1
|
Gan M, Liu N, Li W, Chen M, Bai Z, Liu D, Liu S. Metabolic targeting of regulatory T cells in oral squamous cell carcinoma: new horizons in immunotherapy. Mol Cancer 2024; 23:273. [PMID: 39696340 DOI: 10.1186/s12943-024-02193-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a prevalent oral malignancy, which poses significant health risks with a high mortality rate. Regulatory T cells (Tregs), characterized by their immunosuppressive capabilities, are intricately linked to OSCC progression and patient outcomes. The metabolic reprogramming of Tregs within the OSCC tumor microenvironment (TME) underpins their function, with key pathways such as the tryptophan-kynurenine-aryl hydrocarbon receptor, PI3K-Akt-mTOR and nucleotide metabolism significantly contributing to their suppressive activities. Targeting these metabolic pathways offers a novel therapeutic approach to reduce Treg-mediated immunosuppression and enhance anti-tumor responses. This review explores the metabolic dependencies and pathways that sustain Treg function in OSCC, highlighting key metabolic adaptations such as glycolysis, fatty acid oxidation, amino acid metabolism and PI3K-Akt-mTOR signaling pathway that enable Tregs to thrive in the challenging conditions of the TME. Additionally, the review discusses the influence of the oral microbiome on Treg metabolism and evaluates potential therapeutic strategies targeting these metabolic pathways. Despite the promising potential of these interventions, challenges such as selectivity, toxicity, tumor heterogeneity, and resistance mechanisms remain. The review concludes with perspectives on personalized medicine and integrative approaches, emphasizing the need for continued research to translate these findings into effective clinical applications for OSCC treatment.
Collapse
Affiliation(s)
- Menglai Gan
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Nanshu Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Wenting Li
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Mingwei Chen
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Zhongyu Bai
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Dongjuan Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China.
| | - Sai Liu
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China.
| |
Collapse
|
2
|
Repas J, Frlic T, Snedec T, Kopitar AN, Sourij H, Janež A, Pavlin M. Physiologically Achievable Concentration of 2-Deoxy-D-Glucose Stimulates IFN-γ Secretion in Activated T Cells In Vitro. Int J Mol Sci 2024; 25:10384. [PMID: 39408714 PMCID: PMC11476708 DOI: 10.3390/ijms251910384] [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: 07/25/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
2-deoxy-D-glucose (2DG) is a glycolysis and protein N-glycosylation inhibitor with promising anti-tumor and immunomodulatory effects. However, 2DG can also suppress T cell function, including IFN-γ secretion. Few human T cell studies have studied low-dose 2DG, which can increase IFN-γ in a Jurkat clone. We therefore investigated 2DG's effect on IFN-γ in activated human T cells from PBMCs, with 2DG treatment commenced either concurrently with activation or 48 h after activation. Concurrent 2DG treatment decreased IFN-γ secretion in a dose-dependent manner. However, 2DG treatment of pre-activated T cells had a hormetic effect on IFN-γ, with 0.15-0.6 mM 2DG (achievable in vivo) increasing and >2.4 mM 2DG reducing its secretion. In contrast, IL-2 levels declined monotonously with increasing 2DG concentration. Lower 2DG concentrations reduced PD-1 and increased CD69 expression regardless of treatment timing. The absence of increased T-bet or Eomes expression or IFNG transcription suggests another downstream mechanism. 2DG dose-dependently induced the unfolded protein response, suggesting a possible role in increased IFN-γ secretion, possibly by increasing the ER folding capacity for IFN-γ via increased chaperone expression. Overall, low-dose, short-term 2DG exposure could potentially improve the T cell anti-tumor response.
