Nat Rev Urol 2024; 21:676-686. [PMID: 39112733 DOI: 10.1038/s41585-024-00913-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 11/02/2024]
Abstract
High-risk localized prostate cancer remains a lethal disease with high rates of recurrence, metastases and death, despite attempts at curative local treatment including surgery. Disease recurrence is thought to be a result of failure of local control and occult micrometastases. Neoadjuvant strategies before surgery have been effective in many cancers, but, to date, none has worked in this setting for prostate cancer. Prostate-specific membrane antigen (PSMA)-based theranostics is an exciting and rapidly evolving field in prostate cancer. The novel intravenous radionuclide therapy, [177Lu]Lu-PSMA-617 (lutetium PSMA) has been shown to be effective in treating men with metastatic castration-resistant prostate cancer, targeting cells expressing PSMA throughout the body. When given in a neoadjuvant setting, lutetium PSMA might also improve long-term oncological outcomes in men with high-risk localized disease. A component of radiotherapy is potentially an immunogenic form of cancer cell death. Lutetium PSMA could cause cancer cell death, resulting in release of tumour antigens and induction of a tumour-specific systemic immune response. This targeted radioligand treatment has the potential to treat local and systemic tumour sites by directly targeting cells that express PSMA, but might also act indirectly via this systemic immune response. In selected patients, lutetium PSMA could potentially be combined with systemic immunotherapies to augment the antitumour T cell response, and this might produce long-lasting immunity in prostate cancer.
Collapse
Affiliation(s)
- Renu S Eapen
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia.
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia.
| | - Scott G Williams
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Sean Macdonald
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Simon P Keam
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Nathan Lawrentschuk
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Lewis Au
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Michael S Hofman
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Declan G Murphy
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Paul J Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
4
|
Yu L, Zou R, He J, Qu C. Role of radiation in chimeric antigen receptor T-cell therapy for patients with relapsed/refractory non-Hodgkin lymphoma: Current studies and future prospects. Crit Rev Oncol Hematol 2024; 199:104390. [PMID: 38782146 DOI: 10.1016/j.critrevonc.2024.104390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment approach for patients with relapsed/refractory non-Hodgkin lymphoma (R/R NHL). However, the long-term prognosis has been discouraging. Moreover, the urgent resolution of two critical issues is necessary: minimize tumor burden before CAR-T infusion and control fatal toxicities post CAR-T therapy. By combining radiotherapy (RT), the safety and efficacy of CAR-T can be improved. RT can serve as bridging therapy, reducing the tumor burden before CAR-T infusion, thus enabling safe and successful CAR-T infusion, and as salvage therapy in cases of CAR-T therapy failure. This review aims to discuss the current evidence supporting the use of RT in CAR-T therapy for patients with R/R NHL. Although most studies have shown a positive role of RT in combined modality treatments for patients undergoing CAR-T therapy, the synergy gained from these remains uncertain. Furthermore, the optimal dose/fraction and radiation response require further investigation.
Collapse
Affiliation(s)
- Lingzi Yu
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| | - Rui Zou
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| | - Jiajie He
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| | - Changju Qu
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| |
Collapse
|
5
|
Li L, Chen L, Fan M, Tian Y, Ai H, Yan L, Li F, Lan M, Lai X, Huang Y, Xu P, Feng M, Lang J. A prospective, single-arm trial of PD-1 inhibitors plus chemoradiotherapy for solitary metachronous metastasis nasopharyngeal carcinoma. Oral Oncol 2024; 150:106695. [PMID: 38262250 DOI: 10.1016/j.oraloncology.2024.106695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/25/2024]
Abstract
PURPOSE Initial treatment for Recurrent/Metastatic Nasopharyngeal Carcinoma (R/M NPC) often involves Gemcitabine plus cisplatin with or without PD-1 inhibitors. However, PD-1 inhibitors' effectiveness varies, prompting for better treatments. This study explores effect and safety of combining PD-1 inhibitors with chemoradiotherapy for oligometastatic NPC patients. METHODS Oligometastatic NPC patients underwent radical treatment with PD-1 inhibitors and chemotherapy, followed by concurrent PD-1 inhibitors and chemoradiotherapy, and then maintenance PD-1 inhibitors. Objective response rate (ORR) and disease control rate (DCR) were calculated by irRECIST-1.1, and CTCAE-4.0 was used to evaluate the toxicity. RESULTS The study enrolled 47 patients with a median age of 46. The median follow-up lasted 16.5 months, with metastatic lesions receiving a median radiation dose of 45 Gy. The median courses of PD-1 inhibitors and chemotherapy were 9.5 and 5 respectively. The metastasis sites included lung (40.8 %), liver (21.1 %), mediastinal lymph node (7.9 %), abdominal lymph nodes (3.9 %), bone (21.1 %), adrenal gland (3.9 %), and brain (1.3 %). ORR and DCR were 85.1 % and 100 % at 3 months after radiotherapy. The median survival was not reached yet, and 1 and 2-year OS rates were 93.1 % and 78.4 %. The median PFS was 18 months, with 1 and 2-year PFS rates of 70.2 % and 47.7 % respectively. PD-L1 expression showed a positive correlation for PFS. Twenty-five patients experienced grade 3 or 4 adverse events (AE) that were possibly related to chemotherapy. No grade 5 AE was observed. CONCLUSIONS The synergy of concurrent PD-1 inhibitors and chemoradiotherapy shows promising efficacy and an acceptable toxicity for oligometastasis NPC patients.
Collapse
Affiliation(s)
- Lu Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Lu Chen
- Leshan People's Hospital, Leshan, China
| | - Ming Fan
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yue Tian
- North Sichuan Medical College, Nanchong, China
| | - Hangyu Ai
- Santai County People's Hospital, Mianyang, China
| | - Lu Yan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Mei Lan
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xin Lai
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yecai Huang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Xu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Mei Feng
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China; Department of Medical Oncology, the Third People's Hospital of Sichuan, Chengdu, China.
| | - Jinyi Lang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
6
|
Pontoriero A, Critelli P, Chillari F, Ferrantelli G, Sciacca M, Brogna A, Parisi S, Pergolizzi S. Modulation of Radiation Doses and Chimeric Antigen Receptor T Cells: A Promising New Weapon in Solid Tumors-A Narrative Review. J Pers Med 2023; 13:1261. [PMID: 37623511 PMCID: PMC10455986 DOI: 10.3390/jpm13081261] [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: 06/29/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
Tumor behavior is determined by its interaction with the tumor microenvironment (TME). Chimeric antigen receptor (CART) cell therapy represents a new form of cellular immunotherapy (IT). Immune cells present a different sensitivity to radiation therapy (RT). RT can affect tumor cells both modifying the TME and inducing DNA damage, with different effects depending on the low and high doses delivered, and can favor the expression of CART cells. CART cells are patients' T cells genetically engineered to recognize surface structure and to eradicate cancer cells. High-dose radiation therapy (HDRT, >10-20 Gy/fractions) converts immunologically "cold" tumors into "hot" ones by inducing necrosis and massive inflammation and death. LDRT (low-dose radiation therapy, >5-10 Gy/fractions) increases the expansion of CART cells and leads to non-immunogenetic death. An innovative approach, defined as the LATTICE technique, combines a high dose in higher FDG- uptake areas and a low dose to the tumor periphery. The association of RT and immune checkpoint inhibitors increases tumor immunogenicity and immune response both in irradiated and non-irradiated sites. The aim of this narrative review is to clarify the knowledge, to date, on CART cell therapy and its possible association with radiation therapy in solid tumors.
Collapse
Affiliation(s)
- Antonio Pontoriero
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Paola Critelli
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Federico Chillari
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Giacomo Ferrantelli
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Miriam Sciacca
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Anna Brogna
- Radiotherapy Unit, Medical Physics Unit, A.O.U. “G. Martino”, 98125 Messina, Italy;
| | - Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| |
Collapse
|
7
|
Paganetti H. A review on lymphocyte radiosensitivity and its impact on radiotherapy. Front Oncol 2023; 13:1201500. [PMID: 37601664 PMCID: PMC10435323 DOI: 10.3389/fonc.2023.1201500] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
It is well known that radiation therapy causes lymphopenia in patients and that this is correlated with a negative outcome. The mechanism is not well understood because radiation can have both immunostimulatory and immunosuppressive effects. How tumor dose conformation, dose fractionation, and selective lymph node irradiation in radiation therapy does affect lymphopenia and immune response is an active area of research. In addition, understanding the impact of radiation on the immune system is important for the design and interpretation of clinical trials combining radiation with immune checkpoint inhibitors, both in terms of radiation dose and treatment schedules. Although only a few percent of the total lymphocyte population are circulating, it has been speculated that their increased radiosensitivity may contribute to, or even be the primary cause of, lymphopenia. This review summarizes published data on lymphocyte radiosensitivity based on human, small animal, and in vitro studies. The data indicate differences in radiosensitivity among lymphocyte subpopulations that affect their relative contribution and thus the dynamics of the immune response. In general, B cells appear to be more radiosensitive than T cells and NK cells appear to be the most resistant. However, the reported dose-response data suggest that in the context of lymphopenia in patients, aspects other than cell death must also be considered. Not only absolute lymphocyte counts, but also lymphocyte diversity and activity are likely to be affected by radiation. Taken together, the reviewed data suggest that it is unlikely that radiation-induced cell death in lymphocytes is the sole factor in radiation-induced lymphopenia.
Collapse
Affiliation(s)
- Harald Paganetti
- Department of Radiation Oncology, Massachusetts General Hospital, Boston MA, United States
- Harvard Medical School, Boston MA, United States
| |
Collapse
|
8
|
Khawar MB, Gao G, Rafiq M, Shehzadi A, Afzal A, Abbasi MH, Sheikh N, Afzal N, Ashraf MA, Hamid SE, Shahzaman S, Kawish N, Sun H. Breaking down barriers: The potential of smarter CAR-engineered NK cells against solid tumors. J Cell Biochem 2023; 124:1082-1104. [PMID: 37566723 DOI: 10.1002/jcb.30460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/04/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
Natural killer (NK) cells are considered to be the foremost fighters of our innate immune system against foreign invaders and thus tend to promptly latch onto the virus-infected and tumor/cancerous cells, killing them through phagocytosis. At present, the application of genetically engineered Chimeric antigen receptor (CAR) receptors ensures a guaranteed optimistic response with NK cells and would not allow the affected cells to dodge or escape unchecked. Hence the specificity and uniqueness of CAR-NK cells over CAR-T therapy make them a better immunotherapeutic choice to reduce the load of trafficking of numerous tumor cells near the healthy cell populations in a more intact way than offered by CAR-T immunotherapy. Our review mainly focuses on the preclinical, clinical, and recent advances in clinical research trials and further strategies to achieve an augmented and efficient cure against solid tumors.
Collapse
Affiliation(s)
- Muhammad B Khawar
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental and, Translational Noncoding RNA Research, Yangzhou, China
- Department of Zoology, Applied Molecular Biology and Biomedicine Lab, University of Narowal, Narowal, Pakistan
| | - Guangzhong Gao
- Department of Physiatry, Haian Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nantong, Jiangsu, China
| | - Mussarat Rafiq
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Anila Shehzadi
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Ali Afzal
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | | | - Nadeem Sheikh
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Nimra Afzal
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | | | - Syeda E Hamid
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Sara Shahzaman
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Naseer Kawish
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Haibo Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental and, Translational Noncoding RNA Research, Yangzhou, China
| |
Collapse
|
9
|
Li L, Chen L, Yan L, Guo Y, Li F, Fan M, Lan M, Lai X, Zhou J, Huang Y, Xu P, Lang J, Feng M. Initial analysis of the synergy of programmed cell death-1 (PD-1) inhibitor and concurrent chemoradiotherapy treatment for recurrent/metastatic head and neck squamous cell carcinoma patients. Radiat Oncol 2023; 18:109. [PMID: 37403098 DOI: 10.1186/s13014-023-02310-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 06/26/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Programmed cell death-1 (PD-1) inhibitor was proven to be useful for the recurrent/metastatic head and neck squamous carcinoma (R/M HNSCC) patients. Though both PD-1 inhibitor alone and combination with chemotherapy showed some benefit for PFS and OS, the survival outcome was still not satisfactory. Some studies showed the possible benefit for PD-1 inhibitors combination with radiation for head and neck squamous carcinoma, however there was few studies concerned about synergy of concurrent PD-1 inhibitor combination with chemoradiotherapy for R/M HNSCC. So, we aimed to explore the potential effect and toxicity of the concurrent PD-1 inhibitor and chemoradiotherapy for R/M HNSCC. METHODS We consecutively enrolled the R/M HNSCC patients treated with concurrent PD-1 inhibitor and chemoradiotherapy from August 2018 to April 2022 in Sichuan Cancer hospital. All the patients received the combination of PD-1 inhibitor and chemotherapy, and followed with synergy of concurrent PD-1 inhibitor and chemoradiotherapy, then maintenance PD-1 inhibitor. ORR and DCR was calculated by immune-related Response Evaluation Criteria in Solid Tumors (irRECIST-1.1), and Common terminology criteria for adverse events (CTCAE-4.0) was used to evaluate the toxicity.The Kaplan-Meier method was used to analyze OS and PFS. RESULTS 40 R/M HNSCC patients were enrolled in our stuty. The median follow up time was 14 months. 22 patients had recurrent disease only, 16 patients had metastatic disease only, and 2 patients had both recurrence and metastasis disease. For the recurrent lesions, 23 patients received a median radiation dose of 64 Gy (range 50-70 Gy). 18 patients received a median dose of 45 Gy (range 30-66 Gy) for metastatic lesions. The median courses of PD-1 inhibitors and chemotherapy were 8 and 5 respectively. After the treatment, the ORR and DCR were 70.0% and 100%. The median OS was 19 months (range 6.3-31.7 months), with 1 and 2-years OS rates of 72.8% and 33.3%. The median PFS was 9 months (range 3.1-14.9 months), with 6 and 12 months PFS rates of 75.5% and 41.4% respectively. The PFS had no statistical significance in PD-L1 negative and positive group (7 vs 12 months, p = 0.059). The most common grade 3 or 4 adverse events(AE) were leucopenia (25.0%), neutropenia (17.5%), anemia (10.0%), thrombocytopenia (5.0%), hyponatremia (2.5%), and pneumonia(2.5%). No grade 5 AE was observed. CONCLUSIONS The synergy of concurrent PD-1 inhibitor treatment with chemoradiotherapy shows promise as a treatment strategy and an acceptable toxicity for the R/M HNSCC patients.
