1
|
Roncali L, Hindré F, Samarut E, Lacoeuille F, Rousseau A, Lemée JM, Garcion E, Chérel M. Current landscape and future directions of targeted-alpha-therapy for glioblastoma treatment. Theranostics 2025; 15:4861-4889. [PMID: 40303349 PMCID: PMC12036880 DOI: 10.7150/thno.106081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/02/2025] [Indexed: 05/02/2025] Open
Abstract
Glioblastoma (GB) is the most aggressive malignancy of the central nervous system. Despite two decades of intensive research since the establishment of the standard of care, emerging strategies have yet to produce consistent satisfactory outcomes. Because of its specific localisation and intricate characteristics, GB is a uniquely regulated solid tumour with a strong resistance to therapy. Advances in targeted radionuclide therapy (TRT), particularly with the introduction of a-emitting radionuclides, have unveiled potential avenues for the management of GB. Recent preclinical and clinical studies underscored promising advancements for targeted-α-therapy (TAT), but these therapeutic approaches exhibit a vast design heterogeneity, encompassing diverse radionuclides, vectors, target molecules, and administration modalities. This review seeks to critically assess the therapeutic landscape of GB through the perspective of TAT. Here, the focus is made on the advancements and limitations of in vivo explorations, pilot studies, and clinical trials, to determine the best directions for future investigations. In doing so, we hope to identify existing challenges and draw insights that might pave the way towards a more effective therapeutic approach.
Collapse
Affiliation(s)
- Loris Roncali
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela; E-15782 Santiago de Compostela, Spain
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
| | - François Hindré
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- PRIMEX (Experimental Imagery and Radiobiology Platform), University of Angers, SFR 4208; F-49000 Angers, France
| | - Edouard Samarut
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
- Department of Neurosurgery & Neurotraumatology, University Hospital of Nantes; F-44093 Nantes, France
| | - Franck Lacoeuille
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Nuclear Medicine, University Hospital of Angers; F-49000 Angers, France
| | - Audrey Rousseau
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Pathology, University Hospital of Angers; F-49000 Angers, France
| | - Jean-Michel Lemée
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Neurosurgery, University Hospital of Angers; F-49000 Angers, France
| | - Emmanuel Garcion
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- PACEM (Platform of Cellular and Molecular Analysis), University of Angers, SFR 4208; F-49000 Angers, France
| | - Michel Chérel
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
- Institut de Cancérologie de l'Ouest, Department of Nuclear Medicine; F-44160 Saint-Herblain, France
| |
Collapse
|
2
|
Sarikaya I. Radionuclide treatments of cancer: molecular mechanisms, biological responses, histopathological changes, and role of PET imaging. Nucl Med Commun 2025; 46:193-203. [PMID: 39654504 DOI: 10.1097/mnm.0000000000001941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Radiation treatments [radiotherapy and radionuclide treatments (RNTs)] are one of the main and effective treatment modalities of cancer. Globally, the number of cancer patients treated with radionuclides are much less as compared to number of radiotherapy cases but with the development of new radiotracers, most notably 177 Lu and 225 Ac-labeled prostate-specific membrane antigen ligands, and 223 Ra-dichloride for prostate cancer and 177 Lu-somatostatin analogs for neuroendocrine tumors, there is a significant rise in RNTs in the last decade. As therapeutic applications of nuclear medicine is on the rise, the aim of this review is to summarize biological responses to radiation treatments and molecular mechanisms of radiation-induced cell death (e.g. ionization, DNA damages such as double-strand breaks, DNA repair mechanisms, types of cell deaths such as apoptosis, necrosis, and immunogenic cell death), histopathological changes with radiation treatments, and role of PET imaging in RNTs as part of radionuclide theranostics for selecting and planning patients for RNTs, dosimetry, predicting and assessing response to RNTs, predicting toxicities, and other possible PET findings which may be seen after RNTs such as activation of immune system.
Collapse
Affiliation(s)
- Ismet Sarikaya
- Department of Nuclear Medicine, Faculty of Medicine, Kirklareli University, Kirklareli, Turkey
| |
Collapse
|
3
|
Conte M, Tomaciello M, De Feo MS, Frantellizzi V, Marampon F, De Cristofaro F, De Vincentis G, Filippi L. The Tight Relationship Between the Tumoral Microenvironment and Radium-223. Biomedicines 2025; 13:456. [PMID: 40002869 PMCID: PMC11853176 DOI: 10.3390/biomedicines13020456] [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: 01/19/2025] [Revised: 02/04/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Radium-223 (223Ra) was the first radioactive isotope approved for treating castration-resistant prostate cancer (CRPC) with symptomatic bone metastases without visceral metastatic disease. To better understand the action of 223Ra, its role in the tumor microenvironment represents a crucial aspect. A literature search was conducted using the PubMed/MEDLINE database and studies regarding the relationship between 223Ra and the tumoral microenvironment were considered. The tumoral microenvironment is a complex setting in which complex interactions between cells and molecules occur. Radium-223, as an alpha-emitter, induces double-stranded DNA breaks; to potentiate this effect, it could be used in patients with genetic instability but also in combination with therapies which inhibit DNA repair, modulate the immune response, or control tumor growth. In conclusion, a few studies have taken into consideration the tumoral microenvironment in association with 223Ra. However, its understanding is a priority to better comprehend how to effectively exploit 223Ra and its action mechanism.
Collapse
Affiliation(s)
- Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, “Sapienza” University of Rome, 00161 Rome, Italy; (M.C.); (M.T.); (M.S.D.F.); (V.F.); (F.M.); (F.D.C.); (G.D.V.)
| | - Miriam Tomaciello
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, “Sapienza” University of Rome, 00161 Rome, Italy; (M.C.); (M.T.); (M.S.D.F.); (V.F.); (F.M.); (F.D.C.); (G.D.V.)
| | - Maria Silvia De Feo
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, “Sapienza” University of Rome, 00161 Rome, Italy; (M.C.); (M.T.); (M.S.D.F.); (V.F.); (F.M.); (F.D.C.); (G.D.V.)
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, “Sapienza” University of Rome, 00161 Rome, Italy; (M.C.); (M.T.); (M.S.D.F.); (V.F.); (F.M.); (F.D.C.); (G.D.V.)
| | - Francesco Marampon
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, “Sapienza” University of Rome, 00161 Rome, Italy; (M.C.); (M.T.); (M.S.D.F.); (V.F.); (F.M.); (F.D.C.); (G.D.V.)
| | - Flaminia De Cristofaro
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, “Sapienza” University of Rome, 00161 Rome, Italy; (M.C.); (M.T.); (M.S.D.F.); (V.F.); (F.M.); (F.D.C.); (G.D.V.)
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, “Sapienza” University of Rome, 00161 Rome, Italy; (M.C.); (M.T.); (M.S.D.F.); (V.F.); (F.M.); (F.D.C.); (G.D.V.)
| | - Luca Filippi
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
4
|
Hiraga T. Immune microenvironment of cancer bone metastasis. Bone 2025; 191:117328. [PMID: 39549899 DOI: 10.1016/j.bone.2024.117328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024]
Abstract
Bone is a common and frequent site of metastasis in cancer patients, leading to a significant reduction in quality of life and increased mortality. Bone marrow, the primary site of hematopoiesis, also serves as both a primary and secondary lymphoid organ. It harbors and supports a diverse array of immune cells, thereby creating a distinct immune microenvironment. These immune cells engage in a range of activities, including anti-tumor, pro-tumor, or a combination of both, which influence the development and progression of bone metastases. Rapid advances in cancer immunotherapy have underscored its potential to eradicate bone metastases. However, clinical outcomes have not yet met expectations. To improve the efficacy of immunotherapy, it is crucial to gain a comprehensive and in-depth understanding of the immune microenvironment within bone metastases. This review provides an overview of the current understanding of the role of different immune cells, their anti-tumor and pro-tumor activities, and their overall contribution to bone metastasis.
Collapse
Affiliation(s)
- Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, Shiojiri, Nagano, Japan.
| |
Collapse
|
5
|
Kostopoulos N, Costabile F, Krimitza E, Beghi S, Goia D, Perales-Linares R, Thyfronitis G, LaRiviere MJ, Chong EA, Schuster SJ, Maity A, Koumenis C, Plastaras JP, Facciabene A. Local radiation enhances systemic CAR T-cell efficacy by augmenting antigen crosspresentation and T-cell infiltration. Blood Adv 2024; 8:6308-6320. [PMID: 39213422 DOI: 10.1182/bloodadvances.2024012599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/19/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
ABSTRACT Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 (CART-19) represents a significant advance in the treatment of patients with relapsed or refractory CD19+ B-cell lymphomas. However, a significant portion of patients either relapse or fail to respond. Moreover, many patients have symptomatic disease, requiring bridging radiation therapy (RT) during the period of CAR T-cell manufacturing. To investigate the impact of 1 to 2 fractions of low-dose RT on CART-19 treatment response, we developed a mouse model using A20 lymphoma cells for CART-19 therapy. We found that low-dose fractionated RT had a positive effect on generating abscopal systemic antitumor responses beyond the irradiated site. The combination of RT with CART-19 therapy resulted in additive effects on tumor growth in irradiated masses. Notably, a significant additional increase in antitumor effect was observed in nonirradiated tumors. Mechanistically, our results validate activation of the cyclic guanosine adenosine synthetase/stimulator of interferon genes pathway, tumor-associated antigen crosspriming, and elicitation of epitope spreading. Collectively, our findings suggest that RT may serve as an optimal priming and bridging modality for CAR T-cell therapy, overcoming treatment resistance and improving clinical outcomes in patients with CD19+ hematologic malignancies.
Collapse
Affiliation(s)
- Nektarios Kostopoulos
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| | - Francesca Costabile
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Elisavet Krimitza
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Silvia Beghi
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Denisa Goia
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Renzo Perales-Linares
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - George Thyfronitis
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| | - Michael J LaRiviere
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Elise A Chong
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Stephen J Schuster
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Amit Maity
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - John P Plastaras
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Andrea Facciabene
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA
- The Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
6
|
Xue E, Bracken-Clarke D, Krause H, Adeyelu T, Evans MG, Akbulut D, Quezado M, Gandhi N, Farrell A, Soares HP, Lou E, Phan M, Patel R, Vanderwalde AM, Elliott A, Steuer CE, Saba NF, Lubin DJ, London NR, Gulley JL, Floudas CS. Characterization of Somatostatin Receptor 2 Gene Expression and Immune Landscape in Sinonasal Malignancies. Cancers (Basel) 2024; 16:3931. [PMID: 39682120 DOI: 10.3390/cancers16233931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Olfactory neuroblastoma (ONB), sinonasal undifferentiated carcinoma (SNUC), and sinonasal neuroendocrine carcinoma (SNEC) are rare malignancies arising from the sinonasal tract with limited therapeutic options. The expression of the somatostatin receptor 2 gene (SSTR2), which is expressed in other neuroendocrine neoplasms and is therapeutically actionable, has been reported in these tumors. Here, we analyzed SSTR2 gene expression and its associations with genomic features, established biomarkers predicting of immune response, and the tumor immune microenvironment in a cohort of ONB, SNUC, and SNEC tumor samples (26, 13, and 8 samples, respectively) from a real-world database. SSTR2 gene expression was high in neural-type ONB and low in basal-type ONB and in most of the SNUC and SNEC cases; there was no difference in expression between primary and metastatic tumors. The T cell-inflamed (TCI) score analysis classified 38.5% of SNUC cases as T cell-inflamed compared to only 3.9% of ONB and 0% of SNEC cases; 26.9% of ONB cases were classified as intermediate TCI; and SNEC had the lowest relative immune cell infiltration by deconvolution. In high SSTR2-expressing ONB, there was a higher proportion of infiltrating of Natural Killer cells and dendritic cells by deconvolution. Additionally, high SSTR2-expressing ONB was enriched for proliferation pathways, including E2F and Myc targets and G2M checkpoints. In conclusion, our findings delineate significant differences between these three types of sinonasal malignancies that were examined. In ONB, relative to SNUC and SNEC, the SSTR2 expression profile, combined with its immune profiles, indicates potential novel therapeutic strategies and combinations for this unmet clinical need. Conversely, the inflammatory microenvironment of SNUC may be targetable using immuno-oncologic therapies.
