1
|
Scarpini S, Morigi F, Betti L, Dondi A, Biagi C, Lanari M. Development of a Vaccine against Human Cytomegalovirus: Advances, Barriers, and Implications for the Clinical Practice. Vaccines (Basel) 2021; 9:551. [PMID: 34070277 PMCID: PMC8225126 DOI: 10.3390/vaccines9060551] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 12/13/2022] Open
Abstract
Human cytomegalovirus (hCMV) is one of the most common causes of congenital infection in the post-rubella era, representing a major public health concern. Although most cases are asymptomatic in the neonatal period, congenital CMV (cCMV) disease can result in permanent impairment of cognitive development and represents the leading cause of non-genetic sensorineural hearing loss. Moreover, even if hCMV mostly causes asymptomatic or pauci-symptomatic infections in immunocompetent hosts, it may lead to severe and life-threatening disease in immunocompromised patients. Since immunity reduces the severity of disease, in the last years, the development of an effective and safe hCMV vaccine has been of great interest to pharmacologic researchers. Both hCMV live vaccines-e.g., live-attenuated, chimeric, viral-based-and non-living ones-subunit, RNA-based, virus-like particles, plasmid-based DNA-have been investigated. Encouraging data are emerging from clinical trials, but a hCMV vaccine has not been licensed yet. Major difficulties in the development of a satisfactory vaccine include hCMV's capacity to evade the immune response, unclear immune correlates for protection, low number of available animal models, and insufficient general awareness. Moreover, there is a need to determine which may be the best target populations for vaccine administration. The aim of the present paper is to examine the status of hCMV vaccines undergoing clinical trials and understand barriers limiting their development.
Collapse
Affiliation(s)
- Sara Scarpini
- Specialty School of Pediatrics, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (S.S.); (F.M.); (L.B.)
| | - Francesca Morigi
- Specialty School of Pediatrics, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (S.S.); (F.M.); (L.B.)
| | - Ludovica Betti
- Specialty School of Pediatrics, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (S.S.); (F.M.); (L.B.)
| | - Arianna Dondi
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (C.B.); (M.L.)
| | - Carlotta Biagi
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (C.B.); (M.L.)
| | - Marcello Lanari
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (C.B.); (M.L.)
| |
Collapse
|
2
|
Plotkin SA, Wang D, Oualim A, Diamond DJ, Kotton CN, Mossman S, Carfi A, Anderson D, Dormitzer PR. The Status of Vaccine Development Against the Human Cytomegalovirus. J Infect Dis 2021; 221:S113-S122. [PMID: 32134478 DOI: 10.1093/infdis/jiz447] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Numerous candidate vaccines against cytomegalovirus (CMV) infection and disease are in development. Whereas the previous article [1] provides background and opinions about the issues relating to vaccination, this article provides specifics about the vaccines in active development, as reported at a National Institutes of Health-sponsored meeting in Bethesda on September 4-6, 2018. Here, vaccine developers provide synopses of their candidate vaccines to immunize women to protect against congenital CMV disease and to prevent the consequences of CMV disease in recipients of transplanted organs or hematopoietic stem calls. The projects are presented here roughly in the descending order of their stage of development in the opinion of the first author.
Collapse
Affiliation(s)
- Stanley A Plotkin
- Department of Pediatrics, University of Pennsylvania, Vaxconsult, Doylestown, Pennsylvania, USA
| | - Dai Wang
- Merck & Co., Kenilworth, New Jersey, USA
| | | | - Don J Diamond
- City of Hope National Medical Center, Duarte, California, USA
| | | | | | - Andrea Carfi
- Moderna Therapeutics, Cambridge, Massachusetts, USA
| | | | | |
Collapse
|
3
|
Abstract
The way to a successful vaccine against human cytomegalovirus is hampered by the peculiar biology of this infection. However, some candidate vaccines have been shown to protect seronegative women and transplant recipients, and we should know soon whether they can prevent congenital infection.
Collapse
Affiliation(s)
- Stanley A Plotkin
- Department of Pediatrics, University of Pennsylvania, Vaxconsult, Doylestown, Pennsylvania, USA
| |
Collapse
|
4
|
Where do we Stand after Decades of Studying Human Cytomegalovirus? Microorganisms 2020; 8:microorganisms8050685. [PMID: 32397070 PMCID: PMC7284540 DOI: 10.3390/microorganisms8050685] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 12/26/2022] Open
Abstract
Human cytomegalovirus (HCMV), a linear double-stranded DNA betaherpesvirus belonging to the family of Herpesviridae, is characterized by widespread seroprevalence, ranging between 56% and 94%, strictly dependent on the socioeconomic background of the country being considered. Typically, HCMV causes asymptomatic infection in the immunocompetent population, while in immunocompromised individuals or when transmitted vertically from the mother to the fetus it leads to systemic disease with severe complications and high mortality rate. Following primary infection, HCMV establishes a state of latency primarily in myeloid cells, from which it can be reactivated by various inflammatory stimuli. Several studies have shown that HCMV, despite being a DNA virus, is highly prone to genetic variability that strongly influences its replication and dissemination rates as well as cellular tropism. In this scenario, the few currently available drugs for the treatment of HCMV infections are characterized by high toxicity, poor oral bioavailability, and emerging resistance. Here, we review past and current literature that has greatly advanced our understanding of the biology and genetics of HCMV, stressing the urgent need for innovative and safe anti-HCMV therapies and effective vaccines to treat and prevent HCMV infections, particularly in vulnerable populations.
Collapse
|
5
|
Human Cytomegalovirus Congenital (cCMV) Infection Following Primary and Nonprimary Maternal Infection: Perspectives of Prevention through Vaccine Development. Vaccines (Basel) 2020; 8:vaccines8020194. [PMID: 32340180 PMCID: PMC7349293 DOI: 10.3390/vaccines8020194] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/13/2020] [Accepted: 04/18/2020] [Indexed: 01/26/2023] Open
Abstract
Congenital cytomegalovirus (cCMV) might occur as a result of the human cytomegalovirus (HCMV) primary (PI) or nonprimary infection (NPI) in pregnant women. Immune correlates of protection against cCMV have been partly identified only for PI. Following either PI or NPI, HCMV strains undergo latency. From a diagnostic standpoint, while the serological criteria for the diagnosis of PI are well-established, those for the diagnosis of NPI are still incomplete. Thus far, a recombinant gB subunit vaccine has provided the best results in terms of partial protection. This partial efficacy was hypothetically attributed to the post-fusion instead of the pre-fusion conformation of the gB present in the vaccine. Future efforts should be addressed to verify whether a new recombinant gB pre-fusion vaccine would provide better results in terms of prevention of both PI and NPI. It is still a matter of debate whether human hyperimmune globulin are able to protect from HCMV vertical transmission. In conclusion, the development of an HCMV vaccine that would prevent a significant portion of PI would be a major step forward in the development of a vaccine for both PI and NPI.
Collapse
|
6
|
Abstract
Vaccines already developed have been enormously successful. However, the development of future vaccines requires solution of a number of immunologic problems, including pathogen variability, short effector memory, evoking functional responses, and identification of antigens that generate protective responses. In addition, different populations may respond differently to the same vaccine because of genetic, age, or environmental factors.
Collapse
|
7
|
Plotkin S. The history of vaccination against cytomegalovirus. Med Microbiol Immunol 2015; 204:247-54. [PMID: 25791890 DOI: 10.1007/s00430-015-0388-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/25/2015] [Indexed: 12/22/2022]
Abstract
Cytomegalovirus vaccine development started in the 1970s with attenuated strains. In the 1980s, one of the strains was shown to be safe and effective in renal transplant patients. Then, attention switched to glycoprotein gB, which was shown to give moderate but transient protection against acquisition of the virus by women. The identification of the pp65 tegument protein as the principal target of cellular immune responses resulted in new approaches, particularly DNA, plasmids to protect hematogenous stem cell recipients. The subsequent discovery of the pentameric protein complex that generates most neutralizing antibodies led to efforts to incorporate that complex into vaccines. At this point, there are many candidate CMV vaccines, including live recombinants, replication-defective virus, DNA plasmids, soluble pentameric proteins, peptides, virus-like particles and vectored envelope proteins.