Collapse
Affiliation(s)
- Lu Li
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Chen
- Chengdu Medical College, Chengdu, China
| | - Lu Yan
- Chengdu Medical College, Chengdu, China
| | - Yueqian Guo
- Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Fang Li
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ming Fan
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Mei Lan
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xin Lai
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jie Zhou
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yecai Huang
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Xu
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinyi Lang
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Mei Feng
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
- Department of Clinical Oncology, the Third People's Hospital of Sichuan, Chengdu, China.
| |
Collapse
|
10
|
Li J, Chen Y, Fan Y, Wang H, Mu W, Liu X. Radiotherapy combined with anti-CEACAM1 immunotherapy to induce survival advantage in glioma. Discov Oncol 2023; 14:32. [PMID: 36928507 PMCID: PMC10020409 DOI: 10.1007/s12672-023-00638-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND We aimed to observe the effect of radiotherapy on the expression of immune checkpoint molecule CEACAM1 in patients with glioma and the therapeutical effect of radiotherapy combined with blockade of CEACAM1 in mice with intracranial gliomas. METHODS The expression of CEACAM1 on T-lymphocytes in the peripheral blood of patients with glioma was detected before and after radiotherapy; GL261 murine glioma cells (stably transfected with the luciferase gene) were implanted in the right caudate nucleus of C57BL/6 mice, and tumour growth was observed using the small animal in vivo imaging system. Mice were divided into 4 groups: (1) the isotype control; (2) the radiotherapy; (3) the anti-CEACAM1 treatment; and (4) the combination therapy. The survival of mice after treatment was recorded; the expression of CEACAM1 on murine glioma cells was detected by immunohistochemistry before and after radiotherapy; flow cytometry was adopted to detect CD8+ T-cells (Treg) (CD4+FoxP3+CD25+) among mouse brain-infiltrating T-cells; serum levels of IFN-γ and IL-10 were detected by ELISA; proliferation and apoptosis were observed by immunohistochemistry; Retrospective RNA-seq data analysis was conducted in a cohort of 325 patients with glioma in the Chinese Glioma Genome Atlas (CGGA) database and 702 patients in The Cancer Genome Atlas (TCGA) database. RESULTS The expression of CEACAM1 on CD4+ and CD8+ T-cells in the peripheral blood of patients with glioma was significantly higher 1 week after radiotherapy than before radiotherapy and was further increased 1 month after radiotherapy. Combined therapy notably inhibited the growth of intracranial tumours in mice and prolonged their survival time, with some mice being capable of surviving long-term (> 90 d). Immunohistochemistry revealed that the expression of CEACAM1 in murine glioma tissues after radiotherapy was elevated in a time-dependent manner. Flow cytometry analysis showed an increase in mouse brain-infiltrating CD8+ T-lymphocytes, a decrease in Treg cells, and an increase in CD8+ T/Treg cells after treatment. ELISA demonstrated the elevated levels of IFN and decreased levels of IL-10 in the serum of mice in the combination therapy group. CONCLUSIONS Radiotherapy combined with CEACAM1 inhibitors resulted in strong and durable anti-tumour immune responses against murine glioma and long-term survival of some mice. Hence, this study is expected to offer new effective immunotherapy strategies against glioma.
Collapse
Affiliation(s)
- Jinhu Li
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan, Shanxi, China
| | - Yi Chen
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan, Shanxi, China
| | - Yimin Fan
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan, Shanxi, China
| | - Hongqin Wang
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan, Shanxi, China
| | - Wei Mu
- Department of Interventional Radiology, Shanxi Provincial People's Hospital, Taiyuan, China.
| | - Xiaodong Liu
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan, Shanxi, China.
| |
Collapse
|
11
|
Yegya-Raman N, Wright CM, LaRiviere MJ, Baron JA, Lee DY, Landsburg DJ, Svoboda J, Nasta SD, Gerson JN, Barta SK, Chong EA, Schuster SJ, Maity A, Facciabene A, Paydar I, Plastaras JP. Salvage radiotherapy for relapsed/refractory non-Hodgkin lymphoma following CD19 chimeric antigen receptor T-cell (CART) therapy. Clin Transl Radiat Oncol 2023; 39:100587. [PMID: 36718252 PMCID: PMC9883177 DOI: 10.1016/j.ctro.2023.100587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/18/2022] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Background and purpose CD19-targeting chimeric antigen receptor T-cell (CART) therapy is a promising treatment for relapsed/refractory non-Hodgkin lymphoma, but most patients experience post-CART progression. We describe our institutional experience of salvage radiotherapy (SRT) in this setting. Materials and methods Of 94 patients who received CART therapy from 2018 to 2020, 21 received SRT for post-CART progression. Patients were divided into two groups: locoregional disease (n = 9 [43 %], all disease encompassable within an RT field) and advanced disease (n = 12 [57 %]). Patterns of failure, progression-free survival (PFS), overall survival (OS), and toxicity were assessed. Results Median time from CART infusion to SRT was 4.0 months (range, 0.6-11.5 months). In the locoregional disease group, 8/9 patients (89 %) were treated with comprehensive SRT to a median dose of 37.5 Gy in a median of 15 fractions. In the advanced disease group, all patients (n = 12) were treated with focal SRT to a median dose of 20.8 Gy in a median of 5 fractions. Median follow-up post-SRT was 15.2 months. In-field response was observed in 8/9 (89 %) in the locoregional disease and 8/9 (89 %) evaluable patients in the advanced disease groups. 17/18 evaluable patients (94 %) patients experienced post-SRT progression, all with a distant component. Median OS was 7.4 months; 21 months for locoregional disease versus 2.4 months for advanced disease (p = 0.0002). Median PFS was 1.1 month, and similarly poor regardless of group. No grade ≥ 3 toxicities occurred. Conclusions SRT post-CART therapy appears safe with encouraging in-field response but high rates of out-of-field progression, even for those presenting with locoregional disease, highlighting the need for integration of novel systemic agents.
Collapse
Affiliation(s)
- Nikhil Yegya-Raman
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Christopher M. Wright
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael J. LaRiviere
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Jonathan A. Baron
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Y. Lee
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel J. Landsburg
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Jakub Svoboda
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Sunita D. Nasta
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - James N. Gerson
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Stefan K. Barta
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Elise A. Chong
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Stephen J. Schuster
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Amit Maity
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Andrea Facciabene
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Ima Paydar
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - John P. Plastaras
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States,Corresponding author at: Department of Radiation Oncology, University of Pennsylvania, PCAM/TRC 4 West, 3400 Civic Center Blvd, Philadelphia, PA 19104, United States.
| |
Collapse
|
12
|
Yu Q, Zhang X, Wang N, Li C, Zhang Y, Zhou J, Wang G, Cao Y. Radiation prior to chimeric antigen receptor T-cell therapy is an optimizing bridging strategy in relapsed/refractory aggressive B-cell lymphoma. Radiother Oncol 2022; 177:53-60. [PMID: 36309153 DOI: 10.1016/j.radonc.2022.10.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/11/2022] [Accepted: 10/15/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE We aimed to analyze the safety and efficacy of a radiation bridging regimen with or without chemotherapy compared with chemotherapy alone prior to CAR T-cell treatment for relapsed/refractory aggressive B-cell lymphoma (r/r ABL). METHODS AND MATERIALS In this study, 45 out of 105 patients enrolled in CD19/22 CAR T-cell "cocktail" clinical trial were excluded, including 34 patients without bridging treatment. Total 60 patients receiving CAR T-cell therapies with bridging regimens as chemotherapy alone (C-CAR-T group, n = 31), and radiotherapy with or without chemotherapy (R-CAR-T group, n = 29) between February 2017 and October 2020 were retrospectively analyzed. RESULTS No significant toxicities were identified in the R-CAR-T group, and no patients in either group experienced CAR-T-related deaths. However, the R-CAR-T group showed a lower incidence of cytokine release syndrome (CRS) of grade ≥ 3 relative to the C-CAR-T group (0% vs 19.4%, P = 0.036). The incidence of neurological toxicity was 9.9% and 6.9% in the C-CAR-T group and R-CAR-T group, respectively (P = 0.697). The R-CAR-T group achieved a higher overall response rate (ORR) at the day 30 assessment (82.8% vs 45.2%, P = 0.0025). Further analyzing the outcomes, the R-CAR-T group presented a better 1-year progression-free survival (PFS) rate than the C-CAR-T group (46.9% vs 22.6%, P = 0.0356). Intriguingly, the bridging radiation regimen extremely improved the 6-month PFS (50.8% vs 16. 7%, P = 0.0369) and 1-year overall survival (OS) (56.3% vs 33.3%, P = 0.0236) rates in patients with bulky disease. The study also found that conducting radiotherapy as a bridging regimen was an independent factor that predicted better PFS (HR: 0.534, 95% CI: 0.289-0.987, P = 0.045). CONCLUSIONS Our results provide and strengthen novel insights that the use of radiotherapy as a bridging strategy was demonstrated to reduce the incidence of severe CRS and improve the PFS of patients. In subgroup analysis, it was confirmed that radiotherapy can improve PFS and OS in patients with bulky disease. These findings open new avenues to improve the efficacy and safety of CAR T-cell therapy.
Collapse
Affiliation(s)
- Qiuxia Yu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaoying Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Na Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Chunrui Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Gaoxiang Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| | - Yang Cao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
13
|
Jiang L, Lyu Q, Abdelhamid AMH, Hui S, Sheng K. An efficient rectangular optimization method for sparse orthogonal collimator based small animal irradiation. Phys Med Biol 2022; 67:10.1088/1361-6560/ac910b. [PMID: 36084625 PMCID: PMC9595432 DOI: 10.1088/1361-6560/ac910b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/09/2022] [Indexed: 11/11/2022]
Abstract
Objective.Intensity-modulated radiotherapy (IMRT) is widely used in clinical radiotherapy, treating varying malignancies with conformal doses. As the test field for clinical translation, preclinical small animal experiments need to mimic the human radiotherapy condition, including IMRT. However, small animal IMRT is a systematic challenge due to the lack of corresponding hardware and software for miniaturized targets.Approach.The sparse orthogonal collimators (SOC) based on the direct rectangular aperture optimization (RAO) substantially simplified the hardware for miniaturization. This study investigates and evaluates a significantly improved RAO algorithm for complex mouse irradiation using SOC. Because the Kronecker product representation of the rectangular aperture is the main limitation of the computational performance, we reformulated matrix multiplication in the data fidelity term using multiplication with small matrices instead of the Kronecker product of the dose loading matrices. Solving the optimization problem was further accelerated using the Fast Iterative Shrinkage-Thresholding Algorithm (FISTA).Main results.Four mouse cases, including a liver, a brain tumor, a concave U-target, and a complex total marrow irradiation (TMI) case, were included in this study with manually delineated targets and OARs. Seven coplanar-field SOC IMRT (sIMRT) plans were compared with idealistic fluence map based IMRT (iIMRT) plans. For the first three cases with simpler and smaller targets, the differences between sIMRT plans and iIMRT plans in the planning target volumes (PTV) statistics are within 1%. For the TMI case, the sIMRT plans are superior in reducing hot spots (also termedDmax) of PTV, kidneys, lungs, heart, and bowel by 20.5%, 31.5%, 24.67%, 20.13%, and 17.78%, respectively. On average, in four cases in this study, the sIMRT plan conformity is comparable to that of the iIMRT's with lightly increased R50 and Integral Dose by 2.23% and 2.78%.Significance.The significantly improved sIMRT optimization method allows fast plan creation in under 1 min for smaller targets and makes complex TMI planning feasible while achieving comparable dosimetry to idealistic IMRT with fluence map optimization.
