283 results on '"Geisbert TW"'
Search Results
2. Potential impact of a 2-person security rule on BioSafety Level 4 laboratory workers.
- Author
-
LeDuc JW, Anderson K, Bloom ME, Carrion R Jr, Feldmann H, Fitch JP, Geisbert JB, Geisbert TW, Holbrook MR, Jahrling PB, Ksiazek TG, LeDuc, James W, Anderson, Kevin, Bloom, Marshall E, Carrion, Ricardo Jr, Feldmann, Heinz, Fitch, J Patrick, Geisbert, Joan B, Geisbert, Thomas W, and Holbrook, Michael R
- Abstract
Directors of all major BioSafety Level 4 (BSL-4) laboratories in the United States met in 2008 to review the current status of biocontainment laboratory operations and to discuss the potential impact of a proposed 2-person security rule on maximum-containment laboratory operations. Special attention was paid to the value and risks that would result from a requirement that 2 persons be physically present in the laboratory at all times. A consensus emerged indicating that a video monitoring system represents a more efficient, economical standard; provides greater assurance that pathogens are properly manipulated; and offers an increased margin of employee safety and institutional security. The 2-person security rule (1 to work and 1 to observe) may decrease compliance with dual responsibilities of safety and security by placing undue pressure on the person being observed to quickly finish the work, and by placing the observer in the containment environment unnecessarily. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
3. Marburg virus Angola infection of rhesus macaques: pathogenesis and treatment with recombinant nematode anticoagulant protein c2.
- Author
-
Geisbert TW, Daddario-DiCaprio KM, Geisbert JB, Young HA, Formenty P, Fritz EA, Laren T, Hensley LE, Geisbert, Thomas W, Daddario-DiCaprio, Kathleen M, Geisbert, Joan B, Young, Howard A, Formenty, Pierre, Fritz, Elizabeth A, Larsen, Tom, and Hensley, Lisa E
- Abstract
Background: The procoagulant tissue factor (TF) is thought to play a role in the coagulation disorders that characterize filoviral infections. In this study, we evaluated the pathogenesis of lethal infection with the Angola strain of Marburg virus (MARV-Ang) in rhesus macaques and tested the efficacy of recombinant nematode anticoagulant protein c2 (rNAPc2), an inhibitor of TF/factor VIIa, as a potential treatment.Methods: Twelve rhesus macaques were challenged with a high dose (1000 pfu) of MARV-Ang. Six macaques were treated with rNAPc2, and 6 macaques served as control animals.Results: All 6 control animals succumbed to MARV-Ang challenge by day 8 (mean, 7.3 days), whereas 5 of 6 rNAPc2-treated animals died on day 9 and 1 rNAPc2-treated animal survived. The disease course for MARV-Ang infection appeared to progress more rapidly in rhesus macaques than has been previously reported for other strains of MARV. In contrast to Ebola virus (EBOV) infection in macaques, up-regulation of TF was not as striking, and deposition of fibrin was a less prominent pathologic feature of disease in these animals.Conclusions: These data show that the pathogenicity of MARV-Ang infection appears to be consistent with the apparent increased human virulence attributed to this strain. The apparent reduced efficacy of rNAPc2 against MARV-Ang infection, compared with its efficacy against EBOV infection, appears to be associated with differences in TF induction and fibrin deposition. [ABSTRACT FROM AUTHOR]- Published
- 2007
- Full Text
- View/download PDF
4. Monoclonal antibody therapy demonstrates increased virulence of a lineage VII strain of Lassa virus in nonhuman primates.
- Author
-
Woolsey C, Cross RW, Prasad AN, Agans KN, Borisevich V, Deer DJ, Dobias NS, Fears AC, Harrison MB, Heinrich ML, Fenton KA, Garry RF, Branco LM, and Geisbert TW
- Subjects
- Humans, Animals, Virulence, Macaca fascicularis, Antibodies, Monoclonal pharmacology, Lassa virus, Lassa Fever
- Abstract
Lassa virus (LASV) is a World Health Organization (WHO) priority pathogen that causes high morbidity and mortality. Recently, we showed that a combination of three broadly neutralizing human monoclonal antibodies known as Arevirumab-3 (8.9F, 12.1F, 37.2D) based on the lineage IV Josiah strain protected 100% of cynomolgus macaques against heterologous challenge with lineage II and III strains of LASV when therapy was initiated beginning at day 8 after challenge. LASV strains from Benin and Togo represent a new lineage VII that are more genetically diverse from lineage IV than strains from lineages II and III. Here, we tested the ability of Arevirumab-3 to protect macaques against a LASV lineage VII Togo isolate when treatment was administered beginning 8 days after exposure. Unexpectedly, only 40% of treated animals survived challenge. In a subsequent study we showed that Arevirumab-3 protected 100% of macaques from lethal challenge when treatment was initiated 7 days after LASV Togo exposure. Based on our transcriptomics data, successful Arevirumab-3 treatment correlated with diminished neutrophil signatures and the predicted development of T cell responses. As the in vitro antiviral activity of Arevirumab-3 against LASV Togo was equivalent to lineage II and III strains, the reduced protection in macaques against Togo likely reflects the faster disease course of LASV Togo in macaques than other strains. This data causes concern regarding the ability of heterologous vaccines and treatments to provide cross protection against lineage VII LASV isolates.
- Published
- 2024
- Full Text
- View/download PDF
5. Therapeutic administration of a cross-reactive mAb targeting the fusion glycoprotein of Nipah virus protects nonhuman primates.
- Author
-
Zeitlin L, Cross RW, Woolsey C, West BR, Borisevich V, Agans KN, Prasad AN, Deer DJ, Stuart L, McCavitt-Malvido M, Kim DH, Pettitt J, Crowe JE Jr, Whaley KJ, Veesler D, Dimitrov A, Abelson DM, Geisbert TW, and Broder CC
- Subjects
- Animals, Antibodies, Monoclonal, Bangladesh, Chlorocebus aethiops, Glycoproteins metabolism, Primates, Clinical Trials, Phase I as Topic, Henipavirus Infections prevention & control, Nipah Virus
- Abstract
No licensed vaccines or therapies exist for patients infected with Nipah virus (NiV), although an experimental human monoclonal antibody (mAb) cross-reactive to the NiV and Hendra virus (HeV) G glycoprotein, m102.4, has been tested in a phase 1 trial and has been provided under compassionate use for both HeV and NiV exposures. NiV is a highly pathogenic zoonotic paramyxovirus causing regular outbreaks in humans and animals in South and Southeast Asia. The mortality rate of NiV infection in humans ranges from 40% to more than 90%, making it a substantial public health concern. The NiV G glycoprotein mediates host cell attachment, and the F glycoprotein facilitates membrane fusion and infection. We hypothesized that a mAb against the prefusion conformation of the F glycoprotein may confer better protection than m102.4. To test this, two potent neutralizing mAbs against NiV F protein, hu1F5 and hu12B2, were compared in a hamster model. Hu1F5 provided superior protection to hu12B2 and was selected for comparison with m102.4 for the ability to protect African green monkeys (AGMs) from a stringent NiV challenge. AGMs were exposed intranasally to the Bangladesh strain of NiV and treated 5 days after exposure with either mAb (25 milligrams per kilogram). Whereas only one of six AGMs treated with m102.4 survived until the study end point, all six AGMs treated with hu1F5 were protected. Furthermore, a reduced 10 milligrams per kilogram dose of hu1F5 also provided complete protection against NiV challenge, supporting the upcoming clinical advancement of this mAb for postexposure prophylaxis and therapy.
- Published
- 2024
- Full Text
- View/download PDF
6. A glass-half-full perspective on negative data in Ebolavirus vaccine studies.
- Author
-
Prasad AN and Geisbert TW
- Published
- 2024
- Full Text
- View/download PDF
7. Oral administration of obeldesivir protects nonhuman primates against Sudan ebolavirus .
- Author
-
Cross RW, Woolsey C, Chu VC, Babusis D, Bannister R, Vermillion MS, Geleziunas R, Barrett KT, Bunyan E, Nguyen AQ, Cihlar T, Porter DP, Prasad AN, Deer DJ, Borisevich V, Agans KN, Martinez J, Harrison MB, Dobias NS, Fenton KA, Bilello JP, and Geisbert TW
- Subjects
- Animals, Administration, Oral, Macaca fascicularis, Adenosine Monophosphate administration & dosage, Adenosine Monophosphate pharmacology, Ebolavirus drug effects, Hemorrhagic Fever, Ebola drug therapy, Hemorrhagic Fever, Ebola prevention & control, Nucleosides administration & dosage, Nucleosides pharmacology, Alanine administration & dosage, Alanine analogs & derivatives, Alanine pharmacology, Prodrugs administration & dosage, Prodrugs pharmacology, Antiviral Agents administration & dosage, Antiviral Agents pharmacology
- Abstract
Obeldesivir (ODV, GS-5245) is an orally administered prodrug of the parent nucleoside of remdesivir (RDV) and is presently in phase 3 trials for COVID-19 treatment. In this work, we show that ODV and its circulating parent nucleoside metabolite, GS-441524, have similar in vitro antiviral activity against filoviruses, including Marburg virus, Ebola virus, and Sudan virus (SUDV). We also report that once-daily oral ODV treatment of cynomolgus monkeys for 10 days beginning 24 hours after SUDV exposure confers 100% protection against lethal infection. Transcriptomics data show that ODV treatment delayed the onset of inflammation and correlated with antigen presentation and lymphocyte activation. Our results offer promise for the further development of ODV to control outbreaks of filovirus disease more rapidly.
- Published
- 2024
- Full Text
- View/download PDF
8. Monoclonal antibody therapy protects nonhuman primates against mucosal exposure to Lassa virus.
- Author
-
Cross RW, Fenton KA, Woolsey C, Prasad AN, Borisevich V, Agans KN, Deer DJ, Dobias NS, Fears AC, Heinrich ML, Geisbert JB, Garry RF, Branco LM, and Geisbert TW
- Subjects
- Animals, Humans, Macaca fascicularis, Immunotherapy, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Lassa virus, Lassa Fever drug therapy, Lassa Fever prevention & control
- Abstract
Lassa fever (LF) is an acute viral illness that causes thousands of deaths annually in West Africa. There are currently no Lassa virus (LASV) vaccines or antivirals approved for human use. Recently, we showed that combinations of broadly neutralizing human monoclonal antibodies (BNhuMAbs) known as Arevirumab-2 or Arevirumab-3 protected up to 100% of cynomolgus macaques against challenge with diverse lineages of LASV when treatment was initiated at advanced stages of disease. This previous work assessed efficacy against parenteral exposure. However, transmission of LASV to humans occurs primarily by mucosal exposure to virus shed from Mastomys rodents. Here, we describe the development of a lethal intranasal exposure macaque model of LF. This model is employed to show that Arevirumab cocktails rescue 100% of macaques from lethal LASV infection when treatment is initiated 8 days after LASV exposure. Our work demonstrates BNhuMAbs have utility in treating LASV infection acquired through mucosal exposure., Competing Interests: Declaration of interests L.M.B., R.F.G., and T.W.G. are named on a provisional World International Property Organization patent entitled “Arenavirus Monoclonal Antibodies and Uses” (International publication number WO 2018/106712 A1 filed on 14 June 2018) and on a US Patent application entitled “Arenavirus Monoclonal Antibodies and Uses” (Publication number US 2022/0089696 A1 filed on 24 March 2022). L.M.B. and R.F.G. are co-founders of Zalgen Labs. L.M.B. receives compensation from Zalgen Labs. L.M.B. is employed by Zalgen Labs and receives compensation from the company., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. A Single-shot ChAd3 Vaccine Provides Protection from Intramuscular and Aerosol Sudan Virus Exposure.
