Search API

VAXNEUVANCE 15 Pneumococcal Vaccine

VAXNEUVANCE™ Pneumococcal Vaccine Clinical Trials, Dosage, Indication, News, Side Effects

The U.S. Food and Drug Administration (FDA) approved Merck's VAXNEUVANCE™ on July 16, 2021, for active immunization for the prevention of invasive pneumococcal disease (IPD) caused by 15 serotypes conjugated to a CRM197 carrier protein, including Streptococcus pneumoniae serotypes 1, 3, 4, 5, 6A, 6B, 7F, 9V, 14, 18C, 19A, 19F, 22F, 23F, and 33F, in adults. On June 22, 2022, the FDA approved (STN: 125741) an expanded indication for VAXNEUVANCE, which now includes children aged six weeks through 17 years. The FDA published a Package Insert, Patient Information sheet, and BLA Clinical Review Memorandum; STN: 125741.

On January 28, 2022, the U.S. CDC confirmed that using PCV20 alone or PCV15 in combination with PPSV23 is expected to reduce pneumococcal disease incidence in adults aged 65 years and older, as well as in those aged 19–64 years with certain underlying conditions. Natalie Banniettis, MD, FAAP, Senior Principal Scientist, Global Clinical Development, Merck Research Laboratories, Merck & Co., presented (V114, VAXNEUVANCE): Pediatric Clinical Development Program on February 24, 2022. Merck states that vaccinating with VAXNEUVANCE pneumococcal polysaccharides may not protect all vaccine recipients.

On December 15, 2021, the European Commission approved VAXNEUVANCE EMEA/H/C/005477 for use by adults. On October 24, 2022, the European Commission approved the expanded approval for infants, children, and adolescents aged 6 weeks to less than 18 years.

For more information on the VAXNEUVANCE vaccine, visit www.merck.com or this Merck webpage. The brand is trademarked in Canada. ATC code: J07AL02. U.S. Patent No. 8,192,746

VAXNEUVANCE Vaccine Revenues

On April 26, 2025, Merck announced that Vaxneuvance produced $230 million in revenues. Growth was primarily driven by higher demand in the Europe and Asia Pacific regions, partially offset by lower demand in the U.S. due to competitive pressure. On April 25, 2024, Merck announced that Vaxneuvance generated $219 million in revenues during the first quarter of 2024, representing a 106% increase from the prior year period. 

VAXNEUVANCE Vaccine Indication

VAXNEUVANCE is indicated for active immunization to prevent invasive disease caused by the S. pneumoniae serotypes contained in the vaccine in adults. According to the U.S. FDA disclosure, more than 90 types of pneumococcal bacteria can affect adults differently than children. Pneumococcal serotypes not in the currently licensed conjugate vaccine, such as 22F and 33F, are commonly associated with invasive pneumococcal disease. For example, serotypes 22F and 33F now account for 13% of the invasive pneumococcal disease seen among adults aged 65 and older in the U.S. and 7-12% of European adult cases.

Additionally, serotype 3 remains one of the leading causes of invasive pneumococcal disease in adults and children, despite being included in currently available pneumococcal vaccines. For example, in the U.S., the U.S. CDC says that 15% of invasive pneumococcal disease among adults aged 65 and older continues to be caused by serotype 3. And this ranges from 12 to 18% of adult cases across European countries.

Children under the age of 2 are particularly vulnerable to pneumococcal infection, and the incidence of invasive pneumococcal disease remains highest in the first year of life. There are 100 different types of pneumococcal bacteria, including serotypes 22F, 33F, and 3, which collectively represent more than a quarter of invasive pneumococcal disease cases in children under 5.

The U.S. CDC Advisory Committee on Immunization Practices (ACIP) met on October 20, 2021, and unanimously voted to update pneumococcal vaccination recommendations for adults 65 and older and those ages 19 to 64 with certain underlying medical conditions or other disease risk factors. In both groups, the ACIP voted to recommend vaccination with a sequential regimen of VAXNEUVANCE followed by PNEUMOVAX 23 or with a single dose of 20-valent pneumococcal conjugate vaccine. These updates would apply to adults who have not received a pneumococcal conjugate vaccine or whose previous pneumococcal vaccination history is unknown.

VAXNEUVANCE Dosage

Merck's VAXNEUVANCE is administered as an intramuscular injection. When PCV15 is used, the CDC recommends a minimum of one year between the administration of PCV15 and PPSV23. A minimum interval of 8 weeks can be considered for adults with an immunocompromising condition, a cochlear implant, or a cerebrospinal fluid leak to minimize the risk of IPD caused by serotypes unique to PPSV23 in these vulnerable groups. Adults who have only received PPSV23 may receive a PCV (either PCV20 or PCV15) ≥ one year after their last PPSV23 dose. When PCV15 is used in individuals with a history of receiving PPSV23, it must be followed by another dose of PPSV23.

VAXNEUVANCE Coadministration

The CDC confirmed PCV15, PCV20, or PPSV23 could be coadministered with QIV in an adult immunization program as concomitant administration (PCV15 or PPSV23 and QIV [Fluarix], PCV20, and adjuvanted QIV [Fluad]) has been demonstrated to be immunogenic and safe. However, slightly lower pneumococcal serotype-specific OPA GMTs or geometric mean concentrations were reported when pneumococcal vaccines were coadministered with QIV than when pneumococcal vaccines were given alone. Currently, there is no data on the coadministration of this vaccine with other vaccines (e.g., tetanus, diphtheria, acellular pertussis vaccine, hepatitis B, or zoster vaccine) among adults. Evaluation of coadministration of PCV15, PCV20, or PPSV23 with COVID-19 vaccines is ongoing.

VAXNEUVANCE Side Effects

Do not administer VAXNEUVANCE to individuals with a severe allergic reaction to any VAXNEUVANCE or diphtheria toxoid component. Additionally, individuals with altered immunocompetence, including those receiving immunosuppressive therapy, may have a reduced immune response to VAXNEUVANCE. The most commonly reported solicited adverse reactions in individuals 18 through 49 years of age were injection site pain (75.8%), fatigue (34.3%), myalgia (28.8%), headache (26.5%), injection site swelling (21.7%), injection site erythema (15.1%) and arthralgia (12.7%). The most commonly reported solicited adverse reactions in individuals 50 years of age and older were injection site pain (66.8%), myalgia (26.9%), fatigue (21.5%), headache (18.9%), injection site swelling (15.4%), injection site erythema (10.9%) and arthralgia (7.7%).

VAERS is a U.S. passive reporting system, meaning it relies on individuals to send in reports of their experiences with a vaccine. However, anyone, including parents and patients, can submit information to VAERS. Furthermore, healthcare providers are required by law to report to VAERS. People are also encouraged to report the adverse side effects of prescription drugs to the FDA. Visit www.fda.gov/medwatch or call 1–800–FDA–1088.

VAXNEUVANCE Availability

As of 2024, VAXNEUVANCE was available in the U.S., the U.K., and Europe. On April 27, 2023, Merck reported higher sales of VAXNEUVANCE, which increased to $106 million, primarily due to continued uptake in the pediatric indication following its launch in the U.S. 

VAXNEUVANCE News

June 13, 2023 - VAXNEUVANCE (V114) was found to be well-tolerated and safe for infants, according to a study published in the journal Pediatrics.

October 24, 2022 - Dr. Eliav Barr, SVP, head of global clinical development and chief medical officer, Merck Research Laboratories, stated in a press release: "With this (EC) approval, we are pleased to bring an important new PCV option to a vulnerable population in Europe, including infants less than one year of age, who typically experience the highest rates of disease."

September 16, 2022 - Dr. Eliav Barr, senior vice president, head of global clinical development, and chief medical officer, Merck Research Laboratories, stated, "We are pleased with the CHMP's positive opinion as it brings us one step closer to our goal of helping to protect against pneumococcal strains that pose a substantial risk to infants and children in Europe."

