1
|
Zhao H. Progress of the application of spatially fractionated radiation therapy in palliative treatment of tumors. Discov Oncol 2025; 16:678. [PMID: 40329010 PMCID: PMC12055688 DOI: 10.1007/s12672-025-02487-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
INTRODUCTION/BACKGROUND Malignant tumors pose a serious threat to human health. As tumor volume increases, conventional external beam radiation therapy (cEBRT) faces challenges in tumor control and normal tissue toxicity. Spatially fractionated radiation therapy (SFRT) has emerged as an alternative approach. MATERIALS AND METHODS This article reviews the history of SFRT, including kilovolt X-ray based GRID, megavolt X-ray based GRID, MLC-shaped GRID, LATTICE radiation therapy (LRT), Bragg-peak based SFRT, microbeam, minibeam, SBRT-PATHY, and ISPART. It also explores its radiobiological mechanisms, such as immunomodulation, bystander and abscopal effects, and vascular response. Clinical studies of SFRT in palliative tumor treatment are summarized, and its limitations and future directions are discussed. RESULTS SFRT has shown high symptom remission rates, significant target volume reduction, and even tumor control and long-term survival in some cases across various tumor types. However, it has limitations like lack of standardized dosimetric parameters, complex implementation, small-scale clinical studies, and uncertain immunomodulatory potential. CONCLUSION Despite limitations, SFRT shows promise as a palliative radiation therapy technique. Future large-scale, multi-center clinical trials are needed to standardize dosimetric parameters, clarify immunomodulatory mechanisms, and simplify the technology for wider application.
Collapse
Affiliation(s)
- Hongfu Zhao
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, 130033, Jilin, China.
| |
Collapse
|
2
|
Knight JA, Trosper N, Misa J, Bernard ME, Fabian D, Kudrimoti M, Yan W, St Clair W, Yang ES, Pokhrel D. Reported Early Clinical Outcomes of Forward-Planned Multileaf Collimator-Based 3-Dimensional Conformal Spatially Fractionated Radiation Therapy Technique for Large and Bulky Tumors. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00383-9. [PMID: 40298857 DOI: 10.1016/j.ijrobp.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE Conventionally fractionated radiation therapy for large, bulky (≥8 cm), unresectable tumors has been hampered by radiation-induced morbidity, but application of spatially fractionated radiation therapy (SFRT) for both palliative and curative intent has been increasingly accepted. We report our clinical use of novel 3-dimensional conformal multileaf collimator (MLC)-based SFRT with same-day computed tomography simulation and forward-planning method, providing a safe, rapidly efficacious reduction in disease burden and pain, with minimal normal-tissue toxicity. METHODS AND MATERIALS Patients with large, unresectable bulky tumors received 15 Gy in 1 fraction to the gross tumor volume (GTV) within an hour of computed tomography simulation, using a forward-planned MLC-based SFRT technique. All patients subsequently received either 30 Gy in 10 fractions, generally 2 days after SFRT for palliative intent, or site-specific, full-prescription doses starting 2 to 3 days after SFRT for curative intent. Patients underwent follow-up examinations and imaging in 3-month intervals to assess tumor response, pain control, and radiation-associated toxicity. RESULTS Between November 2019 and January 2024, 24 large tumors in 23 patients were analyzed. Median follow-up was 6 months (range 3-36 months). After SFRT, 16 patients (69.5%) proceeded with palliative-intent radiation therapy, 6 patients (26.0%) underwent curative-intent radiation therapy, and 1 patient (4.3%) declined further radiation therapy. Seven patients (30.4%) reported acute radiation-associated toxicities. A total of 3 acute grade ≥3 toxicities (13.0%) were reported, but no grade 5 toxicities occurred. Complete or partial response was seen in 14 of 24 (58%) tumors; clinical benefit rate was 79.2%. Twenty of 23 patients (86.9%) reported pain relief from tumor burden. CONCLUSIONS Same-day 3-dimensional MLC-based SFRT method provides fast, safe, and effective management of large, bulky, unresectable tumors for both palliative and therapeutic intents across a wide range of tumor sites and histologies, reducing tumor burden and improving patient comfort and compliance. This method could be useful for rapid SFRT including adaptive treatment. We recommend commissioning and validating this method at other institutions, including community cancer centers, to expand the access of efficient, high-quality SFRT treatment to underserved patient cohorts.
Collapse
Affiliation(s)
- James A Knight
- Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky, 40536 USA
| | - Nick Trosper
- Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky, 40536 USA
| | - Josh Misa
- Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky, 40536 USA
| | - Mark E Bernard
- Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky, 40536 USA
| | - Denise Fabian
- Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky, 40536 USA
| | - Mahesh Kudrimoti
- Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky, 40536 USA
| | - Weisi Yan
- Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky, 40536 USA
| | - William St Clair
- Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky, 40536 USA
| | - Eddy S Yang
- Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky, 40536 USA
| | - Damodar Pokhrel
- Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky, 40536 USA.
| |
Collapse
|
3
|
Manring HR, Fleming JL, Meng W, Gamez ME, Blakaj DM, Chakravarti A. FLASH Radiotherapy: From In Vivo Data to Clinical Translation. Hematol Oncol Clin North Am 2025; 39:237-255. [PMID: 39828472 DOI: 10.1016/j.hoc.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Delivery of radiotherapy (RT) at ultra-high dose rates or FLASH radiotherapy (FLASH-RT) is an emerging treatment option for patients with cancer that could increase survival outcomes and quality of life. In vivo data across a multitude of normal tissues and associated tumors have been published demonstrating the FLASH effect while bringing attention to the need for additional research. Combination of FLASH-RT with other treatment options including spatially fractionated RT, immunotherapy, and usage in the setting of reirradiation could also provide additional benefit. Phase I clinical trials have shown promising results, yet research is warranted before routine clinical use of FLASH-RT.
Collapse
Affiliation(s)
- Heather R Manring
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Jessica L Fleming
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Wei Meng
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Mauricio E Gamez
- Department of Radiation Oncology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Dukagjin M Blakaj
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
4
|
Morris ZS, Demaria S, Monjazeb AM, Formenti SC, Weichselbaum RR, Welsh J, Enderling H, Schoenfeld JD, Brody JD, McGee HM, Mondini M, Kent MS, Young KH, Galluzzi L, Karam SD, Theelen WSME, Chang JY, Huynh MA, Daib A, Pitroda S, Chung C, Serre R, Grassberger C, Deng J, Sodji QH, Nguyen AT, Patel RB, Krebs S, Kalbasi A, Kerr C, Vanpouille-Box C, Vick L, Aguilera TA, Ong IM, Herrera F, Menon H, Smart D, Ahmed J, Gartrell RD, Roland CL, Fekrmandi F, Chakraborty B, Bent EH, Berg TJ, Hutson A, Khleif S, Sikora AG, Fong L. Proceedings of the National Cancer Institute Workshop on combining immunotherapy with radiotherapy: challenges and opportunities for clinical translation. Lancet Oncol 2025; 26:e152-e170. [PMID: 40049206 DOI: 10.1016/s1470-2045(24)00656-9] [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: 08/07/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 03/09/2025]
Abstract
Radiotherapy both promotes and antagonises tumour immune recognition. Some clinical studies show improved patient outcomes when immunotherapies are integrated with radiotherapy. Safe, greater than additive, clinical response to the combination is limited to a subset of patients, however, and how radiotherapy can best be combined with immunotherapies remains unclear. The National Cancer Institute-Immuno-Oncology Translational Network-Society for Immunotherapy of Cancer-American Association of Immunology Workshop on Combining Immunotherapy with Radiotherapy was convened to identify and prioritise opportunities and challenges for radiotherapy and immunotherapy combinations. Sessions examined the immune effects of radiation, barriers to anti-tumour immune response, previous clinical trial data, immunological and computational assessment of response, and next-generation radiotherapy-immunotherapy combinations. Panel recommendations included: developing and implementing patient selection and biomarker-guided approaches; applying mechanistic understanding to optimise delivery of radiotherapy and selection of immunotherapies; using rigorous preclinical models including companion animal studies; embracing data sharing and standardisation, advanced modelling, and multidisciplinary cross-institution collaboration; interrogating clinical data, including negative trials; and incorporating novel clinical endpoints and trial designs.
Collapse
Affiliation(s)
- Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Sandra Demaria
- Weill Cornell Medicine, Department of Radiation Oncology, New York, NY, USA
| | - Arta M Monjazeb
- UC Davis Health, Department of Radiation Oncology, Sacramento, CA, USA
| | - Silvia C Formenti
- Weill Cornell Medicine, Department of Radiation Oncology, New York, NY, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA
| | - James Welsh
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Heiko Enderling
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Joshua D Brody
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Heather M McGee
- Department of Radiation Oncology and Department of Immuno-Oncology, City of Hope, Duarte, CA, USA
| | - Michele Mondini
- Gustave Roussy, Université Paris-Saclay, INSERM U1030, Villejuif, France
| | - Michael S Kent
- Davis School of Veterinary Medicine, University of California, Davis, CA, USA
| | | | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Joe Y Chang
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Mai Anh Huynh
- Brigham and Women's Hospital-Dana-Farber Cancer Institute, Boston, MA, USA
| | - Adi Daib
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Sean Pitroda
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA
| | - Caroline Chung
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Raphael Serre
- Aix Marseille University, SMARTc Unit, Inserm S 911 CRO2, Marseille, France
| | | | - Jie Deng
- Department of Radiation Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Quaovi H Sodji
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Anthony T Nguyen
- Cedars-Sinai Medical Center, Department of Radiation Oncology, Los Angeles, CA, USA
| | - Ravi B Patel
- Department of Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA, USA
| | - Simone Krebs
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medicine, Department of Radiology, New York, NY, USA
| | - Anusha Kalbasi
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, USA
| | - Caroline Kerr
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Logan Vick
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | | | - Irene M Ong
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Fernanda Herrera
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland
| | - Hari Menon
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - DeeDee Smart
- Radiation Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Jalal Ahmed
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robyn D Gartrell
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA; Department of Oncology, Division of Pediatric Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Christina L Roland
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Fatemeh Fekrmandi
- Department of Radiation Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Binita Chakraborty
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Eric H Bent
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tracy J Berg
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Alan Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Samir Khleif
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Andrew G Sikora
- Department of Head and Neck Surgery, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Lawrence Fong
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| |
Collapse
|
5
|
Kut C, Quon H, Chen XS. Emerging Radiotherapy Technologies for Head and Neck Squamous Cell Carcinoma: Challenges and Opportunities in the Era of Immunotherapy. Cancers (Basel) 2024; 16:4150. [PMID: 39766050 PMCID: PMC11674243 DOI: 10.3390/cancers16244150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Radiotherapy (RT) is an integral component in the multidisciplinary management of patients with head and neck squamous cell carcinoma (HNSCC). Significant advances have been made toward optimizing tumor control and toxicity profiles of RT for HNSCC in the past two decades. The development of intensity modulated radiotherapy (IMRT) and concurrent chemotherapy established the standard of care for most patients with locally advanced HNSCC around the turn of the century. More recently, selective dose escalation to the most radioresistant part of tumor and avoidance of the most critical substructures of organs at risk, often guided by functional imaging, allowed even further improvement in the therapeutic ratio of IMRT. Other highly conformal RT modalities, including intensity modulated proton therapy (IMPT) and stereotactic body radiotherapy (SBRT) are being increasingly utilized, although there are gaps in our understanding of the normal tissue complication probabilities and their relative biological effectiveness. There is renewed interest in spatially fractionated radiotherapy (SFRT), such as GRID and LATTICE radiotherapy, in both palliative and definitive settings. The emergence of immune checkpoint inhibitors (ICIs) has revolutionized the treatment of patients with recurrent and metastatic HNSCC. Novel RT modalities, including IMPT, SBRT, and SFRT, have the potential to reduce lymphopenia and immune suppression, stimulate anti-tumor immunity, and synergize with ICIs. The next frontier in the treatment of HNSCC may lie in the exploration of combined modality treatment with new RT technologies and ICIs.
