Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 19236, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31848362

ABSTRACT

Vaccines based on cytomegalovirus (CMV) demonstrate protection in animal models of infectious disease and cancer. Vaccine efficacy is associated with the ability of CMV to elicit and indefinitely maintain high frequencies of circulating effector memory T cells (TEM) providing continuous, life-long anti-pathogen immune activity. To allow for the clinical testing of human CMV (HCMV)-based vaccines we constructed and characterized as a vector backbone the recombinant molecular clone TR3 representing a wildtype genome. We demonstrate that TR3 can be stably propagated in vitro and that, despite species incompatibility, recombinant TR3 vectors elicit high frequencies of TEM to inserted antigens in rhesus macaques (RM). Live-attenuated versions of TR3 were generated by deleting viral genes required to counteract intrinsic and innate immune responses. In addition, we eliminated subunits of a viral pentameric glycoprotein complex thus limiting cell tropism. We show in a humanized mouse model that such modified vectors were able to establish persistent infection but lost their ability to reactivate from latency. Nevertheless, attenuated TR3 vectors preserved the ability to elicit and maintain TEM to inserted antigens in RM. We further demonstrate that attenuated TR3 can be grown in approved cell lines upon elimination of an anti-viral host factor using small interfering RNA, thus obviating the need for a complementing cell line. In sum, we have established a versatile platform for the clinical development of live attenuated HCMV-vectored vaccines and immunotherapies.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus Vaccines , Cytomegalovirus , Animals , Cell Line, Tumor , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/genetics , Cytomegalovirus Vaccines/immunology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred NOD , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
2.
J Virol ; 92(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30045984

ABSTRACT

As human cytomegalovirus (HCMV) is a common cause of disease in newborns and transplant recipients, developing an HCMV vaccine is considered a major public health priority. Yet an HCMV vaccine candidate remains elusive. Although the precise HCMV immune correlates of protection are unclear, both humoral and cellular immune responses have been implicated in protection against HCMV infection and disease. Here we describe a vaccine approach based on the well-characterized modified vaccinia virus Ankara (MVA) vector to stimulate robust HCMV humoral and cellular immune responses by an antigen combination composed of the envelope pentamer complex (PC), glycoprotein B (gB), and phosphoprotein 65 (pp65). We show that in mice, multiantigenic MVA vaccine vectors simultaneously expressing all five PC subunits, gB, and pp65 elicit potent complement-independent and complement-dependent HCMV neutralizing antibodies as well as mouse and human MHC-restricted, polyfunctional T cell responses by the individual antigens. In addition, we demonstrate that the PC/gB antigen combination of these multiantigenic MVA vectors can enhance the stimulation of humoral immune responses that mediate in vitro neutralization of different HCMV strains and antibody-dependent cellular cytotoxicity. These results support the use of MVA to develop a multiantigenic vaccine candidate for controlling HCMV infection and disease in different target populations, such as pregnant women and transplant recipients.IMPORTANCE The development of a human cytomegalovirus (HCMV) vaccine to prevent congenital disease and transplantation-related complications is an unmet medical need. While many HCMV vaccine candidates have been developed, partial success in preventing or controlling HCMV infection in women of childbearing age and transplant recipients has been observed with an approach based on envelope glycoprotein B (gB). We introduce a novel vaccine strategy based on the clinically deployable modified vaccinia virus Ankara (MVA) vaccine vector to elicit potent humoral and cellular immune responses by multiple immunodominant HCMV antigens, including gB, phosphoprotein 65, and all five subunits of the pentamer complex. These findings could contribute to development of a multiantigenic vaccine strategy that may afford more protection against HCMV infection and disease than a vaccine approach employing solely gB.


Subject(s)
Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/immunology , Cytomegalovirus/immunology , Phosphoproteins/immunology , Vaccinia virus/immunology , Viral Envelope Proteins/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Antigens, Viral/genetics , Antigens, Viral/immunology , Base Sequence , Complement System Proteins/genetics , Complement System Proteins/metabolism , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/genetics , Female , Gene Expression Regulation , Humans , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Mice , Phosphoproteins/genetics , Pregnancy , Sequence Alignment , Signal Transduction , Vaccinia virus/genetics , Viral Envelope Proteins/genetics , Viral Matrix Proteins/genetics
3.
Front Immunol ; 9: 991, 2018.
Article in English | MEDLINE | ID: mdl-29867968