Collapse
Affiliation(s)
- Jernej Repas
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
| | - Tjaša Frlic
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
| | - Tadeja Snedec
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
| | - Andreja Nataša Kopitar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Harald Sourij
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Medical University Graz, 8010 Graz, Austria;
| | - Andrej Janež
- Clinical Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia;
| | - Mojca Pavlin
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
- Group for Nano- and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
3
|
Yu R, Zhao F, Xu Z, Zhang G, Du B, Shu Q. Current status and future of cancer vaccines: A bibliographic study. Heliyon 2024; 10:e24404. [PMID: 38293405 PMCID: PMC10826732 DOI: 10.1016/j.heliyon.2024.e24404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 11/24/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
Background Cancer vaccines are an important component of tumour immunotherapy. An increasing number of studies have shown that cancer vaccines have considerable clinical benefits. With the development of tumour precision medicine, cancer vaccines have become important because of their individualised targeting effects. However, few bibliometric studies have conducted comprehensive systematic reviews in this field. This study aimed to assess the scientific output and trends in cancer vaccine research from a global perspective. Methods We collected publications on cancer vaccines from the Web of Science Core Collection database, which was limited to articles and reviews in English. Microsoft Excel, VOS Viewer, and CiteSpace V were used for quantitative and visual analyses. Results A total of 7807 articles were included. From 1991 to 2022, the number of publications increased annually. The United States had the highest number of articles published in this field (48.28 %), the highest citation frequency (183,964 times), and the highest H-index (182). The National Institutes of Health topped the list with 476 articles. Schlom J had the highest number of published articles (128) and was the main investigator in this field. The journal, Cancer Immunology Immunotherapy, had published the highest number of articles in related fields. In recent years, tumour microenvironment, immune checkpoint inhibitors, particle vaccines, tumour antigens, and dendritic cells have become research hotspots related to cancer vaccines. Conclusion Cancer vaccines are a popular research topic in the field of tumour immunotherapy. Related research and publications will enter a boom stage. "Immune checkpoint inhibitors", "tumour microenvironment" and "dendritic cells" may become future research hotspots, while "T-cell suppressor" is a potential puzzle to be solved.
Collapse
Affiliation(s)
- Rui Yu
- The First School of Clinical Medical, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangmin Zhao
- The First School of Clinical Medical, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zeting Xu
- The First School of Clinical Medical, Zhejiang Chinese Medical University, Hangzhou, China
| | - Gaochenxi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Bingqing Du
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qijin Shu
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
4
|
Wang J, Zhao X, Wan YY. Intricacies of TGF-β signaling in Treg and Th17 cell biology. Cell Mol Immunol 2023; 20:1002-1022. [PMID: 37217798 PMCID: PMC10468540 DOI: 10.1038/s41423-023-01036-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Balanced immunity is pivotal for health and homeostasis. CD4+ helper T (Th) cells are central to the balance between immune tolerance and immune rejection. Th cells adopt distinct functions to maintain tolerance and clear pathogens. Dysregulation of Th cell function often leads to maladies, including autoimmunity, inflammatory disease, cancer, and infection. Regulatory T (Treg) and Th17 cells are critical Th cell types involved in immune tolerance, homeostasis, pathogenicity, and pathogen clearance. It is therefore critical to understand how Treg and Th17 cells are regulated in health and disease. Cytokines are instrumental in directing Treg and Th17 cell function. The evolutionarily conserved TGF-β (transforming growth factor-β) cytokine superfamily is of particular interest because it is central to the biology of both Treg cells that are predominantly immunosuppressive and Th17 cells that can be proinflammatory, pathogenic, and immune regulatory. How TGF-β superfamily members and their intricate signaling pathways regulate Treg and Th17 cell function is a question that has been intensely investigated for two decades. Here, we introduce the fundamental biology of TGF-β superfamily signaling, Treg cells, and Th17 cells and discuss in detail how the TGF-β superfamily contributes to Treg and Th17 cell biology through complex yet ordered and cooperative signaling networks.
