Go to The Journal of Clinical Investigation
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Transfers
  • Advertising
  • Job board
  • Contact
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Immunology
    • Metabolism
    • Nephrology
    • Oncology
    • Pulmonology
    • All ...
  • Videos
  • Collections
    • Resource and Technical Advances
    • Clinical Medicine
    • Reviews
    • Editorials
    • Perspectives
    • Top read articles
  • JCI This Month
    • Current issue
    • Past issues

  • Current issue
  • Past issues
  • Specialties
  • In-Press Preview
  • Editorials
  • Viewpoint
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Transfers
  • Advertising
  • Job board
  • Contact

COVID-19s

  • 110 Articles
  • 0 Posts
  • ← Previous
  • 1
  • 2
  • 3
  • 4
  • 5
  • 6
  • …
  • 10
  • 11
  • Next →
Durability of ChAdOx1 nCov-19 vaccination in people living with HIV
Ane Ogbe, … , Teresa Lambe, John Frater
Ane Ogbe, … , Teresa Lambe, John Frater
Published February 22, 2022
Citation Information: JCI Insight. 2022. https://doi.org/10.1172/jci.insight.157031.
View: Text | PDF

Durability of ChAdOx1 nCov-19 vaccination in people living with HIV

  • Text
  • PDF
Abstract

Duration of protection from SARS-CoV-2 infection in people with HIV (PWH) following vaccination is unclear. In a sub-study of the phase 2/3 the COV002 trial (NCT04400838), 54 HIV positive male participants on antiretroviral therapy (undetectable viral loads, CD4+ T cells >350 cells/ul) received two doses of ChAdOx1 nCoV-19 (AZD1222) 4-6 weeks apart and were followed for 6 months. Responses to vaccination were determined by serology (IgG ELISA and MesoScale Discovery (MSD)), neutralisation, ACE-2 inhibition, gamma interferon ELISpot, activation-induced marker (AIM) assay and T cell proliferation. We show that 6 months after vaccination the majority of measurable immune responses were greater than pre-vaccination baseline, but with evidence of a decline in both humoral and cell mediated immunity. There was, however, no significant difference compared to a cohort of HIV-uninfected individuals vaccinated with the same regimen. Responses to the variants of concern were detectable, although were lower than wild type. Pre-existing cross-reactive T cell responses to SARS-CoV-2 spike were associated with greater post-vaccine immunity and correlated with prior exposure to beta coronaviruses. These data support the on-going policy to vaccinate PWH against SARS-CoV-2, and underpin the need for long-term monitoring of responses after vaccination.

Authors

Ane Ogbe, Matthew Pace, Mustapha Bittaye, Timothy Tipoe, Sandra Adele, Jasmini Alagaratnam, Parvinder K. Aley, M. Azim Ansari, Anna Bara, Samantha Broadhead, Anthony Brown, Helen Brown, Federica Cappuccini, Paola Cinardo, Wanwisa Dejnirattisai, Katie Ewer, Henry Fok, Pedro M. Folegatti, Jamie Fowler, Leila Godfrey, Anna L. Goodman, Bethany Jackson, Daniel Jenkin, Mathew Jones, Stephanie Longet, Rebecca A. Makinson, Natalie G. Marchevsky, Moncy Mathew, Andrea Mazzella, Yama F. Mujadidi, Lucia Parolini, Claire Petersen, Emma Plested, Katrina Pollock, Thurkka Rajeswaran, Maheshi N. Ramasamy, Sarah Rhead, Hannah Robinson, Nicola Robinson, Helen Sanders, Sonia Serrano Fandos, Tom Tipton, Anele Waters, Panagiota Zacharopoulou, Eleanor Barnes, Susanna Dunachie, Philip Goulder, Paul Klenerman, Gavin R. Screaton, Alan Winston, Adrian V.S. Hill, Sarah C. Gilbert, Miles Carroll, Andrew J. Pollard, Sarah Fidler, Julie Fox, Teresa Lambe, John Frater