Collapse
Affiliation(s)
- Stanley Plotkin
- Emeritus Professor of Pediatrics, University of Pennsylvania, 4650 Wismer Road, Doylestown, PA, 18902, USA,
| |
Collapse
|
8
|
Human cytomegalovirus vaccine based on the envelope gH/gL pentamer complex. PLoS Pathog 2014; 10:e1004524. [PMID: 25412505 PMCID: PMC4239111 DOI: 10.1371/journal.ppat.1004524] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 10/16/2014] [Indexed: 12/22/2022] Open
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. Human cytomegalovirus (HCMV) fetal infection during pregnancy and infection of immunocompromised patients are both clinical problems considered extremely important by the Institute of Medicine. Limited efficacy against primary HCMV infection was found using a subunit vaccine based on glycoprotein B, an important neutralizing antibody determinant blocking HCMV entry into fibroblasts. The HCMV field has been transformed by the discovery that a five-member (pentamer) protein complex is a required factor for epithelial and endothelial cell entry and indispensable for transmission as shown in non-human primates. Targeting HCMV with antibodies specific to the pentamer may interrupt horizontal and vertical transmission. We describe an innovative vaccine strategy to induce serum neutralizing antibodies of impressive magnitude against HCMV in two animal models. Using an attenuated poxvirus vector system, we demonstrate that co-expression of all five pentamer components is significantly more potent to induce serum neutralizing antibodies than subunit subsets of the complex or glycoprotein B, reaching peak levels comparable to HCMV hyperimmune globulin. A vaccine that elicits systemic and mucosal antibody responses that prevents infection of multiple cell types crucial to natural history of HCMV infection could play a role in preventing congenital HCMV infection and control of infection in immunocompromised patients.
Collapse
|
9
|
Immunobiology of human cytomegalovirus: from bench to bedside. Clin Microbiol Rev 2009; 22:76-98, Table of Contents. [PMID: 19136435 DOI: 10.1128/cmr.00034-08] [Citation(s) in RCA: 469] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
SUMMARY Following primary infection, human cytomegalovirus (HCMV) establishes lifelong latency and periodically reactivates without causing symptoms in healthy individuals. In the absence of an adequate host-derived immune response, this fine balance of permitting viral reactivation without causing pathogenesis is disrupted, and HCMV can subsequently cause invasive disease and an array of damaging indirect immunological effects. Over the last decade, our knowledge of the immune response to HCMV infection in healthy virus carriers and diseased individuals has allowed us to translate these findings to develop better diagnostic tools and therapeutic strategies. The application of these emerging technologies in the clinical setting is likely to provide opportunities for better management of patients with HCMV-associated diseases.
Collapse
|
10
|
|
11
|
Schleiss MR, Heineman TC. Progress toward an elusive goal: current status of cytomegalovirus vaccines. Expert Rev Vaccines 2006; 4:381-406. [PMID: 16026251 DOI: 10.1586/14760584.4.3.381] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Although infection with human cytomegalovirus (CMV) is ubiquitous and generally asymptomatic in most individuals, certain patient populations are at high risk for CMV-associated disease. These include HIV-infected individuals with AIDS, transplant patients, and newborn infants with congenital CMV infection. Immunity to CMV infection, both in the transplant setting and among women of childbearing age, plays a vital role in the control of CMV-induced injury and disease. Although immunity induced by CMV infection is not completely protective against reinfection, there is nevertheless a sound basis on which to believe that vaccination could help control CMV disease in high-risk patient populations. Evidence from several animal models of CMV infection indicates that a variety of vaccine strategies are capable of inducing immune responses sufficient to protect against CMV-associated illness following viral challenge. Vaccination has also proven effective in improving pregnancy outcomes following CMV challenge of pregnant guinea pigs, providing a 'proof-of-principle' relevant to human clinical trials of CMV vaccines. Although there are no licensed vaccines currently available for human CMV, progress toward this goal has been made, as evidenced by ongoing clinical trial testing of a number of immunization strategies. CMV vaccines currently in various stages of preclinical and clinical testing include: protein subunit vaccines; DNA vaccines; vectored vaccines using viral vectors, such as attenuated pox- and alphaviruses; peptide vaccines; and live attenuated vaccines. This review summarizes some of the obstacles that must be overcome in development of a CMV vaccine, and provides an overview of the current state of preclinical and clinical trial evaluation of vaccines for this important public health problem.
Collapse
Affiliation(s)
- Mark R Schleiss
- University of Minnesota School of Medicine, 420 Delaware Street SE, MMC 296, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
12
|
Wang Z, La Rosa C, Lacey SF, Maas R, Mekhoubad S, Britt WJ, Diamond DJ. Attenuated poxvirus expressing three immunodominant CMV antigens as a vaccine strategy for CMV infection. J Clin Virol 2006; 35:324-31. [PMID: 16388983 DOI: 10.1016/j.jcv.2005.09.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2005] [Revised: 08/26/2005] [Accepted: 09/15/2005] [Indexed: 10/25/2022]
Abstract
BACKGROUND Human cytomegalovirus (CMV) infection is an important risk factor in the post-transplant (Tx) recovery phase for both hematopoietic stem cell Tx (HSCT) and solid organ Tx (SOT) recipients. CMV infection may be prevented or controlled by simultaneously inducing both CMV-specific neutralizing antibody (nAb) and cellular immunity. Soluble (s) UL55 (surface glycoprotein), UL83 (tegument protein) and UL123/e4 (nuclear protein) are immunodominant in eliciting both CMV nAb and cellular immunity. An attenuated poxvirus, modified vaccinia Ankara (MVA) was selected to develop this vaccine strategy in Tx recipients, because of its clinical safety record, large foreign gene capacity, and capability to activate strong humoral and cellular immune responses against recombinant antigens. OBJECTIVES A subunit vaccine that targets multiple CMV antigens will be used to gain maximal coverage and protective function against CMV infection. rMVA simultaneously expressing sUL55, UL83 and UL123/e4 will be generated, and humoral and cellular immunity it elicits will be characterized, after murine immunization and in vitro to amplify clinical recall responses. STUDY DESIGN rMVA will be constructed in two steps using UL123/e4-pLW22 followed by sUL55-UL83-pLW51 transfer plasmids. Western blots will be used to characterize expression levels of each antigen. Primary immunity will be evaluated in mouse models, while recall responses to the virally expressed CMV antigens will be assessed in human peripheral blood. RESULTS We generated CMV-MVA via homologous recombination, and demonstrated high expression levels of sUL55, UL83 and UL123/e4 by Western blot. CMV-MVA immunization potently induced both humoral and cellular immunity to sUL55, UL83 and UL123 after murine immunization, and cellular immunity to UL83 and UL123 by in vitro amplification of T cell recall responses in human PBMC. CONCLUSIONS rMVA promotes high level expression of three immunodominant CMV antigens, which is reflected in results of immunization studies in which high titers of UL55-specific antibodies and CD4+ T-help are detected, as well as high levels of UL83-specific and moderate levels of UL123-specific CD8+ CTL.
Collapse
Affiliation(s)
- Zhongde Wang
- Laboratory of Vaccine Research, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Khanna R, Diamond DJ. Human cytomegalovirus vaccine: time to look for alternative options. Trends Mol Med 2005; 12:26-33. [PMID: 16337831 DOI: 10.1016/j.molmed.2005.11.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Revised: 11/02/2005] [Accepted: 11/24/2005] [Indexed: 10/25/2022]
Abstract
In 1999, the Institute of Medicine (IOM) of the National Academy of Sciences (USA) assigned the highest priority for a vaccine to prevent congenital human cytomegalovirus (HCMV) infection, on the basis of the life-time cost to the health care system and the impact of the virus on human suffering. Subsequently, this priority was also endorsed by the US National Vaccine Program Office, which proposed a series of recommendations including support for increased funding by government agencies for HCMV vaccine research. Despite numerous attempts over the past three decades, successful licensure of a HCMV vaccine formulation remains elusive. Here we review the current status of HCMV vaccine studies and, based on our recent understanding of immune regulation of HCMV infection, we argue that a more realistic goal for vaccine strategies should be preventing HCMV disease rather than infection.
