Rapid #Spread and #Control of #MDR Gram-Negative #Bacteria in #COVID19 Patient Care Units (Emerg Infect Dis., abstract)

[Source: US Centers for Disease Control and Prevention (CDC), Emerging Infectious Diseases Journal, full page: (LINK). Abstract, edited.]

Volume 27, Number 4—April 2021 | Research Letter

Rapid Spread and Control of Multidrug-Resistant Gram-Negative Bacteria in COVID-19 Patient Care Units

Ashka Patel, Michele Emerick, Marie K. Cabunoc, Michelle H. Williams, Michael Anne Preas, Gregory Schrank, Ronald Rabinowitz, Paul Luethy, J. Kristie Johnson, and Surbhi Leekha

Author affiliations: University of Maryland Medical Center, Baltimore, Maryland, USA (A. Patel, M. Emerick, M.K. Cabunoc, M.H. Williams, M.A. Preas); University of Maryland School of Medicine, Baltimore (G. Schrank, R. Rabinowitz, P. Luethy, J.K. Johnson, S. Leekha)

Abstract

We describe rapid spread of multidrug-resistant gram-negative bacteria among patients in dedicated coronavirus disease care units in a hospital in Maryland, USA, during May–June 2020. Critical illness, high antibiotic use, double occupancy of single rooms, and modified infection prevention practices were key contributing factors. Surveillance culturing aided in outbreak recognition and control.

Keywords: SARS-CoV-2; COVID-19; Antibiotics; Drugs Resistance; Nosocomial Outbreaks; USA.

——

Development of cycling #probe based real-time #PCR methodology for #influenza A viruses possessing the #PA/I38T amino acid #substitution associated with reduced #baloxavir susceptibility (Antiviral Res., abstract)

[Source: Antiviral Research, full page: (LINK). Abstract, edited.]

Antiviral Research | Available online 10 February 2021, 105036 | In Press, Journal Pre-proof

Development of cycling probe based real-time PCR methodology for influenza A viruses possessing the PA/I38T amino acid substitution associated with reduced baloxavir susceptibility

Hidekazu Osada a,b, Irina Chon a, Wint Wint Phyu a, Keita Wagatsuma a, Nobuo Nagata c, Takashi Kawashim a,d, Isamu Sato e, Tadashi Saito f, Naoki Kodo g, Hironori Masaki h, Norichika Asoh i, Yoshiko Tuchihashi i, Yutaka Shirahige j, Yasuhiko Ono k, Yasushi Shimada l, Hirotsune Hamabata m, Kousuke Saito a, Reiko Saito a,b

a Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan; b Infectious Diseases Research Center of Niigata University in Myanmar, Yangon, Yangon Region, Myanmar; c Hiraoka-Kohen Pediatric Clinic, Sapporo, Hokkaido, Japan; d Kawashima Clinic, Shibukawa, Gunma, Japan; e Yoiko-no-Syounika Sato Pediatric Clinic, Niigata, Niigata, Japan; f Pediatric Department, Tako Central Hospital, Katori, Chiba, Japan; g Kodo Pediatric Clinic, Uji, Kyoto, Japan; h Masaki Respiratory Medicine Clinic, Nagasaki, Nagasaki, Japan; i Juzenkai Hospital, Nagasaki, Nagasaki, Japan; j Shirahige Clinic, Nagasaki, Nagasaki, Japan; k Ono Pediatric Clinic, Isahaya, Nagasaki, Japan; l Shimada Children′s Clinic, Kamiamakusa, Kumamoto, Japan; m Awase Daiichi Clinic, Okinawa, Okinawa, Japan

Received 15 May 2020, Revised 4 February 2021, Accepted 6 February 2021, Available online 10 February 2021.

DOI: https://doi.org/10.1016/j.antiviral.2021.105036

Highlights

  • We established cycling probe Real-time PCR systems to detect influenza A viruses with PA/I38T.
  • Pre-treatment prevalence of the PA/I38T mutant virus was 0.0% (0/129) for A(H1N1)pdm09 and 1.7% (4/229) for A/H3N2.
  • A(H3N2) PA/I38T viruses may be transmitted among humans in closed environments.

