1
|
Tepper SC, Lee L, Fice MP, Jones CM, Buac N, Vijayakumar G, Wang D, Colman MW, Gitelis S, Blank AT. Radiotherapy leads to improved overall survival in patients undergoing resection for Undifferentiated pleomorphic sarcoma. Surg Oncol 2024; 56:102118. [PMID: 39121675 DOI: 10.1016/j.suronc.2024.102118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/15/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND AND OBJECTIVES Undifferentiated pleomorphic sarcoma (UPS) is a frequent subtype within the heterogeneous group of soft tissue sarcomas (STS). The use of radiotherapy (RT) has become an important component of a multimodal approach to treating STS. Key studies have demonstrated that the addition of RT improves rates of local control in STS, though the effect on overall survival (OS) is less clear. Furthermore, there is very limited and conflicting evidence regarding effect of RT on overall survival in UPS. The purposes of this investigation were to examine the association between RT and OS in UPS patients undergoing surgical resection and to determine independent prognostic indicators of OS in this patient population. METHODS This was a retrospective review of patients who underwent surgical treatment for primary UPS from 1993 to 2021. Associations between RT and OS were analyzed with Kaplan-Meier curves and log-rank testing. Cox proportional hazards regression analysis was used to determine independent prognostic factors of OS. RESULTS One hundred and fourteen patients who underwent surgical resection of primary UPS were included in the study. Ninety-six (84.2 %) patients received RT perioperatively. Use of RT was associated with improved OS on log-rank testing (hazard ratio (HR) 0.20; 95 % confidence interval (CI) 0.11-0.36; p < 0.001). On multivariate analysis, RT was an independent predictor of improved OS (HR 0.18; 95 % CI 0.09-0.39; p < 0.001) while metastasis at presentation (HR 4.82; 95 % CI 2.26-10.27; p < 0.001) and older age (HR 1.92; 95 % CI 1.20-3.36; p = 0.02) were predictive of decreased OS. Use of RT was not significantly associated with a lower rate of local recurrence in our cohort (p = 0.49). CONCLUSIONS Use of RT in combination with surgery was an independent prognostic indicator of improved overall survival in UPS patients. Older age and metastasis at presentation were associated with worse overall survival. Based on this and other available studies, treatment for UPS should involve limb-sparing resection when feasible with RT to ensure optimal survival.
Collapse
Affiliation(s)
- Sarah C Tepper
- Midwest Orthopaedics at Rush University Medical Center, Chicago, IL, USA.
| | - Linus Lee
- Department of Orthopedic Surgery, Division of Orthopedic Oncology, Rush University Medical Center, Chicago, IL, USA
| | - Michael P Fice
- Midwest Orthopaedics at Rush University Medical Center, Chicago, IL, USA
| | - Conor M Jones
- Midwest Orthopaedics at Rush University Medical Center, Chicago, IL, USA
| | - Neil Buac
- Department of Orthopedic Surgery, Division of Orthopedic Oncology, Rush University Medical Center, Chicago, IL, USA
| | - Gayathri Vijayakumar
- Department of Orthopedic Surgery, Division of Orthopedic Oncology, Rush University Medical Center, Chicago, IL, USA
| | - Dian Wang
- Department of Radiation Oncology, Rush Medical College, Chicago, IL, USA
| | - Matthew W Colman
- Midwest Orthopaedics at Rush University Medical Center, Chicago, IL, USA
| | - Steven Gitelis
- Department of Orthopedic Surgery, Division of Orthopedic Oncology, Rush University Medical Center, Chicago, IL, USA
| | - Alan T Blank
- Department of Orthopedic Surgery, Division of Orthopedic Oncology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
2
|
Guo S, Yao Y, Tang Y, Xin Z, Wu D, Ni C, Huang J, Wei Q, Zhang T. Radiation-induced tumor immune microenvironments and potential targets for combination therapy. Signal Transduct Target Ther 2023; 8:205. [PMID: 37208386 DOI: 10.1038/s41392-023-01462-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/21/2023] [Accepted: 04/27/2023] [Indexed: 05/21/2023] Open
Abstract
As one of the four major means of cancer treatment including surgery, radiotherapy (RT), chemotherapy, immunotherapy, RT can be applied to various cancers as both a radical cancer treatment and an adjuvant treatment before or after surgery. Although RT is an important modality for cancer treatment, the consequential changes caused by RT in the tumor microenvironment (TME) have not yet been fully elucidated. RT-induced damage to cancer cells leads to different outcomes, such as survival, senescence, or death. During RT, alterations in signaling pathways result in changes in the local immune microenvironment. However, some immune cells are immunosuppressive or transform into immunosuppressive phenotypes under specific conditions, leading to the development of radioresistance. Patients who are radioresistant respond poorly to RT and may experience cancer progression. Given that the emergence of radioresistance is inevitable, new radiosensitization treatments are urgently needed. In this review, we discuss the changes in irradiated cancer cells and immune cells in the TME under different RT regimens and describe existing and potential molecules that could be targeted to improve the therapeutic effects of RT. Overall, this review highlights the possibilities of synergistic therapy by building on existing research.
Collapse
Affiliation(s)
- Siyu Guo
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yihan Yao
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Tang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Zengfeng Xin
- Department of Orthopedic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Dang Wu
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Chao Ni
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jian Huang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Qichun Wei
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Ting Zhang
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
3
|
Ren SN, Zhang ZY, Guo RJ, Wang DR, Chen FF, Chen XB, Fang XD. Application of nanotechnology in reversing therapeutic resistance and controlling metastasis of colorectal cancer. World J Gastroenterol 2023; 29:1911-1941. [PMID: 37155531 PMCID: PMC10122790 DOI: 10.3748/wjg.v29.i13.1911] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 02/02/2023] [Accepted: 03/21/2023] [Indexed: 04/06/2023] Open
Abstract
Colorectal cancer (CRC) is the most common digestive malignancy across the world. Its first-line treatments applied in the routine clinical setting include surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy. However, resistance to therapy has been identified as the major clinical challenge that fails the treatment method, leading to recurrence and distant metastasis. An increasing number of studies have been attempting to explore the underlying mechanisms of the resistance of CRC cells to different therapies, which can be summarized into two aspects: (1) The intrinsic characters and adapted alterations of CRC cells before and during treatment that regulate the drug metabolism, drug transport, drug target, and the activation of signaling pathways; and (2) the suppressive features of the tumor microenvironment (TME). To combat the issue of therapeutic resistance, effective strategies are warranted with a focus on the restoration of CRC cells’ sensitivity to specific treatments as well as reprogramming impressive TME into stimulatory conditions. To date, nanotechnology seems promising with scope for improvement of drug mobility, treatment efficacy, and reduction of systemic toxicity. The instinctive advantages offered by nanomaterials enable the diversity of loading cargoes to increase drug concentration and targeting specificity, as well as offer a platform for trying the combination of different treatments to eventually prevent tumor recurrence, metastasis, and reversion of therapy resistance. The present review intends to summarize the known mechanisms of CRC resistance to chemotherapy, radiotherapy, immunotherapy, and targeted therapy, as well as the process of metastasis. We have also emphasized the recent application of nanomaterials in combating therapeutic resistance and preventing metastasis either by combining with other treatment approaches or alone. In summary, nanomedicine is an emerging technology with potential for CRC treatment; hence, efforts should be devoted to targeting cancer cells for the restoration of therapeutic sensitivity as well as reprogramming the TME. It is believed that the combined strategy will be beneficial to achieve synergistic outcomes contributing to control and management of CRC in the future.
Collapse
Affiliation(s)
- Sheng-Nan Ren
- Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Zhan-Yi Zhang
- Bethune Third Clinical Medical College, Jilin University, Changchun 130021, Jilin Province, China
| | - Rui-Jie Guo
- Bethune Third Clinical Medical College, Jilin University, Changchun 130021, Jilin Province, China
| | - Da-Ren Wang
- Bethune Third Clinical Medical College, Jilin University, Changchun 130021, Jilin Province, China
| | - Fang-Fang Chen
- Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Xue-Bo Chen
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Xue-Dong Fang
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
4
|
García-Domínguez DJ, Sánchez-Margalet V, de la Cruz-Merino L, Hontecillas-Prieto L. Knowing the myeloid-derived suppressor cells : Another enemy of sarcomas patients. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 375:93-116. [PMID: 36967155 DOI: 10.1016/bs.ircmb.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Sarcomas are heterogeneous and aggressive malignant tumors with variable responses to current standard treatments being usually incurable for those patients with metastatic and unresectable diseases. The lack of curative strategies has led to develop new therapies in the treatment of sarcomas where the role of immune system is an evolving field. Most sarcomas often exhibit an immunosuppressive microenvironment, which reduces their capacity to trigger an immune response. Therefore, sarcomas are broadly considered as an "immune cold" tumor, although some studies have described a great immune heterogeneity across sarcoma subtypes. Sarcoma cells, like other tumors, evade their immune destruction through a variety of mechanisms, including expansion and recruitment of myeloid derived suppressor cells (MDSCs). MDSCs are immature myeloid cells that have been correlated with a reduction of the therapeutic efficacy, including immunotherapy, tumor progression and worst prognosis. Consequently, different strategies have been developed in recent years to target MDSCs in cancer treatments. This chapter discusses the role of MDSCs in sarcomas and their current potential as a therapeutic target in these malignancies.
Collapse
|
5
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
6
|
Sun H, Liu J, Hu F, Xu M, Leng A, Jiang F, Chen K. Current research and management of undifferentiated pleomorphic sarcoma/myofibrosarcoma. Front Genet 2023; 14:1109491. [PMID: 36873946 PMCID: PMC9978151 DOI: 10.3389/fgene.2023.1109491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Undifferentiated pleomorphic sarcoma (UPS), once termed as malignant fibrous histiocytoma, has always been diagnosed exclusively in clinical practice because it lacks any defined resemblance to normal mesenchymal tissue. Although myxofibrosarcoma (MFS) has been separated from UPS due to its fibroblastic differentiation with myxoid stroma, UPS and MFS are still identified as a sarcoma group in terms of molecular landscapes. In this review article, we will describe the associated genes and signaling pathways involved in the process of sarcoma genesis and make a summary of conventional management, targeted therapy, immunotherapy, and some novel potential treatments of UPS/MFS. With the progressive advancements in medical technology and a better understanding about the pathogenic mechanism of UPS/MFS in the coming decades, new lights will be shed on the successful management of UPS/MFS.
