1
|
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
|
2
|
Gardner LL, Thompson SJ, O'Connor JD, McMahon SJ. Modelling radiobiology. Phys Med Biol 2024; 69:18TR01. [PMID: 39159658 DOI: 10.1088/1361-6560/ad70f0] [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: 04/25/2024] [Accepted: 08/19/2024] [Indexed: 08/21/2024]
Abstract
Radiotherapy has played an essential role in cancer treatment for over a century, and remains one of the best-studied methods of cancer treatment. Because of its close links with the physical sciences, it has been the subject of extensive quantitative mathematical modelling, but a complete understanding of the mechanisms of radiotherapy has remained elusive. In part this is because of the complexity and range of scales involved in radiotherapy-from physical radiation interactions occurring over nanometres to evolution of patient responses over months and years. This review presents the current status and ongoing research in modelling radiotherapy responses across these scales, including basic physical mechanisms of DNA damage, the immediate biological responses this triggers, and genetic- and patient-level determinants of response. Finally, some of the major challenges in this field and potential avenues for future improvements are also discussed.
Collapse
Affiliation(s)
- Lydia L Gardner
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| | - Shannon J Thompson
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| | - John D O'Connor
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
- Ulster University School of Engineering, York Street, Belfast BT15 1AP, United Kingdom
| | - Stephen J McMahon
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| |
Collapse
|
3
|
Ng AM, MacKinnon KM, Cook AA, D'Alonzo RA, Rowshanfarzad P, Nowak AK, Gill S, Ebert MA. Mechanistic in silico explorations of the immunogenic and synergistic effects of radiotherapy and immunotherapy: a critical review. Phys Eng Sci Med 2024:10.1007/s13246-024-01458-1. [PMID: 39017990 DOI: 10.1007/s13246-024-01458-1] [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: 02/12/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024]
Abstract
Immunotherapy is a rapidly evolving field, with many models attempting to describe its impact on the immune system, especially when paired with radiotherapy. Tumor response to this combination involves a complex spatiotemporal dynamic which makes either clinical or pre-clinical in vivo investigation across the resulting extensive solution space extremely difficult. In this review, several in silico models of the interaction between radiotherapy, immunotherapy, and the patient's immune system are examined. The study included only mathematical models published in English that investigated the effects of radiotherapy on the immune system, or the effect of immuno-radiotherapy with immune checkpoint inhibitors. The findings indicate that treatment efficacy was predicted to improve when both radiotherapy and immunotherapy were administered, compared to radiotherapy or immunotherapy alone. However, the models do not agree on the optimal schedule and fractionation of radiotherapy and immunotherapy. This corresponds to relevant clinical trials, which report an improved treatment efficacy with combination therapy, however, the optimal scheduling varies between clinical trials. This discrepancy between the models can be attributed to the variation in model approach and the specific cancer types modeled, making the determination of the optimum general treatment schedule and model challenging. Further research needs to be conducted with similar data sets to evaluate the best model and treatment schedule for a specific cancer type and stage.
Collapse
Affiliation(s)
- Allison M Ng
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia
| | - Kelly M MacKinnon
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia
- National Centre for Asbestos Related Diseases, The University of Western Australia, Crawley, WA, Australia
| | - Alistair A Cook
- National Centre for Asbestos Related Diseases, The University of Western Australia, Crawley, WA, Australia
- Institute for Respiratory Health, Institute for Respiratory Health, Perth, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Rebecca A D'Alonzo
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia
- National Centre for Asbestos Related Diseases, The University of Western Australia, Crawley, WA, Australia
- Institute for Respiratory Health, Institute for Respiratory Health, Perth, WA, Australia
| | - Pejman Rowshanfarzad
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia
- Centre for Advanced Technologies in Cancer Research (CATCR), Perth, WA, Australia
| | - Anna K Nowak
- National Centre for Asbestos Related Diseases, The University of Western Australia, Crawley, WA, Australia
- Institute for Respiratory Health, Institute for Respiratory Health, Perth, WA, Australia
- Medical School, The University of Western Australia, Crawley, WA, Australia
| | - Suki Gill
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Martin A Ebert
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia.
- Centre for Advanced Technologies in Cancer Research (CATCR), Perth, WA, Australia.
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.
| |
Collapse
|
4
|
Rykkelid AM, Sinha PM, Folefac CA, Horsman MR, Sørensen BS, Søland TM, Schreurs OJF, Malinen E, Edin NFJ. Combination of proton- or X-irradiation with anti-PDL1 immunotherapy in two murine oral cancers. Sci Rep 2024; 14:11569. [PMID: 38773258 PMCID: PMC11109162 DOI: 10.1038/s41598-024-62272-z] [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: 11/17/2023] [Accepted: 05/15/2024] [Indexed: 05/23/2024] Open
Abstract
Combining radiation therapy with immunotherapy is a strategy to improve both treatments. The purpose of this study was to compare responses for two syngeneic head and neck cancer (HNC) tumor models in mice following X-ray or proton irradiation with or without immune checkpoint inhibition (ICI). MOC1 (immunogenic) and MOC2 (less immunogenic) tumors were inoculated in the right hind leg of each mouse (C57BL/6J, n = 398). Mice were injected with anti-PDL1 (10 mg/kg, twice weekly for 2 weeks), and tumors were treated with single-dose irradiation (5-30 Gy) with X-rays or protons. MOC2 tumors grew faster and were more radioresistant than MOC1 tumors, and all mice with MOC2 tumors developed metastases. Irradiation reduced the tumor volume in a dose-dependent manner. ICI alone reduced the tumor volume for MOC1 with 20% compared to controls, while no reduction was seen for MOC2. For MOC1, there was a clear treatment synergy when combining irradiation with ICI for radiation doses above 5 Gy and there was a tendency for X-rays being slightly more biologically effective compared to protons. For MOC2, there was a tendency of protons being more effective than X-rays, but both radiation types showed a small synergy when combined with ICI. Although the responses and magnitudes of the therapeutic effect varied, the optimal radiation dose for maximal synergy appeared to be in the order of 10-15 Gy, regardless of tumor model.
Collapse
Affiliation(s)
- Anne Marit Rykkelid
- Department of Physics, University of Oslo, P.O. Box 1048, 0316, Blindern, Oslo, Norway
| | | | | | - Michael R Horsman
- Experimental Clinical Oncology - Dept. Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Brita Singers Sørensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | - Tine Merete Søland
- Institute of Oral Biology, University of Oslo, P.O. Box 1052, 0316, Blindern, Oslo, Norway
| | | | - Eirik Malinen
- Department of Physics, University of Oslo, P.O. Box 1048, 0316, Blindern, Oslo, Norway
- Department of Radiation Biology, Oslo University Hospital, P.O. Box 4950, 0424, Nydalen, Oslo, Norway
| | - Nina Frederike J Edin
- Department of Physics, University of Oslo, P.O. Box 1048, 0316, Blindern, Oslo, Norway.
| |
Collapse
|
5
|
Adhikarla V, Awuah D, Caserta E, Minnix M, Kuznetsov M, Krishnan A, Wong JYC, Shively JE, Wang X, Pichiorri F, Rockne RC. Designing combination therapies for cancer treatment: application of a mathematical framework combining CAR T-cell immunotherapy and targeted radionuclide therapy. Front Immunol 2024; 15:1358478. [PMID: 38698840 PMCID: PMC11063284 DOI: 10.3389/fimmu.2024.1358478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/21/2024] [Indexed: 05/05/2024] Open
Abstract
Introduction Cancer combination treatments involving immunotherapies with targeted radiation therapy are at the forefront of treating cancers. However, dosing and scheduling of these therapies pose a challenge. Mathematical models provide a unique way of optimizing these therapies. Methods Using a preclinical model of multiple myeloma as an example, we demonstrate the capability of a mathematical model to combine these therapies to achieve maximum response, defined as delay in tumor growth. Data from mice studies with targeted radionuclide therapy (TRT) and chimeric antigen receptor (CAR)-T cell monotherapies and combinations with different intervals between them was used to calibrate mathematical model parameters. The dependence of progression-free survival (PFS), overall survival (OS), and the time to minimum tumor burden on dosing and scheduling was evaluated. Different dosing and scheduling schemes were evaluated to maximize the PFS and optimize timings of TRT and CAR-T cell therapies. Results Therapy intervals that were too close or too far apart are shown to be detrimental to the therapeutic efficacy, as TRT too close to CAR-T cell therapy results in radiation related CAR-T cell killing while the therapies being too far apart result in tumor regrowth, negatively impacting tumor control and survival. We show that splitting a dose of TRT or CAR-T cells when administered in combination is advantageous only if the first therapy delivered can produce a significant benefit as a monotherapy. Discussion Mathematical models are crucial tools for optimizing the delivery of cancer combination therapy regimens with application along the lines of achieving cure, maximizing survival or minimizing toxicity.
Collapse
Affiliation(s)
- Vikram Adhikarla
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Dennis Awuah
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Enrico Caserta
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Megan Minnix
- Department of Molecular Imaging and Therapy, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Maxim Kuznetsov
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Amrita Krishnan
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Jefferey Y. C. Wong
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - John E. Shively
- Department of Molecular Imaging and Therapy, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Xiuli Wang
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Flavia Pichiorri
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Russell C. Rockne
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
6
|
Ma L, Deng L, Peng J, Yu J, Meng X. Chemotherapy-free radiotherapy combined with immune checkpoint inhibitors: a new regimen for locally advanced non-small cell lung cancer? Cancer Biol Med 2024; 20:j.issn.2095-3941.2023.0402. [PMID: 38318930 PMCID: PMC10845940 DOI: 10.20892/j.issn.2095-3941.2023.0402] [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: 11/01/2023] [Accepted: 12/22/2023] [Indexed: 02/07/2024] Open
Abstract
Maintenance immunotherapy after concurrent chemoradiotherapy remains the standard therapeutic approach in patients with unresectable locally advanced non-small cell lung cancer (LA-NSCLC). The efficacy of pembrolizumab without chemotherapy in stage IV NSCLC has incited interest in similar approaches for LA-NSCLC. Several recent investigations involving the synergistic potential of immunotherapy combined with radiotherapy (iRT) have generated encouraging results. This review discusses the existing studies and prospective directions of chemotherapy-free iRT strategies in unresectable LA-NSCLC. Although the initial findings of chemotherapy-free iRT strategies have shown promising efficacy, we must consider the methodologic limitations of current studies and the myriad of challenges that accompany the implementation of chemotherapy-free iRT. These challenges include determining the optimal dose and fractionation, precise target volume delineation, and identification of additional suitable patient cohorts. Furthermore, the feasibility of chemotherapy-free iRT as a novel treatment modality for select patients with LA-NSCLC is contingent upon validation through randomized phase III trials.
