scholarly journals A new application of mTOR inhibitor drugs as potential therapeutic agents for COVID-19

Author(s):  
Habibeh Mashayekhi-Sardoo ◽  
Hesamoddin Hosseinjani

Abstract Since December 2019, the COVID-19 emerging pandemic caused by SARS-CoV-2 has resulted in one of the most important global health threats. Concerning the absence of an approved effective vaccine or drug for the treatment and outcome improvement of COVID-19 patients, and the role of SARS-CoV-2 in activation of mammalian target of rapamycin (mTOR) pathway, we decided to review the previous data regarding the therapeutic effect of mTOR inhibitor drugs in COVID-19 patients. We searched the scientific databases such as Web of Science, Embase, Medline (PubMed), Scopus, and Google Scholar using appropriate keywords to find suitable studies or suggestions until October 2020. The findings of the current study confirmed that mTOR inhibitor drugs through suggested mechanisms such as T cell adjustment, induction of autophagy without apoptosis, reduction of viral replication, restoration of the T-cell function, decrease cytokine storm, and moderation of the mTOR–PI3K–AKT pathway activation bring about a therapeutic impact in COVID-19 patients. Taken together, it is necessary to find a suitable therapy for the COVID-19 pandemic emerging. In this regard, we clarify that it is valuable to consider the therapeutic effect of mTOR inhibitor drugs and metformin by its mTOR inhibition property in the treatment of COVID-19 patients.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2025-2025
Author(s):  
Francesca Chiarini ◽  
Cecilia Grimaldi ◽  
Francesca Ricci ◽  
Pierluigi Tazzari ◽  
Camilla Evangelisti ◽  
...  

Abstract Abstract 2025 Poster Board II-2 Introduction: Recent findings have highlighted that constitutively active phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian Target of Rapamycin (mTOR) signaling is a common feature of T-cell acute lymphoblastic leukemia (T-ALL) where it strongly influences cell proliferation and survival. Pathway activation could be due to several reasons which include Notch1 activation leading to HES1-mediated transcriptional suppression of PTEN gene, PTEN phosphorylation or oxidation, and inactivation of SHIP1 phosphatase. These findings lend compelling weight for the application of PI3K/Akt/mTOR inhibitors in T-ALL. Rapamycin and its analogues have shown some promising effects in pre-clinical models of T-ALL. However, mTOR inhibitors are mainly cytostatic and could hyperactivate Akt due to the existence of feedback loops between mTOR, p70 S6 kinase, PI3K, and Akt. Recently, dual PI3K/mTOR inhibitors have been synthesized. Here, we have analyzed the therapeutic potential of the novel, dual PI3K/mTOR inhibitor, NVP-BEZ235, an orally bioavailable imidazoquinoline derivative, which has entered clinical trials for solid tumors, on both T-ALL cell lines and patient samples. Methods and Patients: We employed a panel of cell lines with up-regulated PI3K/Akt/mTOR signaling, including CEM-R cells [which overexpress high levels of the membrane transporter, 170-kDa P-glycoprotein (P-gp)], MOLT-4 and CEM-S cells (which lack PTEN expression), Jurkat cells (which do not express both PTEN and SHIP1), and RPMI-8402 and BE-13 cells. MOLT-4, CEM, and Jurkat cells have a non-functional p53 pathway. Moreover, both Jurkat and MOLT-4 cells have aberrant Notch1 signaling. Patients samples displayed pathway activation as documented by increased levels of p-Akt, p-4E-BP1, and p-S6 ribosomal protein, as well as low/absent PTEN expression. Results: NVP-BEZ235 was cytotoxic to the panel of cell lines as documented by MTT assays. NVP-BEZ235 IC50 ranged from 80 to 280 nM at 24 h. A comparison between NVP-BEZ235 and the dual PI3K/mTOR inhibitor PI-103, a small synthetic molecule of the pyridofuropyrimidine class with the same targets, demonstrated that NVP-BEZ235 was more effective than PI-103 when employed at equimolar concentrations. NVP-BEZ235 did not significantly affect the proliferation of peripheral blood T-lymphocytes from healthy donors stimulated with phytohemagglutinin and interleukin-2, whereas it blocked leukemic cells in the G1 phase of the cell cycle, and this was accompanied by decreased levels of phosphorylated Retinoblastoma protein. NVP-BEZ235 treatment also resulted in apoptotic cell death (about 20-30% at 6 h of exposure, when employed at 200 nM), as documented by Annexin V/propidium iodide staining and cytofluorimetric analysis. Moreover, NVP-BEZ235 activated caspase-8 and caspase-3, as demonstrated by western blot. Western blot documented a dose- and time-dependent dephosphorylation of Akt and its downstream target, GSK-3β, in response to NVP-BEZ235. mTOR downstream targets were also efficiently dephosphorylated, including p70S6 kinase, S6 ribosomal protein, and 4E-BP1. Remarkably, NVP-BEZ235 targeted the side population (SP, identified by Hoechst 33342 staining and ABCG2 expression) of T-ALL cell lines, which might correspond to leukemia initiating cells, and synergized with several chemotherapeutic agents (dexamethasone, vincristine, cyclophosphamide, Ara-C) currently employed for treating T-ALL patients. NVP-BEZ235 reduced chemoresistance to vincristine induced in Jurkat cells by co-culturing with MS-5 stromal cells which mimic the bone marrow microenvironment. NVP-BEZ235 was cytotoxic (IC50: 10-15 nM at 96 h) to primary lymphoblasts from patients with T-ALL, where the drug dephosphorylated 4E-BP1, at variance with rapamycin. Of note, NVP-BEZ235 targeted the SP also in T-ALL patient samples. Conclusions: NVP-BEZ235 was cytotoxic to T-ALL cell lines and patient lymphoblasts (including SP cells) at concentrations that have been previously reported to be achievable in vivo. Taken together, our findings indicate that longitudinal inhibition at two nodes of the PI3K/Akt/mTOR network with NVP-BEZ235, either alone or in combination with other drugs, may serve as an efficient treatment towards T-ALL cells (including those overexpressing P-gp and independently from p53 status) which require upregulation of this signaling pathway for their survival and growth. Disclosures: No relevant conflicts of interest to declare.