×

Disulfiram inhibits neutrophil extracellular trap formation protecting rodents from acute lung injury and SARS-CoV-2 infection
Jose M. Adrover, … , Robert E. Schwartz, Mikala Egeblad
Jose M. Adrover, … , Robert E. Schwartz, Mikala Egeblad
Published February 8, 2022
Citation Information: JCI Insight. 2022. https://doi.org/10.1172/jci.insight.157342.
View: Text | PDF

Disulfiram inhibits neutrophil extracellular trap formation protecting rodents from acute lung injury and SARS-CoV-2 infection

  • Text
  • PDF
Abstract

Severe acute lung injury has few treatment options and a high mortality rate. Upon injury, neutrophils infiltrate the lungs and form neutrophil extracellular traps (NETs), damaging the lungs and driving an exacerbated immune response. Unfortunately, no drug preventing NET formation has completed clinical development. Here, we report that disulfiram —an FDA-approved drug for alcohol use disorder— dramatically reduced NETs, increased survival, improved blood oxygenation, and reduced lung edema in a transfusion-related acute lung injury (TRALI) mouse model. We then tested whether disulfiram could confer protection in the context of SARS-CoV-2 infection, as NETs are elevated in patients with severe COVID-19. In SARS-CoV-2-infected golden hamsters, disulfiram reduced NETs and perivascular fibrosis in the lungs, and downregulated innate immune and complement/coagulation pathways, suggesting that it could be beneficial for COVID-19 patients. In conclusion, an existing FDA-approved drug can block NET formation and improve disease course in two rodent models of lung injury for which treatment options are limited.

Authors

Jose M. Adrover, Lucia Carrau, Juliane Daßler-Plenker, Yaron Bram, Vasuretha Chandar, Sean Houghton, David Redmond, Joseph R. Merrill, Margaret Shevik, Benjamin R. tenOever, Scott K. Lyons, Robert E. Schwartz, Mikala Egeblad

×

Adaptive immune responses in vaccinated patients with symptomatic SARS-CoV-2 Alpha infection
Han-Sol Park, … , David Sullivan, Kelly A. Gebo
Han-Sol Park, … , David Sullivan, Kelly A. Gebo
Published February 1, 2022
Citation Information: JCI Insight. 2022. https://doi.org/10.1172/jci.insight.155944.
View: Text | PDF

Adaptive immune responses in vaccinated patients with symptomatic SARS-CoV-2 Alpha infection

  • Text
  • PDF
Abstract

Benchmarks for protective immunity from infection or severe disease after SARS-CoV-2 vaccination are still being defined. Here we characterized virus neutralizing and ELISA antibody levels, cellular immune responses, and viral variants in 4 separate groups: Healthy control participants weeks (early) or months (late) following vaccination in comparison to symptomatic SARS-CoV-2 infections after partial or full mRNA vaccination. During the study time, most symptomatic breakthrough infections were caused by the SARS-CoV-2 Alpha variant. Neutralizing antibody levels in the healthy controls were sustained over time against the vaccine parent virus, but decreased against the Alpha variant, whereas IgG titers and T cell responses against the parent virus and Alpha variant declined over time in healthy controls. Both partially and fully vaccinated patients with symptomatic infections had lower virus neutralizing antibody levels against parent virus than the healthy controls, similar IgG antibody titers and similar virus-specific T cell responses measured by IFN-γ. Compared to healthy controls, neutralization activity against the Alpha variant was lower in the partially vaccinated infected patients and tended toward lower in the fully vaccinated infected patients. In this cohort of breakthrough infections, parent virus neutralization was the superior predictor of breakthrough infections with the Alpha variant of SARS-CoV-2.

