Herpes simplex virus

This article is about the virus. For information about the disease caused by the virus, see Herpes simplex.
Herpes simplex virus
TEM micrograph of a herpes simplex virus
Virus classification
Group: Group I (dsDNA)
Order: Herpesvirales
Family: Herpesviridae
Subfamily: Alphaherpesvirinae
Genus: Simplexvirus
Species
  • Herpes simplex virus 1 (HSV-1)
  • Herpes simplex virus 2 (HSV-2)

Herpes simplex virus 1 and 2 (HSV-1 and HSV-2), also known as human herpesvirus 1 and 2 (HHV-1 and HHV-2), are two members of the herpesvirus family, Herpesviridae, that infect humans.[1] Both HSV-1 (which produces most cold sores) and HSV-2 (which produces most genital herpes) are ubiquitous and contagious. They can be spread when an infected person is producing and shedding the virus.

In simple terms, herpes simplex 1 is most commonly known as a "cold sore," while herpes simplex 2 is the one known by the public as "herpes," or "genital herpes." Herpes simplex 1 is known to infect about 95% of the human populace, and is treated less seriously than herpes simplex 2, even though both are incurable.

Symptoms of herpes simplex virus infection include watery blisters in the skin or mucous membranes of the mouth, lips, nose or genitals.[1] Lesions heal with a scab characteristic of herpetic disease. Sometimes, the viruses cause very mild or atypical symptoms during outbreaks. However, they can also cause more troublesome forms of herpes simplex. As neurotropic and neuroinvasive viruses, HSV-1 and -2 persist in the body by becoming latent and hiding from the immune system in the cell bodies of neurons. After the initial or primary infection, some infected people experience sporadic episodes of viral reactivation or outbreaks. In an outbreak, the virus in a nerve cell becomes active and is transported via the neuron's axon to the skin, where virus replication and shedding occur and cause new sores.[2] It is one of the most common sexually transmitted infections.[3]

Transmission

Main article: Herpes simplex

HSV-1 and -2 are transmitted by contact with an infected area of the skin during reactivations of the virus. Herpes simplex virus (HSV)-2 is periodically shed in the human genital tract, most often asymptomatically, and most sexual transmissions occur during asymptomatic shedding.[4] Asymptomatic reactivation means that the virus causes atypical, subtle or hard to notice symptoms that are not identified as an active herpes infection. In one study, daily genital swab samples found HSV-2 at a median of 12–28% of days among those who have had an outbreak, and 10% of days among those suffering from asymptomatic infection, with many of these episodes occurring without visible outbreak ("subclinical shedding").[5]

In another study, 73 subjects were randomized to receive valaciclovir 1 g daily or placebo for 60 days each in a 2-way crossover design. A daily swab of the genital area was self-collected for HSV-2 detection by polymerase chain reaction, in order to compare the effect of valaciclovir 1 g once daily for 60 days versus placebo on asymptomatic viral shedding in immunocompetent, HSV-2 seropositive subjects without a history of symptomatic genital herpes infection. The study found that valaciclovir significantly reduced shedding during subclinical days compared to placebo, showing a 71% reduction. 84% of subjects had no shedding while receiving valaciclovir versus 54% of subjects on placebo. 88% of patients treated with valaciclovir had no recognized signs or symptoms versus 77% for placebo.[6]

For HSV-2, subclinical shedding may account for most of the transmission.[5] Studies on discordant partners (one infected with HSV-2, one not) show that the transmission rate is approximately 5 per 10,000 sexual contacts. (Effect of Condoms on Reducing the Transmission of Herpes Simplex Virus Type 2 From Men to Women. A Wald, AGM Langenberg, K Link, et al JAMA. 2001;285(24):3197) Atypical symptoms are often attributed to other causes such as a yeast infection.[7][8] HSV-1 is often acquired orally during childhood. It may also be sexually transmitted, including contact with saliva, such as kissing and mouth-to-genital contact (oral sex).[9] HSV-2 is primarily a sexually transmitted infection, but rates of HSV-1 genital infections are increasing.[7]

Both viruses may also be transmitted vertically during childbirth, although the real risk is very low.[10] The risk of infection is minimal if the mother has no symptoms or exposed blisters during delivery. The risk is considerable when the mother is infected with the virus for the first time during late pregnancy.[11]

Herpes simplex viruses can affect areas of skin exposed to contact with an infected person. An example of this is herpetic whitlow which is a herpes infection on the fingers. This was a common affliction of dental surgeons prior to the routine use of gloves when conducting treatment on patients.

Virology

Viral structure

3D reconstruction and animation of a tail-like assembly on HSV-1 capsid
3D reconstruction of the Herpes simplex virus type 1 (HSV-1) capsid

Animal herpes viruses all share some common properties. The structure of herpes viruses consists of a relatively large double-stranded, linear DNA genome encased within an icosahedral protein cage called the capsid, which is wrapped in a lipid bilayer called the envelope. The envelope is joined to the capsid by means of a tegument. This complete particle is known as the virion.[12] HSV-1 and HSV-2 each contain at least 74 genes (or open reading frames, ORFs) within their genomes,[13] although speculation over gene crowding allows as many as 84 unique protein coding genes by 94 putative ORFs.[14] These genes encode a variety of proteins involved in forming the capsid, tegument and envelope of the virus, as well as controlling the replication and infectivity of the virus. These genes and their functions are summarized in the table below.