Abstract

Baloxavir marboxil has been used for influenza treatment since March 2018 in Japan. After baloxavir treatment, the most frequently detected substitution is Ile38Thr in polymerase acidic protein (PA/I38T), and this substitution reduces baloxavir susceptibility in influenza A viruses. To rapidly investigate the frequency of PA/I38T in influenza A(H1N1)pdm09 and A(H3N2) viruses in clinical samples, we established a rapid real-time system to detect single nucleotide polymorphisms in PA, using cycling probe real-time PCR. We designed two sets of probes that were labeled with either 6-carboxyfluorescein (FAM) or 6-carboxy-X-rhodamine (ROX) to identify PA/I38 (wild type strain) or PA/I38T, respectively. The established cycling probe real-time PCR system showed a dynamic linear range of 101 to 106 copies with high sensitivity in plasmid DNA controls. This real-time PCR system discriminated between PA/I38T and wild type viruses well. During the 2018/19 season, 377 influenza A-positive clinical samples were collected in Japan before antiviral treatment. Using our cycling probe real-time PCR system, we detected no (0/129, 0.0%) influenza A(H1N1)pdm09 viruses with PA/I38T substitutions and four A(H3N2) (4/229, 1.7%) with PA/I38T substitution prior to treatment. In addition, we found PA/I38T variant in siblings who did not received baloxavir treatment during an infection caused by A(H3N2) that afflicted the entire family. Although human-to-human transmission of PA/I38T variant may have occurred in a closed environment, the prevalence of this variant in influenza A viruses was still limited. Our cycling probe-PCR system is thus useful for antiviral surveillance of influenza A viruses possessing PA/I38T.

Keywords: influenza virus – baloxavir marboxil – PA/I38T substitution – cycling probe real-time PCR – antiviral susceptibility

© 2021 Elsevier B.V. All rights reserved.

Keywords: Seasonal Influenza; H1N1pdm09; H3N2; Antivirals; Drugs Resistance; Baloxavir.

——

#Genetic #Conservation of #SARS-CoV-2 #RNA #Replication Complex in Globally Circulating Isolates and Recently Emerged Variants from #Humans and #Minks Suggests Minimal Pre-Existing #Resistance to #Remdesivir (Antiviral Res., abstract)

[Source: Antiviral Research, full page: (LINK). Abstract, edited.]

Antiviral Research | Available online 5 February 2021, 105033 | In Press, Journal Pre-proof | Research paper

Genetic Conservation of SARS-CoV-2 RNA Replication Complex in Globally Circulating Isolates and Recently Emerged Variants from Humans and Minks Suggests Minimal Pre-Existing Resistance to Remdesivir

Ross Martin, Jiani Li, Aiyippa Parvangada, Jason Perry, Tomas Cihlar, Hongmei Mo, Danielle Porter, Evguenia Svarovskaia, Gilead Sciences, 333 Lakeside Dr, Foster City, CA, USA

Received 19 December 2020, Revised 29 January 2021, Accepted 30 January 2021, Available online 5 February 2021.

DOI: https://doi.org/10.1016/j.antiviral.2021.105033

Highlights

  • Remdesivir (RDV) exhibits potent antiviral activity against SARS-CoV-2 and is currently the only drug approved for the treatment of COVID-19.
  • Little is known about the potential for pre-existing resistance to RDV and the global spread of SARS-CoV-2 and transmission between human and other species may lead to genetic diversification.
  • A large set of SARS-CoV-2 human clinical isolates (>90,000), including the recently emerged United Kingdom and South African SARS-CoV-2 variants, and mink isolates (>300) sequences were investigated for genetic changes in the RNA replication complex since the start of pandemic.
  • Low genetic diversity was observed in RNA replication complex in human clinical isolates and mink isolates.
  • Amino acid substitutions previously identified to cause reduced susceptibility to RDV in-vitro were observed at extremely low frequency (0.002%).
  • Other variants observed in clinical isolate sequences were modeled using Cryo-EM to evaluate proximity to RDV active site and potential impact on effectivity.