Authors

Han-Sol Park, Janna R. Shapiro, Ioannis Sitaras, Bezawit A. Woldemeskel, Caroline Garliss, Amanda Dziedzic, Jaiprasath Sachithanandham, Anne E. Jedlicka, Christopher A. Caputo, Kimberly E. Rousseau, Manjusha Thakar, San Suwanmanee, Pricila Hauk, Lateef Aliyu, Natalia I. Majewska, Sushmita Koley, Bela Patel, Patrick Broderick, Giselle Mosnaim, Sonya L. Heath, Emily S. Spivak, Aarthi Shenoy, Evan M. Bloch, Thomas J. Gniadek, Shmuel Shoham, Arturo Casadevall, Daniel Hanley, Andrea L. Cox, Oliver Laeyendecker, Michael Betenbaugh, Steven M. Cramer, Heba H. Mostafa, Andrew Pekosz, Joel N. Blankson, Sabra L. Klein, Aaron A.R. Tobian, David Sullivan, Kelly A. Gebo

×

Limited induction of SARS-CoV-2-specific T cell responses in children with multisystem inflammatory syndrome compared to COVID-19
Vidisha Singh, … , Christina A. Rostad, Ann Chahroudi
Vidisha Singh, … , Christina A. Rostad, Ann Chahroudi
Published January 19, 2022
Citation Information: JCI Insight. 2022. https://doi.org/10.1172/jci.insight.155145.
View: Text | PDF

Limited induction of SARS-CoV-2-specific T cell responses in children with multisystem inflammatory syndrome compared to COVID-19

  • Text
  • PDF
Abstract

Why Multisystem Inflammatory Syndrome in Children (MIS-C) develops after SARS-CoV-2 infection in a subset of children is unknown. We hypothesized that aberrant virus52 specific T-cell responses contribute to MIS-C pathogenesis. We quantified SARS-CoV-2 reactive T-cells, serologic responses against major viral proteins, and cytokine responses from plasma and peripheral blood mononuclear cells in children with convalescent COVID-19, acute MIS-C, and healthy controls. Children with MIS-C had significantly lower virus-specific CD4+ and CD8+ T-cell responses to major SARS-CoV-2 antigens compared with children convalescing from COVID-19. Further, T-cell responses in participants with MIS-C were similar to or lower than those in healthy controls. Serologic responses against spike receptor binding domain (RBD), full-length spike, and nucleocapsid were similar among convalescent COVID-19 and MIS-C, suggesting functional B cell responses. Cytokine profiling demonstrated predominant Th1 polarization of CD4+ T-cells from children with convalescent COVID-19 and MIS-C, although cytokine production was reduced in MIS-C. Our findings support a role for constrained induction of anti-SARS-CoV-2-specific T-cells in the pathogenesis of MIS-C.

Authors

Vidisha Singh, Veronica Obregon-Perko, Stacey A. Lapp, Anna M. Horner, Alyssa Brooks, Lisa Macoy, Laila Hussaini, Austin Lu, Theda Gibson, Guido Silvestri, Alba Grifoni, Daniela Weiskopf, Alessandro Sette, Evan J. Anderson, Christina A. Rostad, Ann Chahroudi

×

Multiple sclerosis therapies differentially impact SARS-CoV-2 vaccine-induced antibody and T cell immunity and function
Joseph J. Sabatino Jr, … , Scott S. Zamvil, Riley Bove
Joseph J. Sabatino Jr, … , Scott S. Zamvil, Riley Bove
Published January 14, 2022
Citation Information: JCI Insight. 2022. https://doi.org/10.1172/jci.insight.156978.
View: Text | PDF

Multiple sclerosis therapies differentially impact SARS-CoV-2 vaccine-induced antibody and T cell immunity and function

