New Drug Approvals

Home » vaccine

Category Archives: vaccine

DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO .....FOR BLOG HOME CLICK HERE

Blog Stats

  • 4,303,324 hits

Flag and hits

Flag Counter

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 2,821 other subscribers
Follow New Drug Approvals on WordPress.com

Archives

Categories

Recent Posts

Flag Counter

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 2,821 other subscribers
DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO, Born in Mumbai in 1964 and graduated from Mumbai University, Completed his Ph.D from ICT, 1991,Matunga, Mumbai, India, in Organic Chemistry, The thesis topic was Synthesis of Novel Pyrethroid Analogues, Currently he is working with AFRICURE PHARMA, ROW2TECH, NIPER-G, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India as ADVISOR, earlier assignment was with GLENMARK LIFE SCIENCES LTD, as CONSUlTANT, Retired from GLENMARK in Jan2022 Research Centre as Principal Scientist, Process Research (bulk actives) at Mahape, Navi Mumbai, India. Total Industry exp 32 plus yrs, Prior to joining Glenmark, he has worked with major multinationals like Hoechst Marion Roussel, now Sanofi, Searle India Ltd, now RPG lifesciences, etc. He has worked with notable scientists like Dr K Nagarajan, Dr Ralph Stapel, Prof S Seshadri, etc, He did custom synthesis for major multinationals in his career like BASF, Novartis, Sanofi, etc., He has worked in Discovery, Natural products, Bulk drugs, Generics, Intermediates, Fine chemicals, Neutraceuticals, GMP, Scaleups, etc, he is now helping millions, has 9 million plus hits on Google on all Organic chemistry websites. His friends call him Open superstar worlddrugtracker. His New Drug Approvals, Green Chemistry International, All about drugs, Eurekamoments, Organic spectroscopy international, etc in organic chemistry are some most read blogs He has hands on experience in initiation and developing novel routes for drug molecules and implementation them on commercial scale over a 32 PLUS year tenure till date Feb 2023, Around 35 plus products in his career. He has good knowledge of IPM, GMP, Regulatory aspects, he has several International patents published worldwide . He has good proficiency in Technology transfer, Spectroscopy, Stereochemistry, Synthesis, Polymorphism etc., He suffered a paralytic stroke/ Acute Transverse mylitis in Dec 2007 and is 90 %Paralysed, He is bound to a wheelchair, this seems to have injected feul in him to help chemists all around the world, he is more active than before and is pushing boundaries, He has 100 million plus hits on Google, 2.5 lakh plus connections on all networking sites, 100 Lakh plus views on dozen plus blogs, 227 countries, 7 continents, He makes himself available to all, contact him on +91 9323115463, email amcrasto@gmail.com, Twitter, @amcrasto , He lives and will die for his family, 90% paralysis cannot kill his soul., Notably he has 38 lakh plus views on New Drug Approvals Blog in 227 countries......https://newdrugapprovals.wordpress.com/ , He appreciates the help he gets from one and all, Friends, Family, Glenmark, Readers, Wellwishers, Doctors, Drug authorities, His Contacts, Physiotherapist, etc He has total of 32 International and Indian awards

Personal Links

Verified Services

View Full Profile →

Archives

Categories

Flag Counter

RTS,S/AS01, RTS,S Mosquirix


 The World Health Organization (WHO) has announced that the Government of Malawi has immunized the first children with RTS,S/AS01 (RTS,S), the world’s first malaria vaccine, according to the World Record Academy.

Sequence:

1MMAPDPNANP NANPNANPNA NPNANPNANP NANPNANPNA NPNANPNANP51NANPNANPNA NPNANPNANP NANPNANPNA NPNKNNQGNG QGHNMPNDPN101RNVDENANAN NAVKNNNNEE PSDKHIEQYL KKIKNSISTE WSPCSVTCGN151GIQVRIKPGS ANKPKDELDY ENDIEKKICK MEKCSSVFNV VNSRPVTNME201NITSGFLGPL LVLQAGFFLL TRILTIPQSL DSWWTSLNFL GGSPVCLGQN251SQSPTSNHSP TSCPPICPGY RWMCLRRFII FLFILLLCLI FLLVLLDYQG301MLPVCPLIPG STTTNTGPCK TCTTPAQGNS MFPSCCCTKP TDGNCTCIPI351PSSWAFAKYL WEWASVRFSW LSLLVPFVQW FVGLSPTVWL SAIWMMWYWG401PSLYSIVSPF IPLLPIFFCL WVYI

RTS,S/AS01 (RTS,S)

RTS,S/AS01, Mosquirix

Cas 149121-47-1

203-400-Antigen CS (Plasmodium falciparum strain NF54 reduced), 203-L-methionine-204-L-methionine-205-L-alanine-206-L-proline-207-L-aspartic acid-210-L-alanine-211-L-asparagine-313-L-asparagine-329-L-glutamic acid-330-L-glutamine-333-L-lysine-336-L-lysine-339-L-isoleucine-373-L-glutamic acid-396-L-arginine-397-L-proline-398-L-valine-399-L-threonine-400-L-asparagine-, (400→1′)-protein with antigen (hepatitis B virus subtype adw small surface reduced) (9CI) 

Other Names

  • Malaria vaccine RTS,S
  • Mosquirix
  • RTS,S

Protein Sequence

Sequence Length: 424

An external file that holds a picture, illustration, etc. Object name is khvi-16-03-1669415-g002.jpg

Figure 2.

Graphical depiction of circumsporozoite (CSP) and RTS,S structures. CSP comprises an N-terminal region containing a signal peptide sequence and Region I that binds heparin sulfate proteoglycans and has embedded within it a conserved five amino acid (KLKQP) proteolytic cleavage site sequence; a central region containing four-amino acid (NANP/NVDP) repeats; and a C-terminal region containing Region II [a thrombospondin (TSP)-like domain] and a canonical glycosylphosphatidylinositol (GPI) anchor addition sequence. The region of the CSP included in the RTS,S vaccine includes the last 18 NANP repeats and C-terminus exclusive of the GPI anchor addition sequence. Hepatitis B virus surface antigen (HBsAg) monomers self-assemble into virus-like particles and approximately 25% of the HBsAg monomers in RTS,S are genetically fused to the truncated CSP and serve as protein carriers. The CSP fragment in RTS,S contains three known T-cell epitopes: a highly variable CD4 + T-cell epitope before the TSP-like domain (TH2R), a highly variable CD8 + T-cell epitope within the TSP-like domain (TH3R), and a conserved “universal” CD4 + T cell epitope (CS.T3) at the C-terminus. (Figure courtesy of a recent publication16 and open access,
PATENTWO 2009080715

https://patents.google.com/patent/WO2009080715A2/tr

XAMPLES

Example 1Recipe for component for a single pediatric dose of RTS, S malaria vaccine (2 vial formulation)Component AmountRTS,S 25μgNaCl 2.25mgPhosphate buffer (NaZK2) 1OmMMonothioglycerol 125μgWater for Injection Make volume to 250 μLThe above is prepared by adding RTS, S antigen to a mix of Water for Injection, NaCl 150OmM, phosphate buffer (NaZK2) 50OmM (pH 6.8 when diluted x 50) and an aqueous solution of monothioglycerol at 10%. Finally pH is adjusted to 7.0 ± 0.1.This may be provided as a vial together with a separate vial of adjuvant, for example a liposomal formulation of MPL and QS21Component Amount l,2-di-oleoyl-5/?-glycero-3-phosphocholine (DOPC) 500 μgCholesterol 125 μgMPL 25 μgQS21 25 μgNaCl 2.25mg Phosphate buffer (NaZK2) 1 OmMWater for Injection Make volume to250 μLFor administration the adjuvant formulation is added to the component formulation, for example using a syringe, and then shaken. Then the dose is administered in the usual way. The pH of the final liquid formulation is about 6.6 +/- 0.1.Example IAA final pediatric liquid formulation (1 vial) according to the invention may be prepared according to the following recipe.Component AmountRTS,S 25μgNaCl 4.5mgPhosphate buffer (NaZK2) 1OmMMonothioglycerol 125μg1 ,2-di-oleoyl-5/?-glycero-3-phosphocholine (DOPC) 500 μgCholesterol 125 μgMPL 25 μgQS21 25 μgWater for Injection Make volume to500 μLThe pH of the above liquid formulation is either adjusted to 7.0 +/- 0.1 (which is favorable for antigen stability, but not favorable at all for the MPL stability), or to 6.1 +/- 0.1 (which is favorable for MPL stability, but not favorable at all for RT S, S stability). Therefore this formulation is intended for rapid use after preparation.The above is prepared by adding RTS, S antigen to a mix of Water for Injection, NaCl 150OmM, phosphate buffer (NaZK2) 50OmM (pH 6.8 when diluted x 50) and an aqueous solution of monothioglycerol at 10%. Then a premix of liposomes containing MPL with QS21 is added, and finally pH is adjusted. Example IBA final adult dose (1 vial formulation) for the RTS, S according to the invention may be prepared as follows:Component AmountRTS,S 50μgNaCl 4.5mgPhosphate buffer (NaZK2) 1OmMMonothioglycerol 250μg1 ,2-di-oleoyl-5/?-glycero-3-phosphocholine (DOPC) 1000 μgCholesterol 250 μgMPL 50 μgQS21 50 μgWater for Injection Make volume to500 μLExample 1CExample 1C may prepared by putting Example 1, IA or IB in an amber vial, for example flushed with nitrogen before filing.

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter 

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

/////////////////////////////////////////////////////////////////////////////////////////////////////

WHO recommends groundbreaking malaria vaccine for children at risk

Historic RTS,S/AS01 recommendation can reinvigorate the fight against malaria6 October 2021https://www.who.int/news/item/06-10-2021-who-recommends-groundbreaking-malaria-vaccine-for-children-at-risk

The World Health Organization (WHO) is recommending widespread use of the RTS,S/AS01 (RTS,S) malaria vaccine among children in sub-Saharan Africa and in other regions with moderate to high P. falciparum malaria transmission. The recommendation is based on results from an ongoing pilot programme in Ghana, Kenya and Malawi that has reached more than 800 000 children since 2019.

“This is a historic moment. The long-awaited malaria vaccine for children is a breakthrough for science, child health and malaria control,” said WHO Director-General Dr Tedros Adhanom Ghebreyesus. “Using this vaccine on top of existing  tools to prevent malaria could save tens of thousands of young lives each year.”

Malaria remains a primary cause of childhood illness and death in sub-Saharan Africa. More than 260 000 African children under the age of five die from malaria annually.

In recent years, WHO and its partners have been reporting a stagnation in progress against the deadly disease.

“For centuries, malaria has stalked sub-Saharan Africa, causing immense personal suffering,” said Dr Matshidiso Moeti, WHO Regional Director for Africa. “We have long hoped for an effective malaria vaccine and now for the first time ever, we have such a vaccine recommended for widespread use. Today’s recommendation offers a glimmer of hope for the continent which shoulders the heaviest burden of the disease and we expect many more African children to be protected from malaria and grow into healthy adults.”

WHO recommendation for the RTS,S malaria vaccine

Based on the advice of two WHO global advisory bodies, one for immunization and the other for malaria, the Organization recommends that:

WHO recommends that in the context of comprehensive malaria control the RTS,S/AS01 malaria vaccine be used for the prevention of P. falciparum malaria in children living in regions with moderate to high transmission as defined by WHO.  RTS,S/AS01 malaria vaccine should be provided in a schedule of 4 doses in children from 5 months of age for the reduction of malaria disease and burden.

Summary of key findings of the malaria vaccine pilots

Key findings of the pilots informed the recommendation based on data and insights generated from two years of vaccination in child health clinics in the three pilot countries, implemented under the leadership of the Ministries of Health of Ghana, Kenya and Malawi. Findings include:

  • Feasible to deliver: Vaccine introduction is feasible, improves health and saves lives, with good and equitable coverage of RTS,S seen through routine immunization systems. This occurred even in the context of the COVID-19 pandemic.
  • Reaching the unreached: RTS,S increases equity in access to malaria prevention.
    • Data from the pilot programme showed that more than two-thirds of children in the 3 countries who are not sleeping under a bednet are benefitting from the RTS,S vaccine.
    • Layering the tools results in over 90% of children benefitting from at least one preventive intervention (insecticide treated bednets or the malaria vaccine).
  • Strong safety profile: To date, more than 2.3 million doses of the vaccine have been administered in 3 African countries – the vaccine has a favorable safety profile.
  • No negative impact on uptake of bednets, other childhood vaccinations, or health seeking behavior for febrile illness. In areas where the vaccine has been introduced, there has been no decrease in the use of insecticide-treated nets, uptake of other childhood vaccinations or health seeking behavior for febrile illness.
  • High impact in real-life childhood vaccination settings: Significant reduction (30%) in deadly severe malaria, even when introduced in areas where insecticide-treated nets are widely used and there is good access to diagnosis and treatment.
  • Highly cost-effective: Modelling estimates that the vaccine is cost effective in areas of moderate to high malaria transmission.

Next steps for the WHO-recommended malaria vaccine will include funding decisions from the global health community for broader rollout, and country decision-making on whether to adopt the vaccine as part of national malaria control strategies.

Financial support

Financing for the pilot programme has been mobilized through an unprecedented collaboration among three key global health funding bodies: Gavi, the Vaccine Alliance; the Global Fund to Fight AIDS, Tuberculosis and Malaria; and Unitaid.

Note to editors:

  • The malaria vaccine, RTS,S, acts against P. falciparum, the most deadly malaria parasite globally, and the most prevalent in Africa.
  • The Malaria Vaccine Implementation Programme is generating evidence and experience on the feasibility, impact and safety of the RTS,S malaria vaccine in real-life, routine settings in selected areas of Ghana, Kenya and Malawi.
  • Pilot malaria vaccine introductions are led by the Ministries of Health of Ghana, Kenya and Malawi.
  • The pilot programme will continue in the 3 pilot countries to understand the added value of the 4th vaccine dose, and to measure longer-term impact on child deaths.
  • The Malaria Vaccine Implementation Programme is coordinated by WHO and supported by in-country and international partners, including PATH, UNICEF and GSK, which is donating up to 10 million doses of the vaccine for the pilot.
  • The RTS,S malaria vaccine is the result of 30 years of research and development by GSK and through a partnership with PATH, with support from a network of African research centres.
  • The Bill & Melinda Gates Foundation provided catalytic funding for late-stage development of RTS,S between 2001 and 2015.

RTS,S/AS01 (trade name Mosquirix) is a recombinant protein-based malaria vaccine. In October 2021, the vaccine was endorsed by the World Health Organization (WHO) for “broad use” in children, making it the first malaria vaccine candidate, and first vaccine to address parasitic infection, to receive this recommendation.[3][4][5]

The RTS,S vaccine was conceived of and created in the late 1980s by scientists working at SmithKline Beecham Biologicals (now GlaxoSmithKline (GSK) Vaccines) laboratories in Belgium.[6] The vaccine was further developed through a collaboration between GSK and the Walter Reed Army Institute of Research in the U.S. state of Maryland[7] and has been funded in part by the PATH Malaria Vaccine Initiative and the Bill and Melinda Gates Foundation. Its efficacy ranges from 26 to 50% in infants and young children.

Approved for use by the European Medicines Agency (EMA) in July 2015,[1] it is the world’s first licensed malaria vaccine and also the first vaccine licensed for use against a human parasitic disease of any kind.[8] On 23 October 2015, WHO’s Strategic Advisory Group of Experts on Immunization (SAGE) and the Malaria Policy Advisory Committee (MPAC) jointly recommended a pilot implementation of the vaccine in Africa.[9] This pilot project for vaccination was launched on 23 April 2019 in Malawi, on 30 April 2019 in Ghana, and on 13 September 2019 in Kenya.[10][11]

Background

Main article: Malaria vaccine

Potential malaria vaccines have been an intense area of research since the 1960s.[12] SPf66 was tested extensively in endemic areas in the 1990s, but clinical trials showed it to be insufficiently effective.[13] Other vaccine candidates, targeting the blood-stage of the malaria parasite’s life cycle, have also been insufficient on their own.[14] Among several potential vaccines under development that target the pre-erythrocytic stage of the disease, RTS,S has shown the most promising results so far.[15]

Approval history

The EMA approved the RTS,S vaccine in July 2015, with a recommendation that it be used in Africa for babies at risk of getting malaria. RTS,S was the world’s first malaria vaccine to get approval for this use.[16][8] Preliminary research suggests that delayed fractional dosing could increase the vaccine’s efficacy up to 86%.[17][18]

On 17 November 2016, WHO announced that the RTS,S vaccine would be rolled out in pilot projects in three countries in sub-Saharan Africa. The pilot program, coordinated by WHO, will assess the extent to which the vaccine’s protective effect shown in advanced clinical trials can be replicated in real-life settings. Specifically, the programme will evaluate the feasibility of delivering the required four doses of the vaccine; the impact of the vaccine on lives saved; and the safety of the vaccine in the context of routine use.[19]

Vaccinations by the ministries of health of Malawi, Ghana, and Kenya began in April and September 2019 and target 360,000 children per year in areas where vaccination would have the highest impact. The results are planned to be used by the World Health Organization to advise about a possible future deployment of the vaccine.[10][11][20] In 2021 it was reported that the vaccine together with other anti-malaria medication when given at the most vulnerable season could reduce deaths and illness from the disease by 70%.[21][22]

Funding

RTS,S has been funded, most recently, by the non-profit PATH Malaria Vaccine Initiative (MVI) and GlaxoSmithKline with funding from the Bill and Melinda Gates Foundation.[23] The RTS,S-based vaccine formulation had previously been demonstrated to be safe, well tolerated, immunogenic, and to potentially confer partial efficacy in both malaria-naive and malaria-experienced adults as well as children.[24]

Components and mechanism

 

The RTS,S vaccine is based on a protein construct first developed by GlaxoSmithKline in 1986. It was named RTS because it was engineered using genes from the repeat (‘R’) and T-cell epitope (‘T’) of the pre-erythrocytic circumsporozoite protein (CSP) of the Plasmodium falciparum malaria parasite together with a viral surface antigen (‘S’) of the hepatitis B virus (HBsAg).[7] This protein was then mixed with additional HBsAg to improve purification, hence the extra “S”.[7] Together, these two protein components assemble into soluble virus-like particles similar to the outer shell of a hepatitis B virus.[25]

A chemical adjuvant (AS01, specifically AS01E) was added to increase the immune system response.[26] Infection is prevented by inducing humoral and cellular immunity, with high antibody titers, that block the parasite from infecting the liver.[27]

The T-cell epitope of CSP is O-fucosylated in Plasmodium falciparum[28][29] and Plasmodium vivax,[30] while the RTS,S vaccine produced in yeast is not.

References

  1. Jump up to:a b “Mosquirix H-W-2300”European Medicines Agency (EMA). Retrieved 4 March 2021.
  2. ^ “RTS,S Malaria Vaccine: 2019 Partnership Award Honoree”YouTube. Global Health Technologies Coalition. Retrieved 6 October 2021.
  3. ^ Davies L (6 October 2021). “WHO endorses use of world’s first malaria vaccine in Africa”The Guardian. Retrieved 6 October2021.
  4. ^ Drysdale C, Kelleher K. “WHO recommends groundbreaking malaria vaccine for children at risk” (Press release). Geneva: World Health Organization. Retrieved 6 October 2021.
  5. ^ Mandavilli A (6 October 2021). “A ‘Historical Event’: First Malaria Vaccine Approved by W.H.O.” New York Times. Retrieved 6 October 2021.
  6. ^ “HYBRID PROTEIN BETWEEN CS FROM PLASMODIUM AND HBsAG”.
  7. Jump up to:a b c Heppner DG, Kester KE, Ockenhouse CF, Tornieporth N, Ofori O, Lyon JA, et al. (March 2005). “Towards an RTS,S-based, multi-stage, multi-antigen vaccine against falciparum malaria: progress at the Walter Reed Army Institute of Research”Vaccine23 (17–18): 2243–50. doi:10.1016/j.vaccine.2005.01.142PMID 15755604Archived from the original on 23 July 2018.
  8. Jump up to:a b Walsh F (24 July 2015). “Malaria vaccine gets ‘green light'”BBC NewsArchived from the original on 21 July 2020. Retrieved 25 July 2015.
  9. ^ Stewart S (23 October 2015). “Pilot implementation of first malaria vaccine recommended by WHO advisory groups” (Press release). Geneva: World Health OrganizationArchived from the original on 19 September 2021.
  10. Jump up to:a b Alonso P (19 June 2019). “Letter to partners – June 2019”(Press release). Wuxi: World Health Organization. Retrieved 22 October 2019.
  11. Jump up to:a b “Malaria vaccine launched in Kenya: Kenya joins Ghana and Malawi to roll out landmark vaccine in pilot introduction” (Press release). Homa Bay: World Health Organization. 13 September 2019. Retrieved 22 October 2019.
  12. ^ Hill AV (October 2011). “Vaccines against malaria”Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences366 (1579): 2806–14. doi:10.1098/rstb.2011.0091PMC 3146776PMID 21893544.
  13. ^ Graves P, Gelband H (April 2006). Graves PM (ed.). “Vaccines for preventing malaria (SPf66)”The Cochrane Database of Systematic Reviews (2): CD005966. doi:10.1002/14651858.CD005966PMC 6532709PMID 16625647.
  14. ^ Graves P, Gelband H (October 2006). Graves PM (ed.). “Vaccines for preventing malaria (blood-stage)”The Cochrane Database of Systematic Reviews (4): CD006199. doi:10.1002/14651858.CD006199PMC 6532641PMID 17054281.
  15. ^ Graves P, Gelband H (October 2006). Graves PM (ed.). “Vaccines for preventing malaria (pre-erythrocytic)”The Cochrane Database of Systematic Reviews (4): CD006198. doi:10.1002/14651858.CD006198PMC 6532586PMID 17054280.
  16. ^ “First malaria vaccine receives positive scientific opinion from EMA”European Medicines Agency. 24 July 2015. Retrieved 24 July 2015.
  17. ^ Birkett A (16 September 2016). “A vaccine for malaria elimination?”PATH.
  18. ^ Regules JA, Cicatelli SB, Bennett JW, Paolino KM, Twomey PS, Moon JE, et al. (September 2016). “Fractional Third and Fourth Dose of RTS,S/AS01 Malaria Candidate Vaccine: A Phase 2a Controlled Human Malaria Parasite Infection and Immunogenicity Study”The Journal of Infectious Diseases214 (5): 762–71. doi:10.1093/infdis/jiw237PMID 27296848.
  19. ^ “Malaria: The malaria vaccine implementation programme (MVIP)”.
  20. ^ “WHO | MVIP countries: Ghana, Kenya and Malawi”.
  21. ^ Chandramohan D, Zongo I, Sagara I, Cairns M, Yerbanga RS, Diarra M, et al. (September 2021). “Seasonal Malaria Vaccination with or without Seasonal Malaria Chemoprevention”The New England Journal of Medicine385 (11): 1005–1017. doi:10.1056/NEJMoa2026330PMID 34432975.
  22. ^ Roxby P (26 August 2021). “Trial suggests malaria sickness could be cut by 70%”BBC NewsArchived from the original on 3 October 2021. Retrieved 26 August 2021.
  23. ^ Stein R (18 October 2011). “Experimental malaria vaccine protects many children, study shows”Washington Post.
  24. ^ Regules JA, Cummings JF, Ockenhouse CF (May 2011). “The RTS,S vaccine candidate for malaria”Expert Review of Vaccines10 (5): 589–99. doi:10.1586/erv.11.57PMID 21604980S2CID 20443829.
  25. ^ Rutgers T, Gordon D, Gathoye AM, Hollingdale M, Hockmeyer W, Rosenberg M, De Wilde M (September 1988). “Hepatitis B Surface Antigen as Carrier Matrix for the Repetitive Epitope of the Circumsporozoite Protein of Plasmodium Falciparum”Nature Biotechnology6 (9): 1065–1070. doi:10.1038/nbt0988-1065S2CID 39880644.
  26. ^ RTS,S Clinical Trials Partnership (July 2015). “Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial”Lancet386 (9988): 31–45. doi:10.1016/S0140-6736(15)60721-8PMC 5626001PMID 25913272.
  27. ^ Foquet L, Hermsen CC, van Gemert GJ, Van Braeckel E, Weening KE, Sauerwein R, et al. (January 2014). “Vaccine-induced monoclonal antibodies targeting circumsporozoite protein prevent Plasmodium falciparum infection”The Journal of Clinical Investigation124 (1): 140–4. doi:10.1172/JCI70349PMC 3871238PMID 24292709.
  28. ^ Swearingen KE, Lindner SE, Shi L, Shears MJ, Harupa A, Hopp CS, et al. (April 2016). “Interrogating the Plasmodium Sporozoite Surface: Identification of Surface-Exposed Proteins and Demonstration of Glycosylation on CSP and TRAP by Mass Spectrometry-Based Proteomics”PLOS Pathogens12 (4): e1005606. doi:10.1371/journal.ppat.1005606PMC 4851412PMID 27128092.
  29. ^ Lopaticki S, Yang AS, John A, Scott NE, Lingford JP, O’Neill MT, et al. (September 2017). “Protein O-fucosylation in Plasmodium falciparum ensures efficient infection of mosquito and vertebrate hosts”Nature Communications8 (1): 561. Bibcode:2017NatCo…8..561Ldoi:10.1038/s41467-017-00571-yPMC 5601480PMID 28916755.
  30. ^ Swearingen KE, Lindner SE, Flannery EL, Vaughan AM, Morrison RD, Patrapuvich R, et al. (July 2017). “Proteogenomic analysis of the total and surface-exposed proteomes of Plasmodium vivax salivary gland sporozoites”PLOS Neglected Tropical Diseases11 (7): e0005791. doi:10.1371/journal.pntd.0005791PMC 5552340PMID 28759593.