The genomes of HSV-1 and HSV-2 are complex and contain two unique regions called the long unique region (UL) and the short unique region (US). Of the 74 known ORFs, UL contains 56 viral genes, whereas US contains only 12.[13] Transcription of HSV genes is catalyzed by RNA polymerase II of the infected host.[13] Immediate early genes, which encode proteins that regulate the expression of early and late viral genes, are the first to be expressed following infection. Early gene expression follows, to allow the synthesis of enzymes involved in DNA replication and the production of certain envelope glycoproteins. Expression of late genes occurs last; this group of genes predominantly encode proteins that form the virion particle.[13]

Five proteins from (UL) form the viral capsid; UL6, UL18, UL35, UL38 and the major capsid protein UL19.[12]

Cellular entry

A simplified diagram of HSV replication

Entry of HSV into a host cell involves several glycoproteins on the surface of the enveloped virus binding to their transmembrane receptors on the cell surface. Many of these receptors are then pulled inwards by the cell, which is thought to open a ring of three gHgL heterodimers stabilizing a compact conformation of the gB glycoprotein, so that it springs out and punctures the cell membrane.[15] The envelope covering the virus particle then fuses with the cell membrane, creating a pore through which the contents of the viral envelope enters the host cell.

The sequential stages of HSV entry are analogous to those of other viruses. At first, complementary receptors on the virus and the cell surface bring the viral and cell membranes into proximity. Interactions of these molecules then form a stable entry pore through which the viral envelope contents are introduced to the host cell. The virus can also be endocytosed after binding to the receptors, and the fusion could occur at the endosome. In electron micrographs the outer leaflets of the viral and cellular lipid bilayers have been seen merged together;[16] this hemifusion may be on the usual path to entry or it may usually be an arrested state more likely to be captured than a transient entry mechanism.

In the case of a herpes virus, initial interactions occur when two viral envelope glycoprotein called glycoprotein C (gC) and glycoprotein B (gB) bind to a cell surface particle called heparan sulfate. Next, the major receptor binding protein, glycoprotein D (gD), binds specifically to at least one of three known entry receptors.[17] These cell receptors include herpesvirus entry mediator (HVEM), nectin-1 and 3-O sulfated heparan sulfate. The nectin receptors usually produce cell-cell adhesion, so provide a strong point of attachment for the virus to the host cell.[15] These interactions bring the membrane surfaces into mutual proximity and allow for other glycoproteins embedded in the viral envelope to interact with other cell surface molecules. Once bound to the HVEM, gD changes its conformation and interacts with viral glycoproteins H (gH) and L (gL), which form a complex. The interaction of these membrane proteins may result in a hemifusion state. gB interaction with the gH/gL complex creates an entry pore for the viral capsid.[16] gB interacts with glycosaminoglycans on the surface of the host cell.

Genetic inoculation

After the viral capsid enters the cellular cytoplasm, it is transported to the cell nucleus. Once attached to the nucleus at a nuclear entry pore, the capsid ejects its DNA contents via the capsid portal. The capsid portal is formed by twelve copies of portal protein, UL6, arranged as a ring; the proteins contain a leucine zipper sequence of amino acids which allow them to adhere to each other.[18] Each icosahedral capsid contains a single portal, located in one vertex.[19][20] The DNA exits the capsid in a single linear segment.[21]

Immune evasion

HSV evades the immune system through interference with MHC class I antigen presentation on the cell surface, by blocking TAP or the transporter associated with antigen processing induced by the secretion of ICP-47 by HSV.[22] In the host cell, TAP transports digested viral antigen epitope peptides from the cytosol to the endoplasmic reticulum, allowing these epitopes to be combined with MHC class I molecules and presented on the surface of the cell. Viral epitope presentation with MHC class I is a requirement for activation of cytotoxic T-lymphocytes (CTLs), the major effectors of the cell-mediated immune response against virally-infected cells. ICP-47 prevents initiation of a CTL-response against HSV, allowing the virus to survive for a protracted period in the host.

Replication

Micrograph showing the viral cytopathic effect of HSV (multi-nucleation, ground glass chromatin).

Following infection of a cell, a cascade of herpes virus proteins, called immediate-early, early, and late, are produced. Research using flow cytometry on another member of the herpes virus family, Kaposi's sarcoma-associated herpesvirus, indicates the possibility of an additional lytic stage, delayed-late.[23] These stages of lytic infection, particularly late lytic, are distinct from the latency stage. In the case of HSV-1, no protein products are detected during latency, whereas they are detected during the lytic cycle.

The early proteins transcribed are used in the regulation of genetic replication of the virus. On entering the cell, an α-TIF protein joins the viral particle and aids in immediate-early transcription. The virion host shutoff protein (VHS or UL41) is very important to viral replication.[24] This enzyme shuts off protein synthesis in the host, degrades host mRNA, helps in viral replication, and regulates gene expression of viral proteins. The viral genome immediately travels to the nucleus but the VHS protein remains in the cytoplasm.[25][26]

The late proteins form the capsid and the receptors on the surface of the virus. Packaging of the viral particles — including the genome, core and the capsid - occurs in the nucleus of the cell. Here, concatemers of the viral genome are separated by cleavage and are placed into pre-formed capsids. HSV-1 undergoes a process of primary and secondary envelopment. The primary envelope is acquired by budding into the inner nuclear membrane of the cell. This then fuses with the outer nuclear membrane releasing a naked capsid into the cytoplasm. The virus acquires its final envelope by budding into cytoplasmic vesicles.[27]

Latent infection

HSVs may persist in a quiescent but persistent form known as latent infection, notably in neural ganglia.[1] HSV-1 tends to reside in the trigeminal ganglia, while HSV-2 tends to reside in the sacral ganglia, but these are tendencies only, not fixed behavior. During latent infection of a cell, HSVs express latency associated transcript (LAT) RNA. LAT regulates the host cell genome and interferes with natural cell death mechanisms. By maintaining the host cells, LAT expression preserves a reservoir of the virus, which allows subsequent, usually symptomatic, periodic recurrences or "outbreaks" characteristic of non-latency. Whether or not recurrences are symptomatic, viral shedding occurs to infect a new host. A protein found in neurons may bind to herpes virus DNA and regulate latency. Herpes virus DNA contains a gene for a protein called ICP4, which is an important transactivator of genes associated with lytic infection in HSV-1.[28] Elements surrounding the gene for ICP4 bind a protein known as the human neuronal protein Neuronal Restrictive Silencing Factor (NRSF) or human Repressor Element Silencing Transcription Factor (REST). When bound to the viral DNA elements, histone deacetylation occurs atop the ICP4 gene sequence to prevent initiation of transcription from this gene, thereby preventing transcription of other viral genes involved in the lytic cycle.[28][29] Another HSV protein reverses the inhibition of ICP4 protein synthesis. ICP0 dissociates NRSF from the ICP4 gene and thus prevents silencing of the viral DNA.[30]

