1
|
Jahanshahi F, Jazayeri SB, Eraghi MM, Reis LO, Hamidikia M, Amiri S, Aghamir SMK. A narrative review on adverse drug reactions of COVID-19 treatments on the kidney. Open Med (Wars) 2024; 19:20230867. [PMID: 38584847 PMCID: PMC10996932 DOI: 10.1515/med-2023-0867] [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: 03/23/2022] [Revised: 11/01/2022] [Accepted: 11/18/2023] [Indexed: 04/09/2024] Open
Abstract
Studies showed that the respiratory is not the only system affected by coronavirus 2, while cardiovascular, digestive, and nervous systems, as well as essential organs such as the kidneys, can be affected by this virus. In this review, we have studied the epidemiology, clinical, and laboratory findings on COVID-19 infection renal involvement, mortality, physiopathology, remaining renal sequels after recovery, underlying renal disease, and renal injury due to its treatment. Also, protective measures for kidney injury are explained in three levels. Evidence of viral particles and genome in the urine and renal tubular cells and signs of damage such as microangiopathy, hypercoagulopathy, and fibrosis are found in COVID-19 patients. The result of this study showed, in hospitalized COVID-19 patients, that the rate of acute kidney injury (AKI) was up to 46%, with a mortality ranging from 11 to 96%. A considerable proportion of patients with AKI would remain on renal replacement therapy. Proteinuria and hematuria are observed in 87 and 75% patients, and increased Cr and glomerular filtration rate (GFR) <60 ml/min per 1.73 m2 are observed in 29.6 and 35.3% of the patients, respectively. Remedsivir is considered to have adverse effects on GFR. COVID-19 patients need special attention to prevent AKI. Those with underlying chronic kidney disease or AKI need proper and explicit evaluation and treatment to improve their prognosis and decrease mortality, which should not be limited to the hospitalization period.
Collapse
Affiliation(s)
- Fatemeh Jahanshahi
- Research Committee Member, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Urology Research Center, Tehran University of Medical Sciences, Sina Hospital, Tehran, Iran
| | - Seyed Behnam Jazayeri
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mirahmadi Eraghi
- Urology Research Center, Tehran University of Medical Sciences, Sina Hospital, Tehran, Iran
- School of Medicine, Qeshm International Branch, Islamic Azad University, Qeshm, Iran
| | - Leonardo Oliveira Reis
- UroScience and Department of Surgery (Urology), School of Medical Sciences, University of Campinas, Unicamp, and Pontifical Catholic University of Campinas, PUC-Campinas, Campinas, São Paulo, Brazil
| | - Mahtab Hamidikia
- Research Committee Member, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Amiri
- Rasool Akram Medical Complex, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
2
|
de Almeida Marques DP, Andrade LAF, Reis EVS, Clarindo FA, Moraes TDFS, Lourenço KL, De Barros WA, Costa NEM, Andrade LMD, Lopes-Ribeiro Á, Coêlho Maciel MS, Corrêa-Dias LC, de Almeida IN, Arantes TS, Litwinski VCV, de Oliveira LC, Serafim MSM, Maltarollo VG, Guatimosim SC, Silva MM, Tsuji M, Ferreira RS, Barreto LV, Barbosa-Stancioli EF, da Fonseca FG, De Fátima Â, Coelho-Dos-Reis JGA. New anti-SARS-CoV-2 aminoadamantane compounds as antiviral candidates for the treatment of COVID-19. Virus Res 2024; 340:199291. [PMID: 38065303 PMCID: PMC10733093 DOI: 10.1016/j.virusres.2023.199291] [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: 10/07/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Here, the antiviral activity of aminoadamantane derivatives were evaluated against SARS-CoV-2. The compounds exhibited low cytotoxicity to Vero, HEK293 and CALU-3 cells up to a concentration of 1,000 µM. The inhibitory concentration (IC50) of aminoadamantane was 39.71 µM in Vero CCL-81 cells and the derivatives showed significantly lower IC50 values, especially for compounds 3F4 (0.32 µM), 3F5 (0.44 µM) and 3E10 (1.28 µM). Additionally, derivatives 3F5 and 3E10 statistically reduced the fluorescence intensity of SARS-CoV-2 protein S from Vero cells at 10 µM. Transmission microscopy confirmed the antiviral activity of the compounds, which reduced cytopathic effects induced by the virus, such as vacuolization, cytoplasmic projections, and the presence of myelin figures derived from cellular activation in the face of infection. Additionally, it was possible to observe a reduction of viral particles adhered to the cell membrane and inside several viral factories, especially after treatment with 3F4. Moreover, although docking analysis showed favorable interactions in the catalytic site of Cathepsin L, the enzymatic activity of this enzyme was not inhibited significantly in vitro. The new derivatives displayed lower predicted toxicities than aminoadamantane, which was observed for either rat or mouse models. Lastly, in vivo antiviral assays of aminoadamantane derivatives in BALB/cJ mice after challenge with the mouse-adapted strain of SARS-CoV-2, corroborated the robust antiviral activity of 3F4 derivative, which was higher than aminoadamantane and its other derivatives. Therefore, aminoadamantane derivatives show potential broad-spectrum antiviral activity, which may contribute to COVID-19 treatment in the face of emerging and re-emerging SARS-CoV-2 variants of concern.
Collapse
Affiliation(s)
- Daisymara Priscila de Almeida Marques
- Laboratório de Virologia Básica e Aplicada (LVBA), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luis Adan Flores Andrade
- Laboratório de Virologia Básica e Aplicada (LVBA), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Centro Tecnológico de Vacinas (CT Vacinas), Belo Horizonte, MG, Brazil
| | - Erik Vinicius Sousa Reis
- Laboratório de Virologia Básica e Aplicada (LVBA), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Felipe Alves Clarindo
- Laboratório de Virologia Básica e Aplicada (LVBA), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Thaís de Fátima Silva Moraes
- Laboratório de Virologia Básica e Aplicada (LVBA), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Karine Lima Lourenço
- Laboratório de Virologia Básica e Aplicada (LVBA), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Centro Tecnológico de Vacinas (CT Vacinas), Belo Horizonte, MG, Brazil
| | - Wellington Alves De Barros
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Nathália Evelyn Morais Costa
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lídia Maria de Andrade
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ágata Lopes-Ribeiro
- Laboratório de Virologia Básica e Aplicada (LVBA), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mariella Sousa Coêlho Maciel
- Laboratório de Virologia Básica e Aplicada (LVBA), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Laura Cardoso Corrêa-Dias
- Laboratório de Virologia Básica e Aplicada (LVBA), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isabela Neves de Almeida
- Departamento de Análises Clínicas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil; Laboratório de Micobacterioses, Faculdade de Medicina, Universidade Federal de, Minas Gerais, Belo Horizonte, MG, Brazil
| | - Thalita Souza Arantes
- Centro de Microscopia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vivian Costa Vasconcelos Litwinski
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, Brazil
| | - Leonardo Camilo de Oliveira
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, Brazil
| | - Mateus Sá Magalhães Serafim
- Laboratório de Virus, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, Brazil
| | - Vinicius Gonçalves Maltarollo
- Departamento de Produtos Farmacêuticos da Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, Brazil
| | - Silvia Carolina Guatimosim
- Departamento de Fisiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, Brazil
| | - Mário Morais Silva
- Departamento de Fisiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, Brazil
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rafaela Salgado Ferreira
- Laboratório de Modelagem Molecular e Planejamento de Fármacos, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, Brazil
| | - Luiza Valença Barreto
- Laboratório de Modelagem Molecular e Planejamento de Fármacos, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, Brazil
| | - Edel Figueiredo Barbosa-Stancioli
- Laboratório de Virologia Básica e Aplicada (LVBA), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flávio Guimarães da Fonseca
- Laboratório de Virologia Básica e Aplicada (LVBA), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Centro Tecnológico de Vacinas (CT Vacinas), Belo Horizonte, MG, Brazil
| | - Ângelo De Fátima
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | | |
Collapse
|
3
|
Lotfi A, Hajian P, Abbasi L, Gargari MK, Fard NNG, Naderi D. A Review on Role of Inflammation in Coronavirus Disease. Endocr Metab Immune Disord Drug Targets 2024; 24:1488-1505. [PMID: 38303532 DOI: 10.2174/0118715303265274231204075802] [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: 07/20/2023] [Revised: 09/25/2023] [Accepted: 10/25/2023] [Indexed: 02/03/2024]
Abstract
The respiratory illness known as COVID-19 is caused by the novel coronavirus, SARS-CoV-2. While the precise pathogenic mechanism of COVID-19 remains unclear, the occurrence of a cytokine storm subsequent to viral infection plays a pivotal role in the initiation and advancement of the disease. The infection of SARS-CoV-2 induces a state of immune system hyperactivity, leading to an excessive production of inflammatory cytokines. Consequently, the identification of the various signaling pathways implicated in the inflammation induced by COVID-19 will enable researchers to investigate new targets for therapeutic intervention.
Collapse
Affiliation(s)
| | - Pouran Hajian
- Department of Anesthesiology, Besat Hospital, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Laleh Abbasi
- Guilan University of Medical Sciences, Rasht, Iran
| | | | - Najmeh Nameh Goshay Fard
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Delaram Naderi
- Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
4
|
Janoff EN, Brown ST, Belitskaya-Levy I, Curtis JL, Bonomo RA, Miller EK, Goldberg AM, Zehm L, Wills A, Hutchinson C, Dumont LJ, Gleason T, Shih MC, ADD Caitlin MS in CCTC website. Design of VA CoronavirUs Research and Efficacy Studies-1 (VA CURES-1): A double-blind, randomized placebo-controlled trial of COVID-19 convalescent plasma in hospitalized patients with early respiratory compromise. Contemp Clin Trials Commun 2023; 35:101190. [PMID: 37560085 PMCID: PMC10407261 DOI: 10.1016/j.conctc.2023.101190] [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: 11/07/2022] [Revised: 07/07/2023] [Accepted: 07/15/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Effective therapeutics for severe acute respiratory syndrome CoronaVirus-2 (SARS-CoV-2) infection are evolving. Under Emergency Use Authorization, COVID-19 convalescent plasma (CCP) was widely used in individuals hospitalized for COVID-19, but few randomized controlled trials supported its efficacy to limit respiratory failure or death. METHODS VA CoronavirUs Research and Efficacy Studies-1 (VA CURES-1) was a double-blind, multi-site, placebo-controlled, randomized clinical trial evaluating the efficacy and safety of CCP with conventional therapy in hospitalized Veterans with SARS-CoV-2 infection and early respiratory compromise (requirement for oxygen). Participants (planned sample size 702) were randomized 1:1 to receive CCP with high titer neutralizing activity or 0.9% saline, stratified by site and age (≥65 versus <65 years old). Participants were followed daily during initial hospitalization and at Days 15, 22 and 28. OUTCOMES The composite primary outcome was acute hypoxemic respiratory failure or all-cause death by Day 28. Secondary outcomes by day 28 included time-to-recovery, clinical severity, mortality, rehospitalization for COVID-19, and adverse events. Serial respiratory and blood samples were collected for safety, virologic and immunologic analyses and future studies. Key variables in predicting the success of CURES-1 were: (1) enrollment early in the course of severe infection; (2) use of plasma with high neutralizing activity; (3) reliance on unambiguous, clinically meaningful outcomes. CURES-1 was terminated for futility due to perceived inability to enroll in the lull between the Alpha and Delta waves of the SARS CoV-2 epidemic. CONCLUSIONS VA CURES-1 was a large multi-site trial designed to provide conclusive information about the efficacy of CCP in well-characterized patients at risk for progression of COVID-19. It utilized a rigorous study design with relevant initial timing, quality of product and outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04539275.
Collapse
Affiliation(s)
- Edward N. Janoff
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
- University of Colorado Denver School of Medicine, Aurora, CO, USA
| | - Sheldon T. Brown
- James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
- Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Ilana Belitskaya-Levy
- Department of Veterans Affairs, Cooperative Studies Program Coordinating Center, Palo Alto, CA, USA
| | - Jeffrey L. Curtis
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
- University of Michigan Medical School, Ann Arbor, MI, USA
| | - Robert A. Bonomo
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
- Case VA CARES, Case Western Reserve University School of Medicine, USA
| | - Elliott K. Miller
- Department of Veterans Affairs, Cooperative Studies Program Clinical Research Pharmacy Coordinating Center, Albuquerque, NM, USA
| | - Alexa M. Goldberg
- Department of Veterans Affairs, Cooperative Studies Program Clinical Research Pharmacy Coordinating Center, Albuquerque, NM, USA
| | - Lisa Zehm
- Department of Veterans Affairs, Cooperative Studies Program Coordinating Center, Palo Alto, CA, USA
| | - Ashlea Wills
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | | | - Larry J. Dumont
- University of Colorado Denver School of Medicine, Aurora, CO, USA
- Vitalant Research Institute, Denver, CO, USA
| | - Theresa Gleason
- Department of Veterans Affairs, Clinical Science Research and Development Service, Washington, DC, USA
| | - Mei-Chiung Shih
- Department of Veterans Affairs, Cooperative Studies Program Coordinating Center, Palo Alto, CA, USA
- Stanford University School of Medicine, Palo Alto, CA, USA
| | - ADD Caitlin MS in CCTC website
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
- University of Colorado Denver School of Medicine, Aurora, CO, USA
- James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
- Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Department of Veterans Affairs, Cooperative Studies Program Coordinating Center, Palo Alto, CA, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
- University of Michigan Medical School, Ann Arbor, MI, USA
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
- Case VA CARES, Case Western Reserve University School of Medicine, USA
- Department of Veterans Affairs, Cooperative Studies Program Clinical Research Pharmacy Coordinating Center, Albuquerque, NM, USA
- Vitalant Research Institute, Denver, CO, USA
- Department of Veterans Affairs, Clinical Science Research and Development Service, Washington, DC, USA
- Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
5
|
Singh A, Kaur A, Chowdhary A. Fungal pathogens and COVID-19. Curr Opin Microbiol 2023; 75:102365. [PMID: 37625261 DOI: 10.1016/j.mib.2023.102365] [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: 05/09/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/27/2023]
Abstract
COVID-19 pandemic highlighted the complications of secondary fungal infections that occurred globally in severe cases of coronavirus disease managed in the intensive care units. Furthermore, varied underlying host factors, such as preexisting immunosuppression, the use of immunomodulatory agents, and invasive procedures predisposing lung tissues to fungal colonization and proliferation, caused increased susceptibility to fungal infections in COVID-19 patient populations. These invasive fungal infections directly impact the overall length of hospitalization and mortality. The most commonly reported fungal infections in patients with COVID-19 include aspergillosis, invasive candidiasis, and mucormycosis. An overall worldwide increase in the prevalence of candidiasis and aspergillosis was observed in COVID-19 patients , whereas outbreaks of mucormycosis were mainly recorded from India. Diagnostic challenges and limited antifungal treatment options make secondary fungal infections among COVID-19 patients more burdensome, which results in improper management and increased mortality.