- Author
-
Honko AN, Hunegnaw R, Moliva JI, Ploquin A, Dulan CNM, Murray T, Carr D, Foulds KE, Geisbert JB, Geisbert TW, Johnson JC, Wollen-Roberts SE, Trefry JC, Stanley DA, and Sullivan NJ
- Abstract
Infection with Sudan virus (SUDV) is characterized by an aggressive disease course with case fatality rates between 40-100% and no approved vaccines or therapeutics. SUDV causes sporadic outbreaks in sub-Saharan Africa, including a recent outbreak in Uganda which has resulted in over 100 confirmed cases in one month. Prior vaccine and therapeutic efforts have historically prioritized Ebola virus (EBOV), leading to a significant gap in available treatments. Two vaccines, Erbevo
® and Zabdeno® /Mvabea® , are licensed for use against EBOV but are ineffective against SUDV. Recombinant adenovirus vector vaccines have been shown to be safe and effective against filoviruses, but efficacy depends on having low seroprevalence to the vector in the target human population. For this reason, and because of an excellent safety and immunogenicity profile, ChAd3 was selected as a superior vaccine vector. Here, a ChAd3 vaccine expressing the SUDV glycoprotein (GP) was evaluated for immunogenicity and efficacy in nonhuman primates. We demonstrate that a single dose of ChAd3-SUDV confers acute and durable protection against lethal SUDV challenge with a strong correlation between the SUDV GP-specific antibody titers and survival outcome. Additionally, we show that a bivalent ChAd3 vaccine encoding the GP from both EBOV and SUDV protects against both parenteral and aerosol lethal SUDV challenge. Our data indicate that the ChAd3-SUDV vaccine is a suitable candidate for a prophylactic vaccination strategy in regions at high risk of filovirus outbreaks. One Sentence Summary: A single-dose of ChAd3 vaccine protected macaques from lethal challenge with Sudan virus (SUDV) by parenteral and aerosol routes of exposure.- Published
- 2024
- Full Text
- View/download PDF
10. Thermostable bivalent filovirus vaccine protects against severe and lethal Sudan ebolavirus and marburgvirus infection.
- Author
-
To A, Wong TAS, Ball AH, Lieberman MM, Yalley-Ogunro J, Cabus M, Nezami S, Paz F, Elyard HA, Borisevich V, Agans KN, Deer DJ, Woolsey C, Cross RW, Geisbert TW, Donini O, and Lehrer AT
- Subjects
- Animals, Humans, Vaccines, Combined, Sudan, Antibodies, Viral, Macaca fascicularis, Water, Ebolavirus, Marburgvirus, Hemorrhagic Fever, Ebola, Viral Vaccines
- Abstract
Although two vaccines for Zaire ebolavirus (EBOV) have been licensed and deployed successfully to combat recurring outbreaks of Ebolavirus Disease in West Africa, there are no vaccines for two other highly pathogenic members of the Filoviridae, Sudan ebolavirus (SUDV) and Marburg marburgvirus (MARV). The results described herein document the immunogenicity and protective efficacy in cynomolgus macaques of a single-vial, thermostabilized (lyophilized) monovalent (SUDV) and bivalent (SUDV & MARV) protein vaccines consisting of recombinant glycoproteins (GP) formulated with a clinical-grade oil-in-water nanoemulsion adjuvant (CoVaccine HT™). Lyophilized formulations of the vaccines were reconstituted with Water for Injection and used to immunize groups of cynomolgus macaques before challenge with a lethal dose of a human SUDV or MARV isolate. Sera collected after each of the three immunizations showed near maximal GP-binding IgG concentrations starting as early as the second dose. Most importantly, the vaccine candidates (monovalent or bivalent) provided 100% protection against severe and lethal filovirus disease after either SUDV or MARV infection. Although mild, subclinical infection was observed in a few macaques, all vaccinated animals remained healthy and survived the filovirus challenge. These results demonstrate the value that thermostabilized protein vaccines could provide for addressing an important gap in preparedness for future filovirus outbreaks., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Axel T. Lehrer and Oreola Donini are named inventors on a patent covering multivalent filovirus subunit vaccines (U.S. Patent 11,433,129B2). Jake Yalley-Ogunro, Mehtap Cabus, Sara Nezami, Fabian Paz, Hanne Andersen Elyard are current employees of BIOQUAL, Inc. Oreola Donini is a current employee of Soligenix Inc. All other authors declare no competing interests., (Copyright © 2023. Published by Elsevier Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
11. Single-dose VSV-based vaccine protects cynomolgus macaques from disease after Taï Forest virus infection.
- Author
-
Fletcher P, O'Donnell KL, Doratt BM, Malherbe DC, Clancy CS, Rhoderick JF, Feldmann F, Hanley PW, Ksiazek TG, Geisbert TW, Messaoudi I, and Marzi A
- Subjects
- Animals, Humans, Macaca fascicularis, Antibodies, Viral, Forests, Ebolavirus, Hemorrhagic Fever, Ebola, Viral Vaccines
- Abstract
Taï Forest virus (TAFV) is a lesser-known ebolavirus that causes lethal infections in chimpanzees and is responsible for a single human case. Limited research has been done on this human pathogen; however, with the recent emergence of filoviruses in West Africa, further investigation and countermeasure development against this virus is warranted. We developed a vesicular stomatitis virus (VSV)-based vaccine expressing the TAFV glycoprotein as the viral antigen and assessed it for protective efficacy in nonhuman primates (NHPs). Following a single high-dose vaccination, NHPs developed antigen-specific binding and neutralizing antibodies as well as modest T cell responses. Importantly, all vaccinated NHPs were uniformly protected from disease after lethal TAFV challenge while the naïve control group succumbed to the disease. Histopathologic lesions consistent with filovirus disease were present in control NHPs but were not observed in vaccinated NHPs. Transcriptional analysis of whole blood samples obtained after vaccination and challenge was performed to gain insight into molecular underpinnings conferring protection. Differentially expressed genes (DEG) detected 7 days post-vaccination were enriched to processes associated with innate immunity and antiviral responses. Only a small number of DEG was detected in vaccinated NHPs post-challenge while over 1,000 DEG were detected in control NHPs at end-stage disease which mapped to gene ontology terms indicative of defense responses and inflammation. Taken together, this data demonstrates the effective single-dose protection of the VSV-TAFV vaccine, and its potential for use in outbreaks.
- Published
- 2023
- Full Text
- View/download PDF
12. The Therapeutic Monoclonal Antibody Bamlanivimab Does Not Enhance SARS-CoV-2 Infection by FcR-Mediated Mechanisms.
- Author
-
Cross RW, Wiethoff CM, Brown-Augsburger P, Berens S, Blackbourne J, Liu L, Wu X, Tetreault J, Dodd C, Sina R, Witcher DR, Newcomb D, Frost D, Wilcox A, Borisevich V, Agans KN, Woolsey C, Prasad AN, Deer DJ, Geisbert JB, Dobias NS, Fenton KA, Strifler B, Ebert P, Higgs R, Beall A, Chanda S, Riva L, Yin X, and Geisbert TW
- Abstract
As part of the non-clinical safety package characterizing bamlanivimab (SARS-CoV-2 neutralizing monoclonal antibody), the risk profile for antibody-dependent enhancement of infection (ADE) was evaluated in vitro and in an African green monkey (AGM) model of COVID-19. In vitro ADE assays in primary human macrophage, Raji, or THP-1 cells were used to evaluate enhancement of viral infection. Bamlanivimab binding to C1q, FcR, and cell-based effector activity was also assessed. In AGMs, the impact of bamlanivimab pretreatment on viral loads and clinical and histological pathology was assessed to evaluate enhanced SARS-CoV-2 replication or pathology. Bamlanivimab did not increase viral replication in vitro, despite a demonstrated effector function. In vivo, no significant differences were found among the AGM groups for weight, temperature, or food intake. Treatment with bamlanivimab reduced viral loads in nasal and oral swabs and BAL fluid relative to control groups. Viral antigen was not detected in lung tissue from animals treated with the highest dose of bamlanivimab. Bamlanivimab did not induce ADE of SARS-CoV-2 infection in vitro or in an AGM model of infection at any dose evaluated. The findings suggest that high-affinity monoclonal antibodies pose a low risk of mediating ADE in patients and support their safety profile as a treatment of COVID-19 disease.
- Published
- 2023
- Full Text
- View/download PDF
13. The Mucin-Like Domain of the Ebola Glycoprotein Does Not Impact Virulence or Pathogenicity in Ferrets.
- Author
-
Halfmann PJ, Borisevich V, Levine CB, Mire CE, Fenton KA, Geisbert TW, Kawaoka Y, and Cross RW
- Subjects
- Animals, Humans, Mucins, Virulence, Ferrets, Glycoproteins genetics, Glycoproteins metabolism, Hemorrhagic Fever, Ebola, Ebolavirus
- Abstract
Background: Ebola virus (EBOV) is considered among the most dangerous viruses with case fatality rates approaching 90% depending on the outbreak. While several viral proteins (VPs) including VP24, VP35, and the soluble glycoprotein are understood to contribute to virulence, less is known of the contribution of the highly variable mucin-like domain (MLD) of EBOV. Early studies have defined a potential role in immune evasion of the MLD by providing a glycan shield to critical glycoprotein residues tied to viral entry. Nonetheless, little is known as to what direct role the MLD plays in acute EBOV disease (EVD)., Methods: We generated an infectious EBOV clone that lacks the MLD and assessed its virulence in ferrets compared with wild-type (WT) virus., Results: No differences in growth kinetics were observed in vitro, nor were there any differences in time to death, viremia, or clinical picture in ferrets infected with recombinant EBOV (rEBOV)-WT or rEBOV-Δmucin., Conclusions: The EBOV MLD does not play a critical role in acute pathogenesis of EVD in ferrets., Competing Interests: Potential conflicts of interest. The authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
14. Modelling Marburg Virus Disease in Syrian Golden Hamsters: Contrasted Virulence Between Angola and Ci67 Strains.
- Author
-
Cross RW, Fenton KA, Foster SL, Geisbert JB, and Geisbert TW
- Subjects
- Cricetinae, Humans, Guinea Pigs, Animals, Mesocricetus, Virulence, Angola, Marburg Virus Disease, Marburgvirus
- Abstract
Background: Marburg virus (MARV) has caused numerous sporadic outbreaks of severe hemorrhagic fever in humans. Human case fatality rates of Marburg virus disease (MVD) outbreaks range from 20% to 90%. Viral genotypes of MARV can differ by over 20%, suggesting variable virulence between lineages may accompany this genetic divergence. Comparison of existing animal models of MVD employing different strains of MARV support differences in virulence across MARV genetic lineages; however, there are few systematic comparisons in models that recapitulate human disease available., Methods: We compared features of disease pathogenesis in uniformly lethal hamster models of MVD made possible through serial adaptation in rodents., Results: No further adaptation from a previously reported guinea pig-adapted (GPA) isolate of MARV-Angola was necessary to achieve uniform lethality in hamsters. Three passages of GPA MARV-Ci67 resulted in uniform lethality, where 4 passages of a GPA Ravn virus was 75% lethal. Hamster-adapted MARV-Ci67 demonstrated delayed time to death, protracted weight loss, lower viral burden, and slower histologic alteration compared to GPA MARV-Angola., Conclusions: These data suggest isolate-dependent virulence differences are maintained even after serial adaptation in rodents and may serve to guide choice of variant and model used for development of vaccines or therapeutics for MVD., Competing Interests: Potential conflicts of interest. All authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
15. A Recombinant Vesicular Stomatitis Virus-Based Vaccine Provides Postexposure Protection Against Bundibugyo Ebolavirus Infection.