Collapse
Affiliation(s)
- Rajiv Khanna
- Tumour Immunology Laboratory, Cooperative Research Centre for Vaccine Technology, Division of Infectious Diseases and Immunology, Queensland Institute of Medical Research, 300 Herston Road, Brisbane, 4006, Australia
| | | |
Collapse
|
14
|
Rios LS, Vallochi AL, Muccioli C, Campos-Machado MA, Belfort R, Rizzo LV. Cytokine profile in response to Cytomegalovirus associated with immune recovery syndrome after highly active antiretroviral therapy. Can J Ophthalmol 2005; 40:711-20. [PMID: 16391635 DOI: 10.1016/s0008-4182(05)80087-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Several changes have occurred in the presentation and course of cytomegalovirus (CMV) retinitis in patients with AIDS since the introduction of HAART (highly active antiretroviral therapy). In some individuals who take HAART, retinitis is kept under control even after the discontinuation of anti-CMV therapy. However, many of these patients develop intraocular inflammation. Uveitis, cataract, vitreitis, cystoid macular edema, epiretinal membrane, and disc edema may occur in patients with immune recovery syndrome (IRS). METHODS We evaluated the CMV-specific immune response in 55 patients by assessing CMV-specific lymphocyte proliferation, cytotoxicity, and cytokine production and correlated it with the clinical outcome. RESULTS Our data suggest that control of CMV retinitis is associated with acquisition of cytotoxic and lymphoproliferative responses to CMV. In addition, the upsurge of macular and disc edema seems associated with the production of interleukin-4 and tumor necrosis factor-alpha, whereas vitreitis is associated with the production of interleukin-2 and interferon-gamma. INTERPRETATION The type of T-cell response that develops after HAART may determine the side effects of immune recovery and these effects are predictable based on the lymphokine profile produced by CMV-specific cells.
Collapse
Affiliation(s)
- Lilia S Rios
- Department of Immunology, Biomedical Sciences Institute, University of São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
15
|
Gaytant MA, Galama JMD, Semmekrot BA, Melchers WJG, Sporken JMM, Oosterbaan HP, van Dop PA, Huisman A, Merkus HMWM, Steegers EAP. The incidence of congenital cytomegalovirus infections in The Netherlands. J Med Virol 2005; 76:71-5. [PMID: 15778958 DOI: 10.1002/jmv.20325] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cytomegalovirus (CMV) is one of the most common causes of congenital infection without an effective treatment or an effective vaccine available to date. The emphasis has to be on preventive strategies, which rely on the epidemiological situation. The incidence of congenital CMV infections, however, is not known for The Netherlands. Therefore, a prospective virological study was carried out in a population of 7,524 pregnant women and 7,793 newborns. CMV-specific IgG antibodies were determined in cord blood by ELISA. When CMV antibodies were present, a CMV specific PCR was performed on the throat swab. A positive PCR was confirmed by urine culture. In addition, the seroepidemiology for CMV was investigated in the metropolitan region (Amsterdam and Rotterdam) which has a different ethnic composition. Congenital CMV infection was found in 7 infants (0.9 per 1,000). None had symptoms at birth or during 24 month follow-up. Carriage or CMV was 41%, with a variation between 35% and 100% depending on ethnicity. The ethnic composition in the south-eastern region was different from that in large cities, but similar to that in the rest of the country. The incidence of congenital CMV infections in The Netherlands is the lowest described to date, which does not justify special preventive policies.
Collapse
Affiliation(s)
- Michael A Gaytant
- Department of Obstetrics and Gynaecology, Radboud University Nijmegen Medical Centre, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Yue Y, Zhou SS, Barry PA. Antibody responses to rhesus cytomegalovirus glycoprotein B in naturally infected rhesus macaques. J Gen Virol 2004; 84:3371-3379. [PMID: 14645918 DOI: 10.1099/vir.0.19508-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Rhesus cytomegalovirus (RhCMV) exhibits strong parallels with human CMV (HCMV) in terms of nucleic and amino acid identities, natural history, and mechanisms of persistence and pathogenesis in its natural host, rhesus macaques (Macaca mulatta). To determine whether this non-human primate model would be useful to assess vaccine strategies for HCMV, host immune responses to RhCMV glycoprotein B (gB) were evaluated in RhCMV-infected monkeys. Total protein extracts were prepared from cells transiently transfected with an expression plasmid for either the full-length gB or a derivative (gBDelta, 1-680 aa) lacking both the transmembrane domain and cytoplasmic tail. Western blot analysis showed identical reactivity of macaque sera with full-length gB and its derivative gBDelta, indicating that the immunodominant epitopes of gB are contained in the extracellular portion of the protein. Using gBDelta extract as a solid phase, a sensitive and specific ELISA was established to characterize gB antibody responses in monkeys acutely and chronically infected with RhCMV. During primary infection (seroconversion), gB-specific antibodies developed concurrently and in parallel with total RhCMV-specific antibodies. However, during chronic infection gB-specific antibody responses were variable. A strong correlation was observed between neutralizing and gB-specific antibody levels in RhCMV-seropositive monkeys. Taken together, the results of this study indicate that, similar to host humoral responses to HCMV gB, anti-gB antibodies are an integral part of humoral immunity to RhCMV infection and probably play an important protective role in limiting the extent of RhCMV infection. Thus, the rhesus macaque model of HCMV infection is relevant for testing gB-based immune therapies.
Collapse
Affiliation(s)
- Yujuan Yue
- Center for Comparative Medicine, University of California, Davis, County Road 98 & Hutchison Drive, Davis, CA 95616, USA
| | - Shan Shan Zhou
- Center for Comparative Medicine, University of California, Davis, County Road 98 & Hutchison Drive, Davis, CA 95616, USA
| | - Peter A Barry
- Center for Comparative Medicine, University of California, Davis, County Road 98 & Hutchison Drive, Davis, CA 95616, USA
| |
Collapse
|
17
|
Gaytant MA, Steegers EAP, Semmekrot BA, Merkus HMMW, Galama JMD. Congenital cytomegalovirus infection: review of the epidemiology and outcome. Obstet Gynecol Surv 2002; 57:245-56. [PMID: 11961482 DOI: 10.1097/00006254-200204000-00024] [Citation(s) in RCA: 170] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
UNLABELLED Cytomegalovirus (CMV) is one of the most common viral causes of congenital infection. A future decision to lower its incidence by vaccination will depend on epidemiological conditions within a country and on the safety of the vaccine to be used, because a life vaccine may cause latency and subsequent reactivation that still may harm the fetus. The aim was to review the epidemiological studies published so far, with respect to factors that affect the incidence of congenital CMV infection, and factors that may influence its outcome, such as preexisting maternal immunity. The study included the data of 19 studies that were retrieved from a MEDLINE search during the period 1977 to 1997. The incidence of congenital CMV infection varied between 0.15% and 2.0% and seemed to correlate with the level of preexisting immunity in the population. Although preexisting maternal immunity was reported to strongly reduce transmission, the severity of congenital CMV infection (symptoms at birth and or sequelae later in life) was not significantly greater after virus transmission due to a primary infection of the mother as compared with recurrence or reinfection. The data indicate that preexisting immunity of the mother does not significantly mitigate the outcome of congenital infection. Moreover, life vaccines may bear a serious risk when transmittable to the fetus. TARGET AUDIENCE Obstetricians and Gynecologists, Family Physicians. LEARNING OBJECTIVES After completion of this article, the reader will be able to describe the natural course of a CMV infection, to list the potential sequelae of a congenital CMV infection, to outline potential strategies to prevent transmission of CMV, and to summarize the diagnostic work up of a patient with a potential CMV infection.
