51
|
Current advances in immune checkpoint inhibitor combinations with radiation therapy or cryotherapy for breast cancer. Breast Cancer Res Treat 2021; 191:229-241. [PMID: 34714450 DOI: 10.1007/s10549-021-06408-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE Immune checkpoint inhibition (ICI) has demonstrated clinically significant efficacy when combined with chemotherapy in triple negative breast cancer (TNBC). Although many patients derived benefit, others do not respond to immunotherapy, therefore relying upon innovative combinations to enhance response. Local therapies such as radiation therapy (RT) and cryotherapy are immunogenic and potentially optimize responses to immunotherapy. Strategies combining these therapies and ICI are actively under investigation. This review will describe the rationale for combining ICI with targeted local therapies in breast cancer. METHODS A literature search was performed to identify pre-clinical and clinical studies assessing ICI combined with RT or cryotherapy published as of August 2021 using PubMed and ClinicalTrials.gov. RESULTS Published studies of ICI with RT and IPI have demonstrated safety and signals of early efficacy. CONCLUSION RT and cryotherapy are local therapies that can be integrated safely with ICI and has shown promise in early trials. Randomized phase II studies testing both of these approaches, such as P-RAD (NCT04443348) and ipilimumab/nivolumab/cryoablation for TNBC (NCT03546686) are current enrolling. The results of these studies are paramount as they will provide long term data on the safety and efficacy of these regimens.
Collapse
|
52
|
Fujiwara K, Saung MT, Jing H, Herbst B, Zarecki M, Muth S, Wu A, Bigelow E, Chen L, Li K, Jurcak N, Blair AB, Ding D, Wichroski M, Blum J, Cheadle N, Koenitzer J, Zheng L. Interrogating the immune-modulating roles of radiation therapy for a rational combination with immune-checkpoint inhibitors in treating pancreatic cancer. J Immunother Cancer 2021; 8:jitc-2019-000351. [PMID: 32675194 PMCID: PMC7368549 DOI: 10.1136/jitc-2019-000351] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Radiation therapy (RT) has the potential to enhance the efficacy of immunotherapy, such as checkpoint inhibitors, which has dramatically altered the landscape of treatments for many cancers, but not yet for pancreatic ductal adenocarcinoma (PDAC). Our prior studies demonstrated that PD ligand-1 and indoleamine 2,3-dioxygenase 1 (IDO1) were induced on tumor epithelia of PDACs following neoadjuvant therapy including RT, suggesting RT may prime PDAC for PD-1 blockade antibody (αPD-1) or IDO1 inhibitor (IDO1i) treatments. In this study, we investigated the antitumor efficacy of the combination therapies with radiation and PD-1 blockade or IDO1 inhibition or both. METHODS We developed and used a mouse syngeneic orthotopic model of PDAC suitable for hypofractionated RT experiments. RESULTS The combination therapy of αPD-1 and RT improved survival. The dual combination of RT/IDO1i and triple combination of RT/αPD-1/IDO1i did not improve survival compared with RT/αPD-1, although all of these combinations offer similar local tumor control. RT/αPD-1 appeared to result in the best systemic interferon-γ response compared with other treatment groups and the highest local expression of immune-activation genes, including Cd28 and Icos. CONCLUSION Our RT model allows examining the immune-modulatory effects of RT alone and in combination with immune-checkpoint inhibitors in the pancreas/local microenvironment. This study highlights the importance of choosing the appropriate immune-modulatory agents to be combined with RT to tip the balance toward antitumor adaptive immune responses.
Collapse
Affiliation(s)
- Kenji Fujiwara
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,JSPS Overseas Research Fellow, Japan Society for the Promotion of Science, Tokyo, Japan
| | - May Tun Saung
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hao Jing
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Brian Herbst
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - MacKenzie Zarecki
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Stephen Muth
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Annie Wu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Elaine Bigelow
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Linda Chen
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Hepato-Bilio-Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Keyu Li
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Hepato-Bilio-Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Neolle Jurcak
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alex B Blair
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ding Ding
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | | - Jordan Blum
- Bristol Myers Squibb Co, Princeton, New Jersey, USA
| | | | | | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States .,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
53
|
Jagodinsky JC, Morris ZS. Priming and Propagating Anti-tumor Immunity: Focal Hypofractionated Radiation for in Situ Vaccination and Systemic Targeted Radionuclide Theranostics for Immunomodulation of Tumor Microenvironments. Semin Radiat Oncol 2021; 30:181-186. [PMID: 32381297 DOI: 10.1016/j.semradonc.2019.12.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent preclinical and clinical studies have elucidated mechanisms whereby radiation therapy influences the anti-tumor immune response. Immunogenic cell death and phenotypic changes in tumor cells surviving radiation may underlie this effect and contribute to the capacity of radiation to elicit an in situ tumor vaccine effect. In situ vaccination is a therapeutic strategy that seeks to convert a patient's own tumor into a source of enhanced antigen recognition for the purpose of augmenting a systemic anti-tumor immune response. Capitalizing on the in situ vaccine effect of radiation, several groups have demonstrated anti-tumor efficacy in preclinical models by combining radiation with immune checkpoint blockade. Local delivery of immune adjuvants and/or immune stimulatory cytokines via direct injection into the radiated tumor microenvironment may further increase the in situ vaccine capacity of radiation therapy. However, recent studies suggest that in some contexts this effect is antagonized by the presence of distant untreated sites of disease that may dampen the systemic immune response generated by in situ vaccination through a phenomenon termed concomitant immune tolerance. Concomitant immune tolerance may be overcome by delivering radiation to all sites of metastatic disease, however this is often not possible to safely achieve using external beam radiation therapy without considerable risk of lymphopenia that would negate the immune effects of in situ vaccination. For patients with widespread metastatic disease, alternative strategies may include systemic treatment with targeted radionuclide therapies alone or in combination with an external beam radiation therapy-based in situ vaccine approach.
Collapse
Affiliation(s)
- Justin C Jagodinsky
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| |
Collapse
|
54
|
Asperud J, Arous D, Edin NFJ, Malinen E. Spatially fractionated radiotherapy: tumor response modelling including immunomodulation. Phys Med Biol 2021; 66. [PMID: 34298527 DOI: 10.1088/1361-6560/ac176b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/23/2021] [Indexed: 01/20/2023]
Abstract
A mathematical tumor response model has been developed, encompassing the interplay between immune cells and cancer cells initiated by either partial or full tumor irradiation. The iterative four-compartment model employs the linear-quadratic radiation response theory for four cell types: active and inactive cytotoxic T lymphocytes (immune cells, CD8+T cells in particular), viable cancer cells (undamaged and reparable cells) and doomed cells (irreparably damaged cells). The cell compartment interactions are calculated per day, with total tumor volume (TV) as the main quantity of interest. The model was fitted to previously published data on syngeneic xenografts (67NR breast carcinoma and Lewis lung carcinoma; (Markovskyet al2019Int. J. Radiat. Oncol. Biol. Phys.103697-708)) subjected to single doses of 10 or 15 Gy by 50% (partial) or 100% (full) TV irradiation. The experimental data included effects from anti-CD8+antibodies and immunosuppressive drugs. Using a new optimization method, promising fits were obtained where the lowest and highest root-mean-squared error values were observed for anti-CD8+treatment and unirradiated control data, respectively, for both cell types. Additionally, predictive capabilities of the model were tested by using the estimated model parameters to predict scenarios for higher doses and different TV irradiation fractions. Here, mean relative deviations in the range of 19%-34% from experimental data were found. However, more validation data is needed to conclude on the model's predictive capabilities. In conclusion, the model was found useful in evaluating the impact from partial and full TV irradiation on the immune response and subsequent tumor growth. The model shows potential to support and guide spatially fractionated radiotherapy in future pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Jonas Asperud
- Department of Physics, University of Oslo, PO Box 1048 Blindern, N-0316 Oslo, Norway
| | - Delmon Arous
- Department of Physics, University of Oslo, PO Box 1048 Blindern, N-0316 Oslo, Norway.,Department of Medical Physics, The Norwegian Radium Hospital, Oslo University Hospital, PO Box 4953 Nydalen, N-0424 Oslo, Norway
| | | | - Eirik Malinen
- Department of Physics, University of Oslo, PO Box 1048 Blindern, N-0316 Oslo, Norway.,Department of Medical Physics, The Norwegian Radium Hospital, Oslo University Hospital, PO Box 4953 Nydalen, N-0424 Oslo, Norway
| |
Collapse
|
55
|
Hijioka S, Morizane C, Ikeda M, Ishii H, Okusaka T, Furuse J. Current status of medical treatment for gastroenteropancreatic neuroendocrine neoplasms and future perspectives. Jpn J Clin Oncol 2021; 51:1185-1196. [PMID: 34038547 PMCID: PMC8326384 DOI: 10.1093/jjco/hyab076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/03/2021] [Indexed: 12/16/2022] Open
Abstract
Neuroendocrine neoplasms (NENs) constitute a heterogeneous group of tumors. In this review, we summarize the results of various clinical trials that have been conducted to investigate the efficacy and safety of various therapeutic options for NENs. Based on the encouraging results obtained from these trials, various therapeutic options have been established for the treatment of NENs, including somatostatin analogs (SSAs), molecularly targeted drugs and cytotoxic agents. In addition, peptide receptor radionucleotide therapy has recently been evaluated for the treatment of various NENs. We also discuss the approach for selecting the appropriate drugs and sequence of treatment with the various drug classes, as recommended by different treatment guidelines. Finally, we discuss the scope for future research in this field, especially into the merits of combination therapy with molecularly targeted drugs plus SSAs, along with ongoing studies.
Collapse
Affiliation(s)
- Susumu Hijioka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroshi Ishii
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
56
|
Trappetti V, Fazzari JM, Fernandez-Palomo C, Scheidegger M, Volarevic V, Martin OA, Djonov VG. Microbeam Radiotherapy-A Novel Therapeutic Approach to Overcome Radioresistance and Enhance Anti-Tumour Response in Melanoma. Int J Mol Sci 2021; 22:7755. [PMID: 34299373 PMCID: PMC8303317 DOI: 10.3390/ijms22147755] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 12/19/2022] Open
Abstract
Melanoma is the deadliest type of skin cancer, due to its invasiveness and limited treatment efficacy. The main therapy for primary melanoma and solitary organ metastases is wide excision. Adjuvant therapy, such as chemotherapy and targeted therapies are mainly used for disseminated disease. Radiotherapy (RT) is a powerful treatment option used in more than 50% of cancer patients, however, conventional RT alone is unable to eradicate melanoma. Its general radioresistance is attributed to overexpression of repair genes in combination with cascades of biochemical repair mechanisms. A novel sophisticated technique based on synchrotron-generated, spatially fractionated RT, called Microbeam Radiation Therapy (MRT), has been shown to overcome these treatment limitations by allowing increased dose delivery. With MRT, a collimator subdivides the homogeneous radiation field into an array of co-planar, high-dose microbeams that are tens of micrometres wide and spaced a few hundred micrometres apart. Different preclinical models demonstrated that MRT has the potential to completely ablate tumours, or significantly improve tumour control while dramatically reducing normal tissue toxicity. Here, we discuss the role of conventional RT-induced immunity and the potential for MRT to enhance local and systemic anti-tumour immune responses. Comparative gene expression analysis from preclinical tumour models indicated a specific gene signature for an 'MRT-induced immune effect'. This focused review highlights the potential of MRT to overcome the inherent radioresistance of melanoma which could be further enhanced for future clinical use with combined treatment strategies, in particular, immunotherapy.
Collapse
Affiliation(s)
- Verdiana Trappetti
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (V.T.); (J.M.F.); (C.F.-P.); (M.S.); (O.A.M.)
| | - Jennifer M. Fazzari
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (V.T.); (J.M.F.); (C.F.-P.); (M.S.); (O.A.M.)
| | - Cristian Fernandez-Palomo
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (V.T.); (J.M.F.); (C.F.-P.); (M.S.); (O.A.M.)
| | - Maximilian Scheidegger
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (V.T.); (J.M.F.); (C.F.-P.); (M.S.); (O.A.M.)
| | - Vladislav Volarevic
- Department of Genetics, Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Olga A. Martin
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (V.T.); (J.M.F.); (C.F.-P.); (M.S.); (O.A.M.)
