Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Epidemics ; 44: 100698, 2023 09.
Article in English | MEDLINE | ID: mdl-37354657

ABSTRACT

BACKGROUND: There is an urgent need to develop a cytomegalovirus (CMV) vaccine as it remains the leading cause of birth defects in the United States. While several CMV vaccine candidates are currently in late-stage clinical trials, the most effective vaccination program remains an open research question. METHODS: To take into account the critical uncertainties when evaluating the vaccine impact on both vertical (congenital) and horizontal CMV transmissions, we developed a CMV agent-based model representative of the US population and contact network structures. RESULTS: We evaluated 648 vaccination scenarios under various assumptions of vaccination age, vaccine efficacy, protection duration, and vaccination coverage. The optimal age of vaccination under all scenarios is shown to be during early childhood. However, a relatively modest benefit was also seen with vaccination of females of reproduction age (around age of 25) assuming near universal coverage and long vaccine-mediated protection. CONCLUSIONS: This study highlights the important need for a pediatric vaccination program in mitigating CMV in the United States. Our model is poised to investigate further location-based vaccine effectiveness questions in future planning of both clinical trials as well as eventual program implementation.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus Vaccines , Female , Child , Humans , Child, Preschool , United States/epidemiology , Cytomegalovirus Infections/epidemiology , Cytomegalovirus Infections/prevention & control , Vaccination , Computer Simulation , Cytomegalovirus Vaccines/therapeutic use , Forecasting
2.
PLoS Pathog ; 19(1): e1011107, 2023 01.
Article in English | MEDLINE | ID: mdl-36662906

ABSTRACT

Cytomegalovirus (CMV) is a leading cause of infant hearing loss and neurodevelopmental delay, but there are no clinically licensed vaccines to prevent infection, in part due to challenges eliciting neutralizing antibodies. One of the most well-studied targets for CMV vaccines is the viral fusogen glycoprotein B (gB), which is required for viral entry into host cells. Within gB, antigenic domain 2 site 1 (AD-2S1) is a target of potently neutralizing antibodies, but gB-based candidate vaccines have yet to elicit robust responses against this region. We mapped the genealogy of B cells encoding potently neutralizing anti-gB AD-2S1 antibodies from their inferred unmutated common ancestor (UCA) and characterized the binding and function of early lineage ancestors. Surprisingly, we found that a single amino acid heavy chain mutation A33N, which was an improbable mutation rarely generated by somatic hypermutation machinery, conferred broad CMV neutralization to the non-neutralizing UCA antibody. Structural studies revealed that this mutation mediated key contacts with the gB AD-2S1 epitope. Collectively, these results provide insight into potently neutralizing gB-directed antibody evolution in a single donor and lay a foundation for using this B cell-lineage directed approach for the design of next-generation CMV vaccines.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Cytomegalovirus Infections , Cytomegalovirus Vaccines , Cytomegalovirus , Humans , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/therapeutic use , Mutation , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
3.
Annu Rev Virol ; 9(1): 491-520, 2022 09 29.
Article in English | MEDLINE | ID: mdl-35704747

ABSTRACT

Human cytomegalovirus (HCMV) infection, the most common cause of congenital disease globally, affecting an estimated 1 million newborns annually, can result in lifelong sequelae in infants, such as sensorineural hearing loss and brain damage. HCMV infection also leads to a significant disease burden in immunocompromised individuals. Hence, an effective HCMV vaccine is urgently needed to prevent infection and HCMV-associated diseases. Unfortunately, despite more than five decades of vaccine development, no successful HCMV vaccine is available. This review summarizes what we have learned from acquired natural immunity, including innate and adaptive immunity; the successes and failures of HCMV vaccine human clinical trials; the progress in related animal models; and the analysis of protective immune responses during natural infection and vaccination settings. Finally, we propose novel vaccine strategies that will harness the knowledge of protective immunity and employ new technology and vaccine concepts to inform next-generation HCMV vaccine development.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus Vaccines , Adaptive Immunity , Animals , Cytomegalovirus , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/therapeutic use , Humans , Immunity, Innate , Infant, Newborn , Vaccine Development
4.
J Clin Invest ; 132(16)2022 08 15.
Article in English | MEDLINE | ID: mdl-35763348