Collapse
Affiliation(s)
- Haitao Sun
- Department of Spine Surgery, Naval Hospital of Eastern Theater Command, Zhoushan, China
| | - Jilu Liu
- Department of Spine Surgery, Naval Hospital of Eastern Theater Command, Zhoushan, China
| | - Fangyuan Hu
- Department of Spine Surgery, Naval Hospital of Eastern Theater Command, Zhoushan, China
| | - Meng Xu
- Department of Spine Surgery, Naval Hospital of Eastern Theater Command, Zhoushan, China
| | - Ao Leng
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Kefu Chen
- The No.988th hospital of Joint Logistic Support Force of PLA, Zhengzhou, China.,Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, China
| |
Collapse
|
7
|
Som A, Rosenboom JG, Chandler A, Sheth RA, Wehrenberg-Klee E. Image-guided intratumoral immunotherapy: Developing a clinically practical technology. Adv Drug Deliv Rev 2022; 189:114505. [PMID: 36007674 PMCID: PMC10456124 DOI: 10.1016/j.addr.2022.114505] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 07/14/2022] [Accepted: 08/17/2022] [Indexed: 02/07/2023]
Abstract
Immunotherapy has revolutionized the contemporary oncology landscape, with durable responses possible across a range of cancer types. However, the majority of cancer patients do not respond to immunotherapy due to numerous immunosuppressive barriers. Efforts to overcome these barriers and increase systemic immunotherapy efficacy have sparked interest in the local intratumoral delivery of immune stimulants to activate the local immune response and subsequently drive systemic tumor immunity. While clinical evaluation of many therapeutic candidates is ongoing, development is hindered by a lack of imaging confirmation of local delivery, insufficient intratumoral drug distribution, and a need for repeated injections. The use of polymeric drug delivery systems, which have been widely used as platforms for both image guidance and controlled drug release, holds promise for delivery of intratumoral immunoadjuvants and the development of an in situ cancer vaccine for patients with metastatic cancer. In this review, we explore the current state of the field for intratumoral delivery and methods for optimizing controlled drug release, as well as practical considerations for drug delivery design to be optimized for clinical image guided delivery particularly by CT and ultrasound.
Collapse
Affiliation(s)
- Avik Som
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, United States; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, United States
| | - Jan-Georg Rosenboom
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, United States; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, United States; Department of Gastroenterology, Brigham and Women's Hospital, United States
| | - Alana Chandler
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, United States; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, United States; Department of Gastroenterology, Brigham and Women's Hospital, United States
| | - Rahul A Sheth
- Department of Interventional Radiology, M.D. Anderson Cancer Center, United States
| | - Eric Wehrenberg-Klee
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, United States.
| |
Collapse
|
8
|
Chan Wah Hak CML, Rullan A, Patin EC, Pedersen M, Melcher AA, Harrington KJ. Enhancing anti-tumour innate immunity by targeting the DNA damage response and pattern recognition receptors in combination with radiotherapy. Front Oncol 2022; 12:971959. [PMID: 36106115 PMCID: PMC9465159 DOI: 10.3389/fonc.2022.971959] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Radiotherapy is one of the most effective and frequently used treatments for a wide range of cancers. In addition to its direct anti-cancer cytotoxic effects, ionising radiation can augment the anti-tumour immune response by triggering pro-inflammatory signals, DNA damage-induced immunogenic cell death and innate immune activation. Anti-tumour innate immunity can result from recruitment and stimulation of dendritic cells (DCs) which leads to tumour-specific adaptive T-cell priming and immunostimulatory cell infiltration. Conversely, radiotherapy can also induce immunosuppressive and anti-inflammatory mediators that can confer radioresistance. Targeting the DNA damage response (DDR) concomitantly with radiotherapy is an attractive strategy for overcoming radioresistance, both by enhancing the radiosensitivity of tumour relative to normal tissues, and tipping the scales in favour of an immunostimulatory tumour microenvironment. This two-pronged approach exploits genomic instability to circumvent immune evasion, targeting both hallmarks of cancer. In this review, we describe targetable DDR proteins (PARP (poly[ADP-ribose] polymerase); ATM/ATR (ataxia-telangiectasia mutated and Rad3-related), DNA-PKcs (DNA-dependent protein kinase, catalytic subunit) and Wee1 (Wee1-like protein kinase) and their potential intersections with druggable immunomodulatory signalling pathways, including nucleic acid-sensing mechanisms (Toll-like receptors (TLR); cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) and retinoic acid-inducible gene-I (RIG-I)-like receptors), and how these might be exploited to enhance radiation therapy. We summarise current preclinical advances, recent and ongoing clinical trials and the challenges of therapeutic combinations with existing treatments such as immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Antonio Rullan
- Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Emmanuel C. Patin
- Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Malin Pedersen
- Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Alan A. Melcher
- Translational Immunotherapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Kevin J. Harrington
- Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
9
|
Zhang Y, Xu Z, Chen H, Sun X, Zhang Z. Survival comparison between postoperative and preoperative radiotherapy for stage I-III non-inflammatory breast cancer. Sci Rep 2022; 12:14288. [PMID: 35995985 PMCID: PMC9395522 DOI: 10.1038/s41598-022-18251-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 08/08/2022] [Indexed: 11/09/2022] Open
Abstract
To compare the survival benefit between preoperative and postoperative radiotherapy for stage I-III non-inflammatory breast cancer patients, we conducted a retrospective cohort study using surveillance, epidemiology and end results databases. Our study recruited patients who had been diagnosed with stage I-III breast cancer and underwent surgery and radiotherapy. The overall survival was calculated by Kaplan-Meier method. Cox risk model was used to determine the impact of radiotherapy according to stage, molecular subtype and other risk factors. Propensity score matching was used to balance measurable confounding factors. Of all the 411,279 enrolled patients varying from 1975 to 2016, 1712 patients received preoperative radiotherapy, and 409,567 patients received postoperative radiotherapy. Compared with the postoperative radiotherapy group, the preoperative radiotherapy group showed significantly higher risks of overall mortality and breast cancer-specific mortality. Survival differences in treatment sequences were correlated with stage, molecular subtypes and other risk factors. According to the results of this study, preoperative radiotherapy did not show a survival advantage, and postoperative radiotherapy is still the primary treatment. However, preoperative radiotherapy also has some theoretical advantages, such as phase reduction and recurrence reduction. Therefore, it is still worthy of further exploration.
Collapse
Affiliation(s)
- Yuxi Zhang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Zhipeng Xu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu, China
| | - Hui Chen
- Department of Radiation Oncology, Jiangsu Province Hospital, Nanjing, China
| | - Xinchen Sun
- Department of Radiation Oncology, Jiangsu Province Hospital, Nanjing, China.
| | - Zhaoyue Zhang
- Department of Radiation Oncology, Jiangsu Province Hospital, Nanjing, China.
| |
Collapse
|
10
|
Baldi GG, Gronchi A, Tazzari M, Stacchiotti S. Immunotherapy in soft tissue sarcoma: current evidence and future perspectives in a variegated family of different tumour. Expert Rev Anticancer Ther 2022; 22:491-503. [PMID: 35412415 DOI: 10.1080/14737140.2022.2065986] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION In the last few years steps forward in the knowledge of the biology of soft tissue sarcomas (STS) has led to the development of new therapeutic strategies, including immunotherapy. AREAS COVERED This review outlines the recent findings on immunological features and provides a synopsis of the results of clinical trials with different immunotherapy approaches in STS, discussing criticisms and how the efficacy of immunotherapy could be improved. EXPERT OPINION The heterogeneity of STS has limited generalized approaches of immunotherapy in the disease. Clinical decisions should encompass a comprehensive characterization of the tumour microenvironment (TME), marked by intra-histotype diversity. Profiling of immune cells, checkpoint molecules and antigen target/HLA expression is deemed to re-shape the classical histotype classification for a selection of the most appropriate immune-based treatment. In a synergistic view, tumour-directed treatments, designed on the genetic and epigenetic histotype make-up, should be monitored for their immunomodulant effect and applied to ensure or amplify immunotherapy response. In light of the dynamic nature of the TME, this immunomonitoring should be conducted at baseline and during treatment, for improved therapeutic decisions and rational sequence of treatment combination, pursuing an immunological marker approach by histotype guidance.
Collapse
Affiliation(s)
- Giacomo G Baldi
- Department of Medical Oncology, Hospital of Prato, Prato, Italy
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marcella Tazzari
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Silvia Stacchiotti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
11
|
Milic M, Mondini M, Deutsch E. How to Improve SBRT Outcomes in NSCLC: From Pre-Clinical Modeling to Successful Clinical Translation. Cancers (Basel) 2022; 14:cancers14071705. [PMID: 35406477 PMCID: PMC8997119 DOI: 10.3390/cancers14071705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Despite major research and clinical efforts, lung cancer remains the leading cause of cancer-related death. Stereotactic body radiotherapy (SBRT) has emerged as a major treatment modality for lung cancer in the last decade. Additional research is needed to elucidate underlying mechanisms of resistance and to develop improved therapeutic strategies. Clinical progress relies on accurate preclinical modelling of human disease in order to yield clinically meaningful results; however, successful translation of pre-clinical research is still lagging behind. In this review, we summarize the major clinical developments of radiation therapy for non-small-cell lung cancer (NSCLC), and we discuss the pre-clinical research models at our disposal, highlighting ongoing translational challenges and future perspectives. Abstract Despite major research and clinical efforts, lung cancer remains the leading cause of cancer-related death. While the delivery of conformal radiotherapy and image guidance of stereotactic body radiotherapy (SBRT) have revolutionized the treatment of early-stage non-small-cell lung cancer (NSCLC), additional research is needed to elucidate underlying mechanisms of resistance and identify novel therapeutic combinations. Clinical progress relies on the successful translation of pre-clinical work, which so far has not always yielded expected results. Improved clinical modelling involves characterizing the preclinical models and selecting appropriate experimental designs that faithfully mimic precise clinical scenarios. Here, we review the current role of SBRT and the scope of pre-clinical armamentarium at our disposal to improve successful clinical translation of pre-clinical research in the radiation oncology of NSCLC.
Collapse
Affiliation(s)
- Marina Milic
- Gustave Roussy, Université Paris-Saclay, INSERM U1030, F-94805 Villejuif, France;
| | - Michele Mondini
- Gustave Roussy, Université Paris-Saclay, INSERM U1030, F-94805 Villejuif, France;
- Correspondence: (M.M.); (E.D.)
| | - Eric Deutsch
- Gustave Roussy, Université Paris-Saclay, INSERM U1030, F-94805 Villejuif, France;
- Gustave Roussy, Département d’Oncologie-Radiothérapie, F-94805 Villejuif, France
- Correspondence: (M.M.); (E.D.)
| |
Collapse
|
12
|
Qiu X, He H, Zeng H, Tong X, Liu Q. The Role of Radiotherapy in Soft Tissue Sarcoma on Extremities With Lymph Nodes Metastasis: An IPTW Propensity Score Analysis of the SEER Database. Front Oncol 2021; 11:751180. [PMID: 34745979 PMCID: PMC8566919 DOI: 10.3389/fonc.2021.751180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Background Soft tissue sarcomas on extremities with regional lymph nodes metastasis (STSE-RLNM) is a devastating situation. Optimizing therapeutic approaches is vital but hampered by a shortage of randomized trials. We used a population-level database to evaluate radiotherapy’s impact on sarcoma-specific survival (SSS) and overall survival (OS) for surgery for STSE-RLNM. Methods We retrospectively screened data from the SEER database (2004–2015), and 265 patients with STSE-RLNM who received surgery, with (134) or without (131) radiotherapy, were enrolled in this study. A propensity-score-matched analysis with the inverse probability of treatment weighting (IPTW) Kaplan–Meier curve was created. The log-rank test and Cox regression analysis were performed to compare SSS and OS in patients with and without radiotherapy. Further analysis of radiotherapy time was conducted, and the Kaplan–Meier curve and the log-rank test were done. Landmark analysis was introduced to attenuate the immortal bias. Results In the original unadjusted cohort, the radiotherapy + surgery group is associated with improved SSS [hazard ratio (HR), 0.66; 95% CI, 0.47–0.91; p = 0.011] and OS (HR, 0.64; 95% CI, 0.47–0.88; p = 0.006). This significant treatment effect was also noted in IPTW-adjusted Cox regression either on SSS (HR, 0.65; 95% CI, 0.45–0.93; p = 0.020) or on OS (HR, 0.64; 95% CI, 0.46–0.91; p = 0.013). The Kaplan–Meier curve and log-rank test showed that pre- and postoperative radiotherapy was not related to SSS (p = 0.980 or OS (p = 0.890). Conclusion Radiotherapy and surgery has a significant benefit on the prognosis of patients with STSE-RLNM compared to surgery alone. These findings should be considered when making treatment decisions for them.