- Peter MacCallum Cancer Centre, Division of Radiation Oncology, Melbourne, VIC 3000, Australia
- University of Melbourne, Parkville, VIC 3010, Australia
| | - Valentin G. Djonov
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (V.T.); (J.M.F.); (C.F.-P.); (M.S.); (O.A.M.)
| |
Collapse
|
57
|
Current Prospects for Treatment of Solid Tumors via Photodynamic, Photothermal, or Ionizing Radiation Therapies Combined with Immune Checkpoint Inhibition (A Review). Pharmaceuticals (Basel) 2021; 14:ph14050447. [PMID: 34068491 PMCID: PMC8151935 DOI: 10.3390/ph14050447] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/21/2022] Open
Abstract
Photodynamic therapy (PDT) causes selective damage to tumor cells and vasculature and also triggers an anti-tumor immune response. The latter fact has prompted the exploration of PDT as an immune-stimulatory adjuvant. PDT is not the only cancer treatment that relies on electromagnetic energy to destroy cancer tissue. Ionizing radiation therapy (RT) and photothermal therapy (PTT) are two other treatment modalities that employ photons (with wavelengths either shorter or longer than PDT, respectively) and also cause tissue damage and immunomodulation. Research on the three modalities has occurred in different “silos”, with minimal interaction between the three topics. This is happening at a time when immune checkpoint inhibition (ICI), another focus of intense research and clinical development, has opened exciting possibilities for combining PDT, PTT, or RT with ICI to achieve improved therapeutic benefits. In this review, we surveyed the literature for studies that describe changes in anti-tumor immunity following the administration of PDT, PTT, and RT, including efforts to combine each modality with ICI. This information, collected all in one place, may make it easier to recognize similarities and differences and help to identify new mechanistic hypotheses toward the goal of achieving optimized combinations and tumor cures.
Collapse
|
58
|
Ukleja J, Kusaka E, Miyamoto DT. Immunotherapy Combined With Radiation Therapy for Genitourinary Malignancies. Front Oncol 2021; 11:663852. [PMID: 34041029 PMCID: PMC8141854 DOI: 10.3389/fonc.2021.663852] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy drugs have recently been approved by the Food and Drug Administration for the treatment of several genitourinary malignancies, including bladder cancer, renal cancer, and prostate cancer. Preclinical data and early clinical trial results suggest that immune checkpoint inhibitors can act synergistically with radiation therapy to enhance tumor cell killing at local irradiated sites and in some cases at distant sites through an abscopal effect. Because radiation therapy is commonly used in the treatment of genitourinary malignancies, there is great interest in testing the combination of immunotherapy with radiation therapy in these cancers to further improve treatment efficacy. In this review, we discuss the current evidence and biological rationale for combining immunotherapy with radiation therapy, as well as emerging data from ongoing and planned clinical trials testing the efficacy and tolerability of this combination in the treatment of genitourinary malignancies. We also outline outstanding questions regarding sequencing, dose fractionation, and biomarkers that remain to be addressed for the optimal delivery of this promising treatment approach.
Collapse
Affiliation(s)
- Jacob Ukleja
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Erika Kusaka
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - David T. Miyamoto
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Massachusetts General Hospital Cancer Center, Charlestown, MA, United States
| |
Collapse
|
59
|
Gallage S, García-Beccaria M, Szydlowska M, Rahbari M, Mohr R, Tacke F, Heikenwalder M. The therapeutic landscape of hepatocellular carcinoma. MED 2021; 2:505-552. [PMID: 35590232 DOI: 10.1016/j.medj.2021.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/23/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023]
|
60
|
Jagodinsky JC, Jin WJ, Bates AM, Hernandez R, Grudzinski JJ, Marsh IR, Chakravarty I, Arthur IS, Zangl LM, Brown RJ, Nystuen EJ, Emma SE, Kerr C, Carlson PM, Sriramaneni RN, Engle JW, Aluicio-Sarduy E, Barnhart TE, Le T, Kim K, Bednarz BP, Weichert JP, Patel RB, Morris ZS. Temporal analysis of type 1 interferon activation in tumor cells following external beam radiotherapy or targeted radionuclide therapy. Theranostics 2021; 11:6120-6137. [PMID: 33995649 PMCID: PMC8120207 DOI: 10.7150/thno.54881] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/26/2021] [Indexed: 12/15/2022] Open
Abstract
Rationale: Clinical interest in combining targeted radionuclide therapies (TRT) with immunotherapies is growing. External beam radiation therapy (EBRT) activates a type 1 interferon (IFN1) response mediated via stimulator of interferon genes (STING), and this is critical to its therapeutic interaction with immune checkpoint blockade. However, little is known about the time course of IFN1 activation after EBRT or whether this may be induced by decay of a TRT source. Methods: We examined the IFN1 response and expression of immune susceptibility markers in B78 and B16 melanomas and MOC2 head and neck cancer murine models using qPCR and western blot. For TRT, we used 90Y chelated to NM600, an alkylphosphocholine analog that exhibits selective uptake and retention in tumor cells including B78 and MOC2. Results: We observed significant IFN1 activation in all cell lines, with peak activation in B78, B16, and MOC2 cell lines occurring 7, 7, and 1 days, respectively, following RT for all doses. This effect was STING-dependent. Select IFN response genes remained upregulated at 14 days following RT. IFN1 activation following STING agonist treatment in vitro was identical to RT suggesting time course differences between cell lines were mediated by STING pathway kinetics and not DNA damage susceptibility. In vivo delivery of EBRT and TRT to B78 and MOC2 tumors resulted in a comparable time course and magnitude of IFN1 activation. In the MOC2 model, the combination of 90Y-NM600 and dual checkpoint blockade therapy reduced tumor growth and prolonged survival compared to single agent therapy and cumulative dose equivalent combination EBRT and dual checkpoint blockade therapy. Conclusions: We report the time course of the STING-dependent IFN1 response following radiation in multiple murine tumor models. We show the potential of TRT to stimulate IFN1 activation that is comparable to that observed with EBRT and this may be critical to the therapeutic integration of TRT with immunotherapies.
Collapse
MESH Headings
- Animals
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/physiopathology
- Carcinoma, Squamous Cell/radiotherapy
- Cell Line, Tumor
- Combined Modality Therapy
- Dose-Response Relationship, Radiation
- Female
- Gene Expression Regulation, Neoplastic/radiation effects
- Gene Knockout Techniques
- Head and Neck Neoplasms/pathology
- Immune Checkpoint Inhibitors
- Interferon Type I/biosynthesis
- Interferon Type I/genetics
- Interferon Type I/physiology
- Lymphocytes/drug effects
- Lymphocytes/radiation effects
- Melanoma, Experimental/immunology
- Melanoma, Experimental/physiopathology
- Melanoma, Experimental/radiotherapy
- Membrane Proteins/agonists
- Membrane Proteins/deficiency
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Mice
- Mice, Inbred C57BL
- Neoplasm Proteins/agonists
- Neoplasm Proteins/physiology
- Radiopharmaceuticals/pharmacokinetics
- Radiopharmaceuticals/therapeutic use
- Time Factors
- Tumor Protein, Translationally-Controlled 1
- Tumor Stem Cell Assay
- Up-Regulation
- Yttrium Radioisotopes/pharmacokinetics
- Yttrium Radioisotopes/therapeutic use
Collapse
Affiliation(s)
- Justin C. Jagodinsky
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Won Jong Jin
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Amber M. Bates
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Reinier Hernandez
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Joseph J. Grudzinski
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Ian R. Marsh
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Ishan Chakravarty
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Ian S. Arthur
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Luke M. Zangl
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Ryan J. Brown
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Erin J. Nystuen
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Sarah E. Emma
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Caroline Kerr
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Peter M. Carlson
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Raghava N. Sriramaneni
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Jonathan W. Engle
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Eduardo Aluicio-Sarduy
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Todd E. Barnhart
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Trang Le
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Bryan P. Bednarz
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Jamey P. Weichert
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Ravi B. Patel
- Department of Radiation Oncology, University of Pittsburgh School Hillman Cancer Center, Pittsburgh, PA
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
61
|
Jagodinsky JC, Medeiros G, Raj HH, Razuan A, Locsin A, Dempsey TG, Tang B, Chakravarty I, Clark PA, Sriramaneni RN, Jin WJ, Lan KH, Das RK, Miller JR, Suarez-Gonzalez D, Morris ZS. A multipurpose brachytherapy catheter to enable intratumoral injection. Brachytherapy 2021; 20:900-910. [PMID: 33785280 DOI: 10.1016/j.brachy.2020.10.012] [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: 06/16/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 11/30/2022]
Abstract
PURPOSE To create and test a multipurpose brachytherapy catheter prototype enabling intratumoral injection and brachytherapy after a single catheter insertion. METHODS AND MATERIALS The design of the prototype consists of an outer tube and an inner syringe tube that can be filled with injectable agent. The outer sheath and inner syringe tube were constructed using polytetrafluoroethylene tubing, and the other components were 3D printed using dental resin and polylactic acid material. To demonstrate functionality, we injected in vitro phantoms with dyed saline. For proof of concept, we demonstrated the potential for the prototype to deliver cell therapy, enhance tumor delineation, deliver tattoo ink for pathology marking, avoid toxicity through local delivery of chemotherapy, and facilitate combination brachytherapy and immunotherapy. RESULTS The prototype enables accurate injection in vitro and in vivo without altering dosimetry. To illustrate the potential for delivery of cell therapies, we injected luciferase-expressing splenocytes and confirmed their delivery with bioluminescence imaging. To demonstrate feasibility of radiographically visualizing injected material, we delivered iohexol contrast intratumorally and confirmed tumor retention using Faxitron x-ray imaging. In addition, we show the potential of intratumoral administration to reduce toxicity associated with cyclophosphamide compared with systemic administration. To demonstrate feasibility, we treated tumor-bearing mice with brachytherapy (192Ir source, 2 Gy to 5 mm) in combination with intratumoral injection of 375,000 U of interleukin 2 and observed no increased toxicity. CONCLUSIONS These results demonstrate that a prototype multipurpose brachytherapy catheter enables accurate intratumoral injection and support the feasibility of combining intratumoral injection with brachytherapy.
Collapse
Affiliation(s)
- Justin C Jagodinsky
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| | - Gabriella Medeiros
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI
| | - Hayley H Raj
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI
| | - Amira Razuan
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI
| | - Alexis Locsin
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI
| | - Tirhas G Dempsey
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI
| | - Beixiao Tang
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI
| | - Ishan Chakravarty
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Paul A Clark
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Raghava N Sriramaneni
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Won Jong Jin
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Keng-Hsueh Lan
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Rupak K Das
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Jessica R Miller
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Darilis Suarez-Gonzalez
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
62
|
Patient-Specific Lymphocyte Loss Kinetics as Biomarker of Spleen Dose in Patients Undergoing Radiation Therapy for Upper Abdominal Malignancies. Adv Radiat Oncol 2021; 6:100545. [PMID: 33665481 PMCID: PMC7897770 DOI: 10.1016/j.adro.2020.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/26/2020] [Accepted: 08/03/2020] [Indexed: 12/24/2022] Open
Abstract
Purpose Radiation therapy (RT)-induced lymphopenia (RIL) is linked with inferior survival in esophageal and pancreatic cancers. Previous work has demonstrated a correlation between spleen dose and RIL risk. The present study correlates spleen dose-volume parameters with fractional lymphocyte loss rate (FLL) and total percent change in absolute lymphocyte count (%ΔALC) and suggests spleen dose constraints to reduce RIL risk. Methods and Materials This registry-based study included 140 patients who underwent RT for pancreatic (n = 67), gastroesophageal (n = 61), or biliary tract (n = 12) adenocarcinoma. Patient-specific parameters of lymphocyte loss kinetics, including FLL and %ΔALC, were calculated based on serial ALCs obtained during RT. Spearman's rho was used to correlate spleen dose-volume parameters with %ΔALC, end-treatment ALC, and FLL. Multivariable logistic regression was used to identify predictors of ≥grade 3 and grade 4 RIL. Results Spleen dose-volume parameters, including mean spleen dose (MSD), all correlated with %ΔALC, end-treatment ALC, and FLL. Controlling for baseline ALC and planning target volume (PTV), an increase in any spleen dose-volume parameter increased the odds of developing ≥grade 3 lymphopenia. Each 1-Gy increase in MSD increased the odds of ≥grade 3 RIL by 18.6%, and each 100-cm3 increase in PTV increased the odds of ≥grade 3 lymphopenia by 20%. Patients with baseline ALC < 1500 cells/μL had a high risk of ≥grade 3 RIL regardless of MSD or PTV. FLL was an equally good predictor of ≥grade 3 lymphopenia as any spleen dose-volume parameter. Conclusions In patients undergoing RT for upper abdominal malignancies, higher spleen dose is associated with higher per-fraction lymphocyte loss rates, higher total %ΔALC, and increased odds of severe lymphopenia. Spleen dose constraints should be individualized based on baseline ALC and PTV size to minimize RIL risk, although our findings require validation in larger, ideally prospective data sets.
