201
|
Kumari S, Mukherjee S, Sinha D, Abdisalaam S, Krishnan S, Asaithamby A. Immunomodulatory Effects of Radiotherapy. Int J Mol Sci 2020; 21:E8151. [PMID: 33142765 PMCID: PMC7663574 DOI: 10.3390/ijms21218151] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Radiation therapy (RT), an integral component of curative treatment for many malignancies, can be administered via an increasing array of techniques. In this review, we summarize the properties and application of different types of RT, specifically, conventional therapy with x-rays, stereotactic body RT, and proton and carbon particle therapies. We highlight how low-linear energy transfer (LET) radiation induces simple DNA lesions that are efficiently repaired by cells, whereas high-LET radiation causes complex DNA lesions that are difficult to repair and that ultimately enhance cancer cell killing. Additionally, we discuss the immunogenicity of radiation-induced tumor death, elucidate the molecular mechanisms by which radiation mounts innate and adaptive immune responses and explore strategies by which we can increase the efficacy of these mechanisms. Understanding the mechanisms by which RT modulates immune signaling and the key players involved in modulating the RT-mediated immune response will help to improve therapeutic efficacy and to identify novel immunomodulatory drugs that will benefit cancer patients undergoing targeted RT.
Collapse
Affiliation(s)
- Sharda Kumari
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Shibani Mukherjee
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Debapriya Sinha
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Salim Abdisalaam
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, FL 32224, USA;
| | - Aroumougame Asaithamby
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| |
Collapse
|
202
|
Ji D, Song C, Li Y, Xia J, Wu Y, Jia J, Cui X, Yu S, Gu J. Combination of radiotherapy and suppression of Tregs enhances abscopal antitumor effect and inhibits metastasis in rectal cancer. J Immunother Cancer 2020; 8:jitc-2020-000826. [PMID: 33106387 PMCID: PMC7592256 DOI: 10.1136/jitc-2020-000826] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2020] [Indexed: 12/18/2022] Open
Abstract
Background Distant metastasis is the major cause of mortality in patients with locally advanced rectal cancer (LARC) following neoadjuvant chemoradiotherapy. Local radiotherapy can trigger an abscopal response to metastatic tumor cells. However, the abscopal effect is a rare event. CD4+ regulatory T (Treg) cell is a highly immune-suppressive subset which impedes immune surveillance against cancer, prevents the development of effective antitumor immunity and promotes tumor progression. We assume that the exploitation of the proimmunogenic effects of radiotherapy with anti-CD25 or anti-Cytotoxic T-Lymphocyte Associated Protein 4 (anti-CTLA4) monoclonal antibodies (mAbs) may enhance the local and abscopal effects in rectal cancer and improve the therapeutic outcome. Methods mRNA expression profiling of 81 pretreatment biopsy samples from LARC patients who received neoadjuvant radiotherapy (nRT) was performed to analyze the correlation between gene expression and prognosis. A retrospective analysis of patients with rectal cancer with distant metastasis or synchronous extracolonic cancers was performed to evaluate the abscopal effect of radiotherapy on rectal cancer. Two different dual-tumor mouse models were established to investigate the efficacy of single dose and dose-fractionated radiotherapy combined with anti-CD25 or anti-CTLA4 and anti-Programmed cell death 1 ligand 1 (anti-PD1) mAbs on the local tumor growth and liver metastasis. The univariate Cox regression analysis, one-way analysis of variance, Dunnett’s test, a mixed-effect linear model and Kaplan-Meier survival analysis were used to calculate p values. Results The proportion of Tregs in pre-nRT biopsies was negatively correlated with prognosis (p=0.007). The retrospective analysis showed that regressing liver metastases were infiltrated by CD8+ T cells. In contrast, stable/progressing metastases and synchronous extracolonic cancers were characterized by PD1+ T cells and Tregs infiltration. Animal experiment results demonstrated that the combination of radiotherapy and anti-CD25/CTLA4 mAb resulted in a significant increase in CD8+ T cells and CD8+/CD4+ ratio in primary and secondary tumors compared with the irradiation alone group (all p<0.05 or p<0.01). The combined treatment was able to decrease Tregs, PD1+CD8+ and PD1+CD4+ T cells (p<0.05), suppress locally irradiated and distal unirradiated tumor growth, and improve overall survival rate. Radiotherapy in conjunction with anti-CTLA4 reduced liver metastasis (p<0.05). Conclusions These data indicated that radiotherapy plus depletion of Tregs was able to improve the antitumor response and generate an abscopal effect.
Collapse
Affiliation(s)
- Dengbo Ji
- Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
| | - Can Song
- School of Life Sciences, Tsinghua University, Beijing, China.,Peking-Tsinghua Center for Life Science, Peking University, Bejing, China
| | - Yongheng Li
- Department of Radiation Oncology, Key laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jinhong Xia
- Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanjing Wu
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinying Jia
- Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xinxin Cui
- Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
| | - Songmao Yu
- Department of Radiation Oncology, Key laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jin Gu
- Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China .,Peking-Tsinghua Center for Life Science, Peking University, Bejing, China.,Department of Gastrointestinal Surgery, Peking University S.G. Hospital, Beijing, China
| |
Collapse
|
203
|
Barsoumian HB, Ramapriyan R, Younes AI, Caetano MS, Menon H, Comeaux NI, Cushman TR, Schoenhals JE, Cadena AP, Reilly TP, Chen D, Masrorpour F, Li A, Hong DS, Diab A, Nguyen QN, Glitza I, Ferrarotto R, Chun SG, Cortez MA, Welsh J. Low-dose radiation treatment enhances systemic antitumor immune responses by overcoming the inhibitory stroma. J Immunother Cancer 2020; 8:jitc-2020-000537. [PMID: 33106386 PMCID: PMC7592253 DOI: 10.1136/jitc-2020-000537] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2020] [Indexed: 12/19/2022] Open
Abstract
Background Despite some successes with checkpoint inhibitors for treating cancer, most patients remain refractory to treatment, possibly due to the inhibitory nature of the tumor stroma that impedes the function and entry of effector cells. We devised a new technique of combining immunotherapy with radiotherapy (XRT), more specifically low-dose XRT, to overcome the stroma and maximize systemic outcomes. Methods We bilaterally established 344SQ lung adenocarcinoma tumors in 129Sv/Ev mice. Primary and secondary tumors were irradiated with either high-dose or low-dose of XRT with systemic anti-programmed cell death protein 1 and anti-cytotoxic T-lymphocyte associated protein 4 administration. Survival and tumor growth were monitored for the various groups, and secondary tumors were phenotyped by flow cytometry for immune populations. Tumor growth factor-beta (TGF-β) cytokine levels were assessed locally after low-dose XRT, and specific immune-cell depletion experiments were conducted to identify the major contributors to the observed systemic antitumor effect. Results Through our preclinical and clinical studies, we observed that when tumor burden was high, there was a necessity of combining high-dose XRT to ‘prime’ T cells at the primary tumor site, with low-dose XRT directed to secondary (metastatic) tumors to ‘modulate the stroma’. Low-dose XRT improved the antitumor outcomes of checkpoint inhibitors by favoring M1 macrophage polarization, enhancing natural killer (NK) cell infiltration, and reducing TGF-β levels. Depletion of CD4+ T cells and NK cells abrogated the observed antitumor effect. Conclusion Our data extend the benefits of low-dose XRT to reprogram the tumor environment and improve the infiltration and function of effector immune cells into secondary tumors.
Collapse
Affiliation(s)
| | - Rishab Ramapriyan
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ahmed I Younes
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mauricio S Caetano
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hari Menon
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nathan I Comeaux
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Taylor R Cushman
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jonathan E Schoenhals
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alexandra P Cadena
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Dawei Chen
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fatemeh Masrorpour
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ailin Li
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David S Hong
- Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Adi Diab
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Quynh-Nhu Nguyen
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Isabella Glitza
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Renata Ferrarotto
- Thoracic Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephen G Chun
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Maria Angelica Cortez
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - James Welsh
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
204
|
Tinganelli W, Durante M. Carbon Ion Radiobiology. Cancers (Basel) 2020; 12:E3022. [PMID: 33080914 PMCID: PMC7603235 DOI: 10.3390/cancers12103022] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
Radiotherapy using accelerated charged particles is rapidly growing worldwide. About 85% of the cancer patients receiving particle therapy are irradiated with protons, which have physical advantages compared to X-rays but a similar biological response. In addition to the ballistic advantages, heavy ions present specific radiobiological features that can make them attractive for treating radioresistant, hypoxic tumors. An ideal heavy ion should have lower toxicity in the entrance channel (normal tissue) and be exquisitely effective in the target region (tumor). Carbon ions have been chosen because they represent the best combination in this direction. Normal tissue toxicities and second cancer risk are similar to those observed in conventional radiotherapy. In the target region, they have increased relative biological effectiveness and a reduced oxygen enhancement ratio compared to X-rays. Some radiobiological properties of densely ionizing carbon ions are so distinct from X-rays and protons that they can be considered as a different "drug" in oncology, and may elicit favorable responses such as an increased immune response and reduced angiogenesis and metastatic potential. The radiobiological properties of carbon ions should guide patient selection and treatment protocols to achieve optimal clinical results.
Collapse
Affiliation(s)
- Walter Tinganelli
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforchung, Planckstraße 1, 64291 Darmstadt, Germany;
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforchung, Planckstraße 1, 64291 Darmstadt, Germany;
- Institut für Festkörperphysik, Technische Universität Darmstadt, Hochschulstraße 8, 64289 Darmstadt, Germany
| |
Collapse
|
205
|
Zhao X, Kong Y, Zhang L. Anti-PD-1 Immunotherapy Combined With Stereotactic Body Radiation Therapy and GM-CSF as Salvage Therapy in a PD-L1-Negative Patient With Refractory Metastatic Esophageal Squamous Cell Carcinoma: A Case Report and Literature Review. Front Oncol 2020; 10:1625. [PMID: 33014817 PMCID: PMC7493754 DOI: 10.3389/fonc.2020.01625] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/27/2020] [Indexed: 12/17/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a malignancy with poor prognosis, which is often diagnosed at a late stage. Effective treatment options are limited when patients fail standard systemic therapy. The application of PD-1 inhibitors have led to a paradigm shift in the treatment of ESCC, but its efficacy as monotherapy is limited. Previous studies have shown that the antitumor effects may be reinforced when a PD-1 inhibitor is combined with radiotherapy or GM-CSF. This study aimed to report a case of a patient about advanced unresectable ESCC negative expression of PD-L1, who experienced tumor progression after chemoradiotherapy and targeted therapy.A significant systemic effect was seen after PD-1 inhibitor combined with GM-CSF and stereotactic body radiotherapy (SBRT) for metastatic lesions, however, severe pneumonia occurred after the triple-combination therapy. This study also reviewed several reports about the efficacy and safety of combination therapy.
Collapse
Affiliation(s)
- Xiangrong Zhao
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| | - Yuehong Kong
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| | - Liyuan Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| |
Collapse
|
206
|
Bip inhibition in glioma stem cells promotes radiation-induced immunogenic cell death. Cell Death Dis 2020; 11:786. [PMID: 32963254 PMCID: PMC7508950 DOI: 10.1038/s41419-020-03000-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 08/07/2020] [Indexed: 12/31/2022]
Abstract
Tumor regression in sites distant to the irradiated field are thought to be associated with emission of damage-associated molecular patterns (DAMPs) molecules and generation of immunogenic cell death (ICD). Glioma stem cells (GSCs) are resistant to high doses of radiation, and ultimately select the outgrowth of a more aggressive tumor. This study showed high-dose IR triggered fewer DAMPs molecules exposure and release in GSCs comparing to matched non-GSCs. Downregulation of binding immunoglobulin protein (Bip) promoted IR-mediated endoplasmic reticulum stress to generate DAMPs molecules by PERK and IRE1-α phosphorylation, and increased dendritic cells mature and effector T lymphocytes activation. GSCs treated with Bip knockdown and IR efficiently prevented tumor generation, and reduced post-radiotherapy tumor recurrence. These data suggest that Bip plays a critical role in inhibition of IR-induced ICD in GSCs, and Bip inhibition may be a promising strategy on adjuvant therapy by ameliorating tumor immune microenvironment.