Collapse
Affiliation(s)
- Xinzhu Qiu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Hongbo He
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Hao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaopeng Tong
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Qing Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Lin YJ, Wei KC, Chen PY, Lim M, Hwang TL. Roles of Neutrophils in Glioma and Brain Metastases. Front Immunol 2021; 12:701383. [PMID: 34484197 PMCID: PMC8411705 DOI: 10.3389/fimmu.2021.701383] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/19/2021] [Indexed: 12/16/2022] Open
Abstract
Neutrophils, which are the most abundant circulating leukocytes in humans, are the first line of defense against bacterial and fungal infections. Recent studies have reported the role and importance of neutrophils in cancers. Glioma and brain metastases are the most common malignant tumors of the brain. The tumor microenvironment (TME) in the brain is complex and unique owing to the brain-blood barrier or brain-tumor barrier, which may prevent drug penetration and decrease the efficacy of immunotherapy. However, there are limited studies on the correlation between brain cancer and neutrophils. This review discusses the origin and functions of neutrophils. Additionally, the current knowledge on the correlation between neutrophil-to-lymphocyte ratio and prognosis of glioma and brain metastases has been summarized. Furthermore, the implications of tumor-associated neutrophil (TAN) phenotypes and the functions of TANs have been discussed. Finally, the potential effects of various treatments on TANs and the ability of neutrophils to function as a nanocarrier of drugs to the brain TME have been summarized. However, further studies are needed to elucidate the complex interactions between neutrophils, other immune cells, and brain tumor cells.
Collapse
Affiliation(s)
- Ya-Jui Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Taiwan
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, Chang Gung Medical Foundation, New Taipei, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Pin-Yuan Chen
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Michael Lim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| |
Collapse
|
14
|
Darmusey L, Pérot G, Thébault N, Le Guellec S, Desplat N, Gaston L, Delespaul L, Lesluyes T, Darbo E, Gomez-Brouchet A, Richard E, Baud J, Leroy L, Coindre JM, Blay JY, Chibon F. ATRX Alteration Contributes to Tumor Growth and Immune Escape in Pleomorphic Sarcomas. Cancers (Basel) 2021; 13:2151. [PMID: 33946962 PMCID: PMC8124877 DOI: 10.3390/cancers13092151] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/14/2021] [Accepted: 04/21/2021] [Indexed: 12/17/2022] Open
Abstract
Whole genome and transcriptome sequencing of a cohort of 67 leiomyosarcomas has been revealed ATRX to be one of the most frequently mutated genes in leiomyosarcomas after TP53 and RB1. While its function is well described in the alternative lengthening of telomeres mechanism, we wondered whether its alteration could have complementary effects on sarcoma oncogenesis. ATRX alteration is associated with the down-expression of genes linked to differentiation in leiomyosarcomas, and to immunity in an additional cohort of 60 poorly differentiated pleomorphic sarcomas. In vitro and in vivo models showed that ATRX down-expression increases tumor growth rate and immune escape by decreasing the immunity load of active mast cells in sarcoma tumors. These data indicate that an alternative to unsuccessful targeting of the adaptive immune system in sarcoma could target the innate system. This might lead to a better outcome for sarcoma patients in terms of ATRX status.
Collapse
Affiliation(s)
- Lucie Darmusey
- INSERM U1037, Cancer Research Center in Toulouse (CRCT), OncoSarc, 31000 Toulouse, France; (L.D.); (G.P.); (N.T.); (S.L.G.); (L.D.); (T.L.); (A.G.-B.); (L.L.)
- IUCT-Oncopole, Institut Claudius Régaud, Department of Pathology, 31000 Toulouse, France
- University of Toulouse 3, Paul Sabatier, 31000 Toulouse, France
| | - Gaëlle Pérot
- INSERM U1037, Cancer Research Center in Toulouse (CRCT), OncoSarc, 31000 Toulouse, France; (L.D.); (G.P.); (N.T.); (S.L.G.); (L.D.); (T.L.); (A.G.-B.); (L.L.)
- Centre Hospitalier Universitaire (CHU) de Toulouse, IUCT-Oncopole, 31000 Toulouse, France
| | - Noémie Thébault
- INSERM U1037, Cancer Research Center in Toulouse (CRCT), OncoSarc, 31000 Toulouse, France; (L.D.); (G.P.); (N.T.); (S.L.G.); (L.D.); (T.L.); (A.G.-B.); (L.L.)
- IUCT-Oncopole, Institut Claudius Régaud, Department of Pathology, 31000 Toulouse, France
| | - Sophie Le Guellec
- INSERM U1037, Cancer Research Center in Toulouse (CRCT), OncoSarc, 31000 Toulouse, France; (L.D.); (G.P.); (N.T.); (S.L.G.); (L.D.); (T.L.); (A.G.-B.); (L.L.)
- IUCT-Oncopole, Institut Claudius Régaud, Department of Pathology, 31000 Toulouse, France
| | - Nelly Desplat
- Inserm UMR1218, Action, Institut Bergonié, 33000 Bordeaux, France; (N.D.); (E.D.); (E.R.); (J.B.); (J.-M.C.)
| | - Laëtitia Gaston
- CHU de Bordeaux, Department of Medical Genetics, 33000 Bordeaux, France;
| | - Lucile Delespaul
- INSERM U1037, Cancer Research Center in Toulouse (CRCT), OncoSarc, 31000 Toulouse, France; (L.D.); (G.P.); (N.T.); (S.L.G.); (L.D.); (T.L.); (A.G.-B.); (L.L.)
- University of Bordeaux, 33000 Bordeaux, France
| | - Tom Lesluyes
- INSERM U1037, Cancer Research Center in Toulouse (CRCT), OncoSarc, 31000 Toulouse, France; (L.D.); (G.P.); (N.T.); (S.L.G.); (L.D.); (T.L.); (A.G.-B.); (L.L.)
- University of Bordeaux, 33000 Bordeaux, France
| | - Elodie Darbo
- Inserm UMR1218, Action, Institut Bergonié, 33000 Bordeaux, France; (N.D.); (E.D.); (E.R.); (J.B.); (J.-M.C.)
- University of Bordeaux, 33000 Bordeaux, France
- CNRS UMR5800, LaBRI, 33400 Talence, France
| | - Anne Gomez-Brouchet
- INSERM U1037, Cancer Research Center in Toulouse (CRCT), OncoSarc, 31000 Toulouse, France; (L.D.); (G.P.); (N.T.); (S.L.G.); (L.D.); (T.L.); (A.G.-B.); (L.L.)
- IUCT-Oncopole, Institut Claudius Régaud, Department of Pathology, 31000 Toulouse, France
- Centre Hospitalier Universitaire (CHU) de Toulouse, IUCT-Oncopole, 31000 Toulouse, France
| | - Elodie Richard
- Inserm UMR1218, Action, Institut Bergonié, 33000 Bordeaux, France; (N.D.); (E.D.); (E.R.); (J.B.); (J.-M.C.)
| | - Jessica Baud
- Inserm UMR1218, Action, Institut Bergonié, 33000 Bordeaux, France; (N.D.); (E.D.); (E.R.); (J.B.); (J.-M.C.)
| | - Laura Leroy
- INSERM U1037, Cancer Research Center in Toulouse (CRCT), OncoSarc, 31000 Toulouse, France; (L.D.); (G.P.); (N.T.); (S.L.G.); (L.D.); (T.L.); (A.G.-B.); (L.L.)
- IUCT-Oncopole, Institut Claudius Régaud, Department of Pathology, 31000 Toulouse, France
| | - Jean-Michel Coindre
- Inserm UMR1218, Action, Institut Bergonié, 33000 Bordeaux, France; (N.D.); (E.D.); (E.R.); (J.B.); (J.-M.C.)
- Institut Bergonie, Department of Pathology, 33000 Bordeaux, France
| | - Jean-Yves Blay
- Centre Léon Bérard, Department of Medical Oncology, 69000 Lyon, France;
- Inserm U1052, CNRS 5286, Cancer Research Center of Lyon, University Claude Bernard Lyon 1, 69000 Lyon, France
| | - Frédéric Chibon
- INSERM U1037, Cancer Research Center in Toulouse (CRCT), OncoSarc, 31000 Toulouse, France; (L.D.); (G.P.); (N.T.); (S.L.G.); (L.D.); (T.L.); (A.G.-B.); (L.L.)
- IUCT-Oncopole, Institut Claudius Régaud, Department of Pathology, 31000 Toulouse, France
- Inserm UMR1218, Action, Institut Bergonié, 33000 Bordeaux, France; (N.D.); (E.D.); (E.R.); (J.B.); (J.-M.C.)
| |
Collapse
|
15
|
The Lymph-Sparing Quotient: A Retrospective Risk Analysis on Extremity Radiation for Soft Tissue Sarcoma Treatment. Cancers (Basel) 2021; 13:cancers13092113. [PMID: 33925683 PMCID: PMC8125651 DOI: 10.3390/cancers13092113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Soft tissue sarcomas, a heterogenous group of tumors with a mesenchymal origin, are mostly located in the extremities and are commonly treated with surgery and radiotherapy. Using opportunities of reducing long-term therapy-related side effects in soft tissue sarcoma treatment is an important task for all physicians involved in soft tissue sarcoma treatment. The extent of lymph-sparing volume in adjuvant radiation therapy of extremity soft tissue sarcoma as a risk factor for lymphedema was analyzed in this study. Patients with a low lymph-sparing volume showed an increased risk of lymphedema in this retrospective study. Maximizing the potential oncologically justifiable lymph-sparing volume should be considered to reduce the risk of high-grade lymphedema when applying RT to extremities. Abstract Radiation therapy (RT) for extremity soft tissue sarcoma is associated with lymphedema risk. In this study, we analyzed the influence of lymph-sparing volume on the lymphedema occurrence in patients who received adjuvant extremity RT. The lymph-sparing quotient (LSQ) was calculated by dividing the lymph-sparing volume by the total extremity volume with double weightingfor the narrowest lymph-sparing region. A total of 34 patients were enrolled in this analysis. The median applied total radiation dose was 66.3 Gy in 36 fractions. Acute lymphedema appeared in 12 patients (35%). Most of them (n = 8) were lymphedema grade 1 and five patients had grade 2 to 3 lymphedema. Chronic lymphedema appeared in 22 patients (65%). 17 of these patients had at least a grade 2 lymphedema. In 13 of 14 patients with an LSQ ≤ 0.2 and 11 of 20 patients with an LSQ > 0.2, an acute or chronic lymphedema ≥ grade 2 was observed. A Kaplan–Meier Analysis of the two groups with the endpoint of a two-year lymph edema-free survival (=2-YLEFS) was estimated with an univariate, significant result (2-YLEFS LSQ ≤ 0.2 vs. LSQ > 0.2: 0% vs. 39%; p = 0.006; hazard ratio LSQ ≤ 0.2 vs. > 0.2 2-YLEFS 2.822 (p = 0.013); 95% confidence interval (CI): 1.24–6.42). Maximizing the potential oncologically-justifiable lymph-sparing volume should be considered to reduce the risk of high-grade lymphedema when applying RT to extremities.