Collapse
Affiliation(s)
- Ashutosh Singh
- Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India; National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Amtoj Kaur
- Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Anuradha Chowdhary
- Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India; National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India.
| |
Collapse
|
6
|
Mahmoud IS, Jarrar YB, Febrimarsa. Modulation of IRAK enzymes as a therapeutic strategy against SARS-CoV-2 induced cytokine storm. Clin Exp Med 2023; 23:2909-2923. [PMID: 37061574 PMCID: PMC10105542 DOI: 10.1007/s10238-023-01064-7] [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: 03/08/2023] [Accepted: 04/02/2023] [Indexed: 04/17/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the current pandemic coronavirus disease 2019 (COVID-19). Dysregulated and excessive production of cytokines and chemokines, known as cytokine storm, is frequently seen in patients with severe COVID-19 disease and it can provoke a severe systematic inflammation in the patients. The IL-1R/TLRs/IRAKs signaling network is a key pathway in immune cells that plays a central role in regulating innate immunity and inflammatory responses via stimulating the expression and production of various proinflammatory molecules including cytokines. Modulation of IRAKs activity has been proposed to be a promising strategy in the treatment of inflammatory disorders. In this review, we highlight the biochemical properties of IRAKs and their role in regulating inflammatory molecular signaling pathways and discuss the potential targeting of IRAKs to suppress the SARS-CoV-2-induced cytokine storm in COVID-19 patients.
Collapse
Affiliation(s)
- Ismail Sami Mahmoud
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan.
| | - Yazun Bashir Jarrar
- Department of Basic Medical Sciences, Faculty of Medicine, Al-Balqa Applied University, As-Salt, Jordan
| | - Febrimarsa
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway, Republic of Ireland
| |
Collapse
|
7
|
Prabowo NA, Megantara MA, Apriningsih H. The role of N-acetylcysteine in decreasing neutrophil-lymphocyte ratio in COVID-19 patients: A double-blind, randomized controlled trial. NARRA J 2023; 3:e121. [PMID: 38454976 PMCID: PMC10919724 DOI: 10.52225/narraj.v3i2.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/09/2023] [Indexed: 03/09/2024]
Abstract
N-acetylcysteine has antioxidant and anti-inflammatory activities that could potentially improve the clinical outcomes of coronavirus disease 2019 (COVID-19) patients. N-acetylcysteine potentially inhibits NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) inflammasome and results in control oxidative stress and cytokine release in COVID-19 patients. The aim of this study was to assess the effect of N-acetylcysteine in reducing the neutrophil-lymphocyte ratio (NLR) in COVID-19 patients. A randomized controlled clinical trial was conducted among severe and moderate COVID-19 patients. The treatment group received oral 1200 mg daily of N-acetylcysteine (three times a day) and the standard care for COVID-19, while the control group received standard care for COVID-19 and a placebo. The NLR was determined on the first day of admission and after the seventh day of treatment. A paired Student t-test was used to compare the NLR before and after treatment while independent Student t-test was used to compare the NLR between treatment and control groups. A total of 40 severe and moderate COVID-19 were enrolled, 20 people in each group, with a mean age was 44.68±13.24 years old. The mean NLR on the first day was 9.44 in the treatment group and 8.84 in the control group. After the seventh day, the mean NLR was 4.27 and 11.54 in the treatment group and control group, respectively. The mean changes of NLR (the pre-treatment compared to post-treatment) in the treatment and control group were reduced 4.05 and increased 3.34, respectively. The NLR in treatment group significantly decreased compared to the control group (p<0.001). In conclusion, N-acetylcysteine 1200 mg daily could reduce the NLR in severe and moderate COVID-19 patients.
Collapse
Affiliation(s)
- Nurhasan A. Prabowo
- Department of Internal Medicine, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
- Department of Internal Medicine, Universitas Sebelas Maret Hospital, Kartasura, Indonesia
| | - Marcelino A. Megantara
- Department of Internal Medicine, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Hendrastutik Apriningsih
- Department of Internal Medicine, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
- Department of Internal Medicine, Universitas Sebelas Maret Hospital, Kartasura, Indonesia
| |
Collapse
|
8
|
Filippi-Arriaga F, Aguilera C, Guillén E, Bellas L, Pérez E, Vendrell L, Agustí A, Cereza G. Unknown adverse drug reactions from spontaneous reports in a hospital setting: characterization, follow-up, and contribution to the pharmacovigilance system. Front Pharmacol 2023; 14:1211786. [PMID: 37492089 PMCID: PMC10364048 DOI: 10.3389/fphar.2023.1211786] [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: 05/02/2023] [Accepted: 06/30/2023] [Indexed: 07/27/2023] Open
Abstract
Introduction: Post-marketing identification and report of unknown adverse drug reactions (ADRs) are crucial for patient safety. However, complete information on unknown ADRs seldom is available at the time of spontaneous ADR reports and this can hamper their contribution to the pharmacovigilance system. Methods: In order to characterize the seriousness and outcome of unknown ADRs at the time of report and at follow-up, and analyze their contribution to generate pharmacovigilance regulatory actions, a retrospective observational study of those identified in the spontaneous ADR reports of patients assisted at a hospital (January, 2016-December, 2021) was carried out. Information on demographic, clinical and complementary tests was retrieved from patients' hospital medical records. To evaluate the contribution to pharmacovigilance system we reviewed the European Union SmPCs, the list of the pharmacovigilance signals discussed by the Pharmacovigilance Risk Assessment Committee, and its recommendations reports on safety signals. Results: A total of 15.2% of the spontaneous reported cases during the study contained at least one unknown drug-ADR pair. After exclusions, 295 unknown drug-ADR pairs were included, within them the most frequently affected organs or systems were: skin and subcutaneous tissue (34, 11.5%), hepatobiliary disorders (28, 9.5%), cardiac disorders (28, 9.5%) and central nervous system disorders (27, 9.2%). The most frequent ADRs were pemphigus (7, 2.4%), and cytolytic hepatitis, sudden death, cutaneous vasculitis and fetal growth restriction with 6 (2%) each. Vaccines such as covid-19 and pneumococcus (68, 21.3%), antineoplastics such as paclitaxel, trastuzumab and vincristine (39, 12.2%) and immunosuppressants such as methotrexate and tocilizumab (35, 11%) were the most frequent drug subgroups involved. Sudden death due to hydroxychloroquine alone or in combination (4, 1.4%) and hypertransaminasemia by vincristine (n = 3, 1%) were the most frequent unknown drug-ADR pairs. A total of 269 (91.2%) of them were serious. Complementary tests were performed in 82.7% of unknown-ADR pairs and helped to reinforce their association in 18.3% of them. A total of 18 (6.1%) unknown drug-ADR pairs were evaluated by the EMA, in 8 (2.7%) the information was added to the drug's SmPC and in 1 case the risk prevention material was updated. Conclusion: Identification and follow-up of unknown ADRs can be of great relevance for patient safety and for the enrichment of the pharmacovigilance system.
Collapse
Affiliation(s)
- Francesca Filippi-Arriaga
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Cristina Aguilera
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain
- Immunomediated Diseases and Innovative Therapies Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Elena Guillén
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Clinical Pharmacology, Area Medicament, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Lucía Bellas
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Eulàlia Pérez
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Lourdes Vendrell
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Antònia Agustí
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain
- Immunomediated Diseases and Innovative Therapies Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Gloria Cereza
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain
- Catalan Centre of Pharmacovigilance, Directorate-General for Healthcare Planning and Regulation, Ministry of Health, Government of Catalonia, Barcelona, Spain
| |
Collapse
|
9
|
Hong H, Friedland A, Hu M, Anstrom KJ, Halabi S, McKinnon JE, Amaravadi R, Rojas-Serrano J, Abella BS, Portillo-Vázquez AM, Woods CW, Hernandez AF, Boulware DR, Naggie S, Rajasingham R. Safety and efficacy of hydroxychloroquine as prophylactic against COVID-19 in healthcare workers: a meta-analysis of randomised clinical trials. BMJ Open 2023; 13:e065305. [PMID: 37328184 PMCID: PMC10276967 DOI: 10.1136/bmjopen-2022-065305] [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: 06/02/2022] [Accepted: 05/31/2023] [Indexed: 06/18/2023] Open
Abstract
OBJECTIVE We studied the safety and efficacy of hydroxychloroquine (HCQ) as pre-exposure prophylaxis for COVID-19 in healthcare workers (HCWs), using a meta-analysis of randomised controlled trials (RCTs). DATA SOURCES PubMed and EMBASE databases were searched to identify randomised trials studying HCQ. STUDY SELECTION Ten RCTs were identified (n=5079 participants). DATA EXTRACTION AND SYNTHESIS The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were used in this systematic review and meta-analysis between HCQ and placebo using a Bayesian random-effects model. A pre-hoc statistical analysis plan was written. MAIN OUTCOMES The primary efficacy outcome was PCR-confirmed SARS-CoV-2 infection and the primary safety outcome was incidence of adverse events. The secondary outcome included clinically suspected SARS-CoV-2 infection. RESULTS Compared with placebo, HCWs randomised to HCQ had no significant difference in PCR-confirmed SARS-CoV-2 infection (OR 0.92, 95% credible interval (CI): 0.58, 1.37) or clinically suspected SARS-CoV-2 infection (OR 0.78, 95% CI: 0.57, 1.10), but significant difference in adverse events (OR 1.35, 95% CI: 1.03, 1.73). CONCLUSIONS AND RELEVANCE Our meta-analysis of 10 RCTs investigating the safety and efficacy of HCQ as pre-exposure prophylaxis in HCWs found that compared with placebo, HCQ does not significantly reduce the risk of confirmed or clinically suspected SARS-CoV-2 infection, while HCQ significantly increases adverse events. PROSPERO REGISTRATION NUMBER CRD42021285093.
Collapse
Affiliation(s)
- Hwanhee Hong
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Anne Friedland
- Department of Infectious Disease, UNC School of Medicine, Chapel Hill, North Carolina, USA
| | - Mengyi Hu
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Kevin J Anstrom
- Collaborative Studies Coordinating Center, University of North Carolina System, Chapel Hill, North Carolina, USA
| | - Susan Halabi
- Duke Clinical Research Institute, Durham, North Carolina, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - John E McKinnon
- Division of Infectious Diseases, Henry Ford Hospital, Detroit, Michigan, USA
| | - Ravi Amaravadi
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jorge Rojas-Serrano
- Interstitial Lung Disease and Rheumatology Units, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Benjamin S Abella
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | - David R Boulware
- Division of Infectious Diseases & International Medicine, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Susanna Naggie
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Radha Rajasingham
- Division of Infectious Diseases & International Medicine, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| |
Collapse
|
10
|
Adzic-Vukicevic T, Mladenovic M, Jovanovic S, Soldatović I, Radovanovic-Spurnic A. Invasive fungal disease in COVID-19 patients: a single-center prospective observational study. Front Med (Lausanne) 2023; 10:1084666. [PMID: 37359005 PMCID: PMC10288186 DOI: 10.3389/fmed.2023.1084666] [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: 10/30/2022] [Accepted: 05/05/2023] [Indexed: 06/28/2023] Open
Abstract
Background Invasive fungal diseases (IFDs) are caused by fungal infections that manifest as serious secondary infections in patients with COVID-19. The increased morbidity and mortality rates are most frequently observed in patients with COVID-19-associated pulmonary aspergillosis (CAPA) and COVID-19-associated candidiasis (CAC). CAPA is the most frequently encountered infection with an incidence rate of 0.7-7.7%, while CAC is a less common and less studied fungal infection in COVID-19 patients. Materials and methods The present article is a prospective observational single-center study that was conducted between 1 September 2021 and 24 December 2021, involving 6,335 patients who were admitted to COVID Hospital "Batajnica," University Clinical Center of Serbia, Belgrade. Results Of the 6,335 patients hospitalized during the four-month period of the study, 120 patients (1.86%) who had a proven diagnosis of IFD were included in the study. These patients were divided into two groups: CAPA patients (n = 63) and CAC patients (n = 56); however, one of the 120 patients was diagnosed with Cryptoccocus neoformans infection. The mean age of the study population was 65.7 ± 13.9 years, and 78 (65.5%) of them were men. The patients were identified to have the following non-malignant comorbidities: arterial hypertension in 62 (52.1%) patients, diabetes mellitus in 34 (28.65), pre-existing lung damage similar to that observed in COPD and asthma in 20 (16.8%), and chronic renal insufficiency in 13 (10.9%) patients. The hematological malignancies were found to be the most prevalent malignancies and were identified in 20 (16.8%) patients, particularly in CAPA patients [11 (17.5%); p < 0.041]. Fiberoptic bronchoscopy with bronchoalveolar lavage fluid (BALF) and microscopic examination confirmed the presence of fungal infections in 17 (14.3%) patients. Serology testing was also performed in the majority of cases. Antibodies against Aspergillus spp. and Candida spp. were predominantly found in CAPA patients (p < 0.001). The patients were also tested for the presence of (1-3)-β-D glucan (p < 0.019), galactomannan, and mannan in the specimens. Blood cultures were found to be positive in 45 (37.8%) patients, mostly in CAC patients. Mechanical ventilation was applied in 41 (34.5%) patients, while a non-invasive technique, such as continuous positive airway pressure (CPAP) or high-flow nasal cannula (HFNC), was used in 20 (16.8%) patients. The following antifungals were administered: echinocandins in 42 (35.3%), voriconazole in 30 (25.2%), and fluconazole in 27 (22.7%) patients. Most of the patients received systemic corticosteroids (mainly methylprednisolone), while 11 (9.16%) received favipiravir, 32 (26.67%) remdesivir, 8 (6.67%) casirivimab/imdevimab, and 5 (4.16%) sotrovimab. The outcome was lethal in 76 (63.9%) patients, predominantly CAC patients (p < 0.001). Conclusion Invasive fungal disease is a severe complication associated with COVID-19 and accounts for increased mortality in these patients. Early identification and appropriate treatment may provide a favorable outcome.