Abstract

Remdesivir (RDV) exhibits potent antiviral activity against SARS-CoV-2 and is currently the only drug approved for the treatment of COVID-19. However, little is currently known about the potential for pre-existing resistance to RDV and the possibility of SARS-CoV-2 genetic diversification that might impact RDV efficacy as the virus continue to spread globally. In this study, >90,000 SARS-CoV-2 sequences from globally circulating clinical isolates, including sequences from recently emerged United Kingdom and South Africa variants, and >300 from mink isolates were analyzed for genetic diversity in the RNA replication complex (nsp7, nsp8, nsp10, nsp12, nsp13, and nsp14) with a focus on the RNA-dependent RNA polymerase (nsp12), the molecular target of RDV. Overall, low genetic variation was observed with only 12 amino acid substitutions present in the entire RNA replication complex in ≥0.5% of analyzed sequences with the highest overall frequency (82.2%) observed for nsp12 P323L that consistently increased over time. Low sequence variation in the RNA replication complex was also observed among the mink isolates. Importantly, the coronavirus Nsp12 mutations previously selected in vitro in the presence of RDV were identified in only 2 isolates (0.002%) within all the analyzed sequences. In addition, among the sequence variants observed in ≥0.5% clinical isolates, including P323L, none were located near the established polymerase active site or sites critical for the RDV mechanism of inhibition. In summary, the low diversity and high genetic stability of the RNA replication complex observed over time and in the recently emerged SARS-CoV-2 variants suggests a minimal global risk of pre-existing SARS-CoV-2 resistance to RDV.

Keywords: SARS-CoV-2 – COVID-19 – nsp12 RdRp – Remdesivir – RDV – genetic diversity – resistance

© 2021 Elsevier B.V. All rights reserved.

Keywords: SARS-CoV-2; COVID-19; Antivirals; Drugs resistance; Remdesivir.

——

#COVID19–Associated Pulmonary #Aspergillosis, March–August 2020 (Emerg Infect Dis., abstract)

[Source: US Centers for Disease Control and Prevention (CDC), Emerging Infectious Diseases Journal, full page: (LINK). Abstract, edited.]

Volume 27, Number 4—April 2021 | Research

COVID-19–Associated Pulmonary Aspergillosis, March–August 2020

Jon Salmanton-García  , Rosanne Sprute, Jannik Stemler, Michele Bartoletti, Damien Dupont, Maricela Valerio, Carolina García-Vidal, Iker Falces-Romero, Marina Machado, Sofía de la Villa, Maria Schroeder, Irma Hoyo, Frank Hanses, Kennio Ferreira-Paim, Daniele Roberto Giacobbe, Jacques F. Meis, Jean-Pierre Gangneux, Azucena Rodríguez-Guardado, Spinello Antinori, Ertan Sal, Xhorxha Malaj, Danila Seidel, Oliver A. Cornely1, Philipp Koehler1, and The FungiScope European Confederation of Medical Mycology/The International Society for Human and Animal Mycology Working Group2

Author affiliations: University of Cologne, Cologne, Germany (J. Salmanton-García, R. Sprute, J. Stemler, E. Sal, X. Malaj, D. Seidel, O.A. Cornely, P. Koehler); L’Azienda Ospedaliero-Universitaria di Bologna Policlinico S. Orsola—Malpighi, Bologna, Italy (M. Bartoletti); Hospices Civils de Lyon, Lyon, France (D. Dupont); Université Claude Bernard Lyon 1, Lyon (D. Dupont); Centre de Recherche en Neurosciences de Lyon, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Lyon (D. Dupont); Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain (M. Valerio, M. Machado, S. de la Villa); Institute of Biomedical Research August Pi i Sunyer, Barcelona, Spain (C. García-Vidal); Hospital Universitario La Paz, Madrid (I. Falces-Romero); University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M. Schroeder); Centro Médico ABC, Mexico City, Mexico (I. Hoyo); University Hospital Regensburg, Regensburg, Germany (F. Hanses); Federal University of Triângulo Mineiro, Uberaba, Brazil (K. Ferreira-Paim); Istituto di Ricovero e Cura a Carattere Scientifico San Martino Polyclinic Hospital, Genoa, Italy (D.R. Giacobbe); Canisius Wilhelmina Hospital, Nijmegen, the Netherlands (J.F. Meis); Federal University of Paraná, Curitiba, Brazil (J.F. Meis); University of Rennes I, Institut National de la Santé et de la Recherche Médicale, École des Hautes Études en Santé Publique, Institut de Recherche en Santé, Environnement et Travail, Rennes, France (J.-P. Gangneux); Hospital de Cabueñes, Gijón, Spain (A. Rodríguez-Guardado); Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain (A. Rodríguez-Guardado); University of Milan, Milan, Italy (S. Antinori); German Centre for Infection Research, Cologne (O.A. Cornely)

