90 results on '"Robbiani DF"'
Search Results
2. AID-Dependent Activation of a MYC Transgene Induces Multiple Myeloma in a Conditional Mouse Model of Post-Germinal Center Malignancies
- Author
-
Chesi, M, Robbiani, D, Sebag, M, Chng, W, Affer, M, Tiedemann, R, Valdez, R, Palmer, S, Haas, S, Stewart, A, Fonseca, R, Kremer, R, Cattoretti, G, Bergsagel, P, Robbiani, DF, Chng, WJ, Palmer, SE, Haas, SS, Stewart, AK, Bergsagel, PL, CATTORETTI, GIORGIO, Chesi, M, Robbiani, D, Sebag, M, Chng, W, Affer, M, Tiedemann, R, Valdez, R, Palmer, S, Haas, S, Stewart, A, Fonseca, R, Kremer, R, Cattoretti, G, Bergsagel, P, Robbiani, DF, Chng, WJ, Palmer, SE, Haas, SS, Stewart, AK, Bergsagel, PL, and CATTORETTI, GIORGIO
- Abstract
By misdirecting the activity of Activation-Induced Deaminase (AID) to a conditional MYC transgene, we have achieved sporadic, AID-dependent MYC activation in germinal center B cells of Vk*MYC mice. Whereas control C57BL/6 mice develop benign monoclonal gammopathy with age, all Vk*MYC mice progress to an indolent multiple myeloma associated with the biological and clinical features highly characteristic of the human disease. Furthermore, antigen-dependent myeloma could be induced by immunization with a T-dependent antigen. Consistent with these findings in mice, more frequent MYC rearrangements, elevated levels of MYC mRNA, and MYC target genes distinguish human patients with multiple myeloma from individuals with monoclonal gammopathy, implicating a causal role for MYC in the progression of monoclonal gammopathy to multiple myeloma.
- Published
- 2008
3. Ultrapotent IgA dimeric antibodies neutralize emerging Omicron variants.
- Author
-
Zuo F, Cao Y, Sun R, Wang Q, Simonelli L, Du L, Bertoglio F, Schubert M, Guerra C, Cavalli A, Hust M, Robbiani DF, Varani L, Abolhassani H, Xie XS, Hammarström L, Marcotte H, and Pan-Hammarström Q
- Abstract
Competing Interests: Y.C. and X.S.X. have applied for a provisional patent for a series of antibodies, including BD55-5514 (SA55) used in this study. They are also founders of Singlomics Biopharmaceuticals. Y.C., X.S.X., L.H., H.M., and Q.P.-H. filed a US patent on the use of secretory IgA antibodies for therapy against SARS-CoV-2 (Secretory IgA Antibodies Against COVID Infection, US2024/0209065A1). All other authors declare no competing interests.
- Published
- 2025
- Full Text
- View/download PDF
4. Autoantibodies neutralizing type I IFNs underlie severe tick-borne encephalitis in ∼10% of patients.
- Author
-
Gervais A, Marchal A, Fortova A, Berankova M, Krbkova L, Pychova M, Salat J, Zhao S, Kerrouche N, Le Voyer T, Stiasny K, Raffl S, Schieber Pachart A, Fafi-Kremer S, Gravier S, Robbiani DF, Abel L, MacDonald MR, Rice CM, Weissmann G, Kamal Eldin T, Robatscher E, Erne EM, Pagani E, Borghesi A, Puel A, Bastard P, Velay A, Martinot M, Hansmann Y, Aberle JH, Ruzek D, Cobat A, Zhang SY, and Casanova JL
- Subjects
- Humans, Female, Male, Middle Aged, Adult, Encephalitis Viruses, Tick-Borne immunology, Aged, Austria epidemiology, Czech Republic, Encephalitis, Tick-Borne immunology, Interferon Type I immunology, Autoantibodies immunology, Antibodies, Neutralizing immunology
- Abstract
Tick-borne encephalitis (TBE) virus (TBEV) is transmitted to humans via tick bites. Infection is benign in >90% of the cases but can cause mild (<5%), moderate (<4%), or severe (<1%) encephalitis. We show here that ∼10% of patients hospitalized for severe TBE in cohorts from Austria, Czech Republic, and France carry auto-Abs neutralizing IFN-α2, -β, and/or -ω at the onset of disease, contrasting with only ∼1% of patients with moderate and mild TBE. These auto-Abs were found in two of eight patients who died and none of 13 with silent infection. The odds ratios (OR) for severe TBE in individuals with these auto-Abs relative to those without them in the general population were 4.9 (95% CI: 1.5-15.9, P < 0.0001) for the neutralization of only 100 pg/ml IFN-α2 and/or -ω, and 20.8 (95% CI: 4.5-97.4, P < 0.0001) for the neutralization of 10 ng/ml IFN-α2 and -ω. Auto-Abs neutralizing type I IFNs accounted for ∼10% of severe TBE cases in these three European cohorts., (© 2024 Gervais et al.)
- Published
- 2024
- Full Text
- View/download PDF
5. Human antibodies in Mexico and Brazil neutralizing tick-borne flaviviruses.
- Author
-
Cervantes Rincón T, Kapoor T, Keeffe JR, Simonelli L, Hoffmann HH, Agudelo M, Jurado A, Peace A, Lee YE, Gazumyan A, Guidetti F, Cantergiani J, Cena B, Bianchini F, Tamagnini E, Moro SG, Svoboda P, Costa F, Reis MG, Ko AI, Fallon BA, Avila-Rios S, Reyes-Téran G, Rice CM, Nussenzweig MC, Bjorkman PJ, Ruzek D, Varani L, MacDonald MR, and Robbiani DF
- Subjects
- Humans, Brazil, Mexico, Antibodies, Viral immunology, Animals, Encephalitis Viruses, Tick-Borne immunology, Flavivirus immunology, Epitopes immunology, Antibodies, Monoclonal immunology, Ticks virology, Ticks immunology, Female, Male, Antibodies, Neutralizing immunology
- Abstract
Flaviviruses such as dengue virus (DENV), Zika virus (ZIKV), and yellow fever virus (YFV) are spread by mosquitoes and cause human disease and mortality in tropical areas. In contrast, Powassan virus (POWV), which causes severe neurologic illness, is a flavivirus transmitted by ticks in temperate regions of the Northern hemisphere. We find serologic neutralizing activity against POWV in individuals living in Mexico and Brazil. Monoclonal antibodies P002 and P003, which were derived from a resident of Mexico (where POWV is not reported), neutralize POWV lineage I by recognizing an epitope on the virus envelope domain III (EDIII) that is shared with a broad range of tick- and mosquito-borne flaviviruses. Our findings raise the possibility that POWV, or a flavivirus closely related to it, infects humans in the tropics., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. Prevalence of Powassan Virus Seropositivity Among People with History of Lyme Disease and Non-Lyme Community Controls in the Northeastern United States.
- Author
-
Kapoor T, Murray L, Kuvaldina M, Jiang CS, Peace AA, Agudelo M, Jurado A, Robbiani DF, Klemens O, Lattwein E, Sabalza M, Fallon BA, and MacDonald MR
- Subjects
- Animals, Humans, United States epidemiology, Prevalence, Retrospective Studies, Prospective Studies, New England epidemiology, Antibodies, Viral, Immunoglobulin G, Encephalitis Viruses, Tick-Borne, Encephalitis, Tick-Borne veterinary, Lyme Disease epidemiology, Lyme Disease veterinary, Ixodes
- Abstract
Introduction: Lyme disease (LD) affects ∼476,000 people each year in the United States. Symptoms are variable and include rash and flu-like symptoms. Reasons for the wide variation in disease outcomes are unknown. Powassan virus (POWV) is a tick-borne flavivirus that causes disease ranging from asymptomatic infection to encephalitis, neurologic damage, and death. POWV and LD geographic case distributions overlap, with Ixodes species ticks as the common vectors. Clinical ramifications of coinfection or sequential infection are unknown. Objectives: This study's primary objective was to determine the prevalence of POWV-reactive antibodies in sera samples collected from previously studied cohorts of individuals with self-reported LD history residing in the Northeastern United States. As a secondary objective, we studied clinical differences between people with self-reported LD history and low versus high POWV antibody levels. Methods: We used an enzyme-linked immunosorbent assay (ELISA) to quantify IgG directed at the POWV envelope (E) protein domain III in 538 samples from individuals with self-reported LD history and 16 community controls. The samples were also tested with an ELISA assay to quantify IgG directed at the POWV NS1 protein. Results: The percentage of individuals with LD history and possible evidence of POWV exposure varied depending on the assay utilized. We found no significant difference in clinical symptoms between those with low or high POWV IgG levels in the in-house assay. Congruence of the EDIII and NS1 assays was low with only 12% of those positive in the in-house EDIII ELISA testing positive in the POWV NS1 ELISA. Conclusions: The results highlight the difficulty in flavivirus diagnostic testing, particularly in the retrospective detection of flavivirus exposure. The findings suggest that a prospective study with symptomatic patients using approved clinical testing is necessary to address the incidence and clinical implications of LD and POWV co-infection or sequential infection.
- Published
- 2024
- Full Text
- View/download PDF
7. Author Correction: SARS-CoV-2 neutralizing antibody structures inform therapeutic strategies.
- Author
-
Barnes CO, Jette CA, Abernathy ME, Dam KA, Esswein SR, Gristick HB, Malyutin AG, Sharaf NG, Huey-Tubman KE, Lee YE, Robbiani DF, Nussenzweig MC, West AP Jr, and Bjorkman PJ
- Published
- 2024
- Full Text
- View/download PDF
8. Conversion of monoclonal IgG to dimeric and secretory IgA restores neutralizing ability and prevents infection of Omicron lineages.
- Author
-
Marcotte H, Cao Y, Zuo F, Simonelli L, Sammartino JC, Pedotti M, Sun R, Cassaniti I, Hagbom M, Piralla A, Yang J, Du L, Percivalle E, Bertoglio F, Schubert M, Abolhassani H, Sherina N, Guerra C, Borte S, Rezaei N, Kumagai-Braesch M, Xue Y, Su C, Yan Q, He P, Grönwall C, Klareskog L, Calzolai L, Cavalli A, Wang Q, Robbiani DF, Hust M, Shi Z, Feng L, Svensson L, Chen L, Bao L, Baldanti F, Xiao J, Qin C, Hammarström L, Yang X, Varani L, Xie XS, and Pan-Hammarström Q
- Subjects
- Animals, Mice, Humans, Immunoglobulin G, Immunoglobulin A, Administration, Intranasal, Mice, Transgenic, Immunoglobulin A, Secretory, Antibodies, Monoclonal
- Abstract
The emergence of Omicron lineages and descendent subvariants continues to present a severe threat to the effectiveness of vaccines and therapeutic antibodies. We have previously suggested that an insufficient mucosal immunoglobulin A (IgA) response induced by the mRNA vaccines is associated with a surge in breakthrough infections. Here, we further show that the intramuscular mRNA and/or inactivated vaccines cannot sufficiently boost the mucosal secretory IgA response in uninfected individuals, particularly against the Omicron variant. We thus engineered and characterized recombinant monomeric, dimeric, and secretory IgA1 antibodies derived from four neutralizing IgG monoclonal antibodies (mAbs 01A05, rmAb23, DXP-604, and XG014) targeting the receptor-binding domain of the spike protein. Compared to their parental IgG antibodies, dimeric and secretory IgA1 antibodies showed a higher neutralizing activity against different variants of concern (VOCs), in part due to an increased avidity. Importantly, the dimeric or secretory IgA1 form of the DXP-604 antibody significantly outperformed its parental IgG antibody, and neutralized the Omicron lineages BA.1, BA.2, and BA.4/5 with a 25- to 75-fold increase in potency. In human angiotensin converting enzyme 2 (ACE2) transgenic mice, a single intranasal dose of the dimeric IgA DXP-604 conferred prophylactic and therapeutic protection against Omicron BA.5. Thus, dimeric or secretory IgA delivered by nasal administration may potentially be exploited for the treatment and prevention of Omicron infection, thereby providing an alternative tool for combating immune evasion by the current circulating subvariants and, potentially, future VOCs., Competing Interests: Competing interests statement:Y.C. and X.S.X. are listed as inventors on a patent on DXP-604 antibody (PCT/CN2021/093305) for Peking University. H.M., Y.C., L.H., X.S.X., and Q.P.-H. have filed a patent on DXP-604 IgA antibodies. All other authors declare that they have no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