ABSTRACT

Human cytomegalovirus (HCMV) is the most common congenital infection and a leading cause of stillbirth, neurodevelopmental impairment, and pediatric hearing loss worldwide. Development of a maternal vaccine or therapeutic to prevent congenital HCMV has been hindered by limited knowledge of the immune responses that protect against HCMV transmission in utero. To identify protective antibody responses, we measured HCMV-specific IgG binding and antiviral functions in paired maternal and cord blood sera from HCMV-seropositive transmitting (n = 41) and non-transmitting (n = 40) mother-infant dyads identified via a large, US-based, public cord blood bank. We found that high-avidity IgG binding to HCMV and antibody-dependent cellular phagocytosis (ADCP) were associated with reduced risk of congenital HCMV infection. We also determined that HCMV-specific IgG activation of FcγRI and FcγRII was enhanced in non-transmitting dyads and that increased ADCP responses were mediated through both FcγRI and FcγRIIA expressed on human monocytes. These findings suggest that engagement of FcγRI/FcγRIIA and Fc effector functions including ADCP may protect against congenital HCMV infection. Taken together, these data can guide future prospective studies on immune correlates against congenital HCMV transmission and inform HCMV vaccine and immunotherapeutic development.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus Vaccines , Herpesviridae Infections , Antibodies, Viral , Antibody Formation , Child , Cytomegalovirus , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/therapeutic use , Herpesviridae Infections/drug therapy , Humans , Immunoglobulin G , Prospective Studies
5.
Int J Biol Sci ; 17(11): 2899-2911, 2021.
Article in English | MEDLINE | ID: mdl-34345215

ABSTRACT

Human cytomegalovirus (HCMV), a ubiquitous in humans, has a high prevalence rate. Young people are susceptible to HCMV infection in developing countries, while older individuals are more susceptible in developed countries. Most patients have no obvious symptoms from the primary infection. Studies have indicated that the virus has gradually adapted to the host immune system. Therefore, the control of HCMV infection requires strong immune modulation. With the recent advances in immunotherapy, its application to HCMV infections is receiving increasing attention. Here, we discuss the immune response to HCMV infection, the immune escape mechanism, and the different roles that HCMV plays in various types of immunotherapy, including vaccines, adoptive cell therapy, checkpoint blockade therapy, and targeted antibodies.


Subject(s)
Cytomegalovirus Infections/prevention & control , Immunotherapy/methods , Animals , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/therapeutic use , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive
6.
Int J Mol Sci ; 22(16)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34445434

ABSTRACT

Human Cytomegalovirus (HCMV) infection is widespread and can result in severe sequelae in susceptible populations. Primary HCMV infection of naïve individuals results in life-long latency characterized by frequent and sporadic reactivations. HCMV infection elicits a robust antibody response, including neutralizing antibodies that can block the infection of susceptible cells in vitro and in vivo. Thus, antibody products and vaccines hold great promise for the prevention and treatment of HCMV, but to date, most attempts to demonstrate their safety and efficacy in clinical trials have been unsuccessful. In this review we summarize publicly available data on these products and highlight new developments and approaches that could assist in successful translation of HCMV immunotherapies.


Subject(s)
Cytomegalovirus Infections/prevention & control , Immunotherapy/methods , Animals , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Clinical Trials as Topic , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/therapeutic use , Humans
7.
BMC Med ; 18(1): 174, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32611419

