346 results on '"Monath TP"'
Search Results
2. Does restricted distribution limit access and coverage of yellow fever vaccine in the United States?
- Author
-
Giesberg Ja, Monath Tp, and Fierros Eg
- Subjects
Risk ,Letter ,Yellow fever vaccine ,lcsh:Medicine ,Health Services Accessibility ,lcsh:Infectious and parasitic diseases ,Yellow Fever ,Humans ,Medicine ,Research article ,lcsh:RC109-216 ,Limit (mathematics) ,Problem Solving ,business.industry ,Viral Vaccine ,Vaccination ,Yellow fever ,lcsh:R ,Viral Vaccines ,Restricted distribution ,medicine.disease ,Virology ,United States ,Immunology ,business ,medicine.drug ,Research Article - Published
- 1998
3. Yellow Fever Vaccine—Reply to S. Arya
- Author
-
Monath Tp, Giesberg Ja, and Fierros Eg
- Subjects
Microbiology (medical) ,Epidemiology ,business.industry ,lcsh:R ,Yellow fever vaccine ,lcsh:Medicine ,Virology ,United States ,lcsh:Infectious and parasitic diseases ,Infectious Diseases ,Medicine ,Research article ,lcsh:RC109-216 ,business ,medicine.drug - Published
- 1999
4. Gastritis in urease-immunized mice after Helicobacter felis challenge may be due to residual bacteria
- Author
-
Ermak, TH, primary, Ding, R, additional, Ekstein, B, additional, Hill, J, additional, Myers, GA, additional, Lee, CK, additional, Pappo, J, additional, Kleanthous, HK, additional, and Monath, TP, additional
- Published
- 1997
- Full Text
- View/download PDF
5. Therapeutic immunization against Helicobacter mustelae in naturally infected ferrets
- Author
-
Cuenca, R, primary, Blanchard, TG, additional, Czinn, SJ, additional, Nedrud, JG, additional, Monath, TP, additional, Lee, CK, additional, and Redline, RW, additional
- Published
- 1996
- Full Text
- View/download PDF
6. Development of antigen-specific memory CD8+ T cells following live-attenuated chimeric West Nile virus vaccination.
- Author
-
Smith HL, Monath TP, Pazoles P, Rothman AL, Casey DM, Terajima M, Ennis FA, Guirakhoo F, Green S, Smith, Heidi L, Monath, Thomas P, Pazoles, Pamela, Rothman, Alan L, Casey, Diane M, Terajima, Masanori, Ennis, Francis A, Guirakhoo, Farshad, and Green, Sharone
- Abstract
ChimeriVax-WN02 is a novel live-attenuated West Nile virus (WNV) vaccine containing modified WNV premembrane (prM) and envelope (E) sequences inserted into the yellow fever 17D vaccine genome. We investigated the induction and evolution of CD8(+) T cell responses to a WNV envelope epitope, which is a dominant target in naturally infected HLA-A*02-positive individuals. WNV epitope-specific CD8(+) T cells were detected by HLA tetramer staining in 22 of 23 donors tested, with peak frequencies occurring between days 14 and 28. WNV epitope-specific T cells evolved from an effector phenotype to a long-lived memory phenotype. In the majority of donors, CD8(+) T cells were able to lyse targets expressing WNV envelope protein and produced macrophage inflammatory protein 1ß, interferon γ, and/or tumor necrosis factor α following envelope peptide stimulation. WNV E-specific CD8(+) T cell responses were detected for up to 1 year after vaccination. The evolution of this WNV-specific T cell response is similar to that observed in established, highly immunogenic vaccines. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
7. Interferon-gamma-mediated antiviral immunity against orthopoxvirus infection is provided by gamma delta T cells.
- Author
-
Agrati C, Castilletti C, De Santis R, Cimini E, Bordi L, Malkovsky M, Poccia F, Capobianchi MR, Abate G, Eslick J, Newman FK, Frey SE, Belshe RB, Monath TP, and Hoft DF
- Published
- 2006
- Full Text
- View/download PDF
8. Flow-cytometric detection of vaccinia-induced memory effector CD4(+), CD8(+), and gamma delta TCR(+) T cells capable of antigen-specific expansion and effector functions.
- Author
-
Abate G, Eslick J, Newman FK, Frey SE, Belshe RB, Monath TP, and Hoft DF
- Abstract
We developed a carboxyfluorescein succinimidyl ester (CFSE)-based flow-cytometric assay that can detect different subsets of vaccinia-specific T cells capable of both antigen-specific expansion and protective effector functions. Proliferation and effector functions were detected by CFSE dilution and intracellular staining, respectively. Absolute numbers of CD4(+)/CFSE(lo)/interferon (IFN)- gamma (+), CD8(+)/CFSE(lo)/IFN- gamma (+), CD8(+)/CFSE(lo)/granzyme A(+), and CD8(+)/CFSE(lo)/CD107a(+) T cells present after in vitro stimulation with live vaccinia were significantly higher in immunized individuals (P<.05). These CD4(+) and CD8(+) T cell increases were >2 log higher than increases detectable by standard lymphoproliferation and cytotoxicity assays. Vaccinia-specific CD8(+)/CFSE(lo)/IFN- gamma (+) and granzyme A(+) T cell responses were significantly correlated with the results of standard (51)Cr-release cytolytic assays (P<.05). Furthermore, vaccinia induced antigen-specific memory gamma delta T cells. We demonstrate that vaccinia induces robust memory effector CD4(+), CD8(+), and gamma delta T cells, all of which are relevant for protection against smallpox. CFSE-based flow-cytometric assays will be useful in evaluating cell-mediated immune responses induced by new smallpox vaccines. Copyright © 2005 Infectious Diseases Society of America [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
9. New developments in flavivirus vaccines with special attention to yellow fever.
- Author
-
Pugachev KV, Guirakhoo F, Monath TP, Pugachev, Konstantin V, Guirakhoo, Farshad, and Monath, Thomas P
- Published
- 2005
- Full Text
- View/download PDF
10. Helicobacter pylori reinfection is common in Peruvian adults after antibiotic eradication therapy.
- Author
-
Soto G, Bautista CT, Roth DE, Gilman RH, Velapatiño B, Ogura M, Dailide G, Razuri M, Meza R, Katz U, Monath TP, Berg DE, Taylor DN, and Gastrointestinal Physiology Working Group in Peru
- Abstract
To characterize posttreatment recurrence of Helicobacter pylori in Peru, 192 adults with H. pylori-positive gastric biopsy specimens were monitored by (14)C-Urea breath test, after eradication of H. pylori by use of amoxicillin, clarithromycin, and omeprazole. The cumulative risk of recurrence at 18 months was 30.3% (95% confidence interval, 21.4%-39.3%). Randomly amplified polymorphic DNA patterns and DNA sequence data established that, among 28 pairs of H. pylori isolates from pretreatment and recurrent infections, 6 (21%) were genetically similar, suggesting recrudescence of the previous infection, and 22 (79%) were different, suggesting reinfection with a new strain that differed from that involved in the initial infection. Eating mainly outside of the home was a risk factor for infection with a new strain (adjusted relative risk [RR], 5.07), whereas older age was a protective factor (adjusted RR, 0.20). Although an increase in the anti-H. pylori IgG antibody titer corresponded to recurrence, pretreatment and recurrent infections were similar with respect to quantitative culture colony counts and histologic characteristics, suggesting that neither prior eradication nor the memory immune response measurably alters the risk or burden of recurrent infection. Although eradication with antibiotics was successful, the high rate of reinfection suggests that treatment is unlikely to have a lasting public health effect in this setting. Copyright © 2003 The University of Chicago [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
11. Yellow fever vaccine: recommendations of the Advisory Committee on Immunization Practices (ACIP), 2002.
- Author
-
Cetron MS, Marfin AA, Julian KG, Gubler DJ, Sharp DJ, Barwick RS, Weld LH, Chen R, Clover RD, Deseda-Tous J, Marchessault V, Offit PA, and Monath TP
- Abstract
This report updates CDC's recommendations for using yellow fever vaccine (CDC. Yellow Fever Vaccine: Recommendations of the Advisory Committee on Immunizations Practices: MMWR 1990;39[No. RR-6]1--6). The 2002 recommendations include new or updated information regarding 1) reports of yellow fever vaccine-associated viscerotropic disease (previously reported as febrile multiple organ system failure); 2) use of yellow fever vaccine for pregnant women and persons infected with human immunodeficiency virus (HIV); and 3) concurrent use of yellow fever vaccine with other vaccines. A link to this report and other information related to yellow fever can be accessed at the website for Travelers' Health, Division of Global Migration and Quarantine, National Center for Infectious Diseases, CDC, at http://www.cdc.gov/travel/index.htm, and through the website for the Division of Vector-Borne Infectious Diseases, National Center for Infectious Diseases, CDC, at http://www.cdc.gov/ncidod/dvbid/yellowfever/index.htm. [ABSTRACT FROM AUTHOR]
- Published
- 2002
12. Studies of glucose metabolism in rhesus monkeys after Venezuelan equine encephalitis virus infection
- Author
-
Bowen Gs, Kemp Ge, Rayfield Ej, and Monath Tp
- Subjects
Blood Glucose ,Encephalomyelitis, Equine ,Male ,medicine.medical_treatment ,Virulence ,Carbohydrate metabolism ,Vaccines, Attenuated ,Arbovirus ,Glucagon ,Virus ,Encephalitis Virus, Venezuelan Equine ,Virology ,medicine ,Animals ,Insulin ,biology ,Histology ,Encephalomyelitis, Venezuelan Equine ,Viral Vaccines ,Glucose Tolerance Test ,medicine.disease ,Macaca mulatta ,Infectious Diseases ,biology.protein ,Female ,Antibody - Abstract
Previous studies have demonstrated a diabetogenic effect of Venezuelan equine encephalitis (VEE) virus in hamsters. A preliminary study was conducted in which five 2- to 3-year-old rhesus monkeys were infected with the virulent Trinidad donkey strain of VEE virus and their carbohydrate metabolism was studied over 10 months. All animals developed mild clinical illness (rhinorrhea, cough, fever), were viremic, and developed antibodies. As compared with the results of preinoculation intravenous glucose tolerance tests (IVGTT), the monkeys had abnormally high glucose values by 2 months postinoculation (PI), progressively diminished insulin responses between 8 days and 5 months PI, and significantly lower glucagon curves 2, 5, and 10 months PI. Pancreatic histology and insulin content were normal. A second, controlled study was conducted of glucose and insulin metabolism in somewhat older (3- to 8-year-old) rhesus monkeys after they were infected with both the Trinidad donkey strain of VEE virus and the attenuated VEE vaccine (TC-83). Groups of six monkeys received the virulent virus and the TC-83 vaccine, and five animals were sham-inoculated with saline. Monkeys inoculated with virulent virus became viremic, and 50% became febrile without overt signs of illness, whereas those given TC-83 virus remained afebrile and did not become viremic, but five of six developed antibodies. Intravenous glucose tolerance tests were performed and serum immunoreactive insulin responses to glucose administration measured before infection and 2 and 5 months later. No significant and consistent alterations of glucose or insulin responses were detected in the infected or control groups. Although several animals had preinoculation anti-islet cell antibodies, none developed new antibodies during the study.