July 28, 2022 - The U.S. CDC's ACIP unanimously voted to provisionally recommend using VAXNEUVANCE as an option to the currently available 13-valent pneumococcal conjugate vaccine (PCV13) for children under 19 years according to the presently recommended PCV13 dosing and schedules.

December 15, 2021 - Merck announced today that the European Commission has approved VAXNEUVANCE for active immunization to prevent invasive disease and pneumonia caused by Streptococcus pneumoniae in individuals 18 years of age and older. The approval enables the marketing of VAXNEUVANCE in all 27 member states of the European Union, as well as in Iceland, Norway, and Liechtenstein.

December 1, 2Merck announced that the U.S. FDA has accepted for Priority Review a supplemental Biologics License Application for VAXNEUVANCE to prevent invasive pneumococcal disease in children six weeks through 17 years of age. Additionally, the FDA has set a target action date under the Prescription Drug User Fee Act of April 1, 2022.

October 20, 2021 - Merck announced the U.S. Centers for Disease Control and Prevention's Advisory Committee on Immunization Practices (ACIP) unanimously voted in favor of updates to pneumococcal vaccination recommendations for adults 65 years and older and adults ages 19 to 64 with certain underlying medical conditions (e.g., chronic conditions such as diabetes, chronic heart disease, chronic lung disease, or chronic liver disease, as well as HIV) or other disease risk factors (e.g., smoking, alcoholism). In both groups, the ACIP voted to provisionally recommend vaccination with either a sequential regimen of VAXNEUVANCE followed by PNEUMOVAX23 or a single dose of 20-valent pneumococcal conjugate vaccine. These updates would apply to adults who have not received a pneumococcal conjugate vaccine or whose previous pneumococcal vaccination history is unknown.

October 15, 2021 - Merck announced that the European Medicines Agency's Committee for Medicinal Products for Human Use (CHMP) had recommended the approval of VAXNEUVANCE™.

August 25, 2021 - Merck announced topline results from the pivotal PNEU-PED (V114-029) study evaluating the immunogenicity, safety, and tolerability of VAXNEUVANCE™ in 1,720 healthy infants enrolled between 42-90 days of age. In the PNEU-PED study, primary endpoints demonstrated that VAXNEUVANCE had a safety profile generally comparable to PCV13 following receipt of any vaccine dose. Secondary endpoints demonstrated statistically superior immune responses for VAXNEUVANCE compared to PCV13 for shared serotypes three and unique serotypes 22F and 33F, based on prespecified criteria. Additionally, VAXNEUVANCE showed non-inferior immune responses to antigens contained in several routinely used pediatric vaccines when administered concomitantly with VAXNEUVANCE or PCV13.

July 16, 2021 - Merck today announced the U.S. Food and Drug Administration approved VAXNEUVANCE™ (Pneumococcal 15-valent Conjugate Vaccine) (pronounced VAKS-noo-vans) for active immunization for the prevention of invasive disease caused by Streptococcus pneumoniae serotypes 1, 3, 4, 5, 6A, 6B, 7F, 9V, 14, 18C, 19A, 19F, 22F, 23F and 33F in adults 18 years of age and older.

May 20, 2021 - Merck announced that V114, the company's investigational 15-valent pneumococcal conjugate vaccine, met its primary immunogenicity and safety endpoints in the V114 Phase 3 pediatric clinical program.

January 12, 2Merck announced that the U.S. FDA accepted for priority review a Biologics License Application (BLA) for V114, Merck's investigational 15-valent pneumococcal conjugate vaccine, for the prevention of invasive pneumococcal disease in adults 18 years of age and older. Additionally, the FDA established a target action date under the Prescription Drug User Fee Act, as of July 18, 2021. The European Medicines Agency is also reviewing an application for licensure of V114 in adults.

October 20, 2020 - Merck announced Positive topline results from Two Additional Phase 3 Adult Studies Evaluating V114, Merck's Investigational 15-valent Pneumococcal Conjugate Vaccine. In the PNEU-PATH (V114-016) study, healthy adults 50 or older received V114 or PCV13, followed by PNEUMOVAX® 23 one year later. Following vaccination with PNEUMOVAX 23 (month 13), immune responses were comparable in vaccination groups for the 15 serotypes in V114. Results also showed that at 30 days post-vaccination with either V114 or PCV13 (day 30), immune responses were comparable for both groups across the 13 serotypes shared by the conjugate vaccines and higher in the V114 group for serotypes 22F and 33F, the two serotypes not included in PCV13.

January 30, 2019 - Merck announced that V114 received a Breakthrough Therapy Designation from the U.S. Food and Drug Administration (FDA) to prevent invasive IPD caused by the vaccine serotypes in pediatric patients six weeks to 18 years of age.

VAXNEUVANCE 15 Clinical Trials

Merck's V114 Pneumococcal Vaccine has undergone many clinical trials, and Merck continues to test the vaccine candidate under many scenarios. The registered 28 clinical trials can be accessed here.

0 min read
Availability: 
USA
Generic: 
V114
Clinical Trial: 
https://www.merckclinicaltrials.com/
Drug Class: 
Conjugate Vaccine
Condition: 
Last Reviewed: 
Saturday, July 5, 2025 - 07:35
Brand: 
VAXNEUVANCE 15
Abbreviation: 
PPV 15
Status: 
Manufacturer Country ID: 
FDA First In Class: 
Yes
Kosher: 
Yes
Rate Vaccine: 
H2lVXRyg
Location tags: 

Zabdeno (Ad26.ZEBOV) and Mvabea (MVA-BN-Filo) Ebola Vaccine

Ebola Vaccine Zabdeno® and Mvabea® Clinical Trials, Dosage, Indication, News

The J&J Innovative Medicine Ebola vaccine consists of two components, Zabdeno® (Ad26.ZEBOV) and Mvabea® (MVA-BN-Filo). Zabdeno is given first, and Mvabea is administered approximately eight weeks later as a vaccine booster. This prime-boost vaccination method is an established approach for preventing infectious diseases during Ebola outbreaks for individuals at risk of exposure and preventively before episodes, says J&J.

J&J developed Zabdeno, a monovalent vaccine designed to provide active, specific, acquired immunity to the Ebola virus. The vaccine is based on an adenovirus type 26 (Ad26) vector expressing the glycoprotein of the Ebola virus Mayinga variant. Bavarian Nordic's Mvabea (MVA-BN-Filo) is a multivalent vaccine preparation designed to provide active acquired immunity to Sudan, Ebola, Marburg, and Tai Forest (formerly Côte d'Ivoire ebolavirus). The Zabdeno and Mvabea combination was evaluated under the European Medicines Agency (EMA) (EMEA/H/C/005337) accelerated assessment. However, as a precautionary measure, a Zabdeno booster vaccination should be considered for individuals at imminent risk of exposure to the Ebola virus, for example, healthcare professionals and those living in or visiting areas with an ongoing Ebola virus outbreak who completed the Zabdeno, Mvabea 2-dose primary vaccination regimen.

On June 4, 2021, J&J welcomed a new recommendation by the WHO's Strategic Advisory Group of Experts on Immunization (SAGE) that supports the use of the Ebola vaccine regimen both during outbreaks for individuals at risk of Ebola exposure and preventively in the absence of an outbreak, as protection against Ebola virus disease (EVD) is considered a significant public health interest. Accordingly, African countries began evaluating a single dose of Bavarian Nordic's Mvabea (MVA-BN-Filo) to treat the Marburg virus disease in August 2021. 

