1
|
Ma G, Yang Y, Li S, Li L, Meng X, Rong Y, Tang M, Cheng Q, Guo H, Li Q, Jin X. Modulating systemic anti-inflammatory response mitigates osteoarthritis progression and associated pain after low-dose radiotherapy. Int Immunopharmacol 2025; 158:114815. [PMID: 40347883 DOI: 10.1016/j.intimp.2025.114815] [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: 12/18/2024] [Revised: 04/27/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Osteoarthritis (OA) is a degenerative joint disease, often accompanied by inflammation. It has reported that low-dose radiotherapy (LDRT) has anti-inflammatory effect for benign pathologies and has been used for clinical treatment of OA in some European regions. However, the underlying molecular mechanisms of LDRT alleviating OA are only poorly understood. Herein, it is verified that LDRT improved the locomotor ability of OA rats, increased the locomotor distance and speed in the in vivo experiments. Moreover, LDRT decreased the degree of cartilage damage, attenuated the synovial inflammation, and promoted macrophage polarization towards M2 type. For the in vitro experiments, LDRT promoted macrophage polarization towards M2-type, which enhanced cell growth and adjusted the inflammatory factors in chondrocyte. Additionally, it is found LDRT could ameliorate OA pain through inhibiting spinal cord inflammation. Take together, our study suggests that LDRT could ameliorate OA symptoms and relieve OA-related pain by exerting its anti-inflammatory effect.
Collapse
Affiliation(s)
- Guorong Ma
- Gansu Provincial Hospital, Lanzhou 730000, China; Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000,China; The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou 730000,China
| | - Yongze Yang
- Gansu Provincial Hospital, Lanzhou 730000, China; The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou 730000,China
| | - Shuzhi Li
- Gansu Provincial Hospital, Lanzhou 730000, China; The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou 730000,China
| | - Linjing Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000,China
| | - Xin Meng
- Department of Emergency, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004,China
| | - Yao Rong
- Gansu Provincial Hospital, Lanzhou 730000, China
| | | | | | - Hongzhang Guo
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou 730000,China.
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000,China.
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000,China.
| |
Collapse
|
2
|
Bargh-Dawson H, Box C, Civale J, Birdsey GM, Bamber JC, Harris E. Radiotherapeutic enhancement using ultrasound-stimulated microbubbles: a critical review. Int J Radiat Biol 2025:1-21. [PMID: 40397648 DOI: 10.1080/09553002.2025.2498980] [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: 03/28/2025] [Accepted: 04/23/2025] [Indexed: 05/23/2025]
Abstract
PURPOSE Ultrasound stimulated microbubbles (USMB) are proposed as radioenhancing agents. Acting mechanically, they are attractive because their effects can be localized to the tumor, limiting the potential for normal tissue toxicity. Extensive preclinical research in models of human cancers has demonstrated increased tumor control when USMB are combined with radiotherapy compared with radiation alone, which has led to recent Phase I trials. The leading theory on the radioenhancement mechanism of action (MOA) is that USMB act as vascular disrupting agents, but others are proposed. MATERIALS AND METHODS Current literature was reviewed with a focus on the role of the tumor vasculature on radiotherapy response, the bioeffects of USMB on the vasculature, and studies of USMB as radioenhancers. Additionally, the possible interplay between USMB as vascular modulators, and radiation-induced anti-tumor immunity, is explored. RESULTS Whilst most preclinical evidence compellingly describes the radioenhancement effect, only one study considers the immune cell infiltration post USMB plus radiotherapy, with non-significant findings. Clinical studies demonstrate the safety of USMB. As a monotherapy, USMB can alter tumor immune microenvironments and induce a variety of bioeffects on the vasculature, depending on the stimulatory acoustic parameters. Treatment parameters used to study the effects of USMB alone, and with radiotherapy, vary in the literature making direct comparisons difficult. CONCLUSIONS Further work exploring USMB for radioenhancement is warranted. Elucidation of the MOA is required to support clinical translation, particularly with a view to optimize treatment parameters.
Collapse
Affiliation(s)
- Hannah Bargh-Dawson
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Carol Box
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - John Civale
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Graeme M Birdsey
- Imperial College London, National Heart and Lung Institute, London, UK
| | - Jeffrey C Bamber
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Emma Harris
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| |
Collapse
|
3
|
Liu Z, Jiang X, Ke Z, Wang W, Tang J, Dai Y. PAR2 deficiency impairs antitumor immunity and attenuates anti-PD1 efficacy in colorectal cancer. Pharmacol Res 2025; 215:107721. [PMID: 40174816 DOI: 10.1016/j.phrs.2025.107721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
A T cell-inflamed tumor microenvironment is predictive of better prognosis and clinical response to immunotherapy. Proteinase-activated receptor 2 (PAR2), a member of G-protein coupled receptors is involved in inflammatory process and the progression of various cancers. However, the role of PAR2 in modulating the tumor microenvironment remains unclear. Here, we found that PAR2 high-expression was associated with a favorable prognosis in patients with colorectal cancer. Intriguingly, PAR2 expression in human colorectal cancer was mainly confined to tumor cells and was significantly associated with CD8+ T cell infiltration. Tumor-intrinsic PAR2 deficiency blunted antitumor immune responses to promote tumor growth and attenuated the therapeutic efficacy of anti-PD1 in a mouse model of colon cancer. Tumors with downregulated PAR2 showed decreased CD8+ T cell infiltration and impaired effector function. Mechanistically, PAR2 activation in tumor cells induced CXCL9 and CXCL10 production via PI3K/AKT/mTOR signaling, thereby enhancing CD8+ T cell recruitment in the tumor microenvironment. In addition, PAR2 was essential for dendritic cell activation and differentiation towards conventional type 1 subset. PAR2 deficiency in dendritic cells markedly impaired their ability to prime CD8+ T cells and control tumor growth in vivo. Thus, our findings identify new roles for PAR2 in promoting antitumor immunity and provide a promising target to improve immunotherapy efficacy in colorectal cancer.
Collapse
Affiliation(s)
- Zilin Liu
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Xuehui Jiang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Ziliang Ke
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Weihong Wang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Jianqiang Tang
- Department of General Surgery, Peking University First Hospital, Beijing, China.
| | - Yun Dai
- Department of Gastroenterology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
4
|
Mansouri A, Ozair A, Bhanja D, Wilding H, Mashiach E, Haque W, Mikolajewicz N, de Macedo Filho L, Mahase SS, Machtay M, Metellus P, Dhermain F, Sheehan J, Kondziolka D, Lunsford LD, Niranjan A, Minniti G, Li J, Kalkanis SN, Wen PY, Kotecha R, McDermott MW, Bettegowda C, Woodworth GF, Brown PD, Sahgal A, Ahluwalia MS. Stereotactic radiosurgery for patients with brain metastases: current principles, expanding indications and opportunities for multidisciplinary care. Nat Rev Clin Oncol 2025; 22:327-347. [PMID: 40108412 DOI: 10.1038/s41571-025-01013-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
The management of brain metastases is challenging and should ideally be coordinated through a multidisciplinary approach. Stereotactic radiosurgery (SRS) has been the cornerstone of management for most patients with oligometastatic central nervous system involvement (one to four brain metastases), and several technological and therapeutic advances over the past decade have broadened the indications for SRS to include polymetastatic central nervous system involvement (>4 brain metastases), preoperative application and fractionated SRS, as well as combinatorial approaches with targeted therapy and immune-checkpoint inhibitors. For example, improved imaging and frameless head-immobilization technologies have facilitated fractionated SRS for large brain metastases or postsurgical cavities, or lesions in proximity to organs at risk. However, these opportunities come with new challenges and questions, including the implications of tumour histology as well as the role and sequencing of concurrent systemic treatments. In this Review, we discuss these advances and associated challenges in the context of ongoing clinical trials, with insights from a global group of experts, including recommendations for current clinical practice and future investigations. The updates provided herein are meaningful for all practitioners in clinical oncology.
Collapse
Affiliation(s)
- Alireza Mansouri
- Department of Neurosurgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA.
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA.
- Department of Neurosurgery, Penn State College of Medicine, Pennsylvania State University, Hershey, PA, USA.
| | - Ahmad Ozair
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Debarati Bhanja
- Department of Neurological Surgery, NYU Langone Health, New York University, New York, NY, USA
| | - Hannah Wilding
- Department of Neurosurgery, Penn State College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Elad Mashiach
- Department of Neurological Surgery, NYU Langone Health, New York University, New York, NY, USA
| | - Waqas Haque
- Division of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Nicholas Mikolajewicz
- Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Leonardo de Macedo Filho
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Neurosurgery, Penn State College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Sean S Mahase
- Department of Radiation Oncology, Penn State Cancer Institute, Hershey, PA, USA
| | - Mitchell Machtay
- Department of Radiation Oncology, Penn State Cancer Institute, Hershey, PA, USA
| | - Philippe Metellus
- Department of Neurosurgery, Ramsay Santé, Hôpital Privé Clairval, Marseille, France
| | - Frédéric Dhermain
- Radiation Therapy Department, Institut Gustave Roussy, Villejuif, France
| | - Jason Sheehan
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, USA
| | - Douglas Kondziolka
- Department of Neurological Surgery, NYU Langone Health, New York University, New York, NY, USA
| | - L Dade Lunsford
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ajay Niranjan
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Giuseppe Minniti
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza IRCCS Neuromed, Pozzilli, Italy
| | - Jing Li
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven N Kalkanis
- Department of Neurosurgery, Henry Ford Health System, Detroit, MI, USA
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Michael W McDermott
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Department of Neurosurgery, Miami Neuroscience Institute, Baptist Health South Florida, Miami, FL, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Brain Tumour Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
- University of Maryland-Medicine Institute for Neuroscience Discovery, Baltimore, MD, USA
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Arjun Sahgal
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Manmeet S Ahluwalia
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA.
| |
Collapse
|
5
|
Reddy SU, Sham R, Smith K, Gaire B, Vancura A, Vancurova I. Immune checkpoint protein PD-L1 promotes transcription of angiogenic and oncogenic proteins IL-8, Bcl3, and STAT1 in ovarian cancer cells. J Biol Chem 2025; 301:108339. [PMID: 39988077 PMCID: PMC11982968 DOI: 10.1016/j.jbc.2025.108339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/29/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025] Open
Abstract
Immunotherapies blocking cell surface signaling of the immune checkpoint PD-L1 have shown great promise in several cancers, but the results have been disappointing in ovarian cancer (OC). One of the main underlying mechanisms likely consists of the cell-intrinsic intracellular functions of PD-L1, which are incompletely understood. The expression of PD-L1 in OC cells is induced by interferon-γ (IFNγ), a pleiotropic cytokine produced in response to chemotherapy or immune checkpoint blockade. We have recently shown that IFNγ induces expression of the proto-oncogene Bcl3, the proangiogenic chemokine interleukin-8 (IL-8)-CXCL8, and the transcription factor STAT1, resulting in increased OC cell proliferation and migration. Here, we report that IFNγ-induced expression of PD-L1 results in PD-L1 recruitment to IL-8, Bcl3, and STAT1 promoters. The occupancy of PD-L1 at IL-8, Bcl3, and STAT1 promoters is associated with increased histone acetylation and RNA polymerase II recruitment to these promoters. Suppression of IFNγ-induced PD-L1 decreases the expression of IL-8, Bcl3, and PD-L1 and increases apoptosis in OC cells. Together, these findings demonstrate that PD-L1 promotes transcription of IL-8, Bcl3, and STAT1, thus providing a novel function of PD-L1 in cancer cells, and suggesting that the increased IL-8, Bcl3, and STAT1 expression mediated by PD-L1 might contribute to the limited effectiveness of cancer immunotherapies targeting the surface expression of PD-L1 in OC.
Collapse
Affiliation(s)
- Suprataptha U Reddy
- Department of Biological Sciences, St John's University, New York, New York, USA
| | - Rachel Sham
- Department of Biological Sciences, St John's University, New York, New York, USA
| | - Khalani Smith
- Department of Biological Sciences, St John's University, New York, New York, USA
| | - Bijaya Gaire
- Department of Biological Sciences, St John's University, New York, New York, USA
| | - Ales Vancura
- Department of Biological Sciences, St John's University, New York, New York, USA
| | - Ivana Vancurova
- Department of Biological Sciences, St John's University, New York, New York, USA.
| |
Collapse
|
6
|
Zhang A, Fan L, Liu Q, Zuo X, Zhu J. Immunological Effects of Proton Radiotherapy: New Opportunities and Challenges in Cancer Therapy. CANCER INNOVATION 2025; 4:e70003. [PMID: 40061827 PMCID: PMC11885950 DOI: 10.1002/cai2.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/23/2024] [Accepted: 01/16/2025] [Indexed: 03/14/2025]
Abstract
Radiation therapy can be categorised by particle type into photon, proton and heavy ion therapies. Proton radiotherapy is highlighted due to its unique physical properties, such as the Bragg peak and minimal exit dose, which offer superior dose distribution. This makes proton radiotherapy especially advantageous for treating tumours near vital organs with complex structures, such as gliomas near the brain, nasopharyngeal carcinoma near the brainstem and mediastinal tumours near the heart. Proton irradiation can induce distant effects through immunogenicity within the target area. The reduced low-dose zone outside the target provides better lymphatic system protection and immune benefits. Additionally, combining proton radiotherapy with immunotherapy may offer further biological advantages. These features make proton radiotherapy a promising option in cancer treatment. This article may aid in the understanding of proton radiotherapy and its immune effects and lead to new effective options for tumour treatment.