- Author
-
Woolsey C, Strampe J, Fenton KA, Agans KN, Martinez J, Borisevich V, Dobias NS, Deer DJ, Geisbert JB, Cross RW, Connor JH, and Geisbert TW
- Subjects
- Animals, Antibodies, Viral, Vesiculovirus genetics, Glycoproteins genetics, Macaca fascicularis, Immunoglobulin G, Hemorrhagic Fever, Ebola, Ebolavirus, Vesicular Stomatitis prevention & control, Viral Vaccines, Ebola Vaccines
- Abstract
Background: The filovirus Bundibugyo virus (BDBV) causes severe disease with a mortality rate of approximately 20%-51%. The only licensed filovirus vaccine in the United States, Ervebo, consists of a recombinant vesicular stomatitis virus (rVSV) vector that expresses Ebola virus (EBOV) glycoprotein (GP). Ervebo was shown to rapidly protect against fatal Ebola disease in clinical trials; however, the vaccine is only indicated against EBOV. Recent outbreaks of other filoviruses underscore the need for additional vaccine candidates, particularly for BDBV infections., Methods: To examine whether the rVSV vaccine candidate rVSVΔG/BDBV-GP could provide therapeutic protection against BDBV, we inoculated seven cynomolgus macaques with 1000 plaque-forming units of BDBV, administering rVSVΔG/BDBV-GP vaccine to 6 of them 20-23 minutes after infection., Results: Five of the treated animals survived infection (83%) compared to an expected natural survival rate of 21% in this macaque model. All treated animals showed an early circulating immune response, while the untreated animal did not. Surviving animals showed evidence of both GP-specific IgM and IgG production, while animals that succumbed did not produce significant IgG., Conclusions: This small, proof-of-concept study demonstrated early treatment with rVSVΔG/BDBV-GP provides a survival benefit in this nonhuman primate model of BDBV infection, perhaps through earlier initiation of adaptive immunity., Competing Interests: Conflict of Interest . T. W. G. claims intellectual property regarding recombinant VSV-based vaccines for the prevention and treatment of filovirus infections. All other authors report no potential conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
16. A Highly Attenuated Panfilovirus VesiculoVax Vaccine Rapidly Protects Nonhuman Primates Against Marburg Virus and 3 Species of Ebola Virus.
- Author
-
Woolsey C, Borisevich V, Agans KN, O'Toole R, Fenton KA, Harrison MB, Prasad AN, Deer DJ, Gerardi C, Morrison N, Cross RW, Eldridge JH, Matassov D, and Geisbert TW
- Subjects
- Humans, Animals, Vaccines, Attenuated, Macaca fascicularis, Vesiculovirus genetics, Vesicular stomatitis Indiana virus, Antibodies, Viral, Ebolavirus, Hemorrhagic Fever, Ebola, Marburgvirus, Viral Vaccines, Ebola Vaccines
- Abstract
Background: The family Filoviridae consists of several virus members known to cause significant mortality and disease in humans. Among these, Ebola virus (EBOV), Marburg virus (MARV), Sudan virus (SUDV), and Bundibugyo virus (BDBV) are considered the deadliest. The vaccine, Ervebo, was shown to rapidly protect humans against Ebola disease, but is indicated only for EBOV infections with limited cross-protection against other filoviruses. Whether multivalent formulations of similar recombinant vesicular stomatitis virus (rVSV)-based vaccines could likewise confer rapid protection is unclear., Methods: Here, we tested the ability of an attenuated, quadrivalent panfilovirus VesiculoVax vaccine (rVSV-Filo) to elicit fast-acting protection against MARV, EBOV, SUDV, and BDBV. Groups of cynomolgus monkeys were vaccinated 7 days before exposure to each of the 4 viral pathogens. All subjects (100%) immunized 1 week earlier survived MARV, SUDV, and BDBV challenge; 80% survived EBOV challenge. Survival correlated with lower viral load, higher glycoprotein-specific immunoglobulin G titers, and the expression of B-cell-, cytotoxic cell-, and antigen presentation-associated transcripts., Conclusions: These results demonstrate multivalent VesiculoVax vaccines are suitable for filovirus outbreak management. The highly attenuated nature of the rVSV-Filo vaccine may be preferable to the Ervebo "delta G" platform, which induced adverse events in a subset of recipients., Competing Interests: Conflict of interest. C. G., N. M., and D. M. are employees of Auro Vaccines. J. E. H. is a former employee of Auro Vaccines. T. W. G. claims intellectual property regarding recombinant vesicular stomatitis virus–based vaccines for the prevention and treatment of filovirus infections. All other authors report no potential conflicts. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
17. Pathogenesis of Aerosolized Ebola Virus Variant Makona in Nonhuman Primates.
- Author
-
Prasad AN, Fenton KA, Agans KN, Borisevich V, Woolsey C, Comer JE, Dobias NS, Peel JE, Deer DJ, Geisbert JB, Lawrence WS, Cross RW, and Geisbert TW
- Subjects
- Humans, Animals, Macaca fascicularis, Aerosols, Viral Load, Ebolavirus, Hemorrhagic Fever, Ebola
- Abstract
Background: Highly pathogenic filoviruses such as Ebola virus (EBOV) hold capacity for delivery by artificial aerosols, and thus potential for intentional misuse. Previous studies have shown that high doses of EBOV delivered by small-particle aerosol cause uniform lethality in nonhuman primates (NHPs), whereas only a few small studies have assessed lower doses in NHPs., Methods: To further characterize the pathogenesis of EBOV infection via small-particle aerosol, we challenged cohorts of cynomolgus monkeys with low doses of EBOV variant Makona, which may help define risks associated with small particle aerosol exposures., Results: Despite using challenge doses orders of magnitude lower than previous studies, infection via this route was uniformly lethal across all cohorts. Time to death was delayed in a dose-dependent manner between aerosol-challenged cohorts, as well as in comparison to animals challenged via the intramuscular route. Here, we describe the observed clinical and pathological details including serum biomarkers, viral burden, and histopathological changes leading to death., Conclusions: Our observations in this model highlight the striking susceptibility of NHPs, and likely humans, via small-particle aerosol exposure to EBOV and emphasize the need for further development of diagnostics and postexposure prophylactics in the event of intentional release via deployment of an aerosol-producing device., Competing Interests: Potential conflicts of interest. All authors: No reported conflicts of interest., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
18. Limited Benefit of Postexposure Prophylaxis With VSV-EBOV in Ebola Virus-Infected Rhesus Macaques.
- Author
-
Bushmaker T, Feldmann F, Lovaglio J, Saturday G, Griffin AJ, O'Donnell KL, Strong JE, Sprecher A, Kobinger G, Geisbert TW, Marzi A, and Feldmann H
- Subjects
- Humans, Animals, Macaca mulatta, Vesiculovirus, Vesicular stomatitis Indiana virus, Ebolavirus, Hemorrhagic Fever, Ebola, Ebola Vaccines
- Abstract
Vesicular stomatitis virus-Ebola virus (VSV-EBOV) vaccine has been successfully used in ring vaccination approaches during EBOV disease outbreaks demonstrating its general benefit in short-term prophylactic vaccination, but actual proof of its benefit in true postexposure prophylaxis (PEP) for humans is missing. Animal studies have indicated PEP efficacy when VSV-EBOV was used within hours of lethal EBOV challenge. Here, we used a lower EBOV challenge dose and a combined intravenous and intramuscular VSV-EBOV administration to improve PEP efficacy in the rhesus macaque model. VSV-EBOV treatment 1 hour after EBOV challenge resulted in delayed disease progression but little benefit in outcome. Thus, we could not confirm previous results indicating questionable benefit of VSV-EBOV for EBOV PEP in a nonhuman primate model., Competing Interests: Potential conflicts of interest. H. F. and T. W. G. claim intellectual property for vesicular stomatitis virus–based filovirus vaccines. All other authors report no potential conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (Published by Oxford University Press on behalf of Infectious Diseases Society of America 2023.)
- Published
- 2023
- Full Text
- View/download PDF
19. Perspectives on Advancing Countermeasures for Filovirus Disease: Report From a Multisector Meeting.
- Author
-
Sprecher A, Cross R, Marzi A, Martins KA, Wolfe D, Montgomery JM, Spiropoulou CF, Cihlar T, Ahuka-Mundeke S, Nyhuis T, Teicher C, Crozier I, Strong J, Kobinger G, Woolsey C, Geisbert TW, Feldmann H, and Muyembe JJ
- Subjects
- Humans, Disease Outbreaks prevention & control, Africa, Filoviridae, Hemorrhagic Fever, Ebola prevention & control, Hemorrhagic Fever, Ebola epidemiology, Filoviridae Infections, Ebolavirus
- Abstract
Although there are now approved treatments and vaccines for Ebola virus disease, the case fatality rate remains unacceptably high even when patients are treated with the newly approved therapeutics. Furthermore, these countermeasures are not expected to be effective against disease caused by other filoviruses. A meeting of subject-matter experts was held during the 10th International Filovirus Symposium to discuss strategies to address these gaps. Several investigational therapeutics, vaccine candidates, and combination strategies were presented. The greatest challenge was identified to be the implementation of well-designed clinical trials of safety and efficacy during filovirus disease outbreaks. Preparing for this will require agreed-upon common protocols for trials intended to bridge multiple outbreaks across all at-risk countries. A multinational research consortium including at-risk countries would be an ideal mechanism to negotiate agreement on protocol design and coordinate preparation. Discussion participants recommended a follow-up meeting be held in Africa to establish such a consortium., Competing Interests: Potential conflicts of interest. T. C. is a full-time employee of Gilead Sciences and holds stock or stock options in the company. T. N. holds stock or stock options in Mapp Biopharmaceutical. H. F. and T. W. G. claim intellectual property for VSV-based viral hemorrhagic fever vaccines. G. K. has received grants or contracts from the Defense Advanced Research Projects Agency for a phase 1 clinical trial of an Ebola vaccine boost (ended in March 2023). All other authors report no potential conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (Published by Oxford University Press on behalf of Infectious Diseases Society of America 2023.)
- Published
- 2023
- Full Text
- View/download PDF
20. Long-term Prophylaxis Against Aerosolized Marburg Virus in Nonhuman Primates With an Afucosylated Monoclonal Antibody.