Collapse
|
207
|
How to improve clinical research in a department of radiation oncology. Bull Cancer 2020; 107:991-998. [PMID: 32950241 DOI: 10.1016/j.bulcan.2020.06.007] [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: 04/14/2020] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Radiation therapy is a core modality for cancer treatment. Around 40% of cancer cures include the use of radiotherapy, either as a single strategy or combined with other treatments. In the past decade, substantial technical advances and novel insights into radiobiological properties have considerably improved patients' outcomes. This study overviewed the landscape of clinical research at our radiotherapy department. METHODS We surveyed our institutional database of clinical trials to collect information for completed or ongoing radiation therapy clinical trials, from 2005 to December 2017 at the Lucien Neuwirth cancer institute. RESULTS A total of 31 clinical trials were undertaken during the study period, of which 4 studies (12.9%) were industry-sponsored and 3 studies (9.7%) were launched by our radiotherapy unit. The vast majority of clinical trials (83.9%) were dedicated to unique organ localization, especially urological cancer (prostate or bladder) (42%). We also observed a shift towards more phase II trials during the study period as well as a special focus on elderly population. Over the last decade, the number of included patients increased by a 5.3 fold input, with 135 inclusions before 2011 and 720 inclusions after 2011. DISCUSSION This study provided an observational and comprehensive analysis of radiotherapy research. From a monocentric point-of-view, these results reflected the on-going progress of worldwide radiotherapy research. Based on a 13-years' experience, this study aimed at highlighting essential cues to ensure efficient and perennial research.
Collapse
|
208
|
Nguyen TT, Nguyen TTD, Ta QTH, Vo VG. Advances in non and minimal-invasive transcutaneous delivery of immunotherapy for cancer treatment. Biomed Pharmacother 2020; 131:110753. [PMID: 33152919 DOI: 10.1016/j.biopha.2020.110753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/20/2022] Open
Abstract
Cancer research has focused on figuring out what was the difference between cancer cells and the tissues within which cancer arose and developing targeted treatments for those differences. With FDA-approved treatments for more ten different cancers and more than thousand new clinical trials, immunotherapy has recently emerged as the most promising area of cancer research by improving efficacy and controlling the adverse effects. Transcutaneous delivery drug delivery offers a number of advantages for the patient because of not only its noninvasive and convenient nature but also factors such as avoidance of first-pass metabolism and prevention of gastrointestinal degradation. The purpose of this review was to highlight technological recent approaches to non and minimal-invasive delivery of immunotherapy for cancer treatment. Finally, some practical considerations and discussions for future studies in the field of transdermal immunomodulation are also included.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Faculty of Pharmacy, Ho Chi Minh City University of Technology (HUTECH), Ho Chi Minh City 700000, Viet Nam
| | - Thi Thuy Dung Nguyen
- Faculty of Environmental and Food Engineering, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Viet Nam
| | - Qui Thanh Hoai Ta
- Institute of Research and Development, Duy Tan University, Danang 550000, Viet Nam
| | - Van Giau Vo
- Bionanotechnology Research Group, Ton Duc Thang University, Ho Chi Minh City 700000, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Viet Nam.
| |
Collapse
|
209
|
Champiat S, Tselikas L, Farhane S, Raoult T, Texier M, Lanoy E, Massard C, Robert C, Ammari S, De Baère T, Marabelle A. Intratumoral Immunotherapy: From Trial Design to Clinical Practice. Clin Cancer Res 2020; 27:665-679. [PMID: 32943460 DOI: 10.1158/1078-0432.ccr-20-0473] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/10/2020] [Accepted: 09/14/2020] [Indexed: 11/16/2022]
Abstract
Systemic immunotherapies such as immune checkpoint blockade targeted at PD(L)1 and CTLA4 have demonstrated their ability to provide durable tumor responses and long-term overall survival benefits for some patients in several solid tumor types. However, a majority of patients remain resistant to these treatments and a significant proportion of them develop severe autoimmune and inflammatory adverse events. Preclinical studies have demonstrated that intratumoral injections of immunostimulatory products (oncolytics, pattern recognition receptor agonists,…) that are able to trigger type I IFN release and enhance tumor antigen presentation on immune cells could generate a strong antitumor immunity and overcome the resistance to systemic immune checkpoint blockade therapies. The intratumoral immunotherapy strategies that are currently in clinical development offer a unique therapeutic and exploratory setting to better understand the immune contexture across tumor lesions of patients with metastatic cancer. Also these local therapeutic products could turn cold tumors into hot and improve the response rates to cancer immunotherapies while diminishing their systemic exposure and toxicities. Intratumoral immunotherapies could prime or boost the immunity against tumors and therefore radically change the combinatorial therapeutic strategies currently pursued for metastatic and local cancers to improve their long-term survival. We aimed to review and discuss the scientific rationale for intratumoral immunotherapy, the challenges raised by this strategy in terms of drug development within clinical trials and the current state-of-the-art regarding the clinical practice of this innovative approach.
Collapse
Affiliation(s)
- Stéphane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France.,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France
| | - Lambros Tselikas
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France.,Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Siham Farhane
- Gustave Roussy Immunotherapy Program (GRIP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thibault Raoult
- Service de Promotion des Etudes Cliniques (SPEC), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Matthieu Texier
- Service de Biostatistiques et d'Epidémiologie (SBE), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Emilie Lanoy
- Service de Biostatistiques et d'Epidémiologie (SBE), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Christophe Massard
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Caroline Robert
- Département de Médecine Oncologique (DMO), Gustave Roussy, Université Paris Saclay, Villejuif, France.,Université Paris Saclay, Saint-Aubin, France
| | - Samy Ammari
- Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thierry De Baère
- Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France.,Université Paris Saclay, Saint-Aubin, France
| | - Aurélien Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France. .,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France.,Gustave Roussy Immunotherapy Program (GRIP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| |
Collapse
|
210
|
Candas-Green D, Xie B, Huang J, Fan M, Wang A, Menaa C, Zhang Y, Zhang L, Jing D, Azghadi S, Zhou W, Liu L, Jiang N, Li T, Gao T, Sweeney C, Shen R, Lin TY, Pan CX, Ozpiskin OM, Woloschak G, Grdina DJ, Vaughan AT, Wang JM, Xia S, Monjazeb AM, Murphy WJ, Sun LQ, Chen HW, Lam KS, Weichselbaum RR, Li JJ. Dual blockade of CD47 and HER2 eliminates radioresistant breast cancer cells. Nat Commun 2020; 11:4591. [PMID: 32929084 PMCID: PMC7490264 DOI: 10.1038/s41467-020-18245-7] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/11/2020] [Indexed: 12/19/2022] Open
Abstract
Although the efficacy of cancer radiotherapy (RT) can be enhanced by targeted immunotherapy, the immunosuppressive factors induced by radiation on tumor cells remain to be identified. Here, we report that CD47-mediated anti-phagocytosis is concurrently upregulated with HER2 in radioresistant breast cancer (BC) cells and RT-treated mouse syngeneic BC. Co-expression of both receptors is more frequently detected in recurrent BC patients with poor prognosis. CD47 is upregulated preferentially in HER2-expressing cells, and blocking CD47 or HER2 reduces both receptors with diminished clonogenicity and augmented phagocytosis. CRISPR-mediated CD47 and HER2 dual knockouts not only inhibit clonogenicity but also enhance macrophage-mediated attack. Dual antibody of both receptors synergizes with RT in control of syngeneic mouse breast tumor. These results provide the evidence that aggressive behavior of radioresistant BC is caused by CD47-mediated anti-phagocytosis conjugated with HER2-prompted proliferation. Dual blockade of CD47 and HER2 is suggested to eliminate resistant cancer cells in BC radiotherapy.
Collapse
Affiliation(s)
- Demet Candas-Green
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Bowen Xie
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie Huang
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Ming Fan
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Cheikh Menaa
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Yanhong Zhang
- Department of Pathology, Kaiser Permanente Medical Center Vallejo and Vacaville, Vallejo, CA, USA
| | - Lu Zhang
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Di Jing
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Soheila Azghadi
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Weibing Zhou
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Liu
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Nian Jiang
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Tao Li
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Tianyi Gao
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Colleen Sweeney
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
- NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Rulong Shen
- Department of Pathology, Ohio State University, Columbus, OH, USA
| | - Tzu-Yin Lin
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Chong-Xian Pan
- NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
- Department of Internal Medicine, University of California Davis, Sacramento, CA, USA
| | - Omer M Ozpiskin
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Gayle Woloschak
- Department of Radiation Oncology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - David J Grdina
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA
| | - Andrew T Vaughan
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
- NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Ji Ming Wang
- Chemoattractant Receptor and Signal Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Shuli Xia
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Arta M Monjazeb
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
- NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - William J Murphy
- NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
- Department of Dermatology, University of California Davis, Sacramento, CA, USA
| | - Lun-Quan Sun
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
- NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
- NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA.
- NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
211
|
A review of radiation induced abscopal effect: combining radiotherapy and immunotherapy to treat the untreated distant metastatic tumours. JOURNAL OF RADIOTHERAPY IN PRACTICE 2020. [DOI: 10.1017/s1460396920000680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractBackground:Radiotherapy is an effective and significant mode of definitive cancer treatment with well-established local tumour control success, especially in the treatment of localised tumours. Although, for metastatic disease, the role of radiotherapy has generally been limited to palliation of symptoms. In the treatment of metastatic diseases settings, the radiation therapy technique has always been confronted with the challenge of the simultaneous treatment of all of the various distant metastatic tumour sites, however, some recent evidence suggests that radiotherapy can potentially induce anticancer immune responses whose effectors potentially migrate to distant metastatic tumours to provoke their regression in cancer patients. Thus, unirradiated distant metastatic tumour sites can exhibit a delayed therapeutic response termed the abscopal effect.Materials and methods:This paper reports on a review of the abscopal effect, including its biological mechanism, the effect of radiation dose and fractionation regime and the timing of immunotherapy administration on radiotherapy-induced abscopal effect, the enhancement of radiotherapy-induced abscopal effects with immunotherapy, the effect of the location of the irradiated tumour and the radiotherapy of multiple tumour sites on the likelihood and effectiveness of inducing abscopal responses in the preclinical and clinical settings and also reports on some evidence of clinical observations in patients.Conclusions:Although abscopal effects of radiotherapy are still relatively rare in patients, it has gained a lot of interest due to recent development and use of immunotherapy strategies incorporating combinations of targeted immunomodulators and immune checkpoint blockade with radiation therapy. The enhancement of cancer immunotherapy could potentially enable the translation of the concept of abscopal effect into the clinics as a new strategy to induce therapeutically effective anti-tumour immune responses in cancer patients. The combination of radiotherapy and immunotherapy has the potential to expand the role of radiotherapy from a purely local tumour control treatment to play a significant role in advanced and metastatic tumour control and this could likely lead to a paradigm shift in the treatment of patients with metastatic cancer.