9. Neutralisation activity of mucosal IgA against XBB sublineages and BA.2.86.
- Author
-
Zuo F, Cao Y, Sun R, Yisimayi A, Du L, Bertoglio F, Schubert M, Guerra C, Cavalli A, Hust M, Robbiani DF, Abolhassani H, Xie XS, Hammarström L, Marcotte H, and Pan-Hammarström Q
- Subjects
- Humans, Antibodies, Neutralizing, Immunoglobulin A, Antibodies, Viral
- Abstract
Competing Interests: YC and XSX have applied for a provisional patent for a series of antibodies, which includes BD55–5514 (SA55) used as a standard monoclonal IgA antibody in this study; they are also the founders of Singlomics Biopharmaceuticals. YC, XSX, LH, HM, and QP-H filed a US patent on the use of secretory IgA antibodies for therapy against SARS-CoV-2 (Secretory IgA Antibodies Against COVID Infection). All other authors declare no competing interests. This work was supported by the EU Horizon 2020 Research and Innovation Program (ATAC, 101003650; awarded to MH, DFR, LH, HM, and QP-H), the Swedish Research Council (2019-01302 and 2020-06116, awarded to QP-H), the Knut and Alice Wallenberg Foundation (KAW2020.0102, awarded to LH and QP-H), the Petrus och Augusta Hedlunds stiftelse (awarded to HM and FZ), the Stiftelsen Clas Groschinskys Minnesfond (awarded to HM and FZ), the Magnus Bergvalls Stiftelse (awarded to FZ), and the Eva & Oscar Ahréns Stiftelse (awarded to FZ). LH, HM, and QP-H are joint last authors.
- Published
- 2024
- Full Text
- View/download PDF
10. Analytical Ultracentrifugation Detects Quaternary Rearrangements and Antibody-Induced Conformational Selection of the SARS-CoV-2 Spike Trimer.
- Author
-
Guerrini G, Mehn D, Fumagalli F, Gioria S, Pedotti M, Simonelli L, Bianchini F, Robbiani DF, Varani L, and Calzolai L
- Subjects
- Humans, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Epitopes chemistry, Epitopes immunology, Ultracentrifugation, Protein Domains, SARS-CoV-2 chemistry, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus immunology
- Abstract
Analytical ultracentrifugation (AUC) analysis shows that the SARS-CoV-2 trimeric Spike (S) protein adopts different quaternary conformations in solution. The relative abundance of the "open" and "close" conformations is temperature-dependent, and samples with different storage temperature history have different open/close distributions. Neutralizing antibodies (NAbs) targeting the S receptor binding domain (RBD) do not alter the conformer populations; by contrast, a NAb targeting a cryptic conformational epitope skews the Spike trimer toward an open conformation. The results highlight AUC, which is typically applied for molecular mass determination of biomolecules as a powerful tool for detecting functionally relevant quaternary protein conformations.
- Published
- 2023
- Full Text
- View/download PDF
11. A combination of two resistance mechanisms is critical for tick-borne encephalitis virus escape from a broadly neutralizing human antibody.
- Author
-
Svoboda P, Haviernik J, Bednar P, Matkovic M, Cervantes Rincón T, Keeffe J, Palus M, Salat J, Agudelo M, Nussenzweig MC, Cavalli A, Robbiani DF, and Ruzek D
- Subjects
- Humans, Antibodies, Viral, Epitopes, Antibodies, Monoclonal, Encephalitis Viruses, Tick-Borne, Encephalitis, Tick-Borne
- Abstract
Tick-borne encephalitis virus (TBEV) is a flavivirus that causes human neuroinfections and represents a growing health problem. The human monoclonal antibody T025 targets envelope protein domain III (EDIII) of TBEV and related tick-borne flaviviruses, potently neutralizing TBEV in vitro and in preclinical models, representing a promising candidate for clinical development. We demonstrate that TBEV escape in the presence of T025 or T028 (another EDIII-targeting human monoclonal antibody) results in virus variants of reduced pathogenicity, characterized by distinct sets of amino acid changes in EDII and EDIII that are jointly needed to confer resistance. EDIII substitution K311N impairs formation of a salt bridge critical for T025-epitope interaction. EDII substitution E230K is not on the T025 epitope but likely induces quaternary rearrangements of the virus surface because of repulsion of positively charged residues on the adjacent EDI. A combination of T025 and T028 prevents virus escape and improves neutralization., Competing Interests: Declaration of interests The Rockefeller University filed a patent application in connection with antibodies T025 and T028, on which M.A., D.F.R., and M.C.N. are inventors., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Heterologous inactivated virus/mRNA vaccination response to BF.7, BQ.1.1, and XBB.1.
- Author
-
Zuo F, Sun R, Abolhassani H, Du L, Wang Y, Vlachiotis S, Bertoglio F, Schubert M, Rezaei N, Chavoshzadeh Z, Guerra C, Cavalli A, Andréll J, Kumagai-Braesch M, Xue Y, Cao Y, Hust M, Robbiani DF, Xie XS, Hammarström L, Marcotte H, and Pan-Hammarström Q
- Abstract
Competing Interests: X.S.X. and Y.C. are the inventors of the provisional patent applications for BD series neutralizing antibodies and are founders of Singlomics Biopharmaceuticals. All other authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
13. Autoantibodies against chemokines post-SARS-CoV-2 infection correlate with disease course.
- Author
-
Muri J, Cecchinato V, Cavalli A, Shanbhag AA, Matkovic M, Biggiogero M, Maida PA, Moritz J, Toscano C, Ghovehoud E, Furlan R, Barbic F, Voza A, De Nadai G, Cervia C, Zurbuchen Y, Taeschler P, Murray LA, Danelon-Sargenti G, Moro S, Gong T, Piffaretti P, Bianchini F, Crivelli V, Podešvová L, Pedotti M, Jarrossay D, Sgrignani J, Thelen S, Uhr M, Bernasconi E, Rauch A, Manzo A, Ciurea A, Rocchi MBL, Varani L, Moser B, Bottazzi B, Thelen M, Fallon BA, Boyman O, Mantovani A, Garzoni C, Franzetti-Pellanda A, Uguccioni M, and Robbiani DF
- Subjects
- Humans, SARS-CoV-2, Autoantibodies, Post-Acute COVID-19 Syndrome, Chemokines, COVID-19
- Abstract
Infection with severe acute respiratory syndrome coronavirus 2 associates with diverse symptoms, which can persist for months. While antiviral antibodies are protective, those targeting interferons and other immune factors are associated with adverse coronavirus disease 2019 (COVID-19) outcomes. Here we discovered that antibodies against specific chemokines were omnipresent post-COVID-19, were associated with favorable disease outcome and negatively correlated with the development of long COVID at 1 yr post-infection. Chemokine antibodies were also present in HIV-1 infection and autoimmune disorders, but they targeted different chemokines compared with COVID-19. Monoclonal antibodies derived from COVID-19 convalescents that bound to the chemokine N-loop impaired cell migration. Given the role of chemokines in orchestrating immune cell trafficking, naturally arising chemokine antibodies may modulate the inflammatory response and thus bear therapeutic potential., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
14. Human neutralizing antibodies to cold linear epitopes and subdomain 1 of the SARS-CoV-2 spike glycoprotein.
- Author
-
Bianchini F, Crivelli V, Abernathy ME, Guerra C, Palus M, Muri J, Marcotte H, Piralla A, Pedotti M, De Gasparo R, Simonelli L, Matkovic M, Toscano C, Biggiogero M, Calvaruso V, Svoboda P, Cervantes Rincón T, Fava T, Podešvová L, Shanbhag AA, Celoria A, Sgrignani J, Stefanik M, Hönig V, Pranclova V, Michalcikova T, Prochazka J, Guerrini G, Mehn D, Ciabattini A, Abolhassani H, Jarrossay D, Uguccioni M, Medaglini D, Pan-Hammarström Q, Calzolai L, Fernandez D, Baldanti F, Franzetti-Pellanda A, Garzoni C, Sedlacek R, Ruzek D, Varani L, Cavalli A, Barnes CO, and Robbiani DF
- Subjects
- Humans, Animals, Mice, SARS-CoV-2, Epitopes, Spike Glycoprotein, Coronavirus, Antibodies, Viral, Neutralization Tests, Antibodies, Neutralizing, COVID-19
- Abstract
Emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants diminishes the efficacy of vaccines and antiviral monoclonal antibodies. Continued development of immunotherapies and vaccine immunogens resilient to viral evolution is therefore necessary. Using coldspot-guided antibody discovery, a screening approach that focuses on portions of the virus spike glycoprotein that are both functionally relevant and averse to change, we identified human neutralizing antibodies to highly conserved viral epitopes. Antibody fp.006 binds the fusion peptide and cross-reacts against coronaviruses of the four genera, including the nine human coronaviruses, through recognition of a conserved motif that includes the S2' site of proteolytic cleavage. Antibody hr2.016 targets the stem helix and neutralizes SARS-CoV-2 variants. Antibody sd1.040 binds to subdomain 1, synergizes with antibody rbd.042 for neutralization, and, similar to fp.006 and hr2.016, protects mice expressing human angiotensin-converting enzyme 2 against infection when present as a bispecific antibody. Thus, coldspot-guided antibody discovery reveals donor-derived neutralizing antibodies that are cross-reactive with Orthocoronavirinae, including SARS-CoV-2 variants.
- Published
- 2023
- Full Text
- View/download PDF
15. Human neutralizing antibodies to cold linear epitopes and to subdomain 1 of SARS-CoV-2.
- Author
-
Bianchini F, Crivelli V, Abernathy ME, Guerra C, Palus M, Muri J, Marcotte H, Piralla A, Pedotti M, De Gasparo R, Simonelli L, Matkovic M, Toscano C, Biggiogero M, Calvaruso V, Svoboda P, Rincón TC, Fava T, Podešvová L, Shanbhag AA, Celoria A, Sgrignani J, Stefanik M, Hönig V, Pranclova V, Michalcikova T, Prochazka J, Guerrini G, Mehn D, Ciabattini A, Abolhassani H, Jarrossay D, Uguccioni M, Medaglini D, Pan-Hammarström Q, Calzolai L, Fernandez D, Baldanti F, Franzetti-Pellanda A, Garzoni C, Sedlacek R, Ruzek D, Varani L, Cavalli A, Barnes CO, and Robbiani DF
- Abstract
Emergence of SARS-CoV-2 variants diminishes the efficacy of vaccines and antiviral monoclonal antibodies. Continued development of immunotherapies and vaccine immunogens resilient to viral evolution is therefore necessary. Using coldspot-guided antibody discovery, a screening approach that focuses on portions of the virus spike that are both functionally relevant and averse to change, we identified human neutralizing antibodies to highly conserved viral epitopes. Antibody fp.006 binds the fusion peptide and cross-reacts against coronaviruses of the four genera , including the nine human coronaviruses, through recognition of a conserved motif that includes the S2' site of proteolytic cleavage. Antibody hr2.016 targets the stem helix and neutralizes SARS-CoV-2 variants. Antibody sd1.040 binds to subdomain 1, synergizes with antibody rbd.042 for neutralization and, like fp.006 and hr2.016, protects mice when present as bispecific antibody. Thus, coldspot-guided antibody discovery reveals donor-derived neutralizing antibodies that are cross-reactive with Orthocoronavirinae , including SARS-CoV-2 variants., One Sentence Summary: Broadly cross-reactive antibodies that protect from SARS-CoV-2 variants are revealed by virus coldspot-driven discovery.
