1
|
Lu H, Sun Y, Gao Y, Xiao L, Zhou J, Yin X, Guo W, Fan K. Clinical application of 192Ir High-Dose-Rate brachytherapy in metastatic lymph nodes of the neck. Discov Oncol 2023; 14:219. [PMID: 38038833 PMCID: PMC10692036 DOI: 10.1007/s12672-023-00827-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/12/2023] [Indexed: 12/02/2023] Open
Abstract
OBJECTIVE The objective of this study was to investigate the safety and effectiveness of high-dose-rate brachytherapy as a treatment modality for recurrent or residual neck metastatic lymph nodes following external radiotherapy. METHODS 38 patients with 52 metastatic lymph nodes recurring or residual after previous external radiotherapy was completed to metastatic lymph nodes in the neck were collected from January 2019 to February 2022. High-dose-rate brachytherapy with 192Ir was performed with a prescribed dose of 20-30 Gy/1f (effective biological dose of 60-120 Gy), and imaging was performed at 1, 3, and 6 months after treatment to assess the local control rate and adverse effects of treatment. RESULTS All 38 patients received completed treatment, and they were followed up for 6 months. 52 patients with neck lymph node metastases had an objective response rate. (Complete response, CR + Partial response, PR) of 76.9%, which comprised 89.5% (34/38) for lymph nodes ≤ 3 cm and 42.9% (4/14) for > 3 cm, P = 0.028. P > 0.05 for CR + PR versus stable disease, SD + progressive disease, PD for lymph nodes between different subdivisions of the neck. Using the Radiation Therapy Oncology Group (RTOG) Acute Toxicity Scoring System, there were 6 cases of acute radioskin injuries of degree I and 4 cases of degree II with a 60% symptomatic relief rate. CONCLUSIONS High-dose-rate brachytherapy serves as a safe and effective method in treating recurrent residual neck metastatic lymph nodes in the field after external radiotherapy, exerting tolerable adverse effects.
Collapse
Affiliation(s)
- Hongling Lu
- Department of Radiation Oncology, Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine-Hebei Province, No. 31, Huanghe West Road, Yunhe District, Cangzhou, 061000, Hebei, China
| | - Yunchuan Sun
- Department of Radiation Oncology, Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine-Hebei Province, No. 31, Huanghe West Road, Yunhe District, Cangzhou, 061000, Hebei, China.
| | - Yan Gao
- Department of Radiation Oncology, Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine-Hebei Province, No. 31, Huanghe West Road, Yunhe District, Cangzhou, 061000, Hebei, China
| | - Li Xiao
- Department of Radiation Oncology, Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine-Hebei Province, No. 31, Huanghe West Road, Yunhe District, Cangzhou, 061000, Hebei, China
| | - Jianxi Zhou
- Department of Radiation Oncology, Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine-Hebei Province, No. 31, Huanghe West Road, Yunhe District, Cangzhou, 061000, Hebei, China
| | - Xiaoming Yin
- Department of Radiation Oncology, Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine-Hebei Province, No. 31, Huanghe West Road, Yunhe District, Cangzhou, 061000, Hebei, China
| | - Wei Guo
- Department of Radiation Oncology, Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine-Hebei Province, No. 31, Huanghe West Road, Yunhe District, Cangzhou, 061000, Hebei, China
| | - Kui Fan
- Department of Radiation Oncology, Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine-Hebei Province, No. 31, Huanghe West Road, Yunhe District, Cangzhou, 061000, Hebei, China
| |
Collapse
|
2
|
Zhao D, Mo Y, Neganova ME, Aleksandrova Y, Tse E, Chubarev VN, Fan R, Sukocheva OA, Liu J. Dual effects of radiotherapy on tumor microenvironment and its contribution towards the development of resistance to immunotherapy in gastrointestinal and thoracic cancers. Front Cell Dev Biol 2023; 11:1266537. [PMID: 37849740 PMCID: PMC10577389 DOI: 10.3389/fcell.2023.1266537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023] Open
Abstract
Successful clinical methods for tumor elimination include a combination of surgical resection, radiotherapy, and chemotherapy. Radiotherapy is one of the crucial components of the cancer treatment regimens which allow to extend patient life expectancy. Current cutting-edge radiotherapy research is focused on the identification of methods that should increase cancer cell sensitivity to radiation and activate anti-cancer immunity mechanisms. Radiation treatment activates various cells of the tumor microenvironment (TME) and impacts tumor growth, angiogenesis, and anti-cancer immunity. Radiotherapy was shown to regulate signaling and anti-cancer functions of various TME immune and vasculature cell components, including tumor-associated macrophages, dendritic cells, endothelial cells, cancer-associated fibroblasts (CAFs), natural killers, and other T cell subsets. Dual effects of radiation, including metastasis-promoting effects and activation of oxidative stress, have been detected, suggesting that radiotherapy triggers heterogeneous targets. In this review, we critically discuss the activation of TME and angiogenesis during radiotherapy which is used to strengthen the effects of novel immunotherapy. Intracellular, genetic, and epigenetic mechanisms of signaling and clinical manipulations of immune responses and oxidative stress by radiotherapy are accented. Current findings indicate that radiotherapy should be considered as a supporting instrument for immunotherapy to limit the cancer-promoting effects of TME. To increase cancer-free survival rates, it is recommended to combine personalized radiation therapy methods with TME-targeting drugs, including immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Deyao Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingyi Mo
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Margarita E. Neganova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russia
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Chernogolovka, Russia
| | - Yulia Aleksandrova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russia
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Chernogolovka, Russia
| | - Edmund Tse
- Department of Hepatology, Royal Adelaide Hospital, CALHN, Adelaide, SA, Australia
| | - Vladimir N. Chubarev
- Sechenov First Moscow State Medical University, Sechenov University, Moscow, Russia
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Olga A. Sukocheva
- Department of Hepatology, Royal Adelaide Hospital, CALHN, Adelaide, SA, Australia
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Levis M, Gastino A, De Giorgi G, Mantovani C, Bironzo P, Mangherini L, Ricci AA, Ricardi U, Cassoni P, Bertero L. Modern Stereotactic Radiotherapy for Brain Metastases from Lung Cancer: Current Trends and Future Perspectives Based on Integrated Translational Approaches. Cancers (Basel) 2023; 15:4622. [PMID: 37760591 PMCID: PMC10526239 DOI: 10.3390/cancers15184622] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/01/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Brain metastases (BMs) represent the most frequent metastatic event in the course of lung cancer patients, occurring in approximately 50% of patients with non-small-cell lung cancer (NSCLC) and in up to 70% in patients with small-cell lung cancer (SCLC). Thus far, many advances have been made in the diagnostic and therapeutic procedures, allowing improvements in the prognosis of these patients. The modern approach relies on the integration of several factors, such as accurate histological and molecular profiling, comprehensive assessment of clinical parameters and precise definition of the extent of intracranial and extracranial disease involvement. The combination of these factors is pivotal to guide the multidisciplinary discussion and to offer the most appropriate treatment to these patients based on a personalized approach. Focal radiotherapy (RT), in all its modalities (radiosurgery (SRS), fractionated stereotactic radiotherapy (SRT), adjuvant stereotactic radiotherapy (aSRT)), is the cornerstone of BM management, either alone or in combination with surgery and systemic therapies. We review the modern therapeutic strategies available to treat lung cancer patients with brain involvement. This includes an accurate review of the different technical solutions which can be exploited to provide a "state-of-art" focal RT and also a detailed description of the systemic agents available as effective alternatives to SRS/SRT when a targetable molecular driver is present. In addition to the validated treatment options, we also discuss the future perspective for focal RT, based on emerging clinical reports (e.g., SRS for patients with many BMs from NSCLC or SRS for BMs from SCLC), together with a presentation of innovative and promising findings in translational research and the combination of novel targeted agents with SRS/SRT.