Collapse
Affiliation(s)
- Anhang Zhang
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| | - Liyuan Fan
- Department of Radiation OncologyQilu Hospital of Shandong UniversityJinanShandongChina
| | - Qi Liu
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| | - Xiaoxin Zuo
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| | - Jian Zhu
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| |
Collapse
|
7
|
Yin Z, Zhang H, Zhang K, Yue J, Tang R, Wang Y, Deng Q, Yu Q. Impacts of combining PD-L1 inhibitor and radiotherapy on the tumour immune microenvironment in a mouse model of esophageal squamous cell carcinoma. BMC Cancer 2025; 25:474. [PMID: 40087599 PMCID: PMC11909915 DOI: 10.1186/s12885-025-13801-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND The combination of radiation with immune checkpoint inhibitors (ICIs) has been demonstrated to display synergistic effects in solid cancers. Nevertheless, the anti-tumor effect of combining radiation with programmed cell death 1 ligand 1 (PD-L1) inhibitor in esophageal squamous cell carcinoma (ESCC) has remained unclear. Therefore, the objectives of our study were to evaluate the anti-tumor effects of PD-L1 inhibitors combined with radiotherapy in a mouse model of ESCC and to depict the immune landscape within the tumor microenvironment (TME). METHODS Murine ESCC cells (mEC25) were injected subcutaneously into the right flanks of C57BL/6 mice. Tumor-bearing mice were exposed to different treatments: IgG antibody (control), anti-PD-L1 antibody, radiation, or radiation + anti-PD-L1 antibody. Tumor growth and survival time of mice were monitored. Tumour immune microenvironment was assessed by flow cytometry, including CD4+T cells, CD8+T cells, regulatory T cells (Tregs), tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and the activation and exhaustion of CD8+T cell. In addition, transcriptomic analysis was used to examine the changes in immune gene expression in the TME. RESULTS Radiotherapy combined with anti-PD-L1 inhibitors (radioimmunotherapy) synergistically enhanced anti-tumor immune response, leading to decreased tumor growth and prolonged survival of tumor-bearing mice. The radioimmunotherapy increased the infiltration of CD8+ T cells, the ratio of CD8+ T cells to Tregs, the population of central memory CD8+ T cells (TCM), interferon-gamma (IFN-γ) secretion of tumor-infiltrating CD8+ T cells, and reduced the accumulation of M2-type TAMs and Tregs in the TME in mouse model. In addition, the radioimmunotherapy induced anti-tumor immune response in the spleen and tumor-draining lymph node (TDLN). Moreover, transcriptomic analysis suggested that the radioimmunotherapy promoted the activation of immune regulatory pathways and increased the expression of cytokines such as CXCL9 and CXCL10, thus creating an immunoinflammatory tumor microenvironment. CONCLUSIONS Our research revealed that anti-PD-L1 inhibitors combined with radiotherapy caused systemic anti-tumor immunity by reshaping the immune microenvironment in a mouse model of ESCC.
Collapse
Affiliation(s)
- Zihao Yin
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Hongfang Zhang
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Ke Zhang
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Jing Yue
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Rongjun Tang
- Hyperthermia Center, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Yaping Wang
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
| | - Qinghua Deng
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China.
| | - Qingqing Yu
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China.
| |
Collapse
|
8
|
Castro F, Pinto ML, Leite Pereira C, Serre K, Costa ÂM, Cavadas B, Barbosa MA, Vermaelen K, León S, Serrano D, Gärtner F, Calvo A, Gonçalves RM, De Wever O, Oliveira MJ. Chitosan/γ-PGA nanoparticles and IFN-γ immunotherapy: A dual approach for triple-negative breast cancer treatment. J Control Release 2025; 379:621-635. [PMID: 39832747 DOI: 10.1016/j.jconrel.2025.01.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/08/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Interferon-γ (IFN-γ) is a key mediator in antitumor immunity and immunotherapy responses, yet its clinical applications remain restricted to chronic granulomatous disease and malignant osteopetrosis. IFN-γ effectiveness as a standalone treatment has shown limited success in clinical trials and its potential for synergistic effects when combined with immunotherapies is under clinical exploration. A particularly compelling combination is that of IFN-γ with Toll-like receptor (TLR) agonists that holds significant promise for cancer treatment. Previously, we demonstrated chitosan (Ch)/poly(γ-glutamic acid) (γ-PGA) nanoparticles (NPs), known to activate TLRs, as adjuvants to radiotherapy by remodeling breast tumor microenvironment and systemic immunosuppression. These immunomodulatory abilities make Ch/γ-PGA NPs promising adjuvants to IFN-γ-based therapies. Here, we addressed the synergistic therapeutic potential of combining Ch/γ-PGA NPs with IFN-γ therapy in the 4T1 orthotopic breast tumor mouse model. While control animals (placebo-treated) had progressive tumor growth and lung metastases, those treated with either NPs or IFN-γ alone had a significant slower tumor growth. Remarkably, primary tumor growth was halted throughout the duration of the treatment when both treatments were combined. Although the animals did not achieve durable complete responses upon treatment withdrawal, it was notable that the NPs plus IFN-γ group presented a lower lung metastatic burden compared to other groups. Systemically, the combination therapy slightly attenuated immunosuppression and the percentage of splenic myeloid cells, while increased the percentage of T helper 1 cells and of cytotoxic T cells. Overall, this proof-of-concept study suggests that Ch/γ-PGA NPs potentiate IFN-γ effects to reduce tumor progression, presenting a novel approach for anticancer strategies.
Collapse
Affiliation(s)
- Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | - Marta Laranjeiro Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Catarina Leite Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Karine Serre
- GIMM - Gulbenkian Institute for Molecular Medicine, Avenida Prof. Egas Moniz, Lisboa, Portugal
| | - Ângela Margarida Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Bruno Cavadas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Mário Adolfo Barbosa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Karim Vermaelen
- Tumor Immunology Laboratory, Department of Pulmonary Medicine and Immuno-Oncology Network Ghent, Ghent University Hospital, Ghent, Belgium; CRIG - Cancer Research Institute Ghent, Ghent University, Belgium
| | - Sergio León
- IdiSNA - Navarra Institute for Health Research, Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, Pamplona, Spain; CIBERONC (-) Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Diego Serrano
- IdiSNA - Navarra Institute for Health Research, Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, Pamplona, Spain
| | - Fátima Gärtner
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Alfonso Calvo
- IdiSNA - Navarra Institute for Health Research, Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, Pamplona, Spain; CIBERONC (-) Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Raquel Madeira Gonçalves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Olivier De Wever
- CRIG - Cancer Research Institute Ghent, Ghent University, Belgium; LECR - Laboratory Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Belgium
| | - Maria José Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
9
|
Zeng X, Jin X, Leng J, Zhang S, Wang Y, Chen J, Zhang S, Teng L, Hu Z, Zhou S, Zeng Z, Long J. High-dose radiation induces dendritic cells maturation by promoting immunogenic cell death in nasopharyngeal carcinoma. Front Immunol 2025; 16:1554018. [PMID: 40040692 PMCID: PMC11876370 DOI: 10.3389/fimmu.2025.1554018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 01/31/2025] [Indexed: 03/06/2025] Open
Abstract
Aim and background Due to the radiosensitivity and deep anatomical location of nasopharyngeal carcinoma (NPC), radiotherapy serves as the cornerstone of standardized treatment for this malignancy. Beyond its cytotoxic effects, radiotherapy can serve as an immunological adjuvant by inducing immunogenic cell death (ICD). Dendritic cells (DCs), as potent antigen-presenting cells, play a critical role in tumor immunotherapy, but their exact role in the ICD process of NPC remains unclear. The effects of high-dose radiation (≥2 Gy) on DCs and the type of immune response it elicits in NPC have not been fully elucidated. Methods An in vitro study was conducted to assess whether ICD of NPC 5-8F cells induced by high-dose radiation could regulate the immune response of DCs. Specifically, the maturation and antigen-presenting capacity of DCs were evaluated following co-culture with NPC cells exposed to high-dose radiation. Results High-dose radiation was found to induce ICD in NPC 5-8F cells, as evidenced by increased pro-inflammatory factor levels and reduced anti-inflammatory factor levels in the cell culture supernatant. Co-culture with NPC cells exposed to high-dose radiation for 15 minutes significantly enhanced the expression of surface molecules on DCs, promoting their immune sensitization. Conclusion High-dose radiation-induced apoptosis of NPC 5-8F cells is a form of ICD, which plays an important role in regulating DC immune function. These findings provide insight into the immunomodulatory effects of radiotherapy in NPC and its potential to enhance tumor immunotherapy through DC activation.
Collapse
Affiliation(s)
- Xianlin Zeng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Xianhuai Jin
- Department of Oncology, Guiyang Public Health Clinical Center, Guiyang, Guizhou, China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ji Leng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shuai Zhang
- Department of Interventional Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yun Wang
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Jin Chen
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shichao Zhang
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Lijing Teng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Zuquan Hu
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shi Zhou
- Department of Interventional Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhu Zeng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Jinhua Long
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
10
|
Iwamoto FM, Tanguturi SK, Nayak L, Wang TJ, Desai A, Lustig RA, Bagley S, Wong ET, Hertan LM, McCluskey C, Hayden J, Muzikansky A, Nakhawa S, Japo J, Bossi CC, Meylan M, Tian Y, Barlow GL, Speliakos P, Ayoub G, Meredith DM, Ligon KL, Haas-Kogan D, Huang K, Wucherpfennig KW, Wen PY, Reardon DA. Re-Irradiation Plus Pembrolizumab: A Phase II Study for Patients with Recurrent Glioblastoma. Clin Cancer Res 2025; 31:316-327. [PMID: 39513953 DOI: 10.1158/1078-0432.ccr-24-1629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/15/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Radiotherapy may enhance antitumor immune responses by several mechanisms, including induction of immunogenic cell death. We performed a phase 2 study of pembrolizumab with re-irradiation in patients with recurrent glioblastoma. PATIENTS AND METHODS Sixty patients with recurrent glioblastoma received pembrolizumab with re-irradiation alone (cohort A, bevacizumab-naïve; n = 30) or with bevacizumab continuation (cohort B, n = 30). Dual primary endpoints, including the overall response rate and overall survival (OS) at either 12 (OS-12; cohort A) or 6 months (OS-6; cohort B), were assessed per cohort relative to historic benchmarks. Paired paraffin-embedded formalin-fixed tumor samples were assessed for immunologic biomarkers by IHC using digital quantification and co-detection-by-indexing (CODEX). RESULTS Study therapy was well tolerated, with most toxicities being grade ≤3. For cohort B, the primary endpoint of OS-6 was achieved (57%); however, survival was not improved for cohort A patients. The overall response rate was 3.3% and 6.7% for cohorts A and B, respectively. CODEX analysis of paired tumor samples from five patients revealed an increase of activated T cells in the tumor microenvironment after study therapy. CONCLUSIONS Compared with historic controls, re-irradiation plus pembrolizumab seemed to improve survival among bevacizumab-refractory patients but not among bevacizumab-naïve patients. CODEX revealed evidence of intratumoral infiltration of activated immune effector cells. A randomized, properly controlled trial of PD-1 blockade plus re-irradiation is warranted to further evaluate this regimen for bevacizumab-refractory patients, but alternative approaches are needed for bevacizumab-naïve patients.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Female
- Middle Aged
- Male
- Glioblastoma/therapy
- Glioblastoma/pathology
- Glioblastoma/mortality
- Glioblastoma/drug therapy
- Aged
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/therapy
- Neoplasm Recurrence, Local/mortality
- Adult
- Re-Irradiation/methods
- Re-Irradiation/adverse effects
- Bevacizumab/administration & dosage
- Brain Neoplasms/therapy
- Brain Neoplasms/pathology
- Brain Neoplasms/mortality
- Treatment Outcome
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/administration & dosage
- Combined Modality Therapy
Collapse
Affiliation(s)
- Fabio M Iwamoto
- Department of Neurology, Columbia University Medical Center, New York, New York
| | - Shyam K Tanguturi
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lakshmi Nayak
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tony J Wang
- Department of Radiation Oncology, Columbia University Medical Center, New York, New York
| | - Arati Desai
- Department of Medical Oncology, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Robert A Lustig
- Department of Radiation Oncology, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Stephen Bagley
- Department of Medical Oncology, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Eric T Wong
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Lauren M Hertan
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Christine McCluskey
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Julia Hayden
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alona Muzikansky
- Department of Biostatistics, Massachusetts General Hospital, Boston, Massachusetts
| | - Shreya Nakhawa
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Julia Japo
- Department of Neuropathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Connor C Bossi
- Department of Neuropathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Maxime Meylan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ye Tian
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Graham L Barlow
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Paul Speliakos
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Georges Ayoub
- Department of Neuropathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David M Meredith
- Department of Neuropathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Keith L Ligon
- Department of Neuropathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kun Huang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
11
|
Chen Y, Qi F, Sun C, Jiang P, Xue X, Yang X, Li X, He X, Wang Y, Zhang T. Navigating the landscape of neoadjuvant immunotherapy for NSCLC: progress and controversies. Ther Adv Med Oncol 2025; 17:17588359241312501. [PMID: 39781239 PMCID: PMC11707791 DOI: 10.1177/17588359241312501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Recently, attention has increasingly centered on non-small-cell lung cancer (NSCLC) with immune checkpoint inhibitors application. Numerous clinical studies have underscored the potential of immunotherapy in treating resectable NSCLC, highlighting its role in improving patient outcomes. However, despite these promising results, there is ongoing debate regarding the efficacy of immunological combination therapy strategies, the prevalence of treatment-related side effects, the identification of predictive biomarkers, and various other challenges within the neoadjuvant context. Careful consideration is essential to maximize the benefits of immunotherapy for patients with resectable NSCLC. This article offers a detailed overview of recent advancements in neoadjuvant immunotherapy for resectable NSCLC. By examining these developments, we aim to provide new perspectives and valuable insights into the benefits and challenges of applying neoadjuvant immunotherapy in clinical settings.