The virus can be reactivated by illnesses such as colds and influenza, eczema, emotional and physical stress, gastric upset, fatigue or injury, by menstruation and possibly exposure to bright sunlight. Genital Herpes may be reactivated by friction.

HSV-1 viral genome

The open reading frames (ORFs) of HSV-1[13][31]
Gene Protein Function/description Gene Protein Function/description
UL1 Glycoprotein L Surface and membrane UL38 UL38; VP19C Capsid assembly and DNA maturation
UL2 UL2 Uracil-DNA glycosylase UL39 UL39; RR-1; ICP6 Ribonucleotide reductase (Large subunit)
UL3 UL3 unknown UL40 UL40; RR-2 Ribonucleotide reductase (Small subunit)
UL4 UL4 unknown UL41 UL41; VHS Tegument protein; Virion host shutoff[24]
UL5 UL5 DNA replication UL42 UL42 DNA polymerase processivity factor
UL6 Portal protein UL-6 Twelve of these proteins constitute the capsid portal ring through which DNA enters and exits the capsid.[18][19][20] UL43 UL43 Membrane protein
UL7 UL7 Virion maturation UL44 Glycoprotein C Surface and membrane
UL8 UL8 DNA virus helicase-primase complex-associated protein UL45 UL45 Membrane protein; C-type lectin[32]
UL9 UL9 Replication origin-binding protein UL46 VP11/12 Tegument proteins
UL10 Glycoprotein M Surface and membrane UL47 UL47; VP13/14 Tegument protein
UL11 UL11 virion exit and secondary envelopment UL48 VP16 (Alpha-TIF) Virion maturation; activate IE genes by interacting with the cellular transcription factors Oct-1 and HCF. Binds to the sequence 5'TAATGARAT3'.
UL12 UL12 Alkaline exonuclease UL49 UL49A Envelope protein
UL13 UL13 Serine-threonine protein kinase UL50 UL50 dUTP diphosphatase
UL14 UL14 Tegument protein UL51 UL51 Tegument protein
UL15 Terminase Processing and packaging of DNA UL52 UL52 DNA helicase/primase complex protein
UL16 UL16 Tegument protein UL53 Glycoprotein K Surface and membrane
UL17 UL17 Processing and packaging DNA UL54 IE63; ICP27 Transcriptional regulation
UL18 VP23 Capsid protein UL55 UL55 Unknown
UL19 VP5 Major capsid protein UL56 UL56 Unknown
UL20 UL20 Membrane protein US1 ICP22; IE68 Viral replication
UL21 UL21 Tegument protein[33] US2 US2 Unknown
UL22 Glycoprotein H Surface and membrane US3 US3 Serine/threonine-protein kinase
UL23 Thymidine kinase Peripheral to DNA replication US4 Glycoprotein G Surface and membrane
UL24 UL24 unknown US5 Glycoprotein J Surface and membrane
UL25 UL25 Processing and packaging DNA US6 Glycoprotein D Surface and membrane
UL26 P40; VP24; VP22A Capsid protein US7 Glycoprotein I Surface and membrane
UL27 Glycoprotein B Surface and membrane US8 Glycoprotein E Surface and membrane
UL28 ICP18.5 Processing and packaging DNA US9 US9 Tegument protein
UL29 UL29; ICP8 Major DNA-binding protein US10 US10 Capsid/Tegument protein
UL30 DNA polymerase DNA replication US11 US11; Vmw21 Binds DNA and RNA
UL31 UL31 Nuclear matrix protein US12 ICP47; IE12 Inhibits MHC class I pathway by preventing binding of antigen to TAP
UL32 UL32 Envelope glycoprotein RS1 ICP4; IE175 Major transcriptional activator. Essential for progression beyond the immediate-early phase of infection. IEG transcription repressor.
UL33 UL33 Processing and packaging DNA ICP0 ICP0; IE110; α0 E3 ubiquitin ligase that activates viral gene transcription by opposing chromatinization of the viral genome and counteracts intrinsic- and interferon-based antiviral responses.[34]
UL34 UL34 Inner nuclear membrane protein LRP1 LRP1 Latency-related protein
UL35 VP26 Capsid protein LRP2 LRP2 Latency-related protein
UL36 UL36 Large tegument protein RL1 RL1; ICP34.5 Neurovirulence factor. Antagonizes PKR by de-phosphorylating eIF4a. Binds to BECN1 and inactivates autophagy.
UL37 UL37 Capsid assembly LAT none Latency-associated transcript