Collapse
Affiliation(s)
- Lu Jiang
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Qihui Lyu
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Amr M H Abdelhamid
- Department of Radiation Oncology, City of Hope Medical Center, Duarte, CA, United States of America
| | - Susanta Hui
- Department of Radiation Oncology, City of Hope Medical Center, Duarte, CA, United States of America
| | - Ke Sheng
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, United States of America
| |
Collapse
|
14
|
An International Consensus on the Design of Prospective Clinical–Translational Trials in Spatially Fractionated Radiation Therapy for Advanced Gynecologic Cancer. Cancers (Basel) 2022; 14:cancers14174267. [PMID: 36077802 PMCID: PMC9454841 DOI: 10.3390/cancers14174267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Spatially fractionated radiation therapy (SFRT) delivers intentionally heterogenous dose to tumors. This is a major departure from current radiation therapy, which strives for uniform dose. Early pilot experience suggests promising treatment outcomes with SFRT in patients with challenging bulky tumors, including gynecologic cancer. Well-conducted prospective multi-institutional clinical trials are now needed to further test SFRT as a treatment modality. However, clinical trial development is hampered by the variabilities in SFRT approach and the overall unfamiliarity with heterogeneous dosing. A broad consensus among SFRT experts, potential investigators, and the wider radiation oncology community is needed so that clinical trials in SFRT can be successfully designed and carried out. We developed an international consensus guideline for the design parameters of clinical/translational trials in SFRT for gynecologic cancer. High-to-moderate consensus was achieved, and harmonized fundamental design parameters for SFRT trials in advanced gynecologic cancer were defined. Abstract Despite the unexpectedly high tumor responses and limited treatment-related toxicities observed with SFRT, prospective multi-institutional clinical trials of SFRT are still lacking. High variability of SFRT technologies and methods, unfamiliar complex dose and prescription concepts for heterogeneous dose and uncertainty regarding systemic therapies present major obstacles towards clinical trial development. To address these challenges, the consensus guideline reported here aimed at facilitating trial development and feasibility through a priori harmonization of treatment approach and the full range of clinical trial design parameters for SFRT trials in gynecologic cancer. Gynecologic cancers were evaluated for the status of SFRT pilot experience. A multi-disciplinary SFRT expert panel for gynecologic cancer was established to develop the consensus through formal panel review/discussions, appropriateness rank voting and public comment solicitation/review. The trial design parameters included eligibility/exclusions, endpoints, SFRT technology/technique, dose/dosimetric parameters, systemic therapies, patient evaluations, and embedded translational science. Cervical cancer was determined as the most suitable gynecologic tumor for an SFRT trial. Consensus emphasized standardization of SFRT dosimetry/physics parameters, biologic dose modeling, and specimen collection for translational/biological endpoints, which may be uniquely feasible in cervical cancer. Incorporation of brachytherapy into the SFRT regimen requires additional pre-trial pilot investigations. Specific consensus recommendations are presented and discussed.
Collapse
|
15
|
Randrian V, Pernot S, Le Malicot K, Catena V, Baumgaertner I, Tacher V, Forestier J, Hautefeuille V, Tabouret-Viaud C, Gagnaire A, Mitry E, Guiu B, Aparicio T, Smith D, Dhomps A, Tasu JP, Perdrisot R, Edeline J, Capron C, Cheze-Le Rest C, Emile JF, Laurent-Puig P, Bejan-Angoulvant T, Sokol H, Lepage C, Taieb J, Tougeron D. FFCD 1709-SIRTCI phase II trial: Selective internal radiation therapy plus Xelox, Bevacizumab and Atezolizumab in liver-dominant metastatic colorectal cancer. Dig Liver Dis 2022; 54:857-863. [PMID: 35610167 DOI: 10.1016/j.dld.2022.04.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/09/2022] [Accepted: 04/22/2022] [Indexed: 12/24/2022]
Abstract
Immune checkpoint inhibitors (ICI) have high efficacy in metastatic colorectal cancer (mCRC) with microsatellite instability (MSI) but not in microsatellite stable (MSS) tumour due to the low tumour mutational burden. Selective internal radiation therapy (SIRT) could enhance neoantigen production thus triggering systemic anti-tumoral immune response (abscopal effect). In addition, Oxalipatin can induce immunogenic cell death and Bevacizumab can decrease the exhaustion of tumour infiltrating lymphocyte. In combination, these treatments could act synergistically to sensitize MSS mCRCs to ICI SIRTCI is a prospective, multicentre, open-label, phase II, non-comparative single-arm study evaluating the efficacy and safety of SIRT plus Xelox, Bevacizumab and Atezolizumab (anti-programmed death-ligand 1) in patients with liver-dominant MSS mCRC. The primary objective is progression-free survival at 9 months. The main inclusion criteria are patients with MSS mCRC with liver-dominant disease, initially unresectable disease and with no prior oncologic treatment for metastatic disease. The trial started in November 2020 and has included 10 out of the 52 planned patients.
Collapse
Affiliation(s)
- Violaine Randrian
- Service d'Hépato-gastroentérologie, CHU de Poitiers et Université de Poitiers, Poitiers 86021, France
| | - Simon Pernot
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Karine Le Malicot
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, University of Burgundy and Franche Comté, Dijon, France
| | - Vittorio Catena
- Department of Radiology, Institut Bergonié, Bordeaux, France
| | | | - Vania Tacher
- University of Paris Est Créteil, Unité INSERM 955, Equipe 18, AP-HP, Hôpitaux Universitaires Henri Mondor, Créteil F-94010, France
| | - Julien Forestier
- Department of Medical Oncology, Hôpital Edouard Herriot, Lyon Cedex 03 69437, France
| | - Vincent Hautefeuille
- Department of Hepato-Gastroenterology and Digestive Oncology, Amiens University Hospital, Amiens, France
| | - Claire Tabouret-Viaud
- Department of Nuclear Medicine, Unicancer-Georges François Leclerc Cancer Center, Dijon, France
| | - Alice Gagnaire
- Department of Hepato-Gastroenterology and Digestive Oncology, Dijon University Hospital, BP 87900 21079 Dijon, EPICAD LNC-UMR1231, Burgundy and Franche-Comte University, Dijon, France
| | - Emmanuel Mitry
- Medical Oncology Department, Paoli-Calmettes Institut, Marseille, France
| | - Boris Guiu
- Hôpital St-Eloi (CHU Montpellier), Université de Montpellier, Montpellier, France
| | - Thomas Aparicio
- AP-HP, Gastroenterology and Digestive Oncology Department, Saint Louis Hospital, 1 avenue Claude Vellefaux, Université de Paris, Paris F-75010, France
| | - Denis Smith
- Service d'Oncologie médicale, Haut-Lévèque Hospital, CHU Bordeaux, Bordeaux, France
| | - Anthony Dhomps
- Nuclear Medicine, University Hospital of Lyon, Pierre Bénite, France
| | - Jean-Pierre Tasu
- Radiology Department, University Hospital Centre Poitiers, Poitiers, France; LATIM, INSERM UMR 1101, Université de Brest, CHU Morvan, 2 avenue FOCH, 29 609 Brest cedex, France
| | - Rémy Perdrisot
- Nuclear Medicine, Poitiers University Hospital, Poitiers France
| | - Julien Edeline
- Medical Oncology, Centre Eugène Marquis, Rennes 35000, France
| | - Claude Capron
- Service d'immunologie, AP-HP, Hôpital Ambroise Paré, Paris, France
| | - Catherine Cheze-Le Rest
- LATIM, INSERM UMR 1101, Université de Brest, CHU Morvan, 2 avenue FOCH, 29 609 Brest cedex, France; Nuclear Medicine, Poitiers University Hospital, Poitiers France
| | - Jean-François Emile
- Department of Pathology, APHP-Hôpital Ambroise Paré, Boulogne-Billancourt, France
| | - Pierre Laurent-Puig
- Department of Biology, Georges Pompidou Hospital, APHP, Université de Paris, Paris, France
| | | | - Harry Sokol
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Gastroentérologie, Paris, Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas and Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Come Lepage
- Department of Hepato-Gastroenterology and Digestive Oncology, Dijon University Hospital, BP 87900 21079 Dijon, EPICAD LNC-UMR1231, Burgundy and Franche-Comte University, Dijon, France
| | - Julien Taieb
- Service de Gastroentérologie et d'Oncologie Digestive, Hôpital Européen George Pompidou, Université de Paris, AP-HP, Paris, France
| | - David Tougeron
- Service d'Hépato-gastroentérologie, CHU de Poitiers et Université de Poitiers, Poitiers 86021, France.
| |
Collapse
|
16
|
Vascular bursts-mediated tumor accumulation and deep penetration of spherical nucleic acids for synergistic radio-immunotherapy. JOURNAL OF CONTROLLED RELEASE : OFFICIAL JOURNAL OF THE CONTROLLED RELEASE SOCIETY 2022; 348:1050-1065. [PMID: 35750133 DOI: 10.1016/j.jconrel.2022.06.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 11/22/2022]
Abstract
While nanomedicines have attracted great interests for tumor therapy, their targeting and intra-tumoral penetrating efficiencies have been questioned. Here, we report a two-step low-dose radiotherapy (RT) strategy to realize significant accumulation and deep penetration of spherical nucleic acids (SNAs)-based nanomedicine for synergistic radio-immunotherapy. The first step RT was employed to recruit large amounts of macrophages into tumor. The tumor infiltrated macrophages not only served as nanoparticles drug depots, but also elicited dynamic bursts extravasation to enhance nanoparticles accumulation. We optimized the spatiotemporal combination of RT and SNAs administration for higher level of SNAs delivery, and the delivered SNAs promote M2-to-M1 phenotype switch of macrophages to increase phagocytosis of nanoparticles by 6-fold, resulting in positive feedback with even higher accumulation and intra-tumor penetration of SNAs. Through vascular bursts and macrophage repolarization, as high as 25-fold enhancement of nanoparticles accumulation was achieved as compared to passive targeting of nanoparticles, and the nanoparticles were eventually distributed throughout the tumor tissue with efficient deep penetration. Finally, SNAs in tumor simultaneously sensitized the second dose of RT and remodeled tumor immune microenvironment, resulting in a synergistic anticancer therapy in combination of anti-PD-L1 antibody (αPD-L1) with no noticeable side effects caused by either RT or αPD-L1.
Collapse
|
17
|
Jiang Y, Wen W, Yang F, Han D, Zhang W, Qin W. Prospect of Prostate Cancer Treatment: Armed CAR-T or Combination Therapy. Cancers (Basel) 2022; 14:cancers14040967. [PMID: 35205714 PMCID: PMC8869943 DOI: 10.3390/cancers14040967] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/29/2022] [Accepted: 02/12/2022] [Indexed: 02/06/2023] Open
Abstract
The incidence rate of prostate cancer is higher in male cancers. With a hidden initiation of disease and long duration, prostate cancer seriously affects men's physical and mental health. Prostate cancer is initially androgen-dependent, and endocrine therapy can achieve good results. However, after 18-24 months of endocrine therapy, most patients eventually develop castration-resistant prostate cancer (CRPC), which becomes metastatic castration resistant prostate cancer (mCRPC) that is difficult to treat. Chimeric Antigen Receptor T cell (CAR-T) therapy is an emerging immune cell therapy that brings hope to cancer patients. CAR-T has shown considerable advantages in the treatment of hematologic tumors. However, there are still obstacles to CAR-T treatment of solid tumors because the physical barrier and the tumor microenvironment inhibit the function of CAR-T cells. In this article, we review the progress of CAR-T therapy in the treatment of prostate cancer and discuss the prospects and challenges of armed CAR-T and combined treatment strategies. At present, there are still many obstacles in the treatment of prostate cancer with CAR-T, but when these obstacles are solved, CAR-T cells can become a favorable weapon for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Yao Jiang
- Department of Urology, First Affiliated Hospital of Air Force Military Medical University, Xi’an 710032, China; (Y.J.); (F.Y.); (D.H.)
| | - Weihong Wen
- Department of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Correspondence: (W.W.); (W.Q.)
| | - Fa Yang
- Department of Urology, First Affiliated Hospital of Air Force Military Medical University, Xi’an 710032, China; (Y.J.); (F.Y.); (D.H.)
| | - Donghui Han
- Department of Urology, First Affiliated Hospital of Air Force Military Medical University, Xi’an 710032, China; (Y.J.); (F.Y.); (D.H.)
| | - Wuhe Zhang
- Department of Urology, Air Force 986 Hospital, Xi’an 710054, China;
| | - Weijun Qin
- Department of Urology, First Affiliated Hospital of Air Force Military Medical University, Xi’an 710032, China; (Y.J.); (F.Y.); (D.H.)
- Correspondence: (W.W.); (W.Q.)
| |
Collapse
|
18
|
Safarzadeh Kozani P, Safarzadeh Kozani P, Rahbarizadeh F. Addressing the obstacles of CAR T cell migration in solid tumors: wishing a heavy traffic. Crit Rev Biotechnol 2021; 42:1079-1098. [PMID: 34957875 DOI: 10.1080/07388551.2021.1988509] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has been recognized as one of the most prosperous treatment options against certain blood-based malignancies. However, the same clinical and commercial success have been out of range in the case of solid tumors. The main contributing factor in this regard is the hostile environment the tumor cells impose that results in the exhaustion of immune effector cells alongside the abrogation of their infiltration capacity. The discovery of the underlying mechanisms and the development of reliable counterstrategies to overcome the inaccessibility of CAR-Ts to their target cells might correlate with encouraging clinical outcomes in advanced solid tumors. Here, we highlight the successive physical and metabolic barriers that systemically administered CAR-Ts face on their journey toward their target cells. Moreover, we propose meticulously-devised countertactics and combination therapies that can be applied to maximize the therapeutic benefits of CAR-T therapies against solid tumors.