Collapse
Affiliation(s)
- Carmen Kut
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD 21287, USA; (C.K.); (H.Q.)
| | - Harry Quon
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD 21287, USA; (C.K.); (H.Q.)
| | - Xuguang Scott Chen
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Grams MP, Mateus CQ, Mashayekhi M, Mutter RW, Djonov V, Fazzari JM, Xiao H, Frechette KM, Wentworth AJ, Morris JM, Klebel B, Thull JC, Guenzel RM, Wismayer DJS, Lucien F, Park SS, Lester SC. Minibeam Radiation Therapy Treatment (MBRT): Commissioning and First Clinical Implementation. Int J Radiat Oncol Biol Phys 2024; 120:1423-1434. [PMID: 39002850 DOI: 10.1016/j.ijrobp.2024.06.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/22/2024] [Accepted: 06/29/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE Minibeam radiation therapy (MBRT) is characterized by the delivery of submillimeter-wide regions of high "peak" and low "valley" doses throughout a tumor. Preclinical studies have long shown the promise of this technique, and we report here the first clinical implementation of MBRT. METHODS AND MATERIALS A clinical orthovoltage unit was commissioned for MBRT patient treatments using 3-, 4-, 5-, 8-, and 10-cm diameter cones. The 180 kVp output was spatially separated into minibeams using a tungsten collimator with 0.5 mm wide slits spaced 1.1 mm on center. Percentage depth dose (PDD) measurements were obtained using film dosimetry and plastic water for both peak and valley doses. PDDs were measured on the central axis for offsets of 0, 0.5, and 1 cm. The peak-to-valley ratio was calculated at each depth for all cones and offsets. To mitigate the effects of patient motion on delivered dose, patient-specific 3-dimensional-printed collimator holders were created. These conformed to the unique anatomy of each patient and affixed the tungsten collimator directly to the body. Two patients were treated with MBRT; both received 2 fractions. RESULTS Peak PDDs decreased gradually with depth. Valley PDDs initially increased slightly with depth, then decreased gradually beyond 2 cm. The peak-to-valley ratios were highest at the surface for smaller cone sizes and offsets. In vivo film dosimetry confirmed a distinct delineation of peak and valley doses in both patients treated with MBRT with no dose blurring. Both patients experienced prompt improvement in symptoms and tumor response. CONCLUSIONS We report commissioning results, treatment processes, and the first 2 patients treated with MBRT using a clinical orthovoltage unit. While demonstrating the feasibility of this approach is a crucial first step toward wider translation, clinical trials are needed to further establish safety and efficacy.
Collapse
Affiliation(s)
- Michael P Grams
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | | | | | - Robert W Mutter
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | | | | | - Huaping Xiao
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Brandon Klebel
- Division of Engineering, Mayo Clinic, Rochester, Minnesota
| | - Jack C Thull
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Fabrice Lucien
- Department of Urology, Mayo Clinic, Rochester, Minnesota; Division of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Sean S Park
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Scott C Lester
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
7
|
Bryan JN, Maitz CA. Translational History and Hope of Immunotherapy of Canine Tumors. Clin Cancer Res 2024; 30:4272-4285. [PMID: 39042399 PMCID: PMC11444889 DOI: 10.1158/1078-0432.ccr-23-2266] [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: 02/29/2024] [Revised: 05/13/2024] [Accepted: 07/01/2024] [Indexed: 07/24/2024]
Abstract
Companion dogs have served an important role in cancer immunotherapy research. Sharing similar environments and diets with humans, dogs naturally develop many of the same cancers. These shared exposures, coupled with dogs' diverse genetic makeup, make them ideal subjects for studying cancer therapies. Tumors like osteosarcoma, hemangiosarcoma, soft-tissue sarcoma, and non-Hodgkin lymphoma occur with greater frequency than their counterpart disease in humans. Canine brain tumors allow the study of therapy strategies with imaging, surgery, and radiotherapy equipment in veterinary patients with near-human geometry. Nonspecific immunostimulants, autologous and allogeneic vaccines, immune checkpoint inhibitors, and cellular therapies used in treating canine cancers have been tested in veterinary clinical trials. These treatments have not only improved outcomes for dogs but have also provided valuable insights for human cancer treatment. Advancements in radiation technology and the development of tools to characterize canine immune responses have further facilitated the ability to translate veterinary clinical trial results to human applications. Advancements in immunotherapy of canine tumors have directly supported translation to human clinical trials leading to approved therapies for patients with cancer around the world. The study of immunotherapy in dogs has been and will continue to be a promising avenue for advancing human cancer treatment.
Collapse
Affiliation(s)
- Jeffrey N. Bryan
- Comparative Oncology, Radiobiology, and Epigenetics Laboratory, Department of Veterinary Medicine and Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Charles A. Maitz
- Comparative Oncology, Radiobiology, and Epigenetics Laboratory, Department of Veterinary Medicine and Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| |
Collapse
|
8
|
Jagodinsky JC, Vera JM, Jin WJ, Shea AG, Clark PA, Sriramaneni RN, Havighurst TC, Chakravarthy I, Allawi RH, Kim K, Harari PM, Sondel PM, Newton MA, Crittenden MR, Gough MJ, Miller JR, Ong IM, Morris ZS. Intratumoral radiation dose heterogeneity augments antitumor immunity in mice and primes responses to checkpoint blockade. Sci Transl Med 2024; 16:eadk0642. [PMID: 39292804 PMCID: PMC11522033 DOI: 10.1126/scitranslmed.adk0642] [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: 07/31/2023] [Revised: 04/03/2024] [Accepted: 08/08/2024] [Indexed: 09/20/2024]
Abstract
Radiation therapy (RT) activates multiple immunologic effects in the tumor microenvironment (TME), with diverse dose-response relationships observed. We hypothesized that, in contrast with homogeneous RT, a heterogeneous RT dose would simultaneously optimize activation of multiple immunogenic effects in a single TME, resulting in a more effective antitumor immune response. Using high-dose-rate brachytherapy, we treated mice bearing syngeneic tumors with a single fraction of heterogeneous RT at a dose ranging from 2 to 30 gray. When combined with dual immune checkpoint inhibition in murine models, heterogeneous RT generated more potent antitumor responses in distant, nonirradiated tumors compared with any homogeneous dose. The antitumor effect after heterogeneous RT required CD4 and CD8 T cells and low-dose RT to a portion of the tumor. At the 3-day post-RT time point, dose heterogeneity imprinted the targeted TME with spatial differences in immune-related gene expression, antigen presentation, and susceptibility of tumor cells to immune-mediated destruction. At a later 10-day post-RT time point, high-, moderate-, or low-RT-dose regions demonstrated distinct infiltrating immune cell populations. This was associated with an increase in the expression of effector-associated cytokines in circulating CD8 T cells. Consistent with enhanced adaptive immune priming, heterogeneous RT promoted clonal expansion of effector CD8 T cells. These findings illuminate the breadth of dose-dependent effects of RT on the TME and the capacity of heterogeneous RT to promote antitumor immunity when combined with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Justin C. Jagodinsky
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Jessica M. Vera
- Department of Statistics and Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
- Sage Bionetworks, 2901 Third Ave. Suite 330, Seattle, WA 98121, USA
| | - Won Jong Jin
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Amanda G. Shea
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Paul A. Clark
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Raghava N. Sriramaneni
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Thomas C. Havighurst
- Department of Statistics and Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Ishan Chakravarthy
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Raad H. Allawi
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - KyungMann Kim
- Department of Statistics and Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Paul M. Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Paul M. Sondel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Michael A. Newton
- Department of Statistics and Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Marka R. Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR 97213, USA
- Oregon Clinic, Portland, OR 97232, USA
| | - Michael J. Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR 97213, USA
| | - Jessica R. Miller
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Irene M. Ong
- Department of Statistics and Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
9
|
Bekker RA, Obertopp N, Redler G, Penagaricano J, Caudell JJ, Yamoah K, Pilon-Thomas S, Moros EG, Enderling H. Spatially fractionated GRID radiation potentiates immune-mediated tumor control. Radiat Oncol 2024; 19:121. [PMID: 39272128 PMCID: PMC11401399 DOI: 10.1186/s13014-024-02514-6] [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: 02/06/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Tumor-immune interactions shape a developing tumor and its tumor immune microenvironment (TIME) resulting in either well-infiltrated, immunologically inflamed tumor beds, or immune deserts with low levels of infiltration. The pre-treatment immune make-up of the TIME is associated with treatment outcome; immunologically inflamed tumors generally exhibit better responses to radio- and immunotherapy than non-inflamed tumors. However, radiotherapy is known to induce opposing immunological consequences, resulting in both immunostimulatory and inhibitory responses. In fact, it is thought that the radiation-induced tumoricidal immune response is curtailed by subsequent applications of radiation. It is thus conceivable that spatially fractionated radiotherapy (SFRT), administered through GRID blocks (SFRT-GRID) or lattice radiotherapy to create areas of low or high dose exposure, may create protective reservoirs of the tumor immune microenvironment, thereby preserving anti-tumor immune responses that are pivotal for radiation success. METHODS We have developed an agent-based model (ABM) of tumor-immune interactions to investigate the immunological consequences and clinical outcomes after 2 Gy × 35 whole tumor radiation therapy (WTRT) and SFRT-GRID. The ABM is conceptually calibrated such that untreated tumors escape immune surveillance and grow to clinical detection. Individual ABM simulations are initialized from four distinct multiplex immunohistochemistry (mIHC) slides, and immune related parameter rates are generated using Latin Hypercube Sampling. RESULTS In silico simulations suggest that radiation-induced cancer cell death alone is insufficient to clear a tumor with WTRT. However, explicit consideration of radiation-induced anti-tumor immunity synergizes with radiation cytotoxicity to eradicate tumors. Similarly, SFRT-GRID is successful with radiation-induced anti-tumor immunity, and, for some pre-treatment TIME compositions and modeling parameters, SFRT-GRID might be superior to WTRT in providing tumor control. CONCLUSION This study demonstrates the pivotal role of the radiation-induced anti-tumor immunity. Prolonged fractionated treatment schedules may counteract early immune recruitment, which may be protected by SFRT-facilitated immune reservoirs. Different biological responses and treatment outcomes are observed based on pre-treatment TIME composition and model parameters. A rigorous analysis and model calibration for different tumor types and immune infiltration states is required before any conclusions can be drawn for clinical translation.