- Published
- 1980
13. Yellow Fever recommendations for tourists to kenya: a flawed risk assessment.
- Author
-
Monath TP, Gershman M, Hill DR, Marano N, Staples JE, and Wilder-Smith A
- Published
- 2009
- Full Text
- View/download PDF
14. Improving vaccine coverage in Africa.
- Author
-
Fasina FO, Kaplan B, Kahn LH, and Monath TP
- Published
- 2008
- Full Text
- View/download PDF
15. Sofosbuvir Off-label Treatment of Yellow Fever Patients During an Outbreak in Brazil, 2018: A Cohort Study.
- Author
-
Rezende IM, Mendonça DC, Costa TA, de Oliveria GFG, Arruda MS, Gonçalves AP, Alves PA, Calzavara-Silva CE, Martins-Filho OA, Teixeira-Carvalho A, Bonjardim CA, Monath TP, LaBeaud AD, Drumond BP, Pascoal-Xavier MA, Pereira LS, and Ramalho DB
- Abstract
We enrolled 21 patients with laboratory-confirmed yellow fever (YF), hospitalized at Eduardo de Menezes Hospital, Brazil, to be treated with sofosbuvir, a drug approved for hepatitis C. Given the absence of specific YF antiviral treatments, the off-label nonrandomized sofosbuvir treatment aimed to address high disease severity and the risk of fatal outcomes. Patients received a daily dose of 400 mg sofosbuvir from 4 to 10 days post-symptom onset. YF viral load (VL) comparisons were made between treated and nontreated patients who either survived or died. The genomic VL for the treated group steadily decreased after day 7 post-symptom onset, suggesting that sofosbuvir might reduce YF VL. This study underscores the urgent need for YF antiviral therapies, advocating for randomized clinical trials to further explore sofosbuvir's role in YF treatment., (© The Author(s) 2024. Published by Oxford University Press on behalf of Infectious Diseases Society of America.)
- Published
- 2024
- Full Text
- View/download PDF
16. Evaluation of humoral immune response after yellow fever infection: an observational study on patients from the 2017-2018 sylvatic outbreak in Brazil.
- Author
-
Gonçalves AP, Almeida LT, Rezende IMd, Fradico JRB, Pereira LS, Ramalho DB, Pascoal Xavier MA, Calzavara Silva CE, Monath TP, LaBeaud AD, Drumond BP, Campi-Azevedo AC, Martins-Filho OA, Teixeira-Carvalho A, and Alves PA
- Subjects
- Humans, Brazil epidemiology, Male, Female, Adult, Middle Aged, Vaccination, Neutralization Tests, Young Adult, Aged, Adolescent, Yellow Fever immunology, Yellow Fever epidemiology, Yellow Fever virology, Yellow fever virus immunology, Yellow fever virus genetics, Immunity, Humoral, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, Antibodies, Viral blood, Antibodies, Viral immunology, Disease Outbreaks, Yellow Fever Vaccine immunology
- Abstract
Between 2016 and 2018, Brazil experienced major sylvatic yellow fever (YF) outbreaks that caused hundreds of casualties, with Minas Gerais (MG) being the most affected state. These outbreaks provided a unique opportunity to assess the immune response triggered by the wild-type (WT) yellow fever virus (YFV) in humans. The plaque reduction neutralization test (PRNT) is currently the standard method to assess the humoral immune response to YFV by measuring neutralizing antibodies (nAbs). The present study aimed to evaluate the humoral immune response of patients from the 2017-2018 sylvatic YF outbreak in MG with different disease outcomes by using PRNTs with a WT YFV strain, isolated from the 2017-2018 outbreak, and a vaccine YFV strain. Samples from naturally infected YF patients were tested, in comparison with healthy vaccinees. Results showed that both groups presented different levels of nAb against the WT and vaccine strains, and the levels of neutralization against the strains varied homotypically and heterotypically. Results based on the geometric mean titers (GMTs) suggest that the humoral immune response after a natural infection of YFV can reach higher levels than that induced by vaccination (GMT of patients against WT YFV compared to GMT of vaccinees, P < 0.0001). These findings suggest that the humoral immune responses triggered by the vaccine and WT strains of YFV are different, possibly due to genetic and antigenic differences between these viruses. Therefore, current means of assessing the immune response in naturally infected YF individuals and immunological surveillance methods in areas with intense viral circulation may need to be updated.IMPORTANCEYellow fever is a deadly febrile disease caused by the YFV. Despite the existence of effective vaccines, this disease still represents a public health concern worldwide. Much is known about the immune response against the vaccine strains of the YFV, but recent studies have shown that it differs from that induced by WT strains. The extent of this difference and the mechanisms behind it are still unclear. Thus, studies aimed to better understand the immune response against this virus are relevant and necessary. The present study evaluated levels of neutralizing antibodies of yellow fever patients from recent outbreaks in Brazil, in comparison with healthy vaccinees, using plaque reduction neutralization tests with WT and vaccine YFV strains. Results showed that the humoral immune response in naturally infected patients was higher than that induced by vaccination, thus providing new insights into the immune response triggered against these viruses., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
17. Japanese Encephalitis: Risk of Emergence in the United States and the Resulting Impact.
- Author
-
Monath TP
- Subjects
- Humans, United States epidemiology, Animals, Horses, Swine, Agriculture, Encephalitis, Japanese epidemiology, Encephalitis, Japanese prevention & control, Encephalitis Virus, Japanese, Encephalitis, Viral, Chiroptera, Culicidae, Vaccines, Zika Virus, Zika Virus Infection
- Abstract
Japanese encephalitis virus is a mosquito-borne member of the Flaviviridae family. JEV is the leading cause of viral encephalitis in Asia and is characterized by encephalitis, high lethality, and neurological sequelae in survivors. The virus also causes severe disease in swine, which are an amplifying host in the transmission cycle, and in horses. US agricultural authorities have recently recognized the threat to the swine industry and initiated preparedness activities. Other mosquito-borne viruses exotic to the Western Hemisphere have been introduced and established in recent years, including West Nile, Zika, and chikungunya viruses, and JEV has recently invaded continental Australia for the first time. These events amply illustrate the potential threat of JEV to US health security. Susceptible indigenous mosquito vectors, birds, feral and domestic pigs, and possibly bats, constitute the receptive ecological ingredients for the spread of JEV in the US. Fortunately, unlike the other virus invaders mentioned above, an inactivated whole virus JE vaccine (IXIARO
® ) has been approved by the US Food and Drug Administration for human use in advance of a public health emergency, but there is no veterinary vaccine. This paper describes the risks and potential consequences of the introduction of JEV into the US, the need to integrate planning for such an event in public health policy, and the requirement for additional countermeasures, including antiviral drugs and an improved single dose vaccine that elicits durable immunity in both humans and livestock.- Published
- 2023
- Full Text
- View/download PDF
18. Immunological correlates of protection afforded by PHV02 live, attenuated recombinant vesicular stomatitis virus vector vaccine against Nipah virus disease.
- Author
-
Monath TP, Nichols R, Feldmann F, Griffin A, Haddock E, Callison J, Meade-White K, Okumura A, Lovaglio J, Hanley PW, Clancy CS, Shaia C, Rida W, and Fusco J
- Subjects
- Animals, Chlorocebus aethiops, Antibodies, Neutralizing, Nipah Virus, Ebola Vaccines, Henipavirus Infections prevention & control, Vesicular Stomatitis, Hemorrhagic Fever, Ebola
- Abstract
Introduction: Immune correlates of protection afforded by PHV02, a recombinant vesicular stomatitis (rVSV) vector vaccine against Nipah virus (NiV) disease, were investigated in the African green monkey (AGM) model. Neutralizing antibody to NiV has been proposed as the principal mediator of protection against future NiV infection., Methods: Two approaches were used to determine the correlation between neutralizing antibody levels and outcomes following a severe (1,000 median lethal doses) intranasal/intratracheal (IN/IT) challenge with NiV (Bangladesh): (1) reduction in vaccine dose given 28 days before challenge and (2) challenge during the early phase of the antibody response to the vaccine., Results: Reduction in vaccine dose to very low levels led to primary vaccine failure rather than a sub-protective level of antibody. All AGMs vaccinated with the nominal clinical dose (2 × 10
7 pfu) at 21, 14, or 7 days before challenge survived. AGMs vaccinated at 21 days before challenge had neutralizing antibodies (geometric mean titer, 71.3). AGMs vaccinated at 7 or 14 days before challenge had either undetectable or low neutralizing antibody titers pre-challenge but had a rapid rise in titers after challenge that abrogated the NiV infection. A simple logistic regression model of the combined studies was used, in which the sole explanatory variable was pre-challenge neutralizing antibody titers. For a pre-challenge titer of 1:5, the predicted survival probability is 100%. The majority of animals with pre-challenge neutralizing titer of ≥1:20 were protected against pulmonary infiltrates on thoracic radiograms, and a majority of those with titers ≥1:40 were protected against clinical signs of illness and against a ≥fourfold antibody increase following challenge (indicating sterile immunity). Controls receiving rVSV-Ebola vaccine rapidly succumbed to NiV challenge, eliminating the innate immunity stimulated by the rVSV vector as a contributor to survival in monkeys challenged as early as 7 days after vaccination., Discussion and Conclusion: It was concluded that PHV02 vaccine elicited a rapid onset of protection and that any detectable level of neutralizing antibody was a functional immune correlate of survival., Competing Interests: TM is employed by Crozet Biopharma LLC. RN and JF are employed by Public Health Vaccines Inc. WR is an independent consultant. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Monath, Nichols, Feldmann, Griffin, Haddock, Callison, Meade-White, Okumura, Lovaglio, Hanley, Clancy, Shaia, Rida and Fusco.)- Published
- 2023
- Full Text
- View/download PDF
19. Characterization and Investigation of Risk Factors for Late-Relapsing Hepatitis After Yellow Fever.
- Author
-
Rezende IM, McClure MA, Pereira LS, Fradico JRB, Cenachi ARC, Moura AS, Paladino LLA, Dutra MRT, Alves PA, Xavier MAP, Said RFDC, Ramalho DB, Gama TDP, Martins-Filho OA, Monath TP, Teixeira-Carvalho A, Drumond BP, and LaBeaud AD
- Subjects
- Humans, Disease Outbreaks, Risk Factors, Brazil epidemiology, Disease Progression, Yellow Fever complications, Yellow Fever epidemiology, Hepatitis epidemiology, Hepatitis A epidemiology, Yellow Fever Vaccine
- Abstract
Background: Late-relapsing hepatitis after yellow fever (LHep-YF) during the convalescent phase of the disease has been described during recent yellow fever (YF) outbreaks in Brazil. LHep-YF is marked by a rebound in liver enzymes and nonspecific clinical manifestations around 46-60 days after YF symptom onset., Methods: Here we have characterized the clinical course and risk factors for LHep-YF using data from a representative cohort of patients who survived YF in Brazil, 2017-2018. A total of 221 YF-positive patients were discharged from the infectious disease reference hospital in Minas Gerais and were followed up at 30, 45, and 60 days post-symptom onset., Results: From 46 to 60 days post-symptom onset, 16% of YF patients (n = 36/221) exhibited a rebound of aminotransferases (aspartate aminotransferase or alanine aminotransferase >500 IU/L), alkaline phosphatase, and total bilirubin levels. Other etiologies of liver inflammation such as infectious hepatitis, autoimmune hepatitis, and metabolic liver disease were ruled out. Jaundice, fatigue, headache, and low platelet levels were associated with LHep-YF. Demographic factors, clinical manifestations, laboratory tests, ultrasound findings, and viral load during the acute phase of YF were not associated with the occurrence of LHep-YF., Conclusions: These findings provide new data on the clinical course of Late-relapsing hepatitis during the convalescent phase of YF and highlight the need for extended patient follow-up after acute YF., Competing Interests: Potential conflicts of interest. A. R. C. C. reports consulting fees from the International Horizon Scanning Initiative’s Horizon Scanning System tracked by ECRI, a global, independent authority on healthcare technology and safety. B. P. D. reports unpaid roles as financial council and vice-president of the Brazilian Society for Virology and as virology chair for the Brazilian Society for Microbiology. All other authors report no potential 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.)
