Herpes simplex research

Herpes simplex research includes all medical research that attempts to prevent, treat, or cure herpes, as well as fundamental research about the nature of herpes. Examples of particular herpes research include, drug development and herpes vaccines. HSV-1 and HSV-2 are commonly thought of as oral and genital herpes respectively, but other members in the herpes family include chickenpox (varicella/zoster), cytomegalovirus (CMV), and Epstein-Barr (EBV).

Vaccine research

Various vaccine candidates have been developed, the first ones in the 1920s, but none has been successful to date.[1]

Due to the genetic similarity of both herpes simplex virus types (HSV-1 and HSV-2), the development of a prophylactic-therapeutic vaccine that proves effective against one type of the virus, would likely prove effective for the other virus type, or at least provide most of the necessary fundamentals. As of 2016, several vaccine candidates are in different stages of clinical trials.

An ideal herpes vaccine should induce immune responses adequate to prevent infection. Short of this ideal, a candidate vaccine might be considered successful if it (a) mitigates primary clinical episodes, (b) prevents colonization of the ganglia, (c) helps reduce the frequency or severity of recurrences, and (d) reduces viral shedding in actively infected or asymptomatic individuals.[2]

Vaccine candidate

The chart below is an attempt to list all known proposed vaccines and their characteristics, please update with any missing information on vaccines only.

Vaccine Company Lead Researcher Vaccine Type Status
HSV-2 ICP0‾ HSV-2 0ΔNLS[3] Rational Vaccines RVx William Halford[4] Live, Attenuated Interferon Sensitive Phase I
dl5-29 / ACAM-529 / HSV-529 Sanofi Pasteur David Knipe[5] Live, Attenuated Replication-Defective HSV Phase I
Admedus[6] Admedus Ian Frazer DNA vaccine: codon optimized Phase II
HerpV Agenus ? Peptide vaccine/QS-21 adjuvant Phase II[7]
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[8] GenVec ? ? Preclinical[9]
Mymetics[10] Mymetics ? ? Preclinical[10]
Vitaherpavac & Herpovax ? ? ? ?
NE-HSV2[11] NanoBio[12] ? ? Preclinical
GeneVax prime/[13] Profectus BioSciences ? ? ?
Biomedical Research Models ? ? ? ?
Tomegavax ? ? ? ?
Herpevac GlaxoSmithKline ? Sub Unit gD2t with alum/MPL adjuvant[14] Discontinued, failed in Phase III trial stage[15]
PaxVax[16] ? ? ? Discontinued
Amgen BioVex ? ? ? ?
AuRX ? ? Live, Attenuated Inactive
Zostavax

(VZV, shingles)

Merck ? Live, Attenuated In Production
Varivax,[17] Varilrix[18] (Varicella, C.Pox) Merck, GlaxoSmithKline ? Live, Attenuated In Production
Shingrix, GSK1437173A

(VZV, shingles)

GlaxoSmithKline ? Sub Unit gE with AS01 adjuvant system[19][20] Phase III[21]

Attenuated vaccines

Depiction shows mice who were vaccinated with two different vaccine candidates, then are exposed to a common wild-type HSV strain usually found in humans. A generic subunit gD vaccine does confer only weak protection, with 1 of 4 mice surviving after a month. The effect of Halford's HSV-2 ICP0 vaccine, achieving sterilizing immunity in vaccinated mice with 5 of 5 mice surviving after a month.

Live, attenuated variant of the HSV-2 vaccine

Diverse subunit HSV vaccines (e.g. Herpevac) have failed to protect humans from acquiring genital herpes in several clinical trials. The success of the chickenpox vaccine demonstrates that a live and appropriately attenuated α-herpesvirus may be used to safely control human disease. Dr. William Halford at the Southern Illinois University (SIU) School of Medicine is presently testing a live-attenuated HSV-2 ICP0‾ vaccine.[22][23][24] Already proven as safe and effective in studies on animals, eliciting 10 to 100 times greater protection against genital herpes than a glycoprotein D subunit vaccine, Halford's vaccine is now currently involved in clinical trials. In 2016, Halford had promising results injecting his vaccine in 20 human subjects, 17 of which got all 3 shots, (the other 3, only 2 shots). All 20 of the participants self-reported an improvement in symptoms, but only 17 received and completed all three dosages.[25] Blot tests showed a clear antibody response, which can not be instigated by a placebo effect.