Abstract

Pneumonia caused by severe acute respiratory syndrome coronavirus 2 emerged in China at the end of 2019. Because of the severe immunomodulation and lymphocyte depletion caused by this virus and the subsequent administration of drugs directed at the immune system, we anticipated that patients might experience fungal superinfection. We collected data from 186 patients who had coronavirus disease–associated pulmonary aspergillosis (CAPA) worldwide during March–August 2020. Overall, 182 patients were admitted to the intensive care unit (ICU), including 180 with acute respiratory distress syndrome and 175 who received mechanical ventilation. CAPA was diagnosed a median of 10 days after coronavirus disease diagnosis. Aspergillus fumigatus was identified in 80.3% of patient cultures, 4 of which were azole-resistant. Most (52.7%) patients received voriconazole. In total, 52.2% of patients died; of the deaths, 33.0% were attributed to CAPA. We found that the cumulative incidence of CAPA in the ICU ranged from 1.0% to 39.1%.

Keywords: SARS-CoV-2; COVID-19; Intensive Care; Aspergillosis.

——

Single-dose #intranasal subunit #vaccine rapidly clears secondary #sepsis in a high-dose pneumonic #plague infection (Vaccine, abstract)

[Source: Vaccine, full page: (LINK). Abstract, edited.]

Vaccine | Available online 30 January 2021 | In Press, Corrected Proof

Single-dose intranasal subunit vaccine rapidly clears secondary sepsis in a high-dose pneumonic plague infection

Christina D’Arco a, Alison A. McCormick b, Paul M. Arnaboldi a,c

a Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, United States; b Department of Biology and Pharmaceutical Sciences, College of Pharmacy, Touro University California, Vallejo, CA 94592, United States; c Biopeptides, Corp., East Setauket, NY 11733, United States

Received 27 March 2020, Revised 12 January 2021, Accepted 16 January 2021, Available online 30 January 2021.

DOI: https://doi.org/10.1016/j.vaccine.2021.01.040

Abstract

Yersinia pestis, the causative agent of plague, has killed millions throughout human history. Though public health initiatives have reduced the number of plague cases, it remains endemic in many areas of the world. It also remains a significant threat for use as a biological weapon. Naturally occurring multi-drug antibiotic resistance has been observed in Y. pestis, and resistant strains have been engineered for use as a biological weapon. Vaccines represent our best means of protection against the threat of antibiotic resistant plague. We have developed a vaccine consisting of two Y. pestis virulence factors, LcrV (V) and F1, conjugated to Tobacco Mosaic Virus (TMV), a safe, non-replicating plant virus that can be administered mucosally, providing complete protection against pneumonic plague, the deadliest form of the disease and the one most likely to be seen in a biological attack. A single intranasal (i.n.) dose of TMV-F1 + TMV-V (TMV-F1/V) protected 88% of mice against lethal challenge with 100 LD50 of Y. pestis CO92pgm-, while immunization with rF1 + rV without TMV was not protective. Serum and tissues were collected at various timepoints after challenge to assess bacterial clearance, histopathology, cytokine production, and antibody production. Overall, TMV-F1/V immunized mice showed a significant reduction in histopathology, bacterial burden, and inflammatory cytokine production following challenge compared to rF1 + rV vaccinated and unvaccinated mice. Pneumonic challenge resulted in systemic dissemination of the bacteria in all groups, but only TMV-F1/V immunized mice rapidly cleared bacteria from the spleen and liver. There was a direct correlation between pre-challenge serum F1 titers and recovery in all immunized mice, strongly suggesting a role for antibody in the neutralization and/or opsonization of Y. pestis in this model. Mucosal administration of a single dose of a Y. pestis TMV-based subunit vaccine, without any additional adjuvant, can effectively protect mice from lethal infection.