2012 ◽  
Vol 23 (15) ◽  
pp. 2963-2972 ◽  
Author(s):  
Yuning Zhou ◽  
Qingding Wang ◽  
Zheng Guo ◽  
Heidi L. Weiss ◽  
B. Mark Evers

The nuclear factor of activated T-cell (NFAT) proteins are a family of transcription factors (NFATc1–c4) involved in the regulation of cell differentiation. We identified REDD1, a negative regulator of mammalian target of rapamycin (mTOR) through the tuberous sclerosis complex (TSC1/2 complex), as a new molecular target of NFATc3. We show that treatment with a combination of phorbol 12-myristate 13-acetate (PMA) plus ionophore A23187 (Io), which induces NFAT activation, increased REDD1 mRNA and protein expression and inhibited mTOR signaling; pretreatment with the calcineurin inhibitor cyclosporin A (CsA), an antagonist of NFAT signaling, decreased REDD1 induction and mTOR inhibition. Knockdown of NFATc3, not NFATc1, NFATc2, or NFATc4, attenuated PMA/Io-induced REDD1 expression. Treatment with PMA/Io increased REDD1 promoter activity and increased NFATc3 binding to the REDD1 promoter. Overexpression of NFATc3 increased REDD1 mRNA and protein expression and increased PMA/Io-mediated REDD1 promoter activity. Treatment with PMA/Io increased expression of the goblet cell differentiation marker MUC2; these changes were attenuated by pretreatment with CsA or knockdown of REDD1 or NFATc3. Overexpression of NFATc3 increased, while knockdown of TSC2 decreased, MUC2 expression. We provide evidence showing NFATc3 inhibits mTOR via induction of REDD1. Our results suggest a role for the NFATc3/REDD1/TSC2 axis in the regulation of intestinal cell differentiation.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 674-674 ◽  
Author(s):  
Greg M Delgoffe ◽  
Thomas P. Kole ◽  
Robert J Cotter ◽  
Jonathan Powell