Collapse
|
212
|
Connor MJ, Smith A, Miah S, Shah TT, Winkler M, Khoo V, Ahmed HU. Targeting Oligometastasis with Stereotactic Ablative Radiation Therapy or Surgery in Metastatic Hormone-sensitive Prostate Cancer: A Systematic Review of Prospective Clinical Trials. Eur Urol Oncol 2020; 3:582-593. [PMID: 32891600 DOI: 10.1016/j.euo.2020.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/01/2020] [Accepted: 07/09/2020] [Indexed: 12/31/2022]
Abstract
CONTEXT Metastasis-directed therapy (MDT) in the form of stereotactic ablative radiation therapy (SABR), or in combination with surgical metastasectomy, may have a role in cancer control and disease progression. OBJECTIVE To perform a systematic review of MDT (surgery or SABR) for oligometastatic (up to 10 metastases, recurrent or de novo) hormone-sensitive prostate cancer in addition to or following primary prostate gland treatment. EVIDENCE ACQUISITION Medline, Embase, Cochrane Review Database, and clinical trial Databases were systematically searched for clinical trials reporting oncological outcomes and safety. The risk of bias was assessed with the Cochrane 2.0 or ROBINS-I tool. EVIDENCE SYNTHESIS From 1025 articles identified, four clinical trials met the prespecified criteria. These included two randomised and two nonrandomised clinical trials (n=169). Baseline prostate-specific antigen level, age, and metastasis ranged from 2.0 to 17.0 ng/ml, 43 to 75 yr, and one to seven lesions, respectively. Nodal, bone, nodal and bone, and visceral metastases were present in 49.7% (84/169), 33.7% (57/169), 15.9% (27/169), and 0.5% (1/169) of patients, respectively. Diagnostic conventional imaging was used in 43.7% (74/169) and positron emission tomography/computerised tomography in 56.2% (95/169) of patients. SABR and surgical metastasectomy with SABR were used in 78.3% (94/120) and 21.6% (26/120) of patients, respectively. Early progression-free survival ranged from 19% to 60%. Local control was reported as 93-100%. Grade II and III SABR toxicities were reported in 8% (8/100) and 1% (1/100) of patients, respectively. Grade IIIa and IIIb surgical complications were reported in 7.69% (2/26) and 0% (0/26) of patients, respectively. CONCLUSIONS MDT is a promising experimental therapeutic approach in men with hormone-sensitive oligometastatic prostate cancer. Randomised comparative studies are required to ascertain its role and optimal timing in oligometastatic recurrence and efficacy in de novo synchronous disease. PATIENT SUMMARY We looked at the evidence regarding the use of surgery or radiotherapy at target areas of cancer spread in men with newly diagnosed or relapsed advanced (metastatic) prostate cancer. Evidence supports both treatment options as promising approaches, but further large trials are required.
Collapse
Affiliation(s)
- Martin J Connor
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College, London, UK; Imperial Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK.
| | - Ailbe Smith
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College, London, UK
| | - Saiful Miah
- Department of Urology, Addenbrooke's Hospital, Cambridge, UK
| | - Taimur T Shah
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College, London, UK; Imperial Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Mathias Winkler
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College, London, UK; Imperial Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Vincent Khoo
- Department of Clinical Oncology, The Royal Marsden Hospital & Institute of Cancer Research, London, UK
| | - Hashim U Ahmed
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College, London, UK; Imperial Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
213
|
Gómez V, Mustapha R, Ng K, Ng T. Radiation therapy and the innate immune response: Clinical implications for immunotherapy approaches. Br J Clin Pharmacol 2020; 86:1726-1735. [PMID: 32388875 PMCID: PMC7444780 DOI: 10.1111/bcp.14351] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy is an essential component of cancer care, contributing up to 40% of curative cancer treatment regimens. It creates DNA double-strand breaks causing cell death in highly replicating tumour cells. However, tumours can develop acquired resistance to therapy. The efficiency of radiation treatment has been increased by means of combining it with other approaches such as chemotherapy, molecule-targeted therapies and, in recent years, immunotherapy (IT). Cancer-cell apoptosis after radiation treatment causes an immunological reaction that contributes to eradicating the tumour via antigen presentation and subsequent T-cell activation. By contrast, radiotherapy also contributes to the formation of an immunosuppressive environment that hinders the efficacy of the therapy. Innate immune cells from myeloid and lymphoid origin show a very active role in both acquired resistance and antitumourigenic mechanisms. Therefore, many efforts are being made in order to reach a better understanding of the innate immunity reactions after radiation therapy (RT) and the design of new combinatorial IT strategies focused in these particular populations.
Collapse
Affiliation(s)
- Valentí Gómez
- UCL Cancer InstituteUniversity College LondonLondonUK
- Cancer Research UK City of London CentreUK
| | - Rami Mustapha
- School of Cancer and Pharmaceutical SciencesKing's College LondonLondonUK
- Cancer Research UK King's Health Partners CentreUK
| | - Kenrick Ng
- UCL Cancer InstituteUniversity College LondonLondonUK
- Department of Medical OncologyUniversity College Hospitals NHS Foundation TrustUK
| | - Tony Ng
- UCL Cancer InstituteUniversity College LondonLondonUK
- Cancer Research UK City of London CentreUK
- School of Cancer and Pharmaceutical SciencesKing's College LondonLondonUK
- Cancer Research UK King's Health Partners CentreUK
| |
Collapse
|
214
|
Ochoa de Olza M, Bourhis J, Irving M, Coukos G, Herrera FG. High versus low dose irradiation for tumor immune reprogramming. Curr Opin Biotechnol 2020; 65:268-283. [PMID: 32882511 DOI: 10.1016/j.copbio.2020.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022]
Abstract
Local administration of ionizing radiation to tumors can promote anticancer immune responses that lead to the abscopal regression of distant metastases, especially in patients receiving systemic immune-checkpoint inhibitors. Growing preclinical evidence indicates that high-dose irradiation administered locally to destroy malignant lesions, can promote the release of danger-associated molecular patterns that lead to the recruitment of immune cells, thus inducing a systemic response against tumor antigens that protects against local disease relapse and also mediates distant antineoplastic effects. An accumulating body of preclinical evidence supports also the implementation of low-dose irradiation to induce tumor immune reprogramming. Here, we provide the rationale for a clinical research agenda to refine future clinical practice based on innovative combinations of radiation-immunotherapy.
Collapse
Affiliation(s)
- Maria Ochoa de Olza
- Department of Oncology, Lausanne University Hospital, and University of Lausanne, Lausanne, Switzerland
| | - Jean Bourhis
- Radiation Oncology Service, Lausanne University Hospital, and University of Lausanne, Lausanne, Switzerland; Department of Oncology, Lausanne University Hospital, and University of Lausanne, Lausanne, Switzerland
| | - Melita Irving
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Department of Oncology, Lausanne University Hospital, and University of Lausanne, Lausanne, Switzerland
| | - Fernanda G Herrera
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Radiation Oncology Service, Lausanne University Hospital, and University of Lausanne, Lausanne, Switzerland; Department of Oncology, Lausanne University Hospital, and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
215
|
Jagodinsky JC, Harari PM, Morris ZS. The Promise of Combining Radiation Therapy With Immunotherapy. Int J Radiat Oncol Biol Phys 2020; 108:6-16. [PMID: 32335187 PMCID: PMC7442714 DOI: 10.1016/j.ijrobp.2020.04.023] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/30/2020] [Accepted: 04/13/2020] [Indexed: 12/17/2022]
Abstract
The development of immunotherapy in oncology builds upon many years of scientific investigation into the cellular mechanics underlying interactions between tumor cells and immune cell populations. The past decade has brought an accelerating pace to the clinical investigation of new immunotherapy agents, particularly in the setting of metastatic disease. The integration of immunotherapy into phase 3 clinical trial design has lagged in settings of advanced locoregional disease, where combination with radiation therapy may be critical. Yet, such may be the settings where immunotherapies have their greatest potential to affect patient survival and achieve curative outcomes. In this review, we discuss the interaction of radiation with the immune system and the potential to augment antitumor immunity through combined-modality approaches that integrate radiation and immunotherapies. The dynamics of cellular and tumor response to radiation offer unique opportunities for beneficial interplay with immunotherapy that may go unrecognized with conventional screening and monotherapy clinical testing of novel pharmaceutical agents. Using immune checkpoint blockade as a primary example, we discuss recent preclinical and clinical studies that illustrate the potential synergy of such therapies in combination with radiation, and we highlight the potential clinical value of such interactions. For various immunotherapy agents, their greatest clinical effect may rest in combination with radiation, and efforts to facilitate systematic investigation of this approach are highly warranted.
Collapse
Affiliation(s)
- Justin C Jagodinsky
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
216
|
Bozorgmehr F, Chung I, Christopoulos P, Krisam J, Schneider MA, Brückner L, Mueller DW, Thomas M, Rieken S. Thoracic radiotherapy plus Durvalumab in elderly and/or frail NSCLC stage III patients unfit for chemotherapy - employing optimized (hypofractionated) radiotherapy to foster durvalumab efficacy: study protocol of the TRADE-hypo trial. BMC Cancer 2020; 20:806. [PMID: 32842974 PMCID: PMC7447611 DOI: 10.1186/s12885-020-07264-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Non-small cell lung cancer is the most common cause of cancer death worldwide, highlighting the need for novel therapeutic concepts. In particular, there is still a lack of treatment strategies for the group of elderly and frail patients, who are frequently not capable of receiving standard therapy regimens. Despite comprising the majority of lung cancer patients, this group is underrepresented in clinical trials. This applies also to elderly and frail patients suffering from unresectable stage III NSCLC, who are unfit for chemotherapy, and, therefore, cannot receive the standard therapy comprising of radiochemotherapy and the recently approved subsequent durvalumab consolidation therapy. These patients often receive radiotherapy only, which raises the concern of undertreatment. The TRADE-hypo trial aims at optimizing treatment of this patient group by combining radiotherapy with concomitant durvalumab administration, thereby employing the immune-promoting effects of radiotherapy, and determining safety, feasibility, and efficacy of this treatment. METHODS/ DESIGN In this prospective phase II clinical trial, durvalumab therapy will be combined with either conventionally fractionated (CON-group) or hypofractionated (HYPO-group) thoracic radiotherapy. A safety stop-and-go lead-in phase will assess safety of hypofractionated radiotherapy with respect to severe pneumonitis in small patient cohorts before opening full enrollment. Tumor tissue, blood and stool samples will be collected before and during the study period to investigate the immunological mechanisms responsible for checkpoint inhibitor efficacy and immune-promoting effects of radiotherapy. DISCUSSION Preclinical data suggests that irradiation-induced immunogenicity can be further increased if applied in a hypofractionated setting, potentially boosting the expected synergistic effect with immune checkpoint inhibition in restoring the immune anti-tumor response. If proven safe and efficient, a hypofractionated radiation schedule can provide a considerably more practicable option for the patient. Taking into consideration the intend to develop a combination treatment strategy that can be made available to patients soon after proving to be efficient and the potentially elevated toxicity of a hypofractionated radiotherapy approach, this trial was designed as a two-trials-in-one design. An accompanying translational research program is planned striving to gain insights into the tumor-host biology and to identify suitable biomarkers to predict therapy response. TRIAL REGISTRATION Clinicaltrials.gov , NCT04351256 . Registered 17 April 2020, Eudra-CT, 2019-002192-33 . Registered 24 October 2019.