Collapse
Affiliation(s)
- Junying Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xingqi Zhao
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yisong Y Wan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
5
|
Gaballa MR, Banerjee P, Milton DR, Jiang X, Ganesh C, Khazal S, Nandivada V, Islam S, Kaplan M, Daher M, Basar R, Alousi A, Mehta R, Alatrash G, Khouri I, Oran B, Marin D, Popat U, Olson A, Tewari P, Jain N, Jabbour E, Ravandi F, Kantarjian H, Chen K, Champlin R, Shpall E, Rezvani K, Kebriaei P. Blinatumomab maintenance after allogeneic hematopoietic cell transplantation for B-lineage acute lymphoblastic leukemia. Blood 2022; 139:1908-1919. [PMID: 34914826 PMCID: PMC8952188 DOI: 10.1182/blood.2021013290] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/29/2021] [Indexed: 11/20/2022] Open
Abstract
Patients with B-lineage acute lymphoblastic leukemia (ALL) are at high-risk for relapse after allogeneic hematopoietic cell transplantation (HCT). We conducted a single-center phase 2 study evaluating the feasibility of 4 cycles of blinatumomab administered every 3 months during the first year after HCT in an effort to mitigate relapse in high-risk ALL patients. Twenty-one of 23 enrolled patients received at least 1 cycle of blinatumomab and were included in the analysis. The median time from HCT to the first cycle of blinatumomab was 78 days (range, 44 to 105). Twelve patients (57%) completed all 4 treatment cycles. Neutropenia was the only grade 4 adverse event (19%). Rates of cytokine release (5% G1) and neurotoxicity (5% G2) were minimal. The cumulative incidence of acute graft-versus-host disease (GVHD) grades 2 to 4 and 3 to 4 were 33% and 5%, respectively; 2 cases of mild (10%) and 1 case of moderate (5%) chronic GVHD were noted. With a median follow-up of 14.3 months, the 1-year overall survival (OS), progression-free survival (PFS), and nonrelapse mortality (NRM) rates were 85%, 71%, and 0%, respectively. In a matched analysis with a contemporary cohort of 57 patients, we found no significant difference between groups regarding blinatumomab's efficacy. Correlative studies of baseline and posttreatment samples identified patients with specific T-cell profiles as "responders" or "nonresponders" to therapy. Responders had higher proportions of effector memory CD8 T-cell subsets. Nonresponders were T-cell deficient and expressed more inhibitory checkpoint molecules, including T-cell immunoglobulin and mucin domain 3 (TIM3). We found that blinatumomab postallogeneic HCT is feasible, and its benefit is dependent on the immune milieu at time of treatment. This paper is posted on ClinicalTrials.gov, study ID: NCT02807883.
Collapse
Affiliation(s)
- Mahmoud R Gaballa
- Bone Marrow Transplant and Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Pinaki Banerjee
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | | | - Xianli Jiang
- Department of Bioinformatics & Computational Biology; and
| | - Christina Ganesh
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Sajad Khazal
- Department of Pediatric Stem Cell Transplantation & Cellular Therapy and
| | | | - Sanjida Islam
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Mecit Kaplan
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - May Daher
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Rafet Basar
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Amin Alousi
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Rohtesh Mehta
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Gheath Alatrash
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Issa Khouri
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Betul Oran
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - David Marin
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Uday Popat
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Amanda Olson
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Priti Tewari
- Department of Pediatric Stem Cell Transplantation & Cellular Therapy and
| | - Nitin Jain
- Department of Leukemia, MD Anderson Cancer Center, University of Texas, Houston, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, MD Anderson Cancer Center, University of Texas, Houston, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, MD Anderson Cancer Center, University of Texas, Houston, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, MD Anderson Cancer Center, University of Texas, Houston, Houston, TX, USA
| | - Ken Chen
- Department of Bioinformatics & Computational Biology; and
| | - Richard Champlin
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| | - Partow Kebriaei
- Department of Stem Cell Transplantation & Cellular Therapy, Houston
| |
Collapse
|
6
|