Collapse
Affiliation(s)
- Elisabetta Xue
- Center for Immuno-Oncology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| | - Dara Bracken-Clarke
- Center for Immuno-Oncology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| | | | | | | | - Dilara Akbulut
- Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| | - Martha Quezado
- Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| | | | | | - Heloisa P Soares
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Emil Lou
- Department of Medicine, Division of Hematology Oncology and Transplant, University of Minnesota, Minneapolis, MN 55414, USA
| | - Minh Phan
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Rusha Patel
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
| | | | | | - Conor E Steuer
- Department of Hematology-Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30308, USA
| | - Nabil F Saba
- Department of Hematology-Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30308, USA
| | - Daniel J Lubin
- Department of Hematology-Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30308, USA
| | - Nyall R London
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Sinonasal and Skull Base Tumor Program, Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD 20892, USA
| | - James L Gulley
- Center for Immuno-Oncology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| | - Charalampos S Floudas
- Center for Immuno-Oncology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| |
Collapse
|
7
|
Lunj S, Smith TAD, Reeves KJ, Currell F, Honeychurch J, Hoskin P, Choudhury A. Immune effects of α and β radionuclides in metastatic prostate cancer. Nat Rev Urol 2024; 21:651-661. [PMID: 39192074 DOI: 10.1038/s41585-024-00924-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/29/2024]
Abstract
External beam radiotherapy is used for radical treatment of organ-confined prostate cancer and to treat lesions in metastatic disease whereas molecular radiotherapy with labelled prostate-specific membrane antigen ligands and radium-223 (223Ra) is indicated for metastatic prostate cancer and has demonstrated substantial improvements in symptom control and overall survival compared with standard-of-care treatment. Prostate cancer is considered an immunologically cold tumour, so limited studies investigating the treatment-induced effects on the immune response have been completed. However, emerging data support the idea that radiotherapy induces an immune response in prostate cancer, but whether the response is an antitumour or pro-tumour response is dependent on the radiotherapy regime and is also cell-line dependent. In vitro data demonstrate that single-dose radiotherapy regimes induce a greater immune-suppressive profile than fractionated regimes; less is known about the immune response induced by molecular radiotherapy agents, but evidence suggests that these agents might induce an immune-suppressive systemic immune response, indicated by increased expression of inhibitory checkpoint molecules such as programmed cell death 1 ligand 1 and 2, and that these changes could be associated with clinical response. Different radiotherapy modalities can induce distinct immune profiles, which can either activate or suppress immune-mediated tumour killing and the current preclinical models used for prostate cancer research are not yet optimal for studying the complexity of the radiotherapy-induced immune response.
Collapse
Affiliation(s)
- Sapna Lunj
- Division of Cancer Sciences, Oglesby Cancer Research Building, University of Manchester, Manchester, UK.
| | - Tim Andrew Davies Smith
- Nuclear Futures Institute, School of Computer Science and Engineering, Bangor University, Bangor, UK
| | - Kimberley Jayne Reeves
- Division of Cancer Sciences, Paterson Building, University of Manchester, Manchester, UK
| | - Fred Currell
- The Dalton Cumbria Facility and the Department of Chemistry, University of Manchester, Manchester, UK
| | - Jamie Honeychurch
- Division of Cancer Sciences, Paterson Building, University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Peter Hoskin
- Division of Cancer Sciences, Paterson Building, University of Manchester, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, Oglesby Cancer Research Building, University of Manchester, Manchester, UK
- The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
8
|
Shea AG, Idrissou MB, Torres AI, Chen T, Hernandez R, Morris ZS, Sodji QH. Immunological effects of radiopharmaceutical therapy. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1331364. [PMID: 39355211 PMCID: PMC11440989 DOI: 10.3389/fnume.2024.1331364] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/14/2024] [Indexed: 10/03/2024]
Abstract
Radiation therapy (RT) is a pillar of cancer therapy used by more than half of all cancer patients. Clinically, RT is mostly delivered as external beam radiation therapy (EBRT). However, the scope of EBRT is limited in the metastatic setting, where all sites of disease need to be irradiated. Such a limitation is attributed to radiation-induced toxicities, for example on bone marrow and hematologic toxicities, resulting from a large EBRT field. Radiopharmaceutical therapy (RPT) has emerged as an alternative to EBRT for the irradiation of all sites of metastatic disease. While RPT can reduce tumor burden, it can also impact the immune system and anti-tumor immunity. Understanding these effects is crucial for predicting and managing treatment-related hematological toxicities and optimizing their integration with other therapeutic modalities, such as immunotherapies. Here, we review the immunomodulatory effects of α- and β-particle emitter-based RPT on various immune cell lines, such as CD8+ and CD4+ T cells, natural killer (NK) cells, and regulatory T (Treg) cells. We briefly discuss Auger electron-emitter (AEE)-based RPT, and finally, we highlight the combination of RPT with immune checkpoint inhibitors, which may offer potential therapeutic synergies for patients with metastatic cancers.
Collapse
Affiliation(s)
- Amanda G. Shea
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Malick Bio Idrissou
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Ana Isabel Torres
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Tessa Chen
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Reiner Hernandez
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Quaovi H. Sodji
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
9
|
Zhou L, Liu Y, Wu Y, Yang X, Spring Kong FM, Lu Y, Xue J. Low-dose radiation therapy mobilizes antitumor immunity: New findings and future perspectives. Int J Cancer 2024; 154:1143-1157. [PMID: 38059788 DOI: 10.1002/ijc.34801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023]
Abstract
Radiotherapy has unique immunostimulatory and immunosuppressive effects. Although high-dose radiotherapy has been found to have systemic antitumor effects, clinically significant abscopal effects were uncommon on the basis of irradiating single lesion. Low-dose radiation therapy (LDRT) emerges as a novel approach to enhance the antitumor immune response due to its role as a leverage to reshape the tumor immune microenvironment (TIME). In this article, from bench to bedside, we reviewed the possible immunomodulatory role of LDRT on TIME and systemic tumor immune environment, and outlined preclinical evidence and clinical application. We also discussed the current challenges when LDRT is used as a combination therapy, including the optimal dose, fraction, frequency, and combination of drugs. The advantage of low toxicity makes LDRT potential to be applied in multiple lesions to amplify antitumor immune response in polymetastatic disease, and its intersection with other disciplines might also make it a direction for radiotherapy-combined modalities.
Collapse
Affiliation(s)
- Laiyan Zhou
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Disaster Medical Center, Sichuan University, Chengdu, China
| | - Yuanxin Liu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanjun Wu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Yang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feng-Ming Spring Kong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Yang M, Liu H, Lou J, Zhang J, Zuo C, Zhu M, Zhang X, Yin Y, Zhang Y, Qin S, Zhang H, Fan X, Dang Y, Cheng C, Cheng Z, Yu F. Alpha-Emitter Radium-223 Induces STING-Dependent Pyroptosis to Trigger Robust Antitumor Immunity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307448. [PMID: 37845027 DOI: 10.1002/smll.202307448] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/25/2023] [Indexed: 10/18/2023]
Abstract
Radium-223 (223 Ra) is the first-in-class alpha-emitter to mediate tumor eradication, which is commonly thought to kill tumor cells by directly cleaving double-strand DNA. However, the immunogenic characteristics and cell death modalities triggered by 223 Ra remain unclear. Here, it is reported that the 223 Ra irradiation induces the pro-inflammatory damage-associated molecular patterns including calreticulin, HMGB1, and HSP70, hallmarks of tumor immunogenicity. Moreover, therapeutic 223 Ra retards tumor progression by triggering pyroptosis, an immunogenic cell death. Mechanically, 223 Ra-induced DNA damage leads to the activation of stimulator of interferon genes (STING)-mediated DNA sensing pathway, which is critical for NLRP3 inflammasome-dependent pyroptosis and subsequent DCs maturation as well as T cell activation. These findings establish an essential role of STING in mediating alpha-emitter 223 Ra-induced antitumor immunity, which provides the basis for the development of novel cancer therapeutic strategies and combinatory therapy.
Collapse
Affiliation(s)
- Mengdie Yang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, China
| | - Haipeng Liu
- Clinical Translation Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jingjing Lou
- Department of Nuclear Medicine, Pudong Medical Center, Fudan University, Shanghai, 201399, China
| | - Jiajia Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, China
| | - Changjing Zuo
- Department of Nuclear Medicine, the First Affiliated Hospital of Navy Medical University (Changhai Hospital), Shanghai, 200433, China
| | - Mengqin Zhu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xiaoyi Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yuzhen Yin
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yu Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, China
| | - Shanshan Qin
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, China
| | - Han Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xin Fan
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yifang Dang
- Clinical Translation Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Chao Cheng
- Department of Nuclear Medicine, the First Affiliated Hospital of Navy Medical University (Changhai Hospital), Shanghai, 200433, China
| | - Zhen Cheng
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China
| | - Fei Yu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, China
| |
Collapse
|
11
|
Barsegian V, Möckel D, Buehler S, Müller SP, Kreissl MC, Ostheim P, Horn PA, Lindemann M. Lymphocyte Function at Baseline Could Be a New Predictor of Tumor Burden following Six Cycles of Radium-223 Therapy in Patients with Metastasized, Castration-Resistant Prostate Cancer. Cancers (Basel) 2024; 16:886. [PMID: 38473247 DOI: 10.3390/cancers16050886] [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: 01/24/2024] [Revised: 02/09/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Previous data indicate that one cycle of treatment with radium-223 (223Ra) did not significantly impair lymphocyte function in patients with metastasized, castration-resistant prostate cancer. The aim of the current study was to assess in 21 patients whether six cycles of this therapy had an effect on lymphocyte proliferation and interferon-γ and interleukin (IL)-10 ELISpot results. Lymphocyte proliferation after stimulation with microbial antigens and the production of interferon-γ continuously decreased after six cycles of radionuclide therapy, reaching statistical significance (p < 0.05) at months 1, 2, 4, and/or 6 after therapy. One month after the last cycle of therapy, 67% of patients showed a decrease in tumor burden. The tumor burden correlated negatively with IL-10 secretion at baseline, e.g., after stimulation with tetanus antigen (p < 0.0001, r = -0.82). As determined by receiver operating characteristic (ROC) curve analysis, tetanus-specific IL-10 spots at baseline had the highest predictive value (p = 0.005) for tumor burden at month 6, with an area under the curve (AUC) of 0.90 (sensitivity 100%, specificity 78%). In conclusion, we observed an additive effect of treatment with 223Ra on immune function and found that IL-10 secretion at baseline predicted tumor burden at month 6 after treatment.
Collapse
Affiliation(s)
- Vahé Barsegian
- Institute of Nuclear Medicine, Helios Kliniken, 19055 Schwerin, Germany
| | - Daniel Möckel
- Institute of Nuclear Medicine, Helios Kliniken, 19055 Schwerin, Germany
| | - Sebastian Buehler
- Institute of Nuclear Medicine, Helios Kliniken, 19055 Schwerin, Germany
| | - Stefan P Müller
- Department of Nuclear Medicine, University Hospital, 45147 Essen, Germany
| | - Michael C Kreissl
- Division of Nuclear Medicine, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
| | | | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital, 19055 Essen, Germany
| | - Monika Lindemann
- Institute for Transfusion Medicine, University Hospital, 19055 Essen, Germany
| |
Collapse
|
12
|
Karlsson J, Schatz CA, Wengner AM, Hammer S, Scholz A, Cuthbertson A, Wagner V, Hennekes H, Jardine V, Hagemann UB. Targeted thorium-227 conjugates as treatment options in oncology. Front Med (Lausanne) 2023; 9:1071086. [PMID: 36726355 PMCID: PMC9885765 DOI: 10.3389/fmed.2022.1071086] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/15/2022] [Indexed: 01/10/2023] Open
Abstract
Targeted alpha therapy (TAT) is a promising approach for addressing unmet needs in oncology. Inherent properties make α-emitting radionuclides well suited to cancer therapy, including high linear energy transfer (LET), penetration range of 2-10 cell layers, induction of complex double-stranded DNA breaks, and immune-stimulatory effects. Several alpha radionuclides, including radium-223 (223Ra), actinium-225 (225Ac), and thorium-227 (227Th), have been investigated. Conjugation of tumor targeting modalities, such as antibodies and small molecules, with a chelator moiety and subsequent radiolabeling with α-emitters enables specific delivery of cytotoxic payloads to different tumor types. 223Ra dichloride, approved for the treatment of patients with metastatic castration-resistant prostate cancer (mCRPC) with bone-metastatic disease and no visceral metastasis, is the only approved and commercialized alpha therapy. However, 223Ra dichloride cannot currently be complexed to targeting moieties. In contrast to 223Ra, 227Th may be readily chelated, which allows radiolabeling of tumor targeting moieties to produce targeted thorium conjugates (TTCs), facilitating delivery to a broad range of tumors. TTCs have shown promise in pre-clinical studies across a range of tumor-cell expressing antigens. A clinical study in hematological malignancy targeting CD22 has demonstrated early signs of activity. Furthermore, pre-clinical studies show additive or synergistic effects when TTCs are combined with established anti-cancer therapies, for example androgen receptor inhibitors (ARI), DNA damage response inhibitors such as poly (ADP)-ribose polymerase inhibitors or ataxia telangiectasia and Rad3-related kinase inhibitors, as well as immune checkpoint inhibitors.
