1
|
Hovhannisyan L, Riether C, Aebersold DM, Medová M, Zimmer Y. CAR T cell-based immunotherapy and radiation therapy: potential, promises and risks. Mol Cancer 2023; 22:82. [PMID: 37173782 PMCID: PMC10176707 DOI: 10.1186/s12943-023-01775-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
CAR T cell-based therapies have revolutionized the treatment of hematological malignancies such as leukemia and lymphoma within the last years. In contrast to the success in hematological cancers, the treatment of solid tumors with CAR T cells is still a major challenge in the field and attempts to overcome these hurdles have not been successful yet. Radiation therapy is used for management of various malignancies for decades and its therapeutic role ranges from local therapy to a priming agent in cancer immunotherapy. Combinations of radiation with immune checkpoint inhibitors have already proven successful in clinical trials. Therefore, a combination of radiation therapy may have the potential to overcome the current limitations of CAR T cell therapy in solid tumor entities. So far, only limited research was conducted in the area of CAR T cells and radiation. In this review we will discuss the potential and risks of such a combination in the treatment of cancer patients.
Collapse
Affiliation(s)
- Lusine Hovhannisyan
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, 3010, Switzerland
| | - Carsten Riether
- Department of Medical Oncology, Inselspital, University Hospital and University of Bern, Bern, 3010, Switzerland
| | - Daniel M Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland
| | - Michaela Medová
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland
| | - Yitzhak Zimmer
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland.
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland.
| |
Collapse
|
2
|
Hu F, Zheng XH, Li T, She HL, Zhang SF. Brain Perfusion Abnormalities after Radiotherapy Measured by 3-Dimensional Arterial Spin Labeling MRI and Correlations with Cognitive Impairment. Radiat Res 2022; 197:324-331. [PMID: 35104874 DOI: 10.1667/rade-21-00143.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 12/10/2021] [Indexed: 11/03/2022]
Abstract
The purpose of this study was to use a 3-dimensional arterial spin labeling (3D ASL) magnetic resonance (MR) method to measure cerebral blood flow (CBF) before and after radiotherapy, and correlate changes with time after receiving radiotherapy and cognitive function. Patients with nasopharyngeal carcinoma receiving radiotherapy at our institution were recruited for the study. Participants were divided into three groups: Pre-radiotherapy control (PC) group, acute reaction period (ARP) group, and delayed reaction period (DRP)group. Thirty-four patients were included in the study. Compared with the PC group, the ARP group exhibited significantly decreased perfusion in the left anterior cingulate cortex (ACC) and right putamen, and increased perfusion in the right cerebellum (Crus 1), right inferior occipital gyrus, left lingual gyrus, left precuneus, and left calcarine gyrus. in the DRP group, increased perfusion was noted in the right cerebellum (Crus 1) and decreased perfusion in the left superior frontal gyrus. CBF differences were observed in several brain areas in the DRP group as compared to the ARP group (P < 0.001). Total Montreal Cognitive Assessment score, and subdomain language and delayed memory recall scores were significantly lower in the ARP and DRP groups than in the PC group (P < 0.05). Data suggest that ASL allows for non-invasive detection of radiation-induced whole-brain CBF changes, which is transient, dynamic and complicated and may be a factor contributing to cognitive impairment induced by radiotherapy for nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Fang Hu
- Medical Imaging and Inspection Institute, Xiangnan University, Chenzhou, Hunan Province, P. R. China.,Department of Radiology, the Affiliated Hospital of Xiangnan University, Chenzhou, Hunan Province, China.,Key Laboratory of Medical Imaging and Artifical Intelligence of Hunan Province, Xiangnan University, Chenzhou 423000, China.,Chenzhou Cognitive Degeneration Brain Disease Early Warning Technology Research and Development Center, Affiliated Hospital of Xiangnan University, Chenzhou 423000, China
| | - Xin-Hui Zheng
- Medical Imaging and Inspection Institute, Xiangnan University, Chenzhou, Hunan Province, P. R. China
| | - Tao Li