Collapse
Affiliation(s)
- Yuzhu Chen
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Fei Qi
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Chenhao Sun
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Peng Jiang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiangyu Xue
- Department of Biochemistry and Molecular Biology, Heilongjiang Provincial Science and Technology Innovation Team in Higher Education Institutes for Infection and Immunity, Harbin Medical University, Harbin, China
| | - Xiaomei Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
- Joint Laboratory for Precision Diagnosis and Treatment Translational Research in Malignant Tumors, Gynecologic Oncology Basic and Clinical Research Laboratory, Capital Medical University, Beijing, China
| | - Xiaomi Li
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xin He
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yishuo Wang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Tongmei Zhang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, No. 9 Beiguan Street, Tongzhou District, Beijing 101149, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Hosseinalizadeh H, Wang LS, Mirzaei H, Amoozgar Z, Tian L, Yu J. Emerging combined CAR-NK cell therapies in cancer treatment: Finding a dancing partner. Mol Ther 2025:S1525-0016(24)00895-5. [PMID: 39754357 DOI: 10.1016/j.ymthe.2024.12.057] [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: 03/21/2024] [Revised: 09/21/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025] Open
Abstract
In recent decades, immunotherapy with chimeric antigen receptors (CARs) has revolutionized cancer treatment and given hope where other cancer therapies have failed. CAR-natural killer (NK) cells are NK cells that have been engineered ex vivo with a CAR on the cell membrane with high specificity for specific target antigens of tumor cells. The impressive results of several studies suggest that CAR-NK cell therapy has significant potential and successful performance in cancer treatment. Despite its effectiveness, CAR-NK cell therapy can have significant challenges when it comes to treating cancer. These challenges include tumor heterogeneity, antigen escape, an immunosuppressive tumor microenvironment, limited tissue migration from blood, exhaustion of CAR-NK cells, and inhibition by immunosuppressive checkpoint molecule signaling, etc. In CAR-T cell therapy, the use of combined approaches has shown encouraging outcomes for tumor regression and improved cancer treatment compared to single therapies. Therefore, to overcome these significant challenges in CAR-NK cells, innovative combination therapies of CAR-NK cells with other conventional therapies (e.g., chemotherapy and radiotherapy) or other immunotherapies are needed to counteract the above challenges and thereby increase the activity of CAR-NK cells. This review comprehensively discusses various cancer-treatment approaches in combination with CAR-NK cell therapy in the hope of providing valuable insights that may improve cancer treatment in the near future.
Collapse
Affiliation(s)
- Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Li-Shu Wang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lei Tian
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| | - Jianhua Yu
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
13
|
Ruan J, Zhou D, Zhang Y, Zhao D, Wei C, Hu K, Zhang F, Hou X, Zhang W. Hyper-fractionated radiotherapy as a bridging strategy to enhance CAR-T efficacy by regulating T-cell co-stimulatory molecules in relapsed/refractory diffuse large B-cell lymphoma. Front Immunol 2024; 15:1481080. [PMID: 39687615 PMCID: PMC11646978 DOI: 10.3389/fimmu.2024.1481080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/13/2024] [Indexed: 12/18/2024] Open
Abstract
Background Bridging therapy can prevent patients from disease progression while waiting for CAR-T cell preparation. Hyper-fractionated radiotherapy can achieve an effective target dose within a short period, minimize radiation damage, and may modify immune environment compared to conventional radiotherapy. Aims This study aims to investigate the efficacy and safety of bridging hyper-fractionated radiotherapy in combination with CAR-T therapy for relapsed/refractory diffuse large B-cell lymphoma. The potential mechanisms were explored. Methods This is a prospective pilot study. After T-cell collection, the patients underwent hyper-fractionated radiotherapy at lesion sites with 1.5 Gy twice daily for 10 days before CAR-T cell infusion. Peripheral blood immune cell subsets and quantitative serum proteomics were assessed before radiotherapy and after radiotherapy before CAR-T cell infusion. Results A total of 13 patients have been enrolled. The median follow-up time was 6 (3-24) months after CAR-T infusion. At 3-month follow-up, 9/13(69%) patients had CR, 1/13(8%) patient had PR, 1/13(8%) patient remained SD, and 2/13(15%) patients died of disease progression. The local recurrence rate was 1/13(8%). Seven patients have been followed up for more than 6 months, and they remain in CR. The median PFS and OS were not reached. No grade 3-4 CRS or ICANS were reported. After hyper-fractionated radiotherapy, peripheral PD1+CD8+T/T ratio significantly decreased while quantitative serum proteomics profiling showed a decrease in sCD28. Conclusion Hyper-fractionated radiotherapy can rapidly control tumor progression sites without delaying the infusion time. This approach can improve the ORR and does not increase the incidence of CRS and ICANS. The mechanism may be related to the regulation of T-cell co-stimulatory molecules, which demands further exploration.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/radiotherapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Male
- Middle Aged
- Female
- Immunotherapy, Adoptive/methods
- Adult
- Aged
- Prospective Studies
- Receptors, Chimeric Antigen/immunology
- Dose Fractionation, Radiation
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Pilot Projects
- Neoplasm Recurrence, Local/radiotherapy
- Treatment Outcome
- Combined Modality Therapy
Collapse
Affiliation(s)
- Jing Ruan
- Department of Hematology, Peking Union Medical College Hospital, Beijing, China
| | - Daobin Zhou
- Department of Hematology, Peking Union Medical College Hospital, Beijing, China
| | - Yan Zhang
- Department of Hematology, Peking Union Medical College Hospital, Beijing, China
| | - Danqing Zhao
- Department of Hematology, Peking Union Medical College Hospital, Beijing, China
| | - Chong Wei
- Department of Hematology, Peking Union Medical College Hospital, Beijing, China
| | - Ke Hu
- Department of Radiotherapy, Peking Union Medical College Hospital, Beijing, China
| | - Fuquan Zhang
- Department of Radiotherapy, Peking Union Medical College Hospital, Beijing, China
| | - Xiaorong Hou
- Department of Radiotherapy, Peking Union Medical College Hospital, Beijing, China
| | - Wei Zhang
- Department of Hematology, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
14
|
Hu J, Zhang J, Wan S, Zhang P. Neoadjuvant immunotherapy for non-small cell lung cancer: Opportunities and challenges. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:224-239. [PMID: 39834585 PMCID: PMC11742355 DOI: 10.1016/j.pccm.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Indexed: 01/22/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment landscape for resectable non-small cell lung cancer. Numerous trials have explored the use of ICIs, either as monotherapy or in combination with other therapies, in the neoadjuvant setting for stage I-III non-small cell lung cancer. Most trials have demonstrated neoadjuvant immunotherapy to be safe and to have remarkable efficacy, with a high pathological response rate and significantly improved event-free survival. This review summarizes the findings of Phase I-III clinical trials investigating various neoadjuvant regimens, including ICI monotherapy, ICI therapy combined with chemotherapy, ICI plus anti-angiogenic therapy, dual ICI therapy, and ICI therapy in combination with radiotherapy or chemoradiotherapy. We discuss the benefits and outcomes associated with each approach. Despite the results being promising, several unresolved issues remain, including identification of reliable biomarkers, the appropriate duration of therapy, the optimal treatment regimen for tumors with high programmed cell death ligand 1 (PD-L1) expression, the false-negative pathological complete response rate, and the role of digital pathology in assessing the response to treatment. Resistance to immunotherapy, in particular, remains a significant barrier to effective use of ICIs. Given the critical influence of the tumor microenvironment (TME) on the response to treatment, we examine the characteristics of the TME in both responsive and resistant tumors as well as the dynamic changes that occur in the TME in response to neoadjuvant immunotherapy. We also summarize the mechanisms underlying T cell responses following neoadjuvant immunotherapy and provide a perspective on strategies to enhance the understanding of tumor heterogeneity, therapy-driven TME remodeling, and overcoming resistance to therapy. Finally, we propose future directions for advancements in personalized neoadjuvant immunotherapy.
Collapse
Affiliation(s)
- Junjie Hu
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Jing Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Shiyue Wan
- Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- The 1st School of Medicine, the 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Shihezi University Medical College, Shihezi, Xinjiang 832000, China
| |
Collapse
|
15
|
Beatty GL, Jaffee EM. Exogenous or in situ vaccination to trigger clinical responses in pancreatic cancer. Carcinogenesis 2024; 45:826-835. [PMID: 39514560 PMCID: PMC11584293 DOI: 10.1093/carcin/bgae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/16/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a lethal disease for which remarkable therapeutic resistance is the norm. Conventional immunotherapies, like immune checkpoint inhibitors, show limited efficacy in PDA due to a remarkably immunosuppressive tumor microenvironment (TME) and systemic inflammation. This review discusses the potential of both exogenous and in situ vaccination strategies to overcome these barriers and enhance anti-tumor immunity in PDA. Exogenous vaccines, including whole-cell, dendritic cell, peptide, and nucleic acid-based vaccines, have shown varying degrees of promise but face challenges related to antigen selection, production complexities, and patient-specific factors. In contrast, in situ vaccination strategies leverage conventional cytotoxic therapies, such as chemotherapy and radiation therapy, to induce immunogenic cell death and modulate the TME with the aim to stimulate anti-tumor immunity. While preclinical studies support the use of in situ vaccination, balancing the stimulatory and inhibitory effects is likely fundamental to eliciting productive anti-tumor responses in patients. Ongoing research seeks to identify new innovative strategies that can harness the endogenous immune response and trigger in situ vaccination. Overall, while both vaccination approaches offer significant potential, further research and clinical trials will be needed to optimize these strategies for improving patient outcomes in PDA.
Collapse
Affiliation(s)
- Gregory L Beatty
- Abramson Cancer Center, University of Pennsylvania, Perelman School of Medicine, 3400 Civic Center Blvd, South Pavilion, Rm 8-107, Philadelphia, PA 19104, United States
| | - Elizabeth M Jaffee
- The Sidney Kimmel Comprehensive Cancer Center, The Bloomberg Kimmel Institute for Immunotherapy, The Cancer Convergence Institute, Johns Hopkins University School of Medicine, 4M07 Bunting Blaustein Cancer Research Building, 1650 Orleans Street, Baltimore, MD 21287, United States
| |
Collapse
|
16
|
Chhipa AS, Boscaro V, Gallicchio M, Patel S. The curious case of type I interferon signaling in cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189204. [PMID: 39477031 DOI: 10.1016/j.bbcan.2024.189204] [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: 06/23/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Cytokines are the crucial signaling proteins that mediate the crosstalks between the cells of tumor microenvironment (TME). Interferon-1 (IFN-1) are the important cytokines that are widely known for their tumor suppressive roles comprising of cancer cell intrinsic and extrinsic mechanisms. Despite having known antitumor effects, IFN-1 are also reported to have tumor promoting functions under varying circumstances. This dichotomy in the functions of IFN-1 is largely attributed to the acute and chronic activation of IFN-1 signaling in TME. The chronic activation of IFN-1 signaling in tumor cells results in altered stimulation of downstream pathways that result in the expression of tumor promoting proteins, while the acute IFN-1 signaling activation maintains its tumor inhibiting functions. In the present review, we have discussed the anti- and pro-tumor actions of IFN-1 signaling under acute and chronic IFN-1 signaling activation. We have also discussed the downstream changes in signaling components that result in tumor supportive functions of a constitutive IFN-1 signaling. We have further discussed the possible strategies to overcome the detrimental effects of chronic IFN-1 pathway activation and to successfully employ IFN-1 for their beneficial anti-tumor effects.
