Try a new search

Format these results:

Searched for:

person:duerrr02

in-biosketch:yes

Total Results:

67


Antibody Titers against Mpox Virus after Vaccination [Letter]

Kottkamp, Angelica C; Samanovic, Marie I; Duerr, Ralf; Oom, Aaron L; Belli, Hayley M; Zucker, Jane R; Rosen, Jennifer B; Mulligan, Mark J; ,
PMID: 38091537
ISSN: 1533-4406
CID: 5589312

Selective adaptation of SARS-CoV-2 Omicron under booster vaccine pressure: a multicentre observational study

Duerr, Ralf; Dimartino, Dacia; Marier, Christian; Zappile, Paul; Wang, Guiqing; François, Fritz; Ortigoza, Mila B; Iturrate, Eduardo; Samanovic, Marie I; Mulligan, Mark J; Heguy, Adriana
BACKGROUND:High rates of vaccination and natural infection drive immunity and redirect selective viral adaptation. Updated boosters are installed to cope with drifted viruses, yet data on adaptive evolution under increasing immune pressure in a real-world situation are lacking. METHODS:Cross-sectional study to characterise SARS-CoV-2 mutational dynamics and selective adaptation over >1 year in relation to vaccine status, viral phylogenetics, and associated clinical and demographic variables. FINDINGS/RESULTS:The study of >5400 SARS-CoV-2 infections between July 2021 and August 2022 in metropolitan New York portrayed the evolutionary transition from Delta to Omicron BA.1-BA.5 variants. Booster vaccinations were implemented during the Delta wave, yet booster breakthrough infections and SARS-CoV-2 re-infections were almost exclusive to Omicron. In adjusted logistic regression analyses, BA.1, BA.2, and BA.5 had a significant growth advantage over co-occurring lineages in the boosted population, unlike BA.2.12.1 or BA.4. Selection pressure by booster shots translated into diffuse adaptive evolution in Delta spike, contrasting with strong, receptor-binding motif-focused adaptive evolution in BA.2-BA.5 spike (Fisher Exact tests; non-synonymous/synonymous mutation rates per site). Convergent evolution has become common in Omicron, engaging spike positions crucial for immune escape, receptor binding, or cleavage. INTERPRETATION/CONCLUSIONS:Booster shots are required to cope with gaps in immunity. Their discriminative immune pressure contributes to their effectiveness but also requires monitoring of selective viral adaptation processes. Omicron BA.2 and BA.5 had a selective advantage under booster vaccination pressure, contributing to the evolution of BA.2 and BA.5 sublineages and recombinant forms that predominate in 2023. FUNDING/BACKGROUND:The study was supported by NYU institutional funds and partly by the Cancer Center Support Grant P30CA016087 at the Laura and Isaac Perlmutter Cancer Center.
PMCID:10623172
PMID: 37866115
ISSN: 2352-3964
CID: 5609742

Plasmatic HIV-1 soluble gp120 is associated with correlates of immune dysfunction and inflammation in ART-treated individuals with undetectable viremia

Benlarbi, Mehdi; Richard, Jonathan; Bourassa, Catherine; Tolbert, William D; Chartrand-Lefebvre, Carl; Gendron-Lepage, Gabrielle; Sylla, Mohamed; El-Far, Mohamed; Messier-Peet, Marc; Guertin, Camille; Turcotte, Isabelle; Fromentin, Rémi; Verly, Myriam Maude; Prévost, Jérémie; Clark, Andrew; Mothes, Walther; Kaufmann, Daniel E; Maldarelli, Frank; Chomont, Nicolas; Bégin, Philippe; Tremblay, Cécile; Baril, Jean-Guy; Trottier, Benoit; Trottier, Sylvie; Duerr, Ralf; Pazgier, Marzena; Durand, Madeleine; Finzi, Andrés
BACKGROUND:Chronic inflammation persists in some people living with HIV (PLWH) during antiretroviral therapy and is associated with premature aging. The gp120 subunit of HIV-1 envelope sheds and can be detected in plasma, showing immunomodulatory properties even in the absence of detectable viremia. We evaluated whether plasmatic soluble gp120 (sgp120) and a family of gp120-specific anti-cluster A antibodies, linked to CD4 depletion in vitro, contribute to chronic inflammation, immune dysfunction, and sub-clinical cardiovascular disease in participants of the Canadian HIV and Aging cohort (CHACS) with undetectable viremia. METHODS:Cross-sectional assessment of sgp120 and anti-cluster A antibodies was performed in 386 individuals from CHACS. Their association with pro-inflammatory cytokines and subclinical coronary artery disease was assessed using linear regression models. RESULTS:High levels of sgp120 and anti-cluster A antibodies inversely correlated with CD4 count and CD4:CD8 ratio. The presence of sgp120 was associated with increased levels of IL-6. In participants with detectable atherosclerotic plaque and detectable sgp120, anti-cluster A antibodies and their combination with sgp120 levels correlated positively with the total volume of atherosclerotic plaques. CONCLUSIONS:sgp120 may act as a pan toxin causing immune dysfunction and sustained inflammation in a subset of PLWH, contributing to the development of premature comorbidities.
PMID: 38035854
ISSN: 1537-6613
CID: 5616992