Replication-defective HSV-2 vaccine

Principle of HSV529

David Knipe, a Professor at Harvard Medical School has developed dl5-29. The dl5-29 vaccine is also known under the name ACAM-529[26] or HSV-529, a replication-defective vaccine that has proved successful in preventing both HSV-2 and HSV-1 infections and in combating the virus in already-infected hosts, in animal models.[27] The HSV-529 is a leading vaccine candidate which has been investigated in numerous research publications, and is endorsed by many researchers in the field (i.a. Lynda A. Morrison and Jeffrey Cohen).[28] It has also been shown that the vaccine induces strong HSV-2-specific antibody and T-cell responses, protects against challenge with a wild-type HSV-2 virus, reduces the severity of recurrent disease, provides cross-protection against HSV-1.[29] The ongoing trials would prove if a durable immune response in humans is to be succesfully achieved or if the vaccine is too over attenuated to do the same. The vaccine is now being researched and developed by Sanofi Pasteur.[30] Both Sanofi Pasteur and the clinical-stage immunotherapy company Immune Design have entered a broad collaboration, which will explore the potential of various combinations of agents against HSV-2, including an adjuvanted trivalent vaccine candidate G103, consisting of recombinantly-expressed viral proteins.[31]

Glycoprotein gD- and DNA-based vaccines

Admedus's vaccine

Professor Ian Frazer developed an experimental vaccine with his team at Coridon, a biotechnology company he founded in 2000. The company, now known under the name Admedus Vaccines, is researching DNA technology for vaccines with prophylactic and therapeutic potential. What's different about this vaccine is the way that response is being created. Instead of introducing a weakened version of the herpes virus, this vaccine uses a small section of DNA to produce T-cells and stimulate the immune response.[32] The new vaccine candidate is designed to prevent new infections, and to treat those who already have the infection. In February 2014, it was announced that Frazer's new vaccine against genital herpes has passed human safety trials in a trial of 20 Australians.[33] In October 2014, Admedus announced success in creating a positive T-cell response in 95% of participants.[34] Further research is required to determine if the vaccine can prevent transmission. In July 2014, Admedus increased its stake in Frazer's vaccines by 16.2%. In addition, $18.4 million was posted as funds raised towards Phase II vaccine testing and research.[35]

The HSV-2 Phase II trial began in April 2015.[36] Interim results were published on March 4, 2016 and based on the results of a scheduled, blinded, pooled analysis of data from the first 20 patients to receive at least three vaccinations in the randomised, placebo controlled Phase II study with the following results:

Admedus plans an analysis of the unblinded data from these 20 study participants to be performed during Q3 2016.[37][38]

Genocea's vaccine

Genocea Biosciences has developed GEN-003, a first-in-class protein subunit T cell-enabled therapeutic vaccine, or immunotherapy, designed to reduce the duration and severity of clinical symptoms associated with moderate-to-severe HSV-2, and to control transmission of the infection. GEN-003 includes the antigens ICP4 and gD2, as well as the proprietary adjuvant Matrix-M.

GEN-003 is concluding Phase IIa clinical trials. In December 2015, Genocea announced interim data showing a 58% decrease in viral shedding and a 69% decrease in genital lesions. They also showed one of the doses stopped outbreaks for at least 6 months.[39]

GEN-003 is undergoing a Phase IIb clinical trial in the United States.