Collapse
Affiliation(s)
- Abu Sufiyan Chhipa
- Department of Pharmacology, Institute of Pharmacy, Nirma University, 382481 Ahmedabad, India; Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Valentina Boscaro
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | | | - Snehal Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, 382481 Ahmedabad, India.
| |
Collapse
|
17
|
Li S, Chen K, Sun Z, Chen M, Pi W, Zhou S, Yang H. Radiation drives tertiary lymphoid structures to reshape TME for synergized antitumour immunity. Expert Rev Mol Med 2024; 26:e30. [PMID: 39438247 PMCID: PMC11505612 DOI: 10.1017/erm.2024.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 04/24/2024] [Accepted: 07/15/2024] [Indexed: 10/25/2024]
Abstract
Radiotherapy (RT) plays a key role in the tumour microenvironment (TME), impacting the immune response via cellular and humoral immunity. RT can induce local immunity to modify the TME. It can stimulate dendritic cell maturation and T-cell infiltration. Moreover, B cells, macrophages and other immune cells may also be affected. Tertiary lymphoid structure (TLS) is a unique structure within the TME and a class of aggregates containing T cells, B cells and other immune cells. The maturation of TLS is determined by the presence of mature dendritic cells, the density of TLS is determined by the number of immune cells. TLS maturation and density both affect the antitumour immune response in the TME. This review summarized the recent research on the impact and the role of RT on TLS, including the changes of TLS components and formation conditions and the mechanism of how RT affects TLS and transforms the TME. RT may promote TLS maturation and density to modify the TME regarding enhanced antitumour immunity.
Collapse
Affiliation(s)
- Shuling Li
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Kuifei Chen
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Zhenwei Sun
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Meng Chen
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Wenhu Pi
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Suna Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Haihua Yang
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| |
Collapse
|
18
|
Wu B, Zhang B, Li B, Wu H, Jiang M. Cold and hot tumors: from molecular mechanisms to targeted therapy. Signal Transduct Target Ther 2024; 9:274. [PMID: 39420203 PMCID: PMC11491057 DOI: 10.1038/s41392-024-01979-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/20/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment, particularly through immune checkpoint blockade (ICB), which has shown notable clinical benefits across various tumor types. Despite the transformative impact of ICB treatment in cancer therapy, only a minority of patients exhibit a positive response to it. In patients with solid tumors, those who respond well to ICB treatment typically demonstrate an active immune profile referred to as the "hot" (immune-inflamed) phenotype. On the other hand, non-responsive patients may exhibit a distinct "cold" (immune-desert) phenotype, differing from the features of "hot" tumors. Additionally, there is a more nuanced "excluded" immune phenotype, positioned between the "cold" and "hot" categories, known as the immune "excluded" type. Effective differentiation between "cold" and "hot" tumors, and understanding tumor intrinsic factors, immune characteristics, TME, and external factors are critical for predicting tumor response and treatment results. It is widely accepted that ICB therapy exerts a more profound effect on "hot" tumors, with limited efficacy against "cold" or "altered" tumors, necessitating combinations with other therapeutic modalities to enhance immune cell infiltration into tumor tissue and convert "cold" or "altered" tumors into "hot" ones. Therefore, aligning with the traits of "cold" and "hot" tumors, this review systematically delineates the respective immune characteristics, influencing factors, and extensively discusses varied treatment approaches and drug targets based on "cold" and "hot" tumors to assess clinical efficacy.
Collapse
Affiliation(s)
- Bo Wu
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bo Zhang
- Department of Youth League Committee, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bowen Li
- Department of Pancreatic and Gastrointestinal Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Haoqi Wu
- Department of Gynaecology and Obstetrics, The Second Hospital of Dalian Medical University, Dalian, China
| | - Meixi Jiang
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
19
|
Stilpeanu RI, Secara BS, Cretu-Stancu M, Bucur O. Oncolytic Viruses as Reliable Adjuvants in CAR-T Cell Therapy for Solid Tumors. Int J Mol Sci 2024; 25:11127. [PMID: 39456909 PMCID: PMC11508774 DOI: 10.3390/ijms252011127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Although impactful scientific advancements have recently been made in cancer therapy, there remains an opportunity for future improvements. Immunotherapy is perhaps one of the most cutting-edge categories of therapies demonstrating potential in the clinical setting. Genetically engineered T cells express chimeric antigen receptors (CARs), which can detect signals expressed by the molecules present on the surface of cancer cells, also called tumor-associated antigens (TAAs). Their effectiveness has been extensively demonstrated in hematological cancers; therefore, these results can establish the groundwork for their applications on a wide range of requirements. However, the application of CAR-T cell technology for solid tumors has several challenges, such as the existence of an immune-suppressing tumor microenvironment and/or inadequate tumor infiltration. Consequently, combining therapies such as CAR-T cell technology with other approaches has been proposed. The effectiveness of combining CAR-T cell with oncolytic virus therapy, with either genetically altered or naturally occurring viruses, to target tumor cells is currently under investigation, with several clinical trials being conducted. This narrative review summarizes the current advancements, opportunities, benefits, and limitations in using each therapy alone and their combination. The use of oncolytic viruses offers an opportunity to address the existing challenges of CAR-T cell therapy, which appear in the process of trying to overcome solid tumors, through the combination of their strengths. Additionally, utilizing oncolytic viruses allows researchers to modify the virus, thus enabling the targeted delivery of specific therapeutic agents within the tumor environment. This, in turn, can potentially enhance the cytotoxic effect and therapeutic potential of CAR-T cell technology on solid malignancies, with impactful results in the clinical setting.
Collapse
MESH Headings
- Humans
- Neoplasms/therapy
- Neoplasms/immunology
- Oncolytic Viruses/genetics
- Oncolytic Viruses/immunology
- Immunotherapy, Adoptive/methods
- Oncolytic Virotherapy/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Animals
- Tumor Microenvironment/immunology
- T-Lymphocytes/immunology
- Combined Modality Therapy/methods
- Adjuvants, Immunologic
- Antigens, Neoplasm/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Ruxandra Ilinca Stilpeanu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
| | - Bianca Stefania Secara
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
| | | | - Octavian Bucur
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
- Genomics Research and Development Institute, 020021 Bucharest, Romania
- Viron Molecular Medicine Institute, Boston, MA 02108, USA
| |
Collapse
|
20
|
Paquette B, Oweida A. Combination of radiotherapy and immunotherapy in duality with the protumoral action of radiation. Cancer Radiother 2024; 28:484-492. [PMID: 39304400 DOI: 10.1016/j.canrad.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 09/22/2024]
Abstract
Radiotherapy is widely used to treat various cancers. Its combination with immune checkpoint inhibitors is intensively studied preclinically and clinically. Although the first results were very encouraging, the number of patients who respond positively remains low, and the therapeutic benefit is often temporary. This review summarizes how radiation can stimulate an antitumor immune response and its combination with immunotherapy based on inhibiting immune checkpoints. We will provide an overview of radiotherapy parameters that should be better controlled to avoid downregulating the antitumor immune response. The low response rate of combining radiotherapy and immunotherapy could, at least in part, be caused by the stimulation of cancer cell invasion and metastasis development that occur at similar doses and number of radiation fractions. To end on a positive note, we explore how a targeted inhibition of the inflammatory cytokines induced by radiation with a cyclooxygenase-2 inhibitor could both support an antitumor immune response and block radiation-induced metastasis formation.
Collapse
Affiliation(s)
- Benoît Paquette
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Quebec, Canada.
| | - Ayman Oweida
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
21
|
Ghemrawi R, Abuamer L, Kremesh S, Hussien G, Ahmed R, Mousa W, Khoder G, Khair M. Revolutionizing Cancer Treatment: Recent Advances in Immunotherapy. Biomedicines 2024; 12:2158. [PMID: 39335671 PMCID: PMC11429153 DOI: 10.3390/biomedicines12092158] [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: 06/19/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer immunotherapy has emerged as a transformative approach in oncology, utilizing the body's immune system to specifically target and destroy malignant cells. This review explores the scope and impact of various immunotherapeutic strategies, including monoclonal antibodies, chimeric antigen receptor (CAR)-T cell therapy, checkpoint inhibitors, cytokine therapy, and therapeutic vaccines. Monoclonal antibodies, such as Rituximab and Trastuzumab, have revolutionized treatment paradigms for lymphoma and breast cancer by offering targeted interventions that reduce off-target effects. CAR-T cell therapy presents a potentially curative option for refractory hematologic malignancies, although challenges remain in effectively treating solid tumors. Checkpoint inhibitors have redefined the management of cancers like melanoma and lung cancer; however, managing immune-related adverse events and ensuring durable responses are critical areas of focus. Cytokine therapy continues to play a vital role in modulating the immune response, with advancements in cytokine engineering improving specificity and reducing systemic toxicity. Therapeutic vaccines, particularly mRNA-based vaccines, represent a frontier in personalized cancer treatment, aiming to generate robust, long-lasting immune responses against tumor-specific antigens. Despite these advancements, the field faces significant challenges, including immune resistance, tumor heterogeneity, and the immunosuppressive tumor microenvironment. Future research should address these obstacles through emerging technologies, such as next-generation antibodies, Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)-based gene editing, and AI-driven drug discovery. By integrating these novel approaches, cancer immunotherapy holds the promise of offering more durable, less toxic, and highly personalized treatment options, ultimately improving patient outcomes and survival rates.
Collapse
Affiliation(s)
- Rose Ghemrawi
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Lama Abuamer
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Sedra Kremesh
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Ghadeer Hussien
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Rahaf Ahmed
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Walaa Mousa
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Ghalia Khoder
- Department of Pharmaceutics and Pharmaceuticals Technology, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Mostafa Khair
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates
| |
Collapse
|
22
|
Romero Fernandez J, Cordoba Largo S, Benlloch Rodriguez R, Gil Haro B. The Effects of Gynecological Tumor Irradiation on the Immune System. Cancers (Basel) 2024; 16:2804. [PMID: 39199577 PMCID: PMC11352652 DOI: 10.3390/cancers16162804] [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: 07/04/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Radiobiology has evolved from a mechanistic model based on DNA damage and response factors into a more complex model that includes effects on the immune system and the tumor microenvironment (TME). Irradiation has an immunomodulatory effect that can manifest as increased anti-tumor immunity or immunosuppression. Irradiation promotes an inflammatory microenvironment through the release of pro-inflammatory cytokines and endothelial damage, which recruit immune system cells to the irradiated area. Radiation-induced immunogenic cell death (ICD), characterized by the release of damage-associated molecular patterns (DAMPs) and tumor antigens, triggers an anti-tumor immune response of both innate and adaptive immunity. Anti-tumor immunity can manifest at a distance from the irradiated area, a phenomenon known as the abscopal effect (AE), which involves dendritic cells and CD8+ T cells. Irradiation also produces an immunosuppressive effect mediated by tumor-associated macrophages (TAMs) and regulatory T lymphocytes (Tregs), which counterbalances the immunostimulatory effect. In this work, we review the mechanisms involved in the radiation-induced immune response, which support the combined treatment of RT and immunotherapy, focusing, where possible, on gynecologic cancer.
Collapse
Affiliation(s)
- Jesus Romero Fernandez
- Radiation Oncology Department, Hospital Universitario Puerta de Hierro, C. Joaquín Rodrigo 1, 28222 Majadahonda, Spain; (S.C.L.); (R.B.R.); (B.G.H.)
| | | | | | | |
Collapse
|
23
|
Reddy SU, Sadia FZ, Vancura A, Vancurova I. IFNγ-Induced Bcl3, PD-L1 and IL-8 Signaling in Ovarian Cancer: Mechanisms and Clinical Significance. Cancers (Basel) 2024; 16:2676. [PMID: 39123403 PMCID: PMC11311860 DOI: 10.3390/cancers16152676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
IFNγ, a pleiotropic cytokine produced not only by activated lymphocytes but also in response to cancer immunotherapies, has both antitumor and tumor-promoting functions. In ovarian cancer (OC) cells, the tumor-promoting functions of IFNγ are mediated by IFNγ-induced expression of Bcl3, PD-L1 and IL-8/CXCL8, which have long been known to have critical cellular functions as a proto-oncogene, an immune checkpoint ligand and a chemoattractant, respectively. However, overwhelming evidence has demonstrated that these three genes have tumor-promoting roles far beyond their originally identified functions. These tumor-promoting mechanisms include increased cancer cell proliferation, invasion, angiogenesis, metastasis, resistance to chemotherapy and immune escape. Recent studies have shown that IFNγ-induced Bcl3, PD-L1 and IL-8 expression is regulated by the same JAK1/STAT1 signaling pathway: IFNγ induces the expression of Bcl3, which then promotes the expression of PD-L1 and IL-8 in OC cells, resulting in their increased proliferation and migration. In this review, we summarize the recent findings on how IFNγ affects the tumor microenvironment and promotes tumor progression, with a special focus on ovarian cancer and on Bcl3, PD-L1 and IL-8/CXCL8 signaling. We also discuss promising novel combinatorial strategies in clinical trials targeting Bcl3, PD-L1 and IL-8 to increase the effectiveness of cancer immunotherapies.