- Published
- 2022
- Full Text
- View/download PDF
16. Anti-chemokine antibodies after SARS-CoV-2 infection correlate with favorable disease course.
- Author
-
Muri J, Cecchinato V, Cavalli A, Shanbhag AA, Matkovic M, Biggiogero M, Maida PA, Moritz J, Toscano C, Ghovehoud E, Furlan R, Barbic F, Voza A, Nadai G, Cervia C, Zurbuchen Y, Taeschler P, Murray LA, Danelon-Sargenti G, Moro S, Gong T, Piffaretti P, Bianchini F, Crivelli V, Podešvová L, Pedotti M, Jarrossay D, Sgrignani J, Thelen S, Uhr M, Bernasconi E, Rauch A, Manzo A, Ciurea A, Rocchi MBL, Varani L, Moser B, Bottazzi B, Thelen M, Fallon BA, Boyman O, Mantovani A, Garzoni C, Franzetti-Pellanda A, Uguccioni M, and Robbiani DF
- Abstract
Infection by SARS-CoV-2 leads to diverse symptoms, which can persist for months. While antiviral antibodies are protective, those targeting interferons and other immune factors are associated with adverse COVID-19 outcomes. Instead, we discovered that antibodies against specific chemokines are omnipresent after COVID-19, associated with favorable disease, and predictive of lack of long COVID symptoms at one year post infection. Anti-chemokine antibodies are present also in HIV-1 infection and autoimmune disorders, but they target different chemokines than those in COVID-19. Monoclonal antibodies derived from COVID- 19 convalescents that bind to the chemokine N-loop impair cell migration. Given the role of chemokines in orchestrating immune cell trafficking, naturally arising anti-chemokine antibodies associated with favorable COVID-19 may be beneficial by modulating the inflammatory response and thus bear therapeutic potential., One-Sentence Summary: Naturally arising anti-chemokine antibodies associate with favorable COVID-19 and predict lack of long COVID.
- Published
- 2022
- Full Text
- View/download PDF
17. A dark side to NS1 antibodies?
- Author
-
Robbiani DF and Růžek D
- Abstract
The NS1 protein of flaviviruses is taking center stage. Recent work has made it an attractive target for development of vaccines and immunotherapeutics. Cavazzoni and colleagues (2021. J. Exp. Med.https://doi.org/10.1084/jem.20210580) now reveal a dark side to NS1, linking it to the development of self-reactive antibodies., (© 2021 Robbiani and Růžek.)
- Published
- 2021
- Full Text
- View/download PDF
18. Publisher Correction: Bispecific IgG neutralizes SARS-CoV-2 variants and prevents escape in mice.
- Author
-
De Gasparo R, Pedotti M, Simonelli L, Nickl P, Muecksch F, Cassaniti I, Percivalle E, Lorenzi JCC, Mazzola F, Magrì D, Michalcikova T, Haviernik J, Honig V, Mrazkova B, Polakova N, Fortova A, Tureckova J, Iatsiuk V, Di Girolamo S, Palus M, Zudova D, Bednar P, Bukova I, Bianchini F, Mehn D, Nencka R, Strakova P, Pavlis O, Rozman J, Gioria S, Sammartino JC, Giardina F, Gaiarsa S, Pan-Hammarström Q, Barnes CO, Bjorkman PJ, Calzolai L, Piralla A, Baldanti F, Nussenzweig MC, Bieniasz PD, Hatziioannou T, Prochazka J, Sedlacek R, Robbiani DF, Ruzek D, and Varani L
- Published
- 2021
- Full Text
- View/download PDF
19. Mapping mutations to the SARS-CoV-2 RBD that escape binding by different classes of antibodies.
- Author
-
Greaney AJ, Starr TN, Barnes CO, Weisblum Y, Schmidt F, Caskey M, Gaebler C, Cho A, Agudelo M, Finkin S, Wang Z, Poston D, Muecksch F, Hatziioannou T, Bieniasz PD, Robbiani DF, Nussenzweig MC, Bjorkman PJ, and Bloom JD
- Subjects
- Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Binding Sites, COVID-19 immunology, Epitopes, HLA Antigens immunology, Humans, Immune Evasion genetics, Models, Molecular, Mutation, Neutralization Tests, Protein Domains, SARS-CoV-2 chemistry, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus chemistry, Antibodies, Viral immunology, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology
- Abstract
Monoclonal antibodies targeting a variety of epitopes have been isolated from individuals previously infected with SARS-CoV-2, but the relative contributions of these different antibody classes to the polyclonal response remains unclear. Here we use a yeast-display system to map all mutations to the viral spike receptor-binding domain (RBD) that escape binding by representatives of three potently neutralizing classes of anti-RBD antibodies with high-resolution structures. We compare the antibody-escape maps to similar maps for convalescent polyclonal plasmas, including plasmas from individuals from whom some of the antibodies were isolated. While the binding of polyclonal plasma antibodies are affected by mutations across multiple RBD epitopes, the plasma-escape maps most resemble those of a single class of antibodies that target an epitope on the RBD that includes site E484. Therefore, although the human immune system can produce antibodies that target diverse RBD epitopes, in practice the polyclonal response to infection is skewed towards a single class of antibodies targeting an epitope that is already undergoing rapid evolution.
- Published
- 2021
- Full Text
- View/download PDF
20. Broad and potent neutralizing human antibodies to tick-borne flaviviruses protect mice from disease.
- Author
-
Agudelo M, Palus M, Keeffe JR, Bianchini F, Svoboda P, Salát J, Peace A, Gazumyan A, Cipolla M, Kapoor T, Guidetti F, Yao KH, Elsterová J, Teislerová D, Chrdle A, Hönig V, Oliveira T, West AP, Lee YE, Rice CM, MacDonald MR, Bjorkman PJ, Růžek D, Robbiani DF, and Nussenzweig MC
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal genetics, Antibodies, Neutralizing administration & dosage, Antibodies, Neutralizing genetics, Antibodies, Viral administration & dosage, Antibodies, Viral genetics, Cells, Cultured, Cohort Studies, Cross Reactions immunology, Encephalitis Viruses, Tick-Borne drug effects, Encephalitis Viruses, Tick-Borne physiology, Encephalitis, Tick-Borne prevention & control, Encephalitis, Tick-Borne virology, Epitopes immunology, Female, Humans, Immunoglobulin G administration & dosage, Mice, Inbred BALB C, Sequence Homology, Amino Acid, Survival Analysis, Viral Envelope Proteins genetics, Viral Envelope Proteins immunology, Mice, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Encephalitis Viruses, Tick-Borne immunology, Encephalitis, Tick-Borne immunology, Immunoglobulin G immunology
- Abstract
Tick-borne encephalitis virus (TBEV) is an emerging human pathogen that causes potentially fatal disease with no specific treatment. Mouse monoclonal antibodies are protective against TBEV, but little is known about the human antibody response to infection. Here, we report on the human neutralizing antibody response to TBEV in a cohort of infected and vaccinated individuals. Expanded clones of memory B cells expressed closely related anti-envelope domain III (EDIII) antibodies in both groups of volunteers. However, the most potent neutralizing antibodies, with IC50s below 1 ng/ml, were found only in individuals who recovered from natural infection. These antibodies also neutralized other tick-borne flaviviruses, including Langat, louping ill, Omsk hemorrhagic fever, Kyasanur forest disease, and Powassan viruses. Structural analysis revealed a conserved epitope near the lateral ridge of EDIII adjoining the EDI-EDIII hinge region. Prophylactic or early therapeutic antibody administration was effective at low doses in mice that were lethally infected with TBEV., Competing Interests: Disclosures: M. Agudelo, D.F. Robbiani, and M.C. Nussenzweig reported a patent to Broadly Neutralizing Antibodies to Tick-Borne Encephalitis and Related Viruses (US 63/118,461) pending. M.C. Nussenzweig reported personal fees from Celldex outside the submitted work. Additionally, M.C. Nussenzweig is a Frontier Bioscience SAB member. No other disclosures were reported., (© 2021 Agudelo et al.)
- Published
- 2021
- Full Text
- View/download PDF
21. Bispecific IgG neutralizes SARS-CoV-2 variants and prevents escape in mice.
- Author
-
De Gasparo R, Pedotti M, Simonelli L, Nickl P, Muecksch F, Cassaniti I, Percivalle E, Lorenzi JCC, Mazzola F, Magrì D, Michalcikova T, Haviernik J, Honig V, Mrazkova B, Polakova N, Fortova A, Tureckova J, Iatsiuk V, Di Girolamo S, Palus M, Zudova D, Bednar P, Bukova I, Bianchini F, Mehn D, Nencka R, Strakova P, Pavlis O, Rozman J, Gioria S, Sammartino JC, Giardina F, Gaiarsa S, Pan-Hammarström Q, Barnes CO, Bjorkman PJ, Calzolai L, Piralla A, Baldanti F, Nussenzweig MC, Bieniasz PD, Hatziioannou T, Prochazka J, Sedlacek R, Robbiani DF, Ruzek D, and Varani L
- Subjects
- Angiotensin-Converting Enzyme 2 antagonists & inhibitors, Angiotensin-Converting Enzyme 2 genetics, Angiotensin-Converting Enzyme 2 metabolism, Animals, Antibodies, Bispecific therapeutic use, Antibodies, Monoclonal immunology, Antibodies, Neutralizing therapeutic use, Body Weight, COVID-19 prevention & control, Dependovirus genetics, Disease Models, Animal, Epitopes, B-Lymphocyte chemistry, Epitopes, B-Lymphocyte immunology, Female, Humans, Immune Evasion genetics, Mice, Mice, Inbred C57BL, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus antagonists & inhibitors, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus immunology, COVID-19 Drug Treatment, Antibodies, Bispecific immunology, Antibodies, Neutralizing immunology, COVID-19 immunology, COVID-19 virology, Immunoglobulin G immunology, SARS-CoV-2 immunology
- Abstract
Neutralizing antibodies that target the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein are among the most promising approaches against COVID-19
1,2 . A bispecific IgG1-like molecule (CoV-X2) has been developed on the basis of C121 and C135, two antibodies derived from donors who had recovered from COVID-193 . Here we show that CoV-X2 simultaneously binds two independent sites on the RBD and, unlike its parental antibodies, prevents detectable spike binding to the cellular receptor of the virus, angiotensin-converting enzyme 2 (ACE2). Furthermore, CoV-X2 neutralizes wild-type SARS-CoV-2 and its variants of concern, as well as escape mutants generated by the parental monoclonal antibodies. We also found that in a mouse model of SARS-CoV-2 infection with lung inflammation, CoV-X2 protects mice from disease and suppresses viral escape. Thus, the simultaneous targeting of non-overlapping RBD epitopes by IgG-like bispecific antibodies is feasible and effective, and combines the advantages of antibody cocktails with those of single-molecule approaches.- Published
- 2021
- Full Text
- View/download PDF
22. miR-15a/16-1 deletion in activated B cells promotes plasma cell and mature B-cell neoplasms.