Collapse
Affiliation(s)
- Tatjana Adzic-Vukicevic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Covid Hospital Batajnica, University Clinical Center of Serbia, Belgrade, Serbia
- Clinic for Pulmonology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Milos Mladenovic
- Covid Hospital Batajnica, University Clinical Center of Serbia, Belgrade, Serbia
| | - Snezana Jovanovic
- Covid Hospital Batajnica, University Clinical Center of Serbia, Belgrade, Serbia
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Belgrade, Serbia
| | - Ivan Soldatović
- Institute for Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Radovanovic-Spurnic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Covid Hospital Batajnica, University Clinical Center of Serbia, Belgrade, Serbia
- Center for Microbiology, University Clinical Center of Serbia, Belgrade, Serbia
| |
Collapse
|
11
|
Tsotsolis S, Kotoulas SC, Lavrentieva A. Invasive Pulmonary Aspergillosis in Coronavirus Disease 2019 Patients Lights and Shadows in the Current Landscape. Adv Respir Med 2023; 91:185-202. [PMID: 37218799 DOI: 10.3390/arm91030016] [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: 02/07/2023] [Revised: 04/17/2023] [Accepted: 04/30/2023] [Indexed: 05/24/2023]
Abstract
Invasive pulmonary aspergillosis (IPA) presents a known risk to critically ill patients with SARS-CoV-2; quantifying the global burden of IPA in SARS-CoV-2 is extremely challenging. The true incidence of COVID-19-associated pulmonary aspergillosis (CAPA) and the impact on mortality is difficult to define because of indiscriminate clinical signs, low culture sensitivity and specificity and variability in clinical practice between centers. While positive cultures of upper airway samples are considered indicative for the diagnosis of probable CAPA, conventional microscopic examination and qualitative culture of respiratory tract samples have quite low sensitivity and specificity. Thus, the diagnosis should be confirmed with serum and BAL GM test or positive BAL culture to mitigate the risk of overdiagnosis and over-treatment. Bronchoscopy has a limited role in these patients and should only be considered when diagnosis confirmation would significantly change clinical management. Varying diagnostic performance, availability, and time-to-results turnaround time are important limitations of currently approved biomarkers and molecular assays for the diagnosis of IA. The use of CT scans for diagnostic purposes is controversial due to practical concerns and the complex character of lesions presented in SARS-CoV-2 patients. The key objective of management is to improve survival by avoiding misdiagnosis and by initiating early, targeted antifungal treatment. The main factors that should be considered upon selection of treatment options include the severity of the infection, concomitant renal or hepatic injury, possible drug interactions, requirement for therapeutic drug monitoring, and cost of therapy. The optimal duration of antifungal therapy for CAPA is still under debate.
Collapse
Affiliation(s)
- Stavros Tsotsolis
- Medical School, Aristotle University of Thessaloniki, Leoforos Agiou Dimitriou, 54124 Thessaloniki, Greece
| | | | - Athina Lavrentieva
- 1st ICU, General Hospital of Thessaloniki "Georgios Papanikolaou", Leoforos Papanikolaou, 57010 Thessaloniki, Greece
| |
Collapse
|
12
|
Regalado-Méndez A, Zavaleta-Avendaño J, Peralta-Reyes E, Natividad R. Convex optimization for maximizing the degradation efficiency of chloroquine in a flow-by electrochemical reactor. J Solid State Electrochem 2023:1-14. [PMID: 37363394 PMCID: PMC10088624 DOI: 10.1007/s10008-023-05452-7] [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: 12/13/2022] [Revised: 02/09/2023] [Accepted: 03/03/2023] [Indexed: 06/28/2023]
Abstract
The degradation efficiency of chloroquine phosphate (CQ), an anti-COVID-19 drug, was investigated in a flow-by electrochemical reactor (FBER) provided with two boron-doped diamond (BDD) electrodes (as cathode and anode) under batch recirculation mode. A central composite rotatable design (CCRD) was run down to model and assess the influence of initial pH in an interval of 3.71 to 11.28, the current density in an interval of 34.32 to 185.68 mA cm-2, and liquid volumetric flow rate in an interval of 0.58 to 1.42 L min-1, and conduct the convex optimization to obtain the maximum degradation efficiency. Experimental results were modeled through a second-order polynomial equation having a determination coefficient (R2) of 0.9705 with a variance coefficient of 1.1%. Optimal operating conditions found (initial pH of 5.38, current density (j) of 34.4 mA cm-2, and liquid flow rate (Q) of 1.42 L min-1) led to a global maximum degradation efficiency, COD removal efficiency, and mineralization efficiency of 89.3, 51.6 and 53.1%, respectively, with an energy consumption of 0.041 kWh L-1 within 9 h of treatment. Additionally, a pseudo-zero-order kinetic model was demonstrated to fit the experimental data and the calculated pseudo-zero-order kinetic constant (kapp) was 13.14 mg L-1 h-1 (2.54 × 10-5 mol dm-3 h-1). Furthermore, the total operating cost was of 0.47 US$ L-1. Finally, this research could be helpful for the treatment of wastewater containing an anti-COVID-19 drug such as CQ. Supplementary Information The online version contains supplementary material available at 10.1007/s10008-023-05452-7.
Collapse
Affiliation(s)
| | | | - Ever Peralta-Reyes
- Investigation Laboratories, Universidad del Mar, Puerto Ángel, 70902 Oaxaca, México
| | - Reyna Natividad
- Chemical Engineering Laboratory, Centro Conjunto de Investigación en Química Sustentable, UAEMex-UNAM, Universidad Autónoma del Estado de México, Estado de México, Toluca, 50200 México
| |
Collapse
|
13
|
Naggie S, Milstone A, Castro M, Collins SP, Lakshmi S, Anderson DJ, Cahuayme-Zuniga L, Turner KB, Cohen LW, Currier J, Fraulo E, Friedland A, Garg J, George A, Mulder H, Olson RE, O'Brien EC, Rothman RL, Shenkman E, Shostak J, Woods CW, Anstrom KJ, Hernandez AF. Hydroxychloroquine for pre-exposure prophylaxis of COVID-19 in health care workers: a randomized, multicenter, placebo-controlled trial Healthcare Worker Exposure Response and Outcomes of Hydroxychloroquine (HERO-HCQ). Int J Infect Dis 2023; 129:40-48. [PMID: 36682681 PMCID: PMC9851717 DOI: 10.1016/j.ijid.2023.01.019] [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: 07/27/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVES To determine whether hydroxychloroquine (HCQ) is safe and effective at preventing COVID-19 infections among health care workers (HCWs). METHODS In a 1: 1 randomized, placebo-controlled, double-blind, parallel-group, superiority trial at 34 US clinical centers, 1360 HCWs at risk for COVID-19 infection were enrolled between April and November 2020. Participants were randomized to HCQ or matched placebo. The HCQ dosing included a loading dose of HCQ 600 mg twice on day 1, followed by 400 mg daily for 29 days. The primary outcome was a composite of confirmed or suspected COVID-19 clinical infection by day 30, defined as new-onset fever, cough, or dyspnea and either a positive SARS-CoV-2 polymerase chain reaction test (confirmed) or a lack of confirmatory testing due to local restrictions (suspected). RESULTS Study enrollment closed before full accrual due to recruitment challenges. The primary end point occurred in 41 (6.0%) participants receiving HCQ and 53 (7.8%) participants receiving placebo. No difference in the proportion of participants experiencing clinical infection (estimated difference of -1.8%, 95% confidence interval -4.6-0.9%, P = 0.20) was identified nor any significant safety issues. CONCLUSION Oral HCQ taken as prescribed appeared safe among HCWs. No significant clinical benefits were observed. The study was not powered to detect a small but potentially important reduction in infection. TRIAL REGISTRATION NCT04334148.
Collapse
Affiliation(s)
- Susanna Naggie
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA.
| | | | - Mario Castro
- University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sean P Collins
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | | | | - Lauren W Cohen
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Judith Currier
- University of California Los Angeles, Los Angeles, California, USA
| | - Elizabeth Fraulo
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Anne Friedland
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Jyotsna Garg
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Anoop George
- Temple University, Philadelphia, Pennsylvania, USA
| | - Hillary Mulder
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Rachel E Olson
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Emily C O'Brien
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | | | | | - Jack Shostak
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Christopher W Woods
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Kevin J Anstrom
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Adrian F Hernandez
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | | |
Collapse
|
14
|
Jeon WJ, Lee HK, Na YG, Jung M, Han SC, Hwang JH, Jung E, Hwang D, Shin JS, Cho CW. Antiviral Lipid Nanocarrier Loaded with Remdesivir Effective Against SARS-CoV-2 in vitro Model. Int J Nanomedicine 2023; 18:1561-1575. [PMID: 37007987 PMCID: PMC10065008 DOI: 10.2147/ijn.s391462] [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: 09/27/2022] [Accepted: 03/10/2023] [Indexed: 03/28/2023] Open
Abstract
Introduction The ongoing SARS-CoV-2 pandemic has affected public health, the economy, and society. This study reported a nanotechnology-based strategy to enhance the antiviral efficacy of the antiviral agent remdesivir (RDS). Results We developed a nanosized spherical RDS-NLC in which the RDS was encapsulated in an amorphous form. The RDS-NLC significantly potentiated the antiviral efficacy of RDS against SARS-CoV-2 and its variants (alpha, beta, and delta). Our study revealed that NLC technology improved the antiviral effect of RDS against SARS-CoV-2 by enhancing the cellular uptake of RDS and reducing SARS-CoV-2 entry in cells. These improvements resulted in a 211% increase in the bioavailability of RDS. Conclusion Thus, the application of NLC against SARS-CoV-2 may be a beneficial strategy to improve the antiviral effects of antiviral agents.
Collapse
Affiliation(s)
- Woo-Jin Jeon
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Hong-Ki Lee
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology (KIT), Jeongeup, Jeollabuk-do, 53212, Republic of Korea
- Human Health Risk Assessment Center, Jeonbuk Branch, Korea Institute of Toxicology (KIT), Jeongeup, Jeollabuk-do, 53212, Republic of Korea
| | - Young-Guk Na
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Minwoo Jung
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Su-Cheol Han
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology (KIT), Jeongeup, Jeollabuk-do, 53212, Republic of Korea
| | - Jeong Ho Hwang
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology (KIT), Jeongeup, Jeollabuk-do, 53212, Republic of Korea
| | - Eunhye Jung
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Dasom Hwang
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Jin Soo Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Correspondence: Jin Soo Shin, Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea, Email
| | - Cheong-Weon Cho
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon, 34134, Republic of Korea
- Cheong-Weon Cho, College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea, Email
| |
Collapse
|
15
|
Affiliation(s)
- Adarsh Bhimraj
- Division of Infectious Diseases, Houston Methodist Hospital, Houston, Texas
| | - Jason C Gallagher
- Department of Pharmacy Practice, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
16
|
Lung Ultrasound Is Useful for Evaluating Lung Damage in COVID-19 Patients Treated with Bamlanivimab and Etesevimab: A Single-Center Pilot Study. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020203. [PMID: 36837405 PMCID: PMC9962749 DOI: 10.3390/medicina59020203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
Background and Objectives: COVID-19 induces massive systemic inflammation. Researchers have spent much time and effort finding an excellent and rapid image tool to evaluate COVID-19 patients. Since the pandemic's beginning, lung ultrasound (LUS) has been identified for this purpose. Monoclonal antibodies (mAb) were used to treat mild patients and prevent respiratory disease worsening. Materials and Methods: We evaluated 15 Caucasian patients with mild COVID-19 who did not require home oxygen, treated with Bamlanivimab and Etesevimab (Group 1). A molecular nose-throat swab test confirmed the diagnosis. All were office patients, and nobody was affected by respiratory failure. They were admitted to receive the single-day infusion of mAb treatment in agreement with the Italian Drug Agency (AIFA) rules for approval. LUS was performed before the drug administration (T0) and after three months (T1). We compared LUS at T1 in other outpatients who came for follow-up and were overlapping at the time of diagnosis for admittance criteria to receive mAb (Group 2). Results: Our COVID-19 outpatients reported no hospitalization in a follow-up visit after recovery. All patients became SARS-CoV-2 negative within one month since T0. LUS score at T0 was 8.23 ± 6.46. At T1 we found a significant decrease in Group 1 LUS score (5.18 ± 4.74; p < 0.05). We also found a significant decrease in the LUS score of Group 1 T1 compared to Group2 T1 (5.18 ± 4.74 vs 7.82 ± 5.21; p < 0.05). Conclusion: Early treatment of the SARS-CoV-2 virus effectively achieves a better recovery from disease and reduces lung involvement after three months as evaluated with LUS. Despite extrapolation to the general population may be done with caution, based on our data this ultrasound method is also effective for evaluating and following lung involvement in COVID-19 patients.
Collapse
|
17
|
Saha P, Biswas SK, Biswas MHA, Ghosh U. An SEQAIHR model to study COVID-19 transmission and optimal control strategies in Hong Kong, 2022. NONLINEAR DYNAMICS 2023; 111:6873-6893. [PMID: 36644569 PMCID: PMC9825089 DOI: 10.1007/s11071-022-08181-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
During the COVID-19 pandemic, one of the major concerns was a medical emergency in human society. Therefore it was necessary to control or restrict the disease spreading among populations in any fruitful way at that time. To frame out a proper policy for controlling COVID-19 spreading with limited medical facilities, here we propose an SEQAIHR model having saturated treatment. We check biological feasibility of model solutions and compute the basic reproduction number ( R 0 ). Moreover, the model exhibits transcritical, backward bifurcation and forward bifurcation with hysteresis with respect to different parameters under some restrictions. Further to validate the model, we fit it with real COVID-19 infected data of Hong Kong from 19th December, 2021 to 3rd April, 2022 and estimate model parameters. Applying sensitivity analysis, we find out the most sensitive parameters that have an effect on R 0 . We estimate R 0 using actual initial growth data of COVID-19 and calculate effective reproduction number for same period. Finally, an optimal control problem has been proposed considering effective vaccination and saturated treatment for hospitalized class to decrease density of the infected class and to minimize implemented cost.