Collapse
|
16
|
Marcus D, Lieverse RIY, Klein C, Abdollahi A, Lambin P, Dubois LJ, Yaromina A. Charged Particle and Conventional Radiotherapy: Current Implications as Partner for Immunotherapy. Cancers (Basel) 2021; 13:1468. [PMID: 33806808 PMCID: PMC8005048 DOI: 10.3390/cancers13061468] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy (RT) has been shown to interfere with inflammatory signals and to enhance tumor immunogenicity via, e.g., immunogenic cell death, thereby potentially augmenting the therapeutic efficacy of immunotherapy. Conventional RT consists predominantly of high energy photon beams. Hypofractionated RT regimens administered, e.g., by stereotactic body radiation therapy (SBRT), are increasingly investigated in combination with cancer immunotherapy within clinical trials. Despite intensive preclinical studies, the optimal dose per fraction and dose schemes for elaboration of RT induced immunogenic potential remain inconclusive. Compared to the scenario of combined immune checkpoint inhibition (ICI) and RT, multimodal therapies utilizing other immunotherapy principles such as adoptive transfer of immune cells, vaccination strategies, targeted immune-cytokines and agonists are underrepresented in both preclinical and clinical settings. Despite the clinical success of ICI and RT combination, e.g., prolonging overall survival in locally advanced lung cancer, curative outcomes are still not achieved for most cancer entities studied. Charged particle RT (PRT) has gained interest as it may enhance tumor immunogenicity compared to conventional RT due to its unique biological and physical properties. However, whether PRT in combination with immune therapy will elicit superior antitumor effects both locally and systemically needs to be further investigated. In this review, the immunological effects of RT in the tumor microenvironment are summarized to understand their implications for immunotherapy combinations. Attention will be given to the various immunotherapeutic interventions that have been co-administered with RT so far. Furthermore, the theoretical basis and first evidences supporting a favorable immunogenicity profile of PRT will be examined.
Collapse
Affiliation(s)
- Damiënne Marcus
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Relinde I. Y. Lieverse
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Carmen Klein
- German Cancer Consortium (DKTK) Core-Center Heidelberg, National Center for Tumor Diseases (NCT), Clinical Cooperation Unit Translational Radiation Oncology, Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120 Heidelberg, Germany; (C.K.); (A.A.)
- Heidelberg Ion-Beam Therapy Center (HIT), Division of Molecular and Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
- National Center for Radiation Oncology (NCRO), Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 222, 69120 Heidelberg, Germany
| | - Amir Abdollahi
- German Cancer Consortium (DKTK) Core-Center Heidelberg, National Center for Tumor Diseases (NCT), Clinical Cooperation Unit Translational Radiation Oncology, Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120 Heidelberg, Germany; (C.K.); (A.A.)
- Heidelberg Ion-Beam Therapy Center (HIT), Division of Molecular and Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
- National Center for Radiation Oncology (NCRO), Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 222, 69120 Heidelberg, Germany
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Ludwig J. Dubois
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| |
Collapse
|
17
|
Desai R, Coxon AT, Dunn GP. Therapeutic applications of the cancer immunoediting hypothesis. Semin Cancer Biol 2021; 78:63-77. [PMID: 33711414 DOI: 10.1016/j.semcancer.2021.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/15/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022]
Abstract
Since the late 19th century, the immune system has increasingly garnered interest as a novel avenue for cancer therapy, particularly given scientific breakthroughs in recent decades delineating the fundamental role of the immune system in tumorigenesis. The immunoediting hypothesis has articulated this role, describing three phases of the tumor-immune system interaction: Elimination, Equilibrium, and Escape wherein tumors progress from active immunologic surveillance and destruction through dynamic immunologic stasis to unfettered growth. The primary goals of immunotherapy are to restrict and revert progression through these phases, thereby improving the immune system's ability to control tumor growth. In this review, we detail the development and foundation of the cancer immunoediting hypothesis and apply this hypothesis to the dynamic immunotherapy field that includes checkpoint blockade, vaccine therapy, and adoptive cell transfer.
Collapse
Affiliation(s)
- Rupen Desai
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew T Coxon
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Gavin P Dunn
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
18
|
Stevens D, Ingels J, Van Lint S, Vandekerckhove B, Vermaelen K. Dendritic Cell-Based Immunotherapy in Lung Cancer. Front Immunol 2021; 11:620374. [PMID: 33679709 PMCID: PMC7928408 DOI: 10.3389/fimmu.2020.620374] [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: 10/22/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer remains the leading cause of cancer-related death worldwide. The advent of immune checkpoint inhibitors has led to a paradigm shift in the treatment of metastatic non-small cell and small cell lung cancer. However, despite prolonged overall survival, only a minority of the patients derive clinical benefit from these treatments suggesting that the full anti-tumoral potential of the immune system is not being harnessed yet. One way to overcome this problem is to combine immune checkpoint blockade with different strategies aimed at inducing or restoring cellular immunity in a tumor-specific, robust, and durable way. Owing to their unique capacity to initiate and regulate T cell responses, dendritic cells have been extensively explored as tools for immunotherapy in many tumors, including lung cancer. In this review, we provide an update on the nearly twenty years of experience with dendritic cell-based immunotherapy in lung cancer. We summarize the main results from the early phase trials and give an overview of the future perspectives within this field.
Collapse
Affiliation(s)
- Dieter Stevens
- Respiratory Medicine - Thoracic Oncology Cluster, Ghent University Hospital, Ghent, Belgium.,Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium
| | - Joline Ingels
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium
| | - Sandra Van Lint
- Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Bart Vandekerckhove
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium.,GMP Cell Therapy Unit, Department of Regenerative Medicine, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Karim Vermaelen
- Respiratory Medicine - Thoracic Oncology Cluster, Ghent University Hospital, Ghent, Belgium.,Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| |
Collapse
|
19
|
The Sarcoma Immune Landscape: Emerging Challenges, Prognostic Significance and Prospective Impact for Immunotherapy Approaches. Cancers (Basel) 2021; 13:cancers13030363. [PMID: 33498238 PMCID: PMC7863949 DOI: 10.3390/cancers13030363] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/19/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Sarcomas are a rare disease with high rates of recurrence and poor prognosis. Important discoveries about the biology of sarcomas have been done during the last decades, without a substantial improvement of systemic treatments. With the agnostic effectivity of immuno-oncological agents in different cancer indications, it is expected that sarcomas can also benefit from these treatments. This article gathers the available data on the specific immune tumor microenvironment of sarcoma and the immunotherapeutic strategies currently under investigation. Abstract Despite significant advances in multidisciplinary treatment strategies including surgery, radiotherapy, targeted therapy and chemotherapy there are yet no substantial improvements in the clinical benefit of patients with sarcomas. Current understanding of the underlying cellular and molecular pathways which govern the dynamic interactions between the tumor stroma, tumor cells and immune infiltrates in sarcoma tissues, led to the clinical development of new therapeutic options based on immunotherapies. Moreover, progress of the treatment of sarcomas also depends on the identification of biomarkers with prognostic and predictive values for selecting patients most likely to benefit from these new therapeutic treatments and also serving as potent therapeutic targets. Novel combinations with radiotherapy, chemotherapy, targeted therapy, vaccines, CAR-T cells and treatments targeting other immune components of the tumor microenvironment are underway aiming to bypass known resistance mechanisms. This review focuses on the role of tumor microenvironment in sarcoma, prognosis and response to novel immunotherapies.
Collapse
|
20
|
Colton M, Cheadle EJ, Honeychurch J, Illidge TM. Reprogramming the tumour microenvironment by radiotherapy: implications for radiotherapy and immunotherapy combinations. Radiat Oncol 2020; 15:254. [PMID: 33148287 PMCID: PMC7640712 DOI: 10.1186/s13014-020-01678-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Radiotherapy (RT) is a highly effective anti-cancer therapy delivered to around 50-60% of patients. It is part of therapy for around 40% of cancer patients who are cured of their disease. Until recently, the focus of this anti-tumour efficacy has been on the direct tumour cytotoxicity and RT-induced DNA damage. Recently, the immunomodulatory effects of RT on the tumour microenvironment have increasingly been recognized. There is now intense interest in potentially using RT to induce an anti-tumour immune response, which has led to rethinking into how the efficacy of RT could be further enhanced. Following the breakthrough of immune check point inhibitors (ICIs), a new era of immuno-oncology (IO) agents has emerged and established immunotherapy as a routine part of cancer treatment. Despite ICI improving outcomes in many cancer types, overall durable responses occur in only a minority of patients. The immunostimulatory effects of RT make combinations with ICI attractive to potentially amplify anti-tumour immunity resulting in increased tumour responses and improved outcomes. In contrast, tumours with profoundly immunosuppressive tumour microenvironments, dominated by myeloid-derived cell populations, remain a greater clinical challenge and RT may potentially further enhance the immunosuppression. To harness the full potential of RT and IO agent combinations, further insights are required to enhance our understanding of the role these immunosuppressive myeloid populations play, how RT influences these populations and how they may be therapeutically manipulated in combination with RT to improve outcomes further. These are exciting times with increasing numbers of IO targets being discovered and IO agents undergoing clinical evaluation. Multidisciplinary research collaborations will be required to establish the optimal parameters for delivering RT (target volume, dose and fractionation) in combination with IO agents, including scheduling to achieve maximal therapeutic efficacy.
Collapse
Affiliation(s)
- Madyson Colton
- Division of Cancer Sciences, Manchester Academic Health Science Centre, NIHR Biomedical Research Centre, University of Manchester, Manchester, UK
| | - Eleanor J Cheadle
- Division of Cancer Sciences, Manchester Academic Health Science Centre, NIHR Biomedical Research Centre, University of Manchester, Manchester, UK
| | - Jamie Honeychurch
- Division of Cancer Sciences, Manchester Academic Health Science Centre, NIHR Biomedical Research Centre, University of Manchester, Manchester, UK
| | - Tim M Illidge
- Division of Cancer Sciences, Manchester Academic Health Science Centre, NIHR Biomedical Research Centre, University of Manchester, Manchester, UK.