- Author
-
Sewastianik T, Straubhaar JR, Zhao JJ, Samur MK, Adler K, Tanton HE, Shanmugam V, Nadeem O, Dennis PS, Pillai V, Wang J, Jiang M, Lin J, Huang Y, Brooks D, Bouxsein M, Dorfman DM, Pinkus GS, Robbiani DF, Ghobrial IM, Budnik B, Jarolim P, Munshi NC, Anderson KC, and Carrasco RD
- Subjects
- Animals, B-Lymphocytes metabolism, B-Lymphocytes pathology, Chromosome Deletion, Chromosome Disorders genetics, Chromosome Disorders pathology, Chromosomes, Human, Pair 13 genetics, Gene Deletion, Gene Expression Regulation, Neoplastic, Humans, Lymphoma, Large B-Cell, Diffuse pathology, Mice, Inbred C57BL, Multigene Family, Multiple Myeloma genetics, Multiple Myeloma pathology, Neoplasms, Plasma Cell pathology, Plasma Cells metabolism, Plasma Cells pathology, Plasmacytoma genetics, Plasmacytoma pathology, Mice, Lymphoma, Large B-Cell, Diffuse genetics, MicroRNAs genetics, Neoplasms, Plasma Cell genetics
- Abstract
Chromosome 13q deletion [del(13q)], harboring the miR-15a/16-1 cluster, is one of the most common genetic alterations in mature B-cell malignancies, which originate from germinal center (GC) and post-GC B cells. Moreover, miR-15a/16 expression is frequently reduced in lymphoma and multiple myeloma (MM) cells without del(13q), suggesting important tumor-suppressor activity. However, the role of miR-15a/16-1 in B-cell activation and initiation of mature B-cell neoplasms remains to be determined. We show that conditional deletion of the miR-15a/16-1 cluster in murine GC B cells induces moderate but widespread molecular and functional changes including an increased number of GC B cells, percentage of dark zone B cells, and maturation into plasma cells. With time, this leads to development of mature B-cell neoplasms resembling human extramedullary plasmacytoma (EP) as well as follicular and diffuse large B-cell lymphomas. The indolent nature and lack of bone marrow involvement of EP in our murine model resembles human primary EP rather than MM that has progressed to extramedullary disease. We corroborate human primary EP having low levels of miR-15a/16 expression, with del(13q) being the most common genetic loss. Additionally, we show that, although the mutational profile of human EP is similar to MM, there are some exceptions such as the low frequency of hyperdiploidy in EP, which could account for different disease presentation. Taken together, our studies highlight the significant role of the miR-15a/16-1 cluster in the regulation of the GC reaction and its fundamental context-dependent tumor-suppression function in plasma cell and B-cell malignancies., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
23. Mutational escape from the polyclonal antibody response to SARS-CoV-2 infection is largely shaped by a single class of antibodies.
- Author
-
Greaney AJ, Starr TN, Barnes CO, Weisblum Y, Schmidt F, Caskey M, Gaebler C, Cho A, Agudelo M, Finkin S, Wang Z, Poston D, Muecksch F, Hatziioannou T, Bieniasz PD, Robbiani DF, Nussenzweig MC, Bjorkman PJ, and Bloom JD
- Abstract
Monoclonal antibodies targeting a variety of epitopes have been isolated from individuals previously infected with SARS-CoV-2, but the relative contributions of these different antibody classes to the polyclonal response remains unclear. Here we use a yeast-display system to map all mutations to the viral spike receptor-binding domain (RBD) that escape binding by representatives of three potently neutralizing classes of anti-RBD antibodies with high-resolution structures. We compare the antibody-escape maps to similar maps for convalescent polyclonal plasma, including plasma from individuals from whom some of the antibodies were isolated. The plasma-escape maps most closely resemble those of a single class of antibodies that target an epitope on the RBD that includes site E484. Therefore, although the human immune system can produce antibodies that target diverse RBD epitopes, in practice the polyclonal response to infection is dominated by a single class of antibodies targeting an epitope that is already undergoing rapid evolution.
- Published
- 2021
- Full Text
- View/download PDF
24. Bispecific antibody neutralizes circulating SARS-CoV-2 variants, prevents escape and protects mice from disease.
- Author
-
De Gasparo R, Pedotti M, Simonelli L, Nickl P, Muecksch F, Cassaniti I, Percivalle E, Lorenzi JCC, Mazzola F, Magrì D, Michalcikova T, Haviernik J, Honig V, Mrazkova B, Polakova N, Fortova A, Tureckova J, Iatsiuk V, Girolamo SD, Palus M, Zudova D, Bednar P, Bukova I, Bianchini F, Mehn D, Nencka R, Strakova P, Pavlis O, Rozman J, Gioria S, Camilla Sammartino J, Giardina F, Gaiarsa S, Hammarström QP, Barnes CO, Bjorkman PJ, Calzolai L, Piralla A, Baldanti F, Nussenzweig MC, Bieniasz PD, Hatziioannou T, Prochazka J, Sedlacek R, Robbiani DF, Ruzek D, and Varani L
- Abstract
Neutralizing antibodies targeting the receptor binding domain (RBD) of the SARS-CoV-2 Spike (S) are among the most promising approaches against coronavirus disease 2019 (COVID-19)
1,2 . We developed a bispecific, IgG1-like molecule (CoV-X2) based on two antibodies derived from COVID-19 convalescent donors, C121 and C1353 . CoV-X2 simultaneously binds two independent sites on the RBD and, unlike its parental antibodies, prevents detectable S binding to Angiotensin-Converting Enzyme 2 (ACE2), the virus cellular receptor. Furthermore, CoV-X2 neutralizes SARS-CoV-2 and its variants of concern, as well as the escape mutants generated by the parental monoclonals. In a novel animal model of SARS-CoV-2 infection with lung inflammation, CoV-X2 protects mice from disease and suppresses viral escape. Thus, simultaneous targeting of non-overlapping RBD epitopes by IgG-like bispecific antibodies is feasible and effective, combining into a single molecule the advantages of antibody cocktails.- Published
- 2021
- Full Text
- View/download PDF
25. Evolution of antibody immunity to SARS-CoV-2.
- Author
-
Gaebler C, Wang Z, Lorenzi JCC, Muecksch F, Finkin S, Tokuyama M, Cho A, Jankovic M, Schaefer-Babajew D, Oliveira TY, Cipolla M, Viant C, Barnes CO, Bram Y, Breton G, Hägglöf T, Mendoza P, Hurley A, Turroja M, Gordon K, Millard KG, Ramos V, Schmidt F, Weisblum Y, Jha D, Tankelevich M, Martinez-Delgado G, Yee J, Patel R, Dizon J, Unson-O'Brien C, Shimeliovich I, Robbiani DF, Zhao Z, Gazumyan A, Schwartz RE, Hatziioannou T, Bjorkman PJ, Mehandru S, Bieniasz PD, Caskey M, and Nussenzweig MC
- Subjects
- Adolescent, Adult, Aged, Antibodies, Monoclonal blood, Antibodies, Monoclonal immunology, Antibodies, Neutralizing blood, Antibodies, Neutralizing genetics, Antibodies, Neutralizing immunology, Antibodies, Viral blood, Antibodies, Viral genetics, Antigens, Viral chemistry, Antigens, Viral genetics, Antigens, Viral immunology, B-Lymphocytes cytology, B-Lymphocytes immunology, Biopsy, COVID-19 blood, Cohort Studies, Fluorescent Antibody Technique, Humans, Immunity, Humoral genetics, Immunoglobulin A immunology, Immunoglobulin G immunology, Immunoglobulin M immunology, Immunologic Memory immunology, Intestines immunology, Middle Aged, Mutation, Somatic Hypermutation, Immunoglobulin, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Time Factors, Young Adult, Antibodies, Viral immunology, COVID-19 immunology, Immunity, Humoral immunology, SARS-CoV-2 immunology
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected 78 million individuals and is responsible for over 1.7 million deaths to date. Infection is associated with the development of variable levels of antibodies with neutralizing activity, which can protect against infection in animal models
1,2 . Antibody levels decrease with time, but, to our knowledge, the nature and quality of the memory B cells that would be required to produce antibodies upon reinfection has not been examined. Here we report on the humoral memory response in a cohort of 87 individuals assessed at 1.3 and 6.2 months after infection with SARS-CoV-2. We find that titres of IgM and IgG antibodies against the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 decrease significantly over this time period, with IgA being less affected. Concurrently, neutralizing activity in plasma decreases by fivefold in pseudotype virus assays. By contrast, the number of RBD-specific memory B cells remains unchanged at 6.2 months after infection. Memory B cells display clonal turnover after 6.2 months, and the antibodies that they express have greater somatic hypermutation, resistance to RBD mutations and increased potency, indicative of continued evolution of the humoral response. Immunofluorescence and PCR analyses of intestinal biopsies obtained from asymptomatic individuals at 4 months after the onset of coronavirus disease 2019 (COVID-19) revealed the persistence of SARS-CoV-2 nucleic acids and immunoreactivity in the small bowel of 7 out of 14 individuals. We conclude that the memory B cell response to SARS-CoV-2 evolves between 1.3 and 6.2 months after infection in a manner that is consistent with antigen persistence.- Published
- 2021
- Full Text
- View/download PDF
26. Antibody potency, effector function, and combinations in protection and therapy for SARS-CoV-2 infection in vivo.
- Author
-
Schäfer A, Muecksch F, Lorenzi JCC, Leist SR, Cipolla M, Bournazos S, Schmidt F, Maison RM, Gazumyan A, Martinez DR, Baric RS, Robbiani DF, Hatziioannou T, Ravetch JV, Bieniasz PD, Bowen RA, Nussenzweig MC, and Sheahan TP
- Subjects
- Animals, COVID-19, Cell Line, Female, Humans, Mesocricetus, Mice, Mice, Inbred BALB C, SARS-CoV-2, Antibodies, Monoclonal, Murine-Derived immunology, Antibodies, Monoclonal, Murine-Derived pharmacology, Antibodies, Neutralizing immunology, Antibodies, Neutralizing pharmacology, Antibodies, Viral immunology, Antibodies, Viral pharmacology, Betacoronavirus immunology, Coronavirus Infections immunology, Coronavirus Infections therapy, Pandemics, Pneumonia, Viral immunology, Pneumonia, Viral therapy
- Abstract
SARS-CoV-2, the causative agent of COVID-19, has been responsible for over 42 million infections and 1 million deaths since its emergence in December 2019. There are few therapeutic options and no approved vaccines. Here, we examine the properties of highly potent human monoclonal antibodies (hu-mAbs) in a Syrian hamster model of SARS-CoV-2 and in a mouse-adapted model of SARS-CoV-2 infection (SARS-CoV-2 MA). Antibody combinations were effective for prevention and in therapy when administered early. However, in vitro antibody neutralization potency did not uniformly correlate with in vivo protection, and some hu-mAbs were more protective in combination in vivo. Analysis of antibody Fc regions revealed that binding to activating Fc receptors contributes to optimal protection against SARS-CoV-2 MA. The data indicate that intact effector function can affect hu-mAb protective activity and that in vivo testing is required to establish optimal hu-mAb combinations for COVID-19 prevention., Competing Interests: Disclosures: R.S. Baric worked with Eli Lilly to develop antibodies for the treatment of COVID-19. D.F. Robbiani reported a patent to coronavirus antibodies pending. M.C. Nussenzweig reported a patent to anti-SARS-2 antibodies pending, and reported that Rockefeller University has applied for a patent on anti-SARS-2 antibodies. These antibodies are being produced for human clinical trials but have not been licensed to any commercial entity. No other disclosures were reported., (© 2020 Schäfer et al.)