The Lancet published a study on September 13, 2021, concluding, 'The Ad26.ZEBOV and MVA-BN-Filo Ebola vaccine regimens were well tolerated in a clinical trial (NCT02509494), with no safety concerns in children. The vaccine induced robust humoral immune responses. Data from two papers published in The Lancet Infectious Diseases demonstrated that Zabdno and Mvabea generated robust humoral immune responses in adults and children, with the immune responses persisting in adults for at least two years. The data also showed that booster vaccination with Ad26.ZEBOV, administered to adults two years after the initial immunization, induced a robust anamnestic response within seven days. On December 14, 2022, a study published by the New England Journal of Medicine concurred with The Lancet's findings. In addition, the JAMA Network published Ebola Vaccines Safe and Immunogenic in Clinical Trials on January 24, 2023. On March 26, 2024, The Lancet Infectious Diseases published results from a study that found the J&J vaccine regimen and booster dose were well tolerated. A similar and robust humoral immune response was observed for participants boosted one year and two years after the first dose, supporting the use of the regimen and flexibility of booster dose administration for prophylactic vaccination in at-risk populations. 

Janssen is now Johnson & Johnson Innovative Medicine.

Zabdeno and Mvabea Vaccine History

The U.S. National Institutes of Health sponsored the initial development of the Mvabea (MVA-BN-Filo) vaccine. The U.S. NIAID, BARDA, and other funding partners supported the development, preclinical, and clinical testing of this investigational vaccine regimen designed to protect against the virus responsible for the 2014-2016 Ebola outbreak in West Africa. On May 29, 2020, the EMA human medicines committee recommended granting Zabdeno and Mvabea a positive opinion for marketing authorizations under exceptional circumstances because the applicant was able to demonstrate that it is not possible to conduct a randomized controlled study that might generate comprehensive clinical data on the efficacy of the latest Ebola vaccine even after authorization. The EMA has approved its use in individuals aged one year or older.

Zabdeno and Mvabea Indication

The Ad26.ZEBOV / MVA-BN-Filo combination is indicated to provide an active acquired immunity to the Ebola virus. Clinical results suggest that Ad26.ZEBOV prime immunization readily induces an immune response enhanced by MVA-BN-Filo boosting. Heterologous 2-dose vaccination with Ad26.ZEBOV and MVA-BN-Filowere were used against the Ebola virus and were well tolerated and immunogenic in healthy volunteers. The vaccine is administered as an intramuscular injection. A study published in 2020 found that the two-dose heterologous regimen with Ad26.ZEBOV and MVA-BN-Filo were safe, well-tolerated, and immunogenic, with humoral and cellular immune responses persisting for one year after vaccination.

Zabdeno and Mvabea Side Effects

The most common side effects found in participants in the clinical trials were injection-site reactions (pain, warmth, and swelling), fatigue, headache, myalgia, arthralgia, and chills reported by the EMA. In a phase 1 study, the reporting of A.E.s was identical to that described for the VAC52150EBL1003 trial. Briefly, solicited A.E.s were recorded in a diary by participants for seven days following each vaccination, and unsolicited A.E.s were collected at all visits until 21 days after dose 2. 

Zabdeno and Mvabea Safety

In November 2023, The Lancet Global Health published: Safety and immunogenicity of the two-dose heterologous Ad26.ZEBOV and MVA-BN-Filo Ebola vaccine regimen in infants: a phase 2, randomized, double-blind, active-controlled trial in Guinea and Sierra Leone. This analysis that included Ad26.ZEBOV and MVA-BN-Filo were well tolerated and induced strong humoral responses in infants younger than one-year-old. There were no safety concerns related to vaccination.

Zabdeno and Mvabea News

March 26, 2024 - The Innovative Medicines Initiative 2 Joint Undertaking and Coalition for Epidemic Preparedness Innovations funded a study: Ad26.ZEBOV, MVA-BN-Filo Ebola virus disease vaccine regimen plus Ad26.ZEBOV booster at one year versus two years in healthcare and front-line workers in the Democratic Republic of the Congo: secondary and exploratory outcomes of an open-label, randomized, phase 2 trial.

January 31, 2023 - The JAMA Network published a review of three vaccine regimens against Zaire Ebola virus disease that safely produced immune responses for up to 12 months, according to two clinical trials of adults and children reported in the New England Journal of Medicine.

December 9, 2021 - The peer-reviewed journal PLOS Pathogens published: Current state of Ebola virus vaccines: A snapshot.

October 4, 2021 - The journal PLOS Medicine published a RESEARCH ARTICLE: Safety and immunogenicity of 2-dose heterologous Ad26.ZEBOV, MVA-BN-Filo Ebola vaccination in healthy and HIV-infected adults: A randomized, placebo-controlled Phase II clinical trial in Africa. Conclusions - Ad26.ZEBOV, MVA-BN-Filo vaccination was well tolerated and immunogenic in healthy and HIV-infected African adults.

September 13, 2021 - A study published by the journal The Lancet assessed the Safety and immunogenicity of a two-dose heterologous vaccine regimen comprising the adenovirus type 26 vector-based vaccine encoding the Ebola virus glycoprotein (Ad26.ZEBOV) and the modified vaccinia Ankara vector-based vaccine, encoding glycoproteins from the Ebola virus, Sudan virus, and Marburg virus, and the nucleoprotein from the Tai Forest virus (MVA-BN-Filo), in a pediatric population in Sierra Leone, Africa. Ebola virus glycoprotein-specific binding antibody responses at 21 days after the second dose of the Ebola virus vaccine were observed in about 97% of the children. And there were no treatment-related deaths.

August 17, 2021 - The World Health Organization Africa confirmed it helped the vaccine deployment from Guinea to Cote d'Ivoire of about 3,000 vaccine doses manufactured by Johnson & Johnson, which will be used to boost the vaccination in areas not experiencing active Ebola transmission.

June 4, 2021 - Johnson & Johnson announced it 'welcomes the Strategic Advisory Group of Experts on Immunization recommendation for the WHO.

November 17, 2020 - Study published by The Lancet: Vaccine - Study Interpretation - The two-dose heterologous regimen with Ad26.ZEBOV and MVA-BN-Filo were safe, well-tolerated, and immunogenic, with humoral and cellular immune responses one year after vaccination. Together, these data support the intended prophylactic indication for the vaccine regimen.

July 1, 2020 - The European Commission adopted the decision granting marketing authorizations to Janssen, a Johnson & Johnson company, for a vaccine against Ebola.

June 12, 2020 - Bavarian Nordic A/S announced that the Company has entered into a new supply contract with Janssen Vaccines & Prevention B.V., part of the Janssen Pharmaceutical Companies of Johnson & Johnson valued at USD 13.9 million. Under the agreement, Bavarian Nordic will manufacture and deliver bulk drug substances of its MVA-BN® Filo vaccine, which Janssen has licensed as part of its Ebola vaccine regimen.

April 19, 2019 - A phase 1 study of healthy volunteers, immunization with Ad26.ZEBOV or MVA-BN-Filo did not result in any vaccine-related serious adverse events.

March 14, 2017 - Immune Responses to Novel Adenovirus Type 26 and Modified Vaccinia Virus Ankara–Vectored Ebola Vaccines at 1 Year.

September 12, 2016 – Bavarian Nordic A/S announced that Janssen Vaccines & Prevention B.V. (Janssen) had completed a submission to the WHO for Emergency Use Assessment and Listing for its Ebola prime-boost vaccine regimen, which includes the MVA-BN® Filo vaccine.

October 22, 2014 – Bavarian Nordic A/S announced a global license and supply agreement for its MVA-BN Filovirus (Ebola and Marburg) vaccine candidate with Crucell Holland B.V., one of the Janssen Pharmaceutical Companies of Johnson & Johnson. 

Zabdeno (Ad26.ZEBOV) and Mvabea (MVA-BN-Filo) Ebola Vaccine Clinical Trials

As a result of these collaborations, more than 6,500 individuals have now participated in clinical studies for the Ebola vaccine across the U.S., Europe, and Africa. So far, the data from these studies suggest that the vaccine stimulates a robust immune response and has a favorable safety profile.