Collapse
Affiliation(s)
- Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran.,Student Research Committee, Medical Biotechnology Research Center, School of Nursing, Midwifery, and Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
19
|
Radiation Priming Chimeric Antigen Receptor T-Cell Therapy in Relapsed/Refractory Diffuse Large B-Cell Lymphoma With High Tumor Burden. J Immunother 2021; 43:32-37. [PMID: 31219975 DOI: 10.1097/cji.0000000000000284] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy demonstrates impressive efficacy in relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL). However, CAR-T therapy-related severe cytokine release syndrome and neurological toxicity limit its clinical application in R/R DLBCL patients with high tumor burden. Here, we conducted a phase II clinical trial testing the efficacy and toxicities of CAR-T therapy in R/R non-Hodgkin lymphoma patients (NCT03196830). Among the enrolled patients, 10 R/R DLBCL patients with high tumor burden were analyzed. Before CAR-T therapy, 4 were treated with intensive combined chemotherapy (C-CAR-cohort), and 6 were exposed to radiotherapy (R-CAR-cohort). Patients in the R-CAR-T-cohort showed a higher overall response rate (100% vs. 25%, P=0.033) and less severe cytokine release syndrome (0% vs. 100%, P=0.0048) and neurotoxicity (0% vs. 75%, P=0.033) incidences than patients in the C-CAR-T-cohort. Furthermore, one case who responded to CAR-T therapy initially and who suffered a relapse shortly was exposed to radiation and achieved complete remission, with an increase in the number of CAR-T copies detected. This study demonstrates that radiotherapy is an optimal debulking regimen to managing R/R DLBCL patients before CAR-T therapy and a promising alternative salvage therapy for patients who suffer a relapse after CAR-T therapy by fuelling CAR-T copies.
Collapse
|
20
|
Mondal P, Kaur B, Natesh J, Meeran SM. The emerging role of miRNA in the perturbation of tumor immune microenvironment in chemoresistance: Therapeutic implications. Semin Cell Dev Biol 2021; 124:99-113. [PMID: 33865701 DOI: 10.1016/j.semcdb.2021.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/16/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023]
Abstract
Chemoresistance is a major hindrance in cancer chemotherapies, a leading cause of tumor recurrence and cancer-related deaths. Cancer cells develop numerous strategies to elude immune attacks and are regulated by immunological factors. Cancer cells can alter the expression of several immune modulators to upregulate the activities of immune checkpoint pathways. Targeting the immune checkpoint inhibitors is a part of the cancer immunotherapy altered during carcinogenesis. These immune modulators have the capability to reprogram the tumor microenvironment, thereby change the efficacy of chemotherapeutics. In general, the sensitivity of drugs is reduced in the immunosuppressive tumor microenvironment, resulting in chemoresistance and tumor relapse. The regulation of microRNAs (miRNAs) is well established in cancer initiation, progression, and therapy. Intriguingly, miRNA affects cancer immune surveillance and immune response by targeting immune checkpoint inhibitors in the tumor microenvironment. miRNAs alter the gene expression at the post-transcriptional level, which modulates both innate and adaptive immune systems. Alteration of tumor immune microenvironment influences drug sensitivity towards cancer cells. Besides, the expression profile of immune-modulatory miRNAs can be used as a potential biomarker to predict the response and clinical outcomes in cancer immunotherapy and chemotherapy. Recent evidences have revealed that cancer-derived immune-modulatory miRNAs might be promising targets to counteract cancer immune escape, thereby increasing drug efficacy. In this review, we have compiled the role of miRNAs in overcoming the chemoresistance by modulating tumor microenvironment and discussed their preclinical and clinical implications.
Collapse
Affiliation(s)
- Priya Mondal
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Bhavjot Kaur
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, India
| | - Jagadish Natesh
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Syed Musthapa Meeran
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
21
|
Paganetti H, Beltran C, Both S, Dong L, Flanz J, Furutani K, Grassberger C, Grosshans DR, Knopf AC, Langendijk JA, Nystrom H, Parodi K, Raaymakers BW, Richter C, Sawakuchi GO, Schippers M, Shaitelman SF, Teo BKK, Unkelbach J, Wohlfahrt P, Lomax T. Roadmap: proton therapy physics and biology. Phys Med Biol 2021; 66. [DOI: 10.1088/1361-6560/abcd16] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022]
|
22
|
Komanduri KV. Chimeric Antigen Receptor T-Cell Therapy in the Management of Relapsed Non-Hodgkin Lymphoma. J Clin Oncol 2021; 39:476-486. [DOI: 10.1200/jco.20.01749] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
23
|
Sang W, Xie L, Wang G, Li J, Zhang Z, Li B, Guo S, Deng C, Dai Y. Oxygen-Enriched Metal-Phenolic X-Ray Nanoprocessor for Cancer Radio-Radiodynamic Therapy in Combination with Checkpoint Blockade Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003338. [PMID: 33643804 PMCID: PMC7887592 DOI: 10.1002/advs.202003338] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/12/2020] [Indexed: 05/05/2023]
Abstract
Radiotherapy (RT) based on DNA damage and reactive oxygen species (ROS) generation has been clinically validated in various types of cancer. However, high dose-dependent induced toxicity to tissues, non-selectivity, and radioresistance greatly limit the application of RT. Herein, an oxygen-enriched X-ray nanoprocessor Hb@Hf-Ce6 nanoparticle is developed for improving the therapeutic effect of RT-radiodynamic therapy (RDT), enhancing modulation of hypoxia tumor microenvironment (TME) and promoting antitumor immune response in combination with programmed cell death protein 1 (PD-1) immune checkpoint blockade. All functional molecules are integrated into the nanoparticle based on metal-phenolic coordination, wherein one high-Z radiosensitizer (hafnium, Hf) coordinated with chlorin e6 (Ce6) modified polyphenols and a promising oxygen carrier (hemoglobin, Hb) is encapsulated for modulation of oxygen balance in the hypoxia TME. Specifically, under single X-ray irradiation, radioluminescence excited by Hf can activate photosensitizer Ce6 for ROS generation by RDT. Therefore, this combinatory strategy induces comprehensive antitumor immune response for cancer eradication and metastasis inhibition. This work presents a multifunctional metal-phenolic nanoplatform for efficient X-ray mediated RT-RDT in combination with immunotherapy and may provide a new therapeutic option for cancer treatment.
Collapse
Affiliation(s)
- Wei Sang
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
- Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
| | - Lisi Xie
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
- Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
| | - Guohao Wang
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
- Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
| | - Jie Li
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
- Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
| | - Zhan Zhang
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
- Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
| | - Bei Li
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
- Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
| | - Sen Guo
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
- Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
| | - Chu‐Xia Deng
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
- Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
| | - Yunlu Dai
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
- Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacauSAR, 999078China
| |
Collapse
|
24
|
Razavi A, Keshavarz-Fathi M, Pawelek J, Rezaei N. Chimeric antigen receptor T-cell therapy for melanoma. Expert Rev Clin Immunol 2021; 17:209-223. [PMID: 33481629 DOI: 10.1080/1744666x.2021.1880895] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION In recent years, chimeric antigen receptor (CAR) T cell therapy has emerged as a cancer treatment. After initial therapeutic success for hematologic malignancies, this approach has been extended for the treatment of solid tumors including melanoma. AREAS COVERED T cells need to be reprogramed to recognize specific antigens expressed only in tumor cells, a difficult problem since cancer cells are simply transformed normal cells. Tumor antigens, namely, CSPG4, CD70, and GD2 have been targeted by CAR-T cells for melanoma. Moreover, different co-stimulatory signaling domains need to be selected to direct T cell fate. In this review, various approaches for the treatment of melanoma and their effectiveness are comprehensively reviewed and the current status, challenges, and future perspective of CAR-T cell therapy for melanoma are discussed. Literature search was accomplished in three databases (PubMed, Google scholar, and Clinicaltrials.gov). Published papers and clinical trials were screened and relevant documents were included by checking pre-defined eligibility criteria. EXPERT OPINION Despite obstacles and the risk of adverse events, CAR T cell therapy could be used for patients with treatment-resistant cancer. Clinical trials are underway to determine the efficacy of this approach for the treatment of melanoma.
Collapse
Affiliation(s)
- Azadehsadat Razavi
- Department of Animal Biology, Faculty of Biology Sciences, University of Kharazmi, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - John Pawelek
- Department of Dermatology and the Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden
| |
Collapse
|
25
|
Immunostimulatory Effects of Radiotherapy for Local and Systemic Control of Melanoma: A Review. Int J Mol Sci 2020; 21:ijms21239324. [PMID: 33297519 PMCID: PMC7730562 DOI: 10.3390/ijms21239324] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
Recently, modern therapies involving immune checkpoint inhibitors, cytokines, and oncolytic virus have been developed. Because of the limited treatment effect of modern therapy alone, the immunostimulatory effect of radiotherapy attracted increasing attention. The combined use of radiotherapy and modern therapy has been examined clinically and non-clinically, and its effectiveness has been confirmed recently. Because melanomas have high immunogenicity, better therapeutic outcomes are desired when using immunotherapy. However, sufficient therapeutic effects have not yet been achieved. Thus far, radiotherapy has been used only for local control of tumors. Although extremely rare, radiotherapy has also been reported for systemic control, i.e., abscopal effect. This is thought to be due to an antitumor immune response. Therefore, we herein summarize past information on not only the mechanism of immune effects on radiotherapy but also biomarkers reported in case reports on abscopal effects. We also reviewed the animal model suitable for evaluating abscopal effects. These results pave the way for further basic research or clinical studies on new treatment methods for melanoma. Currently, palliative radiation is administered to patients with metastatic melanoma for local control. If it is feasible to provide both systemic and local control, the treatment benefit for the patients is very large.
Collapse
|
26
|
Abstract
Since the approval in 2017 and the amazing achievement of Kymriah and Yescarta, the number of basic researchers and clinical trials investigating the safety and efficacy of chimeric antigen receptor-expressing T cells (CAR-T cells) has been relentlessly increasing. Up to now, more than 200 clinical trials are listed on clinical trial database of NIH and the basic research is countless. However, the production of allogeneic CAR-T cells products is still expensive and has toxicity. Thus, more effort is needed to develop reliable off-the-shelf cellular therapeutic methods with safety and efficiency for the treatment of patients with cancer. As a kind of innate effector lymphocyte with potent antitumor activity, natural killer cells (NK cells) have attracted much attention. Until now, basic and clinical research has shown that chimeric antigen receptor-expressing NK cell (CAR-NK) therapy may play a significant anti-tumor role and its safety is higher than CAR-T cell therapy. In this review, we discuss advantages and shortages of employing CAR-NK cells as a novel cellular therapy against cancer.
Collapse
Affiliation(s)
- Jufeng Xia
- Graduate School of Frontier Science, The University of Tokyo, Chiba, Japan.,Department of Immunology, Arai Japan Medical Institute, Tokyo, Japan
| | - Shuichi Minamino
- Department of Immunology, Arai Japan Medical Institute, Tokyo, Japan
| | - Kazuma Kuwabara
- Department of Immunology, Arai Japan Medical Institute, Tokyo, Japan
| |
Collapse
|
27
|
Chen D, Menon H, Verma V, Guo C, Ramapriyan R, Barsoumian H, Younes A, Hu Y, Wasley M, Cortez MA, Welsh J. Response and outcomes after anti-CTLA4 versus anti-PD1 combined with stereotactic body radiation therapy for metastatic non-small cell lung cancer: retrospective analysis of two single-institution prospective trials. J Immunother Cancer 2020; 8:jitc-2019-000492. [PMID: 31996395 PMCID: PMC7057428 DOI: 10.1136/jitc-2019-000492] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND This study compared response rates and outcomes of combined radiotherapy and immunotherapy (iRT) based on the type of checkpoint inhibitor (anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA4) vs antiprogrammed death-1 (PD1)) for metastatic non-small cell lung cancer (mNSCLC). METHODS We retrospectively reviewed two prospective trials of radiation combined with anti-CTLA4 or anti-PD1 for patients with mNSCLC. Patients undergoing non-salvage stereotactic body radiation therapy (SBRT) to lung sites were selected from both trials and grouped by the immunotherapeutic compound received. Endpoints included in-field and out-of-field response rates, and overall response rate (complete or partial response) (all by response evaluation criteria in solid tumors). Progression-free survival (PFS) and overall survival (OS) were estimated with the Kaplan-Meier method. RESULTS Median follow-up times for the 33 patients (n=17 SBRT+anti-CTLA4, n=16 SBRT+anti-PD1) were 19.6 and 19.9 months. Response rates for out-of-field lesions were similar between anti-PD1 (37%) and anti-CTLA4 (24%) (p=0.054). However, global response rates for all lesions were 24% anti-CTLA4 vs 56% anti-PD1 (p=0.194). The PFS was 76% for anti-CTLA4 vs 94% anti-PD1 at 3 months, 52% vs 87% at 6 months, 31% vs 80% at 12 months, and 23% vs 63% at 18 months (p=0.02). Respective OS values were 76% vs 87% at 6 months, 47% vs 80% at 12 months, and 39% vs 66% at 18 months (p=0.08). CONCLUSIONS Both anti-CTLA4 and anti-PD1 agents prompt a similar degree of in-field and out-of-field responses after iRT, although the global response rate and PFS were statistically higher in the anti-PD1 cohort. Further dedicated study and biological mechanistic assessment is required. TRIAL REGISTRATION NUMBERS NCT02239900 and NCT02444741.
