1
|
Chen P, Ren L, Guo Y, Sun Y. Boosting antitumor immunity in breast cancers: Potential of adjuvants, drugs, and nanocarriers. Int Rev Immunol 2024:1-24. [PMID: 39611269 DOI: 10.1080/08830185.2024.2432499] [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: 05/13/2024] [Revised: 10/05/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024]
Abstract
Despite advancements in breast cancer treatment, therapeutic resistance, and tumor recurrence continue to pose formidable challenges. Therefore, a deep knowledge of the intricate interplay between the tumor and the immune system is necessary. In the pursuit of combating breast cancer, the awakening of antitumor immunity has been proposed as a compelling avenue. Tumor stroma in breast cancers contains multiple stromal and immune cells that impact the resistance to therapy and also the expansion of malignant cells. Activating or repressing these stromal and immune cells, as well as their secretions can be proposed for exhausting resistance mechanisms and repressing tumor growth. NK cells and T lymphocytes are the prominent components of breast tumor immunity that can be triggered by adjuvants for eradicating malignant cells. However, stromal cells like endothelial and fibroblast cells, as well as some immune suppressive cells, consisting of premature myeloid cells, and some subsets of macrophages and CD4+ T lymphocytes, can dampen antitumor immunity in favor of breast tumor growth and therapy resistance. This review article aims to research the prospect of harnessing the power of drugs, adjuvants, and nanoparticles in awakening the immune reactions against breast malignant cells. By investigating the immunomodulatory properties of pharmacological agents and the synergistic effects of adjuvants, this review seeks to uncover the mechanisms through which antitumor immunity can be triggered. Moreover, the current review delineates the challenges and opportunities in the translational journey from bench to bedside.
Collapse
Affiliation(s)
- Ping Chen
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| | - Lei Ren
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| | - Youwei Guo
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| | - Yan Sun
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| |
Collapse
|
2
|
Lv Y, Zhu J, Ge S, Jiang T, Xu Y, Yao W, Jiang C. The AXL-mediated modulation of myeloid-derived suppressor cells (MDSC) in nasopharyngeal carcinoma. Med Oncol 2024; 42:17. [PMID: 39592496 DOI: 10.1007/s12032-024-02561-9] [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: 09/19/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024]
Abstract
AXL has ubiquitous expression in multiple cancers, and is strongly linked to both tumor progression, metastasis, and poor prognosis, as well as anti-tumor immune response suppression and induction of tumor resistance to immunotherapy. Therefore, it is a strong target for cancer intervention. Despite the wide application of AXL inhibitors in clinical trials, the role of AXL in the tumor immune microenvironment (TIME) remains undetermined. Herein, we established cell lines with stable AXL knockdown or overexpression using lentiviral infection. Subsequently, we co-cultured the cells with healthy human blood-derived CD33 + PBMCs. After two days of culture, we evaluated the differentiation of PBMCs into MDSCs. Additionally, the culture supernatants were collected from both the co-culture system and the individual cultures of each cell group to measure the concentrations of IL-6 and GM-CSF. Additionally, we subcutaneously administered nasopharyngeal carcinoma (NPC) cells into mice, and evaluated the association between AXL content and MDSC recruitment in the resulting tumors. We demonstrated that AXL is a critical modulator of MDSC differentiation and accumulation in NPC. It modulates IL-6, GM-CSF, and Toll-like receptor contents to achieve the aforementioned actions. Herein, we revealed a strong and direct link between AXL, cytokines in TIME, and MDSC differentiation and accumulation. Our work highlights novel approaches to optimizing existing immunotherapeutic interventions.
Collapse
Affiliation(s)
- Yu Lv
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Jiahui Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Sichen Ge
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Tao Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Yajia Xu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Weige Yao
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Chengyi Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China.
| |
Collapse
|
3
|
Bhardwaj JS, Paliwal S, Singhvi G, Taliyan R. Immunological challenges and opportunities in glioblastoma multiforme: A comprehensive view from immune system lens. Life Sci 2024; 357:123089. [PMID: 39362586 DOI: 10.1016/j.lfs.2024.123089] [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: 01/28/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
Glioblastoma multiforme (GBM), also known as grade IV astrocytoma, is the most common and deadly brain tumour. It has a poor prognosis and a low survival rate. GBM cells' immunological escape mechanism helps them resist advanced multimodal therapy. In physiological homeostasis, brain astrocytes and microglia suppress infections and clear the potential pathogen from the system. However, in severe pathological conditions like cancer, the immune response fails to eliminate mutated and rapidly over-proliferating GBM cells. The malignant cells' interactions with immune cells and the neoplasm's immunosuppressive environment enable the avoidance and their clearance. Immunotherapy efficiently addresses these difficulties, as shown by sufficient evidence. This review discusses how GBM cells inhibit and elude the immune system. These include MHC molecule expression alteration and PD-L1 and CTLA-4 immune checkpoint overexpression. Without co-stimulation, these changes induce effector T-cell tolerance and anergy. The review also covers how MDSCs, TAMs, Herpes Virus Entry Mediators, and Human cytomegalovirus protein decrease the effector immune response against glioblastoma. The latter part discusses various therapies that are available in the market or under clinical trials which revolves around combating resistance against the available multimodal therapies. The recent trends indicate that there are various monoclonal antibodies and peptide-based vaccines that can be utilized to overcome the immune evasion technique harbored by GBM cells. A strategic development of Immunotherapy considering these hallmarks of immune evasion may help in designing a therapy that may prove to be effective in killing the GBM cells thereby, improving the overall survival of GBM-affected patients.
Collapse
Affiliation(s)
- Jayant Singh Bhardwaj
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Shivangi Paliwal
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India.
| |
Collapse
|
4
|
Raymakers L, Demmers TJ, Meijer GJ, Molenaar IQ, van Santvoort HC, Intven MPW, Leusen JHW, Olofsen PA, Daamen LA. The Effect of Radiation Treatment of Solid Tumors on Neutrophil Infiltration and Function: A Systematic Review. Int J Radiat Oncol Biol Phys 2024; 120:845-861. [PMID: 39009323 DOI: 10.1016/j.ijrobp.2024.07.2141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/13/2024] [Accepted: 07/02/2024] [Indexed: 07/17/2024]
Abstract
Radiation therapy (RT) initiates a local and systemic immune response which can induce antitumor immunity and improve immunotherapy efficacy. Neutrophils are among the first immune cells that infiltrate tumors after RT and are suggested to be essential for the initial antitumor immune response. However, neutrophils in tumors are associated with poor outcomes and RT-induced neutrophil infiltration could also change the composition of the tumor microenvironment (TME) in favor of tumor progression. To improve RT efficacy for patients with cancer it is important to understand the interplay between RT and neutrophils. Here, we review the literature on how RT affects the infiltration and function of neutrophils in the TME of solid tumors, using both patients studies and preclinical murine in vivo models. In general, it was found that neutrophil levels increase and reach maximal levels in the first days after RT and can remain elevated up to 3 weeks. Most studies report an immunosuppressive role of neutrophils in the TME after RT, caused by upregulated expression of neutrophil indoleamine 2,3-dioxygenase 1 and arginase 1, as well as neutrophil extracellular trap formation. RT was also associated with increased reactive oxygen species production by neutrophils, which can both improve and inhibit antitumor immunity. In addition, multiple murine models showed improved RT efficacy when depleting neutrophils, suggesting that neutrophils have a protumor phenotype after RT. We conclude that the role of neutrophils should not be overlooked when developing RT strategies and requires further investigation in specific tumor types. In addition, neutrophils can possibly be exploited to enhance RT efficacy by combining RT with neutrophil-targeting therapies.
Collapse
Affiliation(s)
- Léon Raymakers
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Radiation Oncology, University Medical Center Utrecht, UMC Utrecht Cancer Center, Utrecht, The Netherlands
| | - Thijs J Demmers
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gert J Meijer
- Department of Radiation Oncology, University Medical Center Utrecht, UMC Utrecht Cancer Center, Utrecht, The Netherlands
| | - I Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital Nieuwegein, Utrecht University, Utrecht, The Netherlands
| | - Hjalmar C van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital Nieuwegein, Utrecht University, Utrecht, The Netherlands
| | - Martijn P W Intven
- Department of Radiation Oncology, University Medical Center Utrecht, UMC Utrecht Cancer Center, Utrecht, The Netherlands
| | - Jeanette H W Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Patricia A Olofsen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lois A Daamen
- Department of Radiation Oncology, University Medical Center Utrecht, UMC Utrecht Cancer Center, Utrecht, The Netherlands; Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center and St. Antonius Hospital Nieuwegein, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
5
|
Goldmann O, Medina E. Metabolic pathways fueling the suppressive activity of myeloid-derived suppressor cells. Front Immunol 2024; 15:1461455. [PMID: 39534601 PMCID: PMC11554506 DOI: 10.3389/fimmu.2024.1461455] [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: 07/08/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are considered an aberrant population of immature myeloid cells that have attracted considerable attention in recent years due to their potent immunosuppressive activity. These cells are typically absent or present in very low numbers in healthy individuals but become abundant under pathological conditions such as chronic infection, chronic inflammation and cancer. The immunosuppressive activity of MDSC helps to control excessive immune responses that might otherwise lead to tissue damage. This same immunosuppressive activity can be detrimental, particularly in cancer and chronic infection. In the cancer setting, tumors can secrete factors that promote the expansion and recruitment of MDSC, thereby creating a local environment that favors tumor progression by inhibiting the effective immune responses against cancer cells. This has made MDSC a target of interest in cancer therapy, with researchers exploring strategies to inhibit their function or reduce their numbers to improve the efficacy of cancer immunotherapies. In the context of chronic infections, MDSC can lead to persistent infections by suppressing protective immune responses thereby preventing the clearance of pathogens. Therefore, targeting MDSC may provide a novel approach to improve pathogen clearance during chronic infections. Ongoing research on MDSC aims to elucidate the exact processes behind their expansion, recruitment, activation and suppressive mechanisms. In this context, it is becoming increasingly clear that the metabolism of MDSC is closely linked to their immunosuppressive function. For example, MDSC exhibit high rates of glycolysis, which not only provides energy but also generates metabolites that facilitate their immunosuppressive activity. In addition, fatty acid metabolic pathways, such as fatty acid oxidation (FAO), have been implicated in the regulation of MDSC suppressive activity. Furthermore, amino acid metabolism, particularly arginine metabolism mediated by enzymes such as arginase-1, plays a critical role in MDSC-mediated immunosuppression. In this review, we discuss the metabolic signature of MDSC and highlight the therapeutic implications of targeting MDSC metabolism as a novel approach to modulate their immunosuppressive functions.
Collapse
Affiliation(s)
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
6
|
Almeida JS, Sousa LM, Couceiro P, Andrade TF, Alves V, Martinho A, Rodrigues J, Fonseca R, Freitas-Tavares P, Santos-Rosa M, Casanova JM, Rodrigues-Santos P. Peripheral immune profiling of soft tissue sarcoma: perspectives for disease monitoring. Front Immunol 2024; 15:1391840. [PMID: 39502689 PMCID: PMC11536262 DOI: 10.3389/fimmu.2024.1391840] [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: 02/26/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Studying the tumor microenvironment and surrounding lymph nodes is the main focus of current immunological research on soft tissue sarcomas (STS). However, due to the restricted opportunity to examine tumor samples, alternative approaches are required to evaluate immune responses in non-surgical patients. Therefore, the purpose of this study was to evaluate the peripheral immune profile of STS patients, characterize patients accordingly and explore the impact of peripheral immunotypes on patient survival. Blood samples were collected from 55 STS patients and age-matched healthy donors (HD) controls. Deep immunophenotyping and gene expression analysis of whole blood was analyzed using multiparametric flow cytometry and real-time RT-qPCR, respectively. Using xMAP technology, proteomic analysis was also carried out on plasma samples. Unsupervised clustering analysis was used to classify patients based on their immune profiles to further analyze the impact of peripheral immunotypes on patient survival. Significant differences were found between STS patients and HD controls. It was found a contraction of B cells and CD4 T cells compartment, along with decreased expression levels of ICOSLG and CD40LG; a major contribution of suppressor factors, as increased frequency of M-MDSC and memory Tregs, increased expression levels of ARG1, and increased plasma levels of IL-10, soluble VISTA and soluble TIMD-4; and a compromised cytotoxic potential associated with NK and CD8 T cells, namely decreased frequency of CD56dim NK cells, and decreased levels of PRF1, GZMB, and KLRK1. In addition, the patients were classified into three peripheral immunotype groups: "immune-high," "immune-intermediate," and "immune-low." Furthermore, it was found a correlation between these immunotypes and patient survival. Patients classified as "immune-high" exhibited higher levels of immune-related factors linked to cytotoxic/effector activity and longer survival times, whereas patients classified as "immune-low" displayed higher levels of immune factors associated with immunosuppression and shorter survival times. In conclusion, it can be suggested that STS patients have a compromised systemic immunity, and the correlation between immunotypes and survival emphasizes the importance of studying peripheral blood samples in STS. Assessing the peripheral immune response holds promise as a useful method for monitoring and forecasting outcomes in STS.