Collapse
Affiliation(s)
- Michael A Gaytant
- Department of Internal Medicine, Slingerland Hospital Doetinchem, The Netherlands
| | | | | | | | | |
Collapse
|
18
|
Muñoz I, Gutiérrez A, Gimeno C, Farga A, Alberola J, Solano C, Prósper F, García-Conde J, Navarro D. Lack of association between the kinetics of human cytomegalovirus (HCMV) glycoprotein B (gB)-specific and neutralizing serum antibodies and development or recovery from HCMV active infection in patients undergoing allogeneic stem cell transplant. J Med Virol 2001. [DOI: 10.1002/jmv.2004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
19
|
Endrész V, Burián K, Berencsi K, Gyulai Z, Kari L, Horton H, Virok D, Méric C, Plotkin SA, Gönczöl E. Optimization of DNA immunization against human cytomegalovirus. Vaccine 2001; 19:3972-80. [PMID: 11427273 DOI: 10.1016/s0264-410x(01)00116-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The immune responses of mice injected with plasmids VR-gB and VR-gB Delta tm expressing the full-length membrane-anchored, or secreted forms of human cytomegalovirus (HCMV)-glycoprotein B (gB), respectively, and VR-pp65 expressing the HCMV-phosphoprotein 65 (pp65) were analyzed. Pretreatment of mice with the local anesthetic bupivacaine did not enhance antibody production, and IFN-alpha co-expressed with the immunizing plasmids induced a moderate increase in the antibody response. However, antibody response was higher in mice inoculated at three sites in the musculus quadriceps than in mice inoculated at one site with the same dose and in the same muscle. pVR-gB Delta tm induced significantly higher antibody titers than the construct expressing the membrane-anchored form of gB, and priming with pVR-gB Delta tm followed by boosting with the gB subunit resulted in high-titer antibody responses. Immunization with VR-pp65 induced dose-dependent CTL responses in about 50% of the mice at a dose of 50 microg. Co-expression of IFN-alpha did not affect the number of responding mice. These findings might be important for optimization of humoral and cellular immune responses to HCMV after DNA vaccination.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/biosynthesis
- Antigens, Viral/genetics
- Bupivacaine/administration & dosage
- Cytomegalovirus/genetics
- Cytomegalovirus/immunology
- Cytomegalovirus Infections/immunology
- Cytomegalovirus Infections/prevention & control
- Female
- Humans
- Immunization, Secondary
- Immunoglobulin G/biosynthesis
- Injections, Intramuscular
- Interferon-alpha/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Inbred CBA
- Phosphoproteins/genetics
- Phosphoproteins/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/pharmacology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
- Viral Vaccines/pharmacology
Collapse
Affiliation(s)
- V Endrész
- Department of Medical Microbiology, University of Szeged, Szeged, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Gonczol E, Plotkin S. Development of a cytomegalovirus vaccine: lessons from recent clinical trials. Expert Opin Biol Ther 2001; 1:401-12. [PMID: 11727514 DOI: 10.1517/14712598.1.3.401] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cytomegalovirus-caused diseases are preventable. We believe that both neutralising antibodies and cell-mediated immunity are necessary for prevention. Of the CMV proteins, gB and pp65 are the minimum requirements in a vaccine to induced neutralising antibodies and cytotoxic T-lymphocyte (CTL) responses. Immunisation with additional proteins, e.g., gH, gN for neutralising antibodies and IE1exon 4 and pp150 for CTL responses, would strengthen protective immune responses. Approaches to development of a safe and effective cytomegalovirus (CMV) vaccine for the prevention of CMV diseases include: a) a live attenuated vaccine (Towne strain); b) recombinant constructs of the attenuated Towne and the virulent Toledo CMV strains; c) subunit glycoprotein B (gB) adjuvanted with MF59 to induce neutralising antibodies; d) phosphoprotein 65 (pp65) peptide-based vaccines to induce (CTL) for use in therapeutic vaccination; e) canarypox-CMV recombinants, e.g., ALVAC-CMV(gB) and ALVAC-CMV (pp65) to induce neutralising antibodies and CTL responses, respectively; f) DNA plasmids containing the genes for gB and pp65; g) dense bodies containing the key antigens. The attenuated Towne strain, gB/MF59, ALVAC-CMV(gB) and ALVAC-CMV(pp65) approaches have already been tested in clinical trials. The Towne vaccine induced neutralising antibodies and cell-mediated immunity (including CTLs) mitigated CMV disease in seronegative renal transplant recipients and protected against a low-dose virulent CMV challenge in normal volunteers but did not prevent infection in mothers of children excreting CMV. Immunisation with gB/MF59 resulted in high levels of neutralising antibodies in seronegative subjects. ALVAC-CMV(gB) did not induce neutralising antibodies but primed the immune system to a Towne strain challenge, while ALVAC-CMV(pp65) induced long-lasting CTL responses in all originally seronegative volunteers, with CTL precursor frequency similar to naturally seropositive individuals. These results suggest that CMV diseases can be prevented or attenuated and that a vaccine combining ALVAC-CMV(pp65) with gB/MF59 may induce sufficient CTLs and neutralising antibodies to protect against CMV diseases. Meanwhile, other approaches such as DNA peptide and dense body vaccines, should enter Phase I trials. All candidate vaccines will have to demonstrate that immunogenicity provides protection. Combined vaccines containing canarypox (ALVAC) vectors to express CMV-pp65 to induce CTLs and of subunit gB, given together with an appropriate adjuvant to induce neutralising antibodies, should be tested in a target population for the prevention of CMV infection and disease.
Collapse
Affiliation(s)
- E Gonczol
- Wistar Institute/Albert Szent-Gyorgyi Medical University, Aventis Pasteur, Swiftwater, PA, USA
| | | |
Collapse
|
21
|
Alberola J, Tamarit A, Cardeñoso L, Estellés F, Igual R, Navarro D. Longitudinal analysis of human cytomegalovirus glycoprotein B (gB)-specific and neutralizing antibodies in AIDS patients either with or without cytomegalovirus end-organ disease. J Med Virol 2001; 64:35-41. [PMID: 11285566 DOI: 10.1002/jmv.1014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Serum neutralizing and glycoprotein B (gB)-specific antibody levels were monitored prospectively in AIDS patients who either did or did not develop human cytomegalovirus (HCMV) end-organ disease, to delineate further the role of antibodies in protecting against HCMV disease. Antibody levels declined substantially (at least 4-fold) only in patients who developed HCMV disease; this decline in turn occurred concurrently with antigenemia. Nevertheless, AIDS patients who remained free of HCMV disease and did not become antigenemic during the follow-up period maintained stable levels of serum antibodies, with only minor fluctuations. The impact of HAART on the levels of functional anti-HCMV antibodies was investigated in a number of AIDS patients. Serum levels and kinetics of gB and neutralizing antibodies did not differ significantly between patients who responded biologically and virologically to therapy and those who failed to respond. In addition, CD4 + cell counts and HIV viral RNA levels did not correlate with anti-HCMV antibody titers.