HSV-2 viral genome

ORF or feature Comments
a sequence Terminal direct repeat
RL1 Neurovirulence factor
 Exon 1
 Exon 2
RL2 Immediate-early protein; modulator of cell state and gene expression
 Exon 1
 Exon 2
 Exon 3
LAT LAT initiation site; poly(A) site in circularized genome
Start of UL
UL1 Virion surface glycoprotein L
UL2 Uracil-DNA glycosylase
UL3 Nuclear phosphoprotein
UL4
UL5 Component of DNA helicase-primase
UL6 Minor capsid protein
UL7
UL8 Component of DNA helicase-primase
UL9 Ori binding protein
UL10 Virion membrane glycoprotein M
UL11 Myristylated tegument protein
UL12 DNase
UL13 Protein kinase; tegument protein
UL14
UL15 Role in DNA packaging
 Exon 1
 Exon 2
UL16 Proposed initiator CTG codon
UL17
UL18 Capsid protein
UL19 Major capsid protein (start ATG quoted is second possible)
UL20 Virion membrane protein
UL21 Tegument protein
UL22 Virion membrane glycoprotein H
UL23 Thymidine kinase (2 possible poly(A) sites)
UL24
UL25 Virion protein; roles in penetration and virus assembly
UL26 Capsid maturation protease
UL26.5 Capsid assembly protein
UL27 Virion membrane glycoprotein B
UL28 Role in DNA packaging
UL29 Single-stranded DNA binding protein
OriL Origin of DNA replication; location of palindrome given
UL30 DNA polymerase catalytic subunit
UL31
UL32
UL33 Role in DNA packaging
UL34 Membrane-associated phosphoprotein
UL35 Capsid protein
UL36 Very large tegument protein (reiterations omitted for calculation of Ka and Ks)
UL37 Tegument protein
UL38 Capsid protein
UL39 Ribonucleotide reductase large subunit
UL40 Ribonucleotide reductase small subunit
UL41 Tegument protein; host shutoff factor; defective in HSV-2 (HG52) (see text)
UL42 DNA polymerase subunit
UL43 Probable membrane protein
UL44 Virion membrane glycoprotein C
UL45 Tegument/envelope protein
UL46 Tegument protein
UL47 Tegument protein
UL48 Tegument protein; transactivator of immediate-early genes
UL49 Tegument protein
UL49A Probable virion membrane protein
UL50 Deoxyuridine triphosphatase
UL51
UL52 Component of DNA helicase-primase; ATG initiator codon quoted corresponds to HSV-1 (see text)
UL53 Membrane glycoprotein K
UL54 Immediate-early protein; posttranslational regulator of gene expression
UL55
UL56
Start of IRL
LAT LAT initiation and poly(A) sites
RL2 Immediate-early protein; modulator of cell state and gene expression
 Exon 3
 Exon 2
 Exon 1
RL1 Neurovirulence factor
 Exon 2
 Exon 1
a′ sequence Opposite-sense copy of sequence directly repeated at genomic termini
RS1 Immediate-early protein; transcriptional regulator
OriS Origin of DNA replication; limits given are for directly repeated 138 nucleotides
Start of US
US1 Immediate-early protein; intron in 5′ noncoding region
US2
US3 Protein kinase
US4 Virion membrane glycoprotein G
US5 Putative membrane glycoprotein J
US6 Virion membrane glycoprotein D
US7 Virion membrane glycoprotein I
US8 Virion membrane glycoprotein E
US8A Nucleolar protein
US9 Tegument protein
US10 Virion protein
US11 Nucleolar, RNA binding protein
US12 Immediate-early protein; inhibitor of antigen presentation; intron in 5′ noncoding region
Start of TRS
OriS Origin of DNA replication; limits given are for directly repeated 138 nucleotides
RS1 Immediate-early protein; transcriptional regulator
a sequence Terminal direct repeat

Evolution

The herpes simplex 1 genomes can be classified into six clades.[35] Four of these occur in East Africa, one in East Asia and one in Europe and North America. This suggests that the virus may have originated in East Africa. The most recent common ancestor of the Eurasian strains appears to have evolved ~60,000 years ago.[36] The East Asian HSV-1 isolates have an unusual pattern that is currently best explained by the two waves of migration responsible for the peopling of Japan.

The mutation rate has been estimated to be ~1.38×10−7 substitutions/site/year.[35] In clinical setting, the mutations in either the thymidine kinase gene or DNA polymerase gene has caused resistance to aciclovir. However, most of the mutations occur in the thymidine kinase gene rather than the DNA polymerase gene.[37]

Treatment

For more details on treatment of herpes simplex virus, see Herpes simplex.

Herpes viruses establish lifelong infections, and the virus cannot yet be eradicated from the body. Treatment usually involves general-purpose antiviral drugs that interfere with viral replication, reduce the physical severity of outbreak-associated lesions, and lower the chance of transmission to others. Studies of vulnerable patient populations have indicated that daily use of antivirals such as aciclovir[38] and valaciclovir can reduce reactivation rates.[8]

Alzheimer's disease

In the presence of a certain gene variation (APOE-epsilon4 allele carriers), a possible link between HSV-1 (i.e., the virus that causes cold sores or oral herpes) and Alzheimer's disease was reported in 1979.[39] HSV-1 appears to be particularly damaging to the nervous system and increases one’s risk of developing Alzheimer’s disease. The virus interacts with the components and receptors of lipoproteins, which may lead to the development of Alzheimer's disease.[40] This research identifies HSVs as the pathogen most clearly linked to the establishment of Alzheimer’s.[41] According to a study done in 1997, without the presence of the gene allele, HSV-1 does not appear to cause any neurological damage or increase the risk of Alzheimer’s.[42] However, a more recent prospective study published in 2008 with a cohort of 591 people showed a statistically significant difference between patients with antibodies indicating recent reactivation of HSV and those without these antibodies in the incidence of Alzheimer's disease, without direct correlation to the APOE-epsilon4 allele.[43] It should be noted that the trial had a small sample of patients who did not have the antibody at baseline, so the results should be viewed as highly uncertain. In 2011 Manchester University scientists showed that treating HSV1-infected cells with antiviral agents decreased the accumulation of β-amyloid and P-tau, and also decreased HSV-1 replication.[44]

Multiplicity reactivation

Multiplicity reactivation (MR) is the process by which viral genomes containing inactivating damage interact within an infected cell to form a viable viral genome. MR was originally discovered with the bacterial virus bacteriophage T4, but was subsequently also found with pathogenic viruses including influenza virus, HIV-1, adenovirus simian virus 40, vaccinia virus, reovirus, poliovirus and herpes simplex virus.[45]