Collapse
Affiliation(s)
- Jani Sofia Almeida
- Center for Neurosciences and Cell Biology (CNC), Laboratory of Immunology and Oncology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine (FMUC), Institute of Immunology, University of Coimbra, Coimbra, Portugal
- Center for Investigation in Environment, Genetics and Oncobiology (CIMAGO), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Luana Madalena Sousa
- Center for Neurosciences and Cell Biology (CNC), Laboratory of Immunology and Oncology, University of Coimbra, Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Patrícia Couceiro
- Center for Neurosciences and Cell Biology (CNC), Laboratory of Immunology and Oncology, University of Coimbra, Coimbra, Portugal
- Center for Investigation in Environment, Genetics and Oncobiology (CIMAGO), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Tânia Fortes Andrade
- Center for Neurosciences and Cell Biology (CNC), Laboratory of Immunology and Oncology, University of Coimbra, Coimbra, Portugal
| | - Vera Alves
- Faculty of Medicine (FMUC), Institute of Immunology, University of Coimbra, Coimbra, Portugal
- Center for Investigation in Environment, Genetics and Oncobiology (CIMAGO), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - António Martinho
- Portuguese Institute for Blood and Transplantation (IPST), Blood and Transplantation Center of Coimbra, Coimbra, Portugal
| | - Joana Rodrigues
- Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
- Tumor Unit of the Locomotor Apparatus, University Clinic of Orthopedics, Orthopedics Oncology Service, Coimbra Hospital and Universitary Centre (CHUC), Coimbra, Portugal
| | - Ruben Fonseca
- Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
- Tumor Unit of the Locomotor Apparatus, University Clinic of Orthopedics, Orthopedics Oncology Service, Coimbra Hospital and Universitary Centre (CHUC), Coimbra, Portugal
| | - Paulo Freitas-Tavares
- Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
- Tumor Unit of the Locomotor Apparatus, University Clinic of Orthopedics, Orthopedics Oncology Service, Coimbra Hospital and Universitary Centre (CHUC), Coimbra, Portugal
| | - Manuel Santos-Rosa
- Faculty of Medicine (FMUC), Institute of Immunology, University of Coimbra, Coimbra, Portugal
- Center for Investigation in Environment, Genetics and Oncobiology (CIMAGO), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - José Manuel Casanova
- Center for Investigation in Environment, Genetics and Oncobiology (CIMAGO), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
- Tumor Unit of the Locomotor Apparatus, University Clinic of Orthopedics, Orthopedics Oncology Service, Coimbra Hospital and Universitary Centre (CHUC), Coimbra, Portugal
| | - Paulo Rodrigues-Santos
- Center for Neurosciences and Cell Biology (CNC), Laboratory of Immunology and Oncology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine (FMUC), Institute of Immunology, University of Coimbra, Coimbra, Portugal
- Center for Investigation in Environment, Genetics and Oncobiology (CIMAGO), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
| |
Collapse
|
7
|
Mashhouri S, Rahmati A, Azimi A, Fava RA, Ismail IH, Walker J, Elahi S. Targeting Dectin-1 and or VISTA enhances anti-tumor immunity in melanoma but not colorectal cancer model. Cell Oncol (Dordr) 2024; 47:1735-1756. [PMID: 38668817 PMCID: PMC11467025 DOI: 10.1007/s13402-024-00950-w] [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] [Accepted: 04/11/2024] [Indexed: 06/27/2024] Open
Abstract
PURPOSE Acquired resistance to immune checkpoint blockers (ICBs) is a major barrier in cancer treatment, emphasizing the need for innovative strategies. Dectin-1 (gene Clec7a) is a C-type lectin receptor best known for its ability to recognize β-glucan-rich structures in fungal cell walls. While Dectin-1 is expressed in myeloid cells and tumor cells, its significance in cancer remains the subject of controversy. METHODS Using Celc7a-/- mice and curdlan administration to stimulate Dectin-1 signaling, we explored its impact. VISTA KO mice were employed to assess VISTA's role, and bulk RNAseq analyzed curdlan effects on neutrophils. RESULTS Our findings reveal myeloid cells as primary Dectin-1 expressing cells in the tumor microenvironment (TME), displaying an activated phenotype. Strong Dectin-1 co-expression/co-localization with VISTA and PD-L1 in TME myeloid cells was observed. While Dectin-1 deletion lacked protective effects, curdlan stimulation significantly curtailed B16-F10 tumor progression. RNAseq and pathway analyses supported curdlan's role in triggering a cascade of events leading to increased production of pro-inflammatory mediators, potentially resulting in the recruitment and activation of immune cells. Moreover, we identified a heterogeneous subset of Dectin-1+ effector T cells in the TME. Similar to mice, human myeloid cells are the prominent cells expressing Dectin-1 in cancer patients. CONCLUSION Our study proposes Dectin-1 as a potential adjunctive target with ICBs, orchestrating a comprehensive engagement of innate and adaptive immune responses in melanoma. This innovative approach holds promise for overcoming acquired resistance to ICBs in cancer treatment, offering avenues for further exploration and development.
Collapse
Affiliation(s)
- Siavash Mashhouri
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Amirhossein Rahmati
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Ako Azimi
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Roy A Fava
- Department of Veterans Affairs Medical Center, Research Service, White River Junction, VT, USA
- Department of Medicine, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Ismail Hassan Ismail
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Biophysics Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - John Walker
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Shokrollah Elahi
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
8
|
Poyia F, Neophytou CM, Christodoulou MI, Papageorgis P. The Role of Tumor Microenvironment in Pancreatic Cancer Immunotherapy: Current Status and Future Perspectives. Int J Mol Sci 2024; 25:9555. [PMID: 39273502 PMCID: PMC11395109 DOI: 10.3390/ijms25179555] [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: 07/26/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Pancreatic cancer comprises different subtypes, where most cases include ductal adenocarcinoma (PDAC). It is one of the deadliest tumor types, with a poor prognosis. In the majority of patients, the disease has already spread by the time of diagnosis, making full recovery unlikely and increasing mortality risk. Despite developments in its detection and management, including chemotherapy, radiotherapy, and targeted therapies as well as advances in immunotherapy, only in about 13% of PDAC patients does the overall survival exceed 5 years. This may be attributed, at least in part, to the highly desmoplastic tumor microenvironment (TME) that acts as a barrier limiting perfusion, drug delivery, and immune cell infiltration and contributes to the establishment of immunologically 'cold' conditions. Therefore, there is an urgent need to unravel the complexity of the TME that promotes PDAC progression and decipher the mechanisms of pancreatic tumors' resistance to immunotherapy. In this review, we provide an overview of the major cellular and non-cellular components of PDAC TME, as well as their biological interplays. We also discuss the current state of PDAC therapeutic treatments and focus on ongoing and future immunotherapy efforts and multimodal treatments aiming at remodeling the TME to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Fotini Poyia
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Christiana M Neophytou
- Apoptosis and Cancer Chemoresistance Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Maria-Ioanna Christodoulou
- Tumor Immunology and Biomarkers Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Panagiotis Papageorgis
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| |
Collapse
|
9
|
Horowitz NA, Abed Al Wahad A, Bettman NP, Ringelstein-Harlev S, Brenner B, Katz T. Acceleration of non-Hodgkin lymphoma progression during pregnancy in a murine model. Leuk Lymphoma 2024; 65:1370-1373. [PMID: 38781586 DOI: 10.1080/10428194.2024.2353879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/28/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024]
Affiliation(s)
- Netanel A Horowitz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Ali Abed Al Wahad
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Noam P Bettman
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | | | - Benjamin Brenner
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Tami Katz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
10
|
Takacs GP, Garcia JS, Hodges CA, Kreiger CJ, Sherman A, Harrison JK. CSF1R Ligands Expressed by Murine Gliomas Promote M-MDSCs to Suppress CD8 + T Cells in a NOS-Dependent Manner. Cancers (Basel) 2024; 16:3055. [PMID: 39272914 PMCID: PMC11394022 DOI: 10.3390/cancers16173055] [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: 06/08/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor, resulting in poor survival despite aggressive therapies. GBM is characterized by a highly heterogeneous and immunosuppressive tumor microenvironment (TME) made up predominantly of infiltrating peripheral immune cells. One significant immune cell type that contributes to glioma immune evasion is a population of immunosuppressive cells, termed myeloid-derived suppressor cells (MDSCs). Previous studies suggest that a subset of myeloid cells, expressing monocytic (M)-MDSC markers and dual expression of chemokine receptors CCR2 and CX3CR1, utilize CCR2 to infiltrate the TME. This study evaluated the mechanism of CCR2+/CX3CR1+ M-MDSC differentiation and T cell suppressive function in murine glioma models. We determined that bone marrow-derived CCR2+/CX3CR1+ cells adopt an immune suppressive cell phenotype when cultured with glioma-derived factors. Glioma-secreted CSF1R ligands M-CSF and IL-34 were identified as key drivers of M-MDSC differentiation while adenosine and iNOS pathways were implicated in the M-MDSC suppression of T cells. Mining a human GBM spatial RNAseq database revealed a variety of different pathways that M-MDSCs utilize to exert their suppressive function that is driven by complex niches within the microenvironment. These data provide a more comprehensive understanding of the mechanism of M-MDSCs in glioblastoma.
Collapse
Affiliation(s)
| | | | | | | | | | - Jeffrey K. Harrison
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA (J.S.G.); (C.A.H.)
| |
Collapse
|
11
|
Andrew Awuah W, Shah MH, Tan JK, Ranganathan S, Sanker V, Darko K, Tenkorang PO, Adageba BB, Ahluwalia A, Shet V, Aderinto N, Kundu M, Abdul‐Rahman T, Atallah O. Immunotherapeutic advances in glioma management: The rise of vaccine-based approaches. CNS Neurosci Ther 2024; 30:e70013. [PMID: 39215399 PMCID: PMC11364516 DOI: 10.1111/cns.70013] [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: 02/19/2024] [Revised: 07/23/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Gliomas, particularly glioblastoma multiforme (GBM), are highly aggressive brain tumors that present significant challenges in oncology due to their rapid progression and resistance to conventional therapies. Despite advancements in treatment, the prognosis for patients with GBM remains poor, necessitating the exploration of novel therapeutic approaches. One such emerging strategy is the development of glioma vaccines, which aim to stimulate the immune system to target and destroy tumor cells. AIMS This review aims to provide a comprehensive evaluation of the current landscape of glioma vaccine development, analyzing the types of vaccines under investigation, the outcomes of clinical trials, and the challenges and opportunities associated with their implementation. The goal is to highlight the potential of glioma vaccines in advancing more effective and personalized treatments for glioma patients. MATERIALS AND METHODS This narrative review systematically assessed the role of glioma vaccines by including full-text articles published between 2000 and 2024 in English. Databases such as PubMed/MEDLINE, EMBASE, the Cochrane Library, and Scopus were searched using key terms like "glioma," "brain tumor," "glioblastoma," "vaccine," and "immunotherapy." The review incorporated both pre-clinical and clinical studies, including descriptive studies, animal-model studies, cohort studies, and observational studies. Exclusion criteria were applied to omit abstracts, case reports, posters, and non-peer-reviewed studies, ensuring the inclusion of high-quality evidence. RESULTS Clinical trials investigating various glioma vaccines, including peptide-based, DNA/RNA-based, whole-cell, and dendritic-cell vaccines, have shown promising results. These vaccines demonstrated potential in extending survival rates and managing adverse events in glioma patients. However, significant challenges remain, such as therapeutic resistance due to tumor heterogeneity and immune evasion mechanisms. Moreover, the lack of standardized guidelines for evaluating vaccine responses and issues related to ethical considerations, regulatory hurdles, and vaccine acceptance among patients further complicate the implementation of glioma vaccines. DISCUSSION Addressing the challenges associated with glioma vaccines involves exploring combination therapies, targeted approaches, and personalized medicine. Combining vaccines with traditional therapies like radiotherapy or chemotherapy may enhance efficacy by boosting the immune system's ability to fight tumor cells. Personalized vaccines tailored to individual patient profiles present an opportunity for improved outcomes. Furthermore, global collaboration and equitable distribution are critical for ensuring access to glioma vaccines, especially in low- and middle-income countries with limited healthcare resources CONCLUSION: Glioma vaccines represent a promising avenue in the fight against gliomas, offering hope for improving patient outcomes in a disease that is notoriously difficult to treat. Despite the challenges, continued research and the development of innovative strategies, including combination therapies and personalized approaches, are essential for overcoming current barriers and transforming the treatment landscape for glioma patients.
Collapse
Affiliation(s)
| | | | | | | | - Vivek Sanker
- Department of NeurosurgeryTrivandrum Medical CollegeTrivandrumKeralaIndia
| | - Kwadwo Darko
- Department of NeurosurgeryKorle Bu Teaching HospitalAccraGhana
| | | | - Bryan Badayelba Adageba
- Kwame Nkrumah University of Science and Technology School of Medicine and DentistryKumasiGhana
| | | | - Vallabh Shet
- Faculty of MedicineBangalore Medical College and Research InstituteBangaloreKarnatakaIndia
| | - Nicholas Aderinto
- Department of Internal MedicineLAUTECH Teaching HospitalOgbomosoNigeria
| | - Mrinmoy Kundu
- Institute of Medical Sciences and SUM HospitalBhubaneswarOdishaIndia
| | | | - Oday Atallah
- Department of Neurosurgery, Hannover Medical SchoolHannoverGermany
| |
Collapse
|
12
|
Arshi A, Mahmoudi E, Raeisi F, Dehghan Tezerjani M, Bahramian E, Ahmed Y, Peng C. Exploring potential roles of long non-coding RNAs in cancer immunotherapy: a comprehensive review. Front Immunol 2024; 15:1446937. [PMID: 39257589 PMCID: PMC11384988 DOI: 10.3389/fimmu.2024.1446937] [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: 06/10/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Cancer treatment has long been fraught with challenges, including drug resistance, metastasis, and recurrence, making it one of the most difficult diseases to treat effectively. Traditional therapeutic approaches often fall short due to their inability to target cancer stem cells and the complex genetic and epigenetic landscape of tumors. In recent years, cancer immunotherapy has revolutionized the field, offering new hope and viable alternatives to conventional treatments. A particularly promising area of research focuses on non-coding RNAs (ncRNAs), especially long non-coding RNAs (lncRNAs), and their role in cancer resistance and the modulation of signaling pathways. To address these challenges, we performed a comprehensive review of recent studies on lncRNAs and their impact on cancer immunotherapy. Our review highlights the crucial roles that lncRNAs play in affecting both innate and adaptive immunity, thereby influencing the outcomes of cancer treatments. Key observations from our review indicate that lncRNAs can modify the tumor immune microenvironment, enhance immune cell infiltration, and regulate cytokine production, all of which contribute to tumor growth and resistance to therapies. These insights suggest that lncRNAs could serve as potential targets for precision medicine, opening up new avenues for developing more effective cancer immunotherapies. By compiling recent research on lncRNAs across various cancers, this review aims to shed light on their mechanisms within the tumor immune microenvironment.