Abstract Full T cell activation requires antigen recognition (Signal 1) in the context of costimulation (Signal 2). Our group and others have determined that the mammalian Target of Rapamycin (mTOR) plays an important role in integrating costimulatory signals. Specifically, Th1 T cell activation in the absence of mTOR activation leads to tolerance in the form of T cell anergy. mTOR is an evolutionarily-conserved serine/threonine kinase which has been shown to interpret environmental cues in mammalian cells. mTOR is activated by an array of diverse inputs including insulin, amino acids, growth factors and CD28. mTOR signals through two signaling complexes: TORC1 and TORC2. A critical component of TORC1 signaling is the Regulatory Associated Protein of TOR (raptor). One of the central questions in understanding mTOR function is determining how diverse upstream signals can lead to diverse downstream functional consequences. To address this issue in T cells we undertook a proteomic approach to identify novel binding proteins for raptor. Jurkat T cells were either incubated in serum-free media or hyper-activated. Raptor was immunoprecipitated (IP) from the two lysates, separated by SDS-PAGE and silver stained. Next, protein bands that were differentially bound to raptor in the lysates from stimulated versus unstimulated cells were identified. One band located near 90 kDa was excised and using mass spectrometry subsequently determined to be Hsp90, a chaperone protein necessary for the correct folding of many protein “clients”. These findings were next confirmed in primary T cells. Upon activation with anti-CD3 and anti-CD28, IP of raptor led to co-IP of Hsp90 and IP of Hsp90 led to the concomitant precipitation of raptor. To further determine the role of the Hsp90-raptor interaction in T cells we used 17-AAG, an Hsp90 inhibitor. Primary T cells were stimulated in the presence of rapamycin or 17-AAG. Incubation with 17-AAG but not rapamycin led to a decrease in raptor protein levels consistent with the concept that raptor is an Hsp90 client. Functionally, this led to a decrease in TORC1 activation as measured by phosphorylation of S6K-1. More importantly, although17-AAG did not inhibit IL-2 production upon initial stimulation, primary T cells stimulated with anti-CD3 and anti-CD28 in the presence of 17-AAG failed to produce IL-2 upon rechallenge 5 days later; they were anergic. Overall, our findings demonstrate a novel activation-induced interaction between Hsp90 and raptor in T cells. This interaction can regulate the decision between TCR-induced activation and tolerance. Hsp90 inhibitors are currently being evaluated as anti-neoplastic agents, and while such agents do not acutely inhibit T cell function, they may induce anergy in activated Th1 cells. Thus, Hsp90 inhibitors might be incorporated into novel immunosuppressive regimens to treat and prevent GVHD and transplant rejection through the promotion of T cell tolerance.


2005 ◽  
Vol 201 (10) ◽  
pp. 1523-1530 ◽  
Author(s):  
Peter S. Heeger ◽  
Peter N. Lalli ◽  
Feng Lin ◽  
Anna Valujskikh ◽  
Jinbo Liu ◽  
...  

Decay-accelerating factor (Daf) dissociates C3/C5 convertases that assemble on host cells and thereby prevents complement activation on their surfaces. We demonstrate that during primary T cell activation, the absence of Daf on antigen-presenting cells (APCs) and on T cells enhances T cell proliferation and augments the induced frequency of effector cells. The effect is factor D- and, at least in part, C5-dependent, indicating that local alternative pathway activation is essential. We show that cognate T cell–APC interactions are accompanied by rapid production of alternative pathway components and down-regulation of Daf expression. The findings argue that local alternative pathway activation and surface Daf protein function respectively as a costimulator and a negative modulator of T cell immunity and explain previously reported observations linking complement to T cell function. The results could have broad therapeutic implications for disorders in which T cell immunity is important.


2015 ◽  
Vol 43 (4) ◽  
pp. 734-739 ◽  
Author(s):  
Sian M Henson

Aging is accompanied by immune decline leading to increased incidence of infections and malignancies, given the demographic shift of humans towards an older age the identification of strategies for the manipulation of immunity is an important goal. Evidence implicates mammalian target of rapamycin (mTOR) to be a key modulator of aging and the use of mTOR inhibitors has been shown to ameliorate much age-related pathology; however, recent data suggest that senescent CD8+ T-cells function independently of mTOR. This review article will challenge the perceived dogma that mTOR universally controls CD8+ T-cell function.


Gut ◽  
2021 ◽  
pp. gutjnl-2020-322660
Author(s):  
Xiaochuan Dong ◽  
Shumei Song ◽  
Yuan Li ◽  
Yibo Fan ◽  
Lulu Wang ◽  
...  