Collapse
Affiliation(s)
- Pritam Saha
- Department of Applied Mathematics, University of Calcutta, Kolkata, 700009 India
| | | | | | - Uttam Ghosh
- Department of Applied Mathematics, University of Calcutta, Kolkata, 700009 India
| |
Collapse
|
18
|
Gallagher JC, Morgan RL. Successful immunomodulators for the treatment of COVID-19 have opened the pathway for comparative trials. Clin Microbiol Infect 2023; 29:7-9. [PMID: 36257548 PMCID: PMC9575309 DOI: 10.1016/j.cmi.2022.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 01/26/2023]
Affiliation(s)
- Jason C Gallagher
- Temple University, School of Pharmacy, Philadelphia, Pennsylvania, USA.
| | - Rebecca L Morgan
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada; School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
19
|
XAVIER JMA, FIRMINO RT, PEREIRA IF, XAVIER MA, COSTA MCFD, SOARES RDSC, RIBEIRO AIAM. The impact of the COVID-19 pandemic in Brazil: a study in tertiary dental care. Braz Oral Res 2023; 37:025. [PMID: 37018807 DOI: 10.1590/1807-3107bor-2023.vol37.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/19/2022] [Indexed: 04/05/2023] Open
Abstract
The pandemic caused by coronavirus has resonated throughout different levels of health care in Brazil and, in this context, the present research aimed to evaluate this impact on tertiary dental care provided by the Unified Health System (SUS). Therefore, an ecological study was conducted with data obtained from the Hospital Information System processed by the Portal of the Department of Informatics of SUS. The sample consisted of patients of all sexes and age groups, whose Hospital Admission Authorizations (AIHs) were approved for dental tertiary care procedures from January 2015 to December 2020. Descriptive analyses and the ANOVA test with a significance level set at p < 0.05 were used. When the annual mean numbers of AIHs approved were evaluated, findings showed that on an average, the Southeast region authorized a higher number of procedures (p-value < 0.001), however, in the pandemic year (2020), a reduction of approximately 24.5% of these hospitalizations occurred throughout Brazil, with the Midwest being the region most affected (32.12%). A percentage increase occurred in the Surgical Treatment of Oral sinus/Oral nasal Fistula (16.1%), in addition to a significant decrease in performing procedures for Resection of Mouth Lesion (33.4%). In the pandemic year, there was a reduction of 14% in expenditures related to hospital services and 23.26% related to professional services. It was concluded that the data presented demonstrated a significant reduction in AIHs for tertiary dental care in the pandemic year.
Collapse
|
20
|
Zeng Z. Assessment of the potential value of combining western medicine therapies with traditional chinese medicine in the treatment of COVID-19: Mechanistic perspectives. Technol Health Care 2023; 31:169-184. [PMID: 37038790 PMCID: PMC10200170 DOI: 10.3233/thc-236015] [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] [Indexed: 04/12/2023]
Abstract
BACKGROUND The pandemic caused by the novel coronavirus disease (COVID-19) since early 2020 is one of the most significant global health issues in history. Although there is currently no specific treatment for COVID-19, researchers have provided a whole array of potential treatments, both from the Western medicine approach, which is molecular target and pathogenesis based, and from the traditional Chinese medicine (TCM) approach, which is based on the exposure to toxins/pathogens and the balance of the body to combat them for recovery. OBJECTIVE The aim of this research is to find combinations of Western medicine and TCM that may offer better therapeutic efficacy synergystically with a better adverse events profile. The findings of the research may provide a new insight in the development of the treatment of COVID-19. METHODS From the Western medicine perspective, drugs target the mechanisms of viral infection, including the stages of viral entry (Arbidol, Camostat Mesylate, Convalescent Plasma therapy) and viral replication (Lopinavir/Ritonavir, Redemsivir, Ribavirin). Additional therapies target host defenses, preventing cytokine storms (Tocilizumab) and stimulating the immune system (Interferons). On the other hand, TCM also proposed a number of treatment methods for COVID-19 with new scientific approaches identifying their antiviral and immunomodulatory activities. The novel combination of Western medicine and TCM can be proposed by analyzing their respective molecular targets. RESULTS Although TCM is not generally accepted in the Western community because of the general lack of knowledge on their detailed mechanisms, studies and clinical trials suggest that TCM could be beneficial in combating COVID-19. CONCLUSION Based on the principle of combining TCM and Western medicine, two combinations are tested effective in clinical trials, and three possible combinations that might be effective are proposed in the paper.
Collapse
Affiliation(s)
- Zirui Zeng
- International Department, The Affiliated High School of South China Normal University, Guangzhou, Guangdong, China
- University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
21
|
Genetic Study of SARS-CoV-2 Non Structural Protein 12 in COVID-19 Patients Non Responders to Remdesivir. Microbiol Spectr 2022; 10:e0244822. [PMID: 36354320 PMCID: PMC9769853 DOI: 10.1128/spectrum.02448-22] [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] [Indexed: 11/12/2022] Open
Abstract
Remdesivir (RDV) was the first antiviral drug approved by the FDA to treat severe coronavirus disease-2019 (COVID-19) patients. RDV inhibits SARS-CoV-2 replication by stalling the non structural protein 12 (nsp12) subunit of the RNA-dependent RNA polymerase (RdRp). No evidence of global widespread RDV-resistance mutations has been reported, however, defining genetic pathways to RDV resistance and determining emergent mutations prior and subsequent antiviral therapy in clinical settings is necessary. This study identified 57/149 (38.3%) patients who did not respond to one course (5-days) (n = 36/111, 32.4%) or prolonged (5 to 20 days) (n = 21/38, 55.3%) RDV therapy by subgenomic RNA detection. Genetic variants in the nsp12 gene were detected in 29/49 (59.2%) non responder patients by Illumina sequencing, including the de novo E83D mutation that emerged in an immunosuppressed patient after receiving 10 + 8 days of RDV, and the L838I detected at baseline and/or after prolonged RDV treatment in 9/49 (18.4%) non responder subjects. Although 3D protein modeling predicted no interference with RDV, the amino acid substitutions detected in the nsp12 involved changes on the electrostatic outer surface and in secondary structures that may alter antiviral response. It is important for health surveillance to study potential mutations associated with drug resistance as well as the benefit of RDV retreatment, especially in immunosuppressed patients and in those with persistent replication. IMPORTANCE This study provides clinical and microbiologic data of an extended population of hospitalized patients for COVID-19 pneumonia who experienced treatment failure, detected by the presence of subgenomic RNA (sgRNA). The genetic variants found in the nsp12 pharmacological target of RDV bring into focus the importance of monitoring emergent mutations, one of the objectives of the World Health Organization (WHO) for health surveillance. These mutations become even more crucial as RDV keeps being prescribed and new molecules are being repurposed for the treatment of COVID-19. The present article offers new perspectives for the clinical management of non responder patients treated and retreated with RDV and emphasizes the need of further research of the benefit of combinatorial therapies and RDV retreatment, especially in immunosuppressed patients with persistent replication after therapy.
Collapse
|
22
|
Xanthi Z, Vasiliki P, Stavros A. Apheresis and COVID-19 in intensive care unit (ICU). Transfus Apher Sci 2022; 61:103593. [PMID: 36335074 PMCID: PMC9624107 DOI: 10.1016/j.transci.2022.103593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The first known case was identified in Wuhan, China, in December 2019. The disease has since spread worldwide, and on March 2020 the World Health Organization (WHO) declared it as pandemic, causing a public health crisis. Symptoms of COVID-19 are variable, ranging from mild symptoms like fever, cough, and fatigue to severe illness. Elderly patients and those with comorbidities like cardiovascular disease, diabetes, chronic respiratory disease, or cancer are more likely to develop severe forms of the disease. Asymptomatic infections have been well documented. Accumulating evidence suggests that the severity of COVID-19 is due to high levels of circulating inflammatory mediators including cytokines and chemokines leading to cytokine storm syndrome (CSS). Patients are admitted in ICU with severe respiratory failure, but can also develop acute renal failure and multi organ failure. Advances in science and technology have permitted the development of more sophisticated therapies such as extracorporeal organ support (ECOS) therapies that includes renal replacement therapies (RRTs), venoarterial (VA) or veno-venous (VV) extracorporeal membrane Oxygenation (ECMO), extracorporeal CO2 removal (ECCO2R), liver support systems, hemoperfusion, and various blood purification devices, for the treatment of ARDS and septic shock.
Collapse
|
23
|
Özen S, Gülhan B, Yüksek SK, Güney AY, Erat T, Yahşi A, Güneş Ö, Güder L, Mustafaoğlu Ö, Bayraktar P, Üçkardeş F, Emeksiz S, Bayhan Gİ, Parlakay AÖ. Early Short-Term Use of Different Doses of Corticosteroid in Hospitalized Pediatric Patients with Coronavirus Disease 2019 Pneumonia. J PEDIAT INF DIS-GER 2022. [DOI: 10.1055/s-0042-1759529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Abstract
Objective Encouraged by reports of favorable outcomes following the use of corticosteroids in patients with moderate-to-severe coronavirus 2019 (COVID-19) pneumonia, we aimed to present our experience with early short-term corticosteroid use at our center in pediatric patients with COVID-19 pneumonia.
Methods One hundred and twenty-nine pediatric patients were included in the study. Patients were divided into four groups according to the type and dose of corticosteroids given: Group 1 (those receiving dexamethasone 0.15 mg/kg/d); Group 2 (those receiving methylprednisolone 1 mg/kg/d); Group 3 (those receiving methylprednisolone 2 mg/kg/d); and Group 4 (those receiving pulse methylprednisolone 10–30 mg/kg/d).
Results Of 129 patients, 19 (14.7%) patients were assigned to Group 1, 30 (23.3%) patients to Group 2, 30 (23.3%) patients to Group 3, and 50 (38.8%) patients to Group 4. Thirty-two (24.8%) patients were followed in the pediatric intensive care unit (PICU), of whom 13 (10%) required mechanical ventilation, and 7 (%5.4) died. In Group 4, the hospitalization length was significantly longer than in other groups (p < 0.001, p < 0.001). No significant difference was found among the groups in terms of mortality (p = 0.15). The most common comorbidity was obesity (33%). A significant association was found between the presence of comorbidity and mortality (p < 0.001). All patients who died had an underlying disease. Cerebral palsy was the most common underlying disease among the patients who died. Worsening of lymphopenia was significant in patients with severe COVID-19 pneumonia at the time of transfer to the PICU (p = 0.011).
Conclusion Although children usually have a milder course of COVID-19 than adults, underlying diseases and obesity increase the severity of disease manifestations also in children. Further studies are needed to define the exact role of corticosteroids in COVID-19 patients.
Collapse
Affiliation(s)
- Seval Özen
- Pediatric Infectious Diseases Unit, Ankara City Hospital, Ankara, Turkey
| | - Belgin Gülhan
- Pediatric Infectious Diseases Unit, Ankara City Hospital, Ankara, Turkey
| | | | - Ahmet Yasin Güney
- Pediatric Infectious Diseases Unit, Ankara City Hospital, Ankara, Turkey
| | - Tuğba Erat
- Pediatric Infectious Diseases Unit, Ankara City Hospital, Ankara, Turkey
| | - Aysun Yahşi
- Pediatric Infectious Diseases Unit, Ankara City Hospital, Ankara, Turkey
| | - Ömer Güneş
- Pediatric Infectious Diseases Unit, Ankara City Hospital, Ankara, Turkey
| | - Latife Güder
- Pediatric Infectious Diseases Unit, Ankara City Hospital, Ankara, Turkey
| | - Özlem Mustafaoğlu
- Pediatric Infectious Diseases Unit, Ankara City Hospital, Ankara, Turkey
| | - Pınar Bayraktar
- Pediatric Infectious Diseases Unit, Ankara City Hospital, Ankara, Turkey
| | - Fatih Üçkardeş
- Adıyaman University Department of Biostatistics and Medical Informatics, Adıyaman, Turkey
| | - Serhat Emeksiz
- Department of Pediatric Intensive Care Unit, Ankara City Hospital, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Gülsüm İclal Bayhan
- Pediatric Infectious Diseases Unit, Ankara City Hospital, Yildirim Beyazit University, Ankara, Turkey
| | - Aslınur Özkaya Parlakay
- Pediatric Infectious Diseases Unit, Ankara City Hospital, Yildirim Beyazit University, Ankara, Turkey
| |
Collapse
|
24
|
Vulturar DM, Neag MA, Vesa ȘC, Maierean AD, Gherman D, Buzoianu AD, Orăsan OH, Todea DA. Therapeutic Efficacy and Outcomes of Remdesivir versus Remdesivir with Tocilizumab in Severe SARS-CoV-2 Infection. Int J Mol Sci 2022; 23:ijms232214462. [PMID: 36430945 PMCID: PMC9698366 DOI: 10.3390/ijms232214462] [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: 08/16/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) generated many challenges to find an effective drug combination for hospitalized patients with severe forms of coronavirus disease 2019 (COVID-19) pneumonia. We conducted a retrospective cohort study, including 182 patients with severe COVID-19 pneumonia hospitalized between March and October 2021 in a Pneumology Hospital from Cluj-Napoca, Romania. Among patients treated with standard of care, 100 patients received remdesivir (R group) and 82 patients received the combination of remdesivir plus tocilizumab (RT group). We compared the clinical outcomes, the inflammatory markers, superinfections, oxygen requirement, intensive care unit (ICU) admission and mortality rate before drug administration and 7 days after in R group and RT group. Borg score and oxygen support showed an improvement in the R group (p < 0.005). Neutrophiles, C-reactive protein (CRP) and serum ferritin levels decreased significantly in RT group but with a higher rate of superinfection in this group. ICU admission and death did not differ significantly between groups. The combination of remdesivir plus tocilizumab led to a significantly improvement in the inflammatory markers and a decrease in the oxygen requirement. Although the superinfection rate was higher in RT group than in R group, no significant difference was found in the ICU admission and mortality rate between the groups.