ABSTRACT

The development of a vaccine against human cytomegalovirus (CMV) has been a subject of long-term medical interest. The research during recent years identified CMV as an attractive vaccine vector against infectious diseases and tumors. The immune response to CMV persists over a lifetime and its unique feature is the inflationary T cell response to certain viral epitopes. CMV encodes numerous genes involved in immunoevasion, which are non-essential for virus growth in vitro. The deletion of those genes results in virus attenuation in vivo, which enables us to dramatically manipulate its virulence and the immune response. We have previously shown that the murine CMV (MCMV) expressing RAE-1γ, one of the cellular ligands for the NKG2D receptor, is highly attenuated in vivo but retains the ability to induce a strong CD8+ T cell response. Here, we demonstrate that recombinant MCMV expressing high affinity NKG2D ligand murine UL16 binding protein-like transcript (MULT-1) (MULT-1MCMV) inserted in the place of its viral inhibitor is dramatically attenuated in vivo in a NK cell-dependent manner, both in immunocompetent adult mice and in immunologically immature newborns. MULT-1MCMV was more attenuated than the recombinant virus expressing RAE-1γ. Despite the drastic sensitivity to innate immune control, MULT-1MCMV induced an efficient CD8+ T cell response to viral and vectored antigens. By using in vitro assay, we showed that similar to RAE-1γMCMV, MULT-1 expressing virus provided strong priming of CD8+ T cells. Moreover, MULT-1MCMV was able to induce anti-viral antibodies, which after passing the transplacental barrier protect offspring of immunized mothers from challenge infection. Altogether, this study further supports the concept that CMV expressing NKG2D ligand possesses excellent characteristics to serve as a vaccine or vaccine vector.


Subject(s)
Carrier Proteins/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/immunology , Histocompatibility Antigens Class I/genetics , Muromegalovirus/genetics , Animals , Animals, Newborn , CD8-Positive T-Lymphocytes/immunology , Carrier Proteins/immunology , Cytomegalovirus Vaccines/genetics , Disease Models, Animal , Female , Genetic Vectors/immunology , Histocompatibility Antigens Class I/immunology , Immunity, Maternally-Acquired , Immunocompetence , Killer Cells, Natural/immunology , Membrane Proteins , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology
4.
Adv Exp Med Biol ; 1045: 271-296, 2018.
Article in English | MEDLINE | ID: mdl-29896672

ABSTRACT

The development of a cytomegalovirus (CMV) vaccine has become a top priority due to its potential cost-effectiveness and associated public health benefits. However, there are a number of challenges facing vaccine development including the following: (1) CMV has many mechanisms for evading immune responses , and natural immunity is not perfect, (2) the immune correlates for protection are unclear, (3) a narrow range of CMV hosts limits the value of animal models, and (4) the placenta is a specialized organ formed transiently and its immunological status changes with time. In spite of these limitations, several types of CMV vaccine candidate, including live-attenuated, DISC , subunit, DNA, vectored, and peptide vaccines, have been developed or are currently under development. The recognition of the pentameric complex as the major neutralization target and identification of various strategies to block viral immune response evasion mechanisms have opened new avenues to CMV vaccine development. Here, we discuss the immune correlates for protection, the characteristics of the various vaccine candidates and their clinical trials, and the relevant animal models.


Subject(s)
Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/immunology , Cytomegalovirus/immunology , Animals , Cytomegalovirus/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/genetics , Humans , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
5.
J Virol ; 92(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29769344

ABSTRACT

Human cytomegalovirus (HCMV) represents a major cause of clinical complications during pregnancy as well as immunosuppression, and the licensing of a protective HCMV vaccine remains an unmet global need. Here, we designed and validated novel Sendai virus (SeV) vectors delivering the T cell immunogens IE-1 and pp65. To enhance vector safety, we used a replication-deficient strain (rdSeV) that infects target cells in a nonproductive manner while retaining viral gene expression. In this study, we explored the impact that transduction with rdSeV has on human dendritic cells (DCs) by comparing it to the parental, replication-competent Sendai virus strain (rcSeV) as well as the poxvirus strain modified vaccinia Ankara (MVA). We found that wild-type SeV is capable of replicating to high titers in DCs while rdSeV infects cells abortively. Due to the higher degree of attenuation, IE-1 and pp65 protein levels mediated by rdSeV after infection of DCs were markedly reduced compared to those of the parental Sendai virus recombinants, but antigen-specific restimulation of T cell clones was not negatively affected by this. Importantly, rdSeV showed reduced cytotoxic effects compared to rcSeV and MVA and was capable of mediating DC maturation as well as secretion of alpha interferon and interleukin-6. Finally, in a challenge model with a murine cytomegalovirus (MCMV) strain carrying an HCMV pp65 peptide, we found that viral replication was restricted if mice were previously vaccinated with rdSeV-pp65. Taken together, these data demonstrate that rdSeV has great potential as a vector system for the delivery of HCMV immunogens.IMPORTANCE HCMV is a highly prevalent betaherpesvirus that establishes lifelong latency after primary infection. Congenital HCMV infection is the most common viral complication in newborns, causing a number of late sequelae ranging from impaired hearing to mental retardation. At the same time, managing HCMV reactivation during immunosuppression remains a major hurdle in posttransplant care. Since options for the treatment of HCMV infection are still limited, the development of a vaccine to confine HCMV-related morbidities is urgently needed. We generated new vaccine candidates in which the main targets of T cell immunity during natural HCMV infection, IE-1 and pp65, are delivered by a replication-deficient, Sendai virus-based vector system. In addition to classical prophylactic vaccine concepts, these vectors could also be used for therapeutic applications, thereby expanding preexisting immunity in high-risk groups such as transplant recipients or for immunotherapy of glioblastomas expressing HCMV antigens.