- The Christie NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
21
|
Yuan S, Liu Y, Till B, Song Y, Wang Z. Pretreatment Peripheral B Cells Are Associated With Tumor Response to Anti-PD-1-Based Immunotherapy. Front Immunol 2020; 11:563653. [PMID: 33162976 PMCID: PMC7584071 DOI: 10.3389/fimmu.2020.563653] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/13/2020] [Indexed: 12/26/2022] Open
Abstract
Identification of reliable biomarkers to predict efficacy of immune checkpoint inhibitors and to monitor relapse in cancer patients receiving this therapy remains one of the main objectives of cancer immunotherapy research. We found that the pretreatment B cell number in the peripheral blood differed significantly between responders and non-responders to anti-PD-1-based immunotherapy. Patients with various cancer types achieving a clinical response had a significantly lower number of B cells compared with those with progressive disease. Patients who progressed from partial response to progressive disease exhibited a gradually increased number of circulating B cells. Our findings suggest that B cells represent a promising biomarker for anti-PD-1-based immunotherapy responses and inhibit the effect of PD-1 blockade immunotherapy. Thus, preemptive strategies targeting B cells may increase the efficacy of PD-1 blockade immunotherapy in patients with solid tumors.
Collapse
Affiliation(s)
- Shumin Yuan
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yuqing Liu
- Third Affiliated Hospital of Xinxiang Medical College, Xinxiang, China
| | - Brian Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Yongping Song
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Zibing Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
22
|
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: 0.6] [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
|
23
|
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: 112] [Impact Index Per Article: 22.4] [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
|
24
|
Morrissey ME, Byrne R, Nulty C, McCabe NH, Lynam-Lennon N, Butler CT, Kennedy S, O'Toole D, Larkin J, McCormick P, Mehigan B, Cathcart MC, Lysaght J, Reynolds JV, Ryan EJ, Dunne MR, O'Sullivan J. The tumour microenvironment of the upper and lower gastrointestinal tract differentially influences dendritic cell maturation. BMC Cancer 2020; 20:566. [PMID: 32552799 PMCID: PMC7302160 DOI: 10.1186/s12885-020-07012-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
Background Only 10–30% of oesophageal and rectal adenocarcinoma patients treated with neoadjuvant chemoradiotherapy have a complete pathological response. Inflammatory and angiogenic mediators in the tumour microenvironment (TME) may enable evasion of anti-tumour immune responses. Methods The TME influence on infiltrating dendritic cells (DCs) was modelled by treating immature monocyte-derived DCs with Tumour Conditioned Media (TCM) from distinct gastrointestinal sites, prior to LPS-induced maturation. Results Cell line conditioned media from gastrointestinal cell lines inhibited LPS-induced DC markers and TNF-α secretion. TCM generated from human tumour biopsies from oesophageal, rectal and colonic adenocarcinoma induced different effects on LPS-induced DC markers - CD54, CD80, HLA-DR, CD86 and CD83 were enhanced by oesophageal cancer; CD80, CD86 and CD83 were enhanced by rectal cancer, whereas CD54, HLA-DR, CD86, CD83 and PD-L1 were inhibited by colonic cancer. Notably, TCM from all GI cancer types inhibited TNF-α secretion. Additionally, TCM from irradiated biopsies inhibited DC markers. Profiling the TCM showed that IL-2 levels positively correlated with maturation marker CD54, while Ang-2 and bFGF levels negatively correlated with CD54. Conclusion This study identifies that there are differences in DC maturational capacity induced by the TME of distinct gastrointestinal cancers. This could potentially have implications for anti-tumour immunity and response to radiotherapy.
Collapse
Affiliation(s)
- Maria E Morrissey
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Róisín Byrne
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Celina Nulty
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Niamh H McCabe
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Clare T Butler
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Susan Kennedy
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Dermot O'Toole
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | | | | | | | - Mary-Clare Cathcart
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Joanne Lysaght
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - John V Reynolds
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland.,Oesophageal Unit, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Elizabeth J Ryan
- Centre for Colorectal Disease, Education and Research Centre, St. Vincent's University Hospital, Elm Park, Dublin 4, Ireland.,Department of Biological Sciences, Health Research Institute, University of Limerick, Castletroy, Co., Limerick, Ireland
| | - Margaret R Dunne
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Jacintha O'Sullivan
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
25
|
Abstract
Soft-tissue sarcoma (sts) is a rare mesenchymal malignancy that accounts for less than 1% of all adult tumours. Despite the successful advancement of localized therapies such as surgery and radiotherapy, these tumours can, for many, recur-often with metastatic disease. In the advanced setting, the role of systemic therapies is modest and is associated with poor survival. With the discovery of immunotherapies in other tumour types such as melanoma and lung cancer, interest has been renewed in exploring immunotherapy in sts. The biology of some stss makes them ripe for immunotherapy intervention; for example, some stss might have chromosomal translocations resulting in pathognomonic fusion products that have been shown to express cancer/testis antigens. Here, we present a targeted review of the published data and ongoing clinical trials for immunotherapies in patients with sarcoma, which comprise immune checkpoint inhibitors, adoptive cell therapies, and cancer vaccines.
Collapse
Affiliation(s)
- O Ayodele
- Toronto Sarcoma Program, Princess Margaret Cancer Centre and Mount Sinai Hospital, Toronto, ON
| | - A R Abdul Razak
- Toronto Sarcoma Program, Princess Margaret Cancer Centre and Mount Sinai Hospital, Toronto, ON
| |
Collapse
|
26
|
Melo AM, Maher SG, O'Leary SM, Doherty DG, Lysaght J. Selective effects of radiotherapy on viability and function of invariant natural killer T cells in vitro. Radiother Oncol 2020; 145:128-136. [PMID: 31962255 DOI: 10.1016/j.radonc.2019.12.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/09/2019] [Accepted: 12/19/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE Immunotherapies involving the adoptive transfer of ex vivo expanded autologous invariant natural killer (iNKT) cells are a potential option for cancer patients and are under investigation in clinical trials. Most cancer patients receive radiotherapy at some point during their treatment. We investigated the effects of therapeutic doses of radiation on the viability and function of human primary cultures of iNKT cells in vitro. MATERIALS AND METHODS iNKT cell lines generated from 6 healthy donors were subjected to therapeutically-relevant doses of radiation. Cell cycle arrest and cell death were assessed by flow cytometry. Double strand DNA breaks were analysed by measuring phosphorylated histone H2AX expression by fluorescence microscopy. Cytolytic degranulation, cytokine production and cytotoxicity by antigen-stimulated iNKT cells were assessed by flow cytometry. RESULTS Radiation inhibited viability of iNKT cells in a dose-dependent manner. Radiation caused double strand DNA breaks, which were rapidly repaired, and affected the cell cycle at high doses. Moderate doses of radiation did not inhibit degranulation or cytotoxicity by iNKT cells, but induced perforin expression and inhibited proliferation and interferon-γ production by surviving iNKT cells. DISCUSSION Exposure of iNKT cell to radiation can negatively affect their viability and function.
Collapse
Affiliation(s)
- Ashanty M Melo
- Department of Immunology, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland; Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Stephen G Maher
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Seónadh M O'Leary
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Derek G Doherty
- Department of Immunology, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland.
| | - Joanne Lysaght
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| |
Collapse
|
27
|
Castiello L, Aricò E, D'Agostino G, Santodonato L, Belardelli F. In situ Vaccination by Direct Dendritic Cell Inoculation: The Coming of Age of an Old Idea? Front Immunol 2019; 10:2303. [PMID: 31611878 PMCID: PMC6773832 DOI: 10.3389/fimmu.2019.02303] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/11/2019] [Indexed: 12/18/2022] Open
Abstract
For more than 25 years, dendritic cell (DC) based vaccination has flashily held promises to represent a therapeutic approach for cancer treatment. While the vast majority of studies has focused on the use of antigen loaded DC, the intratumoral delivery of unloaded DC aiming at in situ vaccination has gained much less attention. Such approach grounds on the ability of inoculated DC to internalize and process antigens directly released by tumor (usually in combination with cell-death-inducing agents) to activate broad patient-specific antitumor T cell response. In this review, we highlight the recent studies in both solid and hematological tumors showing promising clinical results and discuss the main pitfalls and advantages of this approach for endogenous cancer vaccination. Lastly, we discuss how in situ vaccination by DC inoculation may fit with current immunotherapy approaches to expand and prolong patient response.
Collapse
Affiliation(s)
- Luciano Castiello
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Eleonora Aricò
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | | - Laura Santodonato
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Filippo Belardelli
- Consiglio Nazionale Delle Ricerche, Institute of Translational Pharmacology, Rome, Italy
| |
Collapse
|
28
|
Piretto E, Delitala M, Kim PS, Frascoli F. Effects of mutations and immunogenicity on outcomes of anti-cancer therapies for secondary lesions. Math Biosci 2019; 315:108238. [PMID: 31401294 DOI: 10.1016/j.mbs.2019.108238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/02/2019] [Accepted: 08/03/2019] [Indexed: 12/30/2022]
Abstract
Cancer development is driven by mutations and selective forces, including the action of the immune system and interspecific competition. When administered to patients, anti-cancer therapies affect the development and dynamics of tumours, possibly with various degrees of resistance due to immunoediting and microenvironment. Tumours are able to express a variety of competing phenotypes with different attributes and thus respond differently to various anti-cancer therapies. In this paper, a mathematical framework incorporating a system of delay differential equations for the immune system activation cycle and an agent-based approach for tumour-immune interaction is presented. The focus is on those metastatic, secondary solid lesions that are still undetected and non-vascularised. By using available experimental data, we analyse the effects of combination therapies on these lesions and investigate the role of mutations on the rates of success of common treatments. Findings show that mutations, growth properties and immunoediting influence therapies' outcomes in nonlinear and complex ways, affecting cancer lesion morphologies, phenotypical compositions and overall proliferation patterns. Cascade effects on final outcomes for secondary lesions are also investigated, showing that actions on primary lesions could sometimes result in unexpected clearances of secondary tumours. This outcome is strongly dependent on the clonal composition of the primary and secondary masses and is shown to allow, in some cases, the control of the disease for years.
Collapse
Affiliation(s)
- Elena Piretto
- Department of Mathematical Sciences, Politecnico di Torino, Turin, Italy; Department of Mathematics, Universitá di Torino, Turin, Italy; Department of Mathematics, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Marcello Delitala
- Department of Mathematical Sciences, Politecnico di Torino, Turin, Italy
| | - Peter S Kim
- School of Mathematics and Statistics, University of Sydney, Sydney, New South Wales, Australia
| | - Federico Frascoli
- Department of Mathematics, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia.
| |
Collapse
|
29
|
|
30
|
Shevtsov M, Sato H, Multhoff G, Shibata A. Novel Approaches to Improve the Efficacy of Immuno-Radiotherapy. Front Oncol 2019; 9:156. [PMID: 30941308 PMCID: PMC6433964 DOI: 10.3389/fonc.2019.00156] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/25/2019] [Indexed: 12/31/2022] Open
Abstract
Radiotherapy (RT) has been applied for decades as a treatment modality in the management of various types of cancer. Ionizing radiation induces tumor cell death, which in turn can either elicit protective anti-tumor immune responses or immunosuppression in the tumor micromilieu that contributes to local tumor recurrence. Immunosuppression is frequently accompanied by the attraction of immunosuppressive cells such as myeloid-derived suppressor cells (MDSCs), M2 tumor-associated macrophages (TAMs), T regulatory cells (Tregs), N2 neutrophils, and by the release of immunosuppressive cytokines (TGF-β, IL-10) and chemokines. Immune checkpoint pathways, particularly of the PD-1/PD-L1 axis, have been determined as key regulators of cancer immune escape. While IFN-dependent upregulation of PD-L1 has been extensively investigated, up-to-date studies indicated the importance of DNA damage signaling in the regulation of PD-L1 expression following RT. DNA damage dependent PD-L1 expression is upregulated by ATM/ATR/Chk1 kinase activities and cGAS/STING-dependent pathway, proving the role of DNA damage signaling in PD-L1 induced expression. Checkpoint blockade immunotherapies (i.e., application of anti-PD-1 and anti-PD-L1 antibodies) combined with RT were shown to significantly improve the objective response rates in therapy of various primary and metastatic malignancies. Further improvements in the therapeutic potential of RT are based on combinations of RT with other immunotherapeutic approaches including vaccines, cytokines and cytokine inducers, and an adoptive immune cell transfer (DCs, NK cells, T cells). In the current review we provide immunological rationale for a combination of RT with various immunotherapies as well as analysis of the emerging preclinical evidences for these therapies.