Collapse
Affiliation(s)
- Farastuk Bozorgmehr
- Department of Thoracic Oncology, Thoraxklinik at University Hospital of Heidelberg, Röntgenstraße 1, 69126 Heidelberg, Germany
- Translational Lung Research Center Heidelberg TLRCH, Member of the German Center for Lung Research DZL, Im Neuenheimer Feld 156, 69120 Heidelberg, Germany
| | - Inn Chung
- Department of Thoracic Oncology, Thoraxklinik at University Hospital of Heidelberg, Röntgenstraße 1, 69126 Heidelberg, Germany
- Translational Lung Research Center Heidelberg TLRCH, Member of the German Center for Lung Research DZL, Im Neuenheimer Feld 156, 69120 Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at University Hospital of Heidelberg, Röntgenstraße 1, 69126 Heidelberg, Germany
- Translational Lung Research Center Heidelberg TLRCH, Member of the German Center for Lung Research DZL, Im Neuenheimer Feld 156, 69120 Heidelberg, Germany
| | - Johannes Krisam
- Institute of Medical Biometry and Informatics, University Hospital of Heidelberg, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany
| | - Marc A. Schneider
- Translational Lung Research Center Heidelberg TLRCH, Member of the German Center for Lung Research DZL, Im Neuenheimer Feld 156, 69120 Heidelberg, Germany
- Thoraxklinik at University Hospital of Heidelberg, Translational Research Unit (STF), Röntgenstraße 1, 69126 Heidelberg, Germany
| | - Lena Brückner
- Department of Thoracic Oncology, Thoraxklinik at University Hospital of Heidelberg, Röntgenstraße 1, 69126 Heidelberg, Germany
- Translational Lung Research Center Heidelberg TLRCH, Member of the German Center for Lung Research DZL, Im Neuenheimer Feld 156, 69120 Heidelberg, Germany
| | - Daniel Wilhelm Mueller
- Institute of Clinical Cancer Research IKF GmbH at Northwest Hospital, Steinbacher Hohl 2-26, 60488 Frankfurt am Main, Germany
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik at University Hospital of Heidelberg, Röntgenstraße 1, 69126 Heidelberg, Germany
- Translational Lung Research Center Heidelberg TLRCH, Member of the German Center for Lung Research DZL, Im Neuenheimer Feld 156, 69120 Heidelberg, Germany
| | - Stefan Rieken
- Department of Radiation Oncology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| |
Collapse
|
217
|
Powerski M, Drewes R, Omari J, Relja B, Surov A, Pech M. Intra-hepatic Abscopal Effect Following Radioembolization of Hepatic Metastases. Cardiovasc Intervent Radiol 2020; 43:1641-1649. [PMID: 32808201 PMCID: PMC7591411 DOI: 10.1007/s00270-020-02612-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/02/2020] [Indexed: 01/05/2023]
Abstract
Purpose To search for abscopal effects (AE) distant to the site of radiation after sequential Yittrium-90 (Y-90) radioembolization (RE) of liver malignancies. Methods and Materials In this retrospective analysis, all patients treated by RE between 2007 and 2018 (n = 907) were screened for the following setting/conditions: sequential RE of left and right liver lobe in two sessions, liver-specific MRI (MRI1) acquired max. 10 days before or after first RE (RE1), liver-specific MRI (MRI2) acquired with a minimum time interval of 20 days after MRI1, but before second RE (RE2). No systemic tumor therapies between MRI1 and MRI2. No patients with liver cirrhosis. Metastases > 5 mm in untreated liver lobes were compared in MRI1 and MRI2 and rated as follows: same size or larger in MRI2 = no abscopal effect (NAE); > 30% shrinkage without Y-90 contamination in SPECT/CT = abscopal effect (AE). Results Ninety six of 907 patients met aforementioned criteria. Median time-frame between RE1 and MRI2 was 34 (20–64) days. These 96 cases had 765 metastases which were evaluable (median 5(1–40) metastases per patient). Four patients could be identified with at least one shrinking metastasis of the untreated site: one patient with breast cancer (3 metastases: 0 NAE; 3 AE), one patient with prostate cancer (6 metastases: 3 NAE; 3 metastases > 30% shrinkage but possible Y-90 contamination) and two patients with shrinkage of one metastasis each but less than 30%. Conclusion Our retrospective study documents AE after RE of liver tumors in 1 out of 96 cases, 3 other cases remain unclear.
Collapse
Affiliation(s)
- Maciej Powerski
- Department of Radiology and Nuclear Medicine, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Ralph Drewes
- Department of Radiology and Nuclear Medicine, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Jazan Omari
- Department of Radiology and Nuclear Medicine, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Borna Relja
- Department of Radiology and Nuclear Medicine, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Alexey Surov
- Department of Radiology and Nuclear Medicine, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Maciej Pech
- Department of Radiology and Nuclear Medicine, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany.,2nd Department of Radiology, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214, Gdansk, Poland
| |
Collapse
|
218
|
Kalanxhi E, Meltzer S, Ree AH. Immune-Modulating Effects of Conventional Therapies in Colorectal Cancer. Cancers (Basel) 2020; 12:E2193. [PMID: 32781554 PMCID: PMC7464272 DOI: 10.3390/cancers12082193] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/22/2022] Open
Abstract
Biological heterogeneity and low inherent immunogenicity are two features that greatly impact therapeutic management and outcome in colorectal cancer. Despite high local control rates, systemic tumor dissemination remains the main cause of treatment failure and stresses the need for new developments in combined-modality approaches. While the role of adaptive immune responses in a small subgroup of colorectal tumors with inherent immunogenicity is indisputable, the challenge remains in identifying the optimal synergy between conventional treatment modalities and immune therapy for the majority of the less immunogenic cases. In this context, cytotoxic agents such as radiation and certain chemotherapeutics can be utilized to enhance the immunogenicity of an otherwise immunologically silent disease and enable responsiveness to immune therapy. In this review, we explore the immunological characteristics of colorectal cancer, the effects that standard-of-care treatments have on the immune system, and the opportunities arising from combining immune checkpoint-blocking therapy with immune-modulating conventional treatments.
Collapse
Affiliation(s)
- Erta Kalanxhi
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway; (E.K.); (S.M.)
| | - Sebastian Meltzer
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway; (E.K.); (S.M.)
| | - Anne Hansen Ree
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway; (E.K.); (S.M.)
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| |
Collapse
|
219
|
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
|
220
|
Outcomes of Hypofractional Tomotherapy in Patients with Stage III Nonsmall Cell Lung Cancer Who Are Not Eligible for Surgery or Concurrent Chemoradiation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9168424. [PMID: 32685544 PMCID: PMC7345954 DOI: 10.1155/2020/9168424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/21/2020] [Indexed: 12/04/2022]
Abstract
Purpose We assessed the clinical outcomes and toxicities following hypofractionation with helical tomographic intensity-modulated radiotherapy technology (tomotherapy) in patients with stage III non-small cell lung cancer (NSCLC) who were not candidates for surgery or concurrent chemoradiation. Methods Forty-three patients with stage III NSCLC who were treated between 2011 and 2017 were enrolled. The prescription doses for gross target volume and clinical target volume were 70 Gy and 60 Gy (respectively) delivered in 15–25 fractions over 3–5 weeks. Results The median overall survival (OS) time was 34.23 (range 11.33–99.33) months. The estimated 1-, 2-, and 3-year OS rates were 97.7%, 74.4%, and 55.9%, respectively; the corresponding progression-free survival (PFS) rates were 79.1%, 53.5%, and 36.1%, respectively. The local disease recurrence, regional disease recurrence, and distant metastasis rates at 3 years were 4.7%, 11.62%, and 55.81%, respectively. On multivariate analysis, dose regimen (<19 f vs. ≥19 f) was an independent prognostic factor affecting OS, PFS, and DM (p < 0.05). Seven patients developed grade 1-2 acute radiation pneumonia (RP), 5 patients developed grade 1-2 late RP, while 3 patients developed grade 3 late RP. None of the patients developed grade 4-5 radiation lung injury. Conclusion Tomotherapy may be an effective treatment option for patients with stage III NSCLC. It may be a viable alternative to surgery with lower incidence of side effects.
Collapse
|
221
|
Zucker A, Winter A, Lumley D, Karwowski P, Jung MK, Kao J. Prognostic role of baseline neutrophil-to-lymphocyte ratio in metastatic solid tumors. Mol Clin Oncol 2020; 13:25. [PMID: 32774855 DOI: 10.3892/mco.2020.2095] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/15/2020] [Indexed: 01/04/2023] Open
Abstract
High baseline neutrophil-to-lymphocyte ratio (NLR) has been associated with poor survival in a number of solid tumors, but has not been extensively investigated in the context of radiation oncology. Developing more robust models to predict survival would inform patient care for patients with metastatic solid tumors. The present study was undertaken to evaluate the effect of baseline NLR (using 4 as a cutoff) on survival in 320 consecutive patients with metastatic cancer who were referred to a single radiation oncologist between 2012 and 2015, with a median follow-up of 20.6 months. The median NLR was 4.4 (interquartile range, 2.8-7.2). Patients with a baseline NLR ≤4 had a median survival of 9.3 months compared to 4.1 months for NLR >4 (P<0.001). The number of active tumors, Eastern Cooperative Oncology Group performance status score, baseline albumin, primary tumor site, liver metastases and baseline NLR predicted overall survival on both univariate and multivariate analysis (P<0.05 for all). After adjusting for known prognostic factors for advanced solid tumors, baseline NLR >4 independently predicted adverse survival in this cohort.
Collapse
Affiliation(s)
- Amanda Zucker
- College of Osteopathic Medicine, New York Institute of Technology, Glen Head, NY 11545, USA
| | - Alex Winter
- College of Osteopathic Medicine, New York Institute of Technology, Glen Head, NY 11545, USA
| | - Dean Lumley
- College of Osteopathic Medicine, New York Institute of Technology, Glen Head, NY 11545, USA
| | - Pawel Karwowski
- College of Osteopathic Medicine, New York Institute of Technology, Glen Head, NY 11545, USA
| | - Min-Kyung Jung
- College of Osteopathic Medicine, New York Institute of Technology, Glen Head, NY 11545, USA
| | - Johnny Kao
- Department of Radiation Oncology, Good Samaritan Hospital Medical Center, West Islip, NY 11795, USA
| |
Collapse
|
222
|
Huang J, Li JJ. Multiple Dynamics in Tumor Microenvironment Under Radiotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1263:175-202. [PMID: 32588328 DOI: 10.1007/978-3-030-44518-8_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The tumor microenvironment (TME) is an evolutionally low-level and embryonically featured tissue comprising heterogenic populations of malignant and stromal cells as well as noncellular components. Under radiotherapy (RT), the major modality for the treatment of malignant diseases [1], TME shows an adaptive response in multiple aspects that affect the efficacy of RT. With the potential clinical benefits, interests in RT combined with immunotherapy (IT) are intensified with a large scale of clinical trials underway for an array of cancer types. A better understanding of the multiple molecular aspects, especially the cross talks of RT-mediated energy reprogramming and immunoregulation in the irradiated TME (ITME), will be necessary for further enhancing the benefit of RT-IT modality. Coming studies should further reveal more mechanistic insights of radiation-induced instant or permanent consequence in tumor and stromal cells. Results from these studies will help to identify critical molecular pathways including cancer stem cell repopulation, metabolic rewiring, and specific communication between radioresistant cancer cells and the infiltrated immune active lymphocytes. In this chapter, we will focus on the following aspects: radiation-repopulated cancer stem cells (CSCs), hypoxia and re-oxygenation, reprogramming metabolism, and radiation-induced immune regulation, in which we summarize the current literature to illustrate an integrated image of the ITME. We hope that the contents in this chapter will be informative for physicians and translational researchers in cancer radiotherapy or immunotherapy.
Collapse
Affiliation(s)
- Jie Huang
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA. .,NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
223
|
Loubersac T, Guimas V, Rio E, Libois V, Rigaud J, Supiot S. [Oligorecurrent prostate cancer: current management and perspectives]. Bull Cancer 2020; 107:S35-S40. [PMID: 32620205 DOI: 10.1016/s0007-4551(20)30276-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Oligometastatic prostate cancer (PCa) is an intense area of research thanks to the development of novel PET tracers such as 18F-choline or 68Ga-PSMA. Several retrospective studies in patients with hormone-sensitive oligorecurrent PCa (usually up to 5 metastases with a controlled primary tumor) showed PSA response and a low toxicity profile of metastasis-directed therapies (MDT) such as Stereotactic Body Radiation Therapy (SBRT) or salvage lymph node dissection. More recently, randomized phase 2 studies showed that SBRT can delay the introduction of androgen deprivation, decrease biochemical relapses and increase overall survival. Regarding oligoprogressive metastatic castration-resistant PCa, limited data is however available. Based on these studies the European Association of Urology and the American Society of Radiotherapy EAU now recommend using MDT instead of observation. Several studies are undergoing in France and worldwide in order to confirm the exact role of MDT.
Collapse
Affiliation(s)
- Thomas Loubersac
- Urologie, Hôtel-Dieu, CHU de Nantes, 1, place Alexis-Ricordeau, 44093 Nantes, France
| | - Valentine Guimas
- Service de radiothérapie, Institut de cancérologie de l'Ouest, boulevard Professeur-Jacques-Monod, 44800 Nantes Saint-Herblain, France
| | - Emmanuel Rio
- Service de radiothérapie, Institut de cancérologie de l'Ouest, boulevard Professeur-Jacques-Monod, 44800 Nantes Saint-Herblain, France
| | - Vincent Libois
- Service de radiothérapie, Institut de cancérologie de l'Ouest, boulevard Professeur-Jacques-Monod, 44800 Nantes Saint-Herblain, France
| | - Jérome Rigaud
- Urologie, Hôtel-Dieu, CHU de Nantes, 1, place Alexis-Ricordeau, 44093 Nantes, France
| | - Stéphane Supiot
- Service de radiothérapie, Institut de cancérologie de l'Ouest, boulevard Professeur-Jacques-Monod, 44800 Nantes Saint-Herblain, France.
| |
Collapse
|
224
|
Wang H, Li X, Peng R, Wang Y, Wang J. Stereotactic ablative radiotherapy for colorectal cancer liver metastasis. Semin Cancer Biol 2020; 71:21-32. [PMID: 32629077 DOI: 10.1016/j.semcancer.2020.06.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/24/2022]
Abstract
Survival improvement of colorectal liver metastasis (CRLM) benefits from systemic therapy and metastasis-directed local therapy. Stereotactic ablative body radiotherapy (SABR), as a new efficient metastasis-directed local therapy with a systematic impact, plays a vital role in CRLM multidisciplinary treatment. SABR leads to a dramatic immunological change in the tumor microenvironment (TME) via differential activation of cytoprotective and cytotoxic pathways in malignant and non-malignant cells, in addition to direct tumor cell death. The synergy of SABR and immunotherapy might increase the abscopal response rate of out-field lesions by targeting different steps of the immune-mediated response, in addition to direct intratumoral cell death. The clinical treatment and efficacy of SABR, its influence on TME, and potential molecular underpinnings of which are the topic of this review.