Emerging Bone Marrow Microenvironment-Driven Mechanisms of Drug Resistance in Acute Myeloid Leukemia: Tangle or Chance? Cancers (Basel) 2021; 13:cancers13215319. [PMID: 34771483 PMCID: PMC8582363 DOI: 10.3390/cancers13215319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Despite high rates of remission obtained with conventional chemotherapy, the persistence of leukemic cells after treatments, eventually exiting in disease relapse, remains the main challenge in acute myeloid leukemia (AML). Increasing evidence indicates that, besides AML cell mutations, stromal and immune cells, as leukemic microenvironment components, may protect AML cells from therapies. Here, we will recapitulate emerging bone marrow (BM) microenvironment-dependent mechanisms of therapy resistance. The understanding of these processes will help find new drug combinations and conceive novel and more effective treatments. Abstract Acute myeloid leukemia (AML) has been considered for a long time exclusively driven by critical mutations in hematopoietic stem cells. Recently, the contribution of further players, such as stromal and immune bone marrow (BM) microenvironment components, to AML onset and progression has been pointed out. In particular, mesenchymal stromal cells (MSCs) steadily remodel the leukemic niche, not only favoring leukemic cell growth and development but also tuning their responsiveness to treatments. The list of mechanisms driven by MSCs to promote a leukemia drug-resistant phenotype has progressively expanded. Moreover, the relative proportion and the activation status of immune cells in the BM leukemic microenvironment may vary by influencing their reactivity against leukemic cells. In that, the capacity of the stroma to re-program immune cells, thus promoting and/or hampering therapeutic efficacy, is emerging as a crucial aspect in AML biology, adding an extra layer of complexity. Current treatments for AML have mainly focused on eradicating leukemia cells, with little consideration for the leukemia-damaged BM niche. Increasing evidence on the contribution of stromal and immune cells in response to therapy underscores the need to hold the mutual interplay, which takes place in the BM. A careful dissection of these interactions will help provide novel applications for drugs already under experimentation and open a wide array of opportunities for new drug discovery.
Collapse
|
7
|
Hossain SM, Lynch-Sutherland CF, Chatterjee A, Macaulay EC, Eccles MR. Can Immune Suppression and Epigenome Regulation in Placenta Offer Novel Insights into Cancer Immune Evasion and Immunotherapy Resistance? EPIGENOMES 2021; 5:16. [PMID: 34968365 PMCID: PMC8594685 DOI: 10.3390/epigenomes5030016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/18/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is the second leading cause of mortality and morbidity in the developed world. Cancer progression involves genetic and epigenetic alterations, accompanied by aggressive changes, such as increased immune evasion, onset of metastasis, and drug resistance. Similar to cancer, DNA hypomethylation, immune suppression, and invasive cell behaviours are also observed in the human placenta. Mechanisms that lead to the acquisition of invasive behaviour, immune evasion, and drug and immunotherapy resistance are presently under intense investigations to improve patient outcomes. Here, we review current knowledge regarding the similarities between immune suppression and epigenome regulation, including the expression of repetitive elements (REs), endogenous retroviruses (ERVs) and transposable elements (TEs) in cells of the placenta and in cancer, which are associated with changes in immune regulation and invasiveness. We explore whether immune suppression and epigenome regulation in placenta offers novel insights into immunotherapy resistance in cancer, and we also discuss the implications and the knowledge gaps relevant to these findings, which are rapidly being accrued in these quite disparate research fields. Finally, we discuss potential linkages between TE, ERV and RE activation and expression, regarding mechanisms of immune regulation in placenta and cancer. A greater understanding of the role of immune suppression and associated epigenome regulation in placenta could help to elucidate some comparable mechanisms operating in cancer, and identify potential new therapeutic targets for treating cancer.
Collapse
Affiliation(s)
- Sultana Mehbuba Hossain
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand; (S.M.H.); (C.F.L.-S.); (A.C.); (E.C.M.)
| | - Chiemi F. Lynch-Sutherland
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand; (S.M.H.); (C.F.L.-S.); (A.C.); (E.C.M.)
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand; (S.M.H.); (C.F.L.-S.); (A.C.); (E.C.M.)