- Published
- 2021
- Full Text
- View/download PDF
27. Enhanced SARS-CoV-2 neutralization by dimeric IgA.
- Author
-
Wang Z, Lorenzi JCC, Muecksch F, Finkin S, Viant C, Gaebler C, Cipolla M, Hoffmann HH, Oliveira TY, Oren DA, Ramos V, Nogueira L, Michailidis E, Robbiani DF, Gazumyan A, Rice CM, Hatziioannou T, Bieniasz PD, Caskey M, and Nussenzweig MC
- Subjects
- Animals, Biomarkers blood, COVID-19 blood, COVID-19 immunology, COVID-19 virology, Cell Line, Tumor, Chlorocebus aethiops, Convalescence, HEK293 Cells, Host-Pathogen Interactions, Humans, Protein Multimerization, Vero Cells, Antibodies, Neutralizing blood, Antibodies, Viral blood, COVID-19 diagnosis, Immunoglobulin A blood, SARS-CoV-2 immunology
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19), primarily infects cells at mucosal surfaces. Serum neutralizing antibody responses are variable and generally low in individuals that suffer mild forms of COVID-19. Although potent immunoglobulin G (IgG) antibodies can neutralize the virus, less is known about secretory antibodies such as IgA that might affect the initial viral spread and transmissibility from the mucosa. Here, we characterize the IgA response to SARS-CoV-2 in a cohort of 149 convalescent individuals after diagnosis with COVID-19. IgA responses in plasma generally correlated with IgG responses. Furthermore, clones of IgM-, IgG-, and IgA-producing B cells were derived from common progenitor cells. Plasma IgA monomers specific to SARS-CoV-2 proteins were demonstrated to be twofold less potent than IgG equivalents. However, IgA dimers, the primary form of antibody in the nasopharynx, were, on average, 15 times more potent than IgA monomers against the same target. Thus, dimeric IgA responses may be particularly valuable for protection against SARS-CoV-2 and for vaccine efficacy., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution License 4.0 (CC BY).)
- Published
- 2021
- Full Text
- View/download PDF
28. The hSSB1 orthologue Obfc2b is essential for skeletogenesis but dispensable for the DNA damage response in vivo.
- Author
-
Feldhahn N, Ferretti E, Robbiani DF, Callen E, Deroubaix S, Selleri L, Nussenzweig A, and Nussenzweig MC
- Published
- 2021
- Full Text
- View/download PDF
29. SARS-CoV-2 neutralizing antibody structures inform therapeutic strategies.
- Author
-
Barnes CO, Jette CA, Abernathy ME, Dam KA, Esswein SR, Gristick HB, Malyutin AG, Sharaf NG, Huey-Tubman KE, Lee YE, Robbiani DF, Nussenzweig MC, West AP Jr, and Bjorkman PJ
- Subjects
- Angiotensin-Converting Enzyme 2 chemistry, Angiotensin-Converting Enzyme 2 metabolism, Angiotensin-Converting Enzyme 2 ultrastructure, Antibodies, Neutralizing chemistry, Antibodies, Neutralizing immunology, Binding Sites genetics, Binding Sites immunology, Cell Line, Cryoelectron Microscopy, Humans, Models, Molecular, Mutation, Receptors, Coronavirus chemistry, Receptors, Coronavirus metabolism, Receptors, Coronavirus ultrastructure, SARS-CoV-2 chemistry, SARS-CoV-2 metabolism, SARS-CoV-2 ultrastructure, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Spike Glycoprotein, Coronavirus ultrastructure, Antibodies, Neutralizing therapeutic use, Antibodies, Neutralizing ultrastructure, COVID-19 immunology, SARS-CoV-2 immunology, COVID-19 Drug Treatment
- Abstract
The coronavirus disease 2019 (COVID-19) pandemic presents an urgent health crisis. Human neutralizing antibodies that target the host ACE2 receptor-binding domain (RBD) of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) spike protein
1-5 show promise therapeutically and are being evaluated clinically6-8 . Here, to identify the structural correlates of SARS-CoV-2 neutralization, we solved eight new structures of distinct COVID-19 human neutralizing antibodies5 in complex with the SARS-CoV-2 spike trimer or RBD. Structural comparisons allowed us to classify the antibodies into categories: (1) neutralizing antibodies encoded by the VH3-53 gene segment with short CDRH3 loops that block ACE2 and bind only to 'up' RBDs; (2) ACE2-blocking neutralizing antibodies that bind both up and 'down' RBDs and can contact adjacent RBDs; (3) neutralizing antibodies that bind outside the ACE2 site and recognize both up and down RBDs; and (4) previously described antibodies that do not block ACE2 and bind only to up RBDs9 . Class 2 contained four neutralizing antibodies with epitopes that bridged RBDs, including a VH3-53 antibody that used a long CDRH3 with a hydrophobic tip to bridge between adjacent down RBDs, thereby locking the spike into a closed conformation. Epitope and paratope mapping revealed few interactions with host-derived N-glycans and minor contributions of antibody somatic hypermutations to epitope contacts. Affinity measurements and mapping of naturally occurring and in vitro-selected spike mutants in 3D provided insight into the potential for SARS-CoV-2 to escape from antibodies elicited during infection or delivered therapeutically. These classifications and structural analyses provide rules for assigning current and future human RBD-targeting antibodies into classes, evaluating avidity effects and suggesting combinations for clinical use, and provide insight into immune responses against SARS-CoV-2.- Published
- 2020
- Full Text
- View/download PDF
30. Measuring SARS-CoV-2 neutralizing antibody activity using pseudotyped and chimeric viruses.
- Author
-
Schmidt F, Weisblum Y, Muecksch F, Hoffmann HH, Michailidis E, Lorenzi JCC, Mendoza P, Rutkowska M, Bednarski E, Gaebler C, Agudelo M, Cho A, Wang Z, Gazumyan A, Cipolla M, Caskey M, Robbiani DF, Nussenzweig MC, Rice CM, Hatziioannou T, and Bieniasz PD
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Neutralizing blood, Antibodies, Viral blood, Betacoronavirus genetics, COVID-19, Cell Line, Chimera genetics, Chimera immunology, Chlorocebus aethiops, Coronavirus Infections virology, HEK293 Cells, HIV-1 genetics, HIV-1 immunology, Humans, Neutralization Tests methods, Pandemics, Pneumonia, Viral virology, Recombination, Genetic, SARS-CoV-2, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Vero Cells, Vesicular stomatitis Indiana virus genetics, Vesicular stomatitis Indiana virus immunology, Antibodies, Neutralizing analysis, Antibodies, Viral analysis, Betacoronavirus immunology, Coronavirus Infections immunology, Immunoassay methods, Pneumonia, Viral immunology
- Abstract
The emergence of SARS-CoV-2 and the ensuing explosive epidemic of COVID-19 disease has generated a need for assays to rapidly and conveniently measure the antiviral activity of SARS-CoV-2-specific antibodies. Here, we describe a collection of approaches based on SARS-CoV-2 spike-pseudotyped, single-cycle, replication-defective human immunodeficiency virus type-1 (HIV-1), and vesicular stomatitis virus (VSV), as well as a replication-competent VSV/SARS-CoV-2 chimeric virus. While each surrogate virus exhibited subtle differences in the sensitivity with which neutralizing activity was detected, the neutralizing activity of both convalescent plasma and human monoclonal antibodies measured using each virus correlated quantitatively with neutralizing activity measured using an authentic SARS-CoV-2 neutralization assay. The assays described herein are adaptable to high throughput and are useful tools in the evaluation of serologic immunity conferred by vaccination or prior SARS-CoV-2 infection, as well as the potency of convalescent plasma or human monoclonal antibodies., Competing Interests: Disclosures: F. Schmidt reported a patent to VSV/SARS-CoV-2 chimeric virus pending. Y. Weisblum reported a patent to patent on VSV/SARS-CoV-2 chimeric virus pending. D.F. Robbiani reported a patent to monoclonal antibodies against SARS-CoV-2 pending. M.C. Nussenzweig reported a patent to anti-SARS-2 antibodies pending, "Rockefeller University," and is an inventor on the anti-SARS-2 antibody patent that has been submitted by the Rockefeller University. P.D. Bieniasz reported a patent to VSV/SARS-CoV-2 patent pending. No other disclosures were reported., (© 2020 Schmidt et al.)
- Published
- 2020
- Full Text
- View/download PDF
31. Escape from neutralizing antibodies by SARS-CoV-2 spike protein variants.
- Author
-
Weisblum Y, Schmidt F, Zhang F, DaSilva J, Poston D, Lorenzi JC, Muecksch F, Rutkowska M, Hoffmann HH, Michailidis E, Gaebler C, Agudelo M, Cho A, Wang Z, Gazumyan A, Cipolla M, Luchsinger L, Hillyer CD, Caskey M, Robbiani DF, Rice CM, Nussenzweig MC, Hatziioannou T, and Bieniasz PD
- Subjects
- Angiotensin-Converting Enzyme 2 metabolism, Antibodies, Monoclonal immunology, Base Sequence, COVID-19 virology, Epitopes genetics, Epitopes immunology, Genes, Reporter, Humans, Immunization, Passive, Neutralization Tests, Protein Domains, Protein Isoforms immunology, Reassortant Viruses immunology, Receptors, Virus metabolism, SARS-CoV-2 genetics, SARS-CoV-2 physiology, Selection, Genetic, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, Vesiculovirus genetics, Virus Replication, COVID-19 Serotherapy, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 immunology, COVID-19 therapy, COVID-19 Vaccines immunology, Mutation, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus immunology
- Abstract
Neutralizing antibodies elicited by prior infection or vaccination are likely to be key for future protection of individuals and populations against SARS-CoV-2. Moreover, passively administered antibodies are among the most promising therapeutic and prophylactic anti-SARS-CoV-2 agents. However, the degree to which SARS-CoV-2 will adapt to evade neutralizing antibodies is unclear. Using a recombinant chimeric VSV/SARS-CoV-2 reporter virus, we show that functional SARS-CoV-2 S protein variants with mutations in the receptor-binding domain (RBD) and N-terminal domain that confer resistance to monoclonal antibodies or convalescent plasma can be readily selected. Notably, SARS-CoV-2 S variants that resist commonly elicited neutralizing antibodies are now present at low frequencies in circulating SARS-CoV-2 populations. Finally, the emergence of antibody-resistant SARS-CoV-2 variants that might limit the therapeutic usefulness of monoclonal antibodies can be mitigated by the use of antibody combinations that target distinct neutralizing epitopes., Competing Interests: YW Rockefeller University has applied for a patent relating to the replication compentent VSV/SARS-CoV-2 chimeric virus on which YW is listed as an inventor (US patent 63/036,124), FS Rockefeller University has applied for a patent relating to the replication compentent VSV/SARS-CoV-2 chimeric virus on which FS is listed as an inventor (US patent 63/036,124), FZ, JD, DP, JL, FM, MR, HH, EM, CG, MA, AC, ZW, AG, MC, LL, CH, MC, CR No competing interests declared, DR Rockefeller University has applied for a patent relating to SARS-CoV-2 monoclonal antibodies on which DFR is listed as an inventor, MN Rockefeller University has applied for a patent relating to SARS-CoV-2 monoclonal antibodies on which MCN is listed as an inventor, TH Rockefeller University has applied for a patent relating to the replication compentent VSV/SARS-CoV-2 chimeric virus on which TH is listed as an inventor (US patent 63/036,124), PB Rockefeller University has applied for a patent relating to the replication compentent VSV/SARS-CoV-2 chimeric virus on which PDB is listed as an inventor (US patent 63/036,124), (© 2020, Weisblum et al.)
- Published
- 2020
- Full Text
- View/download PDF
32. ReScan, a Multiplex Diagnostic Pipeline, Pans Human Sera for SARS-CoV-2 Antigens.
- Author
-
Zamecnik CR, Rajan JV, Yamauchi KA, Mann SA, Loudermilk RP, Sowa GM, Zorn KC, Alvarenga BD, Gaebler C, Caskey M, Stone M, Norris PJ, Gu W, Chiu CY, Ng D, Byrnes JR, Zhou XX, Wells JA, Robbiani DF, Nussenzweig MC, DeRisi JL, and Wilson MR
- Subjects
- Antibodies, Viral blood, COVID-19 blood, Female, Humans, Male, Middle Aged, Peptide Library, Protein Array Analysis, Proteome immunology, Reproducibility of Results, SARS-CoV-2 immunology, Sensitivity and Specificity, Viral Proteins immunology, Antigens, Viral immunology, COVID-19 diagnosis, COVID-19 Serological Testing methods, SARS-CoV-2 isolation & purification
- Abstract
Comprehensive understanding of the serological response to SARS-CoV-2 infection is important for both pathophysiologic insight and diagnostic development. Here, we generate a pan-human coronavirus programmable phage display assay to perform proteome-wide profiling of coronavirus antigens enriched by 98 COVID-19 patient sera. Next, we use ReScan, a method to efficiently sequester phage expressing the most immunogenic peptides and print them onto paper-based microarrays using acoustic liquid handling, which isolates and identifies nine candidate antigens, eight of which are derived from the two proteins used for SARS-CoV-2 serologic assays: spike and nucleocapsid proteins. After deployment in a high-throughput assay amenable to clinical lab settings, these antigens show improved specificity over a whole protein panel. This proof-of-concept study demonstrates that ReScan will have broad applicability for other emerging infectious diseases or autoimmune diseases that lack a valid biomarker, enabling a seamless pipeline from antigen discovery to diagnostic using one recombinant protein source., Competing Interests: The authors declare no competing interests., (© 2020 The Author(s).)
- Published
- 2020
- Full Text
- View/download PDF
33. Neutralizing hepatitis B.
- Author
-
Robbiani DF
- Subjects
- Broadly Neutralizing Antibodies, Humans, Hepatitis B prevention & control, Hepatitis B virus immunology
- Abstract
Despite an effective vaccine, hepatitis B virus (HBV) remains a major public health threat since chronic infection leads to liver disease and cancer. Hehle et al. (https://doi.org/10.1084/jem.20200840) discovered human-derived antibodies that potently neutralize the virus. Will this help a cure?, (© 2020 Robbiani.)
- Published
- 2020
- Full Text
- View/download PDF
34. Antibody potency, effector function and combinations in protection from SARS-CoV-2 infection in vivo .
- Author
-
Schäfer A, Muecksch F, Lorenzi JCC, Leist SR, Cipolla M, Bournazos S, Schmidt F, Gazumyan A, Baric RS, Robbiani DF, Hatziioannou T, Ravetch JV, Bieniasz PD, Nussenzweig MC, and Sheahan TP
- Abstract
SARS-CoV-2, the causative agent of COVID-19, is responsible for over 24 million infections and 800,000 deaths since its emergence in December 2019. There are few therapeutic options and no approved vaccines. Here we examine the properties of highly potent human monoclonal antibodies (hu-mAbs) in a mouse adapted model of SARS-CoV-2 infection (SARS-CoV-2 MA). In vitro antibody neutralization potency did not uniformly correlate with in vivo activity, and some hu-mAbs were more potent in combination in vivo . Analysis of antibody Fc regions revealed that binding to activating Fc receptors is essential for optimal protection against SARS-CoV-2 MA. The data indicate that hu-mAb protective activity is dependent on intact effector function and that in vivo testing is required to establish optimal hu-mAb combinations for COVID-19 prevention.
- Published
- 2020
- Full Text
- View/download PDF
35. Enhanced SARS-CoV-2 Neutralization by Secretory IgA in vitro.
- Author
-
Wang Z, Lorenzi JCC, Muecksch F, Finkin S, Viant C, Gaebler C, Cipolla M, Hoffman HH, Oliveira TY, Oren DA, Ramos V, Nogueira L, Michailidis E, Robbiani DF, Gazumyan A, Rice CM, Hatziioannou T, Bieniasz PD, Caskey M, and Nussenzweig MC
- Abstract
SARS-CoV-2 primarily infects cells at mucosal surfaces. Serum neutralizing antibody responses are variable and generally low in individuals that suffer mild forms of the illness. Although potent IgG antibodies can neutralize the virus, less is known about secretory antibodies such as IgA that might impact the initial viral spread and transmissibility from the mucosa. Here we characterize the IgA response to SARS-CoV-2 in a cohort of 149 individuals. IgA responses in plasma generally correlate with IgG responses and clones of IgM, IgG and IgA producing B cells that are derived from common progenitors are evident. Plasma IgA monomers are 2-fold less potent than IgG equivalents. However, IgA dimers, the primary form in the nasopharynx, are on average 15 times more potent than IgA monomers. Thus, secretory IgA responses may be particularly valuable for protection against SARS-CoV-2 and for vaccine efficacy.
- Published
- 2020
- Full Text
- View/download PDF
36. Structural classification of neutralizing antibodies against the SARS-CoV-2 spike receptor-binding domain suggests vaccine and therapeutic strategies.
- Author
-
Barnes CO, Jette CA, Abernathy ME, Dam KA, Esswein SR, Gristick HB, Malyutin AG, Sharaf NG, Huey-Tubman KE, Lee YE, Robbiani DF, Nussenzweig MC, West AP Jr, and Bjorkman PJ
- Abstract
The COVID-19 pandemic presents an urgent health crisis. Human neutralizing antibodies (hNAbs) that target the host ACE2 receptor-binding domain (RBD) of the SARS-CoV-2 spike
1-5 show therapeutic promise and are being evaluated clincally6-8 . To determine structural correlates of SARS-CoV-2 neutralization, we solved 8 new structures of distinct COVID-19 hNAbs5 in complex with SARS-CoV-2 spike trimer or RBD. Structural comparisons allowed classification into categories: (1) VH3-53 hNAbs with short CDRH3s that block ACE2 and bind only to "up" RBDs, (2) ACE2-blocking hNAbs that bind both "up" and "down" RBDs and can contact adjacent RBDs, (3) hNAbs that bind outside the ACE2 site and recognize "up" and "down" RBDs, and (4) Previously-described antibodies that do not block ACE2 and bind only "up" RBDs9 . Class 2 comprised four hNAbs whose epitopes bridged RBDs, including a VH3-53 hNAb that used a long CDRH3 with a hydrophobic tip to bridge between adjacent "down" RBDs, thereby locking spike into a closed conformation. Epitope/paratope mapping revealed few interactions with host-derived N -glycans and minor contributions of antibody somatic hypermutations to epitope contacts. Affinity measurements and mapping of naturally-occurring and in vitro-selected spike mutants in 3D provided insight into the potential for SARS-CoV-2 escape from antibodies elicited during infection or delivered therapeutically. These classifications and structural analyses provide rules for assigning current and future human RBD-targeting antibodies into classes, evaluating avidity effects, suggesting combinations for clinical use, and providing insight into immune responses against SARS-CoV-2.- Published
- 2020
- Full Text
- View/download PDF
37. Structures of Human Antibodies Bound to SARS-CoV-2 Spike Reveal Common Epitopes and Recurrent Features of Antibodies.
- Author
-
Barnes CO, West AP Jr, Huey-Tubman KE, Hoffmann MAG, Sharaf NG, Hoffman PR, Koranda N, Gristick HB, Gaebler C, Muecksch F, Lorenzi JCC, Finkin S, Hägglöf T, Hurley A, Millard KG, Weisblum Y, Schmidt F, Hatziioannou T, Bieniasz PD, Caskey M, Robbiani DF, Nussenzweig MC, and Bjorkman PJ
- Subjects
- Antibodies, Neutralizing blood, Antibodies, Neutralizing isolation & purification, Antibodies, Viral immunology, Antibodies, Viral isolation & purification, Betacoronavirus immunology, COVID-19, Coronavirus Infections blood, Coronavirus Infections therapy, Cross Reactions, Cryoelectron Microscopy, Epitope Mapping, Epitopes, Humans, Immunization, Passive, Immunoglobulin Fab Fragments blood, Immunoglobulin Fab Fragments isolation & purification, Immunoglobulin Fab Fragments ultrastructure, Immunoglobulin G blood, Immunoglobulin G isolation & purification, Immunoglobulin G ultrastructure, Middle East Respiratory Syndrome Coronavirus chemistry, Middle East Respiratory Syndrome Coronavirus immunology, Models, Molecular, Pandemics, Pneumonia, Viral blood, Severe acute respiratory syndrome-related coronavirus chemistry, Severe acute respiratory syndrome-related coronavirus immunology, SARS-CoV-2, Spike Glycoprotein, Coronavirus immunology, COVID-19 Serotherapy, Antibodies, Neutralizing chemistry, Betacoronavirus chemistry, Coronavirus Infections immunology, Immunoglobulin Fab Fragments chemistry, Immunoglobulin G chemistry, Pneumonia, Viral immunology, Spike Glycoprotein, Coronavirus chemistry
- Abstract
Neutralizing antibody responses to coronaviruses mainly target the receptor-binding domain (RBD) of the trimeric spike. Here, we characterized polyclonal immunoglobulin Gs (IgGs) and Fabs from COVID-19 convalescent individuals for recognition of coronavirus spikes. Plasma IgGs differed in their focus on RBD epitopes, recognition of alpha- and beta-coronaviruses, and contributions of avidity to increased binding/neutralization of IgGs over Fabs. Using electron microscopy, we examined specificities of polyclonal plasma Fabs, revealing recognition of both S1
A and RBD epitopes on SARS-CoV-2 spike. Moreover, a 3.4 Å cryo-electron microscopy (cryo-EM) structure of a neutralizing monoclonal Fab-spike complex revealed an epitope that blocks ACE2 receptor binding. Modeling based on these structures suggested different potentials for inter-spike crosslinking by IgGs on viruses, and characterized IgGs would not be affected by identified SARS-CoV-2 spike mutations. Overall, our studies structurally define a recurrent anti-SARS-CoV-2 antibody class derived from VH3-53/VH3-66 and similarity to a SARS-CoV VH3-30 antibody, providing criteria for evaluating vaccine-elicited antibodies., Competing Interests: Declaration of Interests In connection with this work, The Rockefeller University has filed a provisional patent application on which D.F.R. and M.C.N. are inventors., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
38. A Combination of Human Broadly Neutralizing Antibodies against Hepatitis B Virus HBsAg with Distinct Epitopes Suppresses Escape Mutations.
- Author
-
Wang Q, Michailidis E, Yu Y, Wang Z, Hurley AM, Oren DA, Mayer CT, Gazumyan A, Liu Z, Zhou Y, Schoofs T, Yao KH, Nieke JP, Wu J, Jiang Q, Zou C, Kabbani M, Quirk C, Oliveira T, Chhosphel K, Zhang Q, Schneider WM, Jahan C, Ying T, Horowitz J, Caskey M, Jankovic M, Robbiani DF, Wen Y, de Jong YP, Rice CM, and Nussenzweig MC
- Subjects
- Animals, Antibodies, Monoclonal immunology, Cell Line, Tumor, Child, Preschool, Disease Models, Animal, Epitopes immunology, Female, HEK293 Cells, Hep G2 Cells, Hepatitis B, Chronic drug therapy, Hepatitis B, Chronic immunology, Humans, Infant, Mice, Mice, Knockout, Protein Conformation, Broadly Neutralizing Antibodies immunology, Hepatitis B Antibodies immunology, Hepatitis B Surface Antigens immunology, Hepatitis B virus immunology
- Abstract
Although there is no effective cure for chronic hepatitis B virus (HBV) infection, antibodies are protective and correlate with recovery from infection. To examine the human antibody response to HBV, we screened 124 vaccinated and 20 infected, spontaneously recovered individuals. The selected individuals produced shared clones of broadly neutralizing antibodies (bNAbs) that targeted 3 non-overlapping epitopes on the HBV S antigen (HBsAg). Single bNAbs protected humanized mice against infection but selected for resistance mutations in mice with prior established infection. In contrast, infection was controlled by a combination of bNAbs targeting non-overlapping epitopes with complementary sensitivity to mutations that commonly emerge during human infection. The co-crystal structure of one of the bNAbs with an HBsAg peptide epitope revealed a stabilized hairpin loop. This structure, which contains residues frequently mutated in clinical immune escape variants, provides a molecular explanation for why immunotherapy for HBV infection may require combinations of complementary bNAbs., Competing Interests: Declaration of Interests Q.W. and M.C.N. have a provisional patent application with the U.S. Patent and Trademark Office (62898735). Other authors have no conflicts of interest to declare., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
39. Convergent antibody responses to SARS-CoV-2 in convalescent individuals.
- Author
-
Robbiani DF, Gaebler C, Muecksch F, Lorenzi JCC, Wang Z, Cho A, Agudelo M, Barnes CO, Gazumyan A, Finkin S, Hägglöf T, Oliveira TY, Viant C, Hurley A, Hoffmann HH, Millard KG, Kost RG, Cipolla M, Gordon K, Bianchini F, Chen ST, Ramos V, Patel R, Dizon J, Shimeliovich I, Mendoza P, Hartweger H, Nogueira L, Pack M, Horowitz J, Schmidt F, Weisblum Y, Michailidis E, Ashbrook AW, Waltari E, Pak JE, Huey-Tubman KE, Koranda N, Hoffman PR, West AP Jr, Rice CM, Hatziioannou T, Bjorkman PJ, Bieniasz PD, Caskey M, and Nussenzweig MC
- Subjects
- Adolescent, Adult, Aged, Antibodies, Monoclonal analysis, Antibodies, Monoclonal immunology, Antibodies, Neutralizing analysis, Antibodies, Viral analysis, Antibody Specificity, COVID-19, COVID-19 Vaccines, Coronavirus Infections prevention & control, Enzyme-Linked Immunosorbent Assay, Female, Humans, Male, Middle Aged, Neutralization Tests, Pandemics, SARS-CoV-2, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus immunology, Viral Vaccines immunology, Young Adult, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Betacoronavirus immunology, Coronavirus Infections immunology, Pneumonia, Viral immunology
- Abstract
During the coronavirus disease-2019 (COVID-19) pandemic, severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) has led to the infection of millions of people and has claimed hundreds of thousands of lives. The entry of the virus into cells depends on the receptor-binding domain (RBD) of the spike (S) protein of SARS-CoV-2. Although there is currently no vaccine, it is likely that antibodies will be essential for protection. However, little is known about the human antibody response to SARS-CoV-2
1-5 . Here we report on 149 COVID-19-convalescent individuals. Plasma samples collected an average of 39 days after the onset of symptoms had variable half-maximal pseudovirus neutralizing titres; titres were less than 50 in 33% of samples, below 1,000 in 79% of samples and only 1% of samples had titres above 5,000. Antibody sequencing revealed the expansion of clones of RBD-specific memory B cells that expressed closely related antibodies in different individuals. Despite low plasma titres, antibodies to three distinct epitopes on the RBD neutralized the virus with half-maximal inhibitory concentrations (IC50 values) as low as 2 ng ml-1 . In conclusion, most convalescent plasma samples obtained from individuals who recover from COVID-19 do not contain high levels of neutralizing activity. Nevertheless, rare but recurring RBD-specific antibodies with potent antiviral activity were found in all individuals tested, suggesting that a vaccine designed to elicit such antibodies could be broadly effective.- Published
- 2020
- Full Text
- View/download PDF
40. Convergent Antibody Responses to SARS-CoV-2 Infection in Convalescent Individuals.
- Author
-
Robbiani DF, Gaebler C, Muecksch F, Lorenzi JCC, Wang Z, Cho A, Agudelo M, Barnes CO, Gazumyan A, Finkin S, Hagglof T, Oliveira TY, Viant C, Hurley A, Hoffmann HH, Millard KG, Kost RG, Cipolla M, Gordon K, Bianchini F, Chen ST, Ramos V, Patel R, Dizon J, Shimeliovich I, Mendoza P, Hartweger H, Nogueira L, Pack M, Horowitz J, Schmidt F, Weisblum Y, Michailidis E, Ashbrook AW, Waltari E, Pak JE, Huey-Tubman KE, Koranda N, Hoffman PR, West AP Jr, Rice CM, Hatziioannou T, Bjorkman PJ, Bieniasz PD, Caskey M, and Nussenzweig MC
- Abstract
During the COVID-19 pandemic, SARS-CoV-2 infected millions of people and claimed hundreds of thousands of lives. Virus entry into cells depends on the receptor binding domain (RBD) of the SARS-CoV-2 spike protein (S). Although there is no vaccine, it is likely that antibodies will be essential for protection. However, little is known about the human antibody response to SARS-CoV-2
1-5 . Here we report on 149 COVID-19 convalescent individuals. Plasmas collected an average of 39 days after the onset of symptoms had variable half-maximal neutralizing titers ranging from undetectable in 33% to below 1:1000 in 79%, while only 1% showed titers >1:5000. Antibody cloning revealed expanded clones of RBD-specific memory B cells expressing closely related antibodies in different individuals. Despite low plasma titers, antibodies to three distinct epitopes on RBD neutralized at half-maximal inhibitory concentrations (IC50 s) as low as single digit ng/mL. Thus, most convalescent plasmas obtained from individuals who recover from COVID-19 do not contain high levels of neutralizing activity. Nevertheless, rare but recurring RBD-specific antibodies with potent antiviral activity were found in all individuals tested, suggesting that a vaccine designed to elicit such antibodies could be broadly effective., Competing Interests: Declaration of conflict: In connection with this work The Rockefeller University has filed a provisional patent application on which D.F.R. and M.C.N. are inventors.- Published
- 2020
- Full Text
- View/download PDF
41. Structural basis for Zika envelope domain III recognition by a germline version of a recurrent neutralizing antibody.
- Author
-
Esswein SR, Gristick HB, Jurado A, Peace A, Keeffe JR, Lee YE, Voll AV, Saeed M, Nussenzweig MC, Rice CM, Robbiani DF, MacDonald MR, and Bjorkman PJ
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Dengue Virus immunology, Dengue Virus pathogenicity, Epitopes immunology, Germ Cells immunology, Humans, Infant, Newborn, Protein Domains immunology, Viral Vaccines immunology, West Nile virus immunology, West Nile virus pathogenicity, Yellow fever virus immunology, Yellow fever virus pathogenicity, Zika Virus isolation & purification, Zika Virus Infection diagnosis, Zika Virus Infection virology, Antibodies, Neutralizing isolation & purification, Antibodies, Viral immunology, Viral Envelope Proteins immunology, Zika Virus immunology, Zika Virus Infection immunology
- Abstract
Recent epidemics demonstrate the global threat of Zika virus (ZIKV), a flavivirus transmitted by mosquitoes. Although infection is usually asymptomatic or mild, newborns of infected mothers can display severe symptoms, including neurodevelopmental abnormalities and microcephaly. Given the large-scale spread, symptom severity, and lack of treatment or prophylaxis, a safe and effective ZIKV vaccine is urgently needed. However, vaccine design is complicated by concern that elicited antibodies (Abs) may cross-react with other flaviviruses that share a similar envelope protein, such as dengue virus, West Nile virus, and yellow fever virus. This cross-reactivity may worsen symptoms of a subsequent infection through Ab-dependent enhancement. To better understand the neutralizing Ab response and risk of Ab-dependent enhancement, further information on germline Ab binding to ZIKV and the maturation process that gives rise to potently neutralizing Abs is needed. Here we use binding and structural studies to compare mature and inferred-germline Ab binding to envelope protein domain III of ZIKV and other flaviviruses. We show that affinity maturation of the light-chain variable domain is important for strong binding of the recurrent VH3-23/VK1-5 neutralizing Abs to ZIKV envelope protein domain III, and identify interacting residues that contribute to weak, cross-reactive binding to West Nile virus. These findings provide insight into the affinity maturation process and potential cross-reactivity of VH3-23/VK1-5 neutralizing Abs, informing precautions for protein-based vaccines designed to elicit germline versions of neutralizing Abs., Competing Interests: Competing interest statement: D.F.R., M.C.N., and The Rockefeller University have filed a patent application for antibody Z004.
- Published
- 2020
- Full Text
- View/download PDF
42. A combination of two human monoclonal antibodies limits fetal damage by Zika virus in macaques.
- Author
-
Van Rompay KKA, Coffey LL, Kapoor T, Gazumyan A, Keesler RI, Jurado A, Peace A, Agudelo M, Watanabe J, Usachenko J, Singapuri A, Immareddy R, Ardeshir A, Stuart JB, Bournazos S, Ravetch JV, Balderes PJ, Lorenz IC, Esswein SR, Keeffe JR, Bjorkman PJ, Wang Q, Rice CM, MacDonald MR, Nussenzweig MC, and Robbiani DF
- Subjects
- Animals, Animals, Newborn, Antibodies, Monoclonal genetics, Antibodies, Monoclonal immunology, Antibodies, Neutralizing administration & dosage, Antibodies, Neutralizing genetics, Antibodies, Neutralizing immunology, Disease Models, Animal, Drug Therapy, Combination, Female, Fetus immunology, Fetus virology, HEK293 Cells, Humans, Immunoglobulin Fc Fragments administration & dosage, Immunoglobulin Fc Fragments genetics, Immunoglobulin Fc Fragments immunology, Immunoglobulin G administration & dosage, Immunoglobulin G genetics, Immunoglobulin G immunology, Pregnancy, Pregnancy Complications, Infectious immunology, Pregnancy Complications, Infectious virology, Protein Engineering, RNA, Viral isolation & purification, Recombinant Proteins administration & dosage, Recombinant Proteins genetics, Recombinant Proteins immunology, Zika Virus genetics, Zika Virus pathogenicity, Zika Virus Infection immunology, Zika Virus Infection transmission, Zika Virus Infection virology, Antibodies, Monoclonal administration & dosage, Infectious Disease Transmission, Vertical prevention & control, Pregnancy Complications, Infectious prevention & control, Zika Virus immunology, Zika Virus Infection prevention & control
- Abstract
Human infection by Zika virus (ZIKV) during pregnancy can lead to vertical transmission and fetal aberrations, including microcephaly. Prophylactic administration of antibodies can diminish or prevent ZIKV infection in animal models, but whether passive immunization can protect nonhuman primates and their fetuses during pregnancy has not been determined. Z004 and Z021 are neutralizing monoclonal antibodies to domain III of the envelope (EDIII) of ZIKV. Together the two antibodies protect nonpregnant macaques against infection even after Fc modifications to prevent antibody-dependent enhancement (ADE) in vitro and extend their half-lives. Here we report on prophylactic coadministration of the Fc-modified antibodies to pregnant rhesus macaques challenged three times with ZIKV during first and second trimester. The two antibodies did not entirely eliminate maternal viremia but limited vertical transmission, protecting the fetus from neurologic damage. Thus, maternal passive immunization with two antibodies to EDIII can shield primate fetuses from the harmful effects of ZIKV., Competing Interests: Competing interest statement: The Rockefeller University, D.F.R., and M.C.N. have filed a patent application for antibodies Z004 and Z021., (Copyright © 2020 the Author(s). Published by PNAS.)