Collapse
Affiliation(s)
- Mario Levis
- Radiation Oncology Unit, Department of Oncology, University of Turin, 10126 Turin, Italy; (M.L.); (A.G.); (G.D.G.); (C.M.); (U.R.)
| | - Alessio Gastino
- Radiation Oncology Unit, Department of Oncology, University of Turin, 10126 Turin, Italy; (M.L.); (A.G.); (G.D.G.); (C.M.); (U.R.)
| | - Greta De Giorgi
- Radiation Oncology Unit, Department of Oncology, University of Turin, 10126 Turin, Italy; (M.L.); (A.G.); (G.D.G.); (C.M.); (U.R.)
| | - Cristina Mantovani
- Radiation Oncology Unit, Department of Oncology, University of Turin, 10126 Turin, Italy; (M.L.); (A.G.); (G.D.G.); (C.M.); (U.R.)
| | - Paolo Bironzo
- Oncology Unit, Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy;
| | - Luca Mangherini
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.M.); (A.A.R.); (P.C.)
| | - Alessia Andrea Ricci
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.M.); (A.A.R.); (P.C.)
| | - Umberto Ricardi
- Radiation Oncology Unit, Department of Oncology, University of Turin, 10126 Turin, Italy; (M.L.); (A.G.); (G.D.G.); (C.M.); (U.R.)
| | - Paola Cassoni
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.M.); (A.A.R.); (P.C.)
| | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.M.); (A.A.R.); (P.C.)
| |
Collapse
|
4
|
Baude J, Limagne E, Ladjohounlou R, Mirjolet C. Combining radiotherapy and NK cell-based therapies: The time has come. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:31-60. [PMID: 37438020 DOI: 10.1016/bs.ircmb.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Natural killer (NK) cells are innate lymphoid cells that play an essential role in the anti-tumor response through immunosurveillance, multiple mechanisms of cytotoxicity and the synthesis of cytokines modulating the immune tumor microenvironment (TME). After the dramatic advances in immunotherapy targeting T cells including the success of checkpoint inhibitors or autologous chimeric antigen receptor (CAR) expressing T cells in clinical practice, NK cells have gained growing interest for the development of new therapies. Although NK cells have shown promising responses in leukemia patients, the effects of NK-targeted therapies are currently limited in the treatment of solid tumors. Thus, radiotherapy could provide a valuable solution to improve treatments targeting NK cells. Indeed, ionizing radiations represent a powerful immuno-modulator that can either induce a pro-inflammatory and anti-tumor TME, or conversely lead to immunosuppression of effector immune cells in favor of tumor growth and therapeutic escape, depending on how it is delivered and tumor models. However, the effects of ionizing radiation on NK cells are only partially understood. Therefore, we review the effects of radiotherapy on the NK cell-mediated anti-tumor response, and propose potential strategies to reinvigorate NK cells by combining radiotherapy with NK cell-targeted therapies.
Collapse
Affiliation(s)
- Jérémy Baude
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Unicancer, Dijon, France
| | - Emeric Limagne
- TIReCS Team, UMR INSERM 1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, Dijon, France; University of Bourgogne Franche-Comté, Dijon, France
| | - Riad Ladjohounlou
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Unicancer, Dijon, France; TIReCS Team, UMR INSERM 1231, Dijon, France
| | - Céline Mirjolet
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Unicancer, Dijon, France; TIReCS Team, UMR INSERM 1231, Dijon, France.
| |
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
|
Gan YX, Li GH, Ou X, Wang CH, Du QH. Case Report: Chemotherapy and Radiotherapy Combined With DC-CIK for Pulmonary and Mediastinal Metastases From Nasopharyngeal Carcinoma. Front Oncol 2022; 12:778643. [PMID: 35251965 PMCID: PMC8891562 DOI: 10.3389/fonc.2022.778643] [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: 10/14/2021] [Accepted: 01/25/2022] [Indexed: 12/22/2022] Open
Abstract
IntroductionThe optimal treatment for pulmonary and mediastinal metastasis of nasopharyngeal carcinoma (NPC) is still controversial, and the therapeutic effect is poor recently. In one case, we demonstrated a long-term survival after postoperative chemoradiotherapy combined with dendritic cell and cytokine-induced killer (DC-CIK) immunotherapy for pulmonary and mediastinal metastases from NPC.Baseline CharacteristicsA 53-year-old woman was admitted to our hospital in June 2008. Pathological biopsy revealed a poorly differentiated squamous cell carcinoma located in the nasopharynx with the invasion of internal pterygoid muscles, the sphenoid bone, and unilateral neck lymph node metastasis. No distant metastases were observed. The stage of NPC was T3N1M0 III (AJCC8). The patient received concurrent chemoradiotherapy for primary lesion and neck lymph nodes and achieved complete remission (CR) of the disease after 3 months. Follow-up at 3-month intervals was carried out. Pulmonary and mediastinal lymph node metastases were found in July 2009. The patient then underwent right upper lobectomy and mediastinal lymph node dissection and five cycles of gemcitabine and cisplatin (GP) regimen chemotherapy, following radiotherapy and DC-CIK immunotherapy.ResultsAfter a follow-up time of 13 years, no tumor recurrence or metastasis and severe adverse reactions were found.ConclusionPostoperative chemotherapy and radiotherapy in combination with DC-CIK immunotherapy may produce a synergistic therapeutic effect on patients with mediastinal lymph node metastasis from NPC.
Collapse
|
7
|
Berry MR, Fan TM. Target-Based Radiosensitization Strategies: Concepts and Companion Animal Model Outlook. Front Oncol 2021; 11:768692. [PMID: 34746010 PMCID: PMC8564182 DOI: 10.3389/fonc.2021.768692] [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: 09/01/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
External beam radiotherapy is indicated in approximately 50-60% of human cancer patients. The prescribed dose of ionizing radiation that can be delivered to a tumor is determined by the sensitivity of the normal surrounding tissues. Despite dose intensification provided by highly conformal radiotherapy, durable locoregional tumor control remains a clinical barrier for recalcitrant tumor histologies, and contributes to cancer morbidity and mortality. Development of target-based radiosensitization strategies that selectively sensitizes tumor tissue to ionizing radiation is expected to improve radiotherapy efficacy. While exploration of radiosensitization strategies has vastly expanded with technological advances permitting the precise and conformal delivery of radiation, maximal clinical benefit derived from radiotherapy will require complementary discoveries that exploit molecularly-based vulnerabilities of tumor cells, as well as the assessment of investigational radiotherapy strategies in animal models that faithfully recapitulate radiobiologic responses of human cancers. To address these requirements, the purpose of this review is to underscore current and emerging concepts of molecularly targeted radiosensitizing strategies and highlight the utility of companion animal models for improving the predictive value of radiotherapy investigations.
Collapse
Affiliation(s)
- Matthew R Berry
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, United States.,Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
8
|
Heeran AB, Dunne MR, Morrissey ME, Buckley CE, Clarke N, Cannon A, Donlon NE, Nugent TS, Durand M, Dunne C, Larkin JO, Mehigan B, McCormick P, Lynam-Lennon N, O’Sullivan J. The Protein Secretome Is Altered in Rectal Cancer Tissue Compared to Normal Rectal Tissue, and Alterations in the Secretome Induce Enhanced Innate Immune Responses. Cancers (Basel) 2021; 13:571. [PMID: 33540635 PMCID: PMC7867296 DOI: 10.3390/cancers13030571] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Locally advanced rectal cancer is treated with neoadjuvant-chemoradiotherapy; however, only ~22% of patients achieve a complete response, and resistance mechanisms are poorly understood. The role of inflammation and immune cell biology in this setting is under-investigated. In this study, we profiled the inflammatory protein secretome of normal (non-cancer) (n = 8) and malignant rectal tissue (n = 12) pre- and post-radiation in human ex vivo explant models and examined the influence of these untreated and treated secretomes on dendritic cell biology (n = 8 for cancer and normal). These resultant profiles were correlated with patient clinical characteristics. Nineteen factors were secreted at significantly higher levels from the rectal cancer secretome when compared to the normal rectal secretome; Flt-1, P1GF, IFN-γ, IL-6, IL-10, CCL20, CCL26, CCL22, CCL3, CCL4, CCL17, GM-CSF, IL-12/IL-23p40, IL-17A, IL-1α, IL-17A/F, IL-1RA, TSLP and CXCL10 (p < 0.05). Radiation was found to have differential effects on normal rectal tissue and rectal cancer tissue with increased IL-15 and CCL22 secretion following radiation from normal rectal tissue explants (p < 0.05), while no significant alterations were observed in the irradiated rectal cancer tissue. Interestingly, however, the irradiated rectal cancer secretome induced the most potent effect on dendritic cell maturation via upregulation of CD80 and PD-L1. Patient's visceral fat area correlated with secreted factors including CCL20, suggesting that obesity status may alter the tumour microenvironment (TME). These results suggest that radiation does not have a negative effect on the ability of the rectal cancer TME to induce an immune response. Understanding these responses may unveil potential therapeutic targets to enhance radiation response and mitigate normal tissue injury. Tumour irradiation in this cohort enhances innate immune responses, which may be harnessed to improve patient treatment outcome.