Collapse
|
13
|
Sun Q, Li J, Ding Z, Liu Z. Radiopharmaceuticals heat anti-tumor immunity. Theranostics 2023; 13:767-786. [PMID: 36632233 PMCID: PMC9830438 DOI: 10.7150/thno.79806] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 01/06/2023] Open
Abstract
Radiopharmaceutical therapy (RPT) has proven to be an effective cancer treatment with minimal toxicity. With several RPT agents approved by FDA, the remarkable potential of this therapy is now being recognized, and the anti-tumor immunity induced by RPT is beginning to be noticed. This review evaluates the potential of RPT for immune activation, including promoting the release of danger associated-molecular pattern molecules that recruit inflammatory cells into the tumor microenvironment, and activating antigen-presenting cells and cytotoxic T cells. We also discuss the progress of combining RPT with immunotherapy to increase efficacy.
Collapse
Affiliation(s)
- Qi Sun
- Peking University-Tsinghua University Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jiyuan Li
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | | | - Zhibo Liu
- Peking University-Tsinghua University Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China,Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China,Changping Laboratory, Beijing 102206, China,✉ Corresponding author: Zhibo Liu:
| |
Collapse
|
14
|
Kerr CP, Grudzinski JJ, Nguyen TP, Hernandez R, Weichert JP, Morris ZS. Developments in Combining Targeted Radionuclide Therapies and Immunotherapies for Cancer Treatment. Pharmaceutics 2022; 15:128. [PMID: 36678756 PMCID: PMC9865370 DOI: 10.3390/pharmaceutics15010128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
Targeted radionuclide therapy (TRT) and immunotherapy are rapidly growing classes of cancer treatments. Basic, translational, and clinical research are now investigating therapeutic combinations of these agents. In comparison to external beam radiation therapy (EBRT), TRT has the unique advantage of treating all disease sites following intravenous injection and selective tumor uptake and retention-a particularly beneficial property in metastatic disease settings. The therapeutic value of combining radiation therapy with immune checkpoint blockade to treat metastases has been demonstrated in preclinical studies, whereas results of clinical studies have been mixed. Several clinical trials combining TRT and immune checkpoint blockade have been initiated based on preclinical studies combining these with EBRT and/or TRT. Despite the interest in translation of TRT and immunotherapy combinations, many questions remain surrounding the mechanisms of interaction and the optimal approach to clinical implementation of these combinations. This review highlights the mechanisms of interaction between anti-tumor immunity and radiation therapy and the status of basic and translational research and clinical trials investigating combinations of TRT and immunotherapies.
Collapse
Affiliation(s)
- Caroline P. Kerr
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Joseph J. Grudzinski
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Thanh Phuong Nguyen
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Reinier Hernandez
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jamey P. Weichert
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachary S. Morris
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
15
|
Fabian KP, Kowalczyk JT, Reynolds ST, Hodge JW. Dying of Stress: Chemotherapy, Radiotherapy, and Small-Molecule Inhibitors in Immunogenic Cell Death and Immunogenic Modulation. Cells 2022; 11:cells11233826. [PMID: 36497086 PMCID: PMC9737874 DOI: 10.3390/cells11233826] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/11/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Innovative strategies to re-establish the immune-mediated destruction of malignant cells is paramount to the success of anti-cancer therapy. Accumulating evidence suggests that radiotherapy and select chemotherapeutic drugs and small molecule inhibitors induce immunogenic cell stress on tumors that results in improved immune recognition and targeting of the malignant cells. Through immunogenic cell death, which entails the release of antigens and danger signals, and immunogenic modulation, wherein the phenotype of stressed cells is altered to become more susceptible to immune attack, radiotherapies, chemotherapies, and small-molecule inhibitors exert immune-mediated anti-tumor responses. In this review, we discuss the mechanisms of immunogenic cell death and immunogenic modulation and their relevance in the anti-tumor activity of radiotherapies, chemotherapies, and small-molecule inhibitors. Our aim is to feature the immunological aspects of conventional and targeted cancer therapies and highlight how these therapies may be compatible with emerging immunotherapy approaches.
Collapse
|
16
|
Kleinendorst SC, Oosterwijk E, Bussink J, Westdorp H, Konijnenberg MW, Heskamp S. Combining Targeted Radionuclide Therapy and Immune Checkpoint Inhibition for Cancer Treatment. Clin Cancer Res 2022; 28:3652-3657. [PMID: 35471557 PMCID: PMC9433955 DOI: 10.1158/1078-0432.ccr-21-4332] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/03/2022] [Accepted: 04/11/2022] [Indexed: 01/07/2023]
Abstract
The development of immunotherapy, in particular immune checkpoint inhibitors (ICI), has revolutionized cancer treatment in the past decades. However, its efficacy is still limited to subgroups of patients with cancer. Therefore, effective treatment combination strategies are needed. Here, radiotherapy is highly promising, as it can induce immunogenic cell death, triggering the release of pro-inflammatory cytokines, thereby creating an immunogenic phenotype and sensitizing tumors to ICI. Recently, targeted radionuclide therapy (TRT) has attained significant interest for cancer treatment. In this approach, a tumor-targeting radiopharmaceutical is used to specifically deliver a therapeutic radiation dose to all tumor cells, including distant metastatic lesions, while limiting radiation exposure to healthy tissue. However, fundamental differences between TRT and conventional radiotherapy make it impossible to directly extrapolate the biological effects from conventional radiotherapy to TRT. In this review, we present a comprehensive overview of studies investigating the immunomodulatory effects of TRT and the efficacy of combined TRT-ICI treatment. Preclinical studies have evaluated a variety of murine cancer models in which α- or β-emitting radionuclides were directed to a diverse set of targets. In addition, clinical trials are ongoing to assess safety and efficacy of combined TRT-ICI in patients with cancer. Taken together, research indicates that combining TRT and ICI might improve therapeutic response in patients with cancer. Future research has to disclose what the optimal conditions are in terms of dose and treatment schedule to maximize the efficacy of this combined approach.
Collapse
Affiliation(s)
- Simone C. Kleinendorst
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Egbert Oosterwijk
- Department of Urology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Harm Westdorp
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands.,Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mark W. Konijnenberg
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands.,Corresponding Author: Sandra Heskamp, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, the Netherlands. Phone: 243-614-511; E-mail:
| |
Collapse
|
17
|
Kiess AP, Hobbs RF, Bednarz B, Knox SJ, Meredith R, Escorcia FE. ASTRO's Framework for Radiopharmaceutical Therapy Curriculum Development for Trainees. Int J Radiat Oncol Biol Phys 2022; 113:719-726. [PMID: 35367328 DOI: 10.1016/j.ijrobp.2022.03.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 10/18/2022]
Abstract
In 2017, the American Society for Radiation Oncology (ASTRO) board of directors prioritized radiopharmaceutical therapy (RPT) as a leading area for new therapeutic development, and the ASTRO RPT workgroup was created. Herein, the workgroup has developed a framework for RPT curriculum development upon which education leaders can build to integrate this modality into radiation oncology resident education. Through this effort, the workgroup aims to provide a guide to ensure robust training in an emerging therapeutic area within the context of existing radiation oncology training in radiation biology, medical physics, and clinical radiation oncology. The framework first determines the core RPT knowledge required to select patients, prescribe, safely administer, and manage related adverse events. Then, it defines the most important topics for preparing residents for clinical RPT planning and delivery. This framework is designed as a tool to supplement the current training that exists for radiation oncology residents. The final document was approved by the ASTRO board of directors in the fall of 2021.
Collapse
Affiliation(s)
- Ana P Kiess
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland.
| | - Robert F Hobbs
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Bryan Bednarz
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Susan J Knox
- Department of Radiation Oncology, Stanford University Medical Center, Stanford, California
| | - Ruby Meredith
- Department of Radiation Oncology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Freddy E Escorcia
- Molecular Imaging Branch, Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
18
|
Labadie KP, Hamlin DK, Kenoyer A, Daniel SK, Utria AF, Ludwig AD, Kenerson HL, Li L, Sham JG, Chen DL, Orozco JJ, Yeung RS, Orvig C, Li Y, Wilbur DS, Park JO. Glypican-3-Targeted 227Th α-Therapy Reduces Tumor Burden in an Orthotopic Xenograft Murine Model of Hepatocellular Carcinoma. J Nucl Med 2022; 63:1033-1038. [PMID: 34772791 PMCID: PMC9258570 DOI: 10.2967/jnumed.121.262562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/08/2021] [Indexed: 01/03/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a significant cause of morbidity and mortality worldwide, with limited therapeutic options for advanced disease. Targeted α-therapy is an emerging class of targeted cancer therapy in which α-particle-emitting radionuclides, such as 227Th, are delivered specifically to cancer tissue. Glypican-3 (GPC3) is a cell surface glycoprotein highly expressed on HCC. In this study, we describe the development and in vivo efficacy of a 227Th-labeled GPC3-targeting antibody conjugate (227Th-octapa-αGPC3) for treatment of HCC in an orthotopic murine model. Methods: The chelator p-SCN-Bn-H4octapa-NCS (octapa) was conjugated to a GPC3-targeting antibody (αGPC3) for subsequent 227Th radiolabeling (octapa-αGPC3). Conditions were varied to optimize radiolabeling of 227Th. In vitro stability was evaluated by measuring the percentage of protein-bound 227Th by γ-ray spectroscopy. An orthotopic athymic Nu/J murine model using HepG2-Red-FLuc cells was developed. Biodistribution and blood clearance of 227Th-octapa-αGPC3 were evaluated in tumor-bearing mice. The efficacy of 227Th-octapa-αGPC3 was assessed in tumor-bearing animals with serial measurement of serum α-fetoprotein at 23 d after injection. Results: Octapa-conjugated αGPC3 provided up to 70% 227Th labeling yield in 2 h at room temperature. In the presence of ascorbate, at least 97.8% of 227Th was bound to αGPC3-octapa after 14 d in phosphate-buffered saline. In HepG2-Red-FLuc tumor-bearing mice, highly specific GPC3 targeting was observed, with significant 227Th-octapa-αGPC3 accumulation in the tumor over time and minimal accumulation in normal tissue. Twenty-three days after treatment, a significant reduction in tumor burden was observed in mice receiving a 500 kBq/kg dose of 227Th-octapa-αGPC3 by tail-vein injection. No acute off-target toxicity was observed, and no animals died before termination of the study. Conclusion:227Th-octapa-αGPC3 was observed to be stable in vitro; maintain high specificity for GPC3, with favorable biodistribution in vivo; and result in significant antitumor activity without significant acute off-target toxicity in an orthotopic murine model of HCC.