Collapse
|
63
|
Yonezawa N, Murakami H, Demura S, Kato S, Miwa S, Yoshioka K, Shinmura K, Yokogawa N, Shimizu T, Oku N, Kitagawa R, Handa M, Annen R, Kurokawa Y, Fushimi K, Mizukoshi E, Tsuchiya H. Abscopal Effect of Frozen Autograft Reconstruction Combined with an Immune Checkpoint Inhibitor Analyzed Using a Metastatic Bone Tumor Model. Int J Mol Sci 2021; 22:1973. [PMID: 33671258 PMCID: PMC7922593 DOI: 10.3390/ijms22041973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/08/2021] [Accepted: 02/14/2021] [Indexed: 01/10/2023] Open
Abstract
We evaluated the abscopal effect of re-implantation of liquid nitrogen-treated tumor-bearing bone grafts and the synergistic effect of anti-PD-1 (programmed death-1) therapy using a bone metastasis model, created by injecting MMT-060562 cells into the bilateral tibiae of 6-8-week-old female C3H mice. After 2 weeks, the lateral tumors were treated by excision, cryotreatment using liquid nitrogen, excision with anti-PD-1 treatment, and cryotreatment with anti-PD-1 treatment. Anti-mouse PD-1 4H2 was injected on days 1, 6, 12, and 18 post-treatment. The mice were euthanized after 3 weeks; the abscopal effect was evaluated by focusing on growth inhibition of the abscopal tumor. The re-implantation of frozen autografts significantly inhibited the growth of the remaining abscopal tumors. However, a more potent abscopal effect was observed in the anti-PD-1 antibody group. The number of CD8+ T cells infiltrating the abscopal tumor and tumor-specific interferon-γ (IFN-γ)-producing spleen cells increased in the liquid nitrogen-treated group compared with those in the excision group, with no significant difference. The number was significantly higher in the anti-PD-1 antibody-treated group than in the non-treated group. Overall, re-implantation of tumor-bearing frozen autograft has an abscopal effect on abscopal tumor growth, although re-implantation of liquid nitrogen-treated bone grafts did not induce a strong T-cell response or tumor-suppressive effect.
Collapse
Affiliation(s)
- Noritaka Yonezawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Hideki Murakami
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan;
| | - Satoru Demura
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Satoshi Kato
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Shinji Miwa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Katsuhito Yoshioka
- Department of Orthopaedic Surgery, National Hospital Organization Kanazawa Medical Center, Kanazawa, Ishikawa 920-8650, Japan;
| | - Kazuya Shinmura
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Noriaki Yokogawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Takaki Shimizu
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Norihiro Oku
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Ryo Kitagawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Makoto Handa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Ryohei Annen
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Yuki Kurokawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Kazumi Fushimi
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa, Ishikawa 920-8641, Japan; (K.F.); (E.M.)
| | - Eishiro Mizukoshi
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa, Ishikawa 920-8641, Japan; (K.F.); (E.M.)
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| |
Collapse
|
64
|
Cuzzubbo S, Mangsbo S, Nagarajan D, Habra K, Pockley AG, McArdle SEB. Cancer Vaccines: Adjuvant Potency, Importance of Age, Lifestyle, and Treatments. Front Immunol 2021; 11:615240. [PMID: 33679703 PMCID: PMC7927599 DOI: 10.3389/fimmu.2020.615240] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
Although the discovery and characterization of multiple tumor antigens have sparked the development of many antigen/derived cancer vaccines, many are poorly immunogenic and thus, lack clinical efficacy. Adjuvants are therefore incorporated into vaccine formulations to trigger strong and long-lasting immune responses. Adjuvants have generally been classified into two categories: those that ‘depot’ antigens (e.g. mineral salts such as aluminum hydroxide, emulsions, liposomes) and those that act as immunostimulants (Toll Like Receptor agonists, saponins, cytokines). In addition, several novel technologies using vector-based delivery of antigens have been used. Unfortunately, the immune system declines with age, a phenomenon known as immunosenescence, and this is characterized by functional changes in both innate and adaptive cellular immunity systems as well as in lymph node architecture. While many of the immune functions decline over time, others paradoxically increase. Indeed, aging is known to be associated with a low level of chronic inflammation—inflamm-aging. Given that the median age of cancer diagnosis is 66 years and that immunotherapeutic interventions such as cancer vaccines are currently given in combination with or after other forms of treatments which themselves have immune-modulating potential such as surgery, chemotherapy and radiotherapy, the choice of adjuvants requires careful consideration in order to achieve the maximum immune response in a compromised environment. In addition, more clinical trials need to be performed to carefully assess how less conventional form of immune adjuvants, such as exercise, diet and psychological care which have all be shown to influence immune responses can be incorporated to improve the efficacy of cancer vaccines. In this review, adjuvants will be discussed with respect to the above-mentioned important elements.
Collapse
Affiliation(s)
- Stefania Cuzzubbo
- Université de Paris, PARCC, INSERM U970, 75015, Paris, France.,Laboratoire de Recherches Biochirurgicales (Fondation Carpentier), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France
| | - Sara Mangsbo
- Ultimovacs AB, Uppsala, Sweden.,Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Divya Nagarajan
- Department of Immunology, Genetics and Clinical pathology Rudbeck laboratories, Uppsala University, Uppsala, Sweden
| | - Kinana Habra
- The School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Alan Graham Pockley
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Stephanie E B McArdle
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
65
|
The Potentiation of Anti-Tumor Immunity by Tumor Abolition with Alpha Particles, Protons, or Carbon Ion Radiation and Its Enforcement by Combination with Immunoadjuvants or Inhibitors of Immune Suppressor Cells and Checkpoint Molecules. Cells 2021; 10:cells10020228. [PMID: 33503958 PMCID: PMC7912488 DOI: 10.3390/cells10020228] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/24/2022] Open
Abstract
The delivery of radiation therapy (RT) for cancer with intent to cure has been optimized and standardized over the last 80 years. Both preclinical and clinical work emphasized the observation that radiation destroys the tumor and exposes its components to the immune response in a mode that facilitates the induction of anti-tumor immunity or reinforces such a response. External beam photon radiation is the most prevalent in situ abolition treatment, and its use exposed the “abscopal effect”. Particle radiotherapy (PRT), which has been in various stages of research and development for 70 years, is today available for the treatment of patients in the form of alpha particles, proton, or carbon ion radiotherapy. Charged particle radiotherapy is based on the acceleration of charged species, such as protons or carbon-12, which deposit their energy in the treated tumor and have a higher relative biological effectiveness compared with photon radiation. In this review, we will bring evidence that alpha particles, proton, or carbon ion radiation can destroy tumors and activate specific anti-tumor immune responses. Radiation may also directly affect the distribution and function of immune cells such as T cells, regulatory T cells, and mononuclear phagocytes. Tumor abolition by radiation can trigger an immune response against the tumor. However, abolition alone rarely induces effective anti-tumor immunity resulting in systemic tumor rejection. Immunotherapy can complement abolition to reinforce the anti-tumor immunity to better eradicate residual local and metastatic tumor cells. Various methods and agents such as immunoadjuvants, suppressor cell inhibitors, or checkpoint inhibitors were used to manipulate the immune response in combination with radiation. This review deals with the manifestations of particle-mediated radiotherapy and its correlation with immunotherapy of cancer.
Collapse
|
66
|
Abstract
Radiopharmaceutical therapy or targeted radionuclide therapy (TRT) is a well-established class of cancer therapeutics that includes a growing number of FDA-approved drugs and a promising pipeline of experimental therapeutics. Radiobiology is fundamental to a mechanistic understanding of the therapeutic capacity of these agents and their potential toxicities. However, the field of radiobiology has historically focused on external beam radiation. Critical differences exist between TRT and external beam radiotherapy with respect to dosimetry, dose rate, linear energy transfer, duration of treatment delivery, fractionation, range, and target volume. These distinctions simultaneously make it difficult to extrapolate from the radiobiology of external beam radiation to that of TRT and pose considerable challenges for preclinical and clinical studies investigating TRT. Here, we discuss these challenges and explore the current understanding of the radiobiology of radiopharmaceuticals.
Collapse
Affiliation(s)
- Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Andrew Z Wang
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC
| | - Susan J Knox
- Department of Radiation Oncology, Stanford University, Palo Alto, CA.
| |
Collapse
|
67
|
Wisdom AJ, Mowery YM, Hong CS, Himes JE, Nabet BY, Qin X, Zhang D, Chen L, Fradin H, Patel R, Bassil AM, Muise ES, King DA, Xu ES, Carpenter DJ, Kent CL, Smythe KS, Williams NT, Luo L, Ma Y, Alizadeh AA, Owzar K, Diehn M, Bradley T, Kirsch DG. Single cell analysis reveals distinct immune landscapes in transplant and primary sarcomas that determine response or resistance to immunotherapy. Nat Commun 2020; 11:6410. [PMID: 33335088 PMCID: PMC7746723 DOI: 10.1038/s41467-020-19917-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy fails to cure most cancer patients. Preclinical studies indicate that radiotherapy synergizes with immunotherapy, promoting radiation-induced antitumor immunity. Most preclinical immunotherapy studies utilize transplant tumor models, which overestimate patient responses. Here, we show that transplant sarcomas are cured by PD-1 blockade and radiotherapy, but identical treatment fails in autochthonous sarcomas, which demonstrate immunoediting, decreased neoantigen expression, and tumor-specific immune tolerance. We characterize tumor-infiltrating immune cells from transplant and primary tumors, revealing striking differences in their immune landscapes. Although radiotherapy remodels myeloid cells in both models, only transplant tumors are enriched for activated CD8+ T cells. The immune microenvironment of primary murine sarcomas resembles most human sarcomas, while transplant sarcomas resemble the most inflamed human sarcomas. These results identify distinct microenvironments in murine sarcomas that coevolve with the immune system and suggest that patients with a sarcoma immune phenotype similar to transplant tumors may benefit most from PD-1 blockade and radiotherapy.
Collapse
Affiliation(s)
- Amy J Wisdom
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Yvonne M Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA.
- Duke Cancer Institute, Durham, NC, 27708, USA.
| | - Cierra S Hong
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Jonathon E Himes
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Barzin Y Nabet
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Department of Oncology Biomarker Development, Genentech, South San Francisco, CA, 94080, USA
| | - Xiaodi Qin
- Duke Cancer Institute, Durham, NC, 27708, USA
| | | | - Lan Chen
- Merck & Co., Inc, Kenilworth, NJ, 07033, USA
| | - Hélène Fradin
- Duke Center for Genomic and Computational Biology, Durham, NC, 27708, USA
| | - Rutulkumar Patel
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Alex M Bassil
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | | | - Daniel A King
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Eric S Xu
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - David J Carpenter
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Collin L Kent
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | | | - Nerissa T Williams
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Kouros Owzar
- Duke Cancer Institute, Durham, NC, 27708, USA
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, 27710, USA
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, 94305, USA
| | - Todd Bradley
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Pediatric Genomic Medicine, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - David G Kirsch
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA.
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA.
- Duke Cancer Institute, Durham, NC, 27708, USA.
| |
Collapse
|
68
|
Lin Y, Lu R, Hou J, Zhou GG, Fu W. IFNgamma-inducible CXCL10/CXCR3 axis alters the sensitivity of HEp-2 cells to ionizing radiation. Exp Cell Res 2020; 398:112382. [PMID: 33253709 DOI: 10.1016/j.yexcr.2020.112382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 01/06/2023]
Abstract
Radiotherapy is a conventional approach for anti-cancer treatment, killing tumor cells through damaging cellular DNA. While increasing studies have demonstrated that tumors generated the tolerance to radiation and tumor immune system was found to be correlated to radiotherapy resistance. Therefore, it is critical to identify potential immune factors associated with the efficacy of radiotherapy. Here in this study, we evaluated the sensitivities of different tumor cells to radiation and determined HEp-2 cells as the radio-resistant tumor cells for further investigation. IFNgamma as a key regulator of host immune response showed the potential to sensitize tumors to ionizing radiation (IR). Besides, IFNgamma-induced CXC chemokine ligand 10 (CXCL10) was found to be necessary for effective IR-induced killing of cultured HEp-2 cells. Increased clonogenic survival was observed in CXCL10-depleted HEp-2 cells and CXCL10-KO cells. Additionally, the loss of CXCL10 in HEp-2 cells showed less progression of the G0/G1 phase to G2/M when exposed to IR (8 Gy). Local IR (20 Gy) to nude mice bearing HEp-2 tumors significantly reduced tumor burden, while fewer effects on tumor burden in mice carrying CXCL10-KO tumors were observed. We furtherly evaluated the possible roles the chemokine receptor CXCR3 plays in mediating the sensitivity of cultured HEp-2 cells to IR. Altered expression of CXCR3 in HEp-2 cells affected IR-induced killing of HEp-2 cells. Our data suggest the IFNgamma-activated CXCL10/CXCR3 axis may contribute to the effective radiation-induced killing of HEp-2 cells in vitro.