Keywords: Yersinia pestis – Plague – Mucosal vaccine – Subunit vaccine – Tobacco Mosaic Virus

© 2021 Elsevier Ltd. All rights reserved.

Keywords: Yersinia Pestis; Antibiotics; Drugs Resistance; Vaccines; Plague; Pneumonic plague; Animal models.

—–

Continuation versus discontinuation of #RAS #inhibitors in patients admitted to #hospital with #COVID19: a prospective, randomised, open-label trial (Lancet Resp Med., abstract)

[Source: Lancet Respiratory Medicine, full page: (LINK). Abstract, edited.]

Continuation versus discontinuation of renin–angiotensin system inhibitors in patients admitted to hospital with COVID-19: a prospective, randomised, open-label trial

Jordana B Cohen, MD, Thomas C Hanff, MD, Preethi William, MD, Prof Nancy Sweitzer, MD, Nelson R Rosado-Santander, MD, Carola Medina, MD, Juan E Rodriguez-Mori, MD, Nicolás Renna, MD, Tara I Chang, MD, Vicente Corrales-Medina, MD, Prof Jaime F Andrade-Villanueva, MD, Prof Alejandro Barbagelata, MD, Roberto Cristodulo-Cortez, MD, Omar A Díaz-Cucho, MD, Jonas Spaak, MD, Carlos E Alfonso, MD, Renzo Valdivia-Vega, MD, Mirko Villavicencio-Carranza, MD, Ricardo J Ayala-García, MD, Carlos A Castro-Callirgos, MD, Prof Luz A González-Hernández, MD, Eduardo F Bernales-Salas, MD, Johanna C Coacalla-Guerra, MD, Cynthia D Salinas-Herrera, MD, Prof Liliana Nicolosi, MD, Mauro Basconcel, MD, James B Byrd, MD, Tiffany Sharkoski, MPH, Luis E Bendezú-Huasasquiche, MD, Jesse Chittams, MS, Daniel L Edmonston, MD, Charles R Vasquez, MD, Julio A Chirinos, MD

Published: January 07, 2021 | DOI: https://doi.org/10.1016/S2213-2600(20)30558-0

Summary

Background

Biological considerations suggest that renin–angiotensin system inhibitors might influence the severity of COVID-19. We aimed to evaluate whether continuing versus discontinuing renin–angiotensin system inhibitors (angiotensin-converting enzyme inhibitors or angiotensin receptor blockers) affects outcomes in patients admitted to hospital with COVID-19.

Methods

The REPLACE COVID trial was a prospective, randomised, open-label trial done at 20 large referral hospitals in seven countries worldwide. Eligible participants were aged 18 years and older who were admitted to hospital with COVID-19 and were receiving a renin–angiotensin system inhibitor before admission. Individuals with contraindications to continuation or discontinuation of renin–angiotensin system inhibitor therapy were excluded. Participants were randomly assigned (1:1) to continuation or discontinuation of their renin–angiotensin system inhibitor using permuted block randomisation, with allocation concealed using a secure web-based randomisation system. The primary outcome was a global rank score in which participants were ranked across four hierarchical tiers incorporating time to death, duration of mechanical ventilation, time on renal replacement or vasopressor therapy, and multiorgan dysfunction during the hospitalisation. Primary analyses were done in the intention-to-treat population. The REPLACE COVID trial is registered with ClinicalTrials.gov, NCT04338009.