Other vaccines

A study from the Albert Einstein College of Medicine, where glycoprotein D (gD-2) was deleted from the herpes cell, showed positive results when tested in mice.[40] Researchers deleted gD-2 from the herpes virus, which is responsible for herpes microbes entering in and out of cells. The vaccine is still in early stages of development and more research needs to be conducted before receiving FDA approval for clinical trials.[41]

Research conducted by the NanoBio Corporation indicates that an enhanced protection from HSV-2 is a result of mucosal immunity which can be elicited by their intranasal nanoemulsion vaccine. NanoBio published results showing efficiency in studies conducted in both the prophylactic and the therapeutic guinea pig model. This included preventing infection and viral latency in 92% of animals vaccinated and a reduction in recurrent legions by 64% and viral shedding by 53%. NanoBio hopes to raise funds in 2016 to enter into Phase I clinical testing.[42]

Profectus BioSciences intends to use its PBS Vax therapeutic vaccine technology to engineer a vaccine for HSV-2.[43] The vaccine is in early development and much is unknown about its viability.

Biomedical Research Models, a Worcester-based biopharmaceutical company has been awarded a fund for the development of a novel vaccine platform to combat mucosally transmitted pathogens such as HSV-2.[44]

The company Tomegavax is researching to utilize cytomegalovirus (CMV) vectors in the development of a therapeutic vaccine against herpes simplex virus 2 (HSV-2), the causative agent of genital herpes. It has been awarded a grant by the NIH for this purpose.[45]

HerpV, a genital herpes vaccine candidate manufactured by the company Agenus, announced Phase II clinical trial results in June 2014. Results showed up to a 75% reduction in viral load and a weak reduction in viral shedding by 14%.[46] These results were achieved after a series of vaccinations and a booster dose after six months, signalling the vaccine may take time to become effective. Further testing results are to show if the vaccine is a viable candidate against genital herpes.[47] There has not been any recent announcement by Agenus regarding the vaccine HerpV, which seems to have been taken off from the company's research product pipeline.[48]

Mymetics is developing a pre-clinical preventative vaccine for HSV 1 and 2 using its virosome technology.[49] Not much is known about this vaccine or its viability.

Vical had been awarded grant funding from the National Institute of Allergy and Infectious Diseases division of the NIH to develop a plasmid DNA-based vaccine to inhibit recurring lesions in patients latently infected with herpes simplex virus type 2 (HSV-2). The plasmid DNA encoding the HSV-2 antigens was formulated with Vaxfectin, Vical's proprietary cationic lipid adjuvant. Vical is concluding Phase I clinical trials, while reporting data showing the vaccine candidate failed to meet the primary endpoint.[50] The San Diego-based company was forced to concede that their herpes strategy had misfired, with their vaccine failing to perform as well as a placebo.[51] However, that may have changed, since 20 June 2016, when Vical released phase I/II results at ASM.

Discontinued vaccines

One vaccine that was under trial was Herpevac, a vaccine against HSV-2. The National Institutes of Health (NIH) in the United States conducted phase III trials of Herpevac.[52] In 2010, it was reported that, after 8 years of study in more than 8,000 women in the United States and Canada, there was no sign of positive results against the sexually transmitted disease caused by HSV-2[53] (and this despite earlier favorable interim reports[52]).

PaxVax, a specialty vaccine company, partnered with Spector Lab at the UC San Diego Department of Cellular and Molecular Medicine regarding the development of a genital herpes viral vector vaccine. The vaccine was in the pre-clinical stage.[54] The vaccine is no longer listed on their website as a present endeavour and has likely been discontinued.[55]

A private company called BioVex began Phase I clinical trials for ImmunoVEX, another proposed vaccine, in March 2010.[56] The Company had commenced clinical testing in the UK with its vaccine candidate for the prevention and potentially the treatment of genital herpes. The biopharmaceutical company Amgen bought BioVex[57] and their proposed Immunovex vaccine appears to have been discontinued, furthermore it has been removed from the company's research pipeline.[58]

A live, attenuated vaccine (which was proven very effective in clinical trials in Mexico) by the company AuRx has failed to proceed to a Phase III trial in the year 2006, due to financial reasons. The AuRx therapy was shown to be safe and decrease the occurrence of lesions by 86% after one year.[59] The fact that a live, attenuated vaccine induced better protection from HSV infection and symptoms is not new, because live-attenuated vaccines account for the most of the successful vaccines in the use until today. However, governmental and corporate bodies seem to support the more recent but possibly less effective approaches such as Glycoprotein and DNA based vaccines.