Collapse
Affiliation(s)
- Kevin P. Labadie
- Department of Surgery, University of Washington, Seattle, Washington
| | - Donald K. Hamlin
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - Aimee Kenoyer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sara K. Daniel
- Department of Surgery, University of Washington, Seattle, Washington
| | - Alan F. Utria
- Department of Surgery, University of Washington, Seattle, Washington
| | - Andrew D. Ludwig
- Department of Surgery, University of Washington, Seattle, Washington
| | - Heidi L. Kenerson
- Department of Surgery, University of Washington, Seattle, Washington
| | - Lily Li
- Life Sciences Division, TRIUMF, and Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan G. Sham
- Department of Surgery, University of Washington, Seattle, Washington
| | - Delphine L. Chen
- Department of Radiology, University of Washington, Seattle, Washington
| | - Johnnie J. Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Raymond S. Yeung
- Department of Surgery, University of Washington, Seattle, Washington
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yawen Li
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - D. Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - James O. Park
- Department of Surgery, University of Washington, Seattle, Washington
| |
Collapse
|
19
|
King AP, Lin FI, Escorcia FE. Why bother with alpha particles? Eur J Nucl Med Mol Imaging 2021; 49:7-17. [PMID: 34175980 DOI: 10.1007/s00259-021-05431-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022]
Abstract
The approval of 223RaCl2 for cancer therapy in 2013 has heralded a resurgence of interest in the development of α-particle emitting radiopharmaceuticals. In the last decade, over a dozen α-emitting radiopharmaceuticals have entered clinical trials, spawned by strong preclinical studies. In this article, we explore the potential role of α-particle therapy in cancer treatment. We begin by providing a background for the basic principles of therapy with α-emitters, and we explore recent breakthroughs in therapy with α-emitting radionuclides, including conjugates with small molecules and antibodies. Finally, we discuss some outstanding challenges to the clinical adoption of α-therapies and potential strategies to address them.
Collapse
Affiliation(s)
- A Paden King
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20817, USA
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20817, USA
| | - Frank I Lin
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20817, USA
| | - Freddy E Escorcia
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20817, USA.
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20817, USA.
| |
Collapse
|
20
|
Perrin J, Capitao M, Allard M, Chouin N, Gouard S, Marionneau-Lambot S, Louvet C, Donnadieu E, Bruchertseifer F, Morgenstern A, Chérel M, Gaschet J, Guilloux Y. Targeted alpha particle therapy remodels the tumor microenvironment and improves efficacy of immunotherapy. Int J Radiat Oncol Biol Phys 2021; 112:790-801. [PMID: 34699930 DOI: 10.1016/j.ijrobp.2021.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/01/2021] [Accepted: 10/15/2021] [Indexed: 11/18/2022]
Abstract
PURPOSE Tumor microenvironment (TME) can severely impair immunotherapy efficacy by repressing the immune system. In a Multiple Myeloma (MM) murine model, we investigated the impact of Targeted alpha-particle therapy (TAT) on the immune TME. TAT was combined with an adoptive cell transfer of CD8 T-cells (ACT), and the mechanisms of action of this combination were assessed at the tumor site. METHODS This combination treatment was conducted in a syngeneic MM murine model grafted subcutaneously. TAT was delivered by i.v. injection of a bismuth-213 radiolabelled anti-CD138 antibody. To strengthen anti-tumor immune response, TAT was combined with an ACT of tumor specific CD8+ OT-1 T-cells. The tumors were collected and the immune TME analyzed by flow cytometry, immunohistochemistry and ex vivo T-cell motility assay on tumor slices. The chemokine and cytokine productions were also assessed by RT-qPCR. RESULTS Tumor specific CD8+ OT-1 T-cells infiltrated the tumors after ACT. However only treatment with TAT resulted in regulatory CD4 T-cell drop and transient increased production of IL-2, CCL-5 and IFNγ within the tumor. Moreover, OT-1 T-cell recruitment and motility were increased on tumor slices from TAT-treated mice as observed by ex vivo time lapse, contributing to a more homogeneous distribution of OT-1 T-cells in the tumor. Subsequently, the tumor cells increased PD-L1 expression, anti-tumor cytokine production decreased and OT-1 T-cells overexpressed exhaustion markers, suggesting an exhaustion of the immune response. CONCLUSION Combining TAT and ACT seems to transiently remodel the cold TME, improving ACT efficiency. The immune response then leads to the establishment of other tumor cell resistance mechanisms.
Collapse
Affiliation(s)
- Justine Perrin
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - Marisa Capitao
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - Mathilde Allard
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - Nicolas Chouin
- CRCINA, INSERM, CNRS, ONIRIS, Université de Nantes, Université d'Angers, Nantes, France
| | - Sebastien Gouard
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | | | - Cédric Louvet
- Centre de Recherche en Transplantation et Immunologie, INSERM, Université de Nantes, Nantes, France
| | - Emmanuel Donnadieu
- INSERM, U1016, Institut Cochin, CNRS, UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | | | - Michel Chérel
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France; Nuclear Medicine Unit, ICO Cancer Center Gauducheau, Saint Herblain, France
| | - Joëlle Gaschet
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - Yannick Guilloux
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.
| |
Collapse
|
21
|
Lejeune P, Cruciani V, Berg-Larsen A, Schlicker A, Mobergslien A, Bartnitzky L, Berndt S, Zitzmann-Kolbe S, Kamfenkel C, Stargard S, Hammer S, Jørgensen JS, Jackerott M, Nielsen CH, Schatz CA, Hennekes H, Karlsson J, Cuthbertson AS, Mumberg D, Hagemann UB. Immunostimulatory effects of targeted thorium-227 conjugates as single agent and in combination with anti-PD-L1 therapy. J Immunother Cancer 2021; 9:jitc-2021-002387. [PMID: 34615703 PMCID: PMC8496392 DOI: 10.1136/jitc-2021-002387] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2021] [Indexed: 12/25/2022] Open
Abstract
Background Targeted thorium-227 conjugates (TTCs) are an emerging class of targeted alpha therapies (TATs). Their unique mode of action (MoA) is the induction of difficult-to-repair clustered DNA double-strand breaks. However, thus far, their effects on the immune system are largely unknown. Here, we investigated the immunostimulatory effects of the mesothelin-targeted thorium-227 conjugate (MSLN-TTC) in vitro and in vivo in monotherapy and in combination with an inhibitor of the immune checkpoint programmed death receptor ligand 1 (PD-L1) in immunocompetent mice. Methods The murine cell line MC38 was transfected with the human gene encoding for MSLN (hMSLN) to enable binding of the non-cross-reactive MSLN-TTC. The immunostimulatory effects of MSLN-TTC were studied in vitro on human cancer cell lines and MC38-hMSLN cells. The efficacy and MoA of MSLN-TTC were studied in vivo as monotherapy or in combination with anti-PD-L1 in MC38-hMSLN tumor-bearing immunocompetent C57BL/6 mice. Experiments were supported by RNA sequencing, flow cytometry, immunohistochemistry, mesoscale, and TaqMan PCR analyses to study the underlying immunostimulatory effects. In vivo depletion of CD8+ T cells and studies with Rag2/Il2Rg double knockout C57BL/6 mice were conducted to investigate the importance of immune cells to the efficacy of MSLN-TTC. Results MSLN-TTC treatment induced upregulation of DNA sensing pathway transcripts (IL-6, CCL20, CXCL10, and stimulator of interferon genes (STING)-related genes) in vitro as determined by RNASeq analysis. The results, including phospho-STING activation, were confirmed on the protein level. Danger-associated molecular pattern molecules were upregulated in parallel, leading to dendritic cell (DC) activation in vitro. MSLN-TTC showed strong antitumor activity (T:C 0.38, p<0.05) as a single agent in human MSLN-expressing MC38 tumor-bearing immunocompetent mice. Combining MSLN-TTC with anti-PD-L1 further enhanced the efficacy (T:C 0.08, p<0.001) as evidenced by the increased number of tumor-free surviving animals. MSLN-TTC monotherapy caused migration of CD103+ cDC1 DCs and infiltration of CD8+ T cells into tumors, which was enhanced on combination with anti-PD-L1. Intriguingly, CD8+ T-cell depletion decreased antitumor efficacy. Conclusions These in vitro and in vivo data on MSLN-TTC demonstrate that the MoA of TTCs involves activation of the immune system. The findings are of relevance for other targeted radiotherapies and may guide clinical combination strategies.
Collapse
|
22
|
Yang Y, Lima RC, Gemini-Piperni S, Alencar LMR, Santos-Oliveira R. Graphene Quantum Dots for Molecular Radiotherapy: Radiolabeled Graphene Quantum Dots with Radium ( 223Ra) Showed Potent Effect Against Bone Cancer. J Biomed Nanotechnol 2021; 17:1858-1865. [PMID: 34688331 DOI: 10.1166/jbn.2021.3150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The necessity of new drugs with special attention for the therapy of cancer is increasing each day. Despite their properties, alpha therapeutic radiopharmaceuticals, especially based on the use of radium (223Ra) are good choices, due to the highest and differential cytotoxicity, low adverse effects, and higher bioaccumulation on tumor sites. The use of graphene quantum dots as the carrier for 223Ra is a promising approach since graphene quantum dots has low toxicity, high biocompatibility, and adequate size for tumor penetration. In this study, we developed, characterized, radiolabeled with 223Ra, and evaluated in vitro and in vivo graphene quantum dots radiolabeled with radium (223Ra) for bone cancer. The results showed that 223Ra is incorporated into the graphene quantum dot following the Fajans-Paneth-Hahn Law. The cell viability showed a potent effect on osteosarcoma cells (MG63 and SAOS2) but a lower effect in normal fibroblast cells (hFB), corroborating the preferential targeting. Also, the results showed a more prominent effect on MG63 than SAOS2 cells, corroborating the targeting for more undifferentiated cells. The in vivo results demonstrated a renal excretion, associated with fecal excretion and accumulation in bone. The results corroborate the efficacy of 223RaGQDs and open new perspectives for the use of use 223RaGQDs, in several other diseases.
Collapse
Affiliation(s)
- Yang Yang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Ruana Cardoso Lima
- Federal University of Maranhão, Laboratory of Biophysics and Nanosystems, São Luis, 65080-805, Brazil
| | - Sara Gemini-Piperni
- 3Universidade do Grande Rio, Laboratório de Estudos ósseos e Biologia Celular, Rio de Janeiro, 25071202, Brazil
| | | | - Ralph Santos-Oliveira
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Laboratory of Nanoradiopharmaceticals and Synthesis of Novel Radiopharmaceuticals, Rio de Janeiro, 21941906, Brazil
| |
Collapse
|
23
|
Fabian KP, Wolfson B, Hodge JW. From Immunogenic Cell Death to Immunogenic Modulation: Select Chemotherapy Regimens Induce a Spectrum of Immune-Enhancing Activities in the Tumor Microenvironment. Front Oncol 2021; 11:728018. [PMID: 34497771 PMCID: PMC8419351 DOI: 10.3389/fonc.2021.728018] [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: 06/21/2021] [Accepted: 07/29/2021] [Indexed: 12/22/2022] Open
Abstract
Cancer treatment has rapidly entered the age of immunotherapy, and it is becoming clear that the effective therapy of established tumors necessitates rational multi-combination immunotherapy strategies. But even in the advent of immunotherapy, the clinical role of standard-of-care chemotherapy regimens still remains significant and may be complementary to emerging immunotherapeutic approaches. Depending on dose, schedule, and agent, chemotherapy can induce immunogenic cell death, resulting in the release of tumor antigens to stimulate an immune response, or immunogenic modulation, sensitizing surviving tumor cells to immune cell killing. While these have been previously defined as distinct processes, in this review we examine the published mechanisms supporting both immunogenic cell death and immunogenic modulation and propose they be reclassified as similar effects termed "immunogenic cell stress." Treatment-induced immunogenic cell stress is an important result of cytotoxic chemotherapy and future research should consider immunogenic cell stress as a whole rather than just immunogenic cell death or immunogenic modulation. Cancer treatment strategies should be designed specifically to take advantage of these effects in combination immunotherapy, and novel chemotherapy regimens should be designed and investigated to potentially induce all aspects of immunogenic cell stress.