- Published
- 2023
- Full Text
- View/download PDF
20. Biographical Feature: Marc S. Collett (23 May 1951-11 June 2022): the Battle against Viral Disease Has Lost a Valiant Warrior, and the World Has Lost a Splendid Human Being.
- Author
-
Glassman KF, Brugge JS, Purchio AF, Kakach LT, Fuerst TR, Woods RM, Stover CK, Chunduru SK, Hincks JR, McKinlay MA, Nash CH, Pevear DC, Monath TP, Rice CM, and Peters P
- Published
- 2023
- Full Text
- View/download PDF
21. A novel RBD-protein/peptide vaccine elicits broadly neutralizing antibodies and protects mice and macaques against SARS-CoV-2.
- Author
-
Wang S, Wang CY, Kuo HK, Peng WJ, Huang JH, Kuo BS, Lin F, Liu YJ, Liu Z, Wu HT, Ding S, Hou KL, Cheng J, Yang YT, Jiang MH, Wang MS, Chen T, Xia WG, Lin E, Hung CH, Chen HJ, Shih Z, Lin YL, Ryan V, Hu MM, Heppner DG, Malherbe DC, Periasamy S, Kuzmina N, Subramani C, Hellerstein M, Monath TP, Rumyantsev A, Bukreyev A, and Guirakhoo F
- Subjects
- Rats, Mice, Humans, Animals, SARS-CoV-2, COVID-19 Vaccines, Broadly Neutralizing Antibodies, Pandemics prevention & control, Rats, Sprague-Dawley, Spike Glycoprotein, Coronavirus, Antibodies, Neutralizing, Vaccines, Subunit genetics, Mice, Inbred BALB C, Macaca mulatta, Antibodies, Viral, COVID-19 prevention & control, Viral Vaccines
- Abstract
The development of safe and effective vaccines to respond to COVID-19 pandemic/endemic remains a priority. We developed a novel subunit protein-peptide COVID-19 vaccine candidate (UB-612) composed of: (i) receptor binding domain of SARS-CoV-2 spike protein fused to a modified single-chain human IgG1 Fc; (ii) five synthetic peptides incorporating conserved helper and cytotoxic T lymphocyte (Th/CTL) epitopes derived from SARS-CoV-2 structural proteins (three from S2 subunit, one from membrane and one from nucleocapsid), and one universal Th peptide; (iii) aluminum phosphate as adjuvant. The immunogenicity and protective immunity induced by UB-612 vaccine were evaluated in four animal models: Sprague-Dawley rats, AAV-hACE2 transduced BALB/c mice, rhesus and cynomolgus macaques. UB-612 vaccine induced high levels of neutralizing antibody and T-cell responses, in all animals. The immune sera from vaccinated animals neutralized the SARS-CoV-2 original wild-type strains and multiple variants of concern, including Delta and Omicron. The vaccination significantly reduced viral loads, lung pathology scores, and disease progression after intranasal and intratracheal challenge with SARS-CoV-2 in mice, rhesus and cynomolgus macaques. UB-612 has been tested in primary regimens in Phase 1 and Phase 2 clinical studies and is currently being evaluated in a global pivotal Phase 3 clinical study as a single dose heterologous booster.
- Published
- 2022
- Full Text
- View/download PDF
22. Yellow fever resurgence: An avoidable crisis?
- Author
-
Lindsey NP, Horton J, Barrett ADT, Demanou M, Monath TP, Tomori O, Van Herp M, Zeller H, Fall IS, Cibrelus L, and Erin Staples J
- Published
- 2022
- Full Text
- View/download PDF
23. High Neutralizing Antibody Levels Against Severe Acute Respiratory Syndrome Coronavirus 2 Omicron BA.1 and BA.2 After UB-612 Vaccine Booster.
- Author
-
Guirakhoo F, Wang S, Wang CY, Kuo HK, Peng WJ, Liu H, Wang L, Johnson M, Hunt A, Hu MM, Monath TP, Rumyantsev A, and Goldblatt D
- Subjects
- Antibodies, Neutralizing, Antibodies, Viral, Humans, SARS-CoV-2, COVID-19 prevention & control, Viral Vaccines
- Abstract
The highly transmissible severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant has caused high rates of breakthrough infections in those previously vaccinated with ancestral strain coronavirus disease 2019 (COVID-19) vaccines. Here, we demonstrate that a booster dose of UB-612 vaccine candidate delivered 7-9 months after primary vaccination increased neutralizing antibody levels by 131-, 61-, and 49-fold against ancestral SARS-CoV-2 and the Omicron BA.1 and BA.2 variants, respectively. Based on the receptor-binding domain protein binding antibody responses, the UB-612 third-dose booster may lead to an estimated approximately 95% efficacy against symptomatic COVID-19 caused by the ancestral strain. Our results support UB-612 as a potential potent booster against current and emerging SARS-CoV-2 variants., Competing Interests: Potential conflicts of interest. F. G., S. W., L. W., M. M. H., T. M., and A. R. are employees of Vaxxinity Inc (Dallas, Texas). C. Y. W., W.-J. P., H.-K. K., and H. L. are employees of United Biomedical Inc Asia (Hsinchu, Taiwan). All other authors report no potential 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) 2022. 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
- 2022
- Full Text
- View/download PDF
24. Estimation of the Minimal Rift Valley Fever Virus Protective Neutralizing Antibody Titer in Human Volunteers Immunized with MP-12 Vaccine Based on Protection in a Mouse Model of Disease.
- Author
-
Watts DM, Westover JLB, Palermo PM, Bailey KW, Morrill JC, Bettinger GE, Monath TP, Smith DR, Peters CJ, Pittman PR, Orbegozo J, and Gowen BB
- Subjects
- Mice, Humans, Animals, Healthy Volunteers, Vaccines, Attenuated, Antibodies, Viral, Antibodies, Neutralizing, Mice, Inbred BALB C, Disease Models, Animal, Rift Valley fever virus, Rift Valley Fever, Viral Vaccines
- Abstract
The Rift Valley fever virus (RVFV) MP-12 vaccine is a promising human and veterinary vaccine. Although the vaccine elicited neutralizing antibody (nAb) in human volunteers, the minimal antibody titer that is needed to afford protection is unknown. Therefore, this study was conducted to determine the minimal nAb titer elicited by the RVFV MP-12 vaccine in human volunteers that protected mice against lethal RVFV challenge as a surrogate assessment of the protective efficacy of the vaccine. Among volunteers who were vaccinated with the MP-12 vaccine during a phase II trial, sera with antibody titers of 1:20 collected 5 years post-vaccination (PV), 1:40 titer collected 2 years PV, and 1:80 titer collected 1 year PV was passively transferred to groups of BALB/c mice. Blood samples were obtained 1 day after passive transfer to determine the RVFV neutralizing nAb titer before challenge with pathogenic RVFV (strain ZH501). Our results indicated that 1 day after passive transfer of the immune sera, an approximate 4-fold reduction in circulating nAb titers was detected in the mice. The presence of RVFV nAb titers in the range of 1:5 to 1:20 were generally protective (75-100% survival). These results suggested that circulating titers of 1:5 or higher offer a high degree of protection by MP-12-elicited antibody in human volunteers. Also, the findings highlighted the value of using the BALB/c mouse RVFV challenge model as a surrogate for evaluating the protective nAb responses elicited by MP-12 and possible use for evaluating the efficacy of other RVFV vaccine candidates.
- Published
- 2022
- Full Text
- View/download PDF
25. Vaccines to Prevent Coccidioidomycosis: A Gene-Deletion Mutant of Coccidioides Posadasii as a Viable Candidate for Human Trials.
- Author
-
Galgiani JN, Shubitz LF, Orbach MJ, Mandel MA, Powell DA, Klein BS, Robb EJ, Ohkura M, Seka DJ, Tomasiak TM, and Monath TP
- Abstract
Coccidioidomycosis is an endemic fungal infection that is reported in up to 20,000 persons per year and has an economic impact close to $1.5 billion. Natural infection virtually always confers protection from future exposure, and this suggests that a preventative vaccine strategy is likely to succeed. We here review progress toward that objective. There has been ongoing research to discover a coccidioidal vaccine over the past seven decades, including one phase III clinical trial, but for reasons of either efficacy or feasibility, a safe and effective vaccine has not yet been developed. This review first summarizes the past research to develop a coccidioidal vaccine. It then details the evidence that supports a live, gene-deletion vaccine candidate as suitable for further development as both a veterinary and a human clinical product. Finally, a plausible vaccine development plan is described which would be applicable to this vaccine candidate and also useful to other future candidates. The public health and economic impact of coccidioidomycosis fully justifies a public private partnership for vaccine development, and the development of a vaccine for this orphan disease will likely require some degree of public funding.
- Published
- 2022
- Full Text
- View/download PDF
26. Recombinant vesicular stomatitis vaccine against Nipah virus has a favorable safety profile: Model for assessment of live vaccines with neurotropic potential.
- Author
-
Monath TP, Nichols R, Tussey L, Scappaticci K, Pullano TG, Whiteman MD, Vasilakis N, Rossi SL, Campos RK, Azar SR, Spratt HM, Seaton BL, Archambault WT, Costecalde YV, Moore EH, Hawks RJ, and Fusco J
- Subjects
- Animals, Disease Models, Animal, Ebola Vaccines, Glycoproteins genetics, Hemorrhagic Fever, Ebola prevention & control, Humans, Mice, Nipah Virus genetics, Vaccines, Attenuated adverse effects, Vaccines, Synthetic adverse effects, Henipavirus Infections prevention & control, Vesicular Stomatitis prevention & control, Viral Vaccines adverse effects
- Abstract
Nipah virus (NiV) disease is a bat-borne zoonosis responsible for outbreaks with high lethality and is a priority for vaccine development. With funding from the Coalition of Epidemic Preparedness Innovations (CEPI), we are developing a chimeric vaccine (PHV02) composed of recombinant vesicular stomatitis virus (VSV) expressing the envelope glycoproteins of both Ebola virus (EBOV) and NiV. The EBOV glycoprotein (GP) mediates fusion and viral entry and the NiV attachment glycoprotein (G) is a ligand for cell receptors, and stimulates neutralizing antibody, the putative mediator of protection against NiV. PHV02 is identical in construction to the registered Ebola vaccine (Ervebo) with the addition of the NiV G gene. NiV ephrin B2 and B3 receptors are expressed on neural cells and the wild-type NiV is neurotropic and causes encephalitis in affected patients. It was therefore important to assess whether the NiV G alters tropism of the rVSV vector and serves as a virulence factor. PHV02 was fully attenuated in adult hamsters inoculated by the intramuscular (IM) route, whereas parental wild-type VSV was 100% lethal. Two rodent models (mice, hamsters) were infected by the intracerebral (IC) route with graded doses of PHV02. Comparator active controls in various experiments included rVSV-EBOV (representative of Ebola vaccine) and yellow fever (YF) 17DD commercial vaccine. These studies showed PHV02 to be more neurovirulent than both rVSV-EBOV and YF 17DD in infant animals. PHV02 was lethal for adult hamsters inoculated IC but not for adult mice. In contrast YF 17DD retained virulence for adult mice inoculated IC but was not virulent for adult hamsters. Because of the inconsistency of neurovirulence patterns in the rodent models, a monkey neurovirulence test (MNVT) was performed, using YF 17DD as the active comparator because it has a well-established profile of quantifiable microscopic changes in brain centers and a known reporting rate of neurotropic adverse events in humans. In the MNVT PHV02 was significantly less neurovirulent than the YF 17DD vaccine reference control, indicating that the vaccine will have an acceptable safety profile for humans. The findings are important because they illustrate the complexities of phenotypic assessment of novel viral vectors with tissue tropisms determined by transgenic proteins, and because it is unprecedented to use a heterologous comparator virus (YF vaccine) in a regulatory-enabling study. This approach may have value in future studies of other novel viral vectors., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: BLS is employed by Q2 Solutions and MDW by BioReliance. WTA is employed by VirtuStat, YVC is employed by AmplifyBio, and YVC, EHM, and RJH are employees of Battelle. JF, RN, and KS are employees of, and TPG is a consultant to Public Health Vaccines LLC (PHV), which holds licenses to patents covering aspects of the rVSV-Nipah technology from Public Health Canada and the (US) National Institutes of Health and is engaged in development of the PHV02 vaccine. TPM and LT are members of the PHV Board of Directors. TPM and LT are employees and shareholders of Crozet BioPharma LLC. JF, RN, and KS are also shareholders in Crozet