Collapse
Affiliation(s)
- Asghar Arshi
- Department of Biology, York University, Toronto, ON, Canada
| | - Esmaeil Mahmoudi
- Young Researchers and Elite Club, Islamic Azad University, Shahrekord, Iran
| | | | - Masoud Dehghan Tezerjani
- Department of bioinformatics, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Bahramian
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, United States
| | - Yeasin Ahmed
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, United States
| | - Chun Peng
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
13
|
Xia X, Yang Z, Lu Q, Liu Z, Wang L, Du J, Li Y, Yang DH, Wu S. Reshaping the tumor immune microenvironment to improve CAR-T cell-based cancer immunotherapy. Mol Cancer 2024; 23:175. [PMID: 39187850 PMCID: PMC11346058 DOI: 10.1186/s12943-024-02079-8] [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: 06/14/2024] [Accepted: 08/02/2024] [Indexed: 08/28/2024] Open
Abstract
In many hematologic malignancies, the adoptive transfer of chimeric antigen receptor (CAR) T cells has demonstrated notable success; nevertheless, further improvements are necessary to optimize treatment efficacy. Current CAR-T therapies are particularly discouraging for solid tumor treatment. The immunosuppressive microenvironment of tumors affects CAR-T cells, limiting the treatment's effectiveness and safety. Therefore, enhancing CAR-T cell infiltration capacity and resolving the immunosuppressive responses within the tumor microenvironment could boost the anti-tumor effect. Specific strategies include structurally altering CAR-T cells combined with targeted therapy, radiotherapy, or chemotherapy. Overall, monitoring the tumor microenvironment and the status of CAR-T cells is beneficial in further investigating the viability of such strategies and advancing CAR-T cell therapy.
Collapse
Affiliation(s)
- Xueting Xia
- The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Zongxin Yang
- The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Qisi Lu
- The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Foresea Life Insurance Guangzhou General Hospital, Guangzhou, 511300, China
| | - Zhenyun Liu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Lei Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jinwen Du
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, Mineola, NY, 11501, USA.
| | - Shaojie Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
14
|
Tominaga M, Uto T, Fukaya T, Mitoma S, Riethmacher D, Umekita K, Yamashita Y, Sato K. Crucial role of dendritic cells in the generation of anti-tumor T-cell responses and immunogenic tumor microenvironment to suppress tumor development. Front Immunol 2024; 15:1200461. [PMID: 39206204 PMCID: PMC11349553 DOI: 10.3389/fimmu.2024.1200461] [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: 04/05/2023] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Dendritic cells (DCs) are known as unique professional antigen (Ag)-presenting cells (APCs) to prime naïve T cells for the initiation of adaptive immunity. While DCs are believed to play a pivotal role in generating anti-tumor T-cell responses, the importance of DCs in the protection from the progression of tumors remains elusive. Here, we show how the constitutive deficiency of CD11chi DCs influences the progression of tumors with the use of binary transgenic mice with constitutive loss of CD11chi DCs. Constitutive loss of CD11chi DCs not only enhances the progression of tumors but also reduces the responses of Ag-specific T cells. Furthermore, the congenital deficiency of CD11chi DCs generates the immunosuppressive tumor microenvironment (TME) that correlates with the marked accumulation of myeloid-derived suppressor cells (MDSCs) and the prominent productions of immunosuppressive mediators. Thus, our findings suggest that CD11chi DCs are crucial for generating anti-tumor T-cell responses and immunogenic TME to suppress the development of tumors.
Collapse
Affiliation(s)
- Moe Tominaga
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Tomofumi Uto
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Project for Promotion of Cancer Research and Therapeutic Evolution (P-PROMOTE), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| | - Tomohiro Fukaya
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Project for Promotion of Cancer Research and Therapeutic Evolution (P-PROMOTE), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| | - Shuya Mitoma
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Project for Promotion of Cancer Research and Therapeutic Evolution (P-PROMOTE), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| | - Dieter Riethmacher
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan
| | - Kunihiko Umekita
- Division of Respirology, Rheumatology, Infectious Diseases, and Neurology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yoshihiro Yamashita
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Katsuaki Sato
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Project for Promotion of Cancer Research and Therapeutic Evolution (P-PROMOTE), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
- Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
15
|
Bian X, Liu W, Yang K, Sun C. Therapeutic targeting of PARP with immunotherapy in acute myeloid leukemia. Front Pharmacol 2024; 15:1421816. [PMID: 39175540 PMCID: PMC11338796 DOI: 10.3389/fphar.2024.1421816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
Targeting the poly (ADP-ribose) polymerase (PARP) protein has shown therapeutic efficacy in cancers with homologous recombination (HR) deficiency due to BRCA mutations. Only small fraction of acute myeloid leukemia (AML) cells carry BRCA mutations, hence the antitumor efficacy of PARP inhibitors (PARPi) against this malignancy is predicted to be limited; however, recent preclinical studies have demonstrated that PARPi monotherapy has modest efficacy in AML, while in combination with cytotoxic chemotherapy it has remarkable synergistic antitumor effects. Immunotherapy has revolutionized therapeutics in cancer treatment, and PARPi creates an ideal microenvironment for combination therapy with immunomodulatory agents by promoting tumor mutation burden. In this review, we summarize the role of PARP proteins in DNA damage response (DDR) pathways, and discuss recent preclinical studies using synthetic lethal modalities to treat AML. We also review the immunomodulatory effects of PARPi in AML preclinical models and propose future directions for therapy in AML, including combined targeting of the DDR and tumor immune microenvironment; such combination regimens will likely benefit patients with AML undergoing PARPi-mediated cancer therapy.
Collapse
Affiliation(s)
- Xing Bian
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu’an, China
| | - Wenli Liu
- Food and Drug Inspection Center, Lu’an, China
| | - Kaijin Yang
- Food and Drug Inspection Center, Huai’nan, China
| | - Chuanbo Sun
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu’an, China
| |
Collapse
|
16
|
Harley RJ, Lyden M, Aribindi S, Socolovsky L, Harley EH. Head and Neck Merkel Cell Carcinoma: Therapeutic Benefit of Adjuvant Radiotherapy for Nodal Disease. Laryngoscope 2024; 134:3587-3594. [PMID: 38401116 DOI: 10.1002/lary.31333] [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: 06/22/2023] [Revised: 11/26/2023] [Accepted: 01/23/2024] [Indexed: 02/26/2024]
Abstract
OBJECTIVES To evaluate the therapeutic effect of post-operative radiotherapy (PORT) with respect to nodal status among patients with head and neck Merkel cell carcinoma (HNMCC). METHODS In this retrospective study, we queried Surveillance, Epidemiology, and End Results (SEER) dataset from 2000 through 2019. We included all adult patients who received primary surgical resection for histologically confirmed treatment naive HNMCC. Entropy balancing was used to reweight observations such that there was covariate balance between patients who received PORT and patients who received surgical resection alone. Doubly robust estimation was achieved by incorporating weights into a multivariable cox proportional hazards model. Planned post hoc subgroup analysis was performed to evaluate the impact of PORT by pathological node status. RESULTS Among 752 patients (mean age, 73.3 years [SD 10.8]; 64.2% male; 91.2% White; 41.9% node-positive), 60.4% received PORT. Among node-positive patients, we found that PORT was associated with improved overall survival (OS) (aHR, 0.55; 95% CI, 0.37-0.81; p = 0.003) and improved disease-specific survival (DSS) (aHR, 0.57; 95% CI, 0.35-0.92; p = 0.022). Among node-negative patients, we found that PORT was not associated with OS and was associated with worse DSS (aHR, 2.34; 95% CI, 1.30-4.23; p = 0.005). CONCLUSIONS We found that PORT was associated with improved OS and DSS for node-positive patients and worse DSS for node-negative patients. For HNMCC treated with primary surgical resection, these data confirm the value of PORT for pathologically node-positive patients and support the use of single modality surgical therapy for pathologically node-negative patients without other adverse risk factors. LEVEL OF EVIDENCE 4 Laryngoscope, 134:3587-3594, 2024.
Collapse
Affiliation(s)
- Randall J Harley
- Department of Otolaryngology - Head and Neck Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - Megan Lyden
- Department of Otolaryngology - Head and Neck Surgery, Georgetown University Hospital, Washington, District of Columbia, U.S.A
| | - Seetha Aribindi
- Department of Otolaryngology - Head and Neck Surgery, Georgetown University Hospital, Washington, District of Columbia, U.S.A
| | - Leandro Socolovsky
- Department of Otolaryngology - Head and Neck Surgery, Georgetown University Hospital, Washington, District of Columbia, U.S.A
| | - Earl H Harley
- Department of Otolaryngology - Head and Neck Surgery, Georgetown University Hospital, Washington, District of Columbia, U.S.A
| |
Collapse
|
17
|
Jiménez-Cortegana C, Gutiérrez-García C, Sánchez-Jiménez F, Vilariño-García T, Flores-Campos R, Pérez-Pérez A, Garnacho C, Sánchez-León ML, García-Domínguez DJ, Hontecillas-Prieto L, Palazón-Carrión N, De La Cruz-Merino L, Sánchez-Margalet V. Impact of obesity‑associated myeloid‑derived suppressor cells on cancer risk and progression (Review). Int J Oncol 2024; 65:79. [PMID: 38940351 PMCID: PMC11251741 DOI: 10.3892/ijo.2024.5667] [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: 04/09/2024] [Accepted: 06/12/2024] [Indexed: 06/29/2024] Open
Abstract
Obesity is a chronic disease caused by the accumulation of excessive adipose tissue. This disorder is characterized by chronic low‑grade inflammation, which promotes the release of proinflammatory mediators, including cytokines, chemokines and leptin. Simultaneously, chronic inflammation can predispose to cancer development, progression and metastasis. Proinflammatory molecules are involved in the recruitment of specific cell populations in the tumor microenvironment. These cell populations include myeloid‑derived suppressor cells (MDSCs), a heterogeneous, immature myeloid population with immunosuppressive abilities. Obesity‑associated MDSCs have been linked with tumor dissemination, progression and poor clinical outcomes. A comprehensive literature review was conducted to assess the impact of obesity‑associated MDSCs on cancer in both preclinical models and oncological patients with obesity. A secondary objective was to examine the key role that leptin, the most important proinflammatory mediator released by adipocytes, plays in MDSC‑driven immunosuppression Finally, an overview is provided of the different therapeutic approaches available to target MDSCs in the context of obesity‑related cancer.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Cristian Gutiérrez-García
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Flora Sánchez-Jiménez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Teresa Vilariño-García
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Rocio Flores-Campos
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Antonio Pérez-Pérez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Carmen Garnacho
- Department of Normal and Pathological Histology and Cytology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Maria L. Sánchez-León
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Daniel J. García-Domínguez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Natalia Palazón-Carrión
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Luis De La Cruz-Merino
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, Seville 41013, Spain
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, Seville 41013, Spain
| |
Collapse
|
18
|
Abunawas D, Abbasy A, Afifi M, Moaaz M, Kamal A, Awaad A, Elsherbini B. MYELOID-DERIVED SUPPRESSOR CELLS TWO YEARS AFTER HEPATITIS C VIRUS ERADICATION USING DIRECTLY ACTING ANTIVIRALS. ARQUIVOS DE GASTROENTEROLOGIA 2024; 61:e24004. [PMID: 39046003 DOI: 10.1590/s0004-2803.24612024-004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/06/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Myeloid-derived suppressor cells (MDSCs) have immature morphology, relatively weak phagocytic activity, as well as some immunosuppressive functions. The capacity of MDSCs to inhibit T-cell-mediated immunological responses is their most notable functional characteristic. Down-regulating antitumor immune surveillance is one way that the expansion and activation of MDSCs contribute significantly to the occurrence and progression of tumors. Increased levels of MDSCs in patients with chronic hepatitis C virus (HCV) infection could suppress T-cell responses, promoting viral escape and hepatitis progression. This may make HCV-infected individuals more vulnerable to severe infections, hepatic and extra-hepatic tumors, and a diminished capacity to react to immunization. It is still unknown if effective HCV eradication with directly acting antivirals (DAAs) can restore immune functions and immune surveillance capacity. OBJECTIVE The purpose of this study was to observe the frequency of M-MDSCs (CD33+, CD11b+, and HLA-DR) in patients with a previous history of HCV, 2-3 years after virus eradication using DAA therapy. METHODS This study was conducted on 110 subjects: fifty-five subjects without liver cirrhosis who were treated with HCV using DAAs and attained SVR for a period of 2-3 years and 55 age- and gender-matched healthy controls. The study was conducted during the period from January to July 2022. Patients were recruited from the National Viral Hepatitis Treatment Unit, Alexandria University Hepatology outpatient clinic, and the Alexandria University Tropical Medicine outpatient clinic. The frequencies of MDSCs (CD33+CD11b + HLA-DR-) by flow cytometry were assessed. RESULTS Even after the virus had been eradicated for longer than two years, MDSC levels in HCV-treated individuals were found to be considerably higher. In the HCV-treated group, the median number of MDSCs was 5, with an interquartile range (IQR) of 3.79-7.69. In contrast, the median for the control group was 3.1, with an IQR of 1.4-3.2 (P˂0.001). CONCLUSION Successful DAA therapy leads to slow and partial immunological reconstitution, as demonstrated by the failure to attain normal levels of MDSC's 2 years after successful HCV eradication despite the normalization of laboratory parameters as well as the absence of liver fibrosis. The clinical implications of these findings should be thoroughly studied.