Collapse
Affiliation(s)
- Rebecca A Bekker
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, 33612, USA
| | - Nina Obertopp
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, 33612, USA
| | - Gage Redler
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - José Penagaricano
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Jimmy J Caudell
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Kosj Yamoah
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Shari Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Eduardo G Moros
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Heiko Enderling
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Gaudreault M, Yu KK, Chang D, Kron T, Hardcastle N, Chander S, Yeo A. Automated lattice radiation therapy treatment planning personalised to tumour size and shape. Phys Med 2024; 125:104490. [PMID: 39142028 DOI: 10.1016/j.ejmp.2024.104490] [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: 02/20/2024] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024] Open
Abstract
PURPOSE Lattice radiation therapy (LRT) alternates regions of high and low doses inside the tumour. Whilst this technique reported positive results in tumour size reduction, optimal lattice parameters are still unknown. We introduce an automated LRT planning method personalised to tumour shape and designed to allow investigation of lattice geometry. METHODS Patients with retroperitoneal sarcoma were considered for inclusion. Automation was performed with the Eclipse Scripting Application Interface (v16, Varian Medical Systems, Palo Alto). By iterating over vertex size (V) and centre-to-centre distance (D), vertices were segmented within the gross tumour volume (GTV) in an alternating square pattern. Iterations stopped when the number of inserted vertices was contained between a prespecified lower and upper bound. Forty sets of lattices were considered, produced by varying V and D in five lower/upper bound pairs. Best-scoring sets were determined with a score favouring the maximization of GTV dose uniformity and heterogeneity whilst minimizing the maximum dose to organs at risk. RESULTS Fifty patients with tumour volumes between 150 cm3 and 10,000 cm3 were included. Best-scoring sets were characterised by a low number of vertices (<15). Based on the best-scoring set, the predicted parameters to use for new patients were V = 0.19 (GTV volume)1/3 and D = 2V, in centimetres. The number of vertices (N) to insert in the GTV can be estimated with N ≤ (24 × 3% GTV volume)/(4πV3). CONCLUSIONS The automated LRT treatment planning personalised to tumour size allows investigation of lattice geometry over a large range of GTV volumes.
Collapse
Affiliation(s)
- Mathieu Gaudreault
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia; Sir Peter MacCallum, Department of Oncology, the University of Melbourne, Victoria 3000, Australia.
| | - Kelvin K Yu
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia; Benavides Cancer Institute - University of Santo Tomas Hospital, Manila, Philippines
| | - David Chang
- Sir Peter MacCallum, Department of Oncology, the University of Melbourne, Victoria 3000, Australia; Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Tomas Kron
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia; Sir Peter MacCallum, Department of Oncology, the University of Melbourne, Victoria 3000, Australia; Centre for Medical Radiation Physics, University of Wollongong, NSW 2522, Australia
| | - Nicholas Hardcastle
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia; Sir Peter MacCallum, Department of Oncology, the University of Melbourne, Victoria 3000, Australia; Centre for Medical Radiation Physics, University of Wollongong, NSW 2522, Australia
| | - Sarat Chander
- Sir Peter MacCallum, Department of Oncology, the University of Melbourne, Victoria 3000, Australia; Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Adam Yeo
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia; Sir Peter MacCallum, Department of Oncology, the University of Melbourne, Victoria 3000, Australia
| |
Collapse
|
11
|
Lekmeechai S, Pietras K, Axelsson O. 177Lu-SN201 nanoparticle shows superior anti-tumor efficacy over conventional cancer drugs in 4T1 orthotopic model. Invest New Drugs 2024; 42:471-477. [PMID: 38837077 PMCID: PMC11327194 DOI: 10.1007/s10637-024-01450-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
In the current in-vivo study we demonstrate the potential of the radiolabeled nanoparticle 177Lu-SN201 as an effective anticancer treatment, as evidenced by significantly prolonged survival and reduced tumor burden in the aggressive, triple negative 4T1 murine breast cancer model. We show with high statistical significance that 177Lu-SN201 is superior at suppressing the tumor growth not only compared to vehicle but also to the commonly used cancer drugs paclitaxel, niraparib, carboplatin, and the combination of the immune checkpoint inhibitors anti PD-1 and anti-CTLA-4. The dosing of the standard drugs were based on examples in the literature where good effects have been seen in various mouse models. The treatment is reasonably well-tolerated, as indicated by clinical chemistry of liver and renal function through the measurement of glutamate pyruvate alanine aminotransferase, alanine amino transferase, blood urea nitrogen, and creatinine levels in plasma samples, despite some weight loss. Overall, 177Lu-SN201 presents as a promising therapeutic candidate for cancer treatment.
Collapse
Affiliation(s)
| | - Kristian Pietras
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Bldg 404, Lund, SE-223 81, Sweden
| | - Oskar Axelsson
- Spago Nanomedical AB, Scheelevägen 22, Lund, SE-223 63, Sweden.
| |
Collapse
|
12
|
Takashima ME, Berg TJ, Morris ZS. The Effects of Radiation Dose Heterogeneity on the Tumor Microenvironment and Anti-Tumor Immunity. Semin Radiat Oncol 2024; 34:262-271. [PMID: 38880534 DOI: 10.1016/j.semradonc.2024.04.004] [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: 06/18/2024]
Abstract
Radiotherapy elicits dose- and lineage-dependent effects on immune cell survival, migration, activation, and proliferation in targeted tumor microenvironments. Radiation also stimulates phenotypic changes that modulate the immune susceptibility of tumor cells. This has raised interest in using radiotherapy to promote greater response to immunotherapies. To clarify the potential of such combinations, it is critical to understand how best to administer radiation therapy to achieve activation of desired immunologic mechanisms. In considering the multifaceted process of priming and propagating anti-tumor immune response, radiation dose heterogeneity emerges as a potential means for simultaneously engaging diverse dose-dependent effects in a single tumor environment. Recent work in spatially fractionated external beam radiation therapy demonstrates the expansive immune responses achievable when a range of high to low dose radiation is delivered in a tumor. Brachytherapy and radiopharmaceutical therapies deliver inherently heterogeneous distributions of radiation that may contribute to immunogenicity. This review evaluates the interplay of radiation dose and anti-tumor immune response and explores emerging methodological approaches for investigating the effects of heterogeneous dose distribution on immune responses.
Collapse
Affiliation(s)
- Maya E Takashima
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Tracy J Berg
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| |
Collapse
|
13
|
Sheikh K, Li H, Wright JL, Yanagihara TK, Halthore A. The Peaks and Valleys of Photon Versus Proton Spatially Fractionated Radiotherapy. Semin Radiat Oncol 2024; 34:292-301. [PMID: 38880538 DOI: 10.1016/j.semradonc.2024.04.007] [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: 06/18/2024]
Abstract
Spatially-fractionated radiotherapy (SFRT) delivers high doses to small areas of tumor while sparing adjacent tissue, including intervening disease. In this review, we explore the evolution of SFRT technological advances, contrasting approaches with photon and proton beam radiotherapy. We discuss unique dosimetric considerations and physical properties of SFRT, as well as review the preclinical literature that provides an emerging understanding of biological mechanisms. We emphasize crucial areas of future study and highlight clinical trials that are underway to assess SFRT's safety and efficacy, with a focus on immunotherapeutic synergies. The review concludes with practical considerations for SFRT's clinical application, advocating for strategies that leverage its unique dosimetric and biological properties for improved patient outcomes.
Collapse
Affiliation(s)
- Khadija Sheikh
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Radiation Oncology, The Johns Hopkins Proton Center, Washington, DC.
| | - Heng Li
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Radiation Oncology, The Johns Hopkins Proton Center, Washington, DC
| | - Jean L Wright
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Radiation Oncology, The Johns Hopkins Proton Center, Washington, DC
| | - Theodore K Yanagihara
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Aditya Halthore
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Radiation Oncology, The Johns Hopkins Proton Center, Washington, DC
| |
Collapse
|
14
|
Jenkins SV, Johnsrud AJ, Dings RPM, Griffin RJ. Bystander Effects in Spatially Fractionated Radiation Therapy: From Molecule To Organism To Clinical Implications. Semin Radiat Oncol 2024; 34:284-291. [PMID: 38880537 PMCID: PMC11185274 DOI: 10.1016/j.semradonc.2024.05.004] [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] [Indexed: 06/18/2024]
Abstract
The standard of care for radiation therapy is numerous, low-dose fractions that are distributed homogeneously throughout the tumor. An alternative strategy under scrutiny is to apply spatially fractionated radiotherapy (high and low doses throughout the tumor) in one or several fractions, either alone or followed by conventional radiation fractionation . Spatial fractionation allows for significant sparing of normal tissue, and the regions of tumor or normal tissue that received sublethal doses can give rise to beneficial bystander effects in both cases. Bystander effects are broadly defined as biological responses that are significantly greater than would be anticipated based on the radiation dose received. Typically these effects are initiated by diffusion of reactive oxygen species and secretion of various cytokines. As demonstrated in the literature, spatial fractionation related bystander effects can occur locally from cell to cell and in what are known as "cohort effects," which tend to take the form of restructuring of the vasculature, enhanced immune infiltration, and development of immunological memory. Other bystander effects can take place at distant sites in what are known as "abscopal effects." While these events are rare, they are mediated by the immune system and can result in the eradication of secondary and metastatic disease. Currently, due to the complexity and variability of these bystander effects, they are not thoroughly understood, but as knowledge improves they may present significant opportunities for improved clinical outcomes.
Collapse
Affiliation(s)
- Samir V Jenkins
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR..
| | | | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR
| |
Collapse
|
15
|
Parisi S, Sciacca M, Critelli P, Ferrantelli G, Chillari F, Venuti V, Napoli C, Shteiwi I, Siragusa C, Brogna A, Pontoriero A, Ferini G, Santacaterina A, Pergolizzi S. Lattice radiotherapy in inflammatory breast cancer: report of a first case treated with curative aim. Radiat Oncol J 2024; 42:160-165. [PMID: 38946079 PMCID: PMC11215509 DOI: 10.3857/roj.2024.00038] [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: 01/08/2024] [Revised: 01/30/2024] [Accepted: 02/16/2024] [Indexed: 07/02/2024] Open
Abstract
Inflammatory breast cancer (IBC) is a rare, aggressive form of breast cancer characterized by poor prognosis. The treatment requires a multidisciplinary approach, with neoadjuvant chemotherapy, surgery, and radiation therapy (RT). Particularly, high doses of conventional RT have been historically delivered in the adjuvant setting after chemotherapy and mastectomy or as radical treatment in patients ineligible for surgery. Here, we report the case of a 49-year-old woman patient with IBC unsuitable for surgery and treated with a combination of lattice RT and fractionated external beam RT concurrent with trastuzumab, with a curative aim. One year after RT, the patient showed a complete response and tolerable toxicities. This is the first reported case of a not-operable IBC patient treated with this particular kind of RT.