- Published
- 2022
- Full Text
- View/download PDF
27. A multitope SARS-CoV-2 vaccine provides long-lasting B cell and T cell immunity against Delta and Omicron variants.
- Author
-
Wang CY, Hwang KP, Kuo HK, Peng WJ, Shen YH, Kuo BS, Huang JH, Liu H, Ho YH, Lin F, Ding S, Liu Z, Wu HT, Huang CT, Lee YJ, Liu MC, Yang YC, Lu PL, Tsai HC, Lee CH, Shi ZY, Liu CE, Liao CH, Chang FY, Chen HC, Wang FD, Hou KL, Cheng J, Wang MS, Yang YT, Chiu HC, Jiang MH, Shih HY, Shen HY, Chang PY, Lan YR, Chen CT, Lin YL, Liang JJ, Liao CC, Chou YC, Morris MK, Hanson CV, Guirakhoo F, Hellerstein M, Yu HJ, King CC, Kemp T, Heppner DG, and Monath TP
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Antibodies, Neutralizing, Antibodies, Viral, Humans, Immunization, Passive, Middle Aged, SARS-CoV-2, T-Lymphocytes, Young Adult, COVID-19 Serotherapy, COVID-19 prevention & control, COVID-19 therapy, COVID-19 Vaccines
- Abstract
BackgroundThe Delta and Omicron variants of SARS-CoV-2 are currently responsible for breakthrough infections due to waning immunity. We report phase I/II trial results of UB-612, a multitope subunit vaccine containing S1-RBD-sFc protein and rationally designed promiscuous peptides representing sarbecovirus conserved helper T cell and cytotoxic T lymphocyte epitopes on the nucleocapsid (N), membrane (M), and spike (S2) proteins.MethodWe conducted a phase I primary 2-dose (28 days apart) trial of 10, 30, or 100 μg UB-612 in 60 healthy young adults 20 to 55 years old, and 50 of them were boosted with 100 μg of UB-612 approximately 7 to 9 months after the second dose. A separate placebo-controlled and randomized phase II study was conducted with 2 doses of 100 μg of UB-612 (n = 3,875, 18-85 years old). We evaluated interim safety and immunogenicity of phase I until 14 days after the third (booster) dose and of phase II until 28 days after the second dose.ResultsNo vaccine-related serious adverse events were recorded. The most common solicited adverse events were injection site pain and fatigue, mostly mild and transient. In both trials, UB-612 elicited respective neutralizing antibody titers similar to a panel of human convalescent sera. The most striking findings were long-lasting virus-neutralizing antibodies and broad T cell immunity against SARS-CoV-2 variants of concern (VoCs), including Delta and Omicron, and a strong booster-recalled memory immunity with high cross-reactive neutralizing titers against the Delta and Omicron VoCs.ConclusionUB-612 has presented a favorable safety profile, potent booster effect against VoCs, and long-lasting B and broad T cell immunity that warrants further development for both primary immunization and heterologous boosting of other COVID-19 vaccines.Trial RegistrationClinicalTrials.gov: NCT04545749, NCT04773067, and NCT04967742.FundingUBI Asia, Vaxxinity Inc., and Taiwan Centers for Disease Control, Ministry of Health and Welfare.
- Published
- 2022
- Full Text
- View/download PDF
28. A Randomized Dose-Escalating Phase I Trial of a Replication-Deficient Lymphocytic Choriomeningitis Virus Vector-Based Vaccine Against Human Cytomegalovirus.
- Author
-
Schwendinger M, Thiry G, De Vos B, Leroux-Roels G, Bruhwyler J, Huygens A, Ganeff C, Buchinger H, Orlinger KK, Pinschewer DD, Monath TP, and Lilja AE
- Subjects
- Adult, Antibodies, Neutralizing, Antibodies, Viral, Cytomegalovirus genetics, Humans, Immunization, Secondary, Lymphocytic choriomeningitis virus genetics, Cytomegalovirus Vaccines, Vaccines
- Abstract
Background: A vaccine (HB-101) consisting of 2 nonreplicating lymphocytic choriomeningitis virus (LCMV) vectors expressing the human cytomegalovirus antigens glycoprotein B (gB) and the 65-kD phosphoprotein (pp65), respectively, is in development to prevent cytomegalovirus infection., Methods: HB-101 was tested in cytomegalovirus-naive, healthy adults in a randomized, double-blind, placebo-controlled, dose-escalation Phase I trial. Fifty-four subjects received low, medium, or high dose of HB-101 or placebo by intramuscular administration at Month 0, 1, and 3. Safety and immunogenicity were the respective primary and secondary endpoints. Subjects were followed for 12 months after the initial immunization., Results: Vaccination was associated with transient mild to moderate adverse events. HB-101 administration induced dose-dependent gB- and pp65-specific cellular responses, dominated by pp65-specific CD8 T cells, a high fraction of which were polyfunctional. Two administrations were sufficient to elicit dose-dependent gB-binding and cytomegalovirus-neutralizing antibodies (Abs). Cytomegalovirus-specific immune responses were boosted after each administration. Only 1 of 42 vaccine recipients mounted a transient LCMV vector-neutralizing Ab response., Conclusions: HB-101 was well tolerated and induced cytomegalovirus-specific polyfunctional CD8 T-cell and neutralizing Ab responses in the majority of subjects. Lack of vector-neutralizing Ab responses should facilitate booster vaccinations. These results justify further clinical evaluation of this vaccine candidate., (© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America.)
- Published
- 2022
- Full Text
- View/download PDF
29. Transcriptomic signatures induced by the Ebola virus vaccine rVSVΔG-ZEBOV-GP in adult cohorts in Europe, Africa, and North America: a molecular biomarker study.
- Author
-
Vianello E, Gonzalez-Dias P, van Veen S, Engele CG, Quinten E, Monath TP, Medaglini D, Santoro F, Huttner A, Dubey S, Eichberg M, Ndungu FM, Kremsner PG, Essone PN, Agnandji ST, Siegrist CA, Nakaya HI, Ottenhoff THM, and Haks MC
- Subjects
- Adult, Africa, Antibodies, Viral, Biomarkers, Europe, Glycoproteins genetics, Humans, North America, Randomized Controlled Trials as Topic, Transcriptome, Vesiculovirus genetics, Ebola Vaccines adverse effects, Ebolavirus genetics, Hemorrhagic Fever, Ebola prevention & control, Vesicular Stomatitis chemically induced
- Abstract
Background: A recombinant vesicular stomatitis virus vector expressing the Zaire Ebola virus glycoprotein (rVSVΔG-ZEBOV-GP) vaccine has been reported as safe, immunogenic, and highly protective in a ring vaccination trial. We aimed to identify transcriptomic immune response biomarker signatures induced by vaccination and associated signatures with its immunogenicity and reactogenicity to better understand the potential mechanisms of action of the vaccine., Methods: 354 healthy adult volunteers were vaccinated in randomised, double-blind, placebo-controlled trials in Europe (Geneva, Switzerland [November, 2014, to January, 2015]) and North America (USA [Dec 5, 2014, to June 23, 2015]), and dose-escalation trials in Africa (Lambaréné, Gabon [November, 2014, to January, 2015], and Kilifi, Kenya [December, 2014, to January, 2015]) using different doses of the recombinant vesicular stomatitis virus vector expressing the Zaire Ebola virus glycoprotein (rVSVΔG-ZEBOV-GP; 3 × 10
5 to 1 × 108 plaque-forming units [pfu]). Longitudinal transcriptomic responses (days 0, 1, 2, 3, 7, 14, and 28) were measured in whole blood using a targeted gene expression profiling platform (dual-colour reverse-transcriptase multiplex ligation-dependent probe amplification) focusing on 144 immune-related genes. The effect of time and dose on transcriptomic response was also assessed. Logistic regression with lasso regularisation was applied to identify host signatures with optimal discriminatory capability of vaccination at day 1 or day 7 versus baseline, whereas random-effects models and recursive feature elimination combined with regularised logistic regression were used to associate signatures with immunogenicity and reactogenicity., Findings: Our results indicated that perturbation of gene expression peaked on day 1 and returned to baseline levels between day 7 and day 28. The magnitude of the response was dose-dependent, with vaccinees receiving a high dose (≥9 × 106 pfu) of rVSVΔG-ZEBOV-GP exhibiting the largest amplitude. The most differentially expressed genes that were significantly upregulated following vaccination consisted of type I and II interferon-related genes and myeloid cell-associated markers, whereas T cell, natural killer cell, and cytotoxicity-associated genes were downregulated. A gene signature associated with immunogenicity (common to all four cohorts) was identified correlating gene expression profiles with ZEBOV-GP antibody titres and a gene signatures associated with reactogenicity (Geneva cohort) was identified correlating gene expression profiles with an adverse event (ie, arthritis)., Interpretation: Collectively, our results identify and cross-validate immune-related transcriptomic signatures induced by rVSVΔG-ZEBOV-GP vaccination in four cohorts of adult participants from different genetic and geographical backgrounds. These signatures will aid in the rational development, testing, and evaluation of novel vaccines and will allow evaluation of the effect of host factors such as age, co-infection, and comorbidity on responses to vaccines., Funding: Innovative Medicines Initiative 2 Joint Undertaking., Competing Interests: Declaration of interests TPM is an employee of NewLink Genetics Corporation. SD and ME are employees of Merck Sharp & Dohme. All other authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
30. High dose of vesicular stomatitis virus-vectored Ebola virus vaccine causes vesicular disease in swine without horizontal transmission.
- Author
-
Morozov I, Monath TP, Meekins DA, Trujillo JD, Sunwoo SY, Urbaniak K, Kim IJ, Narayanan SK, Indran SV, Ma W, Wilson WC, O'Connor C, Dubey S, Troth SP, Coller BA, Nichols R, Martin BK, Feldmann H, and Richt JA
- Subjects
- Africa, Animals, Chlorocebus aethiops, Ebolavirus genetics, Female, Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola virology, Humans, Male, Models, Animal, RNA, Viral, Swine, Vaccination methods, Vaccines, Synthetic adverse effects, Vaccines, Synthetic immunology, Vero Cells, Vesiculovirus genetics, Ebola Vaccines adverse effects, Ebola Vaccines immunology, Ebolavirus immunology, Vesicular Stomatitis transmission, Vesicular Stomatitis virology, Vesicular stomatitis Indiana virus immunology, Vesiculovirus immunology
- Abstract
ABSTRACT The recent impact of Ebola virus disease (EVD) on public health in Africa clearly demonstrates the need for a safe and efficacious vaccine to control outbreaks and mitigate its threat to global health. ERVEBO® is an effective recombinant Vesicular Stomatitis Virus (VSV)-vectored Ebola virus vaccine (VSV-EBOV) that was approved by the FDA and EMA in late 2019 for use in prevention of EVD. Since the parental virus VSV, which was used to construct VSV-EBOV, is pathogenic for livestock and the vaccine virus may be shed at low levels by vaccinated humans, widespread deployment of the vaccine requires investigation into its infectivity and transmissibility in VSV-susceptible livestock species. We therefore performed a comprehensive clinical analysis of the VSV-EBOV vaccine virus in swine to determine its infectivity and potential for transmission. A high dose of VSV-EBOV resulted in VSV-like clinical signs in swine, with a proportion of pigs developing ulcerative vesicular lesions at the nasal injection site and feet. Uninoculated contact control pigs co-mingled with VSV-EBOV-inoculated pigs did not become infected or display any clinical signs of disease, indicating the vaccine is not readily transmissible to naïve pigs during prolonged close contact. In contrast, virulent wild-type VSV Indiana had a shorter incubation period and was transmitted to contact control pigs. These results indicate that the VSV-EBOV vaccine causes vesicular illness in swine when administered at a high dose. Moreover, the study demonstrates the VSV-EBOV vaccine is not readily transmitted to uninfected pigs, encouraging its safe use as an effective human vaccine.