Subject(s)
Antigens, Viral , Cytomegalovirus Vaccines , Cytomegalovirus , Genetic Vectors , Phosphoproteins , Sendai virus , Transduction, Genetic , Viral Matrix Proteins , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Chlorocebus aethiops , Cricetinae , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus Vaccines/genetics , Cytomegalovirus Vaccines/immunology , Humans , Mice , Mice, Transgenic , Phosphoproteins/genetics , Phosphoproteins/immunology , Vero Cells , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology
6.
Vaccine ; 36(12): 1689-1699, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29456015

ABSTRACT

A cytomegalovirus (CMV) vaccine that is effective at preventing congenital infection and reducing CMV disease in transplant patients remains a high priority as no approved vaccines exist. While the precise correlates of protection are unknown, neutralizing antibodies and antigen-specific T cells have been implicated in controlling infection. We demonstrate that the immunization of mice and nonhuman primates (NHPs) with lipid nanoparticles (LNP) encapsulating modified mRNA encoding CMV glycoproteins gB and pentameric complex (PC) elicit potent and durable neutralizing antibody titers. Since the protective correlates in pregnant women and transplant recipients may differ, we developed an additional mRNA vaccine expressing the immunodominant CMV T cell antigen pp65. Administration of pp65 vaccine with PC and gB elicited robust multi-antigenic T cell responses in mice. Our data demonstrate that mRNA/LNP is a versatile platform that enables the development of vaccination strategies that could prevent CMV infection and consequent disease in different target populations.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/immunology , Cytomegalovirus/immunology , Immunity, Cellular , Immunity, Humoral , Vaccines, Synthetic/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line , Cytokines/metabolism , Cytomegalovirus/genetics , Cytomegalovirus Vaccines/genetics , Female , Gene Expression , Humans , Mice , Neutralization Tests , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vaccination , Vaccines, Synthetic/genetics
7.
J Virol Methods ; 251: 30-37, 2018 01.
Article in English | MEDLINE | ID: mdl-28989096

ABSTRACT

Neutralizing antibodies (NAb) interfering with glycoprotein complex-mediated virus entry into host cells are thought to contribute to the protection against herpesvirus infection. However, using herpesvirus glycoprotein complexes as vaccine antigens can be complicated by the necessity of expressing multiple subunits simultaneously to allow efficient complex assembly and formation of conformational NAb epitopes. By using a novel bacterial artificial chromosome (BAC) clone of the clinically deployable Modified Vaccinia Ankara (MVA) vector and exploiting ribosomal skipping mediated by 2A peptides, MVA vectors were generated that expressed self-processing subunits of the human cytomegalovirus (HCMV) pentamer complex (PC) composed of gH, gL, UL128, UL130, and UL131A. These MVA vectors expressed 2A-linked HCMV PC subunits that were efficiently cleaved and transported to the cell surface as protein complexes forming conformational neutralizing epitopes. In addition, vaccination of mice by only two immunizations with these MVA vectors resulted in potent HCMV NAb responses that remained stable over a period of at least six months. This method of eliciting NAb by 2A-linked, self-processing HCMV PC subunits could contribute to develop a HCMV vaccine candidate and may serve as a template to facilitate the development of subunit vaccine strategies against other herpesviruses.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigens, Viral/immunology , Cytomegalovirus Vaccines/immunology , Cytomegalovirus/immunology , Glycoproteins/immunology , Viral Structural Proteins/immunology , Animals , Antigens, Viral/genetics , Cytomegalovirus/genetics , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/genetics , Genetic Vectors , Glycoproteins/genetics , Immunization Schedule , Mice , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Viral Structural Proteins/genetics
8.
J Exp Med ; 214(7): 1889-1899, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28566275

ABSTRACT

Cytomegalovirus (CMV)-based vaccines have shown remarkable efficacy in the rhesus macaque model of acquired immune deficiency syndrome, enabling 50% of vaccinated monkeys to clear a subsequent virulent simian immunodeficiency virus challenge. The protective vaccine elicited unconventional CD8 T cell responses that were entirely restricted by MHC II or the nonclassical MHC I molecule, MHC-E. These unconventional responses were only elicited by a fibroblast-adapted rhesus CMV vector with limited tissue tropism; a repaired vector with normal tropism elicited conventional responses. Testing whether these unusual protective CD8 T responses could be elicited in humans requires vaccinating human subjects with a fibroblast-adapted mutant of human CMV (HCMV). In this study, we describe the CD8 T cell responses of human subjects vaccinated with two fibroblast-adapted HCMV vaccines. Most responses were identified as conventional classically MHC I restricted, and we found no evidence for MHC II or HLA-E restriction. These results indicate that fibroblast adaptation alone is unlikely to explain the unconventional responses observed in macaques.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/immunology , Cytomegalovirus/immunology , Fibroblasts/immunology , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Cells, Cultured , Cytomegalovirus/physiology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/genetics , Epitopes/immunology , Fibroblasts/virology , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , K562 Cells , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Male , Microscopy, Fluorescence , Mutation , Vaccination
9.
Hum Vaccin Immunother ; 13(12): 2763-2771, 2017 12 02.
Article in English | MEDLINE | ID: mdl-28494195