ABSTRACT

BACKGROUND: Infection with cytomegalovirus (CMV) is highly prevalent worldwide and can cause severe disease in immunocompromised persons and congenitally infected infants. The disease burden caused by congenital CMV infection is high, especially in resource-limited countries. Vaccines are currently under development for various target groups. METHODS: We evaluated the impact of vaccination strategies and hygiene intervention using transmission models. Model parameters were estimated from a cross-sectional serological population study (n=5179) and a retrospective birth cohort (n=31,484), providing information on the age- and sex-specific CMV prevalence and on the birth prevalence of congenital CMV (cCMV). RESULTS: The analyses show that vertical transmission and infectious reactivation are the main drivers of transmission. Vaccination strategies aimed at reducing transmission from mother to child (vaccinating pregnant women or women of reproductive age) can yield substantial reductions of cCMV in 20 years (31.7-71.4% if 70% of women are effectively vaccinated). Alternatively, hygiene intervention aimed at preventing CMV infection and re-infection of women of reproductive age from young children is expected to reduce cCMV by less than 2%. The effects of large-scale vaccination on CMV prevalence can be substantial, owing to the moderate transmissibility of CMV at the population level. However, as CMV causes lifelong infection, the timescale on which reductions in CMV prevalence are expected is in the order of several decades. Elimination of CMV infection in the long run is only feasible for a vaccine with a long duration of protection and high vaccination coverage. CONCLUSIONS: Vaccination is an effective intervention to reduce the birth prevalence of cCMV. Population-level reductions in CMV prevalence can only be achieved on a long timescale. Our results stress the value of vaccinating pregnant women and women of childbearing age and provide support for the development of CMV vaccines and early planning of vaccination scenarios and rollouts.


Subject(s)
Cytomegalovirus Infections/transmission , Cytomegalovirus Vaccines/therapeutic use , Cytomegalovirus/immunology , Cross-Sectional Studies , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/pharmacology , Female , Humans , Male , Retrospective Studies
8.
Ann Intern Med ; 172(5): 306-316, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32040960

ABSTRACT

Background: Triplex vaccine was developed to enhance cytomegalovirus (CMV)-specific T cells and prevent CMV reactivation early after hematopoietic stem cell transplant (HCT). Objective: To determine the safety and efficacy of Triplex. Design: First-in-patient, phase 2 trial. (ClinicalTrials.gov: NCT02506933). Setting: 3 U.S. HCT centers. Participants: 102 CMV-seropositive HCT recipients at high risk for CMV reactivation. Intervention: Intramuscular injections of Triplex or placebo were given on days 28 and 56 after HCT. Triplex is a recombinant attenuated poxvirus (modified vaccinia Ankara) expressing immunodominant CMV antigens. Measurements: The primary outcomes were CMV events (CMV DNA level ≥1250 IU/mL, CMV viremia requiring antiviral treatment, or end-organ disease), nonrelapse mortality, and severe (grade 3 or 4) graft-versus-host disease (GVHD), all evaluated through 100 days after HCT, and grade 3 or 4 adverse events (AEs) within 2 weeks after vaccination that were probably or definitely attributable to injection. Results: A total of 102 patients (51 per group) received the first vaccination, and 91 (89.2%) received both vaccinations (46 Triplex and 45 placebo). Reactivation of CMV occurred in 5 Triplex (9.8%) and 10 placebo (19.6%) recipients (hazard ratio, 0.46 [95% CI, 0.16 to 1.4]; P = 0.075). No Triplex recipient died of nonrelapse causes during the first 100 days or had serious AEs, and no grade 3 or 4 AEs related to vaccination were observed within 2 weeks after vaccination. Incidence of severe acute GVHD after injection was similar between groups (hazard ratio, 1.1 [CI, 0.53 to 2.4]; P = 0.23). Levels of long-lasting, pp65-specific T cells with effector memory phenotype were significantly higher in Triplex than placebo recipients. Limitation: The lower-than-expected incidence of CMV events in the placebo group reduced the power of the trial. Conclusion: No vaccine-associated safety concerns were identified. Triplex elicited and amplified CMV-specific immune responses, and fewer Triplex-vaccinated patients had CMV viremia. Primary Funding Source: National Cancer Institute and Helocyte.