Collapse
Affiliation(s)
- Silvana Parisi
- Radiotherapy Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Miriam Sciacca
- Radiotherapy Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Paola Critelli
- Radiotherapy Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Giacomo Ferrantelli
- Radiotherapy Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Federico Chillari
- Radiotherapy Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Valeria Venuti
- Radiotherapy Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Claudio Napoli
- Radiotherapy Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Issa Shteiwi
- Radiotherapy Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Carmelo Siragusa
- Medical Physics Unit, University Hospital of Messina, Messina, Italy
| | - Anna Brogna
- Medical Physics Unit, University Hospital of Messina, Messina, Italy
| | - Antonio Pontoriero
- Radiotherapy Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | | | | | - Stefano Pergolizzi
- Radiotherapy Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| |
Collapse
|
16
|
Prezado Y, Grams M, Jouglar E, Martínez-Rovira I, Ortiz R, Seco J, Chang S. Spatially fractionated radiation therapy: a critical review on current status of clinical and preclinical studies and knowledge gaps. Phys Med Biol 2024; 69:10TR02. [PMID: 38648789 DOI: 10.1088/1361-6560/ad4192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/22/2024] [Indexed: 04/25/2024]
Abstract
Spatially fractionated radiation therapy (SFRT) is a therapeutic approach with the potential to disrupt the classical paradigms of conventional radiation therapy. The high spatial dose modulation in SFRT activates distinct radiobiological mechanisms which lead to a remarkable increase in normal tissue tolerances. Several decades of clinical use and numerous preclinical experiments suggest that SFRT has the potential to increase the therapeutic index, especially in bulky and radioresistant tumors. To unleash the full potential of SFRT a deeper understanding of the underlying biology and its relationship with the complex dosimetry of SFRT is needed. This review provides a critical analysis of the field, discussing not only the main clinical and preclinical findings but also analyzing the main knowledge gaps in a holistic way.
Collapse
Affiliation(s)
- Yolanda Prezado
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, F-91400, Orsay, France
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, F-91400, Orsay, France
- New Approaches in Radiotherapy Lab, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña, E-15706, Spain
- Oportunius Program, Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, A Coruña, Spain
| | - Michael Grams
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, United States of America
| | - Emmanuel Jouglar
- Institut Curie, PSL Research University, Department of Radiation Oncology, F-75005, Paris and Orsay Protontherapy Center, F-91400, Orsay, France
| | - Immaculada Martínez-Rovira
- Physics Department, Universitat Auto`noma de Barcelona, E-08193, Cerdanyola del Valle`s (Barcelona), Spain
| | - Ramon Ortiz
- University of California San Francisco, Department of Radiation Oncology, 1600 Divisadero Street, San Francisco, CA 94143, United States of America
| | - Joao Seco
- Division of Biomedical physics in Radiation Oncology, DKFZ-German Cancer Research Center, Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, Heidelberg, Germany
| | - Sha Chang
- Dept of Radiation Oncology and Department of Biomedical Engineering, University of North Carolina School of Medicine, United States of America
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolin State University, United States of America
| |
Collapse
|
17
|
Wisdom AJ, Barker CA, Chang JY, Demaria S, Formenti S, Grassberger C, Gregucci F, Hoppe BS, Kirsch DG, Marciscano AE, Mayadev J, Mouw KW, Palta M, Wu CC, Jabbour SK, Schoenfeld JD. The Next Chapter in Immunotherapy and Radiation Combination Therapy: Cancer-Specific Perspectives. Int J Radiat Oncol Biol Phys 2024; 118:1404-1421. [PMID: 38184173 DOI: 10.1016/j.ijrobp.2023.12.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/20/2023] [Accepted: 12/30/2023] [Indexed: 01/08/2024]
Abstract
Immunotherapeutic agents have revolutionized cancer treatment over the past decade. However, most patients fail to respond to immunotherapy alone. A growing body of preclinical studies highlights the potential for synergy between radiation therapy and immunotherapy, but the outcomes of clinical studies have been mixed. This review summarizes the current state of immunotherapy and radiation combination therapy across cancers, highlighting existing challenges and promising areas for future investigation.
Collapse
Affiliation(s)
- Amy J Wisdom
- Harvard Radiation Oncology Program, Boston, Massachusetts
| | - Christopher A Barker
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joe Y Chang
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Clemens Grassberger
- Department of Radiation Oncology, University of Washington, Fred Hutch Cancer Center, Seattle, Washington
| | - Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Bradford S Hoppe
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida
| | - David G Kirsch
- Department of Radiation Oncology, University of Toronto, Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ariel E Marciscano
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jyoti Mayadev
- Department of Radiation Oncology, UC San Diego School of Medicine, San Diego, California
| | - Kent W Mouw
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Manisha Palta
- Department of Radiation Oncology, Duke Cancer Center, Durham, North Carolina
| | - Cheng-Chia Wu
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
| | - Jonathan D Schoenfeld
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
18
|
Mayr NA, Mohiuddin M, Snider JW, Zhang H, Griffin RJ, Amendola BE, Hippe DS, Perez NC, Wu X, Lo SS, Regine WF, Simone CB. Practice Patterns of Spatially Fractionated Radiation Therapy: A Clinical Practice Survey. Adv Radiat Oncol 2024; 9:101308. [PMID: 38405319 PMCID: PMC10885580 DOI: 10.1016/j.adro.2023.101308] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/26/2023] [Indexed: 02/27/2024] Open
Abstract
Purpose Spatially fractionated radiation therapy (SFRT) is increasingly used for bulky advanced tumors, but specifics of clinical SFRT practice remain elusive. This study aimed to determine practice patterns of GRID and Lattice radiation therapy (LRT)-based SFRT. Methods and Materials A survey was designed to identify radiation oncologists' practice patterns of patient selection for SFRT, dosing/planning, dosimetric parameter use, SFRT platforms/techniques, combinations of SFRT with conventional external beam radiation therapy (cERT) and multimodality therapies, and physicists' technical implementation, delivery, and quality procedures. Data were summarized using descriptive statistics. Group comparisons were analyzed with permutation tests. Results The majority of practicing radiation oncologists (United States, 100%; global, 72.7%) considered SFRT an accepted standard-of-care radiation therapy option for bulky/advanced tumors. Treatment of metastases/recurrences and nonmetastatic primary tumors, predominantly head and neck, lung cancer and sarcoma, was commonly practiced. In palliative SFRT, regimens of 15 to 18 Gy/1 fraction predominated (51.3%), and in curative-intent treatment of nonmetastatic tumors, 15 Gy/1 fraction (28.0%) and fractionated SFRT (24.0%) were most common. SFRT was combined with cERT commonly but not always in palliative (78.6%) and curative-intent (85.7%) treatment. SFRT-cERT time sequencing and cERT dose adjustments were variable. In curative-intent treatment, concurrent chemotherapy and immunotherapy were found acceptable by 54.5% and 28.6%, respectively. Use of SFRT dosimetric parameters was highly variable and differed between GRID and LRT. SFRT heterogeneity dosimetric parameters were more commonly used (P = .008) and more commonly thought to influence local control (peak dose, P = .008) in LRT than in GRID therapy. Conclusions SFRT has already evolved as a clinical practice pattern for advanced/bulky tumors. Major treatment approaches are consistent and follow the literature, but SFRT-cERT combination/sequencing and clinical utilization of dosimetric parameters are variable. These areas may benefit from targeted education and standardization, and knowledge gaps may be filled by incorporating identified inconsistencies into future clinical research.
Collapse
Affiliation(s)
- Nina A. Mayr
- College of Human Medicine, Michigan State University, East Lansing, Michigan
| | - Majid Mohiuddin
- Radiation Oncology Consultants and Northwestern Proton Center, Warrenville, Illinois
| | - James W. Snider
- Radiation Oncology, South Florida Proton Therapy Institute, Delray Beach, Florida
| | - Hualin Zhang
- Department of Radiation Oncology, University of Southern California, Los Angeles, California
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | | | - Xiaodong Wu
- Executive Medical Physics Associates, Miami, Florida
| | - Simon S. Lo
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, Washington
| | - William F. Regine
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Charles B. Simone
- Department of Radiation Oncology, New York Proton Center, New York, New York
| |
Collapse
|
19
|
Lukas L, Zhang H, Cheng K, Epstein A. Immune Priming with Spatially Fractionated Radiation Therapy. Curr Oncol Rep 2023; 25:1483-1496. [PMID: 37979032 PMCID: PMC10728252 DOI: 10.1007/s11912-023-01473-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE OF REVIEW This review aims to summarize the current preclinical and clinical evidence of nontargeted immune effects of spatially fractionated radiation therapy (SFRT). We then highlight strategies to augment the immunomodulatory potential of SFRT in combination with immunotherapy (IT). RECENT FINDINGS The response of cancer to IT is limited by primary and acquired immune resistance, and strategies are needed to prime the immune system to increase the efficacy of IT. Radiation therapy can induce immunologic effects and can potentially be used to synergize the effects of IT, although the optimal combination of radiation and IT is largely unknown. SFRT is a novel radiation technique that limits ablative doses to tumor subvolumes, and this highly heterogeneous dose deposition may increase the immune-rich infiltrate within the targeted tumor with enhanced antigen presentation and activated T cells in nonirradiated tumors. The understanding of nontargeted effects of SFRT can contribute to future translational strategies to combine SFRT and IT. Integration of SFRT and IT is an innovative approach to address immune resistance to IT with the overall goal of improving the therapeutic ratio of radiation therapy and increasing the efficacy of IT.
Collapse
Affiliation(s)
- Lauren Lukas
- Department of Radiation Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Hualin Zhang
- Department of Radiation Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Karen Cheng
- Department of Radiation Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Alan Epstein
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
20
|
Strati P, Spiotto MT. Incorporating Immunotherapy with Radiotherapy for Lymphomas. LYMPHATICS 2023; 1:273-286. [PMID: 39917366 PMCID: PMC11800356 DOI: 10.3390/lymphatics1030018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Radiotherapy and/or chemotherapy have been used for nearly 100 years to treat lymphoma. Recently, immunotherapy has been incorporated into the treatment of lymphomas. Here, we will review both the role of immunotherapy in lymphoma as well as the feasibility of incorporating immunotherapies with conventional lymphoma treatments, especially radiotherapy. Immunotherapy agents include checkpoint inhibitors that target the PD-1/PD-L1 axis, CTLA-4, or CD47. In addition, other immunotherapy agents such as bi-specific antibodies and CD19 CAR-T cell therapy are being implemented in various non-Hodgkin's lymphomas. Extrapolating from observations in other disease sites and incorporating immunotherapy with conventional treatments of lymphoma, including radiotherapy, may have opposing effects. Radiotherapy may stimulate anti-tumor immune responses that synergize with immunotherapies. In contrast, radiotherapy, as well as chemotherapy, may also induce local and systemic immune dysfunction which reduces the efficacy of immunotherapies. With newer radiation treatment techniques and limited radiation fields, it is likely that the efficacy of immunotherapy can be maintained when included with conventional treatments. Therefore, there remains an unmet need to better understand the role of immunotherapy alone and in combination with current treatments in lymphoma patients.
Collapse
Affiliation(s)
- Paolo Strati
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael T. Spiotto
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
21
|
Zhang H, Mi J, Xin Q, Cao W, Song C, Zhang N, Yuan C. Recent research and clinical progress of CTLA-4-based immunotherapy for breast cancer. Front Oncol 2023; 13:1256360. [PMID: 37860188 PMCID: PMC10582933 DOI: 10.3389/fonc.2023.1256360] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023] Open
Abstract
Breast cancer is characterized by a high incidence rate and its treatment challenges, particularly in certain subtypes. Consequently, there is an urgent need for the development of novel therapeutic approaches. Immunotherapy utilizing immune checkpoint inhibitors (ICIs) is currently gaining momentum for the treatment of breast cancer. Substantial progress has been made in clinical studies employing cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) inhibitors for breast cancer, but the cure rates are relatively low. To improve the efficacy of CTLA-4-based therapy for breast cancer, further research is imperative to explore more effective immune-based treatment strategies. In addition to monotherapy, CTLA-4 inhibitors are also being investigated in combination with other ICIs or alternative medications. However, it should be noted that immune-based treatments may cause adverse events. This review focuses on the mechanisms of CTLA-4 inhibitor monotherapy or combination therapy in breast cancer. We systematically summarize the latest research and clinical advances in CTLA-4-based immunotherapy for breast cancer, providing new perspectives on the treatment of breast cancer. In addition, this review highlights the immune-related adverse events (irAEs) associated with CTLA-4 inhibitors, providing insights into the development of appropriate clinical tumor immunotherapy regimens and intervention strategies.