Further reading

  • Wilby KJ, Lau TT, Gilchrist SE, Ensom MH (March 2012). “Mosquirix (RTS,S): a novel vaccine for the prevention of Plasmodium falciparum malaria”. The Annals of Pharmacotherapy46 (3): 384–93. doi:10.1345/aph.1Q634PMID 22408046.
  • Asante KP, Abdulla S, Agnandji S, Lyimo J, Vekemans J, Soulanoudjingar S, et al. (October 2011). “Safety and efficacy of the RTS,S/AS01E candidate malaria vaccine given with expanded-programme-on-immunisation vaccines: 19 month follow-up of a randomised, open-label, phase 2 trial”. The Lancet. Infectious Diseases11 (10): 741–9. doi:10.1016/S1473-3099(11)70100-1PMID 21782519.

External links

Vaccine description
TargetP. falciparum; to a lesser extent Hepatitis B
Vaccine typeProtein subunit
Clinical data
Trade namesMosquirix
Routes of
administration
intramuscular injection (0.5 mL)[1]
Legal status
Legal statusIn general: ℞ (Prescription only)

A poster advertising trials of the RTS,S vaccine[2]

malaria vaccine is a vaccine that is used to prevent malaria. The only approved vaccine as of 2021, is RTS,S, known by the brand name Mosquirix.[1] It requires four injections.[1]

Research continues with other malaria vaccines. The most effective malaria vaccine is R21/Matrix-M, with a 77% efficacy rate shown in initial trials, and significantly higher antibody levels than with the RTS,S vaccine.[2] It is the first vaccine that meets the World Health Organization‘s (WHO) goal of a malaria vaccine with at least 75% efficacy.[3][2]

Approved vaccines

RTS,S

Main article: RTS,S

RTS,S (developed by PATH Malaria Vaccine Initiative (MVI) and GlaxoSmithKline (GSK) with support from the Bill and Melinda Gates Foundation) is the most recently developed recombinant vaccine. It consists of the P. falciparum circumsporozoite protein (CSP) from the pre-erythrocytic stage. The CSP antigen causes the production of antibodies capable of preventing the invasion of hepatocytes and additionally elicits a cellular response enabling the destruction of infected hepatocytes. The CSP vaccine presented problems in the trial stage, due to its poor immunogenicity. RTS,S attempted to avoid these by fusing the protein with a surface antigen from hepatitis B, hence creating a more potent and immunogenic vaccine. When tested in trials an emulsion of oil in water and the added adjuvants of monophosphoryl A and QS21 (SBAS2), the vaccine gave protective immunity to 7 out of 8 volunteers when challenged with P. falciparum.[4]

RTS,S/AS01 (commercial name Mosquirix),[5] was engineered using genes from the outer protein of P. falciparum malaria parasite and a portion of a hepatitis B virus plus a chemical adjuvant to boost the immune response. Infection is prevented by inducing high antibody titers that block the parasite from infecting the liver.[6] In November 2012, a Phase III trial of RTS,S found that it provided modest protection against both clinical and severe malaria in young infants.[7]

As of October 2013, preliminary results of a Phase III clinical trial indicated that RTS,S/AS01 reduced the number of cases among young children by almost 50 percent and among infants by around 25 percent. The study ended in 2014. The effects of a booster dose were positive, even though overall efficacy seems to wane with time. After four years reductions were 36 percent for children who received three shots and a booster dose. Missing the booster dose reduced the efficacy against severe malaria to a negligible effect. The vaccine was shown to be less effective for infants. Three doses of vaccine plus a booster reduced the risk of clinical episodes by 26 percent over three years, but offered no significant protection against severe malaria.[8]

In a bid to accommodate a larger group and guarantee a sustained availability for the general public, GSK applied for a marketing license with the European Medicines Agency (EMA) in July 2014.[9] GSK treated the project as a non-profit initiative, with most funding coming from the Gates Foundation, a major contributor to malaria eradication.[10]

On 24 July 2015, Mosquirix received a positive opinion from the European Medicines Agency (EMA) on the proposal for the vaccine to be used to vaccinate children aged 6 weeks to 17 months outside the European Union.[11][12][1] A pilot project for vaccination was launched on 23 April 2019, in Malawi, on 30 April 2019, in Ghana, and on 13 September 2019, in Kenya.[13][14]

In October 2021, the vaccine was endorsed by the World Health Organization for “broad use” in children, making it the first malaria vaccine to receive this recommendation.[15][16][17]

Agents under development

A completely effective vaccine is not available for malaria, although several vaccines are under development. Multiple vaccine candidates targeting the blood-stage of the parasite’s life cycle have been insufficient on their own.[18] Several potential vaccines targeting the pre-erythrocytic stage are being developed, with RTS,S the only approved option so far.[19][7]

R21/Matrix-M

The most effective malaria vaccine is R21/Matrix-M, with 77% efficacy shown in initial trials. It is the first vaccine that meets the World Health Organization’s goal of a malaria vaccine with at least 75% efficacy.[3] It was developed through a collaboration involving the University of Oxford, the Kenya Medical Research Institute, the London School of Hygiene & Tropical MedicineNovavax, the Serum Institute of India, and the Institut de Recherche en Sciences de la Santé in NanoroBurkina Faso. The R21 vaccine uses a circumsporozoite protein (CSP) antigen, at a higher proportion than the RTS,S vaccine. It includes the Matrix-M adjuvant that is also utilized in the Novavax COVID-19 vaccine.[20]

A Phase II trial was reported in April 2021, with a vaccine efficacy of 77% and antibody levels significantly higher than with the RTS,S vaccine. A Phase III trial is planned with 4,800 children across four African countries. If the vaccine is approved, over 200 million doses can be manufactured annually by the Serum Institute of India.[2]

Nanoparticle enhancement of RTS,S

In 2015, researchers used a repetitive antigen display technology to engineer a nanoparticle that displayed malaria specific B cell and T cell epitopes. The particle exhibited icosahedral symmetry and carried on its surface up to 60 copies of the RTS,S protein. The researchers claimed that the density of the protein was much higher than the 14% of the GSK vaccine.[21][22]

PfSPZ vaccine

Main article: PfSPZ Vaccine

The PfSPZ vaccine is a candidate malaria vaccine developed by Sanaria using radiation-attenuated sporozoites to elicit an immune response. Clinical trials have been promising, with trials taking place in Africa, Europe, and the US protecting over 80% of volunteers.[23] It has been subject to some criticism regarding the ultimate feasibility of large-scale production and delivery in Africa, since it must be stored in liquid nitrogen.

The PfSPZ vaccine candidate was granted fast track designation by the U.S. Food and Drug Administration in September 2016.[24]

In April 2019, a phase 3 trial in Bioko was announced, scheduled to start in early 2020.[25]

saRNA vaccine against PMIF

A patent was published in February 2021 for a Self-amplifying RNA (saRNA) vaccine that targets the protein PMIF, which is produced by the plasmodium parasite to inhibit the body’s T-cell response. The vaccine has been tested in mice and is described as, “probably the highest level of protection that has been seen in a mouse model” according to Richard Bucala, co-inventor of the vaccine. There are plans for phase one tests in humans later in 2021.[26]

Other developments

  • SPf66 is a synthetic peptide based vaccine developed by Manuel Elkin Patarroyo team in Colombia, and was tested extensively in endemic areas in the 1990s. Clinical trials showed it to be insufficiently effective, with 28% efficacy in South America and minimal or no efficacy in Africa.[27]
  • The CSP (Circum-Sporozoite Protein) was a vaccine developed that initially appeared promising enough to undergo trials. It is also based on the circumsporozoite protein, but additionally has the recombinant (Asn-Ala-Pro15Asn-Val-Asp-Pro)2-Leu-Arg(R32LR) protein covalently bound to a purified Pseudomonas aeruginosa toxin (A9). However at an early stage a complete lack of protective immunity was demonstrated in those inoculated. The study group used in Kenya had an 82% incidence of parasitaemia whilst the control group only had an 89% incidence. The vaccine intended to cause an increased T-lymphocyte response in those exposed, this was also not observed.[citation needed]
  • The NYVAC-Pf7 multi-stage vaccine attempted to use different technology, incorporating seven P.falciparum antigenic genes. These came from a variety of stages during the life cycle. CSP and sporozoite surface protein 2 (called PfSSP2) were derived from the sporozoite phase. The liver stage antigen 1 (LSA1), three from the erythrocytic stage (merozoite surface protein 1, serine repeat antigen and AMA-1) and one sexual stage antigen (the 25-kDa Pfs25) were included. This was first investigated using Rhesus monkeys and produced encouraging results: 4 out of the 7 antigens produced specific antibody responses (CSP, PfSSP2, MSP1 and PFs25). Later trials in humans, despite demonstrating cellular immune responses in over 90% of the subjects, had very poor antibody responses. Despite this following administration of the vaccine some candidates had complete protection when challenged with P.falciparum. This result has warranted ongoing trials.[citation needed]
  • In 1995 a field trial involving [NANP]19-5.1 proved to be very successful. Out of 194 children vaccinated none developed symptomatic malaria in the 12-week follow up period and only 8 failed to have higher levels of antibody present. The vaccine consists of the schizont export protein (5.1) and 19 repeats of the sporozoite surface protein [NANP]. Limitations of the technology exist as it contains only 20% peptide and has low levels of immunogenicity. It also does not contain any immunodominant T-cell epitopes.[28]
  • A chemical compound undergoing trials for treatment of tuberculosis and cancer—the JmJc inhibitor ML324 and the antitubercular clinical candidate SQ109—is potentially a new line of drugs to treat malaria and kill the parasite in its infectious stage. More tests still need to be carried out before the compounds would be approved as a viable treatment.[29]

Considerations

The task of developing a preventive vaccine for malaria is a complex process. There are a number of considerations to be made concerning what strategy a potential vaccine should adopt.

Parasite diversity

P. falciparum has demonstrated the capability, through the development of multiple drug-resistant parasites, for evolutionary change. The Plasmodium species has a very high rate of replication, much higher than that actually needed to ensure transmission in the parasite’s life cycle. This enables pharmaceutical treatments that are effective at reducing the reproduction rate, but not halting it, to exert a high selection pressure, thus favoring the development of resistance. The process of evolutionary change is one of the key considerations necessary when considering potential vaccine candidates. The development of resistance could cause a significant reduction in efficacy of any potential vaccine thus rendering useless a carefully developed and effective treatment.[30]

Choosing to address the symptom or the source

The parasite induces two main response types from the human immune system. These are anti-parasitic immunity and anti-toxic immunity.

  • “Anti-parasitic immunity” addresses the source; it consists of an antibody response (humoral immunity) and a cell-mediated immune response. Ideally a vaccine would enable the development of anti-plasmodial antibodies in addition to generating an elevated cell-mediated response. Potential antigens against which a vaccine could be targeted will be discussed in greater depth later. Antibodies are part of the specific immune response. They exert their effect by activating the complement cascade, stimulating phagocytic cells into endocytosis through adhesion to an external surface of the antigenic substances, thus ‘marking’ it as offensive. Humoral or cell-mediated immunity consists of many interlinking mechanisms that essentially aim to prevent infection entering the body (through external barriers or hostile internal environments) and then kill any micro-organisms or foreign particles that succeed in penetration. The cell-mediated component consists of many white blood cells (such as monocytesneutrophilsmacrophageslymphocytesbasophilsmast cellsnatural killer cells, and eosinophils) that target foreign bodies by a variety of different mechanisms. In the case of malaria both systems would be targeted to attempt to increase the potential response generated, thus ensuring the maximum chance of preventing disease.[citation needed]
  • “Anti-toxic immunity” addresses the symptoms; it refers to the suppression of the immune response associated with the production of factors that either induce symptoms or reduce the effect that any toxic by-products (of micro-organism presence) have on the development of disease. For example, it has been shown that Tumor necrosis factor-alpha has a central role in generating the symptoms experienced in severe P. falciparum malaria. Thus a therapeutic vaccine could target the production of TNF-a, preventing respiratory distress and cerebral symptoms. This approach has serious limitations as it would not reduce the parasitic load; rather it only reduces the associated pathology. As a result, there are substantial difficulties in evaluating efficacy in human trials.

Taking this information into consideration an ideal vaccine candidate would attempt to generate a more substantial cell-mediated and antibody response on parasite presentation. This would have the benefit of increasing the rate of parasite clearance, thus reducing the experienced symptoms and providing a level of consistent future immunity against the parasite.

Potential targets

See also: PfSPZ Vaccine

Parasite stageTarget
SporozoiteHepatocyte invasion; direct anti-sporozite
HepatozoiteDirect anti-hepatozoite.
Asexual erythrocyticAnti-host erythrocyte, antibodies blocking invasion; anti receptor ligand, anti-soluble toxin
GametocytesAnti-gametocyte. Anti-host erythrocyte, antibodies blocking fertilisation, antibodies blocking egress from the mosquito midgut.

By their very nature, protozoa are more complex organisms than bacteria and viruses, with more complicated structures and life cycles. This presents problems in vaccine development but also increases the number of potential targets for a vaccine. These have been summarised into the life cycle stage and the antibodies that could potentially elicit an immune response.

The epidemiology of malaria varies enormously across the globe, and has led to the belief that it may be necessary to adopt very different vaccine development strategies to target the different populations. A Type 1 vaccine is suggested for those exposed mostly to P. falciparum malaria in sub-Saharan Africa, with the primary objective to reduce the number of severe malaria cases and deaths in infants and children exposed to high transmission rates. The Type 2 vaccine could be thought of as a ‘travellers’ vaccine’, aiming to prevent all cases of clinical symptoms in individuals with no previous exposure. This is another major public health problem, with malaria presenting as one of the most substantial threats to travellers’ health. Problems with the available pharmaceutical therapies include costs, availability, adverse effects and contraindications, inconvenience and compliance, many of which would be reduced or eliminated entirely if an effective (greater than 85–90%) vaccine was developed.[citation needed]

The life cycle of the malaria parasite is particularly complex, presenting initial developmental problems. Despite the huge number of vaccines available, there are none that target parasitic infections. The distinct developmental stages involved in the life cycle present numerous opportunities for targeting antigens, thus potentially eliciting an immune response. Theoretically, each developmental stage could have a vaccine developed specifically to target the parasite. Moreover, any vaccine produced would ideally have the ability to be of therapeutic value as well as preventing further transmission and is likely to consist of a combination of antigens from different phases of the parasite’s development. More than 30 of these antigens are being researched[when?] by teams all over the world in the hope of identifying a combination that can elicit immunity in the inoculated individual. Some of the approaches involve surface expression of the antigen, inhibitory effects of specific antibodies on the life cycle and the protective effects through immunization or passive transfer of antibodies between an immune and a non-immune host. The majority of research into malarial vaccines has focused on the Plasmodium falciparum strain due to the high mortality caused by the parasite and the ease of a carrying out in vitro/in vivo studies. The earliest vaccines attempted to use the parasitic circumsporozoite protein (CSP). This is the most dominant surface antigen of the initial pre-erythrocytic phase. However, problems were encountered due to low efficacy, reactogenicity and low immunogenicity.[citation needed]

  • The initial stage in the life cycle, following inoculation, is a relatively short “pre-erythrocytic” or “hepatic” phase. A vaccine at this stage must have the ability to protect against sporozoites invading and possibly inhibiting the development of parasites in the hepatocytes (through inducing cytotoxic T-lymphocytes that can destroy the infected liver cells). However, if any sporozoites evaded the immune system they would then have the potential to be symptomatic and cause the clinical disease.
  • The second phase of the life cycle is the “erythrocytic” or blood phase. A vaccine here could prevent merozoite multiplication or the invasion of red blood cells. This approach is complicated by the lack of MHC molecule expression on the surface of erythrocytes. Instead, malarial antigens are expressed, and it is this towards which the antibodies could potentially be directed. Another approach would be to attempt to block the process of erythrocyte adherence to blood vessel walls. It is thought that this process is accountable for much of the clinical syndrome associated with malarial infection; therefore a vaccine given during this stage would be therapeutic and hence administered during clinical episodes to prevent further deterioration.
  • The last phase of the life cycle that has the potential to be targeted by a vaccine is the “sexual stage”. This would not give any protective benefits to the individual inoculated but would prevent further transmission of the parasite by preventing the gametocytes from producing multiple sporozoites in the gut wall of the mosquito. It therefore would be used as part of a policy directed at eliminating the parasite from areas of low prevalence or to prevent the development and spread of vaccine-resistant parasites. This type of transmission-blocking vaccine is potentially very important. The evolution of resistance in the malaria parasite occurs very quickly, potentially making any vaccine redundant within a few generations. This approach to the prevention of spread is therefore essential.
  • Another approach is to target the protein kinases, which are present during the entire lifecycle of the malaria parasite. Research is underway on this, yet production of an actual vaccine targeting these protein kinases may still take a long time.[31]
  • Report of a vaccine candidate capable to neutralize all tested strains of Plasmodium falciparum, the most deadly form of the parasite causing malaria, was published in Nature Communications by a team of scientists from the University of Oxford in 2011.[32] The viral vector vaccine, targeting a full-length P. falciparum reticulocyte-binding protein homologue 5 (PfRH5) was found to induce an antibody response in an animal model. The results of this new vaccine confirmed the utility of a key discovery reported from scientists at the Wellcome Trust Sanger Institute, published in Nature.[33] The earlier publication reported P. falciparum relies on a red blood cell surface receptor, known as ‘basigin’, to invade the cells by binding a protein PfRH5 to the receptor.[33] Unlike other antigens of the malaria parasite which are often genetically diverse, the PfRH5 antigen appears to have little genetic diversity. It was found to induce very low antibody response in people naturally exposed to the parasite.[32] The high susceptibility of PfRH5 to the cross-strain neutralizing vaccine-induced antibody demonstrated a significant promise for preventing malaria in the long and often difficult road of vaccine development. According to Professor Adrian Hill, a Wellcome Trust Senior Investigator at the University of Oxford, the next step would be the safety tests of this vaccine. At the time (2011) it was projected that if these proved successful, the clinical trials in patients could begin within two to three years.[34]
  • PfEMP1, one of the proteins known as variant surface antigens (VSAs) produced by Plasmodium falciparum, was found to be a key target of the immune system’s response against the parasite. Studies of blood samples from 296 mostly Kenyan children by researchers of Burnet Institute and their cooperators showed that antibodies against PfEMP1 provide protective immunity, while antibodies developed against other surface antigens do not. Their results demonstrated that PfEMP1 could be a target to develop an effective vaccine which will reduce risk of developing malaria.[35][36]
  • Plasmodium vivax is the common malaria species found in India, Southeast Asia and South America. It is able to stay dormant in the liver and reemerge years later to elicit new infections. Two key proteins involved in the invasion of the red blood cells (RBC) by P. vivax are potential targets for drug or vaccine development. When the Duffy binding protein (DBP) of P. vivax binds the Duffy antigen (DARC) on the surface of RBC, process for the parasite to enter the RBC is initiated. Structures of the core region of DARC and the receptor binding pocket of DBP have been mapped by scientists at the Washington University in St. Louis. The researchers found that the binding is a two-step process which involves two copies of the parasite protein acting together like a pair of tongs which “clamp” two copies of DARC. Antibodies that interfere with the binding, by either targeting the key region of the DARC or the DBP will prevent the infection.[37][38]
  • Antibodies against the Schizont Egress Antigen-1 (PfSEA-1) were found to disable the parasite ability to rupture from the infected red blood cells (RBCs) thus prevent it from continuing with its life cycle. Researchers from Rhode Island Hospital identified Plasmodium falciparum PfSEA-1, a 244 kd malaria antigen expressed in the schizont-infected RBCs. Mice vaccinated with the recombinant PfSEA-1 produced antibodies which interrupted the schizont rupture from the RBCs and decreased the parasite replication. The vaccine protected the mice from lethal challenge of the parasite. Tanzanian and Kenyan children who have antibodies to PfSEA-1 were found to have fewer parasites in their blood stream and milder case of malaria. By blocking the schizont outlet, the PfSEA-1 vaccine may work synergistically with vaccines targeting the other stages of the malaria life cycle such as hepatocyte and RBC invasion.[39][40]

Mix of antigenic components

Increasing the potential immunity generated against Plasmodia can be achieved by attempting to target multiple phases in the life cycle. This is additionally beneficial in reducing the possibility of resistant parasites developing. The use of multiple-parasite antigens can therefore have a synergistic or additive effect.

One of the most successful vaccine candidates in clinical trials[which?][when?] consists of recombinant antigenic proteins to the circumsporozoite protein.[41] (This is discussed in more detail below.)[where?]

Delivery system

 

The selection of an appropriate system is fundamental in all vaccine development, but especially so in the case of malaria. A vaccine targeting several antigens may require delivery to different areas and by different means in order to elicit an effective response. Some adjuvants can direct the vaccine to the specifically targeted cell type—e.g. the use of Hepatitis B virus in the RTS,S vaccine to target infected hepatocytes—but in other cases, particularly when using combined antigenic vaccines, this approach is very complex. Some methods that have been attempted include the use of two vaccines, one directed at generating a blood response and the other a liver-stage response. These two vaccines could then be injected into two different sites, thus enabling the use of a more specific and potentially efficacious delivery system.