- Maurice Wilkins Centre for Molecular Biodiscovery, Level 2, 3A Symonds Street, Auckland 1010, New Zealand
| | - Erin C. Macaulay
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand; (S.M.H.); (C.F.L.-S.); (A.C.); (E.C.M.)
| | - Michael R. Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand; (S.M.H.); (C.F.L.-S.); (A.C.); (E.C.M.)
- Maurice Wilkins Centre for Molecular Biodiscovery, Level 2, 3A Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
8
|
Host immune system modulation in Ph+ acute lymphoblastic leukemia patients treated with dasatinib and blinatumomab. Blood 2021; 138:2290-2293. [PMID: 34297829 DOI: 10.1182/blood.2021011822] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/04/2021] [Indexed: 11/20/2022] Open
|
9
|
Rajaratinam H, Rasudin NS, Al Astani TAD, Mokhtar NF, Yahya MM, Wan Zain WZ, Asma-Abdullah N, Mohd Fuad WE. Breast cancer therapy affects the expression of antineonatal Nav1.5 antibodies in the serum of patients with breast cancer. Oncol Lett 2021; 21:108. [PMID: 33376541 PMCID: PMC7751336 DOI: 10.3892/ol.2020.12369] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/05/2020] [Indexed: 11/06/2022] Open
Abstract
Neonatal Nav1.5 (nNav1.5) is the alternative splice variant of Nav1.5 and it has been widely associated with the progression of breast cancer. The immunological context of nNav1.5 with respect to breast cancer metastases remains unexplored. The presence of antibodies against nNav1.5 may highlight the immunogenicity of nNav1.5. Hence, the aim of the present study was to detect the presence of antineonatal Nav1.5 antibodies (antinNav1.5-Ab) in the serum of patients with breast cancer and to elucidate the effects of breast cancer therapy on its expression. A total of 32 healthy female volunteers and 64 patients with breast cancer were randomly recruited into the present study as the control and breast cancer group, respectively. Patients with breast cancer were divided equally based on their pre- and ongoing-treatment status. Serum samples were tested with in-house indirect enzyme-linked immunosorbent assay (ELISA) to detect antinNav1.5-Ab, CD25 (T regulatory cell marker) using an ELISA kit and Luminex assay to detect the expression of metastasis-associated cytokines, such as vascular endothelial growth factor (VEGF), interleukin (IL)-6, IL-10, IL-8, chemokine (C-C motif) ligand 2 and tumor necrosis factor-alpha (TNF-α) The mean difference in the expression of antinNav1.5-Ab among the three groups (control, pretreatment and ongoing-treatment) was significant (P=0.0005) and the pretreatment breast cancer group exhibited the highest expression. The concentration of CD25 was highest in the pretreatment breast cancer group compared with the control and ongoing-treatment groups. There was a significant positive correlation between antinNav1.5-Ab and IL-6 in the pretreatment group (r=0.7260; P=0.0210) and a significant negative correlation between antinNav1.5-Ab and VEGF in the ongoing-treatment group (r=-0.842; P-value=0.0040). The high expression of antinNav1.5-Ab in the pretreatment group was in accordance with the uninterrupted presence of metastasis and highlighted the immunogenicity of nNav1.5 whereas the low expression of antinNav1.5-Ab in the ongoing-treatment group reflected the efficacy of breast cancer therapy in eliminating metastases. The augmented manifestation of T regulatory cells in the pretreatment group highlighted the functional role of nNav1.5 in promoting metastasis. The parallel expression of antinNav1.5-Ab with the imbalanced expression of cytokines promoting metastasis (IL-8, IL-6 and TNF-α) and cytokines that prevent metastasis (IL-10) indicated the role of nNav1.5 in breast cancer growth. The expression of antinNav1.5-Ab in accordance to the metastatic microenvironment indicates the immunogenicity of the protein and highlights the influence of breast cancer therapy on its expression level.