- Published
- 2021
- Full Text
- View/download PDF
31. Introduction and Tribute to Charlie Calisher.
- Author
-
Monath TP and Murphy FA
- Abstract
It is a great pleasure to contribute a few words of introduction to this Special Issue of MDPI's Diseases entitled "Recent Studies of Arthropod-, Bat-, and Rodent-Borne Viruses: A Theme Issue in Honor of Professor Charles H [...].
- Published
- 2021
- Full Text
- View/download PDF
32. Immunogenicity and safety of fractional doses of yellow fever vaccines: a randomised, double-blind, non-inferiority trial.
- Author
-
Juan-Giner A, Kimathi D, Grantz KH, Hamaluba M, Kazooba P, Njuguna P, Fall G, Dia M, Bob NS, Monath TP, Barrett AD, Hombach J, Mulogo EM, Ampeire I, Karanja HK, Nyehangane D, Mwanga-Amumpaire J, Cummings DAT, Bejon P, Warimwe GM, and Grais RF
- Subjects
- Adult, Antibodies, Neutralizing immunology, Antibodies, Neutralizing isolation & purification, Double-Blind Method, Female, Humans, Kenya, Male, Seroconversion, Uganda, Yellow Fever prevention & control, Yellow Fever Vaccine adverse effects, Yellow Fever Vaccine immunology, Off-Label Use, Yellow Fever Vaccine administration & dosage
- Abstract
Background: Stocks of yellow fever vaccine are insufficient to cover exceptional demands for outbreak response. Fractional dosing has shown efficacy, but evidence is limited to the 17DD substrain vaccine. We assessed the immunogenicity and safety of one-fifth fractional dose compared with standard dose of four WHO-prequalified yellow fever vaccines produced from three substrains., Methods: We did this randomised, double-blind, non-inferiority trial at research centres in Mbarara, Uganda, and Kilifi, Kenya. Eligible participants were aged 18-59 years, had no contraindications for vaccination, were not pregnant or lactating, had no history of yellow fever vaccination or infection, and did not require yellow fever vaccination for travel. Eligible participants were recruited from communities and randomly assigned to one of eight groups, corresponding to the four vaccines at standard or fractional dose. The vaccine was administered subcutaneously by nurses who were not masked to treatment, but participants and other study personnel were masked to vaccine allocation. The primary outcome was proportion of participants with seroconversion 28 days after vaccination. Seroconversion was defined as post-vaccination neutralising antibody titres at least 4 times pre-vaccination measurement measured by 50% plaque reduction neutralisation test (PRNT
50 ). We defined non-inferiority as less than 10% decrease in seroconversion in fractional compared with standard dose groups 28 days after vaccination. The primary outcome was measured in the per-protocol population, and safety analyses included all vaccinated participants. This trial is registered with ClinicalTrials.gov, NCT02991495., Findings: Between Nov 6, 2017, and Feb 21, 2018, 1029 participants were assessed for inclusion. 69 people were ineligible, and 960 participants were enrolled and randomly assigned to vaccine manufacturer and dose (120 to Bio-Manguinhos-Fiocruz standard dose, 120 to Bio-Manguinhos-Fiocruz fractional dose, 120 to Chumakov Institute of Poliomyelitis and Viral Encephalitides standard dose, 120 to Chumakov Institute of Poliomyelitis and Viral Encephalitides fractional dose, 120 to Institut Pasteur Dakar standard dose, 120 to Institut Pasteur Dakar fractional dose, 120 to Sanofi Pasteur standard dose, and 120 to Sanofi Pasteur fractional dose). 49 participants had detectable PRNT50 at baseline and 11 had missing PRNT50 results at baseline or 28 days. 900 were included in the per-protocol analysis. 959 participants were included in the safety analysis. The absolute difference in seroconversion between fractional and standard doses by vaccine was 1·71% (95% CI -2·60 to 5·28) for Bio-Manguinhos-Fiocruz, -0·90% (-4·24 to 3·13) for Chumakov Institute of Poliomyelitis and Viral Encephalitides, 1·82% (-2·75 to 5·39) for Institut Pasteur Dakar, and 0·0% (-3·32 to 3·29) for Sanofi Pasteur. Fractional doses from all four vaccines met the non-inferiority criterion. The most common treatment-related adverse events were headache (22·2%), fatigue (13·7%), myalgia (13·3%) and self-reported fever (9·0%). There were no study-vaccine related serious adverse events., Interpretation: Fractional doses of all WHO-prequalified yellow fever vaccines were non-inferior to the standard dose in inducing seroconversion 28 days after vaccination, with no major safety concerns. These results support the use of fractional dosage in the general adult population for outbreak response in situations of vaccine shortage., Funding: The study was funded by Médecins Sans Frontières Foundation, Wellcome Trust (grant no. 092654), and the UK Department for International Development. Vaccines were donated in kind., (Copyright © 2021 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
33. The Brighton Collaboration standardized template for collection of key information for benefit-risk assessment of viral vector vaccines.
- Author
-
Condit RC, Kim D, Robertson JS, Excler JL, Gurwith M, Monath TP, Pavlakis G, Fast PE, Smith J, Smith ER, Chen RT, and Kochhar S
- Subjects
- Animals, Genetic Vectors, Humans, Internet, Risk Assessment, Drug Evaluation, Preclinical standards, Vaccines, Attenuated adverse effects, Viral Vaccines adverse effects
- Abstract
Many of the vaccines under development for COVID-19 involve the use of viral vectors. The Brighton Collaboration Benefit-Risk Assessment of Vaccines by Technology (BRAVATO, formerly the Viral Vector Vaccine Safety Working Group, V3SWG) working group has prepared a standardized template to describe the key considerations for the benefit-risk assessment of viral vector vaccines. This will facilitate key stakeholders to anticipate potential safety issues and interpret or assess safety data. This would also help improve communication and public acceptance of licensed viral vector vaccines., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper, (Copyright © 2020 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
34. The Origin of COVID-19 and Why It Matters.
- Author
-
Morens DM, Breman JG, Calisher CH, Doherty PC, Hahn BH, Keusch GT, Kramer LD, LeDuc JW, Monath TP, and Taubenberger JK
- Subjects
- Animals, Betacoronavirus classification, Betacoronavirus genetics, COVID-19, Chiroptera virology, Coronavirus Infections prevention & control, Coronavirus Infections transmission, Humans, Pandemics prevention & control, Pneumonia, Viral prevention & control, Pneumonia, Viral transmission, Public Health, SARS-CoV-2, Betacoronavirus isolation & purification, Coronavirus Infections etiology, Pneumonia, Viral etiology
- Abstract
The COVID-19 pandemic is among the deadliest infectious diseases to have emerged in recent history. As with all past pandemics, the specific mechanism of its emergence in humans remains unknown. Nevertheless, a large body of virologic, epidemiologic, veterinary, and ecologic data establishes that the new virus, SARS-CoV-2, evolved directly or indirectly from a β-coronavirus in the sarbecovirus (SARS-like virus) group that naturally infect bats and pangolins in Asia and Southeast Asia. Scientists have warned for decades that such sarbecoviruses are poised to emerge again and again, identified risk factors, and argued for enhanced pandemic prevention and control efforts. Unfortunately, few such preventive actions were taken resulting in the latest coronavirus emergence detected in late 2019 which quickly spread pandemically. The risk of similar coronavirus outbreaks in the future remains high. In addition to controlling the COVID-19 pandemic, we must undertake vigorous scientific, public health, and societal actions, including significantly increased funding for basic and applied research addressing disease emergence, to prevent this tragic history from repeating itself.
- Published
- 2020
- Full Text
- View/download PDF
35. The Brighton Collaboration standardized template for collection of key information for benefit-risk assessment of nucleic acid (RNA and DNA) vaccines.
- Author
-
Kim D, Robertson JS, Excler JL, Condit RC, Fast PE, Gurwith M, Pavlakis G, Monath TP, Smith J, Wood D, Smith ER, Chen RT, and Kochhar S
- Subjects
- COVID-19 Vaccines, Coronavirus Infections genetics, Coronavirus Infections prevention & control, Humans, Public Opinion, Risk Assessment standards, Vaccines, DNA genetics, Viral Vaccines adverse effects, Risk Assessment methods, Vaccines, DNA adverse effects, Vaccines, DNA standards, Viral Vaccines genetics, Viral Vaccines standards
- Abstract
Nucleic acid (DNA and RNA) vaccines are among the most advanced vaccines for COVID-19 under development. The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) has prepared a standardized template to describe the key considerations for the benefit-risk assessment of nucleic acid vaccines. This will facilitate the assessment by key stakeholders of potential safety issues and understanding of overall benefit-risk. The structured assessment provided by the template can also help improve communication and public acceptance of licensed nucleic acid vaccines., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
36. Theoretical risk of genetic reassortment should not impede development of live, attenuated Rift Valley fever (RVF) vaccines commentary on the draft WHO RVF Target Product Profile.
- Author
-
Monath TP, Kortekaas J, Watts DM, Christofferson RC, Desiree LaBeaud A, Gowen B, Peters CJ, Smith DR, Swanepoel R, Morrill JC, Ksiazek TG, Pittman PR, Bird BH, and Bettinger G
- Abstract
In November 2019, The World Health Organization (WHO) issued a draft set of Target Product Profiles (TPPs) describing optimal and minimally acceptable targets for vaccines against Rift Valley fever (RVF), a Phlebovirus with a three segmented genome, in both humans and ruminants. The TPPs contained rigid requirements to protect against genomic reassortment of live, attenuated vaccines (LAVs) with wild-type RVF virus (RVFV), which place undue constraints on development and regulatory approval of LAVs. We review the current LAVs in use and in development, and conclude that there is no evidence that reassortment between LAVs and wild-type RVFV has occurred during field use, that such a reassortment event if it occurred would have no untoward consequence, and that the TPPs should be revised to provide a more balanced assessment of the benefits versus the theoretical risks of reassortment., Competing Interests: The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Dr. Monath is a consultant to the Sabin Vaccine Institute, which owns the license to the MP-12 live attenuated RVFV vaccine. Dr. Bird is developing a live, attenuated Rift Valley fever vaccine, DDVax®, with funding from the Coalition for Epidemic Preparedness Innovations. Dr. Kortekaas is developing the RVFV-4s live attenuated vaccine against RVFV, with funding from the Coalition for Epidemic Preparedness Innovations. Drs. Watts is developing a live attenuated MP12-NSm deletion veterinary vaccine against RVFV, with support from USAID, and is collaborating with the vaccine manufacturer, M.C.I Santé Animale, Mohammedia, Morocco. Drs. Peters, Morrill, Bettinger, and Pittman were formerly engaged in development of the MP-12 live RVFV vaccine, with support from the US Army; however, there are no current financial relationships related to this work., (© 2020 The Author(s).)