Collapse
Affiliation(s)
- Hongsheng Zhang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jintao Mi
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qi Xin
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weiwei Cao
- Department of Clinical Laboratory, People’s Hospital of Deyang City, Deyang, China
| | - Chunjiao Song
- Department of Clinical Laboratory, People’s Hospital of Deyang City, Deyang, China
| | - Naidan Zhang
- Department of Clinical Laboratory, People’s Hospital of Deyang City, Deyang, China
| | - Chengliang Yuan
- Department of Clinical Laboratory, People’s Hospital of Deyang City, Deyang, China
| |
Collapse
|
22
|
Grams MP, Deufel CL, Kavanaugh JA, Corbin KS, Ahmed SK, Haddock MG, Lester SC, Ma DJ, Petersen IA, Finley RR, Lang KG, Spreiter SS, Park SS, Owen D. Clinical aspects of spatially fractionated radiation therapy treatments. Phys Med 2023; 111:102616. [PMID: 37311338 DOI: 10.1016/j.ejmp.2023.102616] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/06/2023] [Accepted: 05/30/2023] [Indexed: 06/15/2023] Open
Abstract
PURPOSE To provide clinical guidance for centers wishing to implement photon spatially fractionated radiation therapy (SFRT) treatments using either a brass grid or volumetric modulated arc therapy (VMAT) lattice approach. METHODS We describe in detail processes which have been developed over the course of a 3-year period during which our institution treated over 240 SFRT cases. The importance of patient selection, along with aspects of simulation, treatment planning, quality assurance, and treatment delivery are discussed. Illustrative examples involving clinical cases are shown, and we discuss safety implications relevant to the heterogeneous dose distributions. RESULTS SFRT can be an effective modality for tumors which are otherwise challenging to manage with conventional radiation therapy techniques or for patients who have limited treatment options. However, SFRT has several aspects which differ drastically from conventional radiation therapy treatments. Therefore, the successful implementation of an SFRT treatment program requires the multidisciplinary expertise and collaboration of physicians, physicists, dosimetrists, and radiation therapists. CONCLUSIONS We have described methods for patient selection, simulation, treatment planning, quality assurance and delivery of clinical SFRT treatments which were built upon our experience treating a large patient population with both a brass grid and VMAT lattice approach. Preclinical research and patient trials aimed at understanding the mechanism of action are needed to elucidate which patients may benefit most from SFRT, and ultimately expand its use.
Collapse
Affiliation(s)
- Michael P Grams
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA.
| | - Christopher L Deufel
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - James A Kavanaugh
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Kimberly S Corbin
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Safia K Ahmed
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Michael G Haddock
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Scott C Lester
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Daniel J Ma
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Ivy A Petersen
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Randi R Finley
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Karen G Lang
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Sheri S Spreiter
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Sean S Park
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Dawn Owen
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| |
Collapse
|
23
|
Tubin S, Vozenin M, Prezado Y, Durante M, Prise K, Lara P, Greco C, Massaccesi M, Guha C, Wu X, Mohiuddin M, Vestergaard A, Bassler N, Gupta S, Stock M, Timmerman R. Novel unconventional radiotherapy techniques: Current status and future perspectives - Report from the 2nd international radiation oncology online seminar. Clin Transl Radiat Oncol 2023; 40:100605. [PMID: 36910025 PMCID: PMC9996385 DOI: 10.1016/j.ctro.2023.100605] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 02/25/2023] Open
Abstract
•Improvement of therapeutic ratio by novel unconventional radiotherapy approaches.•Immunomodulation using high-dose spatially fractionated radiotherapy.•Boosting radiation anti-tumor effects by adding an immune-mediated cell killing.
Collapse
Affiliation(s)
- S. Tubin
- Medaustron Center for Ion Therapy, Marie-Curie Strasse 5, Wiener Neustadt 2700, Austria
| | - M.C. Vozenin
- Radiation Oncology Laboratory, Radiation Oncology Service, Oncology Department, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Y. Prezado
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay 91400, France
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay 91400, France
| | - M. Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Planckstraße 1, Darmstadt 64291, Germany
- Technsiche Universität Darmstadt, Institute for Condensed Matter Physics, Darmstadt, Germany
| | - K.M. Prise
- Patrick G Johnston Centre for Cancer Research Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| | - P.C. Lara
- Canarian Comprehensive Cancer Center, San Roque University Hospital & Fernando Pessoa Canarias University, C/Dolores de la Rocha 9, Las Palmas GC 35001, Spain
| | - C. Greco
- Department of Radiation Oncology Champalimaud Foundation, Av. Brasilia, Lisbon 1400-038, Portugal
| | - M. Massaccesi
- UOC di Radioterapia Oncologica, Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - C. Guha
- Montefiore Medical Center Radiation Oncology, 111 E 210th St, New York, NY, United States
| | - X. Wu
- Executive Medical Physics Associates, 19470 NE 22nd Road, Miami, FL 33179, United States
| | - M.M. Mohiuddin
- Northwestern Medicine Cancer Center Warrenville and Northwestern Medicine Proton Center, 4455 Weaver Pkwy, Warrenville, IL 60555, United States
| | - A. Vestergaard
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - N. Bassler
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - S. Gupta
- The Loop Immuno-Oncology Laboratory, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States
| | - M. Stock
- Medaustron Center for Ion Therapy, Marie-Curie Strasse 5, Wiener Neustadt 2700, Austria
- Karl Landsteiner University of Health Sciences, Marie-Curie Strasse 5, Wiener Neustadt 2700, Austria
| | - R. Timmerman
- Department of Radiation Oncology, University of Texas, Southwestern Medical Center, Inwood Road Dallas, TX 2280, United States
| |
Collapse
|
24
|
Bertho A, Iturri L, Brisebard E, Juchaux M, Gilbert C, Ortiz R, Sebrie C, Jourdain L, Lamirault C, Ramasamy G, Pouzoulet F, Prezado Y. Evaluation of the Role of the Immune System Response After Minibeam Radiation Therapy. Int J Radiat Oncol Biol Phys 2023; 115:426-439. [PMID: 35985455 DOI: 10.1016/j.ijrobp.2022.08.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/23/2022] [Accepted: 08/05/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE Minibeam radiation therapy (MBRT) is an innovative technique that uses a spatial dose modulation. The dose distribution consists of high doses (peaks) in the path of the minibeam and low doses (valleys). The underlying biological mechanism associated with MBRT efficacy remains currently unclear and thus we investigated the potential role of the immune system after treatment with MBRT. METHODS AND MATERIALS Rats bearing an orthotopic glioblastoma cell line were treated with 1 fraction of high dose conventional radiation therapy (30 Gy) or 1 fraction of the same mean dose in MBRT. Both immunocompetent (F344) and immunodeficient (Nude) rats were analyzed in survival studies. Systemic and intratumoral immune cell population changes were studied with flow cytometry and immunohistochemistry (IHC) 2 and 7 days after the irradiation. RESULTS The absence of response of Nude rats after MBRT suggested that T cells were key in the mode of action of MBRT. An inflammatory phenotype was observed in the blood 1 week after irradiation compared with conventional irradiation. Tumor immune cell analysis by flow cytometry showed a substantial infiltration of lymphocytes, specifically of CD8 T cells and B cells in both conventional and MBRT-treated animals. IHC revealed that MBRT induced a faster recruitment of CD8 and CD4 T cells. Animals that were cured by radiation therapy did not suffer tumor growth after reimplantation of tumoral cells, proving the long-term immunity response generated after a high dose of radiation. CONCLUSIONS Our findings show that MBRT can elicit a robust antitumor immune response in glioblastoma while avoiding the high toxicity of a high dose of conventional radiation therapy.
Collapse
Affiliation(s)
- Annaig Bertho
- CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Institut Curie, Université PSL, Orsay, France; CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Université Paris-Saclay, Orsay, France.
| | - Lorea Iturri
- CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Institut Curie, Université PSL, Orsay, France; CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Université Paris-Saclay, Orsay, France
| | | | - Marjorie Juchaux
- CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Institut Curie, Université PSL, Orsay, France; CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Université Paris-Saclay, Orsay, France
| | - Cristèle Gilbert
- CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Institut Curie, Université PSL, Orsay, France; CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Université Paris-Saclay, Orsay, France
| | - Ramon Ortiz
- CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Institut Curie, Université PSL, Orsay, France; CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Université Paris-Saclay, Orsay, France
| | - Catherine Sebrie
- Service Hospitalier Frédéric Joliot, CEA, CNRS, Inserm, BIOMAPS Université Paris-Saclay, Orsay, France
| | - Laurene Jourdain
- Service Hospitalier Frédéric Joliot, CEA, CNRS, Inserm, BIOMAPS Université Paris-Saclay, Orsay, France
| | - Charlotte Lamirault
- Département de Recherche Translationnelle, CurieCoreTech-Experimental Radiotherapy (RadeXp), Institut Curie, PSL University, Paris, France
| | - Gabriel Ramasamy
- Département de Recherche Translationnelle, CurieCoreTech-Experimental Radiotherapy (RadeXp), Institut Curie, PSL University, Paris, France
| | - Frédéric Pouzoulet
- Département de Recherche Translationnelle, CurieCoreTech-Experimental Radiotherapy (RadeXp), Institut Curie, PSL University, Paris, France; Inserm U1288, Laboratoire de Recherche Translationnelle en Oncologie, Institut Curie, PSL University, Université Paris-Saclay, Orsay, France
| | - Yolanda Prezado
- CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Institut Curie, Université PSL, Orsay, France; CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Université Paris-Saclay, Orsay, France
| |
Collapse
|
25
|
Ji X, Jiang W, Wang J, Zhou B, Ding W, Liu S, Huang H, Chen G, Sun X. Application of individualized multimodal radiotherapy combined with immunotherapy in metastatic tumors. Front Immunol 2023; 13:1106644. [PMID: 36713375 PMCID: PMC9877461 DOI: 10.3389/fimmu.2022.1106644] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/22/2022] [Indexed: 01/14/2023] Open
Abstract
Radiotherapy is one of the mainstays of cancer treatment. More than half of cancer patients receive radiation therapy. In addition to the well-known direct tumoricidal effect, radiotherapy has immunomodulatory properties. When combined with immunotherapy, radiotherapy, especially high-dose radiotherapy (HDRT), exert superior systemic effects on distal and unirradiated tumors, which is called abscopal effect. However, these effects are not always effective for cancer patients. Therefore, many studies have focused on exploring the optimized radiotherapy regimens to further enhance the antitumor immunity of HDRT and reduce its immunosuppressive effect. Several studies have shown that low-dose radiotherapy (LDRT) can effectively reprogram the tumor microenvironment, thereby potentially overcoming the immunosuppressive stroma induced by HDRT. However, bridging the gap between preclinical commitment and effective clinical delivery is challenging. In this review, we summarized the existing studies supporting the combined use of HDRT and LDRT to synergistically enhance antitumor immunity, and provided ideas for the individualized clinical application of multimodal radiotherapy (HDRT+LDRT) combined with immunotherapy.