- Author
-
Abelson D, Barajas J, Stuart L, Kim D, Marimuthu A, Hu C, Yamamoto B, Ailor E, Whaley KJ, Vu H, Agans KN, Borisevich V, Deer DJ, Dobias NS, Woolsey C, Prasad AN, Peel JE, Lawrence WS, Cross RW, Geisbert TW, Fenton KA, and Zeitlin L
- Subjects
- Animals, Humans, Antibodies, Monoclonal, Primates, Aerosols, Marburgvirus, Marburg Virus Disease
- Abstract
Marburg virus (MARV) causes a hemorrhagic fever disease in human and nonhuman primates with high levels of morbidity and mortality. Concerns about weaponization of aerosolized MARV have spurred the development of nonhuman primate (NHP) models of aerosol exposure. To address the potential threat of aerosol exposure, a monoclonal antibody that binds MARV glycoprotein was tested, MR186YTE, for its efficacy as a prophylactic. MR186YTE was administered intramuscularly to NHPs at 15 or 5 mg/kg 1 month prior to MARV aerosol challenge. Seventy-five percent (3/4) of the 15 mg/kg dose group and 50% (2/4) of the 5 mg/kg dose group survived. Serum analyses showed that the NHP dosed with 15 mg/kg that succumbed to infection developed an antidrug antibody response and therefore had no detectable MR186YTE at the time of challenge. These results suggest that intramuscular dosing of mAbs may be a clinically useful prophylaxis for MARV aerosol exposure., Competing Interests: Potential conflicts of interest. D. A., J. B., L. S., D. K., A. M., C. H., E. A., and B. Y. are employees of Mapp. L. Z. and K. W. are employees and owners of Mapp. H. V. is an employee of Integrated Biotherapeutics. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
21. Natural History of Nonhuman Primates After Oral Exposure to Ebola Virus Variant Makona.
- Author
-
Prasad AN, Agans KN, Geisbert JB, Borisevich V, Deer DJ, Dobias NS, Comer JE, Woolsey C, Fenton KA, Geisbert TW, and Cross RW
- Subjects
- Humans, Animals, Disease Models, Animal, Viremia, Macaca fascicularis, Biomarkers, Ebolavirus, Hemorrhagic Fever, Ebola
- Abstract
Background: The primary route of infection by Ebola virus (EBOV) is through contact of mucosal surfaces. Few studies have explored infection of nonhuman primates (NHPs) via the oral mucosa, which is a probable portal of natural infection in humans., Methods: To further characterize the pathogenesis of EBOV infection via the oral exposure route, we challenged cohorts of cynomolgus monkeys with low doses of EBOV variant Makona., Results: Infection with 100 or 50 PFU of EBOV Makona via the oral route resulted in 50% and 83% lethality, respectively. Animals that progressed to fatal disease exhibited lymphopenia, marked coagulopathy, high viral loads, and increased levels of serum markers of inflammation and hepatic/renal injury. Survival in these cohorts was associated with milder fluctuations in leukocyte populations, lack of coagulopathy, and reduced or absent serum markers of inflammation and/or hepatic/renal function. Surprisingly, 2 surviving animals from the 100- and 50-PFU cohorts developed transient low-level viremia in the absence of other clinical signs of disease. Conversely, all animals in the 10 PFU cohort remained disease free and survived to the study end point., Conclusions: Our observations highlight the susceptibility of NHPs, and by extension, likely humans, to relatively low doses of EBOV via the oral route., Competing Interests: Potential conflicts of interest. All authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
22. Revisiting the minimum incubation period of Zaire ebolavirus.
- Author
-
Kofman AD, Haberling DL, Mbuyi G, Martel LD, Whitesell AN, Van Herp M, Makaya G, Corvil S, Abedi AA, Ngoma PM, Mbuyi F, Mossoko M, Koivogui E, Soke N, Gbamou N, Fonjungo PN, Keita L, Keita S, Shoemaker TR, Richards GA, Montgomery JM, Breman JG, Geisbert TW, Choi MJ, and Rollin PE
- Abstract
Competing Interests: JGB was president of the American Society of Tropical Medicine and Hygiene from 2019–2020. All other authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
23. Animal Models for Henipavirus Research.
- Author
-
Pigeaud DD, Geisbert TW, and Woolsey C
- Subjects
- Cricetinae, Animals, Humans, Guinea Pigs, Horses, Mice, Dogs, Ferrets, Disease Models, Animal, Primates, Henipavirus Infections, Hendra Virus, Nipah Virus
- Abstract
Hendra virus (HeV) and Nipah virus (NiV) are zoonotic paramyxoviruses in the genus Henipavirus (HNV) that emerged nearly thirty years ago. Outbreaks of HeV and NiV have led to severe respiratory disease and encephalitis in humans and animals characterized by a high mortality rate. Despite the grave threat HNVs pose to public health and global biosecurity, no approved medical countermeasures for human use currently exist against HeV or NiV. To develop candidate vaccines and therapeutics and advance the field's understanding of HNV pathogenesis, animal models of HeV and NiV have been instrumental and remain indispensable. Various species, including rodents, ferrets, and nonhuman primates (NHPs), have been employed for HNV investigations. Among these, NHPs have demonstrated the closest resemblance to human HNV disease, although other animal models replicate some key disease features. Here, we provide a comprehensive review of the currently available animal models (mice, hamsters, guinea pigs, ferrets, cats, dogs, nonhuman primates, horses, and swine) to support HNV research. We also discuss the strengths and limitations of each model for conducting pathogenesis and transmission studies on HeV and NiV and for the evaluation of medical countermeasures.
- Published
- 2023
- Full Text
- View/download PDF
24. A human monoclonal antibody combination rescues nonhuman primates from advanced disease caused by the major lineages of Lassa virus.
- Author
-
Cross RW, Heinrich ML, Fenton KA, Borisevich V, Agans KN, Prasad AN, Woolsey C, Deer DJ, Dobias NS, Rowland MM, Lathigra R, Borrega R, Geisbert JB, Garry RF, Branco LM, and Geisbert TW
- Subjects
- Animals, Humans, Africa, Western, Antibodies, Monoclonal, Antibodies, Neutralizing, Macaca fascicularis, Lassa virus, Lassa Fever prevention & control
- Abstract
There are no approved treatments for Lassa fever (LF), which is responsible for thousands of deaths each year in West Africa. A major challenge in developing effective medical countermeasures against LF is the high diversity of circulating Lassa virus (LASV) strains with four recognized lineages and four proposed lineages. The recent resurgence of LASV in Nigeria caused by genetically distinct strains underscores this concern. Two LASV lineages (II and III) are dominant in Nigeria. Here, we show that combinations of two or three pan-lineage neutralizing human monoclonal antibodies (8.9F, 12.1F, 37.D) known as Arevirumab-2 or Arevirumab-3 can protect up to 100% of cynomolgus macaques against challenge with both lineage II and III LASV isolates when treatment is initiated at advanced stages of disease on day 8 after LASV exposure. This work demonstrates that it may be possible to develop postexposure interventions that can broadly protect against most strains of LASV.
- Published
- 2023
- Full Text
- View/download PDF
25. Alpha-1-antitrypsin and its variant-dependent role in COVID-19 pathogenesis.
- Author
-
Stevens CS, Oguntuyo KY, Kowdle S, Brambilla L, Haas G, Gowlikar A, Siddiquey MN, Schilke RM, Woolard MD, Zhang H, Acklin JA, Ikegame S, Huang CT, Lim JK, Cross RW, Geisbert TW, Ivanov SS, Kamil JP, and Lee B
- Abstract
Rationale: SARS-CoV-2 entry into host cells is facilitated by endogenous and exogenous proteases that proteolytically activate the spike glycoprotein and antiproteases inhibiting this process. Understanding the key actors in viral entry is crucial for advancing knowledge of virus tropism, pathogenesis, and potential therapeutic targets., Objectives: We aimed to investigate the role of naïve serum and alpha-1-antitrypsin (AAT) in inhibiting protease-mediated SARS-CoV-2 entry and explore the implications of AAT deficiency on susceptibility to different SARS-CoV-2 variants., Findings: Our study demonstrates that naïve serum exhibits significant inhibition of SARS-CoV-2 entry, with AAT identified as the major serum protease inhibitor potently restricting entry. Using pseudoparticles, replication-competent pseudoviruses, and authentic SARS-CoV-2, we show that AAT inhibition occurs at low concentrations compared with those in serum and bronchoalveolar tissues, suggesting physiological relevance. Furthermore, sera from subjects with an AAT-deficient genotype show reduced ability to inhibit entry of both Wuhan-Hu-1 (WT) and B.1.617.2 (Delta) but exhibit no difference in inhibiting B.1.1.529 (Omicron) entry., Conclusions: AAT may have a variant-dependent therapeutic potential against SARS-CoV-2. Our findings highlight the importance of further investigating the complex interplay between proteases, antiproteases, and spike glycoprotein activation in SARS-CoV-2 and other respiratory viruses to identify potential therapeutic targets and improve understanding of disease pathogenesis.
- Published
- 2023
- Full Text
- View/download PDF
26. A Recombinant Chimeric Cedar Virus-Based Surrogate Neutralization Assay Platform for Pathogenic Henipaviruses.
- Author
-
Amaya M, Yin R, Yan L, Borisevich V, Adhikari BN, Bennett A, Malagon F, Cer RZ, Bishop-Lilly KA, Dimitrov AS, Cross RW, Geisbert TW, and Broder CC
- Subjects
- Humans, Animals, Viral Envelope Proteins genetics, Glycoproteins genetics, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Henipavirus Infections, Hendra Virus genetics, Nipah Virus genetics
- Abstract
The henipaviruses, Nipah virus (NiV), and Hendra virus (HeV) can cause fatal diseases in humans and animals, whereas Cedar virus is a nonpathogenic henipavirus. Here, using a recombinant Cedar virus (rCedV) reverse genetics platform, the fusion (F) and attachment (G) glycoprotein genes of rCedV were replaced with those of NiV-Bangladesh (NiV-B) or HeV, generating replication-competent chimeric viruses (rCedV-NiV-B and rCedV-HeV), both with and without green fluorescent protein (GFP) or luciferase protein genes. The rCedV chimeras induced a Type I interferon response and utilized only ephrin-B2 and ephrin-B3 as entry receptors compared to rCedV. The neutralizing potencies of well-characterized cross-reactive NiV/HeV F and G specific monoclonal antibodies against rCedV-NiV-B-GFP and rCedV-HeV-GFP highly correlated with measurements obtained using authentic NiV-B and HeV when tested in parallel by plaque reduction neutralization tests (PRNT). A rapid, high-throughput, and quantitative fluorescence reduction neutralization test (FRNT) using the GFP-encoding chimeras was established, and monoclonal antibody neutralization data derived by FRNT highly correlated with data derived by PRNT. The FRNT assay could also measure serum neutralization titers from henipavirus G glycoprotein immunized animals. These rCedV chimeras are an authentic henipavirus-based surrogate neutralization assay that is rapid, cost-effective, and can be utilized outside high containment.
- Published
- 2023
- Full Text
- View/download PDF
27. Natural history of nonhuman primates after conjunctival exposure to Ebola virus.
- Author
-
Cross RW, Prasad AN, Woolsey CB, Agans KN, Borisevich V, Dobias NS, Comer JE, Deer DJ, Geisbert JB, Rasmussen AL, Lipkin WI, Fenton KA, and Geisbert TW
- Subjects
- Animals, Macaca fascicularis, Conjunctiva, Primates, Ebolavirus, Hemorrhagic Fever, Ebola
- Abstract
Transmission of Ebola virus (EBOV) primarily occurs via contact exposure of mucosal surfaces with infected body fluids. Historically, nonhuman primate (NHP) challenge studies have employed intramuscular (i.m.) or small particle aerosol exposure, which are largely lethal routes of infection, but mimic worst-case scenarios such as a needlestick or intentional release, respectively. When exposed by more likely routes of natural infection, limited NHP studies have shown delayed onset of disease and reduced mortality. Here, we performed a series of systematic natural history studies in cynomolgus macaques with a range of conjunctival exposure doses. Challenge with 10,000 plaque forming units (PFU) of EBOV was uniformly lethal, whereas 5/6 subjects survived lower dose challenges (100 or 500 PFU). Conjunctival challenge resulted in a protracted time-to death compared to i.m. Asymptomatic infection was observed in survivors with limited detection of EBOV replication. Inconsistent seropositivity in survivors may suggest physical or natural immunological barriers are sufficient to prevent widespread viral dissemination., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
28. Combination treatment of mannose and GalNAc conjugated small interfering RNA protects against lethal Marburg virus infection.