Collapse
Affiliation(s)
- Lin Ma
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430000, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Liufu Deng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianfeng Peng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Jinming Yu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430000, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
7
|
Ledzewicz U, Schättler H. Optimal dosage protocols for mathematical models of synergy of chemo- and immunotherapy. Front Immunol 2024; 14:1303814. [PMID: 38313433 PMCID: PMC10834764 DOI: 10.3389/fimmu.2023.1303814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/26/2023] [Indexed: 02/06/2024] Open
Abstract
The release of tumor antigens during traditional cancer treatments such as radio- or chemotherapy leads to a stimulation of the immune response which provides synergistic effects these treatments have when combined with immunotherapies. A low-dimensional mathematical model is formulated which, depending on the values of its parameters, encompasses the 3 E's (elimination, equilibrium, escape) of tumor immune system interactions. For the escape situation, optimal control problems are formulated which aim to revert the process to the equilibrium scenario. Some numerical results are included.
Collapse
Affiliation(s)
- Urszula Ledzewicz
- Institute of Mathematics, Lodz University of Technology, Lodz, , Poland
- Department of Mathematics and Statistics, Southern Illinois University Edwardsville, Edwardsville, IL, United States
| | - Heinz Schättler
- Department of Electrical and Systems Engineering, Washington University, St. Louis, MO, United States
| |
Collapse
|
8
|
Mohsin N, Enderling H, Brady-Nicholls R, Zahid MU. Simulating tumor volume dynamics in response to radiotherapy: Implications of model selection. J Theor Biol 2024; 576:111656. [PMID: 37952611 DOI: 10.1016/j.jtbi.2023.111656] [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: 06/02/2022] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
From the beginning of the usage of radiotherapy (RT) for cancer treatment, mathematical modeling has been integral to understanding radiobiology and for designing treatment approaches and schedules. There has been extensive modeling of response to RT with the inclusion of various degrees of biological complexity. In this study, we compare three models of tumor volume dynamics: (1) exponential growth with RT directly reducing tumor volume, (2) logistic growth with direct tumor volume reduction, and (3) logistic growth with RT reducing the tumor carrying capacity with the objective of understanding the implications of model selection and informing the process of model calibration and parameterization. For all three models, we: examined the rates of change in tumor volume during and RT treatment course; performed parameter sensitivity and identifiability analyses; and investigated the impact of the parameter sensitivity on the tumor volume trajectories. In examining the tumor volume dynamics trends, we coined a new metric - the point of maximum reduction of tumor volume (MRV) - to quantify the magnitude and timing of the expected largest impact of RT during a treatment course. We found distinct timing differences in MRV, dependent on model selection. The parameter identifiability and sensitivity analyses revealed the interdependence of the different model parameters and that it is only possible to independently identify tumor growth and radiation response parameters if the underlying tumor growth rate is sufficiently large. Ultimately, the results of these analyses help us to better understand the implications of model selection while simultaneously generating falsifiable hypotheses about MRV timing that can be tested on longitudinal measurements of tumor volume from pre-clinical or clinical data with high acquisition frequency. Although, our study only compares three particular models, the results demonstrate that caution is necessary in selecting models of response to RT, given the artifacts imposed by each model.
Collapse
Affiliation(s)
- Nuverah Mohsin
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Heiko Enderling
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Renee Brady-Nicholls
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States.
| | - Mohammad U Zahid
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States.
| |
Collapse
|
9
|
Backlund E, Grozman V, Egyhazi Brage S, Lewensohn R, Lindberg K, Helgadottir H. Radiotherapy with or without immunotherapy in metastatic melanoma: efficacy and tolerability. Acta Oncol 2023; 62:1921-1930. [PMID: 37966921 DOI: 10.1080/0284186x.2023.2280766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/03/2023] [Indexed: 11/17/2023]
Abstract
INTRODUCTION Radiotherapy (RT) is primarily considered as a palliative treatment in patients with metastatic melanoma. However, observations suggest that when RT is combined with immune checkpoint inhibitors (ICI), it can induce an immune response leading to an anti-tumoral effect also distant from the irradiated area - a phenomenon called 'abscopal effect'. The frequency and circumstances of abscopal effect among metastatic melanoma patients remains uncertain and further research is necessary. MATERIAL AND METHOD This retrospective study included all metastatic melanoma patients who received non-stereotactic RT in Stockholm, Sweden in 2015-2020. Patients were grouped depending on if RT was given at start of ICI (RT + ICI(start)), at ICI progression (RT + ICI(salvage)) or without ICI (RT(only)). Response rates in irradiated (RR(irradiated)) and overall response rates in non-irradiated (ORR(non-irradiated)) metastases were evaluated together with survival and toxicity in each cohort. RESULTS In the RT + ICI(start) (n = 47), RT + ICI(salvage) (n = 41) and RT(only) (n = 55) cohorts, RR(irradiated) was 70.7%, 67.5% and 43.1% (p = 0.018) while the ORR(non-irradiated) was 36.1%, 14.8% and 0.0% (p = 0.003), and the median overall survival was 18.2, 15.0 and 7.2 months, respectively (p = 0.014). Local response to RT was in all cohorts associated with longer survival (p < 0.001). The frequency of grade ≥3 immune-related adverse events was 17.0% and 19.5% in the RT + ICI(start) and RT + ICI(salvage) cohorts. No increased frequency of RT-related adverse events was seen in the RT + ICI cohorts, compared to the RT(only) cohort. CONCLUSION This retrospective study showed that melanoma patients receiving RT in combination with ICI had a superior antitumoral response in both irradiated and non-irradiated lesions as compared to patients receiving only RT. Additionally, a subgroup of patients receiving RT when progressing on ICI experienced tumor regression also in non-irradiated areas.
Collapse
Affiliation(s)
- Ellen Backlund
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Vitali Grozman
- Department of Diagnostic Radiology, Karolinska University Hospital, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm
| | | | - Rolf Lewensohn
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Karin Lindberg
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Hildur Helgadottir
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Hamis S, Somervuo P, Ågren JA, Tadele DS, Kesseli J, Scott JG, Nykter M, Gerlee P, Finkelshtein D, Ovaskainen O. Spatial cumulant models enable spatially informed treatment strategies and analysis of local interactions in cancer systems. J Math Biol 2023; 86:68. [PMID: 37017776 PMCID: PMC10076412 DOI: 10.1007/s00285-023-01903-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 01/13/2023] [Accepted: 03/09/2023] [Indexed: 04/06/2023]
Abstract
Theoretical and applied cancer studies that use individual-based models (IBMs) have been limited by the lack of a mathematical formulation that enables rigorous analysis of these models. However, spatial cumulant models (SCMs), which have arisen from theoretical ecology, describe population dynamics generated by a specific family of IBMs, namely spatio-temporal point processes (STPPs). SCMs are spatially resolved population models formulated by a system of differential equations that approximate the dynamics of two STPP-generated summary statistics: first-order spatial cumulants (densities), and second-order spatial cumulants (spatial covariances). We exemplify how SCMs can be used in mathematical oncology by modelling theoretical cancer cell populations comprising interacting growth factor-producing and non-producing cells. To formulate model equations, we use computational tools that enable the generation of STPPs, SCMs and mean-field population models (MFPMs) from user-defined model descriptions (Cornell et al. Nat Commun 10:4716, 2019). To calculate and compare STPP, SCM and MFPM-generated summary statistics, we develop an application-agnostic computational pipeline. Our results demonstrate that SCMs can capture STPP-generated population density dynamics, even when MFPMs fail to do so. From both MFPM and SCM equations, we derive treatment-induced death rates required to achieve non-growing cell populations. When testing these treatment strategies in STPP-generated cell populations, our results demonstrate that SCM-informed strategies outperform MFPM-informed strategies in terms of inhibiting population growths. We thus demonstrate that SCMs provide a new framework in which to study cell-cell interactions, and can be used to describe and perturb STPP-generated cell population dynamics. We, therefore, argue that SCMs can be used to increase IBMs' applicability in cancer research.
Collapse
Affiliation(s)
- Sara Hamis
- Tampere Institute for Advanced Study, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland.
| | - Panu Somervuo
- Organismal and Evolutionary Biology Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - J Arvid Ågren
- Department of Evolutionary Biology, Uppsala University, Uppsala, Sweden
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
| | - Dagim Shiferaw Tadele
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
- Department for Medical Genetics, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Juha Kesseli
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Jacob G Scott
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
- Case Western Reserve School of Medicine, Cleveland, OH, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Philip Gerlee
- Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
- Mathematical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Dmitri Finkelshtein
- Department of Mathematics, Faculty of Science and Engineering, Swansea University, Swansea, UK
| | - Otso Ovaskainen
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Organismal and Evolutionary Biology Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Department of Biology, Centre for Biodiversity Dynamics, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
11
|
Gonzalez-Crespo I, Gomez-Caamano A, Pouso OL, Fenwick JD, Pardo-Montero J. A Biomathematical Model of Tumor Response to Radioimmunotherapy With αPDL1 and αCTLA4. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:808-821. [PMID: 35544486 DOI: 10.1109/tcbb.2022.3174454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
There is evidence of synergy between radiotherapy and immunotherapy. Radiotherapy can increase liberation of tumor antigens, causing activation of antitumor T-cells. This effect can be boosted with immunotherapy. Radioimmunotherapy has potential to increase tumor control rates. Biomathematical models of response to radioimmunotherapy may help on understanding of the mechanisms affecting response, and assist clinicians on the design of optimal treatment strategies. In this work we present a biomathematical model of tumor response to radioimmunotherapy. The model uses the linear-quadratic response of tumor cells to radiation (or variation of it), and builds on previous developments to include the radiation-induced immune effect. We have focused this study on the combined effect of radiotherapy and αPDL1/ αCTLA4 therapies. The model can fit preclinical data of volume dynamics and control obtained with different dose fractionations and αPDL1/ αCTLA4. A biomathematical study of optimal combination strategies suggests that a good understanding of the involved biological delays, the biokinetics of the immunotherapy drug, and the interplay between them, may be of paramount importance to design optimal radioimmunotherapy schedules. Biomathematical models like the one we present can help to interpret experimental data on the synergy between radiotherapy and immunotherapy, and to assist in the design of more effective treatments.