To increase, accelerate or modify the development of an immune response to a vaccine candidate it is often necessary to combine the antigenic substance to be delivered with an adjuvant or specialised delivery system. These terms are often used interchangeably in relation to vaccine development; however in most cases a distinction can be made. An adjuvant is typically thought of as a substance used in combination with the antigen to produce a more substantial and robust immune response than that elicited by the antigen alone. This is achieved through three mechanisms: by affecting the antigen delivery and presentation, by inducing the production of immunomodulatory cytokines, and by affecting the antigen presenting cells (APC). Adjuvants can consist of many different materials, from cell microparticles to other particulated delivery systems (e.g. liposomes).

Adjuvants are crucial in affecting the specificity and isotype of the necessary antibodies. They are thought to be able to potentiate the link between the innate and adaptive immune responses. Due to the diverse nature of substances that can potentially have this effect on the immune system, it is difficult to classify adjuvants into specific groups. In most circumstances they consist of easily identifiable components of micro-organisms that are recognised by the innate immune system cells. The role of delivery systems is primarily to direct the chosen adjuvant and antigen into target cells to attempt to increase the efficacy of the vaccine further, therefore acting synergistically with the adjuvant.

There is increasing concern that the use of very potent adjuvants could precipitate autoimmune responses, making it imperative that the vaccine is focused on the target cells only. Specific delivery systems can reduce this risk by limiting the potential toxicity and systemic distribution of newly developed adjuvants.

Studies into the efficacy of malaria vaccines developed to date[when?] have illustrated that the presence of an adjuvant is key in determining any protection gained against malaria. A large number of natural and synthetic adjuvants have been identified throughout the history of vaccine development. Options identified thus far for use combined with a malaria vaccine include mycobacterial cell walls, liposomes, monophosphoryl lipid A and squalene.

History

Individuals who are exposed to the parasite in endemic countries develop acquired immunity against disease and death. Such immunity does not however prevent malarial infection; immune individuals often harbour asymptomatic parasites in their blood. This does, however, imply that it is possible to create an immune response that protects against the harmful effects of the parasite.

Research shows that if immunoglobulin is taken from immune adults, purified and then given to individuals who have no protective immunity, some protection can be gained.[42]

Irradiated mosquitoes

In 1967, it was reported that a level of immunity to the Plasmodium berghei parasite could be given to mice by exposing them to sporozoites that had been irradiated by x-rays.[43] Subsequent human studies in the 1970s showed that humans could be immunized against Plasmodium vivax and Plasmodium falciparum by exposing them to the bites of significant numbers of irradiated mosquitos.[44]

From 1989 to 1999, eleven volunteers recruited from the United States Public Health ServiceUnited States Army, and United States Navy were immunized against Plasmodium falciparum by the bites of 1001–2927 mosquitoes that had been irradiated with 15,000 rads of gamma rays from a Co-60 or Cs-137 source.[45] This level of radiation is sufficient to attenuate the malaria parasites so that, while they can still enter hepatic cells, they cannot develop into schizonts nor infect red blood cells.[45] Over a span of 42 weeks, 24 of 26 tests on the volunteers showed that they were protected from malaria.[46]

References

  1. Jump up to:a b c d e “Mosquirix: Opinion on medicine for use outside EU”European Medicines Agency (EMA)Archived from the original on 23 November 2019. Retrieved 22 November 2019.
  2. Jump up to:a b c “Malaria vaccine becomes first to achieve WHO-specified 75% efficacy goal”EurekAlert!. 23 April 2021. Retrieved 24 April2021.
  3. Jump up to:a b Roxby P (23 April 2021). “Malaria vaccine hailed as potential breakthrough”BBC News. Retrieved 24 April 2021.
  4. ^ “RTS,S malaria candidate vaccine reduces malaria by approximately one-third in African infants”malariavaccine.org. Malaria Vaccine Initiative Path. Archived from the original on 23 March 2013. Retrieved 19 March 2013.
  5. ^ “Commercial name of RTS,S”. Archived from the original on 5 April 2012. Retrieved 20 October 2011.
  6. ^ Foquet L, Hermsen CC, van Gemert GJ, Van Braeckel E, Weening KE, Sauerwein R, et al. (January 2014). “Vaccine-induced monoclonal antibodies targeting circumsporozoite protein prevent Plasmodium falciparum infection”The Journal of Clinical Investigation124 (1): 140–4. doi:10.1172/JCI70349PMC 3871238PMID 24292709.
  7. Jump up to:a b Agnandji ST, Lell B, Fernandes JF, Abossolo BP, Methogo BG, Kabwende AL, et al. (December 2012). “A phase 3 trial of RTS,S/AS01 malaria vaccine in African infants”The New England Journal of Medicine367 (24): 2284–95. doi:10.1056/NEJMoa1208394PMID 23136909.
  8. ^ Borghino D (27 April 2015). “Malaria vaccine candidate shown to prevent thousands of cases”http://www.gizmag.com. Retrieved 11 June 2016.
  9. ^ “GSK announces EU regulatory submission of malaria vaccine candidate RTS,S” (Press release). GSK. 24 July 2014. Archived from the original on 4 December 2016. Retrieved 30 July 2015.
  10. ^ Kelland K (7 October 2013). “GSK aims to market world’s first malaria vaccine”Reuters. Retrieved 9 December 2013.
  11. ^ “First malaria vaccine receives positive scientific opinion from EMA” (Press release). European Medicines Agency (EMA). 24 July 2015. Retrieved 30 July 2015.
  12. ^ “GSK’s malaria candidate vaccine, Mosquirix (RTS,S), receives positive opinion from European regulators for the prevention of malaria in young children in sub-Saharan Africa” (Press release). GSK. 24 July 2015. Archived from the original on 28 July 2015. Retrieved 30 July 2015.
  13. ^ Alonso P (19 June 2019). “Letter to partners – June 2019”(Press release). Wuxi: World Health Organization. Retrieved 22 October 2019.
  14. ^ “Malaria vaccine launched in Kenya: Kenya joins Ghana and Malawi to roll out landmark vaccine in pilot introduction” (Press release). Homa Bay: World Health Organization. 13 September 2019. Retrieved 22 October 2019.
  15. ^ Davies L (6 October 2021). “WHO endorses use of world’s first malaria vaccine in Africa”The Guardian. Retrieved 6 October2021.
  16. ^ “WHO recommends groundbreaking malaria vaccine for children at risk” (Press release). World Health Organization. Retrieved 6 October 2021.
  17. ^ Mandavilli A (6 October 2021). “A ‘Historical Event’: First Malaria Vaccine Approved by W.H.O.” The New York Times. Retrieved 6 October 2021.
  18. ^ Graves P, Gelband H (October 2006). “Vaccines for preventing malaria (blood-stage)”The Cochrane Database of Systematic Reviews (4): CD006199. doi:10.1002/14651858.CD006199PMC 6532641PMID 17054281.
  19. ^ Graves P, Gelband H (October 2006). “Vaccines for preventing malaria (pre-erythrocytic)”The Cochrane Database of Systematic Reviews (4): CD006198. doi:10.1002/14651858.CD006198PMC 6532586PMID 17054280.
  20. ^ Lowe D (23 April 2021). “Great Malaria Vaccine News”Science Translational Medicine. Retrieved 24 April 2021.
  21. ^ “Researcher’s nanoparticle key to new malaria vaccine”Research & Development. 4 September 2014. Retrieved 12 June2016.
  22. ^ Burkhard P, Lanar DE (2 December 2015). “Malaria vaccine based on self-assembling protein nanoparticles”Expert Review of Vaccines14 (12): 1525–7. doi:10.1586/14760584.2015.1096781PMC 5019124PMID 26468608.
  23. ^ “Nature report describes complete protection after 10 weeks with three doses of PfSPZ- CVac” (Press release). 15 February 2017.
  24. ^ “SANARIA PfSPZ VACCINE AGAINST MALARIA RECEIVES FDA FAST TRACK DESIGNATION” (PDF). Sanaria Inc. 22 September 2016. Archived from the original (PDF) on 23 October 2016. Retrieved 23 January 2017.
  25. ^ Butler D (April 2019). “Promising malaria vaccine to be tested in first large field trial”. Naturedoi:10.1038/d41586-019-01232-4PMID 32291409.
  26. ^ “First vaccine to fully immunize against malaria builds on pandemic-driven RNA tech”academictimes.com. 25 February 2021. Retrieved 1 March 2021.
  27. ^ Graves P, Gelband H (April 2006). “Vaccines for preventing malaria (SPf66)”The Cochrane Database of Systematic Reviews(2): CD005966. doi:10.1002/14651858.CD005966PMC 6532709PMID 16625647.
  28. ^ Ratanji KD, Derrick JP, Dearman RJ, Kimber I (April 2014). “Immunogenicity of therapeutic proteins: influence of aggregation”Journal of Immunotoxicology11 (2): 99–109. doi:10.3109/1547691X.2013.821564PMC 4002659PMID 23919460.
  29. ^ Reuters Staff (15 January 2021). “South African scientists discover new chemicals that kill malaria parasite”Reuters. Retrieved 2 February 2021.
  30. ^ Kennedy DA, Read AF (December 2018). “Why the evolution of vaccine resistance is less of a concern than the evolution of drug resistance”Proceedings of the National Academy of Sciences of the United States of America115 (51): 12878–12886. doi:10.1073/pnas.1717159115PMC 6304978PMID 30559199.
  31. ^ Zhang VM, Chavchich M, Waters NC (March 2012). “Targeting protein kinases in the malaria parasite: update of an antimalarial drug target”Current Topics in Medicinal Chemistry12 (5): 456–72. doi:10.2174/156802612799362922PMID 22242850. Archived from the original on 30 May 2013. Retrieved 23 March2020.
  32. Jump up to:a b Douglas AD, Williams AR, Illingworth JJ, Kamuyu G, Biswas S, Goodman AL, et al. (December 2011). “The blood-stage malaria antigen PfRH5 is susceptible to vaccine-inducible cross-strain neutralizing antibody”Nature Communications2 (12): 601. Bibcode:2011NatCo…2..601Ddoi:10.1038/ncomms1615PMC 3504505PMID 22186897.
  33. Jump up to:a b Crosnier C, Bustamante LY, Bartholdson SJ, Bei AK, Theron M, Uchikawa M, et al. (November 2011). “Basigin is a receptor essential for erythrocyte invasion by Plasmodium falciparum”Nature480 (7378): 534–7. Bibcode:2011Natur.480..534Cdoi:10.1038/nature10606PMC 3245779PMID 22080952.
  34. ^ Martino M (21 December 2011). “New candidate vaccine neutralizes all tested strains of malaria parasite”fiercebiotech.com. FierceBiotech. Retrieved 23 December 2011.
  35. ^ Parish T (2 August 2012). “Lifting malaria’s deadly veil: Mystery solved in quest for vaccine”. Burnet Institute. Retrieved 14 August2012.
  36. ^ Chan JA, Howell KB, Reiling L, Ataide R, Mackintosh CL, Fowkes FJ, et al. (September 2012). “Targets of antibodies against Plasmodium falciparum-infected erythrocytes in malaria immunity”The Journal of Clinical Investigation122 (9): 3227–38. doi:10.1172/JCI62182PMC 3428085PMID 22850879.
  37. ^ Mullin E (13 January 2014). “Scientists capture key protein structures that could aid malaria vaccine design”. fiercebiotechresearch.com. Retrieved 16 January 2014.
  38. ^ Batchelor JD, Malpede BM, Omattage NS, DeKoster GT, Henzler-Wildman KA, Tolia NH (January 2014). “Red blood cell invasion by Plasmodium vivax: structural basis for DBP engagement of DARC”PLOS Pathogens10 (1): e1003869. doi:10.1371/journal.ppat.1003869PMC 3887093PMID 24415938.
  39. ^ Mullin E (27 May 2014). “Antigen Discovery could advance malaria vaccine”. fiercebiotechresearch.com. Retrieved 22 June2014.
  40. ^ Raj DK, Nixon CP, Nixon CE, Dvorin JD, DiPetrillo CG, Pond-Tor S, et al. (May 2014). “Antibodies to PfSEA-1 block parasite egress from RBCs and protect against malaria infection”Science344(6186): 871–7. Bibcode:2014Sci…344..871Rdoi:10.1126/science.1254417PMC 4184151PMID 24855263.
  41. ^ Plassmeyer ML, Reiter K, Shimp RL, Kotova S, Smith PD, Hurt DE, et al. (September 2009). “Structure of the Plasmodium falciparum circumsporozoite protein, a leading malaria vaccine candidate”The Journal of Biological Chemistry284 (39): 26951–63. doi:10.1074/jbc.M109.013706PMC 2785382PMID 19633296.
  42. ^ “Immunoglobulin Therapy & Other Medical Therapies for Antibody Deficiencies”Immune Deficiency Foundation. Retrieved 30 September 2019.
  43. ^ Nussenzweig RS, Vanderberg J, Most H, Orton C (October 1967). “Protective immunity produced by the injection of x-irradiated sporozoites of plasmodium berghei”. Nature216 (5111): 160–2. Bibcode:1967Natur.216..160Ndoi:10.1038/216160a0PMID 6057225S2CID 4283134.
  44. ^ Clyde DF (May 1975). “Immunization of man against falciparum and vivax malaria by use of attenuated sporozoites”. The American Journal of Tropical Medicine and Hygiene24 (3): 397–401. doi:10.4269/ajtmh.1975.24.397PMID 808142.
  45. Jump up to:a b Hoffman SL, Goh LM, Luke TC, Schneider I, Le TP, Doolan DL, et al. (April 2002). “Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites”The Journal of Infectious Diseases185 (8): 1155–64. doi:10.1086/339409PMID 11930326.
  46. ^ Hoffman SL, Goh LM, Luke TC, Schneider I, Le TP, Doolan DL, et al. (April 2002). “Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites”The Journal of Infectious Diseases185 (8): 1155–64. doi:10.1086/339409PMID 11930326.

Further reading

External links

Screened cup of malaria-infected mosquitoes which will infect a volunteer in a clinical trial
Vaccine description
TargetMalaria
Vaccine typeProtein subunit
Clinical data
Trade namesMosquirix
Routes of
administration
Intramuscular[1]
ATC codeNone
Legal status
Legal statusEU: Rx-only [1]
Identifiers
CAS Number149121-47-1
ChemSpidernone

//////////////RTS,S/AS01, Mosquirix, malaria vaccine, gsk, VACCINE, RTS,S, APPROVALS 2021

NEW DRUG APPROVALS

ONE TIME

$10.00

Soberana 02, FINLAY-FR-2


IFV - Home

Soberana 02

FINLAY-FR-2

cas 2543416-58-4

A SARS-CoV-2 vaccine comprising a conjugate of the spike protein RBD domain with tetanus toxoid (Finlay Vaccine Institute of Cuba)
Soberana 02
, is a conjugate vaccine developed by Instituto Finlay de Vacunas.[517]

Cuba[518]

Iran[517]

517 Zimmer, Carl; Corum, Jonathan; Wee, Sui-Lee. “Coronavirus Vaccine Tracker”. The New York Times. Retrieved 30 June 2021.

518 Sesin, Carmen (14 May 2021). “Cuba begins mass Covid-19 vaccine inoculation before concluding trials”. NBC News. Retrieved 2 July 2021.

Soberana 02, technical name FINLAY-FR-2, is a COVID-19 vaccine produced by the Finlay Institute, a Cuban epidemiological research institute. It is a conjugate vaccine. This candidate followed a previous one called SOBERANA-01 (FINLAY-FR-1).[2] Professor Ihosvany Castellanos Santos said that the antigen is safe because it contains parts instead of the whole live virus, and therefore it does not require extra refrigeration, like other candidates in the world.[3] According to the WHO candidate landscape vaccine document, this vaccine requires two doses, the second one being administered 28 days after the first shot.[4]

The name of the vaccine, Soberana, is a Spanish word that means “sovereign”.[5]

An overview of current COVID-19 vaccine platforms - ScienceDirect

Efficacy

It has shown an efficacy of 62% after only two doses, according to BioCubaFarma, though a pre-print or details of the study have not been released.[6][7][8]

Pharmacology

FINLAY-FR-2 is a conjugate vaccine. It consists of the receptor binding domain of the SARS-CoV-2 spike protein conjugated chemically to tetanus toxoid.[2]

Manufacturing

The spike protein subunit is produced in Chinese hamster ovary cell culture.[2] In a pre-print article scientists from Cuba explain details of the vaccines technology and production.[9][non-primary source needed]

 
  Production  Deliveries  Planned Production  Potential Production

Deliveries (0)Effective production (implies deliveries) (1)

  1. Cuba[10][11]

Planned production

  1. Iran

Potential Production

  1. Ghana
  2. Argentina

In Cuba

The Cuban government says it is planning to produce 100 million doses of its vaccine to respond to its own demand and that of other countries.[12][13] Cuba has also suggested that, once it’s approved, it will offer the vaccine to tourists visiting the country.[14][15][16]

The production of the first batch of about 100,000 doses will start in April.[17] José Moya, representative of the World Health Organization and the Pan American Health Organization (PAHO) in Cuba, suggested that after the vaccine passes all clinical stages, it could be included as part of PAHO’s Revolving Fund.[18]

The roll-out began with an “Interventional Trial”[19] that consisted of inoculating 150,000 at-risk participants which seems to be defined as health-care workers.[20][21] On April 11, 2021, the Ministry of Public Health of Cuba announced that 75,000 health-care workers were inoculated with their first dose of either of the two Cuba’s Phase III vaccines (the other being Abdala).[22][23]

Outside Cuba

Vietnam, Iran, Venezuela, Argentina,[24][25][26] Pakistan, India, the African Union, Jamaica and Suriname[27] have expressed interest in purchasing the vaccine, although they are waiting on Phase 3 results.[28][29]

Iran has signed an agreement to manufacture the vaccine[30] and Argentina is negotiating one.[24][25][26] Additionally, the Cuban government offered a “transfer of technology” to Ghana and will also supply “active materials” needed to make the vaccine.[31][32][33]

While the price is currently unknown, the commercialization strategy of the vaccine will be a combination of the “impact on health” and the capability of Cuba’s system to financially support “the production of vaccines and drugs for the country”, per the director of the Finlay Institute, Vicente Vérez.[34]

Clinical trials

Phase I

FINLAY-FR-2, which started being developed in October 2020, had 40 volunteers for its Phase I, according to the Cuban Public Registry of Clinical Trials, with an open, sequential and adaptive study to assess safety, reactogenicity and explore immunogenicity of the vaccine.[35]

Phase II

Phase IIa involved 100 Cubans, and phase IIb of the vaccine will have 900 volunteers between 19 and 80 years.[36][37] Vicente Vérez, director general of the Finlay Vaccine Institute, said that the vaccine has shown to give an immune response after 14 days.[38] The second phase has been supervised by Iranian officials from the Pasteur Institute.[5]

Phase III

Phase III commenced at the beginning of March as originally scheduled,[39][15] and “ready to publish” results are expected by June.[40][41][42] The trial volunteers are divided into three groups: some will receive two doses of the vaccine 28 days apart, another group will get two doses plus a third immune booster (Soberana Plus[43][44][45]), and the third a placebo.[39]

Although the trials involve thousands of adult volunteers recruited in Havana,[46] Cuba’s public health officials have said that they will also need to conduct phase III trials abroad because the island doesn’t have an outbreak of sufficient scale to produce meaningful statistics on vaccine protection.[5][14]

On March 13, 2021, the Cuban Biotechnology and Pharmaceutical Industries Business Group (BioCubaFarma) announced on social media that it had sent 100,000 doses of its Soberana 02 coronavirus vaccine candidate to the Pasteur Institute of Iran for clinical testing, “as part of the collaboration with other countries in the development of COVID-19 vaccines.” [47]

On April 26, 2021, it was reported that a Phase III conducted by the Pasteur Institute of Iran was approved to be started in Iran[48][49][50] It was previously reported that the Institute will host Phase 3 but the pre-requisites were “technology transfer and joint production”.[51][5]

Mexico plans to host a phase 3 trial.[52]

Interventional Study

The “Interventional Study” is set both in Havana,[53] Cuba’s capital and Santiago de Cuba, Cuba’s second most populous city [54][55] and in other provinces.[56] On May 6, 2021, the Finlay Institute of Vaccines announced on social media that the following adverse events have been observed: injection site pain (20%), inflammation at the injection site (5%), and general discomfort (5%).[57][58]