Collapse
Affiliation(s)
- Harishini Rajaratinam
- School of Health Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Nur Syahmina Rasudin
- School of Health Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Tengku Ahmad Damitri Al Astani
- Department of Chemical Pathology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
- Breast Cancer Awareness and Research (BestARi) Unit, Hospital Universiti Sains Malaysia (HUSM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Noor Fatmawati Mokhtar
- Institute for Research in Molecular Medicine (INFORMM), Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Maya Mazuwin Yahya
- Department of Surgery, School of Medical Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Wan Zainira Wan Zain
- Department of Surgery, School of Medical Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Nurul Asma-Abdullah
- School of Health Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Wan Ezumi Mohd Fuad
- School of Health Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| |
Collapse
|
10
|
Bayati F, Mohammadi M, Valadi M, Jamshidi S, Foma AM, Sharif-Paghaleh E. The Therapeutic Potential of Regulatory T Cells: Challenges and Opportunities. Front Immunol 2021; 11:585819. [PMID: 33519807 PMCID: PMC7844143 DOI: 10.3389/fimmu.2020.585819] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/27/2020] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cells (Tregs) are an immunosuppressive subgroup of CD4+ T cells which are identified by the expression of forkhead box protein P3 (Foxp3). The modulation capacity of these immune cells holds an important role in both transplantation and the development of autoimmune diseases. These cells are the main mediators of self-tolerance and are essential for avoiding excessive immune reactions. Tregs play a key role in the induction of peripheral tolerance that can prevent autoimmunity, by protecting self-reactive lymphocytes from the immune reaction. In contrast to autoimmune responses, tumor cells exploit Tregs in order to prevent immune cell recognition and anti-tumor immune response during the carcinogenesis process. Recently, numerous studies have focused on unraveling the biological functions and principles of Tregs and their primary suppressive mechanisms. Due to the promising and outstanding results, Tregs have been widely investigated as an alternative tool in preventing graft rejection and treating autoimmune diseases. On the other hand, targeting Tregs for the purpose of improving cancer immunotherapy is being intensively evaluated as a desirable and effective method. The purpose of this review is to point out the characteristic function and therapeutic potential of Tregs in regulatory immune mechanisms in transplantation tolerance, autoimmune diseases, cancer therapy, and also to discuss that how the manipulation of these mechanisms may increase the therapeutic options.
Collapse
Affiliation(s)
- Fatemeh Bayati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research & Development Department, Aryogen Pharmed, Karaj, Iran
| | - Mahsa Mohammadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Maryam Valadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeid Jamshidi
- Research & Development Department, Aryogen Pharmed, Karaj, Iran
| | - Arron Munggela Foma
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Sharif-Paghaleh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
11
|
Dwarakanath BS, Farooque A, Gupta S. Targeting regulatory T cells for improving cancer therapy: Challenges and prospects. Cancer Rep (Hoboken) 2018; 1:e21105. [PMID: 32729245 DOI: 10.1002/cnr2.1105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/22/2018] [Accepted: 04/07/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Regulatory T cells (Tregs) play a central role in immune responses to infectious agents and tumors. Paradoxically, Tregs protect self-cells from the immune response as a part of peripheral tolerance and prevents autoimmune disorders, whereas during the process of carcinogenesis, they are exploited by tumor cells for protection against antitumor immune responses. Therefore, Tregs are often considered as a major obstacle in anticancer therapy. The objective of this review is to provide a current understanding on Tregs as a potential cellular target for achieving therapeutic gain and discuss various approaches that are implicated at preclinical and clinical scenario. RECENT FINDINGS Several approaches like immunotherapy and adjuvant chemotherapy, which reduce Tregs population, have been found to be useful in improving local tumor control. Our recent observations with the glycolytic inhibitor, 2-deoxy-D-glucose, established as an adjuvant in radiotherapy and chemotherapy of tumors also show that potential of 2-deoxy-D-glucose to improve local tumor control is linked with its ability to reduce the Tregs pool. CONCLUSIONS Several published studies and emerging evidences indicate that suppression of Treg numbers, infiltration into the tumors, and function can improve the cancer therapy by enhancing the antitumor immunity.
Collapse
Affiliation(s)
| | | | - Seema Gupta
- Department of Oncology, Georgetown University, Washington, DC, USA
| |
Collapse
|