1
|
Li L, Wang X, Jiang M, Li L, Wang D, Li Y. Advancements in a novel model of autophagy and immune network regulation in radioresistance of cancer stem cells. Biomed Pharmacother 2024; 179:117420. [PMID: 39255736 DOI: 10.1016/j.biopha.2024.117420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024] Open
Abstract
Radiotherapy, a precise modality for treating malignant tumors, has undergone rapid advancements in primary and clinical research. The mechanisms underlying tumor radioresistance have become significant research. With the introduction and in-depth study of cancer stem cells (CSCs) theory, CSCs have been identified as the primary factor contributing to the development of tumor radioresistance. The "stemness" of CSCs is a biological characteristic of a small subset of cells within tumor tissues, characterized by self-renewal solid ability. This characteristic leads to resistance to radiotherapy, chemotherapy, and targeted therapies, driving tumor recurrence and metastasis. Another study revealed that cellular autophagy plays a pivotal role in maintaining the "stemness" of CSCs. Autophagy is a cellular mechanism that degrades proteins and organelles to generate nutrients and energy in response to stress. This process maintains cellular homeostasis and contributes to CSCs radioresistance. Furthermore, ionizing radiation (IR) facilitates epithelial-to-mesenchymal transition (EMT), vascular regeneration, and other tumor processes by influencing the infiltration of M2-type tumor-associated macrophages (TAMs). IR promotes the activation of the classical immunosuppressive "switch," PD-1/PD-L1, which diminishes T-cell secretion, leading to immune evasion and promoting radioresistance. Interestingly, recent studies have found that the immune pathway PD-1/PD-L1 is closely related to cellular autophagy. However, the interrelationships between immunity, autophagy, and radioresistance of CSCs and the regulatory mechanisms involved remain unclear. Consequently, this paper reviews recent research to summarize these potential connections, aiming to establish a theoretical foundation for future studies and propose a new model for the network regulation of immunity, autophagy, and radioresistance of tumor cells.
Collapse
Affiliation(s)
- Leyao Li
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Xin Wang
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Mei Jiang
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Lei Li
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Di Wang
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Yajun Li
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China.
| |
Collapse
|
2
|
Yilmaz MT, Gok A, Gedik ME, Caglayan A, Yedekci FY, Aydin Dilsiz S, Gunaydin G, Akyol A, Hurmuz P. The Impact of Dose Rate on the Tumor Microenvironment Using Flattening-filter-free Beams. Clin Oncol (R Coll Radiol) 2024; 36:390-398. [PMID: 38570205 DOI: 10.1016/j.clon.2024.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 03/03/2024] [Accepted: 03/13/2024] [Indexed: 04/05/2024]
Abstract
AIMS Recently, dose delivery technology has rapidly evolved with flattening filter-free beams (FFF), and the biological effects of high dose rates are a matter of interest. We hypothesized that FFF beams at different dose rates obtained with modern linear accelerators have different effects on the TME. MATERIALS AND METHODS The B16-F10 melanoma syngeneic tumor model was established, and mice were randomized to 2 different doses (2 Gy and 10 Gy) and 3 different dose rates (1 Gy/min, 6 Gy/min, and 14 Gy/min) along with the control group. Euthanasia was performed on the seventh day after RT, and intracardiac blood was collected for a comet assay. Tumors were harvested and examined histomorphologically and immunohistochemically. Statistical analyses were performed using SPSS software version 23 (SPSS Inc., Chicago, IL, USA). RESULTS The daily growth rate was uniform, and no difference was observed between tumor volumes across all three dose rates for each dose. Deoxyribonucleic acid (DNA) damage in blood mononuclear cells was not affected by dose or dose rate. In the TME histomorphological examination, the number of mitosis is less in the 10 Gy arm, whereas the pleomorphism score was greater. Nevertheless, varying dose rates had no effect on the number of mitosis or the pleomorphism score. The severity of the inflammation, cell densities in the TME, and expression of immunohistochemical markers were comparable across all doses and dose rates. CONCLUSION In our study involving the B16-F10 syngeneic tumor model, varying dose rates obtained with FFF beams had no effect on tumor volume, blood mononuclear cell DNA damage, or TME parameters. However, in order to fully understand the biological impacts of novel techniques, our study should be validated with alternative preclinical setups.
Collapse
Affiliation(s)
- M T Yilmaz
- Hacettepe University Faculty of Medicine, Department of Radiation Oncology, Ankara, Turkey.
| | - A Gok
- Hacettepe University, Stem Cell Research and Application Center, Ankara, Turkey.
| | - M E Gedik
- Hacettepe University Faculty of Medicine, Basic Oncology Department, Ankara, Turkey.
| | - A Caglayan
- Hacettepe University Faculty of Pharmacy, Department of Toxicology, Ankara, Turkey.
| | - F Y Yedekci
- Hacettepe University Faculty of Medicine, Department of Radiation Oncology, Ankara, Turkey.
| | - S Aydin Dilsiz
- Hacettepe University Faculty of Pharmacy, Department of Toxicology, Ankara, Turkey.
| | - G Gunaydin
- Hacettepe University Faculty of Medicine, Basic Oncology Department, Ankara, Turkey.
| | - A Akyol
- Hacettepe University Faculty of Medicine, Pathology Department, Ankara, Turkey.