Collapse
Affiliation(s)
- Dania Abunawas
- Academic fellow, Immunology and Allergy department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Amany Abbasy
- Lecturer of Tropical Medicine, Tropical Medicine department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mohamed Afifi
- Professor of Microbiology and Immunology, Allergy and Immunology department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Mai Moaaz
- Professor of Immunology and Allergy, Immunology and Allergy department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Ahmed Kamal
- Lecturer of Internal Medicine and Hepatology, Internal Medicine department, Hepatology Unit, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ashraf Awaad
- Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Basem Elsherbini
- Lecturer of Immunology and Allergy, Immunology and Allergy department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
19
|
Taghizadeh-Hesary F. "Reinforcement" by Tumor Microenvironment: The Seventh "R" of Radiobiology. Int J Radiat Oncol Biol Phys 2024; 119:727-733. [PMID: 38032584 DOI: 10.1016/j.ijrobp.2023.09.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/11/2023] [Accepted: 09/16/2023] [Indexed: 12/01/2023]
Affiliation(s)
- Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Clinical Oncology Department, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Liang H, Cui M, Tu J, Chen X. Advancements in osteosarcoma management: integrating immune microenvironment insights with immunotherapeutic strategies. Front Cell Dev Biol 2024; 12:1394339. [PMID: 38915446 PMCID: PMC11194413 DOI: 10.3389/fcell.2024.1394339] [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: 03/01/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024] Open
Abstract
Osteosarcoma, a malignant bone tumor predominantly affecting children and adolescents, presents significant therapeutic challenges, particularly in metastatic or recurrent cases. Conventional surgical and chemotherapeutic approaches have achieved partial therapeutic efficacy; however, the prognosis for long-term survival remains bleak. Recent studies have highlighted the imperative for a comprehensive exploration of the osteosarcoma immune microenvironment, focusing on the integration of diverse immunotherapeutic strategies-including immune checkpoint inhibitors, tumor microenvironment modulators, cytokine therapies, tumor antigen-specific interventions, cancer vaccines, cellular therapies, and antibody-based treatments-that are directly pertinent to modulating this intricate microenvironment. By targeting tumor cells, modulating the tumor microenvironment, and activating host immune responses, these innovative approaches have demonstrated substantial potential in enhancing the effectiveness of osteosarcoma treatments. Although most of these novel strategies are still in research or clinical trial phases, they have already demonstrated significant potential for individuals with osteosarcoma, suggesting the possibility of developing new, more personalized and effective treatment options. This review aims to provide a comprehensive overview of the current advancements in osteosarcoma immunotherapy, emphasizing the significance of integrating various immunotherapeutic methods to optimize therapeutic outcomes. Additionally, it underscores the imperative for subsequent research to further investigate the intricate interactions between the tumor microenvironment and the immune system, aiming to devise more effective treatment strategies. The present review comprehensively addresses the landscape of osteosarcoma immunotherapy, delineating crucial scientific concerns and clinical challenges, thereby outlining potential research directions.
Collapse
Affiliation(s)
- Hang Liang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Cui
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Takacs GP, Garcia JS, Hodges CA, Kreiger CJ, Sherman A, Harrison JK. Glioma-derived M-CSF and IL-34 license M-MDSCs to suppress CD8 + T cells in a NOS-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597474. [PMID: 38895268 PMCID: PMC11185662 DOI: 10.1101/2024.06.05.597474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor, resulting in poor survival despite aggressive therapies. GBM is characterized by a highly heterogeneous and immunosuppressive tumor microenvironment (TME) made up predominantly of infiltrating peripheral immune cells. One significant immune cell type that contributes to glioma immune evasion is a population of immunosuppressive cells, termed myeloid-derived suppressor cells (MDSCs). Previous studies suggest that a subset of myeloid cells, expressing monocytic (M)-MDSC markers and dual expression of chemokine receptors CCR2 and CX3CR1, utilize CCR2 to infiltrate the TME. This study evaluated the mechanism of CCR2+/CX3CR1+ M-MDSC differentiation and T cell suppressive function in murine glioma models. We determined that bone marrow-derived CCR2+/CX3CR1+ cells adopt an immune suppressive cell phenotype when cultured with glioma-derived factors. Glioma secreted CSF1R ligands M-CSF and IL-34 were identified as key drivers of M-MDSC differentiation while adenosine and iNOS pathways were implicated in M-MDSC suppression of T cells. Mining a human GBM spatial RNAseq database revealed a variety of different pathways that M-MDSCs utilize to exert their suppressive function that are driven by complex niches within the microenvironment. These data provide a more comprehensive understanding of the mechanism of M-MDSCs in glioblastoma.
Collapse
Affiliation(s)
- Gregory P. Takacs
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| | - Julia S. Garcia
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| | - Caitlyn A. Hodges
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| | - Christian J. Kreiger
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| | - Alexandra Sherman
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| | - Jeffrey K. Harrison
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| |
Collapse
|
22
|
Mandal SK, Yadav P, Sheth RA. The Neuroimmune Axis and Its Therapeutic Potential for Primary Liver Cancer. Int J Mol Sci 2024; 25:6237. [PMID: 38892423 PMCID: PMC11172507 DOI: 10.3390/ijms25116237] [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: 02/05/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
The autonomic nervous system plays an integral role in motion and sensation as well as the physiologic function of visceral organs. The nervous system additionally plays a key role in primary liver diseases. Until recently, however, the impact of nerves on cancer development, progression, and metastasis has been unappreciated. This review highlights recent advances in understanding neuroanatomical networks within solid organs and their mechanistic influence on organ function, specifically in the liver and liver cancer. We discuss the interaction between the autonomic nervous system, including sympathetic and parasympathetic nerves, and the liver. We also examine how sympathetic innervation affects metabolic functions and diseases like nonalcoholic fatty liver disease (NAFLD). We also delve into the neurobiology of the liver, the interplay between cancer and nerves, and the neural regulation of the immune response. We emphasize the influence of the neuroimmune axis in cancer progression and the potential of targeted interventions like neurolysis to improve cancer treatment outcomes, especially for hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
| | | | - Rahul A. Sheth
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1471, Houston, TX 77030-4009, USA; (S.K.M.); (P.Y.)
| |
Collapse
|
23
|
Ju SH, Song M, Lim JY, Kang YE, Yi HS, Shong M. Metabolic Reprogramming in Thyroid Cancer. Endocrinol Metab (Seoul) 2024; 39:425-444. [PMID: 38853437 PMCID: PMC11220218 DOI: 10.3803/enm.2023.1802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/25/2024] [Accepted: 03/12/2024] [Indexed: 06/11/2024] Open
Abstract
Thyroid cancer is a common endocrine malignancy with increasing incidence globally. Although most cases can be treated effectively, some cases are more aggressive and have a higher risk of mortality. Inhibiting RET and BRAF kinases has emerged as a potential therapeutic strategy for the treatment of thyroid cancer, particularly in cases of advanced or aggressive disease. However, the development of resistance mechanisms may limit the efficacy of these kinase inhibitors. Therefore, developing precise strategies to target thyroid cancer cell metabolism and overcome resistance is a critical area of research for advancing thyroid cancer treatment. In the field of cancer therapeutics, researchers have explored combinatorial strategies involving dual metabolic inhibition and metabolic inhibitors in combination with targeted therapy, chemotherapy, and immunotherapy to overcome the challenge of metabolic plasticity. This review highlights the need for new therapeutic approaches for thyroid cancer and discusses promising metabolic inhibitors targeting thyroid cancer. It also discusses the challenges posed by metabolic plasticity in the development of effective strategies for targeting cancer cell metabolism and explores the potential advantages of combined metabolic targeting.
Collapse
Affiliation(s)
- Sang-Hyeon Ju
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - Minchul Song
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - Joung Youl Lim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - Yea Eun Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Korea
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hyon-Seung Yi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Korea
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Minho Shong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| |
Collapse
|
24
|
Liu Q, Zhou X, Liu K, Wang Y, Liu C, Gao C, Cai Q, Sun C. Exploring risk factors for autoimmune diseases complicated by non-hodgkin lymphoma through regulatory T cell immune-related traits: a Mendelian randomization study. Front Immunol 2024; 15:1374938. [PMID: 38863695 PMCID: PMC11165099 DOI: 10.3389/fimmu.2024.1374938] [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: 01/23/2024] [Accepted: 05/08/2024] [Indexed: 06/13/2024] Open
Abstract
Background The effect of immune cells on autoimmune diseases (ADs) complicated by non-Hodgkin lymphoma (NHL) has been widely recognized, but a causal relationship between regulatory T cell (Treg) immune traits and ADs complicated by NHL remains debated. Methods Aggregate data for 84 Treg-related immune traits were downloaded from the Genome-Wide Association Study (GWAS) catalog, and GWAS data for diffuse large B-cell lymphoma (DLBCL; n=315243), follicular lymphoma (FL; n=325831), sjögren's syndrome (SS; n=402090), rheumatoid arthritis (RA; n=276465), dermatopolymyositis (DM; n=311640), psoriasis (n=407876), atopic dermatitis (AD; n=382254), ulcerative colitis (UC; n=411317), crohn's disease(CD; n=411973) and systemic lupus erythematosus (SLE; n=307587) were downloaded from the FinnGen database. The inverse variance weighting (IVW) method was mainly used to infer any causal association between Treg-related immune traits and DLBCL, FL, SS, DM, RA, Psoriasis, AD, UC, CD and SLE, supplemented by MR-Egger, weighted median, simple mode, and weighted mode. Moreover, we performed sensitivity analyses to assess the validity of the causal relationships. Results There was a potential genetic predisposition association identified between CD39+ CD8br AC, CD39+ CD8br % T cell, and the risk of DLBCL (OR=1.51, p<0.001; OR=1.25, p=0.001) (adjusted FDR<0.1). Genetic prediction revealed potential associations between CD25++ CD8br AC, CD28- CD25++ CD8br % T cell, CD39+ CD8br % CD8br, and the risk of FL (OR=1.13, p=0.022; OR=1.28, p=0.042; OR=0.90, p=0.016) (adjusted FDR>0.1). Furthermore, SLE and CD exhibited a genetically predicted potential association with the CD39+ CD8+ Tregs subset. SS and DM were possibly associated with an increase in the quantity of the CD4+ Tregs subset; RA may have reduced the quantity of the CD39+ CD8+ Tregs subset, although no causal relationship was identified. Sensitivity analyses supported the robustness of our findings. Conclusions There existed a genetically predicted potential association between the CD39+ CD8+ Tregs subset and the risk of DLBCL, while SLE and CD were genetically predicted to be potentially associated with the CD39+ CD8+ Tregs subset. The CD39+ CD8+ Tregs subset potentially aided in the clinical diagnosis and treatment of SLE or CD complicated by DLBCL.
