2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Clinical Use of Propranolol Reduces Biomarkers of Proliferation in Gastric Cancer

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Gastric cancer has one of the highest mortality rate in the world, but the treatment is still limited. Building on previous studies, mechanistic studies on propranolol in gastric cancer mice models and gastric cancer patients were performed. Propranolol inhibited the in vitro proliferation of gastric cancer cells in a time- and concentration-dependent manner. Consistent findings were observed in MFC tumors engrafted 615 mice, which were treated with propranolol at 10 mg/kg daily for 14 days. Propranolol inhibited the phosphorylation of AKT, MEK, and ERK proteins than control in mice tumor tissues respectively (p-AKT 26.16 vs. 56.82, P = 0.0196, p-MEK 28.27 vs. 59.28, P = 0.1102, p-ERK 48.2 vs. 107.4, P = 0.0062). Propranolol had antiproliferative activity in gastric cancer patients receiving 60 mg daily for 7 days prior to surgery(ki67 44.8 vs 125.3 for placebo; P = 0.02). Phosphorylated AKT, MEK, and ERK did not differ between propranolol and placebo treatment in gastric cancer patients. The expression of molecules on CD8 + T cells was not changed both in mice model and patients nor was there a statistically significant difference in CD8 + T cell subsets in patients, although suggestion of an effect was evident. These results prove that propranolol may inhibit the growth of gastric cancer in mice model and patients and the possible mechanism was via inhibiting the AKT and MAPK pathways, but the frequency of tumor infiltration CD8 + T cells did not increase significantly.

          Related collections

          Most cited references20

          • Record: found
          • Abstract: found
          • Article: not found

          Acetylcholine-synthesizing T cells relay neural signals in a vagus nerve circuit.

          Neural circuits regulate cytokine production to prevent potentially damaging inflammation. A prototypical vagus nerve circuit, the inflammatory reflex, inhibits tumor necrosis factor-α production in spleen by a mechanism requiring acetylcholine signaling through the α7 nicotinic acetylcholine receptor expressed on cytokine-producing macrophages. Nerve fibers in spleen lack the enzymatic machinery necessary for acetylcholine production; therefore, how does this neural circuit terminate in cholinergic signaling? We identified an acetylcholine-producing, memory phenotype T cell population in mice that is integral to the inflammatory reflex. These acetylcholine-producing T cells are required for inhibition of cytokine production by vagus nerve stimulation. Thus, action potentials originating in the vagus nerve regulate T cells, which in turn produce the neurotransmitter, acetylcholine, required to control innate immune responses.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Propranolol for infantile haemangiomas: insights into the molecular mechanisms of action.

            Infantile haemangiomas (IH) are the most common benign tumours of infancy. Although most IH are innocuous and 85-90% regress spontaneously, some may become life- or function-threatening and require immediate treatment. Previous standard therapeutic options include physical measures (laser surgery, cryosurgery) and systemic corticosteroids, in severe cases also vincristine, alpha-interferon or cyclophosphamide, all bearing the risk of serious side-effects. Oral propranolol is a very recent therapeutic option for complicated IH with impressive efficacy and generally good tolerance. The effects of propranolol on IH were discovered by chance, and very little is known about its mechanisms of action in IH. Here we present a summary of current knowledge of how propranolol interferes with endothelial cells, vascular tone, angiogenesis and apoptosis. Early, intermediate and long-term effects of propranolol on IH can be attributed to three different pharmacological targets. Early effects (brightening of the haemangioma surface within 1-3 days after start of therapy) are attributable to vasoconstriction due to decreased release of nitric oxide. Intermediate effects are due to the blocking of proangiogenic signals (vascular endothelial growth factor, basic fibroblast growth factor, matrix metalloproteinase 2/9) and result in growth arrest. Long-term effects of propranolol are characterized by induction of apoptosis in proliferating endothelial cells, and result in tumour regression.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              β-Adrenergic Signaling in Mice Housed at Standard Temperatures Suppresses an Effector Phenotype in CD8 + T Cells and Undermines Checkpoint Inhibitor Therapy

              The immune context of tumors has significant prognostic value and is predictive of responsiveness to several forms of therapy, including immunotherapy. We report here that CD8+ T-cell frequency and functional orientation within the tumor microenvironment is regulated by β2-adrenergic receptor (β-AR) signaling in host immune cells. We used three strategies-physiologic (manipulation of ambient thermal environment), pharmacologic (β-blockers), and genetic (β2-AR knockout mice) to reduce adrenergic stress signaling in two widely studied preclinical mouse tumor models. Reducing β-AR signaling facilitated conversion of tumors to an immunologically active tumor microenvironment with increased intratumoral frequency of CD8+ T cells with an effector phenotype and decreased expression of programmed death receptor-1 (PD-1), in addition to an elevated effector CD8+ T-cell to CD4+ regulatory T-cell ratio (IFNγ+CD8+:Treg). Moreover, this conversion significantly increased the efficacy of anti-PD-1 checkpoint blockade. These data highlight the potential of adrenergic stress and norepinephrine-driven β-AR signaling to regulate the immune status of the tumor microenvironment and support the strategic use of clinically available β-blockers in patients to improve responses to immunotherapy. Cancer Res; 77(20); 5639-51. ©2017 AACR.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Oncol
                Front Oncol
                Front. Oncol.
                Frontiers in Oncology
                Frontiers Media S.A.
                2234-943X
                26 April 2021
                2021
                : 11
                : 628613
                Affiliations
                [1] 1 Department of Clinical Pharmacology, Xiangya Hospital, Central South University , Changsha, China
                [2] 2 Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics , Changsha, China
                [3] 3 National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University , Changsha, China
                [4] 4 Department of Pharmacy, Affiliated Hospital of Nantong University , Nantong, China
                [5] 5 Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University , Changsha, China
                [6] 6 USF Taneja College of Pharmacy , Tampa, FL, United States
                Author notes

                Edited by: Rui Liao, First Affiliated Hospital of Chongqing Medical University, China

                Reviewed by: Ram Mohan Ram Kumar, Rajiv Gandhi Centre for Biotechnology, India; Dong-Dong Wu, Henan University, China

                *Correspondence: Yijing He, heyijing@ 123456csu.edu.cn ; Howard L. McLeod, hmcleod1965@ 123456gmail.com

                This article was submitted to Gastrointestinal Cancers, a section of the journal Frontiers in Oncology

                †These authors have contributed equally to this work

                Article
                10.3389/fonc.2021.628613
                8108985
                33981600
                50407986-eff7-41a3-bd1a-a73d9871f0ca
                Copyright © 2021 Hu, Liao, Li, Hu, Chen, Zhang, Wang, Chen, Song, Liu, Zhang, Li, McLeod and He

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 12 November 2020
                : 29 March 2021
                Page count
                Figures: 5, Tables: 5, Equations: 0, References: 20, Pages: 9, Words: 3509
                Funding
                Funded by: Foundation for Innovative Research Groups of the National Natural Science Foundation of China 10.13039/501100012659
                Categories
                Oncology
                Original Research

                Oncology & Radiotherapy
                gastric,propranolol,anti-proliferation,cd8+ t cell,immunity,akt/mapk
                Oncology & Radiotherapy
                gastric, propranolol, anti-proliferation, cd8+ t cell, immunity, akt/mapk

                Comments

                Comment on this article