| | - P Hurmuz
- Hacettepe University Faculty of Medicine, Department of Radiation Oncology, Ankara, Turkey.
| |
Collapse
|
3
|
Skalina KA, Małachowska B, Sindhu KK, Thompson M, Nehlsen AD, Salgado LR, Dovey Z, Hasan S, Guha C, Tang J. Combining theranostic/particle therapy with immunotherapy for the treatment of GU malignancies. BJUI COMPASS 2024; 5:334-344. [PMID: 38481668 PMCID: PMC10927934 DOI: 10.1002/bco2.316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/10/2023] [Indexed: 11/01/2024] Open
Abstract
Particle therapy and radiopharmaceuticals are emerging fields in the treatment of genitourinary cancers. With these novel techniques and the ever-growing immunotherapy options, the combinations of these therapies have the potential to improve current cancer cure rates. However, the most effective sequence and combination of these therapies is unknown and is a question that is actively being explored in multiple ongoing clinical trials. Here, we review the immunological effects of particle therapy and the available radiopharmaceuticals and discuss how best to combine these therapies.
Collapse
Affiliation(s)
- Karin A. Skalina
- Department of Radiation OncologyMontefiore Medical Center/Albert Einstein College of MedicineBronxNew YorkUSA
| | - Beata Małachowska
- Department of Radiation OncologyMontefiore Medical Center/Albert Einstein College of MedicineBronxNew YorkUSA
| | - Kunal K. Sindhu
- Department of Radiation OncologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Marcher Thompson
- Department of Radiation OncologyAIS Cancer Center/Adventist HealthBakersfieldCaliforniaUSA
| | - Anthony D. Nehlsen
- Department of Radiation OncologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Lucas Resende Salgado
- Department of Radiation OncologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Zachary Dovey
- Department of UrologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | - Chandan Guha
- Department of Radiation OncologyMontefiore Medical Center/Albert Einstein College of MedicineBronxNew YorkUSA
| | - Justin Tang
- Department of Radiation OncologyMontefiore Medical Center/Albert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
4
|
Najafi A, Keykhaee M, Kazemi MH, Karimi MY, Khorramdelazad H, Aghamohamadi N, Bolouri MR, Ghaffari-Nazari H, Mirsharif ES, Karimi M, Dehghan Manshadi HR, Mahdavi SR, Safari E, Jalali SA, Falak R, Khoobi M. Catalase-gold nanoaggregates manipulate the tumor microenvironment and enhance the effect of low-dose radiation therapy by reducing hypoxia. Biomed Pharmacother 2023; 167:115557. [PMID: 37757491 DOI: 10.1016/j.biopha.2023.115557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Radiotherapy as a standard method for cancer treatment faces tumor recurrence and antitumoral unresponsiveness. Suppressive tumor microenvironment (TME) and hypoxia are significant challenges affecting efficacy of radiotherapy. Herein, a versatile method is introduced for the preparation of pH-sensitive catalase-gold cross-linked nanoaggregate (Au@CAT) having acceptable stability and selective activity in tumor microenvironment. Combining Au@CAT with low-dose radiotherapy enhanced radiotherapy effects via polarizing protumoral immune cells to the antitumoral landscape. This therapeutic approach also attenuated hypoxia, confirmed by downregulating hypoxia hallmarks, such as hypoxia-inducible factor α-subunits (HIF-α), vascular endothelial growth factor (VEGF), and EGF. Catalase stability against protease digestion was improved significantly in Au@CAT compared to the free catalase. Moreover, minimal toxicity of Au@CAT on normal cells and increased reactive oxygen species (ROS) were confirmed in vitro compared with radiotherapy. Using the nanoaggregates combined with radiotherapy led to a significant reduction of immunosuppressive infiltrating cells such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (T-regs) compared to the other groups. While, this combined therapy could significantly increase the frequency of CD8+ cells as well as M1 to M2 macrophages (MQs) ratio. The combination therapy also reduced the tumor size and increased survival rate in mice models of colorectal cancer (CRC). Our results indicate that this innovative nanocomposite could be an excellent system for catalase delivery, manipulating the TME and providing a potential therapeutic strategy for treating CRC.