Collapse
Affiliation(s)
- Hao Wang
- Department of Radiation Oncology, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China
| | - Xuemin Li
- Department of Radiation Oncology, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China
| | - Ran Peng
- Department of Radiation Oncology, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China
| | - Yuxia Wang
- Department of Radiation Oncology, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China.
| |
Collapse
|
225
|
Hong WX, Haebe S, Lee AS, Westphalen CB, Norton JA, Jiang W, Levy R. Intratumoral Immunotherapy for Early-stage Solid Tumors. Clin Cancer Res 2020; 26:3091-3099. [PMID: 32071116 PMCID: PMC7439755 DOI: 10.1158/1078-0432.ccr-19-3642] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/07/2020] [Accepted: 02/14/2020] [Indexed: 12/31/2022]
Abstract
The unprecedented benefits of immunotherapy in advanced malignancies have resulted in increased interests in exploiting immune stimulatory agents in earlier-stage solid tumors in the neoadjuvant setting. However, systemic delivery of immunotherapies may cause severe immune-related side-effects and hamper the development of combination treatments. Intratumoral delivery of neoadjuvant immunotherapy provides a promising strategy in harnessing the power of immunotherapy while minimizing off-target toxicities. The direct injection of immune stimulating agents into the tumor primes the local tumor-specific immunity to generate a systemic, durable clinical response. Intratumoral immunotherapy is a highly active area of investigation resulting in a plethora of agents, for example, immune receptor agonists, non-oncolytic and oncolytic viral therapies, being tested in preclinical and clinical settings. Currently, more than 20 neoadjuvant clinical trials exploring distinct intratumoral immune stimulatory agents and their combinations are ongoing. Practical considerations, including appropriate timing and optimal local delivery of immune stimulatory agents play an important role in safety and efficacy of this approach. Here, we discuss promising approaches in drug delivery technologies and opportunity for combining intratumoral immunotherapy with other cancer treatments and summarize the recent preclinical and clinical evidences that highlighted its promise as a part of routine oncologic care.
Collapse
Affiliation(s)
- Wan Xing Hong
- Department of Surgery, Stanford University School of Medicine, Stanford, California
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | - Sarah Haebe
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
- Department of Medicine III, University Hospital, LMU, Munich, Germany
| | - Andrew S Lee
- Department of Pathology, Stanford University School of Medicine, Stanford, California
- Shenzhen Bay Laboratory, Cancer Research Institute, Shenzhen, China
| | - C Benedikt Westphalen
- Department of Medicine III, University Hospital, LMU, Munich, Germany
- Comprehensive Cancer Center Munich, Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), DKTK Partner Site, Munich, Germany
| | - Jeffrey A Norton
- Department of Surgery, Stanford University School of Medicine, Stanford, California
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ronald Levy
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California.
| |
Collapse
|
226
|
Wang J, Li Z, Wang Z, Yu Y, Li D, Li B, Ding J. Nanomaterials for Combinational Radio–Immuno Oncotherapy. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910676. [DOI: 10.1002/adfm.201910676] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 03/09/2020] [Indexed: 08/29/2023]
Affiliation(s)
- Juan Wang
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
- Department of Radiation OncologyCancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Zhongmin Li
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
- Department of Gastrointestinal, Colorectal, and Anal SurgeryChina–Japan Union Hospital of Jilin University 126 Xiantai Street Changchun 130012 P. R. China
| | - Zhongtang Wang
- Department of Radiation OncologyCancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Yonghua Yu
- Department of Radiation OncologyCancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Di Li
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
| | - Baosheng Li
- Department of Radiation OncologyCancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
| |
Collapse
|
227
|
Li JJ. Mitigating Coronavirus-Induced Acute Respiratory Distress Syndrome by Radiotherapy. iScience 2020; 23:101215. [PMID: 32512383 PMCID: PMC7260547 DOI: 10.1016/j.isci.2020.101215] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 01/08/2023] Open
Abstract
The acute respiratory distress syndrome (ARDS) induced by SARS-CoV-2-mediated cytokine storm (CS) in lungs leads to the high mortality in COVID-19 patients. To reduce ARDS, an ideal approach is to diminish virus loading by activating immune cells for CS prevention or to suppress the overactive cytokine-releasing immune cells for CS inhibition. Here, a potential radiation-mediated CS regulation is raised by reevaluating the radiation-mediated pneumonia control in the 1920s, with the following latent advantages of lung radiotherapy (LR) in treatment of COVID-19: (1) radiation accesses poorly circulated tissue more efficiently than blood-delivered medications; (2) low-dose radiation (LDR)-mediated metabolic rewiring and immune cell activation inhibit virus loading; (3) pre-consumption of immune reserves by LDR decreases CS severity; (4) higherdose radiation (HDR) within lung-tolerable doses relieves CS by eliminating in situ overactive cytokine-releasing cells. Thus, LDR and HDR or combined with antiviral and life-supporting modalities may mitigate SARS-CoV-2 and other virus-mediated ARDS.
Collapse
Affiliation(s)
- Jian Jian Li
- Department of Radiation Oncology, NCI-designated Comprehensive Cancer Center, University of California at Davis School of Medicine, 4501 X Street, Suite G0140, Sacramento, CA 95817, USA.
| |
Collapse
|
228
|
Radiation-induced bystander and abscopal effects: important lessons from preclinical models. Br J Cancer 2020; 123:339-348. [PMID: 32581341 PMCID: PMC7403362 DOI: 10.1038/s41416-020-0942-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 03/10/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy is a pivotal component in the curative treatment of patients with localised cancer and isolated metastasis, as well as being used as a palliative strategy for patients with disseminated disease. The clinical efficacy of radiotherapy has traditionally been attributed to the local effects of ionising radiation, which induces cell death by directly and indirectly inducing DNA damage, but substantial work has uncovered an unexpected and dual relationship between tumour irradiation and the host immune system. In clinical practice, it is, therefore, tempting to tailor immunotherapies with radiotherapy in order to synergise innate and adaptive immunity against cancer cells, as well as to bypass immune tolerance and exhaustion, with the aim of facilitating tumour regression. However, our understanding of how radiation impacts on immune system activation is still in its early stages, and concerns and challenges regarding therapeutic applications still need to be overcome. With the increasing use of immunotherapy and its common combination with ionising radiation, this review briefly delineates current knowledge about the non-targeted effects of radiotherapy, and aims to provide insights, at the preclinical level, into the mechanisms that are involved with the potential to yield clinically relevant combinatorial approaches of radiotherapy and immunotherapy.
Collapse
|
229
|
Arina A, Gutiontov SI, Weichselbaum RR. Radiotherapy and Immunotherapy for Cancer: From "Systemic" to "Multisite". Clin Cancer Res 2020; 26:2777-2782. [PMID: 32047000 PMCID: PMC10759929 DOI: 10.1158/1078-0432.ccr-19-2034] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/28/2020] [Accepted: 02/06/2020] [Indexed: 11/16/2022]
Abstract
In the era of cancer immunotherapy, there is significant interest in combining conventional cancer therapies, such as radiotherapy, with drugs that stimulate the immune system. The observation that ionizing radiation applied to murine tumors delays the growth of distant tumors ("abscopal effect") and that this effect is potentiated by immunostimulatory drugs, led to clinical trials in which often only one lesion is irradiated in combination with immunotherapy drugs. The results of these initial clinical trials combining radio therapy and immunotherapy show that a meaningful abscopal effect is still infrequent. Recent preclinical data suggest that preexistent intratumoral T cells can survive radiation and contribute to its therapeutic effect. In this review, we discuss possible mechanisms underlying the preclinical/clinical discrepancies regarding the abscopal effect, and we propose the irradiation of multiple or all tumor sites in combination with systemic immunotherapy as a possible avenue to increase the efficacy of radio-immunotherapy.
Collapse
Affiliation(s)
- Ainhoa Arina
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois.
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Stanley I Gutiontov
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois.
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| |
Collapse
|
230
|
Datta NR, Kok HP, Crezee H, Gaipl US, Bodis S. Integrating Loco-Regional Hyperthermia Into the Current Oncology Practice: SWOT and TOWS Analyses. Front Oncol 2020; 10:819. [PMID: 32596144 PMCID: PMC7303270 DOI: 10.3389/fonc.2020.00819] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Moderate hyperthermia at temperatures between 40 and 44°C is a multifaceted therapeutic modality. It is a potent radiosensitizer, interacts favorably with a host of chemotherapeutic agents, and, in combination with radiotherapy, enforces immunomodulation akin to “in situ tumor vaccination.” By sensitizing hypoxic tumor cells and inhibiting repair of radiotherapy-induced DNA damage, the properties of hyperthermia delivered together with photons might provide a tumor-selective therapeutic advantage analogous to high linear energy transfer (LET) neutrons, but with less normal tissue toxicity. Furthermore, the high LET attributes of hyperthermia thermoradiobiologically are likely to enhance low LET protons; thus, proton thermoradiotherapy would mimic 12C ion therapy. Hyperthermia with radiotherapy and/or chemotherapy substantially improves therapeutic outcomes without enhancing normal tissue morbidities, yielding level I evidence reported in several randomized clinical trials, systematic reviews, and meta-analyses for various tumor sites. Technological advancements in hyperthermia delivery, advancements in hyperthermia treatment planning, online invasive and non-invasive MR-guided thermometry, and adherence to quality assurance guidelines have ensured safe and effective delivery of hyperthermia to the target region. Novel biological modeling permits integration of hyperthermia and radiotherapy treatment plans. Further, hyperthermia along with immune checkpoint inhibitors and DNA damage repair inhibitors could further augment the therapeutic efficacy resulting in synthetic lethality. Additionally, hyperthermia induced by magnetic nanoparticles coupled to selective payloads, namely, tumor-specific radiotheranostics (for both tumor imaging and radionuclide therapy), chemotherapeutic drugs, immunotherapeutic agents, and gene silencing, could provide a comprehensive tumor-specific theranostic modality akin to “magic (nano)bullets.” To get a realistic overview of the strength (S), weakness (W), opportunities (O), and threats (T) of hyperthermia, a SWOT analysis has been undertaken. Additionally, a TOWS analysis categorizes future strategies to facilitate further integration of hyperthermia with the current treatment modalities. These could gainfully accomplish a safe, versatile, and cost-effective enhancement of the existing therapeutic armamentarium to improve outcomes in clinical oncology.
Collapse
Affiliation(s)
- Niloy R Datta
- Centre for Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - H Petra Kok
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hans Crezee
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stephan Bodis
- Centre for Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
231
|
Chulpanova DS, Kitaeva KV, Green AR, Rizvanov AA, Solovyeva VV. Molecular Aspects and Future Perspectives of Cytokine-Based Anti-cancer Immunotherapy. Front Cell Dev Biol 2020; 8:402. [PMID: 32582698 PMCID: PMC7283917 DOI: 10.3389/fcell.2020.00402] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/01/2020] [Indexed: 12/11/2022] Open
Abstract
Cytokine-based immunotherapy is a promising field in the cancer treatment, since cytokines, as proteins of the immune system, are able to modulate the host immune response toward cancer cell, as well as directly induce tumor cell death. Since a low dose monotherapy with some cytokines has no significant therapeutic results and a high dose treatment leads to a number of side effects caused by the pleiotropic effect of cytokines, the problem of understanding the influence of cytokines on the immune cells involved in the pro- and anti-tumor immune response remains a pressing one. Immune system cells carry CD makers on their surface which can be used to identify various populations of cells of the immune system that play different roles in pro- and anti-tumor immune responses. This review discusses the functions and specific CD markers of various immune cell populations which are reported to participate in the regulation of the immune response against the tumor. The results of research studies and clinical trials investigating the effect of cytokine therapy on the regulation of immune cell populations and their surface markers are also discussed. Current trends in the development of cancer immunotherapy, as well as the role of cytokines in combination with other therapeutic agents, are also discussed.