Collapse
Affiliation(s)
- J Alberola
- Department of Microbiology, School of Medicine, University of Valencia, Valencia, Spain
| | | | | | | | | | | |
Collapse
|
22
|
Marshall GS, Li M, Stout GG, Louthan MV, Duliège AM, Burke RL, Hunt LA. Antibodies to the major linear neutralizing domains of cytomegalovirus glycoprotein B among natural seropositives and CMV subunit vaccine recipients. Viral Immunol 2001; 13:329-41. [PMID: 11016597 DOI: 10.1089/08828240050144653] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The gB protein (gpUL55) of human cytomegalovirus (CMV) contains C-terminal (AD-1) and N-terminal (AD-2) linear immunodominant neutralizing domains. To measure antibodies to these epitopes, a modified protein (delta-gB) lacking heavily glycosylated intervening regions, the transmembrane domain, and the cytoplasmic domain, was expressed in recombinant baculovirus-infected cells. Eighty-six percent of 600 naturally CMV-seropositive individuals and 93% of 121 gB vaccine recipients had antibodies to delta-gB as detected by enzyme-linked immunosorbent assay (ELISA). The antibody level in vaccinees (median optical density [OD] = 1.73) exceeded that in natural seropositives (median OD = 0.94; p < .0001). Eleven percent of 95 natural seropositives and 7% of 120 gB vaccinees lacked A-gB antibodies but had neutralizing activity. Among subjects with delta-gB antibody, there were weak correlations between antibody level and neutralizing titer. These data suggest that antibodies to linear neutralizing gB domains are highly prevalent in naturally-infected individuals and regularly develop in gB vaccinees. However, for some individuals, discontinuous and/or linear epitopes not represented on delta-gB may be more important in the generation of neutralizing responses.
Collapse
Affiliation(s)
- G S Marshall
- Department of Pediatrics, University of Louisville School of Medicine, Kentucky 40202, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Like varicella zoster virus (VZV), human cytomegalovirus (HCMV) causes disease after both primary and recurrent infections. The former is more serious, particularly in pregnant women, who may transmit the virus to their offspring, with a high risk of mental retardation and deafness. Various experimental vaccines are in development, ranging from live, attenuated HCMV, subunit envelope glycoprotein, poxvirus vectors with CMV genes inserted, and plasmid DANN.
Collapse
Affiliation(s)
- S A Plotkin
- University of Pennsylvania Wistar Institute, Doylestown, USA
| |
Collapse
|
24
|
Halwachs-Baumann G, Wilders-Truschnig M, Enzinger G, Eibl M, Linkesch W, Dornbusch HJ, Santner BI, Marth E, Kessler HH. Cytomegalovirus diagnosis in renal and bone marrow transplant recipients: the impact of molecular assays. J Clin Virol 2001; 20:49-57. [PMID: 11163583 DOI: 10.1016/s1386-6532(00)00155-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) infections are a major threat in transplant recipients. In recent years, new assays for routine CMV diagnosis, based on molecular techniques, have become available. OBJECTIVE The impact of molecular assays for CMV diagnosis in transplant recipient was evaluated. STUDY DESIGN A total of 51 transplant recipients were screened for CMV infection. Serological (AxSYM CMV IgG and recombinant CMV IgM assays), antigenemia, CMV DNA (qualitative in house PCR and the quantitative COBAS AMPLICOR CMV MONITOR Test), and CMV mRNA (NucliSens CMV pp67 Test) tests were compared. RESULTS In 11/20 bone marrow transplant (BMT) recipients and 10/31 renal transplant (RTX) recipients there was no evidence of active CMV infection. Ten RTX recipients and one BMT recipient were antigenemia positive, 21 RTX and seven BMT recipients were PCR positive (qualitative CMV PCR). There were more BMT recipients CMV DNA positive in serum (7/21) than antigenemia positive (1/21). CMV mRNA was found positive in two BMT recipients (one case with no other evidence of CMV infection, the other one CMV DNA positive and antigenemia negative). The only antigenemia positive BMT recipient was found negative for CMV mRNA, but positive in all other tests. Eight RTX recipients were found positive for CMV mRNA. Six of them were also antigenemia positive and five of those were also found positive for CMV IgM. One CMV mRNA positive RTX recipient was CMV IgM positive but antigenemia negative and the other one CMV mRNA positive RTX recipient was found negative in all other tests. Two antigenemia positive RTX recipients were found negative for mRNA and CMV IgM. CONCLUSION Antigenemia was found to be a good screening test for CMV infection in RTX recipients. In BMT recipients, tests based on molecular techniques appeared to be superior compared to antigenemia.
Collapse
Affiliation(s)
- G Halwachs-Baumann
- Department of Laboratory Medicine, Karl-Franzens-University, Graz, Auenbruggerplatz 15, A-8036 Graz, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Pepperl S, Münster J, Mach M, Harris JR, Plachter B. Dense bodies of human cytomegalovirus induce both humoral and cellular immune responses in the absence of viral gene expression. J Virol 2000; 74:6132-46. [PMID: 10846097 PMCID: PMC112112 DOI: 10.1128/jvi.74.13.6132-6146.2000] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2000] [Accepted: 04/10/2000] [Indexed: 11/20/2022] Open
Abstract
Infection of fibroblast cell cultures with human cytomegalovirus (HCMV) leads to the production of significant amounts of defective enveloped particles, termed dense bodies (DB). These noninfectious structures contain major antigenic determinants which are responsible for induction of both the humoral and the cellular immune response against HCMV. We tested the hypothesis that, by virtue of their unique antigenic and structural properties, DB could induce a significant immune response in the absence of infectious virus. Mice were immunized with gradient-purified DB, which were either left untreated or subjected to sequential rounds of sonication and freeze-thawing to prevent cellular entry. Titers of neutralizing antibodies induced by DB were in a range comparable to levels present in convalescent human sera. The virus-neutralizing antibody response was surprisingly durable, with neutralizing antibodies detected 12 months following primary immunization. The HCMV-specific major histocompatibility complex class I-restricted cytolytic T-cell (CTL) response was assayed using mice transgenic for the human HLA-A2 molecule. Immunization with DB led to high levels of HCMV-specific CTL in the absence of de novo viral protein synthesis. Maximal total cytolytic activity in mice immunized with DB was nearly as efficient as the cytolytic activity induced by a standard immunization with murine cytomegalovirus. Furthermore, DB induced a typical T-helper 1 (Th1)-dominated immune response in mice, as determined by cytokine and immunoglobulin G isotype analysis. Induction of humoral and cellular immune responses was achieved without the concomitant use of adjuvant. We thus propose that DB can serve as a basis for the future development of a recombinant nonreplicating vaccine against HCMV. Finally, such particles could be engineered for efficient delivery of antigens from other pathogens to the immune system.
Collapse
Affiliation(s)
- S Pepperl
- Institute for Virology, University of Mainz, Germany
| | | | | | | | | |
Collapse
|
26
|
|
27
|
Alberola J, Tamarit A, Igual R, Navarro D. Early neutralizing and glycoprotein B (gB)-specific antibody responses to human cytomegalovirus (HCMV) in immunocompetent individuals with distinct clinical presentations of primary HCMV infection. J Clin Virol 2000; 16:113-22. [PMID: 10720815 DOI: 10.1016/s1386-6532(00)00061-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Antibodies with functional anti-Human Cytomegalovirus (HCMV) activity are likely to be involved in preventing virus dissemination and thus may contribute to minimize the clinical manifestations of infection. OBJECTIVES To investigate the role of humoral immunity in modulating the clinical expression of primary Human Cytomegalovirus (HCMV) infection in immunocompetent persons. STUDY DESIGN Neutralizing (NA) and glycoprotein B (gB)-specific antibodies were quantitated in acute-phase and late-convalescence phase sera from 19 individuals who developed either HCMV mononucleosis (12) or oligosymptomatic hepatitis (seven). RESULTS The levels of NA in sera drawn early after infection were significantly lower in the former patients than in the latter (P=0. 032). This difference was not related to either the total serum IgG levels and anti-HCMV IgGs avidity or to the presence of higher viral loads in blood, as assessed by detecting serum HCMV DNA by PCR, in patients experiencing mononucleosis. Increased NA titers were seen in all available late-convalescence sera. In these sera, median NA levels were not significantly different among the study groups. Antibodies to HCMV gB of both IgG and IgM classes were detected in all acute-phase sera analyzed. Median anti-gB IgG and IgM titers did not differ significantly between study groups. Likewise, the IgG subclass reactivity pattern against gB was found to be similar for both groups. CONCLUSIONS The data revealed that an intense and early antibody response to gB developed in patients undergoing primary HCMV infection irrespective of the clinical manifestation of the disease. In contrast, a deficient NA response was observed in patients with HCMV mononucleosis versus that observed in patients displaying a milder form of disease-suggesting that the strength of NA response to HCMV generated early after infection might determine the severity of primary HCMV infection.