When HSV particles are exposed to doses of a DNA damaging agent that would be lethal in single infections, but are then allowed to undergo multiple infection (i.e. two or more viruses per host cell), MR is observed. Enhanced survival of HSV-1 due to MR occurs upon exposure to different DNA damaging agents, including methyl methanesulfonate,[46] trimethylpsoralen (which causes inter-strand DNA cross-links),[47][48] and UV light.[49] After treatment of genetically marked HSV with trimethylpsoralen, recombination between the marked viruses increases, suggesting that trimethylpsoralen damages stimulate recombination.[47] MR of HSV appears to partially depend on the host cell recombinational repair machinery since skin fibroblast cells defective in a component of this machinery (i.e. cells from Bloom’s syndrome patients) are deficient in MR.[49] These observations suggest that MR in HSV infections involves genetic recombination between damaged viral genomes resulting in production of viable progeny viruses. HSV-1, upon infecting host cells, induces inflammation and oxidative stress.[50] Thus it appears that the HSV genome may be subjected to oxidative DNA damage during infection, and that MR may enhance viral survival and virulence under these conditions.

Use as an anti-cancer agent

Herpes simplex virus is considered as a potential therapy for cancer and has been extensively clinically tested to assess its oncolytic (cancer killing) ability.[51] Interim overall survival data from Amgen's phase 3 trial of a genetically-attenuated herpes virus suggests efficacy against melanoma.[52]

Use in neuronal connection tracing

Herpes simplex virus is also used as a transneuronal tracer defining connections among neurons by virtue of traversing synapses.[53]

Other related outcomes

Herpes simplex virus is likely the most common cause of Mollaret's meningitis,[54] and, in worse case scenarios, can lead to a potentially fatal case of herpes simplex encephalitis.[55]

Research

There exist commonly used vaccines to some herpesviruses, but only veterinary, such as HVT/LT (Turkey herpesvirus vector laryngotracheitis vaccine), interestingly however, it prevents atherosclerosis (which histologically mirrors atherosclerosis in humans) in target animals vaccinated.[56][57]

Vaccine Company Lead Researcher Vaccine Type Status
HSV-2 ICP0‾ HSV-2 0ΔNLS[58] Rational Vaccines RVx William Halford[59] Live, Attenuated Interferon Sensitive Phase I
dl5-29 / ACAM-529 / HSV-529 Sanofi Pasteur David Knipe[60] Live, Attenuated Replication-Defective HSV Phase I
Admedus[61] Admedus Ian Frazer DNA vaccine: codon optimized Phase II
HerpV Agenus ? Peptide vaccine/QS-21 adjuvant Phase II[62]
Gen-003 Genocea ? Sub Unit gD2/ICP4 with Matrix M2 adjuvant Phase II
Vical Vical ? DNA vaccine: gD2+UL46/Vaxfectin adjuvant Phase II
Einstein Einstein Med College William Jacobs Jr Live, Attenuated HSV-2 deleted in gD2 Preclinical
GV2207[63] GenVec ? ? Preclinical[64]
Mymetics[65] Mymetics ? ? Preclinical[65]
Vitaherpavac & Herpovax ? ? ? ?
NE-HSV2[66] NanoBio[67] ? ? Preclinical
GeneVax prime/[68] Profectus BioSciences ? ? ?
Biomedical Research Models ? ? ? ?
Tomegavax ? ? ? ?
Herpevac GlaxoSmithKline ? Sub Unit gD2t with alum/MPL adjuvant[69] Discontinued, failed in Phase III trial stage[70]
PaxVax[71] ? ? ? Discontinued
Amgen BioVex ? ? ? ?
AuRX ? ? Live, Attenuated Inactive
Zostavax

(VZV, shingles)

Merck ? Live, Attenuated In Production
Varivax,[72] Varilrix[73] (Varicella, C.Pox) Merck, GlaxoSmithKline ? Live, Attenuated In Production
Shingrix, GSK1437173A

(VZV, shingles)

GlaxoSmithKline ? Sub Unit gE with AS01 adjuvant system[74][75] Phase III[76]