Collapse
Affiliation(s)
- Daria S Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Kristina V Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, United Kingdom
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, United Kingdom
| | - Valeriya V Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
232
|
Nanoparticle delivery of immunostimulatory oligonucleotides enhances response to checkpoint inhibitor therapeutics. Proc Natl Acad Sci U S A 2020; 117:13428-13436. [PMID: 32493746 PMCID: PMC7306768 DOI: 10.1073/pnas.2001569117] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Checkpoint inhibitor (CPI) immunotherapies have revolutionized the treatment of a wide array of cancers, but their utility remains limited to a subset of patients with favorable disease phenotypes. We show that the generation of peptide-based nanocomplexes carrying immunostimulatory oligonucleotides dramatically increases the potency of certain of these compounds to stimulate toll-like receptor signaling. The administration of immunostimulatory nanocomplexes carrying CpG oligonucleotides generates antitumor effects and enhances the efficacy of checkpoint inhibitor antibody therapy in mouse models of cancer, and the nanocomplex formulation enables drastic reductions in the dose required to generate therapeutic effects. The recent advent of immune checkpoint inhibitor (CPI) antibodies has revolutionized many aspects of cancer therapy, but the efficacy of these breakthrough therapeutics remains limited, as many patients fail to respond for reasons that still largely evade understanding. An array of studies in human patients and animal models has demonstrated that local signaling can generate strongly immunosuppressive microenvironments within tumors, and emerging evidence suggests that delivery of immunostimulatory molecules into tumors can have therapeutic effects. Nanoparticle formulations of these cargoes offer a promising way to maximize their delivery and to enhance the efficacy of checkpoint inhibitors. We developed a modular nanoparticle system capable of encapsulating an array of immunostimulatory oligonucleotides that, in some cases, greatly increase their potency to activate inflammatory signaling within immune cells in vitro. We hypothesized that these immunostimulatory nanoparticles could suppress tumor growth by activating similar signaling in vivo, and thereby also improve responsiveness to immune checkpoint inhibitor antibody therapies. We found that our engineered nanoparticles carrying a CpG DNA ligand of TLR9 can suppress tumor growth in several animal models of various cancers, resulting in an abscopal effect on distant tumors, and improving responsiveness to anti-CTLA4 treatment with combinatorial effects after intratumoral administration. Moreover, by incorporating tumor-homing peptides, immunostimulatory nucleotide-bearing nanoparticles facilitate antitumor efficacy after systemic intravenous (i.v.) administration.
Collapse
|
233
|
Green M, Van Nest SJ, Soisson E, Huber K, Liao Y, McBride W, Dominello MM, Burmeister J, Joiner MC. Three discipline collaborative radiation therapy (3DCRT) special debate: We should treat all cancer patients with hypofractionation. J Appl Clin Med Phys 2020; 21:7-14. [PMID: 32602186 PMCID: PMC7324689 DOI: 10.1002/acm2.12954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Michael Green
- Department of Radiation OncologyUniversity of MichiganAnn ArborMIUSA
| | | | - Emilie Soisson
- Department of RadiologyUniversity of VermontBurlingtonVTUSA
| | - Kathryn Huber
- Department of Radiation OncologyTufts Medical CenterBostonMAUSA
| | - Yixiang Liao
- Department of Radiation OncologyRush University Medical CenterChicagoILUSA
| | - William McBride
- Department of Radiation OncologyUniversity of California at Los Angeles (UCLA)Los AngelesCAUSA
| | | | - Jay Burmeister
- Department of OncologyWayne State University School of MedicineDetroitMIUSA
- Gershenson Radiation Oncology CenterBarbara Ann Karmanos Cancer InstituteDetroitMIUSA
| | - Michael C. Joiner
- Department of OncologyWayne State University School of MedicineDetroitMIUSA
| |
Collapse
|
234
|
Rao G, Sonabend A, Heimberger AB. Are radiation and response biomarkers the missing elements for efficacious immunotherapy for glioma patients? Neuro Oncol 2020; 22:590-591. [PMID: 31967642 DOI: 10.1093/neuonc/noaa018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Ganesh Rao
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adam Sonabend
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Neurosurgery, Northwestern Medicine, Chicago, Illinois
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
235
|
Chargari C, Levy A, Paoletti X, Soria JC, Massard C, Weichselbaum RR, Deutsch E. Methodological Development of Combination Drug and Radiotherapy in Basic and Clinical Research. Clin Cancer Res 2020; 26:4723-4736. [PMID: 32409306 DOI: 10.1158/1078-0432.ccr-19-4155] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/14/2020] [Accepted: 05/12/2020] [Indexed: 01/03/2023]
Abstract
Newer technical improvements in radiation oncology have been rapidly implemented in recent decades, allowing an improved therapeutic ratio. The development of strategies using local and systemic treatments concurrently, mainly targeted therapies, has however plateaued. Targeted molecular compounds and immunotherapy are increasingly being incorporated as the new standard of care for a wide array of cancers. A better understanding of possible prior methodology issues is therefore required and should be integrated into upcoming early clinical trials including individualized radiotherapy-drug combinations. The outcome of clinical trials is influenced by the validity of the preclinical proofs of concept, the impact on normal tissue, the robustness of biomarkers and the quality of the delivery of radiation. Herein, key methodological aspects are discussed with the aim of optimizing the design and implementation of future precision drug-radiotherapy trials.
Collapse
Affiliation(s)
- Cyrus Chargari
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Institut de Recherche Biomédicale des Armées, Brétigny sur Orge, France
| | - Antonin Levy
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Xavier Paoletti
- University of Versailles St. Quentin, France
- Institut Curie INSERM U900, Biostatistics for Personalized Medicine Team, St. Cloud, France
| | | | - Christophe Massard
- Université Paris-Sud, Orsay, France
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
236
|
Eguren-Santamaria I, Sanmamed MF, Goldberg SB, Kluger HM, Idoate MA, Lu BY, Corral J, Schalper KA, Herbst RS, Gil-Bazo I. PD-1/PD-L1 Blockers in NSCLC Brain Metastases: Challenging Paradigms and Clinical Practice. Clin Cancer Res 2020; 26:4186-4197. [PMID: 32354698 DOI: 10.1158/1078-0432.ccr-20-0798] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/26/2020] [Accepted: 04/28/2020] [Indexed: 11/16/2022]
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized the management of advanced non-small cell lung cancer (NSCLC). However, most pivotal phase III trials systematically excluded patients with active brain metastases, precluding the generalization of the results. Although theoretically restricted from crossing the blood-brain barrier, the novel pharmacokinetic/pharmacodynamic profiles of anti-PD-1/PD-L1 drugs have prompted studies to evaluate their activity in patients with NSCLC with active central nervous system (CNS) involvement. Encouraging results have suggested that ICI could be active in the CNS in selected patients with driver-negative advanced NSCLC with high PD-L1 expression and low CNS disease burden. Single-agent CNS response rates around 30% have been reported. Beyond this particular setting, anti-PD-1/PD-L1 antibodies have been evaluated in patients receiving local therapy for brain metastases (BM), addressing concerns about potential neurologic toxicity risks associated with radiotherapy, more specifically, radionecrosis (RN). Accordingly, a variety of clinical and imaging strategies are being appropriately developed to evaluate tumor response and to rule out pseudoprogression or radionecrosis. Our purpose is to critically summarize the advances regarding the role of systemic anti-PD-1/PD-L1 antibodies for the treatment of NSCLC BM. Data were collected from the PubMed database, reference lists, and abstracts from the latest scientific meetings. Recent reports suggest anti-PD-1/PD-L1 agents are active in a subset of patients with NSCLC with BM showing acceptable toxicity. These advances are expected to change soon the management of these patients but additional research is required to address concerns regarding radionecrosis and the appropriate sequencing of local and systemic therapy combinations.
Collapse
Affiliation(s)
- Iñaki Eguren-Santamaria
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain.,University of Navarra, Center for Applied Medical Research, Program of Immunology and Immunotherapy, Pamplona, Spain
| | - Miguel F Sanmamed
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain.,University of Navarra, Center for Applied Medical Research, Program of Immunology and Immunotherapy, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Sarah B Goldberg
- Yale University School of Medicine and Yale Cancer Center, New Haven, Connecticut
| | - Harriet M Kluger
- Yale University School of Medicine and Yale Cancer Center, New Haven, Connecticut
| | - Miguel A Idoate
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Benjamin Y Lu
- Yale University School of Medicine and Yale Cancer Center, New Haven, Connecticut
| | - Jesús Corral
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Roy S Herbst
- Yale University School of Medicine and Yale Cancer Center, New Haven, Connecticut
| | - Ignacio Gil-Bazo
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain. .,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,University of Navarra, Center for Applied Medical Research, Program of Solid Tumors, Pamplona, Spain
| |
Collapse
|
237
|
Tubin S, Yan W, Mourad WF, Fossati P, Khan MK. The future of radiation-induced abscopal response: beyond conventional radiotherapy approaches. Future Oncol 2020; 16:1137-1151. [PMID: 32338046 DOI: 10.2217/fon-2020-0063] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Advances in the immunological pharmaceuticals, such as checkpoint inhibitors and agonists, have positive implications for the future of the radiotherapy abscopal response. A once rare phenomenon, whereby distant nonirradiated tumor sites regressed after radiotherapy alone, may become more common when combined with the immune modulating agents. Radiotherapy can increase neoantigen expression, increased tumor PD-L1 expression, increase MHC class I expression, reverse exhausted CD8 T cells and increase tumor-infiltrating tumors within the tumor microenvironment. These changes in the tumor and the tumor microenvironment after radiotherapy could potentiate responses to anti-CTL-4, anti-PD-L1/PD-1 and other immunotherapy agents. Thus, advances in checkpoint inhibitors have increased interest in re-evaluation of the role of conventional radiotherapy approaches on the immune system. We reviewed newer nonconventional approaches such as SBRT-PATHY, GRID, FLASH, carbon ion and proton therapy and their role in eliciting immune responses. We believe that combining these novel radiation methods may enhance the outcome with the newly US FDA approved immune modulating agents.
Collapse
Affiliation(s)
- Slavisa Tubin
- MedAustron Center for Ion Therapy and Research, Marie Curie Strasse 5, A-2700 Wiener Neustadt, Austria
| | - Weisi Yan
- Department of Radiation Oncology, Thomas Jefferson University, 11th St, Philadelphia, PA 19107, USA
| | - Waleed F Mourad
- Department of Radiation Medicine, Markey Cancer Center, University of Kentucky, Medical Center, MN 150 - Lexington, KY 40536-0298, USA
| | - Piero Fossati
- MedAustron Center for Ion Therapy and Research, Marie Curie Strasse 5, A-2700 Wiener Neustadt, Austria
| | - Mohammad K Khan
- Department of Radiation Oncology, Emory University School of Medicine, Winship Cancer Institute, 1365-C Clifton Road, Atlanta, GA 30322, USA
| |
Collapse
|
238
|
Heigener DF, Reck M. Immune Checkpoint Inhibition in Non-metastatic Non-small Cell Lung Cancer: Chance for Cure? Drugs 2020; 79:1937-1945. [PMID: 31749060 DOI: 10.1007/s40265-019-01222-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Immune checkpoint inhibition of programmed-death receptor 1 (PD-1) or its ligand (PD-L1) has become a standard in the treatment of metastatic non-small cell lung cancer, either as monotherapy or in combination. Recently, it could be shown that immunotherapy works as consolidation after chemoradiotherapy in locally advanced disease if the tumours express PD-L1. A significant and meaningful survival benefit for consolidation with durvalumab after chemoradiotherapy compared to chemoradiotherapy alone was observed in the PACIFIC trial. In addition, there is a growing body of evidence that this treatment modality is also effective in a neoadjuvant setting in early stages, whereas the role as adjuvant treatment after surgery needs to be determined. The impact of combination therapies in non-metastatic stages-either neoadjuvant or adjuvant-needs to be evaluated in future trials. It is yet unclear whether PD-L1 and tumour mutational burden are predictive biomarkers as randomised trials are missing.