Emergence and dissemination of SARS-CoV-2 XBB.1.5 in New York

Gámbaro, Fabiana; Duerr, Ralf; Dimartino, Dacia; Marier, Christian; Iturrate, Eduardo; Mulligan, Mark J.; Heguy, Adriana; Dellicour, Simon
The recombinant SARS-CoV-2 Omicron XBB.1.5 variant was first detected in New York City (NYC) and rapidly became the predominant variant in the area by early 2023. The increased occurrence of circulating variants within the SARS-CoV-2 XBB-sublineage prompted the modification of COVID-19 mRNA vaccines by Moderna and Pfizer-BioNTech. This update, implemented in mid-September 2023, involved the incorporation of a monovalent XBB.1.5 component. Considering that NYC probably played a central role in the emergence of the XBB.1.5 variant, we conducted phylogeographic analysis to investigate the emergence and spread of this variant in the metropolitan area. Our analysis confirms that XBB.1.5 emerged within or near the NYC area and indicates that XBB.1.5 had a diffusion velocity similar to that of the variant Alpha in the same study area. Additionally, the analysis of 2,392 genomes collected in the context of the genomic surveillance program at NYU Langone Health system showed that there was no increased proportion of XBB.1.5, relative to all cocirculating variants, in the boosted compared to unvaccinated individuals. This study provides a comprehensive description of the emergence and dissemination of XBB.1.5.
SCOPUS:85194000862
ISSN: 2057-1577
CID: 5660512

Emergence and dissemination of SARS-CoV-2 XBB.1.5 in New York

Gámbaro, Fabiana; Duerr, Ralf; Dimartino, Dacia; Marier, Christian; Iturrate, Eduardo; Mulligan, Mark J; Heguy, Adriana; Dellicour, Simon
The recombinant SARS-CoV-2 Omicron XBB.1.5 variant was first detected in New York City (NYC) and rapidly became the predominant variant in the area by early 2023. The increased occurrence of circulating variants within the SARS-CoV-2 XBB-sublineage prompted the modification of COVID-19 mRNA vaccines by Moderna and Pfizer-BioNTech. This update, implemented in mid-September 2023, involved the incorporation of a monovalent XBB.1.5 component. Considering that NYC probably played a central role in the emergence of the XBB.1.5 variant, we conducted phylogeographic analysis to investigate the emergence and spread of this variant in the metropolitan area. Our analysis confirms that XBB.1.5 emerged within or near the NYC area and indicates that XBB.1.5 had a diffusion velocity similar to that of the variant Alpha in the same study area. Additionally, the analysis of 2,392 genomes collected in the context of the genomic surveillance program at NYU Langone Health system showed that there was no increased proportion of XBB.1.5, relative to all cocirculating variants, in the boosted compared to unvaccinated individuals. This study provides a comprehensive description of the emergence and dissemination of XBB.1.5.
PMCID:11108082
PMID: 38774310
ISSN: 2057-1577
CID: 5654532