Collapse
MESH Headings
- Adolescent
- Adult
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibody Affinity
- Child
- Child, Preschool
- Cytomegalovirus Infections/blood
- Cytomegalovirus Infections/immunology
- Cytomegalovirus Infections/virology
- DNA, Viral/blood
- Female
- Hepatitis, Viral, Human/blood
- Hepatitis, Viral, Human/immunology
- Hepatitis, Viral, Human/virology
- Humans
- Immunocompetence/immunology
- Immunoglobulin G/immunology
- Infectious Mononucleosis/blood
- Infectious Mononucleosis/immunology
- Infectious Mononucleosis/virology
- Male
- Neutralization Tests
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- J Alberola
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 17, Valencia, Spain
| | | | | | | |
Collapse
|
28
|
|
29
|
Hwang ES, Kwon KB, Park JW, Kim DJ, Park CG, Cha CY. Induction of neutralizing antibody against human cytomegalovirus (HCMV) with DNA-mediated immunization of HCMV glycoprotein B in mice. Microbiol Immunol 1999; 43:307-10. [PMID: 10338203 DOI: 10.1111/j.1348-0421.1999.tb02409.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Immunization was accomplished by inoculating pcGB containing human cytomegalovirus (HCMV) glycoprotein B (gB) gene into BALB/c mice intramuscularly. IgM antibody was detected in all the immunized group. IgG antibody was also found in all the tested mice with a mean peak antibody titer of 1:262 in three-times immunized groups. IgG antibody appeared at 2 weeks postinoculation, raised peak levels at 7 weeks postinoculation and persisted over 6 months. Neutralizing antibody was developed, and the percent reduction of input infectivity in 1:100 diluted sera was 74.5 % in three-times immunized groups. This study suggested that DNA vaccine using the gene encoding HCMV gB is a candidate method for developing immunity to HCMV.
Collapse
Affiliation(s)
- E S Hwang
- Department of Microbiology, Seoul National University College of Medicine, Republic of Korea
| | | | | | | | | | | |
Collapse
|
30
|
Affiliation(s)
- S A Plotkin
- Pasteur Mérieux Connaught, Doylestown, PA 18901, USA.
| |
Collapse
|
31
|
Endresz V, Kari L, Berencsi K, Kari C, Gyulai Z, Jeney C, Pincus S, Rodeck U, Méric C, Plotkin SA, Gönczöl E. Induction of human cytomegalovirus (HCMV)-glycoprotein B (gB)-specific neutralizing antibody and phosphoprotein 65 (pp65)-specific cytotoxic T lymphocyte responses by naked DNA immunization. Vaccine 1999; 17:50-8. [PMID: 10078607 DOI: 10.1016/s0264-410x(98)00145-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Plasmids expressing the human cytomegalovirus (HCMV) glycoprotein B (gB) (UL55) or phosphoprotein 65 (pp65) (UL83) were constructed and evaluated for their ability to induce immune responses in mice. The full-length gB as well as a truncated form expressing amino acids 1-680 of gB, and lacking the fragment encoding amino acids 681 907 including the transmembrane domain of gB (gB680) were evaluated. Immunization of mice with plasmids coding for gB or gB680 induced ELISA and neutralizing antibodies, with the highest titres in mice immunized with the gB680 plasmid. Mice immunized with the gB plasmid predominantly produced IgG2a gB-specific antibody, while the gB680 plasmid raised mostly IgG1 anti-gB antibody. Mice immunized with the pp65 plasmid developed pp65-specific cytotoxic T lymphocytes (CTL) and ELISA antibodies. Immunization with a mixture of both gB and pp65 plasmids raised antibodies to both proteins and pp65-specific CTL, indicating a lack of interference between these two plasmids. These results suggest that DNA immunization is a useful approach for vaccination against HCMV disease.
Collapse
Affiliation(s)
- V Endresz
- The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Alberola J, Domínguez V, Cardeñoso L, López-Aldeguer J, Blanes M, Estellés F, Ricart C, Pastor A, Igual R, Navarro D. Antibody response to human cytomegalovirus (HCMV) glycoprotein B (gB) in AIDS patients with HCMV end-organ disease. J Med Virol 1998; 55:272-80. [PMID: 9661835 DOI: 10.1002/(sici)1096-9071(199808)55:4<272::aid-jmv4>3.0.co;2-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Human cytomegalovirus (HCMV)-specific antibody responses in HIV-1 infected individuals either with or without HCMV end-organ disease were examined to determine the whether development of HCMV disease was associated with a particular deficit in the antibody response. Antiwhole HCMV, anti-glycoprotein B (gB), and neutralizing antibody levels were higher in HIV-1 infected individuals than in healthy immunocompetent subjects, particularly in patients with AIDS either with or without HCMV-associated disease. Irrespective of location and spread of HCMV disease, patients who had received anti-HCMV therapy prior to sampling exhibited significantly higher anti-gB and neutralizing antibody titers than those who remained untreated. Likewise, patients with HCMV disease who were antigenemic or viremic had significantly lower anti-gB and neutralizing antibody titers than those who tested negative in either assay. Patients with untreated HCMV disease had significantly lower antibody titers than AIDS patients without disease. Analysis of the IgG subclass antibody responses to gB revealed no significant differences among HIV-1 infected individuals. These results suggest that levels of detectable anti-gB and HCMV neutralizing antibodies are inversely related to systemic viral load. Thus, antibodies with such specificities may be relevant in preventing the establishment of HCMV-associated disease or in modulating its progression.
Collapse
Affiliation(s)
- J Alberola
- Department of Microbiology, School of Medicine, University of Valencia, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Navarro D, Lennette E, Tugizov S, Pereira L. Humoral immune response to functional regions of human cytomegalovirus glycoprotein B. J Med Virol 1997. [DOI: 10.1002/(sici)1096-9071(199708)52:4<451::aid-jmv18>3.0.co;2-j] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
34
|
Khattab BA, Lindenmaier W, Frank R, Link H. Three T-cell epitopes within the C-terminal 265 amino acids of the matrix protein pp65 of human cytomegalovirus recognized by human lymphocytes. J Med Virol 1997; 52:68-76. [PMID: 9131460 DOI: 10.1002/(sici)1096-9071(199705)52:1<68::aid-jmv11>3.0.co;2-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although a T-cell response in human cytomegalovirus (HCMV)-immune individuals exists against the most abundantly expressed protein pp65 of the virus matrix, less is known about the determinants that evoke this response. The aim of the study was to identify regions within HCMV pp65 (ppUL83) that contain sequences for the cellular immune response by the use of three recombinant overlapping beta-galactosidase pp65 fusion proteins (C74, C35, and C47), covering the C-terminal 265 amino acids of the entire pp65 sequence. Two T-cell epitope determinants were recognized by human lymphocytes of healthy, HCMV-seropositive, human leukocyte antigen (HLA)-typed individuals. One T-cell determinant (amino acids [aa] 303-388) was localized in the mid-region of the entire pp65 sequence and a second T-cell determinant (aa 477-561) within the C-terminal region. By fine mapping with synthetic hexadecamer peptides three T-cell epitopes were identified within these two regions: P10-I (aa 361-376) in the mid-region, P3-II (aa 485-499), and P6-II (aa 509-524) in the C-terminal region. Inhibition studies with monoclonal antibodies to HLA class I or class II revealed a class II restricted response to peptides P10-I or P6-II, respectively. P10-I responders shared the HLA-DR11 allele and P6-II responders the -DR3 allele. Therefore, these T-cell epitopes of HCMV pp65 might be presented in association with particular HLA class II alleles.