Subject(s)
Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/therapeutic use , Cytomegalovirus , Hematopoietic Stem Cell Transplantation/adverse effects , Viremia/prevention & control , Aged , Cytomegalovirus/immunology , Double-Blind Method , Female , Humans , Male , Middle Aged
9.
Curr Hematol Malig Rep ; 15(2): 90-102, 2020 04.
Article in English | MEDLINE | ID: mdl-31981100

ABSTRACT

PURPOSE OF REVIEW: CMV DNA polymerase inhibitors such as ganciclovir and foscarnet have dramatically reduced the burden of CMV infection in the HCT recipient. However, their use is often limited by toxicities and resistance. Agents with novel mechanisms and favorable toxicity profiles are critically needed. We review recent developments in CMV antivirals and immune-based approaches to mitigating CMV infection. RECENT FINDINGS: Letermovir, an inhibitor of the CMV terminase complex, was approved in 2017 for primary CMV prophylaxis in adult seropositive allogeneic HCT recipients. Maribavir, an inhibitor of the CMV UL97 kinase, is currently in two phase 3 treatment studies. Adoptive immunotherapy using third-party T cells has proven safe and effective in preliminary studies. Vaccine development continues, with several promising candidates currently under study. No longer limited to DNA polymerase inhibitors, the prevention and treatment of CMV infections in the HCT recipient is a rapidly evolving field which should translate into improvements in CMV-related outcomes.


Subject(s)
Antiviral Agents/therapeutic use , Cytomegalovirus Infections/therapy , Cytomegalovirus/drug effects , Hematopoietic Stem Cell Transplantation , Immunotherapy/trends , Molecular Targeted Therapy/trends , Opportunistic Infections/therapy , Animals , Antiviral Agents/adverse effects , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/therapeutic use , Drug Resistance, Viral , Foscarnet/therapeutic use , Ganciclovir/therapeutic use , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunocompromised Host , Immunosuppressive Agents/adverse effects , Immunotherapy/adverse effects , Immunotherapy, Adoptive/trends , Molecular Targeted Therapy/adverse effects , Opportunistic Infections/immunology , Opportunistic Infections/virology , Risk Factors , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Treatment Outcome
10.
J Infect Dis ; 221(5): 715-720, 2020 02 18.
Article in English | MEDLINE | ID: mdl-31593588

ABSTRACT

BACKGROUND: Infection with multiple cytomegalovirus (CMV) strains (mixed infection) was reported in a variety of hosts. As the virus genetic diversity in primary CMV infection and the changes over time remain incompletely defined, we examined CMV diversity and changes in diversity over time in healthy adolescent females who participated in a phase 2 CMV gB/MF59 vaccine trial. METHODS: CMV genetic diversity was determined by genotyping of 5 genes-gB (UL55), gH (UL75), gN (UL73), US28, and UL144-in urine, saliva, and plasma samples from 15 study subjects. RESULTS: At the time of primary infection, 5 of 12 (42%) urine samples had multiple virus strains, and 50% of vaccine recipients were infected with gB1 genotype (vaccine strain). Mixed infection was documented in all 15 subjects within 3 months after primary infection, and the majority had different CMV genotypes in different compartments. Changes in genotypes over time were observed in all subjects. CONCLUSIONS: Infection with multiple CMV genotypes was common during primary infection and further diversification occurred over time. Infection with gB1 genotype in vaccine recipients suggests a lack of strain-specific protection from the vaccine. As only 5 polymorphic genes were assessed, this study likely underestimated the true genetic diversity in primary CMV infection.