Collapse
Affiliation(s)
| | | | | | - Ivana Vancurova
- Department of Biological Sciences, St. John’s University, New York, NY 11439, USA; (S.U.R.); (F.Z.S.); (A.V.)
| |
Collapse
|
24
|
Yu L, Zou R, He J, Qu C. Role of radiation in chimeric antigen receptor T-cell therapy for patients with relapsed/refractory non-Hodgkin lymphoma: Current studies and future prospects. Crit Rev Oncol Hematol 2024; 199:104390. [PMID: 38782146 DOI: 10.1016/j.critrevonc.2024.104390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment approach for patients with relapsed/refractory non-Hodgkin lymphoma (R/R NHL). However, the long-term prognosis has been discouraging. Moreover, the urgent resolution of two critical issues is necessary: minimize tumor burden before CAR-T infusion and control fatal toxicities post CAR-T therapy. By combining radiotherapy (RT), the safety and efficacy of CAR-T can be improved. RT can serve as bridging therapy, reducing the tumor burden before CAR-T infusion, thus enabling safe and successful CAR-T infusion, and as salvage therapy in cases of CAR-T therapy failure. This review aims to discuss the current evidence supporting the use of RT in CAR-T therapy for patients with R/R NHL. Although most studies have shown a positive role of RT in combined modality treatments for patients undergoing CAR-T therapy, the synergy gained from these remains uncertain. Furthermore, the optimal dose/fraction and radiation response require further investigation.
Collapse
Affiliation(s)
- Lingzi Yu
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| | - Rui Zou
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| | - Jiajie He
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| | - Changju Qu
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| |
Collapse
|
25
|
Lerouge L, Ruch A, Pierson J, Thomas N, Barberi-Heyob M. Non-targeted effects of radiation therapy for glioblastoma. Heliyon 2024; 10:e30813. [PMID: 38778925 PMCID: PMC11109805 DOI: 10.1016/j.heliyon.2024.e30813] [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: 02/07/2024] [Revised: 04/05/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Radiotherapy is recommended for the treatment of brain tumors such as glioblastoma (GBM) and brain metastases. Various curative and palliative scenarios suggest improved local-regional control. Although the underlying mechanisms are not yet clear, additional therapeutic effects have been described, including proximity and abscopal reactions at the treatment site. Clinical and preclinical data suggest that the immune system plays an essential role in regulating the non-targeted effects of radiotherapy for GBM. This article reviews current biological mechanisms for regulating the non-targeted effects caused by external and internal radiotherapy, and how they might be applied in a clinical context. Optimization of therapeutic regimens requires assessment of the complexity of the host immune system on the activity of immunosuppressive or immunostimulatory cells, such as glioma-associated macrophages and microglia. This article also discusses recent preclinical models adapted to post-radiotherapy responses. This narrative review explores and discusses the current status of immune responses both locally via the "bystander effect" and remotely via the "abscopal effect". Preclinical and clinical observations demonstrate that unirradiated cells, near or far from the irradiation site, can control the tumor response. Nevertheless, previous studies do not address the problem in its global context, and present gaps regarding the link between the role of the immune system in the control of non-targeted effects for different types of radiotherapy and different fractionation schemes applied to GBM. This narrative synthesis of the scientific literature should help to update and critique available preclinical and medical knowledge. Indirectly, it will help formulate new research projects based on the synthesis and interpretation of results from a non-systematic selection of published studies.
Collapse
Affiliation(s)
- Lucie Lerouge
- Department of Biology, Signals and Systems in Cancer and Neuroscience, CRAN, UMR7039, Université de Lorraine, CNRS, 54500 Vandœuvre-lès-Nancy, France
| | - Aurélie Ruch
- Department of Biology, Signals and Systems in Cancer and Neuroscience, CRAN, UMR7039, Université de Lorraine, CNRS, 54500 Vandœuvre-lès-Nancy, France
| | - Julien Pierson
- Department of Biology, Signals and Systems in Cancer and Neuroscience, CRAN, UMR7039, Université de Lorraine, CNRS, 54500 Vandœuvre-lès-Nancy, France
| | - Noémie Thomas
- Department of Biology, Signals and Systems in Cancer and Neuroscience, CRAN, UMR7039, Université de Lorraine, CNRS, 54500 Vandœuvre-lès-Nancy, France
| | - Muriel Barberi-Heyob
- Department of Biology, Signals and Systems in Cancer and Neuroscience, CRAN, UMR7039, Université de Lorraine, CNRS, 54500 Vandœuvre-lès-Nancy, France
| |
Collapse
|
26
|
Martín-Antonio B, Blanco B, González-Murillo Á, Hidalgo L, Minguillón J, Pérez-Chacón G. Newer generations of multi-target CAR and STAb-T immunotherapeutics: NEXT CART Consortium as a cooperative effort to overcome current limitations. Front Immunol 2024; 15:1386856. [PMID: 38779672 PMCID: PMC11109416 DOI: 10.3389/fimmu.2024.1386856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Adoptive T cellular immunotherapies have emerged as relevant approaches for treating cancer patients who have relapsed or become refractory (R/R) to traditional cancer treatments. Chimeric antigen receptor (CAR) T-cell therapy has improved survival in various hematological malignancies. However, significant limitations still impede the widespread adoption of these therapies in most cancers. To advance in this field, six research groups have created the "NEXT Generation CART MAD Consortium" (NEXT CART) in Madrid's Community, which aims to develop novel cell-based immunotherapies for R/R and poor prognosis cancers. At NEXT CART, various basic and translational research groups and hospitals in Madrid concur to share and synergize their basic expertise in immunotherapy, gene therapy, and immunological synapse, and clinical expertise in pediatric and adult oncology. NEXT CART goal is to develop new cell engineering approaches and treatments for R/R adult and pediatric neoplasms to evaluate in multicenter clinical trials. Here, we discuss the current limitations of T cell-based therapies and introduce our perspective on future developments. Advancement opportunities include developing allogeneic products, optimizing CAR signaling domains, combining cellular immunotherapies, multi-targeting strategies, and improving tumor-infiltrating lymphocytes (TILs)/T cell receptor (TCR) therapy. Furthermore, basic studies aim to identify novel tumor targets, tumor molecules in the tumor microenvironment that impact CAR efficacy, and strategies to enhance the efficiency of the immunological synapse between immune and tumor cells. Our perspective of current cellular immunotherapy underscores the potential of these treatments while acknowledging the existing hurdles that demand innovative solutions to develop their potential for cancer treatment fully.
Collapse
Affiliation(s)
- Beatriz Martín-Antonio
- Department of Experimental Hematology, Instituto de Investigación Sanitaria-Fundación Jiménez Diaz (IIS-FJD), Madrid, Spain
| | - Belén Blanco
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - África González-Murillo
- Department of Pediatric Hematology and Oncology, Advanced Therapies Unit, Fundación Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Laura Hidalgo
- Cellular Biotechnology Unit, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Jordi Minguillón
- La Paz Hospital Institute for Health Research (IdiPAZ), Hospital Universitario La Paz. Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Gema Pérez-Chacón
- Immunity, Immunopathology and Emergent Therapies Group. Instituto de Investigaciones Biomedicas Sols-Morreale. CSIC-UAM, Madrid, Spain
| |
Collapse
|
27
|
Hou Y, Yang K, Wang L, Wang J, Huang X, Piffko A, Luo SZ, Yu X, Rao E, Martinez C, Bugno J, Mack M, Vokes EE, Pitroda SP, Chmura SJ, Weichselbaum RR, Liang HL. Radiotherapy Enhances Metastasis Through Immune Suppression by Inducing PD-L1 and MDSC in Distal Sites. Clin Cancer Res 2024; 30:1945-1958. [PMID: 38427437 PMCID: PMC11062826 DOI: 10.1158/1078-0432.ccr-23-3206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/22/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
PURPOSE Radiotherapy (RT) is a widely employed anticancer treatment. Emerging evidence suggests that RT can elicit both tumor-inhibiting and tumor-promoting immune effects. The purpose of this study is to investigate immune suppressive factors of radiotherapy. EXPERIMENTAL DESIGN We used a heterologous two-tumor model in which adaptive concomitant immunity was eliminated. RESULTS Through analysis of PD-L1 expression and myeloid-derived suppressor cells (MDSC) frequencies using patient peripheral blood mononuclear cells and murine two-tumor and metastasis models, we report that local irradiation can induce a systemic increase in MDSC, as well as PD-L1 expression on dendritic cells and myeloid cells, and thereby increase the potential for metastatic dissemination in distal, nonirradiated tissue. In a mouse model using two distinct tumors, we found that PD-L1 induction by ionizing radiation was dependent on elevated chemokine CXCL10 signaling. Inhibiting PD-L1 or MDSC can potentially abrogate RT-induced metastasis and improve clinical outcomes for patients receiving RT. CONCLUSIONS Blockade of PD-L1/CXCL10 axis or MDSC infiltration during irradiation can enhance abscopal tumor control and reduce metastasis.
Collapse
Affiliation(s)
- Yuzhu Hou
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University; Xi’an, ShaanXi 710061, China
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Jiaai Wang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Xiaona Huang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Andras Piffko
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Sean Z. Luo
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: Biomedical Engineering program, Northwestern University; Evanston, IL 60201, USA
| | - Xinshuang Yu
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: Department of Oncology, First Affiliated Hospital of Shandong, First Medical University and Shandong Provincial Qianfoshan Hospital; Jinan, Shandong 250014, China
| | - Enyu Rao
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: Cancer Institute, Xuzhou Medical University; Xuzhou, Jiangsu 221004, China
| | - Carlos Martinez
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- Current address: University of Illinois at Chicago, Chicago, IL, 60607 USA
| | - Jason Bugno
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
- The Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, IL 600637, USA
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, 93042 Regensburg, Germany
| | - Everett E. Vokes
- Department of Medicine, University of Chicago, Chicago, IL, 60637 USA
| | - Sean P. Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Steven J. Chmura
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
| | - Ralph R. Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| | - Hua Laura Liang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, IL 60637 USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, IL 60637 USA
| |
Collapse
|
28
|
Song D, Ding Y. A new target of radiotherapy combined with immunotherapy: regulatory T cells. Front Immunol 2024; 14:1330099. [PMID: 38259489 PMCID: PMC10800811 DOI: 10.3389/fimmu.2023.1330099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Radiotherapy is one important treatment for malignant tumours. It is widely believed today that radiotherapy has not only been used as a local tumour treatment method, but also can induce systemic anti-tumour responses by influencing the tumour microenvironment, but its efficacy is limited by the tumour immunosuppression microenvironment. With the advancement of technology, immunotherapy has entered a golden age of rapid development, gradually occupying a place in clinical tumour treatment. Regulatory T cells (Tregs) widely distributing in the tumour microenvironment play an important role in mediating tumour development. This article analyzes immunotherapy, the interaction between Tregs, tumours and radiotherapy. It briefly introduces immunotherapies targeting Tregs, aiming to provide new strategies for radiotherapy combined with Immunotherapy.
Collapse
Affiliation(s)
| | - Yun Ding
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
29
|
He J, Yan Y, Zhang J, Wei Z, Li H, Xing L. Synergistic treatment strategy: combining CAR-NK cell therapy and radiotherapy to combat solid tumors. Front Immunol 2023; 14:1298683. [PMID: 38162672 PMCID: PMC10755030 DOI: 10.3389/fimmu.2023.1298683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Immunotherapy, notably chimeric antigen receptor (CAR) modified natural killer (NK) cell therapy, has shown exciting promise in the treatment of hematologic malignancies due to its unique advantages including fewer side effects, diverse activation mechanisms, and wide availability. However, CAR-NK cell therapies have demonstrated limited efficacy against solid tumors, primarily due to challenges posed by the solid tumor microenvironment. In contrast, radiotherapy, a well-established treatment modality, has been proven to modulate the tumor microenvironment and facilitate immune cell infiltration. With these observations, we hypothesize that a novel therapeutic strategy integrating CAR-NK cell therapy with radiotherapy could enhance the ability to treat solid tumors. This hypothesis aims to address the obstacles CAR-NK cell therapies face within the solid tumor microenvironment and explore the potential efficacy of their combination with radiotherapy. By capitalizing on the synergistic advantages of CAR-NK cell therapy and radiotherapy, we posit that this could lead to improved prognoses for patients with solid tumors.
Collapse
Affiliation(s)
- Jie He
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yushan Yan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jun Zhang
- Asclepius (Soochow) Technology Company Group, Suzhou, Jiangsu, China
| | - Zhiming Wei
- Asclepius (Soochow) Technology Company Group, Suzhou, Jiangsu, China
| | - Huashun Li
- Asclepius (Soochow) Technology Company Group, Suzhou, Jiangsu, China
| | - Ligang Xing
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
30
|
Lau APY, Khavkine Binstock SS, Thu KL. CD47: The Next Frontier in Immune Checkpoint Blockade for Non-Small Cell Lung Cancer. Cancers (Basel) 2023; 15:5229. [PMID: 37958404 PMCID: PMC10649163 DOI: 10.3390/cancers15215229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/18/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
The success of PD-1/PD-L1-targeted therapy in lung cancer has resulted in great enthusiasm for additional immunotherapies in development to elicit similar survival benefits, particularly in patients who do not respond to or are ineligible for PD-1 blockade. CD47 is an immunosuppressive molecule that binds SIRPα on antigen-presenting cells to regulate an innate immune checkpoint that blocks phagocytosis and subsequent activation of adaptive tumor immunity. In lung cancer, CD47 expression is associated with poor survival and tumors with EGFR mutations, which do not typically respond to PD-1 blockade. Given its prognostic relevance, its role in facilitating immune escape, and the number of agents currently in clinical development, CD47 blockade represents a promising next-generation immunotherapy for lung cancer. In this review, we briefly summarize how tumors disrupt the cancer immunity cycle to facilitate immune evasion and their exploitation of immune checkpoints like the CD47-SIRPα axis. We also discuss approved immune checkpoint inhibitors and strategies for targeting CD47 that are currently being investigated. Finally, we review the literature supporting CD47 as a promising immunotherapeutic target in lung cancer and offer our perspective on key obstacles that must be overcome to establish CD47 blockade as the next standard of care for lung cancer therapy.