Collapse
Affiliation(s)
- B A Khattab
- Department of Hematology and Oncology, Medical School, Hannover, Germany
| | | | | | | |
Collapse
|
35
|
Berencsi K, Gönczöl E, Endresz V, Kough J, Takeda S, Gyulay Z, Plotkin SA, Rando RF. The N-terminal 303 amino acids of the human cytomegalovirus envelope glycoprotein B (UL55) and the exon 4 region of the major immediate early protein 1 (UL123) induce a cytotoxic T-cell response. Vaccine 1996; 14:369-74. [PMID: 8735546 DOI: 10.1016/0264-410x(95)00227-r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We reported earlier that an adenovirus (Ad) recombinant expressing the full-length human cytomegalovirus (HCMV) glycoprotein B (gB) gene induces gB-specific cytotoxic T lymphocyte (CTL) responses in CBA (H-2k) mice (Berencsi et al., J. Gen. Virol. 74, 257-2512, 1993). Here we show that mice immunized with Ad recombinant viruses expressing truncated forms of the gB gene containing the first 700 (Ad-700), 465 (Ad-465) or 303 (Ad-303) amino acids of gB or an Ad construct containing exon 4 (E4) of the HCMV immediate early 1 (IEI) gene (Ad-IEI (E4)) demonstrate HCMV-specific CTL responses. These data suggest the importance of the first 303 amino acids of the gB polypeptide and the IEI E4 product in designing a vaccine to induce anti-HCMV CTL responses.
Collapse
Affiliation(s)
- K Berencsi
- Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Reuman PD, Rathore MH, Ayoub EM. Developments in childhood immunization. CURRENT PROBLEMS IN PEDIATRICS 1996; 26:107-37. [PMID: 8790972 DOI: 10.1016/s0045-9380(96)80026-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- P D Reuman
- Department of Pediatrics, School of Medicine, University of Florida, Gainesville, USA
| | | | | |
Collapse
|
37
|
Prieur E, Betbeder D, Niedergang F, Major M, Alcover A, Davignon JL, Davrinche C. Combination of human cytomegalovirus recombinant immediate-early protein (IE1) with 80 nm cationic biovectors: protection from proteolysis and potentiation of presentation to CD4+ T-cell clones in vitro. Vaccine 1996; 14:511-20. [PMID: 8782349 DOI: 10.1016/0264-410x(95)00233-q] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We have shown in a previous study that the proliferative CD4+ T-cell response to the regulatory immediate-early protein IE1 was a major component of the overall anti viral response in human cytomegalovirus (HCMV) seropositive blood donors. This viral antigen may be valuable in subunit vaccine design, since anti IE1 CD4+ T cells might provide help for production of antibodies and cytotoxic T lymphocytes (CTL) responses, and could take part in the control of viral infection. Preliminary to the elaboration of future vaccine formulations, we developed immunogenic complexes resulting from the combination of a purified recombinant protein derived from the fusion of Escherichia coli glutathione-S-transferase (GST) and a large C-terminal fragment (e4) of IE1, with new 80 nm cationic synthetic particles called Biovectors. We have shown that the antigen GST-e4 was stably complexed to vectors and that, contrary to the soluble form, it was protected from proteolysis in cell culture medium. By confocal microscopy we observed that the synthetic vectors were internalized by lymphoblastoid B cells, providing a significant enhancement of antigen delivery in antigen presenting cells (APC). Indeed, we demonstrated that the previous combination of antigen with particles, significantly enhanced the proliferation of specific CD4+ T-cell clones directed against IE1 in vitro, when either HLA-matched isolated peripheral blood mononuclear cells or EBV transformed B cell lines were used as APC. The relevance of these observations to the use of these new vectors for vaccine design against HCMV is discussed.
Collapse
Affiliation(s)
- E Prieur
- Inserm U395, IFR 30, UPS, CNRS, CHU, Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
38
|
Bron D, Snoeck R, Lagneaux L. Section Review: Biologicals & Immunologicals: New insights into the pathogenesis and treatment of cytomegalovirus. Expert Opin Investig Drugs 1996. [DOI: 10.1517/13543784.5.3.337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
39
|
|
40
|
|
41
|
Greis KD, Gibson W, Hart GW. Site-specific glycosylation of the human cytomegalovirus tegument basic phosphoprotein (UL32) at serine 921 and serine 952. J Virol 1994; 68:8339-49. [PMID: 7966627 PMCID: PMC237303 DOI: 10.1128/jvi.68.12.8339-8349.1994] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The virion basic phosphoprotein (BPP), UL32, of the human cytomegalovirus (HCMV) is a 149-kDa tegument protein that represents about 15% of the virion protein mass and is modified by O-linked N-acetylglucosamine (O-GlcNAc). O-GlcNAc has been postulated to mediate subunit-subunit interaction in many different types of intracellular protein complexes, while BPP may play a role in viral assembly and/or envelopment. This report describes the identification of the major O-GlcNAc attachment sites on the HCMV (AD169) BPP. Because the amount of BPP isolated from infectious virions was insufficient to determine the site(s) of glycosylation, the full-length protein has been characterized following overexpression in recombinant baculovirus-infected insect cells. The recombinant protein (rBPP) was electrophoretically (by sodium dodecyl sulfate-polyacrylamide gel electrophoresis) and immunologically (by Western immunoassaying) indistinguishable from the BPP isolated from HCMV virions. In addition, the rBPP was modified by O-GlcNAc, and a comparison of the tryptic glycopeptides from the rBPP and native virion BPP indicated that their O-GlcNAc sites are the same. Furthermore, the major sites of O-GlcNAc attachment to the rBPP were mapped on high-performance liquid chromatography-purified glycopeptides by gas phase microsequencing, manual Edman degradation, and electrospray-mass spectrometry. The results demonstrate that the major sites of O-GlcNAc attachment to the BPP are Ser-921 and Ser-952. Identification of these sites will now enable mutagenesis studies to determine the influence of O-GlcNAc on the intracellular location, protein-protein interaction, and biological function of BPP. Finally, the fidelity of the addition of O-GlcNAc to rBPP in insect cells compared with native virion BPP is documented to demonstrate the possible general applicability of the baculovirus expression system to study O-GlcNAc on other low-abundance proteins.
Collapse
Affiliation(s)
- K D Greis
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, School of Medicine and Dentistry 35294
| | | | | |
Collapse
|
42
|
Marshall GS, Stout GG, Knights ME, Rabalais GP, Ashley R, Miller H, Rossier E. Ontogeny of glycoprotein gB-specific antibody and neutralizing activity during natural cytomegalovirus infection. J Med Virol 1994; 43:77-83. [PMID: 8083653 DOI: 10.1002/jmv.1890430115] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The envelope glycoprotein gB (gpUL55) is a candidate for inclusion in subunit cytomegalovirus (CMV) vaccines, although data on gB antibody responses after natural infection are limited. [35S]-labeled gB was partially purified from cells infected with an adenovirus recombinant expressing gB and used in radioimmunoprecipitation assays to characterize responses in solid organ transplant recipients with primary (n = 11) or secondary (n = 8) CMV infection. Seropositive transplant patients without evidence of infection (n = 5) and healthy seroconverters (n = 7) were also studied. gB antibody developed concurrently with CMV-specific IgG, IgM, and neutralizing activity in transplant patients with primary infection. Sustained boosts in gB antibody were seen in patients with secondary infection, and healthy seroconverters developed early gB responses. These data imply that gB antibody is an integral part of the humoral response to CMV infection, and, in view of experimental data regarding immunogenicity, support a role for gB in subunit vaccines.