Collapse
Affiliation(s)
| | | | - James W. Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
24
|
Creemers JHA, van der Doelen MJ, van Wilpe S, Hermsen R, Duiveman-de Boer T, Somford DM, Janssen MJR, Sedelaar JPM, Mehra N, Textor J, Westdorp H. Immunophenotyping Reveals Longitudinal Changes in Circulating Immune Cells During Radium-223 Therapy in Patients With Metastatic Castration-Resistant Prostate Cancer. Front Oncol 2021; 11:667658. [PMID: 34084750 PMCID: PMC8167220 DOI: 10.3389/fonc.2021.667658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/15/2021] [Indexed: 11/13/2022] Open
Abstract
Background Radium-223 improves overall survival (OS) in men with bone metastatic castration-resistant prostate cancer (mCRPC). While the exact mechanism behind this survival benefit remains unclear, radium-induced immunological mechanisms might contribute to the OS advantage. We performed a comprehensive evaluation of the immunological changes in mCRPC patients by phenotyping the peripheral blood mononuclear cells (PBMCs) during radium-223 therapy. Materials and Methods In this prospective, single-arm, exploratory study, PBMCs of 30 mCRPC patients were collected before, during, and after treatment with radium-223. Lymphocyte and monocyte counts were analyzed to get insight into general immune cell trends. Next, we analyzed changes in T cell subsets, myeloid-derived suppressor cells (MDSCs), and immune checkpoint expression using linear regression models. Per subset, the 6-month change (% of baseline) was determined. Bootstrapped 95% confidence intervals were used to measure the degree of uncertainty of our findings. Results We observed a substantial decrease in absolute lymphocyte counts (-0.12 * 10^9 cells/L per injection, 95% CI: -0.143 - -0.102). Simultaneously, an increase was observed in the proportion of T cells that expressed costimulatory (ICOS) or inhibitory (TIM-3, PD-L1, and PD-1) checkpoint molecules. Moreover, the fraction of two immunosuppressive subsets – the regulatory T cells and the monocytic MDSCs – increased throughout treatment. These findings were not more pronounced in patients with an alkaline phosphatase response during therapy. Conclusion Immune cell subsets in patients with mCRPC changed during radium-223 therapy, which warrants further research into the possible immunological consequences of these changes.
Collapse
Affiliation(s)
- Jeroen H A Creemers
- Department of Tumor Immunology, Radboudumc, Nijmegen, Netherlands.,Oncode Institute, Nijmegen, Netherlands
| | - Maarten J van der Doelen
- Department of Medical Oncology, Radboudumc, Nijmegen, Netherlands.,Department of Urology, Radboudumc, Nijmegen, Netherlands
| | - Sandra van Wilpe
- Department of Tumor Immunology, Radboudumc, Nijmegen, Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, Netherlands
| | - Rick Hermsen
- Department of Nuclear Medicine, Canisius-Wilhelmina Hospital, Nijmegen, Netherlands
| | | | - Diederik M Somford
- Department of Urology, Canisius-Wilhelmina Hospital, Nijmegen, Netherlands
| | - Marcel J R Janssen
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, Netherlands
| | | | - Niven Mehra
- Department of Tumor Immunology, Radboudumc, Nijmegen, Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, Netherlands
| | - Johannes Textor
- Department of Tumor Immunology, Radboudumc, Nijmegen, Netherlands.,Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen, Netherlands
| | - Harm Westdorp
- Department of Tumor Immunology, Radboudumc, Nijmegen, Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, Netherlands
| |
Collapse
|
25
|
Abstract
Significance: Genomic instability, a hallmark of cancer, renders cancer cells susceptible to genomic stress from both endogenous and exogenous origins, resulting in the increased tendency to accrue DNA damage, chromosomal instability, or aberrant DNA localization. Apart from the cell autonomous tumor-promoting effects, genomic stress in cancer cells could have a profound impact on the tumor microenvironment. Recent Advances: Recently, it is increasingly appreciated that harnessing genomic stress could provide a promising strategy to revive antitumor immunity, and thereby offer new therapeutic opportunities in cancer treatment. Critical Issues: Genomic stress is closely intertwined with antitumor immunity via mechanisms involving the direct crosstalk with DNA damage response components, upregulation of immune-stimulatory/inhibitory ligands, release of damage-associated molecular patterns, increase of neoantigen repertoire, and activation of DNA sensing pathways. A better understanding of these mechanisms will provide molecular basis for exploiting the genomic stress to boost antitumor immunity. Future Directions: Future research should pay attention to the heterogeneity between individual cancers in the genomic instability and the associated immune response, and how to balance the toxicity and benefit by specifying the types, potency, and treatment sequence of genomic stress inducer in therapeutic practice. Antioxid. Redox Signal. 34, 1128-1150.
Collapse
Affiliation(s)
- Congying Pu
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Siyao Tao
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jun Xu
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Min Huang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
26
|
Damiana TST, Dalm SU. Combination Therapy, a Promising Approach to Enhance the Efficacy of Radionuclide and Targeted Radionuclide Therapy of Prostate and Breast Cancer. Pharmaceutics 2021; 13:pharmaceutics13050674. [PMID: 34067215 PMCID: PMC8151894 DOI: 10.3390/pharmaceutics13050674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022] Open
Abstract
In recent years, radionuclide therapy (RT) and targeted radionuclide therapy (TRT) have gained great interest in cancer treatment. This is due to promising results obtained in both preclinical and clinical studies. However, a complete response is achieved in only a small percentage of patients that receive RT or TRT. As a consequence, there have been several strategies to improve RT and TRT outcomes including the combination of these treatments with other well-established anti-cancer therapies, for example, chemotherapy. Combinations of RT and TRT with other therapies with distinct mechanisms of action represent a promising strategy. As for prostate cancer and breast cancer, the two most prevalent cancer types worldwide, several combination-based therapies have been evaluated. In this review, we will provide an overview of the RT and TRT agents currently used or being investigated in combination with hormone therapy, chemotherapy, immunotherapy, and external beam radiation therapy for the treatment of prostate cancer and breast cancer.
Collapse
|
27
|
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
|
28
|
Abstract
As a single organ distributed diffusely throughout the body, bones represent both a unique challenge and unique opportunity for the treatment of symptomatic metastatic disease. While the multifocality of bone metastases often prevents effective complete treatment with focal radiotherapy, the similar pathophysiology of these diffuse sites of disease opens the door to targeted systemic therapy. The relatively rapid dose fall-off from beta- or alpha-emitting particles, if correctly and reliably targeted to osseous metastases, might reduce tumor burden and enhance pain control or improve survival. Radioisotopes have thus been studied keenly with the first generation of primarily beta-emitting radioisotopes, strontium-89 and samarium-153, which reached early FDA approval based on successful endpoints of pain control. More recently, an alpha-emitting therapy, radium-223, has demonstrated a successful endpoint of improved overall survival in patients with a burden of symptomatic, metastatic castrate-resistant prostate cancer (mCRPC) confined to the bones. With this discovery, an additional survival-improving tool beyond systemic and hormonal agents was added to the treatment arsenal for mCRPC for suitable candidates. With an improved understanding of the optimization of hormonal and systemic therapies in the context of mCRPC, there is lingering uncertainty regarding the safety and efficacy of combinatorial use of alpha and beta-emitting therapies with the current generation of systemic agents. In this narrative review, we will highlight the current understanding of the relative utility and clinical paradigms involving alpha- and beta-emitting radioisotopes. We discuss fundamental mechanisms for antineoplastic activity, initial clinical trials validating their use, the use of concurrent antiresorptive therapies to provide bone protection, and ongoing clinical trials targeted at best utilization of these agents in the broader context of mCRPC treatment.
Collapse
Affiliation(s)
- Andrew W Smith
- Departments of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Benjamin A Greenberger
- Sidney Kimmel Medical College & Cancer Center at Thomas Jefferson University, Philadelphia, PA
| | - Robert B Den
- Sidney Kimmel Medical College & Cancer Center at Thomas Jefferson University, Philadelphia, PA
| | - Richard G Stock
- Departments of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY..
| |
Collapse
|
29
|
Guerra Liberal FDC, O'Sullivan JM, McMahon SJ, Prise KM. Targeted Alpha Therapy: Current Clinical Applications. Cancer Biother Radiopharm 2020; 35:404-417. [PMID: 32552031 DOI: 10.1089/cbr.2020.3576] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
α-Emitting radionuclides have been approved for cancer treatment since 2013, with increasing degrees of success. Despite this clinical utility, little is known regarding the mechanisms of action of α particles in this setting, and accurate assessments of the dosimetry underpinning their effectiveness are lacking. However, targeted alpha therapy (TAT) is gaining more attention as new targets, synthetic chemistry approaches, and α particle emitters are identified, constructed, developed, and realized. From a radiobiological perspective, α particles are more effective at killing cells compared to low linear energy transfer radiation. Also, from these direct effects, it is now evident from preclinical and clinical data that α emitters are capable of both producing effects in nonirradiated bystander cells and stimulating the immune system, extending the biological effects of TAT beyond the range of α particles. The short range of α particles makes them a potent tool to irradiate single-cell lesions or treat solid tumors by minimizing unwanted irradiation of normal tissue surrounding the cancer cells, assuming a high specificity of the radiopharmaceutical and good stability of its chemical bonds. Clinical approval of 223RaCl2 in 2013 was a major milestone in the widespread application of TAT as a safe and effective strategy for cancer treatment. In addition, 225Ac-prostate specific membrane antigen treatment benefit in metastatic castrate-resistant prostate cancer patients, refractory to standard therapies, is another game-changing piece in the short history of TAT clinical application. Clinical applications of TAT are growing with different radionuclides and combination therapies, and in different clinical settings. Despite the remarkable advances in TAT dosimetry and imaging, it has not yet been used to its full potential. Labeled 227Th and 225Ac appear to be promising candidates and could represent the next generation of agents able to extend patient survival in several clinical scenarios.
Collapse
Affiliation(s)
- Francisco D C Guerra Liberal
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom.,Faculdade de Ciências e Tenclonogia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Joe M O'Sullivan
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom.,Clinical Oncology, Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Stephen J McMahon
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Kevin M Prise
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
30
|
Hagemann UB, Wickstroem K, Hammer S, Bjerke RM, Zitzmann-Kolbe S, Ryan OB, Karlsson J, Scholz A, Hennekes H, Mumberg D, Cuthbertson AS. Advances in Precision Oncology: Targeted Thorium-227 Conjugates As a New Modality in Targeted Alpha Therapy. Cancer Biother Radiopharm 2020; 35:497-510. [PMID: 32255671 PMCID: PMC7475103 DOI: 10.1089/cbr.2020.3568] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Targeted α therapy (TAT) offers the potential for the targeted delivery of potent α-particle-emitting radionuclides that emit high linear energy transfer radiation. This leads to a densely ionizing radiation track over a short path. Localized radiation induces cytotoxic, difficult-to-repair, clustered DNA double-strand breaks (DSBs). To date, radium-223 (223Ra) is the only TAT approved for the treatment of patients with metastatic castration-resistant prostate cancer. Thorium-227 (227Th), the progenitor nuclide of 223Ra, offers promise as a wider-ranging alternative due to the availability of efficient chelators, such as octadentate 3,2-hydroxypyridinone (3,2-HOPO). The 3,2-HOPO chelator can be readily conjugated to a range of targeting moieties, enabling the generation of new targeted thorium-227 conjugates (TTCs). This review provides a comprehensive overview of the advances in the preclinical development of TTCs for hematological cancers, including CD22-positive B cell cancers and CD33-positive leukemia, as well as for solid tumors overexpressing renal cell cancer antigen CD70, membrane-anchored glycoprotein mesothelin in mesothelioma, prostate-specific membrane antigen in prostate cancer, and fibroblast growth factor receptor 2. As the mechanism of action for TTCs is linked to the formation of DSBs, the authors also report data supporting combinations of TTCs with inhibitors of the DNA damage response pathways, including those of the ataxia telangiectasia and Rad3-related protein, and poly-ADP ribose polymerase. Finally, emerging evidence suggests that TTCs induce immunogenic cell death through the release of danger-associated molecular patterns. Based on encouraging preclinical data, clinical studies have been initiated to investigate the safety and tolerability of TTCs in patients with various cancers.