Collapse
Affiliation(s)
- Alireza Najafi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Keykhaee
- Department of Pharmaceutical Biomaterials and Medical Biomaterial Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Kazemi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Khorramdelazad
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Nazanin Aghamohamadi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Bolouri
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haniyeh Ghaffari-Nazari
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Milad Karimi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Seied Rabi Mahdavi
- Radiation Biology Research Center& Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Safari
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Jalali
- Immunology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mehdi Khoobi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Liu C, Wang X, Qin W, Tu J, Li C, Zhao W, Ma L, Liu B, Qiu H, Yuan X. Combining radiation and the ATR inhibitor berzosertib activates STING signaling and enhances immunotherapy via inhibiting SHP1 function in colorectal cancer. Cancer Commun (Lond) 2023; 43:435-454. [PMID: 36855844 PMCID: PMC10091106 DOI: 10.1002/cac2.12412] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/03/2023] [Accepted: 02/17/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) have shown a moderate response in colorectal cancer (CRC) with deficient mismatch repair (dMMR) functions and poor response in patients with proficient MMR (pMMR). pMMR tumors are generally immunogenically "cold", emphasizing combination strategies to turn the "cold" tumor "hot" to enhance the efficacy of ICIs. ATR inhibitors (ATRi) have been proven to cooperate with radiation to promote antitumor immunity, but it is unclear whether ATRi could facilitate the efficacy of IR and ICI combinations in CRCs. This study aimed to investigate the efficacy of combining ATRi, irradiation (IR), and anti-PD-L1 antibodies in CRC mouse models with different microsatellite statuses. METHODS The efficacy of combining ATRi, IR, and anti-PD-L1 antibodies was evaluated in CRC tumors. The tumor microenvironment and transcriptome signatures were investigated under different treatment regimens. The mechanisms were explored via cell viability assay, flow cytometry, immunofluorescence, immunoblotting, co-immunoprecipitation, and real-time quantitative PCR in multiple murine and human CRC cell lines. RESULTS Combining ATRi berzosertib and IR enhanced CD8+ T cell infiltration and enhanced the efficacy of anti-PD-L1 therapy in mouse CRC models with different microsatellite statuses. The mechanistic study demonstrated that IR + ATRi could activate both the canonical cGAS-STING-pTBK1/pIRF3 axis by increasing cytosolic double-stranded DNA levels and the non-canonical STING signaling by attenuating SHP1-mediated inhibition of the TRAF6-STING-p65 axis, via promoting SUMOylation of SHP1 at lysine 127. By boosting the STING signaling, IR + ATRi induced type I interferon-related gene expression and strong innate immune activation and reinvigorated the cold tumor microenvironment, thus facilitating immunotherapy. CONCLUSIONS The combination of ATRi and IR could facilitate anti-PD-L1 therapy by promoting STING signaling in CRC models with different microsatellite statuses. The new combination strategy raised by our study is worth investigating in the management of CRC.
Collapse
Affiliation(s)
- Chaofan Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xi Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Wan Qin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Chunya Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Li Ma
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Hong Qiu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| |
Collapse
|
6
|
Entezam A, Fielding A, Bradley D, Fontanarosa D. Absorbed dose calculation for a realistic CT-derived mouse phantom irradiated with a standard Cs-137 cell irradiator using a Monte Carlo method. PLoS One 2023; 18:e0280765. [PMID: 36730280 PMCID: PMC9928120 DOI: 10.1371/journal.pone.0280765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 01/07/2023] [Indexed: 02/03/2023] Open
Abstract
Computed tomography (CT) derived Monte Carlo (MC) phantoms allow dose determination within small animal models that is not feasible with in-vivo dosimetry. The aim of this study was to develop a CT-derived MC phantom generated from a mouse with a xenograft tumour that could then be used to calculate both the dose heterogeneity in the tumour volume and out of field scattered dose for pre-clinical small animal irradiation experiments. A BEAMnrc Monte-Carlo model has been built of our irradiation system that comprises a lead collimator with a 1 cm diameter aperture fitted to a Cs-137 gamma irradiator. The MC model of the irradiation system was validated by comparing the calculated dose results with dosimetric film measurement in a polymethyl methacrylate (PMMA) phantom using a 1D gamma-index analysis. Dose distributions in the MC mouse phantom were calculated and visualized on the CT-image data. Dose volume histograms (DVHs) were generated for the tumour and organs at risk (OARs). The effect of the xenographic tumour volume on the scattered out of field dose was also investigated. The defined gamma index analysis criteria were met, indicating that our MC simulation is a valid model for MC mouse phantom dose calculations. MC dose calculations showed a maximum out of field dose to the mouse of 7% of Dmax. Absorbed dose to the tumour varies in the range 60%-100% of Dmax. DVH analysis demonstrated that tumour received an inhomogeneous dose of 12 Gy-20 Gy (for 20 Gy prescribed dose) while out of field doses to all OARs were minimized (1.29 Gy-1.38 Gy). Variation of the xenographic tumour volume exhibited no significant effect on the out of field scattered dose to OARs. The CT derived MC mouse model presented here is a useful tool for tumour dose verifications as well as investigating the doses to normal tissue (in out of field) for preclinical radiobiological research.