Collapse
Affiliation(s)
- Dawei Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Hari Menon
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Vivek Verma
- Department of radiation oncology, Allegheny General Hospital, Houston, Texas, United States
| | - Chunxiao Guo
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, China
| | - Rishab Ramapriyan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Hampartsoum Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Ahmed Younes
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Yun Hu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Mark Wasley
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - James Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
| |
Collapse
|
28
|
Schepisi G, Conteduca V, Casadei C, Gurioli G, Rossi L, Gallà V, Cursano MC, Brighi N, Lolli C, Menna C, Farolfi A, Burgio SL, Altavilla A, Martinelli G, De Giorgi U. Potential Application of Chimeric Antigen Receptor (CAR)-T Cell Therapy in Renal Cell Tumors. Front Oncol 2020; 10:565857. [PMID: 33072597 PMCID: PMC7538692 DOI: 10.3389/fonc.2020.565857] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
Currently, renal cell carcinoma is characterized by encouraging benefits from immunotherapy that have led to significant results in treatment outcome. The approval of nivolumab primarily as second-line monotherapy and, more recently, the approval of new combination therapies as first-line treatment have confirmed the importance of immunotherapy in this type of tumor. In this context, the chimeric antigen receptor (CAR)-T represents a further step forward in the field of immunotherapy. Initially tested on hematological malignancies, this new therapeutic approach is also becoming a topic of great interest for solid tumors. Although the treatment has several advantages over previous T-cell receptor-dependent immunotherapy, it is facing some obstacles in solid tumors such as a hostile tumor microenvironment and on-tumor/off-tumor toxicities. Several strategies are under investigation to overcome these problems, but the approval of CAR-T cell therapy is still some way off. In renal cancer, the significant advantages obtained from immune checkpoint inhibitors represent a good starting point, but the potential nephrological toxicity of CAR-T cell therapy represents an important risk. In this review, we provide the rationale and preliminary results of CAR-T cell therapy in renal cell malignancies.
Collapse
Affiliation(s)
- Giuseppe Schepisi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Vincenza Conteduca
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Chiara Casadei
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Giorgia Gurioli
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Lorena Rossi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Valentina Gallà
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | | | - Nicole Brighi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Cristian Lolli
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Cecilia Menna
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Alberto Farolfi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Salvatore Luca Burgio
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Amelia Altavilla
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Giovanni Martinelli
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| |
Collapse
|
29
|
Boutros C, Chaput-Gras N, Lanoy E, Larive A, Mateus C, Routier E, Sun R, Tao YG, Massard C, Bahleda R, Schwob D, Ibrahim N, Khoury Abboud RM, Caramella C, Lancia A, Cassard L, Roy S, Soria JC, Robert C, Deutsch E. Dose escalation phase 1 study of radiotherapy in combination with anti-cytotoxic-T-lymphocyte-associated antigen 4 monoclonal antibody ipilimumab in patients with metastatic melanoma. J Immunother Cancer 2020; 8:e000627. [PMID: 32819972 PMCID: PMC7443273 DOI: 10.1136/jitc-2020-000627] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND A synergy between radiotherapy and anti-cytotoxic-T-lymphocyte-associated antigen 4 (anti-CTLA-4) monoclonal antibody has been demonstrated preclinically. The Mel-Ipi-Rx phase 1 study aimed to determine the maximum tolerated dose (MTD) and safety profile of radiotherapy combined with ipilimumab in patients with metastatic melanoma. PATIENTS AND METHODS A 3+3 dose escalation design was used with 9, 15, 18 and 24 Gy dose of radiotherapy at week 4 combined with 10 mg/kg ipilimumab every 3 weeks for four doses. Patients with evidence of clinical benefit at week 12 were eligible for maintenance with ipilimumab 10 mg/kg every 12 weeks starting at week 24 until severe toxicity or disease progression. The database lock occurred on April 30, 2019. Tumor growth rate of irradiated lesions and non-irradiated lesions were analyzed to assess the systemic immunologic antitumor response. Blood immune monitoring was performed before and during treatment to determine if radiotherapy could modify ipilimumab pharmacodynamics. RESULTS 19 patients received ipilimumab between August 2011 and July 2015. Nine patients received the four doses of ipilimumab. All patients received the combined radiotherapy. Grade 3 adverse events occurred in nine patients, the most common being colitis and hepatitis. No drug-related death occurred. Dose limiting toxicity occurred in two of six patients in the cohort receiving 15 Gy. The MTD was 9 Gy. Two patients had complete response, three had partial response response and seven had stable disease, giving an objective response rate of 31% and a clinical benefit rate of 75% at week 24. The median duration of follow-up was 5.8 years (Q1=4.5; Q3=6.8). The median overall survival (95% CI) was estimated at 0.9 years (0.5-2). The median progression-free survival (PFS) (95% CI) was 0.4 (0.2-1.4). Radiotherapy combined with ipilimumab was associated with increased CD4+ and CD8+ICOS+ T cells. Increased CD8+ was significantly associated with PFS. CONCLUSION When combined with ipilimumab at 10 mg/kg, the MTD of radiotherapy was 9 Gy. This combination of ipilimumab and radiotherapy appears to be associated with antitumor activity. Increased CD8+ was significantly associated with PFS. Thus, immune biomarkers may be useful for early response evaluation. TRIAL REGISTRATION NUMBER NCT01557114.
Collapse
Affiliation(s)
- Celine Boutros
- Dermatology Unit, Outpatient Clinic, Department of Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nathalie Chaput-Gras
- Laboratoire d'immunomonitoring En Oncologie, University Paris-Saclay, Faculty of Pharmacy, Gustave Roussy Cancer Campus, Villejuif, France
| | - Emilie Lanoy
- Biostatistic and Epidemiology Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Alicia Larive
- Biostatistic and Epidemiology Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Christine Mateus
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, Villejuif, UK
| | - Emilie Routier
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, Villejuif, UK
| | - Roger Sun
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Yun Gan Tao
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Christophe Massard
- DITEP, University Paris-Saclay, Faculty of Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Rastilav Bahleda
- DITEP, University Paris-Saclay, Faculty of Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Dominique Schwob
- Biostatistic and Epidemiology Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nathalie Ibrahim
- Outpatient Clinic, Department of Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Caroline Caramella
- Department of Radiology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Andrea Lancia
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Radiation Oncology, Polyclinic San Matteo Pavia Fondazione IRCCS, Pavia, Italy
| | - Lydie Cassard
- Laboratoire d'immunomonitoring En Oncologie, University Paris-Saclay, Faculty of Pharmacy, Gustave Roussy Cancer Campus, Villejuif, France
| | - Severine Roy
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, Villejuif, UK
| | - J-C Soria
- DITEP, INSERM Unit U981, University Paris-Saclay, Faculty of Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Caroline Robert
- Dermatology Unit, Department of Medicine, University Paris-Saclay, Faculty of Medicine, INSERM Unit U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - Eric Deutsch
- Department of Radiation Oncology, Radiomics Team, Molecular Radiotherapy INSERM U1030, University Paris-Saclay, Faculty of Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
30
|
Donnadieu E, Dupré L, Pinho LG, Cotta-de-Almeida V. Surmounting the obstacles that impede effective CAR T cell trafficking to solid tumors. J Leukoc Biol 2020; 108:1067-1079. [PMID: 32620049 PMCID: PMC7586996 DOI: 10.1002/jlb.1mr0520-746r] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Innovative immunotherapies based on immune checkpoint targeting antibodies and engineered T cells are transforming the way we approach cancer treatment. However, although these T cell centered strategies result in marked and durable responses in patients across many different tumor types, they provide therapeutic efficacy only in a proportion of patients. A major challenge of immuno‐oncology is thereby to identify mechanisms responsible for resistance to cancer immunotherapy in order to overcome them via adapted strategies that will ultimately improve intrinsic efficacy and response rates. Here, we focus on the barriers that restrain the trafficking of chimeric antigen receptor (CAR)‐expressing T cells to solid tumors. Upon infusion, CAR T cells need to home into malignant sites, navigate within complex tumor environments, form productive interactions with cancer cells, deliver their cytotoxic activities, and finally persist. We review the accumulating evidence that the microenvironment of solid tumors contains multiple obstacles that hinder CAR T cells in the dynamic steps underlying their trafficking. We focus on how these hurdles may in part account for the failure of CAR T cell clinical trials in human carcinomas. Given the engineered nature of CAR T cells and possibilities to modify the tumor environment, there are ample opportunities to augment CAR T cell ability to efficiently find and combat tumors. We present some of these strategies, which represent a dynamic field of research with high potential for clinical applicability.
Collapse
Affiliation(s)
- Emmanuel Donnadieu
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Loïc Dupré
- INSERM, UMR1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France.,CNRS, UMR5282, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Lia Gonçalves Pinho
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Vinicius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| |
Collapse
|
31
|
Sherry AD, Bezzerides M, Khattab MH, Luo G, Ancell KK, Kirschner AN. An autoimmune-based, paraneoplastic neurologic syndrome following checkpoint inhibition and concurrent radiotherapy for merkel cell carcinoma: case report. Strahlenther Onkol 2020; 196:664-670. [PMID: 32006066 DOI: 10.1007/s00066-020-01582-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 01/11/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE Merkel cell carcinoma is highly sensitive to both radiation and immunotherapy. Moreover, concurrent radioimmunotherapy may capitalize on anti-tumor immune activity and improve Merkel cell treatment response, although an enhanced immune system may cross-react with native tissues and lead to significant sequelae. METHODS Here we present a case study of a patient with metastatic Merkel cell carcinoma treated with radiotherapy concurrent with pembrolizumab. RESULTS After radioimmunotherapy, the patient developed sensory neuropathy, visual hallucinations, and mixed motor neuron findings. Neurologic dysfunction progressed to profound gastrointestinal dysmotility necessitating parenteral nutrition and intubation with eventual expiration. CONCLUSION This case represents a unique autoimmune paraneoplastic neurologic syndrome, likely specific to neuroendocrine tumors and motivated by concurrent radioimmunotherapy. Recognition of the potential role of radioimmunotherapy may provide an advantage in anticipating these severe sequelae.
Collapse
MESH Headings
- Aged
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Autoimmune Diseases of the Nervous System/etiology
- Autoimmune Diseases of the Nervous System/immunology
- Axilla
- Carboplatin/administration & dosage
- Carcinoma, Merkel Cell/drug therapy
- Carcinoma, Merkel Cell/radiotherapy
- Carcinoma, Merkel Cell/secondary
- Combined Modality Therapy
- Deglutition Disorders/etiology
- Etoposide/administration & dosage
- Fatal Outcome
- Fingers
- Hallucinations/etiology
- Humans
- Lymphatic Metastasis/diagnostic imaging
- Lymphatic Metastasis/radiotherapy
- Male
- Neuralgia/drug therapy
- Neuralgia/etiology
- Palliative Care
- Paraneoplastic Syndromes, Nervous System/etiology
- Paraneoplastic Syndromes, Nervous System/immunology
- Parenteral Nutrition, Total
- Pneumonia, Aspiration/etiology
- Positron Emission Tomography Computed Tomography
- Radioimmunotherapy/adverse effects
- Radiotherapy, High-Energy
- Radiotherapy, Intensity-Modulated/adverse effects
- Skin Neoplasms/drug therapy
- Skin Neoplasms/radiotherapy
- Skin Neoplasms/secondary
Collapse
Affiliation(s)
| | | | - Mohamed H Khattab
- Department of Radiation Oncology, Vanderbilt University Medical Center, 2220 Pierce Avenue, PRB-B1003, Nashville, TN, USA
| | - Guozhen Luo
- Department of Radiation Oncology, Vanderbilt University Medical Center, 2220 Pierce Avenue, PRB-B1003, Nashville, TN, USA
| | - Kristin K Ancell
- Department of Medicine, Division of Hematology Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Austin N Kirschner
- Department of Radiation Oncology, Vanderbilt University Medical Center, 2220 Pierce Avenue, PRB-B1003, Nashville, TN, USA.
| |
Collapse
|
32
|
Guan L, Wu W, Pang H, Duan D, Li S. Anti-GPC3 single-chain scFv antibody acts as an agent for radio-immunoimaging in diagnosing hepatocellular carcinoma. Am J Transl Res 2019; 11:7422-7431. [PMID: 31934289 PMCID: PMC6943449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/05/2019] [Indexed: 06/10/2023]
Abstract
Glypican-3 (GPC3) over-expresses in hepatocellular carcinoma (HCC), but not expresses or under-expresses in normal adult hepatocytes. Therefore, GPC3 acts as a potential target for diagnosis and treatment of HCC. This study aimed to conduct radio-immunoimaging using GPC3 as a target in order, and to explore its potential for diagnosing and treating HCC. Humanized single-chain antibody scFv for HCC was established using phage antibody library. E.coli HB2151 was infected with recombinant phage antibodies that are considered to be strongly positive by phage ELISA. Then, the soluble antibodies were obtained post IPTG induction. Soluble antibodies were detected using SDS-PAGE assay. Anti-GPC3 single-chain antibodies were labeled using 131I, and then the distribution of radioactive markers in nude mice were analyzed in vivo by radio-immunoimaging. The results indicated that the size of soluble scFv products was 30 kD after purifying anti-GPC3 scFv antibodies that are successfully screened from phage antibody library. Anti-GPC3 phage antibodies could specifically bind to HCC cells. The ratios of radioactive tumor/blood and tumor/muscle for 131I labeled anti-GPC3 monoclonal antibodies were increased gradually, achieving the highest at 48 h. Radio-immunoimaging showed that the radioactive uptake of tumor sites remained the strongest at 48 h, and the ratio of target to non-target was the highest. In conclusion, the established anti-GPC3 scFv antibody had the potential to become an agent for radio-immunoimaging in diagnosing HCC and act as a targeted antibody for further radio-immunotherapy of HCC.