Herpes simplex virus

A research paper providing an overview of the relatively recent state of research, can be found on this page.

Pharmaceutical drugs

Since the introduction of the nucleoside analogs decades ago, treatment of herpes simplex virus (HSV) infections has not seen much innovation, except for the development of their respective prodrugs (Aciclovir, Famciclovir, Valacilovir..). Drawbacks such as as poor bioavailability or limited effectiveness of these drugs require further research effort of new pharmaceutical drugs against the herpes simplex disease. The inhibitors of the Helicase-primase complex of HSV represent a very innovative approach to the treatment of herpesvirus disease.[60] Even though new categories of drugs may seem promising, these would still be only more effective therapeutic drugs, because the permanent removal of herpes simplex symptoms in patients is highly unlikely, due to the fact that HSV is neurotropic and the virus reservoirs lie within neural tissue.

Pharmaceutical Drug Company Lead Researcher Type Status
Aciclovir patents expired Schaeffer & B. Elion nucleic acid analogue In Production
Valaciclovir patents expired ? nucleic acid analogue In Production
Famciclovir patents expired ? nucleic acid analogue In Production
AIC316 AICuris ? helicase-primase inhibitor Phase II[61]
ASP2151 Astellas Pharma Inc Kiyomitsu Katsumata[62] helicase-primase inhibitor ?

Notable progress

Researchers have made a Hammerhead ribozyme that targets and cleaves the mRNA of essential genes in HSV-1. The hammerhead, which targets the mRNA of the UL20 gene, greatly reduced the level of HSV-1 ocular infection in rabbits, and reduced the viral yield in vivo.[63] The gene-targeting approach uses a specially designed RNA enzyme to inhibit strains of the herpes simplex virus. The enzyme disables a gene responsible for producing a protein involved in the maturation and release of viral particles in an infected cell. The technique appears to be effective in experiments with mice and rabbits, but further research is required before it can be attempted in people infected with herpes.[64]

In 2016, researchers showed that the genome editing technology known as CRISPR/Cas can be used to limit viral replication in multiple strains of herpesviruses, in some cases even eliminating the infection altogether.[65] The researchers tested three different strains of herpesviruses: Epstein-Barr virus (EBV), the cause of mononucleosis and some cancers; Herpes simplex viruses (HSV-1) and (HSV-2), which cause cold sores and genital herpes respectively; and Human cytomegalovirus (HCMV), which causes congenital herpes.The results indicated that CRISPR can be used to eliminate replication in all three strains of the virus, but that the technology was so far only successful in actually eradicating EBV. The authors think this may be because the EBV genome is located in dividing cells that are easily accessible to CRISPR. Comparatively, the HSV-1 genome targeted by CRISPR is located in closed-off, non-replicating neurons, which makes reaching the genome much more challenging.[66]

Another possibility to eradicate the HSV-1 variant is being pursued by a team at Duke University. By figuring out how to switch all copies of the virus in the host from latency to their active stage at the same time, rather than the way the virus copies normally stagger their activity stage, leaving some dormant somewhere at all times, it is thought that immune system could kill the entire infected cell population, since they can no longer hide in the nerve cells. This is a potentially risky approach especially for patients with widespread infections as there is the possibility of significant tissue damage from the immune response. One class of drugs called antagomir could trigger reactivation. These are chemically engineered oligonucleotides or short segments of RNA, that can be made to mirror their target genetic material, namely herpes microRNAs. They could be engineered to attach and thus 'silence' the microRNA, thus rendering the virus incapable of keeping latent in their host.[67] Professor Cullen believes a drug could be developed to block the microRNA whose job it is to suppress HSV-1 into latency.[68]