Collapse
Affiliation(s)
- Amir Entezam
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
- * E-mail:
| | - Andrew Fielding
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Chemistry and Physics, Faculty of Science, Queensland University of Technology, Brisbane, QLD, Australia
| | - David Bradley
- Centre for Applied Physics and Radiation Technologies, Sunway University, PJ, Malaysia
- Department of Physics, University of Surrey, Guildford, United Kingdom
| | - Davide Fontanarosa
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
7
|
Abscopal Response in Metastatic Melanoma: Real-World Data of a Retrospective, Multicenter Study. Cancers (Basel) 2022; 14:cancers14174213. [PMID: 36077747 PMCID: PMC9454568 DOI: 10.3390/cancers14174213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/02/2022] [Accepted: 08/11/2022] [Indexed: 11/28/2022] Open
Abstract
Objective: To evaluate the incidence of the abscopal response (AR) in patients with metastatic melanoma requiring palliative radiotherapy (RT). Patients and methods: Patients treated for metastatic melanoma between January 1998 and February 2020 in four oncology departments were screened. Patients with progression under immune checkpoint inhibitors or without ongoing systemic treatment, and requiring palliative RT were considered. The AR was defined as an objective response according to RECIST and/or iRECIST for at least one non-irradiated metastasis at distance (≥10 cm) from the irradiated lesion. Primary endpoint was the rate of AR. Secondary endpoints were overall survival (OS), progression-free survival (PFS), local control (LC) of the irradiated lesion, and toxicity as assessed by CTCAE v5. Results: Over the period considered, 118 patients were included and analyzed. Fifteen patients (12.7%) had an AR. With a median follow-up of 7.7 months (range, 0.2−242.2), median OS and PFS after RT were significantly longer in patients with an AR compared to those without: 28 vs. 6.6 months (p < 0.01) and not reached vs. 3.2 months, respectively. No grade ≥2 toxicity was reported. Patients who developed an AR were more likely to be treated with immunotherapy (93.3% vs. 55.9%, p = 0.02). In multivariate analysis, they had a higher number of irradiated metastases treated concomitantly (HR = 16.9, p < 0.01) and a higher rate of mild infections during RT (HR = 403.5, p < 0.01). Conclusions: AR in metastatic melanoma seems to be highly prognostic of overall survival, although it is a rare phenomenon. It may be promoted by multiple concomitant treatments with RT and immunotherapy and by acute inflammatory events such as infection.
Collapse
|
8
|
Wang B, Hu J, Zhang J, Zhao L. Radiation therapy regulates TCF-1 to maintain CD8+T cell stemness and promotes anti-tumor immunotherapy. Int Immunopharmacol 2022; 107:108646. [DOI: 10.1016/j.intimp.2022.108646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 11/05/2022]
|
9
|
Xie D, Jin X, Sun R, Zhang M, Wang J, Xiong X, Zhang X, Zhao M. Relapse Mechanism and Treatment Strategy After Chimeric Antigen Receptor T-Cell Therapy in Treating B-Cell Hematological Malignancies. Technol Cancer Res Treat 2022; 21:15330338221118413. [PMID: 35989682 PMCID: PMC9403467 DOI: 10.1177/15330338221118413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Over the past few decades, immunotherapy has revolutionized the modern medical oncology field. Chimeric antigen receptor (CAR)-T cell therapy has a promising curative effect in the treatment of hematological malignancies. Anti-CD19 CAR-T cells are the most mature CAR-T cells recently studied and in recent years it has achieved a complete remission rate of approximately 90% in the treatment of B-cell acute lymphoblastic leukemia (B-ALL). Although CAR-T cell therapy has greatly alleviated the disease in patients with leukemia or lymphoma, some of them still relapse after treatment. Therefore, in this article, we discuss the factors that may contribute to disease relapse following CAR-T cell therapy and summarize potential strategies to overcome these obstacles, thus providing the possibility of improving standard treatment regimens.
Collapse
Affiliation(s)
- Danni Xie
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Xin Jin
- Department of Hematology, 66571Tianjin First Central Hospital, Tianjin, China
| | - Rui Sun
- 481107Nankai University School of Medicine, Tianjin, China
| | - Meng Zhang
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Jiaxi Wang
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Xia Xiong
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Xiaomei Zhang
- 481107Nankai University School of Medicine, Tianjin, China
| | - Mingfeng Zhao
- The First Central Clinical College of Tianjin Medical University, Tianjin, China.,Department of Hematology, 66571Tianjin First Central Hospital, Tianjin, China.,481107Nankai University School of Medicine, Tianjin, China
| |
Collapse
|
10
|
He H, Xu T, Li P, Jia G, Li X, Song Q. Anti-PD-1 Immunotherapy Combined With Stereotactic Body Radiation Therapy and GM-CSF as Salvage Therapy in a PD-L1-Positive Patient With Refractory Metastatic Thyroid Hürthle Cell Carcinoma: A Case Report and Literature Review. Front Oncol 2021; 11:782646. [PMID: 34888252 PMCID: PMC8650693 DOI: 10.3389/fonc.2021.782646] [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: 09/24/2021] [Accepted: 11/03/2021] [Indexed: 12/21/2022] Open
Abstract
Thyroid Hürthle cell carcinoma, known as thyroid eosinophilic carcinoma, is a rare pathological type of differentiated thyroid cancer (DTC), representing 3-4% of all thyroid cancers. However, given the high risk of invasion and metastasis, thyroid Hürthle cell carcinoma has a relatively poor prognosis. Traditional treatment methods have limited effects on patients with metastatic thyroid cancers. Developing a valuable therapy for advanced thyroid carcinomas is an unfilled need, and immunotherapy could represent another choice for these tumors. We herein reported the case of a patient with recurrent advanced thyroid Hürthle cell cancer and positive programmed death-ligand 1 (PD-L1) expression, who suffered tumor progression after re-surgery, radiotherapy, and targeted therapy. It is encouraging that PD-1 inhibitors in combination with GM-CSF and stereotactic body irradiation (SBRT) on metastatic disease have a significant anti-tumor effect.
