Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add filters

Language
Document Type
Year range
1.
Eur Heart J ; 43(Suppl 2), 2022.
Article in English | PubMed Central | ID: covidwho-2107437

ABSTRACT

Background/Introduction: Hyperinflammatory responses to SARS-CoV-2 can cause myocarditis and cardiac dysfunction including congestive heart failure [1]. SARS-CoV-2 RNA induces type I interferon (IFN-I), activating IFN regulatory factors (IRFs) and downstream IFN stimulated genes (ISGs) to initiate inflammatory processes. SARS-CoV-2 variants may develop immune escape, undercutting benefits of vaccinations. These challenges highlight the need of variant-independent therapies to improve COVID-19 outcomes. Apabetalone is an epigenetic BD2-selective BET inhibitor in phase 3 trials for cardiovascular disease [2]. Apabetalone has the potential to treat COVID-19. It counters inflammatory signals caused by cytokine storm (CS), preventing cardiac dysfunction associated with severe COVID-19 symptoms in cardiac organoids [3]. It also downregulates angiotensin-converting enzyme 2 (ACE2) expression, the main host cell receptor for SARS-CoV-2 spike protein thus impeding propagation of wild-type SARS-CoV-2 [3,4]. Purpose: 1) Evaluate apabetalone's effect on inflammatory processes induced by viral-RNA mimetic in human lung cells;2) Assess apabetalone's ability to prevent binding of the highly contagious delta variant spike protein to human lung cells. Methods: Inflammatory gene expression was examined by real-time PCR in apabetalone treated human bronchial epithelial cells (Calu-3) stimulated with poly I:C, a well-accepted viral RNA mimetic that elicits inflammatory signals similar to SARS-CoV-2 RNA [5]. Binding of SARS-CoV-2 delta or wild-type spike protein to apabetalone treated Calu-3 cells was determined by flow cytometry. Results: In Calu-3 cells, apabetalone dose-dependently downregulated poly I:C induced transcription of key COVID-19 associated cytokines (IL6, CXCL10, CCL2) to a similar extent as baricitinib (up to 86%, p<0.0001), an anti-inflammatory agent in emergency use for COVID-19 treatment. Moreover, apabetalone but not baricitinib diminished IL1B mRNA levels (up to 66%, p<0.0001). Apabetalone and baricitinib opposed poly I:C induced expression of IFNB1 (an IFN-I), IRF1 and IRF9 (upstream regulators) as well as IFIT1 and IFIT2 (downstream ISGs that regulate CXCL10 expression;up to 90%, p<0.0001). Clinically relevant doses of apabetalone did not alter expression of anti-viral IFITM2, an ISG that blocks SARS-CoV-2, particularly omicron, endosomal entry [6]. Therefore, apabetalone counters the expression of inflammatory factors with roles in CS and IFN-I signaling in response to poly I:C. Additionally, apabetalone reduced delta and wild-type spike protein binding to unstimulated Calu-3 cells (up to 72%, p<0.0001). Conclusions: Apabetalone's dual anti-viral and anti-inflammatory mechanism positions it as a variant-independent COVID-19 therapeutic. Together with an established safety profile from >2000 treatment-years with apabetalone, the data provide rationale for an ongoing clinical trial (NCT04894266) which includes analysis of cardiac damage. Funding Acknowledgement: Type of funding sources: Private company. Main funding source(s): Resverlogix Corp

2.
European Heart Journal ; 42(SUPPL 1):3267, 2021.
Article in English | EMBASE | ID: covidwho-1554126

ABSTRACT

Background/Introduction: SARS-CoV-2 causes life threatening COVID- 19 complications including acute coronary syndrome, venous thromboembolism, hyperinflammation and damage in multiple tissues. The SARSCoV- 2 spike protein binds cell surface receptors including angiotensinconverting enzyme 2 (ACE2) for entry into host cells to initiate infection. Host cell dipeptidyl peptidase-4 (DPP4 / CD26) is implicated as a cofactor in uptake. Recent evidence indicates expression of factors involved in SARS-CoV-2 uptake into host cells is regulated by BET proteins, epigenetic readers modulating gene expression. Apabetalone, the most clinically advanced BET inhibitor (BETi), is in phase 3 trials for cardiovascular disease (CVD) (a, b). In cultured human cardiomyocytes, apabetalone suppressed infection with SARS-CoV-2 and prevented dysfunction of cardiac organoids induced by the cytokine-storm that arises in patients with severe symptoms (c). However, anti-viral properties of apabetalone in other cell types are not known. Purpose: To examine effects of apabetalone on SARS-CoV-2 infection in cell culture via downregulated expression of cell surface receptors involved in viral entry. Cell systems used mimic initial sites of infection in the lung as well as cell types contributing to complications in late stages of infection. Methods: Gene expression was measured by real-time PCR, protein levels by immunoblot or flow cytometry, and binding of recombinant SARSCoV- 2 spike protein by flow cytometry. Infection with SARS-CoV-2 was determined in a BSL3 facility. Infectivity was quantified by determining levels of viral spike protein amongst total cells via imaging on an Operetta CLS. Results: In Calu-3, a human bronchial epithelial cell line, apabetalone dose-dependently downregulated ACE2 gene expression (up to 98%), reduced ACE2 protein levels (up to 84%) and diminished binding of SARSCoV- 2 spike protein (up to 77%, p<0.001 for all parameters). Further, apabetalone abolished infection of Calu-3 cells with live SARS-CoV-2, which was comparable to other antiviral agents. Apabetalone-driven ACE2 downregulation was also observed in extrapulmonary cell types including HepG2, Huh-7 or primary hepatocytes (up to 90%, p<0.001 for all cell types), and Vero E6, a monkey kidney epithelial cell line (up to 38%, p<0.05). DPP4/CD26, a potential cofactor for SARS-CoV-2 uptake, was also downregulated by apabetalone in Calu-3 cells (mRNA ∼65% and protein ∼40%, p<0.001), which may be synergistic with ACE2 reductions to impede SARS-CoV-2 infection. Conclusions: Apabetalone, an investigational drug for CVD, reduced cell surface receptors (ACE2 and DPP4) involved in SARS-CoV-2 uptake into host cells and dramatically attenuated SARS-CoV-2 infection/propagation in vitro. Our results suggest apabetalone can mitigate SARS-CoV-2 replication in multiple organs, which together with an established safety profile supports clinical evaluation of apabetalone to treat.

SELECTION OF CITATIONS
SEARCH DETAIL