Collapse
Affiliation(s)
- Qi Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xintong Zhou
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kunjing Liu
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing, China
| | - Yimin Wang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China
| | - Chundi Gao
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China
| | - Qingqing Cai
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| |
Collapse
|
25
|
Wu LY, Park SH, Jakobsson H, Shackleton M, Möller A. Immune Regulation and Immune Therapy in Melanoma: Review with Emphasis on CD155 Signalling. Cancers (Basel) 2024; 16:1950. [PMID: 38893071 PMCID: PMC11171058 DOI: 10.3390/cancers16111950] [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: 03/28/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Melanoma is commonly diagnosed in a younger population than most other solid malignancies and, in Australia and most of the world, is the leading cause of skin-cancer-related death. Melanoma is a cancer type with high immunogenicity; thus, immunotherapies are used as first-line treatment for advanced melanoma patients. Although immunotherapies are working well, not all the patients are benefitting from them. A lack of a comprehensive understanding of immune regulation in the melanoma tumour microenvironment is a major challenge of patient stratification. Overexpression of CD155 has been reported as a key factor in melanoma immune regulation for the development of therapy resistance. A more thorough understanding of the actions of current immunotherapy strategies, their effects on immune cell subsets, and the roles that CD155 plays are essential for a rational design of novel targets of anti-cancer immunotherapies. In this review, we comprehensively discuss current anti-melanoma immunotherapy strategies and the immune response contribution of different cell lineages, including tumour endothelial cells, myeloid-derived suppressor cells, cytotoxic T cells, cancer-associated fibroblast, and nature killer cells. Finally, we explore the impact of CD155 and its receptors DNAM-1, TIGIT, and CD96 on immune cells, especially in the context of the melanoma tumour microenvironment and anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Li-Ying Wu
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Su-Ho Park
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haakan Jakobsson
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
| | - Mark Shackleton
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
- School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Andreas Möller
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
26
|
Mahale A, Routholla G, Lavanya S, Sharma P, Ghosh B, Kulkarni OP. Pharmacological blockade of HDAC6 attenuates cancer progression by inhibiting IL-1β and modulating immunosuppressive response in OSCC. Int Immunopharmacol 2024; 132:111921. [PMID: 38547770 DOI: 10.1016/j.intimp.2024.111921] [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: 10/19/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 05/01/2024]
Abstract
Interleukin-1-beta (IL-1β) one of the biomarkers for oral squamous cell carcinoma (OSCC), is upregulated in tumor-microenvironment (TME) and associated with poor patient survival. Thus, a novel modulator of IL-1β would be of great therapeutic value for OSCC treatment. Here we report regulation of IL-1β and TME by histone deacetylase-6 (HDAC6)-inhibitor in OSCC. We observed significant upregulation of HDAC6 in 4-nitroquniline (4-NQO)-induced OSCC in mice and 4-NQO & Lipopolysaccharide (LPS) stimulated OSCC and fibroblast cells. Tubastatin A (TSA)-attenuated the OSCC progression in mice as observed improvement in the histology over tongue and esophagus, with reduced tumor burden. TSA treatment to 4-NQO mice attenuated protein expression of HDAC6, pro-and-mature-IL-1β and pro-and-cleaved-caspase-1 and ameliorated acetylated-tubulin. In support of our experimental work, human TCGA analysis revealed HDAC6 and IL-1β were upregulated in the primary tumor, with different tumor stages and grades. We found TSA modulate TME, indicated by downregulation of CD11b+Gr1+-Myeloid-derived suppressor cells, CD11b+F4/80+CD206+ M2-macrophages and increase in CD11b+F4/80+MHCII+ M1-macrophages. TSA significantly reduced the gene expression of HDAC6, IL-1β, Arginase-1 and iNOS in isolated splenic-MDSCs. FaDu-HTB-43 and NIH3T3 cells stimulated with LPS and 4-NQO exhibit higher IL-1β levels in the supernatant. Interestingly, immunoblot analysis of the cell lysate, we observed that TSA does not alter the expression as well as activation of IL-1β and caspase-1 but the acetylated-tubulin was found to be increased. Nocodazole pre-treatment proved that TSA inhibited the lysosomal exocytosis of IL-1β through tubulin acetylation. In conclusion, HDAC6 inhibitors attenuated TME and cancer progression through the regulation of IL-1β in OSCC.
Collapse
Affiliation(s)
- Ashutosh Mahale
- Metabolic Disorders and Neuroscience Research laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Ganesh Routholla
- Epigenetic Research Laboratory, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - S Lavanya
- Metabolic Disorders and Neuroscience Research laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Pravesh Sharma
- Metabolic Disorders and Neuroscience Research laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Onkar Prakash Kulkarni
- Metabolic Disorders and Neuroscience Research laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India.
| |
Collapse
|
27
|
Hu T, Cheng B, Matsunaga A, Zhang T, Lu X, Fang H, Mori SF, Fang X, Wang G, Xu H, Shi H, Cowell JK. Single-cell analysis defines highly specific leukemia-induced neutrophils and links MMP8 expression to recruitment of tumor associated neutrophils during FGFR1 driven leukemogenesis. Exp Hematol Oncol 2024; 13:49. [PMID: 38730491 PMCID: PMC11084112 DOI: 10.1186/s40164-024-00514-6] [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: 02/14/2024] [Accepted: 04/14/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Leukemias driven by activated, chimeric FGFR1 kinases typically progress to AML which have poor prognosis. Mouse models of this syndrome allow detailed analysis of cellular and molecular changes occurring during leukemogenesis. We have used these models to determine the effects of leukemia development on the immune cell composition in the leukemia microenvironment during leukemia development and progression. METHODS Single cell RNA sequencing (scRNA-Seq) was used to characterize leukemia associated neutrophils and define gene expression changes in these cells during leukemia progression. RESULTS scRNA-Seq revealed six distinct subgroups of neutrophils based on their specific differential gene expression. In response to leukemia development, there is a dramatic increase in only two of the neutrophil subgroups. These two subgroups show specific gene expression signatures consistent with neutrophil precursors which give rise to immature polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Analysis of gene expression in these precursor cells identified pathways that were specifically upregulated, the most pronounced of which involved matrix metalloproteinases Mmp8 and Mmp9, during leukemia progression. Pharmacological inhibition of MMPs using Ilomastat preferentially restricted in vitro migration of neutrophils from leukemic mice and led to a significantly improved survival in vivo, accompanied by impaired PMN-MDSC recruitment. As a result, levels of T-cells were proportionally increased. In clinically annotated TCGA databases, MMP8 was shown to act as an independent indicator for poor prognosis and correlated with higher neutrophil infiltration and poor pan-cancer prognosis. CONCLUSION We have defined specific leukemia responsive neutrophil subgroups based on their unique gene expression profile, which appear to be the precursors of neutrophils specifically associated with leukemia progression. An important event during development of these neutrophils is upregulation MMP genes which facilitated mobilization of these precursors from the BM in response to cancer progression, suggesting a possible therapeutic approach to suppress the development of immune tolerance.
Collapse
Affiliation(s)
- Tianxiang Hu
- Georgia Cancer Center, 1410 Laney Walker Blvd, 30912, Augusta, GA, USA.
| | - Bo Cheng
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Atsuko Matsunaga
- Georgia Cancer Center, 1410 Laney Walker Blvd, 30912, Augusta, GA, USA
| | - Ting Zhang
- Georgia Cancer Center, 1410 Laney Walker Blvd, 30912, Augusta, GA, USA
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Xiaocui Lu
- Georgia Cancer Center, 1410 Laney Walker Blvd, 30912, Augusta, GA, USA
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hui Fang
- Georgia Cancer Center, 1410 Laney Walker Blvd, 30912, Augusta, GA, USA
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Stephanie F Mori
- Georgia Cancer Center, 1410 Laney Walker Blvd, 30912, Augusta, GA, USA
| | - Xuexiu Fang
- Georgia Cancer Center, 1410 Laney Walker Blvd, 30912, Augusta, GA, USA
| | - Gavin Wang
- Georgia Cancer Center, 1410 Laney Walker Blvd, 30912, Augusta, GA, USA
- University of Georgia, Athens, GA, USA
| | - Hongyan Xu
- Department of Biostatistics, Data Science and Epidemiology, School of Public Health, Augusta University, 30912, Augusta, GA, USA
| | - Huidong Shi
- Georgia Cancer Center, 1410 Laney Walker Blvd, 30912, Augusta, GA, USA.
| | - John K Cowell
- Georgia Cancer Center, 1410 Laney Walker Blvd, 30912, Augusta, GA, USA.
| |
Collapse
|
28
|
Yang Y, Chen Y, Liu Z, Chang Z, Sun Z, Zhao L. Concomitant NAFLD Facilitates Liver Metastases and PD-1-Refractory by Recruiting MDSCs via CXCL5/CXCR2 in Colorectal Cancer. Cell Mol Gastroenterol Hepatol 2024; 18:101351. [PMID: 38724007 PMCID: PMC11227024 DOI: 10.1016/j.jcmgh.2024.04.008] [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: 01/12/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND & AIMS Both nonalcoholic fatty liver disease (NAFLD) and colorectal cancer (CRC) are prevalent worldwide. The effects of concomitant NAFLD on the risk of colorectal liver metastasis (CRLM) and its mechanisms have not been definitively elucidated. METHODS We observed the effect of concomitant NAFLD on CRLM in the mouse model and explored the underlying mechanisms of specific myeloid-derived suppressor cells (MDSCs) recruitment and then tested the therapeutic application based on the mechanisms. Finally we validated our findings in the clinical samples. RESULTS Here we prove that in different mouse models, NAFLD induces F4/80+ Kupffer cells to secret chemokine CXCL5 and then recruits CXCR2+ MDSCs to promote the growth of CRLM. CRLM with NAFLD background is refractory to the anti-PD-1 monoclonal antibody treatment, but when combined with Reparixin, an inhibitor of CXCR1/2, dual therapy cures the established CRLM in mice with NAFLD. Our clinical studies also indicate that fatty liver diseases increase the infiltration of CXCR2+ MDSCs, as well as the hazard of liver metastases in CRC patients. CONCLUSIONS Collectively, our findings highlight the significance of selective CXCR2+/CD11b+/Gr-1+ subset myeloid cells in favoring the development of CRLM with NAFLD background and identify a pharmaceutical medicine that is already available for the clinical trials and potential treatment.
Collapse
Affiliation(s)
- Yue Yang
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Huaiyin District, Jinan, China; Shandong First Medical University and Shandong Academy of Medical Sciences, Huaiyin District, Jinan, China
| | - Yunsong Chen
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Huaiyin District, Jinan, China
| | - Zhaogang Liu
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Huaiyin District, Jinan, China
| | - Zhibin Chang
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Huaiyin District, Jinan, China; Shandong First Medical University and Shandong Academy of Medical Sciences, Huaiyin District, Jinan, China
| | - Zhicheng Sun
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Huaiyin District, Jinan, China; Shandong First Medical University and Shandong Academy of Medical Sciences, Huaiyin District, Jinan, China
| | - Lei Zhao
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Huaiyin District, Jinan, China; Shandong First Medical University and Shandong Academy of Medical Sciences, Huaiyin District, Jinan, China.
| |
Collapse
|
29
|
Wieboldt R, Sandholzer M, Carlini E, Lin CW, Börsch A, Zingg A, Lardinois D, Herzig P, Don L, Zippelius A, Läubli H, Mantuano NR. Engagement of sialylated glycans with Siglec receptors on suppressive myeloid cells inhibits anticancer immunity via CCL2. Cell Mol Immunol 2024; 21:495-509. [PMID: 38448555 PMCID: PMC11061307 DOI: 10.1038/s41423-024-01142-0] [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: 08/14/2023] [Accepted: 01/30/2024] [Indexed: 03/08/2024] Open
Abstract
The overexpression of sialic acids on glycans, called hypersialylation, is a common alteration found in cancer cells. Sialylated glycans can enhance immune evasion by interacting with sialic acid-binding immunoglobulin-like lectin (Siglec) receptors on tumor-infiltrating immune cells. Here, we investigated the effect of sialylated glycans and their interaction with Siglec receptors on myeloid-derived suppressor cells (MDSCs). We found that MDSCs derived from the blood of lung cancer patients and tumor-bearing mice strongly express inhibitory Siglec receptors and are highly sialylated. In murine cancer models of emergency myelopoiesis, Siglec-E knockout in myeloid cells resulted in prolonged survival and increased tumor infiltration of activated T cells. Targeting suppressive myeloid cells by blocking Siglec receptors or desialylation strongly reduced their suppressive potential. We further identified CCL2 as a mediator involved in T-cell suppression upon interaction between sialoglycans and Siglec receptors on MDSCs. Our results demonstrated that sialylated glycans inhibit anticancer immunity by modulating CCL2 expression.
Collapse
Affiliation(s)
- Ronja Wieboldt
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Michael Sandholzer
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Emanuele Carlini
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Chia-Wei Lin
- Functional Genomics Center Zurich, ETH Zurich, Zurich, Switzerland
| | - Anastasiya Börsch
- Bioinformatics Core Facility, Department of Biomedicine, University of Basel and Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Andreas Zingg
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Didier Lardinois
- Department of Thoracic Surgery, University Hospital Basel, Basel, Switzerland
| | - Petra Herzig
- Laboratory of Cancer Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Leyla Don
- Laboratory of Cancer Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Alfred Zippelius
- Laboratory of Cancer Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- Division of Oncology, University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland.
- Division of Oncology, University Hospital Basel, Basel, Switzerland.
| | - Natalia Rodrigues Mantuano
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland.
| |
Collapse
|
30
|
Sheikhlary S, Lopez DH, Moghimi S, Sun B. Recent Findings on Therapeutic Cancer Vaccines: An Updated Review. Biomolecules 2024; 14:503. [PMID: 38672519 PMCID: PMC11048403 DOI: 10.3390/biom14040503] [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: 02/23/2024] [Revised: 04/06/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer remains one of the global leading causes of death and various vaccines have been developed over the years against it, including cell-based, nucleic acid-based, and viral-based cancer vaccines. Although many vaccines have been effective in in vivo and clinical studies and some have been FDA-approved, there are major limitations to overcome: (1) developing one universal vaccine for a specific cancer is difficult, as tumors with different antigens are different for different individuals, (2) the tumor antigens may be similar to the body's own antigens, and (3) there is the possibility of cancer recurrence. Therefore, developing personalized cancer vaccines with the ability to distinguish between the tumor and the body's antigens is indispensable. This paper provides a comprehensive review of different types of cancer vaccines and highlights important factors necessary for developing efficient cancer vaccines. Moreover, the application of other technologies in cancer therapy is discussed. Finally, several insights and conclusions are presented, such as the possibility of using cold plasma and cancer stem cells in developing future cancer vaccines, to tackle the major limitations in the cancer vaccine developmental process.