- Medical Imaging and Inspection Institute, Xiangnan University, Chenzhou, Hunan Province, P. R. China.,Department of Radiology, the Affiliated Hospital of Xiangnan University, Chenzhou, Hunan Province, China.,Key Laboratory of Medical Imaging and Artifical Intelligence of Hunan Province, Xiangnan University, Chenzhou 423000, China.,Chenzhou Cognitive Degeneration Brain Disease Early Warning Technology Research and Development Center, Affiliated Hospital of Xiangnan University, Chenzhou 423000, China
| | - Hua-Long She
- Department of Radiology, the Affiliated Hospital of Xiangnan University, Chenzhou, Hunan Province, China.,Key Laboratory of Medical Imaging and Artifical Intelligence of Hunan Province, Xiangnan University, Chenzhou 423000, China.,Chenzhou Cognitive Degeneration Brain Disease Early Warning Technology Research and Development Center, Affiliated Hospital of Xiangnan University, Chenzhou 423000, China
| | - Sheng-Fu Zhang
- Department of Anus and Intestine Surgery, the First People's Hospital of Chenzhou, Chenzhou, Hunan Province, China.,Medical Imaging Center, the First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong Province, China
| |
Collapse
|
3
|
Abstract
Immunotherapy (IO) has become a standard treatment in patients with metastatic and locally advanced non-small cell lung cancer (NSCLC), and is now being tested in patients with early stage disease. IO agents currently in use for lung cancer target PD-1, PD-L1, and CTLA-4. While survival and tumor control have improved with IO, many patients have limited or short responses to IO. Therefore, methods to improve the systemic response to IO are needed. Radiation therapy (RT) is an integral component of lung cancer treatment, and may improve systemic response to IO by increasing antigen presentation, increasing co-stimulatory signaling, increasing T-cells recruitment, upregulating PD-L1, increasing tumor stromal lymphocyte infiltration, and altering the microenvironment. IO after definitive chemoradiation is now standard treatment in unresectable stage III NSCLC following publication of the PACIFIC clinical trial. For early stage NSCLC, IO is being investigated in conjunction with stereotactic body radiotherapy (SBRT). The benefit of adding RT to IO in patients with metastatic disease may be especially pronounced in patients with low baseline PD-L1 expression, potentially when delivered as a short course of SBRT, as supported by the PEMBRO-RT clinical trial. Current and ongoing clinical trials are evaluating the optimal radiation dose, timing, and sequencing of RT with IO.
Collapse
Affiliation(s)
- William G Breen
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | | | - Haidong Dong
- Departments of Urology and Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | | |
Collapse
|
4
|
Xia WY, Feng W, Zhang CC, Shen YJ, Zhang Q, Yu W, Cai XW, Fu XL. Radiotherapy for non-small cell lung cancer in the immunotherapy era: the opportunity and challenge-a narrative review. Transl Lung Cancer Res 2020; 9:2120-2136. [PMID: 33209631 PMCID: PMC7653139 DOI: 10.21037/tlcr-20-827] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022]
Abstract
Immunotherapy has radically changed the clinical management of patients with cancer in recent years. Immune checkpoint inhibitors (ICIs) reversing the immunosuppressive effects of the tumor microenvironment are one type of immunotherapy, several of which are approved by the US Food and Drug Administration (FDA) as first-line treatments for patients with non-small cell lung cancer (NSCLC). However, response rates to ICIs are around 19-47% among patients with advanced NSCLC. As a result, the development of combined ICI and radiotherapy has begun with the aim of strengthening patients' antitumor immunity. Radiotherapy with substantial technological improvements not only achieves local tumor control through the induction of deoxyribonucleic acid (DNA) damage in irradiated regions, but also has the potential to mediate immunostimulatory effects that could result in tumor regression beyond irradiated regions. At present, numerous preclinical and clinical research are investigating the efficiency and safety of combining ICI with radiotherapy. The PACIFIC trial showed that combining chemoradiotherapy with ICI could improve clinical outcomes. In this review, we summarize the rationale for combining radiotherapy with immunotherapy. We also discuss the opportunities and challenges of combination therapy, including the timing of radiotherapy, optimal dose and fractionations, radiotherapy target and target volume, acquired resistance, patient selection, and radioimmunotherapy toxicity.