Subject(s)
Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/therapeutic use , Cytomegalovirus/genetics , Polymorphism, Genetic , Vaccination , Adolescent , Coinfection/diagnosis , Coinfection/virology , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/virology , Double-Blind Method , Female , Genotype , Humans , Membrane Glycoproteins/blood , Membrane Glycoproteins/genetics , Membrane Glycoproteins/urine , Real-Time Polymerase Chain Reaction , Receptors, Chemokine/blood , Receptors, Chemokine/genetics , Saliva/virology , Viral Envelope Proteins/blood , Viral Envelope Proteins/genetics , Viral Envelope Proteins/urine , Viral Load , Viral Proteins/blood , Viral Proteins/genetics , Viral Proteins/urine
11.
Proc Natl Acad Sci U S A ; 116(26): 13036-13041, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31189602

ABSTRACT

Human cytomegalovirus (HCMV) causes severe disease in infants and immunocompromised people. There is no approved HCMV vaccine, and vaccine development strategies are complicated by evidence of both persistent infection and reinfection of people with prior immunity. The greatest emphasis has been placed on reducing transmission to seronegative pregnant women to prevent vertical transmission and its potentially severe sequelae. Increasing evidence suggests that the earliest host-HCMV interactions establish conditions for viral persistence, including evasion of host immune responses to the virus. Using a nonhuman primate model of HCMV infection, we show that rhesus macaques immunized against viral interleukin-10 (IL-10) manifest delayed rhesus cytomegalovirus (RhCMV) acquisition and altered immune responses to the infection when it does occur. Among animals with the greatest antiviral IL-10-neutralizing activity, the timing of RhCMV seroconversion was delayed by an average of 12 weeks. After acquisition, such animals displayed an antibody response to the new infection, which peaked as expected after 2 weeks but then declined rapidly. In contrast, surprisingly, vaccination with glycoprotein B (gB) protein had no discernible impact on these outcomes. Our results demonstrate that viral IL-10 is a key regulator of successful host immune responses to RhCMV. Viral IL-10 is, therefore, an important target for vaccine strategies against cytomegalovirus (CMV). Furthermore, given the immunoregulatory function of viral IL-10, targeting this protein may prove synergistic with other vaccine therapies and targets. Our study also provides additional evidence that the earliest host-CMV interactions can have a significant impact on the nature of persistent infection.


Subject(s)
Antigens, Viral/immunology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/pharmacology , Cytomegalovirus/immunology , Infectious Disease Transmission, Vertical/prevention & control , Interleukin-10/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigens, Viral/administration & dosage , Cytomegalovirus Infections/blood , Cytomegalovirus Infections/transmission , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/immunology , Cytomegalovirus Vaccines/therapeutic use , Disease Models, Animal , Female , Host-Pathogen Interactions/immunology , Humans , Immunity, Mucosal , Immunogenicity, Vaccine , Interleukin-10/administration & dosage , Macaca mulatta , Pregnancy , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Viral Envelope Proteins/administration & dosage , Virus Shedding/immunology
12.
J Clin Virol ; 116: 58-61, 2019 07.
Article in English | MEDLINE | ID: mdl-31132546

ABSTRACT

The natural history of cytomegalovirus (CMV) infection in transplant patients has been well established. This virus may originate from the recipient, the donor or both. When pre-transplant IgG antibodies in the recipient are taken into account, three types of infection are possible: primary, reactivation or reinfection. The risks of high viral load and end-organ disease are highest after primary infection and lowest after reactivation. Serial monitoring of patients by quantitative polymerase chain reaction for CMV DNA allows antiviral drugs to be deployed for pre-emptive therapy or an antiviral drug may be given prophylactically. Both of these strategies are effective, but pre-emptive therapy has the advantage that randomised allocation of a new drug or placebo given prophylactically may show a reduced need for pre-emptive valganciclovir. In this review, I will consider what has been learned from use of ganciclovir and valganciclovir and apply this information to clinical trials that have evaluated maribavir, brincidofovir and letermovir. In addition, pre-emptive therapy has the advantage of facilitating the discovery of vaccines against CMV using a pharmacodynamic approach. Briefly, patients awaiting transplantation are given vaccine or placebo pre-transplant. When they proceed to transplantation, various parameters of viral load can be compared to determine if the vaccine has an effect against CMV when compared to patients randomised to receive placebo. If there is evidence of control of CMV, this can be related to immune responses induced by the vaccine to define a correlate of protection. This review will summarise the published evidence available.