Collapse
Affiliation(s)
- Damiana-Maria Vulturar
- Department of Pneumology, Iuliu Hațieganu University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania
| | - Maria Adriana Neag
- Pharmacology, Toxicology and Clinical Pharmacology Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Correspondence:
| | - Ștefan Cristian Vesa
- Pharmacology, Toxicology and Clinical Pharmacology Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Anca-Diana Maierean
- Department of Pneumology, Iuliu Hațieganu University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania
| | - Diana Gherman
- Department of Radiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Anca Dana Buzoianu
- Pharmacology, Toxicology and Clinical Pharmacology Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Olga Hilda Orăsan
- 5th Department Internal Medicine, 4th Medical Clinic, Iuliu Hațieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania
| | - Doina-Adina Todea
- Department of Pneumology, Iuliu Hațieganu University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania
| |
Collapse
|
25
|
Clinical Protocol for Early Treatment of COVID-19 in a real-world scenario: results of a series of patients. MEDICINA CLÍNICA PRÁCTICA 2022. [PMCID: PMC9492515 DOI: 10.1016/j.mcpsp.2022.100346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Introduction Despite the advance in vaccination, the SARS-CoV-2 infection remains a challenge for the medical community. Outpatient and hospital therapy for COVID-19 are still improving. Our study aimed to report the results of a series of patients with COVID-19 who participated in an outpatient treatment protocol since the first clinical manifestation. Methods A case series report of individuals aged ≥ 18 years with clinical symptoms and a confirmed test for COVID-19 submitted to a treatment protocol. Patients were enrolled between May and September 2020 and followed for at least 15 days. The assessed clinical outcomes were the need for hospitalization, admission to the intensive care unit, orotracheal intubation, and death. Results We studied a 116 patients. The mean age was 48 ± 14 years. Females formed 53%. The main comorbidities wereobesity (15.5%), systemic arterial hypertension (10.3%) ,type II diabetes (6%), and lung diseases (6.0%). Temperature > 37.7 °C (51.7%), cough (55.2%), myalgia (37.1%), headache (37.9%), and fatigue (34.5%) were the most frequent signs and symptoms. According to different disease staging, the most administered drugs were: azithromycin, ivermectin, corticosteroid, antibiotics, and anticoagulants. There was no death, and hospitalization accounted for only 8.6% of the patients (1 in ICU); none required orotracheal intubation. The mean length of hospital stay was 5.8 days.
Collapse
|
26
|
COVID-19-Associated Mucormycosis: A Matter of Concern Amid the SARS-CoV-2 Pandemic. Vaccines (Basel) 2022; 10:vaccines10081266. [PMID: 36016154 PMCID: PMC9415927 DOI: 10.3390/vaccines10081266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 12/16/2022] Open
Abstract
Mucormycosis is an invasive fungal infection caused by fungi belonging to order Mucorales. Recently, with the increase in COVID-19 infections, mucormycosis infections have become a matter of concern globally, because of the high morbidity and mortality rates associated with them. Due to the association of mucormycosis with COVID-19 disease, it has been termed COVID-19-associated mucormycosis (CAM). In the present review, we focus on mucormycosis incidence, pathophysiology, risk factors, immune dysfunction, interactions of Mucorales with endothelial cells, and the possible role of iron in Mucorales growth. We review the limitations associated with current diagnostic procedures and the requirement for more specific, cost-effective, convenient, and sensitive assays, such as PCR-based assays and monoclonal antibody-based assays for the effective diagnosis of mucormycosis. We discuss the current treatment options involving antifungal drug therapies, adjunctive therapy, surgical treatment, and their limitations. We also review the importance of nutraceuticals-based therapy for the prevention as well as treatment of mucormycosis. Our review also highlights the need to explore the potential of novel immunotherapeutics, which include antibody-based therapy, cytokine-based therapy, and combination/synergistic antifungal therapy, as treatment options for mucormycosis. In summary, this review provides a complete overview of COVID-19-associated mucormycosis, addressing the current research gaps and future developments required in the field.
Collapse
|
27
|
Aguilera C, Danés I, Guillén E, Vimes A, Bosch M, Cereza G, Sánchez-Montalvá A, Campos-Varela I, Miarons M, Mestre-Torres J, Agustí A. Safety of Drugs Used during the First Wave of COVID-19: A Hospital-Registry-Based Study. Diagnostics (Basel) 2022; 12:diagnostics12071612. [PMID: 35885517 PMCID: PMC9316110 DOI: 10.3390/diagnostics12071612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/02/2022] [Accepted: 06/29/2022] [Indexed: 12/23/2022] Open
Abstract
The emergency of the coronavirus disease 2019 (COVID-19) pandemic led to the off-label use of drugs without data on their toxicity profiles in patients with COVID-19, or on their concomitant use. Patients included in the COVID-19 Patient Registry of a tertiary hospital during the first wave were analyzed to evaluate the adverse drug reactions (ADRs) with the selected treatments. Twenty-one percent of patients (197 out of 933) had at least one ADR, with a total of 240 ADRs. Patients with ADRs were more commonly treated with multiple drugs for COVID-19 infection than patients without ADRs (p < 0.001). They were younger (median 62 years vs. 70.1 years; p < 0.001) and took less medication regularly (69.5% vs. 75.7%; p = 0.031). The most frequent ADRs were gastrointestinal (67.1%), hepatobiliary (10.8%), and cardiac disorders (3.3%). Drugs more frequently involved included lopinavir/ritonavir (82.2%), hydroxychloroquine (72.1%), and azithromycin (66.5%). Although most ADRs recovered without sequelae, fatal cases were described, even though the role of the disease could not be completely ruled out. In similar situations, efforts should be made to use the drugs in the context of clinical trials, and to limit off-label use to those drugs with a better benefit/risk profile in specific situations and for patients at high risk of poor disease prognosis.
Collapse
Affiliation(s)
- Cristina Aguilera
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (E.G.); (A.V.); (M.B.); (A.A.)
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain;
- Immunomediated Diseases and Innovative Therapies Group, Vall d’Hebron Research Institute, 08035 Barcelona, Spain
| | - Immaculada Danés
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (E.G.); (A.V.); (M.B.); (A.A.)
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain;
- Immunomediated Diseases and Innovative Therapies Group, Vall d’Hebron Research Institute, 08035 Barcelona, Spain
- Correspondence:
| | - Elena Guillén
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (E.G.); (A.V.); (M.B.); (A.A.)
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain;
| | - Alba Vimes
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (E.G.); (A.V.); (M.B.); (A.A.)
| | - Montserrat Bosch
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (E.G.); (A.V.); (M.B.); (A.A.)
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain;
- Immunomediated Diseases and Innovative Therapies Group, Vall d’Hebron Research Institute, 08035 Barcelona, Spain
| | - Gloria Cereza
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain;
- Immunomediated Diseases and Innovative Therapies Group, Vall d’Hebron Research Institute, 08035 Barcelona, Spain
- Catalan Institute of Pharmacology Foundation, Vall d’Hebron Hospital Universitari, 08035 Barcelona, Spain
| | - Adrián Sánchez-Montalvá
- International Health and Tuberculosis Unit, Infectious Diseases Department, National Referral Centre for Tropical Diseases, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain;
| | - Isabel Campos-Varela
- Liver Unit, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Marta Miarons
- Pharmacy Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain;
| | - Jaume Mestre-Torres
- Internal Medicine Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain;
| | - Antònia Agustí
- Clinical Pharmacology Service, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (E.G.); (A.V.); (M.B.); (A.A.)
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain;
- Immunomediated Diseases and Innovative Therapies Group, Vall d’Hebron Research Institute, 08035 Barcelona, Spain
| |
Collapse
|
28
|
Domán M, Bányai K. COVID-19-Associated Fungal Infections: An Urgent Need for Alternative Therapeutic Approach? Front Microbiol 2022; 13:919501. [PMID: 35756020 PMCID: PMC9218862 DOI: 10.3389/fmicb.2022.919501] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/19/2022] [Indexed: 12/19/2022] Open
Abstract
Secondary fungal infections may complicate the clinical course of patients affected by viral respiratory diseases, especially those admitted to intensive care unit. Hospitalized COVID-19 patients are at increased risk of fungal co-infections exacerbating the prognosis of disease due to misdiagnosis that often result in treatment failure and high mortality rate. COVID-19-associated fungal infections caused by predominantly Aspergillus and Candida species, and fungi of the order Mucorales have been reported from several countries to become significant challenge for healthcare system. Early diagnosis and adequate antifungal therapy is essential to improve clinical outcomes, however, drug resistance shows a rising trend highlighting the need for alternative therapeutic agents. The purpose of this review is to summarize the current knowledge on COVID-19-associated mycoses, treatment strategies and the most recent advancements in antifungal drug development focusing on peptides with antifungal activity.
Collapse
Affiliation(s)
- Marianna Domán
- Veterinary Medical Research Institute, Budapest, Hungary
| | - Krisztián Bányai
- Veterinary Medical Research Institute, Budapest, Hungary.,Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary
| |
Collapse
|
29
|
Li JT, Zhang YD, Song XR, Li RJ, Yang WL, Tian M, Zhang SF, Cao GH, Song LL, Chen YM, Liu CH. The mechanism and effects of remdesivir-induced developmental toxicity in zebrafish: Blood flow dysfunction and behavioral alterations. J Appl Toxicol 2022; 42:1688-1700. [PMID: 35560222 DOI: 10.1002/jat.4336] [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: 04/24/2021] [Revised: 04/01/2022] [Accepted: 04/30/2022] [Indexed: 11/11/2022]
Abstract
The antiviral drug remdesivir has been used to treat the growing number of coronavirus disease 2019 (COVID-19) patients. However, the drug is mainly excreted through urine and feces and introduced into the environment to affect non-target organisms, including fish, which has raised concerns about potential ecotoxicological effects on aquatic organisms. Moreover, studies on the ecological impacts of remdesivir on aquatic environments have not been reported. Here, we aimed to explore the toxicological impacts of microinjection of remdesivir on zebrafish early embryonic development and larvae and the associated mechanism. We found that 100 μM remdesivir delayed epiboly and impaired convergent movement of embryos during gastrulation, and dose-dependent increases in mortality and malformation were observed in remdesivir-treated embryos. Moreover, 10-100 μM remdesivir decreased blood flow and swimming velocity and altered the behavior of larvae. In terms of molecular mechanisms, eighty differentially expressed genes (DEGs) were identified by transcriptome analysis in the remdesivir-treated group. Some of these DEGs, such as manf, kif3a, hnf1ba, rgn, prkcz, egr1, fosab, nr4a1, and ptgs2b, were mainly involved in early embryonic development, neuronal developmental disorders, vascular disease and the blood flow pathway. These data reveal that remdesivir can impair early embryonic development, blood flow and behavior of zebrafish embryos/larvae, probably due to alterations at the transcriptome level. This study suggests that it is important to avoid the discharge of remdesivir to aquatic ecosystems and provides a theoretical foundation to hinder remdesivir-induced ecotoxicity to aquatic environments.
Collapse
Affiliation(s)
- Ji-Tong Li
- Henan Neurodevelopment Engineering Research Center for Children; Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China.,Department of Nephrology and Rheumatology, Children's Hospital Affiliated to Zhengzhou University; Zhengzhou Key Laboratory of Pediatric Kidney Disease Research, Zhengzhou, China
| | - Yao-Dong Zhang
- Henan Neurodevelopment Engineering Research Center for Children; Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Xiao-Rui Song
- Henan Neurodevelopment Engineering Research Center for Children; Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Rui-Jing Li
- Henan Neurodevelopment Engineering Research Center for Children; Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Wei-Li Yang
- Henan Neurodevelopment Engineering Research Center for Children; Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Ming Tian
- Department of Nephrology and Rheumatology, Children's Hospital Affiliated to Zhengzhou University; Zhengzhou Key Laboratory of Pediatric Kidney Disease Research, Zhengzhou, China
| | - Shu-Feng Zhang
- Department of Nephrology and Rheumatology, Children's Hospital Affiliated to Zhengzhou University; Zhengzhou Key Laboratory of Pediatric Kidney Disease Research, Zhengzhou, China
| | - Guang-Hai Cao
- Department of Nephrology and Rheumatology, Children's Hospital Affiliated to Zhengzhou University; Zhengzhou Key Laboratory of Pediatric Kidney Disease Research, Zhengzhou, China
| | - Lu-Lu Song
- School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yu-Ming Chen
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Cui-Hua Liu
- Henan Neurodevelopment Engineering Research Center for Children; Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China.,Department of Nephrology and Rheumatology, Children's Hospital Affiliated to Zhengzhou University; Zhengzhou Key Laboratory of Pediatric Kidney Disease Research, Zhengzhou, China
| |
Collapse
|
30
|
Mir I, Aamir S, Shah SRH, Shahid M, Amin I, Afzal S, Nawaz A, Khan MU, Idrees M. Immune-related therapeutics: an update on antiviral drugs and vaccines to tackle the COVID-19 pandemic. Osong Public Health Res Perspect 2022; 13:84-100. [PMID: 35538681 PMCID: PMC9091641 DOI: 10.24171/j.phrp.2022.0024] [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: 01/17/2022] [Revised: 03/15/2022] [Accepted: 04/10/2022] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic rapidly spread globally. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes COVID-19, is a positive-sense single-stranded RNA virus with a reported fatality rate ranging from 1% to 7%, and people with immune-compromised conditions, children, and older adults are particularly vulnerable. Respiratory failure and cytokine storm-induced multiple organ failure are the major causes of death. This article highlights the innate and adaptive immune mechanisms of host cells activated in response to SARS-CoV-2 infection and possible therapeutic approaches against COVID-19. Some potential drugs proven to be effective for other viral diseases are under clinical trials now for use against COVID-19. Examples include inhibitors of RNA-dependent RNA polymerase (remdesivir, favipiravir, ribavirin), viral protein synthesis (ivermectin, lopinavir/ ritonavir), and fusion of the viral membrane with host cells (chloroquine, hydroxychloroquine, nitazoxanide, and umifenovir). This article also presents the intellectual groundwork for the ongoing development of vaccines in preclinical and clinical trials, explaining potential candidates (live attenuated-whole virus vaccines, inactivated vaccines, subunit vaccines, DNAbased vaccines, protein-based vaccines, nanoparticle-based vaccines, virus-like particles and mRNA-based vaccines). Designing and developing an effective vaccine (both prophylactic and therapeutic) would be a long-term solution and the most effective way to eliminate the COVID-19 pandemic.