Generation of quality-controlled SARS-CoV-2 variant stocks

de Vries, Maren; Ciabattoni, Grace O; Rodriguez-Rodriguez, Bruno A; Crosse, Keaton M; Papandrea, Dominick; Samanovic, Marie I; Dimartino, Dacia; Marier, Christian; Mulligan, Mark J; Heguy, Adriana; Desvignes, Ludovic; Duerr, Ralf; Dittmann, Meike
One of the main challenges in the fight against coronavirus disease 2019 (COVID-19) stems from the ongoing evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into multiple variants. To address this hurdle, research groups around the world have independently developed protocols to isolate these variants from clinical samples. These isolates are then used in translational and basic research-for example, in vaccine development, drug screening or characterizing SARS-CoV-2 biology and pathogenesis. However, over the course of the COVID-19 pandemic, we have learned that the introduction of artefacts during both in vitro isolation and subsequent propagation to virus stocks can lessen the validity and reproducibility of data. We propose a rigorous pipeline for the generation of high-quality SARS-CoV-2 variant clonal isolates that minimizes the acquisition of mutations and introduces stringent controls to detect them. Overall, the process includes eight stages: (i) cell maintenance, (ii) isolation of SARS-CoV-2 from clinical specimens, (iii) determination of infectious virus titers by plaque assay, (iv) clonal isolation by plaque purification, (v) whole-virus-genome deep-sequencing, (vi and vii) amplification of selected virus clones to master and working stocks and (viii) sucrose purification. This comprehensive protocol will enable researchers to generate reliable SARS-CoV-2 variant inoculates for in vitro and in vivo experimentation and will facilitate comparisons and collaborative work. Quality-controlled working stocks for most applications can be generated from acquired biorepository virus within 1 month. An additional 5-8 d are required when virus is isolated from clinical swab material, and another 6-7 d is needed for sucrose-purifying the stocks.
PMID: 37833423
ISSN: 1750-2799
CID: 5604402

Dynamics and durability of HIV-1 neutralization are determined by viral replication

Schommers, Philipp; Kim, Dae Sung; Schlotz, Maike; Kreer, Christoph; Eggeling, Ralf; Hake, Anna; Stecher, Melanie; Park, Juyeon; Radford, Caelan E; Dingens, Adam S; Ercanoglu, Meryem S; Gruell, Henning; Odidika, Stanley; Dahlhaus, Marten; Gieselmann, Lutz; Ahmadov, Elvin; Lawong, Rene Y; Heger, Eva; Knops, Elena; Wyen, Christoph; Kümmerle, Tim; Römer, Katja; Scholten, Stefan; Wolf, Timo; Stephan, Christoph; Suárez, Isabelle; Raju, Nagarajan; Adhikari, Anurag; Esser, Stefan; Streeck, Hendrik; Duerr, Ralf; Nanfack, Aubin J; Zolla-Pazner, Susan; Geldmacher, Christof; Geisenberger, Otto; Kroidl, Arne; William, Wiston; Maganga, Lucas; Ntinginya, Nyanda Elias; Georgiev, Ivelin S; Vehreschild, Jörg J; Hoelscher, Michael; Fätkenheuer, Gerd; Lavinder, Jason J; Bloom, Jesse D; Seaman, Michael S; Lehmann, Clara; Pfeifer, Nico; Georgiou, George; Klein, Florian
Human immunodeficiency virus type 1 (HIV-1)-neutralizing antibodies (nAbs) that prevent infection are the main goal of HIV vaccine discovery. But as no nAb-eliciting vaccines are yet available, only data from HIV-1 neutralizers-persons with HIV-1 who naturally develop broad and potent nAbs-can inform about the dynamics and durability of nAb responses in humans, knowledge which is crucial for the design of future HIV-1 vaccine regimens. To address this, we assessed HIV-1-neutralizing immunoglobulin G (IgG) from 2,354 persons with HIV-1 on or off antiretroviral therapy (ART). Infection with non-clade B viruses, CD4+ T cell counts <200 µl-1, being off ART and a longer time off ART were independent predictors of a more potent and broad neutralization. In longitudinal analyses, we found nAb half-lives of 9.3 and 16.9 years in individuals with no- or low-level viremia, respectively, and 4.0 years in persons who newly initiated ART. Finally, in a potent HIV-1 neutralizer, we identified lower fractions of serum nAbs and of nAb-encoding memory B cells after ART initiation, suggesting that a decreasing neutralizing serum activity after antigen withdrawal is due to lower levels of nAbs. These results collectively show that HIV-1-neutralizing responses can persist for several years, even at low antigen levels, suggesting that an HIV-1 vaccine may elicit a durable nAb response.
PMCID:10667105
PMID: 37957379
ISSN: 1546-170x
CID: 5611062

Vaccine Equity: Lessons Learned Exploring Facilitators and Barriers to COVID-19 Vaccination in Urban Black Communities