Collapse
Affiliation(s)
- Wu-Yan Xia
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wen Feng
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chen-Chen Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yu-Jia Shen
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qin Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wen Yu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xu-Wei Cai
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | | |
Collapse
|
5
|
Hübbe ML, Jæhger DE, Andresen TL, Andersen MH. Leveraging Endogenous Dendritic Cells to Enhance the Therapeutic Efficacy of Adoptive T-Cell Therapy and Checkpoint Blockade. Front Immunol 2020; 11:578349. [PMID: 33101304 PMCID: PMC7546347 DOI: 10.3389/fimmu.2020.578349] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/26/2020] [Indexed: 01/15/2023] Open
Abstract
Adoptive cell therapy (ACT), based on treatment with autologous tumor infiltrating lymphocyte (TIL)-derived or genetically modified chimeric antigen receptor (CAR) T cells, has become a potentially curative therapy for subgroups of patients with melanoma and hematological malignancies. To further improve response rates, and to broaden the applicability of ACT to more types of solid malignancies, it is necessary to explore and define strategies that can be used as adjuvant treatments to ACT. Stimulation of endogenous dendritic cells (DCs) alongside ACT can be used to promote epitope spreading and thereby decrease the risk of tumor escape due to target antigen downregulation, which is a common cause of disease relapse in initially responsive ACT treated patients. Addition of checkpoint blockade to ACT and DC stimulation might further enhance response rates by counteracting an eventual inactivation of infused and endogenously primed tumor-reactive T cells. This review will outline and discuss therapeutic strategies that can be utilized to engage endogenous DCs alongside ACT and checkpoint blockade, to strengthen the anti-tumor immune response.
Collapse
Affiliation(s)
- Mie Linder Hübbe
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Copenhagen, Denmark
| | - Ditte Elisabeth Jæhger
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Thomas Lars Andresen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Copenhagen, Denmark
| |
Collapse
|
6
|
Huang J, Li JJ. Multiple Dynamics in Tumor Microenvironment Under Radiotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1263:175-202. [PMID: 32588328 DOI: 10.1007/978-3-030-44518-8_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The tumor microenvironment (TME) is an evolutionally low-level and embryonically featured tissue comprising heterogenic populations of malignant and stromal cells as well as noncellular components. Under radiotherapy (RT), the major modality for the treatment of malignant diseases [1], TME shows an adaptive response in multiple aspects that affect the efficacy of RT. With the potential clinical benefits, interests in RT combined with immunotherapy (IT) are intensified with a large scale of clinical trials underway for an array of cancer types. A better understanding of the multiple molecular aspects, especially the cross talks of RT-mediated energy reprogramming and immunoregulation in the irradiated TME (ITME), will be necessary for further enhancing the benefit of RT-IT modality. Coming studies should further reveal more mechanistic insights of radiation-induced instant or permanent consequence in tumor and stromal cells. Results from these studies will help to identify critical molecular pathways including cancer stem cell repopulation, metabolic rewiring, and specific communication between radioresistant cancer cells and the infiltrated immune active lymphocytes. In this chapter, we will focus on the following aspects: radiation-repopulated cancer stem cells (CSCs), hypoxia and re-oxygenation, reprogramming metabolism, and radiation-induced immune regulation, in which we summarize the current literature to illustrate an integrated image of the ITME. We hope that the contents in this chapter will be informative for physicians and translational researchers in cancer radiotherapy or immunotherapy.