Herpes has been used in research with HeLa cells to determine its ability to assist in the treatment of malignant tumors. A study conducted using suicide gene transfer by a cytotoxic approach examined a way to eradicate malignant tumors.[69] Gene therapy is based on the cytotoxic genes that directly or indirectly kill tumor cells regardless of its gene expression. In this case the study uses the transfer of the Herpes simplex virus type I thymidine kinase (HSVtk) as the cytotoxic gene. Hela cells were used in these studies because they have very little ability to communicate through gap junctions.[70] The Hela cells involved were grown in a monolayer culture and then infected with the HSV virus. The HSV mRNA was chosen because it is known to share characteristics with normal eukaryotic mRNA.[71]

The HSVtk expression results in the phosphorylation of drug nucleoside analogues; in this case the drug ganciclovir, an anitiviral medication used to treat and prevent cytomegaloviruses, converts it into the nucleoside analogue triphosphates. Once granciclovir is phosphorylated through HSV-tk it is then incorporating DNA strands when the cancer cells are multiplying.[70] The nucleotide from the ganciclovir is what inhibits the DNA polymerization and the replication process. This causes the cell to die via apoptosis.[69]

Apoptosis is regulated with the help of miRNAs, which are small non-coding RNAs that negatively regulate gene expression.[72] These miRNAs play a critical role in developing the timing, differentiation and cell death. The miRNAs effect on apoptosis has affected cancer development by the regulation of cell proliferation, as well as cell transformation. Avoidance of apoptosis is critical for the success of malignant tumors, and one way for miRNAs to possibly influence cancer development is to regulate apoptosis. In order to support this claim, Hela cells were used for the experiment discussed.

The cytotoxic drug used, ganciclovir, is capable of destroying via apoptosis transduced cells and non-transduced cells from the cellular gap junction. This technique is known as the "bystander effect,” which has suggested to scientists that the effect of some therapeutic agents may be enhanced by diffusion through gap junctional intercellular communication (GJIC) or cell coupling. GJIC is an important function in the maintaining of tissue homeostasis and it is a critical factor in balance of cells dying and surviving.

When Hela cells were transfected with the HSV-tk gene, and were then put in a culture with nontransfected cells, only the HSV-tk transfected Hela cells were killed by the granciclovir, leaving the nonviral cells unharmed.[70] The Hela cells were transfected with the encoding for the gap junction protenin connexin 43 (Cx43) to provide a channel that permits ions and other molecules to move between neighboring cells. Both Hela cells with the HSV-tk and without the HSV-tk were destroyed. This result has led to the evidence needed to state that the bystander effect in the HSV-tk gene therapy is possibly due to the Cx-mediated GJIC.