ABSTRACT

T cell immunity is critical in controlling human cytomegalovirus (HCMV) infection in transplant recipients, and T cells targeting viral immediate early proteins such as IE1, IE2 and pp65 have been speculated to be more effective against reactivation. Here we report efforts to construct replication incompetent adenovirus 6 vectors expressing these viral antigens as vaccine candidates. To reduce the potential liabilities of these viral proteins as vaccine antigens, we introduced mutations to inactivate their reported functions including their nuclear localization signals. The modifications greatly reduced their localization to the nuclei, thus limiting their interactions with cellular proteins important for cell cycle modulation and transactivation. The immunogenicity of modified pp65, IE1 and IE2 vaccines was comparable to their wild-type counterparts in mice and the immunogenicity of the modified antigens was demonstrated in non-human primates.


Subject(s)
Antigens, Viral/immunology , Cytomegalovirus Vaccines/immunology , Cytomegalovirus/immunology , Defective Viruses/genetics , Drug Carriers , Genetic Vectors , Mastadenovirus/genetics , Animals , Antigens, Viral/genetics , Cytomegalovirus/genetics , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/adverse effects , Cytomegalovirus Vaccines/genetics , Female , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutant Proteins/genetics , Mutant Proteins/immunology , Phosphoproteins/genetics , Phosphoproteins/immunology , Primates , Trans-Activators/genetics , Trans-Activators/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology
10.
Virus Genes ; 53(4): 650-655, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28391502

ABSTRACT

Human cytomegalovirus (HCMV) is an important opportunistic pathogen in immunocompromised patients and a major cause of congenital birth defects when acquired in utero. In the 1990s, four chimeric viruses were constructed by replacing genome segments of the high passage Towne strain with segments of the low passage Toledo strain, with the goal of obtaining live attenuated vaccine candidates that remained safe but were more immunogenic than the overly attenuated Towne vaccine. The chimeras were found to be safe when administered to HCMV-seronegative human volunteers, but to differ significantly in their ability to induce seroconversion. This suggests that chimera-specific genetic differences impacted the ability to replicate or persist in vivo and the consequent ability to induce an antibody response. To identify specific genomic breakpoints between Towne and Toledo sequences and establish whether spontaneous mutations or rearrangements had occurred during construction of the chimeras, complete genome sequences were determined. No major deletions or rearrangements were observed, although a number of unanticipated mutations were identified. However, no clear association emerged between the genetic content of the chimeras and the reported levels of vaccine-induced HCMV-specific humoral or cellular immune responses, suggesting that multiple genetic determinants are likely to impact immunogenicity. In addition to revealing the genome organization of the four vaccine candidates, this study provided an opportunity to probe the genetics of HCMV attenuation in humans. The results may be valuable in the future design of safe live or replication-defective vaccines that optimize immunogenicity and efficacy.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/immunology , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Recombination, Genetic , Antibodies, Viral/immunology , Cytomegalovirus/classification , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/genetics , Genome, Viral , Genomics , Humans , Immunization
11.
J Virol ; 90(17): 7902-19, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27334585

ABSTRACT

UNLABELLED: Congenital cytomegalovirus (CMV) infection is a leading cause of mental retardation and deafness in newborns. The guinea pig is the only small animal model for congenital CMV infection. A novel CMV vaccine was investigated as an intervention strategy against congenital guinea pig cytomegalovirus (GPCMV) infection. In this disabled infectious single-cycle (DISC) vaccine strategy, a GPCMV mutant virus was used that lacked the ability to express an essential capsid gene (the UL85 homolog GP85) except when grown on a complementing cell line. In vaccinated animals, the GP85 mutant virus (GP85 DISC) induced an antibody response to important glycoprotein complexes considered neutralizing target antigens (gB, gH/gL/gO, and gM/gN). The vaccine also generated a T cell response to the pp65 homolog (GP83), determined via a newly established guinea pig gamma interferon enzyme-linked immunosorbent spot assay. In a congenital infection protection study, GP85 DISC-vaccinated animals and a nonvaccinated control group were challenged during pregnancy with wild-type GPCMV (10(5) PFU). The pregnant animals carried the pups to term, and viral loads in target organs of pups were analyzed. Based on live pup births in the vaccinated and control groups (94.1% versus 63.6%), the vaccine was successful in reducing mortality (P = 0.0002). Additionally, pups from the vaccinated group had reduced CMV transmission, with 23.5% infected target organs versus 75.9% in the control group. Overall, these preliminary studies indicate that a DISC CMV vaccine strategy has the ability to induce an immune response similar to that of natural virus infection but has the increased safety of a non-replication-competent virus, which makes this approach attractive as a CMV vaccine strategy. IMPORTANCE: Congenital CMV infection is a leading cause of mental retardation and deafness in newborns. An effective vaccine against CMV remains an elusive goal despite over 50 years of CMV research. The guinea pig, with a placenta structure similar to that in humans, is the only small animal model for congenital CMV infection and recapitulates disease symptoms (e.g., deafness) in newborn pups. In this report, a novel vaccine strategy against congenital guinea pig cytomegalovirus (GPCMV) infection was developed, characterized, and tested for efficacy. This disabled infectious single-cycle (DISC) vaccine strategy induced a neutralizing antibody or a T cell response to important target antigens. In a congenital infection protection study, animals were protected against CMV in comparison to the nonvaccinated group (52% reduction of transmission). This novel vaccine was more effective than previously tested gB-based vaccines and most other strategies involving live virus vaccines. Overall, the DISC vaccine is a safe and promising approach against congenital CMV infection.