Collapse
Affiliation(s)
- Aisling B. Heeran
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Margaret R. Dunne
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Maria E. Morrissey
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Croí E. Buckley
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Niamh Clarke
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Aoife Cannon
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Noel E. Donlon
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Timothy S. Nugent
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Michael Durand
- GEMS, St. James’s Hospital, D08 NHY1 Dublin 8, Ireland; (M.D.); (C.D.); (J.O.L.); (B.M.); (P.M.)
| | - Cara Dunne
- GEMS, St. James’s Hospital, D08 NHY1 Dublin 8, Ireland; (M.D.); (C.D.); (J.O.L.); (B.M.); (P.M.)
| | - John O. Larkin
- GEMS, St. James’s Hospital, D08 NHY1 Dublin 8, Ireland; (M.D.); (C.D.); (J.O.L.); (B.M.); (P.M.)
| | - Brian Mehigan
- GEMS, St. James’s Hospital, D08 NHY1 Dublin 8, Ireland; (M.D.); (C.D.); (J.O.L.); (B.M.); (P.M.)
| | - Paul McCormick
- GEMS, St. James’s Hospital, D08 NHY1 Dublin 8, Ireland; (M.D.); (C.D.); (J.O.L.); (B.M.); (P.M.)
| | - Niamh Lynam-Lennon
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Jacintha O’Sullivan
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| |
Collapse
|
9
|
Khan A, Dias F, Neekhra S, Singh B, Srivastava R. Designing and Immunomodulating Multiresponsive Nanomaterial for Cancer Theranostics. Front Chem 2021; 8:631351. [PMID: 33585406 PMCID: PMC7878384 DOI: 10.3389/fchem.2020.631351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/22/2020] [Indexed: 01/14/2023] Open
Abstract
Cancer has been widely investigated yet limited in its manifestation. Cancer treatment holds innovative and futuristic strategies considering high disease heterogeneity. Chemotherapy, radiotherapy and surgery are the most explored pillars; however optimal therapeutic window and patient compliance recruit constraints. Recently evolved immunotherapy demonstrates a vital role of the host immune system to prevent metastasis recurrence, still undesirable clinical response and autoimmune adverse effects remain unresolved. Overcoming these challenges, tunable biomaterials could effectively control the co-delivery of anticancer drugs and immunomodulators. Current status demands a potentially new approach for minimally invasive, synergistic, and combinatorial nano-biomaterial assisted targeted immune-based treatment including therapeutics, diagnosis and imaging. This review discusses the latest findings of engineering biomaterial with immunomodulating properties and implementing novel developments in designing versatile nanosystems for cancer theranostics. We explore the functionalization of nanoparticle for delivering antitumor therapeutic and diagnostic agents promoting immune response. Through understanding the efficacy of delivery system, we have enlightened the applicability of nanomaterials as immunomodulatory nanomedicine further advancing to preclinical and clinical trials. Future and present ongoing improvements in engineering biomaterial could result in generating better insight to deal with cancer through easily accessible immunological interventions.
Collapse
Affiliation(s)
- Amreen Khan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Centre for Research in Nanotechnology and Science, Indian Institute of Technology Bombay, Mumbai, India
| | - Faith Dias
- Department of Chemical Engineering, Thadomal Shahani Engineering College, Mumbai, India
| | - Suditi Neekhra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Barkha Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Centre for Research in Nanotechnology and Science, Indian Institute of Technology Bombay, Mumbai, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
10
|
Bear AS, Vonderheide RH, O'Hara MH. Challenges and Opportunities for Pancreatic Cancer Immunotherapy. Cancer Cell 2020; 38:788-802. [PMID: 32946773 PMCID: PMC7738380 DOI: 10.1016/j.ccell.2020.08.004] [Citation(s) in RCA: 384] [Impact Index Per Article: 76.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is among the most immune-resistant tumor types. Its unique genomic landscape shaped by oncogenic drivers promotes immune suppression from the earliest stages of tumor inception to subvert adaptive T cell immunity. Single-agent immune modulators have thus far proven clinically ineffective, and multi-modal therapies targeting mechanisms of immunotherapy resistance are likely needed. Here, we review novel immunotherapy strategies currently under investigation to (1) confer antigen specificity, (2) enhance T cell effector function, and (3) neutralize immunosuppressive elements within the tumor microenvironment that may be rationally combined to untangle the web of immune resistance in PDA and other tumors.
Collapse
Affiliation(s)
- Adham S Bear
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert H Vonderheide
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| | - Mark H O'Hara
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA. mark.o'
| |
Collapse
|
11
|
Banstola A, Jeong JH, Yook S. Immunoadjuvants for cancer immunotherapy: A review of recent developments. Acta Biomater 2020; 114:16-30. [PMID: 32777293 DOI: 10.1016/j.actbio.2020.07.063] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/14/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy evolved as a new treatment modality to eradicate tumor cells and has gained in popularity after its successful clinical transition. By activating antigen-presenting cells (APCs), and thus, inducing innate or adaptive immune responses, immunoadjuvants have become promising tools for cancer immunotherapy. Different types of immunoadjuvants such as toll-like receptor (TLR) agonists, exosomes, and metallic and plant-derived immunoadjuvants have been studied for their immunological effects. However, the clinical use of immunoadjuvants is limited by short response rates and various side-effects. The rapid progress made in the development of nanoparticle systems as immunoadjuvant carrier vehicles has provided potential carriers for cancer immunotherapy. In this review article, we describe different types of immunoadjuvants, their limitations, modes of action, and the reasons for their clinical adoption. In addition, we review recent progress made in the nanoparticle-based immunoadjuvant field and on the combined use of nanoparticle-based immunoadjuvants and chemotherapy, phototherapy, radiation therapy, and immune checkpoint inhibitor-based therapy. STATEMENT OF SIGNIFICANCE: Cancer immunotherapy emerged as a new hope for treating malignant tumors. Different types of immunoadjuvants serve as an important tool for cancer immunotherapy by activating an innate or adaptive immune response. Limitation of free immunoadjuvant has paved the path for the development of nanoparticle-based immunoadjuvant therapy with the hope of prolonging the therapeutic efficacy. This review highlights the recent advancement made in nanoparticle-based immunoadjuvant therapy in modulating the adaptive and innate immune system. The application of the combinatorial approach of chemotherapy, phototherapy, radiation therapy adds synergy in nanoparticle-based immunoadjuvant therapy. It will broaden the reader's understanding on the recent progress made in immunotherapy with the aid of immunoadjuvant-based nanosystem.
Collapse
Affiliation(s)
- Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea.
| |
Collapse
|
12
|
Glynne-Jones R, Hall M, Nagtegaal ID. The optimal timing for the interval to surgery after short course preoperative radiotherapy (5 ×5 Gy) in rectal cancer - are we too eager for surgery? Cancer Treat Rev 2020; 90:102104. [PMID: 33002819 DOI: 10.1016/j.ctrv.2020.102104] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/04/2020] [Accepted: 09/06/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND The improved overall survival (OS) after short course preoperative radiotherapy (SCPRT) using 5 × 5 Gy reported in the early rectal cancer trials could not be replicated in subsequent phase III trials. This original survival advantage is attributed to poor quality of surgery and the large differential in local recurrence rates, with and without SCPRT. Immuno-modulation during and after SCPRT and its clinical implications have been poorly investigated. We propose an alternative explanation for this survival benefit in terms of immunological mechanisms induced by SCPRT and the timing of surgery, which may validate the concept of consolidation chemotherapy. MATERIAL AND METHODS We reviewed randomized controlled trials (RCTs) and studies of SCPRT from 1985 to 2019. We aimed to examine the precise timing of surgery in days following SCPRT and identify evidence for immune modulation, neo-antigens and memory cell induction by radiation. RESULTS Considerable variability is reported in randomised trials for median overall treatment time (OTT) from start of SCPRT to surgery (8-14 days). Only three early trials showed a benefit in terms of OS from SCPRT, although the level of benefit in preventing local recurrence was consistent across all trials. Different patterns of immune effects are observed within days after SCPRT depending on the OTT, but human leukocyte antigen (HLA)-1 expression was not upregulated. CONCLUSIONS SCPRT has a substantial immune-stimulatory potential. The importance of the timing of surgery after SCPRT may have been underestimated. An optimal interval for surgery after 5 × 5 Gy may lead to better outcomes, which is possibly exploited in total neoadjuvant therapy schedules using consolidation chemotherapy. Individual patient meta-analyses from appropriate SCPRT trials examining outcomes for each day and prospective trials are needed to clarify the validity of this hypothesis. The interaction of SCPRT with tumour adaptive immunology, in particular the kinetics and timing, should be examined further.
Collapse
Affiliation(s)
- R Glynne-Jones
- Radiotherapy Department, Mount Vernon Centre for Cancer Treatment, Mount Vernon Hospital, Northwood HA6 2RN, United Kingdom.
| | - M Hall
- Department of Medical Oncology, Mount Vernon Centre for Cancer Treatment, Mount Vernon Hospital, Northwood HA6 2RN, United Kingdom
| | - I D Nagtegaal
- Department of Pathology, Radboudumc, PO BOX 9101, 6500 HB Nijmegen, the Netherlands
| |
Collapse
|
13
|
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
|
14
|
Piper M, Mueller AC, Karam SD. The interplay between cancer associated fibroblasts and immune cells in the context of radiation therapy. Mol Carcinog 2020; 59:754-765. [PMID: 32363633 DOI: 10.1002/mc.23205] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023]
Abstract
Fibroblasts are a key component of the tumor microenvironment (TME) that can serve as a scaffold for tumor cell migration and augment the tumor's ability to withstand harsh conditions. When activated by external or endogenous stimuli, normal fibroblasts become cancer associated fibroblasts (CAFs), a heterogeneous group of stromal cells in the tumor that are phenotypically and epigenetically different from normal fibroblasts. Dynamic crosstalk between cancer cells, immune cells, and CAFs through chemokines and surface signaling makes the TME conducive to tumor growth. When activated, CAFs promote tumorigenesis and metastasis through several phenomena including regulation of tumor immunity, metabolic reprogramming of the TME, extracellular matrix remodeling and contraction, and induction of therapeutic resistance. Ionizing radiation (radiation theraphy [RT]) is a potent immunological stimulant that has been shown to increase cytotoxic Teff infiltration and IFN-I stimulated genes. RT, however, is unable to overcome the infiltration and activation of immunosuppressive cells which can contribute to tumor progression. Another paradox of RT is that, while very effective at killing cancer cells, it can contribute to the formation of CAFs. This review examines how the interplay between CAFs and immune cells during RT contributes to organ fibrosis, immunosuppression, and tumor growth. We focus on targeting mechanistic pathways of CAF formation as a potentially effective strategy not only for preventing organ fibrosis, but also in hampering tumor progression in response to RT.