BackgroundGastric adenocarcinoma (GAC) is a lethal disease with limited therapeutic options. Genetic alterations in chromatin remodelling gene AT-rich interactive domain 1A (ARID1A) and mTOR pathway activation occur frequently in GAC. Targeting the mechanistic target of rapamycin (mTOR) pathway in unselected patients has failed to show survival benefit. A deeper understanding of GAC might identify a subset that can benefit from mTOR inhibition.MethodsGenomic alterations in ARID1A were analysed in GAC. Mouse gastric epithelial cells from CK19-Cre-Arid1Afl/fl and wild-type mice were used to determine the activation of oncogenic genes due to loss of Arid1A. Functional studies were performed to determine the significance of loss of ARID1A and the sensitivity of ARID1A-deficient cancer cells to mTOR inhibition in GAC.ResultsMore than 30% of GAC cases had alterations (mutations or deletions) of ARID1A and ARID1A expression was negatively associated with phosphorylation of S6 and SOX9 in GAC tissues and patient-derived xenografts (PDXs). Activation of mTOR signalling (increased pS6) and SOX9 nuclear expression were strongly increased in Arid1A−/− mouse gastric tissues which could be curtailed by RAD001, an mTOR inhibitor. Knockdown of ARID1A in GAC cell lines increased pS6 and nuclear SOX9 and increased sensitivity to an mTOR inhibitor which was further amplified by its combination with fluorouracil both in vitro and in vivo in PDXs.ConclusionsThe loss of ARID1A activates pS6 and SOX9 in GAC, which can be effectively targeted by an mTOR inhibitor. Therefore, our studies suggest a new therapeutic strategy of clinically targeting the mTOR pathway in patients with GAC with ARID1A deficiency.


2016 ◽  
Vol 2016 ◽  
pp. 1-11 ◽  
Author(s):  
Hallvard Holdaas ◽  
Paolo De Simone ◽  
Andreas Zuckermann

Malignancy after solid organ transplantation remains a major cause of posttransplant mortality. The mammalian target of rapamycin (mTOR) inhibitor class of immunosuppressants exerts various antioncogenic effects, and the mTOR inhibitor everolimus is licensed for the treatment of several solid cancers. In kidney transplantation, evidence from registry studies indicates a lower rate ofde novomalignancy under mTOR inhibition, with some potentially supportive data from randomized trials of everolimus. Case reports and small single-center series have suggested that switch to everolimus may be beneficial following diagnosis of posttransplant malignancy, particularly for Kaposi’s sarcoma and nonmelanoma skin cancer, but prospective studies are lacking. A systematic review has shown mTOR inhibition to be associated with a significantly lower rate of hepatocellular carcinoma (HCC) recurrence versus standard calcineurin inhibitor therapy. One meta-analysis has concluded that patients with nontransplant HCC experience a low but significant survival benefit under everolimus monotherapy, so far unconfirmed in a transplant population. Data are limited in heart transplantation, although observational data and case reports have indicated that introduction of everolimus is helpful in reducing the recurrence of skin cancers. Overall, it can be concluded that, in certain settings, everolimus appears a promising option to lessen the toll of posttransplant malignancy.


Blood ◽  
2010 ◽  
Vol 115 (23) ◽  
pp. 4758-4769 ◽  
Author(s):  
Hēth R. Turnquist ◽  
Jon Cardinal ◽  
Camila Macedo ◽  
Brian R. Rosborough ◽  
Tina L. Sumpter ◽  
...  

Abstract Prolonged inhibition of the kinase, mammalian target of rapamycin (mTOR), during myeloid dendritic cell (DC) generation confers resistance to maturation. Recently, however, mTOR inhibition immediately before Toll-like receptor ligation has been found to exert proinflammatory effects on myeloid cells, notably enhanced IL-12p40/p70 production. We show, for the first time, that mouse or human DCs generated under mTOR inhibition exhibit markedly enhanced IL-12p70 production after lipopolysaccharide (LPS) stimulation, despite impaired costimulatory molecule expression and poor T-cell stimulatory ability. Consistent with this finding, we reveal that increased IL-12p40 production occurs predominantly in CD86lo immature DCs. High IL-12p40/p70 production by CD86lo DC resulted from failed down-regulation of glycogen synthase kinase-3 (GSK-3) activity and could not be ascribed to enhanced Akt function. Despite high IL-12p70 secretion, rapamycin-conditioned, LPS-stimulated DCs remained poor T-cell stimulators, failing to enhance allogeneic Th1 cell responses. We also report that inhibition of GSK-3 impedes the ability of LPS-stimulated DCs to induce forkhead box p3 in CD4+CD25− T cells, as does the absence of IL-12p40/p70. Thus, GSK-3 activity in DC is regulated via signaling linked to mTOR and modulates their capacity both to produce IL-12p40/p70 and induce forkhead box p3 in CD4+ T cells under inflammatory conditions.


Sign in / Sign up

Export Citation Format

Share Document