Collapse
|
12
|
Gehre S, Meyer F, Sengedorj A, Grottker F, Reichardt CM, Alomo J, Borgmann K, Frey B, Fietkau R, Rückert M, Gaipl US. Clonogenicity-based radioresistance determines the expression of immune suppressive immune checkpoint molecules after hypofractionated irradiation of MDA-MB-231 triple-negative breast cancer cells. Front Oncol 2023; 13:981239. [PMID: 37152024 PMCID: PMC10157086 DOI: 10.3389/fonc.2023.981239] [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: 06/29/2022] [Accepted: 03/28/2023] [Indexed: 05/09/2023] Open
Abstract
Only a subset of patients with triple-negative breast cancer (TNBC) benefits from a combination of radio- (RT) and immunotherapy. Therefore, we aimed to examine the impact of radioresistance and brain metastasizing potential on the immunological phenotype of TNBC cells following hypofractionated RT by analyzing cell death, immune checkpoint molecule (ICM) expression and activation of human monocyte-derived dendritic cells (DCs). MDA-MB-231 triple-negative breast cancer tumor cells were used as model system. Apoptosis was the dominant cell death form of brain metastasizing tumor cells, while Hsp70 release was generally significantly increased following RT and went along with necrosis induction. The ICMs PD-L1, PD-L2, HVEM, ICOS-L, CD137-L and OX40-L were found on the tumor cell surfaces and were significantly upregulated by RT with 5 x 5.2 Gy. Strikingly, the expression of immune suppressive ICMs was significantly higher on radioresistant clones compared to their respective non-radioresistant ones. Although hypofractionated RT led to significant cell death induction and release of Hsp70 in all tumor cell lines, human monocyte-derived DCs were not activated after co-incubation with RT-treated tumor cells. We conclude that radioresistance is a potent driver of immune suppressive ICM expression on the surface of TNBC MDA-MB-231 cells. This mechanism is generally known to predominantly influence the effector phase, rather than the priming phase, of anti-tumor immune responses.
Collapse
Affiliation(s)
- Simon Gehre
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Felix Meyer
- Laboratory of Radiobiology and Experimental Radiooncology, Department of Radiotherapy and Radiation Oncology, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Azzaya Sengedorj
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Fridolin Grottker
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Clara M. Reichardt
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Jannik Alomo
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Kerstin Borgmann
- Laboratory of Radiobiology and Experimental Radiooncology, Department of Radiotherapy and Radiation Oncology, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Michael Rückert
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Udo S. Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- *Correspondence: Udo S. Gaipl,
| |
Collapse
|
13
|
Hormuth DA, Farhat M, Christenson C, Curl B, Chad Quarles C, Chung C, Yankeelov TE. Opportunities for improving brain cancer treatment outcomes through imaging-based mathematical modeling of the delivery of radiotherapy and immunotherapy. Adv Drug Deliv Rev 2022; 187:114367. [PMID: 35654212 PMCID: PMC11165420 DOI: 10.1016/j.addr.2022.114367] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/25/2022] [Accepted: 05/25/2022] [Indexed: 11/01/2022]
Abstract
Immunotherapy has become a fourth pillar in the treatment of brain tumors and, when combined with radiation therapy, may improve patient outcomes and reduce the neurotoxicity. As with other combination therapies, the identification of a treatment schedule that maximizes the synergistic effect of radiation- and immune-therapy is a fundamental challenge. Mechanism-based mathematical modeling is one promising approach to systematically investigate therapeutic combinations to maximize positive outcomes within a rigorous framework. However, successful clinical translation of model-generated combinations of treatment requires patient-specific data to allow the models to be meaningfully initialized and parameterized. Quantitative imaging techniques have emerged as a promising source of high quality, spatially and temporally resolved data for the development and validation of mathematical models. In this review, we will present approaches to personalize mechanism-based modeling frameworks with patient data, and then discuss how these techniques could be leveraged to improve brain cancer outcomes through patient-specific modeling and optimization of treatment strategies.
Collapse
Affiliation(s)
- David A Hormuth
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Maguy Farhat
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Chase Christenson
- Departments of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Brandon Curl
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - C Chad Quarles
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Caroline Chung
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Thomas E Yankeelov
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Diagnostic Medicine, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Oncology, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Imaging Physics, MD Anderson Cancer Center, Houston, TX 77230, USA
| |
Collapse
|
14
|
Ghaffari-Nazari H, Alimohammadi M, Alimohammadi R, Rostami E, Bakhshandeh M, Webster TJ, Mahmoodi Chalbatani G, Tavakkol-Afshari J, Amir Jalali S. Radiation dose and schedule influence the abscopal effect in a bilateral murine CT26 tumor model. Int Immunopharmacol 2022; 108:108737. [DOI: 10.1016/j.intimp.2022.108737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 11/05/2022]
|
15
|
Bekker RA, Kim S, Pilon-Thomas S, Enderling H. Mathematical modeling of radiotherapy and its impact on tumor interactions with the immune system. Neoplasia 2022; 28:100796. [PMID: 35447601 PMCID: PMC9043662 DOI: 10.1016/j.neo.2022.100796] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/27/2022] [Accepted: 04/01/2022] [Indexed: 11/01/2022]
Abstract
Radiotherapy is a primary therapeutic modality widely utilized with curative intent. Traditionally tumor response was hypothesized to be due to high levels of cell death induced by irreparable DNA damage. However, the immunomodulatory aspect of radiation is now widely accepted. As such, interest into the combination of radiotherapy and immunotherapy is increasing, the synergy of which has the potential to improve tumor regression beyond that observed after either treatment alone. However, questions regarding the timing (sequential vs concurrent) and dose fractionation (hyper-, standard-, or hypo-fractionation) that result in improved anti-tumor immune responses, and thus potentially enhanced tumor inhibition, remain. Here we discuss the biological response to radiotherapy and its immunomodulatory properties before giving an overview of pre-clinical data and clinical trials concerned with answering these questions. Finally, we review published mathematical models of the impact of radiotherapy on tumor-immune interactions. Ranging from considering the impact of properties of the tumor microenvironment on the induction of anti-tumor responses, to the impact of choice of radiation site in the setting of metastatic disease, these models all have an underlying feature in common: the push towards personalized therapy.
Collapse
|
16
|
Sung W, Hong TS, Poznansky MC, Paganetti H, Grassberger C. Mathematical Modeling to Simulate the Effect of Adding Radiation Therapy to Immunotherapy and Application to Hepatocellular Carcinoma. Int J Radiat Oncol Biol Phys 2022; 112:1055-1062. [PMID: 34774999 PMCID: PMC9059476 DOI: 10.1016/j.ijrobp.2021.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/21/2021] [Accepted: 11/07/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE To develop a comprehensive framework to simulate the response to immune checkpoint inhibitors (ICIs) in combination with radiation therapy (RT) and to apply the framework for investigating ICI-RT combination regimen in patients with hepatocellular carcinoma (HCC). METHODS AND MATERIALS The mechanistic mathematical model is based on dynamic biological interactions between the immune system and the tumor using input data from patient blood samples and outcomes of clinical trials. The cell compartments are described by ordinary differential equations and represent irradiated and nonirradiated tumor cells and lymphocytes. The effect of ICI is modeled using an immune activation term that is based on tumor size changes observed in a phase 1/2 clinical trial for HCC. Simulated combination regimen are based on ongoing ICI-RT trials. RESULTS The proposed framework successfully describes tumor volume trajectories observed in early-stage clinical trials of durvalumab monotherapy in patients with HCC. For ICI-RT treatment regimen the irradiated tumor fraction is the most important parameter for the efficacy. For 90% of the tumor cells being irradiated, adding RT to ICI yields an increase in clinical benefit from 33% to 71% in nonirradiated tumor sites. The model agrees with clinical data showing an association of outcome with initial tumor volume and lymphocyte counts. We demonstrate model application in clinical trial design to predict progression-free survival curves, showing that the cohort size to show significant improvement heavily depends on the irradiated tumor fraction. CONCLUSIONS We present a framework extending radiation cell kill models to include circulating lymphocytes and the effect of ICIs and enable simulation of combination strategies. The simulations predict that a significant amount of the benefit from RT in combination with ICI stems from the reduction in irradiated tumor burden and associated immune suppression. This aspect needs to be included in the interpretation of outcomes and the design of novel combination trials.
Collapse
Affiliation(s)
- Wonmo Sung
- Division of Biophysics, Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts; Department of Biomedical Engineering and Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Mark C Poznansky
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, Massachusetts
| | - Harald Paganetti
- Division of Biophysics, Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Clemens Grassberger
- Division of Biophysics, Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
17
|
Swamy K. Stereotactic Body Radiotherapy Immunological Planning-A Review With a Proposed Theoretical Model. Front Oncol 2022; 12:729250. [PMID: 35155221 PMCID: PMC8826062 DOI: 10.3389/fonc.2022.729250] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 01/03/2022] [Indexed: 12/20/2022] Open
Abstract
In the stereotactic body radiotherapy (SBRT) and immunotherapy era, we are moving toward an “immunological radiation plan”, i.e., radiation scheduling with abscopal effect as a vital endpoint as well. The literature review of part A enumerates the advantages of the intermediate dose of SBRT 6–10 Gy per fraction, appropriate use of dose painting, proper timing with immunotherapy, and the potential of immunoadjuvants to maximize cell kill in the irradiated lesions, found to have improved the abscopal effects. Part B summarizes part A, primarily the findings of animal trials, forming the basis of the tenets of the proposed model given in part C to realize the true abscopal potential of the SBRT tumor cell kill of the index lesions. Part C proposes a theoretical model highlighting tumor vasculature integrity as the central theme for converting “abscopal effect by chance” to “abscopal effect by design” using a harmonized combinatorial approach. The proposed model principally deals with the use of SBRT in strategizing increased cell kill in irradiated index tumors along with immunomodulators as a basis for improving the consistency of the abscopal effect. Included is the possible role of integrating immunotherapy just after SBRT, “cyclical” antiangiogenics, and immunoadjuvants/immune metabolites as abscopal effect enhancers of SBRT tumor cell kill. The proposed model suggests convergence research in adopting existing numerous SBRT abscopal enhancing strategies around the central point of sustained vascular integrity to develop decisive clinical trial protocols in the future.