Collapse
|
26
|
Halthore A, Fellows Z, Tran A, Deville C, Wright JL, Meyer J, Li H, Sheikh K. Treatment Planning of Bulky Tumors Using Pencil Beam Scanning Proton GRID Therapy. Int J Part Ther 2022; 9:40-49. [PMID: 36721485 PMCID: PMC9875826 DOI: 10.14338/ijpt-22-00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/02/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose To compare spatially fractionated radiation therapy (GRID) treatment planning techniques using proton pencil-beam-scanning (PBS) and photon therapy. Materials and Methods PBS and volumetric modulated arc therapy (VMAT) GRID plans were retrospectively generated for 5 patients with bulky tumors. GRID targets were arranged along the long axis of the gross tumor, spaced 2 and 3 cm apart, and treated with a prescription of 18 Gy. PBS plans used 2- to 3-beam multiple-field optimization with robustness evaluation. Dosimetric parameters including peak-to-edge ratio (PEDR), ratio of dose to 90% of the valley to dose to 10% of the peak VPDR(D90/D10), and volume of normal tissue receiving at least 5 Gy (V5) and 10 Gy (V10) were calculated. The peak-to-valley dose ratio (PVDR), VPDR(D90/D10), and organ-at-risk doses were prospectively assessed in 2 patients undergoing PBS-GRID with pretreatment quality assurance computed tomography (QACT) scans. Results PBS and VMAT GRID plans were generated for 5 patients with bulky tumors. Gross tumor volume values ranged from 826 to 1468 cm3. Peak-to-edge ratio for PBS was higher than for VMAT for both spacing scenarios (2-cm spacing, P = .02; 3-cm spacing, P = .01). VPDR(D90/D10) for PBS was higher than for VMAT (2-cm spacing, P = .004; 3-cm spacing, P = .002). Normal tissue V5 was lower for PBS than for VMAT (2-cm spacing, P = .03; 3-cm spacing, P = .02). Normal tissue mean dose was lower with PBS than with VMAT (2-cm spacing, P = .03; 3-cm spacing, P = .02). Two patients treated using PBS GRID and assessed with pretreatment QACT scans demonstrated robust PVDR, VPDR(D90/D10), and organs-at-risk doses. Conclusions The PEDR was significantly higher for PBS than VMAT plans, indicating lower target edge dose. Normal tissue mean dose was significantly lower with PBS than VMAT. PBS GRID may result in lower normal tissue dose compared with VMAT plans, allowing for further dose escalation in patients with bulky disease.
Collapse
Affiliation(s)
- Aditya Halthore
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
,Department of Radiation Oncology, The Johns Hopkins Proton Center, Washington, DC, USA
| | - Zachary Fellows
- Department of Radiation Oncology, The Johns Hopkins Proton Center, Washington, DC, USA
| | - Anh Tran
- Department of Radiation Oncology, The Johns Hopkins Proton Center, Washington, DC, USA
| | - Curtiland Deville
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
,Department of Radiation Oncology, The Johns Hopkins Proton Center, Washington, DC, USA
| | - Jean L. Wright
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
,Department of Radiation Oncology, The Johns Hopkins Proton Center, Washington, DC, USA
| | - Jeffrey Meyer
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Heng Li
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
,Department of Radiation Oncology, The Johns Hopkins Proton Center, Washington, DC, USA
| | - Khadija Sheikh
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
,Department of Radiation Oncology, The Johns Hopkins Proton Center, Washington, DC, USA
| |
Collapse
|
27
|
An International Consensus on the Design of Prospective Clinical–Translational Trials in Spatially Fractionated Radiation Therapy for Advanced Gynecologic Cancer. Cancers (Basel) 2022; 14:cancers14174267. [PMID: 36077802 PMCID: PMC9454841 DOI: 10.3390/cancers14174267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Spatially fractionated radiation therapy (SFRT) delivers intentionally heterogenous dose to tumors. This is a major departure from current radiation therapy, which strives for uniform dose. Early pilot experience suggests promising treatment outcomes with SFRT in patients with challenging bulky tumors, including gynecologic cancer. Well-conducted prospective multi-institutional clinical trials are now needed to further test SFRT as a treatment modality. However, clinical trial development is hampered by the variabilities in SFRT approach and the overall unfamiliarity with heterogeneous dosing. A broad consensus among SFRT experts, potential investigators, and the wider radiation oncology community is needed so that clinical trials in SFRT can be successfully designed and carried out. We developed an international consensus guideline for the design parameters of clinical/translational trials in SFRT for gynecologic cancer. High-to-moderate consensus was achieved, and harmonized fundamental design parameters for SFRT trials in advanced gynecologic cancer were defined. Abstract Despite the unexpectedly high tumor responses and limited treatment-related toxicities observed with SFRT, prospective multi-institutional clinical trials of SFRT are still lacking. High variability of SFRT technologies and methods, unfamiliar complex dose and prescription concepts for heterogeneous dose and uncertainty regarding systemic therapies present major obstacles towards clinical trial development. To address these challenges, the consensus guideline reported here aimed at facilitating trial development and feasibility through a priori harmonization of treatment approach and the full range of clinical trial design parameters for SFRT trials in gynecologic cancer. Gynecologic cancers were evaluated for the status of SFRT pilot experience. A multi-disciplinary SFRT expert panel for gynecologic cancer was established to develop the consensus through formal panel review/discussions, appropriateness rank voting and public comment solicitation/review. The trial design parameters included eligibility/exclusions, endpoints, SFRT technology/technique, dose/dosimetric parameters, systemic therapies, patient evaluations, and embedded translational science. Cervical cancer was determined as the most suitable gynecologic tumor for an SFRT trial. Consensus emphasized standardization of SFRT dosimetry/physics parameters, biologic dose modeling, and specimen collection for translational/biological endpoints, which may be uniquely feasible in cervical cancer. Incorporation of brachytherapy into the SFRT regimen requires additional pre-trial pilot investigations. Specific consensus recommendations are presented and discussed.
Collapse
|
28
|
Schneider T, Fernandez-Palomo C, Bertho A, Fazzari J, Iturri L, Martin OA, Trappetti V, Djonov V, Prezado Y. Combining FLASH and spatially fractionated radiation therapy: The best of both worlds. Radiother Oncol 2022; 175:169-177. [PMID: 35952978 DOI: 10.1016/j.radonc.2022.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/23/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022]
Abstract
FLASH radiotherapy (FLASH-RT) and spatially fractionated radiation therapy (SFRT) are two new therapeutical strategies that use non-standard dose delivery methods to reduce normal tissue toxicity and increase the therapeutic index. Although likely based on different mechanisms, both FLASH-RT and SFRT have shown to elicit radiobiological effects that significantly differ from those induced by conventional radiotherapy. With the therapeutic potential having been established separately for each technique, the combination of FLASH-RT and SFRT could therefore represent a winning alliance. In this review, we discuss the state of the art, advantages and current limitations, potential synergies, and where a combination of these two techniques could be implemented today or in the near future.
Collapse
Affiliation(s)
- Tim Schneider
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France; Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
| | | | - Annaïg Bertho
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France; Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
| | - Jennifer Fazzari
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland
| | - Lorea Iturri
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France; Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
| | - Olga A Martin
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland; Division of Radiation Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; University of Melbourne, Parkville, VIC 3010, Australia
| | - Verdiana Trappetti
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland
| | - Yolanda Prezado
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France; Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France.
| |
Collapse
|
29
|
Ji H, Zhou Z. A ‘Hybrid’ Radiotherapy Regimen Designed for Immunomodulation: Combining High-Dose Radiotherapy with Low-Dose Radiotherapy. Cancers (Basel) 2022; 14:cancers14143505. [PMID: 35884565 PMCID: PMC9319172 DOI: 10.3390/cancers14143505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Radiotherapy is an important cancer treatment. Aside from its direct killing effect, it also affects anti-tumor immunity. However, radiotherapy’s immune effect is not clear; it depends on the dose and fraction, cancer type, combined immunotherapy, and many other factors. Studies have focused on the optimal radiotherapy regimen to stimulate anti-tumor immunity, but conflicts exist, especially regarding the best radiation dose and fractions. Interestingly, high-dose radiotherapy and low-dose radiotherapy have complementary effects on stimulating anti-tumor immunity. Preclinical studies supporting this finding have accumulated, but gaps between theory and clinical practice still exist. This review summarizes the evidence supporting the use of this ‘hybrid’ radiotherapy approach to effectively stimulate anti-tumor immunity, explains the immune mechanisms of this combination, raises questions that must be addressed in clinical practice, and provides ideas for designing individualized treatment to increase efficiency in stimulating anti-tumor immunity using high-dose plus low-dose radiotherapy. Abstract Radiotherapy (RT) affects anti-tumor immunity. However, the exact impact of RT on anti-tumor immune response differs among cancer types, RT dose and fractions, patients’ innate immunity, and many other factors. There are conflicting findings on the optimal radiation dose and fractions to stimulate effective anti-tumor immunity. High-dose radiotherapy (HDRT) acts in the same way as a double-edged sword in stimulating anti-tumor immunity, while low-dose radiotherapy (LDRT) seems to play a vital role in modulating the tumor immune microenvironment. Recent preclinical data suggest that a ‘hybrid’ radiotherapy regimen, which refers to combining HDRT with LDRT, can reap the advantages of both. Clinical data have also indicated a promising potential. However, there are still questions to be addressed in order to put this novel combination therapy into clinical practice. For example, the selection of treatment site, treatment volume, the sequencing of high-dose radiotherapy and low-dose radiotherapy, combined immunotherapy, and so on. This review summarizes the current evidence supporting the use of HDRT + LDRT, explains possible immune biology mechanisms of this ‘hybrid’ radiotherapy, raises questions to be considered when working out individualized treatment plans, and lists possible avenues to increase efficiency in stimulating anti-tumor immunity using high-dose plus low-dose radiotherapy.
Collapse
|
30
|
Li CX, Liu Y, Zhang YZ, Li JC, Lai J. Astragalus polysaccharide: a review of its immunomodulatory effect. Arch Pharm Res 2022; 45:367-389. [PMID: 35713852 DOI: 10.1007/s12272-022-01393-3] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 06/12/2022] [Indexed: 12/27/2022]
Abstract
The Astragalus polysaccharide is an important bioactive component derived from the dry root of Astragalus membranaceus. This review aims to provide a comprehensive overview of the research progress on the immunomodulatory effect of Astragalus polysaccharide and provide valuable reference information. We review the immunomodulatory effect of Astragalus polysaccharide on central and peripheral immune organs, including bone marrow, thymus, lymph nodes, spleen, and mucosal tissues. Furthermore, the immunomodulatory effect of Astragalus polysaccharide on a variety of immune cells is summarized. Studies have shown that Astragalus polysaccharide can promote the activities of macrophages, natural killer cells, dendritic cells, T lymphocytes, B lymphocytes and microglia and induce the expression of a variety of cytokines and chemokines. The immunomodulatory effect of Astragalus polysaccharide makes it promising for the treatment of many diseases, including cancer, infection, type 1 diabetes, asthma, and autoimmune disease. Among them, the anticancer effect is the most prominent. In short, Astragalus polysaccharide is a valuable immunomodulatory medicine, but further high-quality studies are warranted to corroborate its clinical efficacy.
