Cargando…

Vaccination Route as a Determinant of Protective Antibody Responses against Herpes Simplex Virus

Herpes simplex viruses (HSV) are significant global health problems associated with mucosal and neurologic disease. Prior experimental vaccines primarily elicited neutralizing antibodies targeting glycoprotein D (gD), but those that advanced to clinical efficacy trials have failed. Preclinical studi...

Descripción completa

Detalles Bibliográficos
Autores principales: Burn Aschner, Clare, Pierce, Carl, Knipe, David M., Herold, Betsy C.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7350019/
https://www.ncbi.nlm.nih.gov/pubmed/32516944
http://dx.doi.org/10.3390/vaccines8020277
_version_ 1783557189891260416
author Burn Aschner, Clare
Pierce, Carl
Knipe, David M.
Herold, Betsy C.
author_facet Burn Aschner, Clare
Pierce, Carl
Knipe, David M.
Herold, Betsy C.
author_sort Burn Aschner, Clare
collection PubMed
description Herpes simplex viruses (HSV) are significant global health problems associated with mucosal and neurologic disease. Prior experimental vaccines primarily elicited neutralizing antibodies targeting glycoprotein D (gD), but those that advanced to clinical efficacy trials have failed. Preclinical studies with an HSV-2 strain deleted in gD (ΔgD-2) administered subcutaneously demonstrated that it elicited a high titer, weakly neutralizing antibodies that activated Fcγ receptors to mediate antibody-dependent cellular cytotoxicity (ADCC), and completely protected mice against lethal disease and latency following vaginal or skin challenge with HSV-1 or HSV-2. Vaccine efficacy, however, may be impacted by dose and route of immunization. Thus, the current studies were designed to compare immunogenicity and efficacy following different routes of vaccination with escalating doses of ΔgD-2. We compared ΔgD-2 with two other candidates: recombinant gD protein combined with aluminum hydroxide and monophosphoryl lipid A adjuvants and a replication-defective virus deleted in two proteins involved in viral replication, dl5-29. Compared to the subcutaneous route, intramuscular and/or intradermal immunization resulted in increased total HSV antibody responses for all three vaccines and boosted the ADCC, but not the neutralizing response to ΔgD and dl5-29. The adjuvanted gD protein vaccine provided only partial protection and failed to elicit ADCC independent of route of administration. In contrast, the increased ADCC following intramuscular or intradermal administration of ΔgD-2 or dl5-29 translated into significantly increased protection. The ΔgD-2 vaccine provided 100% protection at doses as low as 5 × 10(4) pfu when administered intramuscularly or intradermally, but not subcutaneously. However, administration of a combination of low dose subcutaneous ΔgD-2 and adjuvanted gD protein resulted in greater protection than low dose ΔgD-2 alone indicating that gD neutralizing antibodies may contribute to protection. Taken together, these results demonstrate that ADCC provides a more predictive correlate of protection against HSV challenge in mice and support intramuscular or intradermal routes of vaccination.
format Online
Article
Text
id pubmed-7350019
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-73500192020-07-22 Vaccination Route as a Determinant of Protective Antibody Responses against Herpes Simplex Virus Burn Aschner, Clare Pierce, Carl Knipe, David M. Herold, Betsy C. Vaccines (Basel) Article Herpes simplex viruses (HSV) are significant global health problems associated with mucosal and neurologic disease. Prior experimental vaccines primarily elicited neutralizing antibodies targeting glycoprotein D (gD), but those that advanced to clinical efficacy trials have failed. Preclinical studies with an HSV-2 strain deleted in gD (ΔgD-2) administered subcutaneously demonstrated that it elicited a high titer, weakly neutralizing antibodies that activated Fcγ receptors to mediate antibody-dependent cellular cytotoxicity (ADCC), and completely protected mice against lethal disease and latency following vaginal or skin challenge with HSV-1 or HSV-2. Vaccine efficacy, however, may be impacted by dose and route of immunization. Thus, the current studies were designed to compare immunogenicity and efficacy following different routes of vaccination with escalating doses of ΔgD-2. We compared ΔgD-2 with two other candidates: recombinant gD protein combined with aluminum hydroxide and monophosphoryl lipid A adjuvants and a replication-defective virus deleted in two proteins involved in viral replication, dl5-29. Compared to the subcutaneous route, intramuscular and/or intradermal immunization resulted in increased total HSV antibody responses for all three vaccines and boosted the ADCC, but not the neutralizing response to ΔgD and dl5-29. The adjuvanted gD protein vaccine provided only partial protection and failed to elicit ADCC independent of route of administration. In contrast, the increased ADCC following intramuscular or intradermal administration of ΔgD-2 or dl5-29 translated into significantly increased protection. The ΔgD-2 vaccine provided 100% protection at doses as low as 5 × 10(4) pfu when administered intramuscularly or intradermally, but not subcutaneously. However, administration of a combination of low dose subcutaneous ΔgD-2 and adjuvanted gD protein resulted in greater protection than low dose ΔgD-2 alone indicating that gD neutralizing antibodies may contribute to protection. Taken together, these results demonstrate that ADCC provides a more predictive correlate of protection against HSV challenge in mice and support intramuscular or intradermal routes of vaccination. MDPI 2020-06-05 /pmc/articles/PMC7350019/ /pubmed/32516944 http://dx.doi.org/10.3390/vaccines8020277 Text en © 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Burn Aschner, Clare
Pierce, Carl
Knipe, David M.
Herold, Betsy C.
Vaccination Route as a Determinant of Protective Antibody Responses against Herpes Simplex Virus
title Vaccination Route as a Determinant of Protective Antibody Responses against Herpes Simplex Virus
title_full Vaccination Route as a Determinant of Protective Antibody Responses against Herpes Simplex Virus
title_fullStr Vaccination Route as a Determinant of Protective Antibody Responses against Herpes Simplex Virus
title_full_unstemmed Vaccination Route as a Determinant of Protective Antibody Responses against Herpes Simplex Virus
title_short Vaccination Route as a Determinant of Protective Antibody Responses against Herpes Simplex Virus
title_sort vaccination route as a determinant of protective antibody responses against herpes simplex virus
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7350019/
https://www.ncbi.nlm.nih.gov/pubmed/32516944
http://dx.doi.org/10.3390/vaccines8020277
work_keys_str_mv AT burnaschnerclare vaccinationrouteasadeterminantofprotectiveantibodyresponsesagainstherpessimplexvirus
AT piercecarl vaccinationrouteasadeterminantofprotectiveantibodyresponsesagainstherpessimplexvirus
AT knipedavidm vaccinationrouteasadeterminantofprotectiveantibodyresponsesagainstherpessimplexvirus
AT heroldbetsyc vaccinationrouteasadeterminantofprotectiveantibodyresponsesagainstherpessimplexvirus