References

  1. 1 2 3 Ryan KJ, Ray CG (editors) (2004). Sherris Medical Microbiology (4th ed.). McGraw Hill. pp. 555–62. ISBN 0-8385-8529-9.
  2. "Herpes simplex". DermNet NZ — New Zealand Dermatological Society. 2006-09-16. Retrieved 2006-10-15.
  3. Straface, Gianluca; Selmin, Alessia; Zanardo, Vincenzo; De Santis, Marco; Ercoli, Alfredo; Scambia, Giovanni (2012). "Herpes Simplex Virus Infection in Pregnancy". Infectious Diseases in Obstetrics and Gynecology. 2012: 1–6. doi:10.1155/2012/385697. ISSN 1064-7449.
  4. Schiffer JT, Mayer BT, Fong Y, Swan DA, Wald A (2014). "Herpes simplex virus-2 transmission probability estimates based on quantity of viral shedding". J R Soc Interface. 11 (95): 20140160. doi:10.1098/rsif.2014.0160. PMID 24671939.
  5. 1 2 Johnston C, Koelle DM, Wald A (Dec 2011). "HSV-2: in pursuit of a vaccine.". J Clin Invest. 121 (12): 4600–9. doi:10.1172/JCI57148. PMID 22133885.
  6. Sperling RS, Fife KH, Warren TJ, Dix LP, Brennan CA (March 2008). "The effect of daily valacyclovir suppression on herpes simplex virus type 2 viral shedding in HSV-2 seropositive subjects without a history of genital herpes". Sex Transm Dis. 35 (3): 286–90. doi:10.1097/OLQ.0b013e31815b0132. PMID 18157071.
  7. 1 2 Gupta R, Warren T, Wald A (2007). "Genital herpes". Lancet. 370 (9605): 2127–37. doi:10.1016/S0140-6736(07)61908-4. PMID 18156035.
  8. 1 2 Koelle DM, Corey L (2008). "Herpes simplex: insights on pathogenesis and possible vaccines". Annual Review of Medicine. 59: 381–95. doi:10.1146/annurev.med.59.061606.095540. PMID 18186706.
  9. "EVERYTHING YOU NEED TO KNOW ABOUT HERPES".
  10. Corey L, Wald A (2009). "Maternal and Neonatal HSV Infections". New England Journal of Medicine. 361 (14): 1376–85. doi:10.1056/NEJMra0807633. PMC 2780322Freely accessible. PMID 19797284.
  11. Kimberlin DW (2007). "Herpes simplex virus infections of the newborn". Semin. Perinatol. 31 (1): 19–25. doi:10.1053/j.semperi.2007.01.003. PMID 17317423.
  12. 1 2 Mettenleiter TC, Klupp BG, Granzow H (2006). "Herpesvirus assembly: a tale of two membranes". Curr. Opin. Microbiol. 9 (4): 423–9. doi:10.1016/j.mib.2006.06.013. PMID 16814597.
  13. 1 2 3 4 5 McGeoch DJ, Rixon FJ, Davison AJ (2006). "Topics in herpesvirus genomics and evolution". Virus Res. 117 (1): 90–104. doi:10.1016/j.virusres.2006.01.002. PMID 16490275.
  14. Rajcáni J, Andrea V, Ingeborg R (2004). "Peculiarities of herpes simplex virus (HSV) transcription: an overview". Virus Genes. 28 (3): 293–310. doi:10.1023/B:VIRU.0000025777.62826.92. PMID 15266111.
  15. 1 2 Clarke RW (2015). "Forces and Structures of the Herpes Simplex Virus (HSV) Entry Mechanism". ACS Infectious Diseases. 1 (9): 403–415. doi:10.1021/acsinfecdis.5b00059.
  16. 1 2 Subramanian RP, Geraghty RJ (2007). "Herpes simplex virus type 1 mediates fusion through a hemifusion intermediate by sequential activity of glycoproteins D, H, L, and B". Proc. Natl. Acad. Sci. U.S.A. 104 (8): 2903–8. doi:10.1073/pnas.0608374104. PMC 1815279Freely accessible. PMID 17299053.
  17. Akhtar J, Shukla D (2009). "Viral entry mechanisms: Cellular and viral mediators of herpes simplex virus entry". FEBS Journal. 276 (24): 7228–7236. doi:10.1111/j.1742-4658.2009.07402.x. PMC 2801626Freely accessible. PMID 19878306.
  18. 1 2 Cardone G, Winkler DC, Trus BL, Cheng N, Heuser JE, Newcomb WW, Brown JC, Steven AC (May 2007). "Visualization of the Herpes Simplex Virus Portal in situ by Cryo-electron Tomography". Virology. 361 (2): 426–34. doi:10.1016/j.virol.2006.10.047. PMC 1930166Freely accessible. PMID 17188319.
  19. 1 2 Trus BL, Cheng N, Newcomb WW, Homa FL, Brown JC, Steven AC (November 2004). "Structure and Polymorphism of the UL6 Portal Protein of Herpes Simplex Virus Type 1". Journal of Virology. 78 (22): 12668–71. doi:10.1128/JVI.78.22.12668-12671.2004. PMC 525097Freely accessible. PMID 15507654.
  20. 1 2 Nellissery JK, Szczepaniak R, Lamberti C, Weller SK (2007-06-20). "A Putative Leucine Zipper within the Herpes Simplex Virus Type 1 UL6 Protein Is Required for Portal Ring Formation". Journal Virology. 81 (17): 8868–77. doi:10.1128/JVI.00739-07. PMC 1951442Freely accessible. PMID 17581990.
  21. Newcomb WW, Booy FP, Brown JC (2007). "Uncoating the Herpes Simplex Virus Genome". J. Mol. Biol. 370 (4): 633–42. doi:10.1016/j.jmb.2007.05.023. PMC 1975772Freely accessible. PMID 17540405.
  22. Abbas et al (2009) Cellular and Molecular Immunology, Elsevier Inc.
  23. Adang LA, Parsons CH, Kedes DH (2006). "Asynchronous Progression through the Lytic Cascade and Variations in Intracellular Viral Loads Revealed by High-Throughput Single-Cell Analysis of Kaposi's Sarcoma-Associated Herpesvirus Infection". J. Virol. 80 (20): 10073–82. doi:10.1128/JVI.01156-06. PMC 1617294Freely accessible. PMID 17005685.