Subject(s)
Capsid Proteins/genetics , Cytomegalovirus Vaccines/immunology , Mutant Proteins/genetics , Roseolovirus Infections/congenital , Roseolovirus Infections/prevention & control , Roseolovirus/physiology , Virus Replication , Animal Structures/virology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/genetics , Enzyme-Linked Immunospot Assay , Interferon-gamma/metabolism , Roseolovirus/genetics , Survival Analysis , T-Lymphocytes/immunology , Treatment Outcome , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Load
12.
Vaccine ; 34(3): 313-9, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26657184

ABSTRACT

BACKGROUND: Cytomegalovirus (CMV) is a leading cause of congenital infection and an important target for vaccine development. METHODS: CMV seronegative girls between 12 and 17 years of age received CMV glycoprotein B (gB) vaccine with MF59 or saline placebo at 0, 1 and 6 months. Blood and urine were collected throughout the study for evidence of CMV infection based on PCR and/or seroconversion to non-vaccine CMV antigens. RESULTS: 402 CMV seronegative subjects were vaccinated (195 vaccine, 207 placebo). The vaccine was generally well tolerated, although local and systemic adverse events were significantly more common in the vaccine group. The vaccine induced gB antibody in all vaccine recipients with a gB geometric mean titer of 13,400 EU; 95%CI 11,436, 15,700, after 3 doses. Overall, 48 CMV infections were detected (21 vaccine, 27 placebo). In the per protocol population (124 vaccine, 125 placebo) vaccine efficacy was 43%; 95%CI: -36; 76, p=0.20. The most significant difference was after 2 doses, administered as per protocol; vaccine efficacy 45%, 95%CI: -9; 72, p=0.08. CONCLUSION: The vaccine was safe and immunogenic. Although the efficacy did not reach conventional levels of significance, the results are consistent with a previous study in adult women (Pass et al. N Engl J Med 2009;360:1191) using the same formulation.


Subject(s)
Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/adverse effects , Cytomegalovirus Vaccines/immunology , Viral Envelope Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adolescent , Antibodies, Viral/blood , Antigens, Viral/analysis , Blood/virology , Child , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/genetics , DNA, Viral/analysis , DNA, Viral/genetics , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Immunization Schedule , Placebos/administration & dosage , Polymerase Chain Reaction , Polysorbates/administration & dosage , Polysorbates/adverse effects , Squalene/administration & dosage , Squalene/adverse effects , Urine/virology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/adverse effects , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics
13.
J Immunol ; 195(8): 3803-15, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26363059

ABSTRACT

CMV infection is a significant cause of morbidity and mortality in immunocompromised individuals, and the development of a vaccine is of high priority. Glycoprotein B (gB) is a leading vaccine candidate but the glycoprotein H (gH) pentameric complex is now recognized as the major target for neutralizing Abs. However, little is known about the T cell immune response against gH and glycoprotein L (gL) and this is likely to be an important attribute for vaccine immunogenicity. In this study, we examine and contrast the magnitude and phenotype of the T cell immune response against gB, gH, and gL within healthy donors. gB-specific CD4(+) T cells were found in 95% of donors, and 29 epitopes were defined with gB-specific response sizes ranging from 0.02 to 2.88% of the CD4(+) T cell pool. In contrast, only 20% of donors exhibited a T cell response against gH or gL. Additionally, gB-specific CD4(+) T cells exhibited a more cytotoxic phenotype, with high levels of granzyme B expression. Glycoproteins were effectively presented following delivery to APCs but only gB-derived epitopes were presented following endogenous synthesis. gB expression was observed exclusively within vesicular structures colocalizing with HLA-DM whereas gH was distributed evenly throughout the cytoplasm. Grafting of the C-terminal domain from gB onto gH could not transfer this pattern of presentation. These results reveal that gB is a uniquely immunogenic CMV glycoprotein and this is likely to reflect its unique pattern of endogenous Ag presentation. Consideration may be required toward mechanisms that boost cellular immunity to gH and gL within future subunit vaccines.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Epitopes, T-Lymphocyte/immunology , Immunity, Cellular , Viral Envelope Proteins/immunology , Adult , CD4-Positive T-Lymphocytes/pathology , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/pathology , Cytomegalovirus Vaccines/genetics , Cytomegalovirus Vaccines/immunology , Epitopes, T-Lymphocyte/genetics , Female , Granzymes , Humans , Male , Middle Aged , Viral Envelope Proteins/genetics
14.
Biotechnol Bioeng ; 112(12): 2505-15, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26058896