Collapse
Affiliation(s)
- David F Heigener
- Department of Pulmonary Medicine, Helios Klinik Schleswig, Schleswig, Germany
- University of Kiel, School of Medicine, Kiel, Germany
| | - Martin Reck
- Department of Oncology, LungenClinic Grosshansdorf, Woehrendamm 80, Grosshansdorf, Germany.
- Airway Research Center North in the German Center for Lung Research (DZL), Grosshansdorf, Germany.
| |
Collapse
|
239
|
Oliver AJ, Darcy PK, Trapani JA, Kershaw MH, Slaney CY. Cross-talk between tumors at anatomically distinct sites. FEBS J 2020; 288:81-90. [PMID: 32248616 DOI: 10.1111/febs.15316] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/14/2020] [Accepted: 03/27/2020] [Indexed: 12/15/2022]
Abstract
Cancer tissue is not homogenous, and individual metastases at different anatomical locations can differ from the primary tumor and from one another in both their morphology and cellular composition, even within an individual patient. Tumors are composed of cancer cells and a range of other cell types, which, together with a variety of secreted molecules, collectively comprise the tumor microenvironment (TME). Cells of the TME can communicate with each other and with distant tissues in a form of molecular cross-talk to influence their growth and function. Cross-talk between cancer cells and local immune cells is well described and can lead to the induction of local immunosuppression. Recently, it has become apparent that tumors located remotely from each other, can engage in cross-talk that can influence their responsiveness to various therapies, including immunotherapy. In this article, we review studies that describe how tumors systemically communicate with distant tissues through motile cells, extracellular vesicles, and secreted molecules that can affect their function. In addition, we summarize evidence from mouse studies and the clinic that indicate an ability of some tumors to influence the progression and therapeutic responses of other tumors in different anatomical locations.
Collapse
Affiliation(s)
- Amanda J Oliver
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| |
Collapse
|
240
|
Chen VE, Greenberger BA, Taylor JM, Edelman MJ, Lu B. The Underappreciated Role of the Humoral Immune System and B Cells in Tumorigenesis and Cancer Therapeutics: A Review. Int J Radiat Oncol Biol Phys 2020; 108:38-45. [PMID: 32251756 DOI: 10.1016/j.ijrobp.2020.03.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 03/21/2020] [Indexed: 02/07/2023]
Abstract
The advent of immunotherapy has ushered in a new era in both cancer research and cancer treatment strategies. Published reviews have described potential mechanisms for therapeutic synergisms from the combination of radiation therapy and immunotherapy, largely overlooking the role of humoral immunity by only focusing on cellular immunity. Given that these 2 branches of the immune system are highly interdependent, in this review we detail both what has already been established regarding the role of humoral immunity in cancer and propose potential avenues that are ripe for further investigation and potential clinical applications.
Collapse
Affiliation(s)
- Victor E Chen
- Department of Radiation Oncology, Sidney Kimmel Medical College & Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Benjamin A Greenberger
- Department of Radiation Oncology, Sidney Kimmel Medical College & Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - James M Taylor
- Department of Radiation Oncology, Sidney Kimmel Medical College & Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Martin J Edelman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Bo Lu
- Department of Radiation Oncology, Sidney Kimmel Medical College & Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
241
|
Gough MJ, Sharon S, Crittenden MR, Young KH. Using Preclinical Data to Design Combination Clinical Trials of Radiation Therapy and Immunotherapy. Semin Radiat Oncol 2020; 30:158-172. [PMID: 32381295 PMCID: PMC7213059 DOI: 10.1016/j.semradonc.2019.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunotherapies are rapidly entering the clinic as approved treatments for diverse cancer pathologies. Radiation therapy is an integral partner in cancer therapy, commonly as part of complicated multimodality approaches that optimize patient outcomes. Preclinical studies have demonstrated that the success of radiation therapy in tumor control is due in part to immune mechanisms, and that outcomes following radiation therapy can be improved through combination with a range of immunotherapies. However, preclinical models of cancer are very different from patient tumors, and the way these preclinical tumors are treated is often very different from standard of care treatment of patients. This review examines the preclinical and clinical data for the role of the immune system in radiation therapy outcomes, and how to integrate preclinical findings into clinical trials, using ongoing studies as examples.
Collapse
Affiliation(s)
- Michael J Gough
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR.
| | - Shay Sharon
- Department of Oral and Maxillofacial Surgery, Hadassah and Hebrew University Medical Center, Jerusalem, ISRAEL
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR; The Oregon Clinic, Portland, OR
| | - Kristina H Young
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR; The Oregon Clinic, Portland, OR
| |
Collapse
|
242
|
McLaughlin M, Patin EC, Pedersen M, Wilkins A, Dillon MT, Melcher AA, Harrington KJ. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer 2020; 20:203-217. [PMID: 32161398 DOI: 10.1038/s41568-020-0246-1] [Citation(s) in RCA: 443] [Impact Index Per Article: 110.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 12/17/2022]
Abstract
The development of immune checkpoint inhibitors (ICIs) is revolutionizing the way we think about cancer treatment. Even so, for most types of cancer, only a minority of patients currently benefit from ICI therapies. Intrinsic and acquired resistance to ICIs has focused research towards new combination therapy approaches that seek to increase response rates, the depth of remission and the durability of benefit. In this Review, we describe how radiotherapy, through its immunomodulating effects, represents a promising combination partner with ICIs. We describe how recent research on DNA damage response (DDR) inhibitors in combination with radiotherapy may be used to augment this approach. Radiotherapy can kill cancer cells while simultaneously triggering the release of pro-inflammatory mediators and increasing tumour-infiltrating immune cells - phenomena often described colloquially as turning immunologically 'cold' tumours 'hot'. Here, we focus on new developments illustrating the key role of tumour cell-autonomous signalling after radiotherapy. Radiotherapy-induced tumour cell micronuclei activate cytosolic nucleic acid sensor pathways, such as cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING), and propagation of the resulting inflammatory signals remodels the immune contexture of the tumour microenvironment. In parallel, radiation can impact immunosurveillance by modulating neoantigen expression. Finally, we highlight how tumour cell-autonomous mechanisms might be exploited by combining DDR inhibitors, ICIs and radiotherapy.
Collapse
Affiliation(s)
- Martin McLaughlin
- Targeted Therapy Team, The Institute of Cancer Research, London, UK.
| | - Emmanuel C Patin
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Malin Pedersen
- Translational Immunotherapy Team, The Institute of Cancer Research, London, UK
| | | | - Magnus T Dillon
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Alan A Melcher
- Translational Immunotherapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Kevin J Harrington
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| |
Collapse
|
243
|
Mondini M, Levy A, Meziani L, Milliat F, Deutsch E. Radiotherapy-immunotherapy combinations - perspectives and challenges. Mol Oncol 2020; 14:1529-1537. [PMID: 32112478 PMCID: PMC7332212 DOI: 10.1002/1878-0261.12658] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/18/2019] [Accepted: 02/27/2020] [Indexed: 12/23/2022] Open
Abstract
Ionizing radiation has historically been used to treat cancer by killing tumour cells, in particular by inducing DNA damage. This view of radiotherapy (RT) as a simple cytotoxic agent has dramatically changed in recent years, and it is now widely accepted that RT can deeply reshape the tumour environment by modulating the immune response. Such evidence gives a strong rationale for the use of immunomodulators to boost the therapeutic value of RT, introducing the era of ‘immunoradiotherapy’. The increasing amount of preclinical and clinical data concerning the combination of RT with immunomodulators, in particular with immune checkpoint inhibitors such as anti‐PD‐1/PD‐L1 and anti‐CTLA4, reflects the interest of the scientific and medical community concerning immunoradiotherapy. The expectations are enormous since the rationale for performing such combinations is strong, with the possibility to use a local treatment such as RT to amplify a systemic antitumour response, as illustrated by the case of the abscopal effect. Nevertheless, several points remain to be addressed such as the need to find biomarkers to identify patients who will benefit from immunoradiotherapy, the identification of the best sequences/schedules for combination with immunomodulators and mechanisms to overcome resistance. Additionally, the effects of immunoradiotherapy on healthy tissues and related toxicity remain largely unexplored. To answer these critical questions and make immunoradiotherapy keep its promising qualities, large efforts are needed from both the pharmaceutical industry and academic/governmental research. Moreover, because of the work of both these entities, the arsenal of available immunomodulators is quickly expanding, thus opening the field to increasing combinations with RT. We thus forecast that the field of immunoradiotherapy will further expand in the coming years, and it needs to be supported by appropriate investment plans.
Collapse
Affiliation(s)
- Michele Mondini
- Gustave Roussy, Université Paris-Saclay, SIRIC SOCRATE, Villejuif, France.,INSERM, U1030, Labex LERMIT, Villejuif, France
| | - Antonin Levy
- INSERM, U1030, Labex LERMIT, Villejuif, France.,Département de Radiothérapie, Gustave Roussy, Université Paris-Saclay, DHU TORINO, Villejuif, France
| | - Lydia Meziani
- Gustave Roussy, Université Paris-Saclay, SIRIC SOCRATE, Villejuif, France.,INSERM, U1030, Labex LERMIT, Villejuif, France
| | - Fabien Milliat
- Department of Radiobiology and regenerative Medicine (SERAMED), Laboratory of Medical Radiobiology (LRMed), Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Eric Deutsch
- Gustave Roussy, Université Paris-Saclay, SIRIC SOCRATE, Villejuif, France.,INSERM, U1030, Labex LERMIT, Villejuif, France.,Département de Radiothérapie, Gustave Roussy, Université Paris-Saclay, DHU TORINO, Villejuif, France
| |
Collapse
|
244
|
Chi MS, Mehta MP, Yang KL, Lai HC, Lin YC, Ko HL, Wang YS, Liao KW, Chi KH. Putative Abscopal Effect in Three Patients Treated by Combined Radiotherapy and Modulated Electrohyperthermia. Front Oncol 2020; 10:254. [PMID: 32211319 PMCID: PMC7077340 DOI: 10.3389/fonc.2020.00254] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 02/14/2020] [Indexed: 01/08/2023] Open
Abstract
Purpose: True abscopal responses from radiation therapy are extremely rare; the combination of immune checkpoint inhibitors with radiation therapy has led to more reports of the abscopal effect, but even in this setting, the genuine magnitude remains unknown and is still considered generally uncommon. We report the occurrence of what appears to be putative, durable abscopal tumor responses with associated auto-immune systemic reactions resulting from the combination of local radiotherapy (RT) and modulated electrohyperthermia (mEHT). Materials and Methods: Data from advanced cancer patients treated palliatively with RT and mEHT between January and December 2017 were collected as part of a post-marketing safety monitoring program of mEHT therapy. We specified a minimum RT dose of 30 Gy and at least four mEHT treatments for reporting toxicities, which was the primary aim of the larger study. Results: Thirty-three patients treated with RT and mEHT, both applied to the same lesion, were included. The median RT dose was 45.5 Gy in 20 fractions (fxs) and the median number of mEHT treatments was 12 (range, 4–20). Most patients had subsequent systemic therapy after one course of RT and mEHT. Three patients (9.1%) developed autoimmune toxicities. Case number 1 received RT and mEHT only; case number 2 had two cycles of concurrent low dose chemotherapy during RT; and case number 3 received concurrent immune checkpoint inhibitors. None of the three patients received any further systemic treatment due to obvious treatment-related autoimmune reactions which occurred rapidly after RT; one had autoimmune hepatitis, one had dermatitis herpetiformis and the third developed severe myasthenia gravis. Interestingly, what we surmise to be long-lasting abscopal responses outside the irradiated area, were noted in all three patients. Conclusion: RT combined with mEHT could putatively result in enhancing immune responsiveness. These preliminary observational findings lead to the generation of a hypothesis that this combination induces both an in-situ, tumor-specific immune reaction and an anti-self-autoimmune reaction, in at least a small proportion of patients, and of those who experience the auto-immune response, tumor response is a concomitant finding. Mechanisms underlying this phenomenon need to be investigated further.