Collapse
|
31
|
Punnanitinont A, Kannisto ED, Matsuzaki J, Odunsi K, Yendamuri S, Singh AK, Patnaik SK. Sublethal Radiation Affects Antigen Processing and Presentation Genes to Enhance Immunogenicity of Cancer Cells. Int J Mol Sci 2020; 21:2573. [PMID: 32272797 PMCID: PMC7178186 DOI: 10.3390/ijms21072573] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 01/10/2023] Open
Abstract
While immunotherapy in cancer is designed to stimulate effector T cell response, tumor-associated antigens have to be presented on malignant cells at a sufficient level for recognition of cancer by T cells. Recent studies suggest that radiotherapy enhances the anti-cancer immune response and also improves the efficacy of immunotherapy. To understand the molecular basis of such observations, we examined the effect of ionizing X-rays on tumor antigens and their presentation in a set of nine human cell lines representing cancers of the esophagus, lung, and head and neck. A single dose of 7.5 or 15 Gy radiation enhanced the New York esophageal squamous cell carcinoma 1 (NY-ESO-1) tumor-antigen-mediated recognition of cancer cells by NY-ESO-1-specific CD8+ T cells. Irradiation led to significant enlargement of live cells after four days, and microscopy and flow cytometry revealed multinucleation and polyploidy in the cells because of dysregulated mitosis, which was also revealed in RNA-sequencing-based transcriptome profiles of cells. Transcriptome analyses also showed that while radiation had no universal effect on genes encoding tumor antigens, it upregulated the expression of numerous genes involved in antigen processing and presentation pathways in all cell lines. This effect may explain the immunostimulatory role of cancer radiotherapy.
Collapse
Affiliation(s)
- Achamaporn Punnanitinont
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; (A.P.); (E.D.K.); (S.Y.)
| | - Eric D. Kannisto
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; (A.P.); (E.D.K.); (S.Y.)
| | - Junko Matsuzaki
- Department of Gynecologic Oncology, and Center for Cancer Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; (J.M.); (K.O.)
| | - Kunle Odunsi
- Department of Gynecologic Oncology, and Center for Cancer Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; (J.M.); (K.O.)
| | - Sai Yendamuri
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; (A.P.); (E.D.K.); (S.Y.)
| | - Anurag K. Singh
- Department of Radiation Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Santosh K. Patnaik
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; (A.P.); (E.D.K.); (S.Y.)
| |
Collapse
|
32
|
Jadvar H. Targeted α-Therapy in Cancer Management: Synopsis of Preclinical and Clinical Studies. Cancer Biother Radiopharm 2020; 35:475-484. [PMID: 32202923 DOI: 10.1089/cbr.2019.3340] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The approval of 223Ra dichloride (223RaCl2) in 2013 was a principal event in introducing targeted α-therapy as a form of safe and effective management strategy in cancer. There is an increasing interest in research and development of new targeted α-therapy agents spearheaded by advancements in cancer biology, radiochemistry, and availability of clinically relevant α particles. There are active clinical studies on sequencing or combining 223RaCl2 with other drug regimens in the setting of metastatic prostate cancer and in other cancers such as osteosarcoma and bone-dominant breast cancer. Targeted α-therapy strategy is also being actively explored through many preclinical and few early clinical studies using 225Ac, 213Bi, 211At, 227Th, and 212Pb. Investigations incorporating 225Ac are more robust and active at this time with promising results. The author provide a brief synopsis of the preclinical and clinical studies in the rapidly evolving field of targeted α-therapy in cancer management.
Collapse
Affiliation(s)
- Hossein Jadvar
- Division of Nuclear Medicine, Department of Radiology, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
33
|
Galluzzi L, Vitale I, Warren S, Adjemian S, Agostinis P, Martinez AB, Chan TA, Coukos G, Demaria S, Deutsch E, Draganov D, Edelson RL, Formenti SC, Fucikova J, Gabriele L, Gaipl US, Gameiro SR, Garg AD, Golden E, Han J, Harrington KJ, Hemminki A, Hodge JW, Hossain DMS, Illidge T, Karin M, Kaufman HL, Kepp O, Kroemer G, Lasarte JJ, Loi S, Lotze MT, Manic G, Merghoub T, Melcher AA, Mossman KL, Prosper F, Rekdal Ø, Rescigno M, Riganti C, Sistigu A, Smyth MJ, Spisek R, Stagg J, Strauss BE, Tang D, Tatsuno K, van Gool SW, Vandenabeele P, Yamazaki T, Zamarin D, Zitvogel L, Cesano A, Marincola FM. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer 2020; 8:e000337. [PMID: 32209603 PMCID: PMC7064135 DOI: 10.1136/jitc-2019-000337] [Citation(s) in RCA: 670] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
Cells succumbing to stress via regulated cell death (RCD) can initiate an adaptive immune response associated with immunological memory, provided they display sufficient antigenicity and adjuvanticity. Moreover, multiple intracellular and microenvironmental features determine the propensity of RCD to drive adaptive immunity. Here, we provide an updated operational definition of immunogenic cell death (ICD), discuss the key factors that dictate the ability of dying cells to drive an adaptive immune response, summarize experimental assays that are currently available for the assessment of ICD in vitro and in vivo, and formulate guidelines for their interpretation.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York City, New York, USA
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
- Université de Paris, Paris, France
| | - Ilio Vitale
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS, Candiolo, Torino, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Sarah Warren
- NanoString Technologies, Seattle, Washington, USA
| | - Sandy Adjemian
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB-KU Leuven Center for Cancer Biology, KU Leuevn, Leuven, Belgium
| | - Aitziber Buqué Martinez
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - George Coukos
- Ludwig Institute for Cancer Research and Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York City, New York, USA
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM "Molecular Radiotherapy and therapeutic innovation", U1030 Molecular Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France
- SIRIC SOCRATES, DHU Torino, Faculté de Medecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | | | - Richard L Edelson
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
- Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
| | - Jitka Fucikova
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio, Prague, Czech Republic
| | - Lucia Gabriele
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Udo S Gaipl
- Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sofia R Gameiro
- Laboratory of Tumor Immunology and Biology, National Cancer Institute/Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Encouse Golden
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
| | - Jian Han
- iRepertoire, Inc, Huntsville, Alabama, USA
| | - Kevin J Harrington
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital/Institute of Cancer Research National Institute for Health Biomedical Research Centre, London, UK
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, National Cancer Institute/Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Tim Illidge
- University of Manchester, NIHR Manchester Biomedical Research Centre, Christie Hospital, Manchester, UK
| | - Michael Karin
- Department of Pharmacology and Pathology, University of California at San Diego (UCSD) School of Medicine, La Jolla, California, USA
| | - Howard L Kaufman
- Division of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Replimune, Inc, Woburn, Massachusetts, USA
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
| | - Guido Kroemer
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, U1138, Paris, France
- Sorbonne Université, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Juan Jose Lasarte
- Program of Immunology and Immunotherapy, Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain
| | - Sherene Loi
- Division of Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Gwenola Manic
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS, Candiolo, Torino, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, MSKCC, New York City, New York, USA
- Weill Cornell Medical College, New York City, New York, USA
- Parker Institute for Cancer Immunotherapy, MSKCC, New York City, New York, USA
| | | | | | - Felipe Prosper
- Hematology and Cell Therapy, Clinica Universidad de Navarra, Pamplona, Spain
| | - Øystein Rekdal
- Lytix Biopharma, Oslo, Norway
- Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Maria Rescigno
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
- Interdepartmental Research Center of Molecular Biotechnology, University of Torino, Torino, Italy
| | - Antonella Sistigu
- UOSD Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Radek Spisek
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio, Prague, Czech Republic
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec City, Canada
- Institut du Cancer de Montréal, Montréal, Quebec City, Canada
- Faculté de Pharmacie de l'Université de Montréal, Montréal, Quebec City, Canada
| | - Bryan E Strauss
- Centro de Investigação Translacional em Oncologia/LIM24, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Kazuki Tatsuno
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Peter Vandenabeele
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
- Methusalem program, Ghent University, Ghent, Belgium
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
| | - Dmitriy Zamarin
- Department of Medicine, Weill Cornell Medical College, New York City, New York, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Equipe labellisée par la Ligue contre le cancer, Gustave Roussy, Villejuif, France
- Faculty of Medicine, University of Paris Sud/Paris Saclay, Le Kremlin-Bicêtre, France
- INSERM U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | | | | |
Collapse
|
34
|
Prokhnevska N, Emerson DA, Kissick HT, Redmond WL. Immunological Complexity of the Prostate Cancer Microenvironment Influences the Response to Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1210:121-147. [PMID: 31900908 DOI: 10.1007/978-3-030-32656-2_7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Prostate cancer is one of the most common cancers in men and a leading cause of cancer-related death. Recent advances in the treatment of advanced prostate cancer, including the use of more potent and selective inhibitors of the androgen signaling pathway, have provided significant clinical benefit for men with metastatic castration-resistant prostate cancer (mCRPC). However, most patients develop progressive lethal disease, highlighting the need for more effective treatments. One such approach is immunotherapy, which harness the power of the patient's immune system to identify and destroy cancer cells through the activation of cytotoxic CD8 T cells specific for tumor antigens. Although immunotherapy, particularly checkpoint blockade, can induce significant clinical responses in patients with solid tumors or hematological malignancies, minimal efficacy has been observed in men with mCRPC. In the current review, we discuss our current understanding of the immunological complexity of the immunosuppressive prostate cancer microenvironment, preclinical models of prostate cancer, and recent advances in immunotherapy clinical trials to improve outcomes for men with mCRPC.
Collapse
Affiliation(s)
| | - Dana A Emerson
- Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA.,Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | | | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA.
| |
Collapse
|
35
|
Cursano MC, Iuliani M, Casadei C, Stellato M, Tonini G, Paganelli G, Santini D, De Giorgi U. Combination radium-223 therapies in patients with bone metastases from castration-resistant prostate cancer: A review. Crit Rev Oncol Hematol 2020; 146:102864. [PMID: 31986318 DOI: 10.1016/j.critrevonc.2020.102864] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 12/27/2019] [Accepted: 01/04/2020] [Indexed: 01/26/2023] Open
Abstract
Chemotherapeutic agents (docetaxel, cabazitaxel), hormonal therapies (abiraterone, enzalutamide) and radium-223 improve survival in patients with bone metastatic castration-resistant prostate cancer (mCRPC). Combinations of radium-223 with these agents or novel drugs have been investigated in order to improve survival and decrease bone-related morbidity. In mCRPC, clinical and preclinical data indicate that radium-223, abiraterone and enzalutamide have a direct effect on prostate cancer cells and bone microenvironment when administered as single agents. Initial results from studies of radium-223 and abiraterone, enzalutamide or docetaxel demonstrated efficacy without any safety concern in pre-treated mCRPC; however, this safety profile changed when radium-based combination therapies were administered in un-pretreated mCRPC. This review underline the biological rationale for combining radium strategies, investigating their effects on bone in terms of control of skeletal-related events and bone disease progression. The aim is to understand the possible reasons why different radium-based combination treatments can led to different clinical outcomes.
Collapse
Affiliation(s)
- M C Cursano
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128, Rome, Italy.
| | - M Iuliani
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128, Rome, Italy
| | - C Casadei
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014, Meldola, Italy
| | - M Stellato
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128, Rome, Italy
| | - G Tonini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128, Rome, Italy
| | - G Paganelli
- Department of Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014, Meldola, Italy
| | - D Santini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128, Rome, Italy
| | - U De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014, Meldola, Italy
| |
Collapse
|
36
|
Ra-223 Treatment for Bone Metastases in Castrate-Resistant Prostate Cancer: Practical Management Issues for Patient Selection. Am J Clin Oncol 2019; 42:399-406. [PMID: 30844849 PMCID: PMC6445613 DOI: 10.1097/coc.0000000000000528] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bone metastases are common in men with metastatic castrate-resistant prostate cancer (mCRPC), occurring in 30% of patients within 2 years of castrate resistance and in >90% of patients over the disease course. There are 6 US Food and Drug Administration-approved therapies for mCRPC with demonstrated survival benefit. Of these, only radium-223 (Ra-223) specifically targets bone metastases, delays development of skeletal-related events, and improves survival. This review discusses key data from the ALSYMPCA trial, which contributed to the approval of Ra-223. Data from other trials are highlighted to provide further insight into which patients might benefit from Ra-223. Special patient populations are described, as well as other considerations for the administration of Ra-223. Finally, ongoing trials of Ra-223 combined with other therapies for mCRPC are discussed. These include combining Ra-223 with sipuleucel-T or immunooncology agents, to enhance immune responses, and trials in mildly symptomatic or asymptomatic patients. To date, the optimal timing, sequence, and combinations of Ra-223 with other agents are yet to be determined. The goals of this review are to provide insight into practical aspects of patient selection for Ra-223 treatment and to discuss key therapeutic strategies using the 6 approved mCRPC agents in patients with bone metastases. Results from ongoing trials should help guide the practitioner in using Ra-223 in patients with mCRPC.