Collapse
Affiliation(s)
- Miles Piper
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Adam C Mueller
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
15
|
Sufficiency of CD40 activation and immune checkpoint blockade for T cell priming and tumor immunity. Proc Natl Acad Sci U S A 2020; 117:8022-8031. [PMID: 32213589 DOI: 10.1073/pnas.1918971117] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Innate immune receptors such as toll-like receptors (TLRs) provide critical molecular links between innate cells and adaptive immune responses. Here, we studied the CD40 pathway as an alternative bridge between dendritic cells (DCs) and adaptive immunity in cancer. Using an experimental design free of chemo- or radiotherapy, we found CD40 activation with agonistic antibodies (⍺CD40) produced complete tumor regressions in a therapy-resistant pancreas cancer model, but only when combined with immune checkpoint blockade (ICB). This effect, unachievable with ICB alone, was independent of TLR, STING, or IFNAR pathways. Mechanistically, αCD40/ICB primed durable T cell responses, and efficacy required DCs and host expression of CD40. Moreover, ICB drove optimal generation of polyfunctional T cells in this "cold" tumor model, instead of rescuing T cell exhaustion. Thus, immunostimulation via αCD40 is sufficient to synergize with ICB for priming. Clinically, combination αCD40/ICB may extend efficacy in patients with "cold" and checkpoint-refractory tumors.
Collapse
|
16
|
Lai X, Friedman A. Mathematical modeling of cancer treatment with radiation and PD-L1 inhibitor. SCIENCE CHINA MATHEMATICS 2020; 63:465-484. [DOI: 10.1007/s11425-019-1648-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/27/2020] [Indexed: 01/04/2025]
|
17
|
Ye J, Mills BN, Zhao T, Han BJ, Murphy JD, Patel AP, Johnston CJ, Lord EM, Belt BA, Linehan DC, Gerber SA. Assessing the Magnitude of Immunogenic Cell Death Following Chemotherapy and Irradiation Reveals a New Strategy to Treat Pancreatic Cancer. Cancer Immunol Res 2020; 8:94-107. [PMID: 31719057 PMCID: PMC6946873 DOI: 10.1158/2326-6066.cir-19-0373] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/18/2019] [Accepted: 11/07/2019] [Indexed: 12/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to have a dismal prognosis, in part, due to ineffective treatment strategies. The efficacy of some chemotherapies and especially radiotherapy is mediated partially by the immune system. Therefore, we hypothesized that profiling the immune response following chemotherapy and/or irradiation can be used as a readout for treatment efficacy but also to help identify optimal therapeutic schedules for PDAC. Using murine models of PDAC, we demonstrated that concurrent administration of stereotactic body radiotherapy (SBRT) and a modified dose of FOLFIRINOX (mFX) resulted in superior tumor control when compared with single or sequential treatment groups. Importantly, this combined treatment schedule enhanced the magnitude of immunogenic cell death, which in turn amplified tumor antigen presentation by dendritic cells and intratumoral CD8+ T-cell infiltration. Concurrent therapy also resulted in systemic immunity contributing to the control of established metastases. These findings provide a rationale for pursuing concurrent treatment schedules of SBRT with mFX in PDAC.
Collapse
Affiliation(s)
- Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Bradley N Mills
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Tony Zhao
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Booyeon J Han
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Joseph D Murphy
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Ankit P Patel
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Carl J Johnston
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Edith M Lord
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Brian A Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - David C Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Scott A Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, New York.
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
18
|
Targets for improving tumor response to radiotherapy. Int Immunopharmacol 2019; 76:105847. [PMID: 31466051 DOI: 10.1016/j.intimp.2019.105847] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/18/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023]
|
19
|
Feng X, Xu W, Li Z, Song W, Ding J, Chen X. Immunomodulatory Nanosystems. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900101. [PMID: 31508270 PMCID: PMC6724480 DOI: 10.1002/advs.201900101] [Citation(s) in RCA: 245] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/21/2019] [Indexed: 05/15/2023]
Abstract
Immunotherapy has emerged as an effective strategy for the prevention and treatment of a variety of diseases, including cancer, infectious diseases, inflammatory diseases, and autoimmune diseases. Immunomodulatory nanosystems can readily improve the therapeutic effects and simultaneously overcome many obstacles facing the treatment method, such as inadequate immune stimulation, off-target side effects, and bioactivity loss of immune agents during circulation. In recent years, researchers have continuously developed nanomaterials with new structures, properties, and functions. This Review provides the most recent advances of nanotechnology for immunostimulation and immunosuppression. In cancer immunotherapy, nanosystems play an essential role in immune cell activation and tumor microenvironment modulation, as well as combination with other antitumor approaches. In infectious diseases, many encouraging outcomes from using nanomaterial vaccines against viral and bacterial infections have been reported. In addition, nanoparticles also potentiate the effects of immunosuppressive immune cells for the treatment of inflammatory and autoimmune diseases. Finally, the challenges and prospects of applying nanotechnology to modulate immunotherapy are discussed.
Collapse
Affiliation(s)
- Xiangru Feng
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
- University of Science and Technology of ChinaHefei230026P. R. China
| | - Weiguo Xu
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| | - Zhongmin Li
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
- Department of Gastrointestinal Colorectal and Anal SurgeryChina–Japan Union Hospital of Jilin UniversityChangchun130033P. R. China
| | - Wantong Song
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| |
Collapse
|
20
|
Akita H, Takahashi H, Asukai K, Tomokuni A, Wada H, Marukawa S, Yamasaki T, Yanagimoto Y, Takahashi Y, Sugimura K, Yamamoto K, Nishimura J, Yasui M, Omori T, Miyata H, Ochi A, Kagawa A, Soh Y, Taniguchi Y, Ohue M, Yano M, Sakon M. The utility of nutritional supportive care with an eicosapentaenoic acid (EPA)-enriched nutrition agent during pre-operative chemoradiotherapy for pancreatic cancer: Prospective randomized control study. Clin Nutr ESPEN 2019; 33:148-153. [PMID: 31451252 DOI: 10.1016/j.clnesp.2019.06.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 03/10/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Neoadjuvant chemoradiotherapy (NACRT) for pancreatic cancer (PC) is potentially associated with various toxicities, which can lead to impaired nutritional status. Eicosapentaenoic acid (EPA) can reduce proinflammatory cytokines and positively influence cancer cachexia syndrome. The aim of this study is to clarify the utility of EPA enriched nutrition support during NACRT for PC. METHODS We randomly assigned 62 patients with PC that received NACRT to either a nutrition intervention (NI) or a normal diet (ND). Patients in the NI group received 2 bottles/day (550 kcal/day) of an EPA-enriched nutrition supplement during NACRT. The primary endpoints were the before-to-after NACRT ratios (post/pre ratios) of skeletal muscle mass and psoas major muscle area (PMA). The secondary endpoints were the post/pre ratios of other nutritional parameters and treatment-related toxicities. RESULTS Only 14 patients (45.2%) in the NI group consumed more than 50% of the EPA-enriched supplement provided. The post/pre ratio of skeletal muscle mass in the NI group (0.99 ± 0.060) was not significantly different from that of the ND group (0.96 ± 0.079, p = 0.102). However, patients that consumed ≥50% of the EPA-enriched supplement (the good intake group) had significantly higher skeletal muscle mass ratios than patients in the ND group (p = 0.042). The PMA ratio was significantly higher in the NI group (0.96 ± 0.081) than in the ND group (0.89 ± 0.072, p = 0.001). The NI and ND groups were not significantly different in other nutritional parameters or in NACRT-related toxicity. CONCLUSIONS We found that EPA-enriched intake could potentially improve the nutritional status of patients with PC that received NACRT, but it was difficult for many patients to drink, due to its disagreeable taste. University Hospital Medical Information Network (http://www.umin.ac.jp), registration number UMIN000033589, https://upload.umin.ac.jp/cgi-bin/ctr_e/ctr_view.cgi?recptno=R000038300.