Collapse
Affiliation(s)
- Lili Guan
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Wei Wu
- The Chongqing Key Laboratory of Toxicology and Drug Analysis, Chongqing Police CollegeChongqing, China
| | - Hua Pang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Dong Duan
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Shaolin Li
- Department of Radiology, Chongqing Medical UniversityChongqing, China
| |
Collapse
|
33
|
Kemeny HR, Elsamadicy AA, Farber SH, Champion CD, Lorrey SJ, Chongsathidkiet P, Woroniecka KI, Cui X, Shen SH, Rhodin KE, Tsvankin V, Everitt J, Sanchez-Perez L, Healy P, McLendon RE, Codd PJ, Dunn IF, Fecci PE. Targeting PD-L1 Initiates Effective Antitumor Immunity in a Murine Model of Cushing Disease. Clin Cancer Res 2019; 26:1141-1151. [PMID: 31744830 PMCID: PMC7809696 DOI: 10.1158/1078-0432.ccr-18-3486] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 10/02/2019] [Accepted: 11/15/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Although pituitary adenoma is classified as benign, Cushing disease is associated with significant morbidity due to the numerous sequelae of elevated cortisol levels. Successful therapy for Cushing disease remains elusive due to high rates of treatment-refractory recurrence. The frequent emergence of lymphocytic hypophysitis following checkpoint blockade for other cancers, as well as the expression of PD-L1 on pituitary adenomas, suggest a role for immunotherapy. EXPERIMENTAL DESIGN This study confirms PD-L1 expression on functioning pituitary adenomas and is the first to evaluate the efficacy of checkpoint blockade (anti-PD-L1) therapy in a preclinical model of Cushing disease. RESULTS Herein, treatment with anti-PD-L1 was successful in reducing adrenocorticotropic hormone plasma levels, decreasing tumor growth, and increasing survival in our model. Furthermore, tumor-infiltrating T cells demonstrated a pattern of checkpoint expression similar to other checkpoint blockade-susceptible tumors. CONCLUSIONS This suggests that immunotherapy, particularly blockade of the PD1/PD-L1 axis, may be a novel therapeutic option for refractory Cushing disease. Clinical investigation is encouraged.
Collapse
Affiliation(s)
- Hanna R Kemeny
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Duke University School of Medicine, Durham, North Carolina
| | - Aladine A Elsamadicy
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Duke University School of Medicine, Durham, North Carolina
| | - S Harrison Farber
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Duke University School of Medicine, Durham, North Carolina
| | - Cosette D Champion
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Duke University School of Medicine, Durham, North Carolina
| | - Selena J Lorrey
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Pakawat Chongsathidkiet
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Karolina I Woroniecka
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Xiuyu Cui
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Steven H Shen
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Duke University School of Medicine, Durham, North Carolina
| | - Kristen E Rhodin
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Duke University School of Medicine, Durham, North Carolina
| | - Vadim Tsvankin
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Jeffrey Everitt
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Luis Sanchez-Perez
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Patrick Healy
- Department of Biostatistics, Duke University, Durham, North Carolina
| | - Roger E McLendon
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Patrick J Codd
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Ian F Dunn
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Peter E Fecci
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina. .,Duke University School of Medicine, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
34
|
Iijima K, Oozeki M, Ikeda K, Honda H, Ishibashi H, Yamaoka M, Fujieda S, Saitoh H, Goto M, Araki M, Amagai K. A case of small bowel adenocarcinoma wherein nivolumab conferred temporary benefit in disease control. Clin J Gastroenterol 2019; 13:372-376. [PMID: 31701367 DOI: 10.1007/s12328-019-01064-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022]
Abstract
Small bowel adenocarcinomas are rare. There is no definite consensus as to whether they should be treated in a manner similar to gastric or to colon cancer. We report the case of a young woman with a primary jejunal adenocarcinoma, bilateral ovary metastases, and peritoneal dissemination. First- and second-line chemotherapy for the gastric cancer failed. She was then treated with the immune checkpoint inhibitor nivolumab and had temporary improvement in her condition. To the best of our knowledge, this is the first case wherein nivolumab has been used to treat small bowel adenocarcinoma.
Collapse
Affiliation(s)
- Kazutaka Iijima
- Department of Gastroenterology, Ibaraki Prefectural Central Hospital, 6528 Koibuchi, Kasama-City, Ibaraki, 309-1793, Japan.
- Department of Gastroenterology, Rumoi Municipal Hospital, Hokkaido, Japan.
| | - Mitsuharu Oozeki
- Department of Gastroenterology, Ibaraki Prefectural Central Hospital, 6528 Koibuchi, Kasama-City, Ibaraki, 309-1793, Japan
| | - Kaori Ikeda
- Department of Gastroenterology, Ibaraki Prefectural Central Hospital, 6528 Koibuchi, Kasama-City, Ibaraki, 309-1793, Japan
| | - Hiroyuki Honda
- Department of Gastroenterology, Ibaraki Prefectural Central Hospital, 6528 Koibuchi, Kasama-City, Ibaraki, 309-1793, Japan
| | - Hajime Ishibashi
- Department of Gastroenterology, Ibaraki Prefectural Central Hospital, 6528 Koibuchi, Kasama-City, Ibaraki, 309-1793, Japan
| | - Masaharu Yamaoka
- Department of Gastroenterology, Ibaraki Prefectural Central Hospital, 6528 Koibuchi, Kasama-City, Ibaraki, 309-1793, Japan
| | - Shinji Fujieda
- Department of Gastroenterology, Ibaraki Prefectural Central Hospital, 6528 Koibuchi, Kasama-City, Ibaraki, 309-1793, Japan
| | - Hitoaki Saitoh
- Department of Diagnostic Pathology, Ibaraki Prefectural Central Hospital, Ibaraki, Japan
| | - Mitsuhide Goto
- Department of Gastroenterology, Ibaraki Prefectural Central Hospital, 6528 Koibuchi, Kasama-City, Ibaraki, 309-1793, Japan
| | - Masahiro Araki
- Department of Gastroenterology, Ibaraki Prefectural Central Hospital, 6528 Koibuchi, Kasama-City, Ibaraki, 309-1793, Japan
| | - Kenji Amagai
- Department of Gastroenterology, Ibaraki Prefectural Central Hospital, 6528 Koibuchi, Kasama-City, Ibaraki, 309-1793, Japan
| |
Collapse
|
35
|
Woods K, Neph R, Nguyen D, Sheng K. A sparse orthogonal collimator for small animal intensity-modulated radiation therapy. Part II: hardware development and commissioning. Med Phys 2019; 46:5733-5747. [PMID: 31621091 DOI: 10.1002/mp.13870] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/18/2022] Open
Abstract
PURPOSE A dose-modulation device for small animal radiotherapy is required to use clinically analogous treatment techniques, which will likely increase the translatability of preclinical research results. Because the clinically used multileaf collimator (MLC) is impractical for miniaturization, we have developed a simpler, better-suited sparse orthogonal collimator (SOC) for delivering small animal intensity-modulated radiation therapy (IMRT) using a rectangular aperture optimization (RAO) treatment planning system. METHODS The SOC system was modeled in computer-aided design software and fabricated with machined tungsten leaves and three-dimensional (3D) printed leaf housing. A graphical user interface was developed for controlling and calibrating the SOC leaves, which are driven by Arduino-controlled stepper motors. A Winston-Lutz test was performed to assess mechanical alignment, and abutting field and grid dose patterns were created to analyze intra- and intercalibration leaf positioning error. Leaf transmission and penumbra were measured over the full range of gantry angles and leaf positions, respectively. Three SOC test plans were delivered, and film measurements were compared to the intended dose distributions. The differences in maximum, mean, and minimum, as well as pixelwise absolute dose differences, were compared for each structure, and a gamma analysis was performed for the target structures using criteria of 4% dose difference and 0.3 mm distance to agreement. RESULTS The Winston-Lutz test revealed maximum directional offsets between the SOC and primary collimator axes of 0.53 mm at 0° and 0.68 mm over the full 360°. Upper and lower abutting field patterns had maximum dose deviations of 18.8 ± 3.1% and 15.5 ± 2.9%, respectively, and grid patterns showed intra- and intercalibration repeatability of 93% and 91%, respectively. Extremely low midleaf (0.15 ± 0.05%) and interleaf (0.27 ± 0.22%) transmission was measured, with no significant rotational variation. The average penumbra was ~0.8 mm for all leaves at field center, with a range of 0.17 mm for all leaf positions. A highly concave test plan was delivered with a ~ 95% gamma analysis pass rate, and a realistic mouse phantom liver irradiation plan achieved a pass rate of ~98%. A highly complex dose distribution was also created with 551 SOC apertures averaging 2.4 mm in size. CONCLUSIONS A sparse orthogonal collimator was developed and commissioned, with promising preliminary dosimetry results. The SOC design, with its limited moving components and high dose-modulation resolution, is ideal for delivering high-quality small animal IMRT with our RAO-based treatment planning system.
Collapse
Affiliation(s)
- Kaley Woods
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Ryan Neph
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Dan Nguyen
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Ke Sheng
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
36
|
Ganem J, Thureau S, Gouel P, Dubray B, Salaun M, Texte E, Vera P. Prognostic value of post-induction chemotherapy 18F-FDG PET-CT in stage II/III non-small cell lung cancer before (chemo-) radiation. PLoS One 2019; 14:e0222885. [PMID: 31603916 PMCID: PMC6788704 DOI: 10.1371/journal.pone.0222885] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The purpose of our present study was to assess the prognostic impact of FDG PET-CT after induction chemotherapy for patients with inoperable non-small-cell lung cancer (NSCLC). MATERIAL AND METHODS This retrospective study included 50 patients with inoperable stage II/III NSCLC from January 2012 to July 2015. They were treated for curative intent with induction chemotherapy, followed by concomitant chemoradiation therapy or sequential radiation therapy. FDG PET-CT scans were acquired at initial staging (PET1) and after the last cycle of induction therapy (PET2). Five parameters were evaluated on both scans: SUVmax, SUVpeak, SUVmean, TLG, MTV, and their respective deltas. The prognostic value of each parameter for overall survival (OS) and progression-free survival (PFS) was evaluated with Cox proportional-hazards regression models. RESULTS Median follow-up was 19 months. PET1 parameters, clinical and histopathological data were not predictive of the outcome. TLG2 and ΔTLG were prognostic factors for OS. TLG2 was the only prognostic factor for PFS. For OS, log-rank test showed that there was a better prognosis for patients with TLG2< 69g (HR = 7.1, 95%CI 2.8-18, p = 0.002) and for patients with ΔTLG< -81% after induction therapy (HR = 3.8, 95%CI 1.5-9.6, p = 0.02). After 2 years, the survival rate was 89% for the patients with low TLG2 vs 52% for the others. We also evaluated a composite parameter considering both MTV2 and ΔSUVmax. Patients with MTV2> 23cc and ΔSUVmax> -55% had significantly shorter OS than the other patients (HR = 5.7, 95%CI 2.1-15.4, p< 0.01). CONCLUSION Post-induction FDG PET might be an added value to assess the patients' prognosis in inoperable stage II/III NSCLC. TLG, ΔTLG as well as the association of MTV and ΔSUVmax seemed to be valuable parameters, more accurate than clinical, pathological or pretherapeutic imaging data.
Collapse
Affiliation(s)
- Julien Ganem
- Department of Nuclear Medicine, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France
- * E-mail:
| | - Sebastien Thureau
- Department of Nuclear Medicine, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France
- Department of Radiation Oncology and Medical Physics, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France
- QuantIF-LITIS, EA 4108-FR, CNRS, University of Rouen, Rouen, France
| | - Pierrick Gouel
- Department of Nuclear Medicine, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France
- QuantIF-LITIS, EA 4108-FR, CNRS, University of Rouen, Rouen, France
| | - Bernard Dubray
- Department of Radiation Oncology and Medical Physics, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France
- QuantIF-LITIS, EA 4108-FR, CNRS, University of Rouen, Rouen, France
| | - Mathieu Salaun
- QuantIF-LITIS, EA 4108-FR, CNRS, University of Rouen, Rouen, France
- Department of Pneumology, Rouen University Hospital, Rouen, France
| | - Edgar Texte
- Department of Nuclear Medicine, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France
| | - Pierre Vera
- Department of Nuclear Medicine, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France
- QuantIF-LITIS, EA 4108-FR, CNRS, University of Rouen, Rouen, France
| |
Collapse
|
37
|
Wang L, Dou M, Ma Q, Yao R, Liu J. Chimeric antigen receptor (CAR)-modified NK cells against cancer: Opportunities and challenges. Int Immunopharmacol 2019; 74:105695. [PMID: 31254958 DOI: 10.1016/j.intimp.2019.105695] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/30/2019] [Accepted: 06/12/2019] [Indexed: 12/13/2022]
Abstract
NK cells may have great potential in tumor immunotherapy because they can kill tumor cells directly and quickly. Chimeric antigen receptor is a fusion protein composed of extracellular antigen recognition domain, transmembrane domain and intracellular signal domain. Rapid development of CAR-modified T cells has made tremendous achievements in the treatment of malignancies, especially hematological malignancies. However, there are many deficiencies in clinical application of CAR-T cell therapy. Car-modified NK cells have attracted much attention because they may avoid these shortcomings. At present, preclinical and clinical studies have shown that CAR-NK cell therapy may play significant anti-tumor role and it is safer than CAR-T cell therapy. Nevertheless, CAR-NK cell therapy still faces some challenges, such as the expansion and activation of primary NK cells in vitro, the difficulty to store and ship NK cell products and the low transduction efficiency. Thus further research is still needed to optimize CAR-NK cell therapy. Building better CAR-NK cells is important to improve the treatment efficacy and combination therapy offers a novel direction of NK-cell based immunotherapy.