ClinicalTrials.gov: NCT0—9494—Phase 3 Study to Assess the Safety and Immunogenicity of Ebola Candidate Vaccines Ad26.ZEBOV and MVA-BN-Filo (EBOVAC-SaThe lone). It was last posted on June 29, 2020.

0 min read
Availability: 
Limited
Generic: 
Ad26.ZEBOV, MVA-BN-Filo
Clinical Trial: 
https://www.janssen.com/uk/tov/clinical-trials
Drug Class: 
Viral Vector Vaccine
Condition: 
Last Reviewed: 
Friday, March 29, 2024 - 06:10
Brand: 
Zabdeno and Mvabea
Abbreviation: 
Ebola Vaccine Regimen
Status: 
Manufacturer Country ID: 
FDA First In Class: 
Yes
Kosher: 
Yes
Halal: 
Yes
Rate Vaccine: 
MfdFWtdl

VIMKUNYA Chikungunya Vaccine

VIMKUNYA® Chikungunya Vaccine Clinical Trials, Dosage, Efficacy, Indication, Side Effects 

Bavarian Nordic A/S VIMKUNYA® (Chikungunya Vaccine, Recombinant) CHIKV-VLP (PXVX0317) is a single-dose aluminum hydroxide-adjuvanted, non-live chikungunya virus-like particle vaccine (VLP) with a multi-protein structure that mimics the organization and conformation of naturally occurring viruses without the viral genome, promoting a more robust immune response and increased antibody production. Vimkunya vaccination triggers the production of neutralizing antibodies at 22 days and up to 183 days post-vaccination, which is expected to confer protection against chikungunya disease. Studies have shown that VLP vaccines are highly immunogenic, have a proven safety record, and typically elicit high-titer neutralizing antibodies, essential for protecting against the Chikungunya virus (CHIKV). The VLPs consist of CHIKV capsid protein (C) and envelope proteins E1 and E2 derived from the CHIKV Senegal strain 37997. VIMKUNYA™ is the first chikungunya vaccine to leverage VLP technology. It utilizes virus-like particles designed to mimic the chikungunya virus, but without the ability to infect cells, replicate, or cause disease. VIMKUNYA technology is licensed from the U.S. National Institute of Allergy and Infectious Diseases (NIAID) at the U.S. National Institutes of Health (NIH).

On February 14, 2025, the U.S. Food and Drug Administration (FDA) approved VIMKUNYA for injection, the first VLP chikungunya vaccine in the United States, for individuals 12 years of age and older. Concurrent with the approval, the FDA awarded Bavarian Nordic (BN) a Priority Review Voucher (PRV) under the Tropical Disease PRV program, which the Company intends to monetize when appropriate. On August 13, 2024, BN announced that the FDA had accepted and granted a Priority Review and Fast Track for the Biologics License Application for CHIKV VLP for individuals 12 and older. The FDA posted the Briefing Document on November 8, 2019. The FDA's approval of VIMKUNYA was based on data from two phase 3 studies that assessed the vaccine's effectiveness by measuring anti-chikungunya virus neutralizing antibody levels in over 3,500 healthy participants aged 12 years and older. On June 26, 2025, Dr. Lyle Petersen of the U.S. CDC presented this information during the ACIP vaccine meeting.

On March 7, 2025, BN announced that the UK Medicines and Healthcare products Regulatory Agency had validated the marketing authorization application and accepted it for review of VIMKUNYA. On February 28, 2025, BN announced that the European Commission had granted marketing authorization for VIMKUNYA in Europe for active immunization to prevent disease caused by the Chikungunya virus in individuals aged 12 years and older. Bavarian Nordic will launch VIMKUNYA in key European markets in the first half of 2025. The European Medicines Agency (EMA) granted PRIME designation in January 2020 (EMEA-002656-PIP01-19). Vimkunya was reviewed under the EMA's accelerated assessment program.

On May 12, 2025, a Journal Article concluded that CHIK-VLP provides approximately 95% overall short-term seroresponse rates in robust trials among potential travelers to CHIKV endemic areas, with an onset of seroprotection that may be as short as 8 days. On August 15, 2025, a study published in The Journal of Infectious Diseases provides the first in vivo evidence that CHIKV directly infects cartilage-producing chondrocytes, offering insights into the mechanisms of alphaviral arthritis.

Bavarian Nordic A/S is a fully integrated vaccine company headquartered in Denmark that develops, manufactures, and commercializes life-saving vaccines. Gaithersburg, Maryland-based Emergent BioSolutions sold this vaccine to Bavarian Nordic A/S on May 15, 2023. As of November 14, 2025, revenue for the first nine months of 2025 increased by 32% to DKK 4,793 million, reflecting a strong performance in both Travel Health and Public Preparedness. Travel Health revenue increased by 23% to DKK 2,327 million compared to the first nine months of 2024, primarily driven by increased demand for rabies and Tickborne encephalitis vaccines, and supported by the gradual launch of the chikungunya vaccine, Vimkunya®. Vimkunya's revenue was DKK 42 million in the first nine months of 2025.

VIMKUNYA Chikungunya Vaccine Availability 2025

As of 2025, VIMKUNYA is commercially available in various countries. VIMKUNYA is the first chikungunya vaccine approved in Europe, the UK, and the United States for individuals as young as 12 years old. As of November 2025, Vimkunya has also been launched in Sweden, Norway, Finland, Italy, and Spain in October.

On February 25, 2025, Bavarian Nordic announced a strategic partnership with Biological E. Limited to expand access to VIMKUNYA in low—and middle-income countries.

VIMKUNYA Efficacy

On April 19, 2025, The Lancet published results from a pivotal phase 3, randomized, double-blind, placebo-controlled, parallel-group trial, concluding that Vimkunya induces a rapid and robust immune response, supporting the potential for this vaccine to protect individuals aged 12–64 years from disease caused by the chikungunya virus.

U.S. CDC and VIMKUNYA

Bavarian Nordic announced on April 16, 2025, that the U.S. Centers for Disease Control and Prevention's (CDC) Advisory Committee on Immunization Practices (ACIP) voted to recommend VIMKUNYA for injection to prevent disease caused by the chikungunya virus. The ACIP recommends VIMKUNYA for individuals aged 12 and older traveling to a country or territory with a chikungunya outbreak. In addition, VIMKUNYA may be considered for persons traveling to or taking up residence in a country or territory without an outbreak but with an elevated risk for U.S. travelers, particularly if planning extended travel. The ACIP also recommends VIMKUNYA for laboratory workers with potential exposure to chikungunya. The HHS Secretary adopted these ACIP recommendations on May 13, 2025, and they are now official recommendations of the CDC.

VIMKUNYA Chikungunya Vaccine Indication

VIMKUNYA is indicated to prevent the Chikungunya virus, which is spread to people through infected mosquitoes. Symptoms lasting 3-4 weeks include high-grade fever, joint pain, headache, muscle pain, joint swelling, or rash. Chikungunya chronic arthralgia impacts a minority of patients but creates long-term health and work concerns. As of March 2025, Additional clinical studies are required to confirm the clinical profile of VIMKUNYA™, and confirmatory efficacy studies are also planned as part of the post-marketing commitments and requirements as agreed with the U.S. FDA.

VIMKUNYA Chikungunya Vaccine Dosage

VIMKUNYA is administered as a 40 µg dose by a muscular injection. VIMKUNYA is supplied as a single-dose, 1-mL glass prefilled syringe with a 0.8-mL dose volume. People with a lowered immune system, including people receiving medications that affect the immune system, may have a diminished response to VIMKUNYA™ vaccination.

VIMKUNYA Chikungunya Vaccine Side Effects

The most commonly reported adverse event in the clinical study was pain at the injection site, occurring in 12 (23%) of 53 participants who received the unadjuvanted vaccine (Group 1) and 111 (31%) of 356 who received the adjuvanted vaccine. No vaccine-related serious adverse events were reported. People should not receive VIMKUNYA if they are allergic to any of the vaccine ingredients or have had an allergic reaction following a previous dose of VIMKUNYA. 