Collapse
Affiliation(s)
- Maxim Shevtsov
- Center for Translational Cancer Research, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany.,Institute of Cytology, Russian Academy of Sciences (RAS), St. Petersburg, Russia.,First Pavlov State Medical University of St. Petersburg, St. Petersburg, Russia.,Almazov National Medical Research Centre, Polenov Russian Scientific Research Institute of Neurosurgery, St. Petersburg, Russia
| | - Hiro Sato
- Department of Radiation Oncology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Gabriele Multhoff
- Center for Translational Cancer Research, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Atsushi Shibata
- Education and Research Support Center, Graduate School of Medicine, Gunma University, Maebashi, Japan
| |
Collapse
|
31
|
Pender A, Jones RL, Pollack S. Optimising Cancer Vaccine Design in Sarcoma. Cancers (Basel) 2018; 11:E1. [PMID: 30577459 PMCID: PMC6356514 DOI: 10.3390/cancers11010001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/15/2018] [Accepted: 12/18/2018] [Indexed: 12/14/2022] Open
Abstract
Immunotherapeutics are increasingly recognized as a key tool in the armamentarium against malignancy. The success of immune checkpoint-targeting drugs and adoptive cell therapy has refocused attention on the potential anti-cancer effect of eliciting a tumour-specific immunological response. Sarcomas are a rare and diverse group of tumours with a limited prognosis in advanced disease despite systemic therapeutics. Various vaccine strategies including peptide vaccines against cancer testis antigens, dendritic cell vaccines, and viral vectors have been trialled in sarcoma with growing evidence of efficacy. Here, we review the principles of successful vaccine development and how these have been applied thus far to the treatment of sarcoma.
Collapse
Affiliation(s)
- Alexandra Pender
- Sarcoma Unit, Department of Medicine, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK.
| | - Robin L Jones
- Sarcoma Unit, Department of Medicine, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK.
- Division of Clinical Studies, The Institute of Cancer Research, London SW3 6JB, UK.
| | - Seth Pollack
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA 98109, USA.
- Division of Oncology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
32
|
Naghavi AO, Yang GQ, Latifi K, Gillies R, McLeod H, Harrison LB. The Future of Radiation Oncology in Soft Tissue Sarcoma. Cancer Control 2018. [PMCID: PMC6291881 DOI: 10.1177/1073274818815504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Radiotherapy (RT) is an important component of the treatment of soft tissue sarcomas (STS) and has been traditionally incorporated with a homogenous approach despite the reality that STS displays a known heterogeneity in clinicopathologic features and treatment outcomes. In this article, we explore the principle components of personalized medicine, including genomics, radiomics, and treatment response, along with their impact on the future of radiation therapy for STS. We propose a shift in the treatment paradigm for STS from a one-size-fits-all technique to one that implements the tenets of personalized medicine and includes the framework for a potential clinical trial technique in this heterogeneous disease.
Collapse
Affiliation(s)
- Arash O. Naghavi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- These authors contributed equally to this work
| | - George Q. Yang
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- These authors contributed equally to this work
| | - Kujtim Latifi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Howard McLeod
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Louis B. Harrison
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
33
|
van Gulijk M, Dammeijer F, Aerts JGJV, Vroman H. Combination Strategies to Optimize Efficacy of Dendritic Cell-Based Immunotherapy. Front Immunol 2018; 9:2759. [PMID: 30568653 PMCID: PMC6289976 DOI: 10.3389/fimmu.2018.02759] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/09/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells (APCs) that are essential for the activation of immune responses. In various malignancies, these immunostimulatory properties are exploited by DC-therapy, aiming at the induction of effective anti-tumor immunity by vaccination with ex vivo antigen-loaded DCs. Depending on the type of DC-therapy used, long-term clinical efficacy upon DC-therapy remains restricted to a proportion of patients, likely due to lack of immunogenicity of tumor cells, presence of a stromal compartment, and the suppressive tumor microenvironment (TME), thereby leading to the development of resistance. In order to circumvent tumor-induced suppressive mechanisms and unleash the full potential of DC-therapy, considerable efforts have been made to combine DC-therapy with chemotherapy, radiotherapy or with checkpoint inhibitors. These combination strategies could enhance tumor immunogenicity, stimulate endogenous DCs following immunogenic cell death, improve infiltration of cytotoxic T lymphocytes (CTLs) or specifically deplete immunosuppressive cells in the TME, such as regulatory T-cells and myeloid-derived suppressor cells. In this review, different strategies of combining DC-therapy with immunomodulatory treatments will be discussed. These strategies and insights will improve and guide DC-based combination immunotherapies with the aim of further improving patient prognosis and care.
Collapse
Affiliation(s)
- Mandy van Gulijk
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Floris Dammeijer
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Heleen Vroman
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
34
|
Fiore M, Gronchi A. ASO Author Reflections: Evolving Concepts for Radiation Therapy in Extremity and Trunk Soft Tissue Sarcoma. Ann Surg Oncol 2018; 25:942-943. [PMID: 30421054 DOI: 10.1245/s10434-018-7052-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Indexed: 11/18/2022]
Affiliation(s)
- Marco Fiore
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
35
|
Subbiah V, Murthy R, Hong DS, Prins RM, Hosing C, Hendricks K, Kolli D, Noffsinger L, Brown R, McGuire M, Fu S, Piha-Paul S, Naing A, Conley AP, Benjamin RS, Kaur I, Bosch ML. Cytokines Produced by Dendritic Cells Administered Intratumorally Correlate with Clinical Outcome in Patients with Diverse Cancers. Clin Cancer Res 2018; 24:3845-3856. [PMID: 30018119 DOI: 10.1158/1078-0432.ccr-17-2707] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/05/2018] [Accepted: 05/07/2018] [Indexed: 01/08/2023]
Abstract
Purpose: Dendritic cells (DC) initiate adaptive immune responses through the uptake and presentation of antigenic material. In preclinical studies, intratumorally injected activated DCs (aDCs; DCVax-Direct) were superior to immature DCs in rejecting tumors from mice.Experimental Design: This single-arm, open-label phase I clinical trial evaluated the safety and efficacy of aDCs, administered intratumorally, in patients with solid tumors. Three dose levels (2 million, 6 million, and 15 million aDCs per injection) were tested using a standard 3 + 3 dose-escalation trial design. Feasibility, immunogenicity, changes to the tumor microenvironment after direct injection, and survival were evaluated. We also investigated cytokine production of aDCs prior to injection.Results: In total, 39 of the 40 enrolled patients were evaluable. The injections of aDCs were well tolerated with no dose-limiting toxicities. Increased lymphocyte infiltration was observed in 54% of assessed patients. Stable disease (SD; best response) at week 8 was associated with increased overall survival. Increased secretion of interleukin (IL)-8 and IL12p40 by aDCs was significantly associated with survival (P = 0.023 and 0.024, respectively). Increased TNFα levels correlated positively with SD at week 8 (P < 0.01).Conclusions: Intratumoral aDC injections were feasible and safe. Increased production of specific cytokines was correlated with SD and prolonged survival, demonstrating a link between the functional profile of aDCs prior to injection and patient outcomes. Clin Cancer Res; 24(16); 3845-56. ©2018 AACR.
Collapse
Affiliation(s)
- Vivek Subbiah
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ravi Murthy
- Department of Interventional Radiology, Division of Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David S Hong
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert M Prins
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Chitra Hosing
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | - Robert Brown
- Department of Pathology and Laboratory Medicine, UT Health, University of Texas Health Science Center, Houston, Texas
| | - Mary McGuire
- Department of Pathology and Laboratory Medicine, UT Health, University of Texas Health Science Center, Houston, Texas
| | - Siquing Fu
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarina Piha-Paul
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anthony P Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert S Benjamin
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Indreshpal Kaur
- Cell Therapy Labs, GMP Laboratory, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
36
|
Abdo J, Cornell DL, Mittal SK, Agrawal DK. Immunotherapy Plus Cryotherapy: Potential Augmented Abscopal Effect for Advanced Cancers. Front Oncol 2018; 8:85. [PMID: 29644213 PMCID: PMC5882833 DOI: 10.3389/fonc.2018.00085] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/12/2018] [Indexed: 01/10/2023] Open
Abstract
Since the 1920s the gold standard for treating cancer has been surgery, which is typically preceded or followed with chemotherapy and/or radiation, a process that perhaps contributes to the destruction of a patient’s immune defense system. Cryosurgery ablation of a solid tumor is mechanistically similar to a vaccination where hundreds of unique antigens from a heterogeneous population of tumor cells derived from the invading cancer are released. However, releasing tumor-derived self-antigens into circulation may not be sufficient enough to overcome the checkpoint escape mechanisms some cancers have evolved to avoid immune responses. The potentiated immune response caused by blocking tumor checkpoints designed to prevent programmed cell death may be the optimal treatment method for the immune system to recognize these new circulating cryoablated self-antigens. Preclinical and clinical evidence exists for the complementary roles for Cytotoxic T-lymphocyte-associated protein (CTLA-4) and PD-1 antagonists in regulating adaptive immunity, demonstrating that combination immunotherapy followed by cryosurgery provides a more targeted immune response to distant lesions, a phenomenon known as the abscopal effect. We propose that when the host’s immune system has been “primed” with combined anti-CTLA-4 and anti-PD-1 adjuvants prior to cryosurgery, the preserved cryoablated tumor antigens will be presented and processed by the host’s immune system resulting in a robust cytotoxic CD8+ T-cell response. Based on recent investigations and well-described biochemical mechanisms presented herein, a polyvalent autoinoculation of many tumor-specific antigens, derived from a heterogeneous population of tumor cancer cells, would present to an unhindered yet pre-sensitized immune system yielding a superior advantage in locating, recognizing, and destroying tumor cells throughout the body.