Authorizations

 
  Full authorization  Emergency authorization

See also: List of COVID-19 vaccine authorizations § Soberana 02

References

  1. ^ “Cuba’s Soberana Plus against Covid-19 is showing good results”. Prensa Latina. Retrieved 10 May 2021.
  2. Jump up to:a b c Malik JA, Mulla AH, Farooqi T, Pottoo FH, Anwar S, Rengasamy KR (January 2021). “Targets and strategies for vaccine development against SARS-CoV-2”Biomedicine & Pharmacotherapy137: 111254. doi:10.1016/j.biopha.2021.111254PMC 7843096PMID 33550049.
  3. ^ Santos IC (January 2021). “Rapid response to: Covid 19: Hope is being eclipsed by deep frustration”BMJ372: n171. doi:10.1136/bmj.n171.
  4. ^ “Draft landscape and tracker of COVID-19 candidate vaccines”http://www.who.intWorld Health Organization. Retrieved 2021-02-04.
  5. Jump up to:a b c d Rasmussen SE, Eqbali A (12 January 2021). “Iran, Cuba, Under U.S. Sanctions, Team Up for Covid-19 Vaccine Trials”The Wall Street Journal.
  6. ^ “Cuba’s homegrown Covid vaccine shows promise”http://www.ft.com. Retrieved 2021-06-20.
  7. ^ “Cuba encouraged by early efficacy results of homegrown COVID-19 vaccine”http://www.zawya.com. Retrieved 2021-06-20.
  8. ^ Acosta, Nelson (2021-06-20). “Cuba encouraged by early results of homegrown COVID-19 vaccine amid worst outbreak”The Age. Retrieved 2021-06-20.
  9. ^ Valdes-Balbin, Yury; Santana-Mederos, Darielys; Quintero, Lauren; Fernández, Sonsire; Rodriguez, Laura; Ramirez, Belinda Sanchez; Perez, Rocmira; Acosta, Claudia; Méndez, Yanira; Ricardo, Manuel G.; Hernandez, Tays (2021-02-09). “SARS-CoV-2 RBD-Tetanus toxoid conjugate vaccine induces a strong neutralizing immunity in preclinical studies”doi:10.1101/2021.02.08.430146.
  10. ^ Melimopoulos, Elizabeth. “Is Cuba closing in on COVID vaccine sovereignty?”http://www.aljazeera.com. Retrieved 2021-05-07.
  11. ^ “Optimism as Cuba set to test its own Covid vaccine”BBC News. 2021-02-16. Retrieved 2021-05-07.
  12. ^ “Cuba espera fabricar 100 millones de dosis de su candidato vacunal Soberana 02”Nodal (in Spanish). 21 January 2021.
  13. ^ “Vaccino, Cuba pronta a produrre 100 milioni di dosi di ‘Soberana 02′”Dire (in Italian). 21 January 2021.
  14. Jump up to:a b Ribeiro G (4 February 2021). “Cuba to offer coronavirus vaccines to tourists”Brazilian Report.
  15. Jump up to:a b “Coronavirus: Vacuna cubana Soberana 02 alista fase 3 y ensayos”Deutsche Welle (in Spanish). 5 February 2021.
  16. ^ Meredith S (23 February 2021). “‘Sun, sea, sand and Soberana 02’: Cuba open to inoculating tourists with homegrown Covid vaccine”CNBC.
  17. ^ “Coronavirus: Vacuna cubana Soberana 02 alista fase 3 y ensayos”Deutsche Welle (in Spanish). 5 February 2021. Las expectativas sobre Soberana 02 son tales que el titular del organismo estatal que desarrolló la vacuna, Vicente Vérez, confirmó que mientras se aguarden los resultados de la Fase 3 solo en La Habana, en abril se dará inicio a la producción del primer lote, de alrededor de 100 mil dosis.
  18. ^ “Cuba anuncia fase 3 de la vacuna Soberana 02”La Jornada(in Spanish). 7 February 2021. Una vez que superen las etapas clínicas, la OMS podría contar con el fármaco cubano, afirmó Moya, y “pasar a ser parte del grupo de vacunas que se oferten a través del Fondo Rotatorio”, un mecanismo que desde hace cuatro décadas permite gestionar antígenos e insumos a los países de las Américas.
  19. ^ “SOBERANA – INTERVENTION | Registro Público Cubano de Ensayos Clínicos”rpcec.sld.cu. Retrieved 2021-04-11.
  20. ^ “Cuba says it’s ‘betting it safe’ with its own Covid vaccine”NBC News. Retrieved 2021-04-11.
  21. ^ “Cuba begins testing 2nd COVID-19 vaccine on health care workers”medicalxpress.com. Retrieved 2021-04-11.
  22. ^ Ministry of Public Health of Cuba (11 April 2021). “[Translated] “The administration of the 1st dose of the Cuban vaccine candidates #Soberana02 and #Abdala to the 75 thousand health workers and Biocubafarma who are part of the intervention study taking place in #LaHabana has concluded.””Twitter. Retrieved 2021-04-11.
  23. ^ “Cuban scientists, health workers received first anti-Covid-19 dose”http://www.plenglish.com/index.php?o=rn&id=66247&SEO=cuban-scientists-health-workers-received-first-anti-covid-19-dose (in Spanish). Retrieved 2021-04-11.
  24. Jump up to:a b “ILARREGUI (EMBAJADOR EN CUBA): “DURANTE ESTE AÑO PODREMOS TENER VACUNAS CUBANAS EN ARGENTINA””RadioCut. Retrieved 2021-05-07.
  25. Jump up to:a b Argentina, Cadena 3. “Argentina comenzó a negociar con Cuba la vacuna Soberana”Cadena 3 Argentina (in Spanish). Retrieved 2021-05-07.
  26. Jump up to:a b de 2021, 6 de Mayo. “Sin definiciones sobre cuándo podrían llegar, el Gobierno avanza para conseguir las vacunas Soberana y Abdala de Cuba”infobae (in Spanish). Retrieved 2021-05-07.
  27. ^ admin (2021-04-09). “Cuba’s COVID-19 Vaccines Being Sought After by CARICOM Countries”Caribbean News. Retrieved 2021-05-07.
  28. ^ Guenot, Marianne (2021-02-15). “Cuba is working on a homegrown COVID-19 vaccine program. It has a history of fighting disease without help from the West”Business Insider France (in French). Retrieved 2021-05-07.
  29. ^ Página12 (2021-01-22). “Soberana 02: Cuba prepara cien millones de dosis de la vacuna contra el coronavirus | “No somos una multinacional. Nuestro fin es crear salud”, dijo el director del Instituto Finlay de Vacunas”PAGINA12. Retrieved 2021-05-07.
  30. ^ “Cuban coronavirus vaccine to start third clinical trial phase in Iran”Tehran Times. 2021-04-18. Retrieved 2021-05-07.
  31. ^ Banini | 0542440286, Awofisoye Richard. “CEO OF FDA DISCUSSES PRODUCTION OF COVID-19 VACCINE WITH CUBAN AMBASSADOR”http://www.fdaghana.gov.gh. Retrieved 2021-05-05.
  32. ^ “Cuba To Transfer COVID-19 Vaccine Technology To Ghana”http://www.gnbcc.net. Retrieved 2021-05-05.
  33. ^ “Cuban government offers to transfer COVID-19 Soberana 02 vaccine technology to Ghana”Rio Times Online. 16 February 2021.
  34. ^ “Coronavirus: Cuba will produce 100 million doses of its Soberana 02 vaccine”OnCubaNews English. 2021-01-21. Retrieved 2021-05-07.
  35. ^ “SOBERANA 02 | Registro Público Cubano de Ensayos Clínicos”Cuban Registry of Clinical Trials (in Spanish). Retrieved 24 January 2021.
  36. ^ Cuba inicia nova fase de testes com vacina que desenvolve contra covid-19 (in Portuguese), Universo Online, 19 January 2021, Wikidata Q105047566
  37. ^ “Cuba apuesta por crear primera vacuna de América Latina contra el covid-19”France 24 (in Spanish). 2021-01-21. Retrieved 24 January 2021.
  38. ^ “Cuba negotiates with other countries to develop phase 3 of Soberana 02 vaccine”OnCubaNews English. 2020-12-30. Retrieved 24 January 2021.
  39. Jump up to:a b “Cuban-developed vaccine enters Phase III trial”ABS CBN. 5 March 2021.
  40. ^ Mega, Emiliano Rodríguez (2021-04-29). “Can Cuba beat COVID with its homegrown vaccines?”Naturedoi:10.1038/d41586-021-01126-4PMID 33927405.
  41. ^ “Cuban Vaccine Ready in July. Interview with the Cuban Ambassador to the Czech Republic”Pressenza. 2021-03-23. Retrieved 2021-04-29.
  42. ^ Augustin, Ed (2021-05-12). “Cuba deploys unproven homegrown vaccines, hoping to slow an exploding virus outbreak”The New York TimesISSN 0362-4331. Retrieved 2021-05-14.
  43. ^ “L’esempio cubano sui vaccini”http://www.ilfoglio.it (in Italian). Retrieved 2021-05-07.
  44. ^ Avances de las vacunas cubanas contra la COVID-19, retrieved 2021-05-07
  45. ^ Mega, Emiliano Rodríguez (2021-04-29). “Can Cuba beat COVID with its homegrown vaccines?”Naturedoi:10.1038/d41586-021-01126-4PMID 33927405.
  46. ^ Yaffe, Helen. “Cuba’s five COVID-19 vaccines: the full story on Soberana 01/02/Plus, Abdala, and Mambisa”LSE Latin America and Caribbean blog. Retrieved 2021-03-31.
  47. ^ “Cuba sends 100,000 doses of the Soberana 02 vaccine candidate to Iran” oncubanews.com. Retrieved 19 March 2021.
  48. ^ “Iran-Cuba vaccine enters phase three clinical trials”Tehran Times. 2021-04-26. Retrieved 2021-04-28.
  49. ^ “Cuban coronavirus vaccine to start third clinical trial phase in Iran”Tehran Times. 2021-04-18. Retrieved 2021-04-28.
  50. ^ “América Latina apura una vacuna propia. Cuba, adelante; México avanza. Pero no son los únicos”http://www.poresto.net (in Spanish). Retrieved 2021-04-28.
  51. ^ Marsh S (2021-01-09). “Cuba to collaborate with Iran on coronavirus vaccine”Reuters. Retrieved 2021-01-24.
  52. ^ “Mexico Hopes to Work With Cuba on Covid Vaccine Phase 3 Trial”Bloomberg.com. 2021-02-14. Retrieved 2021-05-07.
  53. ^ Marsh, Sarah (2021-03-24). “Nearly all Havana to receive experimental Cuban COVID-19 vaccines”Reuters. Retrieved 2021-04-28.
  54. ^ BioCubaFarma (April 6, 2021). “[Translated] Updating the vaccination process with vaccine candidates #Soberana02 and #Abdala during ongoing clinical trials.#VacunasCubanasCovid19”Twitter (in Spanish). Retrieved 2021-04-11.
  55. ^ “Intervention study with Covid-19 vaccine candidate Abdala begins”Radio Cadena Agramonte. Retrieved 2021-04-28.
  56. ^ “Cuba administers over 62,000 doses in intervention trials”http://www.plenglish.com/index.php?o=rn&id=66012&SEO=cuba-administers-over-62000-doses-in-intervention-trials (in Spanish). Retrieved 2021-04-28.
  57. ^ “[Trnslated] In more than 62 thousand applied doses of #Soberana02 the safety of the vaccine has been demonstrated. Adverse effects have been: 👉 Pain at the injection site (20%). 👉 Redness at the injection site (5%). 👉 Feeling of general malaise (5%)”Twitter. Retrieved 2021-05-07.
  58. ^ “[Translated]In more than 62 thousand applied doses of #Soberana02 the safety of the vaccine has been demonstrated. Adverse effects have been: 👉 Pain at the injection site (20%). 👉 Redness at the injection site (5%). 👉 Feeling of general malaise (5%)”Facebook. Retrieved 2021-05-07.

External links

Scholia has a profile for SOBERANA 02 (Q105047585).
Vaccine description
TargetSARS-CoV-2
Vaccine typeConjugate
Clinical data
Other namesFINLAY-FR-2, SOBERANA PLUS[1]
Routes of
administration
Intramuscular
Legal status
Legal statusFull and Emergency Authorizations: List of Soberana 02 authorizations
Part of a series on the
COVID-19 pandemic
COVID-19 (disease)SARS-CoV-2 (virus)CasesDeaths
showTimeline
showLocations
showInternational response
showMedical response
showEconomic impact and recession
showImpacts
 COVID-19 portal

/////////////////SARS-CoV-2, covid 19, corona virus, vaccine, iran, cuba, Soberana 02, FINLAY-FR-2

 Nature (London, United Kingdom) (2021), 

wdt-3

NEW DRUG APPROVALS

one time

$10.00

CVnCoV, zorecimeran, CureVac COVID-19 vaccine



CVnCoV

cas 2541470-90-8 

An optimized, non-chemical modified mRNA encoding the prefusion-stabilized full-length spike protein of SARS-CoV-2 virus (Curevac)

zorecimeranCureVac COVID-19 vaccine

CureVac/Bayer

GSK

NCT04674189 NCT04449276 NCT04515147 NCT04652102
EudraCT-2020-004066-19

mRNA-based vaccine

PHASE 3

CVnCoVHumoral and cellular responsesCD4+ T-cells, CD8+ T-cellsN/AN/ARhesus macaque[124]

124. Rauch S, Gooch K, Hall Y, Salguero FJ, Dennis MJ, Gleeson FV. et almRNA vaccine CVnCoV protects non-human primates from SARS-CoV-2 challenge infectionbioRxiv. 2020. 2020 12.23.424138

The CureVac COVID-19 vaccine is a COVID-19 vaccine candidate developed by CureVac N.V. and the Coalition for Epidemic Preparedness Innovations (CEPI).[1] The vaccine showed inadequate results in its Phase III trials with only 47% efficacy.[2] The European Medicines Agency stated that: “(…) medicine developers should design studies to demonstrate a rate of efficacy of at least 50%.”[3].

The CVnCov Vaccine (or CV07050101) is in development by CureVac AG. The vaccine uses mRNA technology to create a protein associated with SARS-CoV2, and upon administration and replication, to initiate subsequent immune responses in the body. As of June 2020, the company received regulatory approval from German and Belgian Authorities to commence Phase 1 clinical trials of this vaccine (NCT04449276).

Efficacy

On 16 June 2021,[4] CureVac said its vaccine showed 47% efficacy from its Phase III trial. This was based on interim analysis of 134 COVID cases in its Phase III study conducted in Europe and Latin America. The final analysis for the trials requires a minimum of 80 additional cases.[2]

Pharmacology

CVnCoV is an mRNA vaccine that encodes the full-length, pre-fusion stabilized coronavirus spike protein, and activates the immune system against it.[5][6][7] CVnCoV technology does not interact with the human genome.[6] CVnCoV uses unmodified RNA,[8] unlike the Pfizer–BioNTech COVID-19 vaccine and Moderna COVID-19 vaccine, which both use nucleoside-modified RNA.[9]

Manufacturing

Manufacturing of mRNA vaccines can be performed rapidly in high volume,[10] including use of portable, automated printers (“RNA microfactories”) for which CureVac has a joint development partnership with Tesla.[11]

mRNA vaccines require stringent cold chain refrigeration throughout manufacturing, distribution and storage.[12][13] The CureVac technology for CVnCoV uses a non-modified, more natural mRNA less affected by hydrolysis, enabling storage at 5 °C (41 °F) and relatively simplified cold chain requirements that facilitate up to three months of storage and distribution to world regions that do not have specialized ultracold equipment.[6][10]

CureVac has a European-based network to accelerate manufacturing of CVnCoV, if proven safe and effective, for production of up to 300 million doses in 2021 and 600 million doses in 2022.[10][14] An estimated 405 million doses will be provided to EU states.[14]

Clinical trials

In November 2020, CureVac reported results of a Phase I-II clinical trial that CVnCoV (active ingredient zorecimeran) was well-tolerated, safe, and produced a robust immune response.[15][16]

In December 2020, CureVac began a Phase III clinical trial of CVnCoV with 36,500 participants.[17][18] Bayer will provide clinical trial support and international logistics for the Phase III trial, and may be involved in eventual manufacturing should the vaccine prove to be safe and effective.[19][20] In February 2021, the EU’s CHMP started a rolling review of CVnCoV.[21][22] In April 2021, the same procedure began in Switzerland.[23]

Brand names

The manufacturer currently markets the vaccine under the name CVnCoV.[24] Zorecimeran is the proposed international nonproprietary name (pINN).[25]

References

  1. ^ “CureVac focuses on the development of mRNA-based coronavirus vaccine to protect people worldwide”CureVac(Press release). 15 March 2020. Retrieved 17 February 2021.
  2. Jump up to:a b Burger, Ludwig (16 June 2021). “CureVac fails in pivotal COVID-19 vaccine trial with 47% efficacy”Reuters. Retrieved 17 June 2021.
  3. ^ https://www.ema.europa.eu/en/human-regulatory/overview/public-health-threats/coronavirus-disease-covid-19/treatments-vaccines/vaccines-covid-19/covid-19-vaccines-studies-approval#what-is-the-level-of-efficacy-that-can-be-accepted-for-approval?-section
  4. ^ “CureVac Provides Update on Phase 2b/3 Trial of First-Generation COVID-19 Vaccine Candidate, CVnCoV”. 16 June 2021.
  5. ^ https://www.curevac.com/wp-content/uploads/2020/10/20201023-CureVac-Manuscript-draft-preclinical-data.pdf
  6. Jump up to:a b c Schlake T, Thess A, Fotin-Mleczek M, Kallen KJ (November 2012). “Developing mRNA-vaccine technologies”RNA Biology9(11): 1319–30. doi:10.4161/rna.22269PMC 3597572PMID 23064118.
  7. ^ “Understanding mRNA COVID-19 vaccines”. US Centers for Disease Control and Prevention. 18 December 2020. Retrieved 5 January 2021.
  8. ^ “COVID-19”. CureVac. Retrieved 21 December 2020.
  9. ^ Dolgin, Elie (25 November 2020). “COVID-19 vaccines poised for launch, but impact on pandemic unclear”. Nature Biotechnology: d41587–020–00022-y. doi:10.1038/d41587-020-00022-yPMID 33239758S2CID 227176634.
  10. Jump up to:a b c Nawrat A (3 December 2020). “Q&A with CureVac: resolving the ultra-cold chain logistics of Covid-19 mRNA vaccines”. Pharmaceutical Technology. Retrieved 5 January 2021.
  11. ^ “Tesla to make molecule printers for German COVID-19 vaccine developer CureVac”Reuters. 2 July 2020. Retrieved 19 December 2020.
  12. ^ Kartoglu U, Milstien J (July 2014). “Tools and approaches to ensure quality of vaccines throughout the cold chain”Expert Review of Vaccines13 (7): 843–54. doi:10.1586/14760584.2014.923761PMC 4743593PMID 24865112.
  13. ^ Hanson CM, George AM, Sawadogo A, Schreiber B (April 2017). “Is freezing in the vaccine cold chain an ongoing issue? A literature review”Vaccine35 (17): 2127–2133. doi:10.1016/j.vaccine.2016.09.070PMID 28364920.
  14. Jump up to:a b Kansteiner F (17 November 2020). “CureVac, armed with COVID-19 vaccine deal, plots ‘pandemic-scale’ Euro manufacturing expansion”. FiercePharma, Questex LLC. Retrieved 5 January2021.
  15. ^ “CureVac’s Covid-19 vaccine induces immune response in study”. Clinical Trials Arena. 3 November 2020. Retrieved 5 January 2021.
  16. ^ “CureVac’s COVID-19 vaccine triggers immune response in Phase I trial”Reuters. 2 November 2020. Retrieved 5 January2021.
  17. ^ “Multicenter Clinical Study Evaluating the Efficacy and Safety of Investigational SARS-CoV-2 mRNA Vaccine CVnCoV in Adults 18 Years of Age and Older”. EU Clinical Trials Register. 19 November 2020. Retrieved 5 January 2021. Proposed INN: zorecimeran
  18. ^ “A Study to Determine the Safety and Efficacy of SARS-CoV-2 mRNA Vaccine CVnCoV in Adults”ClinicalTrials.gov. 8 December 2020. NCT04652102. Retrieved 19 December 2020.
  19. ^ Burger L (7 January 2021). “CureVac strikes COVID-19 vaccine alliance with Bayer”Reuters. Retrieved 17 February 2021.
  20. ^ “CureVac and Bayer join forces on COVID-19 vaccine candidate CVnCoV”CureVac (Press release). 7 January 2021. Retrieved 17 February 2021.
  21. ^ “EMA starts rolling review of CureVac’s COVID-19 vaccine (CVnCoV)”European Medicines Agency (EMA) (Press release). 11 February 2021. Retrieved 12 February 2021.
  22. ^ “CureVac Initiates Rolling Submission With European Medicines Agency for COVID-19 Vaccine Candidate, CVnCoV”CureVac(Press release).
  23. ^ “CureVac starts review process in Switzerland for COVID-19 vaccine hopeful”Reuters. 19 April 2021. Retrieved 19 April 2021.
  24. ^ “Celonic and CureVac Announce Agreement to Manufacture over 100 Million Doses of CureVac’s COVID-19 Vaccine Candidate, CVnCoV”CureVac (Press release). 30 March 2021. Retrieved 14 April 2021.
  25. ^ World Health Organization (October 2020). “International Nonproprietary Names for Pharmaceutical Substances (INN). Proposed INN: List 124 – COVID-19 (special edition)” (PDF). WHO Drug Information34 (3): 668–69. Archived (PDF) from the original on 27 November 2020.

External links

Scholia has a profile for zorecimeran (Q97154239).
Vaccine description
TargetSARS-CoV-2
Vaccine typemRNA
Clinical data
Other namesCVnCoV, CV07050101
Routes of
administration
Intramuscular
ATC codeNone
Identifiers
DrugBankDB15844
UNII5TP24STD1S
Part of a series on the
COVID-19 pandemic
COVID-19 (disease)SARS-CoV-2 (virus)
showTimeline
showLocations
showInternational response
showMedical response
showImpact
 COVID-19 portal
  1. Rego GNA, Nucci MP, Alves AH, Oliveira FA, Marti LC, Nucci LP, Mamani JB, Gamarra LF: Current Clinical Trials Protocols and the Global Effort for Immunization against SARS-CoV-2. Vaccines (Basel). 2020 Aug 25;8(3). pii: vaccines8030474. doi: 10.3390/vaccines8030474. [Article]
  2. Speiser DE, Bachmann MF: COVID-19: Mechanisms of Vaccination and Immunity. Vaccines (Basel). 2020 Jul 22;8(3). pii: vaccines8030404. doi: 10.3390/vaccines8030404. [Article]
  3. CureVac & Covid-19 [Link]
  4. Smart Patients [Link]
  5. Regulatory News [Link]

////////////zorecimeran, CVnCoV, CV07050101, CORONA VACCINE, COVID 19, VACCINE, CUREVAC, SARS-CoV-2, CV07050101, SARS-CoV-2 mRNA vaccine

wdt-29

NEW DRUG APPROVALS

one time

$10.00

COVAX-19


Vaxine Pty Ltd company logo

Vaxine Pty Ltd company logo

Vaxine's promising new COVID-19 vaccine candidate

Vaxine’s promising new COVID-19 vaccine candidate

A new multivalent COVID-19 vaccine developed by Australian company Vaxine to tackle the new virus variants could be game-changer in the fight against COVID-19 

The world desperately needs a vaccine that blocks virus transmission and protects against all the variants. Covax-19 vaccine may soon change history”— Sharen Pringle, Vaxine Business Mananager

ADELAIDE, SA, AUSTRALIA, May 16, 2021 /EINPresswire.com/ — Professor Petrovsky, who is the Chairman and Research Director of Australian-based Vaxine Pty Ltd, explains that the two biggest challenges to tackling the COVID-19 pandemic are to develop a vaccine that completely prevents virus transmission something other COVID-19 have not been completely successful in achieving, and the second being to find a vaccine that protects equally against all the evolving immune-escape variants.

Professor Petrovsky has been researching coronavirus vaccines for the last 17 years, having previously published scientific papers on vaccines against both the SARS and MERS coronaviruses, which were highly protective in relevant animal models. He also recently published data from a collaboration with the US Army on development of a promising Ebola vaccine that protected mice against this most lethal disease after just a single vaccine dose. He has now successfully taken the same approach to design a protein-based vaccine against COVID-19.

Studies in a broad range of animal models including mice, hamsters, ferrets and monkeys, have recently revealed the high potential of this vaccine that is currently known as Covax-19(TM), but which likely will be soon rebranded as in its latest iteration it moves into late stage human trials in a number of countries.

Recent breakthrough data generated by Vaxine’s partner, Professor Kaissar Tabynov who leads the International Center for Vaccinology at the Kazakh National Agrarian University has shown that Vaxine’s unique spike protein antigen which is produced using insect cells in culture, was unique in that it not only totally protected hamsters from infection themselves but also prevented them from transmitting the virus to unvaccinated animals that were placed in the same cage two days after the vaccinated animals had been challenged with virus. Protection against transmission was not seen in hamsters given other vaccines making this finding unique to Vaxine’s spike protein antigen.

This hamster data reinforced findings in hamster, ferret and monkey challenge study performed by collaborating US Universities, who showed that two doses of Vaxine’s Covax-19 vaccine provided complete clearance of recoverable virus from the lungs and nose of animals when sampled just days after an infectious challenge.

“COVAX-19 vaccine has now been shown to be highly protective against the original Wuhan strain of the virus in hamster, ferret and monkey infection models performed by independent academic institutions in multiple countries, attesting to the strength of our protein-based vaccine approach”, says Prof. Petrovsky.

“A key element in the success of Covax-19 vaccine is the inclusion of Vaxine’s Advax adjuvant technology which acts as a turbocharger to drive an optimal immune response against the virus” explains Prof. Petrovsky who has been working on this promising vaccine adjuvant technology for the last 20 years with funding support from the US National Institutes of Health.

“We have now shown that our COVAX-19 vaccine can provide effective immunity including an ability to block nasal virus replication and this in turn successfully prevents transmission of the virus to vaccine-naïve animals,” he explains.

Follow on studies to confirm and expand upon these initial findings are currently underway at several US universities as well as Kazakh National Agrarian University, with a manuscript describing some of the initial animal data currently under review at a leading vaccine journal.

In another major breakthrough the team has now developed the vaccine into a multivariant format designed to protect against all the recently described variant strains of COVID-19, with work also underway on the most recently described Indian strains.

While the data is still preliminary says Prof. Petrovsky, the immune responses to the multivalent vaccine in mice are generating equally strong antibody binding activity against all the major virus variants. “This is extremely exciting as the world desperately needs vaccines able to protect against all the new strains of the virus including the UK, South African and Brazilian strains. By contrast , the currently available vaccines are clearly not as strong against some of these variants as they are against the original Wuhan strain” he explains.