Collapse
Affiliation(s)
- Chun-Xiao Li
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Liu
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Zhen Zhang
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing-Chun Li
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jiang Lai
- Department of Anorectal Surgery, Third People's Hospital of Chengdu, Chengdu, China.
| |
Collapse
|
31
|
Jin JY. Prospect of radiotherapy technology development in the era of immunotherapy. JOURNAL OF THE NATIONAL CANCER CENTER 2022; 2:106-112. [PMID: 39034954 PMCID: PMC11256706 DOI: 10.1016/j.jncc.2022.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022] Open
Abstract
Radiotherapy (RT) is one of the important modalities for cancer treatments. Mounting evidence suggests that the host immune system is involved in the tumor cell killing during RT, and future RT technology development should aim to minimize radiation dose to the immune system while maintaining a sufficient dose to the tumor. A brief history of RT technology development is first summarized. Three RT technologies, namely FLASH RT, proton therapy, and spatially fractionated RT (SFRT), are singled out for the era of immunotherapy. Besides the technical aspects, the mechanism of FLASH effect is discussed, which is likely the combined results of the recombination effect, oxygen depletion effect and immune sparing effect. The proton therapy should have the advantage of causing much less immune damage in comparison to X-ray based RT due to the Bragg peak. However, the relative biological effectiveness (RBE) uncertainty and range uncertainty may hinder the translation of this advantage into clinical benefit. Research approaches to overcome these two technical hurdles are discussed. Various SFRT approaches and their application are reviewed. These approaches are categorized as single-field 1D/2D SFRT, multi-field 3D SFRT and quasi-3D SFRT techniques. A 3D SFRT approach, which is achieved by placing the Bragg peak of a proton 2D SFRT field in discrete depths, may have special potential because all 3 technologies (FLASH RT, proton therapy and SFRT) may be used in this approach.
Collapse
Affiliation(s)
- Jian-Yue Jin
- Radiation Oncology, Seidman Cancer Center, University Hospitals, Case Western Reserve University, Cleveland, United States
| |
Collapse
|
32
|
Baxevanis CN, Gritzapis AD, Voutsas IF, Batsaki P, Goulielmaki M, Adamaki M, Zoumpourlis V, Fortis SP. T-Cell Repertoire in Tumor Radiation: The Emerging Frontier as a Radiotherapy Biomarker. Cancers (Basel) 2022; 14:cancers14112674. [PMID: 35681654 PMCID: PMC9179913 DOI: 10.3390/cancers14112674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Radiotherapy constitutes an essential component of the treatment for malignant disease. Besides its direct effect on cancer cells, namely, DNA damage and cell death, ionizing irradiation also mediates indirect antitumor effects that are mostly mediated by the immune system. Investigations into the processes underlying the interaction between radiotherapy and the immune system have uncovered mechanisms that can be exploited to promote the antitumor efficacy of radiotherapy both locally in the irradiated primary tumor and also at distant lesions in non-irradiated tumors. Because of its capacity to stimulate antitumor immunity, radiotherapy is also applied in combination with immune-checkpoint-inhibition-based immunotherapy. This review discusses the important pathways that govern the synergistic interactions between ionizing radiation and antitumor immune reactivity. Unravelling these involved mechanisms is mandatory for the successful application of anticancer radiotherapy and immunotherapy. We also place emphasis on the need for biomarkers that will aid in the selection of patients most likely to benefit from such combined treatments. Abstract Radiotherapy (RT) is a therapeutic modality that aims to eliminate malignant cells through the induction of DNA damage in the irradiated tumor site. In addition to its cytotoxic properties, RT also induces mechanisms that result in the promotion of antitumor immunity both locally within the irradiation field but also at distant tumor lesions, a phenomenon that is known as the “abscopal” effect. Because the immune system is capable of sensing the effects of RT, several treatment protocols have been assessing the synergistic role of radiotherapy combined with immunotherapy, collectively referred to as radioimmunotherapy. Herein, we discuss mechanistic insights underlying RT-based immunomodulation, which also enhance our understanding of how RT regulates antitumor T-cell-mediated immunity. Such knowledge is essential for the discovery of predictive biomarkers and for the improvement of clinical trials investigating the efficacy of radio-immunotherapeutic modalities in cancer patients.
Collapse
Affiliation(s)
- Constantin N. Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Angelos D. Gritzapis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Ioannis F. Voutsas
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Panagiota Batsaki
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Maria Goulielmaki
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Maria Adamaki
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece; (M.A.); (V.Z.)
| | - Vassilios Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece; (M.A.); (V.Z.)
| | - Sotirios P. Fortis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
- Correspondence: ; Tel.: +30-2106409462
| |
Collapse
|
33
|
Friedrich T, Scholz M, Durante M. A predictive biophysical model of the combined action of radiotherapy and immunotherapy in cancer. Int J Radiat Oncol Biol Phys 2022; 113:872-884. [DOI: 10.1016/j.ijrobp.2022.03.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/24/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
|
34
|
Grams MP, Tseung HSWC, Ito S, Zhang Y, Owen D, Park SS, Ahmed SK, Petersen IA, Haddock MG, Harmsen WS, Ma DJ. A Dosimetric Comparison of Lattice, Brass, and Proton Grid Therapy Treatment Plans. Pract Radiat Oncol 2022; 12:e442-e452. [DOI: 10.1016/j.prro.2022.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/28/2022] [Accepted: 03/09/2022] [Indexed: 11/28/2022]
|
35
|
Moghaddasi L, Reid P, Bezak E, Marcu LG. Radiobiological and Treatment-Related Aspects of Spatially Fractionated Radiotherapy. Int J Mol Sci 2022; 23:3366. [PMID: 35328787 PMCID: PMC8954016 DOI: 10.3390/ijms23063366] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/13/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
The continuously evolving field of radiotherapy aims to devise and implement techniques that allow for greater tumour control and better sparing of critical organs. Investigations into the complexity of tumour radiobiology confirmed the high heterogeneity of tumours as being responsible for the often poor treatment outcome. Hypoxic subvolumes, a subpopulation of cancer stem cells, as well as the inherent or acquired radioresistance define tumour aggressiveness and metastatic potential, which remain a therapeutic challenge. Non-conventional irradiation techniques, such as spatially fractionated radiotherapy, have been developed to tackle some of these challenges and to offer a high therapeutic index when treating radioresistant tumours. The goal of this article was to highlight the current knowledge on the molecular and radiobiological mechanisms behind spatially fractionated radiotherapy and to present the up-to-date preclinical and clinical evidence towards the therapeutic potential of this technique involving both photon and proton beams.
Collapse
Affiliation(s)
- Leyla Moghaddasi
- Department of Medical Physics, Austin Health, Ballarat, VIC 3350, Australia;
- School of Physical Sciences, University of Adelaide, Adelaide, SA 5001, Australia;
| | - Paul Reid
- Radiation Health, Environment Protection Authority, Adelaide, SA 5000, Australia;
| | - Eva Bezak
- School of Physical Sciences, University of Adelaide, Adelaide, SA 5001, Australia;
- Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Loredana G. Marcu
- Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
- Faculty of Informatics and Science, University of Oradea, 1 Universitatii Str., 410087 Oradea, Romania
| |
Collapse
|
36
|
Yan D, Cai S, Bai L, Du Z, Li H, Sun P, Cao J, Yi N, Liu SB, Tang Z. Integration of immune and hypoxia gene signatures improves the prediction of radiosensitivity in breast cancer. Am J Cancer Res 2022; 12:1222-1240. [PMID: 35411250 PMCID: PMC8984882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/22/2022] [Indexed: 06/14/2023] Open
Abstract
Immunity and hypoxia are two important factors that affect the response of cancer patients to radiotherapy. At the same time, considering the limited predictive value of a single predictive model and the uncertainty of grouping patients near the cutoff value, we developed and validated a combined model based on immune- and hypoxia-related gene expression profiles to predict the radiosensitivity of breast cancer patients. This study was based on breast cancer data from The Cancer Genome Atlas (TCGA). Spike-and-slab Lasso regression analysis was performed to select three immune-related genes and develop a radiosensitivity model. Lasso Cox regression modeling selected 11 hypoxia-related genes for development of radiosensitivity model. Three independent datasets (Molecular Taxonomy of Breast Cancer International Consortium [METABRIC], E-TABM-158, GSE103746) were used to validate the predictive value of radiosensitivity signatures. In the TCGA dataset, the 10-year survival probabilities of the immune radioresistant (IRR) and hypoxia radioresistant (HRR) groups were 0.189 (0.037, 0.973) and 0.477 (0.293, 0.776), respectively. The 10-year survival probabilities of the immune radiosensitive (IRS) and hypoxia radiosensitive (HRS) groups were 0.778 (0.676, 0.895) and 0.824 (0.723, 0.939), respectively. Based on these two gene signatures, we further constructed a combined model and divided all patients into three groups (IRS/HRS, mixed, IRR/HRR). We identified the IRS/HRS patients most likely to benefit from radiotherapy; the 10-year survival probability was 0.886 (0.806, 0.976). The 10-year survival probability of the IRR/HRR group was 0. In conclusion, a combined model integrating immune- and hypoxia-related gene signatures could effectively predict the radiosensitivity of breast cancer and more accurately identify radiosensitive and radioresistant patients than a single model.
Collapse
Affiliation(s)
- Derui Yan
- Department of Biostatistics, School of Public Health, Medical College of Soochow UniversitySuzhou 215123, Jiangsu, China
- Suzhou Key Laboratory of Medical Biotechnology, Suzhou Vocational Health CollegeSuzhou 215009, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow UniversitySuzhou 215123, Jiangsu, China
| | - Shang Cai
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, China
| | - Lu Bai
- Department of Biostatistics, School of Public Health, Medical College of Soochow UniversitySuzhou 215123, Jiangsu, China
- Suzhou Key Laboratory of Medical Biotechnology, Suzhou Vocational Health CollegeSuzhou 215009, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow UniversitySuzhou 215123, Jiangsu, China
| | - Zixuan Du
- Department of Biostatistics, School of Public Health, Medical College of Soochow UniversitySuzhou 215123, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow UniversitySuzhou 215123, Jiangsu, China
| | - Huijun Li
- Department of Biostatistics, School of Public Health, Medical College of Soochow UniversitySuzhou 215123, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow UniversitySuzhou 215123, Jiangsu, China
| | - Peng Sun
- Department of Otolaryngology, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Jianping Cao
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow UniversitySuzhou 215031, Jiangsu, China
| | - Nengjun Yi
- Department of Biostatistics, University of Alabama at BirminghamBirmingham, AL 35294, USA
| | - Song-Bai Liu
- Suzhou Key Laboratory of Medical Biotechnology, Suzhou Vocational Health CollegeSuzhou 215009, Jiangsu, China
| | - Zaixiang Tang
- Department of Biostatistics, School of Public Health, Medical College of Soochow UniversitySuzhou 215123, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow UniversitySuzhou 215123, Jiangsu, China
| |
Collapse
|
37
|
Mayr NA, Snider JW, Regine WF, Mohiuddin M, Hippe DS, Peñagarícano J, Mohiuddin M, Kudrimoti MR, Zhang H, Limoli CL, Le QT, Simone CB. An International Consensus on the Design of Prospective Clinical-Translational Trials in Spatially Fractionated Radiation Therapy. Adv Radiat Oncol 2022; 7:100866. [PMID: 35198833 PMCID: PMC8843999 DOI: 10.1016/j.adro.2021.100866] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/12/2021] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Spatially fractionated radiation therapy (SFRT), which delivers highly nonuniform dose distributions instead of conventionally practiced homogeneous tumor dose, has shown high rates of clinical response with minimal toxicities in large-volume primary or metastatic malignancies. However, prospective multi-institutional clinical trials in SFRT are lacking, and SFRT techniques and dose parameters remain variable. Agreement on dose prescription, technical administration, and clinical and translational design parameters for SFRT trials is essential to enable broad participation and successful accrual to rigorously test the SFRT approach. We aimed to develop a consensus for the design of multi-institutional clinical trials in SFRT, tailored to specific primary tumor sites, to help facilitate development and enhance the feasibility of such trials. METHODS AND MATERIALS Primary tumor sites with sufficient pilot experience in SFRT were identified, and fundamental trial design questions were determined. For each tumor site, a comprehensive consensus effort was established through disease-specific expert panels. Clinical trial design criteria included eligibility, SFRT technology and technique, dose and fractionation, target- and normal-tissue dose parameters, systemic therapies, clinical trial endpoints, and translational science considerations. Iterative appropriateness rank voting, expert panel consensus reviews and discussions, and public comment posting were used for consensus development. RESULTS Clinical trial criteria were developed for head and neck cancer and soft-tissue sarcoma. Final consensus among the 22 trial design categories each (a total of 163 criteria) was high to moderate overall. Uniform patient cohorts of advanced bulky disease, standardization of SFRT technologies and dosimetry and physics parameters, and collection of translational correlates were considered essential to trial design. Final guideline recommendations and the degree of agreement are presented and discussed. CONCLUSIONS This consensus provides design guidelines for the development of prospective multi-institutional clinical trials testing SFRT in advanced head and neck cancer and soft-tissue sarcoma through in-advance harmonization of the fundamental clinical trial design among SFRT experts, potential investigators, and the SFRT community.