Collapse
Affiliation(s)
- Joe Abdo
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States
| | - David L Cornell
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States.,Department of Surgery, CHI Health Creighton University Medical Center, Omaha, NE, United States
| | - Sumeet K Mittal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States.,Dignity Health, Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States
| |
Collapse
|
37
|
Levy A, Bonvalot S, Bellefqih S, Terrier P, Le Cesne A, Le Péchoux C. Is dose de-escalation possible in sarcoma patients treated with enlarged limb sparing resection? Radiother Oncol 2018; 126:493-498. [DOI: 10.1016/j.radonc.2017.10.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 09/04/2017] [Accepted: 10/20/2017] [Indexed: 12/11/2022]
|
38
|
Walle T, Martinez Monge R, Cerwenka A, Ajona D, Melero I, Lecanda F. Radiation effects on antitumor immune responses: current perspectives and challenges. Ther Adv Med Oncol 2018; 10:1758834017742575. [PMID: 29383033 PMCID: PMC5784573 DOI: 10.1177/1758834017742575] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy (RT) is currently used in more than 50% of cancer patients during the course of their disease in the curative, adjuvant or palliative setting. RT achieves good local control of tumor growth, conferring DNA damage and impacting tumor vasculature and the immune system. Formerly regarded as a merely immunosuppressive treatment, pre- and clinical observations indicate that the therapeutic effect of RT is partially immune mediated. In some instances, RT synergizes with immunotherapy (IT), through different mechanisms promoting an effective antitumor immune response. Cell death induced by RT is thought to be immunogenic and results in modulation of lymphocyte effector function in the tumor microenvironment promoting local control. Moreover, a systemic immune response can be elicited or modulated to exert effects outside the irradiation field (so called abscopal effects). In this review, we discuss the body of evidence related to RT and its immunogenic potential for the future design of novel combination therapies.
Collapse
Affiliation(s)
- Thomas Walle
- Innate Immunity Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Adelheid Cerwenka
- German Cancer Research Center (DKFZ), Research Group Innate Immunity, Heidelberg, Germany
| | - Daniel Ajona
- Division of Oncology, Centre for Applied Biomedical Research (CIMA), Pamplona, SpainIdiSNA, Navarra Institute for Health Research, Pamplona, SpainDepartment of Biochemistry and Genetics, University of Navarra, Pamplona, Spain Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)
| | - Ignacio Melero
- Programme in Immunotherapy, Centre for Applied Biomedical Research (CIMA), Pamplona, SpainDepartment of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)
| | - Fernando Lecanda
- Programme in Solid Tumours and Biomarkers, Division of Oncology, Centre for Applied Biomedical Research (CIMA), IdiSNA, Navarra Institute for Health Research, Department of Histology and Pathology, University of Navarra, School of Medicine, Pamplona, Spain. Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)
| |
Collapse
|
39
|
Coukos G, Tanyi J, Kandalaft LE. Opportunities in immunotherapy of ovarian cancer. Ann Oncol 2017; 27 Suppl 1:i11-i15. [PMID: 27141063 DOI: 10.1093/annonc/mdw084] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer (OC) is the most important cause of gynecological cancer-related mortality, with the majority of women presenting with advanced disease. Although surgery and chemotherapy can improve survival, the 5-year survival rates remain ominously low at 45%. Novel therapies are urgently needed. The presence of T cells in the OC tumor microenvironment is correlated with improved progression-free and overall survival, while the presence of regulatory T cells and expression of T-cell inhibitory molecules is correlated with a poor prognosis. These data indicate that immunotherapy could hold promise in improving the treatment of OC. In this review, we will discuss the rational of immunotherapy, highlight current results with cancer vaccines, adoptive T-cell therapy and immunomodulatory agents and summarize the immune effects of selected chemotherapeutic and radiotherapeutic agents.
Collapse
Affiliation(s)
- G Coukos
- Ludwig Institute for Cancer Research at the University of Lausanne, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
| | - J Tanyi
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, USA
| | - L E Kandalaft
- Ludwig Institute for Cancer Research at the University of Lausanne, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
40
|
Nathenson MJ, Conley AP, Sausville E. Immunotherapy: A New (and Old) Approach to Treatment of Soft Tissue and Bone Sarcomas. Oncologist 2017; 23:71-83. [PMID: 28935774 DOI: 10.1634/theoncologist.2016-0025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 07/14/2017] [Indexed: 12/11/2022] Open
Abstract
Soft tissue and bone sarcomas are a rare and heterogeneous form of cancer. With standard of care treatment options including surgery, radiation, and chemotherapy, the long-term survival is still low for high-risk soft tissue sarcoma patients. New treatment strategies are needed. Immunotherapy offers a new potential treatment paradigm with great promise. Immunotherapy of soft tissue sarcomas dates back to Dr. Coley's first use of toxins in the late 1800s. A variety of strategies of immunotherapy have been tried in soft tissue and bone sarcomas, including various vaccines and cytokines, with limited success. Results of these early clinical trials with vaccines and cytokines were disappointing, but there are reasons to be optimistic. Recent advances, particularly with the use of adoptive T-cell therapy and immune checkpoint inhibitors, have led to a resurgence of this field for all cancer patients. Clinical trials utilizing adoptive T-cell therapy and immune checkpoint inhibitors in soft tissue and bone sarcomas are under way. This paper reviews the current state of evidence for the use of immunotherapy, as well as current immunotherapy strategies (vaccines, adopative T-cell therapy, and immune checkpoint blockade), in soft tissue and bone sarcomas. By understanding the tumor microenviroment of sarcomas and how it relates to their immunoresponsiveness, better immunotherapy clinical trials can be designed, hopefully with improved outcomes for soft tissue and bone sarcoma patients. IMPLICATIONS FOR PRACTICE Immunotherapy is a promising treatment paradigm that is gaining acceptance for the management of several cancers, including melanoma, renal cell carcinoma, prostate cancer, and lung cancer. There is a long history of immunotherapy in the treatment of soft tissue and bone sarcomas, although with little success. It is important to understand past failures to develop future immunotherapy treatment strategies with an improved possibility of success. This article reviews the history of and current state of immunotherapy research in the treatment of soft tissue and bone sarcomas, with particular regard to vaccine trials, adoptive T-cell therapy, and immune checkpoint blockade.
Collapse
Affiliation(s)
- Michael J Nathenson
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Anthony P Conley
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Edward Sausville
- Department of Medicine and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
41
|
Murthy V, Minehart J, Sterman DH. Local Immunotherapy of Cancer: Innovative Approaches to Harnessing Tumor-Specific Immune Responses. J Natl Cancer Inst 2017; 109:4085220. [DOI: 10.1093/jnci/djx097] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/24/2017] [Indexed: 12/12/2022] Open
|
42
|
Candidate immune biomarkers for radioimmunotherapy. Biochim Biophys Acta Rev Cancer 2017; 1868:58-68. [DOI: 10.1016/j.bbcan.2017.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/21/2017] [Accepted: 02/25/2017] [Indexed: 12/25/2022]
|
43
|
Poleszczuk J, Luddy K, Chen L, Lee JK, Harrison LB, Czerniecki BJ, Soliman H, Enderling H. Neoadjuvant radiotherapy of early-stage breast cancer and long-term disease-free survival. Breast Cancer Res 2017; 19:75. [PMID: 28666457 PMCID: PMC5493088 DOI: 10.1186/s13058-017-0870-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 06/20/2017] [Indexed: 02/08/2023] Open
Abstract
Background Compared with surgery alone, postoperative adjuvant radiotherapy (RT) improves relapse-free survival of patients with early-stage breast cancer. We evaluated the long-term overall and disease-free survival rates of neoadjuvant (presurgical) versus adjuvant RT in early-stage breast cancer patients. Methods We used the Surveillance, Epidemiology, and End Results (SEER) database provided by the National Institutes of Health to derive an analytic dataset of 250,195 female patients with early-stage breast cancer who received RT before (n = 2554; 1.02%) or after (n = 247,641; 98.98%) surgery. Disease-free survival, defined as time to diagnosis of a second primary tumor at any location, was calculated from automated patient identification matching of all SEER records. Results Partial and complete mastectomies were performed in 94.4% and 5.6% of patients, respectively. In the largest cohort of estrogen receptor-positive women who underwent partial mastectomy, the HR of developing a second primary tumor after neoadjuvant compared with adjuvant RT was 0.64 (95% CI 0.55–0.75; P < 0.0001). Overall survival was independent of radiation sequence (HR 1; P = 0.95). Neoadjuvant RT also resulted in a lower HR for second primary cancer among estrogen receptor-positive patients who underwent mastectomy compared with those who received adjuvant RT (HR 0.48, 95% CI 0.26–0.87; P = 0.0162). Conclusions Neoadjuvant RT may significantly improve disease-free survival without reducing overall survival, especially for estrogen receptor-positive patients with early-stage breast cancer. This finding warrants further exploration of potential long-term benefits of neoadjuvant radiotherapy for early-stage breast cancer in a controlled, prospective clinical trial setting, with correlative studies done to identify potential mechanisms of superiority. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0870-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jan Poleszczuk
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, SRB 4, Tampa, FL, 33612, USA. .,Department for Mathematical Modeling of Physiological Processes, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland.
| | - Kimberly Luddy
- Department of Cancer Imaging & Metabolism, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Lu Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Jae K Lee
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Louis B Harrison
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Brian J Czerniecki
- Department of Breast Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Hatem Soliman
- Department of Women's Oncology and Experimental Therapeutics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Heiko Enderling
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, SRB 4, Tampa, FL, 33612, USA. .,Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
44
|
Tao Z, Li S, Ichim TE, Yang J, Riordan N, Yenugonda V, Babic I, Kesari S. Cellular immunotherapy of cancer: an overview and future directions. Immunotherapy 2017; 9:589-606. [DOI: 10.2217/imt-2016-0086] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The clinical success of checkpoint inhibitors has led to a renaissance of interest in cancer immunotherapies. In particular, the possibility of ex vivo expanding autologous lymphocytes that specifically recognize tumor cells has attracted much research and clinical trial interest. In this review, we discuss the historical background of tumor immunotherapy using cell-based approaches, and provide some rationale for overcoming current barriers to success of autologous immunotherapy. An overview of adoptive transfer of lymphocytes, tumor infiltrating lymphocytes and dendritic cell therapies is provided. We conclude with discussing the possibility of gene-manipulating immune cells in order to augment therapeutic activity, including silencing of the immune-suppressive zinc finger orphan nuclear receptor, NR2F6, as an attractive means of overcoming tumor-associated immune suppression.