References

  1. Chentoufi AA, Kritzer E, Yu DM, Nesburn AB, Benmohamed L (2012). "Towards a rational design of an asymptomatic clinical herpes vaccine: the old, the new, and the unknown". Clin. Dev. Immunol. 2012: 187585. doi:10.1155/2012/187585. PMC 3324142Freely accessible. PMID 22548113.
  2. Whitley, Richard J.; Roizman, Bernard (15 July 2002). "Herpes simplex viruses: is a vaccine tenable?". J Clin Invest. 110 (2): 145–151. doi:10.1172/JCI16126. PMC 151069Freely accessible. PMID 12122103.
  3. "Introducing RVx". 2016-03-12. Retrieved 2016-08-02.
  4. "Herpes Vaccine Research". Herpes Vaccine Research. Retrieved 2016-08-02.
  5. "Knipe Lab | Harvard Medical School". knipelab.med.harvard.edu. Retrieved 2016-08-02.
  6. "Admedus US Research & Development - Admedus US". www.admedus.com. Retrieved 2016-08-02.
  7. "Biological Efficacy Study of HerpV Vaccine With QS-21 to Treat Subjects With Recurrent Genital Herpes". Retrieved 31 August 2016.
  8. "GV2207 – HSV-2 Immunotherapeutic :: GenVec, Inc. (GNVC)". www.genvec.com. Retrieved 2016-08-16.
  9. "GV2207 – HSV-2 Immunotherapeutic :: GenVec, Inc. (GNVC)". www.genvec.com. Retrieved 2016-08-16.
  10. 1 2 "Herpes Simplex". MyMetics. Retrieved 2016-08-03.
  11. "Nanobio - HSV-2 Vaccine". Retrieved 2 August 2016.
  12. Corporation, NanoBio. "NanoBio's Genital Herpes Vaccine Demonstrates Efficacy In Guinea Pigs As Both A Prophylactic And A Therapeutic Vaccine". Retrieved 2 August 2016.
  13. "PBS Vax™ Therapeutic Vaccines". profectusbiosciences.com. Retrieved 15 August 2016.
  14. "Status of Vaccine Research and Development of Vaccines for Herpes Simplex Virus" (PDF). Retrieved 30 August 2016.
  15. "QUESTIONS AND ANSWERS The Herpevac Trial for Women". Retrieved 30 August 2016.
  16. "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.
  17. "VARIVAX® Varicella Virus Vaccine Live" (PDF). Retrieved 30 August 2016.
  18. "VARILRIX® Live attenuated varicella vaccine" (PDF). Retrieved 23 August 2016.
  19. "Vaccine Detail GSK1437173A". Retrieved 31 August 2016.
  20. "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.
  21. "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.
  22. Halford WP, Püschel R, Gershburg E, Wilber A, Gershburg S, Rakowski B (2011). "A live-attenuated HSV-2 ICP0 virus elicits 10 to 100 times greater protection against genital herpes than a glycoprotein D subunit vaccine". PLoS ONE. 6 (3): e17748. doi:10.1371/journal.pone.0017748. PMC 3055896Freely accessible. PMID 21412438.
  23. Halford WP, Geltz J, Gershburg E. "Pan-HSV-2 IgG Antibody in Vaccinated Mice and Guinea Pigs Correlates with Protection against Herpes Simplex Virus 2". PLoS ONE. 8 (6): e65523. doi:10.1371/journal.pone.0065523. Retrieved 9 March 2014.
  24. Halford, William P.; Püschel, Ringo; Rakowski, Brandon (17 August 2010). "Herpes Simplex Virus 2 ICP0 − Mutant Viruses Are Avirulent and Immunogenic: Implications for a Genital Herpes Vaccine". PLOS ONE. 5 (8): e12251. doi:10.1371/journal.pone.0012251. PMC 2923193Freely accessible. PMID 20808928. Retrieved 2 August 2016 via PLoS Journals.
  25. Royer, Derek J.; Gurung, Hem R.; Jinkins, Jeremy K.; Geltz, Joshua J.; Wu, Jennifer L.; Halford, William P.; Carr, Daniel J. J. (1 June 2016). "A Highly Efficacious Herpes Simplex Virus 1 Vaccine Blocks Viral Pathogenesis and Prevents Corneal Immunopathology via Humoral Immunity". J. Virol. 90 (11): 5514–5529. doi:10.1128/JVI.00517-16. PMC 4934733Freely accessible. PMID 27030264. Retrieved 2 August 2016 via jvi.asm.org.