Subject(s)
Antiviral Agents/therapeutic use , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/therapeutic use , Cytomegalovirus/drug effects , Clinical Trials as Topic , Combined Modality Therapy , Cytomegalovirus/immunology , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/immunology , Humans , Transplantation/adverse effects , Viral Load/drug effects
13.
J Virol ; 92(7)2018 04 01.
Article in English | MEDLINE | ID: mdl-29343580

ABSTRACT

Human cytomegalovirus (HCMV) is the most common congenitally transmitted pathogen worldwide, impacting an estimated 1 million newborns annually. Congenital HCMV (cCMV) infection is a major global contributor to long-term neurologic deficits, including deafness, microcephaly, and neurodevelopmental delay, as well as to fetal loss and occasional infant mortality. Accordingly, design of a maternal vaccine to prevent cCMV continues to be a top public health priority. Nevertheless, we remain without a licensed vaccine. Maternal immunity provides partial protection, as the risk of vertical HCMV transmission from chronically infected mothers is reduced compared to settings in which the mother is newly infected during pregnancy. Therefore, an understanding of the maternal immune correlates of protection against cCMV is critical to informing design of an efficacious maternal vaccine. Although vaccine development is being assiduously pursued by a large number of pharmaceutical manufacturers, biotechnology organizations, and academic researchers, some pessimism has been expressed regarding the issue of whether a vaccine to protect against cCMV is possible. This pessimism is based on observations that natural immunity is not completely protective against maternal reinfection and congenital transmission. However, we assert that optimism regarding vaccine development is indeed justified, on the basis of accruing evidence of immune correlates of protection-readily achievable by vaccination-that are associated with reduced transmission of HCMV to the fetus in seronegative women. In light of the substantial burden on society conferred by cCMV infection, even a modest reduction in the occurrence of this fetal disease is an important public health goal and justifies aggressive clinical evaluation of vaccines currently in the pipeline.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus Vaccines , Cytomegalovirus/immunology , Immunity, Maternally-Acquired/immunology , Infectious Disease Transmission, Vertical/prevention & control , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/immunology , Cytomegalovirus Vaccines/therapeutic use , Female , Humans , Pregnancy
14.
Placenta ; 60 Suppl 1: S73-S79, 2017 12.
Article in English | MEDLINE | ID: mdl-28456432

ABSTRACT

Human cytomegalovirus (HCMV) is the most common congenitally transmitted pathogen worldwide, impacting an estimated 1 million newborns annually. In a subset of infected infants, congenital HCMV causes severe, long-lasting sequelae, including deafness, microcephaly, neurodevelopmental delay, and even death. Accordingly, a maternal vaccine to prevent congenital HCMV infection continues to be a top public health priority. Nevertheless, all vaccines tested to date have failed to meet clinical trial endpoints. Maternal immunity provides partial protection against congenital HCMV transmission, as vertical transmission from seropositive mothers is relatively rare. Therefore, an understanding of the maternal immune correlates of protection against HCMV congenital infection will be critical to inform design of an efficacious maternal vaccine. This review summarizes our understanding of the innate and adaptive immune correlates of protection against congenital transmission of HCMV, and discusses the advantages and applications of a novel nonhuman primate model of congenital CMV transmission to aid in rational vaccine design and evaluation.


Subject(s)
Adaptive Immunity , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Immunity, Innate , Maternal-Fetal Exchange , Placenta/immunology , Pregnancy Complications, Infectious/immunology , Animals , Biomedical Research/methods , Biomedical Research/trends , Cytomegalovirus/isolation & purification , Cytomegalovirus/physiology , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/transmission , Cytomegalovirus Vaccines/therapeutic use , Disease Models, Animal , Female , Humans , Immunity, Cellular , Macaca mulatta , Placenta/virology , Pregnancy , Pregnancy Complications, Infectious/virology
15.
Sci Transl Med ; 8(362): 362ra145, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27797961