- Author
-
Ye X, Holland R, Wood M, Pasetka C, Palmer L, Samaridou E, McClintock K, Borisevich V, Geisbert TW, Cross RW, and Heyes J
- Published
- 2023
- Full Text
- View/download PDF
29. Recombinant vesicular stomatitis virus-vectored vaccine induces long-lasting immunity against Nipah virus disease.
- Author
-
Woolsey C, Borisevich V, Fears AC, Agans KN, Deer DJ, Prasad AN, O'Toole R, Foster SL, Dobias NS, Geisbert JB, Fenton KA, Cross RW, and Geisbert TW
- Subjects
- Animals, Humans, Chlorocebus aethiops, Antibodies, Viral, Vesiculovirus genetics, Nipah Virus genetics, Viral Vaccines genetics, Vesicular Stomatitis, COVID-19
- Abstract
The emergence of the novel henipavirus, Langya virus, received global attention after the virus sickened over three dozen people in China. There is heightened concern that henipaviruses, as respiratory pathogens, could spark another pandemic, most notably the deadly Nipah virus (NiV). NiV causes near-annual outbreaks in Bangladesh and India and induces a highly fatal respiratory disease and encephalitis in humans. No licensed countermeasures against this pathogen exist. An ideal NiV vaccine would confer both fast-acting and long-lived protection. Recently, we reported the generation of a recombinant vesicular stomatitis virus-based (rVSV-based) vaccine expressing the NiV glycoprotein (rVSV-ΔG-NiVBG) that protected 100% of nonhuman primates from NiV-associated lethality within a week. Here, to evaluate the durability of rVSV-ΔG-NiVBG, we vaccinated African green monkeys (AGMs) one year before challenge with an uniformly lethal dose of NiV. The rVSV-ΔG-NiVBG vaccine induced stable and robust humoral responses, whereas cellular responses were modest. All immunized AGMs (whether receiving a single dose or prime-boosted) survived with no detectable clinical signs or NiV replication. Transcriptomic analyses indicated that adaptive immune signatures correlated with vaccine-mediated protection. While vaccines for certain respiratory infections (e.g., COVID-19) have yet to provide durable protection, our results suggest that rVSV-ΔG-NiVBG elicits long-lasting immunity.
- Published
- 2023
- Full Text
- View/download PDF
30. Marburg and Ebola Virus Infections Elicit a Complex, Muted Inflammatory State in Bats.
- Author
-
Jayaprakash AD, Ronk AJ, Prasad AN, Covington MF, Stein KR, Schwarz TM, Hekmaty S, Fenton KA, Geisbert TW, Basler CF, Bukreyev A, and Sachidanandam R
- Subjects
- Humans, Animals, Liver, Hemorrhagic Fever, Ebola veterinary, Chiroptera, Ebolavirus genetics, Filoviridae Infections, Marburgvirus genetics
- Abstract
The Marburg and Ebola filoviruses cause a severe, often fatal, disease in humans and nonhuman primates but have only subclinical effects in bats, including Egyptian rousettes, which are a natural reservoir of Marburg virus. A fundamental question is why these viruses are highly pathogenic in humans but fail to cause disease in bats. To address this question, we infected one cohort of Egyptian rousette bats with Marburg virus and another cohort with Ebola virus and harvested multiple tissues for mRNA expression analysis. While virus transcripts were found primarily in the liver, principal component analysis (PCA) revealed coordinated changes across multiple tissues. Gene signatures in kidney and liver pointed at induction of vasodilation, reduction in coagulation, and changes in the regulation of iron metabolism. Signatures of immune response detected in spleen and liver indicated a robust anti-inflammatory state signified by macrophages in the M2 state and an active T cell response. The evolutionary divergence between bats and humans of many responsive genes might provide a framework for understanding the differing outcomes upon infection by filoviruses. In this study, we outline multiple interconnected pathways that respond to infection by MARV and EBOV, providing insights into the complexity of the mechanisms that enable bats to resist the disease caused by filoviral infections. The results have the potential to aid in the development of new strategies to effectively mitigate and treat the disease caused by these viruses in humans.
- Published
- 2023
- Full Text
- View/download PDF
31. Nonhuman Primate Models for Nipah and Hendra Virus Countermeasure Evaluation.
- Author
-
Mire CE, Satterfield BA, and Geisbert TW
- Subjects
- Animals, Humans, Antiviral Agents pharmacology, Australia, Disease Outbreaks, Primates, Hendra Virus
- Abstract
Hendra and Nipah viruses are henipaviruses that have caused lethal human disease in Australia and Malaysia, Bangladesh, India, and the Philippines, respectively. These viruses are considered Category C pathogens by the US Centers for Disease Control. Nipah virus was recently placed on the World Health Organization Research and Development Blueprint Roadmaps for vaccine and therapeutic development. Given the infrequent and unpredictable nature of henipavirus outbreaks licensure of vaccines and therapeutics will likely require an animal model to demonstrate protective efficacy against henipavirus disease. Studies have shown that nonhuman primates are the most accurate model of human henipavirus disease and would be an important component of any application for licensure of a vaccine or antiviral drug under the US FDA Animal Rule. Nonhuman primate model selection and dosing are discussed regarding vaccine and therapeutic studies against henipaviruses., (© 2023. Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
32. Overview of Experimental Vaccines and Antiviral Therapeutics for Henipavirus Infection.
- Author
-
Satterfield BA, Mire CE, and Geisbert TW
- Subjects
- Humans, Animals, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Australia, Disease Outbreaks, Henipavirus Infections drug therapy, Henipavirus Infections prevention & control, Chiroptera
- Abstract
Hendra virus (HeV) and Nipah virus (NiV) are highly pathogenic paramyxoviruses, which have emerged in recent decades and cause sporadic outbreaks of respiratory and encephalitic disease in Australia and Southeast Asia, respectively. Over two billion people currently live in regions potentially at risk due to the wide range of the Pteropus fruit bat reservoir, yet there are no approved vaccines or therapeutics to protect against or treat henipavirus disease. In recent years, significant progress has been made toward developing various experimental vaccine platforms and therapeutics. Here, we describe these advances for both human and livestock vaccine candidates and discuss the numerous preclinical studies and the few that have progressed to human phase 1 clinical trial and the one approved veterinary vaccine., (© 2023. Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
33. A single-shot ChAd3-MARV vaccine confers rapid and durable protection against Marburg virus in nonhuman primates.
- Author
-
Hunegnaw R, Honko AN, Wang L, Carr D, Murray T, Shi W, Nguyen L, Storm N, Dulan CNM, Foulds KE, Agans KN, Cross RW, Geisbert JB, Cheng C, Ploquin A, Stanley DA, Geisbert TW, Nabel GJ, and Sullivan NJ
- Subjects
- Animals, Humans, Pan troglodytes, Primates, Adenoviridae, Marburgvirus, Ebolavirus, Ebola Vaccines, Marburg Virus Disease prevention & control, Hemorrhagic Fever, Ebola
- Abstract
Marburg virus (MARV) causes a severe hemorrhagic fever disease in primates with mortality rates in humans of up to 90%. MARV has been identified as a category A bioterrorism agent by the Centers for Disease Control and Prevention (CDC) and priority pathogen A by the National Institute of Allergy and Infectious Diseases (NIAID), needing urgent research and development of countermeasures because of the high public health risk it poses. The recent cases of MARV in West Africa underscore the substantial outbreak potential of this virus. The potential for cross-border spread, as had occurred during the 2014-2016 Ebola virus outbreak, illustrates the critical need for MARV vaccines. To support regulatory approval of the chimpanzee adenovirus 3 (ChAd3)-MARV vaccine that has completed phase 1 trials, we showed that the nonreplicating ChAd3 vector, which has a demonstrated safety profile in humans, protected against a uniformly lethal challenge with MARV/Ang. Protective immunity was achieved within 7 days of vaccination and was maintained through 1 year after vaccination. Antigen-specific antibodies were an immune correlate of protection in the acute challenge model, and their concentration was predictive of protection. These results demonstrate that a single-shot ChAd3-MARV vaccine generated a protective immune response that was both rapid and durable with an immune correlate of protection that will support advanced clinical development.
- Published
- 2022
- Full Text
- View/download PDF
34. Lessons from the pandemic: Responding to emerging zoonotic viral diseases-a Keystone Symposia report.
- Author
-
Cable J, Fauci A, Dowling WE, Günther S, Bente DA, Yadav PD, Madoff LC, Wang LF, Arora RK, Van Kerkhove M, Chu MC, Jaenisch T, Epstein JH, Frost SDW, Bausch DG, Hensley LE, Bergeron É, Sitaras I, Gunn MD, Geisbert TW, Muñoz-Fontela C, Krammer F, de Wit E, Nordenfelt P, Saphire EO, Gilbert SC, Corbett KS, Branco LM, Baize S, van Doremalen N, Krieger MA, Clemens SAC, Hesselink R, and Hartman D
- Subjects
- Humans, Pandemics, Disease Outbreaks, COVID-19 epidemiology, Ebolavirus
- Abstract
The COVID-19 pandemic caught the world largely unprepared, including scientific and policy communities. On April 10-13, 2022, researchers across academia, industry, government, and nonprofit organizations met at the Keystone symposium "Lessons from the Pandemic: Responding to Emerging Zoonotic Viral Diseases" to discuss the successes and challenges of the COVID-19 pandemic and what lessons can be applied moving forward. Speakers focused on experiences not only from the COVID-19 pandemic but also from outbreaks of other pathogens, including the Ebola virus, Lassa virus, and Nipah virus. A general consensus was that investments made during the COVID-19 pandemic in infrastructure, collaborations, laboratory and manufacturing capacity, diagnostics, clinical trial networks, and regulatory enhancements-notably, in low-to-middle income countries-must be maintained and strengthened to enable quick, concerted responses to future threats, especially to zoonotic pathogens., (© 2022 New York Academy of Sciences.)
- Published
- 2022
- Full Text
- View/download PDF
35. Natural history of Sudan ebolavirus infection in rhesus and cynomolgus macaques.
- Author
-
Woolsey C, Fears AC, Borisevich V, Agans KN, Dobias NS, Prasad AN, Deer DJ, Geisbert JB, Fenton KA, Geisbert TW, and Cross RW
- Subjects
- Animals, Macaca fascicularis, Macaca mulatta, Uganda, Ebolavirus genetics, Hemorrhagic Fever, Ebola
- Abstract
Due to its high mortality rate and continued re-emergence, Ebolavirus disease (EVD) continues to pose a serious threat to global health. A group of viruses within the genus Ebolavirus causes this severe hemorrhagic disease in humans: Ebola virus (EBOV; species Zaire ebolavirus ), Sudan virus (SUDV; species Sudan ebolavirus ), Bundibugyo virus, and Taï Forest virus. EBOV and SUDV are associated with the highest case fatality rates. While the host response to EBOV has been comprehensively examined, limited data exists for SUDV infection. For medical countermeasure testing, well-characterized SUDV nonhuman primate (NHP) models are thus needed. Here, we describe a natural history study in which rhesus ( N = 11) and cynomolgus macaques ( N = 14) were intramuscularly exposed to a 1000 plaque-forming unit dose of SUDV (Gulu variant). Time-course analyses of various hematological, pathological, serological, coagulation, and transcriptomic findings are reported. SUDV infection was uniformly lethal in cynomolgus macaques (100% mortality), whereas a single rhesus macaque subject (91% mortality) survived to the study endpoint (median time-to-death of ∼8.0 and ∼8.5 days in cynomolgus and rhesus macaques, respectively). Infected macaques exhibited hallmark features of human EVD. The early stage was typified by viremia, granulocytosis, lymphopenia, albuminemia, thrombocytopenia, and decreased expression of HLA-class transcripts. At mid-to-late disease, animals developed fever and petechial rashes, and expressed high levels of pro-inflammatory mediators, pro-thrombotic factors, and markers indicative of liver and kidney injury. End-stage disease was characterized by shock and multi-organ failure. In summary, macaques recapitulate human SUDV disease, supporting these models for use in the development of vaccines and therapeutics.