  24. 1 2 Matis J, Kúdelová M (2001). "Early shutoff of host protein synthesis in cells infected with herpes simplex viruses". Acta Virol. 45 (5–6): 269–77. doi:10.2217/fvl.11.24. PMID 12083325.
  25. Taddeo B, Roizman B (2006). "The Virion Host Shutoff Protein (UL41) of Herpes Simplex Virus 1 Is an Endoribonuclease with a Substrate Specificity Similar to That of RNase A". J. Virol. 80 (18): 9341–5. doi:10.1128/JVI.01008-06. PMC 1563938Freely accessible. PMID 16940547.
  26. Skepper JN, Whiteley A, Browne H, Minson A (June 2001). "Herpes Simplex Virus Nucleocapsids Mature to Progeny Virions by an Envelopment → Deenvelopment → Reenvelopment Pathway". J. Virol. 75 (12): 5697–702. doi:10.1128/JVI.75.12.5697-5702.2001. PMC 114284Freely accessible. PMID 11356979.
  27. Granzow H, Klupp BG, Fuchs W, Veits J, Osterrieder N, Mettenleiter TC (April 2001). "Egress of Alphaherpesviruses: Comparative Ultrastructural Study". J. Virol. 75 (8): 3675–84. doi:10.1128/JVI.75.8.3675-3684.2001. PMC 114859Freely accessible. PMID 11264357.
  28. 1 2 Pinnoji RC, Bedadala GR, George B, Holland TC, Hill JM, Hsia SC (2007). "Repressor element-1 silencing transcription factor/neuronal restrictive silencer factor (REST/NRSF) can regulate HSV-1 immediate-early transcription via histone modification". Virol. J. 4: 56. doi:10.1186/1743-422X-4-56. PMC 1906746Freely accessible. PMID 17555596.
  29. Bedadala GR, Pinnoji RC, Hsia SC (2007). "Early growth response gene 1 (Egr-1) regulates HSV-1 ICP4 and ICP22 gene expression". Cell Res. 17 (6): 546–55. doi:10.1038/cr.2007.44. PMID 17502875.
  30. Roizman B, Gu H, Mandel G (2005). "The first 30 minutes in the life of a virus: unREST in the nucleus". Cell Cycle. 4 (8): 1019–21. doi:10.4161/cc.4.8.1902. PMID 16082207.
  31. Search in UniProt Knowledgebase (Swiss-Prot and TrEMBL) for: HHV1
  32. Wyrwicz LS, Ginalski K, Rychlewski L (2007). "HSV-1 UL45 encodes a carbohydrate binding C-type lectin protein". Cell Cycle. 7 (2): 269–71. doi:10.4161/cc.7.2.5324. PMID 18256535.
  33. Vittone V, Diefenbach E, Triffett D, Douglas MW, Cunningham AL, Diefenbach RJ (2005). "Determination of Interactions between Tegument Proteins of Herpes Simplex Virus Type 1". J. Virol. 79 (15): 9566–71. doi:10.1128/JVI.79.15.9566-9571.2005. PMC 1181608Freely accessible. PMID 16014918.
  34. Matis J, Kúdelová M (2011). "HSV-1 ICP0: paving the way for viral replication". Future Virology. 6 (4): 421–429. doi:10.2217/fvl.11.24. PMC 3133933Freely accessible. PMID 21765858.
  35. 1 2 Kolb AW, Ané C, Brandt CR (2013). "Using HSV-1 genome phylogenetics to track past human migrations". PLOS ONE. 8 (10): e76267. doi:10.1371/journal.pone.0076267.
  36. Bowden R, Sakaoka H, Ward R, Donnelly P (2006). "Patterns of Eurasian HSV-1 molecular diversity and inferences of human migrations". Infect Genet Evol. 6 (1): 63–74. doi:10.1016/j.meegid.2005.01.004.
  37. Hussin A, Md , Nor NS, Ibrahim N (2013). "Phenotypic and genotypic characterization of induced acyclovir-resistant clinical isolates of herpes simplex virus type 1". Antivir Res. 100: 306–313. doi:10.1016/j.antiviral.2013.09.008.
  38. Kimberlin DW, Whitley RJ, Wan W, Powell DA, Storch G, Ahmed A, Palmer A, Sánchez PJ, Jacobs RF, Bradley JS, Robinson JL, Shelton M, Dennehy PH, Leach C, Rathore M, Abughali N, Wright P, Frenkel LM, Brady RC, Van Dyke R, Weiner LB, Guzman-Cottrill J, McCarthy CA, Griffin J, Jester P, Parker M, Lakeman FD, Kuo H, Lee CH, Cloud GA (2011). "Oral acyclovir suppression and neurodevelopment after neonatal herpes". N. Engl. J. Med. 365 (14): 1284–92. doi:10.1056/NEJMoa1003509. PMC 3250992Freely accessible. PMID 21991950.
  39. Middleton PJ, Petric M, Kozak M, Rewcastle NB, McLachlan DR (May 1980). "Herpes-simplex viral genome and senile and presenile dementias of Alzheimer and Pick". Lancet. 315 (8176): 1038. doi:10.1016/S0140-6736(80)91490-7. PMID 6103379.
  40. Dobson CB, Itzhaki RF (1999). "Herpes simplex virus type 1 and Alzheimer's disease". Neurobiol. Aging. 20 (4): 457–65. doi:10.1016/S0197-4580(99)00055-X. PMID 10604441.
  41. Pyles RB (November 2001). "The association of herpes simplex virus and Alzheimer's disease: a potential synthesis of genetic and environmental factors" (PDF). Herpes. 8 (3): 64–8. PMID 11867022.
  42. Itzhaki RF, Lin WR, Shang D, Wilcock GK, Faragher B, Jamieson GA (January 1997). "Herpes simplex virus type 1 in brain and risk of Alzheimer's disease". Lancet. 349 (9047): 241–4. doi:10.1016/S0140-6736(96)10149-5. PMID 9014911.
  43. Letenneur L, Pérès K, Fleury H, Garrigue I, Barberger-Gateau P, Helmer C, Orgogozo JM, Gauthier S, Dartigues JF (2008). "Seropositivity to herpes simplex virus antibodies and risk of Alzheimer's disease: a population-based cohort study.". PLoS ONE. 3 (11): e3637. doi:10.1371/journal.pone.0003637. PMC 2572852Freely accessible. PMID 18982063.