Collapse
Affiliation(s)
- Yunting Lin
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Ruitao Lu
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Jingmei Hou
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Grace Guoying Zhou
- Shenzhen International Institute for Biomedical Research, 1301 Guanguang Rd. 3F Building 1-B, Silver Star Hi-tech Park Longhua District, Shenzhen, Guangdong, 518116, China.
| | - Wenmin Fu
- Shenzhen International Institute for Biomedical Research, 1301 Guanguang Rd. 3F Building 1-B, Silver Star Hi-tech Park Longhua District, Shenzhen, Guangdong, 518116, China.
| |
Collapse
|
69
|
Schaue D, McBride WH. Flying by the seat of our pants: is low dose radiation therapy for COVID-19 an option? Int J Radiat Biol 2020; 96:1219-1223. [PMID: 32401694 PMCID: PMC7725653 DOI: 10.1080/09553002.2020.1767314] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 02/03/2023]
Affiliation(s)
- Dörthe Schaue
- Department of Radiation Oncology, University of California at Los Angeles (UCLA), Los Angeles, CA, USA
| | - William H McBride
- Department of Radiation Oncology, University of California at Los Angeles (UCLA), Los Angeles, CA, USA
| |
Collapse
|
70
|
Dyer BA, Feng CH, Eskander R, Sharabi AB, Mell LK, McHale M, Mayadev JS. Current Status of Clinical Trials for Cervical and Uterine Cancer Using Immunotherapy Combined With Radiation. Int J Radiat Oncol Biol Phys 2020; 109:396-412. [PMID: 32942005 DOI: 10.1016/j.ijrobp.2020.09.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 01/05/2023]
Abstract
Novel therapies combined with radiation continue to be of significant interest in the developmental treatment paradigm of gynecologic cancers. Clinical implementation of immunotherapy in oncology has rapidly changed the treatment landscape, options, paradigm, and outcomes through clinical trials. Immunotherapy has emerged as a therapeutic pillar in the treatment of solid tumors with demonstrable synergistic activity when combined with radiation therapy and chemoradiotherapy by an alteration or enhancement of the immune system. In solid tumors, radiation therapy induces migration of dendritic cells, T cell activation, and proliferation, and increases in tumor-infiltrating lymphocytes. These immunomodulatory effects in conjunction with immune checkpoint blockade are currently under active investigation in the adjuvant, definitive, and metastatic settings. Results from early phase trials demonstrate promising efficacy and overall tolerable toxicity profiles of combined modality treatment. There is significant interest in optimizing the treatment for patients with locally advanced cervical cancer beyond the standard of care-chemoradiation-which has been in place for the last 30 years. The majority of cervical cancer emerges after persistent infection with a high-risk subtype of the human papillomavirus, where viral oncoproteins lead to cellular changes and immortalization. As a result, immune tolerance can develop, resulting in cancer. Knowledge of the mechanism of human papillomavirus-related oncogenesis suggests that immune therapy or checkpoint blockade can reinvigorate an antitumor immune response. Current clinical trials are exploring the therapeutic potential of these approaches. Uterine cancers have been grouped into 4 molecular subclasses by their driver mutations, mutational burden, and copy-number alterations. Of these subgroups, the polymerase epsilon-mutated and microsatellite-unstable may represent up to 40% of endometrial cancers, and they have been shown to be immunogenic. Because of the inherent immunogenicity of these MSI-high tumors, combined immune modulation strategies, including chemotherapy, radiation, and immunotherapy and immune checkpoint inhibitor therapy, are being explored to improve treatment outcomes. In this review, we explore current immunomodulatory and multimodality therapeutic approaches in the treatment of cervical and uterine cancer through ongoing clinical trials investigating the combination of immunotherapy and radiation therapy.
Collapse
Affiliation(s)
- Brandon A Dyer
- Department of Radiation Oncology, University of Washington Medical Center, Seattle, Washington
| | - Christine H Feng
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| | - Ramez Eskander
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Services, University of California San Diego, La Jolla, California
| | - Andrew B Sharabi
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| | - Loren K Mell
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| | - Michael McHale
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Services, University of California San Diego, La Jolla, California
| | - Jyoti S Mayadev
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California.
| |
Collapse
|
71
|
Shamseddine A, Zeidan YH, Kreidieh M, Khalifeh I, Turfa R, Kattan J, Mukherji D, Temraz S, Alqasem K, Amarin R, Al Awabdeh T, Deeba S, Jamali F, Mohamad I, Elkhaldi M, Daoud F, Al Masri M, Dabous A, Hushki A, Jaber O, Khoury C, El Husseini Z, Charafeddine M, Al Darazi M, Geara F. Short-course radiation followed by mFOLFOX-6 plus avelumab for locally-advanced rectal adenocarcinoma. BMC Cancer 2020; 20:831. [PMID: 32873251 PMCID: PMC7466814 DOI: 10.1186/s12885-020-07333-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 08/24/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Current standard practice for locally advanced rectal cancer (LARC) entails a multidisciplinary approach that includes preoperative chemoradiotherapy, followed by total mesorectal excision, and then adjuvant chemotherapy. The latter has been accompanied by low compliance rates and no survival benefit in phase III randomized trials, so the strategy of administering neoadjuvant, rather than adjuvant, chemotherapy has been adapted by many trials, with improvement in pathologic complete response. Induction chemotherapy with oxaliplatin has been shown to have increased efficacy in rectal cancer, while short-course radiation therapy with consolidation chemotherapy increased short-term overall survival rate and decreased toxicity levels, making it cheaper and more convenient than long-course radiation therapy. This led to recognition of total neoadjuvant therapy as a valid treatment approach in many guidelines despite limited available survival data. With the upregulation (PDL-1) expression in rectal tumors after radiotherapy and the increased use of in malignant melanoma, the novel approach of combining immunotherapy with chemotherapy after radiation may have a role in further increasing pCR and improving overall outcomes in rectal cancer. METHODS The study is an open label single arm multi- center phase II trial. Forty-four recruited LARC patients will receive 5Gy x 5fractions of SCRT, followed by 6 cycles of mFOLFOX-6 plus avelumab, before TME is performed. The hypothesis is that the addition of avelumab to mFOLFOX-6, administered following SCRT, will improve pCR and overall outcomes. The primary outcome measure is the proportion of patients who achieve a pCR, defined as no viable tumor cells on the excised specimen. Secondary objectives are to evaluate 3-year progression-free survival, tumor response to treatment (tumor regression grades 0 & 1), density of tumor-infiltrating lymphocytes, correlation of baseline Immunoscore with pCR rates and changes in PD-L1 expression. DISCUSSION Recent studies show an increase in PD-L1 expression and density of CD8+ TILs after CRT in rectal cancer patients, implying a potential role for combinatory strategies using PD-L1- and programmed-death- 1 inhibiting drugs. We aim through this study to evaluate pCR following SCRT, followed by mFOLFOX-6 with avelumab, and then TME procedure in patients with LARC. TRIAL REGISTRATION Trial Registration Number and Date of Registration: ClinicalTrials.gov NCT03503630, April 20, 2018.
Collapse
Affiliation(s)
- Ali Shamseddine
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute- NKBCI, American University of Beirut Medical Center, Beirut, Lebanon.
| | - Youssef H Zeidan
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Malek Kreidieh
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute- NKBCI, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ibrahim Khalifeh
- Department of pathology and laboratory medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rim Turfa
- Department of Medical Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Joseph Kattan
- Department of Medical Oncology, Hôtel Dieu de France, Beirut, Lebanon
| | - Deborah Mukherji
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute- NKBCI, American University of Beirut Medical Center, Beirut, Lebanon
| | - Sally Temraz
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute- NKBCI, American University of Beirut Medical Center, Beirut, Lebanon
| | - Kholoud Alqasem
- Department of Medical Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Rula Amarin
- Department of Medical Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Tala Al Awabdeh
- Department of Medical Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Samer Deeba
- Department of General Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Faek Jamali
- Department of General Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Issa Mohamad
- Department of Medical Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Mousa Elkhaldi
- Department of Medical Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Faiez Daoud
- Department of Surgical Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Mahmoud Al Masri
- Department of Surgical Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Ali Dabous
- Department of Surgical Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Ahmad Hushki
- Gastroenterology Department, King Hussein Cancer Center, Amman, Jordan
| | - Omar Jaber
- Pathology Department, King Hussein Cancer Center, Amman, Jordan
| | - Clement Khoury
- Department of Radiation Oncology, Hotel-Dieu de France Hospital, Beirut, Lebanon
| | - Ziad El Husseini
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute- NKBCI, American University of Beirut Medical Center, Beirut, Lebanon
| | - Maya Charafeddine
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute- NKBCI, American University of Beirut Medical Center, Beirut, Lebanon
| | - Monita Al Darazi
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute- NKBCI, American University of Beirut Medical Center, Beirut, Lebanon
| | - Fady Geara
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
72
|
Jagodinsky JC, Harari PM, Morris ZS. The Promise of Combining Radiation Therapy With Immunotherapy. Int J Radiat Oncol Biol Phys 2020; 108:6-16. [PMID: 32335187 PMCID: PMC7442714 DOI: 10.1016/j.ijrobp.2020.04.023] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/30/2020] [Accepted: 04/13/2020] [Indexed: 12/17/2022]
Abstract
The development of immunotherapy in oncology builds upon many years of scientific investigation into the cellular mechanics underlying interactions between tumor cells and immune cell populations. The past decade has brought an accelerating pace to the clinical investigation of new immunotherapy agents, particularly in the setting of metastatic disease. The integration of immunotherapy into phase 3 clinical trial design has lagged in settings of advanced locoregional disease, where combination with radiation therapy may be critical. Yet, such may be the settings where immunotherapies have their greatest potential to affect patient survival and achieve curative outcomes. In this review, we discuss the interaction of radiation with the immune system and the potential to augment antitumor immunity through combined-modality approaches that integrate radiation and immunotherapies. The dynamics of cellular and tumor response to radiation offer unique opportunities for beneficial interplay with immunotherapy that may go unrecognized with conventional screening and monotherapy clinical testing of novel pharmaceutical agents. Using immune checkpoint blockade as a primary example, we discuss recent preclinical and clinical studies that illustrate the potential synergy of such therapies in combination with radiation, and we highlight the potential clinical value of such interactions. For various immunotherapy agents, their greatest clinical effect may rest in combination with radiation, and efforts to facilitate systematic investigation of this approach are highly warranted.
Collapse
Affiliation(s)
- Justin C Jagodinsky
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
73
|
Dong X, Cheng R, Zhu S, Liu H, Zhou R, Zhang C, Chen K, Mei L, Wang C, Su C, Liu X, Gu Z, Zhao Y. A Heterojunction Structured WO 2.9-WSe 2 Nanoradiosensitizer Increases Local Tumor Ablation and Checkpoint Blockade Immunotherapy upon Low Radiation Dose. ACS NANO 2020; 14:5400-5416. [PMID: 32324373 DOI: 10.1021/acsnano.9b08962] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Radiotherapy (RT) in practical use often suffers from off-target side effects and ineffectiveness against hypoxic tumor microenvironment (TME) as well as remote metastases. With regard to these problems, herein, we provide semiconductor heterojunction structured WO2.9-WSe2-PEG nanoparticles to realize a synergistic RT/photothermal therapy (PTT)/checkpoint blockade immunotherapy (CBT) for enhanced antitumor and antimetastatic effect. Based on the heterojunction structured nanoparticle with high Z element, the nanosystem could realize non-oxygen-dependent reactive oxygen species generation by catalyzing highly expressed H2O2 in TME upon X-ray irradiation, which could further induce immunogenic cell death. Meanwhile, this nanosystem could also induce hyperthermia upon near-infrared irradiation to enhance RT outcome. With the addition of anti-PD-L1 antibody-based CBT, our results give potent evidence that local RT/PTT upon mild temperature and low radiation dose could efficiently ablate local tumors and inhibit tumor metastasis as well as prevent tumor rechallenge. Our study provides not only one kind of radiosensitizer based on semiconductor nanoparticles but also a versatile nanoplatform for simultaneous triple-combined therapy (RT/PTT/CBT) for treating both local and metastasis tumors.