VIMKUNYA Vaccination Recommendation

Chikungunya vaccination is recommended for adults traveling to a country or territory with a chikungunya outbreak. Since 2014, the U.S. CDC has reported Chikungunya virus disease cases among U.S. travelers returning from countries in the Americas where the virus is circulating. As of March 18, 2025, there have been Chikungunya outbreaks in numerous countries across Africa, the Americas, Asia, and Europe.

VIMKUNYA Revenues

Vimkunya's revenue from sales was DKK 5 million in the first quarter of 2025. The vaccine was launched in the U.S. in mid-March 2025, and the first European markets are expected to launch in the second quarter of 2025. On June 18, 2025, Bavarian Nordic announced that it had agreed to sell its Priority Review Voucher for a total cash consideration of $160 million, which will be recognized as other income.

VIMKUNYA Chikungunya Vaccine News

November 14, 2025 - Paul Chaplin, President & Chief Executive Officer of Bavarian Nordic, said: "Our travel health portfolio has shown continued strength, contributing to our best quarter for this business to date. 

July 31, 2025 - Bavarian Nordic A/S announced the completion of the sale of its Priority Review Voucher, for which the Company received a cash consideration of $160 million. 

July 22, 2025 - Paul Chaplin, President and CEO of Bavarian Nordic, said: "The regulatory submission and acceptance by Health Canada represent yet another highlight in our efforts to expand access to our chikungunya vaccine across the globe."

May 9, 2025 - Bavarian Nordic A/S announced its interim financial results and business progress for the first three months of 2025. Revenue for the first three months increased 62% to DKK 1,347 million, reflecting a strong performance in Travel Health and Public Preparedness.. Travel Health revenue increased by 52% to DKK 680 million.

April 16, 2025 - Paul Chaplin, President and CEO of Bavarian Nordic, stated: "The ACIP's recommendation of our chikungunya vaccine for Americans aged 12 and older represents a significant step forward in expanding access to this vaccine in the U.S."

March 27, 2025 - The Lancet published the results from a company-sponsored Phase 3 clinical study, which showed very positive outcomes.

March 18, 2025 - Lee Ann Kimak, Vice President of Commercial for North America at Bavarian Nordic, commented, "As the first virus-like particle single-dose, prefilled syringe chikungunya vaccine, VIMKUNYA™ provides a crucial new tool to help protect at-risk individuals traveling to regions where the virus is spreading."

March 7, 2025 - "Following the approvals of our Chikungunya vaccine in the US and EU last month, we are excited about the opportunity to expand its availability to European countries later this year," said Paul Chaplin, President and CEO of Bavarian Nordic.

March 5, 2025 - The Company stated: We have a leading commercialized portfolio of travel vaccines, and we continue to strengthen our position as a preferred supplier of mpox and smallpox vaccines to governments for public health preparedness.

February 28, 2025: Paul Chaplin, President and CEO of Bavarian Nordic, commented, "As we expand our presence across Europe, this vaccine will help further consolidate our leading position in travel vaccines. We look forward to making the vaccine available in key markets during the first half of 2025."

February 14, 2025 - Paul Chaplin, President and CEO of Bavarian Nordic, commented in a press release, "As climate change continues to expand the reach of mosquito-borne illnesses like chikungunya, this milestone underscores the importance of cutting-edge solutions to safeguard travelers and vulnerable populations."

January 31, 2025 - "The (EMA) recommendation of our chikungunya vaccine for approval in Europe marks a huge milestone in our efforts to deliver protection against this debilitating disease and, once approved, would represent a significant contribution to expanding the availability of vaccines to a broader population, including adolescents aged 12-17 years," said Paul Chaplin, President and CEO of Bavarian Nordic.

January 31, 2025 - Meeting highlights from the Committee for Medicinal Products for Human Use 27-30 January 2025.

August 13, 2024 - "The FDA review, along with the ongoing review of our CHIKV VLP vaccine by EMA, represents the first regulatory reviews of a chikungunya vaccine for adolescents, potentially providing a broader usage by populations at risk of this debilitating disease," commented Paul Chaplin, President and CEO of Bavarian Nordic.

July 18, 2024 - "The MAA submission and review marks a pivotal milestone for Bavarian Nordic in 2024, and we look forward to working closely with EMA throughout the evaluation process to make our chikungunya vaccine available to individuals 12 years of age and older at risk of chikungunya virus infection," said Paul Chaplin, President and CEO of Bavarian Nordic.

June 26, 2024 - "The MAA marks our second major submission in just a few weeks as part of our efforts to launch the Chikungunya vaccine next year in the U.S. and Europe," said Paul Chaplin, President and CEO of Bavarian Nordic, in a press release.

May 15, 2023: Bavarian Nordic acquired the rights to the development-stage Chikungunya vaccine candidate CHIKV VLP and manufacturing development facilities in Bern, Switzerland, and San Diego, California.

November 1, 2022 - Emergent BioSolutions Inc. announced results from a Phase 2 study evaluating the safety and immunogenicity of the CHIKV VLP vaccine candidate in prior recipients of other investigational alphavirus vaccines. The study demonstrated that the CHIKV VLP vaccine candidate was well-tolerated and immunogenic in alphavirus vaccine-naïve participants and participants previously vaccinated against the Venezuelan equine encephalitis virus. 

May 26, 2021 - Emergent BioSolutions Inc. announced two-year persistence data from its Phase 2 clinical study. Two years post-vaccination, SNA responses were 19 times higher than pre-vaccination titers following a single adjuvanted 40 µg dose of the CHIKV VLP vaccine, supporting the persistence of the immune response. All subjects in the single-dose regimen remained seropositive at their one-year and two-year visits. The vaccine candidate was well-tolerated, and no significant safety concerns related to the vaccine were identified.

January 13, 2020—Emergent BioSolutions Inc. announced that it had received an agreement from the European Medicines Agency to pursue its proposed development plan for its chikungunya virus-like particle (CHIKV VLP) vaccine candidate.

April 26, 2019 - Study published by PLOS: Vaccination with a chikungunya virus-like particle vaccine exacerbates disease in aged mice. Conclusions: Unadjuvanted CHIK VLP vaccination elicits immune responses that protect 100% of adult mice against CHIKV infection. However, an improved vaccine/adjuvant combination is still necessary to enhance the protective immunity against CHIKV in older people.

January 28, 2021 - A VLP vaccine for the epidemic Chikungunya virus protects non-human primates against infection.

CHIKV-VLP Clinical Trials

The CHIKV-VLP vaccine candidate is being investigated in various clinical trials in 2025. The approval of Vimkunya in 2025 was based on data from two phase 3 studies that assessed the vaccine's effectiveness by measuring anti-chikungunya virus-neutralizing antibody levels in over 3,500 healthy participants aged 12 years and older. Dr. Victoria Jenkins presented phase 3 clinical trial safety and efficacy data on CHIKV VLP at the Advisory Committee on Immunization Practices meeting on October 23, 2024.

On March 27, 2025, a company-sponsored Phase 3 study found that between May 12 and December 2, 2022, 413 participants were recruited and randomly assigned (1:1) to receive the Vimkunya vaccine (n = 206) or a placebo (n = 207). The coprimary endpoints of immunologic superiority were met, as measured by chikungunya virus SNA titers compared with placebo and the geometric mean titer at day 22. Vimkunya induced a protective seroresponse (SNA NT80≥100, considered the presumptive seroprotective antibody response) in 149 (82%) of 181 participants (95% CI 76·1–87·2) at day 15, in 165 (87%) of 189 participants (81·8–91·3) at day 22, and in 139 (76%) of 184 participants (68·9–81·2) at day 183. Although the early immune response was slightly higher in the 65–74 age group on day 15 than in the 75+ age group, the seroresponse rates on day 22 and day 183 were similar. There were no notable differences in adverse event rates between the groups; most adverse events were of grade 1 or 2 severity and had a short duration. No vaccine-related serious adverse events or deaths occurred.