Collapse
Affiliation(s)
- Haihua He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tangpeng Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ping Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guohua Jia
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangpan Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Entezam A, Fielding A, Moi D, Bradley D, Ratnayake G, Sim L, Kralik C, Fontanarosa D. Investigation of scattered dose in a mouse phantom model for pre-clinical dosimetry studies. Radiat Phys Chem Oxf Engl 1993 2021. [DOI: 10.1016/j.radphyschem.2021.109691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
12
|
Choi SH, Lee DY, Kang S, Lee MK, Lee JH, Lee SH, Lee HL, Lee HY, Jeong YIL. Caffeic Acid Phenethyl Ester-Incorporated Radio-Sensitive Nanoparticles of Phenylboronic Acid Pinacol Ester-Conjugated Hyaluronic Acid for Application in Radioprotection. Int J Mol Sci 2021; 22:6347. [PMID: 34198522 PMCID: PMC8231778 DOI: 10.3390/ijms22126347] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/31/2021] [Accepted: 06/08/2021] [Indexed: 12/30/2022] Open
Abstract
We synthesized phenylboronic acid pinacol ester (PBPE)-conjugated hyaluronic acid (HA) via thiobis(ethylamine) (TbEA) linkage (abbreviated as HAsPBPE conjugates) to fabricate the radiosensitive delivery of caffeic acid phenetyl ester (CAPE) and for application in radioprotection. PBPE was primarily conjugated with TbEA and then PBPE-TbEA conjugates were conjugated again with hyaluronic acid using carbodiimide chemistry. CAPE-incorporated nanoparticles of HAsPBPE were fabricated by the nanoprecipitation method and then the organic solvent was removed by dialysis. CAPE-incorporated HAsPBPE nanoparticles have a small particle size of about 80 or 100 nm and they have a spherical shape. When CAPE-incorporated HAsPBPE nanoparticles were irradiated, nanoparticles became swelled or disintegrated and their morphologies were changed. Furthermore, the CAPE release rate from HAsPBPE nanoparticles were increased according to the radiation dose, indicating that CAPE-incorporated HAsPBPE nanoparticles have radio-sensitivity. CAPE and CAPE-incorporated HAsPBPE nanoparticles appropriately prevented radiation-induced cell death and suppressed intracellular accumulation of reactive oxygen species (ROS). CAPE and CAPE-incorporated HAsPBPE nanoparticles efficiently improved survivability of mice from radiation-induced death and reduced apoptotic cell death. We suggest that HAsPBPE nanoparticles are promising candidates for the radio-sensitive delivery of CAPE.
Collapse
Affiliation(s)
- Seon-Hee Choi
- Biomedical R&D Center, Pusan National University Yangsan Hospital, Gyeongnam 50612, Korea; (S.-H.C.); (H.-L.L.)
| | - Dong-Yeon Lee
- Department of Radiation Oncology, Dongnam Institute of Radiological & Medical Sciences, Pusan 46033, Korea;
| | - Sohi Kang
- Biomaterial R&BD Center, Chonnam National University, Gwangju 61186, Korea;
| | - Min-Kyung Lee
- Department of Dental Hygiene, Dong-Eui University, Pusan 47340, Korea;
| | - Jae-Heun Lee
- Department of Radiological Science, Dong-Eui University, Pusan 47340, Korea; (J.-H.L.); (S.-H.L.)
| | - Sang-Heon Lee
- Department of Radiological Science, Dong-Eui University, Pusan 47340, Korea; (J.-H.L.); (S.-H.L.)
| | - Hye-Lim Lee
- Biomedical R&D Center, Pusan National University Yangsan Hospital, Gyeongnam 50612, Korea; (S.-H.C.); (H.-L.L.)
| | - Hyo-Young Lee
- Department of Radiological Science, Dong-Eui University, Pusan 47340, Korea; (J.-H.L.); (S.-H.L.)
| | - Young-IL Jeong
- Biomedical R&D Center, Pusan National University Yangsan Hospital, Gyeongnam 50612, Korea; (S.-H.C.); (H.-L.L.)
| |
Collapse
|
13
|
Chen C, Liu Y, Cui B. Effect of radiotherapy on T cell and PD-1 / PD-L1 blocking therapy in tumor microenvironment. Hum Vaccin Immunother 2021; 17:1555-1567. [PMID: 33428533 DOI: 10.1080/21645515.2020.1840254] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer is a worldwide problem that threatens human health. Radiotherapy plays an important role in a variety of cancer treatment methods. The administration of radiotherapy can alter the differentiation pathways and functions of T cells, which in turn improves the immune response of T cells. Radiotherapy can also induce up-regulation of PD-L1 expression, which means that it has great potential for enhancing the therapeutic effect of anti-PD-1/PD-L1 inhibitors and reducing the risk of drug resistance toward them. At present, the combination of radiotherapy and anti-PD-1/PD-L1 inhibitors has shown significant therapeutic effects in clinical tumor research. This review focuses on the mechanism of radiotherapy on T cells reported in recent years, as well as related research progress in the application of PD-1/PD-L1 blockers. It will provide a theoretical basis for the rational clinical application of radiotherapy combined with PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Chen Chen
- Department of Colorectal Surgery, The Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Yanlong Liu
- Department of Colorectal Surgery, The Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Binbin Cui
- Department of Colorectal Surgery, The Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| |
Collapse
|
14
|
Harari A, Graciotti M, Bassani-Sternberg M, Kandalaft LE. Antitumour dendritic cell vaccination in a priming and boosting approach. Nat Rev Drug Discov 2020; 19:635-652. [PMID: 32764681 DOI: 10.1038/s41573-020-0074-8] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
Abstract
Mobilizing antitumour immunity through vaccination potentially constitutes a powerful anticancer strategy but has not yet provided robust clinical benefits in large patient populations. Although major hurdles still exist, we believe that currently available strategies for vaccines that target dendritic cells or use them to present antitumour antigens could be integrated into existing clinical practice using prime-boost approaches. In the priming phase, these approaches capitalize on either standard treatment modalities to trigger in situ vaccination and release tumour antigens or vaccination with dendritic cells loaded with tumour lysates or patient-specific neoantigens. In a second boost phase, personalized synthetic vaccines specifically boost T cells that were triggered during the priming phase. This immunotherapy approach has been enabled by the substantial recent improvements in dendritic cell vaccines. In this Perspective, we discuss these improvements, highlight how the prime-boost approach can be translated into clinical practice and provide solutions for various anticipated hurdles.