Collapse
Affiliation(s)
- Xinghua Dong
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100190, China
| | - Ran Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
- College of Mechanical and Electronic Engineering, Shandong University of Science and Technology, Qingdao 266590, P.R. China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Huimin Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
- College of Mechanical and Electronic Engineering, Shandong University of Science and Technology, Qingdao 266590, P.R. China
| | - Ruyi Zhou
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Chenyang Zhang
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Kui Chen
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Linqiang Mei
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Chengyan Wang
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Chunjian Su
- College of Mechanical and Electronic Engineering, Shandong University of Science and Technology, Qingdao 266590, P.R. China
| | - Xiangfeng Liu
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhanjun Gu
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Yuliang Zhao
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
74
|
Starr JS, Sonbol MB, Hobday TJ, Sharma A, Kendi AT, Halfdanarson TR. Peptide Receptor Radionuclide Therapy for the Treatment of Pancreatic Neuroendocrine Tumors: Recent Insights. Onco Targets Ther 2020; 13:3545-3555. [PMID: 32431509 PMCID: PMC7205451 DOI: 10.2147/ott.s202867] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/26/2020] [Indexed: 12/27/2022] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) is a paradigm shifting approach to the treatment of neuroendocrine tumors. Although there are no prospective randomized trials directly studying PRRT in pancreatic neuroendocrine tumors (panNETs), there are data to suggest benefit in this patient population. Collectively, the data, consisting of two prospective and six retrospective studies, show a median PFS ranging from 20 to 39 months and a median OS ranging from 37 to 79 months. There are ongoing (and upcoming) prospective, randomized trials of PRRT in panNETs, which will provide further evidence to support this approach.
Collapse
Affiliation(s)
- Jason S Starr
- Division of Hematology/Oncology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Timothy J Hobday
- Division of Hematology/Oncology, Mayo Clinic, Rochester, MN, USA
| | - Akash Sharma
- Division of Nuclear Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Ayse Tuba Kendi
- Division of Hematology/Oncology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
75
|
Zhou P, Chen L, Yan D, Huang C, Chen G, Wang Z, Zhong L, Luo W, Chen D, Chun C, Zhang S, Li G. Early variations in lymphocytes and T lymphocyte subsets are associated with radiation pneumonitis in lung cancer patients and experimental mice received thoracic irradiation. Cancer Med 2020; 9:3437-3444. [PMID: 32207253 PMCID: PMC7221303 DOI: 10.1002/cam4.2987] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 11/29/2022] Open
Abstract
There were no ideal markers to predict the development of radiation pneumonitis (RP). We want to investigate the value of variations of lymphocytes and T lymphocyte subsets in predicting RP after radiotherapy (RT) of lung cancer based on previous clinical findings. A total of 182 lung cancer patients who received RT were retrospectively analyzed. Circulating lymphocytes and T lymphocyte subsets were measured before, during, and after RT. Patients were evaluated from the start of RT to 6 months post‐RT. A mice model with acute radiation‐induced lung injury was established and circulating lymphocytes were measured weekly until 8 weeks after irradiation. Univariate and multivariate analyses were adopted to identify risk factors of RP. Lymphocyte levels significantly decreased (P < .001) in patients before RP symptoms developed that also was able to be seen in the mice model and the values recovered during remission of symptoms. The decrease in lymphocyte count reflected the severity of RP. Meanwhile, CD4+ T lymphocyte count was significantly lower during the occurrence of symptoms in patients with RP than in those without RP (P < .001), and it improved along with RP recovery. Levels of lymphocytes and CD4+ T lymphocyte subsets proved as independent predictors of RP. Here we showed that lower peripheral blood levels of lymphocytes and CD4+ T lymphocyte were associated with an increased risk of RP, which was validated by this mice model, and thus are associated with differences in radiation‐induced lung toxicity among individuals and help identify those who are susceptible to developing RP after RT.
Collapse
Affiliation(s)
- Pu Zhou
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Lu Chen
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Dong Yan
- Institute for Pathology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Changlin Huang
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Guangpeng Chen
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zhiyi Wang
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Liangzhi Zhong
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wen Luo
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Diangang Chen
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Chui Chun
- Institute for Radiology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shushu Zhang
- Institute for Radiology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Guanghui Li
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
76
|
Cell repopulation, rewiring metabolism, and immune regulation in cancer radiotherapy. RADIATION MEDICINE AND PROTECTION 2020. [DOI: 10.1016/j.radmp.2020.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
77
|
Lee AK, Pan D, Bao X, Hu M, Li F, Li CY. Endogenous Retrovirus Activation as a Key Mechanism of Anti-Tumor Immune Response in Radiotherapy. Radiat Res 2020; 193:305-317. [PMID: 32074012 DOI: 10.1667/rade-20-00013] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The generation of DNA double-strand breaks has historically been taught as the mechanism through which radiotherapy kills cancer cells. Recently, radiation-induced cytosolic DNA release and activation of the cGAS/STING pathway, with ensuing induction of interferon secretion and immune activation, have been recognized as important mechanisms for radiation-mediated anti-tumor efficacy. Here we demonstrate that radiation-induced activation of endogenous retroviruses (ERVs) also plays a major role in regulating the anti-tumor immune response during irradiation. Radiation-induced ERV-associated dsRNA transcription and subsequent activation of the innate antiviral MDA5/MAVS/TBK1 pathway led to downstream transcription of interferon-stimulated genes. Additionally, genetic knockout of KAP1, a chromatin modulator responsible for suppressing ERV transcription sites within the genome, enhanced the effect of radiation-induced anti-tumor response in vivo in two different tumor models. This anti-tumor response was immune-mediated and required an intact host immune system. Our findings indicate that radiation-induced ERV-dsRNA expression and subsequent immune response play critical roles in clinical radiotherapy, and manipulation of epigenetic regulators and the dsRNA-sensing innate immunity pathway could be promising targets to enhance the efficacy of radiotherapy and cancer immunotherapy.
Collapse
Affiliation(s)
- Andrew K Lee
- Department of Pharmacology and Cancer Biology, Duke University Graduate School, Durham, North Carolina
| | | | | | | | | | - Chuan-Yuan Li
- Department of Pharmacology and Cancer Biology, Duke University Graduate School, Durham, North Carolina.,Department of Dermatology.,Department of Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
78
|
Choi JS, Sansoni ER, Lovin BD, Lindquist NR, Phan J, Mayo LL, Ferrarotto R, Su SY. Abscopal Effect Following Immunotherapy and Combined Stereotactic Body Radiation Therapy in Recurrent Metastatic Head and Neck Squamous Cell Carcinoma: A Report of Two Cases and Literature Review. Ann Otol Rhinol Laryngol 2019; 129:517-522. [PMID: 31875405 DOI: 10.1177/0003489419896602] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE We present two patients with recurrent, metastatic head and neck squamous cell carcinoma (R/M HNSCC) after platinum-based chemotherapy and radiotherapy (RT) with complete response via abscopal effect following combined immunotherapy (IT) and stereotactic body radiation therapy (SBRT). We review the literature for patients undergoing combined treatment with IT and RT to identify potential cases of abscopal response. STUDY DESIGN This is a case series with a contemporary review of the literature. METHODS Retrospective chart review identified two patients with potential abscopal responses after IT and RT for R/M HNSCC. The MEDLINE database was queried using the search terms "abscopal AND head and neck squamous cell carcinoma" and "immunotherapy AND stereotactic body radiation therapy." RESULTS Two patients with metastatic HNSCC developed complete responses via a possible abscopal effect following combined SBRT and IT. Interim follow-up of both patients revealed a sustained, complete response. We examine the immunogenic effects of RT and report the first cases of potential abscopal effect for R/M HNSCC. We also review several preclinical studies demonstrating the synergistic efficacy of combined RT and IT with a discussion of possible mechanism. CONCLUSION Observation of abscopal effect with combined IT and RT is currently under investigation through several preclinical studies and trials. To the best of our knowledge, these are the first two reported cases of abscopal effect for patients with HNSCC. We report two patients with R/M HNSCC with sustained, complete response after systemic IT and local RT.
Collapse
Affiliation(s)
- Jonathan S Choi
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Eugene R Sansoni
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Benjamin D Lovin
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Nathan R Lindquist
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Jack Phan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lauren L Mayo
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Renata Ferrarotto
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shirley Y Su
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
79
|
Alcaide-Leon P, Luks TL, Lafontaine M, Lupo JM, Okada H, Clarke JL, Villanueva-Meyer JE. Treatment-induced lesions in newly diagnosed glioblastoma patients undergoing chemoradiotherapy and heat-shock protein vaccine therapy. J Neurooncol 2019; 146:71-78. [PMID: 31728884 DOI: 10.1007/s11060-019-03336-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 11/05/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Treatment-induced lesions represent a great challenge in neuro-oncology. The aims of this study were (i) to characterize treatment induced lesions in glioblastoma patients treated with chemoradiotherapy and heat-shock protein (HSP) vaccine and (ii) to evaluate the diagnostic accuracy of diffusion weighted imaging for differentiation between treatment-induced lesions and tumor progression. METHODS Twenty-seven patients with newly diagnosed glioblastoma treated with HSP vaccine and chemoradiotherapy were included. Serial magnetic resonance imaging evaluation was performed to detect treatment-induced lesions and assess their growth. Quantitative analysis of the apparent diffusion coefficient (ADC) was performed to discriminate treatment-induced lesions from tumor progression. Mann-Whitney U-test and receiver operating characteristic (ROC) curves were used for analysis. RESULTS Thirty-three percent of patients developed treatment-induced lesions. Five treatment-related lesions appeared between end of radiotherapy and the first vaccine administration; 4 lesions within the first 4 months from vaccine initiation and 1 at 3.5 years. Three patients with pathology proven treatment-induced lesions showed a biphasic growth pattern progressed shortly after. ADC ratio between the peripheral enhancing rim and central necrosis showed an accuracy of 0.84 (95% CI 0.63-1) for differentiation between progression and treatment-induced lesions. CONCLUSION Our findings do not support the iRANO recommendation of a 6-month time window in which progressive disease should not be declared after immunotherapy initiation. A biphasic growth pattern of pathologically proven treatment-induced lesions was associated with a dismal prognosis. The presence of lower ADC values in the central necrotic portion of the lesions compared to the enhancing rim shows high specificity for detection of treatment-induced lesions.
Collapse
Affiliation(s)
- Paula Alcaide-Leon
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA. .,Medical Imaging, University Health Network, 399 Bathurst St, Toronto, ON, M5T 2S8, Canada.
| | - Tracy L Luks
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Marisa Lafontaine
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer L Clarke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
80
|
Ellsworth SG, Yalamanchali A, Zhang H, Grossman SA, Hobbs R, Jin JY. Comprehensive Analysis of the Kinetics of Radiation-Induced Lymphocyte Loss in Patients Treated with External Beam Radiation Therapy. Radiat Res 2019; 193:73-81. [PMID: 31675264 DOI: 10.1667/rr15367.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced lymphopenia (RIL) is associated with worse survival in patients with solid tumors, as well as lower response rates to checkpoint inhibitors. While single-fraction total-body irradiation is known to result in exponential decreases in the absolute lymphocyte count (ALC), the kinetics of lymphocyte loss after focal fractionated exposures have not previously been characterized. In the current study, lymphocyte loss kinetics was analyzed among patients undergoing focal fractionated radiotherapy for clinical indications. This registry-based study included 419 patients who received either total-body irradiation (TBI; n = 30), stereotactic body radiation therapy (SBRT; n = 73) or conventionally fractionated chemoradiation therapy (CFRT; n = 316). For each patient, serial ALCs were plotted against radiotherapy fraction number. The initial three weeks of treatment for CFRT patients and the entirety of treatment for SBRT and TBI patients were fit to exponential decay in the form ALC(x) = ae-bx, where ALC(x) is the ALC after x fractions. From those fits, fractional lymphocyte loss (FLL) was calculated as FLL = (1 - e-b) * 100, and multivariable regression was performed to identify significant correlates of FLL. Median linearized R2 when fitting the initial fractions was 0.98, 0.93 and 0.97 for patients receiving TBI, SBRT and CFRT, respectively. In CFRT patients, apparent ALC loss rate slowed after week 3. Fitting ALC loss over the entire CFRT course therefore required the addition of a constant term, "c". For TBI and SBRT patients, treatment ended during the pure exponential decay phase. Initial FLL varied significantly with treatment technique. Mean FLL was 35.5%, 24.3% and 10.77% for patients receiving TBI, SBRT and CFRT, respectively (P < 0.001). Significant correlates of FLL varied by site and included field size, dose per fraction, mean spleen dose, chemotherapy backbone and age. Finally, total percentage ALC loss during radiotherapy was highly correlated with FLL (P < 0.001). Lymphocyte depletion kinetics during the initial phase of fractionated radiotherapy are characterized by pure exponential decay. Initial FLL is strongly correlated with radiotherapy planning parameters and total percentage ALC loss. The two groups with the highest FLL received no concurrent chemotherapy, suggesting that ALC loss can be a consequence of radiotherapy alone. This work may assist in selecting patients for adaptive radiotherapy approaches to mitigate RIL risk.