ABSTRACT

Human cytomegalovirus (HCMV) causes significant disease worldwide. Multiple HCMV vaccines have been tested in man but only partial protection has been achieved. The HCMV gH/gL/UL128/UL130/UL131A complex (Pentamer) is the main target of neutralizing antibodies in HCMV seropositive individuals and raises high titers of neutralizing antibodies in small animals and non-human primates (NHP). Thus, Pentamer is a promising candidate for an effective HCMV vaccine. Development of a Pentamer-based subunit vaccine requires expression of high amounts of a functional and stable complex. We describe here the development of a mammalian expression system for large scale Pentamer production. Several approaches comprising three different CHO-originated cell lines and multiple vector as well as selection strategies were tested. Stable cell pools expressed the HCMV Pentamer at a titer of approximately 60 mg/L at laboratory scale. A FACS-based single cell sorting approach allowed selection of a highly expressing clone producing Pentamer at the level of approximately 400 mg/L in a laboratory scale fed-batch culture. Expression in a 50 L bioreactor led to the production of HCMV Pentamer at comparable titers indicating the feasibility of further scale-up for manufacturing at commercial scale. The CHO-produced HCMV Pentamer bound to a panel of human neutralizing antibodies and raised potently neutralizing immune response in mice. Thus, we have generated an expression system for the large scale production of functional HCMV Pentamer at high titers suitable for future subunit vaccine production.


Subject(s)
CHO Cells , Cytomegalovirus Vaccines/immunology , Gene Expression , Viral Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cricetulus , Cytomegalovirus/genetics , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/genetics , Cytomegalovirus Vaccines/metabolism , Mice , Protein Multimerization , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/metabolism , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
15.
Adv Exp Med Biol ; 848: 131-48, 2015.
Article in English | MEDLINE | ID: mdl-25757619

ABSTRACT

There is no licensed vaccine or cure for human cytomegalovirus (CMV), a ubiquitous ß-herpes virus that infects 60-95 % of adults worldwide. Infection is a major cause of congenital abnormalities in newborns, contributes to development of childhood cerebral palsy and medulloblastoma, can result in severe disease in immunocompromised patients, and is a major impediment during successful organ transplantation. While CMV has been increasingly associated with numerous inflammatory diseases and cancers, only recently has it been correlated with increased risk of heart disease in adults, the number-one killer in the USA. These data, among others, suggest that subclinical CMV infection, or microinfection, in healthy individuals may play more of a causative role than an epiphenomenon in development of CMV-associated pathologies. Due to the myriad of diseases and complications associated with CMV, an efficacious vaccine would be highly valuable in reducing human morbidity and mortality as well as saving billions of dollars in annual health-care costs and disability adjusted life years (DALY) in the developing world. Therefore, the development of a safe efficacious CMV vaccine or immune therapy is paramount to the public health. This review aims to provide a brief overview on aspects of CMV infection and disease and focuses on current vaccine strategies. The use of new synthetic DNA vaccines might offer one such approach to this difficult problem.


Subject(s)
Cloning, Molecular/methods , Cytomegalovirus Infections/therapy , Cytomegalovirus Vaccines/therapeutic use , Immunotherapy, Active/methods , Vaccines, DNA/therapeutic use , Adult , Animals , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/genetics , DNA, Recombinant/genetics , DNA, Recombinant/therapeutic use , Humans , Vaccines, DNA/genetics
16.
Transplantation ; 99(1): 120-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25050468

ABSTRACT

BACKGROUND: Infection with human cytomegalovirus (CMV) is a significant cause of morbidity and mortality in solid organ and hematopoietic stem cell transplant (HSCT) recipients. METHODS: The present study explored the safety, feasibility, and immunogenicity of CMV pp65 messenger RNA-loaded autologous monocyte-derived dendritic cells (DC) as a cellular vaccine for active immunization in healthy volunteers and allogeneic HSCT recipients. Four CMV-seronegative healthy volunteers and three allogeneic HSCT recipients were included in the study. Four clinical-grade autologous monocyte-derived DC vaccines were prepared after a single leukapheresis procedure and administered intradermally at a weekly interval. RESULTS: De novo induction of CMV-specific T-cell responses was detected in three of four healthy volunteers without serious adverse events. Of the HSCT recipients, none developed CMV disease and one of two patients displayed a remarkable threefold increase in CMV pp65-specific T cells on completion of the DC vaccination trial. CONCLUSION: In conclusion, our DC vaccination strategy induced or expanded a CMV-specific cellular response in four of six efficacy-evaluable study subjects, providing a base for its further exploration in larger cohorts.