Collapse
Affiliation(s)
- Alexandre Harari
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Michele Graciotti
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Lana E Kandalaft
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland. .,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
15
|
Radiation-induced bystander and abscopal effects: important lessons from preclinical models. Br J Cancer 2020; 123:339-348. [PMID: 32581341 PMCID: PMC7403362 DOI: 10.1038/s41416-020-0942-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 03/10/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy is a pivotal component in the curative treatment of patients with localised cancer and isolated metastasis, as well as being used as a palliative strategy for patients with disseminated disease. The clinical efficacy of radiotherapy has traditionally been attributed to the local effects of ionising radiation, which induces cell death by directly and indirectly inducing DNA damage, but substantial work has uncovered an unexpected and dual relationship between tumour irradiation and the host immune system. In clinical practice, it is, therefore, tempting to tailor immunotherapies with radiotherapy in order to synergise innate and adaptive immunity against cancer cells, as well as to bypass immune tolerance and exhaustion, with the aim of facilitating tumour regression. However, our understanding of how radiation impacts on immune system activation is still in its early stages, and concerns and challenges regarding therapeutic applications still need to be overcome. With the increasing use of immunotherapy and its common combination with ionising radiation, this review briefly delineates current knowledge about the non-targeted effects of radiotherapy, and aims to provide insights, at the preclinical level, into the mechanisms that are involved with the potential to yield clinically relevant combinatorial approaches of radiotherapy and immunotherapy.
Collapse
|
16
|
Yamada T, Uchino J, Chihara Y, Shimamoto T, Iwasaku M, Tamiya N, Kaneko Y, Kiyomi F, Takayama K. Rationale and design of a phase II trial of durvalumab treatment in patients with NSCLC ineligible for stage III chemoradiotherapy following radiation monotherapy (SPIRAL-RT study). Ther Adv Med Oncol 2020; 12:1758835920927841. [PMID: 32536981 PMCID: PMC7268142 DOI: 10.1177/1758835920927841] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 04/28/2020] [Indexed: 12/18/2022] Open
Abstract
Background: In the PACIFIC study, progression-free survival (PFS) and overall survival (OS) of patients with unresectable, locally advanced, stage III non-small cell lung cancer (NSCLC) were prolonged by durvalumab as maintenance therapy after radical concurrent chemoradiotherapy using platinum-based antitumor agents. However, no data were obtained to reveal the efficacy of durvalumab after radiation monotherapy in patients unsuitable for chemoradiotherapy. Here, we describe an ongoing single-arm, prospective, open-label, multicenter phase II trial of durvalumab in patients with NSCLC ineligible for stage III chemoradiotherapy following radiation monotherapy (SPIRAL-RT study). Methods: Durvalumab at 10 mg/kg body weight is administered every 2 weeks after radiation therapy until individual patients meet the discontinuation criteria. The treatment duration is up to 12 months. The primary endpoint is the 1-year PFS rate. Secondary endpoints are response rate, PFS, OS, and safety. Durvalumab treatment after radiation monotherapy is expected to prolong 1-year PFS rate and have acceptable adverse events. Discussion: We are conducting an intervention study to investigate the safety and efficacy of durvalumab treatment in patients with NSCLC ineligible for stage III chemoradiotherapy following radiation monotherapy.
Collapse
Affiliation(s)
- Tadaaki Yamada
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junji Uchino
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto 602-0857, Japan
| | - Yusuke Chihara
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takayuki Shimamoto
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masahiro Iwasaku
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nobuyo Tamiya
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshiko Kaneko
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Fumiaki Kiyomi
- Statistics and Data Center, Clinical Research Support Center Kyushu, Fukuoka, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
17
|
Ellerin BE, Demandante CGN, Martins JT. Pure abscopal effect of radiotherapy in a salivary gland carcinoma: Case report, literature review, and a search for new approaches. Cancer Radiother 2020; 24:226-246. [PMID: 32192840 DOI: 10.1016/j.canrad.2020.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/15/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
We report the case of an 84-year-old woman with poorly differentiated non-small cell carcinoma of the right parotid who presented with headache, was found to have a primary right parotid gland cancer as well as metastatic disease, and underwent palliative radiotherapy to the primary site. The patient received no chemotherapy or immunotherapy, but both the primary site and several non-irradiated foci in the lungs regressed or completely resolved. The patient remained free of disease for about one year before progression. The case is a rare instance of abscopal regression of metastatic disease in the absence of pharmacologic immunomodulation. A literature review surveys the history of the abscopal effect of radiation therapy, attempts to understand the mechanisms of its successes and failures, and points to new approaches that can inform and improve the outcomes of radioimmunotherapy.