Collapse
Affiliation(s)
- Iqra Mir
- Division of Molecular Virology and Infectious Diseases, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Sania Aamir
- Division of Molecular Virology and Infectious Diseases, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Syed Rizwan Hussain Shah
- Division of Molecular Virology and Infectious Diseases, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Shahid
- Division of Molecular Virology and Infectious Diseases, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Iram Amin
- Division of Molecular Virology and Infectious Diseases, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Samia Afzal
- Division of Molecular Virology and Infectious Diseases, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Amjad Nawaz
- Division of Molecular Virology and Infectious Diseases, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Umer Khan
- University Institute of Medical lab Technology, Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan
| | - Muhammad Idrees
- Division of Molecular Virology and Infectious Diseases, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| |
Collapse
|
31
|
Elfaki I. The Impact of the Coronavirus (COVID-19) Infection on the Drug-Metabolizing Enzymes Cytochrome P450s. Drug Metab Lett 2022; 15:DML-EPUB-122095. [PMID: 35362390 DOI: 10.2174/1872312815666220331142046] [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: 12/13/2021] [Revised: 01/10/2022] [Accepted: 02/21/2022] [Indexed: 11/22/2022]
Abstract
Coronaviruses cause disease in human and animals. In 2019 a novel coronavirus was first characterized in Wuhan, China. It causes acute respiratory disease and designated the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or COVID-19. The COVID-19 spread to all cities of China, and in 2020 to the whole world. Patients with COVID-19 may recover without medical treatment. However, some patients need medical care. The Cytochrome p450s (CYP450s) are large superfamily of enzymes catalyze the metabolism of endogenous substrates and xenobiotics. CYP450s catalyze the biotransformation of 80% of the drug in clinical use. The CYP450 present in liver, lungs, intestine and other tissues. COVID-19 has been reported to decrease the activity of certain isoforms of CYP450s in an isoform specific manner. Furthermore, the COVID-19 infection decreases the liver functions including the drug clearance or detoxification medicated by the CYP450s. The healthcare providers should be aware of this disease-drug interaction when prescribing drugs for treatment of COVID-19 and other comorbidities.
Collapse
Affiliation(s)
- Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Kingdom of Saudi Arabia
| |
Collapse
|
32
|
MUTLU P, MİRİCİ A, GÖNLÜGÜR U, OZTOPRAK B, ÖZER Ş, REŞORLU M, AKÇALI A, ÜLKER ÇAKIR D, DOĞAN CR. Evaluating the clinical, radiological, microbiological, biochemical parameters and the treatment response in COVID-19 pneumonia. JOURNAL OF HEALTH SCIENCES AND MEDICINE 2022. [DOI: 10.32322/jhsm.1035790] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
33
|
Biswas M, Sawajan N, Rungrotmongkol T, Sanachai K, Ershadian M, Sukasem C. Pharmacogenetics and Precision Medicine Approaches for the Improvement of COVID-19 Therapies. Front Pharmacol 2022; 13:835136. [PMID: 35250581 PMCID: PMC8894812 DOI: 10.3389/fphar.2022.835136] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 01/18/2023] Open
Abstract
Many drugs are being administered to tackle coronavirus disease 2019 (COVID-19) pandemic situations without establishing clinical effectiveness or tailoring safety. A repurposing strategy might be more effective and successful if pharmacogenetic interventions are being considered in future clinical studies/trials. Although it is very unlikely that there are almost no pharmacogenetic data for COVID-19 drugs, however, from inferring the pharmacokinetic (PK)/pharmacodynamic(PD) properties and some pharmacogenetic evidence in other diseases/clinical conditions, it is highly likely that pharmacogenetic associations are also feasible in at least some COVID-19 drugs. We strongly mandate to undertake a pharmacogenetic assessment for at least these drug-gene pairs (atazanavir-UGT1A1, ABCB1, SLCO1B1, APOA5; efavirenz-CYP2B6; nevirapine-HLA, CYP2B6, ABCB1; lopinavir-SLCO1B3, ABCC2; ribavirin-SLC28A2; tocilizumab-FCGR3A; ivermectin-ABCB1; oseltamivir-CES1, ABCB1; clopidogrel-CYP2C19, ABCB1, warfarin-CYP2C9, VKORC1; non-steroidal anti-inflammatory drugs (NSAIDs)-CYP2C9) in COVID-19 patients for advancing precision medicine. Molecular docking and computational studies are promising to achieve new therapeutics against SARS-CoV-2 infection. The current situation in the discovery of anti-SARS-CoV-2 agents at four important targets from in silico studies has been described and summarized in this review. Although natural occurring compounds from different herbs against SARS-CoV-2 infection are favorable, however, accurate experimental investigation of these compounds is warranted to provide insightful information. Moreover, clinical considerations of drug-drug interactions (DDIs) and drug-herb interactions (DHIs) of the existing repurposed drugs along with pharmacogenetic (e.g., efavirenz and CYP2B6) and herbogenetic (e.g., andrographolide and CYP2C9) interventions, collectively called multifactorial drug-gene interactions (DGIs), may further accelerate the development of precision COVID-19 therapies in the real-world clinical settings.
Collapse
Affiliation(s)
- Mohitosh Biswas
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
- Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh
| | - Nares Sawajan
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
- Department of Pathology, School of Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | - Thanyada Rungrotmongkol
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Kamonpan Sanachai
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Maliheh Ershadian
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
| | - Chonlaphat Sukasem
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
- Pharmacogenomics and Precision Medicine, The Preventive Genomics and Family Check-up Services Center, Bumrungrad International Hospital, Bangkok, Thailand
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
34
|
Identification of Differentially Expressed Genes in COVID-19 and Integrated Bioinformatics Analysis of Signaling Pathways. Genet Res (Camb) 2022; 2021:2728757. [PMID: 35002537 PMCID: PMC8710042 DOI: 10.1155/2021/2728757] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is acutely infectious pneumonia. Currently, the specific causes and treatment targets of COVID-19 are still unclear. Herein, comprehensive bioinformatics methods were employed to analyze the hub genes in COVID-19 and tried to reveal its potential mechanisms. First of all, 34 groups of COVID-19 lung tissues and 17 other diseases' lung tissues were selected from the GSE151764 gene expression profile for research. According to the analysis of the DEGs (differentially expressed genes) in the samples using the limma software package, 84 upregulated DEGs and 46 downregulated DEGs were obtained. Later, by the Database for Annotation, Visualization, and Integrated Discovery (DAVID), they were enriched in the Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. It was found that the upregulated DEGs were enriched in the type I interferon signaling pathway, AGE-RAGE signaling pathway in diabetic complications, coronavirus disease, etc. Downregulated DEGs were in cellular response to cytokine stimulus, IL-17 signaling pathway, FoxO signaling pathway, etc. Then, based on GSEA, the enrichment of the gene set in the sample was analyzed in the GO terms, and the gene set was enriched in the positive regulation of myeloid leukocyte cytokine production involved in immune response, programmed necrotic cell death, translesion synthesis, necroptotic process, and condensed nuclear chromosome. Finally, with the help of STRING tools, the PPI (protein-protein interaction) network diagrams of DEGs were constructed. With degree ≥13 as the cutoff degree, 3 upregulated hub genes (ISG15, FN1, and HLA-G) and 4 downregulated hub genes (FOXP3, CXCR4, MMP9, and CD69) were screened out for high degree. All these findings will help us to understand the potential molecular mechanisms of COVID-19, which is also of great significance for its diagnosis and prevention.
Collapse
|
35
|
Soliman OM, Moeen SM, Abbas YA, Kamel EZ. The impact of dexamethasone versus methylprednisolone upon neutrophil/lymphocyte ratio in COVID-19 patients admitted to ICU and its implication upon mortality. EGYPTIAN JOURNAL OF ANAESTHESIA 2022. [DOI: 10.1080/11101849.2021.2024985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- OM Soliman
- Faculty of Medicine, Assiut University, Assiut, Egypt
| | - SM Moeen
- Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Yara A. Abbas
- Faculty of Medicine, Assiut University, Assiut, Egypt
| | | |
Collapse
|
36
|
Santos LH, Kronenberger T, Almeida RG, Silva EB, Rocha REO, Oliveira JC, Barreto LV, Skinner D, Fajtová P, Giardini MA, Woodworth B, Bardine C, Lourenço AL, Craik CS, Poso A, Podust LM, McKerrow JH, Siqueira-Neto JL, O'Donoghue AJ, da Silva Júnior EN, Ferreira RS. Structure-based identification of naphthoquinones and derivatives as novel inhibitors of main protease Mpro and papain-like protease PLpro of SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.01.05.475095. [PMID: 35018373 PMCID: PMC8750648 DOI: 10.1101/2022.01.05.475095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The worldwide COVID-19 pandemic caused by the coronavirus SARS-CoV-2 urgently demands novel direct antiviral treatments. The main protease (Mpro) and papain-like protease (PLpro) are attractive drug targets among coronaviruses due to their essential role in processing the polyproteins translated from the viral RNA. In the present work, we virtually screened 688 naphthoquinoidal compounds and derivatives against Mpro of SARS-CoV-2. Twenty-four derivatives were selected and evaluated in biochemical assays against Mpro using a novel fluorogenic substrate. In parallel, these compounds were also assayed with SARS-CoV-2 PLpro. Four compounds inhibited Mpro with half-maximal inhibitory concentration (IC 50 ) values between 0.41 µM and 66 µM. In addition, eight compounds inhibited PLpro with IC 50 ranging from 1.7 µM to 46 µM. Molecular dynamics simulations suggest stable binding modes for Mpro inhibitors with frequent interactions with residues in the S1 and S2 pockets of the active site. For two PLpro inhibitors, interactions occur in the S3 and S4 pockets. In summary, our structure-based computational and biochemical approach identified novel naphthoquinonal scaffolds that can be further explored as SARS-CoV-2 antivirals.
Collapse
|
37
|
Tio SY, Williams E, Worth LJ, Deane AM, Bond K, Slavin MA, Sasadeusz J. Invasive pulmonary aspergillosis in critically ill patients with COVID-19 in Australia: implications for screening and treatment. Intern Med J 2021; 51:2129-2132. [PMID: 34939293 DOI: 10.1111/imj.15602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/15/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022]
Abstract
We report four cases of invasive pulmonary aspergillus co-infection in patients with coronavirus disease 2019 (COVID-19) infection and acute respiratory distress syndrome requiring intensive care unit (ICU) admission. Aspergillus fumigatus and Aspergillus terreus were isolated, with early infection onset following ICU admission. Clinicians should be aware of invasive pulmonary aspergillosis in ICU patients with COVID-19 infection, particularly those receiving dexamethasone. We propose screening of these high-risk patients with twice-weekly fungal culture from tracheal aspirate and, if feasible, Aspergillus polymerase chain reaction. Diagnosis is challenging and antifungal treatment should be considered in critically ill patients who have new or worsening pulmonary changes on chest imaging and mycological evidence of infection.
Collapse
Affiliation(s)
- Shio Yen Tio
- Victorian Infectious Diseases Service (VIDS), The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,National Centre for Infections in Cancer, Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Eloise Williams
- Department of Microbiology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Leon J Worth
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,National Centre for Infections in Cancer, Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Adam M Deane
- The University of Melbourne, Department of Critical Care, Melbourne Medical School, Melbourne, Australia
| | - Katherine Bond
- Department of Microbiology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Monica A Slavin
- Victorian Infectious Diseases Service (VIDS), The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,National Centre for Infections in Cancer, Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Joe Sasadeusz
- Victorian Infectious Diseases Service (VIDS), The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
38
|
Aljuhani O, Al Sulaiman K, Alshabasy A, Eljaaly K, Al Shaya AI, Noureldeen H, Aboudeif M, Al Dosari B, Alkhalaf A, Korayem GB, Aleissa MM, Badreldin HA, Al Harbi S, Alhammad A, Vishwakarma R. Association between tocilizumab and emerging multidrug-resistant organisms in critically ill patients with COVID-19: A multicenter, retrospective cohort study. BMC Infect Dis 2021; 21:1127. [PMID: 34724920 PMCID: PMC8559694 DOI: 10.1186/s12879-021-06813-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/22/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Tocilizumab is an IgG1 class recombinant humanized monoclonal antibody that directly inhibits the IL-6 receptor. Several randomized clinical trials have evaluated its safety and efficacy in patients with coronavirus disease 2019 (COVID-19), and these studies demonstrate conflicting results. Our study aimed to determine the association between tocilizumab treatment and microbial isolation and emergence of multidrug-resistant bacteria in critically ill patients with COVID-19. METHODS A multicenter retrospective cohort study was conducted at two tertiary government hospitals in Saudi Arabia. All critically ill patients admitted to intensive care units with a positive COVID-19 PCR test between March 1 and December 31, 2020, who met study criteria were included. Patients who received tocilizumab were compared to those who did not receive it. RESULTS A total of 738 patients who met our inclusion criteria were included in the analysis. Of these, 262 (35.5%) received tocilizumab, and 476 (64.5%) were included in the control group. Patients who received tocilizumab had higher odds for microbial isolation (OR 1.34; 95% CI 0.91-1.94, p = 0.13); however, the difference was not statistically significant. Development of resistant organisms (OR 1.00; 95% CI 0.51-1.98, p = 0.99) or detection of carbapenem-resistant Enterobacteriaceae (CRE) (OR 0.67; 95% CI 0.29-1.54, p = 0.34) was not statistically significant between the two groups. CONCLUSIONS Tocilizumab use in critically ill patients with COVID-19 is not associated with higher microbial isolation, the emergence of resistant organisms, or the detection of CRE organisms.
Collapse
Affiliation(s)
- Ohoud Aljuhani
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, P. O. Box 80260, Jeddah, 21589, Saudi Arabia.
| | - Khalid Al Sulaiman
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Adel Alshabasy
- Department of Anesthesia and Intensive Care, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Anesthesia and Intensive Care, Ain Shams University, Cairo, Egypt
| | - Khalid Eljaaly
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, P. O. Box 80260, Jeddah, 21589, Saudi Arabia
- College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Abdulrahman I Al Shaya
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haytham Noureldeen
- Department of Anesthesia and Intensive Care, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Anesthesia and Intensive Care, Ain Shams University, Cairo, Egypt
| | - Mohammed Aboudeif
- Department of Anesthesia and Intensive Care, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Bodoor Al Dosari
- Pharmaceutical Care Department, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Amina Alkhalaf
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, P. O. Box 80260, Jeddah, 21589, Saudi Arabia
| | - Ghazwa B Korayem
- College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Muneera M Aleissa
- College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
- Brigham and Women's Hospital, Boston, MA, USA
| | - Hisham A Badreldin
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Shmeylan Al Harbi
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Abdullah Alhammad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ramesh Vishwakarma
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
39
|
Elshafei A, Khidr EG, El-Husseiny AA, Gomaa MH. RAAS, ACE2 and COVID-19; a mechanistic review. Saudi J Biol Sci 2021; 28:6465-6470. [PMID: 34305426 PMCID: PMC8270731 DOI: 10.1016/j.sjbs.2021.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/26/2021] [Accepted: 07/04/2021] [Indexed: 01/08/2023] Open
Abstract
The use of angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) in coronavirus disease 2019 (COVID-19) patients has been claimed as associated with the risk of COVID-19 infection and its subsequent morbidities and mortalities. These claims were resulting from the possibility of upregulating the expression of angiotensin-converting enzyme 2 (ACE2), facilitation of SARS-CoV-2 entry, and increasing the susceptibility of infection in such treated cardiovascular patients. ACE2 and renin-angiotensin-aldosterone system (RAAS) products have a critical function in controlling the severity of lung injury, fibrosis, and failure following the initiation of the disease. This review is to clarify the mechanisms beyond the possible deleterious effects of angiotensin II (Ang II), and the potential protective role of angiotensin 1-7 (Ang 1-7) against pulmonary fibrosis, with a subsequent discussion of the latest updates on ACEIs/ARBs use and COVID-19 susceptibility in the light of these mechanisms and biochemical explanation.