ABSTRACT

Congenital human cytomegalovirus (HCMV) infection occurs in ~0.64% of infants born each year in the United States and is the leading nongenetic cause of childhood neurodevelopmental disabilities. No licensed HCMV vaccine is currently available. Natural immunity to HCMV in women before pregnancy is associated with a reduced risk of fetal infection, suggesting that a vaccine is feasible if it can reproduce immune responses elicited by natural infection. On the basis of this premise, we developed a whole-virus vaccine candidate from the live attenuated AD169 strain, with genetic modifications to improve its immunogenicity and attenuation. We first restored the expression of the pentameric gH/gL/pUL128-131 protein complex, a major target for neutralizing antibodies in natural immunity. We then incorporated a chemically controlled protein stabilization switch in the virus, enabling us to regulate viral replication with a synthetic compound named Shield-1. The virus replicated as efficiently as its parental virus in the presence of Shield-1 but failed to produce progeny upon removal of the compound. The vaccine was immunogenic in multiple animal species and induced durable neutralizing antibodies, as well as CD4+ and CD8+ T cells, to multiple viral antigens in nonhuman primates.


Subject(s)
Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/therapeutic use , Infectious Disease Transmission, Vertical/prevention & control , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cytomegalovirus , Female , Humans , Immunogenicity, Vaccine , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Pregnancy , Rabbits , Vaccines, Attenuated/therapeutic use , Viral Proteins/immunology
16.
Pharmazie ; 71(9): 499-503, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-29441844

ABSTRACT

OBJECTIVE: To evaluate the effectiveness and safety of V-5 immunitor (V5) added to chemotherapy of tuberculosis (TB) patients. METHODS: The databases Medline, Embase, Biosis, Cochrane Central Register of Controlled Trials, SCI, CBM, VIP and CNKI were searched. Randomized controlled trials (RCT) and controlled clinical trials (CCT) of V5 immunitor with or without a placebo-control as adjuvant therapy in the chemotherapy of TB patients were included. Two reviewers independently performed data extraction and quality assessment. Data were analyzed using RevMan 5.3 software by The Cochrane Collaboration. RESULTS: Four studies were included. At the end of the follow-up period, pooled RR (Risk Ratio) and its 95% CI of sputum smear conversion rate were 4.91 (3.32, 7.28) in drug-sensitive, drug-resistant TB patients or HIV-TB co-infection patients. When analyzing inflammation biomarkers including ESR and leukocyte accounts, pooled mean difference and its 95% CI of ESR and leukocyte accounts were -7.62 (-9.55, -5.68) and -2.13 (-2.58, -1.68), respectively. As to body weight, pooled mean difference and its 95% CI were 0.96 (-1.13, 3.05) in TB patients. Two clinical trials were included for analyzing temperature after using V5 immunitor, pooled mean difference and its 95% CI were -0.34 (-0.46, -0.22) in TB patients. These results suggested that V5 immunitor holds important promise in improving sputum conversion to AFB- and inhibiting inflammatory reaction in TB patients, but showed no significant promotion to the increase in body weight based on this meta-analysis. Compared with the control group, V5 immunitor may have some potential in decreasing the temperature of TB patients. No systemic adverse events were reported. CONCLUSION: Added to chemotherapy, V5 immunitor seems to be helpful in the treatment of TB patients in terms of improving sputum conversion and reducing inflammatory reactions.


Subject(s)
Antitubercular Agents/therapeutic use , Cytomegalovirus Vaccines/therapeutic use , Hepatitis B Vaccines/therapeutic use , Tuberculosis, Pulmonary/drug therapy , Antitubercular Agents/adverse effects , Cytomegalovirus Vaccines/adverse effects , Hepatitis B Vaccines/adverse effects , Humans , Randomized Controlled Trials as Topic , Tuberculosis, Pulmonary/microbiology
17.
Immunotherapy ; 7(10): 1059-71, 2015.
Article in English | MEDLINE | ID: mdl-26507225