Collapse
Affiliation(s)
- Sara Sheikhlary
- Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ 85721, USA
| | - David Humberto Lopez
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Sophia Moghimi
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Bo Sun
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| |
Collapse
|
31
|
Chen T, Wang M, Chen Y, Liu Y. Current challenges and therapeutic advances of CAR-T cell therapy for solid tumors. Cancer Cell Int 2024; 24:133. [PMID: 38622705 PMCID: PMC11017638 DOI: 10.1186/s12935-024-03315-3] [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: 11/27/2023] [Accepted: 03/26/2024] [Indexed: 04/17/2024] Open
Abstract
The application of chimeric antigen receptor (CAR) T cells in the management of hematological malignancies has emerged as a noteworthy therapeutic breakthrough. Nevertheless, the utilization and effectiveness of CAR-T cell therapy in solid tumors are still limited primarily because of the absence of tumor-specific target antigen, the existence of immunosuppressive tumor microenvironment, restricted T cell invasion and proliferation, and the occurrence of severe toxicity. This review explored the history of CAR-T and its latest advancements in the management of solid tumors. According to recent studies, optimizing the design of CAR-T cells, implementing logic-gated CAR-T cells and refining the delivery methods of therapeutic agents can all enhance the efficacy of CAR-T cell therapy. Furthermore, combination therapy shows promise as a way to improve the effectiveness of CAR-T cell therapy. At present, numerous clinical trials involving CAR-T cells for solid tumors are actively in progress. In conclusion, CAR-T cell therapy has both potential and challenges when it comes to treating solid tumors. As CAR-T cell therapy continues to evolve, further innovations will be devised to surmount the challenges associated with this treatment modality, ultimately leading to enhanced therapeutic response for patients suffered solid tumors.
Collapse
Affiliation(s)
- Tong Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Mingzhao Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yanchao Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
32
|
Pradhan R, Kundu A, Kundu CN. The cytokines in tumor microenvironment: from cancer initiation-elongation-progression to metastatic outgrowth. Crit Rev Oncol Hematol 2024; 196:104311. [PMID: 38442808 DOI: 10.1016/j.critrevonc.2024.104311] [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: 09/16/2023] [Revised: 02/07/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
It is a well-known fact that cancer can be augmented by infections and inflammation. In fact, chronic inflammation establishes a tumor-supporting-microenvironment (TME), which contributes to neoplastic progression. Presently, extensive research is going on to establish the interrelationship between infection, inflammation, immune response, and cancer. Cytokines are the most essential components in this linkage, which are secreted by immune cells and stromal cells of TME. Cytokines have potential involvement in tumor initiation, elongation, progression, metastatic outgrowth, angiogenesis, and development of therapeutic resistance. They are also linked with increased cancer symptoms along with reduced quality of life in advanced cancer patients. The cancer patients experience multiple symptoms including pain, asthenia, fatigue, anorexia, cachexia, and neurodegenerative disorders etc. Anti-cancer therapeutics can be developed by targeting cytokines along with TME to reduce the immunocompromised state and also modulate the TME. This review article depicts the composition and function of different inflammatory cells within the TME, more precisely the role of cytokines in cancer initiation, elongation, and progression as well as the clinical effects in advanced cancer patients. It also provides an overview of different natural compounds, nanoparticles, and chemotherapeutic agents that can target cytokines along with TME, which finally pave the way for cytokines-targeted anti-cancer therapeutics.
Collapse
Affiliation(s)
- Rajalaxmi Pradhan
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India.
| | - Anushka Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India.
| | - Chanakya Nath Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India.
| |
Collapse
|
33
|
Saleh RO, Ibrahim FM, Pallathadka H, Kaur I, Ahmad I, Ali SHJ, Redhee AH, Ghildiyal P, Jawad MA, Alsaadi SB. Nucleic acid vaccines-based therapy for triple-negative breast cancer: A new paradigm in tumor immunotherapy arena. Cell Biochem Funct 2024; 42:e3992. [PMID: 38551221 DOI: 10.1002/cbf.3992] [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: 01/21/2024] [Revised: 03/04/2024] [Accepted: 03/10/2024] [Indexed: 04/02/2024]
Abstract
Nucleic acid vaccines (NAVs) have the potential to be economical, safe, and efficacious. Furthermore, just the chosen antigen in the pathogen is the target of the immune responses brought on by NAVs. Triple-negative breast cancer (TNBC) treatment shows great promise for nucleic acid-based vaccines, such as DNA (as plasmids) and RNA (as messenger RNA [mRNA]). Moreover, cancer vaccines offer a compelling approach that can elicit targeted and long-lasting immune responses against tumor antigens. Bacterial plasmids that encode antigens and immunostimulatory molecules serve as the foundation for DNA vaccines. In the 1990s, plasmid DNA encoding the influenza A nucleoprotein triggered a protective and targeted cytotoxic T lymphocyte (CTL) response, marking the first instance of DNA vaccine-mediated immunity. Similarly, in vitro transcribed mRNA was first successfully used in animals in 1990. At that point, mice were given an injection of the gene encoding the mRNA sequence, and the researchers saw the production of a protein. We begin this review by summarizing our existing knowledge of NAVs. Next, we addressed NAV delivery, emphasizing the need to increase efficacy in TNBC.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | - Fatma M Ibrahim
- Community Health Nursing, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
- Geriatric Nursing, Mansoura University, Mansoura, Egypt
| | | | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Saad Hayif Jasim Ali
- Department of Medical Laboratory, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq
| | - Ahmed Huseen Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Salim B Alsaadi
- Department of Pharmaceutics, Al-Hadi University College, Baghdad, Iraq
| |
Collapse
|
34
|
Yang Z, Liu L, Zhu Z, Hu Z, Liu B, Gong J, Jin Y, Luo J, Deng Y, Jin Y, Wang G, Yin Y. Tumor-Associated Monocytes Reprogram CD8 + T Cells into Central Memory-Like Cells with Potent Antitumor Effects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304501. [PMID: 38386350 DOI: 10.1002/advs.202304501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 01/09/2024] [Indexed: 02/23/2024]
Abstract
CD8+ T cells are critical for host antitumor responses, whereas persistent antigenic stimulation and excessive inflammatory signals lead to T cell dysfunction or exhaustion. Increasing early memory T cells can improve T cell persistence and empower T cell-mediated tumor eradication, especially for adoptive cancer immunotherapy. Here, it is reported that tumor-associated monocytes (TAMos) are highly correlated with the accumulation of CD8+ memory T cells in human cancers. Further analysis identifies that TAMos selectively reprogram CD8+ T cells into T central memory-like (TCM-like) cells with enhanced recall responses. L-NMMA, a pan nitric oxide synthase inhibitor, can mitigate TAMo-mediated inhibition of T cell proliferation without affecting TCM-like cell generation. Moreover, the modified T cells by TAMo exposure and L-NMMA treatment exhibit long-term persistence and elicit superior antitumor effects in vivo. Mechanistically, the transmembrane protein CD300LG is involved in TAMo-mediated TCM-like cell polarization in a cell-cell contact-dependent manner. Thus, the terminally differentiated TAMo subset (CD300LGhighACElow) mainly contributes to TCM-like cell development. Taken together, these findings establish the significance of TAMos in boosting T-cell antitumor immunity.
Collapse
Affiliation(s)
- Zeliang Yang
- Department of Pathology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Liang Liu
- Department of Pathology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Zhenyu Zhu
- Department of Pathology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Zixi Hu
- Department of Pathology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Bowen Liu
- Department of Pathology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Jingjing Gong
- Department of Pathology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Yuan Jin
- Department of Pathology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Juan Luo
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Yichen Deng
- Department of Pathology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Yan Jin
- Department of Pathology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Guangxi Wang
- Department of Pathology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Yuxin Yin
- Department of Pathology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, 100191, China
| |
Collapse
|
35
|
Levstek L, Janžič L, Ihan A, Kopitar AN. Biomarkers for prediction of CAR T therapy outcomes: current and future perspectives. Front Immunol 2024; 15:1378944. [PMID: 38558801 PMCID: PMC10979304 DOI: 10.3389/fimmu.2024.1378944] [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: 01/30/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy holds enormous potential for the treatment of hematologic malignancies. Despite its benefits, it is still used as a second line of therapy, mainly because of its severe side effects and patient unresponsiveness. Numerous researchers worldwide have attempted to identify effective predictive biomarkers for early prediction of treatment outcomes and adverse effects in CAR T cell therapy, albeit so far only with limited success. This review provides a comprehensive overview of the current state of predictive biomarkers. Although existing predictive metrics correlate to some extent with treatment outcomes, they fail to encapsulate the complexity of the immune system dynamics. The aim of this review is to identify six major groups of predictive biomarkers and propose their use in developing improved and efficient prediction models. These groups include changes in mitochondrial dynamics, endothelial activation, central nervous system impairment, immune system markers, extracellular vesicles, and the inhibitory tumor microenvironment. A comprehensive understanding of the multiple factors that influence therapeutic efficacy has the potential to significantly improve the course of CAR T cell therapy and patient care, thereby making this advanced immunotherapy more appealing and the course of therapy more convenient and favorable for patients.
Collapse
Affiliation(s)
| | | | | | - Andreja Nataša Kopitar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
36
|
Arshad J, Rao A, Repp ML, Rao R, Wu C, Merchant JL. Myeloid-Derived Suppressor Cells: Therapeutic Target for Gastrointestinal Cancers. Int J Mol Sci 2024; 25:2985. [PMID: 38474232 PMCID: PMC10931832 DOI: 10.3390/ijms25052985] [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: 01/02/2024] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Gastrointestinal cancers represent one of the more challenging cancers to treat. Current strategies to cure and control gastrointestinal (GI) cancers like surgery, radiation, chemotherapy, and immunotherapy have met with limited success, and research has turned towards further characterizing the tumor microenvironment to develop novel therapeutics. Myeloid-derived suppressor cells (MDSCs) have emerged as crucial drivers of pathogenesis and progression within the tumor microenvironment in GI malignancies. Many MDSCs clinical targets have been defined in preclinical models, that potentially play an integral role in blocking recruitment and expansion, promoting MDSC differentiation into mature myeloid cells, depleting existing MDSCs, altering MDSC metabolic pathways, and directly inhibiting MDSC function. This review article analyzes the role of MDSCs in GI cancers as viable therapeutic targets for gastrointestinal malignancies and reviews the existing clinical trial landscape of recently completed and ongoing clinical studies testing novel therapeutics in GI cancers.
Collapse
Affiliation(s)
- Junaid Arshad
- University of Arizona Cancer Center, GI Medical Oncology, Tucson, AZ 85724, USA;
| | - Amith Rao
- Banner University Medical Center—University of Arizona, Tucson, AZ 85719, USA; (A.R.)
| | - Matthew L. Repp
- College of Medicine, University of Arizona, Tucson, AZ 85719, USA;
| | - Rohit Rao
- University Hospitals Cleveland Medical Center, Case Western Reserve School of Medicine, Cleveland, OH 44106, USA;
| | - Clinton Wu
- Banner University Medical Center—University of Arizona, Tucson, AZ 85719, USA; (A.R.)
| | - Juanita L. Merchant
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| |
Collapse
|
37
|
Natsuki S, Yoshii M, Tanaka H, Mori T, Deguchi S, Miki Y, Tamura T, Toyokawa T, Lee S, Maeda K. Involvement of CX3CR1 + cells appearing in the abdominal cavity in the immunosuppressive environment immediately after gastric cancer surgery. World J Surg Oncol 2024; 22:74. [PMID: 38433196 PMCID: PMC10910822 DOI: 10.1186/s12957-024-03353-1] [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: 11/29/2023] [Accepted: 02/24/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Gastric cancer is primarily treated by surgery; however, little is known about the changes in the intraperitoneal immune environment and the prognostic impact of surgery. Surgical stress and cancer-associated inflammation cause immune cells to mobilize into the abdominal cavity via numerous cytokines. One such cytokine, CX3CR1, has various immune-related functions that remain to be fully explained. We characterized the intraperitoneal immune environment by investigating CX3CR1+ cells in intraperitoneal lavage fluid during gastric cancer surgery. METHODS Lavage fluid samples were obtained from a total of 41 patients who underwent gastrectomy. The relative expression of various genes was analyzed using quantitative real-time PCR. The association of each gene expression with clinicopathological features and surgical outcomes was examined. The fraction of CX3CR1+ cells was analyzed by flow cytometry. Cytokine profiles in lavage fluid samples were investigated using a cytometric beads array. RESULTS CX3CR1high patients exhibited higher levels of perioperative inflammation in blood tests and more recurrences than CX3CR1low patients. CX3CR1high patients tended to exhibit higher pathological T and N stage than CX3CR1low patients. CX3CR1 was primarily expressed on myeloid-derived suppressor cells and tumor-associated macrophages. In particular, polymorphonuclear myeloid-derived suppressor cells were associated with perioperative inflammation, pathological N, and recurrences. These immunosuppressive cells were associated with a trend toward unfavorable prognosis. Moreover, CX3CR1 expression was correlated with programmed death-1 expression. CONCLUSIONS Our results suggest that CX3CR1+ cells are associated with an acute inflammatory response, tumor-promotion, and recurrence. CX3CR1 expression could be taken advantage of as a beneficial therapeutic target for improving immunosuppressive state in the future. In addition, analysis of intra-abdominal CX3CR1+ cells could be useful for characterizing the immune environment after gastric cancer surgery.