  26. "High-purity preparation of HSV-2 vaccine candidate ACAM529 is immunogenic and efficacious in vivo". PLoS One. Retrieved May 20, 2014.
  27. "Immunization with a replication-defective herpes simplex virus 2 mutant reduces herpes simplex virus 1 infection and prevents ocular disease" (PDF). ScienceDirect. Retrieved May 20, 2014.
  28. "NIH Launches Trial of Investigational Genital Herpes Vaccine". NIAID. Retrieved 17 September 2014.
  29. "Comparative Efficacy and Immunogenicity of Replication-Defective, Recombinant Glycoprotein, and DNA Vaccines for Herpes Simplex Virus 2 Infections in Mice and Guinea Pigs" (PDF). JOURNAL OF VIROLOGY. Retrieved May 20, 2014.
  30. "FOCUS | March 7, 2008 | LICENSING: Herpes Vaccine Developed at HMS Licensed for Preclinical Trials".
  31. "Sanofi Pasteur and Immune Design Enter Broad Collaboration for the Development of a Herpes Simplex Virus Therapy". Retrieved 17 October 2014.
  32. "Potential Cure For Herpes Simplex Virus". HSV Outbreak. Retrieved 19 August 2014.
  33. "Gardasil inventor Professor Ian Frazer's new genital herpes vaccine proves safe in passing first human trials". Retrieved 21 February 2014.
  34. "Admedus vaccine for herpes has success; moves to next clinical trial". Proactive Investors. Retrieved 3 October 2014.
  35. "Admedus increases interest in Prof Ian Frazer's vaccines". Proactive Investors. Retrieved 3 September 2014.
  36. "Admedus has high hopes for Herpes vaccine as dosing commences - Proactiveinvestors (AU)". Retrieved 2 August 2016.
  37. "Interim HSV-2 Clinical Trial Data Encouraging" (PDF). www.admedus.com - Announcements. Admedus Ltd.
  38. "Interim HSV-2 Phase II clinical trial data encouraging" (PDF). www.admedus.com - ASX Announcements. Admedus Ltd.
  39. "Genocea Corporate Overview" (PDF). Genocea. Retrieved 5 March 2016.
  40. "Herpes simplex type 2 virus deleted in glycoprotein D protects against vaginal, skin and neural disease". eLife Science. Retrieved 12 March 2015.
  41. "Radical Vaccine Design Effective Against Herpes Virus". HHMI.org. Retrieved 12 March 2015.
  42. "NanoBio's Genital Herpes Vaccine Demonstrates Efficacy In Guinea Pigs As Both A Prophylactic And A Therapeutic Vaccine". NanoBio Corporation. Retrieved 2 October 2015.
  43. "Pipeline for PBS Vax™ Therapeutic Vaccines". Profectus Biosciences. Retrieved 12 December 2015.
  44. "BioMedical Research Models Inc awarded Grant for Mucosal HSV-2 vaccine". Vaccine Nation (Cameron Bisset). Retrieved 13 June 2014.
  45. "Project Information - NIH RePORTER - NIH Research Portfolio Online Reporting Tools Expenditures and Results". Retrieved 2 August 2016.
  46. "Agenus Vaccine Shows Significant Reduction in Viral Burden after HerpV Generated Immune Activation". Business Wire. Retrieved 10 September 2014.
  47. "Herpes News: Early 2014 Roundup - Just Herpes". Retrieved 2 August 2016.
  48. Agenus Pipeline
  49. "Mymetics HSV Vaccine Candidate". Mymetics. Retrieved 22 April 2016.
  50. "Vical (VICL) Genital Herpes Vaccine Phase 1/2 Missed Primary Endpoint". StreetInsider.com. Retrieved 22 June 2015.
  51. "Vical Reports Top-Line Results From Phase 1/2 Trial of Therapeutic Genital Herpes Vaccine". FierceMarkets. 22 June 2015. Retrieved 23 June 2015.