Collapse
|
37
|
Morris MJ, Corey E, Guise TA, Gulley JL, Kevin Kelly W, Quinn DI, Scholz A, Sgouros G. Radium-223 mechanism of action: implications for use in treatment combinations. Nat Rev Urol 2019; 16:745-756. [PMID: 31712765 PMCID: PMC7515774 DOI: 10.1038/s41585-019-0251-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2019] [Indexed: 12/16/2022]
Abstract
The targeted alpha therapy radium-223 (223Ra) can prolong survival in men with castration-resistant prostate cancer (CRPC) who have symptomatic bone metastases and no known visceral metastases. Preclinical studies demonstrate that 223Ra preferentially incorporates into newly formed bone matrix within osteoblastic metastatic lesions. The emitted high-energy alpha particles induce DNA double-strand breaks that might be irreparable and lead to cell death in nearby exposed tumour cells, osteoblasts and osteoclasts. Consequently, tumour growth and abnormal bone formation are inhibited by these direct effects and by the disruption of positive-feedback loops between tumour cells and the bone microenvironment. 223Ra might also modulate immune responses within the bone. The clinical utility of 223Ra has encouraged the development of other anticancer targeted alpha therapies. A thorough understanding of the mechanism of action could inform the design of new combinatorial treatment strategies that might be more efficacious than monotherapy. On the basis of the current mechanistic knowledge and potential clinical benefits, combination therapies of 223Ra with microtubule-stabilizing cytotoxic drugs and agents targeting the androgen receptor axis, immune checkpoint receptors or DNA damage response proteins are being explored in patients with CRPC and metastatic bone disease.
Collapse
Affiliation(s)
- Michael J Morris
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA.
| | - Eva Corey
- Department of Urology, University of Washington, School of Medicine, Seattle, WA, USA
| | - Theresa A Guise
- Indiana University, School of Medicine, Indianapolis, IN, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - William Kevin Kelly
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - David I Quinn
- Norris Comprehensive Cancer Center, Los Angeles, CA, USA
- Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Arne Scholz
- Bayer AG, Drug Discovery, Pharmaceuticals, Berlin, Germany
| | - George Sgouros
- Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
Tafreshi NK, Doligalski ML, Tichacek CJ, Pandya DN, Budzevich MM, El-Haddad G, Khushalani NI, Moros EG, McLaughlin ML, Wadas TJ, Morse DL. Development of Targeted Alpha Particle Therapy for Solid Tumors. Molecules 2019; 24:molecules24234314. [PMID: 31779154 PMCID: PMC6930656 DOI: 10.3390/molecules24234314] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022] Open
Abstract
Targeted alpha-particle therapy (TAT) aims to selectively deliver radionuclides emitting α-particles (cytotoxic payload) to tumors by chelation to monoclonal antibodies, peptides or small molecules that recognize tumor-associated antigens or cell-surface receptors. Because of the high linear energy transfer (LET) and short range of alpha (α) particles in tissue, cancer cells can be significantly damaged while causing minimal toxicity to surrounding healthy cells. Recent clinical studies have demonstrated the remarkable efficacy of TAT in the treatment of metastatic, castration-resistant prostate cancer. In this comprehensive review, we discuss the current consensus regarding the properties of the α-particle-emitting radionuclides that are potentially relevant for use in the clinic; the TAT-mediated mechanisms responsible for cell death; the different classes of targeting moieties and radiometal chelators available for TAT development; current approaches to calculating radiation dosimetry for TATs; and lead optimization via medicinal chemistry to improve the TAT radiopharmaceutical properties. We have also summarized the use of TATs in pre-clinical and clinical studies to date.
Collapse
Affiliation(s)
- Narges K. Tafreshi
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Michael L. Doligalski
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Christopher J. Tichacek
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Darpan N. Pandya
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA; (D.N.P.); (T.J.W.)
| | - Mikalai M. Budzevich
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Ghassan El-Haddad
- Depts. of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Nikhil I. Khushalani
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Eduardo G. Moros
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Department of Physics, University of South Florida, Tampa, FL 33612, USA
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Mark L. McLaughlin
- Department of Pharmaceutical Sciences, West Virginia University, Health Sciences Center, Morgantown, WV & Modulation Therapeutics Inc., 64 Medical Center Drive, Morgantown, WV 26506, USA;
| | - Thaddeus J. Wadas
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA; (D.N.P.); (T.J.W.)
| | - David L. Morse
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
- Department of Physics, University of South Florida, Tampa, FL 33612, USA
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
- Correspondence: ; Tel.: +1-813-745-8948; Fax: +1-813-745-8375
| |
Collapse
|
39
|
De Vincentis G, Gerritsen W, Gschwend JE, Hacker M, Lewington V, O'Sullivan JM, Oya M, Pacilio M, Parker C, Shore N, Sartor O. Advances in targeted alpha therapy for prostate cancer. Ann Oncol 2019; 30:1728-1739. [PMID: 31418764 PMCID: PMC6927314 DOI: 10.1093/annonc/mdz270] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Amongst therapeutic radiopharmaceuticals, targeted alpha therapy (TαT) can deliver potent and local radiation selectively to cancer cells as well as the tumor microenvironment and thereby control cancer while minimizing toxicity. In this review, we discuss the history, progress, and future potential of TαT in the treatment of prostate cancer, including dosimetry-individualized treatment planning, combinations with small-molecule therapies, and conjugation to molecules directed against antigens expressed by prostate cancer cells, such as prostate-specific membrane antigen (PSMA) or components of the tumor microenvironment. A clinical proof of concept that TαT is efficacious in treating bone-metastatic castration-resistant prostate cancer has been demonstrated by radium-223 via improved overall survival and long-term safety/tolerability in the phase III ALSYMPCA trial. Dosimetry calculation and pharmacokinetic measurements of TαT provide the potential for optimization and individualized treatment planning for a precision medicine-based cancer management paradigm. The ability to combine TαTs with other agents, including chemotherapy, androgen receptor-targeting agents, DNA repair inhibitors, and immuno-oncology agents, is under investigation. Currently, TαTs that specifically target prostate cancer cells expressing PSMA represents a promising therapeutic approach. Both PSMA-targeted actinium-225 and thorium-227 conjugates are under investigation. The described clinical benefit, safety and tolerability of radium-223 and the recent progress in TαT trial development suggest that TαT occupies an important new role in prostate cancer treatment. Ongoing studies with newer dosimetry methods, PSMA targeting, and novel approaches to combination therapies should expand the utility of TαT in prostate cancer treatment.
Collapse
Affiliation(s)
- G De Vincentis
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, Rome, Italy
| | - W Gerritsen
- Department of Medical Oncology, Radboud UMC, Nijmegen, The Netherlands
| | - J E Gschwend
- Department of Urology, Technical University of Munich, Rechts der Isar Medical Center, Munich, Germany
| | - M Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - V Lewington
- Department of Imaging Sciences and Biomedical Engineering, King's College, London, UK
| | - J M O'Sullivan
- Center for Cancer Research and Cell Biology, Queen's University Belfast, Northern Ireland Cancer Center, Belfast City Hospital, Belfast, Northern Ireland
| | - M Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - M Pacilio
- Medical Physics Department, "Policlinico Umberto I" University Hospital, Rome, Italy
| | - C Parker
- The Royal Marsden Hospital, Sutton, UK
| | - N Shore
- Carolina Urologic Research Center, Myrtle Beach
| | - O Sartor
- Tulane Cancer Center, Tulane University, New Orleans, USA.
| |
Collapse
|
40
|
Stelcer E, Konkol M, Głȩboka A, Suchorska WM. Liquid Biopsy in Oligometastatic Prostate Cancer-A Biologist's Point of View. Front Oncol 2019; 9:775. [PMID: 31475117 PMCID: PMC6702517 DOI: 10.3389/fonc.2019.00775] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer (PCa) is the main cause of cancer-related mortality in males and the diagnosis, treatment, and care of these patients places a great burden on healthcare systems globally. Clinically, PCa is highly heterogeneous, ranging from indolent tumors to highly aggressive disease. In many cases treatment-generally either radiotherapy (RT) or surgery-can be curative. Several key genetic and demographic factors such as age, family history, genetic susceptibility, and race are associated with a high incidence of PCa. While our understanding of PCa, which is mainly based on the tools of molecular biology-has improved dramatically in recent years, efforts to better understand this complex disease have led to the identification of a new type of PCa-oligometastatic PCa. Oligometastatic disease should be considered an individual, heterogeneous entity with distinct metastatic phenotypes and, consequently, wide prognostic variability. In general, patients with oligometastatic disease typically present less biologically aggressive tumors whose metastatic potential is more limited and which are slow-growing. These patients are good candidates for more aggressive treatment approaches. The main aim of the presented review was to evaluate the utility of liquid biopsy for diagnostic purposes in PCa and for use in monitoring disease progression and treatment response, particularly in patients with oligometastatic PCa. Liquid biopsies offer a rapid, non-invasive approach whose use t is expected to play an important role in routine clinical practice to benefit patients. However, more research is needed to resolve the many existing discrepancies with regard to the definition and isolation method for specific biomarkers, as well as the need to determine the most appropriate markers. Consequently, the current priority in this field is to standardize liquid biopsy-based techniques. This review will help to improve understanding of the biology of PCa, particularly the recently defined condition known as "oligometastatic PCa". The presented review of the body of evidence suggests that additional research in molecular biology may help to establish novel treatments for oligometastatic PCa. In the near future, the treatment of PCa will require an interdisciplinary approach involving active cooperation among clinicians, physicians, and biologists.
Collapse
Affiliation(s)
- Ewelina Stelcer
- Radiobiology Laboratory, Greater Poland Cancer Centre, Poznan, Poland
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Konkol
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland
- Radiation Oncology Department, Greater Poland Cancer Centre, Poznan, Poland
| | | | - Wiktoria Maria Suchorska
- Radiobiology Laboratory, Greater Poland Cancer Centre, Poznan, Poland
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
41
|
The Mode-of-Action of Targeted Alpha Therapy Radium-223 as an Enabler for Novel Combinations to Treat Patients with Bone Metastasis. Int J Mol Sci 2019; 20:ijms20163899. [PMID: 31405099 PMCID: PMC6720648 DOI: 10.3390/ijms20163899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/12/2022] Open
Abstract
Bone metastasis is a common clinical complication in several cancer types, and it causes a severe reduction in quality of life as well as lowering survival time. Bone metastases proceed through a vicious self-reinforcing cycle that can be osteolytic or osteoblastic in nature. The vicious cycle is characterized by cancer cells residing in bone releasing signal molecules that promote the differentiation of osteoclasts and osteoblasts either directly or indirectly. The increased activity of osteoclasts and osteoblasts then increases bone turnover, which releases growth factors that benefit metastatic cancer cells. In order to improve the prognosis of patients with bone metastases this cycle must be broken. Radium-223 dichloride (radium-223), the first targeted alpha therapy (TAT) approved, is an osteomimetic radionuclide that is incorporated into bone metastases where its high-linear energy transfer alpha radiation disrupts both the activity of bone cells and cancer cells. Therefore, radium-223 treatment has been shown preclinically to directly affect cancer cells in both osteolytic breast cancer and osteoblastic prostate cancer bone metastases as well as to inhibit the differentiation of osteoblasts and osteoclasts. Clinical studies have demonstrated an increase in survival in patients with metastatic castration-resistant prostate cancer. Due to the effectiveness and low toxicity of radium-223, several novel combination treatment strategies are currently eliciting considerable research interest.