Collapse
Affiliation(s)
- Hirofumi Akita
- Department of Surgery, Osaka International Cancer Institute, Japan; Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Japan.
| | | | - Kei Asukai
- Department of Surgery, Osaka International Cancer Institute, Japan
| | - Akira Tomokuni
- Department of Surgery, Osaka International Cancer Institute, Japan
| | - Hiroshi Wada
- Department of Surgery, Osaka International Cancer Institute, Japan
| | - Satoko Marukawa
- Department of Endocrinology and Metabolism, Osaka International Cancer Institute, Japan
| | - Tomoyuki Yamasaki
- Department of Endocrinology and Metabolism, Osaka International Cancer Institute, Japan
| | | | - Yusuke Takahashi
- Department of Surgery, Osaka International Cancer Institute, Japan
| | - Keijiro Sugimura
- Department of Surgery, Osaka International Cancer Institute, Japan
| | | | | | - Masayoshi Yasui
- Department of Surgery, Osaka International Cancer Institute, Japan
| | - Takeshi Omori
- Department of Surgery, Osaka International Cancer Institute, Japan
| | - Hiroshi Miyata
- Department of Surgery, Osaka International Cancer Institute, Japan
| | - Ayami Ochi
- Department of Nursing, Osaka International Cancer Institute, Japan
| | - Ayano Kagawa
- Department of Nursing, Osaka International Cancer Institute, Japan
| | - Yuko Soh
- Department of Nutrition, Osaka International Cancer Institute, Japan
| | - Yuko Taniguchi
- Department of Nutrition, Osaka International Cancer Institute, Japan
| | - Masayuki Ohue
- Department of Surgery, Osaka International Cancer Institute, Japan
| | - Masahiko Yano
- Department of Surgery, Osaka International Cancer Institute, Japan
| | - Masato Sakon
- Department of Surgery, Osaka International Cancer Institute, Japan
| |
Collapse
|
21
|
Template-assisted 192Ir-based stereotactic ablative brachytherapy as a neoadjuvant treatment for operable peripheral non-small cell lung cancer: a phase I clinical trial. J Contemp Brachytherapy 2019; 11:162-168. [PMID: 31139225 PMCID: PMC6536138 DOI: 10.5114/jcb.2019.84613] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/18/2019] [Indexed: 12/02/2022] Open
Abstract
Purpose To evaluate safety, feasibility, and efficacy of template-assisted 192Ir-based stereotactic ablative brachytherapy (SABT), combined with surgery for peripheral non-small cell lung cancer (NSCLC). Material and methods Patients with pathologically confirmed operable peripheral NSCLC, who underwent template-assisted SABT (30 Gy delivered in one fraction) and were scheduled for tumor resection 4-6 weeks after SABT were included in this study. The perioperative adverse reactions of SABT were recorded to evaluate safety and feasibility of SABT for neoadjuvant therapy. Dosimetric data from both simulated and actual plans were collected and compared. Imaging with 18F-fluorodeoxyglucose-positron emission tomography/computed tomography (18F-FDG-PET/CT) and dynamic contrast-enhanced computed tomography were scheduled before SABT and surgery to evaluate the efficacy of the neoadjuvant therapy with SABT. Results Patients did not experience any serious adverse events. None of the patients had a delay in receiving surgery. After 4-6 weeks, the indicators for the efficacy of neoadjuvant therapy significantly decreased in all patients: gross tumor volume (p < 0.001), maximum standardized uptake value (p < 0.001), tumor blood volume (p < 0.001), and tumor blood flow (p = 0.008). Dosimetric parameters in the delivered SABT plan slightly changed from the preoperative simulation, but the difference was not statistically significant (p > 0.05). Conclusions The efficacy of template-assisted SABT for neoadjuvant therapy was significant in operable peripheral NSCLC. Moreover, no serious adverse reactions were observed; when the coplanar template guidance technique was applied, dosimetric parameters were in good agreement between the actual SABT plan and the preoperative simulated plan.
Collapse
|
22
|
Chakwizira A, Ahlstedt J, Nittby Redebrandt H, Ceberg C. Mathematical modelling of the synergistic combination of radiotherapy and indoleamine-2,3-dioxygenase (IDO) inhibitory immunotherapy against glioblastoma. Br J Radiol 2018; 91:20170857. [PMID: 29688039 PMCID: PMC6221783 DOI: 10.1259/bjr.20170857] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Objective: Recent research has shown that combining radiotherapy and immunotherapy can counteract the ability of cancer to evade and suppress the native immune system. To optimise the synergy of the combined therapies, factors such as radiation dose and fractionation must be considered, alongside numerous parameters resulting from the complexity of cancer–immune system interactions. It is instructive to use mathematical models to tackle this problem. Methods: In this work, we adapted a model primarily to describe the synergistic effect between single-fraction radiotherapy and immunotherapy (1-methyl tryptophan) observed in previous experiments with glioblastoma-carrying rats. We also showed how the model can be used to generate hypotheses on the outcome of other treatment fractionation schemes. Results: The model successfully reproduced the results of the experiments. Moreover, it provided support for the hypothesis that, for a given biologically effective dose, the efficacy of the combination therapy and the synergy between the two therapies are favoured by the administration of radiotherapy in a hypofractionated regime. Furthermore, for a double-fraction irradiation regimen, the synergy is favoured by a short time interval between the treatment fractions. Conclusion: It was concluded that the model could be fitted to reproduce the experimental data well within its uncertainties. It was also demonstrated that the fitted model can be used to form hypotheses to be validated by further pre-clinical experiments. Advances in knowledge: The results of this work support the hypothesis that the synergetic action of combined radiotherapy and immunotherapy is favoured by using a hypofractionated radiation treatment regimen, given over a short time interval.
Collapse
Affiliation(s)
- Arthur Chakwizira
- 1 Medical Radiation Physics, Department of Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Jonatan Ahlstedt
- 2 Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Henrietta Nittby Redebrandt
- 2 Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Crister Ceberg
- 1 Medical Radiation Physics, Department of Clinical Sciences Lund, Lund University , Lund , Sweden
| |
Collapse
|
23
|
McGinnis GJ, Friedman D, Young KH, Torres ERS, Thomas CR, Gough MJ, Raber J. Neuroinflammatory and cognitive consequences of combined radiation and immunotherapy in a novel preclinical model. Oncotarget 2018; 8:9155-9173. [PMID: 27893434 PMCID: PMC5354722 DOI: 10.18632/oncotarget.13551] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/14/2016] [Indexed: 11/25/2022] Open
Abstract
Background Cancer patients often report behavioral and cognitive changes following cancer treatment. These effects can be seen in patients who have not yet received treatment or have received only peripheral (non-brain) irradiation. Novel treatments combining radiotherapy (RT) and immunotherapy (IT) demonstrate remarkable efficacy with respect to tumor outcomes by enhancing the proinflammatory environment in the tumor. However, a proinflammatory environment in the brain mediates cognitive impairments in other neurological disorders and may affect brain function in cancer patients receiving these novel treatments. Currently, gaps exist as to whether these treatments impact the brain in individuals with or without tumors and with regard to the underlying mechanisms. Results Combined treatment with precision RT and checkpoint inhibitor IT achieved control of tumor growth. However, BALB/c mice receiving combined treatment demonstrated changes in measures of anxiety levels, regardless of tumor status. C57BL/6J mice with tumors demonstrated increased anxiety, except following combined treatment. Object recognition memory was impaired in C57BL/6J mice without tumors following combined treatment. All mice with tumors showed impaired object recognition, except those treated with RT alone. Mice with tumors demonstrated impaired amygdala-dependent cued fear memory, while maintaining hippocampus-dependent context fear memory. These behavioral alterations and cognitive impairments were accompanied by increased microglial activation in mice receiving immunotherapy alone or combined with RT. Finally, based on tumor status, there were significant changes in proinflammatory cytokines (IFN-γ, IL-6, IL-5, IL-2, IL-10) and a growth factor (FGF-basic). Materials and Methods Here we test the hypothesis that IT combined with peripheral RT have detrimental behavioral and cognitive effects as a result of an enhanced proinflammatory environment in the brain. BALB/c mice with or without injected hind flank CT26 colorectal carcinoma or C57BL/6J mice with or without Lewis Lung carcinoma were used for all experiments. Checkpoint inhibitor IT, using an anti-CTLA-4 antibody, and precision CT-guided peripheral RT alone and combined were used to closely model clinical treatment. We assessed behavioral and cognitive performance and investigated the immune environment using immunohistochemistry and multiplex assays to analyze proinflammatory mediators. Conclusions Although combined treatment achieved tumor growth control, it affected the brain and induced changes in measures of anxiety, cognitive impairments, and neuroinflammation.
Collapse
Affiliation(s)
- Gwendolyn J McGinnis
- Howard Hughes Medical Institute, Oregon Health and Science University, Portland, OR.,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR.,Department of Radiation Medicine, Oregon Health and Science University, Portland, OR
| | - David Friedman
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR
| | - Kristina H Young
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR
| | - Eileen Ruth S Torres
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR
| | - Charles R Thomas
- Department of Radiation Medicine, Oregon Health and Science University, Portland, OR
| | - Michael J Gough
- Department of Radiation Medicine, Oregon Health and Science University, Portland, OR.,Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR.,Department of Radiation Medicine, Oregon Health and Science University, Portland, OR.,Department of Neurology, Oregon Health and Science University, Portland, OR.,Division of Neuroscience, Oregon National Primate Research Center, Portland, OR
| |
Collapse
|
24
|
A Dendritic Cell Vaccine Combined With Radiotherapy Activates the Specific Immune Response in Patients With Esophageal Cancer. J Immunother 2018; 40:71-76. [PMID: 28125513 DOI: 10.1097/cji.0000000000000155] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Dendritic cells (DC) are highly efficient antigen-presenting cells. DC may be used to create DC vaccines against cancer, but the optimal strategies remain to be elucidated. This study aimed to examine the benefits and adverse effects of using esophageal cancer cell antigens to stimulate DC to trigger the specific immune response in patients with esophageal cancer undergoing radiotherapy. This was an observational cohort study performed at Lianshui County People's Hospital between September 2010 and June 2012. Forty patients with esophageal cancer planned to receive radiotherapy were selected, and 28 received the DC vaccine. DC were isolated, loaded with antigens, and intradermally injected after being cultured for 1 week. One week after injection, the patients underwent a delayed-type hypersensitivity test. Serum Th1 cytokines [interleukin (IL)-2, IL-12, and interferon (IFN)-γ] and antigen-specific IFN-γCD8 T cells were tested before and after vaccination. Patients were followed up for 2 years. Adverse events were monitored. Patients in the vaccine group tolerated the DC vaccine. Levels of serum IL-2 (+92.4%), IL-12 (+70.9%), and IFN-γ (+214.3%) as well as the proportion of IFN-γCD8 T cells (3.0-16.4-fold) were significantly increased compared with baseline and the control group (all P<0.05). The 1- (82.1% vs. 50.0%, P=0.04) and 2-year survival (67.8% vs. 33.3%, P=0.04) was improved by vaccination. Only 2 patients showed mild fever. In conclusion, the DC vaccine triggered the specific immune response and induced the secretion of Th1 cytokines. The vaccine may lead to better survival, but this have to be confirmed. Adverse events were rare and mild.