Collapse
Affiliation(s)
- Jie Huang
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA. .,NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
7
|
Yang H, Jin T, Li M, Xue J, Lu B. Synergistic effect of immunotherapy and radiotherapy in non-small cell lung cancer: current clinical trials and prospective challenges. PRECISION CLINICAL MEDICINE 2019; 2:57-70. [PMID: 35694698 PMCID: PMC8985786 DOI: 10.1093/pcmedi/pbz004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 01/31/2019] [Accepted: 02/02/2019] [Indexed: 02/05/2023] Open
Abstract
Lately, the success of ICIs has drastically changed the landscape of cancer treatment, and several immune checkpoint inhibitors (ICIs) have been approved by the US Food and Drug Administration (FDA) for advanced non-small cell lung cancer (NSCLC). However, numerous patients are resistant to ICIs and require additional procedures for better efficacy results. Thus, combination therapy is urgently needed to strengthen the anti-tumor immunity. A variety of preclinical and clinical studies combining ICIs with radiotherapy (RT) have demonstrated that the combination could induce synergistic effects, as RT overcomes the resistance to ICIs. However, the underlying mechanism of the synergistic effect and the optimal arrangement of the combination therapy are indecisive now. Hence, this review was conducted to provide an update on the current clinical trial results and highlighted the ongoing trials. We also discussed the optimal parameters in clinical trials, including radiation dose, radiation fractionation, radiation target field, and sequencing of combination therapy. In this review, we found that combination therapy showed stronger anti-tumor immunity with tolerable toxicities in clinical trials. However, the best combination mode and potential biomarkers for the target patients in combination therapy are still unclear.
Collapse
Affiliation(s)
- Hui Yang
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Jin
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| | - Mengqian Li
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Lu
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
8
|
|
9
|
Abstract
Immunotherapy, particularly immune-checkpoint inhibition, is producing encouraging clinical responses and affecting the way numerous cancers are treated. Yet immune-checkpoint therapy is not effective for many patients, and even those who initially respond can experience relapse, fueling interest in finding new processes or tools to improve the effectiveness of these novel therapeutics. One such tool is radiation. Both preclinical and clinical studies have demonstrated that the systemic effects of immunotherapy can be amplified when it is used in combination with radiation and, conversely, that the immunogenic effects of local irradiation can be amplified and extended to distant sites when used with immunotherapy. We review how stereotactic ablative radiation therapy, a technique specifically indicated for tumors treated with immune-checkpoint inhibitors, can potentiate the effects of immune-checkpoint therapy. We further explore how these novel therapeutics may transform radiation, previously considered a local treatment option, into powerful systemic therapy.
Collapse
|
10
|
Bashiardes S, Tuganbaev T, Federici S, Elinav E. The microbiome in anti-cancer therapy. Semin Immunol 2017; 32:74-81. [PMID: 28431920 DOI: 10.1016/j.smim.2017.04.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/21/2017] [Accepted: 04/05/2017] [Indexed: 02/07/2023]
Abstract
The commensal microbiome constitutes an important modulator of host physiology and risk of disease, including cancer development and progression. Lately, the microbiome has been suggested to modulate the efficacy of anti-cancer treatment. Examples include chemotherapy and total body irradiation-induced barrier function disruption, leading to microbial efflux that drives activation of anti-tumorigenic T cells; Microbiome-driven release of reactive oxygen species contributing to the efficacy of platinum salts; and microbiome-induced immune priming promoting the anti-tumor effects of alkylating chemotherapy and immune checkpoint inhibitors. Furthermore, selected commensals are able to colonize solid tumors. This 'tumor microbiome' may further impact local tumor responses to treatment and potentially be harnessed for tumor-specific targeting and therapeutic delivery. In this review, we present recent advances in understanding of the intricate role of microbiome in modulating efficacy of a number of anti-cancer treatments, and discuss how anti-cancer treatment approaches utilizing the tumor microbiome may enhance oncological treatment efficacy.
Collapse
Affiliation(s)
- Stavros Bashiardes
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Timur Tuganbaev
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Sara Federici
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel.