Collapse
Affiliation(s)
| | | | - J T Martins
- UT Health HOPE Cancer Center, Tyler, TX 75701, USA
| |
Collapse
|
18
|
Abbassi LM, Cao KI, Kirova YM. [Immunotherapy and radiotherapy for management of breast cancer: Rational and overview on clinical practice]. Cancer Radiother 2020; 24:73-80. [PMID: 32046913 DOI: 10.1016/j.canrad.2019.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 06/20/2019] [Accepted: 06/26/2019] [Indexed: 10/25/2022]
Abstract
The discovery of immunotherapy led to understand the major role of immune system during the tumor process. Conventional treatments, such as chemotherapy, are directly tumoricidal. New drugs are developed to target specifically the immune system to make it regain its ability to recognize and eliminate cancer cells. Radiotherapy is used for a long time for its local action, but its systemic role, based on its impact on immunity, is now better known. Breast cancer was wrongly considered poorly immunogenic and put aside the amazing progress in this new area of treatment. In this article, we would like to present the pre-clinical and clinical rationales to associate immunotherapy to radiotherapy in the management of breast cancer.
Collapse
Affiliation(s)
- L M Abbassi
- Département d'oncologie radiothérapie, Institut Curie, 26, rue d'Ulm, 75005 Paris, France.
| | - K I Cao
- Département d'oncologie radiothérapie, Institut Curie, 26, rue d'Ulm, 75005 Paris, France
| | - Y M Kirova
- Département d'oncologie radiothérapie, Institut Curie, 26, rue d'Ulm, 75005 Paris, France
| |
Collapse
|
19
|
Liu PH, Sidi S. Targeting the Innate Immune Kinase IRAK1 in Radioresistant Cancer: Double-Edged Sword or One-Two Punch? Front Oncol 2019; 9:1174. [PMID: 31799178 PMCID: PMC6866135 DOI: 10.3389/fonc.2019.01174] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/18/2019] [Indexed: 01/07/2023] Open
Abstract
Antitumor immunity has emerged as a favorable byproduct of radiation therapy (RT), whereby tumor-associated antigens released from irradiated cells unleash innate and adaptive attacks on tumors located both within and outside the radiation field. RT-induced immune responses further provide actionable targets for overcoming tumor resistance to RT (R-RT); immunotherapy (IT) with checkpoint inhibitors or Toll-like receptor (TLR) agonists can markedly improve, if not synergize with, RT in preclinical models, and several of these drugs are currently investigated as radiosensitizers in patients. In an unbiased chemical-genetic screen in a zebrafish model of tumor R-RT, we unexpectedly found that Interleukin 1 Receptor-Associated Kinase 1 (IRAK1), a core effector of TLR-mediated innate immunity, also functions in live fish and human cancer models to counter RT-induced cell death mediated by the PIDDosome complex (PIDD-RAIDD-caspase-2). IRAK1 acting both as a driver of intrinsic tumor R-RT and as an effector of RT-induced antitumor immunity would, at first glance, pose obvious therapeutic conundrums. IRAK1 inhibitors would be expected to sensitize the irradiated tumor to RT but simultaneously thwart RT-induced antitumor immunity as initiated by stromal dendritic cells. Conversely, TLR agonist-based immunotherapy would be expected to intensify RT-induced antitumor immunity but at the expense of fueling IRAK1-mediated cell survival in the irradiated tumor. We discuss how IRAK1's differential reliance on catalytic activity in the radiation vs. TLR responses might help overcome these hurdles, as well as the crucial importance of developing IRAK1 inhibitors that lack activity against IRAK4, the kinase activity of which is essential for IRAK1 activation in both pathways.
Collapse
Affiliation(s)
- Peter H Liu
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, United States.,Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Samuel Sidi
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, United States.,Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
20
|
Karam SD, Raben D. Radioimmunotherapy for the treatment of head and neck cancer. Lancet Oncol 2019; 20:e404-e416. [DOI: 10.1016/s1470-2045(19)30306-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 03/19/2019] [Accepted: 03/25/2019] [Indexed: 12/27/2022]
|
21
|
Tumor Microenvironment as A "Game Changer" in Cancer Radiotherapy. Int J Mol Sci 2019; 20:ijms20133212. [PMID: 31261963 PMCID: PMC6650939 DOI: 10.3390/ijms20133212] [Citation(s) in RCA: 300] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/19/2019] [Accepted: 06/25/2019] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy (RT), besides cancer cells, also affects the tumor microenvironment (TME): tumor blood vessels and cells of the immune system. It damages endothelial cells and causes radiation-induced inflammation. Damaged vessels inhibit the infiltration of CD8+ T lymphocytes into tumors, and immunosuppressive pathways are activated. They lead to the accumulation of radioresistant suppressor cells, including tumor-associated macrophages (TAMs) with the M2 phenotype, myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs). The area of tumor hypoxia increases. Hypoxia reduces oxygen-dependent DNA damage and weakens the anti-cancer RT effect. It activates the formation of new blood vessels and leads to cancer relapse after irradiation. Irradiation may also activate the immune response through immunogenic cell death induction. This leads to the "in situ" vaccination effect. In this article, we review how changes in the TME affect radiation-induced anticancer efficacy. There is a very delicate balance between the activation of the immune system and the immunosuppression induced by RT. The effects of RT doses on immune system reactions and also on tumor vascularization remain unclear. A better understanding of these interactions will contribute to the optimization of RT treatment, which may prevent the recurrence of cancer.