  52. 1 2 "Herpevac Trial for Women". Archived from the original on 2007-10-20. Retrieved 2008-03-04.
  53. Jon Cohen (15 October 2010). "aPainful Failure of Promising Genital Herpes Vaccine". Science. 330 (6002): 304. doi:10.1126/science.330.6002.304. PMID 20947733.
  54. "PaxVax Signs R&D Collaboration with UC San Diego to Develop a Vaccine to Prevent Herpes Simplex Virus Infections - PaxVax - Socially Responsible Vaccines". Retrieved 2 August 2016.
  55. "The PaxVax Platform - Product Pipeline". Retrieved 5 September 2016.
  56. "BioVex commences dosing in Phase 1 study of ImmunoVEX live attenuated genital herpes vaccine". 5 March 2010. Retrieved 2 August 2016.
  57. "Amgen completes acquisition of BioVex Group". 4 March 2011. Retrieved 2 August 2016 via The Boston Globe.
  58. "Amgen Pipeline". Retrieved 2 August 2016.
  59. "AuRx, Inc". Retrieved 2 August 2016.
  60. Helicase–primase inhibitors as the potential next generation of highly active drugs against herpes simplex viruses
  61. "AiCuris - R&D Pipeline". www.aicuris.com. Retrieved 2016-09-16.
  62. "Effect of ASP2151, a herpesvirus helicase-primase inhibitor, in a guinea pig model of genital herpes.". Retrieved 30 August 2016.
  63. Molecular Therapy (2006-05-01). "Molecular Therapy – Abstract of article: 801. RNA Gene Therapy Targeting Herpes Simplex Virus". Nature.com. Retrieved 2011-04-12.
  64. "University of Florida News –Potential new herpes therapy studied". News.ufl.edu. 2009-02-03. Retrieved 2011-04-12.
  65. Kassabian, Sara. "CRISPR Puts Up a Fight Against Persistent Herpesviruses: A Short Animation". Speaking of Medicine - PLOS Community Blog. PLOS.org. Retrieved 4 August 2016.
  66. van Diemen, FR; et al. "Using CRISPR to combat viral infections: a new way to treat herpes?". PLOS Media YouTube Channel. PLOS.org. Retrieved 4 August 2016.
  67. Fox, Maggie (2008-07-02). "New approach offers chance to finally kill herpes". Reuters. Retrieved 2011-04-12.
  68. Kingsbury, Kathleen (2008-07-02). "A Cure for Cold Sores?". Time. Retrieved 2010-05-04.
  69. 1 2 Martin, Trepel; Charlotte A. Stoneham; Hariklia Eleftherohorinou; Nicholas D. Mazarakis; Renata Pasqualini; Wadih Arap; Amin Hajitou (August 2009). "A Heterotypic Bystander Effect for Tumor Cell Killing after AAVPmediated Vascular-targeted Suicide Gene Transfer". Mol Cancer Ther. 8 (8): 2383–2391. doi:10.1158/1535-7163.MCT-09-0110. PMC 2871293Freely accessible. PMID 19671758.
  70. 1 2 3 Mesnil, M.; Piccoli, C.; Tiraby, G.; Willencke, K.; Yamasaki, H. (March 1996). "Bystander killing of cancer cells by herpes simplex virus thymidine kinase gene is mediated by connexins". Proceedings of the National Academy of Sciences of the United States of America. 93 (5): 1831–1835. doi:10.1073/pnas.93.5.1831. PMC 39867Freely accessible. PMID 8700844.
  71. Stringer, J.R; LOUIS E. HOLLAND; RONALD I. SWANSTROM; 1 KENNETH PIVO; AND EDWARD K. WAGNER (March 1977). "Quantitation of Herpes Simplex Virus Type 1 RNA in Infected HeLa Cells". Journal of Virology. 21 (3): 889–901. PMC 515626Freely accessible. PMID 191652.
  72. Jovanovic, M; MO Hengartner (2006). "miRNAs and apoptosis: RNAs to die for". Nature. 25 (46): 6176–6187. doi:10.1038/sj.onc.1209912. Retrieved 6 April 2012.
This article is issued from Wikipedia - version of the 11/15/2016. The text is available under the Creative Commons Attribution/Share Alike but additional terms may apply for the media files.