Collapse
Affiliation(s)
- Asa P. Y. Lau
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Sharon S. Khavkine Binstock
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Kelsie L. Thu
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
31
|
Tamai Y, Fujiwara N, Tanaka T, Mizuno S, Nakagawa H. Combination Therapy of Immune Checkpoint Inhibitors with Locoregional Therapy for Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:5072. [PMID: 37894439 PMCID: PMC10605879 DOI: 10.3390/cancers15205072] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is estimated to be the fourth leading cause of cancer-related deaths globally, and its overall prognosis is dismal because most cases are diagnosed at a late stage and are unamenable to curative treatment. The emergence of immune checkpoint inhibitors (ICIs) has dramatically improved the therapeutic efficacy for advanced hepatocellular carcinoma; however, their response rates remain unsatisfactory, partly because >50% of HCC exhibit an ICI-nonresponsive tumor microenvironment characterized by a paucity of cytotoxic T cells (immune-cold), as well as difficulty in their infiltration into tumor sites (immune excluded). To overcome this limitation, combination therapies with locoregional therapies, including ablation, transarterial embolization, and radiotherapy, which are usually used for early stage HCCs, have been actively explored to enhance ICI efficacy by promoting the release of tumor-associated antigens and cytokines, and eventually accelerating the so-called cancer-immunity cycle. Various combination therapies have been investigated in early- to late-phase clinical trials, and some have shown promising results. This comprehensive article provides an overview of the immune landscape for HCC to understand ICI efficacy and its limitations and, subsequently, reviews the status of combinatorial therapies of ICIs with locoregional therapy for HCC.
Collapse
Affiliation(s)
- Yasuyuki Tamai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan; (Y.T.); (T.T.); (H.N.)
| | - Naoto Fujiwara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan; (Y.T.); (T.T.); (H.N.)
| | - Takamitsu Tanaka
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan; (Y.T.); (T.T.); (H.N.)
| | - Shugo Mizuno
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan;
| | - Hayato Nakagawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan; (Y.T.); (T.T.); (H.N.)
| |
Collapse
|
32
|
Antelo G, Comas S, Casas F, Valduvieco I, Barreto T, Laplana M, Mases J, Oses G, Mollà M. Clinical outcomes and timing on the combination of focal radiation therapy and immunotherapy for the treatment of brain metastases. Front Immunol 2023; 14:1236398. [PMID: 37915576 PMCID: PMC10616465 DOI: 10.3389/fimmu.2023.1236398] [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: 06/07/2023] [Accepted: 10/05/2023] [Indexed: 11/03/2023] Open
Abstract
Introduction Radiotherapy is one of the standard treatments for brain metastases (BM). Over the past years, the introduction of immunotherapy as routine treatment for solid tumors has forced investigators to review and evaluate how it would interact with radiation. Radiation and Immunotherapy have shown a synergic effect activating the host's immune system and enhancing treatment response. The combinatory effect on BM is currently under investigation. Methods Data published on Pubmed to determine toxicity, survival, treatment characteristics and timing on the combination of radiotherapy and immunotherapy for the treatment of BM has been reviewed. Results Mostly retrospective reviews report an improvement of intracranial progression free survival (iPFS) when combining radioimmunotherapy for BM patients. Two systematic reviews and meta-analysis and one phase II prospective trial also report a benefit on iPFS without an increase of toxicity. Among the published literature, the definition of concurrency is heterogeneous, being one month or even narrowed intervals correlated to better clinical outcomes. Toxicity due to concurrent radioimmunotherapy, specifically symptomatic radionecrosis, is also directly analyzed and reported to be low, similar to the toxicity rates secondary to stereotactic radiosurgery alone. Conclusion Radiation combined with immunotherapy has shown in predominantly retrospective reviews a synergic effect on the treatment of BM. The concurrent combination of radioimmunotherapy is a feasible therapeutic strategy and seems to improve clinical outcomes, especially iPFS, when delivered within <30 days. Larger prospective and randomized studies are needed to establish reliable outcomes, best delivery strategies and toxicity profile.
Collapse
Affiliation(s)
- Gabriela Antelo
- Radiation Oncology Department, Hospital Clinic Barcelona, Barcelona, Spain
| | - Silvia Comas
- Radiation Oncology Department, Institut Catalá d'Oncologia (ICO), Badalona, Badalona, Spain
| | - Francesc Casas
- Radiation Oncology Department, Hospital Clinic Barcelona, Barcelona, Spain
| | - Izaskun Valduvieco
- Radiation Oncology Department, Hospital Clinic Barcelona, Barcelona, Spain
| | - Tanny Barreto
- Radiation Oncology Department, Hospital Clinic Barcelona, Barcelona, Spain
| | - María Laplana
- Radiation Oncology Department, Hospital Clinic Barcelona, Barcelona, Spain
| | - Joel Mases
- Radiation Oncology Department, Hospital Clinic Barcelona, Barcelona, Spain
| | - Gabriela Oses
- Radiation Oncology Department, Hospital Clinic Barcelona, Barcelona, Spain
| | - Meritxell Mollà
- Radiation Oncology Department, Hospital Clinic Barcelona, Barcelona, Spain
| |
Collapse
|
33
|
Ali Mohammad S, Hak A, Pogu SV, Rengan AK. Radiotherapy, photodynamic therapy, and cryoablation-induced abscopal effect: Challenges and future prospects. CANCER INNOVATION 2023; 2:323-345. [PMID: 38090387 PMCID: PMC10686191 DOI: 10.1002/cai2.53] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/10/2022] [Accepted: 12/29/2022] [Indexed: 10/15/2024]
Abstract
Local therapy modalities such as radiation therapy, photodynamic therapy, photothermal therapy, and cryoablation have been used to treat localized tumors for decades. The discovery of the abscopal effect causes a paradigm shift where local therapy also causes systemic effects and leads to the remission of nonirradiated tumors. The abscopal effect of radiation therapy, alone or in combination with other treatments, has been extensively studied over the last six decades. However, the results are unsatisfactory in producing robust, reproducible, and long-lasting systemic effects. Although immunotherapy and radiation therapy are promising in producing the abscopal effect, the abscopal effect's mechanism is still unclear, owing to various factors such as irradiation type and dose and cancer type. This article reviews the research progress, clinical and preclinical evidence of the abscopal effect by various local therapies alone and in combination with chemotherapy and immunotherapy, case reports, and the current challenges in producing the abscopal effect by various local therapies, focusing on radiotherapy, photodynamic therapy, cryoablation, and the prospects for obtaining a robust, reproducible, and long-lasting abscopal effect.
Collapse
Affiliation(s)
| | - Arshadul Hak
- Indian Institute of Technology HyderabadKandi, SangareddyTelanganaIndia
| | - Sunil V. Pogu
- Indian Institute of Technology HyderabadKandi, SangareddyTelanganaIndia
| | - Aravind K. Rengan
- Indian Institute of Technology HyderabadKandi, SangareddyTelanganaIndia
| |
Collapse
|
34
|
Burgaleta AM, Burguete AB, Gutiérrez LR, Nuín EB, Felipe GA, de la Vega FA. Local treatment in oligometastasis from breast cancer: an overview. Clin Transl Oncol 2023; 25:2861-2867. [PMID: 37106239 DOI: 10.1007/s12094-023-03170-0] [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: 01/17/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023]
Abstract
Oligometastasic breast cancer (OMBC) consists of breast cancer patient with a limited number of systemic metastases (≤ 5), all of them candidates for local ablative treatment with the intention of achieving long-term control of the metastasis and, eventually, an increase in survival The first consensus for the management of patients with oligometastatic breast cancer (OMBC) was published in 2007, establishing that a more aggressive multidisciplinary strategy is recommended in order to increase the survival while maintaining a good quality of life. The current scientific evidence is based on observational studies, mainly retrospective, systematic reviews and meta-analyses, and only a randomized nonexclusive study of oligometastatic (OM) published. All trials with Stereotactic Body Radiation Therapy (SBRT) in OM cancer have shown excellent tolerance and good local control, although first trials on Lung SBRT did not prove so excellent tolerance and had some deaths due to bronchus irradiation and secondary hemoptysis. There are multiple ongoing studies researching the benefit of SBRT in oligometastatic breast cancer. Despite the lack of impact on survival seen in the NRG BR-002 Trial, SBRT probably allows the delay of the systemic treatment until progression, and so, improves the quality of life of patients. We have to wait for the results of the ongoing and future studies for clarification of the role of local treatment in OMBC.
Collapse
Affiliation(s)
- Ana Manterola Burgaleta
- Multidisciplinary Breast Cancer Unit, Radiation Oncology Service, University Hospital of Navarre, Pamplona, Spain
- Service of Radiotherapy Oncology, University Hospital of Navarre, Pamplona, Spain
| | | | | | - Erkuden Burillo Nuín
- Service of Radiotherapy Oncology, University Hospital of Navarre, Pamplona, Spain
| | - Gemma Asín Felipe
- Multidisciplinary Breast Cancer Unit, Radiation Oncology Service, University Hospital of Navarre, Pamplona, Spain
- Service of Radiotherapy Oncology, University Hospital of Navarre, Pamplona, Spain
| | - Fernando Arias de la Vega
- Service of Radiotherapy Oncology, University Hospital of Navarre, Pamplona, Spain.
- "Clinical Research Group in Radiation Oncology", Health Research Institute of Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
35
|
Fan X, Li J, Huang B, Lu H, Lu C, Pan M, Wang X, Zhang H, You Y, Wang X, Wang Q, Zhang J. Noninvasive radiomics model reveals macrophage infiltration in glioma. Cancer Lett 2023; 573:216380. [PMID: 37660885 DOI: 10.1016/j.canlet.2023.216380] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/20/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023]
Abstract
Preoperative MRI is an essential diagnostic and therapeutic reference for gliomas. This study aims to evaluate the prognostic aspect of a radiomics biomarker for glioma and further investigate its relationship with tumor microenvironment and macrophage infiltration. We covered preoperative MRI of 664 glioma patients from three independent datasets: Jiangsu Province Hospital (JSPH, n = 338), The Cancer Genome Atlas dataset (TCGA, n = 252), and Repository of Molecular Brain Neoplasia Data (REMBRANDT, n = 74). Incorporating a multistep post-processing workflow, 20 radiomics features (Rads) were selected and a radiomics survival biomarker (RadSurv) was developed, proving highly efficient in risk stratification of gliomas (cut-off = 1.06), as well as lower-grade gliomas (cut-off = 0.64) and glioblastomas (cut-off = 1.80) through three fixed cut-off values. Through immune infiltration analysis, we found a positive correlation between RadSurv and macrophage infiltration (RMΦ = 0.297, p < 0.001; RM2Φ = 0.241, p < 0.001), further confirmed by immunohistochemical-staining (glioblastomas, n = 32) and single-cell sequencing (multifocal glioblastomas, n = 2). In conclusion, RadSurv acts as a strong prognostic biomarker for gliomas, exhibiting a non-negligible positive correlation with macrophage infiltration, especially with M2 macrophage, which strongly suggests the promise of radiomics-based models as a preoperative alternative to conventional genomics for predicting tumor macrophage infiltration and provides clinical guidance for immunotherapy.