Collapse
Affiliation(s)
- Susannah G Ellsworth
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana; Departments of
| | - Anirudh Yalamanchali
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana; Departments of
| | - Hong Zhang
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana; Departments of
| | | | - Robert Hobbs
- Departments of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Jian-Yue Jin
- Department of Radiation Oncology, Case Western Reserve University School of Medicine
| |
Collapse
|
81
|
Petrelli F, De Stefani A, Trevisan F, Parati C, Inno A, Merelli B, Ghidini M, Bruschieri L, Vitali E, Cabiddu M, Borgonovo K, Ghilardi M, Barni S, Ghidini A. Combination of radiotherapy and immunotherapy for brain metastases: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2019; 144:102830. [PMID: 31733443 DOI: 10.1016/j.critrevonc.2019.102830] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 06/15/2019] [Accepted: 10/26/2019] [Indexed: 01/15/2023] Open
Abstract
Radiotherapy (RT) represents a mainstay in the treatment of brain metastases (BMs) from solid tumors. Immunotherapy (IT) has improved survival of metastatic cancer patients across many tumor types. The combination of RT and IT for the treatment of BMs has a strong rationale, but data on efficacy and safety of this combination is still limited. A systematic search of PubMed, the Cochrane Central Register of Controlled Trials, and EMBASE was conducted. 33 studies were included for a total of 1520 patients, most of them with melanoma (87%). Median pooled OS was 15.9 months (95%CI 13.9-18.1). One- and 2-year OS rates were 55.2% (95% CI 49.3-60.9) and 35.7% (95% CI 30.4-41.3), respectively. Addition of IT to RT was associated with improved OS (HR = 0.54, 95%CI 0.44-0.67; P < 0.001). For patients with BMs from solid tumors, addition of concurrent IT to brain RT is able to increase survival and provide long term control.
Collapse
Affiliation(s)
- Fausto Petrelli
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy.
| | - Agostina De Stefani
- Radiotherapy Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Francesca Trevisan
- Radiotherapy Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Chiara Parati
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Alessandro Inno
- Oncology Unit, Ospedale Sacro Cuore don Calabria Cancer Care Center, Via Don A. Sempreboni 5, 37024, Negrar, VR, Italy
| | - Barbara Merelli
- Oncology Unit, ASST Papa Giovanni XXIII, Piazza Oms 1, 24127, Bergamo, Italy
| | - Michele Ghidini
- Oncology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Viale F. Sforza 28, 20122, Milano, Italy
| | - Lorenza Bruschieri
- Radiotherapy Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Elisabetta Vitali
- Radiotherapy Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Mary Cabiddu
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Karen Borgonovo
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Mara Ghilardi
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Sandro Barni
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Antonio Ghidini
- Oncology Unit, Casa di Cura Igea, Via Marcona 69, 20144, Milano, Italy
| |
Collapse
|
82
|
Tabrizi S, McDuff S, Ho AY. Combining Radiation Therapy with Immune Checkpoint Blockadein Breast Cancer. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00327-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
83
|
Steer A, Cordes N, Jendrossek V, Klein D. Impact of Cancer-Associated Fibroblast on the Radiation-Response of Solid Xenograft Tumors. Front Mol Biosci 2019; 6:70. [PMID: 31475157 PMCID: PMC6705217 DOI: 10.3389/fmolb.2019.00070] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 07/29/2019] [Indexed: 01/18/2023] Open
Abstract
Increasing evidence indicates that the heterogeneous tumor stroma supports therapy resistance at multiple levels. Fibroblasts, particularly cancer-associated fibroblasts (CAFs) are critical components of the tumor stroma. However, the impact of CAFs on the outcome of radiotherapy (RT) is poorly understood. Here, we investigated if and how fibroblasts/CAFs modulate the radiation response of malignant tumors by altering cancer cell radiosensitivity or radioresistance in vitro and in vivo. The influence of fibroblasts on cancer cell proliferation, cell death induction and long-term survival after RT was studied using different sets of fibroblasts and cancer cells in an indirect co-culture (2D) system to analyse potential paracrine interactions or a 3D model to study direct interactions. Paracrine signals from embryonic NIH-3T3 fibroblasts promoted MPR31.4 prostate and Py8119 breast cancer cell proliferation. Indirect co-culture with L929 skin fibroblasts induced higher levels of apoptosis in irradiated MPR31.4 cells, while they promoted proliferation of irradiated Py8119 cells. In addition, NIH-3T3 fibroblasts promoted long-term clonogenic survival of both tumor cell types upon irradiation in the 3D co-culture system when compared to non-irradiated controls. Also in vivo, co-implantation of cancer cells and fibroblasts resulted in different effects depending on the respective cell combinations used: co-implantation of MPR31.4 cells with NIH-3T3 fibroblasts or of Py8119 cells with L929 fibroblasts led to increased tumor growth and reduced radiation-induced tumor growth delay when compared to the respective tumors without co-implanted fibroblasts. Taken together, the impact of fibroblasts on cancer cell behavior and radiation sensitivity largely depended on the respective cell types used as they either exerted a pro-tumorigenic and radioresistance-promoting effect, an anti-tumorigenic effect, or no effect. We conclude that the plasticity of fibroblasts allows for such a broad spectrum of activities by the same fibroblast and that this plasticity is at least in part mediated by cancer cell-induced fibroblast activation toward CAFs.
Collapse
Affiliation(s)
- Alizée Steer
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Nils Cordes
- Faculty of Medicine, OncoRay-National Center for Radiation Research in Oncology, Technische Universität Dresden, Dresden, Germany.,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Institute of Radiooncology-OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, Heidelberg, Germany.,German Cancer Research Center (DKFZ)-Partner Site Dresden, Heidelberg, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Diana Klein
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| |
Collapse
|
84
|
Durante M, Formenti S. Harnessing radiation to improve immunotherapy: better with particles? Br J Radiol 2019; 93:20190224. [PMID: 31317768 DOI: 10.1259/bjr.20190224] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The combination of radiotherapy and immunotherapy is one of the most promising strategies for cancer treatment. Recent clinical results support the pre-clinical experiments pointing to a benefit for the combined treatment in metastatic patients. Charged particle therapy (using protons or heavier ions) is considered one of the most advanced radiotherapy techniques, but its cost remains higher than conventional X-ray therapy. The most important question to be addressed to justify a more widespread use of particle therapy is whether they can be more effective than X-rays in combination with immunotherapy. Protons and heavy ions have physical advantages compared to X-rays that lead to a reduced damage to the immune cells, that are required for an effective immune response. Moreover, densely ionizing radiation may have biological advantages, due to different cell death pathways and release of cytokine mediators of inflammation. We will discuss results in esophageal cancer patients showing that charged particles can reduce the damage to blood lymphocytes compared to X-rays, and preliminary in vitro studies pointing to an increased release of immune-stimulating cytokines after heavy ion exposure. Pre-clinical and clinical studies are ongoing to test these hypotheses.
Collapse
Affiliation(s)
- Marco Durante
- GSI Helmholtzzentrum für Schwerionenforschung, Biophysics Department, Darmstadt, Germany.,Technische Universität Darmstadt, Institut für Festkörperphysik, Darmstadt, Germany
| | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
85
|
Trommer M, Yeo SY, Persigehl T, Bunck A, Grüll H, Schlaak M, Theurich S, von Bergwelt-Baildon M, Morgenthaler J, Herter JM, Celik E, Marnitz S, Baues C. Abscopal Effects in Radio-Immunotherapy-Response Analysis of Metastatic Cancer Patients With Progressive Disease Under Anti-PD-1 Immune Checkpoint Inhibition. Front Pharmacol 2019; 10:511. [PMID: 31156434 PMCID: PMC6530339 DOI: 10.3389/fphar.2019.00511] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/24/2019] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoint inhibition (ICI) targeting the programmed death receptor 1 (PD-1) has shown promising results in the fight against cancer. Systemic anti-tumor reactions due to radiation therapy (RT) can lead to regression of non-irradiated lesions (NiLs), termed “abscopal effect” (AbE). Combination of both treatments can enhance this effect. The aim of this study was to evaluate AbEs during anti-PD-1 therapy and irradiation. We screened 168 patients receiving pembrolizumab or nivolumab at our center. Inclusion criteria were start of RT within 1 month after the first or last application of pembrolizumab (2 mg/kg every 3 weeks) or nivolumab (3 mg/kg every 2 weeks) and at least one metastasis outside the irradiation field. We estimated the total dose during ICI for each patient using the linear quadratic (LQ) model expressed as 2 Gy equivalent dose (EQD2) using α/β of 10 Gy. Radiological images were required showing progression or no change in NiLs before and regression after completion of RT(s). Images must have been acquired at least 4 weeks after the onset of ICI or RT. The surface areas of the longest diameters of the short- and long-axes of NiLs were measured. One hundred twenty-six out of 168 (75%) patients received ICI and RT. Fifty-three percent (67/126) were treated simultaneously, and 24 of these (36%) were eligible for lesion analysis. AbE was observed in 29% (7/24). One to six lesions (mean = 3 ± 2) in each AbE patient were analyzed. Patients were diagnosed with malignant melanoma (MM) (n = 3), non-small cell lung cancer (NSCLC) (n = 3), and renal cell carcinoma (RCC) (n = 1). They were irradiated once (n = 1), twice (n = 2), or three times (n = 4) with an average total EQD2 of 120.0 ± 37.7 Gy. Eighty-two percent of RTs of AbE patients were applied with high single doses. MM patients received pembrolizumab, NSCLC, and RCC patients received nivolumab for an average duration of 45 ± 35 weeks. We demonstrate that 29% of the analyzed patients showed AbE. Strict inclusion criteria were applied to distinguish the effects of AbE from the systemic effect of ICI. Our data suggest the clinical existence of systemic effects of irradiation under ICI and could contribute to the development of a broader range of cancer treatments.