- Published
- 2020
- Full Text
- View/download PDF
37. Comparative evaluation of pathogenicity of three isolates of vesicular stomatitis virus (Indiana serotype) in pigs.
- Author
-
Morozov I, Davis AS, Ellsworth S, Trujillo JD, McDowell C, Shivanna V, Dasen EJ, Nichols R, Martin BK, Monath TP, and Richt JA
- Subjects
- Animal Structures pathology, Animal Structures virology, Animals, Blood virology, Serogroup, Swine, Vesiculovirus classification, Virulence, Virus Replication, Virus Shedding, Disease Models, Animal, Vesicular Stomatitis pathology, Vesicular Stomatitis virology, Vesiculovirus growth & development, Vesiculovirus pathogenicity
- Abstract
Vesicular stomatitis (VS) is a notifiable disease of livestock affecting cattle, horses, pigs and humans. Vesicular stomatitis virus (VSV) serotypes Indiana and New Jersey are endemic to Central America; however, they also cause sporadic and scattered outbreaks in various countries in South and North America, including the USA. In order to develop an effective experimental challenge model for VSV, we compared the pathogenicity of three VSV serotype Indiana isolates in 36 4-5 week-old pigs. Two bovine isolates of Central American origin and one equine isolate from the USA were used for the experimental infections. Each pig was inoculated with a single isolate by both the intradermal and intranasal routes. Clinical signs of VSV infection were recorded daily for 10 days post-inoculation (days p.i.). Nasal and tonsillar swab samples and blood were collected to monitor immune responses, virus replication and shedding. Post-challenge, characteristic signs of VS were observed, including vesicles on the nasal planum and coronary bands, lameness, loss of hoof walls and pyrexia. Pigs inoculated with the Central American isolates showed consistently more severe clinical signs in comparison to the pigs infected with the USA isolate. Genomic RNA was isolated from the original challenge virus stocks, sequenced and compared to VSV genomes available in GenBank. Comparative genome analysis demonstrated significant differences between the VSV isolate from the USA and the two Central American isolates. Our results indicate that the Central American isolates of VSV serotype Indiana used in this study are more virulent in swine than the USA VSV serotype Indiana isolate and represent good candidate challenge strains for future VSV studies.
- Published
- 2019
- Full Text
- View/download PDF
38. Summit proceedings: Biomedical countermeasure development for emerging vector-borne viral diseases.
- Author
-
Blackman MA, Marchionni MA, Gilly J, Hepburn M, Innis BL, Barrett ADT, Kester KE, Mascola JR, Cummings JF, Monath TP, Cassetti MC, Kim JH, Saville M, and Thomas SJ
- Subjects
- Animals, Clinical Trials as Topic, Disease Models, Animal, Drug Evaluation, Preclinical, Female, Humans, Influenza, Human etiology, Influenza, Human prevention & control, Pregnancy, Pregnancy Complications, Infectious etiology, Translational Research, Biomedical, Viral Vaccines therapeutic use, Zika Virus Infection etiology, Zika Virus Infection prevention & control, Communicable Diseases, Emerging prevention & control, Disease Vectors, Viral Vaccines pharmacology, Virus Diseases prevention & control
- Abstract
Emerging and re-emerging infectious diseases are an expanding global threat to public health, security, and economies. Increasing populations, urbanization, deforestation, climate change, anti-vaccination movements, war, and international travel are some of the contributing factors to this trend. The recent Ebola, MERS-CoV, and Zika outbreaks demonstrated we are insufficiently prepared to respond with proven safe and effective countermeasures (i.e., vaccines and therapeutics). The State University of New York Upstate Medical University and the Trudeau Institute convened a summit of key opinion and thought leaders in the life sciences and biomedical research and development enterprises to explore global biopreparedness challenges, take an inventory of existing capabilities and capacities related to preparation and response, assess current "gaps," and prospect what could be done to improve our position. Herein we describe the summit proceedings, "Translational Immunology Supporting Biomedical Countermeasure Development for Emerging Vector-borne Viral Diseases," held October 2-3, 2018, at the Trudeau Institute in Saranac Lake, NY., (Copyright © 2019.)
- Published
- 2019
- Full Text
- View/download PDF
39. A short history of Lassa fever: the first 10-15 years after discovery.
- Author
-
Monath TP
- Subjects
- Animals, Antibodies, Viral, Arenavirus genetics, Cross Infection, Disease Outbreaks, Genetic Variation, History, 20th Century, History, 21st Century, Humans, Nigeria, Phylogeny, Rodentia virology, Sierra Leone, Vaccination, Zoonoses, Lassa Fever history, Lassa Fever physiopathology, Lassa Fever therapy, Lassa Fever transmission, Lassa virus genetics, Lassa virus pathogenicity
- Abstract
This brief review is focused on the events surrounding the initial discovery of a new viral hemorrhagic fever in 1969 and the subsequent 10-15 years during which a substantial understanding of the disease was gained. In 1969, a series of sequential life-threating or fatal infections occurred among health care workers in Nigeria and the laboratory scientist who isolated and characterized the causative agent. The agent, Lassa virus was named after the geographical location of the first recognized human case. The new virus was shown to be related to lymphocytic choriomeningitis and to previously unclassified neotropical viruses, including Argentine and Bolivian hemorrhagic fevers, and a new taxonomic grouping, the Arenaviruses, was proposed. In 1970-72, three further epidemics occurred in Nigeria, Liberia and Sierra Leone, the first two involved nosocomial transmission, and the third was a community-based outbreak, during which the rodent reservoir host was identified. In 1976, a long-term research project commenced in Sierra Leone, which produced a rich body of data from prospectively designed studies on the clinical features, transmission, and treatment of the disease., (Copyright © 2019. Published by Elsevier B.V.)
- Published
- 2019
- Full Text
- View/download PDF
40. rVSVΔG-ZEBOV-GP (also designated V920) recombinant vesicular stomatitis virus pseudotyped with Ebola Zaire Glycoprotein: Standardized template with key considerations for a risk/benefit assessment.
- Author
-
Monath TP, Fast PE, Modjarrad K, Clarke DK, Martin BK, Fusco J, Nichols R, Heppner DG, Simon JK, Dubey S, Troth SP, Wolf J, Singh V, Coller BA, and Robertson JS
- Abstract
The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to evaluate the safety and characteristics of live, recombinant viral vector vaccines. A recent publication by the V3SWG described live, attenuated, recombinant vesicular stomatitis virus (rVSV) as a chimeric virus vaccine for HIV-1 (Clarke et al., 2016). The rVSV vector system is being explored as a platform for development of multiple vaccines. This paper reviews the molecular and biological features of the rVSV vector system, followed by a template with details on the safety and characteristics of a rVSV vaccine against Zaire ebolavirus (ZEBOV). The rVSV-ZEBOV vaccine is a live, replication competent vector in which the VSV glycoprotein (G) gene is replaced with the glycoprotein (GP) gene of ZEBOV. Multiple copies of GP are expressed and assembled into the viral envelope responsible for inducing protective immunity. The vaccine (designated V920) was originally constructed by the National Microbiology Laboratory, Public Health Agency of Canada, further developed by NewLink Genetics Corp. and Merck & Co., and is now in final stages of registration by Merck. The vaccine is attenuated by deletion of the principal virulence factor of VSV (the G protein), which also removes the primary target for anti-vector immunity. The V920 vaccine caused no toxicities after intramuscular (IM) or intracranial injection of nonhuman primates and no reproductive or developmental toxicity in a rat model. In multiple studies, cynomolgus macaques immunized IM with a wide range of virus doses rapidly developed ZEBOV-specific antibodies measured in IgG ELISA and neutralization assays and were fully protected against lethal challenge with ZEBOV virus. Over 20,000 people have received the vaccine in clinical trials; the vaccine has proven to be safe and well tolerated. During the first few days after vaccination, many vaccinees experience a mild acute-phase reaction with fever, headache, myalgia, and arthralgia of short duration; this period is associated with a low-level viremia, activation of anti-viral genes, and increased levels of chemokines and cytokines. Oligoarthritis and rash appearing in the second week occur at a low incidence, and are typically mild-moderate in severity and self-limited. V920 vaccine was used in a Phase III efficacy trial during the West African Ebola epidemic in 2015, showing 100% protection against Ebola Virus Disease, and it has subsequently been deployed for emergency control of Ebola outbreaks in central Africa. The template provided here provides a comprehensive picture of the first rVSV vector to reach the final stage of development and to provide a solution to control of an alarming human disease.
- Published
- 2019
- Full Text
- View/download PDF
41. Determinants of antibody persistence across doses and continents after single-dose rVSV-ZEBOV vaccination for Ebola virus disease: an observational cohort study.
- Author
-
Huttner A, Agnandji ST, Combescure C, Fernandes JF, Bache EB, Kabwende L, Ndungu FM, Brosnahan J, Monath TP, Lemaître B, Grillet S, Botto M, Engler O, Portmann J, Siegrist D, Bejon P, Silvera P, Kremsner P, and Siegrist CA
- Subjects
- Adult, Cohort Studies, Female, Humans, Kenya, Male, Middle Aged, Switzerland, Antibodies, Viral blood, Dose-Response Relationship, Drug, Ebola Vaccines immunology, Ebola Vaccines therapeutic use, Ebolavirus drug effects, Hemorrhagic Fever, Ebola drug therapy, Medication Adherence
- Abstract
Background: The recombinant vesicular stomatitis virus (rVSV) vaccine expressing the Zaire Ebola virus (ZEBOV) glycoprotein is efficacious in the weeks following single-dose injection, but duration of immunity is unknown. We aimed to assess antibody persistence at 1 and 2 years in volunteers who received single-dose rVSV-ZEBOV in three previous trials., Methods: In this observational cohort study, we prospectively followed-up participants from the African and European phase 1 rVSV-ZEBOV trials, who were vaccinated once in 2014-15 with 300 000 (low dose) or 10-50 million (high dose) plaque-forming units (pfu) of rVSV-ZEBOV vaccine to assess ZEBOV glycoprotein (IgG) antibody persistence. The primary outcome was ZEBOV glycoprotein-specific IgG geometric mean concentrations (GMCs) measured yearly by ELISA compared with 1 month (ie, 28 days) after immunisation. We report GMCs up to 2 years (Geneva, Switzerland, including neutralising antibodies up to 6 months) and 1 year (Lambaréné, Gabon; Kilifi, Kenya) after vaccination and factors associated with higher antibody persistence beyond 6 months, according to multivariable analyses. Trials and the observational study were registered at ClinicalTrials.gov (Geneva: NCT02287480 and NCT02933931; Kilifi: NCT02296983) and the Pan-African Clinical Trials Registry (Lambaréné PACTR201411000919191)., Findings: Of 217 vaccinees from the original studies (102 from the Geneva study, 75 from the Lambaréné study, and 40 from the Kilifi study), 197 returned and provided samples at 1 year (95 from the Geneva study, 63 from the Lambaréné, and 39 from the Kilifi study) and 90 at 2 years (all from the Geneva study). In the Geneva group, 44 (100%) of 44 participants who had been given a high dose (ie, 10-50 million pfu) of vaccine and who were seropositive at day 28 remained seropositive at 2 years, whereas 33 (89%) of 37 who had been given the low dose (ie, 300 000 pfu) remained seropositive for 2 years (p=0·042). In participants who had received a high dose, ZEBOV glycoprotein IgG GMCs decreased significantly between their peak (at 1-3 months) and month 6 after vaccination in Geneva (p<0·0001) and Lambaréné (p=0·0298) but not in Kilifi (p=0·5833) and subsequently remained stable at all sites apart from Geneva, where GMC in those given a high dose of vaccine increased significantly between 6 months and 1 year (p=0·0264). Antibody persistence was similar at 1 year and at 6 months in those who had received a low dose of vaccine, with lower titres among participants from the Geneva study at 2 years than at 1 year after vaccination (GMC ratio 0·61, 95% CI 0·49-0·77; p<0·0001). In multivariable analyses, predictors of increased IgG GMCs beyond 6 months included high-dose versus low-dose vaccination (Geneva p=0·0133; Lambaréné p=0·008) and vaccine-related arthritis (p=0·0176), but not sex, age, or baseline seropositivity (all p>0·05). Neutralising antibodies seem to be less durable, with seropositivity dropping from 64-71% at 28 days to 27-31% at 6 months in participants from the Geneva study., Interpretation: Antibody responses to single-dose rVSV-ZEBOV vaccination are sustained across dose ranges and settings, a key criterion in countries where booster vaccinations would be impractical., Funding: The Wellcome Trust and Innovative Medicines Initiative 2 Joint Undertaking., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
42. Bunyavirus Taxonomy: Limitations and Misconceptions Associated with the Current ICTV Criteria Used for Species Demarcation.