Subject(s)
Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus/immunology , Dendritic Cells/transplantation , Hematopoietic Stem Cell Transplantation/adverse effects , Phosphoproteins/immunology , RNA, Messenger/biosynthesis , RNA, Viral/biosynthesis , T-Lymphocytes/immunology , Transfection , Viral Matrix Proteins/immunology , Adult , Belgium , Cytomegalovirus/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/adverse effects , Cytomegalovirus Vaccines/genetics , Cytomegalovirus Vaccines/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Feasibility Studies , Female , Healthy Volunteers , Humans , Immunization Schedule , Injections, Intradermal , Male , Middle Aged , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , RNA, Messenger/genetics , RNA, Viral/metabolism , T-Lymphocytes/virology , Time Factors , Transplantation, Homologous , Treatment Outcome , Vaccination , Viral Matrix Proteins/biosynthesis , Viral Matrix Proteins/genetics , Young Adult
17.
PLoS Pathog ; 10(11): e1004524, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25412505

ABSTRACT

Human Cytomegalovirus (HCMV) utilizes two different pathways for host cell entry. HCMV entry into fibroblasts requires glycoproteins gB and gH/gL, whereas HCMV entry into epithelial and endothelial cells (EC) requires an additional complex composed of gH, gL, UL128, UL130, and UL131A, referred to as the gH/gL-pentamer complex (gH/gL-PC). While there are no established correlates of protection against HCMV, antibodies are thought to be important in controlling infection. Neutralizing antibodies (NAb) that prevent gH/gL-PC mediated entry into EC are candidates to be assessed for in vivo protective function. However, these potent NAb are predominantly directed against conformational epitopes derived from the assembled gH/gL-PC. To address these concerns, we constructed Modified Vaccinia Ankara (MVA) viruses co-expressing all five gH/gL-PC subunits (MVA-gH/gL-PC), subsets of gH/gL-PC subunits (gH/gL or UL128/UL130/UL131A), or the gB subunit from HCMV strain TB40/E. We provide evidence for cell surface expression and assembly of complexes expressing full-length gH or gB, or their secretion when the corresponding transmembrane domains are deleted. Mice or rhesus macaques (RM) were vaccinated three times with MVA recombinants and serum NAb titers that prevented 50% infection of human EC or fibroblasts by HCMV TB40/E were determined. NAb responses induced by MVA-gH/gL-PC blocked HCMV infection of EC with potencies that were two orders of magnitude greater than those induced by MVA expressing gH/gL, UL128-UL131A, or gB. In addition, MVA-gH/gL-PC induced NAb responses that were durable and efficacious to prevent HCMV infection of Hofbauer macrophages, a fetal-derived cell localized within the placenta. NAb were also detectable in saliva of vaccinated RM and reached serum peak levels comparable to NAb titers found in HCMV hyperimmune globulins. This vaccine based on a translational poxvirus platform co-delivers all five HCMV gH/gL-PC subunits to achieve robust humoral responses that neutralize HCMV infection of EC, placental macrophages and fibroblasts, properties of potential value in a prophylactic vaccine.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cytomegalovirus Infections , Cytomegalovirus Vaccines , Cytomegalovirus , Multiprotein Complexes , Viral Envelope Proteins , Animals , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/genetics , Cytomegalovirus Vaccines/immunology , Female , Humans , Macaca mulatta , Mice , Mice, Inbred BALB C , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
18.
Virol J ; 11: 104, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24898886

ABSTRACT

BACKGROUND: Human cytomegalovirus (HCMV) causes serious HCMV-related diseases in immunocompromised people. Vaccination is the most effective measure to control infection with the pathogen, yet no vaccine has been licensed till now. We performed a head-to-head comparison of the protective abilities of multiple DNA vaccines in murine model of murine cytomegalovirus (MCMV) infection. METHODS: Five DNA vaccines were constructed. Four encoding MCMV proteins gp34 (m04), p65 (M84), DNA helicase (M105), and immediate-early 1 protein pp89 (IE-1) , respectively, which were reported to induce CD8+ T cell responses, were compared with the one expressing gB (M55), the neutralizing antibody target antigen, for immune protection in BALB/c mice. Mice were immunized with these DNA vaccines 1 to 4 times via intramuscular injection followed by electroporation, and were subsequently infected with a lethal dose (3 × LD50) of highly virulent SG-MCMV. Specific antibodies and IFN-γ secreting splenocytes were detected by immunoblotting and ELISPOT, respectively. Protective abilities in mice provided by the vaccines were evaluated by residual virus titers in organs, survival rate and weight loss. RESULTS: These DNA vaccines, especially m04, M84 and IE-1, could effectively reduce the virus loads in salivary glands and spleens of mice, but they couldn't completely clear the residual virus. Survival rates of 100% in mice after a lethal dose of MCMV infection could be reached by more than one dose of M84 vaccine or two doses of m04 or IE-1 vaccine. Immunization with M55 or M105 DNA at four doses offered mice only 62.5% survival rate after the lethal challenge. CONCLUSIONS: The study demonstrated that DNA vaccines could effectively afford mice protection against infection with a highly virulent MCMV and that the protection offered by induced CD8+ T cell immunity might be superior to that by gB-specific antibodies. These results are valuable references for development and application of HCMV vaccines.