Collapse
|
18
|
Jiang S, Liu Z, Tian Y, Zhuang M, Piao S, Gao Y, Tam A, Hu H, Cheng W. A Comprehensive Evaluation of ZrC Nanoparticle in Combined Photothermal and Radiation Therapy for Treatment of Triple-Negative Breast Cancer. Front Oncol 2021; 11:801352. [PMID: 34993150 PMCID: PMC8724783 DOI: 10.3389/fonc.2021.801352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/24/2021] [Indexed: 01/18/2023] Open
Abstract
Because of the difficulty in treating triple-negative breast cancer (TNBC), the search for treatments has never stopped. Treatment opinions remain limited for triple-negative breast cancer (TNBC). The current treatment approach of using photothermal therapy (PTT) is often imprecise and has limited penetration below the surface of the skin. On the other hand, radiation therapy (RT) has its unavoidable disadvantages, such as side effects or ineffectiveness against hypoxic tumor microenvironment (TME). In this study, we proposed the use of ZrC nanoparticles in conjunction with RT/PTT to enhance antitumor and antimetastatic effect. We modified the ZrC nanoparticle with bovine serum albumin (BSA) and folic acid (FA), sizing desirable about 100nm. The photothermal conversion efficiency was calculated to be 40.51% and sensitizer enhancement ration (SER) was 1.8. With addition of ZrC NPs, more DNA were damaged in γ-H2AX and more ROS were detected with immunofluorescence. In vitro and vivo, the combined therapy with ZrC NPS showed the best effect of tumor cell inhibition and safety. Our results provide evidence that the combination of ZrC NPs, PT, and RT is effective in of TNBC, making it a great potential application for cancer therapy in clinic.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhao Liu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ming Zhuang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shiqi Piao
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yan Gao
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Andrew Tam
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - Hongtao Hu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Wen Cheng, ; Hongtao Hu,
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Wen Cheng, ; Hongtao Hu,
| |
Collapse
|
19
|
Rodríguez Plá M, Dualde Beltrán D, Ferrer Albiach E. Immune Checkpoints Inhibitors and SRS/SBRT Synergy in Metastatic Non-Small-Cell Lung Cancer and Melanoma: A Systematic Review. Int J Mol Sci 2021; 22:ijms222111621. [PMID: 34769050 PMCID: PMC8584181 DOI: 10.3390/ijms222111621] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Several immunotherapy (IT) agents are FDA approved for treatment of melanoma and non-small-cell lung cancer (NSCLC). The addition of stereotactic radiosurgery (SRS) or stereotactic body radiation therapy (SBRT) to immunotherapy looks promising. A systematic review was conducted to evaluate the possible synergistic effects of immune checkpoints inhibitors (ICIs) and stereotactic radiation therapy in melanoma and NSCLC. Materials and methods: Pubmed databases from January 2010 to December 2020 were reviewed to identify English language studies reporting control of local and abscopal effect of the combination of ICI-SBRT/SRS in metastatic NSCLC and melanoma cancer. The inclusion criteria were followed according to PICO criteria. Results: Thirty-nine articles were included of the 2141 initial results. The reported rates for local control were 16.5–100% and 40–94% in brain and extracerebral metastases, respectively. Distant/abscopal response rates were 1–45% in extracerebral metastases. Abscopal effect could not be evaluated in brain metastases because it was not reported in studies. Treatments were well tolerated with few grade 4 toxicities and no grade 5. Conclusions: The combined treatment of ICI-SBRT/SRS achieves high local control and non-negligible abscopal response in patients with extracerebral metastases, with its benefit in cerebral metastases being more controversial. Clinical trials are needed to better characterize the potential synergism.
Collapse
|
20
|
Alfonso JCL, Grass GD, Welsh E, Ahmed KA, Teer JK, Pilon-Thomas S, Harrison LB, Cleveland JL, Mulé JJ, Eschrich SA, Torres-Roca JF, Enderling H. Tumor-immune ecosystem dynamics define an individual Radiation Immune Score to predict pan-cancer radiocurability. Neoplasia 2021; 23:1110-1122. [PMID: 34619428 PMCID: PMC8502777 DOI: 10.1016/j.neo.2021.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 01/10/2023] Open
Abstract
Radiotherapy efficacy is the result of radiation-mediated cytotoxicity coupled with stimulation of antitumor immune responses. We develop an in silico 3-dimensional agent-based model of diverse tumor-immune ecosystems (TIES) represented as anti- or pro-tumor immune phenotypes. We validate the model in 10,469 patients across 31 tumor types by demonstrating that clinically detected tumors have pro-tumor TIES. We then quantify the likelihood radiation induces antitumor TIES shifts toward immune-mediated tumor elimination by developing the individual Radiation Immune Score (iRIS). We show iRIS distribution across 31 tumor types is consistent with the clinical effectiveness of radiotherapy, and in combination with a molecular radiosensitivity index (RSI) combines to predict pan-cancer radiocurability. We show that iRIS correlates with local control and survival in a separate cohort of 59 lung cancer patients treated with radiation. In combination, iRIS and RSI predict radiation-induced TIES shifts in individual patients and identify candidates for radiation de-escalation and treatment escalation. This is the first clinically and biologically validated computational model to simulate and predict pan-cancer response and outcomes via the perturbation of the TIES by radiotherapy.
Collapse
Affiliation(s)
- Juan C L Alfonso
- Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - G Daniel Grass
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Eric Welsh
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kamran A Ahmed
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Shari Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Louis B Harrison
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - John L Cleveland
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - James J Mulé
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Steven A Eschrich
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Javier F Torres-Roca
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Heiko Enderling
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
21
|
Angiogenesis and immune checkpoint dual blockade in combination with radiotherapy for treatment of solid cancers: opportunities and challenges. Oncogenesis 2021; 10:47. [PMID: 34247198 PMCID: PMC8272720 DOI: 10.1038/s41389-021-00335-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/02/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Several immune checkpoint blockades (ICBs) capable of overcoming the immunosuppressive roles of the tumor immune microenvironment have been approved by the US Food and Drug Administration as front-line treatments of various tumor types. However, due to the considerable heterogeneity of solid tumor cells, inhibiting one target will only influence a portion of the tumor cells. One way to enhance the tumor-killing efficiency is to develop a multiagent therapeutic strategy targeting different aspects of tumor biology and the microenvironment to provide the maximal clinical benefit for patients with late-stage disease. One such strategy is the administration of anti-PD1, an ICB, in combination with the humanized monoclonal antibody bevacizumab, an anti-angiogenic therapy, to patients with recurrent/metastatic malignancies, including hepatocellular carcinoma, metastatic renal cell carcinoma, non-small cell lung cancer, and uterine cancer. Radiotherapy (RT), a critical component of solid cancer management, has the capacity to prime the immune system for an adaptive antitumor response. Here, we present an overview of the most recent published data in preclinical and clinical studies elucidating that RT could further potentiate the antitumor effects of immune checkpoint and angiogenesis dual blockade. In addition, we explore opportunities of triple combinational treatment, as well as discuss the challenges of validating biomarkers and the management of associated toxicity.
Collapse
|
22
|
Sahebjam S, Forsyth PA, Tran ND, Arrington JA, Macaulay R, Etame AB, Walko CM, Boyle T, Peguero EN, Jaglal M, Mokhtari S, Enderling H, Raghunand N, Gatewood T, Long W, Dzierzeski JL, Evernden B, Robinson T, Wicklund MC, Kim S, Thompson ZJ, Chen DT, Chinnaiyan P, Yu HHM. Hypofractionated stereotactic re-irradiation with pembrolizumab and bevacizumab in patients with recurrent high-grade gliomas: results from a phase I study. Neuro Oncol 2021; 23:677-686. [PMID: 33173935 DOI: 10.1093/neuonc/noaa260] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Radiotherapy may synergize with programmed cell death 1 (PD1)/PD1 ligand (PD-L1) blockade. The purpose of this study was to determine the recommended phase II dose, safety/tolerability, and preliminary efficacy of combining pembrolizumab, an anti-PD1 monoclonal antibody, with hypofractionated stereotactic irradiation (HFSRT) and bevacizumab in patients with recurrent high-grade gliomas (HGGs). METHODS Eligible subjects with recurrent glioblastoma or anaplastic astrocytoma were treated with pembrolizumab (100 or 200 mg based on dose level Q3W) concurrently with HFSRT (30 Gy in 5 fractions) and bevacizumab 10 mg/kg Q2W. RESULTS Thirty-two patients were enrolled (bevacizumab-naïve, n = 24; bevacizumab-resistant, n = 8). The most common treatment-related adverse events (TRAEs) were proteinuria (40.6%), fatigue (25%), increased alanine aminotransferase (25%), and hypertension (25%). TRAEs leading to discontinuation occurred in 1 patient who experienced a grade 3 elevation of aspartate aminotransferase. In the bevacizumab-naïve cohort, 20 patients (83%) had a complete response or partial response. The median overall survival (OS) and progression-free survival (PFS) were 13.45 months (95% CI: 9.46-18.46) and 7.92 months (95% CI: 6.31-12.45), respectively. In the bevacizumab-resistant cohort, PR was achieved in 5 patients (62%). Median OS was 9.3 months (95% CI: 8.97-18.86) with a median PFS of 6.54 months (95% CI: 5.95-18.86). The majority of patients (n = 20/26; 77%) had tumor-cell/tumor-microenvironment PD-L1 expression <1%. CONCLUSIONS The combination of HFSRT with pembrolizumab and bevacizumab in patients with recurrent HGG is generally safe and well tolerated. These findings merit further investigation of HFSRT with immunotherapy in HGGs.