Collapse
Affiliation(s)
- Ziqi Tao
- The Affiliated XuZhou Center Hospital of Nanjing University of Chinese Medicine, The Affiliated XuZhou Hospital of Medical College of Southeast University, Jiangsu, China
| | - Shuang Li
- Department of Endocrinology, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | | | - Junbao Yang
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Neil Riordan
- Medistem Panama, Inc., City of Knowledge, Clayton, Republic of Panama
| | - Venkata Yenugonda
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Ivan Babic
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Santosh Kesari
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
- John Wayne Cancer Institute, 2200 Santa Monica Blvd, Santa Monica, CA 90404, USA
| |
Collapse
|
45
|
Bockel S, Antoni D, Deutsch É, Mornex F. Immunothérapie et radiothérapie. Cancer Radiother 2017; 21:244-255. [DOI: 10.1016/j.canrad.2016.12.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 12/09/2016] [Accepted: 12/13/2016] [Indexed: 12/15/2022]
|
46
|
Ghosn M, El Rassy E, Kourie HR. Immunotherapies in sarcoma: Updates and future perspectives. World J Clin Oncol 2017; 8:145-150. [PMID: 28439495 PMCID: PMC5385435 DOI: 10.5306/wjco.v8.i2.145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/15/2016] [Accepted: 01/16/2017] [Indexed: 02/06/2023] Open
Abstract
Sarcomas are malignant tumors that are characterized by a wide diversity of subtypes with various cytogenetic profiles. Despite major treatment breakthroughs, standard treatment modalities combining chemotherapy, radiotherapy, and surgery failed to improve overall survival. Therefore, high expectations are foreseen with immunotherapy upon its maturation and better understanding of its mechanism of action. This paper presents a targeted review of the published data and ongoing clinical trials in immunotherapies of sarcomas, mainly adoptive cell therapies, cancer vaccines and immune checkpoint inhibitors.
Collapse
|
47
|
Loi M, Desideri I, Greto D, Mangoni M, Sottili M, Meattini I, Becherini C, Terziani F, Delli Paoli C, Olmetto E, Bonomo P, Livi L. Radiotherapy in the age of cancer immunology: Current concepts and future developments. Crit Rev Oncol Hematol 2017; 112:1-10. [PMID: 28325250 DOI: 10.1016/j.critrevonc.2017.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/24/2016] [Accepted: 02/06/2017] [Indexed: 02/06/2023] Open
Abstract
Major advances in the knowledge of cancer biology and its interactions with tumor immune environment led to the emergence, in the last five years of new immunotherapy-based treatment strategies in cancer patients. At the same time, improvement in radiation technique and progress in radiobiology allowed in the last decade to expand the applications of radiotherapy in a growing number of settings. At present, there are strong theoretical basis to propose immune-enhanced radiation therapy that may represent in the future a new paradigm of treatment, combining the intrinsic power of radiotherapy to elicit a specific, systemic, tumor-directed immune response with modern highly conformal and precise dose delivery, in order to maximize response at the major site of disease and obtain durable disease control. The aim of this review is to describe the principal mechanisms of immune modulation of response to radiation and investigational strategies to harness the potential of radiation-inducible immune response: radiation therapy is expected to be not just a local treatment but the cornerstone of a multimodal strategy that might achieve long-lasting tumor remission at the primary site and systemic efficacy metastatic lesions.
Collapse
Affiliation(s)
- Mauro Loi
- Department of Radiation Oncology, University of Florence, Florence, Italy.
| | - Isacco Desideri
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Daniela Greto
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Monica Mangoni
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Mariangela Sottili
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Icro Meattini
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Carlotta Becherini
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Francesca Terziani
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | | | - Emanuela Olmetto
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Pierluigi Bonomo
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Lorenzo Livi
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
48
|
Kalina JL, Neilson DS, Comber AP, Rauw JM, Alexander AS, Vergidis J, Lum JJ. Immune Modulation by Androgen Deprivation and Radiation Therapy: Implications for Prostate Cancer Immunotherapy. Cancers (Basel) 2017; 9:cancers9020013. [PMID: 28134800 PMCID: PMC5332936 DOI: 10.3390/cancers9020013] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 01/20/2017] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer patients often receive androgen deprivation therapy (ADT) in combination with radiation therapy (RT). Recent evidence suggests that both ADT and RT have immune modulatory properties. First, ADT can cause infiltration of lymphocytes into the prostate, although it remains unclear whether the influx of lymphocytes is beneficial, particularly with the advent of new classes of androgen blockers. Second, in rare cases, radiation can elicit immune responses that mediate regression of metastatic lesions lying outside the field of radiation, a phenomenon known as the abscopal response. In light of these findings, there is emerging interest in exploiting any potential synergy between ADT, RT, and immunotherapy. Here, we provide a comprehensive review of the rationale behind combining immunotherapy with ADT and RT for the treatment of prostate cancer, including an examination of the current clinical trials that employ this combination. The reported outcomes of several trials demonstrate the promise of this combination strategy; however, further scrutiny is needed to elucidate how these standard therapies interact with immune modulators. In addition, we discuss the importance of synchronizing immune modulation relative to ADT and RT, and provide insight into elements that may impact the ability to achieve maximum synergy between these treatments.
Collapse
Affiliation(s)
- Jennifer L Kalina
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, BC V8R 6V5, Canada.
| | - David S Neilson
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, BC V8R 6V5, Canada.
- Department of Biochemistry & Microbiology, University of Victoria, Victoria, BC V8P 5C2, Canada.
| | - Alexandra P Comber
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, BC V8R 6V5, Canada.
- Department of Biochemistry & Microbiology, University of Victoria, Victoria, BC V8P 5C2, Canada.
| | - Jennifer M Rauw
- British Columbia Cancer Agency, Victoria, BC, V8R 6V5, Canada.
- Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| | - Abraham S Alexander
- British Columbia Cancer Agency, Victoria, BC, V8R 6V5, Canada.
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| | - Joanna Vergidis
- British Columbia Cancer Agency, Victoria, BC, V8R 6V5, Canada.
- Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| | - Julian J Lum
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, BC V8R 6V5, Canada.
- Department of Biochemistry & Microbiology, University of Victoria, Victoria, BC V8P 5C2, Canada.
| |
Collapse
|
49
|
Hillman GG, Reich LA, Rothstein SE, Abernathy LM, Fountain MD, Hankerd K, Yunker CK, Rakowski JT, Quemeneur E, Slos P. Radiotherapy and MVA-MUC1-IL-2 vaccine act synergistically for inducing specific immunity to MUC-1 tumor antigen. J Immunother Cancer 2017; 5:4. [PMID: 28116088 PMCID: PMC5240430 DOI: 10.1186/s40425-016-0204-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/12/2016] [Indexed: 11/30/2022] Open
Abstract
Background We previously demonstrated that tumor irradiation potentiates cancer vaccines using genetic modification of tumor cells in murine tumor models. To investigate whether tumor irradiation augments the immune response to MUC1 tumor antigen, we have tested the efficacy of tumor irradiation combined with an MVA-MUC1-IL2 cancer vaccine (Transgene TG4010) for murine renal adenocarcinoma (Renca) cells transfected with MUC1. Methods Established subcutaneous Renca-MUC1 tumors were treated with 8 Gy radiation on day 11 and peritumoral injections of MVA-MUC1-IL2 vector on day 12 and 17, or using a reverse sequence of vaccine followed by radiation. Growth delays were monitored by tumor measurements and histological responses were evaluated by immunohistochemistry. Specific immunity was assessed by challenge with Renca-MUC1 cells. Generation of tumor-specific T cells was detected by IFN-γ production from splenocytes stimulated in vitro with tumor lysates using ELISPOT assays. Results Tumor growth delays observed by tumor irradiation combined with MVA-MUC1-IL-2 vaccine were significantly more prolonged than those observed by vaccine, radiation, or radiation with MVA empty vector. The sequence of cancer vaccine followed by radiation two days later resulted in 55–58% complete responders and 60% mouse long-term survival. This sequence was more effective than that of radiation followed by vaccine leading to 24–30% complete responders and 30% mouse survival. Responding mice were immune to challenge with Renca-MUC1 cells, indicating the induction of specific tumor immunity. Histology studies of regressing tumors at 1 week after therapy, revealed extensive tumor destruction and a heavy infiltration of CD45+ leukocytes including F4/80+ macrophages, CD8+ cytotoxic T cells and CD4+ helper T cells. The generation of tumor-specific T cells by combined therapy was confirmed by IFN-γ secretion in tumor-stimulated splenocytes. An abscopal effect was measured by rejection of an untreated tumor on the contralateral flank to the tumor treated with radiation and vaccine. Conclusions These findings suggest that cancer vaccine given prior to local tumor irradiation augments an immune response targeted at tumor antigens that results in specific anti-tumor immunity. These findings support further exploration of the combination of radiotherapy with cancer vaccines for the treatment of cancer. Electronic supplementary material The online version of this article (doi:10.1186/s40425-016-0204-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gilda G Hillman
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA ; Radiation Oncology Division, Immunology & Microbiology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI 48201 USA
| | - Lyndsey A Reich
- Radiation Oncology Division, Immunology & Microbiology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI 48201 USA
| | - Shoshana E Rothstein
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA
| | - Lisa M Abernathy
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA ; Radiation Oncology Division, Immunology & Microbiology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI 48201 USA ; Present address: Department of Microbiology and Immunology, Indiana University School of Medicine at Notre Dame, South Bend, IN 46617 USA
| | - Matthew D Fountain
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA ; Radiation Oncology Division, Immunology & Microbiology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI 48201 USA
| | - Kali Hankerd
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA
| | - Christopher K Yunker
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA
| | - Joseph T Rakowski
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA
| | - Eric Quemeneur
- Transgene SA, Parc d'Innovation, CS80166, 67405 Illkirch-Graffenstaden Cedex, France
| | - Philippe Slos
- Transgene SA, Parc d'Innovation, CS80166, 67405 Illkirch-Graffenstaden Cedex, France ; Present address: Oncodesign, 20, rue Jean Mazen, 21076 Dijon Cedex, France
| |
Collapse
|
50
|
Nathenson MJ, Sausville E. Looking for answers: the current status of neoadjuvant treatment in localized soft tissue sarcomas. Cancer Chemother Pharmacol 2016; 78:895-919. [PMID: 27206640 PMCID: PMC7577379 DOI: 10.1007/s00280-016-3055-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 05/04/2016] [Indexed: 12/19/2022]
Abstract
PURPOSE Sarcomas are a rare and heterogeneous variant of cancer. The standard of care treatment involves surgical resection with radiation in high-risk patients. Despite appropriate treatment approximately 50 % of patients will suffer and die from recurrent disease. The purpose of this article is to review the current evidence concerning the use of neoadjuvant chemotherapy with or without radiation in soft tissue sarcomas. METHODS An in-depth literature search was conducted using Ovid Medline and PubMed. RESULTS The most active chemotherapeutic agents in sarcoma are anthracyclines and ifosfamide. Adjuvant chemotherapy trials show only minimal benefit. Neoadjuvant chemotherapy offers the potential advantage of reducing the extent of surgery, increasing the limb salvage rate, early exposure of micrometastatic disease to chemotherapy, and assessment of tumor response to chemotherapy. Some retrospective and phase II trials suggest a benefit to neoadjuvant chemotherapy. Unfortunately, no clearly positive phase III prospectively randomized trials exist for neoadjuvant therapy in soft tissue sarcomas. CONCLUSIONS The current neoadjuvant chemotherapy trials that do exist are heterogeneous resulting in conflicting results. However, neoadjuvant chemotherapy with or without radiation can be considered in patients with high-risk disease in an attempt to improve long-term outcomes.
Collapse
Affiliation(s)
- Michael J Nathenson
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 450, Houston, TX, 77030, USA.
| | - Edward Sausville
- Department of Medicine, University of Maryland Medical Center, 22 South Greene Street Suite 9d10, Baltimore, MD, 21201, USA
| |
Collapse
|