| |
Collapse
|
11
|
Radiation-induced decrease of CD8+ dendritic cells contributes to Th1/Th2 shift. Int Immunopharmacol 2017; 46:178-185. [PMID: 28314222 DOI: 10.1016/j.intimp.2017.03.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/09/2017] [Accepted: 03/10/2017] [Indexed: 11/21/2022]
Abstract
Exposure to ionizing radiation (IR) often reduce the helper T (Th) 1 like function, resulting in a Th1/Th2 imbalance, which could affect the efficacy of cancer radiotherapy. As the most potent antigen presenting cells, dendritic cells (DC) can be divided into several subsets with specialized function. However, there is no literature covering the changes of DC subsets and their roles in immune regulation in response to IR. In the present study, we were aimed to investigate the changes of DC subsets after IR and its relationship with Th1/Th2 immunity. We found a significant decrease of BDCA3+DC in the blood of patients treated with radiotherapy. CD8+DC, a mouse equivalent of human BDCA3+DC, was also found decreased in mice spleen, peripheral blood and lymph node tissues after irradiation. As CD8+DC mainly induce Th1 immunity, we tested the changes of Th1/Th2 response and found that IR caused a repression of Th1 immunity, indicating a possible role of CD8+DC in radiation-induced Th1/Th2 imbalance. We also found that a CD8+DC-inducing cytokine, Fms-like tyrosine kinase 3 ligand (FLT3 ligand), restored CD8+DC and reversed Th1/Th2 shift. And then we found that bone marrow cells from irradiated mice differentiated into less CD8+DC, which was also protected by FLT3 ligand. In conclusion, our data showed that IR induced a decrease of CD8+DC and Th1/Th2 shift, which was reversed by Flt3 ligand treatment, suggesting a novel mechanism for radiation-induced immunosuppression.
Collapse
|
12
|
Combination treatment with decitabine and ionizing radiation enhances tumor cells susceptibility of T cells. Sci Rep 2016; 6:32470. [PMID: 27671170 PMCID: PMC5037374 DOI: 10.1038/srep32470] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 08/09/2016] [Indexed: 02/07/2023] Open
Abstract
Decitabine has been found to have anti-metabolic and anti-tumor activities in various tumor cells. Recently, the use of decitabine in combination with other conventional therapies reportedly resulted in improved anti-tumor activity against various tumors. Ionizing radiation (IR) is widely used as a cancer treatment. Decitabine and IR improve immunogenicity and susceptibility of tumor cells to immune cells by up-regulating the expression of various molecules such as major histocompatibility complex (MHC) class I; natural-killer group 2, member D (NKG2D) ligands; and co-stimulatory molecules. However, the effects of combining decitabine and IR therapies are largely unknown. Our results indicate that decitabine or IR treatment upregulates MHC class I, along with various co-stimulatory molecules in target tumor cells. Furthermore, decitabine and IR combination treatment further upregulates MHC class I, along with the co-stimulatory molecules, when compared to the effect of each treatment alone. Importantly, decitabine treatment further enhanced T cell-mediated cytotoxicity and release of IFN- γ against target tumor cells which is induced by IR. Interestingly, decitabine did not affect NKG2D ligand expression or NK cell-mediated cytotoxicity in target tumor cells. These observations suggest that decitabine may be used as a useful immunomodulator to sensitize tumor cells in combination with other tumor therapies.
Collapse
|
13
|
Harnessing the Microbiome to Enhance Cancer Immunotherapy. J Immunol Res 2015; 2015:368736. [PMID: 26101781 PMCID: PMC4458560 DOI: 10.1155/2015/368736] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/10/2015] [Indexed: 12/20/2022] Open
Abstract
The microbiota plays a key role in regulating the innate and adaptive immune system. Herein, we review the immunological aspects of the microbiota in tumor immunity in mice and man, with a focus on toll-like receptor (TLR) agonists, vaccines, checkpoint modulators, chemotherapy, and adoptive T cell transfer (ACT) therapies. We propose innovative treatments that may safely harness the microbiota to enhance T cell-based therapies in cancer patients. Finally, we highlight recent developments in tumor immunotherapy, particularly novel ways to modulate the microbiome and memory T cell responses to human malignancies.