Parameswaran, Lalitha; Jaysing, Anna; Ding, Helen; Wilkenfeld, Marc; Dean, Ranekka; Wilson, Kesi K; Frank, Olivia; Duerr, Ralf; Mulligan, Mark J
COVID-19 vaccines were developed at unparalleled speed, but racial disparities persist in vaccine uptake. This is a cross-sectional survey that was conducted in mid-2021 in ambulatory clinics across Brooklyn, New York. The objectives of the study were to assess: knowledge of COVID-19, healthcare communication and access, attitudes including trust in the process of vaccine development and mistrust due to racial discrimination, and to determine the relationship of the above to vaccine receipt. 58 respondents self-identified as Black non-Hispanic and completed the survey: the majority were women (79%), <50 years old (65%), employed (66%), and had annual household income <$75,000 (59%). The majority reported having some health insurance (97%) and a regular place of healthcare (95%). 60% of respondents reported COVID-19 vaccination receipt. A significant percentage of the vaccinated group compared to the unvaccinated group scored higher on knowledge questions (91% vs. 65%; p = 0.018), felt it was important that others in the community get vaccinated (89% vs. 65%, p = 0.04), and trusted vaccine safety (86% vs. 35%; p < 0.0001) and effectiveness (88% vs. 48%; p < 0.001). The unvaccinated group reported a lower annual household income of <$75,000 (72% vs. 50%; p = 0.0002) and also differed by employment status (p = 0.04). Majority in both groups agreed that racial discrimination interferes with healthcare (78%). In summary, unvaccinated Black non-Hispanic respondents report significant concerns about vaccine safety and efficacy and have greater mistrust in the vaccine development process. The relationship between racial discrimination, mistrust, and vaccine hesitancy needs further study in order to improve vaccine uptake in this population.
PMID: 37391605
ISSN: 2196-8837
CID: 5540652

A neonatal mouse model characterizes transmissibility of SARS-CoV-2 variants and reveals a role for ORF8

Rodriguez-Rodriguez, Bruno A; Ciabattoni, Grace O; Duerr, Ralf; Valero-Jimenez, Ana M; Yeung, Stephen T; Crosse, Keaton M; Schinlever, Austin R; Bernard-Raichon, Lucie; Rodriguez Galvan, Joaquin; McGrath, Marisa E; Vashee, Sanjay; Xue, Yong; Loomis, Cynthia A; Khanna, Kamal M; Cadwell, Ken; Desvignes, Ludovic; Frieman, Matthew B; Ortigoza, Mila B; Dittmann, Meike
Small animal models have been a challenge for the study of SARS-CoV-2 transmission, with most investigators using golden hamsters or ferrets. Mice have the advantages of low cost, wide availability, less regulatory and husbandry challenges, and the existence of a versatile reagent and genetic toolbox. However, adult mice do not robustly transmit SARS-CoV-2. Here we establish a model based on neonatal mice that allows for transmission of clinical SARS-CoV-2 isolates. We characterize tropism, respiratory tract replication and transmission of ancestral WA-1 compared to variants Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), Omicron BA.1 and Omicron BQ.1.1. We identify inter-variant differences in timing and magnitude of infectious particle shedding from index mice, both of which shape transmission to contact mice. Furthermore, we characterize two recombinant SARS-CoV-2 lacking either the ORF6 or ORF8 host antagonists. The removal of ORF8 shifts viral replication towards the lower respiratory tract, resulting in significantly delayed and reduced transmission in our model. Our results demonstrate the potential of our neonatal mouse model to characterize viral and host determinants of SARS-CoV-2 transmission, while revealing a role for an accessory protein in this context.
PMID: 37230979
ISSN: 2041-1723
CID: 5508612

Local and Systemic Antibody Response to SARS-CoV-2 Infection in Critically Ill COVID-19 Patients

Barnett, C.R.; Krolikowski, K.; Tsay, J.J.; Wu, B.G.; Li, Y.; Chang, M.; Kyeremateng, Y.; Brosnahan, S.; Singh, S.; Kocak, I.; Collazo, D.E.; Mukherjee, V.; Lubinsky, A.S.; Postelnicu, R.; Ghedin, E.; Chung, M.; Angel, L.F.; Sulaiman, I.; Duerr, R.; Schluger, R.; Rafeq, S.; Carpenito, J.; Bakker, J.; Amoroso, N.E.; Kaufman, D.A.; Pradhan, D.; Li, H.; Wang, C.; Silverman, G.; Segal, L.N.
ORIGINAL:0017185
ISSN: 1535-4970
CID: 5651662