The European Commission granted marketing authorization upon the recommendation of the Committee for Medicinal Products for Human Use of the European Medicines Agency in January 2025. It was based on results from two phase 3 clinical trials that enrolled more than 3,500 healthy individuals 12 years and older. The studies met their primary endpoints, with results showing that 21 days after vaccination, the vaccine-induced neutralizing antibodies were present in up to 97.8% of the vaccinated individuals, and a rapid immune response began to develop within one week.

ClinicalTrials.gov, NCT03992872 - Phase 2 Findings: Between November 20, 2019, and January 19, 2021, 60 participants (20 [33%] female and 40 [67%] male; 40 (67%) White; median age 47·0 years [IQR 13·5]), 30 previous alphavirus vaccine recipients and 30 alphavirus vaccine-naïve controls, were enrolled, vaccinated with CHIKV VLP, and completed the trial. The anti-CHIKV neutralizing antibody seroconversion rate at day 22 was 100% (95% CI 88·6-100) in both groups. GMTs peaked in previous alphavirus vaccine recipients and alphavirus vaccine-naïve controls at day 22 (2032·5 [95% CI 1413·0-2923·6] and 2299·2 [1598·1-3307·8], respectively) and were similar between the groups on day 22 and all subsequent visits. A higher proportion of previous alphavirus vaccine recipients (93·3% [95% CI 78·7-98·2]) had a four-fold neutralizing antibody increase at day 8 than did alphavirus vaccine-naïve controls (66·7% [48·8-80·8]; p=0·021). There was no statistically significant difference in the incidence of solicited adverse events between the previous alphavirus vaccine recipients and the alphavirus vaccine-naïve controls (53.3% vs. 40.0%, respectively). However, the relatively small sample size of the trial limited the power to detect a significant difference. No serious adverse events were reported with the vaccine. Interpretation: The CHIKV VLP vaccine was well tolerated and demonstrated similar immunogenicity in alphavirus-naïve participants and those who had previously received a heterologous alphavirus vaccine. There were no significant differences in adverse events between the groups. The results of this study support the use of the CHIKV VLP vaccine in individuals with prior exposure to alphavirus vaccines.

According to a study presented in October 2023, the researchers assessed anti-CHIKV serum-neutralizing antibody titers at designated time points throughout the study. They determined the seroresponse rate (SRR) in the two cohorts. In total, 3,254 participants were enrolled in the adult and adolescent trial, of whom 2,790 received a CHIKV VLP dose and 464 received a placebo. The data showed an SRR of 98% on day 22 for the vaccine and 1% for the placebo. A rapid antibody response was also observed in the vaccine group, with an SRR of 47% on day 8 and 97% on day 15. These responses remained durable through day 183, when the SRR reached 86%. For the older adult trial, 413 participants were enrolled — 206 received CHIKV VLP, and 207 received a placebo. On day 22, the SRR was 87% for vaccine recipients and 1% for placebo recipients. By day 15, a rapid antibody response was also observed in the CHIKV VLP group, with a seroprotection rate (SRR) of 82%. Additional data from the study revealed a favorable safety profile for the vaccine, with most adverse events being mild to moderate in severity. The most common adverse events were myalgia, fatigue, and headache.

On June 20, 2023, the Company announced the phase 3 study results, which included 413 participants enrolled and randomized 1:1 to receive either a single intramuscular injection of CHIKV VLP or a placebo. The initial results up to Day 22 post-vaccination showed that CHIKV VLP was immunogenic in healthy adults ≥65 years of age, as demonstrated by a strong induction of CHIKV neutralizing antibodies in 87% of vaccinees with neutralizing antibody titers exceeding the threshold agreed with authorities as a marker of seroprotection, thus meeting the primary endpoints of the study. Notably, most subjects (82%) also had seroprotective neutralizing antibodies on Day 15 post-vaccination, demonstrating a rapid onset of protection for the VLP-based CHIKV vaccine candidate.

On August 6, 2023, Bavarian Nordic (BN) announced positive topline results from a Phase 3 clinical trial (NCT05072080). PXVX0317 met all the coprimary endpoints and was highly immunogenic in the majority of subjects 22 days after a single vaccination. A rapid and durable response was confirmed, with high immunity levels at 2 weeks and 6 months post-immunization.

On November 14, 2022, the Company announced that a planned post-approval multicenter Phase 3b clinical study will evaluate the vaccine candidate's efficacy in preventing CHIKV disease and assess the utility of a model-guided disease surveillance framework to optimize the execution of a field efficacy trial using CHIKV as a model emerging pathogen.

On November 1, 2022, the Company reported a Phase 2 parallel-group study—a gender-matched, open-label study evaluating the safety and immunogenicity of an adjuvanted CHIKV VLP vaccine candidate in individuals who had previously received experimental alphavirus vaccines, compared with a cohort of individuals naïve to alphavirus vaccines. The study enrolled 60 healthy adults at two U.S. sites. The vaccine candidate was well-tolerated, with no notable difference in the incidence of adverse events between the groups. The majority of solicited adverse events were mild or moderate. The most common adverse event was local injection site pain. The seroconversion rate 21 days post-vaccination was 100% in both groups. A higher percentage of prior alphavirus vaccine candidates showed a fourfold or greater increase on study day eight, narrowing the difference between the alphavirus vaccine-naïve group.

A randomized, double-masked, parallel-group, Phase 2 study was conducted at three U.S. sites involving 445 participants. Eligible participants were healthy CHIKV-naïve adults aged 18–45 years. The primary endpoint was the geometric mean titer of the anti-CHIKV neutralizing antibody on day 57 (28 days after the last vaccination). Safety was also assessed.

0 min read
Availability: 
USA, Europe, pending in the UK
Generic: 
CHIKV-VLP
Clinical Trial: 
https://clinicaltrials.gov/ct2/history/NCT04546724?V_11=View
Drug Class: 
Virus-Like Particle Vaccine, alum-adjuvanted
Condition: 
Last Reviewed: 
Saturday, November 15, 2025 - 10:35
Brand: 
VIMKUNYA®
Abbreviation: 
PXVX0317
Status: 
Manufacturer Country ID: 
FDA First In Class: 
Yes

M72/AS01E (M72) Tuberculosis Vaccine

M72/AS01E (M72) Tuberculosis Vaccine Candidate Clinical Trials, Efficacy, Side Effects

M72/AS01E (M72) is a subunit vaccine candidate that contains the M72 recombinant fusion protein derived from two Mycobacterium tuberculosis (Mtb) antigens (Mtb32A and Mtb39A) combined with the GlaxoSmithKline plc (GSK) proprietary Adjuvant System AS01. The Mtb39A and Mtb32A components of the recombinant antigen elicited specific lymphoproliferation and/or interferon-gamma (IFN-γ) production in individuals with latent and active TB. AS01E is the adjuvant in GSK's Shingrix® shingles and Mosquirix™ (RTS,S) vaccines. M72/AS01E is indicated to prevent Pulmonary Tuberculosis (TB) in adolescents and adults. This vaccine aims to prevent latent TB from progressing to active TB rather than preventing TB infection.

The M72/AS01E vaccine candidate has been under development since the early 2000s. In 2020, GSK announced a partnership with the Gates Medical Research Institute (Gate MRI) to develop M72/AS01E further. GSK licensed M72 to the Gates MRI. An adjuvant is an ingredient in some vaccines that can help create a more robust immune response. GSK retains ownership of the adjuvant. It was initially designed and clinically evaluated by the biopharmaceutical company GSK through the proof-of-concept phase (Phase 2b), in partnership with Aeras and the International AIDS Vaccine Initiative (IAVI). It was funded by GSK and, in part, by the Bill & Melinda Gates Foundation. In the Phase 2b clinical trial, this vaccine candidate provided approximately 54% protection against progression to active pulmonary TB for three years in TB-infected HIV-negative adults.