Collapse
Affiliation(s)
- Nina A. Mayr
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, Washington
- Tumor Heterogeneity Imaging and Radiomics Laboratory, University of Washington School of Medicine, Seattle, Washington
| | - James W. Snider
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - William F. Regine
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Majid Mohiuddin
- Radiation Oncology Consultants and Northwestern Proton Center, Warrenville, Illinois
| | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | | - Mahesh R. Kudrimoti
- Department of Radiation Medicine, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Hualin Zhang
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Charles L. Limoli
- Department of Radiation Oncology, University of California School of Medicine, Irvine, Irvine, California
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Charles B. Simone
- Department of Radiation Oncology, New York Proton Center, New York, New York
| |
Collapse
|
38
|
Abstract
AbstractSpatially fractionated radiation therapy (SFRT) challenges some of the classical dogmas in conventional radiotherapy. The highly modulated spatial dose distributions in SFRT have been shown to lead, both in early clinical trials and in small animal experiments, to a significant increase in normal tissue dose tolerances. Tumour control effectiveness is maintained or even enhanced in some configurations as compared with conventional radiotherapy. SFRT seems to activate distinct radiobiological mechanisms, which have been postulated to involve bystander effects, microvascular alterations and/or immunomodulation. Currently, it is unclear which is the dosimetric parameter which correlates the most with both tumour control and normal tissue sparing in SFRT. Additional biological experiments aiming at parametrizing the relationship between the irradiation parameters (beam width, spacing, peak-to-valley dose ratio, peak and valley doses) and the radiobiology are needed. A sound knowledge of the interrelation between the physical parameters in SFRT and the biological response would expand its clinical use, with a higher level of homogenisation in the realisation of clinical trials. This manuscript reviews the state of the art of this promising therapeutic modality, the current radiobiological knowledge and elaborates on future perspectives.
Collapse
|
39
|
Bertho A, Iturri L, Prezado Y. Radiation-induced immune response in novel radiotherapy approaches FLASH and spatially fractionated radiotherapies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 376:37-68. [PMID: 36997269 DOI: 10.1016/bs.ircmb.2022.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The last several years have revealed increasing evidence of the immunomodulatory role of radiation therapy. Radiotherapy reshapes the tumoral microenvironment can shift the balance toward a more immunostimulatory or immunosuppressive microenvironment. The immune response to radiation therapy appears to depend on the irradiation configuration (dose, particle, fractionation) and delivery modes (dose rate, spatial distributions). Although an optimal irradiation configuration (dose, temporal fractionation, spatial dose distribution, etc.) has not yet been determined, temporal schemes employing high doses per fraction appear to favor radiation-induced immune response through immunogenic cell death. Through the release of damage-associated molecular patterns and the sensing of double-stranded DNA and RNA breaks, immunogenic cell death activates the innate and adaptive immune response, leading to tumor infiltration by effector T cells and the abscopal effect. Novel radiotherapy approaches such as FLASH and spatially fractionated radiotherapies (SFRT) strongly modulate the dose delivery method. FLASH-RT and SFRT have the potential to trigger the immune system effectively while preserving healthy surrounding tissues. This manuscript reviews the current state of knowledge on the immunomodulation effects of these two new radiotherapy techniques in the tumor, healthy immune cells and non-targeted regions, as well as their therapeutic potential in combination with immunotherapy.
Collapse
|
40
|
Bazyar S, O’Brien ET, Benefield T, Roberts VR, Kumar RJ, Gupta GP, Zhou O, Lee YZ. Immune-Mediated Effects of Microplanar Radiotherapy with a Small Animal Irradiator. Cancers (Basel) 2021; 14:155. [PMID: 35008319 PMCID: PMC8750301 DOI: 10.3390/cancers14010155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/15/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022] Open
Abstract
Spatially fractionated radiotherapy has been shown to have effects on the immune system that differ from conventional radiotherapy (CRT). We compared several aspects of the immune response to CRT relative to a model of spatially fractionated radiotherapy (RT), termed microplanar radiotherapy (MRT). MRT delivers hundreds of grays of radiation in submillimeter beams (peak), separated by non-radiated volumes (valley). We have developed a preclinical method to apply MRT by a commercial small animal irradiator. Using a B16-F10 murine melanoma model, we first evaluated the in vitro and in vivo effect of MRT, which demonstrated significant treatment superiority relative to CRT. Interestingly, we observed insignificant treatment responses when MRT was applied to Rag-/- and CD8-depleted mice. An immuno-histological analysis showed that MRT recruited cytotoxic lymphocytes (CD8), while suppressing the number of regulatory T cells (Tregs). Using RT-qPCR, we observed that, compared to CRT, MRT, up to the dose that we applied, significantly increased and did not saturate CXCL9 expression, a cytokine that plays a crucial role in the attraction of activated T cells. Finally, MRT combined with anti-CTLA-4 ablated the tumor in half of the cases, and induced prolonged systemic antitumor immunity.
Collapse
Affiliation(s)
- Soha Bazyar
- Department of Radiation Oncology, University of Maryland, Maryland, MD 21201, USA;
| | - Edward Timothy O’Brien
- Department of Physics and Astronomy, The University of North Carolina, Chapel Hill, NC 27514, USA;
| | - Thad Benefield
- Department of Radiology, The University of North Carolina, Chapel Hill, NC 27514, USA;
| | | | - Rashmi J. Kumar
- Medical Scientist Training Program, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA;
| | - Gaorav P. Gupta
- Department of Radiation Oncology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA;
| | - Otto Zhou
- Department of Applied Physics Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA;
| | - Yueh Z. Lee
- Department of Radiology, The University of North Carolina, Chapel Hill, NC 27514, USA;
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| |
Collapse
|
41
|
Jenkins SV, Alimohammadi M, Terry AS, Griffin RJ, Tackett AJ, Leung JW, Vang KB, Byrum SD, Dings RPM. Dysbiotic stress increases the sensitivity of the tumor vasculature to radiotherapy and c-Met inhibitors. Angiogenesis 2021; 24:597-611. [PMID: 33629198 PMCID: PMC8295215 DOI: 10.1007/s10456-021-09771-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/22/2021] [Accepted: 02/03/2021] [Indexed: 10/22/2022]
Abstract
Antibiotic-induced microbial imbalance, or dysbiosis, has systemic and long-lasting effects on the host and response to cancer therapies. However, the effects on tumor endothelial cells are largely unknown. Therefore, the goal of the current study was to generate matched B16-F10 melanoma associated endothelial cell lines isolated from mice with and without antibiotic-induced dysbiosis. After validating endothelial cell markers on a genomic and proteomic level, functional angiogenesis assays (i.e., migration and tube formation) also confirmed their vasculature origin. Subsequently, we found that tumor endothelial cells derived from dysbiotic mice (TEC-Dys) were more sensitive to ionizing radiotherapy in the range of clinically-relevant hypofractionated doses, as compared to tumor endothelial cells derived from orthobiotic mice (TEC-Ortho). In order to identify tumor vasculature-associated drug targets during dysbiosis, we used tandem mass tag mass spectroscopy and focused on the statistically significant cellular membrane proteins overexpressed in TEC-Dys. By these criteria c-Met was the most differentially expressed protein, which was validated histologically by comparing tumors with or without dysbiosis. Moreover, in vitro, c-Met inhibitors Foretinib, Crizotinib and Cabozantinib were significantly more effective against TEC-Dys than TEC-Ortho. In vivo, Foretinib inhibited tumor growth to a greater extent during dysbiosis as compared to orthobiotic conditions. Thus, we surmise that tumor response in dysbiotic patients may be greatly improved by targeting dysbiosis-induced pathways, such as c-Met, distinct from the many targets suppressed due to dysbiosis.
Collapse
Affiliation(s)
- Samir V Jenkins
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Mohammad Alimohammadi
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Alexia S Terry
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Arkansas Children's Research Institute, 13 Children's Way, Little Rock, AR, 72202, USA
| | - Justin W Leung
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kieng B Vang
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Arkansas Children's Research Institute, 13 Children's Way, Little Rock, AR, 72202, USA
| | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot #771, Little Rock, AR, 72205, USA.
| |
Collapse
|
42
|
Current Prospects for Treatment of Solid Tumors via Photodynamic, Photothermal, or Ionizing Radiation Therapies Combined with Immune Checkpoint Inhibition (A Review). Pharmaceuticals (Basel) 2021; 14:ph14050447. [PMID: 34068491 PMCID: PMC8151935 DOI: 10.3390/ph14050447] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/21/2022] Open
Abstract
Photodynamic therapy (PDT) causes selective damage to tumor cells and vasculature and also triggers an anti-tumor immune response. The latter fact has prompted the exploration of PDT as an immune-stimulatory adjuvant. PDT is not the only cancer treatment that relies on electromagnetic energy to destroy cancer tissue. Ionizing radiation therapy (RT) and photothermal therapy (PTT) are two other treatment modalities that employ photons (with wavelengths either shorter or longer than PDT, respectively) and also cause tissue damage and immunomodulation. Research on the three modalities has occurred in different “silos”, with minimal interaction between the three topics. This is happening at a time when immune checkpoint inhibition (ICI), another focus of intense research and clinical development, has opened exciting possibilities for combining PDT, PTT, or RT with ICI to achieve improved therapeutic benefits. In this review, we surveyed the literature for studies that describe changes in anti-tumor immunity following the administration of PDT, PTT, and RT, including efforts to combine each modality with ICI. This information, collected all in one place, may make it easier to recognize similarities and differences and help to identify new mechanistic hypotheses toward the goal of achieving optimized combinations and tumor cures.
Collapse
|