Collapse
Affiliation(s)
- G S Marshall
- Division of Pediatric Infectious Diseases, University of Louisville School of Medicine, KY 40292
| | | | | | | | | | | | | |
Collapse
|
43
|
Bonci A, Bracci L, Caudai C, Lozzi L, Moschettini D, Niccolai N, Scarselli M, Valensin PE, Neri P. Characterization of immunoreactive octapeptides of human-cytomegalovirus gp58. EUROPEAN JOURNAL OF BIOCHEMISTRY 1993; 215:383-7. [PMID: 7688304 DOI: 10.1111/j.1432-1033.1993.tb18044.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We have mapped continuous epitopes, for positions 591-673 of the human cytomegalovirus 58-kDa glycoprotein using overlapping synthetic peptides and human sera. This region contains a fragment previously described as including the dominant site for induction of human-cytomegalovirus antibodies. Since the selected sequence is highly conserved among herpes viruses, we have considered the possible presence of antigenic cross-reactivity, particularly with the Epstein-Barr virus. Several peptides in the studied region were antigenic and two main continuous epitopes have been identified. Serological cross-reactions observed with Epstein-Barr virus are discussed, focusing on the possible implications of structural features and sequence similarity between human-cytomegalovirus and Epstein-Barr-virus glycoproteins.
Collapse
Affiliation(s)
- A Bonci
- Dipartimento di Biologia Molecolare, Università di Siena, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lolli F, Sundqvist VA, Castagna A, Ljungman P, Linde A, Andersson G, Olsson T, Wahren B. T and B cell responses to cytomegalovirus antigens in healthy blood donors and bone marrow transplant recipients. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 1993; 7:55-62. [PMID: 8395936 DOI: 10.1111/j.1574-695x.1993.tb00381.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We measured the production of interferon-gamma (IFN-gamma) from single T cells and the T cell proliferative response to different cytomegalovirus (CMV) antigens in healthy blood donors and bone marrow transplant recipients. The antigens consisted of a CMV nuclear antigen (CMV na) containing the pp65-kDa matrix protein and the immediate early antigens but lacking CMV glycoproteins, and an antigen comprising native CMV glycoproteins (CMV gp). We also measured the IgG antibodies to CMV na and CMV gp. The T cells reacted to CMV na in CMV seropositive blood donors both with the production of IFN-gamma and with proliferation, while bone marrow transplant recipients had a deficient T cell response. After stimulation with CMV gp, no T cell response could be observed in CMV seropositive subjects. IgG antibodies to CMV na coexisted in plasma with similar levels of antibodies to CMV gp.
Collapse
Affiliation(s)
- F Lolli
- Department of Virology, National Bacteriological Laboratory, Solna, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Gupta R, Gonczol E, Manning ML, Starr S, Johnson B, Murphy GF, Plotkin SA. Delayed type hypersensitivity to human cytomegalovirus. J Med Virol 1993; 39:109-17. [PMID: 8387567 DOI: 10.1002/jmv.1890390206] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
A skin test for immunity to human cytomegalovirus (HCMV) is described in which skin induration is measured after intradermal injection of antigen derived from heat-inactivated Towne strain HCMV or of envelope prepared from the virus. Randomly selected healthy young adult males and females were prescreened for evidence of past infection with HCMV using serologic tests. Each individual was inoculated with heat inactivated whole virion HCMV antigen prepared from serum-free supernatants of HCMV-infected MRC-5 cells, non-infected MRC-5 cell lysates, and Candida extract. HCMV seropositive individuals developed positive skin reactions to both the Candida extract and the HCMV test antigen. No response was observed at the MRC-5 cell lysate inoculation site. The envelope antigen also elicited a response in seropositive individuals. Seronegative individuals who were negative to the HCMV intradermal antigen at the start of the study developed a positive response 1 week after subcutaneous immunization with live attenuated Towne strain HCMV. This response also correlated with the onset of in vitro proliferation responses to HCMV antigens by peripheral blood lymphocytes obtained from the immunized individuals. Furthermore, skin test and lymphocyte proliferation responses remained positive when tested up to 93 days post-immunization. In guinea pig experiments with HCMV, those animals immunized with purified HCMV or a virus envelope preparation developed strong skin reactions to intradermal injection of each of those antigens. In contrast, no reaction was observed in immune animals, either to viral capsid antigen or uninfected cell-lysate antigen, and no reactions were observed to any HCMV antigen in non-immune animals.
Collapse
Affiliation(s)
- R Gupta
- University of Pennsylvania, Philadelphia
| | | | | | | | | | | | | |
Collapse
|
46
|
Strategies for the treatment and prevention of cytomegalovirus infections. Int J Antimicrob Agents 1993; 3:187-204. [DOI: 10.1016/0924-8579(93)90012-t] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/1993] [Indexed: 11/19/2022]
|
47
|
Gönczöl E, deTaisne C, Hirka G, Berencsi K, Lin WC, Paoletti E, Plotkin S. High expression of human cytomegalovirus (HCMV)-gB protein in cells infected with a vaccinia-gB recombinant: the importance of the gB protein in HCMV immunity. Vaccine 1991; 9:631-7. [PMID: 1659051 DOI: 10.1016/0264-410x(91)90187-b] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The human cytomegalovirus (HCMV), Towne strain, glycoprotein B (gB) gene was cloned into a vaccinia vector (Copenhagen strain) under the control of the H6 early and late vaccinia promoters (Vac-gB recombinant). The gB protein was expressed in a high percentage of the Vac-gB-infected cells throughout the virus replication cycle. Cytosine-arabinoside (ara-C) did not influence the expression of the gB protein early after infection (5 h), but did inhibit it later in viral replication (7-29 h). The Vac-gB recombinant induced HCMV neutralizing antibodies in guinea-pigs. Cells infected with the Vac-gB recombinant absorbed 50-88% of neutralizing activity of human sera obtained from volunteers previously inoculated with the Towne or Toledo strains and from naturally seropositive individuals.
Collapse
Affiliation(s)
- E Gönczöl
- Wistar Institute of Anatomy and Biology, Philadelphia, PA 19104
| | | | | | | | | | | | | |
Collapse
|
48
|
Kongsuwan K, Prideaux CT, Johnson MA, Sheppard M, Fahey KJ. Nucleotide sequence of the gene encoding infectious laryngotracheitis virus glycoprotein B. Virology 1991; 184:404-10. [PMID: 1840710 DOI: 10.1016/0042-6822(91)90859-a] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The nucleotide sequence of the infectious laryngotracheitis virus (ILTV) gene encoding the 205K complex glycoprotein (gp205) was determined. The gene is contained within a 3-kb EcoRI restriction fragment mapping at approximately map coordinates 0.23 to 0.25 in the UL region of the ILTV genome and is transcribed from right to left. Nucleotide sequence analysis of the DNA fragment identified a single, long open reading frame capable of encoding 873 amino acids. The predicted precursor polypeptide derived from this open reading frame would have a calculated Mr of 98,895 Da and contains nine potential glycosylation sites. Hydropathic analysis indicates the presence of an amino terminal hydrophobic sequence and hydrophobic carboxyl terminal domain which may function as a signal peptide and a membrane anchor sequence, respectively. Comparison of the predicted ILTV gp205 protein sequence with those of other herpesviruses revealed a significant sequence similarity with gB-like glycoproteins. Extensive homology was observed throughout the molecule except for the amino and carboxyl termini. The high homology in predicted primary and secondary structures is consistent with the essential role of the gB family of proteins for viral infectivity and pathogenesis.
Collapse
Affiliation(s)
- K Kongsuwan
- CSIRO Division of Animal Health, Animal Research Laboratory, Parkville, VIC, Australia
| | | | | | | | | |
Collapse
|
49
|
Kameoka J, Endo K, Yoshinaga K, Shiobara S, Mori T, Matsuda T, Furukawa T. Failure to induce cytomegalovirus immunity with its envelope antigen injected without adjuvants. Vaccine 1991; 9:605-6. [PMID: 1663304 DOI: 10.1016/0264-410x(91)90259-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
50
|
Gönczöl E, Plotkin S. Progress in vaccine development for prevention of human cytomegalovirus infection. Curr Top Microbiol Immunol 1990; 154:255-74. [PMID: 2161323 DOI: 10.1007/978-3-642-74980-3_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- E Gönczöl
- Wistar Institute, Philadelphia, PA 19104
| | | |
Collapse
|