Collapse
Affiliation(s)
- Xiao Fan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jintan Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bin Huang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Hongyu Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chenfei Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Minhong Pan
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiefeng Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongjian Zhang
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Xiuxing Wang
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
36
|
Zhao Y, He GS, Li G. Triplet regimen as a novel modality for advanced unresectable hepatocellular carcinoma: A case report and review of literature. World J Clin Cases 2023; 11:6558-6564. [PMID: 37900216 PMCID: PMC10601001 DOI: 10.12998/wjcc.v11.i27.6558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/22/2023] [Accepted: 08/31/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Portal vein tumor thrombus (PVTT) is a common complication, accounting for 44%-62.2% of Hepatocellular carcinoma (HCC), and often indicates the poor prognosis. There is no global consensus for the treatment of unresectable HCC with PVTT. In the present case, we reported a novel strategy of radiotherapy-antiangiogenesis-immune checkpoint blockade combination, which showed better response and prolonged survival. CASE SUMMARY A 51-year-old male diagnosed with HCC (Child-Pugh class A), chronic hepatitis B virus infection and Cheng's type III PVTT, was given radiotherapy to part of the lesion plus targeted therapy as the first-line therapy, and achieved partial remission. After radiotherapy, lenvatinib plus pembrolizumab was used as maintenance therapy, and complete remission was achieved. The patient remains alive 46 months after the diagnosis of the HCC with PVTT. CONCLUSION This case of unresectable HCC patient with PVTT treated by radiation-lenvatinib-pembrolizumab combination therapy shows apparent clinical efficacy, which demonstrates that local radiotherapy plus antiangiogenesis and immune checkpoint blockad could also benefit patients with advanced HCC.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Radiation Oncology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Guo-Sheng He
- Department of Radiation Oncology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Gong Li
- Department of Radiation Oncology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| |
Collapse
|
37
|
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
|
38
|
Su JX, Li SJ, Zhou XF, Zhang ZJ, Yan Y, Liu SL, Qi Q. Chemotherapy-induced metastasis: molecular mechanisms and clinical therapies. Acta Pharmacol Sin 2023; 44:1725-1736. [PMID: 37169853 PMCID: PMC10462662 DOI: 10.1038/s41401-023-01093-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023]
Abstract
Chemotherapy, the most widely accepted treatment for malignant tumors, is dependent on cell death induced by various drugs including antimetabolites, alkylating agents, mitotic spindle inhibitors, antitumor antibiotics, and hormonal anticancer drugs. In addition to causing side effects due to non-selective cytotoxicity, chemotherapeutic drugs can initiate and promote metastasis, which greatly reduces their clinical efficacy. The knowledge of how they induce metastasis is essential for developing strategies that improve the outcomes of chemotherapy. Herein, we summarize the recent findings on chemotherapy-induced metastasis and discuss the underlying mechanisms including tumor-initiating cell expansion, the epithelial-mesenchymal transition, extracellular vesicle involvement, and tumor microenvironment alterations. In addition, the use of combination treatments to overcome chemotherapy-induced metastasis is also elaborated.
Collapse
Affiliation(s)
- Jin-Xuan Su
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Si-Jia Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiao-Feng Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhi-Jing Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yu Yan
- Functional Experimental Teaching Center, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Song-Lin Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Qi Qi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
39
|
Liu X, Zhao Z, Dai W, Liao K, Sun Q, Chen D, Pan X, Feng L, Ding Y, Wei S. The Development of Immunotherapy for the Treatment of Recurrent Glioblastoma. Cancers (Basel) 2023; 15:4308. [PMID: 37686584 PMCID: PMC10486426 DOI: 10.3390/cancers15174308] [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: 07/19/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 09/10/2023] Open
Abstract
Recurrent glioblastoma (rGBM) is a highly aggressive form of brain cancer that poses a significant challenge for treatment in neuro-oncology, and the survival status of patients after relapse usually means rapid deterioration, thus becoming the leading cause of death among patients. In recent years, immunotherapy has emerged as a promising strategy for the treatment of recurrent glioblastoma by stimulating the body's immune system to recognize and attack cancer cells, which could be used in combination with other treatments such as surgery, radiation, and chemotherapy to improve outcomes for patients with recurrent glioblastoma. This therapy combines several key methods such as the use of monoclonal antibodies, chimeric antigen receptor T cell (CAR-T) therapy, checkpoint inhibitors, oncolytic viral therapy cancer vaccines, and combination strategies. In this review, we mainly document the latest immunotherapies for the treatment of glioblastoma and especially focus on rGBM.
Collapse
Affiliation(s)
- Xudong Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (X.L.); (Y.D.)
| | - Zihui Zhao
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China;
| | - Wufei Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering Research, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China;
| | - Kuo Liao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China;
| | - Qi Sun
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Q.S.); (L.F.)
| | - Dongjiang Chen
- Division of Neuro-Oncology, USC Keck Brain Tumor Center, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA;
| | - Xingxin Pan
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Lishuang Feng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Q.S.); (L.F.)
| | - Ying Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (X.L.); (Y.D.)
| | - Shiyou Wei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
40
|
Zhang L, Tang T, Liu L, Li C, Li Y, Geng C. Effect of tumor-infiltrating lymphocytes depending on the presence of postmastectomy radiotherapy on the prognosis in pT1-2N1M0 breast cancer. Front Oncol 2023; 13:1175965. [PMID: 37601690 PMCID: PMC10436467 DOI: 10.3389/fonc.2023.1175965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
Background Currently, it remains unclear regarding the association between tumor-infiltrating lymphocytes (TILs) and the efficacy of postoperative radiotherapy in primary tumors. Here we attempted to investigate the effect of TILs depending on the presence of postmastectomy radiotherapy (PMRT) on the prognosis in pT1-2N1M0 breast cancer. Methods The clinical data of pT1-2N1M0 breast cancer patients undergoing mastectomy and axillary lymph node dissection were retrospectively analyzed. The effect of TILs on the prognosis was assessed based on the infiltration degree (low: TILs ≤10%, high: TILs >10%), and then the prognosis of patients with low and high infiltration of TILs was analyzed based on presence or absence of PMRT. Results Totally 213 patients were eligible for the study, including 162 cases of low infiltration and 51 of high infiltration. High-infiltration patients tended to be ER/PR-negative, HER2-positive, and have high histological grade. The infiltration in triple-negative and HER2-positive subtypes was higher compared with Luminal A subtype. Regarding local-regional recurrence-free survival, recurrence-free survival, and overall survival (OS) rates, the differences were all inapparent whether in high- and low-infiltration patients or in high-infiltration patients with/without PMRT. Compared with those without PMRT, low-infiltration patients with PMRT showed a significantly increased OS rate (92.8% vs. 80.0%, p=0.023). Multivariate analysis further confirmed PMRT as an independent predicator of OS in low-infiltration patients (HR: 0.228, 95%CI: 0.081-0.644, p=0.005). Conclusion High infiltration of TILs in pT1-2N1M0 breast cancer may be associated with clinicopathological factors. Low-infiltration patients, but not high-infiltration patients, may derive survival benefits from PMRT.
Collapse
Affiliation(s)
| | | | | | | | | | - Cuizhi Geng
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
41
|
Li T, Qian X, Liu J, Xue F, Luo J, Yao G, Yan J, Liu X, Xiao B, Li J. Radiotherapy plus immune checkpoint inhibitor in prostate cancer. Front Oncol 2023; 13:1210673. [PMID: 37546397 PMCID: PMC10403272 DOI: 10.3389/fonc.2023.1210673] [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: 04/23/2023] [Accepted: 07/04/2023] [Indexed: 08/08/2023] Open
Abstract
The immune checkpoint inhibitor (ICI) is a promising strategy for treating cancer. However, the efficiency of ICI monotherapy is limited, which could be mainly attributed to the tumor microenvironment of the "cold" tumor. Prostate cancer, a type of "cold" cancer, is the most common cancer affecting men's health. Radiotherapy is regarded as one of the most effective prostate cancer treatments. In the era of immune therapy, the enhanced antigen presentation and immune cell infiltration caused by radiotherapy might boost the therapeutic efficacy of ICI. Here, the rationale of radiotherapy combined with ICI was reviewed. Also, the scheme of radiotherapy combined with immune checkpoint blockades was suggested as a potential option to improve the outcome of patients with prostate cancer.
Collapse
Affiliation(s)
- Tianjie Li
- School of Clinical Medicine, Tsinghua University, Beijing, China
- Department of Urology, Beijing Tsinghua Changung Hospital, Beijing, China
| | - Xinye Qian
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jinyang Liu
- School of Medical, Tsinghua University, Beijing, China
| | - Feng Xue
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jing Luo
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Guanqun Yao
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jun Yan
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Xiaodong Liu
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bo Xiao
- Department of Urology, Beijing Tsinghua Changung Hospital, Beijing, China
| | - Jianxing Li
- Department of Urology, Beijing Tsinghua Changung Hospital, Beijing, China
| |
Collapse
|
42
|
Zhou Z, Guan B, Xia H, Zheng R, Xu B. Particle radiotherapy in the era of radioimmunotherapy. Cancer Lett 2023:216268. [PMID: 37331583 DOI: 10.1016/j.canlet.2023.216268] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/24/2023] [Accepted: 06/11/2023] [Indexed: 06/20/2023]
Abstract
Radiotherapy (RT) is one of the key modalities for cancer treatment, and more than 70% of tumor patients will receive RT during the course of their disease. Particle radiotherapy, such as proton radiotherapy, carbon-ion radiotherapy (CIRT) and boron neutron capture therapy (BNCT), is currently available for the treatment of patients Immunotherapy combined with photon RT has been successfully used in the clinic. The effect of immunotherapy combined with particle RT is an area of interest. However, the molecular mechanisms underlying the effects of combined immunotherapy and particle RT remain largely unknown. In this review, we summarize the properties of different types of particle RT and the mechanisms underlying their radiobiological effects. Additionally, we compared the main molecular players in photon RT and particle RT and the mechanisms involved the RT-mediated immune response.
Collapse
Affiliation(s)
- Zihan Zhou
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Bingjie Guan
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Huang Xia
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Rong Zheng
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China; Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China.
| | - Benhua Xu
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China; Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China.
| |
Collapse
|
43
|
Dabaja B, Spiotto M. Radiation for hematologic malignancies: from cell killing to immune cell priming. Front Oncol 2023; 13:1205836. [PMID: 37384297 PMCID: PMC10299853 DOI: 10.3389/fonc.2023.1205836] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/30/2023] [Indexed: 06/30/2023] Open
Abstract
Over the past half-century, the role of radiotherapy has been revolutionized, in part, by a shift from intent to directly kill cancer cells to the goal of priming anti-tumor immune responses that attack both irradiated and non-irradiated tumors. Stimulation of anti-tumor immunity depends on the interplay between radiation, the tumor microenvironment, and the host immune system, which is a burgeoning concept in cancer immunology. While the interplay of radiotherapy and the immune system has been primarily studied in solid tumors, we are beginning to understand this interplay in hematological malignancies. The intent of this review is to lead readers through some of the important recent advances in immunotherapy and adoptive cell therapy, highlighting the best available evidence in support of incorporating radiation therapy and immunotherapy into the treatment of hematological malignancies. Evidence is presented regarding how radiation therapy 'converses' with the immune system to stimulate and enhance anti-tumor immune responses. This pro-immunogenic role of radiotherapy can be combined with monoclonal antibodies, cytokines and/or other immunostimulatory agents to enhance the regression of hematological malignancies. Furthermore, we will discuss how radiotherapy facilitates the effectiveness of cellular immunotherapies by acting as a "bridge" that facilitated CAR T cell engraftment and activity. These initial studies suggest radiotherapy may help catalyze a shift from using chemotherapy-intensive treatment to treatment that is "chemo-free" by combining with immunotherapy to target both the radiated and non-irradiated disease sites. This "journey" has opened the door for novel uses of radiotherapy in hematological malignancies due to its ability to prime anti-tumor immune responses which can augment immunotherapy and adoptive cell-based therapy.
Collapse
|
44
|
Chi A, Nguyen NP. Mechanistic rationales for combining immunotherapy with radiotherapy. Front Immunol 2023; 14:1125905. [PMID: 37377970 PMCID: PMC10291094 DOI: 10.3389/fimmu.2023.1125905] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Immunotherapy consisted mainly of immune checkpoint inhibitors (ICIs) has led to significantly improved antitumor response. However, such response has been observed only in tumors possessing an overall responsive tumor immune micro-environment (TIME), in which the presence of functional tumor-infiltrating lymphocytes (TILs) is critical. Various mechanisms of immune escape from immunosurveillance exist, leading to different TIME phenotypes in correlation with primary or acquired resistance to ICIs. Radiotherapy has been shown to induce antitumor immunity not only in the irradiated primary tumor, but also at unirradiated distant sites of metastases. Such antitumor immunity is mainly elicited by radiation's stimulatory effects on antigenicity and adjuvanticity. Furthermore, it may be significantly augmented when irradiation is combined with immunotherapy, such as ICIs. Therefore, radiotherapy represents one potential therapeutic strategy to restore anti-tumor immunity in tumors presenting with an unresponsive TIME. In this review, the generation of anti-tumor immunity, its impairment, radiation's immunogenic properties, and the antitumor effects of combining radiation with immunotherapy will be comprehensively discussed.