- Published
- 2020
- Full Text
- View/download PDF
43. Risk of Zika microcephaly correlates with features of maternal antibodies.
- Author
-
Robbiani DF, Olsen PC, Costa F, Wang Q, Oliveira TY, Nery N Jr, Aromolaran A, do Rosário MS, Sacramento GA, Cruz JS, Khouri R, Wunder EA Jr, Mattos A, de Paula Freitas B, Sarno M, Archanjo G, Daltro D, Carvalho GBS, Pimentel K, de Siqueira IC, de Almeida JRM, Henriques DF, Lima JA, Vasconcelos PFC, Schaefer-Babajew D, Azzopardi SA, Bozzacco L, Gazumyan A, Belfort R Jr, Alcântara AP, Carvalho G, Moreira L, Araujo K, Reis MG, Keesler RI, Coffey LL, Tisoncik-Go J, Gale M Jr, Rajagopal L, Adams Waldorf KM, Dudley DM, Simmons HA, Mejia A, O'Connor DH, Steinbach RJ, Haese N, Smith J, Lewis A, Colgin L, Roberts V, Frias A, Kelleher M, Hirsch A, Streblow DN, Rice CM, MacDonald MR, de Almeida ARP, Van Rompay KKA, Ko AI, and Nussenzweig MC
- Subjects
- Animals, Brain embryology, Brain immunology, Brain pathology, Female, Fetus embryology, Fetus immunology, Fetus pathology, Humans, K562 Cells, Macaca mulatta, Macaca nemestrina, Microcephaly pathology, Pregnancy, Pregnancy Complications, Infectious pathology, Zika Virus Infection pathology, Antibodies, Viral immunology, Maternal-Fetal Exchange immunology, Microcephaly immunology, Pregnancy Complications, Infectious immunology, Zika Virus immunology, Zika Virus Infection immunology
- Abstract
Zika virus (ZIKV) infection during pregnancy causes congenital abnormalities, including microcephaly. However, rates vary widely, and the contributing risk factors remain unclear. We examined the serum antibody response to ZIKV and other flaviviruses in Brazilian women giving birth during the 2015-2016 outbreak. Infected pregnancies with intermediate or higher ZIKV antibody enhancement titers were at increased risk to give birth to microcephalic infants compared with those with lower titers (P < 0.0001). Similarly, analysis of ZIKV-infected pregnant macaques revealed that fetal brain damage was more frequent in mothers with higher enhancement titers. Thus, features of the maternal antibodies are associated with and may contribute to the genesis of ZIKV-associated microcephaly., (© 2019 Robbiani et al.)
- Published
- 2019
- Full Text
- View/download PDF
44. A Combination of Two Human Monoclonal Antibodies Prevents Zika Virus Escape Mutations in Non-human Primates.
- Author
-
Keeffe JR, Van Rompay KKA, Olsen PC, Wang Q, Gazumyan A, Azzopardi SA, Schaefer-Babajew D, Lee YE, Stuart JB, Singapuri A, Watanabe J, Usachenko J, Ardeshir A, Saeed M, Agudelo M, Eisenreich T, Bournazos S, Oliveira TY, Rice CM, Coffey LL, MacDonald MR, Bjorkman PJ, Nussenzweig MC, and Robbiani DF
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal chemistry, Antibodies, Neutralizing immunology, Dengue Virus immunology, Epitopes immunology, HEK293 Cells, Humans, Macaca, Mice, Knockout, Protein Domains, Antibodies, Monoclonal immunology, Mutation genetics, Zika Virus immunology
- Abstract
Zika virus (ZIKV) causes severe neurologic complications and fetal aberrations. Vaccine development is hindered by potential safety concerns due to antibody cross-reactivity with dengue virus and the possibility of disease enhancement. In contrast, passive administration of anti-ZIKV antibodies engineered to prevent enhancement may be safe and effective. Here, we report on human monoclonal antibody Z021, a potent neutralizer that recognizes an epitope on the lateral ridge of the envelope domain III (EDIII) of ZIKV and is protective against ZIKV in mice. When administered to macaques undergoing a high-dose ZIKV challenge, a single anti-EDIII antibody selected for resistant variants. Co-administration of two antibodies, Z004 and Z021, which target distinct sites on EDIII, was associated with a delay and a 3- to 4-log decrease in peak viremia. Moreover, the combination of these antibodies engineered to avoid enhancement prevented viral escape due to mutation in macaques, a natural host for ZIKV., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
45. Correction: RAG1/2 induces genomic insertions by mobilizing DNA into RAG1/2-independent breaks.
- Author
-
Rommel PC, Oliveira TY, Nussenzweig MC, and Robbiani DF
- Published
- 2017
- Full Text
- View/download PDF
46. Recurrent Potent Human Neutralizing Antibodies to Zika Virus in Brazil and Mexico.
- Author
-
Robbiani DF, Bozzacco L, Keeffe JR, Khouri R, Olsen PC, Gazumyan A, Schaefer-Babajew D, Avila-Rios S, Nogueira L, Patel R, Azzopardi SA, Uhl LFK, Saeed M, Sevilla-Reyes EE, Agudelo M, Yao KH, Golijanin J, Gristick HB, Lee YE, Hurley A, Caskey M, Pai J, Oliveira T, Wunder EA Jr, Sacramento G, Nery N Jr, Orge C, Costa F, Reis MG, Thomas NM, Eisenreich T, Weinberger DM, de Almeida ARP, West AP Jr, Rice CM, Bjorkman PJ, Reyes-Teran G, Ko AI, MacDonald MR, and Nussenzweig MC
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, Antibodies, Viral blood, Antibodies, Viral immunology, B-Lymphocytes immunology, Brazil, Female, Humans, Immunologic Memory, Leukocytes, Mononuclear immunology, Male, Mexico, Mice, Zika Virus Infection blood, Antibodies, Neutralizing chemistry, Antibodies, Viral chemistry, Zika Virus Infection immunology
- Abstract
Antibodies to Zika virus (ZIKV) can be protective. To examine the antibody response in individuals who develop high titers of anti-ZIKV antibodies, we screened cohorts in Brazil and Mexico for ZIKV envelope domain III (ZEDIII) binding and neutralization. We find that serologic reactivity to dengue 1 virus (DENV1) EDIII before ZIKV exposure is associated with increased ZIKV neutralizing titers after exposure. Antibody cloning shows that donors with high ZIKV neutralizing antibody titers have expanded clones of memory B cells that express the same immunoglobulin VH3-23/VK1-5 genes. These recurring antibodies cross-react with DENV1, but not other flaviviruses, neutralize both DENV1 and ZIKV, and protect mice against ZIKV challenge. Structural analyses reveal the mechanism of recognition of the ZEDIII lateral ridge by VH3-23/VK1-5 antibodies. Serologic testing shows that antibodies to this region correlate with serum neutralizing activity to ZIKV. Thus, high neutralizing responses to ZIKV are associated with pre-existing reactivity to DENV1 in humans., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
47. RAG1/2 induces genomic insertions by mobilizing DNA into RAG1/2-independent breaks.
- Author
-
Rommel PC, Oliveira TY, Nussenzweig MC, and Robbiani DF
- Subjects
- Animals, B-Lymphocytes metabolism, Genomic Instability, Immunoglobulins genetics, Mice, Mice, Inbred C57BL, Receptors, Antigen, T-Cell genetics, Translocation, Genetic, V(D)J Recombination, DNA Damage, DNA-Binding Proteins physiology, Homeodomain Proteins physiology, Mutagenesis, Insertional
- Abstract
The RAG recombinase (RAG1/2) plays an essential role in adaptive immunity by mediating V(D)J recombination in developing lymphocytes. In contrast, aberrant RAG1/2 activity promotes lymphocyte malignancies by causing chromosomal translocations and DNA deletions at cancer genes. RAG1/2 can also induce genomic DNA insertions by transposition and trans-V(D)J recombination, but only few such putative events have been documented in vivo. We used next-generation sequencing techniques to examine chromosomal rearrangements in primary murine B cells and discovered that RAG1/2 causes aberrant insertions by releasing cleaved antibody gene fragments that subsequently reintegrate into DNA breaks induced on a heterologous chromosome. We confirmed that RAG1/2 also mobilizes genomic DNA into independent physiological breaks by identifying similar insertions in human lymphoma and leukemia. Our findings reveal a novel RAG1/2-mediated insertion pathway distinct from DNA transposition and trans-V(D)J recombination that destabilizes the genome and shares features with reported oncogenic DNA insertions., (© 2017 Rommel et al.)
- Published
- 2017
- Full Text
- View/download PDF
48. The cell cycle restricts activation-induced cytidine deaminase activity to early G1.
- Author
-
Wang Q, Kieffer-Kwon KR, Oliveira TY, Mayer CT, Yao K, Pai J, Cao Z, Dose M, Casellas R, Jankovic M, Nussenzweig MC, and Robbiani DF
- Subjects
- Animals, Cell Cycle, Cell Nucleus enzymology, Cells, Cultured, DNA Damage, Deamination, Genes, Immunoglobulin, Mice, Mice, Inbred C57BL, Transcription, Genetic, AICDA (Activation-Induced Cytidine Deaminase), Cytidine Deaminase metabolism, G1 Phase
- Abstract
Activation-induced cytidine deaminase (AID) converts cytosine into uracil to initiate somatic hypermutation (SHM) and class switch recombination (CSR) of antibody genes. In addition, this enzyme produces DNA lesions at off-target sites that lead to mutations and chromosome translocations. However, AID is mostly cytoplasmic, and how and exactly when it accesses nuclear DNA remains enigmatic. Here, we show that AID is transiently in spatial contact with genomic DNA from the time the nuclear membrane breaks down in prometaphase until early G1, when it is actively exported into the cytoplasm. Consistent with this observation, the immunoglobulin (Igh) gene deamination as measured by uracil accumulation occurs primarily in early G1 after chromosomes decondense. Altering the timing of cell cycle-regulated AID nuclear residence increases DNA damage at off-target sites. Thus, the cell cycle-controlled breakdown and reassembly of the nuclear membrane and the restoration of transcription after mitosis constitute an essential time window for AID-induced deamination, and provide a novel DNA damage mechanism restricted to early G1., (© 2017 Wang et al.)
- Published
- 2017
- Full Text
- View/download PDF
49. Mutations, kataegis and translocations in B cells: understanding AID promiscuous activity.
- Author
-
Casellas R, Basu U, Yewdell WT, Chaudhuri J, Robbiani DF, and Di Noia JM
- Subjects
- Animals, Humans, Somatic Hypermutation, Immunoglobulin immunology, Translocation, Genetic, AICDA (Activation-Induced Cytidine Deaminase), B-Lymphocytes immunology, Cytidine Deaminase immunology, Immunoglobulin Class Switching immunology, Lymphoma, B-Cell genetics, Lymphoma, B-Cell immunology, Mutation
- Abstract
As B cells engage in the immune response, they express activation-induced cytidine deaminase (AID) to initiate the hypermutation and recombination of immunoglobulin genes, which are crucial processes for the efficient recognition and disposal of pathogens. However, AID must be tightly controlled in B cells to minimize off-target mutations, which can drive chromosomal translocations and the development of B cell malignancies, such as lymphomas. Recent genomic and biochemical analyses have begun to unravel the mechanisms of how AID-mediated deamination is targeted outside immunoglobulin genes. Here, we discuss the transcriptional and topological features that are emerging as key drivers of AID promiscuous activity.
- Published
- 2016
- Full Text
- View/download PDF
50. A New Way to Diversify Antibodies by DNA Transposition.
- Author
-
Robbiani DF and Nussenzweig MC
- Subjects
- Humans, Antibodies, Monoclonal immunology, Antibody Specificity, Antigenic Variation immunology, Antigens, Protozoan immunology, Malaria immunology, Mutagenesis, Insertional genetics, Plasmodium falciparum immunology, Receptors, Immunologic immunology
- Abstract
While searching for new therapeutics against malaria, Lanzavecchia and colleagues discovered that antibodies can be diversified by DNA sequences encoded outside of antibody genes., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.