Already there have been multiple confirmed cases of vaccine breakthrough where otherwise healthy individuals who have received mRNA, adenovirus or inactivated whole virus vaccines have become infected generally with either the South African or Brazilian variants.

This problem of immune-escape will only get worse over time as more complex variants emerge which is why Vaxine has been putting all its energy into finding a robust solution to this issue before proceeding with Phase 3 clinical trials of its Covax-19 vaccine.

Dr. Petrovsky went on to conclude “Now we have a multivalent formulation of Covax-19 vaccine that is showing high promise in animal studies, we plan to work as fast as we can to advance this new vaccine formulation in human trials, while expanding manufacturing capacity to ensure we are able to produce enough vaccine to meet the enormous global demand that will be attracted by such a successful vaccine.”

“To help us in this task Vaxine is looking to assemble a global network of partner organisations in countries around the world to assist Vaxine with vaccine development, clinical trials, manufacturing, distribution and sales. This is going to be a mammoth effort as we go to war against this insidious virus that continues to wreak havoc around the globe, with WHO recently predicting that the second year of the pandemic is likely to be much worse even than the first, an ominous warning for many countries that still remain poorly prepared and lacking in local vaccine manufacturing capability.

Vaxine wishes to help developing countries to establish their own local state-of-the-art vaccine manufacturing facilities, providing advice on appropriate facility design and undertaking technology transfer of its state of the art protein production technology to such facilities.

Countries in the developing world can no longer afford to sit and wait for outside organisations like COVAX to solve their vaccine supply problems, instead Vaxine proposes to help such countries find their own local solutions to the vaccine supply bottleneck for this.

Sharen Pringle
Vaxine Pty Ltd
437 033 400
email us here……..https://www.einnews.com/pr_news/541113168/covid-19-vaccine-breakthrough

Currently, the Australian influenza vaccine and adjuvant specialist and the Polish protein drug maker have just inked a memorandum of understanding, so the terms of a future contract remain to be defined. However, the technology behind is interesting.

The partners intent to utilize an insect cell-based recombinant spike protein of SARS-CoV–2 in combination with Vaxine’s proprietary Advax™ adjuvant and have already started Phase I testing in Australia with first result expected later this month. The company announced it will use artificial intelligence to evalutate clinical data in real time and announced the ambition to complete Phase II and III trials at the end of this year. “Supported by Microsoft technology, we aim to collect and analyse the COVAX-19™ trial data in real time, rather than waiting until the end of the trial before seeing if the vaccine is working, which is the traditional process,” said Vaxine’s Research Director Professor Nikolai Petrovsky from Flinders University in Adelaide.

Preclinically, Vaxine Pty Ltd’s syntetic spike protein with the company’s non-inflammatory Advax™ adjuvant, induced antibody and T-cell immune responses against the co-administered antigen. In various animal models, Covax-19 vaccination provided robust protection against an infection with the novel coronavirus.

The Phase I of Vaxine Pty Ltd in running since July in 40 healthy volunteers. If results are positive, the Australian vaccine maker is to expand studies and manufacturing to Europe. Under a future agreement Mabion SA would lead clinical development, manufacturing, regulatory negotiations and could exclusively market the vaccine in the EU and – optionally – in the US……..https://european-biotechnology.com/up-to-date/latest-news/news/mabion-to-licence-covid-19-jab-from-vaxine-pty-ltd.html

////////////////COVAX-19, corona virus, covid 19, Vaxine, australia, vaccine

wdt-26

NEW DRUG APPROVALS

ONE TIME

$10.00

IIBR-100


Coronavirus: Israel to start COVID-19 vaccine human trials on November 1 | Al Arabiya English

IIBR-100

Brilife

Recombinant vesicular stomatitis virus (rVSV) vaccine

Israel Institute for Biological Research

Hadassah Medical Center; Sheba Medical Center Hospital

The SARS-CoV-2 virus is responsible for the COVID-19 pandemic. The pandemic emerged from Wuhan Province in China in December 2019 and was declared by the WHO Director-General a Public Health Emergency of International Concern on 30 January 2020.

In this study, a vaccine developed by IIBR for SARS-CoV-2 virus will be assessed for its safety and potential efficacy in volunteers. The study is comprised of two phases, a dose-escalation phase (phase I) during which subjects (18-55 years old) will be randomly allocated to receive a single administration of IIBR-100 100 at low, mid or high dose or saline or two administrations of IIBR-100 at low dose, or saline, 28 days apart.

Based on results obtained during phase I, and cumulative phase I data review, the expansion phase (phase II) has begun, during which larger cohorts as well as elderly age subjects will be randomly allocated to receive a single administration of IIBR-100 at low, mid or high dose or saline, or two administrations of IIBR-100 at low, mid or high dose (prime-boost) or saline, 28 days apart. Additional top-dose (prime-boost) may be implemented when immunogenicity of any prime-boost arm is considered insufficient.

Based on immunogenicity preliminary data and DSMB recommendations, the two administrations of mid, high and top dose (prime-boost) or saline will continue.

The subjects will be followed for a period of up to 12 months post last vaccine administration to assess the safety and efficacy of the vaccine.

https://clinicaltrials.gov/ct2/show/NCT04608305

IIBR-100 also known as Brilife is a COVID-19 vaccine candidate developed by The Israel Institute for Biological Research.[1][2]

References

  1. ^ Clinical trial number NCT04608305 for “Phase I/II Randomized, Multi-Center, Placebo-Controlled, Dose-Escalation Study to Evaluate the Safety, Immunogenicity and Potential Efficacy of an rVSV-SARS-CoV-2-S Vaccine (IIBR-100) in Adults” at ClinicalTrials.gov
  2. ^ Jeffay N (29 December 2020). “As Israel goes vaccine-wild, will the homegrown version lose its shot?”The Times of Israel. Retrieved 1 January 2021.

candidate developed by The Israel Institute for Biological Research.[1][2]

https://www.timesofisrael.com/israeli-institutes-vaccine-candidate-said-highly-effective-in-animal-trials/

Israeli institute’s COVID vaccine candidate said very effective in animal trials

Secretive Israeli research center’s shot shows near 100% efficacy in non-human trials, is on par with US company Moderna’s candidate, TV report says

Israeli researchers at a top secret research center have made progress on a coronavirus vaccine that shows a high level of effectiveness in animals, according to a Friday TV report.

However, there is no guarantee that the vaccine under development will be effective in humans, or will be available soon.

The Israel Institute for Biological Research (IIBR), a secretive unit that works under the Prime Minister’s Office, developed a vaccine that shows close to 100 percent protection against the virus in lab animals, the Channel 12 report said, citing “a security source.”

The vaccine under development is on par in effectiveness with a vaccine being developed by US biotechnology company Moderna, the report said.

Unlike vaccines developed abroad, the domestic vaccine will first be delivered to Israeli citizens, it added. If successful, it was expected to provide protection against the disease with a single dose.

The institute has not started human trials but was preparing to manufacture 10 to 15 million doses, report said.

Hebrew media have reported on potential breakthroughs at the shadowy institute several times before, starting in mid-March, with the Defense Ministry pushing back on some of the claims to tamper expectations.

Magen David Adom medical workers test Israelis for the coronavirus at a drive-through site in Lod, on July 10, 2020. (Yossi Aloni/Flash90)

IIBR said last month that it had completed successful coronavirus vaccine trials on rodents, paving the way for further testing on other animals and then possibly human trials.

In a paper published on the website of bioRxiv, an online repository for papers that haven’t yet been peer-reviewed, the institute, which is based in Ness Ziona, said it hopes to have a finished vaccine in a year, or possibly even earlier.

In the abstract of the report, the researchers say their vaccine, which they tested on hamsters, “results in rapid and potent induction of neutralizing antibodies against SARS-CoV-2,” the virus that causes COVID-19.

Earlier this month a vaccine adviser to the government cautioned that there was no guarantee that the shots being developed will prove widely effective.

In May, the institute confirmed that it had isolated an antibody it believed could be used to develop treatments against the virus. The development would not be useful in the creation of a vaccine, but would rather be a move toward a drug treatment for those who have already contracted the disease.

Tal Zaks, Moderna’s Israeli chief medical officer, described to Channel 12 on Friday the company’s push into Phase 3 testing of its vaccine candidate, which was developed with the National Institutes of Health, and began its first injections Monday.

The trial, the world’s largest vaccine study, plans to test the vaccine on 30,000 volunteers.

There’s still no guarantee that the experimental vaccine, developed by the National Institutes of Health and Moderna Inc., will really offer protection.

“The first time we saw the first model, that the vaccine, even if it’s just in mice, successfully stimulated the immune system to identify the virus and neutralize it, I knew that we hadn’t missed anything, that we had the correct vaccine,” he said.

“And of course the second ‘ah-ha’ moment was when we saw the first clinical results, when it was clear that in humans we weren’t just getting to antibody levels we were seeing in sick people, which is what we aspired to, but we were getting to even higher levels,” Zaks said.

A Nurse gives a volunteer an injection, as the world’s biggest study of a possible COVID-19 vaccine, developed by the US National Institutes of Health and Moderna Inc., gets underway on July 27, 2020, in Binghamton, NY. (AP Photo/Hans Pennink)

Last month Israel signed a deal with Moderna for the potential purchase of its coronavirus vaccine if it ends up proving effective.

Moderna said the vaccination was administered in Savannah, Georgia, the first site to get underway among more than seven dozen trial sites scattered around the country.

Several other vaccines made by China and by Britain’s Oxford University earlier this month began smaller final-stage tests in Brazil and other hard-hit countries.

The massive studies aren’t just to test if the shots work — they’re needed to check each potential vaccine’s safety. And following the same study rules will let scientists eventually compare all the shots.

It normally takes years to create a new vaccine from scratch, but scientists are setting speed records this time around, spurred by knowledge that vaccination is the world’s best hope against the pandemic.

If everything goes right with the final studies, it still will take months for the first data to trickle in from the Moderna test, followed by the Oxford one.

Governments around the world are trying to stockpile millions of doses of those leading candidates so if and when regulators approve one or more vaccines, immunizations can begin immediately. But the first available doses will be rationed, presumably reserved for people at highest risk from the virus.

Coronavirus cases in Israel rose by 1,791 in 24 hours on Friday and the national death toll hit 512, according to the latest Health Ministry figures.

The total case count stood at 70,970, with 320 patients in serious condition, including 98 on ventilators. The number of recovered patients reached 43,850.

Israel has the fifth-highest number of new coronavirus infections per capita in the world, overtaking the United States, according to data compiled by a scientific publication based at Oxford University.

And while Israel has seen the number of new coronavirus cases rocket to more than 2,000 a day in recent weeks, a new Hebrew University report published on Thursday asserted that Israel has managed to gain control of the second wave of the coronavirus, thanks to a recent stabilization in the number of seriously and moderately ill patients.

The curve for seriously and moderately ill patients began to spike in late June before stabilizing in recent days, the researchers reported. They credited the restrictions imposed by the government in recent weeks to limit crowding for helping to flatten the curve.

According to the report, the death toll will climb by roughly 200 in the coming three weeks as a result of the high infection rate over the past month.

Experts have blamed a too-speedy reopening and the lack of an effective contact-tracing program as main factors in the virus resurgence, which has come as new daily coronavirus cases around the world have also reached record highs.

Vaccine description
TargetSARS-CoV-2
Vaccine typeViral vector
Clinical data
Other namesBrilife
Routes of
administration
Intramuscular
Part of a series on the
COVID-19 pandemic
COVID-19 (disease)SARS-CoV-2 virus (variants)
showTimeline
showLocations
showInternational response
showMedical response
showImpact
 COVID-19 portal

//////IIBR-100, Brilife,  COVID-19,  vaccine,  israel, corona virus, covid 19, SARS-CoV-2

wdt-21

NEW DRUG APPROVALS

one time

$10.00

ABDALA, CIGB-66


Cuban president praises progress of Abdala vaccine against Covid-19

ABDALA

CIGB-66, is a COVID-19 vaccine

Cuba says Abdala vaccine 92.28% effective against coronavirus

The announcement came just days after the government said another homegrown vaccine, Soberana 2, has proved to be 62% effective with just two of its three doses.

https://health.economictimes.indiatimes.com/news/industry/cuba-says-abdala-vaccine-92-28-effective-against-coronavirus/83735386

  • June 22, 2021, 10:03 IST

HavanaCuba said on Monday that its three-shot Abdala vaccine against the coronavirus has been proved 92.28% effective in last-stage clinical trials.

The announcement came just days after the government said another homegrown vaccine, Soberana 2, has proved to be 62% effective with just two of its three doses.

“Hit by the pandemic, our scientists at the Finlay Institute and Center for Genetic Engineering and Biotechnology have risen above all the obstacles and given us two very effective vaccines,” President Miguel Diaz-Canel tweeted.

The announcement came from state-run biopharmaceutical corporation BioCubaFarma, which oversees Finlay, the maker of Soberana 2, and the Center for Genetic Engineering and Biotechnology, the producer of Abdala.

Both vaccines are expected to be granted emergency authority by local regulators shortly.

Cuba, whose biotech sector has exported vaccines for decades, has five coronavirus vaccine candidates.

The Caribbean’s largest island is facing its worst Covid-19 outbreak since the start of the pandemic following the arrival of more contagious variants, setting new records for daily coronavirus cases.

The Communist-run country has opted not to import foreign vaccines but to rely on its own. Some experts said it was a risky bet but it appears to have paid off, putting Cuba in position to burnish its scientific reputation, generate much-needed hard currency through exports and strengthen the vaccination drive worldwide.

Several countries from Argentina and Jamaica to Mexico, Vietnam and Venezuela have expressed an interest in buying Cuba’s vaccines. Iran started producing Soberana 2 earlier this year as part of late-phase clinical trials.

Cuba’s authorities have already started administering the experimental vaccines en masse as part of “intervention studies” they hope will slow the spread of the virus.

About a million of the country’s 11.2 million residents have been fully vaccinated to date.

Daily cases have halved in the capital, Havana, since the start of the vaccination campaign a month ago, using Abdala, according to official data.

Cuba has reported a total of 169,365 Covid-19 cases and 1,170 deaths.

ABDALA, technical name CIGB-66, is a COVID-19 vaccine candidate developed by the Center for Genetic Engineering and Biotechnology in Cuba.[1][2] This vaccine candidate, named after a patriotic drama by Cuban independence hero José Martí, is a protein subunit vaccine containing COVID-derived proteins that trigger an immune response.[3] However, none of the clinical trial full results have been published. This candidate followed a previous one called CIGB-669 (MAMBISA).[4]

The vaccine is one of two Cuba-developed COVID-19 vaccines in Phase III trials.[5][6][7]

Clinical research

Phase I/II

In July 2020, CIGB-66 commenced phase I/II clinical trials.[8]

Phase III

The Phase III trial compares 3 doses of the vaccine administered at 0, 14 and 28 days against a placebo, with the primary outcome measuring the proportion of cases reported for each group 14 days after the third dose.

The trial was registered on 18 March 2021. The first dose was administered on 22 March and by April 4, the 48,000 participants had received their first dose,[9][10] and second doses started being administered from April 5.[11][12] Third doses have started being administered on 19 April[13][14][15] and on May 1, 97% of the original participants had received their 3 doses, the others 3% were lost in the process.

Intervention study

124,000 people aged 19 to 80 received 3 doses of the vaccine as part of an intervention study, with the primary outcome measuring the proportion of cases and deaths for the vaccinated compared to the unvaccinated population.[16]

A wider intervention study with the 1.7 million inhabitants of Havana is expected to start in May with the ABDALA and Soberana 2 vaccine.[17]

Efficacy

From May 3, the efficacy of the vaccine will start being evaluated.[18][19][20]

The “first evaluation of efficacy” can begin when there is 50 cases, then there is a second evaluation at 100 cases and a definitive efficacy can “finally be demonstrated” at 150 cases, Cuban Center for Genetic Engineering and Biotechnology director said.[21]

Production outside Cuba

Venezuela has claimed that it will manufacture the vaccine[22] but this claim has not yet materialised.[23] State-owned EspromedBIO will manufacture the vaccine but some “arrangements” are needed to start production.[24] In April, Nicolás Maduro said that a capacity of 2 Million doses per month is hoped to be reach by “August, September approximately”.[25

In June 2021, Vietnam’s Ministry of Health announced that negotiations were ongoing between Cuba and Vietnam for Abdala vaccine production. The Institute of Vaccines and Medical Biologicals (IVAC) was named as the focal point for receiving technology transfer.[26]

References

  1. ^ “ABDALA Clinical Study – Phase III”rpcec.sld.cu. Registro Público Cubano de Ensayos Clínicos. Retrieved 22 March 2021.
  2. ^ “ABDALA Clinical Study”rpcec.sld.cu. Registro Público Cubano de Ensayos Clínicos. Retrieved 22 March 2021.
  3. ^ Yaffe H (31 March 2021). “Cuba’s five COVID-19 vaccines: the full story on Soberana 01/02/Plus, Abdala, and Mambisa”LSE Latin America and Caribbean blog. Retrieved 31 March 2021.
  4. ^ “MAMBISA Study”rpcec.sld.cu. Registro Público Cubano de Ensayos Clínicos. Retrieved 22 March 2021.
  5. ^ “Three-shot Cuban COVID-19 vaccine candidate moves forward in phase III”http://www.bioworld.com. Retrieved 10 April 2021.
  6. ^ “Cuba’s Abdala COVID-19 vaccine enters phase 3 clinical trial – Xinhua | English.news.cn”http://www.xinhuanet.com. Retrieved 10 April 2021.
  7. ^ Zimmer C, Corum J, Wee SL. “Coronavirus Vaccine Tracker”The New York TimesISSN 0362-4331. Retrieved 10 April 2021.
  8. ^ “ABDALA Clinical Study”rpcec.sld.cu. Registro Público Cubano de Ensayos Clínicos. Retrieved 21 March 2021.
  9. ^ BioCubaFarma (4 April 2021). “[Translated] “The application of the 1st dose of #Abdala, in volunteer 48 thousand, of the Phase III Clinical Trial. Next Monday, April 5, the application of the 2nd dose of this vaccine candidate begins. #VcaunasCubanasCovid19 .””Twitter. Retrieved 10 April 2021.
  10. ^ “Covid Check-in: Cuba’s Homegrown Vaccines”AS/COA. Retrieved 10 April 2021.
  11. ^ BioCubaFarma (5 April 2021). “[Translated] “The application of the 2nd dose of the vaccine candidates begins today #Abdala and #Soberana02 , as part of the 3rd phase of the clinical trial. Workers of @Emcomed1 in Havana and eastern provinces, from very early hours they carry out their distribution until the vaccination centers””Twitter (in Spanish). Retrieved 10 April 2021.
  12. ^ “Two Cuban Vaccines Start Second Dose Phase III Trials”Kawsachun News. 5 April 2021. Retrieved 10 April 2021.
  13. ^ “Abdala: Comienza tercera dosis en el Oriente cubano”http://www.cuba.cu (in Spanish). Retrieved 21 April 2021.
  14. ^ BioCubaFarma. “[Translated] “Application of the 3rd dose of the vaccine candidate begins #Abdala in the provinces of Granma, Santiago de Cuba and Guantánamo. The application of the 2nd dose of #Soberana02 within the framework of the EC Phase III.#VacunasCubanasCovid19”Twitter. Retrieved 21 April 2021.
  15. ^ Noticias, Agencia Cubana de. “Convergen múltiples voluntades para éxito de estudio Abdala en Bayamo”ACN (in Spanish). Retrieved 21 April 2021.
  16. ^ “ABDALA-Intervention | Registro Público Cubano de Ensayos Clínicos”rpcec.sld.cu. Retrieved 10 April 2021.
  17. ^ Ministerio de Salud Pública en Cuba. “Sitio oficial de gobierno del Ministerio de Salud Pública en Cuba”Sitio oficial de gobierno del Ministerio de Salud Pública en Cuba (in Spanish). Retrieved 23 April 2021.
  18. ^ “Scientists announce Abdala’s administration of 3rd dose will finish”http://www.plenglish.com/index.php?o=rn&id=66941&SEO=scientists-announce-abdalas-administration-of-3rd-dose-will-finish (in Spanish). Retrieved 2 May 2021.
  19. ^ Noticias, Agencia Cubana de. “Concluye aplicación de vacuna Abdala en Oriente de Cuba”ACN (in Spanish). Retrieved 2 May2021.
  20. ^ “Cuba conclui ensaios clínicos de candidata a vacina contra covid-19”R7.com (in Portuguese). 2 May 2021. Retrieved 2 May2021.
  21. ^ “Abdala cerca de concluir la fase III de ensayos clínicos; Mambisa se alista para avanzar a nueva fase (+Video)”Granma.cu (in Spanish). Retrieved 3 May 2021.
  22. ^ “Cuba says it’s ‘betting it safe’ with its own Covid vaccine”NBC News. Retrieved 10 April 2021.
  23. ^ “Maduro struggles to make his grand vaccine promise”Eminetra.co.uk. 2 May 2021. Retrieved 3 May 2021.
  24. ^ “Venezuela producirá la vacuna cubana anticovid Abdala”http://www.efe.com (in Spanish). Retrieved 3 May 2021.
  25. ^ Apr 11, Reuters /; 2021; Ist, 16:27. “Indonesian President orders Java rescue efforts after quake kills 8 – Times of India”The Times of India. Retrieved 3 May 2021.
  26. ^ Ministry of Health Vietnam (16 June 2021). “Bộ trưởng Bộ Y tế đàm phán với Cuba về hợp tác sản xuất vaccine”giadinh.net.vn(in Vietnamese). Retrieved 17 June 2021.

External links

Scholia has a profile for Abdala (Q106390652).
Vaccine description
TargetSARS-CoV-2
Vaccine typeProtein subunit
Clinical data
Other namesABDALA
Routes of
administration
Intramuscular
Part of a series on the
COVID-19 pandemic
COVID-19 (disease)SARS-CoV-2 virus (variants)
showTimeline
showLocations
showInternational response
showMedical response
showImpact
 COVID-19 portal

//ABDALA, CUBA, CIGB-66,  COVID-19,  vaccine, CORONA VIRUS, SARS-CoV-2

wdt-20

NEW DRUG APPROVALS

ONE TIME

$10.00

QazCovid-in


Kazakhstan joins list of countries with homegrown COVID-19 vaccines
Kazakhstan starts vaccination of volunteers with domestic QazCovid-in  vaccine

QazCovid-in

QazCovid-inVaccinePhase I/II/IIIThe QazCovid-in vaccine is an inactivated vaccine. Inactive viral vaccines are created by propagating viruses in cell culture (such as in Vero cells) and/or by inactivation using a chemical reagent (such as beta-propiolactone or formaldehyde). Upon vaccination, this allows the body to generate a diverse immune response against numerous viral antigens while having no threat of actually being infected because the virus is inactive.NEWS FEED December 31, 2020The Republic of Khazakstan’s QazCovid-in COVID19 vaccine enters phase 3 with an expected 3000 participants. August 28, 2020QazCovid-in, an inactive viral vaccine manufactured by Research Institute for Biological Safety Problems Republic of Kazakhstan enters Phase 1/2 clinical trials.ORGANIZATIONSResearch Institute for Biological Safety Problems, National Scientific Center for Phthisiopulmonology of the Republic of Kazakhstan, City polyclinic No. 4 of the UZO of Almaty, Clinic of the International Institute of Postgraduate Education, City Multidisciplinary Hospital of the Health Department of the Akimat of Zhambyl RegionCOUNTRIES INVOLVED TRIAL PARTICIPANTS

Phase 1: 44

Phase 2: 200

Phase 3: 3000CLINICAL TRIAL NUMBERNCT04530357NCT04691908

QazCovid-in, also known as QazVac, is an inactivated virus vaccine developed by the Research Institute for Biological Safety Problems in Kazakhstan.[499]

Kazakhstan[499]

https://fortune.com/2021/04/26/new-covid-19-vaccine-kazakhstan-qazvac/

A new vaccine on the scene: Kazakhstan begins rollout of homegrown QazVac

The world’s approved COVID-19 vaccines have all come from large economies such as the U.S., China, the U.K., Russia, and India. Until today.