Collapse
Affiliation(s)
- Alexander Chi
- Department of Radiation Oncology, Capital Medical University Xuanwu Hospital, Beijing, China
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Nam Phong Nguyen
- Department of Radiation Oncology, Howard University, Washington, DC, United States
| |
Collapse
|
45
|
Rohila D, Park IH, Pham TV, Jones R, Tapia E, Liu KX, Tamayo P, Yu A, Sharabi AB, Joshi S. Targeting macrophage Syk enhances responses to immune checkpoint blockade and radiotherapy in high-risk neuroblastoma. Front Immunol 2023; 14:1148317. [PMID: 37350973 PMCID: PMC10283071 DOI: 10.3389/fimmu.2023.1148317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/02/2023] [Indexed: 06/24/2023] Open
Abstract
Background Neuroblastoma (NB) is considered an immunologically cold tumor and is usually less responsive to immune checkpoint blockade (ICB). Tumor-associated macrophages (TAMs) are highly infiltrated in NB tumors and promote immune escape and resistance to ICB. Hence therapeutic strategies targeting immunosuppressive TAMs can improve responses to ICB in NB. We recently discovered that spleen tyrosine kinase (Syk) reprograms TAMs toward an immunostimulatory phenotype and enhances T-cell responses in the lung adenocarcinoma model. Here we investigated if Syk is an immune-oncology target in NB and tested whether a novel immunotherapeutic approach utilizing Syk inhibitor together with radiation and ICB could provide a durable anti-tumor immune response in an MYCN amplified murine model of NB. Methods Myeloid Syk KO mice and syngeneic MYCN-amplified cell lines were used to elucidate the effect of myeloid Syk on the NB tumor microenvironment (TME). In addition, the effect of Syk inhibitor, R788, on anti-tumor immunity alone or in combination with anti-PDL1 mAb and radiation was also determined in murine NB models. The underlying mechanism of action of this novel therapeutic combination was also investigated. Results Herein, we report that Syk is a marker of NB-associated macrophages and plays a crucial role in promoting immunosuppression in the NB TME. We found that the blockade of Syk in NB-bearing mice markedly impairs tumor growth. This effect is facilitated by macrophages that become immunogenic in the absence of Syk, skewing the suppressive TME towards immunostimulation and activating anti-tumor immune responses. Moreover, combining FDA-approved Syk inhibitor, R788 (fostamatinib) along with anti-PDL1 mAb provides a synergistic effect leading to complete tumor regression and durable anti-tumor immunity in mice bearing small tumors (50 mm3) but not larger tumors (250 mm3). However, combining radiation to R788 and anti-PDL1 mAb prolongs the survival of mice bearing large NB9464 tumors. Conclusion Collectively, our findings demonstrate the central role of macrophage Syk in NB progression and demonstrate that Syk blockade can "reeducate" TAMs towards immunostimulatory phenotype, leading to enhanced T cell responses. These findings further support the clinical evaluation of fostamatinib alone or with radiation and ICB, as a novel therapeutic intervention in neuroblastoma.
Collapse
Affiliation(s)
- Deepak Rohila
- Division of Pediatric Hematology-Oncology, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - In Hwan Park
- Division of Pediatric Hematology-Oncology, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Timothy V. Pham
- Office of Cancer Genomics, University of California San Diego, San Diego, CA, United States
| | - Riley Jones
- Department of Radiation Medicine and Applied Sciences, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Elisabette Tapia
- Division of Pediatric Hematology-Oncology, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Kevin X. Liu
- Department of Radiation Oncology, Dana Farber Cancer Institute, Boston, MA, United States
| | - Pablo Tamayo
- Office of Cancer Genomics, University of California San Diego, San Diego, CA, United States
| | - Alice Yu
- Division of Pediatric Hematology-Oncology, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Andrew B. Sharabi
- Department of Radiation Medicine and Applied Sciences, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Shweta Joshi
- Division of Pediatric Hematology-Oncology, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
46
|
Eckstein J, Gogineni E, Sidiqi B, Lisser N, Parashar B. Effect of Immunotherapy and Stereotactic Body Radiation Therapy Sequencing on Local Control and Survival in Patients With Spine Metastases. Adv Radiat Oncol 2023; 8:101179. [PMID: 36896213 PMCID: PMC9991541 DOI: 10.1016/j.adro.2023.101179] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 01/09/2023] [Indexed: 01/17/2023] Open
Abstract
Purpose Stereotactic body radiation therapy (SBRT) is commonly used to treat spinal metastases in combination with immunotherapy (IT). The optimal sequencing of these modalities is unclear. This study aimed to investigate whether sequencing of IT and SBRT was associated with differences in local control (LC), overall survival (OS), and toxicity when treating spine metastases. Methods and Materials All patients at our institution who received spine SBRT from 2010 to 2019 with systemic therapy data available were reviewed retrospectively. The primary endpoint was LC. Secondary endpoints were toxicity (fracture and radiation myelitis) and OS. Kaplan-Meier analysis was used to determine whether IT sequencing (before versus after SBRT) and use of IT were associated with LC or OS. Results A total of 191 lesions in 128 patients met inclusion criteria with 50 (26%) lesions in 33 (26%) patients who received IT. Fourteen (11%) patients with 24 (13%) lesions received the first IT dose before SBRT, whereas 19 (15%) patients with 26 (14%) lesions received the first dose after SBRT. LC did not differ between lesions treated with IT before SBRT versus after SBRT (1 year 73% versus 81%, log rank = 0.275, P = .600). Fracture risk was not associated with IT timing (χ2 = 0.137, P = .934) or receipt of IT (χ2 = 0.508, P = .476), and no radiation myelitis events occurred. Median OS was 31.8 versus 6.6 months for the IT after SBRT versus IT before SBRT cohorts, respectively (log rank = 13.193, P < .001). On Cox univariate analysis and multivariate analysis, receipt of IT before SBRT and Karnofsky performance status <80 were associated with worse OS. IT treatment versus none was not associated with any difference in LC (log rank = 1.063, P = .303) or OS (log rank = 1.736, P = .188). Conclusions Sequencing of IT and SBRT was not associated with any difference in LC or toxicity, but delivering IT after SBRT versus before SBRT was associated with improved OS.
Collapse
Affiliation(s)
- Jacob Eckstein
- Department of Radiation Medicine, Zucker School of Medicine, Hofstra, Northwell Health, New York, New York
| | - Emile Gogineni
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Baho Sidiqi
- Department of Radiation Medicine, Zucker School of Medicine, Hofstra, Northwell Health, New York, New York
| | - Noah Lisser
- Department of Radiation Medicine, Zucker School of Medicine, Hofstra, Northwell Health, New York, New York
| | - Bhupesh Parashar
- Department of Radiation Medicine, Zucker School of Medicine, Hofstra, Northwell Health, New York, New York
| |
Collapse
|
47
|
Hannon G, Lesch ML, Gerber SA. Harnessing the Immunological Effects of Radiation to Improve Immunotherapies in Cancer. Int J Mol Sci 2023; 24:7359. [PMID: 37108522 PMCID: PMC10138513 DOI: 10.3390/ijms24087359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Ionizing radiation (IR) is used to treat 50% of cancers. While the cytotoxic effects related to DNA damage with IR have been known since the early 20th century, the role of the immune system in the treatment response is still yet to be fully determined. IR can induce immunogenic cell death (ICD), which activates innate and adaptive immunity against the cancer. It has also been widely reported that an intact immune system is essential to IR efficacy. However, this response is typically transient, and wound healing processes also become upregulated, dampening early immunological efforts to overcome the disease. This immune suppression involves many complex cellular and molecular mechanisms that ultimately result in the generation of radioresistance in many cases. Understanding the mechanisms behind these responses is challenging as the effects are extensive and often occur simultaneously within the tumor. Here, we describe the effects of IR on the immune landscape of tumors. ICD, along with myeloid and lymphoid responses to IR, are discussed, with the hope of shedding light on the complex immune stimulatory and immunosuppressive responses involved with this cornerstone cancer treatment. Leveraging these immunological effects can provide a platform for improving immunotherapy efficacy in the future.
Collapse
Affiliation(s)
- Gary Hannon
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Maggie L. Lesch
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Scott A. Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
48
|
Liu S, Liao Y, Chen Y, Yang H, Hu Y, Chen Z, Fu S, Wu J. Effect of triple therapy with low-dose total body irradiation and hypo-fractionated radiation plus anti-programmed cell death protein 1 blockade on abscopal antitumor immune responses in breast cancer. Int Immunopharmacol 2023; 117:110026. [PMID: 36934673 DOI: 10.1016/j.intimp.2023.110026] [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: 12/27/2022] [Revised: 03/05/2023] [Accepted: 03/09/2023] [Indexed: 03/19/2023]
Abstract
Immunostimulatory effects of radiotherapy can be synergistically augmented with immune checkpoint blockade to act both on irradiated tumor lesions and distant, non-irradiated tumor sites. Our hypothesis was that low-dose total body irradiation (L-TBI) combined with hypo-fractionated radiotherapy (H-RT) and anti-programmed cell death protein 1 (aPD-1) checkpoint blockade would enhance the systemic immune response. We tested the efficacy of this triple therapy (L-TBI + H-RT + aPD-1) in BALB/c mice with bilateral breast cancer xenografts. The L-TBI dose was 0.1 Gy. The primary tumor was treated with H-RT (8 Gy × 3). The PD-1 monoclonal antibody was injected intraperitoneally, and the secondary tumors not receiving H-RT were monitored for response. The triple therapy significantly delayed both primary and secondary tumor growths, improved survival rates, and reduced the number of lung metastasis lesions. It increased the activated dendritic and CD8+ T cell populations and reduced the infiltration of myeloid-derived suppressor cells in the secondary tumor microenvironment relative to other groups. Thus, L-TBI could be a potential therapeutic modality, and when combined with H-RT and aPD-1, the therapeutic effect could be enhanced significantly.
Collapse
Affiliation(s)
- Shuya Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yin Liao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yao Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Hanshan Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yuru Hu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhuo Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China; Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, China.
| |
Collapse
|
49
|
Ottaiano A, Santorsola M, Circelli L, Trotta AM, Izzo F, Perri F, Cascella M, Sabbatino F, Granata V, Correra M, Tarotto L, Stilo S, Fiore F, Martucci N, Rocca AL, Picone C, Muto P, Borzillo V, Belli A, Patrone R, Mercadante E, Tatangelo F, Ferrara G, Di Mauro A, Scognamiglio G, Berretta M, Capuozzo M, Lombardi A, Galon J, Gualillo O, Pace U, Delrio P, Savarese G, Scala S, Nasti G, Caraglia M. Oligo-Metastatic Cancers: Putative Biomarkers, Emerging Challenges and New Perspectives. Cancers (Basel) 2023; 15:1827. [PMID: 36980713 PMCID: PMC10047282 DOI: 10.3390/cancers15061827] [Citation(s) in RCA: 5] [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: 02/23/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Some cancer patients display a less aggressive form of metastatic disease, characterized by a low tumor burden and involving a smaller number of sites, which is referred to as "oligometastatic disease" (OMD). This review discusses new biomarkers, as well as methodological challenges and perspectives characterizing OMD. Recent studies have revealed that specific microRNA profiles, chromosome patterns, driver gene mutations (ERBB2, PBRM1, SETD2, KRAS, PIK3CA, SMAD4), polymorphisms (TCF7L2), and levels of immune cell infiltration into metastases, depending on the tumor type, are associated with an oligometastatic behavior. This suggests that OMD could be a distinct disease with specific biological and molecular characteristics. Therefore, the heterogeneity of initial tumor burden and inclusion of OMD patients in clinical trials pose a crucial methodological question that requires responses in the near future. Additionally, a solid understanding of the molecular and biological features of OMD will be necessary to support and complete the clinical staging systems, enabling a better distinction of metastatic behavior and tailored treatments.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Luisa Circelli
- AMES, Centro Polidiagnostico Strumentale SRL, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy
| | - Anna Maria Trotta
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Francesco Izzo
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Francesco Perri
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Marco Cascella
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
| | - Vincenza Granata
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Marco Correra
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Luca Tarotto
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Salvatore Stilo
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Francesco Fiore
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Nicola Martucci
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Antonello La Rocca
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Carmine Picone
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Paolo Muto
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Valentina Borzillo
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Andrea Belli
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Renato Patrone
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Edoardo Mercadante
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Fabiana Tatangelo
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Gerardo Ferrara
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Annabella Di Mauro
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Giosué Scognamiglio
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | | | - Angela Lombardi
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Ugo Pace
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Paolo Delrio
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Giovanni Savarese
- AMES, Centro Polidiagnostico Strumentale SRL, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy
| | - Stefania Scala
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Guglielmo Nasti
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy
| |
Collapse
|
50
|
Zhang XW, Wu YS, Xu TM, Cui MH. CAR-T Cells in the Treatment of Ovarian Cancer: A Promising Cell Therapy. Biomolecules 2023; 13:biom13030465. [PMID: 36979400 PMCID: PMC10046142 DOI: 10.3390/biom13030465] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Ovarian cancer (OC) is among the most common gynecologic malignancies with a poor prognosis and a high mortality rate. Most patients are diagnosed at an advanced stage (stage III or IV), with 5-year survival rates ranging from 25% to 47% worldwide. Surgical resection and first-line chemotherapy are the main treatment modalities for OC. However, patients usually relapse within a few years of initial treatment due to resistance to chemotherapy. Cell-based therapies, particularly adoptive T-cell therapy and chimeric antigen receptor T (CAR-T) cell therapy, represent an alternative immunotherapy approach with great potential for hematologic malignancies. However, the use of CAR-T-cell therapy for the treatment of OC is still associated with several difficulties. In this review, we comprehensively discuss recent innovations in CAR-T-cell engineering to improve clinical efficacy, as well as strategies to overcome the limitations of CAR-T-cell therapy in OC.
Collapse
|