Subject(s)
Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/immunology , Vaccines, DNA/immunology , Animal Structures/virology , Animals , Antibodies, Viral/blood , Antigens, Viral/genetics , Antigens, Viral/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/genetics , Disease Models, Animal , Electroporation , Female , Injections, Intramuscular , Leukocytes, Mononuclear/immunology , Mice, Inbred BALB C , Muromegalovirus/genetics , Muromegalovirus/immunology , Survival Analysis , Vaccination/methods , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Load
19.
J Clin Invest ; 124(5): 1928-44, 2014 May.
Article in English | MEDLINE | ID: mdl-24691437

ABSTRACT

The most abundantly produced virion protein in human cytomegalovirus (HCMV) is the immunodominant phosphoprotein 65 (pp65), which is frequently included in CMV vaccines. Although it is nonessential for in vitro CMV growth, pp65 displays immunomodulatory functions that support a potential role in primary and/or persistent infection. To determine the contribution of pp65 to CMV infection and immunity, we generated a rhesus CMV lacking both pp65 orthologs (RhCMVΔpp65ab). While deletion of pp65ab slightly reduced growth in vitro and increased defective particle formation, the protein composition of secreted virions was largely unchanged. Interestingly, pp65 was not required for primary and persistent infection in animals. Immune responses induced by RhCMVΔpp65ab did not prevent reinfection with rhesus CMV; however, reinfection with RhCMVΔUS2-11, which lacks viral-encoded MHC-I antigen presentation inhibitors, was prevented. Unexpectedly, induction of pp65b-specific T cells alone did not protect against RhCMVΔUS2-11 challenge, suggesting that T cells targeting multiple CMV antigens are required for protection. However, pp65-specific immunity was crucial for controlling viral dissemination during primary infection, as indicated by the marked increase of RhCMVΔpp65ab genome copies in CMV-naive, but not CMV-immune, animals. Our data provide rationale for inclusion of pp65 into CMV vaccines but also demonstrate that pp65-induced T cell responses alone do not recapitulate the protective effect of natural infection.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Phosphoproteins/immunology , Viral Matrix Proteins/immunology , Animals , Antigen Presentation/immunology , Cell Line , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/pathology , Cytomegalovirus Vaccines/genetics , Cytomegalovirus Vaccines/immunology , Gene Deletion , Humans , Macaca mulatta , Mice , Phosphoproteins/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Viral Matrix Proteins/genetics
20.
Methods Mol Biol ; 1119: 311-48, 2014.
Article in English | MEDLINE | ID: mdl-24639230

ABSTRACT

The development of prophylactic and to lesser extent therapeutic vaccines for the prevention of disease associated with human cytomegalovirus (HCMV) infections has received considerable attention from biomedical researchers and pharmaceutical companies over the previous 15 years, even though attempts to produce such vaccines have been described in the literature for over 40 years. Studies of the natural history of congenital HCMV infection and infection in allograft recipients have suggested that prophylaxis of disease associated with HCMV infection could be possible, particularly in hosts at risk for more severe disease secondary to the lack of preexisting immunity. Provided a substantial understanding of immune response to HCMV together with several animal models that faithfully recapitulate aspects of human infection and immunity, investigators seem well positioned to design and test candidate vaccines. Yet more recent studies of the role of a maternal immunity in the natural history of congenital HCMV infection, including the recognition that reinfection of previously immune women by genetically distinct strains of HCMV occur in populations with a high seroprevalence, have raised several questions about the nature of protective immunity in maternal populations. This finding coupled with observations that have documented a significant incidence of damaging congenital infections in offspring of women with immunity to HCMV prior to conception has suggested that vaccine development based on conventional paradigms of adaptive immunity to viral infections may be of limited value in the prevention of damaging congenital HCMV infections. Perhaps a more achievable goal will be prophylactic vaccines to modify HCMV associated disease in allograft transplant recipients. Although recent descriptions of the results from vaccine trials have been heralded as evidence of an emerging success in the quest for a HCMV vaccine, careful analyses of these studies have also revealed that major hurdles remain to be addressed by current strategies.


Subject(s)
Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/immunology , Molecular Biology/methods , Vaccines, DNA/immunology , Cytomegalovirus/immunology , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/genetics , Female , Humans , Vaccines, DNA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...