Collapse
Affiliation(s)
- Solmaz Sahebjam
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Peter A Forsyth
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Nam D Tran
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - John A Arrington
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Robert Macaulay
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Arnold B Etame
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Christine M Walko
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Theresa Boyle
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Edwin N Peguero
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Michael Jaglal
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Sepideh Mokhtari
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Heiko Enderling
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Natarajan Raghunand
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | - Tyra Gatewood
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Wendy Long
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | | | | | - Timothy Robinson
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | | | - Sungjune Kim
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| | | | - Dung-Tsa Chen
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Prakash Chinnaiyan
- Beaumont Health and Oakland University School of Medicine, Royal Oak, Michigan, USA
| | - Hsiang-Hsuan Michael Yu
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,University of South Florida, Tampa, Florida
| |
Collapse
|
23
|
Friedrich T, Henthorn N, Durante M. Modeling Radioimmune Response-Current Status and Perspectives. Front Oncol 2021; 11:647272. [PMID: 33796470 PMCID: PMC8008061 DOI: 10.3389/fonc.2021.647272] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
The combination of immune therapy with radiation offers an exciting and promising treatment modality in cancer therapy. It has been hypothesized that radiation induces damage signals within the tumor, making it more detectable for the immune system. In combination with inhibiting immune checkpoints an effective anti-tumor immune response may be established. This inversion from tumor immune evasion raises numerous questions to be solved to support an effective clinical implementation: These include the optimum immune drug and radiation dose time courses, the amount of damage and associated doses required to stimulate an immune response, and the impact of lymphocyte status and dynamics. Biophysical modeling can offer unique insights, providing quantitative information addressing these factors and highlighting mechanisms of action. In this work we review the existing modeling approaches of combined ‘radioimmune’ response, as well as associated fields of study. We propose modeling attempts that appear relevant for an effective and predictive model. We emphasize the importance of the time course of drug and dose delivery in view to the time course of the triggered biological processes. Special attention is also paid to the dose distribution to circulating blood lymphocytes and the effect this has on immune competence.
Collapse
Affiliation(s)
- Thomas Friedrich
- Biophysics Department, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| | - Nicholas Henthorn
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Marco Durante
- Biophysics Department, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany.,Institute for Solid State Physics, Technical University Darmstadt, Darmstadt, Germany
| |
Collapse
|
24
|
Lee BM, Seong J. Radiotherapy as an immune checkpoint blockade combination strategy for hepatocellular carcinoma. World J Gastroenterol 2021; 27:919-927. [PMID: 33776363 PMCID: PMC7968135 DOI: 10.3748/wjg.v27.i10.919] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/29/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
In the immune oncology era, the clinical efficacy of immune checkpoint inhibitors (ICIs) against most solid cancers is well known. In hepatocellular carcinoma, the recent success of combination therapy with targeting agents has accelerated the search for novel combination strategies. Radiotherapy (RT), an attractive modality, can be combined with ICIs, which act as strong modulators of the tumor immune microenvironment. Herein, we discuss immune modulation caused by radiation and the current trials of RT–ICI combination treatment as well as future perspectives.
Collapse
Affiliation(s)
- Byung Min Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Jinsil Seong
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, South Korea
| |
Collapse
|
25
|
Johnson KE, Howard GR, Morgan D, Brenner EA, Gardner AL, Durrett RE, Mo W, Al’Khafaji A, Sontag ED, Jarrett AM, Yankeelov TE, Brock A. Integrating transcriptomics and bulk time course data into a mathematical framework to describe and predict therapeutic resistance in cancer. Phys Biol 2020; 18:016001. [PMID: 33215611 PMCID: PMC8156495 DOI: 10.1088/1478-3975/abb09c] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A significant challenge in the field of biomedicine is the development of methods to integrate the multitude of dispersed data sets into comprehensive frameworks to be used to generate optimal clinical decisions. Recent technological advances in single cell analysis allow for high-dimensional molecular characterization of cells and populations, but to date, few mathematical models have attempted to integrate measurements from the single cell scale with other types of longitudinal data. Here, we present a framework that actionizes static outputs from a machine learning model and leverages these as measurements of state variables in a dynamic model of treatment response. We apply this framework to breast cancer cells to integrate single cell transcriptomic data with longitudinal bulk cell population (bulk time course) data. We demonstrate that the explicit inclusion of the phenotypic composition estimate, derived from single cell RNA-sequencing data (scRNA-seq), improves accuracy in the prediction of new treatments with a concordance correlation coefficient (CCC) of 0.92 compared to a prediction accuracy of CCC = 0.64 when fitting on longitudinal bulk cell population data alone. To our knowledge, this is the first work that explicitly integrates single cell clonally-resolved transcriptome datasets with bulk time-course data to jointly calibrate a mathematical model of drug resistance dynamics. We anticipate this approach to be a first step that demonstrates the feasibility of incorporating multiple data types into mathematical models to develop optimized treatment regimens from data.
Collapse
Affiliation(s)
- Kaitlyn E Johnson
- Department of Biomedical Engineering, The University of
Texas at Austin, Austin, TX, 78712, United States of America
| | - Grant R Howard
- Department of Biomedical Engineering, The University of
Texas at Austin, Austin, TX, 78712, United States of America
| | - Daylin Morgan
- Department of Biomedical Engineering, The University of
Texas at Austin, Austin, TX, 78712, United States of America
| | - Eric A Brenner
- Department of Biomedical Engineering, The University of
Texas at Austin, Austin, TX, 78712, United States of America
- Institute for Cellular and Molecular Biology, The
University of Texas at Austin, Austin, TX, 78712, United States of America
| | - Andrea L Gardner
- Department of Biomedical Engineering, The University of
Texas at Austin, Austin, TX, 78712, United States of America
| | - Russell E Durrett
- Department of Biomedical Engineering, The University of
Texas at Austin, Austin, TX, 78712, United States of America
- Institute for Cellular and Molecular Biology, The
University of Texas at Austin, Austin, TX, 78712, United States of America
| | - William Mo
- Department of Biomedical Engineering, The University of
Texas at Austin, Austin, TX, 78712, United States of America
| | - Aziz Al’Khafaji
- Department of Biomedical Engineering, The University of
Texas at Austin, Austin, TX, 78712, United States of America
- Institute for Cellular and Molecular Biology, The
University of Texas at Austin, Austin, TX, 78712, United States of America
| | - Eduardo D Sontag
- Department of Electrical and Computer Engineering,
Northeastern University, Boston, MA, 02115, United States of America
- Department of Bioengineering, Northeastern University,
Boston, MA, 02115, United States of America
- Laboratory of Systems Pharmacology, Program in Therapeutics
Science, Harvard Medical School, Boston, MA, 02115, United States of America
| | - Angela M Jarrett
- Livestrong Cancer Institutes, Dell Medical School, The
University of Texas at Austin, Austin, TX, 78712, United States of America
- Oden Institute for Computational Engineering and Sciences,
The University of Texas at Austin
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of
Texas at Austin, Austin, TX, 78712, United States of America
- Livestrong Cancer Institutes, Dell Medical School, The
University of Texas at Austin, Austin, TX, 78712, United States of America
- Oden Institute for Computational Engineering and Sciences,
The University of Texas at Austin
- Department of Diagnostic Medicine, The University of Texas
at Austin, Austin, TX, 78712, United States of America
- Department of Oncology, The University of Texas at Austin,
Austin, TX, 78712, United States of America
- Department of Imaging Physics, The MD Anderson Cancer
Center Houston, TX, 77030, United States of America
| | - Amy Brock
- Department of Biomedical Engineering, The University of
Texas at Austin, Austin, TX, 78712, United States of America
- Institute for Cellular and Molecular Biology, The
University of Texas at Austin, Austin, TX, 78712, United States of America
- Livestrong Cancer Institutes, Dell Medical School, The
University of Texas at Austin, Austin, TX, 78712, United States of America
| |
Collapse
|
26
|
Wang D, Zhang X, Gao Y, Cui X, Yang Y, Mao W, Li M, Zhang B, Yu J. Research Progress and Existing Problems for Abscopal Effect. Cancer Manag Res 2020; 12:6695-6706. [PMID: 32801902 PMCID: PMC7413699 DOI: 10.2147/cmar.s245426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
Radiation therapy plays a vital role in the treatment of tumours. In particular, the occurrence of the “abscopal effect” brings about a favourable turn for the treatment of patients with advanced metastatic malignant tumours. Because of the abscopal effect, non-irradiated areas are also treated. However, the abscopal effect occurs by chance, not through seeking. Although the abscopal effect has been studied enthusiastically, the desired result does not appear to be achieved. Moreover, its combination with immunotherapy appears to be overwhelming. There is an opinion that abscopal effect is difficult to achieve by irradiation of a single tumour, and irradiation of multiple or total lesions is advocated to increase the possibility of obtaining clinically meaningful outcomes. Obviously, there are still questions about the mechanism, condition and possibility underlying the occurrence of the abscopal effect. Can the abscopal effect truly change the future treatment strategy as the researchers expect? What are the current problems? This article reviewed the research in recent years to explore the progress and controversy surrounding the abscopal effect of radiation therapy.
Collapse
Affiliation(s)
- Di Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Xia Zhang
- Department of Oncology, The Fifth People's Hospital of Dalian, Dalian, People's Republic of China
| | - Yajie Gao
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Xiaonan Cui
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Yanqin Yang
- Department of Radiation Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Weifeng Mao
- The School of Basic Medical Sciences, Dalian Medical University, Dalian, People's Republic of China
| | - Minghuan Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Bin Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| |
Collapse
|
27
|
Sung W, Grassberger C, McNamara AL, Basler L, Ehrbar S, Tanadini-Lang S, Hong TS, Paganetti H. A tumor-immune interaction model for hepatocellular carcinoma based on measured lymphocyte counts in patients undergoing radiotherapy. Radiother Oncol 2020; 151:73-81. [PMID: 32679308 DOI: 10.1016/j.radonc.2020.07.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE The impact of radiation therapy on the immune system has recently gained attention particularly when delivered in combination with immunotherapy. However, it is unclear how different treatment fractionation regimens influence the interaction between the immune system and radiation. The goal of this work was to develop a mathematical model that quantifies both the immune stimulating as well as the immunosuppressive effects of radiotherapy and simulates the effects of different fractionation regimens based on patient data. METHODS AND MATERIALS The framework describes the temporal evolution of tumor cells, lymphocytes, and inactivated dying tumor cells releasing antigens during radiation therapy, specifically modeling how recruited lymphocytes inhibit tumor progression. The parameters of the model were partly taken from the literature and in part extracted from blood samples (circulating lymphocytes: CLs) collected from hepatocellular carcinoma patients undergoing radiotherapy and their outcomes. The dose volume histograms to circulating lymphocytes were calculated with a probability-based model. RESULTS Based on the fitted parameters, the model enabled a study into the depletion and recovery of CLs in patients as a function of fractionation regimen. Our results quantify the ability of short fractionation regimens to lead to shorter periods of lymphocyte depletion and predict faster recovery after the end of treatment. The model shows that treatment breaks between fractions can prolong the period of lymphocyte depletion and should be avoided. CONCLUSIONS This study introduces a mathematical model for tumor-immune interactions using clinically extracted radiotherapy patient data, which can be applied to design trials aimed at minimizing lymphocyte depleting effects in radiation therapy.