On June 28, 2023, Wellcome and the Gates Foundation announced funding to advance M72/AS01E (M72) through a Phase 3 clinical trial (NCT06062238). To support the M72 Phase 3 clinical trial, which will cost an estimated $550 million, Wellcome is providing up to $150 million, and the Gates MRI, a subsidiary of the Gates Foundation, will fund the remainder, about $400 million. On March 19, 2024, Gates MRI announced that the Phase 3 clinical trial was underway at the University of the Witwatersrand, Johannesburg, South Africa, the first of up to 60 study sites in seven countries, recruiting about 26,000 participants. Participants enrolled in the trial will be tested for latent TB using Interferon-Gamma Release Assays (IGRAs), which are a type of blood test. Of the 20,000 total participants, 18,000 will be IGRA-positive, which means they have evidence of latent TB; 1,000 will be people living with HIV; and 1,000 will be IGRA-negative. As of October 2024, Indonesia participates in the M72/AS01E vaccine clinical trial. Should the trial be successful, GSK would provide the adjuvant post-licensure. The estimated primary completion date for this study is April 2028. If M72/AS01E is approved, it could be the first new TB vaccine in 100 years.

M72/AS01E (M72) Tuberculosis Vaccine Indication

The WHO says TB is a preventable and curable disease, and that 25% of the global population is estimated to be infected with Mycobacterium tuberculosis. TB is the leading infectious cause of death worldwide. The BCG vaccine helps protect young children against severe systemic forms of TB but offers limited protection against pulmonary TB among adolescents and adults.

M72/AS01E (M72) Tuberculosis Vaccine Dosage

M72/AS01E: Two doses are administered intramuscularly in the deltoid region of the arm.

M72/AS01E (M72) Tuberculosis Vaccine News

June 10, 2024 - Indonesian Minister of Health Budi Gunadi Sadikin said Indonesia plans to conduct a trial of GSK's TB vaccine candidate.

March 19, 2024 - "The launch of this pivotal Phase 3 trial demonstrates our commitment to harnessing the power of medical innovation to fight diseases like TB that are particularly devastating for low- and middle-income countries," said Emilio A. Emini, Ph.D., CEO of the Gates MRI.

November 16, 2023 - Nicole Frahm, Ph.D. The head of Biomarker Development presented Immune Correlates of Protection for BCG and M72 TB Vaccines.

November 16, 2023 - Linda Han, MD, MPH, presented: Safety and immunogenicity of the investigational TB vaccine M72/AS01E-4 in people living with HIV.

June 28, 2023 - Bill Gates, the co-chair of the Bill & Melinda Gates Foundation, commented in a press release, "Greater investment in safe and effective TB vaccines alongside a suite of new diagnostics and treatments could transform TB care for millions of people, saving lives and lowering the burden of this devastating and costly disease."

May 10, 2022 - AFP reported that the M72 TB vaccine could save tens of thousands of South African lives yearly.

December 20, 2021 - A Multi-country, Epidemiologic Study to Assess the Interferon Gamma Release Assay Positivity and Build Capacity to Conduct a TB Vaccine Efficacy Study in Populations With a High TB Disease Burden.

June 30, 2020 - The WHO published - Vaccines and Immunization: Investigational vaccine candidate M72/AS01E: FAQs.

January 27, 2020 - GSK plc announced that it had licensed its M72/AS01E TB vaccine candidate to the Bill & Melinda Gates Medical Research Institute. Trevor Mundel, President of Global Health at the Bill & Melinda Gates Foundation. "Discovering and developing a new TB vaccine is a Gates Foundation priority, and we look forward to working in partnership with stakeholders in the global health community towards this goal."

October 29, 2019 -  GSK and IAVI reported that GSK's M72/AS01E candidate vaccine significantly reduced the incidence of pulmonary TB in HIV-negative adults with latent TB infection. These results demonstrate an overall vaccine efficacy of 50% during the three years after vaccination. In addition, the candidate vaccine has an acceptable safety and reactogenicity profile.

September 25, 2018 - Results from the Phase 2b Controlled Trial of M72/AS01 E Vaccine to Prevent Tuberculosis. Conclusions: M72/AS01E provided 54% protection against active pulmonary tuberculosis in adults infected with M. tuberculosis, without evident safety concerns. 

M72/AS01E Clinical Trials

Clinical Trial NCT01755598: Study to Evaluate the Efficacy of GlaxoSmithKline Biologicals' Candidate TB Vaccine in Adults. The results showed that administering two doses of M72/AS01E successfully reduced the development of active TB disease with 50% efficacy (90% CI, 12 to 71) in HIV-negative adults with latent Mycobacterium tuberculosis infection. Thirty-nine participants contributed to the primary vaccine efficacy analysis during 3 years of follow-up: 13 participants vaccinated with M72/AS01E developed pulmonary TB, compared with 26 participants in the placebo group.

Clinical Trial NCT06062238: TBV02-301 was First Posted on October 2, 2023 - 20,000 adolescents and adults will be randomized 1:1 to receive two doses of M72/AS01E or placebo. The primary endpoint: preventing bacteriologically confirmed pulmonary TB. This trial will be conducted in 3 cohorts: Interferon gamma release assay-positive Cohort, IGRA-Negative Cohort, and HIV Cohort.

0 min read
Availability: 
N/A/
Generic: 
M72/AS01 E
Drug Class: 
Subunit Vaccine Candidate
Condition: 
Last Reviewed: 
Wednesday, November 12, 2025 - 12:05
Brand: 
M72
Status: 
Manufacturer Country ID: 
FDA First In Class: 
Yes

VPM1002 Tuberculosis Vaccine

VPM1002 Tuberculosis Vaccine

The VPM1002 vaccine candidate is a genetically modified recombinant Bacille Calmette–Guérin (BCG) vaccine derived from the Mycobacterium bovis BCG Danish 1331P (rague) strain, characterized as rBCGäureCHly+. VPM1002 contains weakened tuberculosis (TB)-like bacteria. These are genetically modified so that immune cells can better recognize them. 

The VPM1002 vaccine candidate, expressing listeriolysin and deficient in urease, is generated in the gCG Prague genetic background, which offers a better safety profile than other BCG versions due to the absence of the RD2 genome segment. This vaccine's urease C gene replaces the listeriolysin O encoding gene from Listeria monocytogenes. By reducing urease C, phagosome acidification can occur, promoting phagolysosome fusion while also providing the optimal low pH for listeriolysin O stability.

VPM1002 was created in response and investigated for preventing pulmonary TB in newborns and TB recurrence in adults through post-exposure immunization. VPM1002 facilitates mycobacterial antigens being released into the cytosol while also triggering autophagy, inflammasome activation, and apoptosis because of antigens and bacterial DNA being released into the cytosol of the host cell due to Listeriolysin O expression in this vaccine. In addition, mycobacterial antigens that access the cytosol also improve antigen presentation.

The results from a double-masked, randomized, active-controlled phase 2 study were recently conducted in South Africa and published in The Lancet Infectious Diseases on June 27, 2022. The researcher's Interpretation: VPM1002 was less reactogenic than BCG. Both vaccines were immunogenic, although higher responses th the BCG vaccine.

Serum Institute of India Pvt. Ltd. (SSI) VPM1002 is based on a BCG vaccine developed at the beginning of the 20th century. In 2004, the Max-Planck-Gesellschaft granted Takzine Projekt Management (VPM) the license for the vaccine candidate. In 2012, the company began developing the vaccine with the SSI, which acquired a majority stake in VPM. VPM1002 is currently being studied for efficacy and safety in multiple centers in three clinical trials.—DrugBank Accession Number: DB15801.