Collapse
Affiliation(s)
- Maike Trommer
- Faculty of Medicine and University Hospital Cologne, Department of Radiation Oncology and Cyberknife Center, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Radio Immune-Oncology Consortium, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology (CIO Köln Bonn), University of Cologne, Cologne, Germany
| | - Sin Yuin Yeo
- Faculty of Medicine and University Hospital Cologne, Radio Immune-Oncology Consortium, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Department of Diagnostic and Interventional Radiology, University of Cologne, Cologne, Germany
| | - Thorsten Persigehl
- Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology (CIO Köln Bonn), University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Department of Diagnostic and Interventional Radiology, University of Cologne, Cologne, Germany
| | - Anne Bunck
- Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology (CIO Köln Bonn), University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Department of Diagnostic and Interventional Radiology, University of Cologne, Cologne, Germany
| | - Holger Grüll
- Faculty of Medicine and University Hospital Cologne, Radio Immune-Oncology Consortium, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Department of Diagnostic and Interventional Radiology, University of Cologne, Cologne, Germany
| | - Max Schlaak
- Faculty of Medicine and University Hospital Cologne, Radio Immune-Oncology Consortium, University of Cologne, Cologne, Germany.,Department of Dermatology and Allergology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Sebastian Theurich
- Faculty of Medicine and University Hospital Cologne, Radio Immune-Oncology Consortium, University of Cologne, Cologne, Germany.,Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Gene Center, Cancer- and Immunometabolism Research Group, Ludwig-Maximilians University Munich, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Faculty of Medicine and University Hospital Cologne, Radio Immune-Oncology Consortium, University of Cologne, Cologne, Germany.,Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Janis Morgenthaler
- Faculty of Medicine and University Hospital Cologne, Department of Radiation Oncology and Cyberknife Center, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology (CIO Köln Bonn), University of Cologne, Cologne, Germany
| | - Jan M Herter
- Faculty of Medicine and University Hospital Cologne, Department of Radiation Oncology and Cyberknife Center, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology (CIO Köln Bonn), University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Eren Celik
- Faculty of Medicine and University Hospital Cologne, Department of Radiation Oncology and Cyberknife Center, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology (CIO Köln Bonn), University of Cologne, Cologne, Germany
| | - Simone Marnitz
- Faculty of Medicine and University Hospital Cologne, Department of Radiation Oncology and Cyberknife Center, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Radio Immune-Oncology Consortium, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology (CIO Köln Bonn), University of Cologne, Cologne, Germany
| | - Christian Baues
- Faculty of Medicine and University Hospital Cologne, Department of Radiation Oncology and Cyberknife Center, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Radio Immune-Oncology Consortium, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology (CIO Köln Bonn), University of Cologne, Cologne, Germany
| |
Collapse
|
86
|
The Combination of Stereotactic Body Radiation Therapy and Immunotherapy in Primary Liver Tumors. JOURNAL OF ONCOLOGY 2019; 2019:4304817. [PMID: 31182960 PMCID: PMC6512065 DOI: 10.1155/2019/4304817] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/18/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023]
Abstract
Treatment recommendations for primary liver malignancies, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are complex and require a multidisciplinary approach. Despite surgical options that are potentially curative, options for nonsurgical candidates include systemic therapy, radiotherapy (RT), transarterial chemoembolization (TACE), and radiofrequency ablation (RFA). Stereotactic Body Radiation Therapy (SBRT) is now in routine use for the treatment of lung cancer, and there is growing evidence supporting its use in liver tumors. SBRT has the advantage of delivering ablative radiation doses in a limited number of fractions while minimizing the risk of radiation-induced liver disease (RILD) through highly conformal treatment plans. It should be considered in a multidisciplinary setting for the management of patients with unresectable, locally advanced primary liver malignancies and limited treatment options. Recently, the combination of immunotherapy with SBRT has been proposed to improve antitumor effects through engaging the immune system. This review aims at shedding light on the novel concept of the combination strategy of immune-radiotherapy in liver tumors by exploring the evidence surrounding the use of SBRT and immunotherapy for the treatment of HCC and CCA.
Collapse
|
87
|
Bristow RG, Alexander B, Baumann M, Bratman SV, Brown JM, Camphausen K, Choyke P, Citrin D, Contessa JN, Dicker A, Kirsch DG, Krause M, Le QT, Milosevic M, Morris ZS, Sarkaria JN, Sondel PM, Tran PT, Wilson GD, Willers H, Wong RKS, Harari PM. Combining precision radiotherapy with molecular targeting and immunomodulatory agents: a guideline by the American Society for Radiation Oncology. Lancet Oncol 2019; 19:e240-e251. [PMID: 29726389 DOI: 10.1016/s1470-2045(18)30096-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 10/30/2017] [Accepted: 12/18/2017] [Indexed: 02/07/2023]
Abstract
The practice of radiation oncology is primarily based on precise technical delivery of highly conformal, image-guided external beam radiotherapy or brachytherapy. However, systematic research efforts are being made to facilitate individualised radiation dose prescriptions on the basis of gene-expressssion profiles that reflect the radiosensitivity of tumour and normal tissue. This advance in precision radiotherapy should complement those benefits made in precision cancer medicine that use molecularly targeted agents and immunotherapies. The personalisation of cancer therapy, predicated largely on genomic interrogation, is facilitating the selection of therapies that are directed against driver mutations, aberrant cell signalling, tumour microenvironments, and genetic susceptibilities. With the increasing technical power of radiotherapy to safely increase local tumour control for many solid tumours, it is an opportune time to rigorously explore the potential benefits of combining radiotherapy with molecular targeted agents and immunotherapies to increase cancer survival outcomes. This theme provides the basis and foundation for this American Society for Radiation Oncology guideline on combining radiotherapy with molecular targeting and immunotherapy agents.
Collapse
Affiliation(s)
- Robert G Bristow
- Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, ON, Canada.
| | - Brian Alexander
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Scott V Bratman
- Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, ON, Canada
| | - J Martin Brown
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Kevin Camphausen
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter Choyke
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Deborah Citrin
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joseph N Contessa
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Adam Dicker
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - David G Kirsch
- Department of Radiation Oncology and Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | | | - Quynh-Thu Le
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michael Milosevic
- Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Phuoc T Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Department of Oncology, and Department of Urology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - George D Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, USA
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca K S Wong
- Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
88
|
Administration of Dendritic Cells and Anti-PD-1 Antibody Converts X-ray Irradiated Tumors Into Effective In situ Vaccines. Int J Radiat Oncol Biol Phys 2019; 103:958-969. [DOI: 10.1016/j.ijrobp.2018.11.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 11/05/2018] [Accepted: 11/10/2018] [Indexed: 12/21/2022]
|
89
|
Osborn VW, Lee A, Yamada Y. Stereotactic Body Radiation Therapy for Spinal Malignancies. Technol Cancer Res Treat 2019; 17:1533033818802304. [PMID: 30343661 PMCID: PMC6198394 DOI: 10.1177/1533033818802304] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Stereotactic body radiation therapy and stereotactic radiosurgery have become important treatment options for the treatment of spinal malignancies. A better understanding of dose tolerances with more conformal technology have allowed administration of higher and more ablative doses. In this review, the framework for approaching a patient with spinal metastases and primary tumors will be discussed as well as details on the delivery of this treatment.
Collapse
Affiliation(s)
- Virginia W Osborn
- 1 Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,2 Department of Radiation Oncology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Anna Lee
- 1 Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,2 Department of Radiation Oncology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Yoshiya Yamada
- 1 Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
90
|
Belluomini L, Fiorica F, Frassoldati A. Immune Checkpoint Inhibitors and Radiotherapy in NSCLC Patients: Not Just a Fluke. Oncol Ther 2019; 7:83-91. [PMID: 32700194 PMCID: PMC7360008 DOI: 10.1007/s40487-019-0092-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Indexed: 12/30/2022] Open
Abstract
The discovery of immune checkpoint inhibitors (ICIs) such as programmed cell death protein 1 (PD-1) inhibitors, nivolumab and pembrolizumab, and programmed cell death ligand 1 (PD-L1) inhibitors, atezolizumab and durvalumab, has revolutionized the treatment of advanced non-small cell lung cancer (NSCLC). Concurrent radiotherapy (RT) is of particular interest with regard to the potential role for this combination in many settings. The purpose of this commentary is to evaluate the potential for the combination of immune checkpoint inhibitors and radiotherapy, including analysis of studies that have considered this combination in various settings.
Collapse
Affiliation(s)
| | - Francesco Fiorica
- Radiation Oncology Unit, University Hospital St. Anna, Ferrara, Italy
| | | |
Collapse
|
91
|
Bahig H, Aubin F, Stagg J, Gologan O, Ballivy O, Bissada E, Nguyen-Tan FP, Soulières D, Guertin L, Filion E, Christopoulos A, Lambert L, Tehfe M, Ayad T, Charpentier D, Jamal R, Wong P. Phase I/II trial of Durvalumab plus Tremelimumab and stereotactic body radiotherapy for metastatic head and neck carcinoma. BMC Cancer 2019; 19:68. [PMID: 30642290 PMCID: PMC6332607 DOI: 10.1186/s12885-019-5266-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 01/02/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The efficacy of immunotherapy targeting the PD-1/PD-L1 pathway has previously been demonstrated in metastatic head and neck squamous cell carcinoma (HNSCC). Stereotactic Body Radiotherapy (SBRT) aims at ablating metastatic lesions and may play a synergistic role with immunotherapy. The purpose of this study is to assess the safety and efficacy of triple treatment combination (TTC) consisting of the administration of durvalumab and tremelimumab in combination with SBRT in metastatic HNSCC. METHOD This is a phase I/II single arm study that will include 35 patients with 2-10 extracranial metastatic lesions. Patients will receive durvalumab (1500 mg IV every 4 weeks (Q4W)) and tremelimumab (75 mg IV Q4W for a total of 4 doses) until progression, unacceptable toxicity or patient withdrawal. SBRT to 2-5 metastases will be administered between cycles 2 and 3 of immunotherapy. The safety of the treatment combination will be evaluated through assessment of TTC-related toxicities, defined as grade 3-5 toxicities based on Common Terminology Criteria for Adverse Events (v 4.03), occurring within 6 weeks from SBRT start, and that are definitely, probably or possibly related to the combination of all treatments. We hypothesize that dual targeting of PD-L1 and CTLA-4 pathways combined with SBRT will lead to < 35% grade 3-5 acute toxicities related to TTC. Progression free survival (PFS) will be the primary endpoint of the phase II portion of this study and will be assessed with radiological exams every 8 weeks using the RECIST version 1.1 criteria. DISCUSSION The combination of synergistic dual checkpoints inhibition along with ablative radiation may significantly potentiate the local and systemic disease control. This study constitutes the first clinical trial combining effects of SBRT with dual checkpoint blockade with durvalumab and tremelimumab in the treatment of metastatic HNSCC. If positive, this study would lead to a phase III trial testing this treatment combination against standard of care in metastatic HNSCC. TRIAL REGISTRATION NCT03283605 . Registration date: September 14, 2017; version 1.
Collapse
Affiliation(s)
- Houda Bahig
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Francine Aubin
- Department of Medical Oncology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Olguta Gologan
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
- Pathology Department, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - Olivier Ballivy
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Eric Bissada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
- Department of Otorhinolaryngology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - Felix-Phuc Nguyen-Tan
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Denis Soulières
- Department of Medical Oncology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Louis Guertin
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
- Department of Otorhinolaryngology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - Edith Filion
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Apostolos Christopoulos
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
- Department of Otorhinolaryngology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - Louise Lambert
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Mustapha Tehfe
- Department of Medical Oncology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Tareck Ayad
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
- Department of Otorhinolaryngology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - Danielle Charpentier
- Department of Medical Oncology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Rahima Jamal
- Department of Medical Oncology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Philip Wong
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| |
Collapse
|
92
|
Yilmaz MT, Elmali A, Yazici G. Abscopal Effect, From Myth to Reality: From Radiation Oncologists' Perspective. Cureus 2019; 11:e3860. [PMID: 30899611 PMCID: PMC6414182 DOI: 10.7759/cureus.3860] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The abscopal effect is mediated by a systemic anti-tumor immune response and reflects the regression of non-irradiated metastatic lesions at a distance from the primary site of irradiation. This review will focus on understanding the biological rationale behind the abscopal effect of radiotherapy (RT), which has a recently renewed interest as a result of the successes achieved with immunotherapy and RT in combination. Both RT and immunotherapy are standard components of modern treatment regimens. Combination of these two modalities results in an increased response in the irradiated lesions themselves and the metastatic regions distant from the site of irradiation. We will summarize the abscopal effect of radiotherapy, in particular, the synergistic effect of RT and immunotherapy.
Collapse
Affiliation(s)
| | - Aysenur Elmali
- Radiation Oncology, Hacettepe University Medical School, Ankara, TUR
| | - Gozde Yazici
- Radiation Oncology, Hacettepe University Medical School, Ankara, TUR
| |
Collapse
|
93
|
Dahl O, Dale JE, Brydøy M. Rationale for combination of radiation therapy and immune checkpoint blockers to improve cancer treatment. Acta Oncol 2019; 58:9-20. [PMID: 30632870 DOI: 10.1080/0284186x.2018.1554259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Radiation therapy for cancer is considered to be immunosuppressive. However, the cellular response after radiation therapy may stimulate or suppress an immune response. The effect may vary with the tumor type and occasionally tumor regressions have been observed outside the irradiated volume, both in animal studies and in the clinic. A renewed interest in the role of immunity for the observed effect of radiation came with the current recognized role of immune checkpoint blockers (ICBs) for control of selected cancer types. We therefore here review preclinical studies and clinical reports on the interaction of ICBs and radiation as a basis for further clinical trials. Some tumor types where the combination of these modalities seems especially promising are also proposed.