Collapse
Affiliation(s)
- Wonmo Sung
- Department of Radiation Oncology, Massachusetts General Hospital/Harvard Medical School, United States
| | - Clemens Grassberger
- Department of Radiation Oncology, Massachusetts General Hospital/Harvard Medical School, United States
| | - Aimee Louise McNamara
- Department of Radiation Oncology, Massachusetts General Hospital/Harvard Medical School, United States
| | - Lucas Basler
- Department of Radiation Oncology, Paul Scherrer Institut, Villigen, Switzerland
| | - Stefanie Ehrbar
- Department of Radiation Oncology, University Hospital Zurich, Switzerland
| | | | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital/Harvard Medical School, United States
| | - Harald Paganetti
- Department of Radiation Oncology, Massachusetts General Hospital/Harvard Medical School, United States.
| |
Collapse
|
28
|
López Alfonso JC, Poleszczuk J, Walker R, Kim S, Pilon-Thomas S, Conejo-Garcia JJ, Soliman H, Czerniecki B, Harrison LB, Enderling H. Immunologic Consequences of Sequencing Cancer Radiotherapy and Surgery. JCO Clin Cancer Inform 2020; 3:1-16. [PMID: 30964698 DOI: 10.1200/cci.18.00075] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
PURPOSE Early-stage cancers are routinely treated with surgery followed by radiotherapy (SR). Radiotherapy before surgery (RS) has been widely ignored for some cancers. We evaluate overall survival (OS) and disease-free survival (DFS) with SR and RS for different cancer types and simulate the plausibility of RS- and SR-induced antitumor immunity contributing to outcomes. MATERIALS AND METHODS We analyzed a SEER data set of early-stage cancers treated with SR or RS. OS and DFS were calculated for cancers with sufficient numbers for statistical power (cancers of lung and bronchus, esophagus, rectum, cervix uteri, corpus uteri, and breast). We simulated the immunologic consequences of SR, RS, and radiotherapy alone in a mathematical model of tumor-immune interactions. RESULTS RS improved OS for cancers with low 20-year survival rates (lung: hazard ratio [HR], 0.88; P = .046) and improved DFS for cancers with higher survival (breast: HR = 0.64; P < .001). For rectal cancer, with intermediate 20-year survival, RS improved both OS (HR = 0.89; P = .006) and DFS (HR = 0.86; P = .04). Model simulations suggested that RS could increase OS by eliminating cancer for a broader range of model parameters and radiotherapy-induced antitumor immunity compared with SR for selected parameter combinations. This could create an immune memory that may explain increased DFS after RS for certain cancers. CONCLUSION Study results suggest plausibility that radiation to the bulk of the tumor could induce a more robust immune response and better harness the synergy of radiotherapy and antitumor immunity than postsurgical radiation to the tumor bed. This exploratory study provides motivation for prospective evaluation of immune activation of RS versus SR in controlled clinical studies.
Collapse
Affiliation(s)
- Juan Carlos López Alfonso
- Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jan Poleszczuk
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Rachel Walker
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Sungjune Kim
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Shari Pilon-Thomas
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Jose J Conejo-Garcia
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Hatem Soliman
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Brian Czerniecki
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Louis B Harrison
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Heiko Enderling
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
29
|
Ellerin BE, Demandante CGN, Martins JT. Pure abscopal effect of radiotherapy in a salivary gland carcinoma: Case report, literature review, and a search for new approaches. Cancer Radiother 2020; 24:226-246. [PMID: 32192840 DOI: 10.1016/j.canrad.2020.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/15/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
We report the case of an 84-year-old woman with poorly differentiated non-small cell carcinoma of the right parotid who presented with headache, was found to have a primary right parotid gland cancer as well as metastatic disease, and underwent palliative radiotherapy to the primary site. The patient received no chemotherapy or immunotherapy, but both the primary site and several non-irradiated foci in the lungs regressed or completely resolved. The patient remained free of disease for about one year before progression. The case is a rare instance of abscopal regression of metastatic disease in the absence of pharmacologic immunomodulation. A literature review surveys the history of the abscopal effect of radiation therapy, attempts to understand the mechanisms of its successes and failures, and points to new approaches that can inform and improve the outcomes of radioimmunotherapy.
Collapse
Affiliation(s)
| | | | - J T Martins
- UT Health HOPE Cancer Center, Tyler, TX 75701, USA
| |
Collapse
|
30
|
Massaccesi M, Boldrini L, Piras A, Stimato G, Quaranta F, Azario L, Mattiucci GC, Valentini V. Spatially fractionated radiotherapy (SFRT) targeting the hypoxic tumor segment for the intentional induction of non-targeted effects: An in silico study to exploit a new treatment paradigm. Tech Innov Patient Support Radiat Oncol 2020; 14:11-14. [PMID: 32154394 PMCID: PMC7052565 DOI: 10.1016/j.tipsro.2020.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 02/15/2020] [Accepted: 02/17/2020] [Indexed: 11/25/2022] Open
Abstract
Introduction The possibility of intentionally triggering non targeted effects (NTEs) using spatially fractionated radiotherapy (SFRT) alone or combined with immunotherapy is an intriguing and fascinating area of research. Among different techniques for SFRT, stereotactic body radiotherapy targeting exclusively the central hypoxic segment of bulky tumors, (SBRT-PATHY) might trigger immunogenic cell death more efficiently. This in silico study aims to identify the best possible dosimetric trade-off for prescribing SFRT with volumetric modulated arc (VMAT) based stereotactic radiotherapy (SRT). Material and methods Eight spherical volumes defined "Gross Tumor Volumes" (GTVs) were generated with diameters of 3-10 cm (with incremental steps of 1 cm), simulating tumor lesions. The inner third part of each GTV (GTVcentral) was selected to simulate the central hypoxic area and a ring structure was derived around it to simulate the tumor periphery (GTVperipheral). Volumetric modulated arc radiation treatment (VMAT) plans were calculated to deliver a single fraction of 10 Gy to each GTVcentral with different dose prescription methods: target mean and isodose driven (40, 50, 60, 70, 80 and 90%).The volume of GTVperipheral receiving less than 2 Gy was recorded as dosimetric performance indicator. Results 56 possible dosimetric scenarios were analyzed. The largest percentage of GTVperipheral spared from the dose of 2 Gy was achieved with dose prescription methods to the 70% isodose line for lesions smaller than 6 cm (range 42.9-48.4%) and to the target mean for larger ones (range 52.9-64.5%). Conclusions Optimizing the dose prescription method may reduce the dose to tumor periphery in VMAT-based SFRT, thus potentially sparing tumor infiltrating immune cells. The optimal method may vary according to the size of the lesion. This should be taken into account when designing prospective trials using SFRT.
Collapse
Affiliation(s)
- M Massaccesi
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, UOC di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Roma, Italy
| | - L Boldrini
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, UOC di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Roma, Italy
| | - A Piras
- Università Cattolica del Sacro Cuore, Istituto di Radiologia, Roma, Italy
| | - G Stimato
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, UOC di Fisica Sanitaria, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Roma, Italy
| | - F Quaranta
- Università Cattolica del Sacro Cuore, Istituto di Fisica, Roma, Italy
| | - L Azario
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, UOC di Fisica Sanitaria, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Roma, Italy.,Università Cattolica del Sacro Cuore, Istituto di Fisica, Roma, Italy
| | - G C Mattiucci
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, UOC di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Roma, Italy.,Università Cattolica del Sacro Cuore, Istituto di Radiologia, Roma, Italy
| | - V Valentini
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, UOC di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Roma, Italy.,Università Cattolica del Sacro Cuore, Istituto di Radiologia, Roma, Italy
| |
Collapse
|
31
|
Montaseri G, Alfonso JCL, Hatzikirou H, Meyer-Hermann M. A minimal modeling framework of radiation and immune system synergy to assist radiotherapy planning. J Theor Biol 2020; 486:110099. [PMID: 31790681 DOI: 10.1016/j.jtbi.2019.110099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/15/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023]
Abstract
Recent evidence indicates the ability of radiotherapy to induce local and systemic tumor-specific immune responses as a result of immunogenic cell death. However, fractionation regimes routinely used in clinical practice typically ignore the synergy between radiation and the immune system, and instead attempt to completely eradicate tumors by the direct lethal effect of radiation on cancer cells. This paradigm is expected to change in the near future due to the potential benefits of considering radiation-induced antitumor immunity during treatment planning. Towards this goal, we propose a minimal modeling framework based on key aspects of the tumor-immune system interplay to simulate the effects of radiation on tumors and the immunological consequences of radiotherapy. The impacts of tumor-associated vasculature and intratumoral oxygen-mediated heterogeneity on treatment outcomes are ininvestigated. The model provides estimates of the minimum radiation doses required for tumor eradication given a certain number of treatment fractions. Moreover, estimates of treatment duration for disease control given predetermined fractional radiation doses can be also obtained. Although theoretical in nature, this study motivates the development and establishment of immune-based decision-support tools in radiotherapy planning.
Collapse
Affiliation(s)
- Ghazal Montaseri
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany; Centre for Individualised Infection Medicine (CIIM), Hannover, Germany
| | - Juan Carlos López Alfonso
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany; Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Haralampos Hatzikirou
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany; Centre for Individualised Infection Medicine (CIIM), Hannover, Germany; Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Germany.
| |
Collapse
|
32
|
Van Coillie S, Wiernicki B, Xu J. Molecular and Cellular Functions of CTLA-4. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:7-32. [PMID: 32185705 DOI: 10.1007/978-981-15-3266-5_2] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is an inhibitory receptor belonging to the CD28 immunoglobulin subfamily, expressed primarily by T-cells. Its ligands, CD80 and CD86, are typically found on the surface of antigen-presenting cells and can either bind CD28 or CTLA-4, resulting in a costimulatory or a co-inhibitory response, respectively. Because of its dampening effect, CTLA-4 is a crucial regulator of T-cell homeostasis and self-tolerance. The mechanisms by which CTLA-4 exerts its inhibitory function can be categorized as either cell-intrinsic (affects the CTLA-4 expressing T-cell) or cell-extrinsic (affects secondary cells). Research from the last decade has shown that CTLA-4 mainly acts in a cell-extrinsic manner via its competition with CD28, CTLA-4-mediated trans-endocytosis of CD80 and CD86, and its direct tolerogenic effects on the interacting cell. Nonetheless, intrinsic CTLA-4 signaling has been implicated in T-cell motility and the regulation of CTLA-4 its subcellular localization amongst others. CTLA-4 is well recognized as a key immune checkpoint and has gained significant momentum as a therapeutic target in the field of autoimmunity and cancer. In this chapter, we describe the role of costimulation in immune response induction as well as the main mechanisms by which CTLA-4 can inhibit this process.