Collapse
|
22
|
Daniel P, Sabri S, Chaddad A, Meehan B, Jean-Claude B, Rak J, Abdulkarim BS. Temozolomide Induced Hypermutation in Glioma: Evolutionary Mechanisms and Therapeutic Opportunities. Front Oncol 2019; 9:41. [PMID: 30778375 PMCID: PMC6369148 DOI: 10.3389/fonc.2019.00041] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/16/2019] [Indexed: 12/12/2022] Open
Abstract
Glioma are the most common type of malignant brain tumor, with glioblastoma (GBM) representing the most common and most lethal type of glioma. Surgical resection followed by radiotherapy and chemotherapy using the alkylating agent Temozolomide (TMZ) remain the mainstay of treatment for glioma. While this multimodal regimen is sufficient to temporarily eliminate the bulk of the tumor mass, recurrence is inevitable and often poses major challenges for clinical management due to treatment resistance and failure to respond to targeted therapies. Improved tumor profiling capacity has enabled characterization of the genomic landscape of gliomas with the overarching goal to identify clinically relevant subtypes and inform treatment decisions. Increased tumor mutational load has been shown to correlate with higher levels of neoantigens and is indicative of the potential to induce a durable response to immunotherapy. Following treatment with TMZ, a subset of glioma has been identified to recur with increased tumor mutational load. These hypermutant recurrent glioma represent a subtype of recurrence with unique molecular vulnerabilities. In this review, we will elaborate on the current knowledge regarding the evolution of hypermutation in gliomas and the potential therapeutic opportunities that arise with TMZ-induced hypermutation in gliomas.
Collapse
Affiliation(s)
- Paul Daniel
- Division of Radiation Oncology, Department of Oncology, McGill University, Montréal, QC, Canada
| | - Siham Sabri
- Department of Pathology, McGill University, Montréal, QC, Canada.,Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Ahmad Chaddad
- Division of Radiation Oncology, Department of Oncology, McGill University, Montréal, QC, Canada.,The Laboratory for Imagery, Vision and Artificial Intelligence, École de Technologie Supérieure, Montréal, QC, Canada
| | - Brian Meehan
- Department of Pediatrics, McGill University, Montréal, QC, Canada
| | - Bertrand Jean-Claude
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada.,Department of Medicine, McGill University, Montréal, QC, Canada
| | - Janusz Rak
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada.,Department of Pediatrics, McGill University, Montréal, QC, Canada
| | - Bassam S Abdulkarim
- Division of Radiation Oncology, Department of Oncology, McGill University, Montréal, QC, Canada.,Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
23
|
Wirsdörfer F, de Leve S, Jendrossek V. Combining Radiotherapy and Immunotherapy in Lung Cancer: Can We Expect Limitations Due to Altered Normal Tissue Toxicity? Int J Mol Sci 2018; 20:ijms20010024. [PMID: 30577587 PMCID: PMC6337556 DOI: 10.3390/ijms20010024] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 02/08/2023] Open
Abstract
In recent decades, technical advances in surgery and radiotherapy, as well as breakthroughs in the knowledge on cancer biology, have helped to substantially improve the standard of cancer care with respect to overall response rates, progression-free survival, and the quality of life of cancer patients. In this context, immunotherapy is thought to have revolutionized the standard of care for cancer patients in the long term. For example, immunotherapy approaches such as immune checkpoint blockade are currently increasingly being used in cancer treatment, either alone or in combination with chemotherapy or radiotherapy, and there is hope from the first clinical trials that the appropriate integration of immunotherapy into standard care will raise the success rates of cancer therapy to a new level. Nevertheless, successful cancer therapy remains a major challenge, particularly in tumors with either pronounced resistance to chemotherapy and radiation treatment, a high risk of normal tissue complications, or both, as in lung cancer. Chemotherapy, radiotherapy and immunotherapy have the capacity to evoke adverse effects in normal tissues when administered alone. However, therapy concepts are usually highly complex, and it is still not clear if combining immunotherapy with radio(chemo)therapy will increase the risk of normal tissue complications, in particular since normal tissue toxicity induced by chemotherapy and radiotherapy can involve immunologic processes. Unfortunately, no reliable biomarkers are available so far that are suited to predict the unique normal tissue sensitivity of a given patient to a given treatment. Consequently, clinical trials combining radiotherapy and immunotherapy are attracting major attention, not only regarding efficacy, but also with regard to safety. In the present review, we summarize the current knowledge of radiation-induced and immunotherapy-induced effects in tumor and normal tissue of the lung, and discuss the potential limitations of combined radio-immunotherapy in lung cancer with a focus on the suspected risk for enhanced acute and chronic normal tissue toxicity.
Collapse
Affiliation(s)
- Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45147 Essen, Germany.
| | - Simone de Leve
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45147 Essen, Germany.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45147 Essen, Germany.
| |
Collapse
|