Collapse
Affiliation(s)
- Seiji Natsuki
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Mami Yoshii
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan.
| | | | - Takuya Mori
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Sota Deguchi
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Yuichiro Miki
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Tatsuro Tamura
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Takahiro Toyokawa
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Shigeru Lee
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Kiyoshi Maeda
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| |
Collapse
|
38
|
Sugiyama S, Yumimoto K, Fujinuma S, Nakayama KI. Identification of effective CCR2 inhibitors for cancer therapy using humanized mice. J Biochem 2024; 175:195-204. [PMID: 37947138 DOI: 10.1093/jb/mvad086] [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: 10/22/2023] [Revised: 10/28/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
C-C chemokine receptor type 2 (CCR2) is the receptor for C-C motif chemokine 2 (CCL2) and is associated with various inflammatory diseases and cancer metastasis. Although many inhibitors for CCR2 have been developed, it remains unresolved which inhibitors are the most effective in the clinical setting. In the present study, we compared 10 existing human CCR2 antagonists in a calcium influx assay using human monocytic leukemia cells. Among them, MK0812 was found to be the most potent inhibitor of human CCR2. Furthermore, we generated a human CCR2B knock-in mouse model to test the efficacy of MK0812 against a lung metastasis model of breast cancer. Oral administration of MK0812 to humanized mice did indeed reduce the number of monocytic myeloid-derived suppressor cells and the rate of lung metastasis. These results suggest that MK0812 is the most promising candidate among the commercially available CCR2 inhibitors. We propose that combining these two screening methods may provide an excellent experimental method for identifying effective drugs that inhibit human CCR2.
Collapse
Affiliation(s)
- Shigeaki Sugiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Kanae Yumimoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Shun Fujinuma
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
- Anticancer Strategies Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
39
|
Krizova L, Benesova I, Zemanova P, Spacek J, Strizova Z, Humlova Z, Mikulova V, Petruzelka L, Vocka M. Immunophenotyping of peripheral blood in NSCLC patients discriminates responders to immune checkpoint inhibitors. J Cancer Res Clin Oncol 2024; 150:99. [PMID: 38383923 PMCID: PMC10881622 DOI: 10.1007/s00432-024-05628-2] [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: 11/30/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) dramatically changed the prognosis of patients with NSCLC. Unfortunately, a reliable predictive biomarker is still missing. Commonly used biomarkers, such as PD-L1, MSI, or TMB, are not quite accurate in predicting ICI efficacy. METHODS In this prospective observational cohort study, we investigated the predictive role of erythrocytes, thrombocytes, innate and adaptive immune cells, complement proteins (C3, C4), and cytokines from peripheral blood of 224 patients with stage III/IV NSCLC treated with ICI alone (pembrolizumab, nivolumab, and atezolizumab) or in combination (nivolumab + ipilimumab) with chemotherapy. These values were analyzed for associations with the response to the treatment and survival endpoints. RESULTS Higher baseline Tregs, MPV, hemoglobin, and lower monocyte levels were associated with favorable PFS and OS. Moreover, increased baseline basophils and lower levels of C3 predicted significantly improved PFS. The levels of the baseline immature granulocytes, C3, and monocytes were significantly associated with the occurrence of partial regression at the first restaging. Multiple studied parameters (n = 9) were related to PFS benefit at the time of first restaging as compared to baseline values. In addition, PFS nonbenefit group showed a decrease in lymphocyte count after three months of therapy. The OS benefit was associated with higher levels of lymphocytes, erythrocytes, hemoglobin, MCV, and MPV, and a lower value of NLR after three months of treatment. CONCLUSION Our work suggests that parameters from peripheral venous blood may be potential biomarkers in NSCLC patients on ICI. The baseline values of Tregs, C3, monocytes, and MPV are especially recommended for further investigation.
Collapse
Affiliation(s)
- Ludmila Krizova
- Department of Oncology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 499/2, 128 00, Prague 2, Czech Republic
| | - Iva Benesova
- Department of Immunology, Second Faculty of Medicine, Charles University in Prague and University Hospital in Motol, Prague, Czech Republic
| | - Petra Zemanova
- Department of Oncology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 499/2, 128 00, Prague 2, Czech Republic
| | - Jan Spacek
- Department of Oncology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 499/2, 128 00, Prague 2, Czech Republic
| | - Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University in Prague and University Hospital in Motol, Prague, Czech Republic
| | - Zuzana Humlova
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Veronika Mikulova
- Institute of Medical Biochemistry and Laboratory Diagnostics, Laboratory of Clinical Immunology and Allergology, General University Hospital in Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lubos Petruzelka
- Department of Oncology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 499/2, 128 00, Prague 2, Czech Republic
| | - Michal Vocka
- Department of Oncology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 499/2, 128 00, Prague 2, Czech Republic.
| |
Collapse
|
40
|
Lacinski RA, Dziadowicz SA, Stewart A, Chaharbakhshi E, Akhter H, Pisquiy JJ, Victory JH, Hardham JB, Chew C, Prorock A, Bao Y, Sol-Church K, Hobbs GR, Klein E, Nalesnik MA, Hu G, de Oliveira A, Santiago SP, Lindsey BA. Nanosphere pharmacodynamics improves safety of immunostimulatory cytokine therapy. iScience 2024; 27:108836. [PMID: 38303687 PMCID: PMC10831265 DOI: 10.1016/j.isci.2024.108836] [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: 09/26/2023] [Revised: 11/04/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Systemic administration of interleukin (IL)-12 induces potent anti-tumor immune responses in preclinical cancer models through the systemic activation of effector immune cells and release of proinflammatory cytokines. IL-12-loaded PLGA nanospheres (IL12ns) are hypothesized to improve therapeutic efficacy and thwart unwanted side effects observed in previous human clinical trials. Through the investigation of peripheral blood and local tissue immune responses in healthy BALB/c mice, the immune-protective pharmacodynamics of IL12ns were suggested. Nanospheres increased pro-inflammatory plasma cytokines/chemokines (IFN-γ, IL-6, TNF-α, and CXCL10) without inducing maladaptive transcriptomic signatures in circulating peripheral immune cells. Gene expression profiling revealed activation of pro-inflammatory signaling pathways in systemic tissues, the likely source of these effector cytokines. These data support that nanosphere pharmacodynamics, including shielding IL-12 from circulating immune cells, depositing peripherally in systemic immune tissues, and then slowly eluting bioactive cytokine, thereafter, are essential to safe immunostimulatory therapy.
Collapse
Affiliation(s)
- Ryan A. Lacinski
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Sebastian A. Dziadowicz
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Amanda Stewart
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Edwin Chaharbakhshi
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Halima Akhter
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - John J. Pisquiy
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Jack H. Victory
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Joshua B. Hardham
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Claude Chew
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alyson Prorock
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Yongde Bao
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Katia Sol-Church
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Gerald R. Hobbs
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Edwin Klein
- Division of Laboratory Animal Resources, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Michael A. Nalesnik
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, USA
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Ana de Oliveira
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Stell P. Santiago
- Department of Pathology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Brock A. Lindsey
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
41
|
Jiang Y, Qiao S, Li L, Zhu X. Combination of radiotherapy and Anlotinib enhances benefit from immunotherapy to liver metastasis and abscopal tumor from lung cancer. Int Immunopharmacol 2024; 128:111441. [PMID: 38171056 DOI: 10.1016/j.intimp.2023.111441] [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: 12/01/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
Many studies have shown that liver metastasis can weaken the efficacy of immunotherapy. Immunotherapy combined with radiotherapy or anti-angiogenic therapy has been proven to have synergistic anti-tumor effects. So we devote to explore whether the combination of the three therapies can exert effective anti-tumor effects on liver metastasis. The clinical information of 118 patients with liver metastasis were collected to compare the intrahepatic progression-free survival between immunotherapy and immunotherapy combined with other treatments. We used Lewis lung cancer (LLC) cell to establish a mouse liver metastasis tumor model and record tumor burden and survival. Tumor-infiltrating immune cells detected by flow cytometry. RNA sequencing was performed and the proportion of immune cells were analyzed by TIMER2.0 database. Compared with immunotherapy group, the combination therapy group showed a trend for longer median intrahepatic progression-free survival. Radiotherapy combined with PD-1 inhibitor and Anlotinib can inhibit liver metastasis and subcutaneous tumor growth and prolong the survival compared with other groups in vivo. Compared with the anti-PD-1 treatment group, triple therapy can increase CD4+T, CD8+T, and IFN-γ+CD8+T cells and decrease infiltration of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) in tumors. PPAR signaling pathway were significantly activated and CD8+T and dendritic cells (DC) were increased in the triple therapy group compared to the PD-1 inhibitor combined with Anlotinib group. Radiotherapy combined with PD-1 inhibitor and Anlotinib can effectively exert anti-tumor efficacy and reshape the tumor immune microenvironment by increasing the infiltration of anti-tumor immune cells and reducing the infiltration of immunosuppressive immune cells.
Collapse
Affiliation(s)
- Yuhang Jiang
- Southern Medical University, Guangzhou 510280, China
| | - Simiao Qiao
- Southern Medical University, Guangzhou 510280, China
| | - Luyao Li
- Southern Medical University, Guangzhou 510280, China
| | - Xiaoxia Zhu
- Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
42
|
Farahmandnejad M, Mosaddeghi P, Dorvash M, Sakhteman A, Negahdaripour M, Faridi P. Correlation of Myeloid-Derived Suppressor Cell Expansion with Upregulated Transposable Elements in Severe COVID-19 Unveiled in Single-Cell RNA Sequencing Reanalysis. Biomedicines 2024; 12:315. [PMID: 38397917 PMCID: PMC10887269 DOI: 10.3390/biomedicines12020315] [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: 12/07/2023] [Revised: 01/14/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Some studies have investigated the potential role of transposable elements (TEs) in COVID-19 pathogenesis and complications. However, to the best of our knowledge, there is no study to examine the possible association of TE expression in cell functions and its potential role in COVID-19 immune response at the single-cell level. In this study, we reanalyzed single-cell RNA seq data of bronchoalveolar lavage (BAL) samples obtained from six severe COVID-19 patients and three healthy donors to assess the probable correlation of TE expression with the immune responses induced by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in COVID-19 patients. Our findings indicate that the expansion of myeloid-derived suppressor cells (MDSCs) may be a characteristic feature of COVID-19. Additionally, a significant increase in TE expression in MDSCs was observed. This upregulation of TEs in COVID-19 may be linked to the adaptability of these cells in response to their microenvironments. Furthermore, it appears that the identification of overexpressed TEs by pattern recognition receptors (PRRs) in MDSCs may enhance the suppressive capacity of these cells. Thus, this study emphasizes the crucial role of TEs in the functionality of MDSCs during COVID-19.
Collapse
Affiliation(s)
- Mitra Farahmandnejad
- Quality Control of Drug Products Department, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran;
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Pouria Mosaddeghi
- Medicinal Plants Processing Research Center, School of Pharmacy, Shiraz University of Medical Science, Shiraz 71348-14336, Iran;
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Mohammadreza Dorvash
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia;
| | - Amirhossein Sakhteman
- Proteomics and Bioanalytics, Department of Molecular Life Sciences, School of Life Sciences, Technical University of Munich, 80333 Munich, Germany;
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Pouya Faridi
- Monash Proteomics and Metabolomics Platform, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC 3800, Australia
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
| |
Collapse
|
43
|
Rong N, Wei X, Liu J. The Role of Neutrophil in COVID-19: Positive or Negative. J Innate Immun 2024; 16:80-95. [PMID: 38224674 PMCID: PMC10861219 DOI: 10.1159/000535541] [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: 09/16/2023] [Accepted: 11/27/2023] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND Neutrophils are the first line of defense against pathogens. They are divided into multiple subpopulations during development and kill pathogens through various mechanisms. Neutrophils are considered one of the markers of severe COVID-19. SUMMARY In-depth research has revealed that neutrophil subpopulations have multiple complex functions. Different subsets of neutrophils play an important role in the progression of COVID-19. KEY MESSAGES In this review, we provide a detailed overview of the developmental processes of neutrophils at different stages and their recruitment and activation after SARS-CoV-2 infection, aiming to elucidate the changes in neutrophil subpopulations, characteristics, and functions after infection and provide a reference for mechanistic research on neutrophil subpopulations in the context of SARS-CoV-2 infection. In addition, we have also summarized research progress on potential targeted drugs for neutrophil immunotherapy, hoping to provide information that aids the development of therapeutic drugs for the clinical treatment of critically ill COVID-19 patients.
Collapse
Affiliation(s)
- Na Rong
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China,
| | - Xiaohui Wei
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Chen J, Madina BR, Ahmadi E, Yarovinsky TO, Krady MM, Meehan EV, Wang IC, Ye X, Pitmon E, Ma XY, Almassian B, Nakaar V, Wang K. Cancer immunotherapy with enveloped self-amplifying mRNA CARG-2020 that modulates IL-12, IL-17 and PD-L1 pathways to prevent tumor recurrence. Acta Pharm Sin B 2024; 14:335-349. [PMID: 38261838 PMCID: PMC10792965 DOI: 10.1016/j.apsb.2023.08.034] [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: 05/31/2023] [Revised: 08/01/2023] [Accepted: 08/15/2023] [Indexed: 01/25/2024] Open
Abstract
Targeting multiple immune mechanisms may overcome therapy resistance and further improve cancer immunotherapy for humans. Here, we describe the application of virus-like vesicles (VLV) for delivery of three immunomodulators alone and in combination, as a promising approach for cancer immunotherapy. VLV vectors were designed to deliver single chain interleukin (IL)-12, short-hairpin RNA (shRNA) targeting programmed death ligand 1 (PD-L1), and a dominant-negative form of IL-17 receptor A (dn-IL17RA) as a single payload or as a combination payload. Intralesional delivery of the VLV vector expressing IL-12 alone, as well as the trivalent vector (designated CARG-2020) eradicated large established tumors. However, only CARG-2020 prevented tumor recurrence and provided long-term survival benefit to the tumor-bearing mice, indicating a benefit of the combined immunomodulation. The abscopal effects of CARG-2020 on the non-injected contralateral tumors, as well as protection from the tumor cell re-challenge, suggest immune-mediated mechanism of protection and establishment of immunological memory. Mechanistically, CARG-2020 potently activates Th1 immune mechanisms and inhibits expression of genes related to T cell exhaustion and cancer-promoting inflammation. The ability of CARG-2020 to prevent tumor recurrence and to provide survival benefit makes it a promising candidate for its development for human cancer immunotherapy.