Collapse
|
25
|
NF-kappaB: Two Sides of the Same Coin. Genes (Basel) 2018; 9:genes9010024. [PMID: 29315242 PMCID: PMC5793177 DOI: 10.3390/genes9010024] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/02/2018] [Accepted: 01/05/2018] [Indexed: 01/05/2023] Open
Abstract
Nuclear Factor-kappa B (NF-κB) is a transcription factor family that regulates a large number of genes that are involved in important physiological processes, including survival, inflammation, and immune responses. More recently, constitutive expression of NF-κB has been associated with several types of cancer. In addition, microorganisms, such as viruses and bacteria, cooperate in the activation of NF-κB in tumors, confirming the multifactorial role of this transcription factor as a cancer driver. Recent reports have shown that the NF-κB signaling pathway should receive attention for the development of therapies. In addition to the direct effects of NF-κB in cancer cells, it might also impact immune cells that can both promote or prevent tumor development. Currently, with the rise of cancer immunotherapy, the link among immune cells, inflammation, and cancer is a major focus, and NF-κB could be an important regulator for the success of these therapies. This review discusses the contrasting roles of NF-κB as a regulator of pro- and antitumor processes and its potential as a therapeutic target.
Collapse
|
26
|
Perceived stress moderates the effects of a randomized trial of dance movement therapy on diurnal cortisol slopes in breast cancer patients. Psychoneuroendocrinology 2018; 87:119-126. [PMID: 29059542 DOI: 10.1016/j.psyneuen.2017.10.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/12/2017] [Accepted: 10/12/2017] [Indexed: 12/22/2022]
Abstract
Women with breast cancer are at risk of psychosocial distress and may suffer from aberrant diurnal cortisol rhythms. Dance movement therapy (DMT), a movement-based psychotherapy that incorporates exercise and artistic components, has demonstrated stress reduction effects. This study examined the effects of DMT on the diurnal cortisol rhythms of breast cancer patients undergoing radiotherapy treatment and the role of perceived stress in producing such effects. The study sample comprised 121 Chinese breast cancer patients randomized to the DMT (n=63) and control (n=58) groups. The intervention consisted of six 1.5-h group sessions held twice weekly over the course of radiotherapy. Participants completed validated self-report measures of perceived stress, fatigue, pain, and sleep disturbance and provided five salivary cortisol samples at baseline (Time 1) and post-intervention (Time 2). Moderated mediation analysis was used to evaluate the intervention effect on Time 2 diurnal cortisol slopes. Despite the absence of a significant DMT effect on diurnal cortisol slopes (B=-0.55, 95% CI=-1.20 to 0.08, β=-0.14), baseline perceived stress significantly moderated the intervention effect (B=-0.18, 95% CI=-0.32 to -0.05, β=-0.30). At high levels of baseline perceived stress (1 SD above the mean), the DMT group showed a steeper cortisol slope (M=-7.14) than the control group (M=-5.80) at Time 2. The present findings suggest that DMT might have a beneficial effect on diurnal cortisol slopes in breast cancer patients with high levels of distress.
Collapse
|
27
|
McGinnis GJ, Raber J. CNS side effects of immune checkpoint inhibitors: preclinical models, genetics and multimodality therapy. Immunotherapy 2017; 9:929-941. [PMID: 29338610 PMCID: PMC6161123 DOI: 10.2217/imt-2017-0056] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/09/2017] [Indexed: 02/06/2023] Open
Abstract
Following cancer treatment, patients often report behavioral and cognitive changes. Novel cancer immunotherapeutics have the potential to produce sustained cancer survivorship, meaning patients will live longer with the side effects of treatment. Given the role of inflammatory pathways in mediating behavioral and cognitive impairments seen in cancer, we aim in this review to discuss emerging evidence for the contribution of immune checkpoint blockade to exacerbate these CNS effects. We discuss ongoing studies regarding the ability of immune checkpoint inhibitors to reach the brain and how treatment responses to checkpoint inhibitors may be modulated by genetic factors. We further consider the use of preclinical tumor-models to study the role of tumor status in CNS effects of immune checkpoint inhibitors and multimodality therapy.
Collapse
Affiliation(s)
- Gwendolyn J McGinnis
- Department of Radiation Medicine, Oregon Health & Science University, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, OR, USA
| | - Jacob Raber
- Department of Radiation Medicine, Oregon Health & Science University, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, OR, USA
- Department of Neurology, Oregon Health & Science University, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185th Ave, Beaverton, OR 97006, USA
| |
Collapse
|
28
|
Janiak MK, Wincenciak M, Cheda A, Nowosielska EM, Calabrese EJ. Cancer immunotherapy: how low-level ionizing radiation can play a key role. Cancer Immunol Immunother 2017; 66:819-832. [PMID: 28361232 PMCID: PMC5489643 DOI: 10.1007/s00262-017-1993-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 03/22/2017] [Indexed: 12/17/2022]
Abstract
The cancer immunoediting hypothesis assumes that the immune system guards the host against the incipient cancer, but also "edits" the immunogenicity of surviving neoplastic cells and supports remodeling of tumor microenvironment towards an immunosuppressive and pro-neoplastic state. Local irradiation of tumors during standard radiotherapy, by killing neoplastic cells and generating inflammation, stimulates anti-cancer immunity and/or partially reverses cancer-promoting immunosuppression. These effects are induced by moderate (0.1-2.0 Gy) or high (>2 Gy) doses of ionizing radiation which can also harm normal tissues, impede immune functions, and increase the risk of secondary neoplasms. In contrast, such complications do not occur with exposures to low doses (≤0.1 Gy for acute irradiation or ≤0.1 mGy/min dose rate for chronic exposures) of low-LET ionizing radiation. Furthermore, considerable evidence indicates that such low-level radiation (LLR) exposures retard the development of neoplasms in humans and experimental animals. Here, we review immunosuppressive mechanisms induced by growing tumors as well as immunomodulatory effects of LLR evidently or likely associated with cancer-inhibiting outcomes of such exposures. We also offer suggestions how LLR may restore and/or stimulate effective anti-tumor immunity during the more advanced stages of carcinogenesis. We postulate that, based on epidemiological and experimental data amassed over the last few decades, whole- or half-body irradiations with LLR should be systematically examined for its potential to be a viable immunotherapeutic treatment option for patients with systemic cancer.
Collapse
Affiliation(s)
- Marek K Janiak
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland.
| | - Marta Wincenciak
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| | - Aneta Cheda
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| | - Ewa M Nowosielska
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| | - Edward J Calabrese
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
29
|
Chen J, Tian X, Mei Z, Wang Y, Yao Y, Zhang S, Li X, Wang H, Zhang J, Xie C. The effect of the TLR9 ligand CpG-oligodeoxynucleotide on the protective immune response to radiation-induced lung fibrosis in mice. Mol Immunol 2016; 80:33-40. [PMID: 27825048 DOI: 10.1016/j.molimm.2016.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 10/26/2016] [Accepted: 11/01/2016] [Indexed: 12/18/2022]
Abstract
CpG-oligodeoxynucleotide (CpG-ODN) is not only reported to protect against airway hyper responsiveness but is also known as a potent vaccine adjuvant for anti-tumor therapy. Little is known about the effect of CpG-ODN in mice with radiation-induced lung fibrosis (RILF), a common late stage form of tissue damage that occurs after thorax radiotherapy (RT). Here, we evaluated the immunomodulatory effects of CpG-ODN on the development of RILF. Mice were divided into four groups: (1) RT, single dose of 12Gy to the whole thorax; (2) CpG, only intraperitoneal injection of CpG-ODN for total 5 weeks; (3) RT+CpG, irradiation plus CpG-ODN treatment before and after irradiation for total 5 weeks; and (4) control (CTL): No RT or CpG-ODN treatment. In this study, we found that CpG-ODN treatment attenuated lung fibrosis and collagen deposition by increasing the number of M1 macrophagocytes, levels of Type-2 cytokines and TGF-β. CpG-ODN administration up-regulated the expression of TLR9 and STAT1 phosphorylation and reversed the expression of Type-2 immune response key transcription factor GATA-3. Activation of the JAK-STAT1 signaling pathway further enhanced M1 macrophage differentiation and Type-1 cytokine production. This study reveals the mitigating effect of early exposure to CpG-ODN on lung injury caused by irradiation in mice. The potential mechanism of action may be related to enhancement of Type-1 immunity. In conclusion, CpG-ODN may be a potential therapeutic target to treat RILF.