On Monday, Kazakhstan started rolling out its homegrown vaccine, now known as QazVac. Before a rebranding at the end of last month, it was called QazCovid-in, but the central Asian country’s government decided that name might be a turnoff for the public.

The vaccine was developed by Kazakhstan’s Research Institute for Biological Safety Problems, which claimed 96% efficacy in the second stage of clinical trials. The final phase is still ongoing, with a conclusion expected in July, but Kazakh health authorities decided it was fine to begin the rollout as long as the 3,000-participant Phase III trial was at least halfway finished.

This isn’t an adenovirus vector vaccine like those from Johnson & Johnson and AstraZeneca—though it does share their relatively mild refrigeration requirements—nor is it an mRNA-based jab like the BioNTech/Pfizer and Moderna vaccines. Instead, it uses an inactivated form of the SARS-CoV-2 virus itself, much like China’s CoronaVac and India’s Covaxin, which are both in use, and Valneva’s vaccine, which isn’t there yet. The QazVac regimen comprises two doses, to be administered three weeks apart.

‘Turn the tide’

Health Minister Alexei Tsoi was one of the first QazVac recipients on Monday morning. Tsoi was at the start of this month on the receiving end of a public dressing-down by President Kassym-Jomart Tokayev, who was furious about the sluggish start to the country’s inoculation campaign amid rising case numbers.

“You must turn the tide, otherwise a personnel decision that is going to be very disappointing for you will follow,” Tokayev told Tsoi. The vaccination campaign, which had previously focused on frontline workers, then reportedly sprang to life for others too in the oil-rich country.

Thus far, Kazakhstan’s vaccination drive has been powered by Russia’s Sputnik V, which has been produced locally for the past couple of months (Tokayev opted for the Russian shot, rather than waiting for QazVac). By late last week, just over 800,000 people had received their first dose. Kazakhstan has a population of 18.8 million people; the government plans to inoculate 2 million each month.

Tokayev tweeted Friday that domestic production would provide vaccine availability to all citizens. If so, that would be a remarkable turnaround—Almaty health officials said five weeks ago that the largest Kazakh city had run out of vaccines, and mass vaccination would not be realistic in the near future.

QazVac may have given Tokayev the opportunity to praise Kazakhstan’s scientific prowess, but production remains a bottleneck. The first batch to be distributed runs to only 50,000 doses, and the next tranche, to be produced in May, will be of the same volume.

Tsoi said Monday that the Kazakh government was talking to Turkish manufacturers about increasing production capacity.

QazCovid-in, commercially known as QazVac,[1][2] is a COVID-19 vaccine developed by the Research Institute for Biological Safety Problems in Kazakhstan.[3][4][5] QazCoVac-P is a second COVID-19 vaccine developed by the Kazakh Biosafety Research Institute and in clinical trials.[6]

Clinical research

QazVac is currently in Phase 3 (III) of the Clinical Trial, which is expected to be fully completed by 9 July 2021.[7][8] It is unclear when the first preliminary results will be published.[9][10]

The administration of the vaccine for the general population began at the end of April 2021.[11] The Research Institute Kunsulu Zakarya’s Director General’s justification is that the trial is almost 50% completed and “people who have received [the] vaccine feel well; there have been no side-effects and the effectiveness of the vaccine is high”.[12]

Production

The vaccine was first manufactured by Kazakhstan’s Research Institute of Biological Safety Problems. Production capacity has been capped at 50,000 doses per month.

Beginning in June 2021, the vaccine is slated[13] to be packaged in large bulk to be bottled in Turkey by a major Turkish company.[14][15] This will allow for a production capacity of 500,000-600,000 doses per month.[16] The contract is still being negotiated,[17] despite earlier claims that suggesting the deal had already been finalized.[18][19]

Vaccine innoculation

The first batch of 50,000 doses was delivered on 26 April 2021, and vaccination began shortly after.[20] In June 2021, the capacity will increase to 100,000 doses per month, regardless of the contract for bottling in Turkey.[21]

Authorization

   Full authorization  Emergency authorization

See also: List of COVID-19 vaccine authorizations § QazCovid-in

Characteristics

The vaccine can be stored at standard refrigeration temperatures (2°C-8°C) and is a two-dose régime with the doses administered twenty-one days apart.[22]

References

  1. ^ “Kazakhstan: Officials under fire over vaccination failures | Eurasianet”eurasianet.org. Retrieved 11 April 2021.
  2. ^ INFORM.KZ (31 March 2021). “Vaccination with homegrown QazVac vaccine likely to start in late April”http://www.inform.kz. Retrieved 11 April 2021.
  3. ^ Yergaliyeva A (20 December 2020). “Kazakhstan Begins Vaccinating 3,000 Volunteers With Self-Made QazCovid-in”The Astana Times. Retrieved 2 March2021.
  4. ^ Clinical trial number NCT04691908 for “Immunogenicity, Efficacy and Safety of QazCovid-in® COVID-19 Vaccine” at ClinicalTrials.gov
  5. ^ “Reactogenicity, Safety and Immunogenicity of QazCovid-in® COVID-19 Vaccine – Full Text View – ClinicalTrials.gov”clinicaltrials.gov.
  6. ^ “Kazakh Biosafety Research Institute Begins Clinical Trials of Another Vaccine Against COVID-19”. The Astana Times.
  7. ^ INFORM.KZ (31 March 2021). “Vaccination with homegrown QazVac vaccine likely to start in late April”http://www.inform.kz. Retrieved 11 April 2021.
  8. ^ “QazVac готова и уже на подходе”Время (in Russian). Retrieved 11 April2021.
  9. ^ INFORM.KZ (9 April 2021). “3rd stage of clinical trials of QazCovid-in vaccine to be 50% complete by Apr 15”http://www.inform.kz. Retrieved 11 April 2021.
  10. ^ “Kazakhstan’s COVID-19 vaccine to be bottled in Turkey”http://www.aa.com.tr. Retrieved 11 April 2021.
  11. ^ tengrinews.kz (9 April 2021). “Как правильно применять казахстанскую вакцину QazVac, рассказал ученый”Главные новости Казахстана – Tengrinews.kz (in Russian). Retrieved 11 April 2021.
  12. ^ “QazVac готова и уже на подходе”Время (in Russian). Retrieved 11 April2021.
  13. ^ It’s unclear at which level of preparation the vaccine will be send to Turkey.
  14. ^ MENAFN. “Kazakh COVID-19 vaccine to be bottled in Turkey”menafn.com. Retrieved 11 April 2021.
  15. ^ “QazVac готова и уже на подходе”Время (in Russian). Retrieved 11 April2021.
  16. ^ “Kazakhstan Launches Production of First Homegrown Vaccine, ‘QazVac'”caspiannews.com. Retrieved 26 April 2021.
  17. ^ INFORM.KZ (21 April 2021). “Healthcare Ministry comments on production of QazVac vaccine”http://www.inform.kz. Retrieved 22 April 2021.
  18. ^ “К концу апреля в Казахстане будет выпущено 50000 доз собственной вакцины”“СНГ СЕГОДНЯ” – последние новости стран СНГ читайте на SNG.TODAY. Retrieved 12 April 2021.
  19. ^ “Kazakhstan’s COVID-19 vaccine to be bottled in Turkey”http://www.aa.com.tr. Retrieved 12 April 2021.
  20. ^ contributor, Guest (26 April 2021). “Kazakhstan launches QazVac, its own COVID-19 vaccine”EU Reporter. Retrieved 26 April 2021.
  21. ^ “Казахстанскую вакцину QazVac будут разливать в Турции”informburo.kz(in Russian). 9 April 2021. Retrieved 12 April 2021.
  22. ^ INFORM.KZ (26 April 2021). “Health Minister Alexei Tsoi to be one of the first to get homegrown QazCovid-in vaccine”http://www.inform.kz. Retrieved 26 April 2021.

External links

Scholia has a profile for QazCovid-in (Q99518269).

The QazCovid-in vaccine, an inactivated vaccine, was developed and tested in the Kazakh Research Institute for Biological Safety Problems1. It demonstrated high efficacy, safety, and immunogenicity at 96% in initial Phase I and II trials (NCT04530357), and will now be undergoing upcoming Phase III trials2,3.

  1. The Astana Times: Kazakhstan Begins Vaccinating 3,000 Volunteers With Self-Made QazCovid-in [Link]
  2. The Lancet: COVID-19 response in central Asia [Link]
  3. Economic Research Institute: QazCovid-in [Link]
Vaccine description
TargetSARS-CoV-2
Vaccine typeInactivated
Clinical data
Routes of
administration
Intramuscular
Identifiers
DrugBankDB16441
Part of a series on the
COVID-19 pandemic
COVID-19 (disease)SARS-CoV-2 virus (variants)
showTimeline
showLocations
showInternational response
showMedical response
showImpact
 COVID-19 portal

///////////QazVac, COVID 19, vaccine, QazCovid-in, kazakhastan, SARS-CoV-2, corona virus

NEW DRUG APPROVALS

one time

$10.00

COVIran Barakat


Vaccine description
TargetSARS-CoV-2
Vaccine typeInactivated
Clinical data
Routes of
administration
Intramuscular
ATC codeNone
Legal status
Legal statusEmergency use authorization: IRNFull and Emergency Authorizations: List of COVIran Barakat COVID-19 vaccine authorizations

COVIran Barakat

COVIran Barakat, is an inactivated virus vaccine developed by Shifa Pharmed Industrial Co in Iran.[501

https://en.trend.az/iran/society/3439812.html

BAKU, Azerbaijan, Jun. 14 2021

By Elnur Baghishov – Trend:

An emergency license was issued for the use of the Iranian-made “CovIran Barakat” vaccine against the coronavirus yesterday on June 13, the Iranian Minister of Health and Medical Education Saeed Namaki said, Trend reports citing IRNA.

He made the remark in an event dedicated to the launch of a number of health and medical facilities in Iran’s Markazi Province today on June 14.

Namaki said that moreover, a license for the using of the Iranian-made “Pastor” vaccine against the coronavirus will be issued next week.

“Also, the licenses for the using of Iranian-made “Razi” and “Fakhra” vaccines will be issued in the near future,” he added.

According to the minister, the Iranian population will be vaccinated fully by the end of autumn with the opportunities created in connection with the production of vaccines in Iran.

Reportedly, about 10 million people in Iran are planned to be vaccinated with the “CovIran Barakat’ vaccine next week. The production of “CovIran Barakat” vaccine in Iran is expected to reach 50 million doses per month by the end of the summer.

On June 14, 26 health and medical facilities were launched in Iran’s Markazi Province. A total of 1.45 trillion rials (about $34.5 million) has been spent on these facilities.

Iran continues to monitor the coronavirus situation in the country. According to recent reports from Iranian officials, over 3.03 million people have been infected, and 82,217 people have already died.

Meanwhile, over 2.66 million people have reportedly recovered from the disease.

The country continues to apply strict measures to contain further spread. Reportedly, the disease was brought to Iran by a businessman from Iran’s Qom city, who went on a business trip to China, despite official warnings. The man died later from the disease.

The Islamic Republic only announced its first infections and deaths from the coronavirus on Feb. 19.

The outbreak in the Chinese city of Wuhan – which is an international transport hub – began at a fish market in late December 2019.

The World Health Organization (WHO) on March 11 declared COVID-19 a pandemic. Some sources claim the coronavirus outbreak started as early as November 2019.

A total of 5.2 million people have been vaccinated in Iran so far. About 4.35 million people were vaccinated on the first stage, and 851,000 people were vaccinated on the second stage.

COVIran Barakat is a COVID-19 vaccine developed by Iranian state-owned Shifa Pharmed Industrial Group. It has successfully been tested on animals and has been approved by the Iran Food and Drug Administration for testing on humans.[1][2][3] Phase 2/3 (II/III) clinical trial began on 13 March 2021,[4] and the first participants were inoculated on March 29.[5] Finally, the vaccine consumption license was issued on June 13, 2021.[6] Around 650 people worked in 3 shifts around the clock to develop the vaccine.[7]

Dr. Minoo Mohraz has been selected as the lead of the “Corona vaccine project in Iran”.[8] Dr. Mohraz is an Iranian physician, scientist, and AIDS specialist. She is a Full Professor (Emeritus) of Infectious Diseases at Tehran University of Medical Sciences and head of the Iranian Centre for HIV/AIDS.[9] Dr. Mohraz has also served as within the World Health Organization as an expert on HIV/AIDS in Iran and the Eastern Mediterranean.[10]

This vaccine has been authorised for emergency use by the Iranian authorities. This makes it the first locally developed to be approved for emergency use in the Middle East.[11]

Technology

On 29 December 2020, human trials of Iran’s first domestic COVID-19 vaccine candidate were started. The mechanism of production of this vaccine is based on the inactivated vaccine. In other words, “it is made of a coronavirus that has been weakened or killed by chemicals, similar to how polio immunizations are made.”[12]

Development

Iran’s first domestic COVID-19 vaccine candidate was started

Tayyebeh Mokhber, the first volunteer who receives a shot of COVIran Barakat was the daughter of Mohammad Mokhber director of setad. Minister of Health Saeed Namaki and Vice President for Science and Technology Sorena Sattari participated at the ceremony of vaccine injection. According to reports, there are more than 65,000 Iranians volunteered to test the vaccine and 56 selected people took part in the first phase of human trials which last 45 to 60 days.[13] The initial phase of human-testing for this vaccine started with the injection of 56 volunteers who were at the age of 18-50.[14][15][16]

The second/third group of volunteers were also injected with the vaccine.[17][18] According to the head of the vaccine production team at the Setad, the results show that this vaccine also neutralizes the British mutated COVID-19 virus.[19][20][21]

In March 2021, the Executive Office of Imam Khomeini’s Order began a Phase II–III clinical trial of COVIran Barakat with 280 participants in cities including Tehran, Mashhad, Karaj, Esfahan, Shiraz. According to the allowance of medical equipment department, the second phase coincided with third phase.[22][23] The vaccine has reached its third phase of human-testing;[24] and the first injection(s) of the 3rd phase began 25 April 2021.[25]

As official in charge of manufacturing Iran Barakat vaccines, Mohammad Reza Salehi said, “some neighboring countries tend to enter the third phase of the clinical trial of the Iranian “COVIran Barakat””. They are reviewing recommendations to let them participate.[14]

Production

According to Setad (the Executive Headquarters of Imam’s Directive), under the direct control of the Supreme Leader of Iran, “production of the vaccine developed by one of its companies, Shifa Pharmed, could reach 12 million doses per month, six months after a successful trial ends”.[26] On 15 March 2021, he stated that EIKO has already a capacity of three million doses per month and that by end of June the capacity will be 15-20 million doses per month.[27][28]

On 29 March 2021, the Tehran Times reported that a capacity of three million doses per month was achieved;[citation needed] and the production line of 25 million doses per month of Iran Koo vaccine was discharged on 26 April 2021.[29]

On 10 May 2021, the first product of mass production of the Iranian corona vaccine called “COVIran Barakat” was unveiled in phase one of the vaccine production factory associated with Execution of Imam Khomeini’s Order (EIKO). Therefore, 2 industrial lines have been set up. The first production line is prepared and the second line is being prepared. By the end of September (taking into account the capacity of three million doses of the first line), 20 million doses of Iran Barakat vaccine will be available in the month.[30]

Authorizations

 
  Full authorization  Emergency authorization

See also: List of COVID-19 vaccine authorizations § COVIran Barakat

References

  1. ^ “Iranians demand a COVID-19 vaccine, not politics, from their leaders”Los Angeles Times. 19 January 2021.
  2. ^ “Coronavirus Tzar Forced to Apologize to Clergy”iranwire.
  3. ^ Vahdat, Amir (29 December 2020). “Iran begins human trials for locally made coronavirus vaccine”Times of Israel. Retrieved 30 December 2020.
  4. ^ “IRCT | A double-blind, randomized, placebo-controlled Phase II/III Clinical trial to evaluate the safety and efficacy of COVID-19 inactivated vaccine (Shifa-Pharmed) in a population aged 18 to 75 years”en.irct.ir. Retrieved 2021-04-07.
  5. ^ 3080 (2021-03-30). “Some foreign states willing to cooperate in COVIran Barakat clinical test: Official”IRNA English. Retrieved 2021-04-07.
  6. ^ The vaccine consumption license was issued yjc.ir Retrieved 16 June 2021
  7. ^ دانش فنی واکسن برکت صد درصد ایرانی است/ تلاش ۶۵۰ نفر در ساخت واکسن ایرانی کرونا
  8. ^ “Iranian corona vaccine will arrive by ‘next July'”Persian Bibi (in Persian). 2020-12-06. Retrieved 2020-12-18.
  9. ^ “Good News About AIDS”Young Journalists Club. February 6, 2016. Retrieved 2020-02-26.
  10. ^ “Allameh Tabatabai | Professor Minoo Mehrz: Everything I have is from Tehran University of Medical Sciences”. Tehran University of Medical Sciences Alumni Communication Office. Retrieved March 29, 2017.
  11. ^ “Iran issues license on its coronavirus vaccine”Trend.Az. 2021-06-14. Retrieved 2021-06-14.
  12. ^ “Iran begins first human trial of locally made virus vaccine”health.economictimes.indiatimes. 29 December 2019.
  13. ^ “COVIran Barakat: Iran launches human trials of its COVID vaccine”aljazeera. 29 December 2019.
  14. Jump up to:a b “Some foreign states willing to cooperate in COVIran Barakat clinical test: Official”irna.
  15. ^ Human test of Iranian corona vaccine begins / Minister of Health: We are the first vaccinator in Asia with 100 years of experience tasnimnews.com Retrieved 29 December 2020
  16. ^ End of the injection of phase one (studies) of “Kovoo-Iran Barakat” vaccineyjc.ir Retrieved 16 February 2021
  17. ^ Start of injecting the Iranian corona vaccine to the second group of volunteersmehrnews.com
  18. ^ The injection of “Iranian corona vaccine” to the second group of volunteers began tasnimnews.com
  19. ^ Jalili: The Iranian vaccine neutralizes the British virus yjc.ir
  20. ^ Iranian vaccine succeeds in neutralizing “British mutated virus” isna.ir
  21. ^ “Iran Vaccine Boasts Total Protection Against U.K. Covid Strain”bloomberg.
  22. ^ “Clinical trials of COVIRAN vaccine enter phases 2, 3”isna. 15 March 2021.
  23. ^ “واکسن ایران برکت احتمالا تا پایان خرداد ۱۴۰۰ به دست هموطنان می‌رسد”IRNA. 24 March 2021.
  24. ^ The beginning of the third stage of the human test of COVIran Barakat IRINN, Retrieved 21 April 2021
  25. ^ The third phase of Iran Barakat vaccine was injected YJC, Retrieved 25 April 2021
  26. ^ “Iran starts human testing of first domestic COVID-19 vaccine”reuters. 29 December 2019.
  27. ^ “Iran starts mass-production of homegrown coronavirus vaccine”Tehran Times. 2021-03-15. Retrieved 2021-04-07.
  28. ^ “Iran to kick off production of 3mn doses of COVIRAN”Mehr News Agency. 2021-03-15. Retrieved 2021-04-07.
  29. ^ The production line of 25 million doses per month of Iran Koo vaccine was cleared, Retrieved 4 May 2021
  30. ^ “نخستین محصول تولید انبوه واکسن “کوو ایران برکت” فردا رونمایی می‌شود”irna. 10 May 2021.

External links

Scholia has a profile for COVIran Barakat (Q105217191).

/////////COVIran Barakat, iran, coronavirus,  COVID-19 vaccine, Shifa Pharmed, SARS-CoV-2

wdt-18

NEW DRUG APPROVALS

one time

$10.00

Convidicea (Ad5-nCoV)


A vial of Convidecia vaccine
Vaccine description
TargetSARS-CoV-2
Vaccine typeViral vector
Clinical data
Trade namesConvidecia
Routes of
administration
IntramuscularIntranasal
ATC codeNone
Legal status
Legal statusFull and Emergency authorizations
Identifiers
DrugBankDB15655

Convidicea (Ad5-nCoV)

Recombinant vaccine (adenovirus type 5 vector)

Recombinant Novel Coronavirus Vaccine (Adenovirus Type 5 Vector)

CanSino Biologics, china

see https://covid19.trackvaccines.org/vaccines/2/

AD5-nCOV, trade-named Convidecia, is a single-dose[1] viral vector vaccine for COVID-19 developed by CanSino Biologics. It conducted its Phase III trials in Argentina,[2] Chile,[3] Mexico,[4] Pakistan,[5] Russia,[6] and Saudi Arabia[7] with 40,000 participants.