Collapse
|
14
|
Inflammatory cells of immunosuppressive phenotypes in oral lichen planus have a proinflammatory pattern of expression and are associated with clinical parameters. Clin Oral Investig 2012; 17:1365-73. [DOI: 10.1007/s00784-012-0814-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 07/23/2012] [Indexed: 12/30/2022]
|
15
|
Wang HZ, Qiu SJ, Lv XF, Wang YY, Liang Y, Xiong WF, Ouyang ZB. Diffusion tensor imaging and 1H-MRS study on radiation-induced brain injury after nasopharyngeal carcinoma radiotherapy. Clin Radiol 2011; 67:340-5. [PMID: 22119296 DOI: 10.1016/j.crad.2011.09.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 09/17/2011] [Accepted: 09/27/2011] [Indexed: 11/20/2022]
Abstract
AIM To investigate the metabolic characteristics of the temporal lobes following radiation therapy for nasopharyngeal carcinoma using diffusion tensor imaging (DTI) and proton magnetic resonance spectroscopy ((1)H-MRS). MATERIALS AND METHODS DTI and (1)H-MRS were performed in 48 patients after radiotherapy for nasopharyngeal carcinoma and in 24 healthy, age-matched controls. All patients and controls had normal findings on conventional MRI. Apparent diffusion coefficient (ADC), fractional anisotropy (FA), three eigenvalues λ1, λ2, λ3, N-acetylaspartic acid (NAA)/choline (Cho), NAA/creatinine (Cr), and Cho/Cr were measured in both temporal lobes. Patients were divided into three groups according to time after completion of radiotherapy: group 1, less than 6 months; group 2, 6-12 months; group 3, more than 12 months. Mean values for each parameter were compared using one-way analysis of variance (ANOVA). RESULTS Mean FA in group 1 was significantly lower compared to group 3 and the control group (p < 0.05). Group-wise comparisons of apparent diffusion coefficient (ADC) values among all the groups were not significantly different. Eigenvalue λ1 was significantly lower in groups 1 and 3 compared to the control group (p < 0.05). NAA/Cho and NAA/Cr were significantly lower in each group compared to the control group (p < 0.01 for both). The decrease in NAA/Cho was greatest in group 1. There were no significant between-group differences regarding Cho/Cr. CONCLUSION A combination of DTI and (1)H-MRS can be used to detect radiation-induced brain injury, in patients treated for nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- H-Z Wang
- Department of Medical Imaging Center, Nan Fang Hospital, Southern Medical University, Guangzhou 510515, China
| | | | | | | | | | | | | |
Collapse
|
16
|
Salem ML, Cole DJ. Dendritic cell recovery post-lymphodepletion: a potential mechanism for anti-cancer adoptive T cell therapy and vaccination. Cancer Immunol Immunother 2010; 59:341-353. [PMID: 19921513 PMCID: PMC3070377 DOI: 10.1007/s00262-009-0792-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Accepted: 10/26/2009] [Indexed: 02/07/2023]
Abstract
Adoptive transfer of autologous tumor-reactive T cells holds promise as a cancer immunotherapy. In this approach, T cells are harvested from a tumor-bearing host, expanded in vitro and infused back to the same host. Conditioning of the recipient host with a lymphodepletion regimen of chemotherapy or radiotherapy before adoptive T cell transfer has been shown to substantially improve survival and anti-tumor responses of the transferred cells. These effects are further enhanced when the adoptive T cell transfer is followed by vaccination with tumor antigens in combination with a potent immune adjuvant. Although significant progress has been made toward an understanding of the reasons underlying the beneficial effects of lymphodepletion to T cell adoptive therapy, the precise mechanisms remain poorly understood. Recent studies, including ours, would indicate a more central role for antigen presenting cells, in particular dendritic cells. Unraveling the exact role of these important cells in mediation of the beneficial effects of lymphodepletion could provide novel pathways toward the rational design of more effective anti-cancer immunotherapy. This article focuses on how the frequency, phenotype, and functions of dendritic cells are altered during the lymphopenic and recovery phases post-induction of lymphodepletion, and how they affect the anti-tumor responses of adoptively transferred T cells.
Collapse
Affiliation(s)
- Mohamed Labib Salem
- Surgery Department, Medical University of South Carolina, Charleston, 29425, USA.