Collapse
Affiliation(s)
- Samya Van Coillie
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, 9052, Ghent, Belgium.
| | - Bartosz Wiernicki
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, 9052, Ghent, Belgium
| | - Jie Xu
- Institutes of Biomedical Sciences, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
33
|
Assessing the interactions between radiotherapy and antitumour immunity. Nat Rev Clin Oncol 2019; 16:729-745. [PMID: 31243334 DOI: 10.1038/s41571-019-0238-9] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2019] [Indexed: 12/17/2022]
Abstract
Immunotherapy, specifically the introduction of immune checkpoint inhibitors, has transformed the treatment of cancer, enabling long-term tumour control even in individuals with advanced-stage disease. Unfortunately, only a small subset of patients show a response to currently available immunotherapies. Despite a growing consensus that combining immune checkpoint inhibitors with radiotherapy can increase response rates, this approach might be limited by the development of persistent radiation-induced immunosuppression. The ultimate goal of combining immunotherapy with radiotherapy is to induce a shift from an ineffective, pre-existing immune response to a long-lasting, therapy-induced immune response at all sites of disease. To achieve this goal and enable the adaptation and monitoring of individualized treatment approaches, assessment of the dynamic changes in the immune system at the patient level is essential. In this Review, we summarize the available clinical data, including forthcoming methods to assess the immune response to radiotherapy at the patient level, ranging from serum biomarkers to imaging techniques that enable investigation of immune cell dynamics in patients. Furthermore, we discuss modelling approaches that have been developed to predict the interaction of immunotherapy with radiotherapy, and highlight how they could be combined with biomarkers of antitumour immunity to optimize radiotherapy regimens and maximize their synergy with immunotherapy.
Collapse
|
34
|
Johnson KE, Howard G, Mo W, Strasser MK, Lima EABF, Huang S, Brock A. Cancer cell population growth kinetics at low densities deviate from the exponential growth model and suggest an Allee effect. PLoS Biol 2019; 17:e3000399. [PMID: 31381560 PMCID: PMC6695196 DOI: 10.1371/journal.pbio.3000399] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/15/2019] [Accepted: 07/08/2019] [Indexed: 12/30/2022] Open
Abstract
Most models of cancer cell population expansion assume exponential growth kinetics at low cell densities, with deviations to account for observed slowing of growth rate only at higher densities due to limited resources such as space and nutrients. However, recent preclinical and clinical observations of tumor initiation or recurrence indicate the presence of tumor growth kinetics in which growth rates scale positively with cell numbers. These observations are analogous to the cooperative behavior of species in an ecosystem described by the ecological principle of the Allee effect. In preclinical and clinical models, however, tumor growth data are limited by the lower limit of detection (i.e., a measurable lesion) and confounding variables, such as tumor microenvironment, and immune responses may cause and mask deviations from exponential growth models. In this work, we present alternative growth models to investigate the presence of an Allee effect in cancer cells seeded at low cell densities in a controlled in vitro setting. We propose a stochastic modeling framework to disentangle expected deviations due to small population size stochastic effects from cooperative growth and use the moment approach for stochastic parameter estimation to calibrate the observed growth trajectories. We validate the framework on simulated data and apply this approach to longitudinal cell proliferation data of BT-474 luminal B breast cancer cells. We find that cell population growth kinetics are best described by a model structure that considers the Allee effect, in that the birth rate of tumor cells increases with cell number in the regime of small population size. This indicates a potentially critical role of cooperative behavior among tumor cells at low cell densities with relevance to early stage growth patterns of emerging and relapsed tumors.
Collapse
Affiliation(s)
- Kaitlyn E. Johnson
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - Grant Howard
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - William Mo
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - Michael K. Strasser
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Ernesto A. B. F. Lima
- Institute for Computation Engineering and Sciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Sui Huang
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
- Department of Oncology, Livestrong Cancer Institute, Dell Medical School, The University of Texas at Austin, Austin, Texas, United States of America
| |
Collapse
|
35
|
Brady R, Enderling H. Mathematical Models of Cancer: When to Predict Novel Therapies, and When Not to. Bull Math Biol 2019; 81:3722-3731. [PMID: 31338741 PMCID: PMC6764933 DOI: 10.1007/s11538-019-00640-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/02/2019] [Indexed: 12/27/2022]
Abstract
The number of publications on mathematical modeling of cancer is growing at an exponential rate, according to PubMed records, provided by the US National Library of Medicine and the National Institutes of Health. Seminal papers have initiated and promoted mathematical modeling of cancer and have helped define the field of mathematical oncology (Norton and Simon in J Natl Cancer Inst 58:1735-1741, 1977; Norton in Can Res 48:7067-7071, 1988; Hahnfeldt et al. in Can Res 59:4770-4775, 1999; Anderson et al. in Comput Math Methods Med 2:129-154, 2000. https://doi.org/10.1080/10273660008833042 ; Michor et al. in Nature 435:1267-1270, 2005. https://doi.org/10.1038/nature03669 ; Anderson et al. in Cell 127:905-915, 2006. https://doi.org/10.1016/j.cell.2006.09.042 ; Benzekry et al. in PLoS Comput Biol 10:e1003800, 2014. https://doi.org/10.1371/journal.pcbi.1003800 ). Following the introduction of undergraduate and graduate programs in mathematical biology, we have begun to see curricula developing with specific and exclusive focus on mathematical oncology. In 2018, 218 articles on mathematical modeling of cancer were published in various journals, including not only traditional modeling journals like the Bulletin of Mathematical Biology and the Journal of Theoretical Biology, but also publications in renowned science, biology, and cancer journals with tremendous impact in the cancer field (Cell, Cancer Research, Clinical Cancer Research, Cancer Discovery, Scientific Reports, PNAS, PLoS Biology, Nature Communications, eLife, etc). This shows the breadth of cancer models that are being developed for multiple purposes. While some models are phenomenological in nature following a bottom-up approach, other models are more top-down data-driven. Here, we discuss the emerging trend in mathematical oncology publications to predict novel, optimal, sometimes even patient-specific treatments, and propose a convention when to use a model to predict novel treatments and, probably more importantly, when not to.
Collapse
Affiliation(s)
- Renee Brady
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33647, USA
| | - Heiko Enderling
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33647, USA.
| |
Collapse
|
36
|
Dzobo K, Adotey S, Thomford NE, Dzobo W. Integrating Artificial and Human Intelligence: A Partnership for Responsible Innovation in Biomedical Engineering and Medicine. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 24:247-263. [PMID: 31313972 DOI: 10.1089/omi.2019.0038] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Historically, the term "artificial intelligence" dates to 1956 when it was first used in a conference at Dartmouth College in the US. Since then, the development of artificial intelligence has in part been shaped by the field of neuroscience. By understanding the human brain, scientists have attempted to build new intelligent machines capable of performing complex tasks akin to humans. Indeed, future research into artificial intelligence will continue to benefit from the study of the human brain. While the development of artificial intelligence algorithms has been fast paced, the actual use of most artificial intelligence (AI) algorithms in biomedical engineering and clinical practice is still markedly below its conceivably broader potentials. This is partly because for any algorithm to be incorporated into existing workflows it has to stand the test of scientific validation, clinical and personal utility, application context, and is equitable as well. In this context, there is much to be gained by combining AI and human intelligence (HI). Harnessing Big Data, computing power and storage capacities, and addressing societal issues emergent from algorithm applications, demand deploying HI in tandem with AI. Very few countries, even economically developed states, lack adequate and critical governance frames to best understand and steer the AI innovation trajectories in health care. Drug discovery and translational pharmaceutical research stand to gain from AI technology provided they are also informed by HI. In this expert review, we analyze the ways in which AI applications are likely to traverse the continuum of life from birth to death, and encompassing not only humans but also all animal, plant, and other living organisms that are increasingly touched by AI. Examples of AI applications include digital health, diagnosis of diseases in newborns, remote monitoring of health by smart devices, real-time Big Data analytics for prompt diagnosis of heart attacks, and facial analysis software with consequences on civil liberties. While we underscore the need for integration of AI and HI, we note that AI technology does not have to replace medical specialists or scientists and rather, is in need of such expert HI. Altogether, AI and HI offer synergy for responsible innovation and veritable prospects for improving health care from prevention to diagnosis to therapeutics while unintended consequences of automation emergent from AI and algorithms should be borne in mind on scientific cultures, work force, and society at large.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Medical Campus, Anzio Road, Observatory 7925, Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sampson Adotey
- International Development Innovation Network, D-Lab, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Nicholas E Thomford
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Witness Dzobo
- Pathology and Immunology Department, University Hospital Southampton, Mail Point B, Tremona Road, Southampton, UK.,University of Portsmouth, Faculty of Science, St Michael's Building, White Swan Road, Portsmouth, UK
| |
Collapse
|
37
|
Grapin M, Richard C, Limagne E, Boidot R, Morgand V, Bertaut A, Derangere V, Laurent PA, Thibaudin M, Fumet JD, Crehange G, Ghiringhelli F, Mirjolet C. Optimized fractionated radiotherapy with anti-PD-L1 and anti-TIGIT: a promising new combination. J Immunother Cancer 2019; 7:160. [PMID: 31238970 PMCID: PMC6593525 DOI: 10.1186/s40425-019-0634-9] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/10/2019] [Indexed: 02/06/2023] Open
Abstract
PURPOSE/OBJECTIVE Radiotherapy (RT) induces an immunogenic antitumor response, but also some immunosuppressive barriers. It remains unclear how different fractionation protocols can modulate the immune microenvironment. Clinical studies are ongoing to evaluate immune checkpoint inhibitors (ICI) in association with RT. However, only few trials aim to optimize the RT fractionation to improve efficacy of these associations. Here we sought to characterize the effect of different fractionation protocols on immune response with a view to associating them with ICI. MATERIALS/METHODS Mice bearing subcutaneous CT26 colon tumors were irradiated using a SARRP device according to different radiation schemes with a same biologically effective dose. Mice were monitored for tumor growth. The radiation immune response (lymphoid, myeloid cells, lymphoid cytokines and immune checkpoint targets) was monitored by flow cytometry at different timepoints after treatment and by RNA sequencing analysis (RNAseq). The same radiation protocols were performed with and without inhibitors of immune checkpoints modulated by RT. RESULTS In the absence of ICI, we showed that 18x2Gy and 3x8Gy induced the longest tumor growth delay compared to 1×16.4Gy. While 3x8Gy and 1×16.4Gy induced a lymphoid response (CD8+ T-cells, Regulators T-cells), 18x2Gy induced a myeloid response (myeloid-derived suppressor cells, tumor-associated macrophages 2). The secretion of granzyme B by CD8+ T cells was increased to a greater extent with 3x8Gy. The expression of PD-L1 by tumor cells was moderately increased by RT, but most durably with 18x2Gy. T cell immunoreceptor with Ig and ITIM domains (TIGIT) expression by CD8+ T-cells was increased with 3x8Gy, but decreased with 18x2Gy. These results were also observed with RNAseq. RT was dramatically more effective with 3x8Gy compared to all the other treatments schemes when associated with anti-TIGIT and anti-PD-L1 (9/10 mice in complete response). The association of anti-PD-L1 and RT was also effective in the 18x2Gy group (8/12 mice in complete response). CONCLUSION Each fractionation scheme induced different lymphoid and myeloid responses as well as various modulations of PD-L1 and TIGIT expression. Furthermore, 3x8Gy was the most effective protocol when associated with anti-PD-L1 and anti-TIGIT. This is the first study combining RT and anti-TIGIT with promising results; further studies are warranted.