In February 2021, global data from Phase III trials and 101 COVID cases showed that the vaccine had a 65.7% efficacy in preventing moderate symptoms of COVID-19, and 91% efficacy in preventing severe disease.[8] It has similar efficacy to Johnson & Johnson’s Ad26.COV2.S, another one-shot adenovirus vector vaccine with 66% efficacy in a global trial.[9][1] Convidecia is similar to other viral vector vaccines like AZD1222Gam-COVID-Vac, and Ad26.COV2.S.[10] Its single-dose regimen and normal refrigerator storage requirement (2°to 8 °C) could make it a favorable vaccine option for many countries.[9]

Convidecia is approved for use by some countries in Asia,[11][12][13] Europe,[14][15] and Latin America.[16][17][18] Production capacity for Ad5-NCov should reach 500 million doses in 2021. Manufacturing will take place in China,[19] Malaysia,[13] Mexico,[20] and Pakistan.[21]

Ad5-nCoV is a recombinant adenovirus type-5 vector (Ad5) vaccine currently being investigated for prophylaxis against SARS-CoV-2.1,2 It is being developed by CanSino Biologics Inc., in partnership with the Beijing Institute of Biotechnology, who in March 2020 announced the approval of a phase I clinical trial (ChiCTR2000030906)1 with an expected completion in December 2020. The study will evaluate antibody response in healthy patients between the ages of 18 and 60 who will receive one of three study doses, with follow-up taking place at weeks 2 and 4 and months 3 and 6 post-vaccination.2

  1. Chinese Clinical Trial Register: A phase I clinical trial for recombinant novel coronavirus (2019-COV) vaccine (adenoviral vector) [Link]
  2. Antibody Society: COVID-19 Archives [Link]

Technology

Convidecia is a viral vector vaccine similar to AstraZeneca‘s AZD1222 and Gamaleya‘s Gam-COVID-Vac.[10] Ad5-nCOV can be stored in less extreme cold conditions compared to mRNA vaccines.[22][9]

Efficacy

In February 2021, data released from an interim analysis of Phase III trials with 30,000 participants and 101 COVID cases showed that globally, the vaccine had an efficacy of 65.7% at preventing moderate cases of COVID-19 and 90.98% efficacy at preventing severe cases. In the Pakistan trial subset, the vaccine had an efficacy of 74.8% at preventing symptomatic cases 100% for preventing severe disease.[8]

While the efficacy rates were lower than the Pfizer–BioNTech and Moderna vaccines, its single-dose regimen and normal refrigerator storage requirement (2 to 8 °C) could make it a favorable option for many countries. It has similar efficacy to Johnson & Johnson’s Ad26.COV2.S, another one-shot adenovirus vaccine found to be 66% effective in a global trial.[9][1]

Clinical trials

Phase I-II

In early 2020, Chen Wei led a joint team of the Institute of Biotechnology, the Academy of Military Medical Sciences and CanSino Biologics to develop AD5-nCOV. According to the Chinese state media, the team registered an experimental COVID-19 vaccine for Phase I trial in China on 17 March 2020 to test its safety. The trial was conducted on 108 healthy adults aged 18 to 60 in two medical facilities in WuhanHubei province.[23]

In April, Ad5-nCoV became the first COVID-19 vaccine candidate in the world to begin Phase II trials.[24] The Phase II trial results were published in the peer-reviewed journal The Lancet in August 2020, and noted neutralizing antibody and T cell responses based on statistical analyses of data involving 508 eligible participants.[25] In September, Zeng Guang, chief scientist of the Chinese Center for Disease Control and Prevention said the amount of COVID-19 antibodies in subjects from the Phase I trials remained high six months after the first shot. Zeng said the high levels of antibodies suggested the shots may provide immunity for an extended period of time, although Phase III results were still required.[26] On September 24, CanSino began Phase IIb trials on 481 participants to evaluate the safety and immunogenicity of Ad5-nCoV for children ages 6–17 and elderly individuals ages 56 and above.[27]

In August, China’s National Intellectual Property Administration issued the country’s first COVID-19 vaccine patent to CanSino.[28]

On 16 May 2020, Canadian Prime Minister Justin Trudeau announced Health Canada had approved Phase II trials to be conducted by the Canadian Center for Vaccinology (CCfV) on the COVID-19 vaccine produced by CanSino. Scott Halperin, director of the CCfV said the vaccine would not be the only one going into clinical trials in Canada, and any potential vaccine would not be publicly available until after Phase 3 is complete.[29][30] If the vaccine trials were successful, then the National Research Council would work with CanSino to produce and distribute the vaccine in Canada.[30] In August 2020, the National Research Council disclosed the vaccine had not been approved by Chinese customs to ship to Canada, after which the collaboration between CanSino and the Canadian Center for Vaccinology was abandoned.[31]

Nasal spray trials

In September, CanSino began a Phase I trial in China with 144 adults to determine the safety and immunogenicity of the vaccine to be administered as a nasal spray, in contrast with most COVID-19 vaccine candidates which require intramuscular injection.[32] On June 3, 2021, Chen Wei announced the expansion of clinical trials was approved by the NMPA, in the meantime, they are applying for Emergency Use Listing for the nasal spray.[33]

Phase III

In August, Saudi Arabia confirmed it would begin Phase III trials on 5,000 people for Ad5-nCoV in the cities of Riyadh, Dammam, and Mecca.[7]

In October, Mexico began Phase III trials on 15,000 volunteers.[34][4]

In September, Russia began Phase III trials on 500 volunteers,[35] which Petrovax later received approval from the government to expand to 8,000 more volunteers.[36][6]

In September, Pakistan began Phase III trials on 40,000 volunteers as part of a global multi-center study.[5] As of December, about 13,000 volunteers have participated in trials of Ad5-nCoV.[22]

In November, Chile began Phase III trials on 5,200 volunteers to be managed by University of La Frontera.[37][3]

In December, Argentina’s Fundación Huésped began Phase III trials in 11 health centers in the metropolitan area of Buenos Aires and Mar del Plata.[2]

Combination trials

In April 2021, a new trial was registered in Jiangsu involving one dose of Convidecia followed by a dose of ZF2001 28 or 56 days later using different technologies as a way to further boost efficacy.[38]

Manufacturing

In February, Chen Wei who lead the development of the vaccine, said annual production capacity for Ad5-NCov could reach 500 million doses in 2021.[19]

In February, Mexico received the first batch of active ingredients for Convidecia, which is being packaged in Querétaro by Drugmex.[20]

In Malaysia, final filling and packaging of the vaccine for distribution would be completed by Solution Biologics.[13]

In May, Pakistan began filling and finishing 3 million doses a month at the National Institute of Health, which would be branded as PakVac for domestic distribution.[39]

If the vaccine is approved in Russia, Petrovax said it would produce 10 million doses per month in 2021.[40]

Marketing and deployment

 
  Full authorization  Emergency authorization  Eligible COVAX recipient (ongoing assessment)[41]

See also: List of COVID-19 vaccine authorizations § Convidecia

Asia

On 25 June 2020, China approved the vaccine for limited use by the military.[42] In February 2021, China approved the vaccine for general use.[11]

In February, Malaysia‘s Solution Biologics agreed to supply 3.5 million doses to the government.[43] The doses would be delivered starting in April with 500,000 complete doses, with the rest in bulk to be finished by Solution Biologics.[13]

In October, Indonesia reached an agreement with CanSino to deliver 100,000 doses in November 2020, with the expectation that an additional 15 to 20 million doses would be delivered in 2021.[44]

In February, Pakistan approved the vaccine for emergency use.[45] The country purchased 20 million doses of the vaccine[12] of which the first 3 million doses are to arrive in May.[12]

Europe

In March, Hungary granted emergency use approval for the vaccine.[14]

In March, Moldova authorized use of the vaccine.[46]

North America

In December 2020, Mexico‘s Foreign Minister Marcelo Ebrard signed an agreement for 35 million doses.[47] In February, Mexico approved the vaccine for emergency use.[48] Mexico received active ingredients for 2 million doses with a total of 6 million doses expected to arrive in February.[16]

South America

In June, Argentina approved emergency use of the vaccine and ordered 5.4 million doses.[17]

In June, Brazil announced plans to purchase 60 million doses.[49] In May, Brazil began reviewing the vaccine for emergency use.[50]

In March, Chile signed a deal for 1.8 million doses for delivery between May and June,[51] for which emergency use approval was granted in April.[18]

In June, Ecuador approved emergency use and ordered 6 million doses for delivery between June and August 2021.[52]

References

  1. Jump up to:a b c “It’s not just Johnson & Johnson: China has a single-dose COVID-19 vaccine that has 65% efficacy”Fortune. Retrieved 2021-02-11.
  2. Jump up to:a b “Comenzará en la Argentina un nuevo estudio de vacuna recombinante contra el SARS-CoV-2”infobae (in Spanish). 14 December 2020. Retrieved 2020-12-15.
  3. Jump up to:a b “Gob.cl – Article: Science Minister: “We Work With Maximum Rigor So That Science And Technology Benefit People’S Health””Government of Chile. Retrieved 2020-11-21.
  4. Jump up to:a b “Chinese Covid vaccine trials to be expanded to five more states”Mexico News Daily. 2020-11-10. Retrieved 2020-11-11.
  5. Jump up to:a b “Phase III Trial of A COVID-19 Vaccine of Adenovirus Vector in Adults 18 Years Old and Above – Full Text View – ClinicalTrials.gov”clinicaltrials.gov. Retrieved 2020-10-21.
  6. Jump up to:a b Reuters Staff (2020-12-07). “Russia approves clinical trials for Chinese COVID-19 vaccine Ad5-Ncov: Ifax”Reuters. Retrieved 2020-12-07.
  7. Jump up to:a b Eltahir N (9 August 2020). “CanSino to start Phase III trial of COVID-19 vaccine in Saudi”Reuters. Retrieved 9 August 2020.
  8. Jump up to:a b “CanSinoBIO’s COVID-19 vaccine 65.7% effective in global trials, Pakistan official says”Reuters. 8 February 2021. Retrieved 2021-02-08.
  9. Jump up to:a b c d “China’s CanSino Covid Vaccine Shows 65.7% Efficacy”Bloomberg.com. 2021-02-08. Retrieved 2021-02-10.
  10. Jump up to:a b Zimmer C, Corum J, Wee SL (2020-06-10). “Coronavirus Vaccine Tracker”The New York TimesISSN 0362-4331. Retrieved 2020-12-12.
  11. Jump up to:a b Liu R (2021-02-25). “China approves two more domestic COVID-19 vaccines for public use”Reuters. Retrieved 2021-02-26.
  12. Jump up to:a b c “Pakistan purchases over 30 million COVID doses from China: sources”ARY NEWS. 2021-04-25. Retrieved 2021-04-26.
  13. Jump up to:a b c d “Malaysia to receive CanSino vaccine this month | The Malaysian Insight”http://www.themalaysianinsight.com. Retrieved 2021-04-03.
  14. Jump up to:a b Ashok R (2021-03-22). “UPDATE 2-China’s CanSino Biologics COVID-19 vaccine receives emergency use approval in Hungary”Reuters. Retrieved 2021-03-22.
  15. ^ “Membrii NITAG au venit cu recomandări privind utilizarea vaccinurilor împotriva COVID-19 în Republica Moldova”Ministerul Sănătății, Muncii și Protecţiei Sociale. 2021-03-03. Retrieved 2021-05-21.
  16. Jump up to:a b “‘Our gratitude always’: From China’s CanSino, Mexico welcomes biggest vaccine shipment yet”Reuters. 2021-02-11. Retrieved 2021-02-11.
  17. Jump up to:a b “Argentina issues emergency approval to China’s single-dose Cansino COVID-19 vaccine”Reuters. 2021-06-11. Retrieved 2021-06-11.
  18. Jump up to:a b “ISP Approves Emergency Use And Importation Of Cansino Vaccine To Fight COVID-19”Institute of Public Health of Chile. Retrieved 2021-04-08.
  19. Jump up to:a b “China can hit 500-mln-dose annual capacity of CanSinoBIO COVID-19 vaccine this year”finance.yahoo.com. Retrieved 2021-02-28.
  20. Jump up to:a b Solomon DB (2021-02-28). “China’s CanSino says first vaccines packaged in Mexico will be ready in March”Reuters. Retrieved 2021-03-12.
  21. ^ “Pakistan develops homemade anti-Covid vaccine ‘PakVac'”The Express Tribune. 2021-05-24. Retrieved 2021-05-25.
  22. Jump up to:a b Constable P, Hussain S. “Defying fears and skepticism, thousands in Pakistan volunteer for Chinese vaccine trials”The Washington PostISSN 0190-8286. Retrieved 2021-01-01.
  23. ^ Cui J (23 March 2020). “Human vaccine trial gets underway”China Daily. Retrieved 18 April 2020.
  24. ^ Xie J (15 April 2020). “China Announces Phase 2 of Clinical Trials of COVID-19 Vaccine”Voice of America. Retrieved 18 April2020.
  25. ^ Zhu FC, Guan XH, Li YH, Huang JY, Jiang T, Hou LH, et al. (August 2020). “Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: a randomised, double-blind, placebo-controlled, phase 2 trial”Lancet396 (10249): 479–488. doi:10.1016/S0140-6736(20)31605-6PMC 7836858PMID 32702299.
  26. ^ O’Brien E (2020-09-25). “Covid Antibodies Endure Over Six Months in China Trial Subjects”http://www.bloomberg.com. Retrieved 2020-09-29.
  27. ^ “Phase IIb Clinical Trial of A COVID-19 Vaccine Named Recombinant Novel Coronavirus Vaccine (Adenovirus Type 5 Vector) – Full Text View – ClinicalTrials.gov”clinicaltrials.gov. Retrieved 2020-10-21.
  28. ^ Yu S (17 August 2020). “China grants country’s first COVID-19 vaccine patent to CanSino: state media”Reuters. Retrieved 17 August 2020.
  29. ^ Bogart N (16 May 2020). “Health Canada approves first clinical trial for potential COVID-19 vaccine”CTV News. Retrieved 7 September 2020.
  30. Jump up to:a b Ryan H (May 16, 2020). “Canada’s first COVID-19 vaccine trials approved for Halifax university”CBC News. Retrieved January 4, 2021.
  31. ^ Cooke A (26 August 2020). “Canadian COVID-19 clinical trial scrapped after China wouldn’t ship potential vaccine”CBC News. Retrieved 7 September 2020.
  32. ^ “A Clinical Trial of a Recombinant Adenovirus 5 Vectored COVID-19 Vaccine (Ad5-nCoV) With Two Doses in Healthy Adults – Full Text View – ClinicalTrials.gov”clinicaltrials.gov. Retrieved 25 September 2020.
  33. ^ Cao X, Liu Y (2021-06-04). “陈薇院士:雾化吸入式新冠疫苗正在申请紧急使用”Sci Tech Daily. Chinanews.com. Retrieved 2021-06-04.
  34. ^ “México recibe el primer lote de la vacuna candidata de CanSino Biologics; alistan pruebas”EL CEO (in Spanish). 2020-11-03. Retrieved 2020-11-03.
  35. ^ “Clinical Trial of Recombinant Novel Coronavirus Vaccine (Adenovirus Type 5 Vector) Against COVID-19 – Full Text View – ClinicalTrials.gov”clinicaltrials.gov. Retrieved 2020-10-21.
  36. ^ Bloomberg News (2020-11-25). “Russia’s Richest Man Seeks Global Market for Local Covid-19 Drug – BNN Bloomberg”BNN. Retrieved 2020-11-28.
  37. ^ Yáñez PL (2020-11-15). “Así funcionan las cuatro vacunas que se probarán en Chile”La Tercera. Retrieved 2020-11-17.
  38. ^ “China trials mixing of CanSinoBIO’s and Zhifei Longcom’s COVID-19 vaccines -data”Reuters. 2021-04-19. Retrieved 2021-06-16.
  39. ^ “Covid vaccine: Pakistan starts production of CanSino, China’s single-dose jab”Khaleej Times. Retrieved 2021-05-28.
  40. ^ “Russian Recruits Show ‘No Side Effects’ in Chinese Coronavirus Vaccine Trials”The Moscow Times. 2020-09-21. Retrieved 2020-09-22.
  41. ^ “Status of COVID-19 Vaccines within WHO EUL/PQ evaluation process”World Health Organization (WHO).
  42. ^ Reuters Staff (2020-06-29). “CanSino’s COVID-19 vaccine candidate approved for military use in China”Reuters. Retrieved 2020-12-13.
  43. ^ Reuters Staff (2021-02-04). “Malaysia’s Solution Group to supply 3.5 million doses of CanSino vaccine to government”Reuters. Retrieved 2021-02-04.
  44. ^ Taufiqurrahman M. “Indonesia can be manufacutring hub for COVID-19 vaccine, says Chinese foreign minister”. Jakarta Post. Retrieved 13 October 2020.
  45. ^ Shahzad A (2021-02-12). “Pakistan approves Chinese CanSinoBIO COVID vaccine for emergency use”Reuters. Retrieved 2021-02-12.
  46. ^ “Membrii NITAG au venit cu recomandări privind utilizarea vaccinurilor împotriva COVID-19 în Republica Moldova”Ministerul Sănătății, Muncii și Protecţiei Sociale. 2021-03-03. Retrieved 2021-05-21.
  47. ^ Reuters Staff (2020-12-10). “Mexico agrees to buy 35 million doses of CanSino COVID vaccine”Reuters. Retrieved 2020-12-10.
  48. ^ “Mexico approves China’s CanSino and Sinovac COVID-19 vaccines”Reuters. 10 February 2021.
  49. ^ “Brazil to buy single-shot Chinese COVID-19 vaccine”Reuters. 2021-06-15. Retrieved 2021-06-16.
  50. ^ “Brazil in vaccine talks with Moderna, reviewing CanSino shot”Reuters. 2021-05-19. Retrieved 2021-05-21.
  51. ^ Sherwood D (2021-03-30). “Chile inks deal for 1.8 million doses of CanSino COVID-19 vaccine as inoculation drive plows ahead”Reuters. Retrieved 2021-03-30.
  52. ^ Valencia A. “Ecuador authorizes use of China’s CanSino vaccine against COVID-19”Reuters. Retrieved 2021-06-16.

Further reading

External links

Scholia has a profile for Ad5-nCoV (Q96695265).

/////////Convidicea, Ad5-nCoV, Recombinant vaccine, adenovirus type 5 vector, CanSino Biologics, china, SARS-CoV-2, corona virus, vaccine, covid 19

Convidecia

 
  Full authorization  Emergency authorization  Eligible COVAX recipient (ongoing assessment)[2]

Convidecia is a viral vector vaccine[478] produced by the Chinese company CanSino Biologics and the Beijing Institute of Biotechnology of the Academy of Military Medical Sciences.Full (1)

  1. China[479]

Emergency (8)

  1. Argentina[480]
  2. Chile[481]
  3. Ecuador[482]
  4. Hungary[483][272]
  5. Malaysia[484]
  6. Mexico[436]
  7. Moldova[229]
  8. Pakistan[485]

wdt-17

NEW DRUG APPROVALS

one time

$10.00

EpiVacCorona


Russia approves 2nd coronavirus vaccine "EpiVacCorona"

Origin of EpiVacCorona antigenes

  1. MKIEEGKLVIWINGDKGYNGLAEVGKKFEKDTGIKVTVEHPDKLEEKFPQVAATGDGPDIIFWAHDRFGGYAQSGLLAEITPDKAFQDKLYPFTWDAVRYNGKLIAYPIAVEALSLIYNKDLLPNPPKTWEEIPALDKELKAKGKSALMFNLQEPYFTWPLIAADGGYAFKYENGKYDIKDVGVDNAGAKAGLTFLVDLIKNKHMNADTDYSIAEAAFNKGETAMTINGPWAWSNIDTSKVNYGVTVLPTFKGQPSKPFVGVLSAGINAASPNKELAKEFLENYLLTDEGLEAVNKDKPLGAVALKSYEEELAKDPRIAATMENAQKGEIMPNIPQMSAFWYAVRTAVINAASGRQTVDEALKDAQTNSSSNNNNNNNNNNLGDNGPQNQRNAPRITFGGPSDSTGSNQNGERSGARSKQRRPQGLPNNTASWFTALTQHGKEDLKFPRGQGVPINTNSSPDDQIGYYRRATRRIRGGDGKMKDLSPRWYFYYLGTGPEAGLPYGANKDGIIWVATEGALNTPKDHIGTRNPANNAAIVLQLPQGTTLPKGFYAEGSRGGSQASSRSSSRSRNSSRNSTPGSSRGTSPARMAGNGGDAALALLLLDRLNQLESKMSGKGQQQQGQTVTKKSAAEASKKPRQKRTATKAYNVTQAFGRRGPEQTQGNFGDQELIRQGTDYKHWPQIAQFAPSASAFFGMSRIGMEVTPSGTWLTYTGAIKLDDKDPNFKDQVILLNKHIDAYKTFPPTEPKKDKKKKADETQALPQRQKKQQTVTLLPAADLDDLSKQLQQSMSSADSTQA. “Carrier protein sequence”.

EpiVacCorona

Federal Budgetary Research Institution State Research Center of Virology and Biotechnology

peptide, russia

PATENT https://www.fips.ru/registers-doc-view/fips_servlet?DB=RUPAT&DocNumber=2743594&TypeFile=htmlRU 2 743 594 RU 2 743 593RU 2 743 595 RU 2 738 081 Science (Washington, DC, United States) (2021), 372(6538), 116-117. 

EpiVacCorona (Russian: ЭпиВакКорона, tr. EpiVakKorona) is a peptide-based vaccine against COVID-19 developed by the VECTOR center of Virology.[1][2][3] It consists of three chemically synthesized peptides (short fragments of a viral spike protein) that are conjugated to a large carrier protein. This protein is a fusion product of a viral nucleocapsid protein and a bacterial MBP protein.The third phase of a clinical trial, which should show whether the vaccine is able to protect people from COVID-19 or not, was launched in November 2020 with more than three thousand participants.[2] It is assumed it will be completed in August 2021.[2] According to the vaccine developers, the peptides and the viral part of the chimeric protein should immunize people who received this vaccine against SARS-CoV-2 and trigger the production of protective antibodies. However, some experts in the field have expressed concerns about the selection of peptides for use as vaccine antigens.[3][4] In addition, there are also serious concerns about the vaccine immunogenicity data, which have fueled independent civic research efforts[5][6][7] and criticism by some experts.[3][8][4][9][10] Meanwhile, the EpiVacCorona has received vaccine emergency authorization in a form of government registration and is available for vaccination outside the clinical trials.[11] The vaccine delivered via intramuscular route and aluminum hydroxide serves as an immunological adjuvant.

Description[edit]

Origin of EpiVacCorona antigenes

Composition

The vaccine includes three chemically synthesized short fragments of the viral spike protein – peptides, which, according to the developers of EpiVacCorona represent the protein regions containing B-cell epitopes that should be recognized by the human immune system.

These peptides are represented by following amino acid sequences:

1) CRLFRKSNLKPFERDISTEIYQAGS, 2) CKEIDRLNEVAKNLNESLIDLQE, 3) CKNLNESLIDLQELGKYEQYIK.[1][12][13]

In the vaccine all peptides are conjugated to a carrier protein, which is an expression product of the chimeric gene. This chimeric gene was created by fusion of two genes originating from different organisms, namely a gene encoding a viral nucleocapsid protein and a gene encoding a bacterial maltose-binding protein (MBP). The fusion chimeric gene expressed in Escherichia coli. The sequence of the chimeric protein is available from the patent.[4] The genetic construct of the chimeric gene also includes a short genetic fragment encoding a polyhistidine-tag, which is used to purify the chimeric protein from E. coli lysate. After the purification, the protein is conjugated with three peptides in a way that only one variant of the peptide molecule is attached to each protein molecule. As a result, three types of conjugated molecules are created: chimeric protein with attached peptide number 1, the same protein with peptide number 2, and finally the same protein with peptide number 3. All three types of conjugated molecules are included in the vaccine.[citation needed]

EpiVacCorona: antigens origin and composition

Vaccine antigens and antibodies

According to the developers’ publications,[14][5][6] vaccine antigens are three peptides of the spike protein and a chimeric protein consisting of two parts (viral nucleocapsid protein and bacterial maltose-binding protein). In addition, the polyhistidine-tag – a short peptide that is introduced into a vaccine composition to purify a chimeric protein from a bacterial lysate – is also a vaccine antigen against which antibodies can form in those who have received the vaccine. A person vaccinated with EpiVacCorona can develop antibodies not only to the peptides of the spike protein, but also to other antigens present in the vaccine. According to Anna Popova who is a head of the Federal Service for Supervision of Consumer Rights Protection and Human Welfare, it takes 42 days for those vaccinated with EpiVacCorona to develop immunity.[15]

figure2

Development

Immunogenic peptide screening in rabbits for EpiVacCorona design

Preclinical studies

The primary screening of peptides for the search for the most immunogenic ones was carried out in animals. The level of antibodies that was triggered by each tested peptide after administration to rabbits was measured. In the test, hemocyanin protein was used as a carrier protein for the studied peptides. Further, on six species of animals (mice, rats, rabbits, African green monkeys, rhesus monkeys, guinea pigs), the vaccine was shown to be harmless in terms of such parameters as general toxicity, allergic properties, and mutagenic activity. In four species of animals (hamsters, ferrets, African green monkeys, rhesus monkeys), specific activity was shown: immunogenicity and protective properties against SARS-CoV-2. The main results of preclinical studies are published in the “Bulletin of the Russian Academy of Medical Sciences”.[12][13]

Clinical studies

The studies development timeline was reported in Russian media in January 2021.[16] There are currently two clinical trials of EpiVacCorona registered in the ClinicalTrials.gov database.[17][18][2]

Phase I-II

The trial “Study of the Safety, Reactogenicity and Immunogenicity of “EpiVacCorona” Vaccine for the Prevention of COVID-19 (EpiVacCorona)”[18] was registered in clinical trial database with ClinicalTrials.gov identifier: NCT04780035. Another trial with the same title was registered with ClinicalTrials.gov Identifier: NCT04527575. Results of the trial that included data on 86 participants were published in Russian Journal of Infection and Immunity, indicating preliminary evidence of safety and an immune response.[1] The publication reports preliminary results of the first two phases of clinical trials of the vaccine in volunteers, of which 14 people aged 18-30 years participated in the first phase, and 86 volunteers aged 18-60 years in the second phase. It is claimed that antibodies were formed in 100% of the volunteers, and the vaccine is also claimed to be safe.[1]

EpiVacCorona Vaccine Development Timeline

Phase III

The third phase of a clinical trial, which should show whether the vaccine is able to protect people from COVID-19 or not, was launched in November 2020 with more than three thousand participants planned. It is expected to be completed in September 2021.[2] In the clinical trials database the phase III trial etitled “Study of the Tolerability, Safety, Immunogenicity and Preventive Efficacy of the EpiVacCorona Vaccine for the Prevention of COVID-19[2]” was registered only in March 2021 with ClinicalTrials.gov Identifier: NCT04780035. Phase 3-4 trial was registered in Russia at 18.11.2020 with 4991 participants planned.[19]

Intellectual property

The following patents of the Russian Federation for invention have been published, which protect the EpiVacCorona vaccine:

Peptide immunogens and vaccine composition against coronavirus infection COVID-19 using peptide immunogens” (No. 2738081). There are 7 peptides in patented vaccine compositions.