Findings

Between March 31 and Aug 20, 2020, 152 participants were enrolled and randomly assigned to either continue or discontinue renin–angiotensin system inhibitor therapy (continuation group n=75; discontinuation group n=77). Mean age of participants was 62 years (SD 12), 68 (45%) were female, mean body-mass index was 33 kg/m2 (SD 8), and 79 (52%) had diabetes. Compared with discontinuation of renin–angiotensin system inhibitors, continuation had no effect on the global rank score (median rank 73 [IQR 40–110] for continuation vs 81 [38–117] for discontinuation; β-coefficient 8 [95% CI −13 to 29]). There were 16 (21%) of 75 participants in the continuation arm versus 14 (18%) of 77 in the discontinuation arm who required intensive care unit admission or invasive mechanical ventilation, and 11 (15%) of 75 participants in the continuation group versus ten (13%) of 77 in the discontinuation group died. 29 (39%) participants in the continuation group and 28 (36%) participants in the discontinuation group had at least one adverse event (χ2 test of adverse events between treatment groups p=0·77). There was no difference in blood pressure, serum potassium, or creatinine during follow-up across the two groups.

Interpretation

Consistent with international society recommendations, renin–angiotensin system inhibitors can be safely continued in patients admitted to hospital with COVID-19.

Funding

REPLACE COVID Investigators, REPLACE COVID Trial Social Fundraising Campaign, and FastGrants.

Keywords: SARS-CoV-2; COVID-19; Drugs safety.

—–

High-Throughput Rational #Design of the #Remdesivir Binding Site in the #RdRp of #SARS-CoV-2: Implications for Potential Resistance (iScience, abstract)

[Source: iScience, full page: (LINK). Abstract, edited.]

High-Throughput Rational Design of the Remdesivir Binding Site in the RdRp of SARS-CoV-2: Implications for Potential Resistance

Aditya K. Padhi, Rohit Shukla, Prakash Saudagar, Timir Tripathi

Open Access | Published: December 26, 2020 | DOI: https://doi.org/10.1016/j.isci.2020.101992

Highlights

  • SARS-CoV-2 may acquire mutations in nsp12 to develop remdesivir resistance.
  • The hotspot residues in nsp12 that exhibited the highest potential for mutation were identified.
  • SARS-CoV-2 can undergo positive selection and attain remdesivir resistance with very few mutations.
  • The work is crucial for the understanding and management of drug resistance.

Summary

The use of remdesivir to treat COVID-19 will likely continue before clinical trials are completed. Due to the lengthening pandemic and evolving nature of the virus, predicting potential residues prone to mutation is crucial for the management of remdesivir resistance. Using a rational ligand-based interface design complemented with mutational mapping, we generated a total of 100,000 mutations and provided insight into the functional outcomes of mutations in the remdesivir-binding site in nsp12 subunit of RdRp. After designing 46 residues in the remdesivir-binding site of nsp12, the designs retained 97-98% sequence identity, suggesting that very few mutations in nsp12 are required for SARS-CoV-2 to attain remdesivir resistance. Several mutants displayed decreased binding affinity to remdesivir, suggesting drug resistance. These hotspot residues had a higher probability of undergoing selective mutation and thus conferring remdesivir resistance. Identifying the potential residues prone to mutation improves our understanding of SARS-CoV-2 drug resistance and COVID-19 pathogenesis.

Publication History Accepted: December 21, 2020 – Received in revised form: September 28, 2020 – Received: July 13, 2020

Publication stage In Press Accepted Manuscript

Identification DOI: https://doi.org/10.1016/j.isci.2020.101992

Copyright © 2020

Keywords: SARS-CoV-2; COVID-19; Antivirals; Drugs Resistance; Remdesivir.

—–

Increase in #HA #Carbapenem-Resistant #Acinetobacter baumannii #Infection and #Colonization in an Acute Care #Hospital During a Surge in #COVID19 Admissions — #NJ, February–July 2020 (MMWR Morb Mortal Wkly Rep., abstract)

[Source: US Centers for Disease Control and Prevention (CDC), MMWR Morbidity and Mortality Weekly Report, full page: (LINK). Abstract, edited.]