Collapse
Affiliation(s)
- Ju Chen
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan Hospital of Traditional Chinese Medicine, Foshan 528000, China
| | | | - Elham Ahmadi
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- CaroGen Corporation, Farmington, CT 06030, USA
| | | | | | - Eileen Victoria Meehan
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Isabella China Wang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- The Loomis Chaffee School, Windsor, CT 06095, USA
| | - Xiaoyang Ye
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Elise Pitmon
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | - Kepeng Wang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
45
|
Okamoto M, Yamamoto M. TCR Signals Controlling Adaptive Immunity against Toxoplasma and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1444:177-193. [PMID: 38467980 DOI: 10.1007/978-981-99-9781-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
T cells play a crucial role in adaptive immunity by recognizing and eliminating foreign pathogens and abnormal cells such as cancer cells. T cell receptor (TCR), which is expressed on the surface of T cells, recognizes and binds to specific antigens presented by major histocompatibility complex (MHC) molecules on antigen-presenting cells (APCs). This activation process leads to the proliferation and differentiation of T cells, allowing them to carry out their specific immune response functions. This chapter outlines the TCR signaling pathways that are common to different T cell subsets, as well as the recently elucidated TCR signaling pathway specific to CD8+ T cells and its role in controlling anti-Toxoplasma and anti-tumor immunity.
Collapse
Affiliation(s)
- Masaaki Okamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
- Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.
| |
Collapse
|
46
|
Xie Y, Jiang H. The exploration of mitochondrial-related features helps to reveal the prognosis and immunotherapy methods of colorectal cancer. Cancer Rep (Hoboken) 2024; 7:e1914. [PMID: 37903487 PMCID: PMC10809275 DOI: 10.1002/cnr2.1914] [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: 07/06/2023] [Revised: 09/01/2023] [Accepted: 09/29/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Cancer cell survival, proliferation, and metabolism are all intertwined with mitochondria. However, a complete description of how the features of mitochondria relate to the tumor microenvironment (TME) and immunological landscape of colorectal cancer (CRC) has yet to be made. We performed subgroup analysis on CRC patient data obtained from the databases using non-negative matrix factorization (NMF) clustering. Construct a prognostic model using the mitochondrial-related gene (MRG) risk score, and then compare it to other models for accuracy. Comprehensive analyses of the risk score, in conjunction with the TME and immune landscape, were performed, and the relationship between the model and different types of cell death, radiation and chemotherapy, and drug resistance was investigated. Results from immunohistochemistry and single-cell sequencing were utilized to verify the model genes, and a drug sensitivity analysis was conducted to evaluate possible therapeutic medicines. The pan-cancer analysis is utilized to further investigate the role of genes in a wider range of malignancies. METHODS AND RESULTS We found that CRC patients based on MRG were divided into two groups with significant differences in survival outcomes and TME between groups. The predictive power of the risk score was further shown by building a prognostic model and testing it extensively in both internal and external cohorts. Multiple immune therapeutic responses and the expression of immunological checkpoints demonstrate that the risk score is connected to immunotherapy success. The correlation analysis of the risk score provide more ideas and guidance for prognostic models in clinical treatment. CONCLUSION The TME, immune cell infiltration, and responsiveness to immunotherapy in CRC were all thoroughly evaluated on the basis of MRG features. The comparative validation of multiple queues and models combined with clinical data ensures the effectiveness and clinical practicality of MRG features. Our studies help clinicians create individualized treatment programs for individuals with cancer.
Collapse
Affiliation(s)
- Yun‐hui Xie
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of ChengduAffiliated Hospital of Southwest Jiaotong UniversityChengduChina
| | - Hui‐zhong Jiang
- College of GraduateGuizhou University of Traditional Chinese MedicineGuiyangChina
| |
Collapse
|
47
|
Donkor M, Choe JY, Reid DM, Fiadjoe HK, Quinn B, Ranjan A, Pulse M, Chaudhary P, Basha R, Jones HP. Surgical Primary Tumor Resection Reduces Accumulation of CD11b + Myeloid Cells in the Lungs Augmenting the Efficacy of an Intranasal Cancer Vaccination against Secondary Lung Metastasis. Pharmaceuticals (Basel) 2023; 17:51. [PMID: 38256885 PMCID: PMC10821475 DOI: 10.3390/ph17010051] [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: 12/06/2023] [Revised: 12/23/2023] [Accepted: 12/24/2023] [Indexed: 01/24/2024] Open
Abstract
A hallmark of effective cancer treatment is the prevention of tumor reoccurrence and metastasis to distal organs, which are responsible for most cancer deaths. However, primary tumor resection is expected to be curative as most solid tumors have been shown both experimentally and clinically to accelerate metastasis to distal organs including the lungs. In this study, we evaluated the efficacy of our engineered nasal nano-vaccine (CpG-NP-Tag) in reducing accelerated lung metastasis resulting from primary tumor resection. Cytosine-phosphate-guanine oligonucleotide [CpG ODN]-conjugated nanoparticle [NP] encapsulating tumor antigen [Tag] (CpG-NP-Tag) was manufactured and tested in vivo using a syngeneic mouse mammary tumor model following intranasal delivery. We found that our nasal nano-vaccine (CpG-NP-Tag), compared to control NPs administered after primary mammary tumor resection, significantly reduced lung metastasis in female BALB/c mice subjected to surgery (surgery mice). An evaluation of vaccine efficacy in both surgery and non-surgery mice revealed that primary tumor resection reduces CD11b+ monocyte-derived suppressor-like cell accumulation in the lungs, allowing increased infiltration of vaccine-elicited T cells (IFN-γ CD8+ T cells) in the lungs of surgery mice compared to non-surgery mice. These findings suggest that the combination of the target delivery of a nasal vaccine in conjunction with the standard surgery of primary tumors is a plausible adjunctive treatment against the establishment of lung metastasis.
Collapse
Affiliation(s)
- Michael Donkor
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center, Fort Worth, TX 76107, USA (D.M.R.); (H.K.F.); (A.R.); (P.C.)
| | - Jamie Y. Choe
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center, Fort Worth, TX 76107, USA (D.M.R.); (H.K.F.); (A.R.); (P.C.)
| | - Danielle Marie Reid
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center, Fort Worth, TX 76107, USA (D.M.R.); (H.K.F.); (A.R.); (P.C.)
| | - Hope K. Fiadjoe
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center, Fort Worth, TX 76107, USA (D.M.R.); (H.K.F.); (A.R.); (P.C.)
| | - Byron Quinn
- Department of Biology, Langston University, Langston, OK 73050, USA
| | - Amalendu Ranjan
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center, Fort Worth, TX 76107, USA (D.M.R.); (H.K.F.); (A.R.); (P.C.)
| | - Mark Pulse
- Department of Pharmaceutical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA;
| | - Pankaj Chaudhary
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center, Fort Worth, TX 76107, USA (D.M.R.); (H.K.F.); (A.R.); (P.C.)
| | - Riyaz Basha
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center, Fort Worth, TX 76107, USA (D.M.R.); (H.K.F.); (A.R.); (P.C.)
| | - Harlan P. Jones
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center, Fort Worth, TX 76107, USA (D.M.R.); (H.K.F.); (A.R.); (P.C.)
- Institute for Health Disparities UNTHC 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| |
Collapse
|
48
|
Imodoye SO, Adedokun KA. EMT-induced immune evasion: connecting the dots from mechanisms to therapy. Clin Exp Med 2023; 23:4265-4287. [PMID: 37966552 DOI: 10.1007/s10238-023-01229-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a dynamic program crucial for organismal development and tissue regeneration. Unfortunately, this program is often hijacked by epithelial tumors to facilitate metastasis. Beyond its role in cancer spread, EMT increases cancer cell survival by activating stem cell programs and bypassing apoptotic programs. Importantly, the capacity of EMT to enforce tumor progression by altering the tumor cell phenotype without triggering immune responses opens the intriguing possibility of a mechanistic link between EMT-driven cancers and immune evasion. Indeed, EMT has been acknowledged as a of driver immune evasion, but the mechanisms are still evolving. Here, we review recent insights into the influence of EMT on tumor immune evasion. Specifically, we focus on the mechanistic roles of EMT in immune escape as the basis that may provide a platform for innovative therapeutic approaches in advanced tumors. We summarize promising therapeutic approaches currently in clinical trials and trending preclinical studies aimed at reinvigorating the tumor microenvironment to create immune-permissive conditions that facilitates immune-mediated tumor clearance. We anticipate that this will assist researchers and pharmaceutical companies in understanding how EMT compromises the immune response, potentially paving the way for effective cancer therapies.
Collapse
Affiliation(s)
- Sikiru O Imodoye
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, USA.
| | - Kamoru A Adedokun
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| |
Collapse
|
49
|
CHEN QIUQIANG, GUO XUEJUN, MA WENXUE. Opportunities and challenges of CD47-targeted therapy in cancer immunotherapy. Oncol Res 2023; 32:49-60. [PMID: 38188674 PMCID: PMC10767231 DOI: 10.32604/or.2023.042383] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/09/2023] [Indexed: 01/09/2024] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy for the treatment of cancer, with the tumor microenvironment (TME) playing a pivotal role in modulating the immune response. CD47, a cell surface protein, has been identified as a crucial regulator of the TME and a potential therapeutic target for cancer therapy. However, the precise functions and implications of CD47 in the TME during immunotherapy for cancer patients remain incompletely understood. This comprehensive review aims to provide an overview of CD47's multifaced role in TME regulation and immune evasion, elucidating its impact on various types of immunotherapy outcomes, including checkpoint inhibitors and CAR T-cell therapy. Notably, CD47-targeted therapies offer a promising avenue for improving cancer treatment outcomes, especially when combined with other immunotherapeutic approaches. The review also discusses current and potential CD47-targeted therapies being explored for cancer treatment and delves into the associated challenges and opportunities inherent in targeting CD47. Despite the demonstrated effectiveness of CD47-targeted therapies, there are potential problems, including unintended effects on healthy cells, hematological toxicities, and the development if resistance. Consequently, further research efforts are warranted to fully understand the underlying mechanisms of resistance and to optimize CD47-targeted therapies through innovative combination approaches, ultimately improving cancer treatment outcomes. Overall, this comprehensive review highlights the significance of CD47 as a promising target for cancer immunotherapy and provides valuable insight into the challenges and opportunities in developing effective CD47-targeted therapies for cancer treatment.
Collapse
Affiliation(s)
- QIUQIANG CHEN
- Key Laboratory for Translational Medicine, The First Affiliated Hospital, Huzhou University School of Medicine, Huzhou, 313000, China
| | - XUEJUN GUO
- Department of Hematology, Puyang Youtian General Hospital, Puyang, 457001, China
| | - WENXUE MA
- Department of Medicine, Moores Cancer Center, Sanford Stem Cell Institute, University of California San Diego, La Jolla, San Diego, 92093, USA
| |
Collapse
|
50
|
DeRyckere D, Huelse JM, Earp HS, Graham DK. TAM family kinases as therapeutic targets at the interface of cancer and immunity. Nat Rev Clin Oncol 2023; 20:755-779. [PMID: 37667010 DOI: 10.1038/s41571-023-00813-7] [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] [Accepted: 08/07/2023] [Indexed: 09/06/2023]
Abstract
Novel treatment approaches are needed to overcome innate and acquired mechanisms of resistance to current anticancer therapies in cancer cells and the tumour immune microenvironment. The TAM (TYRO3, AXL and MERTK) family receptor tyrosine kinases (RTKs) are potential therapeutic targets in a wide range of cancers. In cancer cells, TAM RTKs activate signalling pathways that promote cell survival, metastasis and resistance to a variety of chemotherapeutic agents and targeted therapies. TAM RTKs also function in innate immune cells, contributing to various mechanisms that suppress antitumour immunity and promote resistance to immune-checkpoint inhibitors. Therefore, TAM antagonists provide an unprecedented opportunity for both direct and immune-mediated therapeutic activity provided by inhibition of a single target, and are likely to be particularly effective when used in combination with other cancer therapies. To exploit this potential, a variety of agents have been designed to selectively target TAM RTKs, many of which have now entered clinical testing. This Review provides an essential guide to the TAM RTKs for clinicians, including an overview of the rationale for therapeutic targeting of TAM RTKs in cancer cells and the tumour immune microenvironment, a description of the current preclinical and clinical experience with TAM inhibitors, and a perspective on strategies for continued development of TAM-targeted agents for oncology applications.
Collapse
Affiliation(s)
- Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Paediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Justus M Huelse
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Paediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - H Shelton Earp
- Department of Medicine, UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Douglas K Graham
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA.
- Department of Paediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|