| | | |
Collapse
|
17
|
Salem ML, Díaz-Montero CM, Al-Khami AA, El-Naggar SA, Naga O, Montero AJ, Khafagy A, Cole DJ. Recovery from cyclophosphamide-induced lymphopenia results in expansion of immature dendritic cells which can mediate enhanced prime-boost vaccination antitumor responses in vivo when stimulated with the TLR3 agonist poly(I:C). JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:2030-40. [PMID: 19201856 PMCID: PMC3066095 DOI: 10.4049/jimmunol.0801829] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recent preclinical studies suggest that vaccination following adoptive transfer of CD8(+) T cells into a lymphopenic host can augment the therapeutic antitumor responses of the transferred cells. However, the mechanism by which the lymphopenic microenvironment benefits Ag-specific CD8(+) T cell responses remains elusive. We show herein that induction of lymphodepletion by a single 4 mg cyclophosphamide (CTX) treatment induces a marked expansion of immature dendritic cells (DCs) in the peripheral blood on days 8-16 post-CTX (termed restoration phase). In vitro, these DCs were functional, because they showed normal phagocytosis and effective Ag presentation capability upon activation. In vivo, administration of the TLR3 agonist poly(I:C) at the peak of DC expansion (day 12 postlymphopenia) induced inflammatory cytokine production and increases in the number of activated DCs in lymph nodes. Importantly, boosting with gp100(25-33) melanoma peptide combined with poly(I:C) 12 days after an initial priming with the same regimen significantly increased the expansion and the antitumor efficacy of adoptively transferred pmel-1 CD8(+) T cells. These responses were abrogated after depletion of activated DCs during Ag boosting. In conclusion, our data show that CTX treatment induces, during the restoration phase, expansion of immature DCs, which are functional and can be exploited in vivo to foster more effective antitumor adoptive immunotherapy strategies.
Collapse
Affiliation(s)
- Mohamed L Salem
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Tatum AM, Mylin LM, Bender SJ, Fischer MA, Vigliotti BA, Tevethia MJ, Tevethia SS, Schell TD. CD8+ T cells targeting a single immunodominant epitope are sufficient for elimination of established SV40 T antigen-induced brain tumors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:4406-17. [PMID: 18768900 PMCID: PMC2631553 DOI: 10.4049/jimmunol.181.6.4406] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Immunotherapy of established solid tumors is rarely achieved, and the mechanisms leading to success remain to be elucidated. We previously showed that extended control of advanced-stage autochthonous brain tumors is achieved following adoptive transfer of naive C57BL/6 splenocytes into sublethally irradiated line SV11 mice expressing the SV40 T Ag (T Ag) oncoprotein, and was associated with in vivo priming of CD8(+) T cells (T(CD8)) specific for the dominant epitope IV (T Ag residues 404-411). Using donor lymphocytes derived from mice that are tolerant to epitope IV or a newly characterized transgenic mouse line expressing an epitope IV-specific TCR, we show that epitope IV-specific T(CD8) are a necessary component of the donor pool and that purified naive epitope IV-specific T(CD8) are sufficient to promote complete and rapid regression of established tumors. While transfer of naive TCR-IV cells alone induced some initial tumor regression, increased survival of tumor-bearing mice required prior conditioning of the host with a sublethal dose of gamma irradiation and was associated with complete tumor eradication. Regression of established tumors was associated with rapid accumulation of TCR-IV T cells within the brain following initial priming against the endogenous T Ag in the peripheral lymphoid organs. Additionally, persistence of functional TCR-IV cells in both the brain and peripheral lymphoid organs was associated with long-term tumor-free survival. Finally, we show that production of IFN-gamma, but not perforin or TNF-alpha, by the donor lymphocytes is critical for control of autochthonous brain tumors.