ABSTRACT

Cytomegalovirus (CMV) remains a major cause of morbidity and mortality in allogeneic hematopoietic cell transplantation. Advances in surveillance of cytomegalovirus reactivation using sensitive techniques and a preemptive strategy to treat virus reactivation has reduced incidence of cytomegalovirus end organ disease. However, severe immunosuppression associated with extensive T-cell depletion resulting from graft-versus-host disease prevention for cases of mismatched or others such as haploidentical allogeneic hematopoietic cell transplantation (allo-HCT) and graft-versus-host disease therapy itself create clinical challenges in managing cytomegalovirus infection. Novel anticytomegalovirus therapies including newer pharmacologic interventions, vaccines, and adoptive cellular therapies to restore anticytomegalovirus immunity appear promising and are expected to continue to shape our treatment armamentarium. Eradication of CMV disease altogether, rather than simply suppressing viremia, should be the ultimate desirable goal.


Subject(s)
Antiviral Agents/therapeutic use , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/therapeutic use , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Cytomegalovirus/physiology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunosuppression Therapy , Lymphocyte Depletion , Transplantation, Homologous , Virus Activation
18.
Hum Vaccin Immunother ; 11(7): 1788-802, 2015.
Article in English | MEDLINE | ID: mdl-25984886

ABSTRACT

We present an age-structured dynamic transmission model for cytomegalovirus (CMV) in the United States, based on natural history and available data, primarily aiming to combine the available qualitative and quantitative knowledge toward more complex modeling frameworks to better reflect the underlying biology and epidemiology of the CMV infection. The model structure explicitly accounts for primary infections, reactivations and re-infections. Duration of infectiousness and likelihood of reactivation were both assumed to be age-dependent, and natural reduction in the re-infection risk following primary infection was included. We used an empirical social contact matrix (POLYMOD-based) as support for CMV transmission between different age groups. The baseline model reproduced well the age-stratified seroprevalence data (National Health and Nutrition Examination Survey III) used for calibration. The model was further used to explore the potential impact of hypothetical vaccination on reducing congenital CMV infection under various vaccine profiles and vaccination scenarios. Our preliminary model-based simulations suggested that while infant vaccination may represent an attractive way to reduce congenital CMV infection over time, adolescent female vaccination with an adequate routine booster platform may, under certain conditions, provide an alternative. However, for such tools to be considered toward actual decision-making, enhanced validations based on additional studies and data would be further necessary. The modeling framework presented in this paper was designed to be sufficiently general and flexible, such that it can allow for further adaptations to reflect new knowledge or data that may become available in the future.


Subject(s)
Cytomegalovirus Infections/transmission , Cytomegalovirus Vaccines/therapeutic use , Cytomegalovirus/immunology , Immunization Programs/organization & administration , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Child , Child, Preschool , Cytomegalovirus Infections/epidemiology , Female , Forecasting , Health Planning , Humans , Immunization, Secondary , Infant , Male , Middle Aged , Models, Statistical , Reproducibility of Results , Seroepidemiologic Studies , United States/epidemiology , Vaccination , Young Adult
19.
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
20.
Clin Perinatol ; 42(1): 61-75, viii, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25677997

ABSTRACT

Mother-to-child transmission of cytomegalovirus (CMV) and varicella zoster virus (VZV) can lead to severe birth defects and neurologic impairment of infants. Congenital CMV complicates up to 1% of all pregnancies globally. Although antiviral treatment of infants congenitally infected with CMV can ameliorate the CMV-associated hearing loss and developmental delay, interventions to prevent congenital CMV infection and the associated neurologic impairments are still being evaluated. Congenital VZV infection is rare. Active and passive immunization strategies to prevent perinatal CMV infection with similar efficacy to those established to prevent perinatal VZV infections are critically needed in pediatric health.


Subject(s)
Chickenpox Vaccine/therapeutic use , Chickenpox/prevention & control , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/therapeutic use , Pregnancy Complications, Infectious/prevention & control , Antiviral Agents/therapeutic use , Chickenpox/drug therapy , Chickenpox/epidemiology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/epidemiology , Female , Humans , Infant, Newborn , Male , Pregnancy , Pregnancy Complications, Infectious/drug therapy , Pregnancy Complications, Infectious/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...