  • Text
  • PDF
Abstract

BACKGROUND. Vaccine-elicited adaptive immunity is a prerequisite for control of SARS-CoV-2 infection. Multiple sclerosis (MS) disease-modifying therapies (DMTs) differentially target humoral and cellular immunity. A comprehensive comparison of MS DMTs on SARS-CoV-2 vaccine-specific immunity is needed, including quantitative and functional B and T cell responses. METHODS. Spike-specific antibody and T cell responses were measured before and following SARS-CoV-2 vaccination in a cohort of 80 subjects, including healthy controls and MS patients in six DMT groups: untreated, glatiramer acetate (GA), dimethyl fumarate (DMF), natalizumab (NTZ), sphingosine-1-phosphate (S1P) receptor modulators, and anti-CD20 monoclonal antibodies. Anti-spike antibody responses were quantified by Luminex assay, high-resolution spike epitope reactivity was mapped by VirScan, and pseudovirus neutralization was assessed. Spike-specific CD4+ and CD8+ T cell responses were characterized by activation-induced marker (AIM) expression, cytokine production, and tetramer analysis. RESULTS. Anti-spike IgG levels were similar between healthy controls, untreated MS, GA, DMF, and NTZ patients, but were significantly reduced in anti-CD20 and S1P-treated patients. Anti-spike seropositivity in anti-CD20 patients was significantly correlated with CD19+ B cell levels and inversely correlated with cumulative treatment duration. Spike epitope reactivity and pseudovirus neutralization was reduced in anti-CD20 and S1P patients, directly correlating with reduced spike receptor binding domain (RBD) IgG levels. Spike-specific CD4+ and CD8+ T cell reactivity remained robust across all groups except in S1P-treated patients in whom post-vaccine CD4+ T cell responses were attenuated. CONCLUSIONS. These findings from a large MS cohort exposed to a wide spectrum of MS immunotherapies have important implications for treatment-specific COVID-19 clinical guidelines. FUNDING. This work was supported by grants from the NIH 1K08NS107619 (JJS), NMSS TA- 1903-33713 (JJS), K08NS096117 (MRW), Westridge Foundation (MRW), Chan Zuckerberg Biohub (JLD), R01AI159260 (JAH), R01NS092835 (SSZ), R01AI131624 (SSZ), R21NS108159 (SSZ), NMSS RG1701-26628 (SSZ), and the Maisin Foundation (SSZ).

Authors

Joseph J. Sabatino Jr, Kristen Mittl, William M. Rowles, Kira McPolin, Jayant V. Rajan, Matthew T. Laurie, Colin R. Zamecnik, Ravi Dandekar, Bonny D. Alvarenga, Rita P. Loudermilk, Chloe Gerungan, Collin M. Spencer, Sharon A. Sagan, Danillo G. Augusto, Jessa R. Alexander, Joseph L. DeRisi, Jill A. Hollenbach, Michael R. Wilson, Scott S. Zamvil, Riley Bove

×

SARS-CoV-2 BNT162b2 vaccine-induced humoral response and reactogenicity in individuals with prior COVID-19 disease
Steven G. Kelsen, … , Michael R. Ruggieri Sr., Nina T. Gentile
Steven G. Kelsen, … , Michael R. Ruggieri Sr., Nina T. Gentile
Published January 12, 2022
Citation Information: JCI Insight. 2022. https://doi.org/10.1172/jci.insight.155889.
View: Text | PDF

SARS-CoV-2 BNT162b2 vaccine-induced humoral response and reactogenicity in individuals with prior COVID-19 disease

  • Text
  • PDF
Abstract

BACKGROUND. Most subjects with prior COVID-19 disease manifest long–term, protective immune responses against re-infection. Accordingly, we tested the hypothesis that humoral immune and reactogenicity responses to a SARS-CoV-2 mRNA vaccine differ in subjects with and without prior COVID-19. METHODS. Health care workers (n=61) with (n=30) and without (n=31) prior COVID-19 disease received two, 30 µg doses of Pfizer BNT162b2 vaccine 3 weeks apart. Serum IgG antibody against the Spike receptor-binding domain (RBD); serum neutralizing activity; and vaccine reactogenicity were assessed longitudinally every 2 weeks for 56 days after the 1st injection. RESULTS. The COVID group manifested more rapid increases in Spike IgG antibody and serum neutralizing activity post 1st vaccine dose but showed little or no increase after the 2nd dose compared to the infection-naïve group. In fact, Spike IgG was maximum after the 1st dose in 36% of the COVID group versus 0% of the infection-naïve group. Peak IgG antibody was lower but appeared to fall more slowly in the COVID-19 versus the infection-naïve group. Finally, adverse systemic reactions e.g., fever, headache and malaise, were more frequent and lasted longer after both the 1st and 2nd injection in the COVID group than in the infection-naïve group. CONCLUSION. Subjects with prior COVID-19 demonstrate a robust, accelerated humoral immune response to the 1st dose but attenuated response to the 2nd dose of BNT162b2 vaccine compared to controls. The COVID-19 group also experiences greater reactogenicity. Humoral responses and reactogenicity to BNT162b2 differ qualitatively and quantitatively in subjects with prior COVID-19 compared to infection-naive subjects. FUNDING. This work was supported by Institutional Funds.

Authors

Steven G. Kelsen, Alan S. Braverman, Mark O. Aksoy, Jacob A. Hayman, Puja S. Patel, Charu Rajput, Huaqing Zhao, Susan G. Fisher, Michael R. Ruggieri Sr., Nina T. Gentile

×

Single-cell profiling of T and B cell repertoires following SARS-CoV-2 mRNA vaccine
Suhas Sureshchandra, … , Izabela Coimbra Ibraim, Ilhem Messaoudi
Suhas Sureshchandra, … , Izabela Coimbra Ibraim, Ilhem Messaoudi
Published December 22, 2021
Citation Information: JCI Insight. 2021;6(24):e153201. https://doi.org/10.1172/jci.insight.153201.
View: Text | PDF

Single-cell profiling of T and B cell repertoires following SARS-CoV-2 mRNA vaccine

  • Text
  • PDF
Abstract

mRNA vaccines for SARS-CoV-2 have shown exceptional clinical efficacy, providing robust protection against severe disease. However, our understanding of transcriptional and repertoire changes following full vaccination remains incomplete. We used scRNA-Seq and functional assays to compare humoral and cellular responses to 2 doses of mRNA vaccine with responses observed in convalescent individuals with asymptomatic disease. Our analyses revealed enrichment of spike-specific B cells, activated CD4+ T cells, and robust antigen-specific polyfunctional CD4+ T cell responses following vaccination. On the other hand, although clonally expanded CD8+ T cells were observed following both vaccination and natural infection, CD8+ T cell responses were relatively weak and variable. In addition, TCR gene usage was variable, reflecting the diversity of repertoires and MHC polymorphism in the human population. Natural infection induced expansion of CD8+ T cell clones that occupy distinct clusters compared to those induced by vaccination and likely recognize a broader set of viral antigens of viral epitopes presented by the virus not seen in the mRNA vaccine. Our study highlights a coordinated adaptive immune response in which early CD4+ T cell responses facilitate the development of the B cell response and substantial expansion of effector CD8+ T cells, together capable of contributing to future recall responses.

Authors

Suhas Sureshchandra, Sloan A. Lewis, Brianna M. Doratt, Allen Jankeel, Izabela Coimbra Ibraim, Ilhem Messaoudi

×

Clinico-histopathologic and single nuclei RNA sequencing insights into cardiac injury and microthrombi in critical COVID-19
Michael I. Brener, … , Nathan R. Tucker, Emily J. Tsai
Michael I. Brener, … , Nathan R. Tucker, Emily J. Tsai
Published December 14, 2021
Citation Information: JCI Insight. 2021. https://doi.org/10.1172/jci.insight.154633.
View: Text | PDF

Clinico-histopathologic and single nuclei RNA sequencing insights into cardiac injury and microthrombi in critical COVID-19

  • Text
  • PDF
Abstract

Acute cardiac injury is prevalent in critical COVID-19 and associated with increased mortality. Its etiology remains debated, as initially presumed causes--- myocarditis and cardiac necrosis--- have proven uncommon. To elucidate the pathophysiology of COVID-19-associated cardiac injury, we conducted a prospective study of the first 69 consecutive COVID-19 decedents at Columbia University Irving Medical Center in New York City. Of six acute cardiac histopathologic features, microthrombi was the most commonly detected amongst our cohort (n=48, 70%). We tested associations of cardiac microthrombi with biomarkers of inflammation, cardiac injury, and fibrinolysis and with in-hospital antiplatelet therapy, therapeutic anticoagulation, and corticosteroid treatment, while adjusting for multiple clinical factors, including COVID-19 therapies. Higher peak erythrocyte sedimentation rate and c-reactive protein were independently associated with increased odds of microthrombi, supporting an immunothrombotic etiology. Using single nuclei RNA-sequencing analysis on 3 patients with and 4 patients without cardiac microthrombi, we discovered an enrichment of pro-thrombotic/anti-fibrinolytic, extracellular matrix remodeling, and immune-potentiating signaling amongst cardiac fibroblasts in microthrombi-positive, relative to microthrombi-negative, COVID-19 hearts. Non-COVID-19 non-failing hearts were used as reference controls. Our study identifies a specific transcriptomic signature in cardiac fibroblasts as a salient feature of microthrombi-positive COVID-19 hearts. Our findings warrant further mechanistic study as cardiac fibroblasts may represent a potential therapeutic target for COVID-19-associated cardiac microthrombi.

Authors

Michael I. Brener, Michelle L. Hulke, Nobuaki Fukuma, Stephanie Golob, Robert S. Zilinyi, Zhipeng Zhou, Christos Tzimas, Ilaria Russo, Claire McGroder, Ryan D. Pfeiffer, Alexander Chong, Geping Zhang, Daniel Burkhoff, Martin B. Leon, Mathew S. Maurer, Jeffrey W. Moses, Anne-Catrin Uhlemann, Hanina Hibshoosh, Nir Uriel, Matthias J. Szabolcs, Björn Redfors, Charles C. Marboe, Matthew R. Baldwin, Nathan R. Tucker, Emily J. Tsai

×

Duration of viral shedding and culture positivity with post-vaccination SARS-CoV-2 delta variant infections
Mark J. Siedner, … , Jacob E. Lemieux, Jonathan Z. Li
Mark J. Siedner, … , Jacob E. Lemieux, Jonathan Z. Li
Published December 6, 2021
Citation Information: JCI Insight. 2021. https://doi.org/10.1172/jci.insight.155483.
View: Text | PDF

Duration of viral shedding and culture positivity with post-vaccination SARS-CoV-2 delta variant infections

  • Text
  • PDF
Abstract

Isolation guidelines for severe acute respiratory syndrome–cornavirus-2 (SARS-CoV-2) are largely derived from data collected prior to emergence of the delta variant. We followed a cohort of ambulatory patients with post-vaccination breakthrough SARS-CoV-2 infections with longitudinal collection of nasal swabs for SARS-CoV-2 viral load quantification, whole genome sequencing, and viral culture. All delta variant infections (10/10, 100%) in our cohort were symptomatic, compared with 64% (9/14) of non-delta variant infections. Symptomatic delta variant breakthrough infections were characterized by higher initial viral load, longer duration of virologic shedding by PCR, greater likelihood of replication-competent virus at early stages of infection, and longer duration of culturable virus compared to non-delta variants. The duration of time since vaccination was also correlated with both duration of PCR positivity and duration of detection of replication-competent virus. Nonetheless, no individuals with symptomatic delta variant infections had replication-competent virus by day 10 after symptom onset or 24 hours after resolution of symptoms. These data support current US Center for Disease Control isolation guidelines and reinforce the importance of prompt testing and isolation among symptomatic individuals with delta variant breakthrough infections. Additional data are needed to evaluate these relationships among asymptomatic and more severe delta variant breakthrough infections.

Authors

Mark J. Siedner, Julie Boucau, Rebecca F. Gilbert, Rockib Uddin, Jonathan Luu, Sebastien Haneuse, Tammy Vyas, Zahra Reynolds, Surabhi Iyer, Grace C. Chamberlin, Robert H. Goldstein, Crystal M. North, Chana A. Sacks, James Regan, James P. Flynn, Manish C. Choudhary, Jatin M. Vyas, Amy K. Barczak, Jacob E. Lemieux, Jonathan Z. Li

×

Pharmacokinetics of high-titer anti-SARS-CoV-2 human convalescent plasma in high-risk children
Oren Gordon, … , Sabra L. Klein, Sanjay K. Jain
Oren Gordon, … , Sabra L. Klein, Sanjay K. Jain
Published December 2, 2021
Citation Information: JCI Insight. 2021. https://doi.org/10.1172/jci.insight.151518.
View: Text | PDF

Pharmacokinetics of high-titer anti-SARS-CoV-2 human convalescent plasma in high-risk children

  • Text
  • PDF
Abstract

BACKGROUND. While most children experience mild COVID-19, high-risk children with underlying conditions may develop severe disease, requiring interventions. Kinetics of antibodies transferred via COVID-19 convalescent plasma early in disease, have not been characterized. METHODS. In this study (NCT04377672), high-risk children were prospectively enrolled to receive high-titer COVID-19 convalescent plasma (>1:320 anti-spike IgG; Euroimmun). Passive transfer of antibodies and endogenous antibody production were serially evaluated for up to 2 months after transfusion. Commercial and research ELISA assays, virus neutralization assays, high-throughput phage-display assay utilizing a coronavirus epitope library and pharmacokinetic analyses were performed. RESULTS. Fourteen high-risk children (median age 7.5 years) received high-titer COVID-19 convalescent plasma, nine children within five days (range 2-7) of symptom onset and five children within 4 days (range 3-5) after exposure to SARS-CoV-2. There were no serious adverse events related to transfusion. Antibodies to SARS-CoV-2 were transferred from the donor to the recipient, but antibody titers declined by 14-21 days with a 15.1-day t½ for spike protein IgG. Donor plasma had significant neutralization capacity which was transferred to the recipient. However, as early as 30 minutes post-transfusion, recipient plasma had low neutralization capacity. CONCLUSIONS. Convalescent plasma transfused to high-risk children appears to be safe with expected antibody kinetics, regardless of weight or age. However, current use of convalescent plasma in high-risk children achieves low neutralizing capacity.

Authors

Oren Gordon, Mary Katherine Brosnan, Steve Yoon, Dawoon Jung, Kirsten Littlefield, Abhinaya Ganesan, Christopher A. Caputo, Maggie Li, William R. Morgenlander, Stephanie N. Henson, Alvaro A. Ordonez, Patricia De Jesus, Elizabeth W. Tucker, Nadine Peart Akindele, Zexu Ma, Jo Wilson, Camilo A. Ruiz-Bedoya, M. Elizabeth M. Younger, Evan M. Bloch, Shmuel Shoham, David Sullivan, Aaron A.R. Tobian, Kenneth R. Cooke, Ben Larman, Jogarao V.S. Gobburu, Arturo Casadevall, Andrew Pekosz, Howard M. Lederman, Sabra L. Klein, Sanjay K. Jain

×
  • ← Previous
  • 1
  • 2
  • 3
  • 4
  • 5
  • 6
  • …
  • 10
  • 11
  • Next →

No posts were found with this tag.

Advertisement

Copyright © 2023 American Society for Clinical Investigation
ISSN 2379-3708

Sign up for email alerts