Increase in Hospital-Acquired Carbapenem-Resistant Acinetobacter baumannii Infection and Colonization in an Acute Care Hospital During a Surge in COVID-19 Admissions — New Jersey, February–July 2020

Early Release / December 1, 2020 / 69

Stephen Perez, PhD1,2; Gabriel K. Innes, VMD, PhD2; Maroya Spalding Walters, PhD3; Jason Mehr, MPH2; Jessica Arias2; Rebecca Greeley, MPH2; Debra Chew, MD4

Summary

  • What is already known about this topic?
    • Carbapenem-resistant Acinetobacter baumannii (CRAB) causes health care–associated infections that are challenging to contain and often linked to infection prevention and control (IPC) breaches.
  • What is added by this report?
    • A New Jersey hospital reported a cluster of 34 CRAB cases that peaked during a surge in COVID-19 hospitalizations. Strategies to preserve continuity of care led to deviations in IPC practices; CRAB cases decreased when normal operations resumed.
  • What are the implications for public health practice?
    • Hospitals managing surges of patients with COVID-19 might be vulnerable to outbreaks of multidrug-resistant organism (MDRO) infections. Maintaining IPC best practices (e.g., MDRO surveillance and hand hygiene and environmental cleaning audits) to the extent possible could mitigate spread.

Abstract

Carbapenem-resistant Acinetobacter baumannii (CRAB), an opportunistic pathogen primarily associated with hospital-acquired infections, is an urgent public health threat (1). In health care facilities, CRAB readily contaminates the patient care environment and health care providers’ hands, survives for extended periods on dry surfaces, and can be spread by asymptomatically colonized persons; these factors make CRAB outbreaks in acute care hospitals difficult to control (2,3). On May 28, 2020, a New Jersey hospital (hospital A) reported a cluster of CRAB infections during a surge in patients hospitalized with coronavirus disease 2019 (COVID-19). Hospital A and the New Jersey Department of Health (NJDOH) conducted an investigation, and identified 34 patients with hospital-acquired multidrug-resistant CRAB infection or colonization during February–July 2020, including 21 (62%) who were admitted to two intensive care units (ICUs) dedicated to caring for COVID-19 patients. In late March, increasing COVID-19–related hospitalizations led to shortages in personnel, personal protective equipment (PPE), and medical equipment, resulting in changes to conventional infection prevention and control (IPC) practices. In late May, hospital A resumed normal operations, including standard IPC measures, as COVID-19 hospitalizations decreased, lessening the impact of personnel and supply chain shortages on hospital functions. CRAB cases subsequently returned to a pre–COVID-19 baseline of none to two cases monthly. The occurrence of this cluster underscores the potential for multidrug-resistant organisms (MDROs) to spread during events when standard hospital practices might be disrupted; conventional IPC strategies should be reinstated as soon as capacity and resources allow.

(…)

Keywords: Antibiotics; Drugs Resistance; Carbapenem; Acinetobacter baumannii; SARS-CoV-2; COVID-19; Nosocomial Outbreaks; USA.

——

#Antimicrobial #management of respiratory #infections in #SARS-CoV-2 patients: Clinical and ASP conundrums (Open Forum Infect Dis., abstract)

[Source: Open Forum Infectious Diseases, full page: (LINK). Abstract, edited.]

Antimicrobial management of respiratory infections in SARS-CoV-2 patients: Clinical and ASP conundrums

Ellie J C Goldstein, MD, Glenn Tillotson, PhD, Mark Redell, PharmD

Open Forum Infectious Diseases, ofaa517, https://doi.org/10.1093/ofid/ofaa517

Published: 26 October 2020

Abstract

The role of empirical and even directed antimicrobial management of patients hospitalized with SARS- CoV-2 infection is problematic; antibiotics are used frequently among these patients whether to treat confirmed or suspected co-infection or simply symptoms. In the rapidly changing clinical landscape of SARS- CoV-2 there is minimal guidance for selecting appropriate treatment versus non-antimicrobial treatment, and clinicians are pressed to make daily decisions under the stress of absence of data while watching patients deteriorate. We review current data and patterns of antimicrobial use and the potential approach for antimicrobial stewardship in the context of SARS- CoV-2.