Collapse
Affiliation(s)
- Luyao Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266000, China
| | - Mei Dou
- School of Public Health, Qingdao University, Qingdao 266021, Shandong, China
| | - Qingxia Ma
- School of Basic Medical Sciences, Qingdao University, 38 Dengzhou Road, Qingdao 266021, China
| | - Ruixue Yao
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266000, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266000, China.
| |
Collapse
|
38
|
Brokāne L, Jaunalksne I, Tilgase A, Olmane E, Petroška D, Rasa A, Alberts P. Combination treatment with nivolumab and Rigvir of a progressive stage IIC skin melanoma patient. Clin Case Rep 2019; 7:1191-1196. [PMID: 31183092 PMCID: PMC6552946 DOI: 10.1002/ccr3.2182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 04/01/2019] [Accepted: 04/16/2019] [Indexed: 11/30/2022] Open
Abstract
A 35-year-old male patient was diagnosed with stage IIC skin melanoma that rapidly progressed after surgery. Treatment was continued with radiotherapy, which did not stop further spread of disease and the patient was put on a combination of nivolumab and Rigvir. Subsequently, the progression has slowed.
Collapse
Affiliation(s)
| | | | | | - Evija Olmane
- Department of RadiologyPauls Stradiņš Clinical University HospitalRīgaLatvia
| | - Donatas Petroška
- National Center of PathologyAffiliate of Vilnius University Hospital Santaros KlinikosVilniusLithuania
| | | | | |
Collapse
|
39
|
Fan W, Tang W, Lau J, Shen Z, Xie J, Shi J, Chen X. Breaking the Depth Dependence by Nanotechnology-Enhanced X-Ray-Excited Deep Cancer Theranostics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1806381. [PMID: 30698854 DOI: 10.1002/adma.201806381] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/26/2018] [Indexed: 05/12/2023]
Abstract
The advancements in nanotechnology have created multifunctional nanomaterials aimed at enhancing diagnostic accuracy and treatment efficacy for cancer. However, the ability to target deep-seated tumors remains one of the most critical challenges for certain nanomedicine applications. To this end, X-ray-excited theranostic techniques provide a means of overcoming the limits of light penetration and tissue attenuation. Herein, a comprehensive overview of the recent advances in nanotechnology-enhanced X-ray-excited imaging and therapeutic methodologies is presented, with an emphasis on the design of multifunctional nanomaterials for contrast-enhanced computed tomography (CT) imaging, X-ray-excited optical luminescence (XEOL) imaging, and X-ray-excited multimodal synchronous/synergistic therapy. The latter is based on the concurrent use of radiotherapy with chemotherapy, gas therapy, photodynamic therapy, or immunotherapy. Moreover, the featured biomedical applications of X-ray-excited deep theranostics are discussed to highlight the advantages of X-ray in high-sensitivity detection and efficient elimination of malignant tumors. Finally, key issues and technical challenges associated with this deep theranostic technology are identified, with the intention of advancing its translation into the clinic.
Collapse
Affiliation(s)
- Wenpei Fan
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wei Tang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Joseph Lau
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zheyu Shen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
40
|
Menon H, Ramapriyan R, Cushman TR, Verma V, Kim HH, Schoenhals JE, Atalar C, Selek U, Chun SG, Chang JY, Barsoumian HB, Nguyen QN, Altan M, Cortez MA, Hahn SM, Welsh JW. Role of Radiation Therapy in Modulation of the Tumor Stroma and Microenvironment. Front Immunol 2019; 10:193. [PMID: 30828330 PMCID: PMC6384252 DOI: 10.3389/fimmu.2019.00193] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/23/2019] [Indexed: 12/22/2022] Open
Abstract
In recent decades, there has been substantial growth in our understanding of the immune system and its role in tumor growth and overall survival. A central finding has been the cross-talk between tumor cells and the surrounding environment or stroma. This tumor stroma, comprised of various cells, and extracellular matrix (ECM), has been shown to aid in suppressing host immune responses against tumor cells. Through immunosuppressive cytokine secretion, metabolic alterations, and other mechanisms, the tumor stroma provides a complex network of safeguards for tumor proliferation. With recent advances in more effective, localized treatment, radiation therapy (XRT) has allowed for strategies that can effectively alter and ablate tumor stromal tissue. This includes promoting immunogenic cell death through tumor antigen release to increasing immune cell trafficking, XRT has a unique advantage against the tumoral immune evasion mechanisms that are orchestrated by stromal cells. Current studies are underway to elucidate pathways within the tumor stroma as potential targets for immunotherapy and chemoradiation. This review summarizes the effects of tumor stroma in tumor immune evasion, explains how XRT may help overcome these effects, with potential combinatorial approaches for future treatment modalities.
Collapse
Affiliation(s)
- Hari Menon
- Departments of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rishab Ramapriyan
- Departments of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Taylor R. Cushman
- Departments of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vivek Verma
- Department of Radiation Oncology, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Hans H. Kim
- Department of Radiation Medicine, School of Medicine, Oregon Health and Sciences University, Portland, OR, United States
| | | | - Cemre Atalar
- Departments of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ugur Selek
- Department of Radiation Oncology, School of Medicine, Koç University, Istanbul, Turkey
| | - Stephen G. Chun
- Departments of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joe Y. Chang
- Departments of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hampartsoum B. Barsoumian
- Departments of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Quynh-Nhu Nguyen
- Departments of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mehmet Altan
- Thoracic/Head and Neck Medical Oncology, Houston, TX, United States
| | - Maria A. Cortez
- Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stephen M. Hahn
- Departments of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James W. Welsh
- Departments of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
41
|
Ali AI, Oliver AJ, Samiei T, Chan JD, Kershaw MH, Slaney CY. Genetic Redirection of T Cells for the Treatment of Pancreatic Cancer. Front Oncol 2019; 9:56. [PMID: 30809507 PMCID: PMC6379296 DOI: 10.3389/fonc.2019.00056] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/21/2019] [Indexed: 12/12/2022] Open
Abstract
Conventional treatments for pancreatic cancer are largely ineffective, and the prognosis for the vast majority of patients is poor. Clearly, new treatment options are desperately needed. Immunotherapy offers hope for the development of treatments for pancreatic cancer. A central requirement for the efficacy of this approach is the existence of cancer antigen-specific T cells, but these are often not present or difficult to isolate for most pancreatic tumors. Nevertheless, specific T cells can be generated using genetic modification to express chimeric antigen receptors (CAR), which can enable T cell responses against pancreatic tumor cells. CAR T cells can be produced ex vivo and expanded in vitro for infusion into patients. Remarkable responses have been documented using CAR T cells against several malignancies, including leukemias and lymphomas. Based on these successes, the extension of CAR T cell therapy for pancreatic cancer holds great promise. However, there are a number of challenges that limit the full potential of CAR T cell therapies for pancreatic cancer, including the highly immunosuppressive tumor microenvironment (TME). In this article, we will review the recent progress in using CAR T cells in pancreatic cancer preclinical and clinical settings, discuss hurdles for utilizing the full potential of CAR T cell therapy and propose research strategies and future perspectives. Research into the use of CAR T cell therapy in pancreatic cancer setting is rapidly gaining momentum and understanding strategies to overcome the current challenges in the pancreatic cancer setting will allow the development of effective CAR T cell therapies, either alone or in combination with other treatments to benefit pancreatic cancer patients.
Collapse
Affiliation(s)
- Aesha I Ali
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Amanda J Oliver
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Tinaz Samiei
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Jack D Chan
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
42
|
Manca P, Raez LE, Salzberg M, Sanchez J, Hunis B, Rolfo C. The value of immunotherapy in head and neck cancer. Expert Opin Biol Ther 2018; 19:35-43. [PMID: 30537444 DOI: 10.1080/14712598.2019.1556637] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Head and neck squamous cell carcinomas (HNSCC) previously had limited treatment options once patients had progressed on systemic chemotherapy. With recent advances, immunotherapy now plays an important role in the treatment of advanced disease with improved outcomes as compared to cytotoxic chemotherapy. AREAS COVERED This article reviews the effects of the immune system and how it influences the development and response to HNSCC therapy. We additionally provide a summary of immunotherapy treatments available as well as their applicable clinical trials that led to their approval. EXPERT COMMENTARY The challenges that need to be addressed in order to maximize the benefits of immunotherapy in HNSCC are the selection criteria for immune checkpoint inhibitors and the optimization of combination regimens of immunotherapeutics or chemo-immunotherapy. Furthermore, there remains to be a lack of knowledge in how to incorporate molecular biomarkers as predictors of response to HNSCC immunotherapy.
Collapse
Affiliation(s)
- Paolo Manca
- a Medical Oncology Department , Campus Bio-medico University , Rome , Italy
| | - Luis E Raez
- b Memorial Cancer Institute/Florida International University , Hollywood , FL , USA
| | - Matthew Salzberg
- b Memorial Cancer Institute/Florida International University , Hollywood , FL , USA
| | - Jorge Sanchez
- c Departamento de Oncologia , Hospital Edgardo Rebagliati Martins , Lima , Peru
| | - Brian Hunis
- b Memorial Cancer Institute/Florida International University , Hollywood , FL , USA
| | - Christian Rolfo
- d Marlene and Stewart Greenebaum Comprehensive Cancer Center , University of Maryland School of Medicine , Baltimore , Maryland , USA
| |
Collapse
|
43
|
Pouget JP, Georgakilas AG, Ravanat JL. Targeted and Off-Target (Bystander and Abscopal) Effects of Radiation Therapy: Redox Mechanisms and Risk/Benefit Analysis. Antioxid Redox Signal 2018; 29:1447-1487. [PMID: 29350049 PMCID: PMC6199630 DOI: 10.1089/ars.2017.7267] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 01/13/2018] [Accepted: 01/15/2018] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Radiation therapy (from external beams to unsealed and sealed radionuclide sources) takes advantage of the detrimental effects of the clustered production of radicals and reactive oxygen species (ROS). Research has mainly focused on the interaction of radiation with water, which is the major constituent of living beings, and with nuclear DNA, which contains the genetic information. This led to the so-called target theory according to which cells have to be hit by ionizing particles to elicit an important biological response, including cell death. In cancer therapy, the Poisson law and linear quadratic mathematical models have been used to describe the probability of hits per cell as a function of the radiation dose. Recent Advances: However, in the last 20 years, many studies have shown that radiation generates "danger" signals that propagate from irradiated to nonirradiated cells, leading to bystander and other off-target effects. CRITICAL ISSUES Like for targeted effects, redox mechanisms play a key role also in off-target effects through transmission of ROS and reactive nitrogen species (RNS), and also of cytokines, ATP, and extracellular DNA. Particularly, nuclear factor kappa B is essential for triggering self-sustained production of ROS and RNS, thus making the bystander response similar to inflammation. In some therapeutic cases, this phenomenon is associated with recruitment of immune cells that are involved in distant irradiation effects (called "away-from-target" i.e., abscopal effects). FUTURE DIRECTIONS Determining the contribution of targeted and off-target effects in the clinic is still challenging. This has important consequences not only in radiotherapy but also possibly in diagnostic procedures and in radiation protection.
Collapse
Affiliation(s)
- Jean-Pierre Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Alexandros G. Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Jean-Luc Ravanat
- Univ. Grenoble Alpes, CEA, CNRS INAC SyMMES UMR 5819, Grenoble, France
| |
Collapse
|
44
|
Verma V, Cushman TR, Tang C, Welsh JW. Toxicity of radiation and immunotherapy combinations. Adv Radiat Oncol 2018; 3:506-511. [PMID: 30370349 PMCID: PMC6200891 DOI: 10.1016/j.adro.2018.08.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 08/05/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022] Open
Abstract
Purpose Although immunotherapy is a rapidly emerging modality for cancer care, there have been multiple reports of fatal toxicities. There have also been cases of treatment-related deaths with combined non-immunotherapeutic biologic compounds with radiation therapy. Thus, provision of summative information appraising the safety of combinatorial immunotherapy and radiation therapy (iRT) is imperative. Because this has not been well characterized, this review summarizes the available evidence to date. Methods and materials Owing to the heterogeneity and relatively low quantity of published reports, this review was conducted in a narrative rather than systematic format. Results The results of combined iRT, both concurrent and sequential, are discussed for oncologic therapy of the brain, lung, liver, and prostate. Most evidence is from small samples and shorter follow-up but does consist of multiple prospective publications. Most data exist for ipilimumab, with programmed cell death -1 inhibitors emerging in more recent years. With 2 large phase 3 trials as exceptions, there were no instances of iRT-related deaths across all discussed studies. Altogether, grade 3 to 4 toxicities were relatively low in frequency; of the studies that compared iRT with an “immunotherapy only” or “RT only” cohort, none documented a clear increase in high-grade adverse events with combined-modality management. Conclusions Despite the low quantity of data, combined iRT offers encouraging safety profiles. There is no evidence that iRT produces an overt increase in high-grade toxicities. Further data, especially on concurrent iRT, are anticipated from numerous iRT trials that are currently ongoing worldwide.