Collapse
|
42
|
Van den Wyngaert T, Tombal B. The changing role of radium-223 in metastatic castrate-resistant prostate cancer: has the EMA missed the mark with revising the label? THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2019; 63:170-182. [PMID: 31298017 DOI: 10.23736/s1824-4785.19.03205-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Radium-223 (223Ra) is a life-prolonging treatment in symptomatic men with metastatic castrate-resistant prostate cancer (mCRPC) and bone metastases, but no visceral disease, regardless of prior treatment with docetaxel. Together with four other drugs (i.e. abiraterone, cabazitaxel, docetaxel, enzalutamide), it has been available for clinical use since 2013 and has been shown to also provide benefits in quality-of-life and societal benefits. However, in 2018 the European Medicines Agency ruled to restrict the use of radium-223 to a more advanced disease setting after at least two lines of one or the other life-prolonging agent. This decision was triggered by the results of a safety interim analysis of ERA-223, a trial investigating the combination of 223Ra and abiraterone versus abiraterone alone in patients without prior chemotherapy (with the exception of adjuvant treatment) with asymptomatic bone predominant mCRPC. That safety analysis showed an early increased risk of fracture and deaths with the combination treatment. This review critically appraises the available and emerging data with 223Ra treatment in an attempt to assess the appropriateness of the revised label of radium-223.
Collapse
Affiliation(s)
- Tim Van den Wyngaert
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium - .,Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium -
| | - Bertrand Tombal
- Department of Urology, Saint Luc University Clinic, Brussels, Belgium.,Institute of Clinical Research, Catholic University of Louvain, Brussels, Belgium
| |
Collapse
|
43
|
Patel VG, Oh WK. The evolving landscape of immunotherapy in advanced prostate cancer. Immunotherapy 2019; 11:903-912. [PMID: 31161846 DOI: 10.2217/imt-2019-0019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Prostate cancer exists in a clinical continuum of hormone-sensitive to castration-resistant disease. Despite the use of chemotherapy and androgen synthesis inhibitors in the castration-resistant setting, this remains a lethal disease. The advent of immune checkpoint blockade has changed the outlook for cancer treatment and survival for several tumors since its first approval in 2011; however, the clinical benefit in castration-resistant prostate cancer (CRPC) is rather limited. Currently, Sipuleucel-T remains the only immune modality to be approved in CRPC setting. Such immune resistance likely exists due to low immunogenicity of prostate tumor cells and an immunosuppressive tumor microenvironment. In this review, we describe the early experiences of immune checkpoint blockade and therapeutic vaccines in CRPC. We then outline strategies currently being implemented to overcome immune resistance, as well as genomic biomarker investigation to identify patients that may harbor more immunogenic tumors. At last, we preview emerging immunotherapeutic platforms.
Collapse
Affiliation(s)
- Vaibhav G Patel
- Department of Medicine, Division of Hematology & Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10010, USA
| | - William K Oh
- Department of Medicine, Division of Hematology & Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10010, USA
| |
Collapse
|
44
|
Hagemann UB, Ellingsen C, Schuhmacher J, Kristian A, Mobergslien A, Cruciani V, Wickstroem K, Schatz CA, Kneip C, Golfier S, Smeets R, Uran S, Hennekes H, Karlsson J, Bjerke RM, Ryan OB, Mumberg D, Ziegelbauer K, Cuthbertson AS. Mesothelin-Targeted Thorium-227 Conjugate (MSLN-TTC): Preclinical Evaluation of a New Targeted Alpha Therapy for Mesothelin-Positive Cancers. Clin Cancer Res 2019; 25:4723-4734. [DOI: 10.1158/1078-0432.ccr-18-3476] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/25/2019] [Accepted: 05/02/2019] [Indexed: 11/16/2022]
|
45
|
Ozpiskin OM, Zhang L, Li JJ. Immune targets in the tumor microenvironment treated by radiotherapy. Am J Cancer Res 2019; 9:1215-1231. [PMID: 30867826 PMCID: PMC6401500 DOI: 10.7150/thno.32648] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy (RT), the major anti-cancer modality for more than half of cancer patients after diagnosis, has the advantage of local tumor control with relatively less systematic side effects comparing to chemotherapy. However, the efficacy of RT is limited by acquired tumor resistance leading to the risks of relapse and metastasis. To further enhance the efficacy of RT, with the renaissances of targeted immunotherapy (TIT), increasing interests are raised on RT combined with TIT including cancer vaccines, T-cell therapy, and antibody-based immune checkpoint blockers (ICB) such as anti-CTLA-4 and anti-PD1/PD-L1. In achieving a significant synergy between RT and TIT, the dynamics of radiation-induced response in tumor cells and stromal cells, especially the cross-talk between tumor cells and immune cells in the irradiated tumor microenvironment (ITME) as highlighted in recent literature are to be elucidated. The abscopal effect refereeing the RT-induced priming function outside of ITME could be compromised by the immune-suppressive factors such as CD47 and PD-L1 on tumor cells and Treg induced or enhanced in the ITME. Cell surface receptors temporally or permanently induced and bioactive elements released from dead cells could serve antigenic source (radiation-associated antigenic proteins, RAAPs) to the host and have functions in immune regulation on the tumor. This review is attempted to summarize a cluster of factors that are inducible by radiation and targetable by antibodies, or have potential to be immune regulators to synergize tumor control with RT. Further characterization of immune regulators in ITME will deepen our understanding of the interplay among immune regulators in ITME and discover new effective targets for the combined modality with RT and TIT.
Collapse
|
46
|
Ricci M, Frantellizzi V, Bulzonetti N, De Vincentis G. Reversibility of castration resistance status after Radium-223 dichloride treatment: clinical evidence and review of the literature. Int J Radiat Biol 2019; 95:554-561. [PMID: 30557063 DOI: 10.1080/09553002.2019.1558301] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the history of prostate cancer, some of the patients progressed to castration-resistant prostate cancer (CRPC) stage and, although new drugs and treatment protocols have been introduced, CRPC presents poor prognosis. This review is focused on biological mechanisms, underlying CRPC described in scientific literature in order to explain the reversion of resistance to castration. We present the case of a 73-year-old man, affected by bone metastatic CRPC, early treated with Radium-223 with a complete response. After 15 months from Radium-223 treatment, prostate-specific antigen increased with radiological progression. Androgen deprivation therapy was again performed and was effective, despite previous CRPC condition and no known mechanisms that may explain the reversion of this condition. Therefore, to our knowledge, he is the unique described case of the reversion of resistance to castration. Nevertheless, promising aspects may be lack of intrametastatic production of androgen or the suppression of bypass androgen receptor signaling pathways. Furthermore, the cytotoxic action of Radium-223 on cancer stem cell (CSC), due to surrounding clones with high-bone turnover, or the immune response that underlying the abscopal effect, may also modulate the reversion of CRPC after Radium-223. If confirmed by multicenter trials, the reversion of CRPC may impact on the management of prostate cancer.
Collapse
Affiliation(s)
- Maria Ricci
- a Department of Radiological Sciences, Oncology and Anatomical Pathology , Sapienza University of Rome , Rome , Italy
| | - Viviana Frantellizzi
- a Department of Radiological Sciences, Oncology and Anatomical Pathology , Sapienza University of Rome , Rome , Italy.,b PhD Program: Angio-Cardio-Thoracic Pathophisiology and Imaging , "Sapienza" University of Rome , Rome , Italy
| | - Nadia Bulzonetti
- c Department of Radiotherapy , Policlinico Umberto I, Sapienza University of Rome , Rome , Italy
| | - Giuseppe De Vincentis
- a Department of Radiological Sciences, Oncology and Anatomical Pathology , Sapienza University of Rome , Rome , Italy
| |
Collapse
|
47
|
Heinrich D, Bruland Ø, Guise TA, Suzuki H, Sartor O. Alkaline phosphatase in metastatic castration-resistant prostate cancer: reassessment of an older biomarker. Future Oncol 2018; 14:2543-2556. [PMID: 29925281 DOI: 10.2217/fon-2018-0087] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Since most patients with metastatic castration-resistant prostate cancer (mCRPC) have bone metastases, it is important to understand the potential impact of therapies on prognostic biomarkers, such as ALP. Clinical studies involving mCRPC life-prolonging agents (i.e., sipuleucel-T, abiraterone, enzalutamide, docetaxel, cabazitaxel, and radium-223) have shown that baseline ALP level is prognostic for overall survival, and may be a better prognostic marker for overall survival than prostate-specific antigen in patients with bone-dominant mCRPC. Mechanism of action differences between therapies may partly explain ALP dynamics during treatment. ALP changes can be interpreted within the context of other parameters while monitoring disease activity to better understand the underlying pathology. This review evaluates the current role of ALP in mCRPC.
Collapse
Affiliation(s)
- Daniel Heinrich
- Department of Oncology, Akershus University Hospital, Sykehusveien 25, 1478 Lørenskog, Norway
| | - Øyvind Bruland
- Department of Oncology, Oslo University Hospital-Norwegian Radium Hospital, Ullernchausseen 70, 0379 Oslo, Norway
| | - Theresa A Guise
- Department of Medicine, Indiana University School of Medicine, 980 W. Walnut St, Walther Hall, R3, Room C130 Indianapolis, IN 46202, USA
| | - Hiroyoshi Suzuki
- Department of Urology, Toho University Sakura Medical Center, 564-1 Shimazu, Sakura-shi, Chiba 285-8741, Japan
| | - Oliver Sartor
- Departments of Medicine & Urology, Tulane Cancer Center, 1430 Tulane Ave., SL-42, New Orleans, LA 70112, USA
| |
Collapse
|
48
|
Redman JM, Madan RA. Spotlight on atezolizumab and its potential as an oncology agent. Expert Rev Anticancer Ther 2018; 18:719-722. [DOI: 10.1080/14737140.2018.1483241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Jason M. Redman
- Medical Oncology Service Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ravi A. Madan
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
49
|
Wennerberg E, Vanpouille-Box C, Bornstein S, Yamazaki T, Demaria S, Galluzzi L. Immune recognition of irradiated cancer cells. Immunol Rev 2018; 280:220-230. [PMID: 29027232 DOI: 10.1111/imr.12568] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ionizing irradiation has been extensively employed for the clinical management of solid tumors, with therapeutic or palliative intents, for decades. Until recently, radiation therapy (RT) was believed to mediate antineoplastic activity mostly (if not only) as a consequence of cancer cell-intrinsic effects. Indeed, the macromolecular damage imposed to malignant cells by RT initiates one or multiple signal transduction cascades that drive a permanent proliferative arrest (cellular senescence) or regulated cell death. Both these phenomena show a rather linear dose-response correlation. However, RT also mediates consistent immunological activity, not only as an "on-target effect" originating within irradiated cancer cells, but also as an "off-target effect" depending on the interaction between RT and stromal, endothelial, and immune components of the tumor microenvironment. Interestingly, the immunological activity of RT does not exhibit linear dose-response correlation. Here, we discuss the mechanisms whereby RT alters the capacity of the immune system to recognize and eliminate irradiated cancer cells, either as an "on-target" or as on "off-target" effect. In particular, we discuss the antagonism between the immunostimulatory and immunosuppressive effects of RT as we delineate combinatorial strategies to boost the former at the expenses of the latter.
Collapse
Affiliation(s)
- Erik Wennerberg
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | | | - Sophia Bornstein
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Université Paris Descartes/Paris V, Paris, France
| |
Collapse
|
50
|
Targeted alpha therapy using Radium-223: From physics to biological effects. Cancer Treat Rev 2018; 68:47-54. [PMID: 29859504 DOI: 10.1016/j.ctrv.2018.05.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Abstract
With the advance of the use of ionizing radiation in therapy, targeted alpha therapy (TAT) has assumed an important role around the world. This kind of therapy can potentially reduce side effects caused by radiation in normal tissues and increased destructive radiobiological effects in tumor cells. However, in many countries, the use of this therapy is still in a pioneering phase. Radium-223 (223Ra), an alpha-emitting radionuclide, has been the first of its kind to be approved for the treatment of bone metastasis in metastatic castration-resistant prostate cancer. Nevertheless, the interaction mechanism and the direct effects of this radiopharmaceutical in tumor cells are not fully understood neither characterized at a molecular level. In fact, the ways how TAT is linked to radiobiological effects in cancer is not yet revised. Therefore, this review introduces some physical properties of TAT that leads to biological effects and links this information to the hallmarks of cancer. The authors also collected the studies developed with 223Ra to correlate with the three categories reviewed - properties of TAT, 5 R's of radiobiology and hallmarks of cancer- and with the promising future to this radiopharmaceutical.
Collapse
|