Collapse
Affiliation(s)
- Mathieu Grapin
- Department of Radiation Oncology, Unicancer - Georges-Francois Leclerc Cancer Center, 1 rue Professeur Marion 77 980, 21079, Dijon Cedex, BP, France
| | - Corentin Richard
- Cancer Biology Research Platform, Unicancer - Georges-Francois Leclerc Cancer Center, Dijon, France
| | - Emeric Limagne
- Cancer Biology Research Platform, Unicancer - Georges-Francois Leclerc Cancer Center, Dijon, France
| | - Romain Boidot
- Cancer Biology Research Platform, Unicancer - Georges-Francois Leclerc Cancer Center, Dijon, France.,INSERM UMR 1231, Dijon, France
| | - Véronique Morgand
- Department of Radiation Oncology, Unicancer - Georges-Francois Leclerc Cancer Center, 1 rue Professeur Marion 77 980, 21079, Dijon Cedex, BP, France
| | - Aurélie Bertaut
- Methodology, data-management and biostatistics unit, Unicancer - Georges-Francois Leclerc Cancer Center , Dijon, France
| | - Valentin Derangere
- Cancer Biology Research Platform, Unicancer - Georges-Francois Leclerc Cancer Center, Dijon, France
| | - Pierre-Antoine Laurent
- Department of Radiation Oncology, Unicancer - Georges-Francois Leclerc Cancer Center, 1 rue Professeur Marion 77 980, 21079, Dijon Cedex, BP, France
| | - Marion Thibaudin
- Cancer Biology Research Platform, Unicancer - Georges-Francois Leclerc Cancer Center, Dijon, France
| | - Jean David Fumet
- Cancer Biology Research Platform, Unicancer - Georges-Francois Leclerc Cancer Center, Dijon, France.,Methodology, data-management and biostatistics unit, Unicancer - Georges-Francois Leclerc Cancer Center , Dijon, France
| | - Gilles Crehange
- Department of Radiation Oncology, Unicancer - Georges-Francois Leclerc Cancer Center, 1 rue Professeur Marion 77 980, 21079, Dijon Cedex, BP, France
| | - François Ghiringhelli
- Cancer Biology Research Platform, Unicancer - Georges-Francois Leclerc Cancer Center, Dijon, France.,INSERM UMR 1231, Dijon, France.,Department of Medical Oncology, Unicancer - Georges-Francois Leclerc Cancer Center, Dijon, France
| | - Céline Mirjolet
- Department of Radiation Oncology, Unicancer - Georges-Francois Leclerc Cancer Center, 1 rue Professeur Marion 77 980, 21079, Dijon Cedex, BP, France. .,INSERM UMR 1231, Dijon, France.
| |
Collapse
|
38
|
Boustani J, Grapin M, Laurent PA, Apetoh L, Mirjolet C. The 6th R of Radiobiology: Reactivation of Anti-Tumor Immune Response. Cancers (Basel) 2019; 11:E860. [PMID: 31226866 PMCID: PMC6627091 DOI: 10.3390/cancers11060860] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 12/12/2022] Open
Abstract
Historically, the 4Rs and then the 5Rs of radiobiology explained the effect of radiation therapy (RT) fractionation on the treatment efficacy. These 5Rs are: Repair, Redistribution, Reoxygenation, Repopulation and, more recently, intrinsic Radiosensitivity. Advances in radiobiology have demonstrated that RT is able to modify the tumor micro environment (TME) and to induce a local and systemic (abscopal effect) immune response. Conversely, RT is able to increase some immunosuppressive barriers, which can lead to tumor radioresistance. Fractionation and dose can affect the immunomodulatory properties of RT. Here, we review how fractionation, dose and timing shape the RT-induced anti-tumor immune response and the therapeutic effect of RT. We discuss how immunomodulators targeting immune checkpoint inhibitors and the cGAS/STING (cyclic GMP-AMP Synthase/Stimulator of Interferon Genes) pathway can be successfully combined with RT. We then review current trials evaluating the RT/Immunotherapy combination efficacy and suggest new innovative associations of RT with immunotherapies currently used in clinic or in development with strategic schedule administration (fractionation, dose, and timing) to reverse immune-related radioresistance. Overall, our work will present the existing evidence supporting the claim that the reactivation of the anti-tumor immune response can be regarded as the 6th R of Radiobiology.
Collapse
Affiliation(s)
- Jihane Boustani
- Department of Radiation Oncology, Unicancer-Georges-Francois Leclerc Cancer Center, Dijon, France.
| | - Mathieu Grapin
- Department of Radiation Oncology, Unicancer-Georges-Francois Leclerc Cancer Center, Dijon, France.
| | - Pierre-Antoine Laurent
- Department of Radiation Oncology, Unicancer-Georges-Francois Leclerc Cancer Center, Dijon, France.
| | | | - Céline Mirjolet
- Department of Radiation Oncology, Unicancer-Georges-Francois Leclerc Cancer Center, Dijon, France.
- INSERM, U1231 Dijon, France.
| |
Collapse
|
39
|
Spring BQ, Lang RT, Kercher EM, Rizvi I, Wenham RM, Conejo-Garcia JR, Hasan T, Gatenby RA, Enderling H. Illuminating the Numbers: Integrating Mathematical Models to Optimize Photomedicine Dosimetry and Combination Therapies. FRONTIERS IN PHYSICS 2019; 7:46. [PMID: 31123672 PMCID: PMC6529192 DOI: 10.3389/fphy.2019.00046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cancer photomedicine offers unique mechanisms for inducing local tumor damage with the potential to stimulate local and systemic anti-tumor immunity. Optically-active nanomedicine offers these features as well as spatiotemporal control of tumor-focused drug release to realize synergistic combination therapies. Achieving quantitative dosimetry is a major challenge, and dosimetry is fundamental to photomedicine for personalizing and tailoring therapeutic regimens to specific patients and anatomical locations. The challenge of dosimetry is perhaps greater for photomedicine than many standard therapies given the complexity of light delivery and light-tissue interactions as well as the resulting photochemistry responsible for tumor damage and drug-release, in addition to the usual intricacies of therapeutic agent delivery. An emerging multidisciplinary approach in oncology utilizes mathematical and computational models to iteratively and quantitively analyze complex dosimetry, and biological response parameters. These models are parameterized by preclinical and clinical observations and then tested against previously unseen data. Such calibrated and validated models can be deployed to simulate treatment doses, protocols, and combinations that have not yet been experimentally or clinically evaluated and can provide testable optimal treatment outcomes in a practical workflow. Here, we foresee the utility of these computational approaches to guide adaptive therapy, and how mathematical models might be further developed and integrated as a novel methodology to guide precision photomedicine.
Collapse
Affiliation(s)
- Bryan Q. Spring
- Translational Biophotonics Cluster, Northeastern University, Boston, MA, United States
- Department of Physics, Northeastern University, Boston, MA, United States
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Ryan T. Lang
- Translational Biophotonics Cluster, Northeastern University, Boston, MA, United States
- Department of Physics, Northeastern University, Boston, MA, United States
| | - Eric M. Kercher
- Translational Biophotonics Cluster, Northeastern University, Boston, MA, United States
- Department of Physics, Northeastern University, Boston, MA, United States
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Robert M. Wenham
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - José R. Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Robert A. Gatenby
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Heiko Enderling
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| |
Collapse
|
40
|
Yilmaz MT, Elmali A, Yazici G. Abscopal Effect, From Myth to Reality: From Radiation Oncologists' Perspective. Cureus 2019; 11:e3860. [PMID: 30899611 PMCID: PMC6414182 DOI: 10.7759/cureus.3860] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The abscopal effect is mediated by a systemic anti-tumor immune response and reflects the regression of non-irradiated metastatic lesions at a distance from the primary site of irradiation. This review will focus on understanding the biological rationale behind the abscopal effect of radiotherapy (RT), which has a recently renewed interest as a result of the successes achieved with immunotherapy and RT in combination. Both RT and immunotherapy are standard components of modern treatment regimens. Combination of these two modalities results in an increased response in the irradiated lesions themselves and the metastatic regions distant from the site of irradiation. We will summarize the abscopal effect of radiotherapy, in particular, the synergistic effect of RT and immunotherapy.
Collapse
Affiliation(s)
| | - Aysenur Elmali
- Radiation Oncology, Hacettepe University Medical School, Ankara, TUR
| | - Gozde Yazici
- Radiation Oncology, Hacettepe University Medical School, Ankara, TUR
| |
Collapse
|