Collapse
Key Words
- ACE1, angiotensin-converting enzyme 1
- ACE2
- ACE2, angiotensin-converting enzyme 2
- ACEIs
- ACEIs, angiotensin-converting enzyme inhibitors
- AEC-II, alveolar epithelial type II cells
- ARBs
- ARBs, angiotensin receptor blockers
- AT1R, angiotensin type 1 receptor
- AT2R, angiotensin type 2 receptor
- Ang 1-7, angiotensin 1-7
- Ang 1-9, angiotensin 1-9
- AngI, angiotensin I
- AngII, angiotensin II
- Angiotensin 1–7
- Angiotensin II
- COVID-19
- COVID-19, coronavirus disease 2019
- CVD, cardiovascular disease
- ERK, extracellular signal-regulated kinase
- ICU, intensive care unit
- MAPK, mitogen-activated protein kinase
- NLRP3, (NOD, LRR, and pyrin domain-containing protein 3)
- RAAS, renin-angiotensin-aldosterone system
- TGF-β, transforming growth factor-beta
- miR-21, microRNA-21
Collapse
Affiliation(s)
- Ahmed Elshafei
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Emad Gamil Khidr
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed A. El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Maher H. Gomaa
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| |
Collapse
|
40
|
Tsikouras P, Kourti V, Gerede A, Kiosse E, Panopoulou M, Zervoudis S, Bothou A, Iatrakis G, Gaitatzi F, Vatsidou X, Chalkidou A, Nikolettos K, Alexiou A, Peitsidis P, Lambropoulou M, Michalopoulos S, Nikolettos N, Rafailidis P. Impact of SARS-CoV-2 on pregnancy outcomes (Review). MEDICINE INTERNATIONAL 2021; 1:19. [PMID: 36698529 PMCID: PMC9829087 DOI: 10.3892/mi.2021.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/25/2021] [Indexed: 01/28/2023]
Abstract
The impact of the pandemic outbreak associated with coronavirus 2019 disease (COVID-19) on pregnant women is of interest to obstetricians and gynecologists due to the vulnerability of this target group. In pregnant women and their infants, an exceptional clinical management is warranted. Current epidemiological findings provide information regarding the effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on pregnant patients and potential adverse perinatal outcomes. Overall, these findings are a strong indication that an increased antenatal surveillance for pregnant patients infected with COVID-19 is warranted. The aim of the present narrative review was to summarize the data obtained to date regarding the health of women during pregnancy, as well as that of the fetus associated with the risk of severe infection due to COVID-19. The present review aimed to provide further insight into the effects of this pandemic on pregnancy, also providing the experience of the authors on this matter as an example.
Collapse
Affiliation(s)
- Panagiotis Tsikouras
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Vasiliki Kourti
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Aggeliki Gerede
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Eleni Kiosse
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Maria Panopoulou
- Laboratory of Clinical Microbiology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Stefanos Zervoudis
- Technological Educational Institute of Athens and Rea Maternity Hospital, 175 64 Athens, Greece
| | - Anastasia Bothou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - George Iatrakis
- Technological Educational Institute of Athens and Rea Maternity Hospital, 175 64 Athens, Greece
| | - Fotini Gaitatzi
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Xanthi Vatsidou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Anna Chalkidou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Konstantinos Nikolettos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Alexis Alexiou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Panagiotis Peitsidis
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Maria Lambropoulou
- Department of Histology and Embryology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Spyridon Michalopoulos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Nikolaos Nikolettos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Petros Rafailidis
- Second Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
41
|
Sarker M, Hasan A, Rafi M, Hossain M, El-Mageed H, Elsapagh R, Capasso R, Emran T. A Comprehensive Overview of the Newly Emerged COVID-19 Pandemic: Features, Origin, Genomics, Epidemiology, Treatment, and Prevention. BIOLOGICS 2021; 1:357-383. [DOI: 10.3390/biologics1030021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The coronavirus disease 2019 (COVID-19), a life-threatening pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has resulted in massive destruction and is still continuously adding to its death toll. The advent of this global outbreak has not yet been confirmed; however, investigation for suitable prophylaxis against this lethal virus is being carried out by experts all around the globe. The SARS-CoV-2 belongs to the Coronaviridae superfamily, like the other previously occurring human coronavirus variants. To better understand a new virus variant, such as the SARS-CoV-2 delta variant, it is vital to investigate previous virus strains, including their genomic composition and functionality. Our study aimed at addressing the basic overview of the virus’ profile that may provide the scientific community with evidence-based insights into COVID-19. Therefore, this study accomplished a comprehensive literature review that includes the virus’ origin, classification, structure, life cycle, genome, mutation, epidemiology, and subsequent essential factors associated with host–virus interaction. Moreover, we summarized the considerable diagnostic measures, treatment options, including multiple therapeutic approaches, and prevention, as well as future directions that may reduce the impact and misery caused by this devastating pandemic. The observations and data provided here have been screened and accumulated through extensive literature study, hence this study will help the scientific community properly understand this new virus and provide further leads for therapeutic interventions.
Collapse
Affiliation(s)
- Md. Sarker
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - A. Hasan
- Industrial Biotechnology Laboratory, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Md. Rafi
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md. Hossain
- Department of Pharmacy, State University of Bangladesh, 77 Satmasjid Road, Dhanmondi, Dhaka 1205, Bangladesh
| | - H. El-Mageed
- Micro-Analysis and Environmental Research and Community Services Center, Faculty of Science, Beni-Suef University, Beni-Suef City 62521, Egypt
| | - Reem Elsapagh
- Faculty of Pharmacy, Cairo University, Cairo 12613, Egypt
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy
| | - Talha Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| |
Collapse
|
42
|
Liu L, Qu H, Li JJ, Yang YW, Zeng QX, Gong YW, He ZZ, Zhang YH, Zhang W, Liu B, Che LC. Effectiveness of methylprednisolone therapy in patients with a high-risk common type of COVID-19 pneumonia: a retrospective cohort study. Clin Exp Med 2021; 22:487-497. [PMID: 34677701 PMCID: PMC8532093 DOI: 10.1007/s10238-021-00765-1] [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: 09/25/2021] [Accepted: 10/04/2021] [Indexed: 01/08/2023]
Abstract
The optimal timing of glucocorticoid treatment for coronavirus disease 2019 (COVID-19) pneumonia is uncertain. We evaluated the clinical outcomes of methylprednisolone therapy (MPT) for patients with a high-risk common type (HRCT) COVID-19 pneumonia. We conducted a multicenter retrospective cohort study in Northeast China. A comparison was performed between the standard treatment (SDT) group and the SDT + MPT group to determine the efficacy of methylprednisolone in treating HRCT COVID-19 pneumonia. We collected the medical records of 403 patients with HRCT COVID-19 pneumonia (127 in the SDT + MPT group and 276 in the SDT group). None of the patients had received mechanical ventilation or died. Furthermore, there were no side effects associated with MPT. Patients in the SDT + MPT group treated with methylprednisolone received an intravenous injection for a median interval of five days (interquartile range of 3 to 7 days). The trends in lymphocyte count, C-reactive protein, interleukin 6, lactic acid dehydrogenase, respiratory rate, SpO2, PaO2, D-dimer and body temperature were similar between the SDT + MPT and SDT groups. The results for the SDT + MPT group seemed to improve faster than those for the SDT group; however, the results were not statistically significant. Clinical outcomes revealed that the average hospitalized days and the rate of progression to severe type COVID-19 pneumonia in both the SDT + MPT group and the SDT group were 14.56 ± 0.57 days versus 16.55 ± 0.3 days (p = 0.0009) and 21.26% (27/127) versus 32.4% (89/276) (p = 0.0247), respectively. The 16-day nucleic acid negative rate was higher in the SDT + MPT group than in the SDT group, 81.73% (104/127) versus 65.27% (180/276) (p = 0.0006). MPT effectively prevents patients with HRCT COVID-19 pneumonia from progressing to the severe stage.
Collapse
Affiliation(s)
- Lei Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Hang Qu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jun Jian Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Wei Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiu Xi Zeng
- Kang'an Hospital of Mudanjiang, Mudanjiang, China
| | - Yan Wen Gong
- Kang'an Hospital of Mudanjiang, Mudanjiang, China
| | - Zhong Zhi He
- Kang'an Hospital of Mudanjiang, Mudanjiang, China
| | - Yi He Zhang
- Kang'an Hospital of Mudanjiang, Mudanjiang, China
| | - Wei Zhang
- Kang'an Hospital of Mudanjiang, Mudanjiang, China
| | - Bin Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li Chun Che
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
43
|
Abd‐Elsalam S, Noor RA, Badawi R, Khalaf M, Esmail ES, Soliman S, Abd El Ghafar MS, Elbahnasawy M, Moustafa EF, Hassany SM, Medhat MA, Ramadan HK, Eldeen MAS, Alboraie M, Cordie A, Esmat G. Clinical study evaluating the efficacy of ivermectin in COVID-19 treatment: A randomized controlled study. J Med Virol 2021; 93:5833-5838. [PMID: 34076901 PMCID: PMC8242425 DOI: 10.1002/jmv.27122] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/25/2022]
Abstract
Researchers around the world are working at record speed to find the best ways to treat and prevent coronavirus disease 2019 (COVID-19). This study aimed to evaluate the efficacy of ivermectin for the treatment of hospitalized mild to moderate COVID-19 infected patients. This was a randomized open-label controlled study that included 164 patients with COVID-19. Patients were randomized into two groups where Group 1 (Ivermectin group) included patients who received ivermectin 12 mg once daily for 3 days with standard care and Group 2 (control group) included patients who received standard protocol of treatment alone for 14 days. The main outcomes were mortality, the length of hospital stay, and the need for mechanical ventilation. All patients were followed up for 1 month. Overall, 82 individuals were randomized to receive ivermectin plus standard of care and 82 to receive standard of care alone. Patients in the ivermectin group had a shorter length of hospital stay (8.82 ± 4.94 days) than the control group (10.97 ± 5.28 days), but this was not statistically significant (p = 0.085). Three patients (3.7%) in each group required mechanical ventilation (p = 1.00). The death rate was three patients in the ivermectin group (3.7%) versus four patients (4.9%) in the control group without any significant difference between the two groups (p = 1.00). Although there was no statistically significant difference in any endpoints by ivermectin doses (12 mg/day for 3 days); there was an observed trend to reducing hospital stay in the ivermectin-treated group.
Collapse
Affiliation(s)
- Sherief Abd‐Elsalam
- Department of Tropical Medicine and Infectious diseases, Faculty of MedicineTanta UniversityTantaEgypt
| | - Rasha A. Noor
- Department of Internal Medicine, Faculty of MedicineTanta UniversityTantaEgypt
| | - Rehab Badawi
- Department of Tropical Medicine and Infectious diseases, Faculty of MedicineTanta UniversityTantaEgypt
| | - Mai Khalaf
- Department of Tropical Medicine and Infectious diseases, Faculty of MedicineTanta UniversityTantaEgypt
| | - Eslam S. Esmail
- Department of Tropical Medicine and Infectious diseases, Faculty of MedicineTanta UniversityTantaEgypt
| | - Shaimaa Soliman
- Department of Public health and Community Medicine, Faculty of MedicineMenoufia UniversityMenoufiaEgypt
| | - Mohamed S. Abd El Ghafar
- Department of Anesthesia, Surgical Intensive Care, and Pain Medicine, Faculty of MedicineTanta UniversityTantaEgypt
| | - Mohamed Elbahnasawy
- Department of Emergency Medicine and traumatology, Faculty of MedicineTanta UniversityTantaEgypt
| | - Ehab F. Moustafa
- Department of Tropical Medicine and Gastroenterology, Faculty of MedicineAssiut UniversityAssiutEgypt
| | - Sahar M. Hassany
- Department of Tropical Medicine and Gastroenterology, Faculty of MedicineAssiut UniversityAssiutEgypt
| | - Mohammed A. Medhat
- Department of Tropical Medicine and Gastroenterology, Faculty of MedicineAssiut UniversityAssiutEgypt
| | - Haidi Karam‐Allah Ramadan
- Department of Tropical Medicine and Gastroenterology, Faculty of MedicineAssiut UniversityAssiutEgypt
| | | | | | - Ahmed Cordie
- Department of Endemic Medicine, Kasr Alainy School of MedicineCairo UniversityCairoEgypt
| | - Gamal Esmat
- Department of Endemic Medicine, Kasr Alainy School of MedicineCairo UniversityCairoEgypt
| |
Collapse
|
44
|
Elrobaa IH, New KJ. COVID-19: Pulmonary and Extra Pulmonary Manifestations. Front Public Health 2021; 9:711616. [PMID: 34650947 PMCID: PMC8505777 DOI: 10.3389/fpubh.2021.711616] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/30/2021] [Indexed: 01/08/2023] Open
Abstract
Introduction: The coronavirus disease-2019 (COVID-19) pandemic has been the most significant event in 2020, with ~86.8 million cases and 1.88 million deaths worldwide. It is a highly infectious disease, wherein the virus (severe acute respiratory syndrome coronavirus 2) rapidly multiplies and spreads to all parts of the body. Therefore, COVID-19 is not only respiratory disease but also a multisystem disease. Many people, including physicians, incorrectly believe that the disease affects only the respiratory tract. In this study, we aimed to describe COVID-19 manifestations and the underlying pathophysiology to provide the readers with a better understanding of this disease to achieve good management and to control the spread of this disease. Methods: Secondary data were obtained from PubMed, Google Scholar, and Scopus databases. The keywords used for the search were as follows: COVID-19, COVID-19 pulmonary manifestations, COVID-19 extra pulmonary manifestations, and pathophysiology of COVID-19. We collected secondary data from systemic reviews, metaanalyses, case series, and case reports in the form of public data that was published on websites of the government, medical corporations, medical peer-reviewed journals, and medical academies, all of which were indexed in PubMed, Google Scholar, or Scopus. Our questions were as follows: Is COVID-19 a respiratory disease only? and What are the extrapulmonary manifestations of COVID-19? Results: From our data, we found that a patient with COVID-19 may be either asymptomatic or symptomatic. Symptomatic cases may have either pulmonary or extrapulmonary manifestations. Pulmonary manifestations occur as mild, moderate, or severe cases. In mild and moderate cases, extrapulmonary manifestations such as gastroenteritis, fever, or vomiting may present alone. Some of these cases may be missed for diagnosis, and the patient may receive symptomatic treatment without a COVID-19 diagnosis, leading to increased spread of the infection. Extrapulmonary manifestations may occur in severe and critical cases as complications of severe infections (high viral overload) or the cytokine storm, such as in acute kidney injury (AKI), heart failure (HF), and venous thromboembolic (VTE) manifestation. Conclusion: COVID-19 is not a respiratory disease alone; rather, it is a multisystem disease. Pulmonary and extrapulmonary manifestations should be considered for early diagnosis and to control the spread of the infection.