Peptide immunogens and vaccine composition against coronavirus infection COVID-19 using peptide immunogens” (No. 2743593). The patented vaccine composition contains 2 peptides.

Peptide immunogens used as a component of a vaccine composition against coronavirus infection COVID-19″ (No. 2743594). The patented vaccine composition contains 3 peptides.

Vaccine composition against coronavirus infection COVID-19″ (No. 2743595). The patented vaccine composition contains 3 peptides.

In all of these patents, the carrier protein is referred to as a chimeric fusion protein with an amino acid sequence derived from two parts, a bacterial maltose binding protein and a viral nucleocapsid protein.[20]

EpiVacCorona vaccine registration certificate

Authorization

 
  Full authorization  Emergency authorization

See also: List of COVID-19 vaccine authorizations § EpiVacCorona

The VECTOR has received vaccine emergency authorization in a form of government registration in October 2020.[21]

In Russia phase III clinical study is called post-registration study. Therefore, government registration of the vaccine means permission to perform phase III clinical research and public vaccination outside of clinical trials as well.[21] Since December 2020, the vaccine has been released for public vaccination in Russia.[22]

As of March 2021, Turkmenistan is the only foreign state to register EpiVacCorona with full authorization.[23][24]

Russia’s Chief Health Officer Anna Popova said: “In December 2020 the EpiVacCorona documents were presented to the World Health Organization, and we are expecting a decision from WHO.”[25] However, Deutsche Welle reports “As of March 1, the WHO had yet to receive an Expression of Interest (EOI) from EpiVacCorona’s developers, “VECTOR,” to enable WHO experts to evaluate their vaccine.”[26]

Export

The Deputy Director-General of the World Health Organization (WHO) Dr. Soumya Swaminathan during news conference in Geneva that took place in October 2020, told: “We will only be able to have a position on a vaccine when we see results of the phase III clinical trials.”[27] According to the center’s director Rinat Maksyutov, many government and non-government organizations want to test or be involved in the production of the vaccine.[28] As of March 30, Venezuela obtained 1000 doses of the Russian EpiVacCorona vaccine for a trial.[29] Venezuela also has reached a deal to purchase doses of the vaccine, as well as manufacture it locally, Vice President Delcy Rodriguez provided this information on June 4, 2021.[30] Turkmenistan expects to receive EpiVacCorona, as the vaccine has already been approved for use in that country.[31]

Controversy

Independent study of clinical trial participants

Ministry of Health’s response to a request from trial participants to perform independent antibody screening tests

English translation of Ministry of Health’s response to a request from trial participants to perform independent antibody screening tests.

At the start of the Phase III, trial participants and those vaccinated outside the trial began to form a community through the Telegram messenger network. On January 18, 2021, the members of the community turned to the Ministry of Health of the Russian Federation with an open letter, in which they stated that the production of antibodies after vaccination among them is much lower than declared by vaccine developers. Study participants claimed that antibodies were not found in more than 50% of those who documented their participation in the study, although only 25% of the participants should have had a placebo according to the study design. The trial participants also claimed that negative results were obtained using the a special ELISA test developed and recommended by VECTOR for EpiVacCorona detection.[5][6][4] More questions about the quality and protectiveness of antibodies induced by EpiVacCorona appeared along with the first results of a special antibody VECTOR’s test, when, with a positive special test, negative results of all other commercially available tests were otained: LIAISON SARS-CoV-2 S1 / S2 IgG – DiaSorin, IgM / IgG – Mindray, SARS-CoV-2 IgG – Abbott Architect, Anti-SARS-CoV-2 ELISA (IgG) – Euroimmun, Access SARS-CoV-2 IgG (RBD) – Beckman Coulter, “SARS-CoV-2-IgG-ELISA -BEST “-” Vector-Best “,” Anti-RBD IgG “- Gamaleya Research Center.[5][6][4][8] Clinical trial participants conducted their own antibody mini-study that was performed in independent Russian laboratory. The study participants asked Dr. Alexander Chepurnov, the former head of the infectious diseases department at VECTOR, who now works at another medical institute, to check neutralizing antibodies presence in their serum samples.[3] They also sent to Dr. Chepurnov control serum samples from former COVID-19 patients or people vaccinated with another Russian vaccine, Sputnik V, which is known to trigger the production of neutralizing antibodies.[32] All serum samples were blinded before antibody tests. On 23 March 2021, the participants reported the results of their mini-study in an open letter to the Ministry of Health of the Russian Federation.[6][7] According to the letter, even with the help of the VECTOR antibody detection system, antibodies were detected only in 70-75% of those vaccinated with EpiVacCorona. However, the level of antibodies was very low. Moreover, according to the letter, virus-neutralizing antibodies were not detected in the independent research Dr. Alexander Chepurnov laboratory at all.[3][6][7] The trial participants asked Ministry of Health in their open letter to perform independent study for the verification of their findings.[3][6][7] In addition, the letter reports 18 cases of COVID-19 cases as of March 22, 2021 among those who received the vaccine and became ill (sometimes severe) three weeks or later after the second dose of EpiVacCorona.[33][6][7] April 20, 2021 the study participants got a reply, with refusal of performing any additional verification antibody tests or investigation of sever COVID-19 cases among vaccinated individuals. The reply include the following text: “Considering that the listed immunobiological preparations (vaccines) for the prevention of COVID-19 are registered in the prescribed manner, their effectiveness and safety have been confirmed.”

Vaccine criticism by independent experts

Some independent experts criticized the vaccine design[3][4] and clinical data presentation in the publication.[8][9][10] The experts are saying that peptide selection is “crucial” for the innovative peptide approach, which VECTOR uses for EpiVacCorona design. However, some researchers are not convinced that the viral spike protein peptides selected for the vaccine are actually “visible” by human immune system.[3][4][34] They stated that these peptides do not overlap[35] with peptides that have been shown in several publications to contain human linear B cell epitopes in spike protein of SARS-CoV-2.[36][37][38][39][40] Moreover, the study was criticized for the lack of positive control of convalescent plasma samples in reports related to neutralizing antibody titers in vaccinated individuals.[1][10] The same study was also criticized for presence of detectable antibodies in negative controls samples that were not discussed by authors.[1][10] In addition, vaccine developers have been criticized for aggressively advertising their vaccine efficacy prior to the completion of phase III clinical trial. The most substantial criticism came from Dr. Konstantin Chumakov, who currently serves as the Associate Director for Research at the FDA Office of Vaccines Research and Review. Dr. Chumakov said: “I would not be in a hurry to call this peptide formulation a vaccine yet, because its effectiveness has not yet been proven…For the introduction of such a vaccine, the level of evidence must be much higher, and therefore the developers of EpiVacCorona, before launching their vaccine on the market, had to conduct clinical trials and prove that their vaccine actually protects against the disease. However, such tests were not carried out, which is absolutely unacceptable.”[41]

The title page of the “EpiVacCorona” patent with Anna’s Popova name among inventors

Conflict of interest

The vaccine design was protected by several already issued patents (see section above). In each patent one of its co-authors is a namesake of Anna Popova who is a head of the Federal Service for Supervision of Consumer Rights Protection and Human Welfare. This patent authorship represents an issue as far as Anna Popova is a head of the Russian agency that is charged with overseeing vaccine safety and efficacy. As a co-author of these patents, she might have an interest in promoting the vaccine despite its shortcomings.

References

  1. Jump up to:a b c d e f Ryzhikov AB, Ryzhikov EA, Bogryantseva MP, Usova SV, Danilenko ED, Nechaeva EA, Pyankov OV, Pyankova OG, Gudymo AS, Bodnev SA, Onkhonova GS, Sleptsova ES, Kuzubov VI, Ryndyuk NN, Ginko ZI, Petrov VN, Moiseeva AA, Torzhkova PY, Pyankov SA, Tregubchak TV, Antonec DV, Gavrilova EV, Maksyutov RA (2021). “A single blind, placebo-controlled randomized study of the safety, reactogenicity and immunogenicity of the “EpiVacCorona” Vaccine for the prevention of COVID-19, in volunteers aged 18–60 years (phase I–II)”Russian Journal of Infection and Immunity11 (2): 283–296. doi:10.15789/2220-7619-ASB-1699.
  2. Jump up to:a b c d e Federal Budgetary Research Institution State Research Center of Virology and Biotechnology “Vector” (2 March 2021). “Multicenter Double-blind Placebo-controlled Comparative Randomized Study of the Tolerability, Safety, Immunogenicity and Prophylactic Efficacy of the EpiVacCorona Peptide Antigen-based Vaccine for the Prevention of COVID-19, With the Participation of 3000 Volunteers Aged 18 Years and Above (Phase III-IV)”.
  3. Jump up to:a b c d e f g DobrovidovaApr. 6, Olga; 2021; Am, 11:05 (6 April 2021). “Russia’s COVID-19 defense may depend on mystery vaccine from former bioweapons lab—but does it work?”Science | AAAS. Retrieved 24 April 2021.
  4. Jump up to:a b c d e f Dobrovidova, Olga (9 April 2021). “Latest Russian vaccine comes with a big dose of mystery”Science372 (6538): 116–117. doi:10.1126/science.372.6538.116ISSN 0036-8075PMID 33833104S2CID 233191522.
  5. Jump up to:a b c Staff, Reuters (26 March 2021). “Volunteers break rank to raise doubts in trial of Russia’s second COVID-19 vaccine”Reuters. Retrieved 23 April 2021.
  6. Jump up to:a b c d e f g “”ЭпиВакКорона” глазами участников клинических испытаний и ученых-биологов”Троицкий вариант — Наука (in Russian). 23 March 2021. Retrieved 23 April 2021.
  7. Jump up to:a b c d e https://epivakorona.com/openletter.htm
  8. Jump up to:a b c “EpiVacCorona’s race to the finish line Meduza speaks to the developer and manufacturer about concerns surrounding Russia’s latest coronavirus vaccine”meduza.io. Retrieved 23 April2021.
  9. Jump up to:a b “Нет антител, вопросы к составу, непрозрачность данных. Что не так с вакциной “ЭпиВакКорона””BBC News Русская служба (in Russian). Retrieved 23 April 2021.
  10. Jump up to:a b c d “Sputnik V’s ugly cousin Clinical results for Russia’s EpiVacCorona vaccine are finally here, but developers published in an obscure local journal, raising questions and concerns”meduza.io. Retrieved 23 April 2021.
  11. ^ “About 200,000 EpiVacCorona vaccine doses go into civil circulation in Russia”TASS. Retrieved 25 April 2021.
  12. Jump up to:a bhttps://www.researchgate.net/publication/350822775_Immunogenicity_and_protectivity_of_the_peptide_candidate_vaccine_against_SARS-CoV-2
  13. Jump up to:a b Ryzhikov AB, Ryzhikov EA, Bogryantseva MP, Usova SV, Danilenko ED, Imatdinov IR, Nechaeva EA, Pyankov OV, Pyankova OG, Gudymo AS, Bodnev SA, Onkhonova GS, Sleptsova ES, Kuzubov VI, Ryndyuk NN, Ginko ZI, Petrov VN, Moiseeva AA, Torzhkova PY, Pyankov SA, Tregubchak TV, Antonec DV, Sleptsova ES, Gavrilova EV, Maksyutov RA (2021). “Immunogenicity and Protectivityof the Peptide Vaccine againstSARS-CoV-2”Annals of the Russian Academy of Medical Sciences76 (1): 5–19. doi:10.15690/vramn1528.
  14. ^ Ryzhikov, A. B.; Ryzhikov, Е. А.; Bogryantseva, M. P.; Usova, S. V.; Danilenko, E. D.; Nechaeva, E. A.; Pyankov, O. V.; Pyankova, O. G.; Gudymo, A. S. (24 March 2021). “A single blind, placebo-controlled randomized study of the safety, reactogenicity and immunogenicity of the “EpiVacCorona” Vaccine for the prevention of COVID-19, in volunteers aged 18–60 years (phase I–II)”Russian Journal of Infection and Immunity. Retrieved 23 April 2021.
  15. ^ “People vaccinated with Russia’s EpiVacCorona need 42 days to develop immunity – watchdog”TASS. Retrieved 25 April 2021.
  16. ^ “Что ждать от “ЭпиВакКороны”. Все о пептидной вакцине против COVID-19″РИА Новости(in Russian). 1 January 2021. Retrieved 24 April 2021.
  17. ^ s.r.o, Direct Impact. “AIM database substance – EpiVacCorona”AIM. Retrieved 25 April 2021.
  18. Jump up to:a b Federal Budgetary Research Institution State Research Center of Virology and Biotechnology “Vector” (20 February 2021). “Simple, Blind, Placebo-controlled, Randomized Study of the Safety, Reactogenicity and Immunogenicity of Vaccine Based on Peptide Antigens for the Prevention of COVID-19 (EpiVacCorona), in Volunteers Aged 18-60 Years (I-II Phase)”.
  19. ^ Реестр Клинических исследований COV/pept-03/20[1]
  20. ^MKIEEGKLVIWINGDKGYNGLAEVGKKFEKDTGIKVTVEHPDKLEEKFPQVAATGDGPDIIFWAHDRFGGYAQSGLLAEITPDKAFQDKLYPFTWDAVRYNGKLIAYPIAVEALSLIYNKDLLPNPPKTWEEIPALDKELKAKGKSALMFNLQEPYFTWPLIAADGGYAFKYENGKYDIKDVGVDNAGAKAGLTFLVDLIKNKHMNADTDYSIAEAAFNKGETAMTINGPWAWSNIDTSKVNYGVTVLPTFKGQPSKPFVGVLSAGINAASPNKELAKEFLENYLLTDEGLEAVNKDKPLGAVALKSYEEELAKDPRIAATMENAQKGEIMPNIPQMSAFWYAVRTAVINAASGRQTVDEALKDAQTNSSSNNNNNNNNNNLGDNGPQNQRNAPRITFGGPSDSTGSNQNGERSGARSKQRRPQGLPNNTASWFTALTQHGKEDLKFPRGQGVPINTNSSPDDQIGYYRRATRRIRGGDGKMKDLSPRWYFYYLGTGPEAGLPYGANKDGIIWVATEGALNTPKDHIGTRNPANNAAIVLQLPQGTTLPKGFYAEGSRGGSQASSRSSSRSRNSSRNSTPGSSRGTSPARMAGNGGDAALALLLLDRLNQLESKMSGKGQQQQGQTVTKKSAAEASKKPRQKRTATKAYNVTQAFGRRGPEQTQGNFGDQELIRQGTDYKHWPQIAQFAPSASAFFGMSRIGMEVTPSGTWLTYTGAIKLDDKDPNFKDQVILLNKHIDAYKTFPPTEPKKDKKKKADETQALPQRQKKQQTVTLLPAADLDDLSKQLQQSMSSADSTQA. “Carrier protein sequence”.
  21. Jump up to:a b “Russia begins post-registration trials of EpiVacCorona Covid-19 vaccine”http://www.clinicaltrialsarena.com. Retrieved 25 April 2021.
  22. ^ “Вакцина “ЭпиВакКорона” поступила в гражданский оборот”РИА Новости (in Russian). 11 December 2020. Retrieved 23 April 2021.
  23. ^ “Turkmenistan registers vaccines for the prevention of infectious diseases”Turkmenistan Today. 29 January 2021.
  24. ^ “Turkmenistan: Master Berdymukhamedov goes to Moscow | Eurasianet”eurasianet.org. Retrieved 25 April 2021.
  25. ^ “Russia submits EpiVacCorona vaccine documents to WHO – Rospotrebnadzor head Popova”interfax.com. Retrieved 23 April 2021.
  26. ^ Welle (www.dw.com), Deutsche. “Two more Russian vaccines: What we do and don’t know | DW | 09.03.2021”DW.COM. Retrieved 23 April 2021.
  27. ^ “COVID-19 vaccine: WHO in talks with Russia on its second vaccine EpiVacCorona”mint. 16 October 2020. Retrieved 9 June 2021.
  28. ^ “Vector Center says has over 45 inquiries from abroad about its EpiVacCorona vaccine”TASS. Retrieved 25 April 2021.
  29. ^ Foundation, Thomson Reuters. “Venezuela receives doses of Russian EpiVacCorona vaccine for trials”news.trust.org. Retrieved 25 April 2021.
  30. ^ “Venezuela to purchase and manufacture Russia’s EpiVacCorona vaccine”Reuters. 5 June 2021. Retrieved 13 June 2021.
  31. ^ turkmenportal. “Turkmenistan Approves Use of Russia’s EpiVacCorona Vaccine | Society”Business Turkmenistan Information Center. Retrieved 25 April 2021.
  32. ^ Jones, Ian; Roy, Polly (20 February 2021). “Sputnik V COVID-19 vaccine candidate appears safe and effective”The Lancet397 (10275): 642–643. doi:10.1016/S0140-6736(21)00191-4ISSN 0140-6736PMC 7906719PMID 33545098.
  33. ^ “Участники КИ “ЭпиВакКороны” продолжают исследовать эффективность вакцины”pcr.news. Retrieved 24 April 2021.
  34. ^ Li, Yang; Ma, Ming-Liang; Lei, Qing; Wang, Feng; Hong, Wei; Lai, Dan-Yun; Hou, Hongyan; Xu, Zhao-Wei; Zhang, Bo; Chen, Hong; Yu, Caizheng (30 March 2021). “Linear epitope landscape of the SARS-CoV-2 Spike protein constructed from 1,051 COVID-19 patients”Cell Reports34 (13): 108915. doi:10.1016/j.celrep.2021.108915ISSN 2211-1247PMC 7953450PMID 33761319.
  35. ^ “Вакцина “ЭпиВакКорона” в иллюстрациях”Троицкий вариант — Наука (in Russian). 23 March 2021. Retrieved 24 April 2021.
  36. ^ Yi, Zhigang; Ling, Yun; Zhang, Xiaonan; Chen, Jieliang; Hu, Kongying; Wang, Yuyan; Song, Wuhui; Ying, Tianlei; Zhang, Rong; Lu, HongZhou; Yuan, Zhenghong (December 2020). “Functional mapping of B-cell linear epitopes of SARS-CoV-2 in COVID-19 convalescent population”Emerging Microbes & Infections9 (1): 1988–1996. doi:10.1080/22221751.2020.1815591ISSN 2222-1751PMC 7534331PMID 32844713.
  37. ^ Poh, Chek Meng; Carissimo, Guillaume; Wang, Bei; Amrun, Siti Naqiah; Lee, Cheryl Yi-Pin; Chee, Rhonda Sin-Ling; Fong, Siew-Wai; Yeo, Nicholas Kim-Wah; Lee, Wen-Hsin; Torres-Ruesta, Anthony; Leo, Yee-Sin (1 June 2020). “Two linear epitopes on the SARS-CoV-2 spike protein that elicit neutralising antibodies in COVID-19 patients”Nature Communications11 (1): 2806. doi:10.1038/s41467-020-16638-2ISSN 2041-1723PMC 7264175PMID 32483236.
  38. ^ Li, Yang; Lai, Dan-Yun; Zhang, Hai-Nan; Jiang, He-Wei; Tian, Xiaolong; Ma, Ming-Liang; Qi, Huan; Meng, Qing-Feng; Guo, Shu-Juan; Wu, Yanling; Wang, Wei (October 2020). “Linear epitopes of SARS-CoV-2 spike protein elicit neutralizing antibodies in COVID-19 patients”Cellular & Molecular Immunology17 (10): 1095–1097. doi:10.1038/s41423-020-00523-5ISSN 2042-0226PMC 7475724PMID 32895485.
  39. ^ Farrera-Soler, Lluc; Daguer, Jean-Pierre; Barluenga, Sofia; Vadas, Oscar; Cohen, Patrick; Pagano, Sabrina; Yerly, Sabine; Kaiser, Laurent; Vuilleumier, Nicolas; Winssinger, Nicolas (2020). “Identification of immunodominant linear epitopes from SARS-CoV-2 patient plasma”PLOS ONE15 (9): e0238089. doi:10.1371/journal.pone.0238089ISSN 1932-6203PMC 7480855PMID 32903266.
  40. ^ Shrock, Ellen; Fujimura, Eric; Kula, Tomasz; Timms, Richard T.; Lee, I.-Hsiu; Leng, Yumei; Robinson, Matthew L.; Sie, Brandon M.; Li, Mamie Z.; Chen, Yuezhou; Logue, Jennifer (27 November 2020). “Viral epitope profiling of COVID-19 patients reveals cross-reactivity and correlates of severity”Science370 (6520): eabd4250. doi:10.1126/science.abd4250ISSN 1095-9203PMC 7857405PMID 32994364.
  41. ^ “Константин Чумаков: “Даже если человек переболел COVID-19, ему все равно нужно привиться. Иммунный ответ на прививку лучше и долговечнее, чем на саму болезнь””republic.ru (in Russian). Retrieved 24 April 2021.

External links

EpiVacCorona vaccine
Vaccine description
TargetSARS-CoV-2
Vaccine typePeptide subunit
Clinical data
Trade namesEpiVacCorona
Routes of
administration
Intramuscular
ATC codeNone
Legal status
Legal statusRegistered in Russia on 14 October 2020 RU Registered.TU approved.Full list : List of EpiVacCorona COVID-19 vaccine authorizations
Identifiers
DrugBankDB16439
Part of a series on the
COVID-19 pandemic
COVID-19 (disease)SARS-CoV-2 (virus)
showTimeline
showLocations
showInternational response
showMedical response
showImpact
 COVID-19 portal

EpiVacCorona Vaccine, developed by the Vektor State Research Center of Virology and Biotechnology in Russia, is based on peptide-antigens that facilitate immunity to the SARS-CoV-2 virus1. It is currently being tested in Phase I/II clinical trials for safety and immunogenicity (NCT04527575)1,2.

  1. Precision Vaccinations: VACCINE INFO EpiVacCorona Vaccine [Link]
  2. The Pharma Letter: Russia’s EpiVacCorona vaccine post-registration trials started [Link]

//////EpiVacCorona, SARS-CoV-2, RUSSIA, CORONA VIRUS, COVID 19, VACCINE, PEPTIDE

wdt-16

NEW DRUG APPROVALS

ONE TIME

$10.00

%d bloggers like this: