49 results on '"Arunachalam PS"'
Search Results
2. Elucidating allergic reaction mechanisms in response to SARS-CoV-2 mRNA vaccination in adults.
- Author
-
Shah MM, Layhadi JA, Hourcade DE, Fulton WT, Tan TJ, Dunham D, Chang I, Vel MS, Fernandes A, Lee AS, Liu J, Arunachalam PS, Galli SJ, Boyd SD, Pulendran B, Davis MM, O'Hara R, Park H, Mitchell LM, Akk A, Patterson A, Jerath MR, Monroy JM, Ren Z, Kendall PL, Durham SR, Fedina A, Gibbs BF, Agache I, Chinthrajah S, Sindher SB, Heider A, Akdis CA, Shamji MH, Pham CTN, and Nadeau KC
- Subjects
- Humans, Male, Female, Adult, Middle Aged, mRNA Vaccines immunology, Vaccination adverse effects, Hypersensitivity immunology, Hypersensitivity etiology, Immunoglobulin G immunology, Immunoglobulin G blood, Aged, Immunoglobulin E immunology, Immunoglobulin E blood, COVID-19 Vaccines immunology, COVID-19 Vaccines adverse effects, COVID-19 immunology, COVID-19 prevention & control, SARS-CoV-2 immunology, Basophils immunology, Basophils metabolism, Complement Activation immunology
- Abstract
Background: During the COVID-19 pandemic, novel nanoparticle-based mRNA vaccines were developed. A small number of individuals developed allergic reactions to these vaccines although the mechanisms remain undefined., Methods: To understand COVID-19 vaccine-mediated allergic reactions, we enrolled 19 participants who developed allergic events within 2 h of vaccination and 13 controls, nonreactors. Using standard hemolysis assays, we demonstrated that sera from allergic participants induced stronger complement activation compared to nonallergic subjects following ex vivo vaccine exposure., Results: Vaccine-mediated complement activation correlated with anti-polyethelyne glycol (PEG) IgG (but not IgM) levels while anti-PEG IgE was undetectable in all subjects. Depletion of total IgG suppressed complement activation in select individuals. To investigate the effects of vaccine excipients on basophil function, we employed a validated indirect basophil activation test that stratified the allergic populations into high and low responders. Complement C3a and C5a receptor blockade in this system suppressed basophil response, providing strong evidence for complement involvement in vaccine-mediated basophil activation. Single-cell multiome analysis revealed differential expression of genes encoding the cytokine response and Toll-like receptor (TLR) pathways within the monocyte compartment. Differential chromatin accessibility for IL-13 and IL-1B genes was found in allergic and nonallergic participants, suggesting that in vivo, epigenetic modulation of mononuclear phagocyte immunophenotypes determines their subsequent functional responsiveness, contributing to the overall physiologic manifestation of vaccine reactions., Conclusion: These findings provide insights into the mechanisms underlying allergic reactions to COVID-19 mRNA vaccines, which may be used for future vaccine strategies in individuals with prior history of allergies or reactions and reduce vaccine hesitancy., (© 2024 European Academy of Allergy and Clinical Immunology and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
3. A comparative immunological assessment of multiple clinical-stage adjuvants for the R21 malaria vaccine in nonhuman primates.
- Author
-
Arunachalam PS, Ha N, Dennison SM, Spreng RL, Seaton KE, Xiao P, Feng Y, Zarnitsyna VI, Kazmin D, Hu M, Santagata JM, Xie X, Rogers K, Shirreff LM, Chottin C, Spencer AJ, Dutta S, Prieto K, Julien JP, Tomai M, Fox CB, Villinger F, Hill AVS, Tomaras GD, and Pulendran B
- Subjects
- Animals, Macaca mulatta, Adjuvants, Vaccine, Antibodies, Protozoan immunology, Cytokines metabolism, Malaria Vaccines immunology, Adjuvants, Immunologic pharmacology
- Abstract
Authorization of the Matrix-M (MM)-adjuvanted R21 vaccine by three countries and its subsequent endorsement by the World Health Organization for malaria prevention in children are a milestone in the fight against malaria. Yet, our understanding of the innate and adaptive immune responses elicited by this vaccine remains limited. Here, we compared three clinically relevant adjuvants [3M-052 + aluminum hydroxide (Alum) (3M), a TLR7/8 agonist formulated in Alum; GLA-LSQ, a TLR4 agonist formulated in liposomes with QS-21; and MM, the now-approved adjuvant for R21] for their capacity to induce durable immune responses to R21 in macaques. R21 adjuvanted with 3M on a 0, 8, and 23-week schedule elicited anti-circumsporozoite antibody responses comparable in magnitude to the R21/MM vaccine administered using a 0-4-8-week regimen and persisted up to 72 weeks with a half-life of 337 days. A booster dose at 72 weeks induced a recall response similar to the R21/MM vaccination. In contrast, R21/GLA-LSQ immunization induced a lower, short-lived response at the dose used. Consistent with the durable serum antibody responses, MM and 3M induced long-lived plasma cells in the bone marrow and other tissues, including the spleen. Furthermore, whereas 3M stimulated potent and persistent antiviral transcriptional and cytokine signatures after primary and booster immunizations, MM induced enhanced expression of interferon- and T
H 2-related signatures more highly after the booster vaccination. Collectively, these findings provide a resource on the immune responses of three clinically relevant adjuvants with R21 and highlight the promise of 3M as another adjuvant for malarial vaccines.- Published
- 2024
- Full Text
- View/download PDF
4. AS03 adjuvant enhances the magnitude, persistence, and clonal breadth of memory B cell responses to a plant-based COVID-19 vaccine in humans.
- Author
-
Grigoryan L, Feng Y, Bellusci L, Lai L, Wali B, Ellis M, Yuan M, Arunachalam PS, Hu M, Kowli S, Gupta S, Maysel-Auslender S, Maecker HT, Samaha H, Rouphael N, Wilson IA, Moreno AC, Suthar MS, Khurana S, Pillet S, Charland N, Ward BJ, and Pulendran B
- Subjects
- Adult, Humans, Memory B Cells, COVID-19 Vaccines, Antibodies, Viral, Drug Combinations, Influenza Vaccines, Influenza, Human, COVID-19 prevention & control, Polysorbates, Squalene, alpha-Tocopherol
- Abstract
Vaccine adjuvants increase the breadth of serum antibody responses, but whether this is due to the generation of antigen-specific B cell clones with distinct specificities or the maturation of memory B cell clones that produce broadly cross-reactive antibodies is unknown. Here, we longitudinally analyzed immune responses in healthy adults after two-dose vaccination with either a virus-like particle COVID-19 vaccine (CoVLP), CoVLP adjuvanted with AS03 (CoVLP+AS03), or a messenger RNA vaccination (mRNA-1273). CoVLP+AS03 enhanced the magnitude and durability of circulating antibodies and antigen-specific CD4
+ T cell and memory B cell responses. Antigen-specific CD4+ T cells in the CoVLP+AS03 group at day 42 correlated with antigen-specific memory B cells at 6 months. CoVLP+AS03 induced memory B cell responses, which accumulated somatic hypermutations over 6 months, resulting in enhanced neutralization breadth of monoclonal antibodies. Furthermore, the fraction of broadly neutralizing antibodies encoded by memory B cells increased between day 42 and 6 months. These results indicate that AS03 enhances the antigenic breadth of B cell memory at the clonal level and induces progressive maturation of the B cell response.- Published
- 2024
- Full Text
- View/download PDF
5. Impaired innate and adaptive immune responses to BNT162b2 SARS-CoV-2 vaccination in systemic lupus erythematosus.
- Author
-
Sarin KY, Zheng H, Chaichian Y, Arunachalam PS, Swaminathan G, Eschholz A, Gao F, Wirz OF, Lam B, Yang E, Lee LW, Feng A, Lewis MA, Lin J, Maecker HT, Boyd SD, Davis MM, Nadeau KC, Pulendran B, Khatri P, Utz PJ, and Zaba LC
- Subjects
- Humans, BNT162 Vaccine, COVID-19 Vaccines, SARS-CoV-2, Vaccination, COVID-19 prevention & control, Lupus Erythematosus, Systemic
- Abstract
Understanding the immune responses to SARS-CoV-2 vaccination is critical to optimizing vaccination strategies for individuals with autoimmune diseases, such as systemic lupus erythematosus (SLE). Here, we comprehensively analyzed innate and adaptive immune responses in 19 patients with SLE receiving a complete 2-dose Pfizer-BioNTech mRNA vaccine (BNT162b2) regimen compared with a control cohort of 56 healthy control (HC) volunteers. Patients with SLE exhibited impaired neutralizing antibody production and antigen-specific CD4+ and CD8+ T cell responses relative to HC. Interestingly, antibody responses were only altered in patients with SLE treated with immunosuppressive therapies, whereas impairment of antigen-specific CD4+ and CD8+ T cell numbers was independent of medication. Patients with SLE also displayed reduced levels of circulating CXC motif chemokine ligands, CXCL9, CXCL10, CXCL11, and IFN-γ after secondary vaccination as well as downregulation of gene expression pathways indicative of compromised innate immune responses. Single-cell RNA-Seq analysis reveals that patients with SLE showed reduced levels of a vaccine-inducible monocyte population characterized by overexpression of IFN-response transcription factors. Thus, although 2 doses of BNT162b2 induced relatively robust immune responses in patients with SLE, our data demonstrate impairment of both innate and adaptive immune responses relative to HC, highlighting a need for population-specific vaccination studies.
- Published
- 2024
- Full Text
- View/download PDF
6. BCG vaccination stimulates integrated organ immunity by feedback of the adaptive immune response to imprint prolonged innate antiviral resistance.
- Author
-
Lee A, Floyd K, Wu S, Fang Z, Tan TK, Froggatt HM, Powers JM, Leist SR, Gully KL, Hubbard ML, Li C, Hui H, Scoville D, Ruggiero AD, Liang Y, Pavenko A, Lujan V, Baric RS, Nolan GP, Arunachalam PS, Suthar MS, and Pulendran B
- Subjects
- Animals, Mice, Humans, Feedback, Vaccination, Weight Loss, Antiviral Agents, Immunity, Innate, BCG Vaccine, Adaptive Immunity
- Abstract
Bacille Calmette-Guérin (BCG) vaccination can confer nonspecific protection against heterologous pathogens. However, the underlying mechanisms remain mysterious. We show that mice vaccinated intravenously with BCG exhibited reduced weight loss and/or improved viral clearance when challenged with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 B.1.351) or PR8 influenza. Protection was first evident between 14 and 21 d post-vaccination and lasted ∼3 months. Notably, BCG induced a biphasic innate response and robust antigen-specific type 1 helper T cell (T
H 1 cell) responses in the lungs. MyD88 signaling was essential for innate and TH 1 cell responses, and protection against SARS-CoV-2. Depletion of CD4+ T cells or interferon (IFN)-γ activity before infection obliterated innate activation and protection. Single-cell and spatial transcriptomics revealed CD4-dependent expression of IFN-stimulated genes in lung myeloid and epithelial cells. Notably, BCG also induced protection against weight loss after mouse-adapted SARS-CoV-2 BA.5, SARS-CoV and SHC014 coronavirus infections. Thus, BCG elicits integrated organ immunity, where CD4+ T cells feed back on tissue myeloid and epithelial cells to imprint prolonged and broad innate antiviral resistance., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
7. Multi-omics analysis of mucosal and systemic immunity to SARS-CoV-2 after birth.
- Author
-
Wimmers F, Burrell AR, Feng Y, Zheng H, Arunachalam PS, Hu M, Spranger S, Nyhoff LE, Joshi D, Trisal M, Awasthi M, Bellusci L, Ashraf U, Kowli S, Konvinse KC, Yang E, Blanco M, Pellegrini K, Tharp G, Hagan T, Chinthrajah RS, Nguyen TT, Grifoni A, Sette A, Nadeau KC, Haslam DB, Bosinger SE, Wrammert J, Maecker HT, Utz PJ, Wang TT, Khurana S, Khatri P, Staat MA, and Pulendran B
- Subjects
- Adult, Child, Infant, Humans, Child, Preschool, Multiomics, Cytokines metabolism, Interferon-alpha, Immunity, Mucosal, SARS-CoV-2 metabolism, COVID-19
- Abstract
The dynamics of immunity to infection in infants remain obscure. Here, we used a multi-omics approach to perform a longitudinal analysis of immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in infants and young children by analyzing blood samples and weekly nasal swabs collected before, during, and after infection with Omicron and non-Omicron variants. Infection stimulated robust antibody titers that, unlike in adults, showed no sign of decay for up to 300 days. Infants mounted a robust mucosal immune response characterized by inflammatory cytokines, interferon (IFN) α, and T helper (Th) 17 and neutrophil markers (interleukin [IL]-17, IL-8, and CXCL1). The immune response in blood was characterized by upregulation of activation markers on innate cells, no inflammatory cytokines, but several chemokines and IFNα. The latter correlated with viral load and expression of interferon-stimulated genes (ISGs) in myeloid cells measured by single-cell multi-omics. Together, these data provide a snapshot of immunity to infection during the initial weeks and months of life., Competing Interests: Declaration of interests B.P. serves on the External Immunology Board of GSK and on the Scientific Advisory Board of Sanofi, Medicago, Boehringer Ingelheim, Icosavax, and EdJen. F.W. is a consultant for Gilead. A.S. is a consultant for Gritstone Bio, Flow Pharma, Moderna, AstraZeneca, Qiagen, Fortress, Gilead, Sanofi, Merck, RiverVest, MedaCorp, Turnstone, NA Vaccine Institute, Emervax, Gerson Lehrman Group and Guggenheim. LJI has filed for patent protection for various aspects of T cell epitope and vaccine design work., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
8. Author Correction: A ferritin-based COVID-19 nanoparticle vaccine that elicits robust, durable, broad-spectrum neutralizing antisera in non-human primates.
- Author
-
Weidenbacher PA, Sanyal M, Friedland N, Tang S, Arunachalam PS, Hu M, Kumru OS, Morris MK, Fontenot J, Shirreff L, Do J, Cheng YC, Vasudevan G, Feinberg MB, Villinger FJ, Hanson C, Joshi SB, Volkin DB, Pulendran B, and Kim PS
- Published
- 2023
- Full Text
- View/download PDF
9. Addendum: Systems vaccinology of the BNT162b2 mRNA vaccine in humans.
- Author
-
Arunachalam PS, Scott MKD, Hagan T, Li C, Feng Y, Wimmers F, Grigoryan L, Trisal M, Edara VV, Lai L, Chang SE, Feng A, Dhingra S, Shah M, Lee AS, Chinthrajah S, Sindher SB, Mallajosyula V, Gao F, Sigal N, Kowli S, Gupta S, Pellegrini K, Tharp G, Maysel-Auslender S, Hamilton S, Aoued H, Hrusovsky K, Roskey M, Bosinger SE, Maecker HT, Boyd SD, Davis MM, Utz PJ, Suthar MS, Khatri P, Nadeau KC, and Pulendran B
- Subjects
- Humans, Vaccines, Synthetic, mRNA Vaccines, BNT162 Vaccine, Vaccinology
- Published
- 2023
- Full Text
- View/download PDF
10. Durability of immune responses to mRNA booster vaccination against COVID-19.
- Author
-
Arunachalam PS, Lai L, Samaha H, Feng Y, Hu M, Hui HS, Wali B, Ellis M, Davis-Gardner ME, Huerta C, Bechnak K, Bechnak S, Lee M, Litvack MB, Losada C, Grifoni A, Sette A, Zarnitsyna VI, Rouphael N, Suthar MS, and Pulendran B
- Subjects
- Adult, Humans, SARS-CoV-2, COVID-19 Vaccines, Vaccination, RNA, Messenger, Immunity, Antibodies, Viral, Antibodies, Neutralizing, COVID-19 prevention & control
- Abstract
BackgroundMaintaining durable immunity following vaccination represents a major challenge, but whether mRNA booster vaccination improves durability is unknown.MethodsWe measured antibody responses in 55 healthy adults, who received a booster dose of the Pfizer-BioNTech or Moderna vaccine against SARS-CoV-2 and calculated the half-life of the antibody titers. We also measured memory B and T cell responses in a subset of 28 participants. In 13 volunteers who received a second booster vaccine, we measured serum antibody titers and memory B and T cell responses.ResultsThe booster (third immunization) dose at 6 to 10 months increased the half-life of the serum-neutralizing antibody (nAb) titers to 76 days from 56 to 66 days after the primary 2-dose vaccination. A second booster dose (fourth immunization) a year after the primary vaccination further increased the half-life to 88 days. However, despite this modestly improved durability in nAb responses against the ancestral (WA.1) strain, there was a loss of neutralization capacity against the Omicron subvariants BA.2.75.2, BQ.1.1, and XBB.1.5 (48-, 71-, and 66-fold drop in titers, respectively, relative to the WA.1 strain). Although only 45% to 65% of participants demonstrated a detectable nAb titer against the newer variants after the booster (third dose), the response declined to below the detection limit in almost all individuals by 6 months. In contrast, booster vaccination induced antigen-specific memory B and T cells that persisted for at least 6 months.ConclusionThe durability of serum antibody responses improves only marginally following booster immunizations with the Pfizer-BioNTech or Moderna mRNA vaccines.
- Published
- 2023
- Full Text
- View/download PDF
11. Broadly neutralizing antibodies against sarbecoviruses generated by immunization of macaques with an AS03-adjuvanted COVID-19 vaccine.
- Author
-
Feng Y, Yuan M, Powers JM, Hu M, Munt JE, Arunachalam PS, Leist SR, Bellusci L, Kim J, Sprouse KR, Adams LE, Sundaramurthy S, Zhu X, Shirreff LM, Mallory ML, Scobey TD, Moreno A, O'Hagan DT, Kleanthous H, Villinger FJ, Veesler D, King NP, Suthar MS, Khurana S, Baric RS, Wilson IA, and Pulendran B
- Subjects
- Animals, Humans, Mice, Broadly Neutralizing Antibodies, COVID-19 Vaccines, Macaca, SARS-CoV-2, Immunization, Vaccination, Antibodies, Monoclonal, Antibodies, Viral, Antibodies, Neutralizing, Severe acute respiratory syndrome-related coronavirus, COVID-19 prevention & control
- Abstract
The rapid emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants that evade immunity elicited by vaccination has placed an imperative on the development of countermeasures that provide broad protection against SARS-CoV-2 and related sarbecoviruses. Here, we identified extremely potent monoclonal antibodies (mAbs) that neutralized multiple sarbecoviruses from macaques vaccinated with AS03-adjuvanted monovalent subunit vaccines. Longitudinal analysis revealed progressive accumulation of somatic mutation in the immunoglobulin genes of antigen-specific memory B cells (MBCs) for at least 1 year after primary vaccination. Antibodies generated from these antigen-specific MBCs at 5 to 12 months after vaccination displayed greater potency and breadth relative to those identified at 1.4 months. Fifteen of the 338 (about 4.4%) antibodies isolated at 1.4 to 6 months after the primary vaccination showed potency against SARS-CoV-2 BA.1, despite the absence of serum BA.1 neutralization. 25F9 and 20A7 neutralized authentic clade 1 sarbecoviruses (SARS-CoV, WIV-1, SHC014, SARS-CoV-2 D614G, BA.1, and Pangolin-GD) and vesicular stomatitis virus-pseudotyped clade 3 sarbecoviruses (BtKY72 and PRD-0038). 20A7 and 27A12 showed potent neutralization against all SARS-CoV-2 variants and multiple Omicron sublineages, including BA.1, BA.2, BA.3, BA.4/5, BQ.1, BQ.1.1, and XBB. Crystallography studies revealed the molecular basis of broad and potent neutralization through targeting conserved sites within the RBD. Prophylactic protection of 25F9, 20A7, and 27A12 was confirmed in mice, and administration of 25F9 particularly provided complete protection against SARS-CoV-2, BA.1, SARS-CoV, and SHC014 challenge. These data underscore the extremely potent and broad activity of these mAbs against sarbecoviruses.
- Published
- 2023
- Full Text
- View/download PDF
12. A ferritin-based COVID-19 nanoparticle vaccine that elicits robust, durable, broad-spectrum neutralizing antisera in non-human primates.
- Author
-
Weidenbacher PA, Sanyal M, Friedland N, Tang S, Arunachalam PS, Hu M, Kumru OS, Morris MK, Fontenot J, Shirreff L, Do J, Cheng YC, Vasudevan G, Feinberg MB, Villinger FJ, Hanson C, Joshi SB, Volkin DB, Pulendran B, and Kim PS
- Subjects
- Animals, Humans, COVID-19 Vaccines, Ferritins, SARS-CoV-2, Immune Sera, Primates, Antibodies, Neutralizing, Antibodies, Viral, COVID-19 prevention & control, Geranium, Nanoparticles
- Abstract
While the rapid development of COVID-19 vaccines has been a scientific triumph, the need remains for a globally available vaccine that provides longer-lasting immunity against present and future SARS-CoV-2 variants of concern (VOCs). Here, we describe DCFHP, a ferritin-based, protein-nanoparticle vaccine candidate that, when formulated with aluminum hydroxide as the sole adjuvant (DCFHP-alum), elicits potent and durable neutralizing antisera in non-human primates against known VOCs, including Omicron BQ.1, as well as against SARS-CoV-1. Following a booster ~one year after the initial immunization, DCFHP-alum elicits a robust anamnestic response. To enable global accessibility, we generated a cell line that can enable production of thousands of vaccine doses per liter of cell culture and show that DCFHP-alum maintains potency for at least 14 days at temperatures exceeding standard room temperature. DCFHP-alum has potential as a once-yearly (or less frequent) booster vaccine, and as a primary vaccine for pediatric use including in infants., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
13. Spheromers reveal robust T cell responses to the Pfizer/BioNTech vaccine and attenuated peripheral CD8 + T cell responses post SARS-CoV-2 infection.
- Author
-
Gao F, Mallajosyula V, Arunachalam PS, van der Ploeg K, Manohar M, Röltgen K, Yang F, Wirz O, Hoh R, Haraguchi E, Lee JY, Willis R, Ramachandiran V, Li J, Kathuria KR, Li C, Lee AS, Shah MM, Sindher SB, Gonzalez J, Altman JD, Wang TT, Boyd SD, Pulendran B, Jagannathan P, Nadeau KC, and Davis MM
- Subjects
- Humans, CD8-Positive T-Lymphocytes, BNT162 Vaccine, SARS-CoV-2, Vaccination, Antibodies, Viral, COVID-19, Vaccines
- Abstract
T cells are a critical component of the response to SARS-CoV-2, but their kinetics after infection and vaccination are insufficiently understood. Using "spheromer" peptide-MHC multimer reagents, we analyzed healthy subjects receiving two doses of the Pfizer/BioNTech BNT162b2 vaccine. Vaccination resulted in robust spike-specific T cell responses for the dominant CD4
+ (HLA-DRB1∗ 15:01/S191) and CD8+ (HLA-A∗ 02/S691) T cell epitopes. Antigen-specific CD4+ and CD8+ T cell responses were asynchronous, with the peak CD4+ T cell responses occurring 1 week post the second vaccination (boost), whereas CD8+ T cells peaked 2 weeks later. These peripheral T cell responses were elevated compared with COVID-19 patients. We also found that previous SARS-CoV-2 infection resulted in decreased CD8+ T cell activation and expansion, suggesting that previous infection can influence the T cell response to vaccination., Competing Interests: Declaration of interests V.M. and M.M.D. are inventors on a patent application (PCT/US2021/064378) on the spheromer technology described in this work., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
14. Systems biological assessment of the temporal dynamics of immunity to a viral infection in the first weeks and months of life.
- Author
-
Wimmers F, Burrell AR, Feng Y, Zheng H, Arunachalam PS, Hu M, Spranger S, Nyhoff L, Joshi D, Trisal M, Awasthi M, Bellusci L, Ashraf U, Kowli S, Konvinse KC, Yang E, Blanco M, Pellegrini K, Tharp G, Hagan T, Chinthrajah RS, Grifoni A, Sette A, Nadeau KC, Haslam DB, Bosinger SE, Wrammert J, Maecker HT, Utz PJ, Wang TT, Khurana S, Khatri P, Staat MA, and Pulendran B
- Abstract
The dynamics of innate and adaptive immunity to infection in infants remain obscure. Here, we used a multi-omics approach to perform a longitudinal analysis of immunity to SARS-CoV-2 infection in infants and young children in the first weeks and months of life by analyzing blood samples collected before, during, and after infection with Omicron and Non-Omicron variants. Infection stimulated robust antibody titers that, unlike in adults, were stably maintained for >300 days. Antigen-specific memory B cell (MCB) responses were durable for 150 days but waned thereafter. Somatic hypermutation of V-genes in MCB accumulated progressively over 9 months. The innate response was characterized by upregulation of activation markers on blood innate cells, and a plasma cytokine profile distinct from that seen in adults, with no inflammatory cytokines, but an early and transient accumulation of chemokines (CXCL10, IL8, IL-18R1, CSF-1, CX3CL1), and type I IFN. The latter was strongly correlated with viral load, and expression of interferon-stimulated genes (ISGs) in myeloid cells measured by single-cell transcriptomics. Consistent with this, single-cell ATAC-seq revealed enhanced accessibility of chromatic loci targeted by interferon regulatory factors (IRFs) and reduced accessibility of AP-1 targeted loci, as well as traces of epigenetic imprinting in monocytes, during convalescence. Together, these data provide the first snapshot of immunity to infection during the initial weeks and months of life., Competing Interests: Declaration of Interest Alessandro Sette is a consultant for Gritstone Bio, Flow Pharma, Moderna, AstraZeneca, Qiagen, Fortress, Gilead, Sanofi, Merck, RiverVest, MedaCorp, Turnstone, NA Vaccine Institute, Emervax, Gerson Lehrman Group and Guggenheim. LJI has filed for patent protection for various aspects of T cell epitope and vaccine design work.
- Published
- 2023
- Full Text
- View/download PDF
15. Extremely potent pan-sarbecovirus neutralizing antibodies generated by immunization of macaques with an AS03-adjuvanted monovalent subunit vaccine against SARS-CoV-2.
- Author
-
Feng Y, Yuan M, Powers JM, Hu M, Munt JE, Arunachalam PS, Leist SR, Bellusci L, Adams LE, Sundaramurthy S, Shirreff LM, Mallory ML, Scooby TD, Moreno A, O'Hagan DT, Kleanthous H, Villinger FJ, Veesler D, King NP, Suthar MS, Khurana S, Baric RS, Wilson IA, and Pulendran B
- Abstract
The rapid emergence of SARS-CoV-2 variants that evade immunity to vaccination has placed a global health imperative on the development of therapeutic countermeasures that provide broad protection against SARS-CoV-2 and related sarbecoviruses. Here, we identified extremely potent pan-sarbecovirus antibodies from non-human primates vaccinated with an AS03 adjuvanted subunit vaccine against SARS-CoV-2 that recognize conserved epitopes in the receptor binding domain (RBD) with femtomolar affinities. Longitudinal analysis revealed progressive accumulation of somatic mutation in the immunoglobulin genes of antigen-specific memory B cells for at least one year following primary vaccination. 514 monoclonal antibodies (mAbs) were generated from antigen-specific memory B cells. Antibodies isolated at 5 to 12 months following vaccination displayed greater potency and breadth, relative to those identified at 1.4 months. Notably, 15 out of 338 (∼4.4%) antibodies isolated at 1.4∼6 months after the primary vaccination showed extraordinary neutralization potency against SARS-CoV-2 omicron BA.1, despite the absence of BA.1 neutralization in serum. Two of them, 25F9 and 20A7, neutralized authentic clade Ia sarbecoviruses (SARS-CoV, WIV-1, SHC014) and clade Ib sarbecoviruses (SARS-CoV-2 D614G, SARS-CoV-2 BA.1, Pangolin-GD) with half-maximal inhibition concentrations of (0.85 ng/ml, 3 ng/ml, 6 ng/ml, 6 ng/ml, 42 ng/ml, 6 ng/ml) and (13 ng/ml, 2 ng/ml, 18 ng/ml, 9 ng/ml, 6 ng/ml, 345 ng/ml), respectively. Furthermore, 20A7 and 27A12 showed potent neutralization against all SARS-CoV-2 variants of concern and multiple Omicron sublineages, including BA.1, BA.2, BA.3, BA.4/5, BQ.1, BQ.1.1 and XBB variants. X-ray crystallography studies revealed the molecular basis of broad and potent neutralization through targeting conserved RBD sites. In vivo prophylactic protection of 25F9, 20A7 and 27A12 was confirmed in aged Balb/c mice. Notably, administration of 25F9 provided complete protection against SARS-CoV-2, SARS-CoV-2 BA.1, SARS-CoV, and SHC014 challenge, underscoring that these mAbs are promising pan-sarbecovirus therapeutic antibodies., One Sentence Summary: Extremely potent pan-sarbecovirus neutralizing antibodies.
- Published
- 2023
- Full Text
- View/download PDF
16. A ferritin-based COVID-19 nanoparticle vaccine that elicits robust, durable, broad-spectrum neutralizing antisera in non-human primates.
- Author
-
Weidenbacher PA, Sanyal M, Friedland N, Tang S, Arunachalam PS, Hu M, Kumru OS, Morris MK, Fontenot J, Shirreff L, Do J, Cheng YC, Vasudevan G, Feinberg MB, Villinger FJ, Hanson C, Joshi SB, Volkin DB, Pulendran B, and Kim PS
- Abstract
While the rapid development of COVID-19 vaccines has been a scientific triumph, the need remains for a globally available vaccine that provides longer-lasting immunity against present and future SARS-CoV-2 variants of concern (VOCs). Here, we describe DCFHP, a ferritin-based, protein-nanoparticle vaccine candidate that, when formulated with aluminum hydroxide as the sole adjuvant (DCFHP-alum), elicits potent and durable neutralizing antisera in non-human primates against known VOCs, including Omicron BQ.1, as well as against SARS-CoV-1. Following a booster ∼one year after the initial immunization, DCFHP-alum elicits a robust anamnestic response. To enable global accessibility, we generated a cell line that can enable production of thousands of vaccine doses per liter of cell culture and show that DCFHP-alum maintains potency for at least 14 days at temperatures exceeding standard room temperature. DCFHP-alum has potential as a once-yearly booster vaccine, and as a primary vaccine for pediatric use including in infants.
- Published
- 2022
- Full Text
- View/download PDF
17. Durability of immune responses to the booster mRNA vaccination against COVID-19.
- Author
-
Arunachalam PS, Lai L, Samaha H, Feng Y, Hu M, Hui HS, Wali B, Ellis M, Huerta C, Bechnack K, Bechnack S, Lee M, Litvack M, Losada C, Grifoni A, Sette A, Zarnitsyna VI, Rouphael N, Suthar MS, and Pulendran B
- Abstract
Waning immunity to vaccination represents a major challenge in vaccinology. Whether booster vaccination improves the durability of immune responses is unknown. Here we show, using a cohort of 55 adult vaccinees who received the BNT162b2 (Pfizer-BioNTech) or mRNA-1273 (Moderna) vaccine against SARS-CoV-2, that a booster (i.e., 3
rd immunization) dose at 6 - 10 months increased the half-life of serum neutralizing antibody (nAb) titers to 76 days from 56 - 66 days estimated after the primary two-dose vaccination series. A second booster dose (i.e., 4th immunization) more than a year after the primary vaccination increased the half-life further to 88 days. However, despite this modestly improved durability in nAb responses against the Wuhan strain, there was a loss in neutralization capacity against Omicron subvariants, especially the recently emerged variants, BA.2.75.2 and BQ.1.1 (35 and 50-fold drop in titers respectively, relative to the ancestral (WA.1) strain. While only 55 â€" 65% of participants demonstrated a detectable nAb titer against the newer variants after the booster (3rd dose), the response declined to below the detection limit in almost all individuals by 6 months. Notably, even against BA.1 and BA.5, the titers declined rapidly in a third of the vaccinees and were below the detection limit at 6 months. In contrast, booster vaccination induced antigen-specific memory B and T cells that persisted for at least 6 months. Collectively, our data show that the durability of immune responses improves following subsequent booster immunizations; however, the emergence of immune evasive variants reduces the effectiveness of booster doses in preventing infection.- Published
- 2022
- Full Text
- View/download PDF
18. Early immune markers of clinical, virological, and immunological outcomes in patients with COVID-19: a multi-omics study.
- Author
-
Hu Z, van der Ploeg K, Chakraborty S, Arunachalam PS, Mori DAM, Jacobson KB, Bonilla H, Parsonnet J, Andrews JR, Holubar M, Subramanian A, Khosla C, Maldonado Y, Hedlin H, de la Parte L, Press K, Ty M, Tan GS, Blish C, Takahashi S, Rodriguez-Barraquer I, Greenhouse B, Butte AJ, Singh U, Pulendran B, Wang TT, and Jagannathan P
- Subjects
- Humans, Antibodies, Viral, Biomarkers, BNT162 Vaccine, Cytokines metabolism, Disease Progression, RNA, Messenger, SARS-CoV-2, Clinical Trials as Topic, COVID-19
- Abstract
Background: The great majority of severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) infections are mild and uncomplicated, but some individuals with initially mild COVID-19 progressively develop more severe symptoms. Furthermore, there is substantial heterogeneity in SARS-CoV-2-specific memory immune responses following infection. There remains a critical need to identify host immune biomarkers predictive of clinical and immunological outcomes in SARS-CoV-2-infected patients., Methods: Leveraging longitudinal samples and data from a clinical trial (N=108) in SARS-CoV-2-infected outpatients, we used host proteomics and transcriptomics to characterize the trajectory of the immune response in COVID-19 patients. We characterized the association between early immune markers and subsequent disease progression, control of viral shedding, and SARS-CoV-2-specific T cell and antibody responses measured up to 7 months after enrollment. We further compared associations between early immune markers and subsequent T cell and antibody responses following natural infection with those following mRNA vaccination. We developed machine-learning models to predict patient outcomes and validated the predictive model using data from 54 individuals enrolled in an independent clinical trial., Results: We identify early immune signatures, including plasma RIG-I levels, early IFN signaling, and related cytokines (CXCL10, MCP1, MCP-2, and MCP-3) associated with subsequent disease progression, control of viral shedding, and the SARS-CoV-2-specific T cell and antibody response measured up to 7 months after enrollment. We found that several biomarkers for immunological outcomes are shared between individuals receiving BNT162b2 (Pfizer-BioNTech) vaccine and COVID-19 patients. Finally, we demonstrate that machine-learning models using 2-7 plasma protein markers measured early within the course of infection are able to accurately predict disease progression, T cell memory, and the antibody response post-infection in a second, independent dataset., Conclusions: Early immune signatures following infection can accurately predict clinical and immunological outcomes in outpatients with COVID-19 using validated machine-learning models., Funding: Support for the study was provided from National Institute of Health/National Institute of Allergy and Infectious Diseases (NIH/NIAID) (U01 AI150741-01S1 and T32-AI052073), the Stanford's Innovative Medicines Accelerator, National Institutes of Health/National Institute on Drug Abuse (NIH/NIDA) DP1DA046089, and anonymous donors to Stanford University. Peginterferon lambda provided by Eiger BioPharmaceuticals., Competing Interests: ZH, Kv, SC, PA, DM, KJ, HB, JP, JA, MH, AS, CK, YM, HH, Ld, KP, MT, GT, CB, ST, BG, AB, US, BP, TW, PJ No competing interests declared, IR Reviewing editor, eLife, (© 2022, Hu et al.)
- Published
- 2022
- Full Text
- View/download PDF
19. Distinct sensitivities to SARS-CoV-2 variants in vaccinated humans and mice.
- Author
-
Walls AC, VanBlargan LA, Wu K, Choi A, Navarro MJ, Lee D, Avena L, Berrueta DM, Pham MN, Elbashir S, Kraft JC, Miranda MC, Kepl E, Johnson M, Blackstone A, Sprouse K, Fiala B, O'Connor MA, Brunette N, Arunachalam PS, Shirreff L, Rogers K, Carter L, Fuller DH, Villinger F, Pulendran B, Diamond MS, Edwards DK, King NP, and Veesler D
- Subjects
- Animals, Antibodies, Neutralizing, Antibodies, Viral, Humans, Mice, Mice, Inbred BALB C, Primates, SARS-CoV-2, Spike Glycoprotein, Coronavirus, COVID-19 prevention & control, Viral Vaccines
- Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 has led to the development of a large number of vaccines, several of which are now approved for use in humans. Understanding vaccine-elicited antibody responses against emerging SARS-CoV-2 variants of concern (VOCs) in real time is key to inform public health policies. Serum neutralizing antibody titers are the current best correlate of protection from SARS-CoV-2 challenge in non-human primates and a key metric to understand immune evasion of VOCs. We report that vaccinated BALB/c mice do not recapitulate faithfully the breadth and potency of neutralizing antibody responses elicited by various vaccine platforms against VOCs, compared with non-human primates or humans, suggesting caution should be exercised when interpreting data obtained with this animal model., Competing Interests: Declaration of interests A.C.W., N.P.K., and D.V. are named as inventors on patent applications filed by the University of Washington based on the RBD-NP presented in this paper. N.P.K. is a co-founder, shareholder, paid consultant, and chair of the scientific advisory board of Icosavax, Inc., and the King lab has received an unrelated sponsored research agreement from Pfizer. M.S.D. is a consultant for Inbios, Vir Biotechnology, and Carnival Corporation, and on the Scientific Advisory Boards of Moderna and Immunome. The Diamond laboratory has received unrelated funding support in sponsored research agreements from Vir Biotechnology, Kaleido, and Emergent BioSolutions and past support from Moderna not related to these studies. K.W., A.C., and D.K.E are employees of Moderna and hold stock/stock options in the company. D.H.F. has equity interest in HDT Bio., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
20. Durable protection against the SARS-CoV-2 Omicron variant is induced by an adjuvanted subunit vaccine.
- Author
-
Arunachalam PS, Feng Y, Ashraf U, Hu M, Walls AC, Edara VV, Zarnitsyna VI, Aye PP, Golden N, Miranda MC, Green KWM, Threeton BM, Maness NJ, Beddingfield BJ, Bohm RP, Scheuermann SE, Goff K, Dufour J, Russell-Lodrigue K, Kepl E, Fiala B, Wrenn S, Ravichandran R, Ellis D, Carter L, Rogers K, Shirreff LM, Ferrell DE, Deb Adhikary NR, Fontenot J, Hammond HL, Frieman M, Grifoni A, Sette A, O'Hagan DT, Van Der Most R, Rappuoli R, Villinger F, Kleanthous H, Rappaport J, Suthar MS, Veesler D, Wang TT, King NP, and Pulendran B
- Subjects
- Adjuvants, Immunologic pharmacology, Animals, Antibodies, Neutralizing, Antibodies, Viral, COVID-19 Vaccines, Humans, SARS-CoV-2, Vaccines, Subunit, COVID-19 prevention & control, Viral Vaccines
- Abstract
Despite the remarkable efficacy of COVID-19 vaccines, waning immunity and the emergence of SARS-CoV-2 variants such as Omicron represents a global health challenge. Here, we present data from a study in nonhuman primates demonstrating durable protection against the Omicron BA.1 variant induced by a subunit SARS-CoV-2 vaccine comprising the receptor binding domain of the ancestral strain (RBD-Wu) on the I53-50 nanoparticle adjuvanted with AS03, which was recently authorized for use in individuals 18 years or older. Vaccination induced neutralizing antibody (nAb) titers that were maintained at high concentrations for at least 1 year after two doses, with a pseudovirus nAb geometric mean titer (GMT) of 1978 and a live virus nAb GMT of 1331 against the ancestral strain but not against the Omicron BA.1 variant. However, a booster dose at 6 to 12 months with RBD-Wu or RBD-β (RBD from the Beta variant) displayed on I53-50 elicited high neutralizing titers against the ancestral and Omicron variants. In addition, we observed persistent neutralization titers against a panel of sarbecoviruses, including SARS-CoV. Furthermore, there were substantial and persistent memory T and B cell responses reactive to Beta and Omicron variants. Vaccination resulted in protection against Omicron infection in the lung and suppression of viral burden in the nares at 6 weeks after the final booster immunization. Even at 6 months after vaccination, we observed protection in the lung and rapid control of virus in the nares. These results highlight the durable and cross-protective immunity elicited by the AS03-adjuvanted RBD-I53-50 nanoparticle vaccine.
- Published
- 2022
- Full Text
- View/download PDF
21. Adjuvanting a subunit SARS-CoV-2 vaccine with clinically relevant adjuvants induces durable protection in mice.
- Author
-
Grigoryan L, Lee A, Walls AC, Lai L, Franco B, Arunachalam PS, Feng Y, Luo W, Vanderheiden A, Floyd K, Wrenn S, Pettie D, Miranda MC, Kepl E, Ravichandran R, Sydeman C, Brunette N, Murphy M, Fiala B, Carter L, Coffman RL, Novack D, Kleanthous H, O'Hagan DT, van der Most R, McLellan JS, Suthar M, Veesler D, King NP, and Pulendran B
- Abstract
Adjuvants enhance the magnitude and the durability of the immune response to vaccines. However, there is a paucity of comparative studies on the nature of the immune responses stimulated by leading adjuvant candidates. In this study, we compared five clinically relevant adjuvants in mice-alum, AS03 (a squalene-based adjuvant supplemented with α-tocopherol), AS37 (a TLR7 ligand emulsified in alum), CpG1018 (a TLR9 ligand emulsified in alum), O/W 1849101 (a squalene-based adjuvant)-for their capacity to stimulate immune responses when combined with a subunit vaccine under clinical development. We found that all four of the adjuvant candidates surpassed alum with respect to their capacity to induce enhanced and durable antigen-specific antibody responses. The TLR-agonist-based adjuvants CpG1018 (TLR9) and AS37 (TLR7) induced Th1-skewed CD4+ T cell responses, while alum, O/W, and AS03 induced a balanced Th1/Th2 response. Consistent with this, adjuvants induced distinct patterns of early innate responses. Finally, vaccines adjuvanted with AS03, AS37, and CpG1018/alum-induced durable neutralizing-antibody responses and significant protection against the B.1.351 variant 7 months following immunization. These results, together with our recent results from an identical study in non-human primates (NHPs), provide a comparative benchmarking of five clinically relevant vaccine adjuvants for their capacity to stimulate immunity to a subunit vaccine, demonstrating the capacity of adjuvanted SARS-CoV-2 subunit vaccines to provide durable protection against the B.1.351 variant. Furthermore, these results reveal differences between the widely-used C57BL/6 mouse strain and NHP animal models, highlighting the importance of species selection for future vaccine and adjuvant studies., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
22. Mechanisms of innate and adaptive immunity to the Pfizer-BioNTech BNT162b2 vaccine.
- Author
-
Li C, Lee A, Grigoryan L, Arunachalam PS, Scott MKD, Trisal M, Wimmers F, Sanyal M, Weidenbacher PA, Feng Y, Adamska JZ, Valore E, Wang Y, Verma R, Reis N, Dunham D, O'Hara R, Park H, Luo W, Gitlin AD, Kim P, Khatri P, Nadeau KC, and Pulendran B
- Subjects
- Adaptive Immunity, Animals, BNT162 Vaccine, Humans, Immunity, Innate, Mice, Vaccines, Synthetic, mRNA Vaccines, CD8-Positive T-Lymphocytes, Vaccines
- Abstract
Despite the success of the BNT162b2 mRNA vaccine, the immunological mechanisms that underlie its efficacy are poorly understood. Here we analyzed the innate and adaptive responses to BNT162b2 in mice, and show that immunization stimulated potent antibody and antigen-specific T cell responses, as well as strikingly enhanced innate responses after secondary immunization, which was concurrent with enhanced serum interferon (IFN)-γ levels 1 d following secondary immunization. Notably, we found that natural killer cells and CD8
+ T cells in the draining lymph nodes are the major producers of this circulating IFN-γ. Analysis of knockout mice revealed that induction of antibody and T cell responses to BNT162b2 was not dependent on signaling via Toll-like receptors 2, 3, 4, 5 and 7 nor inflammasome activation, nor the necroptosis or pyroptosis cell death pathways. Rather, the CD8+ T cell response induced by BNT162b2 was dependent on type I interferon-dependent MDA5 signaling. These results provide insights into the molecular mechanisms by which the BNT162b2 vaccine stimulates immune responses., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2022
- Full Text
- View/download PDF
23. Immune imprinting, breadth of variant recognition, and germinal center response in human SARS-CoV-2 infection and vaccination.
- Author
-
Röltgen K, Nielsen SCA, Silva O, Younes SF, Zaslavsky M, Costales C, Yang F, Wirz OF, Solis D, Hoh RA, Wang A, Arunachalam PS, Colburg D, Zhao S, Haraguchi E, Lee AS, Shah MM, Manohar M, Chang I, Gao F, Mallajosyula V, Li C, Liu J, Shoura MJ, Sindher SB, Parsons E, Dashdorj NJ, Dashdorj ND, Monroe R, Serrano GE, Beach TG, Chinthrajah RS, Charville GW, Wilbur JL, Wohlstadter JN, Davis MM, Pulendran B, Troxell ML, Sigal GB, Natkunam Y, Pinsky BA, Nadeau KC, and Boyd SD
- Subjects
- Antigens, Viral, Humans, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus, Vaccination, Antibodies, Viral, BNT162 Vaccine, COVID-19 prevention & control, Germinal Center
- Abstract
During the SARS-CoV-2 pandemic, novel and traditional vaccine strategies have been deployed globally. We investigated whether antibodies stimulated by mRNA vaccination (BNT162b2), including third-dose boosting, differ from those generated by infection or adenoviral (ChAdOx1-S and Gam-COVID-Vac) or inactivated viral (BBIBP-CorV) vaccines. We analyzed human lymph nodes after infection or mRNA vaccination for correlates of serological differences. Antibody breadth against viral variants is lower after infection compared with all vaccines evaluated but improves over several months. Viral variant infection elicits variant-specific antibodies, but prior mRNA vaccination imprints serological responses toward Wuhan-Hu-1 rather than variant antigens. In contrast to disrupted germinal centers (GCs) in lymph nodes during infection, mRNA vaccination stimulates robust GCs containing vaccine mRNA and spike antigen up to 8 weeks postvaccination in some cases. SARS-CoV-2 antibody specificity, breadth, and maturation are affected by imprinting from exposure history and distinct histological and antigenic contexts in infection compared with vaccination., Competing Interests: Declaration of interests S.D.B. has consulted for Regeneron, Sanofi, Novartis, and Janssen on topics unrelated to this study and owns stock in AbCellera Biologics. K.C.N. reports grants from the National Institute of Allergy and Infectious Diseases (NIAID), Food Allergy Research & Education (FARE), End Allergies Together (EAT), National Heart Lung and Blood Institute (NHLBI), and National Institute of Environmental Health Sciences (NIEHS). K.C.N. is Director of FARE and World Allergy Organization (WAO) Center of Excellence at Stanford; Adviser at Cour Pharmaceuticals; Cofounder of Before Brands, Alladapt, Latitude, and IgGenix; National Scientific Committee member for the Immune Tolerance Network (ITN) of NIAID; recipient of a Research Sponsorship from Nestle; Consultant and Advisory Board Member at Before Brands, Alladapt, IgGenix, NHLBI, and ProBio; and Data and Safety Monitoring Board member at NHLBI. J.L.W., J.N.W., and G.B.S. are employees of Meso Scale Diagnostics (MSD)., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. Early non-neutralizing, afucosylated antibody responses are associated with COVID-19 severity.
- Author
-
Chakraborty S, Gonzalez JC, Sievers BL, Mallajosyula V, Chakraborty S, Dubey M, Ashraf U, Cheng BY, Kathale N, Tran KQT, Scallan C, Sinnott A, Cassidy A, Chen ST, Gelbart T, Gao F, Golan Y, Ji X, Kim-Schulze S, Prahl M, Gaw SL, Gnjatic S, Marron TU, Merad M, Arunachalam PS, Boyd SD, Davis MM, Holubar M, Khosla C, Maecker HT, Maldonado Y, Mellins ED, Nadeau KC, Pulendran B, Singh U, Subramanian A, Utz PJ, Sherwood R, Zhang S, Jagannathan P, Tan GS, and Wang TT
- Subjects
- Antibodies, Neutralizing, Antibodies, Viral, Antibody Formation, COVID-19 Vaccines, Humans, Prospective Studies, SARS-CoV-2, Spike Glycoprotein, Coronavirus, COVID-19
- Abstract
A damaging inflammatory response is implicated in the pathogenesis of severe coronavirus disease 2019 (COVID-19), but mechanisms contributing to this response are unclear. In two prospective cohorts, early non-neutralizing, afucosylated immunoglobulin G (IgG) antibodies specific to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were associated with progression from mild to more severe COVID-19. To study the biology of afucosylated IgG immune complexes, we developed an in vivo model that revealed that human IgG-Fc-gamma receptor (FcγR) interactions could regulate inflammation in the lung. Afucosylated IgG immune complexes isolated from patients with COVID-19 induced inflammatory cytokine production and robust infiltration of the lung by immune cells. In contrast to the antibody structures that were associated with disease progression, antibodies that were elicited by messenger RNA SARS-CoV-2 vaccines were highly fucosylated and enriched in sialylation, both modifications that reduce the inflammatory potential of IgG. Vaccine-elicited IgG did not promote an inflammatory lung response. These results show that human IgG-FcγR interactions regulate inflammation in the lung and define distinct lung activities mediated by the IgG that are associated with protection against, or progression to, severe COVID-19.
- Published
- 2022
- Full Text
- View/download PDF
25. Antibodies elicited by SARS-CoV-2 infection or mRNA vaccines have reduced neutralizing activity against Beta and Omicron pseudoviruses.
- Author
-
Sievers BL, Chakraborty S, Xue Y, Gelbart T, Gonzalez JC, Cassidy AG, Golan Y, Prahl M, Gaw SL, Arunachalam PS, Blish CA, Boyd SD, Davis MM, Jagannathan P, Nadeau KC, Pulendran B, Singh U, Scheuermann RH, Frieman MB, Vashee S, Wang TT, and Tan GS
- Subjects
- Adult, COVID-19 Vaccines immunology, Female, Humans, Pregnancy, Pregnancy Complications, Infectious immunology, Pregnancy Complications, Infectious prevention & control, Pregnancy Complications, Infectious virology, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus immunology, Vaccines, Synthetic immunology, mRNA Vaccines immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 immunology, COVID-19 prevention & control, COVID-19 virology
- Abstract
Multiple severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants that have mutations associated with increased transmission and antibody escape have arisen over the course of the current pandemic. Although the current vaccines have largely been effective against past variants, the number of mutations found on the Omicron (B.1.1.529) spike protein appear to diminish the protection conferred by preexisting immunity. Using vesicular stomatitis virus (VSV) pseudoparticles expressing the spike protein of several SARS-CoV-2 variants, we evaluated the magnitude and breadth of the neutralizing antibody response over time in individuals after infection and in mRNA-vaccinated individuals. We observed that boosting increases the magnitude of the antibody response to wild-type (D614), Beta, Delta, and Omicron variants; however, the Omicron variant was the most resistant to neutralization. We further observed that vaccinated healthy adults had robust and broad antibody responses, whereas responses may have been reduced in vaccinated pregnant women, underscoring the importance of learning how to maximize mRNA vaccine responses in pregnant populations. Findings from this study show substantial heterogeneity in the magnitude and breadth of responses after infection and mRNA vaccination and may support the addition of more conserved viral antigens to existing SARS-CoV-2 vaccines.
- Published
- 2022
- Full Text
- View/download PDF
26. Safety, immunogenicity, and protection provided by unadjuvanted and adjuvanted formulations of a recombinant plant-derived virus-like particle vaccine candidate for COVID-19 in nonhuman primates.
- Author
-
Pillet S, Arunachalam PS, Andreani G, Golden N, Fontenot J, Aye PP, Röltgen K, Lehmicke G, Gobeil P, Dubé C, Trépanier S, Charland N, D'Aoust MA, Russell-Lodrigue K, Monjure C, Blair RV, Boyd SD, Bohm RP, Rappaport J, Villinger F, Landry N, Pulendran B, and Ward BJ
- Subjects
- Adjuvants, Immunologic administration & dosage, Animals, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, Antibodies, Viral blood, Antibodies, Viral immunology, COVID-19 epidemiology, COVID-19 virology, COVID-19 Vaccines administration & dosage, Disease Models, Animal, Drug Combinations, Drug Compounding methods, Immunity, Humoral, Macaca mulatta, Male, Polysorbates administration & dosage, Recombinant Proteins immunology, Recombinant Proteins metabolism, Spike Glycoprotein, Coronavirus immunology, Spike Glycoprotein, Coronavirus metabolism, Squalene administration & dosage, Treatment Outcome, Vaccines, Virus-Like Particle administration & dosage, alpha-Tocopherol administration & dosage, Adjuvants, Immunologic adverse effects, COVID-19 immunology, COVID-19 prevention & control, COVID-19 Vaccines adverse effects, Immunogenicity, Vaccine immunology, Pandemics prevention & control, Polysorbates adverse effects, SARS-CoV-2 immunology, Squalene adverse effects, Nicotiana metabolism, Vaccination methods, Vaccines, Virus-Like Particle adverse effects, alpha-Tocopherol adverse effects
- Abstract
Although antivirals are important tools to control severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, effective vaccines are essential to control the current coronavirus disease 2019 (COVID-19) pandemic. Plant-derived virus-like particle (VLP) vaccine candidates have previously demonstrated immunogenicity and efficacy against influenza. Here, we report the immunogenicity and protection induced in rhesus macaques by intramuscular injections of a VLP bearing a SARS-CoV-2 spike protein (CoVLP) vaccine candidate formulated with or without Adjuvant System 03 (AS03) or cytidine-phospho-guanosine (CpG) 1018. Although a single dose of the unadjuvanted CoVLP vaccine candidate stimulated humoral and cell-mediated immune responses, booster immunization (at 28 days after priming) and adjuvant administration significantly improved both responses, with higher immunogenicity and protection provided by the AS03-adjuvanted CoVLP. Fifteen micrograms of CoVLP adjuvanted with AS03 induced a polyfunctional interleukin-2 (IL-2)-driven response and IL-4 expression in CD4 T cells. Animals were challenged by multiple routes (i.e., intratracheal, intranasal, and ocular) with a total viral dose of 10
6 plaque-forming units of SARS-CoV-2. Lower viral replication in nasal swabs and bronchoalveolar lavage fluid (BALF) as well as fewer SARS-CoV-2-infected cells and immune cell infiltrates in the lungs concomitant with reduced levels of proinflammatory cytokines and chemotactic factors in the BALF were observed in animals immunized with the CoVLP adjuvanted with AS03. No clinical, pathologic, or virologic evidence of vaccine-associated enhanced disease was observed in vaccinated animals. The CoVLP adjuvanted with AS03 was therefore selected for vaccine development and clinical trials., (© 2021. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
27. A molecular atlas of innate immunity to adjuvanted and live attenuated vaccines, in mice.
- Author
-
Lee A, Scott MKD, Wimmers F, Arunachalam PS, Luo W, Fox CB, Tomai M, Khatri P, and Pulendran B
- Subjects
- Adaptive Immunity, Adjuvants, Immunologic pharmacology, Alum Compounds, Animals, Antibodies, Viral immunology, Epigenomics, Female, Humans, Immunization, Membrane Glycoproteins agonists, Mice, Mice, Inbred C57BL, Monocytes, Myeloid Cells, Ovalbumin, Toll-Like Receptor 7 agonists, Toll-Like Receptor 8 agonists, Vaccination, Adjuvants, Immunologic chemistry, Immunity, Humoral immunology, Immunity, Innate drug effects, Vaccines, Attenuated immunology
- Abstract
Adjuvants hold great potential in enhancing vaccine efficacy, making the understanding and improving of adjuvants critical goals in vaccinology. The TLR7/8 agonist, 3M-052, induces long-lived humoral immunity in non-human primates and is currently being evaluated in human clinical trials. However, the innate mechanisms of 3M-052 have not been fully characterized. Here, we perform flow cytometry, single cell RNA-seq and ATAC-seq to profile the kinetics, transcriptomics and epigenomics of innate immune cells in murine draining lymph nodes following 3M-052-Alum/Ovalbumin immunization. We find that 3M-052-Alum/OVA induces a robust antiviral and interferon gene program, similar to the yellow fever vaccine, which is known to confer long-lasting protection. Activation of myeloid cells in dLNs persists through day 28 and single cell analysis reveals putative TF-gene regulatory programs in distinct myeloid cells and heterogeneity of monocytes. This study provides a comprehensive characterization of the transcriptomics and epigenomics of innate populations in the dLNs after vaccination., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
28. Durability of immune responses to the BNT162b2 mRNA vaccine.
- Author
-
Suthar MS, Arunachalam PS, Hu M, Reis N, Trisal M, Raeber O, Chinthrajah S, Davis-Gardner ME, Manning K, Mudvari P, Boritz E, Godbole S, Henry AR, Douek DC, Halfmann P, Kawaoka Y, Boyd SD, Davis MM, Zarnitsyna VI, Nadeau K, and Pulendran B
- Subjects
- Antibody Formation, BNT162 Vaccine, Humans, Vaccines, Synthetic, mRNA Vaccines, COVID-19 prevention & control, COVID-19 Vaccines
- Abstract
Antibody responses to the Pfizer-BioNTech mRNA vaccine waned substantially 6 months after the second vaccination., Competing Interests: Declaration of interests B.P. serves on the External Immunology Network of GSK and on the Scientific Advisory Board of Sanofi and Medicago. M.S. serves on the advisory board for Moderna., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
29. Structurally and functionally distinct early antibody responses predict COVID-19 disease trajectory and mRNA vaccine response.
- Author
-
Chakraborty S, Gonzalez JC, Sievers BL, Mallajosyula V, Chakraborty S, Dubey M, Ashraf U, Cheng BY, Kathale N, Tran KQT, Scallan C, Sinnott A, Cassidy A, Chen ST, Gelbart T, Gao F, Golan Y, Ji X, Kim-Schulze S, Prahl M, Gaw SL, Gnjatic S, Marron TU, Merad M, Arunachalam PS, Boyd SD, Davis MM, Holubar M, Khosla C, Maecker HT, Maldonado Y, Mellins ED, Nadeau KC, Pulendran B, Singh U, Subramanian A, Utz PJ, Sherwood R, Zhang S, Jagannathan P, Tan GS, and Wang TT
- Abstract
A damaging inflammatory response is strongly implicated in the pathogenesis of severe COVID-19 but mechanisms contributing to this response are unclear. In two prospective cohorts, early non-neutralizing, afucosylated, anti-SARS-CoV-2 IgG predicted progression from mild, to more severe COVID-19. In contrast to the antibody structures that predicted disease progression, antibodies that were elicited by mRNA SARS-CoV-2 vaccines were low in Fc afucosylation and enriched in sialylation, both modifications that reduce the inflammatory potential of IgG. To study the biology afucosylated IgG immune complexes, we developed an in vivo model which revealed that human IgG-FcγR interactions can regulate inflammation in the lung. Afucosylated IgG immune complexes induced inflammatory cytokine production and robust infiltration of the lung by immune cells. By contrast, vaccine elicited IgG did not promote an inflammatory lung response. Here, we show that IgG-FcγR interactions can regulate inflammation in the lung and define distinct lung activities associated with the IgG that predict severe COVID-19 and protection against SARS-CoV-2., One Sentence Summary: Divergent early antibody responses predict COVID-19 disease trajectory and mRNA vaccine response and are functionally distinct in vivo .
- Published
- 2021
- Full Text
- View/download PDF
30. Direct comparison of antibody responses to four SARS-CoV-2 vaccines in Mongolia.
- Author
-
Dashdorj NJ, Wirz OF, Röltgen K, Haraguchi E, Buzzanco AS 3rd, Sibai M, Wang H, Miller JA, Solis D, Sahoo MK, Arunachalam PS, Lee AS, Shah MM, Liu J, Byambabaatar S, Bat-Ulzii P, Enkhbat A, Batbold E, Zulkhuu D, Ochirsum B, Khurelsukh T, Dalantai G, Burged N, Baatarsuren U, Ariungerel N, Oidovsambuu O, Bungert AS, Genden Z, Yagaanbuyant D, Mordorj A, Pulendran B, Chinthrajah S, Nadeau KC, Jardetzky T, Wilbur JL, Wohlstadter JN, Sigal GB, Pinsky BA, Boyd SD, and Dashdorj ND
- Subjects
- Adult, Aged, Angiotensin-Converting Enzyme 2 antagonists & inhibitors, Angiotensin-Converting Enzyme 2 genetics, Angiotensin-Converting Enzyme 2 immunology, Antibodies, Viral biosynthesis, COVID-19 epidemiology, COVID-19 immunology, COVID-19 virology, Female, Gene Expression, Humans, Immune Sera chemistry, Immunogenicity, Vaccine, Male, Middle Aged, Mongolia epidemiology, Retrospective Studies, SARS-CoV-2 immunology, SARS-CoV-2 pathogenicity, Antibodies, Viral blood, BNT162 Vaccine administration & dosage, COVID-19 prevention & control, COVID-19 Vaccines administration & dosage, ChAdOx1 nCoV-19 administration & dosage, Mass Vaccination, SARS-CoV-2 drug effects
- Abstract
Different SARS-CoV-2 vaccines are approved in various countries, but few direct comparisons of the antibody responses they stimulate have been reported. We collected plasma specimens in July 2021 from 196 Mongolian participants fully vaccinated with one of four COVID-19 vaccines: Pfizer/BioNTech, AstraZeneca, Sputnik V, and Sinopharm. Functional antibody testing with a panel of nine SARS-CoV-2 viral variant receptor binding domain (RBD) proteins revealed marked differences in vaccine responses, with low antibody levels and RBD-ACE2 blocking activity stimulated by the Sinopharm and Sputnik V vaccines in comparison to the AstraZeneca or Pfizer/BioNTech vaccines. The Alpha variant caused 97% of infections in Mongolia in June and early July 2021. Individuals who recover from SARS-CoV-2 infection after vaccination achieve high antibody titers in most cases. These data suggest that public health interventions such as vaccine boosting, potentially with more potent vaccine types, may be needed to control COVID-19 in Mongolia and worldwide., Competing Interests: Declaration of interests S.D.B. has consulted for Regeneron, Sanofi, and Novartis on topics unrelated to this study and owns stock in AbCellera Biologics; B.P. and P.S.A. are inventors on a provisional patent application (no. 63/026,577) submitted by the Board of Trustees of the Leland Stanford Junior University, Stanford, CA, that covers the use of “Therapeutic Methods for Treating COVID-19 Infections”; B.P. serves on the External Immunology Network of GSK and on the Scientific Advisory Board of Medicago and Boehringer Ingelheim; and K.C.N. reports grants from National Institute of Allergy and Infectious Diseases (NIAID), Food Allergy Research & Education (FARE), End Allergies Together (EAT), National Heart, Lung, and Blood Institute (NHLBI), and National Institute of Environmental Health Sciences (NIEHS). K.C.N. is Director of FARE and World Allergy Organization (WAO) Center of Excellence at Stanford, Advisor at Cour Pharmaceuticals, Cofounder of Before Brands, Alladapt, Latitude, and IgGenix, National Scientific Committee member for the Immune Tolerance Network (ITN) of NIAID, recipient of a Research Sponsorship from Nestle, Consultant and Advisory Board Member at Before Brands, Alladapt, IgGenix, NHLBI, and ProBio, and Data and Safety Monitoring Board member at NHLBI; R.S. Chinthrajah receives grant support from CoFAR National Institute of Allergy and Infectious Diseases, Aimmune, DBV Technologies, Astellas, AnaptysBio, Novartis, and Regeneron and is an advisory board member for Alladapt Immunotherapeutics Inc., Novartis, and Genentech. J.L.W., J.N.W., and G.B.S. are employees of Meso Scale Diagnostics (MSD). The rest of the authors declare that they have no conflicts of interest relevant to this manuscript., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
31. Elicitation of broadly protective sarbecovirus immunity by receptor-binding domain nanoparticle vaccines.
- Author
-
Walls AC, Miranda MC, Schäfer A, Pham MN, Greaney A, Arunachalam PS, Navarro MJ, Tortorici MA, Rogers K, O'Connor MA, Shirreff L, Ferrell DE, Bowen J, Brunette N, Kepl E, Zepeda SK, Starr T, Hsieh CL, Fiala B, Wrenn S, Pettie D, Sydeman C, Sprouse KR, Johnson M, Blackstone A, Ravichandran R, Ogohara C, Carter L, Tilles SW, Rappuoli R, Leist SR, Martinez DR, Clark M, Tisch R, O'Hagan DT, Van Der Most R, Van Voorhis WC, Corti D, McLellan JS, Kleanthous H, Sheahan TP, Smith KD, Fuller DH, Villinger F, Bloom J, Pulendran B, Baric RS, King NP, and Veesler D
- Abstract
Understanding vaccine-elicited protection against SARS-CoV-2 variants and other sarbecoviruses is key for guiding public health policies. We show that a clinical stage multivalent SARS-CoV-2 spike receptor-binding domain nanoparticle (RBD-NP) vaccine protects mice from SARS-CoV-2 challenge after a single immunization, indicating a potential dose-sparing strategy. We benchmarked serum neutralizing activity elicited by RBD-NPs in non-human primates against a lead prefusion-stabilized SARS-CoV-2 spike (HexaPro) using a panel of circulating mutants. Polyclonal antibodies elicited by both vaccines are similarly resilient to many RBD residue substitutions tested, although mutations at and surrounding position 484 have negative consequences for neutralization. Mosaic and cocktail nanoparticle immunogens displaying multiple sarbecovirus RBDs elicit broad neutralizing activity in mice and protect mice against SARS-CoV challenge even in the absence of SARS-CoV RBD in the vaccine. This study provides proof of principle that multivalent sarbecovirus RBD-NPs induce heterotypic protection and motivates advancing such broadly protective sarbecovirus vaccines to the clinic., Competing Interests: Declaration of interests A.C.W., N.P.K., and D.V. are named as inventors on patent applications filed by the University of Washington based on the studies presented in this paper. N.P.K. is a co-founder, shareholder, paid consultant, and chair of the scientific advisory board of Icosavax, Inc. and has received an unrelated sponsored research agreement from Pfizer. D.C. is an employee of Vir Biotechnology and may hold shares in Vir Biotechnology. D.V. is a consultant for and has received an unrelated sponsored research agreement from Vir Biotechnology, Inc. R.R., D.T.O., and R.V.D.M. are employees of GlaxoSmithKline. C.-L.H. and J.S.M. are inventors on US patent application no. 63/032,502, “Engineered Coronavirus Spike (S) Protein and Methods of Use Thereof”. R.S.B. has collaborative research agreements with Takeda, Pfizer, Eli Lily, Gilead, Ridgeback Biosciences, and VaxArt, unrelated to the current research. The other authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
32. Immune responses to SARS-CoV-2 in the lung.
- Author
-
Arunachalam PS
- Subjects
- Humans, Immunity, Innate, Lung, COVID-19, SARS-CoV-2
- Abstract
Competing Interests: Competing interests: None declared.
- Published
- 2021
- Full Text
- View/download PDF
33. Systems vaccinology of the BNT162b2 mRNA vaccine in humans.
- Author
-
Arunachalam PS, Scott MKD, Hagan T, Li C, Feng Y, Wimmers F, Grigoryan L, Trisal M, Edara VV, Lai L, Chang SE, Feng A, Dhingra S, Shah M, Lee AS, Chinthrajah S, Sindher SB, Mallajosyula V, Gao F, Sigal N, Kowli S, Gupta S, Pellegrini K, Tharp G, Maysel-Auslender S, Hamilton S, Aoued H, Hrusovsky K, Roskey M, Bosinger SE, Maecker HT, Boyd SD, Davis MM, Utz PJ, Suthar MS, Khatri P, Nadeau KC, and Pulendran B
- Subjects
- Adult, Aged, Antibodies, Neutralizing immunology, Autoantibodies immunology, BNT162 Vaccine, COVID-19 Vaccines administration & dosage, Female, Humans, Immunization, Secondary, Male, Middle Aged, Single-Cell Analysis, Spike Glycoprotein, Coronavirus immunology, Transcription, Genetic, Transcriptome genetics, Young Adult, Adaptive Immunity, Antibodies, Viral immunology, COVID-19 immunology, COVID-19 Vaccines immunology, Immunity, Innate, T-Lymphocytes immunology, Vaccinology
- Abstract
The emergency use authorization of two mRNA vaccines in less than a year from the emergence of SARS-CoV-2 represents a landmark in vaccinology
1,2 . Yet, how mRNA vaccines stimulate the immune system to elicit protective immune responses is unknown. Here we used a systems vaccinology approach to comprehensively profile the innate and adaptive immune responses of 56 healthy volunteers who were vaccinated with the Pfizer-BioNTech mRNA vaccine (BNT162b2). Vaccination resulted in the robust production of neutralizing antibodies against the wild-type SARS-CoV-2 (derived from 2019-nCOV/USA_WA1/2020) and, to a lesser extent, the B.1.351 strain, as well as significant increases in antigen-specific polyfunctional CD4 and CD8 T cells after the second dose. Booster vaccination stimulated a notably enhanced innate immune response as compared to primary vaccination, evidenced by (1) a greater frequency of CD14+ CD16+ inflammatory monocytes; (2) a higher concentration of plasma IFNγ; and (3) a transcriptional signature of innate antiviral immunity. Consistent with these observations, our single-cell transcriptomics analysis demonstrated an approximately 100-fold increase in the frequency of a myeloid cell cluster enriched in interferon-response transcription factors and reduced in AP-1 transcription factors, after secondary immunization. Finally, we identified distinct innate pathways associated with CD8 T cell and neutralizing antibody responses, and show that a monocyte-related signature correlates with the neutralizing antibody response against the B.1.351 variant. Collectively, these data provide insights into the immune responses induced by mRNA vaccination and demonstrate its capacity to prime the innate immune system to mount a more potent response after booster immunization., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
34. Adjuvanting a subunit COVID-19 vaccine to induce protective immunity.
- Author
-
Arunachalam PS, Walls AC, Golden N, Atyeo C, Fischinger S, Li C, Aye P, Navarro MJ, Lai L, Edara VV, Röltgen K, Rogers K, Shirreff L, Ferrell DE, Wrenn S, Pettie D, Kraft JC, Miranda MC, Kepl E, Sydeman C, Brunette N, Murphy M, Fiala B, Carter L, White AG, Trisal M, Hsieh CL, Russell-Lodrigue K, Monjure C, Dufour J, Spencer S, Doyle-Meyers L, Bohm RP, Maness NJ, Roy C, Plante JA, Plante KS, Zhu A, Gorman MJ, Shin S, Shen X, Fontenot J, Gupta S, O'Hagan DT, Van Der Most R, Rappuoli R, Coffman RL, Novack D, McLellan JS, Subramaniam S, Montefiori D, Boyd SD, Flynn JL, Alter G, Villinger F, Kleanthous H, Rappaport J, Suthar MS, King NP, Veesler D, and Pulendran B
- Subjects
- Alum Compounds, Animals, Antibodies, Viral immunology, CD4-Positive T-Lymphocytes cytology, CD4-Positive T-Lymphocytes immunology, COVID-19 virology, Clinical Trials, Phase I as Topic, Clinical Trials, Phase II as Topic, Disease Models, Animal, Immunity, Cellular, Immunity, Humoral, Macaca mulatta immunology, Male, Oligodeoxyribonucleotides, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus immunology, Squalene, Adjuvants, Immunologic, Antibodies, Neutralizing immunology, COVID-19 immunology, COVID-19 prevention & control, COVID-19 Vaccines immunology, SARS-CoV-2 immunology, Vaccines, Subunit immunology
- Abstract
The development of a portfolio of COVID-19 vaccines to vaccinate the global population remains an urgent public health imperative
1 . Here we demonstrate the capacity of a subunit vaccine, comprising the SARS-CoV-2 spike protein receptor-binding domain displayed on an I53-50 protein nanoparticle scaffold (hereafter designated RBD-NP), to stimulate robust and durable neutralizing-antibody responses and protection against SARS-CoV-2 in rhesus macaques. We evaluated five adjuvants including Essai O/W 1849101, a squalene-in-water emulsion; AS03, an α-tocopherol-containing oil-in-water emulsion; AS37, a Toll-like receptor 7 (TLR7) agonist adsorbed to alum; CpG1018-alum, a TLR9 agonist formulated in alum; and alum. RBD-NP immunization with AS03, CpG1018-alum, AS37 or alum induced substantial neutralizing-antibody and CD4 T cell responses, and conferred protection against SARS-CoV-2 infection in the pharynges, nares and bronchoalveolar lavage. The neutralizing-antibody response to live virus was maintained up to 180 days after vaccination with RBD-NP in AS03 (RBD-NP-AS03), and correlated with protection from infection. RBD-NP immunization cross-neutralized the B.1.1.7 SARS-CoV-2 variant efficiently but showed a reduced response against the B.1.351 variant. RBD-NP-AS03 produced a 4.5-fold reduction in neutralization of B.1.351 whereas the group immunized with RBD-NP-AS37 produced a 16-fold reduction in neutralization of B.1.351, suggesting differences in the breadth of the neutralizing-antibody response induced by these adjuvants. Furthermore, RBD-NP-AS03 was as immunogenic as a prefusion-stabilized spike immunogen (HexaPro) with AS03 adjuvant. These data highlight the efficacy of the adjuvanted RBD-NP vaccine in promoting protective immunity against SARS-CoV-2 and have led to phase I/II clinical trials of this vaccine (NCT04742738 and NCT04750343).- Published
- 2021
- Full Text
- View/download PDF
35. Systems biological assessment of human immunity to BNT162b2 mRNA vaccination.
- Author
-
Arunachalam PS, Scott MKD, Hagan T, Li C, Feng Y, Wimmers F, Grigoryan L, Trisal M, Edara VV, Lai L, Chang SE, Feng A, Dhingra S, Shah M, Lee AS, Chinthrajah S, Sindher T, Mallajosyula V, Gao F, Sigal N, Kowli S, Gupta S, Pellegrini K, Tharp G, Maysel-Auslender S, Bosinger S, Maecker HT, Boyd SD, Davis MM, Utz PJ, Suthar MS, Khatri P, Nadeau KC, and Pulendran B
- Abstract
The emergency use authorization of two COVID-19 mRNA vaccines in less than a year since the emergence of SARS-CoV-2, represents a landmark in vaccinology1,2. Yet, how mRNA vaccines stimulate the immune system to elicit protective immune responses is unknown. Here we used a systems biological approach to comprehensively profile the innate and adaptive immune responses in 56 healthy volunteers vaccinated with the Pfizer-BioNTech mRNA vaccine. Vaccination resulted in robust production of neutralizing antibodies (nAbs) against the parent strain and the variant of concern, B.1.351, but no induction of autoantibodies, and significant increases in antigen-specific polyfunctional CD4 and CD8 T cells after the second dose. The innate response induced within the first 2 days of booster vaccination was profoundly increased, relative to the response at corresponding times after priming. Thus, there was a striking increase in the: (i) frequency of CD14+CD16+ inflammatory monocytes; (ii) concentration of IFN- y in the plasma, which correlated with enhanced pSTAT3 and pSTAT1 levels in monocytes and T cells; and (iii) transcriptional signatures of innate responses characteristic of antiviral vaccine responses against pandemic influenza, HIV and Ebola, within 2 days following booster vaccination compared to primary vaccination. Consistent with these observations, single-cell transcriptomics analysis of 242,479 leukocytes demonstrated a ~100-fold increase in the frequency of a myeloid cluster, enriched in a signature of interferon-response transcription factors (TFs) and reduced in AP-1 TFs, one day after secondary immunization, at day 21. Finally, we delineated distinct molecular pathways of innate activation that correlate with CD8 T cell and nAb responses and identified an early monocyte-related signature that was associated with the breadth of the nAb response against the B1.351 variant strain. Collectively, these data provide insights into the immune responses induced by mRNA vaccines and demonstrate their capacity to stimulate an enhanced innate response following booster immunization.
- Published
- 2021
- Full Text
- View/download PDF
36. mRNA vaccination compared to infection elicits an IgG-predominant response with greater SARS-CoV-2 specificity and similar decrease in variant spike recognition.
- Author
-
Röltgen K, Nielsen SCA, Arunachalam PS, Yang F, Hoh RA, Wirz OF, Lee AS, Gao F, Mallajosyula V, Li C, Haraguchi E, Shoura MJ, Wilbur JL, Wohlstadter JN, Davis MM, Pinsky BA, Sigal GB, Pulendran B, Nadeau KC, and Boyd SD
- Abstract
During the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, new vaccine strategies including lipid nanoparticle delivery of antigen encoding RNA have been deployed globally. The BioNTech/Pfizer mRNA vaccine BNT162b2 encoding SARS-CoV-2 spike protein shows 95% efficacy in preventing disease, but it is unclear how the antibody responses to vaccination differ from those generated by infection. Here we compare the magnitude and breadth of antibodies targeting SARS-CoV-2, SARS-CoV-2 variants of concern, and endemic coronaviruses, in vaccinees and infected patients. We find that vaccination differs from infection in the dominance of IgG over IgM and IgA responses, with IgG reaching levels similar to those of severely ill COVID-19 patients and shows decreased breadth of the antibody response targeting endemic coronaviruses. Viral variants of concern from B.1.1.7 to P.1 to B.1.351 form a remarkably consistent hierarchy of progressively decreasing antibody recognition by both vaccinees and infected patients exposed to Wuhan-Hu-1 antigens., Competing Interests: Declaration of Interests S.D.B. has consulted for Regeneron, Sanofi, and Novartis on topics unrelated to this study, and owns stock in AbCellera Biologics. K.C.N. reports grants from National Institute of Allergy and Infectious Diseases (NIAID), Food Allergy Research & Education (FARE), End Allergies Together (EAT); National Heart, Lung, and Blood Institute (NHLBI), and National Institute of Environmental Health Sciences (NIEHS). K.C.N. is Director of FARE and World Allergy Organization (WAO) Center of Excellence at Stanford; Advisor at Cour Pharmaceuticals; Cofounder of Before Brands, Alladapt, Latitude, and IgGenix; National Scientific Committee member for the Immune Tolerance Network (ITN) of NIAID; recipient of a Research Sponsorship from Nestle; Consultant and Advisory Board Member at Before Brands, Alladapt, IgGenix, NHLBI, and ProBio; and Data and Safety Monitoring Board member at NHLBI.
- Published
- 2021
- Full Text
- View/download PDF
37. The C3/465 glycan hole cluster in BG505 HIV-1 envelope is the major neutralizing target involved in preventing mucosal SHIV infection.
- Author
-
Charles TP, Burton SL, Arunachalam PS, Cottrell CA, Sewall LM, Bollimpelli VS, Gangadhara S, Dey AK, Ward AB, Shaw GM, Hunter E, Amara RR, Pulendran B, van Gils MJ, and Derdeyn CA
- Subjects
- Animals, Epitopes immunology, Female, HIV Infections immunology, HIV Infections virology, Humans, Immunization, Macaca mulatta, Simian Acquired Immunodeficiency Syndrome immunology, Simian Acquired Immunodeficiency Syndrome virology, Vaccination, Antibodies, Neutralizing immunology, HIV Antibodies immunology, HIV-1 immunology, Polysaccharides immunology, Simian Acquired Immunodeficiency Syndrome prevention & control, Simian Immunodeficiency Virus immunology, env Gene Products, Human Immunodeficiency Virus immunology
- Abstract
Stabilized HIV-1 envelope (Env) trimers elicit tier 2 autologous neutralizing antibody (nAb) responses in immunized animals. We previously demonstrated that BG505 SOSIP.664.T332N gp140 (BG505 SOSIP) immunization of rhesus macaques (RM) provided robust protection against autologous intra-vaginal simian-human immunodeficiency virus (SHIV) challenge that was predicted by high serum nAb titers. Here, we show that nAb in these protected RM targeted a glycan hole proximal to residue 465 in gp120 in all cases. nAb also targeted another glycan hole at residues 241/289 and an epitope in V1 at varying frequencies. Non-neutralizing antibodies directed at N611-shielded epitopes in gp41 were also present but were more prevalent in RM with low nAb titers. Longitudinal analysis demonstrated that nAb broadened in some RM during sequential immunization but remained focused in others, the latter being associated with increases in nAb titer. Thirty-eight monoclonal antibodies (mAbs) isolated from a protected RM with an exceptionally high serum neutralization titer bound to the trimer in ELISA, and four of the mAbs potently neutralized the BG505 Env pseudovirus (PV) and SHIV. The four neutralizing mAbs were clonally related and targeted the 465 glycan hole to varying degrees, mimicking the serum. The data demonstrate that the C3/465 glycan hole cluster was the dominant neutralization target in high titer protected RM, despite other co-circulating neutralizing and non-neutralizing specificities. The isolation of a neutralizing mAb family argues that clonotype expansion occurred during BG505 SOSIP immunization, leading to high titer, protective nAb and setting a desirable benchmark for HIV vaccines., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
38. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans.
- Author
-
Arunachalam PS, Wimmers F, Mok CKP, Perera RAPM, Scott M, Hagan T, Sigal N, Feng Y, Bristow L, Tak-Yin Tsang O, Wagh D, Coller J, Pellegrini KL, Kazmin D, Alaaeddine G, Leung WS, Chan JMC, Chik TSH, Choi CYC, Huerta C, Paine McCullough M, Lv H, Anderson E, Edupuganti S, Upadhyay AA, Bosinger SE, Maecker HT, Khatri P, Rouphael N, Peiris M, and Pulendran B
- Subjects
- COVID-19, Cytokines blood, DNA, Bacterial blood, Dendritic Cells immunology, Dendritic Cells metabolism, Female, Flow Cytometry, HLA-DR Antigens analysis, Humans, Immunity, Immunity, Innate, Immunoglobulins blood, Immunoglobulins immunology, Inflammation Mediators blood, Interferon Type I metabolism, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear metabolism, Lipopolysaccharides blood, Male, Myeloid Cells immunology, Myeloid Cells metabolism, Pandemics, SARS-CoV-2, Signal Transduction, Single-Cell Analysis, Systems Biology, TOR Serine-Threonine Kinases metabolism, Transcription, Genetic, Transcriptome, Betacoronavirus immunology, Coronavirus Infections immunology, Pneumonia, Viral immunology
- Abstract
Coronavirus disease 2019 (COVID-19) represents a global crisis, yet major knowledge gaps remain about human immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We analyzed immune responses in 76 COVID-19 patients and 69 healthy individuals from Hong Kong and Atlanta, Georgia, United States. In the peripheral blood mononuclear cells (PBMCs) of COVID-19 patients, we observed reduced expression of human leukocyte antigen class DR (HLA-DR) and proinflammatory cytokines by myeloid cells as well as impaired mammalian target of rapamycin (mTOR) signaling and interferon-α (IFN-α) production by plasmacytoid dendritic cells. By contrast, we detected enhanced plasma levels of inflammatory mediators-including EN-RAGE, TNFSF14, and oncostatin M-which correlated with disease severity and increased bacterial products in plasma. Single-cell transcriptomics revealed a lack of type I IFNs, reduced HLA-DR in the myeloid cells of patients with severe COVID-19, and transient expression of IFN-stimulated genes. This was consistent with bulk PBMC transcriptomics and transient, low IFN-α levels in plasma during infection. These results reveal mechanisms and potential therapeutic targets for COVID-19., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
39. T cell-inducing vaccine durably prevents mucosal SHIV infection even with lower neutralizing antibody titers.
- Author
-
Arunachalam PS, Charles TP, Joag V, Bollimpelli VS, Scott MKD, Wimmers F, Burton SL, Labranche CC, Petitdemange C, Gangadhara S, Styles TM, Quarnstrom CF, Walter KA, Ketas TJ, Legere T, Jagadeesh Reddy PB, Kasturi SP, Tsai A, Yeung BZ, Gupta S, Tomai M, Vasilakos J, Shaw GM, Kang CY, Moore JP, Subramaniam S, Khatri P, Montefiori D, Kozlowski PA, Derdeyn CA, Hunter E, Masopust D, Amara RR, and Pulendran B
- Subjects
- Animals, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, CD4-Positive T-Lymphocytes drug effects, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Female, Gene Products, gag immunology, Genetic Vectors, Immunity, Cellular immunology, Immunity, Heterologous, Immunogenicity, Vaccine, Immunologic Memory immunology, Macaca mulatta, Mucous Membrane, Vagina, Antibodies, Neutralizing drug effects, Antibodies, Viral drug effects, CD8-Positive T-Lymphocytes drug effects, Gene Products, gag genetics, Immunity, Cellular drug effects, SAIDS Vaccines pharmacology, Simian Acquired Immunodeficiency Syndrome prevention & control, Simian Immunodeficiency Virus immunology
- Abstract
Recent efforts toward an HIV vaccine focus on inducing broadly neutralizing antibodies, but eliciting both neutralizing antibodies (nAbs) and cellular responses may be superior. Here, we immunized macaques with an HIV envelope trimer, either alone to induce nAbs, or together with a heterologous viral vector regimen to elicit nAbs and cellular immunity, including CD8
+ tissue-resident memory T cells. After ten vaginal challenges with autologous virus, protection was observed in both vaccine groups at 53.3% and 66.7%, respectively. A nAb titer >300 was generally associated with protection but in the heterologous viral vector + nAb group, titers <300 were sufficient. In this group, protection was durable as the animals resisted six more challenges 5 months later. Antigen stimulation of T cells in ex vivo vaginal tissue cultures triggered antiviral responses in myeloid and CD4+ T cells. We propose that cellular immune responses reduce the threshold of nAbs required to confer superior and durable protection.- Published
- 2020
- Full Text
- View/download PDF
40. PTAP motif duplication in the p6 Gag protein confers a replication advantage on HIV-1 subtype C.
- Author
-
Sharma S, Arunachalam PS, Menon M, Ragupathy V, Satya RV, Jebaraj J, Aralaguppe SG, Rao C, Pal S, Saravanan S, Murugavel KG, Balakrishnan P, Solomon S, Hewlett I, and Ranga U
- Subjects
- Adult, Amino Acid Motifs, Cells, Cultured, DNA-Binding Proteins genetics, Endosomal Sorting Complexes Required for Transport genetics, Female, HIV Infections genetics, HIV-1 chemistry, HIV-1 genetics, Host-Pathogen Interactions, Humans, Longitudinal Studies, Male, Middle Aged, Protein Interaction Maps, Transcription Factors genetics, gag Gene Products, Human Immunodeficiency Virus chemistry, gag Gene Products, Human Immunodeficiency Virus genetics, DNA-Binding Proteins metabolism, Endosomal Sorting Complexes Required for Transport metabolism, HIV Infections metabolism, HIV Infections virology, HIV-1 physiology, Transcription Factors metabolism, Virus Replication, gag Gene Products, Human Immunodeficiency Virus metabolism
- Abstract
HIV-1 subtype C (HIV-1C) may duplicate longer amino acid stretches in the p6 Gag protein, leading to the creation of an additional Pro-Thr/Ser-Ala-Pro (PTAP) motif necessary for viral packaging. However, the biological significance of a duplication of the PTAP motif for HIV-1 replication and pathogenesis has not been experimentally validated. In a longitudinal study of two different clinical cohorts of select HIV-1 seropositive, drug-naive individuals from India, we found that 8 of 50 of these individuals harbored a mixed infection of viral strains discordant for the PTAP duplication. Conventional and next-generation sequencing of six primary viral quasispecies at multiple time points disclosed that in a mixed infection, the viral strains containing the PTAP duplication dominated the infection. The dominance of the double-PTAP viral strains over a genetically similar single-PTAP viral clone was confirmed in viral proliferation and pairwise competition assays. Of note, in the proximity ligation assay, double-PTAP Gag proteins exhibited a significantly enhanced interaction with the host protein tumor susceptibility gene 101 (Tsg101). Moreover, Tsg101 overexpression resulted in a biphasic effect on HIV-1C proliferation, an enhanced effect at low concentration and an inhibitory effect only at higher concentrations, unlike a uniformly inhibitory effect on subtype B strains. In summary, our results indicate that the duplication of the PTAP motif in the p6 Gag protein enhances the replication fitness of HIV-1C by engaging the Tsg101 host protein with a higher affinity. Our results have implications for HIV-1 pathogenesis, especially of HIV-1C.
- Published
- 2018
- Full Text
- View/download PDF
41. Toll-Like Receptor 9 Activation Rescues Impaired Antibody Response in Needle-free Intradermal DNA Vaccination.
- Author
-
Arunachalam PS, Mishra R, Badarinath K, Selvam D, Payeli SK, Stout RR, and Ranga U
- Abstract
The delivery of plasmid DNA to the skin can target distinct subsets of dermal dendritic cells to confer a superior immune response. The needle-free immunization technology offers a reliable, safe and efficient means to administer intradermal (ID) injections. We report here that the ID injection of DNA vectors using an NF device (NF-ID) elicits a superior cell-mediated immune response, at much lesser DNA dosage, comparable in magnitude to the traditional intramuscular immunization. However, the humoral response is significantly impaired, possibly at the stage of B cell isotype switching. We found that the NF-ID administration deposits the DNA primarily on the epidermis resulting in a rapid loss of the DNA as well as the synthesized antigen due to the faster regeneration rate of the skin layers. Therefore, despite the immune-rich nature of the skin, the NF-ID immunization of DNA vectors may be limited by the impaired humoral response. Additional booster injections are required to augment the antibody response. As an alternative and a viable solution, we rescued the IgG response by coadministration of a Toll-like receptor 9 agonist, among other adjuvants examined. Our work has important implication for the optimization of the emerging needle-free technology for ID immunization.
- Published
- 2016
- Full Text
- View/download PDF
42. Botulinum toxin injection for failed tracheo-oesophageal voice in laryngectomees: the Sunderland experience.
- Author
-
Ramachandran K, Arunachalam PS, Hurren A, Marsh RL, and Samuel PR
- Subjects
- Aged, Aphonia etiology, Aphonia therapy, Carcinoma, Squamous Cell complications, Carcinoma, Squamous Cell surgery, Esophageal Spasm, Diffuse complications, Female, Humans, Laryngeal Neoplasms complications, Laryngeal Neoplasms surgery, Larynx, Artificial, Male, Middle Aged, Speech, Esophageal methods, Treatment Outcome, Voice Disorders complications, Voice Disorders therapy, Anti-Dyskinesia Agents administration & dosage, Botulinum Toxins administration & dosage, Laryngectomy, Speech, Alaryngeal methods
- Abstract
Spasm of the pharyngo-oesophageal segment is one of the important causes of tracheo-oesophageal voice failure. Traditionally it has been managed by either prolonged speech therapy, surgical pharyngeal myotomy or pharyngeal plexus neurectomy with varying degrees of success. Botulinum neurotoxin has been found to be effective in relieving pharyngo-oesophageal segment spasm. Since 1995, we have used botulinum toxin injection on 10 laryngectomees with either aphonia or hypertonicity due to pharyngo-oesophageal segment spasm. Early results were analysed by the Sunderland Surgical Voice Restoration Rating scale. Seven of the 10 patients, who were previously completely aphonic, developed voice following this therapy and are using their valve choice as their only method of communication. Out of the three patients who were treated for hypertonic voice, two did derive some benefit from the procedure. One patient developed a hypotonic voice, which lasted for a few months.
- Published
- 2003
- Full Text
- View/download PDF
43. Role of computerized tomography (CT) scan of the chest in patients with newly diagnosed head and neck cancers.
- Author
-
Arunachalam PS, Putnam G, Jennings P, Messersmith R, and Robson AK
- Subjects
- Aged, Female, Humans, Male, Prospective Studies, Sensitivity and Specificity, Carcinoma, Squamous Cell diagnostic imaging, Carcinoma, Squamous Cell secondary, Head and Neck Neoplasms pathology, Lung Neoplasms diagnostic imaging, Lung Neoplasms secondary, Radiography, Thoracic, Tomography, X-Ray Computed
- Abstract
Chest metastases and second primaries are not uncommon in patients with head and neck cancer. Early detection of a second site of malignant disease may alter prognosis and management. This study assessed the diagnostic yield of chest radiographs compared with computerized tomography (CT) in a series of patients with head and neck cancer. Forty-four consecutive patients with a head and neck squamous cell carcinoma (SCC) attending the head and neck surgery department of Cumberland Infirmary, Carlisle, between January 2000 and December 2000 were included in this prospective study. Patients with lymphomas and localized cancers of the skin and lip were excluded. Thirty men and 14 women, with a mean age of 67 years, were assessed. All had chest radiographs and chest CT at the same time as the CT scan of the primary site. Only one patient had a true positive finding on chest radiograph. Five patients had an abnormal chest CT. Of these, two had multiple lung metastases, and another patient had biopsy-proved bronchogenic carcinoma and underwent surgical excision. The sensitivity and specificity of CT scan was 100% and 95%, as opposed to 33% and 97% for chest radiograph.
- Published
- 2002
- Full Text
- View/download PDF
44. Colloid cyst of third ventricle: a rare cause of episodic vertigo.
- Author
-
Arunachalam PS and Johnson I
- Subjects
- Adult, Cysts diagnosis, Humans, Magnetic Resonance Imaging, Male, Cysts complications, Third Ventricle, Vertigo etiology
- Published
- 2002
- Full Text
- View/download PDF
45. Nasal septal surgery: evaluation of symptomatic and general health outcomes.
- Author
-
Arunachalam PS, Kitcher E, Gray J, and Wilson JA
- Subjects
- Adolescent, Adult, Aged, Chi-Square Distribution, Female, Follow-Up Studies, Humans, Male, Middle Aged, Nasal Obstruction diagnosis, Nasal Septum physiopathology, Patient Satisfaction, Prospective Studies, Severity of Illness Index, Statistics, Nonparametric, Treatment Outcome, Nasal Obstruction surgery, Nasal Septum surgery, Otorhinolaryngologic Surgical Procedures methods, Quality of Life
- Abstract
Although septoplasty and submucous resections are common procedures, there have been very few studies on the outcome of nasal septal surgery. This prospective study of two hundred patients undergoing septal surgery used the Fairley nasal symptom score, the Nottingham health profile, a general health questionnaire and clinical examination of nasal cavities to assess the outcome. A wide range of baseline severity scores was observed. Almost 40% of patients failed to attend for review. Analysis of the outcomes in the remaining 121 patients revealed significant improvement in (a) nasal obstruction in 74%, (b) facial pain in 72%, and (c) catarrh in 64% of patients. There was a lack of correlation between observed postoperative reduction in the number of nasal septal areas deviated and improvement in nasal obstruction. The Nottingham health profile and general health questionnaire scores remained unchanged in a large majority of patients. Postoperative improvement in nasal obstruction was independent of grade of surgeon or concomitant lateral nasal wall surgery. The principal benefits of septal surgery relate to improvement in nasal symptoms. The generic quality-of-life measures such as the Nottingham health profile and general health questionnaire did not show significant improvement in quality of life. Our results support the use of disease-specific instruments to evaluate the outcome of septal surgery.
- Published
- 2001
- Full Text
- View/download PDF
46. Persistent foreign body sensation and pharyngeal pain due to retention of staples: an interesting sequelae of endoscopic stapling procedure.
- Author
-
Arunachalam PS and Cameron DS
- Subjects
- Aged, Foreign-Body Migration etiology, Humans, Male, Foreign-Body Migration diagnosis, Pain, Postoperative etiology, Sutures adverse effects, Zenker Diverticulum surgery
- Abstract
The surgical treatment of a pharyngeal pouch with endoscopic stapling diverticulotomy is a relatively new concept. Long-term results and complications are yet to be fully studied. We describe a patient who developed persistent pharyngeal pain and foreign body sensation due to retention of a clump of staples at the cricopharyngeal sphincter. This complication has not been reported before. This case highlights the need for repeat endoscopy rather than a barium swallow X-ray if the patients are symptomatic after stapling procedures.
- Published
- 2001
- Full Text
- View/download PDF
47. Lemierre's syndrome: a complication of acute oropharyngitis.
- Author
-
Agarwal R, Arunachalam PS, and Bosman DA
- Subjects
- Accessory Nerve Diseases microbiology, Adult, Bacteremia diagnosis, Female, Humans, Jugular Veins diagnostic imaging, Magnetic Resonance Imaging methods, Syndrome, Ultrasonography, Venous Thrombosis diagnosis, Venous Thrombosis microbiology, Vocal Cord Paralysis microbiology, Bacteremia etiology, Fusobacterium Infections diagnosis, Fusobacterium necrophorum isolation & purification, Tonsillitis complications
- Abstract
Lemierre's syndrome is a recognized but infrequently seen complication of acute oropharyngitis. In this case report the patient presented with acute sore throat that led to a bacteraemia with internal jugular vein thrombosis and subsequent cranial nerve palsies.
- Published
- 2000
- Full Text
- View/download PDF
48. Extensive squamous cell carcinoma involving the parotid gland and carotid artery a case report.
- Author
-
Arunachalam PS and Ramachandra CR
- Abstract
Squamous cell carcinomas of the parotid gland are very rare. Majority of them will be metastatic with primaries in the head and neck. Very rarely, they can be from unknown primaries or primary parotid tumor itself. We present a case of the Squamous cell carcinoma with extensive local spread to the scalp, skin of the face, parapharyngeal space and causing occlusion of the internal carotid artery. Brief review of literature regarding primary and secondary type of these tumors is also included. Key words: Squamous cell carcinoma, parotid tumors.
- Published
- 2000
- Full Text
- View/download PDF
49. Quincke's oedema of the uvula associated with mucous retention cyst-a case report.
- Author
-
Arunachalam PS and Ramachandra CR
- Abstract
We report a rare case of recurrent angioedema of uvula (Quincke's Oedema), causing air way obstruction. A brief review of literature and treatment options are alto included. The histoloigical specimen showed an associated mucous retention eyst, which in conjunction with Quinckc's oedema has not been previously reported.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.