Collapse
MESH Headings
- Animals
- Antigens, Polyomavirus Transforming/immunology
- Brain Neoplasms/immunology
- Brain Neoplasms/pathology
- Brain Neoplasms/therapy
- Brain Neoplasms/virology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/transplantation
- CD8-Positive T-Lymphocytes/virology
- Cell Line, Transformed
- Cells, Cultured
- Choroid Plexus Neoplasms/immunology
- Choroid Plexus Neoplasms/pathology
- Choroid Plexus Neoplasms/therapy
- Choroid Plexus Neoplasms/virology
- Cytotoxicity, Immunologic
- Female
- Immunodominant Epitopes/genetics
- Immunodominant Epitopes/immunology
- Immunodominant Epitopes/metabolism
- Immunotherapy, Adoptive/methods
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Rats
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Simian virus 40/immunology
Collapse
Affiliation(s)
- Angela M. Tatum
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Lawrence M. Mylin
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Susan J. Bender
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Matthew A. Fischer
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Beth A. Vigliotti
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - M. Judith Tevethia
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Satvir S. Tevethia
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Todd D. Schell
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| |
Collapse
|
19
|
Weiss L, Reich S, Zeira M, Or R, Resnick IB, Slavin S, Shapira MY. N-acetylcysteine mildly inhibits the graft-vs.-leukemia effect but not the lymphokine activated cells (LAK) activity. Transpl Immunol 2007; 17:198-202. [PMID: 17331847 DOI: 10.1016/j.trim.2006.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2006] [Revised: 10/26/2006] [Accepted: 10/31/2006] [Indexed: 11/26/2022]
Abstract
N-acetylcysteine (NAC) is a known antioxidant and induces modulation of glutathione cellular content effects. It has been suggested that in the context of stem cell transplantation (SCT), NAC can prevent and treat graft-vs.-host disease, veno-occlusive disease and idiopathic pneumonia syndrome. We investigated the possible effect of NAC on graft-vs.-leukemia effect (GVL) and lymphokine activated cells (LAK) activity in murine models. After 10 days of NAC treatment, the cytotoxic activity of the LAK cells did not significantly differ from LAK activity generated from spleen cells obtained from untreated controls. However, NAC mildly suppressed GVL (appearance of leukemia in 8/36 animals treated with NAC as compared to 0/20 in the SCT control group, p=0.023). In spite of this mild suppression of GVL, no negative effect on achievement of donor chimerism was seen. We conclude that NAC usage in SCT may be relatively safe with regard to the GVL effect, yet further clinical studies are warranted.
Collapse
Affiliation(s)
- Lola Weiss
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
20
|
Meng Y, Carpentier AF, Chen L, Boisserie G, Simon JM, Mazeron JJ, Delattre JY. Successful combination of local CpG-ODN and radiotherapy in malignant glioma. Int J Cancer 2005; 116:992-7. [PMID: 15856470 DOI: 10.1002/ijc.21131] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Oligodeoxynucleotides containing CpG motifs (CpG-ODN) display broad immunostimulating activity and are currently under clinical trial in various malignancies, including recurrent glioblastomas. Combining CpG-ODN with another therapy that could induce antigen release might enhance tumor-specific immune response. We investigated whether radiotherapy (RT) could be associated advantageously to intratumoral injections of CpG-ODN. Fisher rats bearing 9L glioma were treated with various combinations of RT and CpG-28, an oligonucleotide with good immunostimulating activity. RT and CpG-28 induced complete tumor remission in one-third of the animals. When both treatments were combined, complete tumor remission was achieved in two-thirds of the animals (p < 0.001 when compared to non-treated rats, p < 0.03 when compared to CpG-28 alone). Such efficacy was not observed in nude mice, underlying the role of T cells in antitumor effects. The combination of both treatments appeared optimal when the delay between RT and CpG-28 administration was <3 days (from 100% survival for a 3 days delay, to 57% survival for a 21 days delay, p < 0.05). Tumor infiltration by immune cells and expression within tumors of the CpG receptor, TLR9, were not modified by irradiation. These results support an attractive strategy of sequential radiotherapy and immunotherapy by CpG-ODN and have potential implications for future clinical trials with CpG-ODN.
Collapse
Affiliation(s)
- Yuxia Meng
- Fédération de neurologie Mazarin and Institut National de la Santé et de la Recherche Médicale, UMR-495, Hôpital de la Salpêtrière, Paris, France
| | | | | | | | | | | | | |
Collapse
|
21
|
Klebanoff CA, Khong HT, Antony PA, Palmer DC, Restifo NP. Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol 2005; 26:111-7. [PMID: 15668127 PMCID: PMC1388277 DOI: 10.1016/j.it.2004.12.003] [Citation(s) in RCA: 355] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Christopher A Klebanoff
- Howard Hughes Medical Institute-National Institutes of Health Research Scholars Program, Bethesda, MD 20814, USA
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1502, USA
| | - Hung T Khong
- University of South Alabama Cancer Research Institute, 307 N. University Blvd./MSB 2015, Mobile, AL 36688-0002, USA
| | - Paul A Antony
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1502, USA
| | - Douglas C Palmer
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1502, USA
- Corresponding author: Palmer, D.C. ()
| | - Nicholas P Restifo
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1502, USA
| |
Collapse
|