Collapse
Affiliation(s)
- Jing Chen
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China
| | - Xiaoli Tian
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China
| | - Zijie Mei
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China
| | - Yacheng Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China
| | - Ye Yao
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China
| | - Shimin Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China
| | - Xin Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China
| | - Hui Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China
| | - Junhong Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China.
| |
Collapse
|
30
|
Abstract
Tumour-promoting inflammation is considered one of the enabling characteristics of cancer development. Chronic inflammatory disease increases the risk of some cancers, and strong epidemiological evidence exists that NSAIDs, particularly aspirin, are powerful chemopreventive agents. Tumour microenvironments contain many different inflammatory cells and mediators; targeting these factors in genetic, transplantable and inducible murine models of cancer substantially reduces the development, growth and spread of disease. Thus, this complex network of inflammation offers targets for prevention and treatment of malignant disease. Much potential exists in this area for novel cancer prevention and treatment strategies, although clinical research to support targeting of cancer-related inflammation and innate immunity in patients with advanced-stage cancer remains in its infancy. Following the initial successes of immunotherapies that modulate the adaptive immune system, we assert that inflammation and innate immunity are important targets in patients with cancer on the basis of extensive preclinical and epidemiological data. The adaptive immune response is heavily dependent on innate immunity, therefore, inhibiting some of the tumour-promoting immunosuppressive actions of the innate immune system might enhance the potential of immunotherapies that activate a nascent antitumour response.
Collapse
Affiliation(s)
- Shanthini M Crusz
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Frances R Balkwill
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
31
|
De Tollenaere C, Lievens Y, Vandecasteele K, Vermaelen K, Surmont V. Unresectable stage III non-small-cell lung cancer: Have we made any progress? World J Respirol 2015; 5:140-151. [DOI: 10.5320/wjr.v5.i2.140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/27/2015] [Accepted: 06/08/2015] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is responsible for the most cancer deaths worldwide with an incidence that is still rising. One third of patients have unresectable stage IIIA or stage IIIB disease. The standard of care for locally advanced disease in patients with good performance status consists of combined modality therapy in particular concurrent chemoradiotherapy. But despite a lot of efforts done in the past, local control and survival of patients with unresectable stage III non-small-cell lung cancer (NSCLC) remains poor. Improving outcomes for patients with unresectable stage III NSCLC has therefore been an area of ongoing research. Research has focused on improving systemic therapy, improving radiation therapy or adding a maintenance therapy to consolidate the initial therapy. Also implementation of newer targeted therapies and immunotherapy has been investigated as well as the option of prophylactic cranial irradiation. This article reviews the latest literature on improving local control and preventing distant metastases. It seems that we have reached a plateau with conventional chemotherapy. Radiotherapy dose escalation did not improve outcome although increasing radiation dose-intensity with new radiotherapy techniques and the use of newer agents, e.g., immunotherapy might be promising. In the future well-designed clinical trials are necessary to prove those promising results.
Collapse
|
32
|
Modulation of dendritic cell function by the radiation-mediated secretory protein γ-synuclein. Cell Death Discov 2015; 1:15011. [PMID: 27551446 PMCID: PMC4979407 DOI: 10.1038/cddiscovery.2015.11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 12/18/2022] Open
Abstract
Recently, γ-synuclein (SNCG), which is also known as breast cancer-specific gene-1, has been demonstrated to be an adverse and aggressive marker in breast cancer. In our previous study, SNCG was significantly upregulated in irradiated human breast cancer cells. The aim of this study was to investigate whether radiation-induced, tumor-derived SNCG can influence dendritic cell (DC) function in immune systems. The phenotypical and functional changes of DCs in the presence or absence of SNCG were investigated by FACS analysis, ELISA, and real-time PCR. The ability of SNCG-treated DCs to influence T cells was also examined by coculturing with T cells. The treatment of DCs with SNCG protein inhibited the surface expression of the co-stimulatory molecules CD40 and CD86, and decreased the mRNA levels of pro-inflammatory cytokines. The SNCG-treated DCs inhibited T-cell proliferation slightly, but distinctively increased the population of regulatory T cells. In addition, the production of TGF-β from T cells was significantly increased when they were cocultured with SNCG-treated DCs. Taken together, these results demonstrate that tumor-derived SNCG contributes to immunosuppressive effects via the inhibition of DC differentiation and activation, thus making it a potential target for cancer treatment.
Collapse
|
33
|
Barker HE, Paget JTE, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer 2015; 15:409-25. [PMID: 26105538 PMCID: PMC4896389 DOI: 10.1038/nrc3958] [Citation(s) in RCA: 1492] [Impact Index Per Article: 149.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Radiotherapy plays a central part in curing cancer. For decades, most research on improving treatment outcomes has focused on modulating radiation-induced biological effects on cancer cells. Recently, we have better understood that components within the tumour microenvironment have pivotal roles in determining treatment outcomes. In this Review, we describe vascular, stromal and immunological changes that are induced in the tumour microenvironment by irradiation and discuss how these changes may promote radioresistance and tumour recurrence. We also highlight how this knowledge is guiding the development of new treatment paradigms in which biologically targeted agents will be combined with radiotherapy.
Collapse
Affiliation(s)
- Holly E. Barker
- Targeted Therapy Team, The Institute of Cancer Research, London, SW3 6JB, UK
| | - James T. E. Paget
- Targeted Therapy Team, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Aadil A. Khan
- Targeted Therapy Team, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Kevin J. Harrington
- Targeted Therapy Team, The Institute of Cancer Research, London, SW3 6JB, UK
| |
Collapse
|
34
|
Datta J, Berk E, Cintolo JA, Xu S, Roses RE, Czerniecki BJ. Rationale for a Multimodality Strategy to Enhance the Efficacy of Dendritic Cell-Based Cancer Immunotherapy. Front Immunol 2015; 6:271. [PMID: 26082780 PMCID: PMC4451636 DOI: 10.3389/fimmu.2015.00271] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/15/2015] [Indexed: 02/03/2023] Open
Abstract
Dendritic cells (DC), master antigen-presenting cells that orchestrate interactions between the adaptive and innate immune arms, are increasingly utilized in cancer immunotherapy. Despite remarkable progress in our understanding of DC immunobiology, as well as several encouraging clinical applications – such as DC-based sipuleucel-T for metastatic castration-resistant prostate cancer – clinically effective DC-based immunotherapy as monotherapy for a majority of tumors remains a distant goal. The complex interplay between diverse molecular and immune processes that govern resistance to DC-based vaccination compels a multimodality approach, encompassing a growing arsenal of antitumor agents which target these distinct processes and synergistically enhance DC function. These include antibody-based targeted molecular therapies, immune checkpoint inhibitors, therapies that inhibit immunosuppressive cellular elements, conventional cytotoxic modalities, and immune potentiating adjuvants. It is likely that in the emerging era of “precision” cancer therapeutics, tangible clinical benefits will only be realized with a multifaceted – and personalized – approach combining DC-based vaccination with adjunctive strategies.
Collapse
Affiliation(s)
- Jashodeep Datta
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Erik Berk
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Jessica A Cintolo
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Shuwen Xu
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Robert E Roses
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Brian J Czerniecki
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA ; Rena Rowen Breast Center, Hospital of the University of Pennsylvania , Philadelphia, PA , USA
| |
Collapse
|
35
|
Ridnour LA, Cheng RYS, Weiss JM, Kaur S, Soto-Pantoja DR, Basudhar D, Heinecke JL, Stewart CA, DeGraff W, Sowers AL, Thetford A, Kesarwala AH, Roberts DD, Young HA, Mitchell JB, Trinchieri G, Wiltrout RH, Wink DA. NOS Inhibition Modulates Immune Polarization and Improves Radiation-Induced Tumor Growth Delay. Cancer Res 2015; 75:2788-99. [PMID: 25990221 DOI: 10.1158/0008-5472.can-14-3011] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/08/2015] [Indexed: 12/24/2022]
Abstract
Nitric oxide synthases (NOS) are important mediators of progrowth signaling in tumor cells, as they regulate angiogenesis, immune response, and immune-mediated wound healing. Ionizing radiation (IR) is also an immune modulator and inducer of wound response. We hypothesized that radiation therapeutic efficacy could be improved by targeting NOS following tumor irradiation. Herein, we show enhanced radiation-induced (10 Gy) tumor growth delay in a syngeneic model (C3H) but not immunosuppressed (Nu/Nu) squamous cell carcinoma tumor-bearing mice treated post-IR with the constitutive NOS inhibitor N(G)-nitro-l-arginine methyl ester (L-NAME). These results suggest a requirement of T cells for improved radiation tumor response. In support of this observation, tumor irradiation induced a rapid increase in the immunosuppressive Th2 cytokine IL10, which was abated by post-IR administration of L-NAME. In vivo suppression of IL10 using an antisense IL10 morpholino also extended the tumor growth delay induced by radiation in a manner similar to L-NAME. Further examination of this mechanism in cultured Jurkat T cells revealed L-NAME suppression of IR-induced IL10 expression, which reaccumulated in the presence of exogenous NO donor. In addition to L-NAME, the guanylyl cyclase inhibitors ODQ and thrombospondin-1 also abated IR-induced IL10 expression in Jurkat T cells and ANA-1 macrophages, which further suggests that the immunosuppressive effects involve eNOS. Moreover, cytotoxic Th1 cytokines, including IL2, IL12p40, and IFNγ, as well as activated CD8(+) T cells were elevated in tumors receiving post-IR L-NAME. Together, these results suggest that post-IR NOS inhibition improves radiation tumor response via Th1 immune polarization within the tumor microenvironment.