- Published
- 2022
- Full Text
- View/download PDF
36. An introduction to the Marburg virus vaccine consortium, MARVAC.
- Author
-
Cross RW, Longini IM, Becker S, Bok K, Boucher D, Carroll MW, Díaz JV, Dowling WE, Draghia-Akli R, Duworko JT, Dye JM, Egan MA, Fast P, Finan A, Finch C, Fleming TR, Fusco J, Geisbert TW, Griffiths A, Günther S, Hensley LE, Honko A, Hunegnaw R, Jakubik J, Ledgerwood J, Luhn K, Matassov D, Meshulam J, Nelson EV, Parks CL, Rustomjee R, Safronetz D, Schwartz LM, Smith D, Smock P, Sow Y, Spiropoulou CF, Sullivan NJ, Warfield KL, Wolfe D, Woolsey C, Zahn R, Henao-Restrepo AM, Muñoz-Fontela C, and Marzi A
- Subjects
- Animals, Humans, Marburg Virus Disease prevention & control, Marburgvirus, Viral Vaccines
- Abstract
The emergence of Marburg virus (MARV) in Guinea and Ghana triggered the assembly of the MARV vaccine "MARVAC" consortium representing leaders in the field of vaccine research and development aiming to facilitate a rapid response to this infectious disease threat. Here, we discuss current progress, challenges, and future directions for MARV vaccines., Competing Interests: T.W.G. holds U.S. patent number 7,635,485 issued to US Government, U.S. patent number 7,838,658 issued to Arbutus Biopharma, U.S. patent number 8,017,130 issued to US Government, U.S. patent number 8,716,464 issued to Arbutus Biopharma, and U.S. patent number 8,796,013 issued to Boston University. N.J.S. has a patent for Chimpanzee Adenoviral vector-based filovirus vaccines (for use in humans) with royalties paid to GlaxoSmithKline Biologicals. K.L.W. is a stockholder of Emergent BioSolutions. R.Z. holds a patent for “Methods and compositions for inducing protective immunity against Marburg virus infection”. All other authors declare no conflict of interest. The conclusions in this report are those of the authors and do not necessarily represent the official position of the U.S. Centers for Disease Control and Prevention.
- Published
- 2022
- Full Text
- View/download PDF
37. Nonhuman Primates Are Protected against Marburg Virus Disease by Vaccination with a Vesicular Stomatitis Virus Vector-Based Vaccine Prepared under Conditions to Allow Advancement to Human Clinical Trials.
- Author
-
Cooper CL, Morrow G, Yuan M, Coleman JW, Hou F, Reiserova L, Li SL, Wagner D, Carpov A, Wallace-Selman O, Valentin K, Choi Y, Wilson A, Kilianski A, Sayeed E, Agans KN, Borisevich V, Cross RW, Geisbert TW, Feinberg MB, Gupta SB, and Parks CL
- Abstract
Vaccines are needed to disrupt or prevent continued outbreaks of filoviruses in humans across Western and Central Africa, including outbreaks of Marburg virus (MARV). As part of a filovirus vaccine product development plan, it is important to investigate dose response early in preclinical development to identify the dose range that may be optimal for safety, immunogenicity, and efficacy, and perhaps demonstrate that using lower doses is feasible, which will improve product access. To determine the efficacious dose range for a manufacturing-ready live recombinant vesicular stomatitis virus vaccine vector (rVSV∆G-MARV-GP) encoding the MARV glycoprotein (GP), a dose-range study was conducted in cynomolgus macaques. Results showed that a single intramuscular injection with as little as 200 plaque-forming units (PFUs) was 100% efficacious against lethality and prevented development of viremia and clinical pathologies associated with MARV Angola infection. Across the vaccine doses tested, there was nearly a 2000-fold range of anti-MARV glycoprotein (GP) serum IgG titers with seroconversion detectable even at the lowest doses. Virus-neutralizing serum antibodies also were detected in animals vaccinated with the higher vaccine doses indicating that vaccination induced functional antibodies, but that the assay was a less sensitive indicator of seroconversion. Collectively, the data indicates that a relatively wide range of anti-GP serum IgG titers are observed in animals that are protected from disease implying that seroconversion is positively associated with efficacy, but that more extensive immunologic analyses on samples collected from our study as well as future preclinical studies will be valuable in identifying additional immune responses correlated with protection that can serve as markers to monitor in human trials needed to generate data that can support vaccine licensure in the future.
- Published
- 2022
- Full Text
- View/download PDF
38. A recombinant VSV-vectored vaccine rapidly protects nonhuman primates against heterologous lethal Lassa fever.
- Author
-
Cross RW, Woolsey C, Prasad AN, Borisevich V, Agans KN, Deer DJ, Geisbert JB, Dobias NS, Fenton KA, and Geisbert TW
- Subjects
- Animals, Humans, Lassa virus, Macaca fascicularis, Vaccines, Synthetic, Lassa Fever prevention & control, Viral Vaccines
- Abstract
Lassa virus (LASV) is recognized by the World Health Organization as one of the top five pathogens likely to cause a severe outbreak. A recent unprecedented resurgence of LASV in Nigeria caused by genetically diverse strains underscores the need for licensed medical countermeasures. Single-injection vaccines that can rapidly control outbreaks and confer long-term immunity are needed. Vaccination of cynomolgus monkeys with a recombinant vesicular stomatitis virus vector expressing the glycoprotein precursor of LASV lineage IV strain Josiah (rVSVΔG-LASV-GPC) induces fast-acting protection in monkeys challenged 3 or 7 days later with a genetically heterologous lineage II isolate of LASV from Nigeria, while nonspecifically vaccinated control animals succumb to challenge. The rVSVΔG-LASV-GPC vaccine induces rapid activation of adaptive immunity and the transcription of natural killer (NK) cell-affiliated mRNAs. This study demonstrates that rVSVΔG-LASV-GPC may provide rapid protection in humans against LASV infections in cases where immediate public-health intervention is required., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
39. Reversion of Ebolavirus Disease from a Single Intramuscular Injection of a Pan-Ebolavirus Immunotherapeutic.
- Author
-
Kuang E, Cross RW, McCavitt-Malvido M, Abelson DM, Borisevich V, Stuart L, Agans KN, Mlakar N, Marimuthu A, Deer DJ, Shestowsky WS, Kim D, Geisbert JB, Zeitlin L, Moyer CL, Roy CJ, Geisbert TW, and Bornholdt ZA
- Abstract
Intravenous (IV) administration of antiviral monoclonal antibodies (mAbs) can be challenging, particularly during an ongoing epidemic, due to the considerable resources required for performing infusions. An ebolavirus therapeutic administered via intramuscular (IM) injection would reduce the burdens associated with IV infusion and allow rapid treatment of exposed individuals during an outbreak. Here, we demonstrate how MBP134, a cocktail of two pan-ebolavirus mAbs, reverses the course of Sudan ebolavirus disease (Gulu variant) with a single IV or IM dose in non-human primates (NHPs) as late as five days post-exposure. We also investigate the utility of adding half-life extension mutations to the MBP134 mAbs, ultimately creating a half-life extended cocktail designated MBP431. When delivered as a post-exposure prophylactic or therapeutic, a single IM dose of MBP431 offered complete or significant protection in NHPs challenged with Zaire ebolavirus . In conjunction with previous studies, these results support the use of MBP431 as a rapidly deployable IM medical countermeasure against every known species of ebolavirus.
- Published
- 2022
- Full Text
- View/download PDF
40. A highly attenuated Vesiculovax vaccine rapidly protects nonhuman primates against lethal Marburg virus challenge.
- Author
-
Woolsey C, Cross RW, Agans KN, Borisevich V, Deer DJ, Geisbert JB, Gerardi C, Latham TE, Fenton KA, Egan MA, Eldridge JH, Geisbert TW, and Matassov D
- Subjects
- Animals, Antibodies, Viral, Glycoproteins, Humans, Macaca fascicularis, Vaccines, Attenuated, Vesiculovirus genetics, Ebolavirus, Hemorrhagic Fever, Ebola, Marburg Virus Disease prevention & control, Marburgvirus, Viral Vaccines
- Abstract
Background: Marburg virus (MARV), an Ebola-like virus, remains an eminent threat to public health as demonstrated by its high associated mortality rate (23-90%) and recent emergence in West Africa for the first time. Although a recombinant vesicular stomatitis virus (rVSV)-based vaccine (Ervebo) is licensed for Ebola virus disease (EVD), no approved countermeasures exist against MARV. Results from clinical trials indicate Ervebo prevents EVD in 97.5-100% of vaccinees 10 days onwards post-immunization., Methodology/findings: Given the rapid immunogenicity of the Ervebo platform against EVD, we tested whether a similar, but highly attenuated, rVSV-based Vesiculovax vector expressing the glycoprotein (GP) of MARV (rVSV-N4CT1-MARV-GP) could provide swift protection against Marburg virus disease (MVD). Here, groups of cynomolgus monkeys were vaccinated 7, 5, or 3 days before exposure to a lethal dose of MARV (Angola variant). All subjects (100%) immunized one week prior to challenge survived; 80% and 20% of subjects survived when vaccinated 5- and 3-days pre-exposure, respectively. Lethality was associated with higher viral load and sustained innate immunity transcriptional signatures, whereas survival correlated with development of MARV GP-specific antibodies and early expression of predicted NK cell-, B-cell-, and cytotoxic T-cell-type quantities., Conclusions/significance: These results emphasize the utility of Vesiculovax vaccines for MVD outbreak management. The highly attenuated nature of rVSV-N4CT1 vaccines, which are clinically safe in humans, may be preferable to vaccines based on the same platform as Ervebo (rVSV "delta G" platform), which in some trial participants induced vaccine-related adverse events in association with viral replication including arthralgia/arthritis, dermatitis, and cutaneous vasculitis., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: The rVSV vector described in this manuscript is the subject of a patent licensed to Auro Vaccines. C.G., T.E.H., M.A.E., J.H.E., and D.M. are paid employees of Auro Vaccines, LLC. Auro Vaccines in-licenses a portfolio of patents previously obtained from Wyeth to support the development of Vesiculovax vectors as vaccines against hemorrhagic fever viruses. The other authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
41. Combination therapy with remdesivir and monoclonal antibodies protects nonhuman primates against advanced Sudan virus disease.