- Author
-
Blitvich BJ, Beaty BJ, Blair CD, Brault AC, Dobler G, Drebot MA, Haddow AD, Kramer LD, LaBeaud AD, Monath TP, Mossel EC, Plante K, Powers AM, Tesh RB, Turell MJ, Vasilakis N, and Weaver SC
- Subjects
- Bunyaviridae genetics, Bunyaviridae pathogenicity, Bunyaviridae Infections diagnosis, Guidelines as Topic, Humans, International Agencies, Species Specificity, Terminology as Topic, Bunyaviridae classification, Bunyaviridae Infections virology, Genome, Viral, Mosquito Vectors virology, Phylogeny
- Abstract
The International Committee on Taxonomy of Viruses (ICTV) has implemented numerous changes to the taxonomic classification of bunyaviruses over the years. Whereas most changes have been justified and necessary because of the need to accommodate newly discovered and unclassified viruses, other changes are a cause of concern, especially the decision to demote scores of formerly recognized species to essentially strains of newly designated species. This practice was first described in the seventh taxonomy report of the ICTV and has continued in all subsequent reports. In some instances, viruses that share less than 75% nucleotide sequence identity across their genomes, produce vastly different clinical presentations, possess distinct vector and host associations, have different biosafety recommendations, and occur in nonoverlapping geographic regions are classified as strains of the same species. Complicating the matter is the fact that virus strains have been completely eliminated from ICTV reports; thus, critically important information on virus identities and their associated biological and epidemiological features cannot be readily related to the ICTV classification. Here, we summarize the current status of bunyavirus taxonomy and discuss the adverse consequences associated with the reclassification and resulting omission of numerous viruses of public health importance from ICTV reports. As members of the American Committee on Arthropod-borne Viruses, we encourage the ICTV Bunyavirus Study Group to reconsider their stance on bunyavirus taxonomy, to revise the criteria currently used for species demarcation, and to list additional strains of public and veterinary importance.
- Published
- 2018
- Full Text
- View/download PDF
43. Assessment of the ability of V920 recombinant vesicular stomatitis-Zaire ebolavirus vaccine to replicate in relevant arthropod cell cultures and vector species.
- Author
-
Bergren NA, Miller MR, Monath TP, and Kading RC
- Subjects
- Aedes immunology, Aedes virology, Animals, Arthropods virology, Chlorocebus aethiops, Culex immunology, Culex virology, Hemorrhagic Fever, Ebola prevention & control, Humans, Vero Cells, Vesicular Stomatitis immunology, Vesicular Stomatitis prevention & control, Vesicular Stomatitis virology, Arthropods immunology, Ebola Vaccines immunology, Ebolavirus immunology, Hemorrhagic Fever, Ebola immunology, Mosquito Vectors immunology, Vaccines, Synthetic immunology, Viral Envelope Proteins immunology
- Abstract
V920, rVSVΔG-ZEBOV-GP, is a recombinant vesicular stomatitis-Zaire ebolavirus vaccine which has shown an acceptable safety profile and provides a protective immune response against Ebola virus disease (EVD) induced by Zaire ebolavirus in humans. The purpose of this study was to determine whether the V920 vaccine is capable of replicating in arthropod cell cultures of relevant vector species and of replicating in live mosquitoes. While the V920 vaccine replicated well in Vero cells, no replication was observed in Anopheles or Aedes mosquito, Culicoides biting midge, or Lutzomyia sand fly cells, nor in live Culex or Aedes mosquitoes following exposure through intrathoracic inoculation or feeding on a high-titer infectious blood meal. The insect taxa selected for use in this study represent actual and potential epidemic vectors of VSV. V920 vaccine inoculated into Cx. quinquefasciatus and Ae. aegypti mosquitoes demonstrated persistence of replication-competent virus following inoculation, consistent with the recognized biological stability of the vaccine, but no evidence for active virus replication in live mosquitoes was observed. Following administration of an infectious blood meal to Ae. aegypti and Cx. quinquefasciatus mosquitoes at a titer several log
10 PFU more concentrated than would be observed in vaccinated individuals, no infection or dissemination of V920 was observed in either mosquito species. In vitro and in vivo data gathered during this study support minimal risk of the vector-borne potential of the V920 vaccine.- Published
- 2018
- Full Text
- View/download PDF
44. Clinical development of a recombinant Ebola vaccine in the midst of an unprecedented epidemic.
- Author
-
Coller BG, Blue J, Das R, Dubey S, Finelli L, Gupta S, Helmond F, Grant-Klein RJ, Liu K, Simon J, Troth S, VanRheenen S, Waterbury J, Wivel A, Wolf J, Heppner DG, Kemp T, Nichols R, and Monath TP
- Subjects
- Adolescent, Adult, Africa epidemiology, Child, Clinical Trials as Topic, Ebolavirus genetics, Europe epidemiology, Hemorrhagic Fever, Ebola epidemiology, Hemorrhagic Fever, Ebola mortality, Hemorrhagic Fever, Ebola therapy, Humans, Immunogenicity, Vaccine, Treatment Outcome, United States epidemiology, Vaccines, Attenuated immunology, Vaccines, Synthetic immunology, Vesiculovirus genetics, Vesiculovirus immunology, Viral Envelope Proteins immunology, Ebola Vaccines immunology, Ebolavirus immunology, Epidemics prevention & control, Hemorrhagic Fever, Ebola prevention & control, Viral Envelope Proteins genetics
- Abstract
The 2014-2016 Ebola outbreak caused over 28,000 cases and 11,000 deaths. Merck & Co. Inc., Kenilworth, NJ USA and NewLink Genetics are working with private and public partners to develop and license an Ebola vaccine that was evaluated extensively during the outbreak. The vaccine referred to as V920 is a recombinant vesicular stomatitis virus (rVSV) in which the VSV-G envelope glycoprotein (GP) is completely replaced by the Zaire ebolavirus GP (rVSVΔG-ZEBOV-GP). Eight Phase I and four Phase II/III clinical trials enrolling approximately 17,000 subjects were conducted in parallel to the outbreak to assess the safety, immunogenicity, and/or efficacy of V920. Immunogenicity data demonstrate that anti-GP antibodies are generally detectable by ELISA by 14days postvaccination with up to 100% seroconversion observed by 28days post dose. In addition, the results of a ring vaccination trial conducted by the WHO and their partners in Guinea suggest robust vaccine efficacy within 10days of receipt of a single dose of vaccine. The vaccine is generally well-tolerated when administered to healthy, non-pregnant adults. The development of this vaccine candidate in the context of this unprecedented epidemic has involved the close cooperation of large number of international partners and highlights what we as a public health community can accomplish when working together towards a common goal. Study identification: V920-001 to V920-012. CLINICALTRIALS.GOV identifiers: NCT02269423; NCT02280408; NCT02374385; NCT02314923; NCT02287480; NCT02283099; NCT02296983; NCT02344407; NCT02378753; NCT02503202., (Copyright © 2017. Published by Elsevier Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
45. Assessing the safety and immunogenicity of recombinant vesicular stomatitis virus Ebola vaccine in healthy adults: a randomized clinical trial.
- Author
-
ElSherif MS, Brown C, MacKinnon-Cameron D, Li L, Racine T, Alimonti J, Rudge TL, Sabourin C, Silvera P, Hooper JW, Kwilas SA, Kilgore N, Badorrek C, Ramsey WJ, Heppner DG, Kemp T, Monath TP, Nowak T, McNeil SA, Langley JM, and Halperin SA
- Subjects
- Adolescent, Adult, Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, Canada, Double-Blind Method, Ebolavirus, Female, Healthy Volunteers, Humans, Immunoglobulin G blood, Male, Membrane Glycoproteins genetics, Middle Aged, Regression Analysis, Vaccination methods, Vaccines, Synthetic administration & dosage, Vesicular stomatitis Indiana virus, Viral Envelope Proteins genetics, Young Adult, Ebola Vaccines administration & dosage, Hemorrhagic Fever, Ebola prevention & control, Membrane Glycoproteins immunology, Viral Envelope Proteins immunology
- Abstract
Background: The 2013-2016 Ebola virus outbreak in West Africa was the most widespread in history. In response, alive attenuated recombinant vesicular stomatitis virus (rVSV) vaccine expressing Zaire Ebolavirus glycoprotein (rVSVΔG-ZEBOV-GP) was evaluated in humans., Methods: In a phase 1, randomized, dose-ranging, observer-blind, placebo-controlled trial, healthy adults aged 18-65 years were randomized into 4 groups of 10 to receive one of 3 vaccine doses or placebo. Follow-up visits spanned 180 days postvaccination for safety monitoring, immunogenicity testing and any rVSV virus shedding., Results: Forty participants were injected with rVSVΔG-ZEBOV-GP vaccine ( n = 30) or saline placebo ( n = 10). No serious adverse events related to the vaccine or participant withdrawals were reported. Solicited adverse events during the 14-day follow-up period were mild to moderate and self-limited, with the exception of injection-site pain and headache. Viremia following vaccination was transient and no longer detectable after study day 3, with no virus shedding in saliva or urine. All vaccinated participants developed serum immunoglobulin G (IgG), as measured by Ebola virus envelope glycoprotein-based enzyme-linked immunosorbent assay (ELISA). Immunogenicity was comparable across all dose groups, and sustained IgG titers were detectable through to the last visit, at study day 180., Interpretation: In this phase 1 study, there were no safety concerns after a single dose of rVSVΔG-ZEBOV-GP vaccine. IgG ELISA showed persistent high titers at 180 days postimmunization. There was a period of reactogenicity, but in general, the vaccine was well tolerated. This study provides evidence of the safety and immunogenicity of rVSVΔG-ZEBOV-GP vaccine and importance of its further investigation. Trial registration: Clinical-Trials.gov no., NCT02374385., Competing Interests: Competing interests: May ElSherif reports grants from the Canadian Institutes of Health Research/Public Health Agency of Canada, during the conduct of the study. Donna MacKinnon-Cameron reports an operating grant from the Canadian Institutes of Health Research/Public Health Agency of Canada, during the conduct of the study. Li Li reports an operating grant from the Canadian Institutes of Health Research/Public Health Agency of Canada, during the conduct of the study. Judie Alimonti reports personal fees from NewLink Genetics, outside the submitted work; and Her Majesty the Queen in right of Canada holds a patent related to the rVSVΔG-ZEBOV-GP vaccine (patents: EU 1 527 087; US 8 012 489; CA 2,493,142). W. Jay Ramsey was an employee of NewLink Genetics during the conduct of the study, and reports personal fees as a salaried employee of NewLink Genetics, outside the submitted work. Donald Gray Heppner reports grants from the Department of Health and Human Services, during the conduct of the study and employment by NewLink Genetics Inc. (which is developing the vaccine described in this article), outside the submitted work. Tracy Kemp was an employee of NewLink Genetics during the conduct of the study; she also reports personal fees as a salaried employee of NewLink Genetics, outside the submitted work. Thomas Monath reports a grant from BARDA and the US Department of Health and Human Services, and financial support for study, personal salary and benefits from New-Link Genetics. Shelly McNeil reports an operating grant from the Canadian Institutes of Health Research/Public Health Agency of Canada, during the conduct of the study; and grants and personal fees from vaccine manufacturers (such as Merck, GlaxoSmithKline Biologicals, Sanofi Pasteur and Pfizer) outside the submitted work. Joanne Langley reports an operating grant from the Canadian Institutes of Health Research/Public Health Agency of Canada, during the conduct of the study, as well as grants and personal fees from vaccine manufacturers (such as Merck, Glaxo-SmithKline Biologicals, Sanofi Pasteur, Pfizer, and Novartis) to conduct vaccine clinical trials, outside the submitted work. Scott Halperin reports an operating grant from the Canadian Institutes of Health Research/Public Health Agency of Canada, during the conduct of the study, and grants and personal fees from vaccine manufacturers (such as Merck, GlaxoSmithKline Biologicals, Sanofi Pasteur, Pfizer and Novartis) outside the submitted work. No other competing interests were declared., (© 2017 Canadian Medical Association or its licensors.)