Collapse
Affiliation(s)
- Lisa A Ridnour
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| | - Robert Y S Cheng
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jonathan M Weiss
- Cancer Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - David R Soto-Pantoja
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Debashree Basudhar
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Julie L Heinecke
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - C Andrew Stewart
- Cancer Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - William DeGraff
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Anastasia L Sowers
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Angela Thetford
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Aparna H Kesarwala
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Howard A Young
- Cancer Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Giorgio Trinchieri
- Cancer Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - Robert H Wiltrout
- Cancer Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - David A Wink
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
36
|
Hekim N, Cetin Z, Nikitaki Z, Cort A, Saygili EI. Radiation triggering immune response and inflammation. Cancer Lett 2015; 368:156-63. [PMID: 25911239 DOI: 10.1016/j.canlet.2015.04.016] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 04/13/2015] [Accepted: 04/15/2015] [Indexed: 12/23/2022]
Abstract
Radiation therapy (RT) is a well-established but still under optimization branch of Cancer Therapy (CT). RT uses electromagnetic waves or charged particles in order to kill malignant cells, by accumulating the energy onto these cells. The issue at stake for RT, as well as for any other Cancer Therapy technique, is always to kill only cancer cells, without affecting the surrounding healthy ones. This perspective of CT is usually described under the terms "specificity" and "selectivity". Specificity and selectivity are the ideal goal, but the ideal is never entirely achieved. Thus, in addition to killing healthy cells, changes and effects are observed in the immune system after irradiation. In this review, we mainly focus on the effects of ionizing radiation on the immune system and its components like bone marrow. Additionally, we are interested in the effects and benefits of low-dose ionizing radiation on the hematopoiesis and immune response. Low dose radiation has been shown to induce biological responses like inflammatory responses, innate immune system activation and DNA repair (adaptive response). This review reveals the fact that there are many unanswered questions regarding the role of radiation as either an immune-activating (low dose) or immunosuppressive (high dose) agent.
Collapse
Affiliation(s)
- Nezih Hekim
- Department of Medical Biochemistry, School of Medicine, SANKO University, Gaziantep, Turkey
| | - Zafer Cetin
- Department of Medical Biology & Genetics, School of Medicine, SANKO University, Gaziantep, Turkey
| | - Zacharenia Nikitaki
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Zografou 15780, Athens, Greece
| | - Aysegul Cort
- Department of Medical Biochemistry, School of Medicine, SANKO University, Gaziantep, Turkey; Department of Nutrition and Dietetics, Faculty of Health Sciences, SANKO University, Gaziantep, Turkey
| | - Eyup Ilker Saygili
- Department of Medical Biochemistry, School of Medicine, SANKO University, Gaziantep, Turkey.
| |
Collapse
|
37
|
Multhoff G, Pockley AG, Schmid TE, Schilling D. The role of heat shock protein 70 (Hsp70) in radiation-induced immunomodulation. Cancer Lett 2015; 368:179-84. [PMID: 25681671 DOI: 10.1016/j.canlet.2015.02.013] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/04/2015] [Accepted: 02/07/2015] [Indexed: 02/08/2023]
Abstract
Despite enormous progress in radiation technologies (high precision image-guided irradiation, proton irradiation, heavy ion irradiation) and radiotherapeutic concepts (hypofractionated irradiation schemes), the clinical outcome of radiotherapy in locally advanced and metastasized tumors and in hypoxic tumors which are radiation-resistant remains unsatisfactory. Given their key influence on a number of biological and immunological parameters, this article considers the influence of irradiation-induced stress proteins on radiation-induced immunomodulation. Depending on its location, the major stress-inducible Heat shock protein 70 (Hsp70) has been found to fulfill multiple roles. On the one hand, increased intracellular Hsp70 levels have been found to play a key role in the recovery from stress such as radio(chemo)therapy, and on the other hand extracellular Hsp70 proteins are potent stimulators of the innate immune system and mediators of anti-tumor immunity. Furthermore, if loaded with tumor-derived peptides, members of the Heat Shock Protein 70 (HSP70) and 90 (HSP90) families can stimulate the adaptive immune system via antigen cross-presentation. An irradiation-induced enhancement of the selective expression of a membrane form of Hsp70 on the surface of tumor cells which can act as a recognition structure for activated NK cells might have significant clinical relevance, in that the outcome of irradiation therapy for advanced tumors could be improved by combining it with cell-based and other immunotherapies that target this membrane form of Hsp70.
Collapse
Affiliation(s)
- Gabriele Multhoff
- Department of Radiation Oncology, Technische Universität München, Klinikum rechts der Isar, Munich, Germany; Helmholtz Center Munich, German Research Center for Environmental Health, CCG - "Innate Immunity in Tumor Biology", Munich, Germany.
| | - Alan G Pockley
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Thomas E Schmid
- Department of Radiation Oncology, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Daniela Schilling
- Department of Radiation Oncology, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| |
Collapse
|
38
|
Abstract
PURPOSE This review focuses on recent advances in the field of combining radiation with immunotherapy for the treatment of malignant diseases, since various combinatorial cancer therapy approaches have lately proven highly successful. RESULTS With initial case reports and anecdotes progressively converting into solid clinical data, interest in cancer immunotherapy (CIT) has risen steeply. Especially immune checkpoint blockade therapies have recently celebrated tremendous successes in the treatment of severe malignancies resistant to conventional treatment strategies. Nevertheless, the high variability of patient responses to CIT remains a major hurdle, clearly indicating an urgent need for improvement. It has been suggested that successful cancer therapy most probably involves combinatorial treatment approaches. Radiotherapy (RT) has been proposed as a powerful partner for CIT due to its broad spectrum of immune modulatory characteristics. Several preclinical studies, supported by an increasing number of clinical observations, have demonstrated synergistic interactions between RT and CIT resulting in significantly improved therapy outcomes. CONCLUSIONS Numerous reports have shown that radiation is capable of tipping the scales from tumor immune evasion to elimination in different tumor types. The next puzzle to be solved is the question of logistics - including types, schedule and dosage of combinatorial RT and CIT strategies.
Collapse
Affiliation(s)
- Klara Soukup
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital - Harvard Medical School , Boston, MA , USA
| | | |
Collapse
|
39
|
Datta J, Terhune JH, Lowenfeld L, Cintolo JA, Xu S, Roses RE, Czerniecki BJ. Optimizing dendritic cell-based approaches for cancer immunotherapy. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2014; 87:491-518. [PMID: 25506283 PMCID: PMC4257036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Dendritic cells (DC) are professional antigen-presenting cells uniquely suited for cancer immunotherapy. They induce primary immune responses, potentiate the effector functions of previously primed T-lymphocytes, and orchestrate communication between innate and adaptive immunity. The remarkable diversity of cytokine activation regimens, DC maturation states, and antigen-loading strategies employed in current DC-based vaccine design reflect an evolving, but incomplete, understanding of optimal DC immunobiology. In the clinical realm, existing DC-based cancer immunotherapy efforts have yielded encouraging but inconsistent results. Despite recent U.S. Federal and Drug Administration (FDA) approval of DC-based sipuleucel-T for metastatic castration-resistant prostate cancer, clinically effective DC immunotherapy as monotherapy for a majority of tumors remains a distant goal. Recent work has identified strategies that may allow for more potent "next-generation" DC vaccines. Additionally, multimodality approaches incorporating DC-based immunotherapy may improve clinical outcomes.
Collapse
Affiliation(s)
- Jashodeep Datta
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Julia H. Terhune
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lea Lowenfeld
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jessica A. Cintolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shuwen Xu
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Robert E. Roses
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Brian J. Czerniecki
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania,To whom all correspondence should be addressed: Brian J. Czerniecki, MD PhD, Department of Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Drive, Philadelphia, PA 19104; Tele: 215-615-1696; Fax: 215-615-0555;
| |
Collapse
|
40
|
Ahmed MM, Guha C, Hodge JW, Jaffee E. Immunobiology of radiotherapy: new paradigms. Radiat Res 2014; 182:123-5. [PMID: 25036983 DOI: 10.1667/rr13849.1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Mansoor M Ahmed
- a Radiation Research Program, National Cancer Institute/National Institutes of Health, Rockville, Maryland 20850
| | | | | | | |
Collapse
|