- Author
-
Cross RW, Bornholdt ZA, Prasad AN, Woolsey C, Borisevich V, Agans KN, Deer DJ, Abelson DM, Kim DH, Shestowsky WS, Campbell LA, Bunyan E, Geisbert JB, Dobias NS, Fenton KA, Porter DP, Zeitlin L, and Geisbert TW
- Subjects
- Adenosine Monophosphate analogs & derivatives, Alanine analogs & derivatives, Animals, Antibodies, Monoclonal, Antibodies, Viral, Humans, Macaca mulatta, Ebolavirus, Hemorrhagic Fever, Ebola drug therapy, Hemorrhagic Fever, Ebola prevention & control, Marburgvirus, Virus Diseases
- Abstract
A major challenge in managing acute viral infections is ameliorating disease when treatment is delayed. Previously, we reported the success of a 2-pronged mAb and antiviral remdesivir therapeutic approach to treat advanced illness in rhesus monkeys infected with Marburg virus (MARV). Here, we explored the benefit of a similar combination therapy for Sudan ebolavirus (Sudan virus; SUDV) infection. Importantly, no licensed anti-SUDV therapeutics currently exist, and infection of rhesus macaques with SUDV results in a rapid disease course similar to MARV with a mean time to death of 8.3 days. When initiation of therapy with either remdesivir or a pan-ebolavirus mAb cocktail (MBP431) was delayed until 6 days after inoculation, only 20% of macaques survived. In contrast, when remdesivir and MBP431 treatment were combined beginning 6 days after inoculation, significant protection (80%) was achieved. Our results suggest that combination therapy may be a viable treatment for patients with advanced filovirus disease that warrants further clinical testing in future outbreaks.
- Published
- 2022
- Full Text
- View/download PDF
42. LY-CoV1404 (bebtelovimab) potently neutralizes SARS-CoV-2 variants.
- Author
-
Westendorf K, Žentelis S, Wang L, Foster D, Vaillancourt P, Wiggin M, Lovett E, van der Lee R, Hendle J, Pustilnik A, Sauder JM, Kraft L, Hwang Y, Siegel RW, Chen J, Heinz BA, Higgs RE, Kallewaard NL, Jepson K, Goya R, Smith MA, Collins DW, Pellacani D, Xiang P, de Puyraimond V, Ricicova M, Devorkin L, Pritchard C, O'Neill A, Dalal K, Panwar P, Dhupar H, Garces FA, Cohen CA, Dye JM, Huie KE, Badger CV, Kobasa D, Audet J, Freitas JJ, Hassanali S, Hughes I, Munoz L, Palma HC, Ramamurthy B, Cross RW, Geisbert TW, Menachery V, Lokugamage K, Borisevich V, Lanz I, Anderson L, Sipahimalani P, Corbett KS, Yang ES, Zhang Y, Shi W, Zhou T, Choe M, Misasi J, Kwong PD, Sullivan NJ, Graham BS, Fernandez TL, Hansen CL, Falconer E, Mascola JR, Jones BE, and Barnhart BC
- Subjects
- Antibodies, Monoclonal chemistry, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Antibodies, Neutralizing chemistry, Antibodies, Neutralizing pharmacology, Antibodies, Neutralizing therapeutic use, Antibodies, Viral, Epitopes, Humans, SARS-CoV-2, COVID-19 Drug Treatment
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-neutralizing monoclonal antibodies (mAbs) can reduce the risk of hospitalization from coronavirus disease 2019 (COVID-19) when administered early. However, SARS-CoV-2 variants of concern (VOCs) have negatively affected therapeutic use of some authorized mAbs. Using a high-throughput B cell screening pipeline, we isolated LY-CoV1404 (bebtelovimab), a highly potent SARS-CoV-2 spike glycoprotein receptor binding domain (RBD)-specific antibody. LY-CoV1404 potently neutralizes authentic SARS-CoV-2, B.1.1.7, B.1.351, and B.1.617.2. In pseudovirus neutralization studies, LY-CoV1404 potently neutralizes variants, including B.1.1.7, B.1.351, B.1.617.2, B.1.427/B.1.429, P.1, B.1.526, B.1.1.529, and the BA.2 subvariant. Structural analysis reveals that the contact residues of the LY-CoV1404 epitope are highly conserved, except for N439 and N501. The binding and neutralizing activity of LY-CoV1404 is unaffected by the most common mutations at these positions (N439K and N501Y). The broad and potent neutralization activity and the relatively conserved epitope suggest that LY-CoV1404 has the potential to be an effective therapeutic agent to treat all known variants., Competing Interests: Declaration of interests Eli Lilly and Company provided resources for this study. AbCellera Biologics Inc. received funding from the U.S. Department of Defense, Defense Advanced Research Projects Agency (DARPA) Pandemic Prevention Platform, agreement D18AC00002. This research was funded in part by the U.S. Government (the views and conclusions contained in this document are those of the authors and should not be interpreted as representing the official policies, either expressed or implied, of the U.S. Government). This research used resources of the Advanced Photon Source, a U.S. Department of Energy (DOE) Office of Science User Facility operated for the DOE Office of Science by Argonne National Laboratory under contract DE-AC02-06CH11357 (https://www.aps.anl.gov/Science/Publications/Acknowledgment-Statement-for-Publications). Use of the Lilly Research Laboratories Collaborative Access Team (LRL-CAT) beamline at Sector 31 of the Advanced Photon Source was provided by Eli Lilly and Company, which operates the facility (http://lrlcat.lilly.com/). This work was supported by the Intramural Program at the National Institutes of Health, National Institute of Allergy and Infectious Diseases, Vaccine Research Center (to B.S.G. and J.R.M.). Operations support of the Galveston National Laboratory was supported by NIAID/NIH grant UC7AI094660. D.F., P.V., A.P., J.H., J.M.S., R.W.S., J.C., I. H., J.J.F., S.H., H.C.P., B.R., B.A.H., R.W.S., J.C., J.M.S., R.E.H., N.K., and B.E.J. are employees and/or stockholders of Eli Lilly and Company. K.W., S.Ž., M.W., E.L., L.K., Y.H., K.J., R.G., M.A.S., D.W.C., D.P., P.X., V.d.P., R.v.d.L., M.R., L.D., C.P., I.L., L.A., P.S., T.L.F., C.L.H., E.F., and B.C.B. are employees and stockholders of AbCellera Biologics Inc. AbCellera Biologics Inc. and the National Institutes of Health have filed patent applications related to the work described herein (US patent application 17/192243 and international patent application PCT/US21/20843, both titled “Anti-Coronavirus Antibodies and Methods of Use”)., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
43. A recombinant VSV-vectored vaccine rapidly protects nonhuman primates against lethal Nipah virus disease.
- Author
-
Foster SL, Woolsey C, Borisevich V, Agans KN, Prasad AN, Deer DJ, Geisbert JB, Dobias NS, Fenton KA, Cross RW, and Geisbert TW
- Subjects
- Animals, Antibodies, Neutralizing, Antibodies, Viral immunology, Biomarkers, Genetic Vectors, Kaplan-Meier Estimate, Neutralization Tests, Outcome Assessment, Health Care, Primate Diseases diagnosis, Primate Diseases mortality, Primate Diseases virology, Vaccination, Viral Load, Henipavirus Infections veterinary, Nipah Virus immunology, Primate Diseases prevention & control, Vaccines, Synthetic immunology, Viral Vaccines immunology
- Abstract
SignificanceConcern has increased about the pandemic potential of Nipah virus (NiV). Similar to SARS-CoV-2, NiV is an RNA virus that is transmitted by respiratory droplets. There are currently no NiV vaccines licensed for human use. While several preventive vaccines have shown promise in protecting animals against lethal NiV disease, most studies have assessed protection 1 mo after vaccination. However, in order to contain and control outbreaks, vaccines that can rapidly confer protection in days rather than months are needed. Here, we show that a recombinant vesicular stomatitis virus vector expressing the NiV glycoprotein can completely protect monkeys vaccinated 7 d prior to NiV exposure and 67% of animals vaccinated 3 d before NiV challenge.
- Published
- 2022
- Full Text
- View/download PDF
44. Asymmetric and non-stoichiometric glycoprotein recognition by two distinct antibodies results in broad protection against ebolaviruses.
- Author
-
Milligan JC, Davis CW, Yu X, Ilinykh PA, Huang K, Halfmann PJ, Cross RW, Borisevich V, Agans KN, Geisbert JB, Chennareddy C, Goff AJ, Piper AE, Hui S, Shaffer KCL, Buck T, Heinrich ML, Branco LM, Crozier I, Holbrook MR, Kuhn JH, Kawaoka Y, Glass PJ, Bukreyev A, Geisbert TW, Worwa G, Ahmed R, and Saphire EO
- Subjects
- Animals, Epitopes, Glycoproteins chemistry, Protein Subunits, Antibodies, Neutralizing, Antibodies, Viral, Ebolavirus, Hemorrhagic Fever, Ebola
- Abstract
Several ebolaviruses cause outbreaks of severe disease. Vaccines and monoclonal antibody cocktails are available to treat Ebola virus (EBOV) infections, but not Sudan virus (SUDV) or other ebolaviruses. Current cocktails contain antibodies that cross-react with the secreted soluble glycoprotein (sGP) that absorbs virus-neutralizing antibodies. By sorting memory B cells from EBOV infection survivors, we isolated two broadly reactive anti-GP monoclonal antibodies, 1C3 and 1C11, that potently neutralize, protect rodents from disease, and lack sGP cross-reactivity. Both antibodies recognize quaternary epitopes in trimeric ebolavirus GP. 1C11 bridges adjacent protomers via the fusion loop. 1C3 has a tripartite epitope in the center of the trimer apex. One 1C3 antigen-binding fragment anchors simultaneously to the three receptor-binding sites in the GP trimer, and separate 1C3 paratope regions interact differently with identical residues on the three protomers. A cocktail of both antibodies completely protected nonhuman primates from EBOV and SUDV infections, indicating their potential clinical value., Competing Interests: Declaration of interests R.A., C.D., and E.O.S. are inventors on a patent relating to the antibodies described in this work. All other authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
45. Current state of Ebola virus vaccines: A snapshot.
- Author
-
Woolsey C and Geisbert TW
- Subjects
- Humans, Ebola Vaccines, Hemorrhagic Fever, Ebola prevention & control
- Abstract
Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: TWG is listed as a co-inventor on U.S. patent numbers 7,635,485; 8,017,130; 8,796,013 related to filovirus vaccines and has received no royalties from any filovirus vaccine. CW has declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
46. Development of a SARS-CoV-2 Vaccine Candidate Using Plant-Based Manufacturing and a Tobacco Mosaic Virus-like Nano-Particle.