Collapse
Affiliation(s)
- Olav Dahl
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Jon Espen Dale
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Marianne Brydøy
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
94
|
Radiation therapy for patients with newly diagnosed metastatic head and neck squamous cell carcinoma. Head Neck 2018; 41:130-138. [DOI: 10.1002/hed.25476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 03/11/2018] [Accepted: 07/05/2018] [Indexed: 11/07/2022] Open
|
95
|
Tsoutsou PG, Zaman K, Martin Lluesma S, Cagnon L, Kandalaft L, Vozenin MC. Emerging Opportunities of Radiotherapy Combined With Immunotherapy in the Era of Breast Cancer Heterogeneity. Front Oncol 2018; 8:609. [PMID: 30619749 PMCID: PMC6305124 DOI: 10.3389/fonc.2018.00609] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
The association of radiotherapy and immunotherapy has recently emerged as an exciting combination that might improve outcomes in many solid tumor settings. In the context of breast cancer, this opportunity is promising and under investigation. Given the heterogeneity of breast cancer, it might be meaningful to study the association of radiotherapy and immunotherapy distinctly among the various breast cancer subtypes. The use of biomarkers, such as tumor infiltrating lymphocytes, which are also associated to breast cancer heterogeneity, might provide an opportunity for tailored studies. This review highlights current knowledge of the association of radiotherapy and immunotherapy in the setting of breast cancer and attempts to highlight the therapeutic opportunities among breast cancer heterogeneity.
Collapse
Affiliation(s)
- Pelagia G Tsoutsou
- Division of Oncology, Radio-oncology Department, Vaudois University Hospital Centre (CHUV), Lausanne, Switzerland.,Radio-Oncology Research Laboratory, Vaudois University Hospital Centre (CHUV), Epalinges, Switzerland.,Radiation Oncology Department, Hôpital Neuchâtelois, La Chaux-de-Fonds, Switzerland
| | - Khalil Zaman
- Department of Oncology, Breast Center, Vaudois University Hospital Centre (CHUV), Lausanne, Switzerland
| | - Silvia Martin Lluesma
- Department of Oncology, Center of Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Laurene Cagnon
- Department of Oncology, Center of Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Lana Kandalaft
- Department of Oncology, Center of Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Marie-Catherine Vozenin
- Radio-Oncology Research Laboratory, Vaudois University Hospital Centre (CHUV), Epalinges, Switzerland
| |
Collapse
|
96
|
Stewart RD, Carlson DJ, Butkus MP, Hawkins R, Friedrich T, Scholz M. A comparison of mechanism-inspired models for particle relative biological effectiveness (RBE). Med Phys 2018; 45:e925-e952. [PMID: 30421808 DOI: 10.1002/mp.13207] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 09/05/2018] [Accepted: 09/13/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND SIGNIFICANCE The application of heavy ion beams in cancer therapy must account for the increasing relative biological effectiveness (RBE) with increasing penetration depth when determining dose prescriptions and organ at risk (OAR) constraints in treatment planning. Because RBE depends in a complex manner on factors such as the ion type, energy, cell and tissue radiosensitivity, physical dose, biological endpoint, and position within and outside treatment fields, biophysical models reflecting these dependencies are required for the personalization and optimization of treatment plans. AIM To review and compare three mechanism-inspired models which predict the complexities of particle RBE for various ion types, energies, linear energy transfer (LET) values and tissue radiation sensitivities. METHODS The review of models and mechanisms focuses on the Local Effect Model (LEM), the Microdosimetric-Kinetic (MK) model, and the Repair-Misrepair-Fixation (RMF) model in combination with the Monte Carlo Damage Simulation (MCDS). These models relate the induction of potentially lethal double strand breaks (DSBs) to the subsequent interactions and biological processing of DSB into more lethal forms of damage. A key element to explain the increased biological effectiveness of high LET ions compared to MV x rays is the characterization of the number and local complexity (clustering) of the initial DSB produced within a cell. For high LET ions, the spatial density of DSB induction along an ion's trajectory is much greater than along the path of a low LET electron, such as the secondary electrons produced by the megavoltage (MV) x rays used in conventional radiation therapy. The main aspects of the three models are introduced and the conceptual similarities and differences are critiqued and highlighted. Model predictions are compared in terms of the RBE for DSB induction and for reproductive cell survival. RESULTS AND CONCLUSIONS Comparisons of the RBE for DSB induction and for cell survival are presented for proton (1 H), helium (4 He), and carbon (12 C) ions for the therapeutically most relevant range of ion beam energies. The reviewed models embody mechanisms of action acting over the spatial scales underlying the biological processing of potentially lethal DSB into more lethal forms of damage. Differences among the number and types of input parameters, relevant biological targets, and the computational approaches among the LEM, MK and RMF models are summarized and critiqued. Potential experiments to test some of the seemingly contradictory aspects of the models are discussed.
Collapse
Affiliation(s)
- Robert D Stewart
- Department of Radiation Oncology, University of Washington School of Medicine, 1959 NE Pacific Street, Box 356043, Seattle, WA, 98195, USA
| | - David J Carlson
- Department of Therapeutic Radiology, Yale University, New Haven, CT, USA
| | - Michael P Butkus
- Department of Therapeutic Radiology, Yale University, New Haven, CT, USA
| | - Roland Hawkins
- Radiation Oncology Center, Ochsner Clinic Foundation, New Orleans, LA, 70121, USA
| | | | | |
Collapse
|
97
|
Song Y, Lee SY, Kim AR, Kim S, Heo J, Shum D, Kim SH, Choi I, Lee YJ, Seo HR. Identification of radiation-induced EndMT inhibitors through cell-based phenomic screening. FEBS Open Bio 2018; 9:82-91. [PMID: 30652076 PMCID: PMC6325571 DOI: 10.1002/2211-5463.12552] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/09/2018] [Accepted: 10/19/2018] [Indexed: 12/24/2022] Open
Abstract
Radiation‐induced pulmonary fibrosis (RIPF) triggers physiological abnormalities. Endothelial‐to‐mesenchymal transition (EndMT) is the phenotypic conversion of endothelial cells to fibroblast‐like cells and is involved in RIPF. In this study, we established a phenomic screening platform to measure radiation‐induced stress fibers and optimized the conditions for high‐throughput screening using human umbilical vein endothelial cells (HUVECs) to develop compounds targeting RIPF. The results of screening indicated that CHIR‐99021 reduced radiation‐induced fibrosis, as evidenced by an enlargement of cell size and increases in actin stress fibers and α‐smooth muscle actin expression. These effects were elicited without inducing serious toxicity in HUVECs, and the cytotoxic effect of ionizing radiation (IR) in nonsmall cell lung cancer was also enhanced. These results demonstrate that CHIR‐99021 enhanced the effects of IR therapy by suppressing radiation‐induced EndMT in lung cancer.
Collapse
Affiliation(s)
- Yeonhwa Song
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| | - Su-Yeon Lee
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| | - A-Ram Kim
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| | - Sanghwa Kim
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| | - Jinyeong Heo
- Assay Development and Screening Institut Pasteur Korea Seongnam-si Korea
| | - David Shum
- Assay Development and Screening Institut Pasteur Korea Seongnam-si Korea
| | - Se-Hyuk Kim
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| | - Inhee Choi
- Medicinal Chemistry Institut Pasteur Korea Seongnam-si Korea
| | - Yoon-Jin Lee
- Division of Radiation Effects Korea Institute of Radiological and Medical Sciences Seoul Korea
| | - Haeng Ran Seo
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| |
Collapse
|
98
|
|
99
|
Deana Y, Burgara-Estrella AJ, Montalvo-Corral M, Angulo-Molina A, Acosta-Elías MA, Silva-Campa E, Sarabia-Sainz JA, Rodríguez-Hernández IC, Pedroza-Montero MR. Effect of gamma irradiation doses in the structural and functional properties of mice splenic cells. Int J Radiat Biol 2018; 95:286-297. [PMID: 30496016 DOI: 10.1080/09553002.2019.1547435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSE Ionizing radiation is nowadays effectively used in cancer treatments. However, the effect of irradiation in immune-system cells is poorly understood and remains controversial. The aim of this work was to determine the effect of γ-irradiation in the structural and functional properties of mice splenic cells. MATERIALS AND METHODS Structural traits of irradiated splenic cells were evaluated by Atomic Force Microscopy and Raman spectroscopy. Functional properties were measured by gene and protein expression by RT-qPCR and ELISA, respectively. The induced cytotoxic effect was evaluated by MTT assay and the phagocytic capability by flow cytometry. RESULTS Membrane roughness and molecular composition of splenic adherent cells are not changed by irradiation doses exposure. An increase in transcription of pro-inflammatory cytokines was observed. While protein expression decreased in IL-2 dose-dependent, relevant differences were identified in the anti-inflammatory marker IL-10 at 27 Gy. An increase of cytotoxicity in irradiated cells at 7 Gy and 27 Gy doses was observed, while phagocytosis was slight increased at 7 Gy dose but not statistically significant. CONCLUSIONS We have demonstrated that γ-irradiation affects the splenic cells and changes the cytokines profile toward a pro-inflammatory phenotype and a tendency to increase the cytotoxicity was found, which implies a stimulation of immune response induced by γ-irradiation.
Collapse
Affiliation(s)
- Yanik Deana
- a Departamento de Investigación en Física , Universidad de Sonora , Hermosillo , México.,b Institute for Chemistry and Bioanalytics , University of Applied Sciences and Arts Northwestern , Muttenz , Switzerland
| | | | - Maricela Montalvo-Corral
- c Departamento de Nutrición , Centro de Investigación en Alimentación y Desarrollo A.C. , Hermosillo , México
| | | | - Mónica A Acosta-Elías
- a Departamento de Investigación en Física , Universidad de Sonora , Hermosillo , México
| | - Erika Silva-Campa
- a Departamento de Investigación en Física , Universidad de Sonora , Hermosillo , México
| | - Jose A Sarabia-Sainz
- a Departamento de Investigación en Física , Universidad de Sonora , Hermosillo , México
| | | | | |
Collapse
|
100
|
Keall PJ, Nguyen DT, O'Brien R, Zhang P, Happersett L, Bertholet J, Poulsen PR. Review of Real-Time 3-Dimensional Image Guided Radiation Therapy on Standard-Equipped Cancer Radiation Therapy Systems: Are We at the Tipping Point for the Era of Real-Time Radiation Therapy? Int J Radiat Oncol Biol Phys 2018; 102:922-931. [PMID: 29784460 PMCID: PMC6800174 DOI: 10.1016/j.ijrobp.2018.04.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/21/2018] [Accepted: 04/05/2018] [Indexed: 01/29/2023]
Abstract
PURPOSE To review real-time 3-dimensional (3D) image guided radiation therapy (IGRT) on standard-equipped cancer radiation therapy systems, focusing on clinically implemented solutions. METHODS AND MATERIALS Three groups in 3 continents have clinically implemented novel real-time 3D IGRT solutions on standard-equipped linear accelerators. These technologies encompass kilovoltage, combined megavoltage-kilovoltage, and combined kilovoltage-optical imaging. The cancer sites treated span pelvic and abdominal tumors for which respiratory motion is present. For each method the 3D-measured motion during treatment is reported. After treatment, dose reconstruction was used to assess the treatment quality in the presence of motion with and without real-time 3D IGRT. The geometric accuracy was quantified through phantom experiments. A literature search was conducted to identify additional real-time 3D IGRT methods that could be clinically implemented in the near future. RESULTS The real-time 3D IGRT methods were successfully clinically implemented and have been used to treat more than 200 patients. Systematic target position shifts were observed using all 3 methods. Dose reconstruction demonstrated that the delivered dose is closer to the planned dose with real-time 3D IGRT than without real-time 3D IGRT. In addition, compromised target dose coverage and variable normal tissue doses were found without real-time 3D IGRT. The geometric accuracy results with real-time 3D IGRT had a mean error of <0.5 mm and a standard deviation of <1.1 mm. Numerous additional articles exist that describe real-time 3D IGRT methods using standard-equipped radiation therapy systems that could also be clinically implemented. CONCLUSIONS Multiple clinical implementations of real-time 3D IGRT on standard-equipped cancer radiation therapy systems have been demonstrated. Many more approaches that could be implemented were identified. These solutions provide a pathway for the broader adoption of methods to make radiation therapy more accurate, impacting tumor and normal tissue dose, margins, and ultimately patient outcomes.
Collapse
Affiliation(s)
- Paul J Keall
- ACRF Image X Institute, University of Sydney, Sydney, Australia.
| | | | - Ricky O'Brien
- ACRF Image X Institute, University of Sydney, Sydney, Australia
| | - Pengpeng Zhang
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Laura Happersett
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jenny Bertholet
- Joint Department of Physics, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Per R Poulsen
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|