Issue Section: Major Article

This content is only available as a PDF.

© The Author(s) 2020. Published by Oxford University Press on behalf of Infectious Diseases Society of America.

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs licence (http://creativecommons.org/licenses/by-nc-nd/4.0/), which permits non-commercial reproduction and distribution of the work, in any medium, provided the original work is not altered or transformed in any way, and that the work is properly cited. For commercial re-use, please contact journals.permissions@oup.com

Keywords: SARS-CoV-2; COVID-19; Antibiotics; Drugs Resistance.

——

In vitro profiling of #laninamivir #resistant substitutions in #N3 to #N9 #avian #influenza #neuraminidase subtypes and their association with in vivo susceptibility (J Virol., abstract)

[Source: Journal of Virology, full page: (LINK). Abstract, edited.]

In vitro profiling of laninamivir-resistant substitutions in N3 to N9 avian influenza neuraminidase subtypes and their association with in vivo susceptibility

Ju Hwan Jeong, Won-Suk Choi, Khristine Joy C. Antigua, Young Ki Choi, Elena A. Govorkova, Richard J. Webby, Yun Hee Baek, Min-Suk Song

DOI: 10.1128/JVI.01679-20

ABSTRACT

Laninamivir (LAN) is a long-acting neuraminidase (NA) inhibitor (NAI) with a similar binding profile in the influenza NA enzyme active site as that of other NAIs, oseltamivir (OS), zanamivir (ZAN), and peramivir, and may share common resistance markers with these NAIs. We screened viruses with NA substitutions previously found during OS and ZAN selection in avian influenza viruses (AIV) of the N3 to N9 subtypes for LAN susceptibility. Of the 72 NA substitutions, 19 conferred resistance to LAN, which ranged from an 11.2- to 549.8-fold decreased inhibitory activity over that of their parental viruses. Ten NA substitutions reduced the susceptibility to all four NAIs, whereas the remaining 26 substitutions yielded susceptibility to one or more NAIs. To determine whether the in vitro susceptibility of multi-NAI resistant AIVs is associated with in vivo susceptibility, we infected BALB/c mice with recombinant AIVs with R292K (ma81k-N3R292K) or Q136K (ma81k-N8Q136K) NA substitutions, which impart in vitro susceptibility only to LAN or OS, respectively. Both ma81k-N3R292K- and ma81k-N8Q136K virus-infected mice exhibited reduced weight loss, mortality, and lung viral titers when treated with their susceptible NAIs, confirming the in vitro susceptibility of these substitutions. Together, LAN resistance profiling of AIVs of a range of NA subtypes improves understanding of NAI-resistance mechanisms. Furthermore, the association of in vitro and in vivo NAI-susceptibility indicates that our models are useful tools for monitoring NAI-susceptibility of AIVs.

IMPORTANCE

The chemical structures of neuraminidase inhibitors (NAIs) possess similarities, but slight differences can result in variable susceptibility of avian influenza viruses (AIVs) carrying resistance-associated NA substitutions. Therefore, comprehensive susceptibility profiling of these substitutions in AIVs is critical for understanding the mechanism of antiviral resistance. In this study, we profiled resistance to the anti-influenza drug laninamivir in AIVs with substitutions known to impart resistance to other NAIs. We found 10 substitutions that conferred resistance to all four NAIs tested. On the other hand, we found the remaining 26 NA substitutions susceptible to at least one or more NAIs and showed for a small selection that in vitro data predicted in vivo behavior. Therefore, our findings highlight the usefulness of screening resistance markers in NA enzyme inhibition assay and animal models of AIV infections.

Copyright © 2020 American Society for Microbiology. All Rights Reserved.

Keywords: Influenza A; Avian Influenza; Antivirals; Drugs Resistance; Laninamivir.

——