Collapse
Affiliation(s)
- Vivek Verma
- Department of Radiation Oncology, Allegheny General Hospital, Pittsburgh, Pennsylvania
| | - Taylor R Cushman
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chad Tang
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James W Welsh
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
45
|
Stereotactic radiotherapy in metastatic breast cancer. Breast 2018; 41:57-66. [DOI: 10.1016/j.breast.2018.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 05/09/2018] [Accepted: 06/21/2018] [Indexed: 12/19/2022] Open
|
46
|
Mujoo K, Hunt CR, Pandita RK, Ferrari M, Krishnan S, Cooke JP, Hahn S, Pandita TK. Harnessing and Optimizing the Interplay between Immunotherapy and Radiotherapy to Improve Survival Outcomes. Mol Cancer Res 2018; 16:1209-1214. [PMID: 29592896 PMCID: PMC6072560 DOI: 10.1158/1541-7786.mcr-17-0743] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/19/2018] [Accepted: 02/13/2018] [Indexed: 01/06/2023]
Abstract
In the past, radiotherapy was primarily used to control local disease, but recent technological advances in accurate, high-dose ionizing radiation (IR) delivery have not only increased local tumor control but in some cases reduced metastatic burden. These "off target" therapeutic effects of IR at nonirradiated tumor sites, also known as abscopal effects, are thought to be mediated by tumor antigen-primed T cells that travel to metastatic sites and promote tumor regression. Similarly, early indications reveal that IR in combination with immune checkpoint inhibitors, such as ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1), can provide superior therapeutic responses. These observations suggest that local radiotherapy results in altered gene expression, exposure of new antigens, or cell death that can interact with immunotherapy. As such, radiotherapy enhancement of immune responses offers a promising synergy with the potential for substantial clinical benefit. This review focuses on the biology that underlies the mechanisms for the interaction between radiation-induced tumor cell death and enhanced immunologic response. Mol Cancer Res; 16(8); 1209-14. ©2018 AACR.
Collapse
Affiliation(s)
- Kalpana Mujoo
- Department of Radiation Oncology, the Houston Methodist Research Institute, Weil Cornell Medical College, Houston, Texas.
| | - Clayton R Hunt
- Department of Radiation Oncology, the Houston Methodist Research Institute, Weil Cornell Medical College, Houston, Texas
| | - Raj K Pandita
- Department of Radiation Oncology, the Houston Methodist Research Institute, Weil Cornell Medical College, Houston, Texas
| | - Mauro Ferrari
- Department of Nanomedicine, the Houston Methodist Research Institute, Weil Cornell Medical College, Houston, Texas
| | - Sunil Krishnan
- Department of Radiation Oncology, the UT MD Anderson Cancer Center, Houston, Texas
| | - John P Cooke
- Department of Cardiovascular Sciences, the Houston Methodist Research Institute, Weil Cornell Medical College, Houston, Texas
| | - Stephen Hahn
- Department of Radiation Oncology, the UT MD Anderson Cancer Center, Houston, Texas
| | - Tej K Pandita
- Department of Radiation Oncology, the Houston Methodist Research Institute, Weil Cornell Medical College, Houston, Texas.
| |
Collapse
|
47
|
Niknam S, Barsoumian HB, Schoenhals JE, Jackson HL, Yanamandra N, Caetano MS, Li A, Younes AI, Cadena A, Cushman TR, Chang JY, Nguyen QN, Gomez DR, Diab A, Heymach JV, Hwu P, Cortez MA, Welsh JW. Radiation Followed by OX40 Stimulation Drives Local and Abscopal Antitumor Effects in an Anti-PD1-Resistant Lung Tumor Model. Clin Cancer Res 2018; 24:5735-5743. [PMID: 29784675 DOI: 10.1158/1078-0432.ccr-17-3279] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/22/2018] [Accepted: 05/15/2018] [Indexed: 12/11/2022]
Abstract
Purpose: Radiation is used extensively to treat localized cancer, but improved understanding of its effects on the immune system has increased interest in its potential systemic (abscopal) effects, particularly in combination with checkpoint inhibitors such as anti-PD1. The majority of patients either do not respond or develop resistance to monotherapy over time. Here, we investigated the efficacy of OX40 (CD134) stimulation as an alternative immunotherapeutic approach in combination with radiotherapy (XRT) in a murine model of anti-PD1-resistant lung tumors.Experimental Design: We established a bilateral tumor model in 129Sv/Ev mice using an anti-PD1-resistant lung tumor cell line. Primary tumors were treated with intratumoral injection of an OX40 agonist antibody, given as adjuvant therapy after XRT (36 Gy in three 12-Gy fractions), whereas secondary tumors were left untreated to investigate abscopal outcomes.Results: The combination of XRT followed by OX40 stimulation effectively inhibited local and systemic antitumor growth, limited lung metastases, and improved survival rates. This treatment regimen augmented CD4+ and CD8+ T-cell expansion. XRT induced the expression of OX40 on T cells in tumors and spleens and increased the percentages of splenic CD103+ dendritic cells.Conclusions: Our data extend the benefits of radiation to systemic disease control, especially when combined with anti-OX40 agonist to promote immunologically mediated abscopal effects. Moreover, this study provides a rational treatment approach and sequence to overcome anti-PD1-resistant poorly immunogenic tumors. Clin Cancer Res; 24(22); 5735-43. ©2018 AACR.
Collapse
Affiliation(s)
- Sharareh Niknam
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jonathan E Schoenhals
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heather L Jackson
- Immuno-oncology and combinations DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Niranjan Yanamandra
- Immuno-oncology and combinations DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Mauricio S Caetano
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ailin Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ahmed I Younes
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexandra Cadena
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Taylor R Cushman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joe Y Chang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Quynh N Nguyen
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel R Gomez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adi Diab
- Department of Thoracic Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John V Heymach
- Department of Thoracic Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria Angelica Cortez
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
48
|
Emambux S, Tachon G, Junca A, Tougeron D. Results and challenges of immune checkpoint inhibitors in colorectal cancer. Expert Opin Biol Ther 2018; 18:561-573. [PMID: 29471676 DOI: 10.1080/14712598.2018.1445222] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Colorectal cancer (CRC) is the third most commonly diagnosed cancer worldwide and clinical outcome has improved substantially during the last two decades with targeted therapies. The immune system has a major role in cancers, especially the CD8 + T cells specific to tumor antigens. However, tumors can escape immune response by different mechanisms including upregulation of inhibitory immune checkpoint receptors, such as well-known Programmed cell Death protein-1 (PD-1)/Programmed cell Death Ligand 1 (PD-L1) interaction, leading CD8 + T cells to a state of anergy. Immunotherapy, with the so-called immune checkpoint inhibitors (CPIs), has recently been approved in treatment of multiple cancers due to its prolonged disease control and acceptable toxicities. The recent groundbreaking success involving anti-PD-1 CPIs in metastatic CRC with deficient mismatch repair system (dMMR) is promising, with several trials ongoing. Major challenges are ahead in order to determine how, when and for which patients we should use these CPIs in CRC. AREAS COVERED This review highlights some promises and challenges concerning personalized immunotherapy in CRC. First results and ongoing breakthrough trials are presented. The crucial role of biomarkers in selecting patient is also discussed. EXPERT OPINION As of now, dMMR and POLE mutations (DNA polymerase ε) with ultramutator phenotype are the most powerful predictive biomarkers of CPI efficacy. The most challenging issue is pMMR mCRC and determination of how to convert a 'nonimmunogenic' neoplasm into an 'immunogenic' neoplasm, a combination of CPIs with radiation or MEK inhibitor probably being the most relevant strategy. Next-generation sequencing (NGS) assays to quantify mutational load could be more reliable predictive biomarkers of CPIs efficacy than PD-L1 expression or immune scores.
Collapse
Affiliation(s)
- Sheik Emambux
- a Department of Medical Oncology , Poitiers University Hospital , Poitiers , France
| | - Gaelle Tachon
- b Department of Cancer biology , Poitiers University Hospital , Poitiers , France
- c INSERM U-1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases group , University of Poitiers , Poitiers , France
- d University of Poitiers , Faculty of medicine , Poitiers France
| | - Audelaure Junca
- d University of Poitiers , Faculty of medicine , Poitiers France
- e Department of Pathology , Poitiers University Hospital , Poitiers , France
| | - David Tougeron
- a Department of Medical Oncology , Poitiers University Hospital , Poitiers , France
- d University of Poitiers , Faculty of medicine , Poitiers France
- f Department of Gastroenterology , Poitiers University Hospital , Poitiers , France
| |
Collapse
|
49
|
Verma V, Cushman TR, Selek U, Tang C, Welsh JW. Safety of Combined Immunotherapy and Thoracic Radiation Therapy: Analysis of 3 Single-Institutional Phase I/II Trials. Int J Radiat Oncol Biol Phys 2018; 101:1141-1148. [PMID: 30012526 DOI: 10.1016/j.ijrobp.2018.04.054] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/11/2018] [Accepted: 04/18/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE The safety of combined immunotherapy and thoracic radiation therapy (iRT) has been understudied. We evaluated toxicities in patients receiving iRT from 3 single-institutional phase 1/2 trials. METHODS AND MATERIALS Clinical/treatment characteristics and toxicities (per the Common Toxicity Criteria for Adverse Events, version 4.0) were extracted. For purposes of this analysis, groupings were made into (1) patients receiving immunotherapy plus stereotactic body radiation therapy (50 Gy/4 fractions or 60 Gy/10 fractions), (2) immunotherapy plus 45 Gy/15 fractions, and (3) twice-daily chemoimmunoradiotherapy (45 Gy in twice-daily fractions). RESULTS None of the 60 patients undergoing immunotherapy plus stereotactic body radiation therapy (50 Gy, n = 49; 60 Gy, n = 11) experienced grade ≥4 events. There were 34 instances of any grade 3 event (in 15 total patients), with 9 pulmonary specific grade 3 events (in 4 patients). In the patients receiving 45 Gy/15 fractions (small cell lung cancers, n = 26; non-small cell lung cancers, n = 27), there were 2 grade 4 events (in the same patient), along with 17 grade 3 toxicities experienced by 10 total patients (2 pulmonary specific). Lastly, in the twice-daily cohort (n = 22), there were 5 grade 4 events (3 of which occurred in 1 patient) and 16 grade 3 toxicities occurring in 8 total patients (half of which were hematologic). CONCLUSIONS Administration of combined iRT is safe in the short term. Toxicities did not appreciably associate with demographics or dosimetry.
Collapse
Affiliation(s)
- Vivek Verma
- Department of Radiation Oncology, Allegheny General Hospital, Pittsburgh, Pennsylvania
| | - Taylor R Cushman
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ugur Selek
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas; Department of Radiation Oncology, School of Medicine, Koc University, Istanbul, Turkey
| | - Chad Tang
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - James W Welsh
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
50
|
Kesarwani P, Prabhu A, Kant S, Kumar P, Graham SF, Buelow KL, Wilson GD, Miller CR, Chinnaiyan P. Tryptophan Metabolism Contributes to Radiation-Induced Immune Checkpoint Reactivation in Glioblastoma. Clin Cancer Res 2018; 24:3632-3643. [PMID: 29691296 DOI: 10.1158/1078-0432.ccr-18-0041] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/06/2018] [Accepted: 04/20/2018] [Indexed: 12/21/2022]
Abstract
Purpose: Immune checkpoint inhibitors designed to revert tumor-induced immunosuppression have emerged as potent anticancer therapies. Tryptophan metabolism represents an immune checkpoint, and targeting this pathway's rate-limiting enzyme IDO1 is actively being investigated clinically. Here, we studied the intermediary metabolism of tryptophan metabolism in glioblastoma and evaluated the activity of the IDO1 inhibitor GDC-0919, both alone and in combination with radiation (RT).Experimental Design: LC/GC-MS and expression profiling was performed for metabolomic and genomic analyses of patient-derived glioma. Immunocompetent mice were injected orthotopically with genetically engineered murine glioma cells and treated with GDC-0919 alone or combined with RT. Flow cytometry was performed on isolated tumors to determine immune consequences of individual treatments.Results: Integrated cross-platform analyses coupling global metabolomic and gene expression profiling identified aberrant tryptophan metabolism as a metabolic node specific to the mesenchymal and classical subtypes of glioblastoma. GDC-0919 demonstrated potent inhibition of this node and effectively crossed the blood-brain barrier. Although GDC-0919 as a single agent did not demonstrate antitumor activity, it had a strong potential for enhancing RT response in glioblastoma, which was further augmented with a hypofractionated regimen. RT response in glioblastoma involves immune stimulation, reflected by increases in activated and cytotoxic T cells, which was balanced by immune checkpoint reactivation, reflected by an increase in IDO1 expression and regulatory T cells (Treg). GDC-0919 mitigated RT-induced Tregs and enhanced T-cell activation.Conclusions: Tryptophan metabolism represents a metabolic node in glioblastoma, and combining RT with IDO1 inhibition enhances therapeutic response by mitigating RT-induced immunosuppression. Clin Cancer Res; 24(15); 3632-43. ©2018 AACR.
Collapse
Affiliation(s)
- Pravin Kesarwani
- Department of Radiation Oncology, Beaumont Health, Royal Oak, Michigan
| | - Antony Prabhu
- Department of Radiation Oncology, Beaumont Health, Royal Oak, Michigan
| | - Shiva Kant
- Department of Radiation Oncology, Beaumont Health, Royal Oak, Michigan
| | - Praveen Kumar
- Metabolomics and Obstetrics/Gynecology, Beaumont Research Institute, Beaumont Health, Royal Oak, Michigan
| | - Stewart F Graham
- Metabolomics and Obstetrics/Gynecology, Beaumont Research Institute, Beaumont Health, Royal Oak, Michigan
| | - Katie L Buelow
- Department of Radiation Oncology, Beaumont Health, Royal Oak, Michigan
| | - George D Wilson
- Department of Radiation Oncology, Beaumont Health, Royal Oak, Michigan
| | - C Ryan Miller
- Department of Pathology & Laboratory Medicine, Neurology, & Pharmacology, Lineberger Comprehensive Cancer Center and Neurosciences Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Prakash Chinnaiyan
- Department of Radiation Oncology, Beaumont Health, Royal Oak, Michigan. .,Oakland University William Beaumont School of Medicine, Royal Oak, Michigan
| |
Collapse
|