  44. Wozniak MA, Frost AL, Preston CM, Itzhaki RF (2011). "Antivirals Reduce the Formation of Key Alzheimer's Disease Molecules in Cell Cultures Acutely Infected with Herpes Simplex Virus Type 1". PLoS ONE. 6 (10): e25152. doi:10.1371/journal.pone.0025152.
  45. Michod RE, Bernstein H, Nedelcu AM (2008). "Adaptive value of sex in microbial pathogens". Infection, Genetics and Evolution. 8 (3): 267–285. doi:10.1016/j.meegid.2008.01.002. PMID 18295550.
  46. Das SK (1982). "Multiplicity reactivation of alkylating agent damaged herpes simplex virus (type I) in human cells". Mutation research. 105 (1–2): 15–18. doi:10.1016/0165-7992(82)90201-9. PMID 6289091.
  47. 1 2 Hall JD, Scherer K (1981). "Repair of psoralen-treated DNA by genetic recombination in human cells infected with herpes simplex virus". Cancer Research. 41 (12 Pt 1): 5033–5038. PMID 6272987.
  48. Coppey J, Sala-Trepat M, Lopez B (1989). "Multiplicity reactivation and mutagenesis of trimethylpsoralen-damaged herpes virus in normal and Fanconi's anaemia cells". Mutagenesis. 4 (1): 67–71. doi:10.1093/mutage/4.1.67. PMID 2541311.
  49. 1 2 Selsky CA, Henson P, Weichselbaum RR, Little JB (1979). "Defective reactivation of ultraviolet light-irradiated herpesvirus by a Bloom's syndrome fibroblast strain". Cancer Research. 39 (9): 3392–3396. PMID 225021.
  50. Valyi-Nagy T, Olson SJ, Valyi-Nagy K, Montine TJ, Dermody TS (2000). "Herpes Simplex Virus Type 1 Latency in the Murine Nervous System is Associated with Oxidative Damage to Neurons". Virology. 278 (2): 309–321. doi:10.1006/viro.2000.0678. PMID 11118355.
  51. Varghese S, Rabkin SD (1 December 2002). "Oncolytic herpes simplex virus vectors for cancer virotherapy". Cancer Gene Therapy. 9 (12): 967–978. doi:10.1038/sj.cgt.7700537. PMID 12522436.
  52. "Amgen Presents Interim Overall Survival Data From Phase 3 Study Of Talimogene Laherparepvec In Patients With Metastatic Melanoma" (Press release). November 18, 2013. Retrieved 30 October 2015.
  53. Norgren, R. B., Jr., & Lehman, M. N. (1998). "Herpes simplex virus as a transneuronal tracer. [Review].". Neurosci Biobehav Rev. 22 (6): 695–708. doi:10.1016/s0149-7634(98)00008-6. PMID 9809305.
  54. Harrisons Principles of Internal Medicine, 19th edition. p. 1179. ISBN 9780071802154.
  55. "Meningitis - Infectious Disease and Antimicrobial Agents". www.antimicrobe.org. Retrieved 2016-03-14.
  56. http://www.bioone.org/doi/abs/10.1637/10383-092412-Reg.1
  57. https://books.google.com/books?id=0ToGnkORSAMC&pg=PA25&lpg=PA25 Role of Herpesvirus in Artherogenesis edited by David P Hajjar, Stephen M Schwartz
  58. "Introducing RVx". 2016-03-12. Retrieved 2016-08-02.
  59. "Herpes Vaccine Research". Herpes Vaccine Research. Retrieved 2016-08-02.
  60. "Knipe Lab | Harvard Medical School". knipelab.med.harvard.edu. Retrieved 2016-08-02.
  61. "Admedus US Research & Development - Admedus US". www.admedus.com. Retrieved 2016-08-02.
  62. "Biological Efficacy Study of HerpV Vaccine With QS-21 to Treat Subjects With Recurrent Genital Herpes". Retrieved 31 August 2016.
  63. "GV2207 – HSV-2 Immunotherapeutic :: GenVec, Inc. (GNVC)". www.genvec.com. Retrieved 2016-08-16.
  64. "GV2207 – HSV-2 Immunotherapeutic :: GenVec, Inc. (GNVC)". www.genvec.com. Retrieved 2016-08-16.
  65. 1 2 "Herpes Simplex". MyMetics. Retrieved 2016-08-03.
  66. "Nanobio - HSV-2 Vaccine". Retrieved 2 August 2016.
  67. Corporation, NanoBio. "NanoBio's Genital Herpes Vaccine Demonstrates Efficacy In Guinea Pigs As Both A Prophylactic And A Therapeutic Vaccine". Retrieved 2 August 2016.
  68. "PBS Vax™ Therapeutic Vaccines". profectusbiosciences.com. Retrieved 15 August 2016.
  69. "Status of Vaccine Research and Development of Vaccines for Herpes Simplex Virus" (PDF). Retrieved 30 August 2016.
  70. "QUESTIONS AND ANSWERS The Herpevac Trial for Women". Retrieved 30 August 2016.
  71. "PaxVax Signs R&D Collaboration with UC San Diego to Develop a Vaccine to Prevent Herpes Simplex Virus Infections". paxvax.com. Retrieved 15 August 2016.
  72. "VARIVAX® Varicella Virus Vaccine Live" (PDF). Retrieved 30 August 2016.
  73. "VARILRIX® Live attenuated varicella vaccine" (PDF). Retrieved 23 August 2016.
  74. "Vaccine Detail GSK1437173A". Retrieved 31 August 2016.
  75. "GSK's candidate Shingles Vaccine Containing Agenus' QS-21 Stimulon® Demonstrates 90% Efficacy against Shingles and 89% Efficacy against Post-Herpetic Neuralgia in Adults aged 70 Years and Older". Retrieved 31 August 2016.
  76. "Cross-vaccination Study of GSK Biologicals' Herpes Zoster Subunit (HZ/su) Vaccine (GSK 1437173A) in Subjects Who Previously Received Placebo in ZOSTER-006 (NCT01165177) and ZOSTER-022 (NCT01165229) Studies.". Retrieved 31 August 2016.

External links

This article is issued from Wikipedia - version of the 12/3/2016. The text is available under the Creative Commons Attribution/Share Alike but additional terms may apply for the media files.