- Author
-
Royal JM, Simpson CA, McCormick AA, Phillips A, Hume S, Morton J, Shepherd J, Oh Y, Swope K, DeBeauchamp JL, Webby RJ, Cross RW, Borisevich V, Geisbert TW, Demarco JK, Bratcher B, Haydon H, and Pogue GP
- Abstract
Stable, effective, easy-to-manufacture vaccines are critical to stopping the COVID-19 pandemic resulting from the coronavirus SARS-CoV-2. We constructed a vaccine candidate CoV-RBD121-NP, which is comprised of the SARS-CoV-2 receptor-binding domain (RBD) of the spike glycoprotein (S) fused to a human IgG1 Fc domain (CoV-RBD121) and conjugated to a modified tobacco mosaic virus (TMV) nanoparticle. In vitro, CoV-RBD121 bound to the host virus receptor ACE2 and to the monoclonal antibody CR3022, a neutralizing antibody that blocks S binding to ACE2. The CoV-RBD121-NP vaccine candidate retained key SARS-CoV-2 spike protein epitopes, had consistent manufacturing release properties of safety, identity, and strength, and displayed stable potency when stored for 12 months at 2-8 °C or 22-28 °C. Immunogenicity studies revealed strong antibody responses in C57BL/6 mice with non-adjuvanted or adjuvanted (7909 CpG) formulations. The non-adjuvanted vaccine induced a balanced Th1/Th2 response and antibodies that recognized both the S1 domain and full S protein from SARS2-CoV-2, whereas the adjuvanted vaccine induced a Th1-biased response. Both adjuvanted and non-adjuvanted vaccines induced virus neutralizing titers as measured by three different assays. Collectively, these data showed the production of a stable candidate vaccine for COVID-19 through the association of the SARS-CoV-2 RBD with the TMV-like nanoparticle.
- Published
- 2021
- Full Text
- View/download PDF
47. Pan-ebolavirus protective therapy by two multifunctional human antibodies.
- Author
-
Gilchuk P, Murin CD, Cross RW, Ilinykh PA, Huang K, Kuzmina N, Borisevich V, Agans KN, Geisbert JB, Zost SJ, Nargi RS, Sutton RE, Suryadevara N, Bombardi RG, Carnahan RH, Bukreyev A, Geisbert TW, Ward AB, and Crowe JE Jr
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Binding Sites, Cell Line, Cryoelectron Microscopy, Ebolavirus ultrastructure, Epitopes immunology, Female, Glycoproteins chemistry, Glycoproteins immunology, Hemorrhagic Fever, Ebola virology, Humans, Hydrogen-Ion Concentration, Mice, Inbred BALB C, Models, Molecular, Primates, Receptors, Fc metabolism, Recombinant Proteins immunology, Viremia immunology, Mice, Antibodies, Viral immunology, Ebolavirus immunology, Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola prevention & control
- Abstract
Ebolaviruses cause a severe and often fatal illness with the potential for global spread. Monoclonal antibody-based treatments that have become available recently have a narrow therapeutic spectrum and are ineffective against ebolaviruses other than Ebola virus (EBOV), including medically important Bundibugyo (BDBV) and Sudan (SUDV) viruses. Here, we report the development of a therapeutic cocktail comprising two broadly neutralizing human antibodies, rEBOV-515 and rEBOV-442, that recognize non-overlapping sites on the ebolavirus glycoprotein (GP). Antibodies in the cocktail exhibited synergistic neutralizing activity, resisted viral escape, and possessed differing requirements for their Fc-regions for optimal in vivo activities. The cocktail protected non-human primates from ebolavirus disease caused by EBOV, BDBV, or SUDV with high therapeutic effectiveness. High-resolution structures of the cocktail antibodies in complex with GP revealed the molecular determinants for neutralization breadth and potency. This study provides advanced preclinical data to support clinical development of this cocktail for pan-ebolavirus therapy., Competing Interests: Declaration of interests J.E.C. has served as a consultant for Eli Lilly, GlaxoSmithKline and Luna Biologics, is a member of the Scientific Advisory Boards of CompuVax and Meissa Vaccines, and is Founder of IDBiologics. The Crowe laboratory at Vanderbilt University Medical Center has received unrelated sponsored research agreements from Takeda Vaccines, IDBiologics, and AstraZeneca. Vanderbilt University has applied for patents concerning ebolavirus antibodies that are related to this work. All other authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
48. Comparison of Zaire and Bundibugyo Ebolavirus Polymerase Complexes and Susceptibility to Antivirals through a Newly Developed Bundibugyo Minigenome System.
- Author
-
Levine CB, Mire CE, and Geisbert TW
- Subjects
- Antibodies, Viral genetics, Antibodies, Viral immunology, Antiviral Agents therapeutic use, Drug Resistance, Viral genetics, Ebolavirus immunology, Genes, Reporter genetics, Genome, Viral genetics, Humans, Viral Proteins metabolism, Ebolavirus genetics, Genetic Engineering methods, Virus Replication genetics
- Abstract
Members of the genus Ebolavirus cause lethal disease in humans, with Zaire ebolavirus (EBOV) being the most pathogenic (up to 90% morality) and Bundibugyo ebolavirus (BDBV) the least pathogenic (∼37% mortality). Historically, there has been a lack of research on BDBV, and there is no means to study BDBV outside of a high-containment laboratory. Here, we describe a minigenome replication system to study BDBV transcription and compare the efficacy of small-molecule inhibitors between EBOV and BDBV. Using this system, we examined the ability of the polymerase complex proteins from EBOV and BDBV to interact and form a functional unit as well as the impact of the genomic untranslated ends, known to contain important signals for transcription (3'-untranslated region) and replication (5'-untranslated region). Various levels of compatibility were observed between proteins of the polymerase complex from each ebolavirus, resulting in differences in genome transcription efficiency. Most pronounced was the effect of the nucleoprotein and the 3'-untranslated region. These data suggest that there are intrinsic specificities in the polymerase complex and untranslated signaling regions that could offer insight regarding observed pathogenic differences. Further adding to the differences in the polymerase complexes, posttransfection/infection treatment with the compound remdesivir (GS-5734) showed a greater inhibitory effect against BDBV than EBOV. The delayed growth kinetics of BDBV and the greater susceptibility to polymerase inhibitors indicate that disruption of the polymerase complex is a viable target for therapeutics. IMPORTANCE Ebolavirus disease is a viral infection and is fatal in 25 to 90% of cases, depending on the viral species and the amount of supportive care available. Two species have caused outbreaks in the Democratic Republic of the Congo, Zaire ebolavirus (EBOV) and Bundibugyo ebolavirus (BDBV). Pathogenesis and clinical outcome differ between these two species, but there is still limited information regarding the viral mechanism for these differences. Previous studies suggested that BDBV replicates slower than EBOV, but it is unknown if this is due to differences in the polymerase complex and its role in transcription and replication. This study details the construction of a minigenome replication system that can be used in a biosafety level 2 laboratory. This system will be important for studying the polymerase complex of BDBV and comparing it with other filoviruses and can be used as a tool for screening inhibitors of viral growth.
- Published
- 2021
- Full Text
- View/download PDF
49. A recombinant Cedar virus based high-throughput screening assay for henipavirus antiviral discovery.
- Author
-
Amaya M, Cheng H, Borisevich V, Navaratnarajah CK, Cattaneo R, Cooper L, Moore TW, Gaisina IN, Geisbert TW, Rong L, and Broder CC
- Subjects
- Cell Line, Genes, Reporter, Henipavirus physiology, Henipavirus Infections virology, Humans, Luciferases genetics, Luciferases metabolism, Recombination, Genetic, Viral Envelope Proteins genetics, Virus Internalization drug effects, Virus Replication drug effects, Antiviral Agents pharmacology, Henipavirus drug effects, Henipavirus Infections drug therapy, High-Throughput Screening Assays, Viral Envelope Proteins metabolism
- Abstract
Nipah virus (NiV) and Hendra virus (HeV) are highly pathogenic, bat-borne paramyxoviruses in the genus Henipavirus that cause severe and often fatal acute respiratory and/or neurologic diseases in humans and livestock. There are currently no approved antiviral therapeutics or vaccines for use in humans to treat or prevent NiV or HeV infection. To facilitate development of henipavirus antivirals, a high-throughput screening (HTS) platform was developed based on a well-characterized recombinant version of the nonpathogenic Henipavirus, Cedar virus (rCedV). Using reverse genetics, a rCedV encoding firefly luciferase (rCedV-Luc) was rescued and its utility evaluated for high-throughput antiviral compound screening. The luciferase reporter gene signal kinetics of rCedV-Luc in different human cell lines was characterized and validated as an authentic real-time measure of viral growth. The rCedV-Luc platform was optimized as an HTS assay that demonstrated high sensitivity with robust Z' scores, excellent signal-to-background ratios and coefficients of variation. Eight candidate compounds that inhibited rCedV replication were identified for additional validation and demonstrated that 4 compounds inhibited authentic NiV-Bangladesh replication. Further evaluation of 2 of the 4 validated compounds in a 9-point dose response titration demonstrated potent antiviral activity against NiV-Bangladesh and HeV, with minimal cytotoxicity. This rCedV reporter can serve as a surrogate yet authentic BSL-2 henipavirus platform that will dramatically accelerate drug candidate identification in the development of anti-henipavirus therapies., (Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
50. Cooperativity mediated by rationally selected combinations of human monoclonal antibodies targeting the henipavirus receptor binding protein.
- Author
-
Doyle MP, Kose N, Borisevich V, Binshtein E, Amaya M, Nagel M, Annand EJ, Armstrong E, Bombardi R, Dong J, Schey KL, Broder CC, Zeitlin L, Kuang EA, Bornholdt ZA, West BR, Geisbert TW, Cross RW, and Crowe JE Jr
- Subjects
- Animals, Antibody Specificity, Chlorocebus aethiops, Cross Reactions, Disease Models, Animal, Drug Therapy, Combination, Ephrin-B2 immunology, Ephrin-B2 metabolism, Ephrin-B3 immunology, Ephrin-B3 metabolism, Female, Henipavirus Infections immunology, Henipavirus Infections metabolism, Henipavirus Infections virology, Host-Pathogen Interactions, Humans, Mesocricetus, Receptors, Virus immunology, Receptors, Virus metabolism, Vero Cells, Antibodies, Monoclonal pharmacology, Antibodies, Neutralizing pharmacology, Antiviral Agents pharmacology, Ephrin-B2 antagonists & inhibitors, Ephrin-B3 antagonists & inhibitors, Henipavirus pathogenicity, Henipavirus Infections prevention & control, Receptors, Virus antagonists & inhibitors
- Abstract
Hendra virus and Nipah virus (NiV), members of the Henipavirus (HNV) genus, are zoonotic paramyxoviruses known to cause severe disease across six mammalian orders, including humans. We isolated a panel of human monoclonal antibodies (mAbs) from the B cells of an individual with prior exposure to equine Hendra virus (HeV) vaccine, targeting distinct antigenic sites. The most potent class of cross-reactive antibodies achieves neutralization by blocking viral attachment to the host cell receptors ephrin-B2 and ephrin-B3, with a second class being enhanced by receptor binding. mAbs from both classes display synergistic activity in vitro. In a stringent hamster model of NiV Bangladesh (NiV
B ) infection, antibodies from both classes reduce morbidity and mortality and achieve synergistic protection in combination. These candidate mAbs might be suitable for use in a cocktail therapeutic approach to achieve synergistic potency and reduce the risk of virus escape., Competing Interests: Declaration of interests J.E.C. has served as a consultant for Luna Biologics, is a member of the Scientific Advisory Board of Meissa Vaccines, and is Founder of IDBiologics. The Crowe laboratory at Vanderbilt University Medical Center has received unrelated sponsored research agreements from Takeda, IDBiologics, and AstraZeneca. E.A.K., Z.A.B., and B.R.W. are employees and shareholders of Mapp. L.Z. is an employee, shareholder, and co-owner of Mapp. C.C.B. is a US federal employee, and C.C.B. and M.A. are co-inventors on US and foreign patents pertaining to Cedar virus and methods of use and recombinant Cedar virus chimeras, whose assignees are the US as represented by the Henry M. Jackson Foundation for the Advancement of Military Medicine (Bethesda, MD, USA). The remaining authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.