Collapse
Affiliation(s)
- Islam H. Elrobaa
- Emergency Medicine Specialist in Hamad Medical Corporation, Qatar and Lecturer in Clinical Education Department, College of Medicine, Qatar University, Doha, Qatar
| | - Karl J. New
- Clinical Physiology, School of Health, Sport, and Professional Practice, Faculty of Life Science and Education, University of South Wales, Treforest, United Kingdom
| |
Collapse
|
45
|
Malani M, Salunke P, Kulkarni S, Jain GK, Sheikh A, Kesharwani P, Nirmal J. Repurposing pharmaceutical excipients as an antiviral agent against SARS-CoV-2. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 33:110-136. [PMID: 34464232 DOI: 10.1080/09205063.2021.1975020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The limited time indorsed to face the COVID-19 emergency and large number of deaths across the globe, poses an unrelenting challenge to find apt therapeutic approaches. However, lead candidate selection to phase III trials of new chemical entity is a time-consuming procedure, and not feasible in pandemic, such as the one we are facing. Drug repositioning, an exploration of existing drug for new therapeutic use, could be an effective alternative as it allows fast-track estimation in phase II-III trials, or even forthright compassionate use. Although, drugs repurposed for COVID-19 pandemic are commercially available, yet the evaluation of their safety and efficacy is tiresome and painstaking. In absence of any specific treatment the easy alternatives such as over the counter products, phytotherapies and home remedies have been largely adopted for prophylaxis and therapy as well. In recent years, it has been demonstrated that several pharmaceutical excipients possess antiviral properties making them prospective candidates against SARS-CoV-2. This review highlights the mechanism of action of various antiviral excipients and their propensity to act against SARs-CoV2. Though, repurposing of pharmaceutical excipients against COVID-19 has the edge over therapeutic agents in terms of safety, cost and fast-track approval trial burdened, this hypothesis needs to be experimentally verified for COVID-19 patients.
Collapse
Affiliation(s)
- Manisha Malani
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS)-Pilani, Hyderabad, India
| | - Prerana Salunke
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS)-Pilani, Hyderabad, India
| | - Shraddha Kulkarni
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS)-Pilani, Hyderabad, India
| | - Gaurav K Jain
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Jayabalan Nirmal
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS)-Pilani, Hyderabad, India
| |
Collapse
|
46
|
Hatzl S, Reisinger AC, Posch F, Prattes J, Stradner M, Pilz S, Eller P, Schoerghuber M, Toller W, Gorkiewicz G, Metnitz P, Rief M, Prüller F, Rosenkranz AR, Valentin T, Krause R, Hoenigl M, Schilcher G. Antifungal prophylaxis for prevention of COVID-19-associated pulmonary aspergillosis in critically ill patients: an observational study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2021; 25:335. [PMID: 34526087 PMCID: PMC8441945 DOI: 10.1186/s13054-021-03753-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Coronavirus disease 19 (COVID-19)-associated pulmonary aspergillosis (CAPA) emerged as important fungal complications in patients with COVID-19-associated severe acute respiratory failure (ARF). Whether mould active antifungal prophylaxis (MAFP) can prevent CAPA remains elusive so far. METHODS In this observational study, we included all consecutive patients admitted to intensive care units with COVID-19-associated ARF between September 1, 2020, and May 1, 2021. We compared patients with versus without antifungal prophylaxis with respect to CAPA incidence (primary outcome) and mortality (secondary outcome). Propensity score adjustment was performed to account for any imbalances in baseline characteristics. CAPA cases were classified according to European Confederation of Medical Mycology (ECMM)/International Society of Human and Animal Mycoses (ISHAM) consensus criteria. RESULTS We included 132 patients, of whom 75 (57%) received antifungal prophylaxis (98% posaconazole). Ten CAPA cases were diagnosed, after a median of 6 days following ICU admission. Of those, 9 CAPA cases were recorded in the non-prophylaxis group and one in the prophylaxis group, respectively. However, no difference in 30-day ICU mortality could be observed. Thirty-day CAPA incidence estimates were 1.4% (95% CI 0.2-9.7) in the MAFP group and 17.5% (95% CI 9.6-31.4) in the group without MAFP (p = 0.002). The respective subdistributional hazard ratio (sHR) for CAPA incidence comparing the MAFP versus no MAFP group was of 0.08 (95% CI 0.01-0.63; p = 0.017). CONCLUSION In ICU patients with COVID-19 ARF, antifungal prophylaxis was associated with significantly reduced CAPA incidence, but this did not translate into improved survival. Randomized controlled trials are warranted to evaluate the efficacy and safety of MAFP with respect to CAPA incidence and clinical outcomes.
Collapse
Affiliation(s)
- Stefan Hatzl
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Graz, Austria.,Division of Haematology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Alexander C Reisinger
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florian Posch
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Juergen Prattes
- Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Martin Stradner
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Stefan Pilz
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Philipp Eller
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Michael Schoerghuber
- Department of Anaesthesiology and Intensive Care Medicine, Medical University Graz, Graz, Austria
| | - Wolfgang Toller
- Department of Anaesthesiology and Intensive Care Medicine, Medical University Graz, Graz, Austria
| | | | - Philipp Metnitz
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Martin Rief
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Florian Prüller
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Alexander R Rosenkranz
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Thomas Valentin
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Robert Krause
- Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
| | - Martin Hoenigl
- Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, Graz, Austria.,Division of Infectious Diseases, University of California San Diego, San Diego, USA
| | - Gernot Schilcher
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
47
|
Smith EL, Gwee A, Roberts JA, Molton JS, Wurzel D, Hughes CM, Rogers B. Prospective Study of Policies and Use of Therapies for COVID-19 Amongst Australian Health Services during 2020. Intern Med J 2021; 52:214-222. [PMID: 34490712 PMCID: PMC8653236 DOI: 10.1111/imj.15510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/20/2021] [Accepted: 09/02/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The COVID-19 pandemic has generated significant debate about how emerging infections can be treated in the absence of evidence-based therapies to combat disease. In particular, the use of off-label therapies outside of a clinical trial setting has been controversial. AIM We aimed to longitudinally study policies and prescribing practices pertaining to therapies for COVID-19 in Australian Health Services in 2020. METHODS Prospective data was collected from participating Australian health services who may care for patients with COVID-19 via an electronic portal. A single informant from each health service was emailed a survey link at regular intervals. Information was sought regarding changes to COVID-19 policy at their service and use of therapies for COVID-19. RESULTS Overall, 78 hospitals were represented from 39 respondents with longitudinal data collection from May to December 2020. All Australian states/territories were represented with the majority of respondents located in a major city (34/39; 87%). Just over half (20/39) of respondents had a written policy for COVID-19 therapy use at their health service at survey enrolment and policies changed frequently throughout the pandemic. Therapy use outside of a clinical trial was reported in 54% of health services, most frequently in Victoria, correlating with higher numbers of COVID-19 cases. At study commencement hydroxychloroquine was most frequently used, with corticosteroids and remdesivir use increasingly throughout the study period. CONCLUSION Our results reflect the reactive nature of prescribing of therapies for COVID-19 and highlight the importance of evidence-based guidelines to assist prescribers. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- E L Smith
- Monash Infectious Diseases, Monash Health, Melbourne, Victoria
| | - A Gwee
- Murdoch Children's Research Institute, Melbourne, Victoria.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria
| | - J A Roberts
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland.,Departments of Pharmacy and Critical Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland
| | | | - D Wurzel
- Department of Respiratory and Sleep Medicine, The Royal Children's Hospital, Melbourne, Victoria.,Allergy and Lung Health Unit, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria
| | - C M Hughes
- Monash Infectious Diseases, Monash Health, Melbourne, Victoria.,Department of Microbiology, Monash Pathology, Monash Health, Melbourne, Victoria
| | - B Rogers
- Monash Infectious Diseases, Monash Health, Melbourne, Victoria.,Centre for Inflammatory Diseases, Monash University, Melbourne, Victoria
| |
Collapse
|
48
|
Luo L, Qiu Q, Huang F, Liu K, Lan Y, Li X, Huang Y, Cui L, Luo H. Drug repurposing against coronavirus disease 2019 (COVID-19): A review. J Pharm Anal 2021; 11:683-690. [PMID: 34513115 PMCID: PMC8416689 DOI: 10.1016/j.jpha.2021.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/16/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
Since December 2019, severe acute respiratory syndrome coronavirus 2 has been found to be the culprit in the coronavirus disease 2019 (COVID-19), causing a global pandemic. Despite the existence of many vaccine programs, the number of confirmed cases and fatalities due to COVID-19 is still increasing. Furthermore, a number of variants have been reported. Because of the absence of approved anti-coronavirus drugs, the treatment and management of COVID-19 has become a global challenge. Under these circumstances, drug repurposing is an effective method to identify candidate drugs with a shorter cycle of clinical trials. Here, we summarize the current status of the application of drug repurposing in COVID-19, including drug repurposing based on virtual computer screening, network pharmacology, and bioactivity, which may be a beneficial COVID-19 treatment. Mechanism of SARS-CoV-2 infection and drug targets were reviewed. Drug repurposing against COVID-19 based on computer virtual screening, network pharmacology, bioactivity were summarized. The use of drug repurposing in COVID-19 was addressed.
Collapse
Affiliation(s)
- Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.,Marine Medical Research Institute of Zhanjiang, Zhanjiang, 524023, Guangdong, China
| | - Qin Qiu
- Graduate School, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Fangfang Huang
- Graduate School, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Kaifeng Liu
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Yongqi Lan
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Xiaoling Li
- Animal Experiment Center, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Yuge Huang
- Department of Pediatrics, the Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Hui Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| |
Collapse
|
49
|
Adebisi YA, Alaran AJ, Okereke M, Oke GI, Amos OA, Olaoye OC, Oladunjoye I, Olanrewaju AY, Ukor NA, Lucero-Prisno DE. COVID-19 and Antimicrobial Resistance: A Review. Infect Dis (Lond) 2021; 14:11786337211033870. [PMID: 34376994 PMCID: PMC8327234 DOI: 10.1177/11786337211033870] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/01/2021] [Indexed: 12/13/2022] Open
Abstract
As the world continues to respond to the coronavirus pandemic (COVID-19), there is a larger hidden threat of antimicrobial resistance (AMR) lurking behind. AMR remains worrisome in that the pathogens causing resistant infections to thrive in hospitals and medical facilities, putting all patients at risk, irrespective of the severity of their medical conditions, further compounding the management of COVID-19. This study aims to provide overview of early findings on COVID-19 and AMR as well as to provide recommendations and lesson learned toward improving antimicrobial stewardship. We conducted a rapid narrative review of published articles by searching PubMed and Google Scholar on COVID-19 and Antimicrobial Resistance with predetermined keywords. Secondary bacterial infections play crucial roles in mortality and morbidity associated with COVID-19. Research has shown that a minority of COVID-19 patients need antibiotics to treat secondary bacterial infections. Current evidence reiterates the need not to give antibiotic therapy or prophylaxis to patients with mild COVID-19 or to patients with suspected or confirmed moderate COVID-19 illness unless it is indicated. The pandemic has also brought to the fore the deficiencies in health systems around the world. This comes with a lot of lessons, one of which is that despite the advances in medicine; we remain incredibly vulnerable to infections with limited or no standard therapies. This is worth thinking in the context of AMR, as the resistant pathogens are evolving and leading us to the era of untreatable infections. There is a necessity for continuous research into understanding and controlling infectious agents, as well as the development of newer functional antimicrobials and the need to strengthen the antimicrobial stewardship programs.
Collapse
Affiliation(s)
| | | | - Melody Okereke
- Faculty of Pharmaceutical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Gabriel Ilerioluwa Oke
- Department of Medical Laboratory Science, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | | | | | - Iyiola Oladunjoye
- Department of Microbiology, University of Ilorin, Ilorin, Kwara State, Nigeria
| | | | | | - Don Eliseo Lucero-Prisno
- Department of Global Health and Development, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
50
|
Zhang JC, Li T. Awake extracorporeal membrane oxygenation support for a critically ill COVID-19 patient: A case report. World J Clin Cases 2021; 9:5963-5971. [PMID: 34368315 PMCID: PMC8316967 DOI: 10.12998/wjcc.v9.i21.5963] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/30/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A critically ill coronavirus disease 2019 (COVID-19) patient complicated by acute respiratory distress syndrome is reported. The patient survived following treatment with awake veno-venous extracorporeal membrane oxygenation (ECMO). CASE SUMMARY A 53-year-old male patient attended our hospital following a cough for 11 d and fever for 9 d. According to his computed tomography (CT) scan and real-time reverse transcription-polymerase chain reaction assay of a throat swab, nucleic acid was positive, confirming that he had COVID-19. He was subsequently transferred to the intensive care unit due to respiratory failure. The patient received antiviral drugs, a small dose of glucocorticoid, and respiratory support, including mechanical ventilation, but the treatment effect was poor. On the 28th day after admission, veno-venous ECMO and prone position ventilation (PPV) were performed, combined with awake ECMO and other comprehensive rehabilitation measures. On the 17th day of ECMO, the patient started to improve and his chest CT and lung compliance improved. ECMO was discontinued after 27 days, and mechanical ventilation was also discontinued after 9 days. The patient was then transferred to the rehabilitation department. CONCLUSION COVID-19 can damage lung tissues and cause evident inflammatory exudation, thus affecting oxygenation function. Awake ECMO, PPV, and comprehensive rehabilitation are effective in patients with critical COVID-19 and respiratory failure.
Collapse
Affiliation(s)
- Jing-Chen Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Tong Li
- Department of Critical Care Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|