- Published
- 2017
- Full Text
- View/download PDF
46. Epidemic arboviral diseases: priorities for research and public health.
- Author
-
Wilder-Smith A, Gubler DJ, Weaver SC, Monath TP, Heymann DL, and Scott TW
- Subjects
- Aedes virology, Animals, Dengue Virus, Global Health, Humans, Vaccines economics, Vaccines immunology, Zika Virus, Arbovirus Infections epidemiology, Arbovirus Infections prevention & control, Epidemics prevention & control, Public Health economics, Research economics
- Abstract
For decades, arboviral diseases were considered to be only minor contributors to global mortality and disability. As a result, low priority was given to arbovirus research investment and related public health infrastructure. The past five decades, however, have seen an unprecedented emergence of epidemic arboviral diseases (notably dengue, chikungunya, yellow fever, and Zika virus disease) resulting from the triad of the modern world: urbanisation, globalisation, and international mobility. The public health emergency of Zika virus, and the threat of global spread of yellow fever, combined with the resurgence of dengue and chikungunya, constitute a wake-up call for governments, academia, funders, and WHO to strengthen programmes and enhance research in aedes-transmitted diseases. The common features of these diseases should stimulate similar research themes for diagnostics, vaccines, biological targets and immune responses, environmental determinants, and vector control measures. Combining interventions known to be effective against multiple arboviral diseases will offer the most cost-effective and sustainable strategy for disease reduction. New global alliances are needed to enable the combination of efforts and resources for more effective and timely solutions., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
47. Responding to the threat of urban yellow fever outbreaks.
- Author
-
Wilder-Smith A and Monath TP
- Subjects
- Aedes, Animals, Humans, Yellow fever virus, Disease Outbreaks, Yellow Fever epidemiology
- Published
- 2017
- Full Text
- View/download PDF
48. A Recombinant Vesicular Stomatitis Virus Ebola Vaccine.
- Author
-
Regules JA, Beigel JH, Paolino KM, Voell J, Castellano AR, Hu Z, Muñoz P, Moon JE, Ruck RC, Bennett JW, Twomey PS, Gutiérrez RL, Remich SA, Hack HR, Wisniewski ML, Josleyn MD, Kwilas SA, Van Deusen N, Mbaya OT, Zhou Y, Stanley DA, Jing W, Smith KS, Shi M, Ledgerwood JE, Graham BS, Sullivan NJ, Jagodzinski LL, Peel SA, Alimonti JB, Hooper JW, Silvera PM, Martin BK, Monath TP, Ramsey WJ, Link CJ, Lane HC, Michael NL, Davey RT Jr, and Thomas SJ
- Subjects
- Adult, Antibodies, Viral blood, Double-Blind Method, Ebola Vaccines administration & dosage, Ebola Vaccines adverse effects, Ebolavirus genetics, Ebolavirus isolation & purification, Enzyme-Linked Immunosorbent Assay, Female, Hemorrhagic Fever, Ebola immunology, Humans, Male, Middle Aged, Recombinant Proteins, Seroconversion, Vaccines, Attenuated immunology, Vesicular stomatitis Indiana virus, Viral Envelope Proteins isolation & purification, Viremia, Ebola Vaccines immunology, Ebolavirus immunology, Hemorrhagic Fever, Ebola prevention & control
- Abstract
Background: The worst Ebola virus disease (EVD) outbreak in history has resulted in more than 28,000 cases and 11,000 deaths. We present the final results of two phase 1 trials of an attenuated, replication-competent, recombinant vesicular stomatitis virus (rVSV)-based vaccine candidate designed to prevent EVD., Methods: We conducted two phase 1, placebo-controlled, double-blind, dose-escalation trials of an rVSV-based vaccine candidate expressing the glycoprotein of a Zaire strain of Ebola virus (ZEBOV). A total of 39 adults at each site (78 participants in all) were consecutively enrolled into groups of 13. At each site, volunteers received one of three doses of the rVSV-ZEBOV vaccine (3 million plaque-forming units [PFU], 20 million PFU, or 100 million PFU) or placebo. Volunteers at one of the sites received a second dose at day 28. Safety and immunogenicity were assessed., Results: The most common adverse events were injection-site pain, fatigue, myalgia, and headache. Transient rVSV viremia was noted in all the vaccine recipients after dose 1. The rates of adverse events and viremia were lower after the second dose than after the first dose. By day 28, all the vaccine recipients had seroconversion as assessed by an enzyme-linked immunosorbent assay (ELISA) against the glycoprotein of the ZEBOV-Kikwit strain. At day 28, geometric mean titers of antibodies against ZEBOV glycoprotein were higher in the groups that received 20 million PFU or 100 million PFU than in the group that received 3 million PFU, as assessed by ELISA and by pseudovirion neutralization assay. A second dose at 28 days after dose 1 significantly increased antibody titers at day 56, but the effect was diminished at 6 months., Conclusions: This Ebola vaccine candidate elicited anti-Ebola antibody responses. After vaccination, rVSV viremia occurred frequently but was transient. These results support further evaluation of the vaccine dose of 20 million PFU for preexposure prophylaxis and suggest that a second dose may boost antibody responses. (Funded by the National Institutes of Health and others; rVSV∆G-ZEBOV-GP ClinicalTrials.gov numbers, NCT02269423 and NCT02280408 .).
- Published
- 2017
- Full Text
- View/download PDF
49. Additive Protection against Congenital Cytomegalovirus Conferred by Combined Glycoprotein B/pp65 Vaccination Using a Lymphocytic Choriomeningitis Virus Vector.
- Author
-
Schleiss MR, Berka U, Watson E, Aistleithner M, Kiefmann B, Mangeat B, Swanson EC, Gillis PA, Hernandez-Alvarado N, Fernández-Alarcón C, Zabeli JC, Pinschewer DD, Lilja AE, Schwendinger M, Guirakhoo F, Monath TP, and Orlinger KK
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, Antigens, Viral genetics, Antigens, Viral immunology, Cytomegalovirus Infections congenital, Cytomegalovirus Vaccines administration & dosage, Disease Models, Animal, Female, Guinea Pigs, Mice, Inbred C57BL, Phosphoproteins genetics, T-Lymphocytes immunology, Vaccines, Combined administration & dosage, Vaccines, Combined immunology, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic immunology, Viral Envelope Proteins genetics, Viral Matrix Proteins genetics, Cytomegalovirus Infections prevention & control, Cytomegalovirus Vaccines immunology, Drug Carriers, Lymphocytic choriomeningitis virus genetics, Phosphoproteins immunology, Viral Envelope Proteins immunology, Viral Matrix Proteins immunology
- Abstract
Subunit vaccines for prevention of congenital cytomegalovirus (CMV) infection based on glycoprotein B (gB) and pp65 are in clinical trials, but it is unclear whether simultaneous vaccination with both antigens enhances protection. We undertook evaluation of a novel bivalent vaccine based on nonreplicating lymphocytic choriomeningitis virus (rLCMV) vectors expressing a cytoplasmic tail-deleted gB [gB(dCt)] and full-length pp65 from human CMV in mice. Immunization with the gB(dCt) vector alone elicited a comparable gB-binding antibody response and a superior neutralizing response to that elicited by adjuvanted subunit gB. Immunization with the pp65 vector alone elicited robust T cell responses. Comparable immunogenicity of the combined gB(dCt) and pp65 vectors with the individual monovalent formulations was demonstrated. To demonstrate proof of principle for a bivalent rLCMV-based HCMV vaccine, the congenital guinea pig cytomegalovirus (GPCMV) infection model was used to compare rLCMV vectors encoding homologs of pp65 (GP83) and gB(dCt), alone and in combination versus Freund's adjuvanted recombinant gB. Both vectors elicited significant immune responses, and no loss of gB immunogenicity was noted with the bivalent formulation. Combined vaccination with rLCMV-vectored GPCMV gB(dCt) and pp65 (GP83) conferred better protection against maternal viremia than subunit or either monovalent rLCMV vaccine. The bivalent vaccine also was significantly more effective in reducing pup mortality than the monovalent vaccines. In summary, bivalent vaccines with rLCMV vectors expressing gB and pp65 elicited potent humoral and cellular responses and conferred protection in the GPCMV model. Further clinical trials of LCMV-vectored HCMV vaccines are warranted., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
50. Unique safety issues associated with virus-vectored vaccines: Potential for and theoretical consequences of recombination with wild type virus strains.
- Author
-
Condit RC, Williamson AL, Sheets R, Seligman SJ, Monath TP, Excler JL, Gurwith M, Bok K, Robertson JS, Kim D, Michael Hendry R, Singh V, Mac LM, and Chen RT
- Subjects
- Animals, Humans, Vaccines, Attenuated adverse effects, Vaccines, Attenuated genetics, Virulence, Viruses, Drug Carriers, Genetic Vectors, Recombination, Genetic, Viral Vaccines adverse effects, Viral Vaccines genetics
- Abstract
In 2003 and 2013, the World Health Organization convened informal consultations on characterization and quality aspects of vaccines based on live virus vectors. In the resulting reports, one of several issues raised for future study was the potential for recombination of virus-vectored vaccines with wild type pathogenic virus strains. This paper presents an assessment of this issue formulated by the Brighton Collaboration. To provide an appropriate context for understanding the potential for recombination of virus-vectored vaccines, we review briefly the current status of virus-vectored vaccines, mechanisms of recombination between viruses, experience with recombination involving live attenuated vaccines in the field, and concerns raised previously in the literature regarding recombination of virus-vectored vaccines with wild type virus strains. We then present a discussion of the major variables that could influence recombination between a virus-vectored vaccine and circulating wild type virus and the consequences of such recombination, including intrinsic recombination properties of the parent virus used as a vector; sequence relatedness of vector and wild virus; virus host range, pathogenesis and transmission; replication competency of vector in target host; mechanism of vector attenuation; additional factors potentially affecting virulence; and circulation of multiple recombinant vectors in the same target population. Finally, we present some guiding principles for vector design and testing intended to anticipate and mitigate the potential for and consequences of recombination of virus-vectored vaccines with wild type pathogenic virus strains., (Copyright © 2016. Published by Elsevier Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.