Collapse
Affiliation(s)
- Mau-Shin Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Ph.D. Degree Program of Biomedical Science and Engineering, National Chiao-Tung University, Hsinchu, Taiwan
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Miami, FL, United States
| | - Kai-Lin Yang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Hung-Chih Lai
- Department of Hematology and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ying-Chu Lin
- Department of Hematology and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Hui-Ling Ko
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yu-Shan Wang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Institute of Molecular Medicine and Bioengineering, National Chiao-Tung University, Hsinchu, Taiwan
| | - Kuang-Wen Liao
- Institute of Molecular Medicine and Bioengineering, National Chiao-Tung University, Hsinchu, Taiwan
| | - Kwan-Hwa Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
245
|
MacManus MP, Hofman MS, Hicks RJ, Campbell BA, Wirth A, Seymour JF, Haynes N, Burbury K. Abscopal Regressions of Lymphoma After Involved-Site Radiation Therapy Confirmed by Positron Emission Tomography. Int J Radiat Oncol Biol Phys 2020; 108:204-211. [PMID: 32151671 DOI: 10.1016/j.ijrobp.2020.02.636] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 02/02/2020] [Accepted: 02/14/2020] [Indexed: 01/17/2023]
Abstract
PURPOSE Patients with abscopal regressions of lymphoma after palliative involved-site radiation therapy (ISRT), detected on sequential 18F-fluorodeoxyglucose positron emission tomography (FDG-PET), were identified by audit. A retrospective analysis was subsequently conducted to estimate the frequency of abscopal regression in follicular lymphoma (FL). METHODS AND MATERIALS Potential cases were identified at multidisciplinary lymphoma meetings and fulfilled these criteria: (1) palliative ISRT given for histologically confirmed lymphoma, (2) >2 lesions visualized on FDG-PET, (3) >1 unirradiated lesion(s) outside ISRT volume, (4) no systemic therapy delivered <2 months before radiation therapy or between radiation therapy and response assessment, (5) complete metabolic response (CMR) in ≥1 unirradiated lesions detected on serial FDG-PET/CT. All ISRT patients with FL treated in 2016 to 2018 were systematically reviewed. RESULTS Seven cases of abscopal regression were identified, including 4 patients with FL. In all cases, a CMR was apparent both within the ISRT volume and in ≥1 unirradiated lesions. One patient each was identified with mantle cell lymphoma (4 Gy in 2 fractions), Hodgkin lymphoma (20 Gy in 3 fractions, then 30 Gy in 15 fractions to the same volume), and Richter transformation of chronic lymphatic leukemia (30 Gy in 10 fractions). The 4 patients with FL received either 4 Gy in 2 fractions (n = 3) or 4 Gy followed 8 months later by 30 Gy in 15 fractions (n = 1). From 2016 to 2018, 29 courses of ISRT were prescribed for multifocal FL, after which 4 of 29 (13.8%) abscopal responses were detected, including in 4 of 9 (44.4%) patients with serial PET scans. Two patients, with relapsed disease after initial abscopal responses, experienced durable CMRs with immunotherapies. CONCLUSIONS In 4 of 7 cases, PET-detected abscopal regression of lymphoma occurred after 4 Gy, in 2 of 7 cases after repeated ISRT to the same volume, and in 2 of 7 was associated with subsequent complete response to immunotherapy, consistent with an immune basis for the abscopal effect. Abscopal regressions in FL appear to be more common than previously suspected.
Collapse
Affiliation(s)
- Michael P MacManus
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.
| | - Michael S Hofman
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Department of Molecular Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Rodney J Hicks
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Department of Molecular Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Belinda A Campbell
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Andrew Wirth
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - John F Seymour
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Department of Molecular Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Nicole Haynes
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Kate Burbury
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
| |
Collapse
|
246
|
Cell repopulation, rewiring metabolism, and immune regulation in cancer radiotherapy. RADIATION MEDICINE AND PROTECTION 2020. [DOI: 10.1016/j.radmp.2020.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
247
|
Demaria S, Formenti SC. The abscopal effect 67 years later: from a side story to center stage. Br J Radiol 2020; 93:20200042. [PMID: 32101479 DOI: 10.1259/bjr.20200042] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
For over a century, ionising radiation has been used to treat cancer based on its cytotoxic effects on tumour cells. Technical progress has enabled more precise targeting of the tumour to reduce normal tissue toxicity while delivering higher radiation doses per fraction of treatment.In 1953, unexpected regression in lesions outside of the irradiated field were noted by an observant physician, RH Mole, who named such phenomenon "abscopal effect" from the Latin ab (position away from) and scopus (mark or target), in an article published in this journal. Clinical abscopal responses have been reported over the years but because of their very rare occurrence they could not be methodically studied, remaining akin to a curiosity. Nevertheless, their occurrence has ignited interest in studying the systemic effects of radiotherapy. Progress in dissecting the mechanisms that govern the function of the immune system in cancer has enabled to study the implication of immunity in the abscopal effect of radiation. It has become clear that ionising radiation activates canonical pathways of response to viral infections, and can stimulate antitumour immunity. These immune stimulatory effects of radiation have become clinically relevant in the current era of cancer immunotherapy, rendering abscopal responses in patients an attainable aim. Here, we will briefly review the parallel evolutions of two separate fields of medicine, radiation therapy and cancer immunology, and discuss their therapeutic partnership.
Collapse
Affiliation(s)
- Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, Stich Radiation Oncology, 525 East 68th Street, Box #169, New York, NY 10065, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, Stich Radiation Oncology, 525 East 68th Street, Box #169, New York, NY 10065, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
248
|
Corrao G, Marvaso G, Ferrara R, Lo Russo G, Gugliandolo SG, Piperno G, Spaggiari L, De Marinis F, Orecchia R, Garassino MC, Jereczek-Fossa BA. Stereotatic radiotherapy in metastatic non-small cell lung cancer: Combining immunotherapy and radiotherapy with a focus on liver metastases. Lung Cancer 2020; 142:70-79. [PMID: 32120227 DOI: 10.1016/j.lungcan.2020.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/17/2020] [Accepted: 02/22/2020] [Indexed: 01/19/2023]
Abstract
Presence of liver metastases correlates with worse survival and response to any treatments. This may be due to the microenvironment of liver which leads tumor to escape from Immune System. Stereotactic Body Radiation Therapy may help to sensitize Immune System and to improve the immunotherapy effect. Interest is being directed toward combining Immune-Checkpoint Inhibitors with radiotherapy to improve response to immunotherapy. However, the mechanisms by which radiation induces anti-tumor T-cells remain unclear. Preclinical studies founded radiotherapy enhances antitumor immune responses, increasing tumor antigen release, and inducing T-cell infiltration. Radiotherapy is under investigation for its ability to enhance responses to immunotherapy. Nevertheless, how to optimally deliver combination therapy regarding dose-fractionation and timing of radiotherapy is unknown. The aim of this review is to explore the role of Stereotactic Body Radiation Therapy in metastatic non-small cell lung cancer, focusing on patients with liver metastases, and the possible immunological implications combining immunotherapy and radiotherapy.
Collapse
Affiliation(s)
- Giulia Corrao
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Giulia Marvaso
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy.
| | - Roberto Ferrara
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133, Milan, Italy
| | - Giuseppe Lo Russo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133, Milan, Italy
| | - Simone Giovanni Gugliandolo
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141, Milan, Italy
| | - Gaia Piperno
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141, Milan, Italy
| | - Lorenzo Spaggiari
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy; Department of Thoracic Surgery, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Filippo De Marinis
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Roberto Orecchia
- Scientific Direction, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Marina Chiara Garassino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133, Milan, Italy
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| |
Collapse
|
249
|
Absolute Lymphocyte Count Predicts Abscopal Responses and Outcomes in Patients Receiving Combined Immunotherapy and Radiation Therapy: Analysis of 3 Phase 1/2 Trials. Int J Radiat Oncol Biol Phys 2020; 108:196-203. [PMID: 32036004 DOI: 10.1016/j.ijrobp.2020.01.032] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/21/2020] [Accepted: 01/27/2020] [Indexed: 10/25/2022]
Abstract
PURPOSE Research to elucidate predictive factors of the abscopal effect is an essential first step toward potentially modifying these factors to increase the incidence of systemic antitumor effects. This study, using data from 3 institutional phase 1/2 trials, examined the predictive capacity of recorded parameters in patients undergoing combined radiation therapy (RT) and immunotherapy and explored outcomes based on those predictive factors. METHODS AND MATERIALS All patients underwent combined immunotherapy and RT and had at least 1 nonirradiated noncontiguous lesion to evaluate out-of-field (abscopal) responses, defined as the best Response Evaluation Criteria in Solid Tumors response. RESULTS Altogether, 153 patients met study criteria, and the median follow-up was 21.1 months. The most common cancer types were non-small cell lung carcinoma (n = 62), small cell lung carcinoma (n = 25), head and neck cancers (n = 16), and renal cell carcinoma (n = 13). Immunotherapies included ipilimumab (n = 98) and pembrolizumab (n = 55). Multivariable linear regression indicated that post-RT absolute lymphocyte count (ALC), when analyzed as a continuous variable, correlated with abscopal responses (P < .001). For post-RT ALC, the abscopal response rate was 34.2% in the cohort with ALC higher than the median value, compared with 3.9% in patients with ALC lower than the median (P < .0001). Corresponding figures for pre-RT ALC were 30.3% versus 7.8%, respectively (P = .0004). Cox multivariate analysis confirmed that lower post-RT ALC also associated with poorer progression-free survival (P = .009) and overall survival (P = .026). CONCLUSIONS Lymphopenia, measured as the continuous variable of post-RT ALC, may affect the occurrence of abscopal responses and thus influence prognosis in patients treated with RT and immunotherapy. Although this hypothesis-generating finding requires corroboration by additional data, it suggests the importance of ALC monitoring and the potential of therapeutic manipulation of this parameter to induce abscopal effects.
Collapse
|
250
|
Benna M, Guy JB, Bosacki C, Jmour O, Ben Mrad M, Ogorodniitchouk O, Soltani S, Lan M, Daguenet E, Mery B, Sotton S, Magné N, Vallard A. Chemoradiation and granulocyte-colony or granulocyte macrophage-colony stimulating factors (G-CSF or GM-CSF): time to think out of the box? Br J Radiol 2020; 93:20190147. [PMID: 31971824 DOI: 10.1259/bjr.20190147] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Concerns have been raised about potential toxic interactions when colony-stimulating factors (CSFs) and chemoradiation are concurrently performed. In 2006, the ASCO guidelines advised against their concomitant use. Nevertheless, with the development of modern radiotherapy techniques and supportive care, the therapeutic index of combined chemotherapy, radiotherapy, and CSFs is worth reassessing. Recent clinical trials testing chemoradiation in lung cancer let investigators free to decide the use of concomitant CSFs or not. No abnormal infield event was reported after the use of modern radiotherapy techniques and concomitant chemotherapy regimens. These elements call for further investigation to set new recommendations in favour of the association of chemoradiation and CSFs. Moreover, radiotherapy could induce anticancer systemic effects mediated by the immune system in vitro and in vivo. With combined CSFs, this effect was reinforced in preclinical and clinical trials introducing innovative radioimmunotherapy models. So far, the association of radiation with CSFs has not been combined with immunotherapy. However, it might play a major role in triggering an immune response against cancer cells, leading to abscopal effects. The present article reassesses the therapeutic index of the combination CSFs-chemoradiation through an updated review on its safety and efficacy. It also provides a special focus on radioimmunotherapy.
Collapse
Affiliation(s)
- Marouan Benna
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Jean-Baptiste Guy
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Claire Bosacki
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Omar Jmour
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Majed Ben Mrad
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | | | - Saïd Soltani
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Meiling Lan
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Elisabeth Daguenet
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Benoîte Mery
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Sandrine Sotton
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Nicolas Magné
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Alexis Vallard
- Department of Radiotherapy, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| |
Collapse
|