Serum Institute of India was founded in 1966 by Dr. Cyrus Poonawalla to manufacture life-saving immuno-biologicals.

VPM1002 Indication

BCG vaccination activates the human immune system against viral infections, reducing the risk of severe disease progression and, in turn, lowering the death rate. VPM1002 is indicated to prevent tuberculosis recurrence. The WHO Global TB Report 2021 provides a comprehensive and up-to-date assessment of the TB epidemic. Previous Global TB reports are accessed at this WHO link.

VPM1002 Dosage

A single dose of VPM1002 calculates the vaccine's efficacy against TB recurrence.

VPM1002 Side Effects

A phase 2 study found that VPM1002 was not associated with serious safety concerns.

VPM1002 Vaccine News 2023

December 1, 2022 - "We decided from the beginning to improve classical BCG genetically in a way to exploit all its advantages. VPM1002 has turned out not only to be specific against the TB pathogen but also to have he ability to train the immunity," commented Prof. Dr. Stefan Kaufmann, the inventor of VPM1002 and head of the Emeritus Group Immunology at the Max Planck Institute for Multidisciplinary Sciences in Goettingen, Germany, in a press release.

June 27, 2022 - The Lancet Infectious Diseases published: A next-generation BCG vaccine moves forward.

April 13, 2022 - University Health Network, Toronto - Efficacy and Safety of VPM1002.

January 28, 2020 - Bacillus Calmette-Guérin Therapy for Bladder Cancer: An Update.

July 27, 2020 - The Serum Institute of India is conducting a phase III clinical trial of BCG vaccine candidate VPM1002 to evaluate its ability to reduce infection and severe disease outcomes.

April 23, 2020 - About 6,000 high-risk individuals will enter a phase 3 clinical trial to test the recombinant BCG vaccine. The Drug Controller General of India approved this study in India.

February 20, 2020 - A clinical study (SAKK 06/14) under the direction of Cyrill A. Rentsch, University Hospital Basel, Switzerland, together with the Swiss Group for Clinical Cancer Research (SAKK), is now investigating whether it is possible to avoid removing bladders from patients who have cancer of the bladder by using VPM1002. 

January 21, 2021 - IAVI announced it is partnering with the study sponsor, Serum Institute of India Pvt. Ltd., and Vakzine Projekt Management GmbH, a German development consulting firm for the pharmaceutical industry and a subsidiary of SIIPL, to conduct the Prime study. A multicenter, double-blind, randomized, active-controlled Phase III study to evaluate the efficacy and safety of the vaccine candidate VPM1002 compared to BCG for preventing Mycobacterium tuberculosis infection in HIV-exposed and HIV-unexposed newborn infants. Study sites are in Gabon, Kenya, South Africa, Tanzania, and Uganda.

September 19, 2020 - The Recombinant Bacille Calmette–Guérin Vaccine VPM1002: Ready for Clinical Efficacy Testing.

August 6, 2019 – The Indian Council of Medical Research announced it had launched India's first large-scale clinical study for new TB vaccines. The TB vaccine in this new clinical study offers a chance to contain the accelerating spread of multidrug-resistant TB. Treating TB requires a multi-drug course of treatment lasting up to 6 months and longer when treating drug-resistant TB.

VPM1002 Vaccine Clinical Trials

The vaccine is being tested in a Phase III clinical trial involving adult volunteers in India. In mice, VPM1002 gave better protection than BCG against aerosol infection with M tuberculosis and improved survival in severe combined immunodeficiency mice. The study by Cotton and colleagues bridged from the original hygromycin-resistant formulation of VPM1002 to a hygromycin-sensitive formulation.

June 2022 - Safety and immunogenicity of VPM1002 versus BCG in South African newborn babies: a randomized, phase 2 non-inferiority double-blind controlled trial - Between June 4, 2015, and October 16, 2017, 416 eligible newborn babies were randomly assigned and received the study vaccine. Seven (2%) of 312 participants in the VPM1002 group had a grade 3–4 vaccine-related adverse reaction or lymphadenopathy of 10 mm or greater in diameter compared with 34 (33%) of 104 participants in the BCG group (risk difference −30·45% [95% CI −39·61% to −21·28%]; pnon-inferiority<0·0001); VPM1002 was thus non-inferior to BCG for the primary outcome. Incidence of severe injection site reactions was lower with VPM1002 than BCG: scarring occurred in 65 (21%) participants in the VPM1002 group versus 77 (74%) participants in the BCG group (p<0·0001); ulceration occurred in one (<1%) versus 15 (14%; p<0·0001); and abscess formation occurred in five (2%) versus 23 (22%; p<0·0001). Restimulated IFNγ concentrations were lower in the VPM1002 group than in the BCG group at week 6, week 12, month 6, and month 12. The percentage of multifunctional CD4+ T cells was higher in the VPM1002 group than the BCG group at day 14 but lower at week 6, week 12, month 6, and month 12. The percentage of multifunctional CD8+ T cells was lower in the VPM1002 group than the BCG group at week 6, week 12, and month 6, but did not differ at other time points. Interpretation: VPM1002 was less reactogenic than BCG and was not associated with any serious safety concerns. Both vaccines were immunogenic, although higher responses th the BCG vaccine. VPM1002 is currently being studied for efficacy and safety in a multicenter phase 3 clinical trial in babies in sub-Saharan Africa.

0 min read
Availability: 
N/A
Generic: 
VPM1002
Drug Class: 
Recombinant Vaccine
Condition: 
Last Reviewed: 
Thursday, November 13, 2025 - 13:10
Brand: 
TBD
Abbreviation: 
rBCG
Status: 
Manufacturer Country ID: 
Kosher: 
Yes

Sabin IPV Polio Vaccine

Sabin IPV Vaccine Description 2022

Sabin Inactivated Poliovirus Vaccine (sIPV) is a liquid trivalent vaccine produced from Sabin poliovirus type 1, 2, and 3 strains grown on Vero cells. The sIPV, the Poliomyelitis Vaccine (Vero Cell), inactivated, Sabin strains, was prequalified by the World Health Organization in June 2022. The sIPV is available for United Nations agencies to purchase to support the global polio endgame strategy.

Sinovac Biotech Ltd. (SINOVAC) is a China-based biopharmaceutical company (NASDAQ: SVA) that focuses on the R&D, manufacturing, and commercialization of vaccines that protect against human infectious diseases.

Sabin IPV Vaccine Indication

The Sabin IPV vaccine is indicated to prevent the Wild Polio Virus in the hope of completely eradicating the disease.

Sabin IPV Vaccine Dosage

The vaccine is administered as three intramuscular injections of (0.5ml) on Day 0, Day 30, and Day 60, respectively, and a single intramuscular injection of vaccine (0.5 ml) at 18 months.

Sabin IPV Vaccine News

December 29, 2022 - Sinovac Biotech Ltd. sales of non-pandemic vaccines increased 37% from $128 million to $ 176 million on broad-based growth.

June 2, 2022 - SINOVAC Biotech Ltd. announced that it received WHO prequalification for its Poliomyelitis Vaccine.

May 13, 2022 - A polio vaccination campaign is scheduled to run over three days using the bivalent oral polio vaccine (bOPV) throughout Bethlehem and Jerusalem.

July 19, 2021 - SINOVAC Biotech Ltd. announced it received a drug registration approval from the National Medical Product Administration of China for its Sabin strain-based inactivated polio vaccine (Vero cells) ("sIPV") on July 12, 2021. The vaccine is expected to be available on the market by the end of 2021. SINOVAC also submitted an application for prequalification to the WHO in early 2020, and the WHO conducted its on-site inspection in February 2021.

April 15, 2021 - The Lancet published - Immunogenicity of two-dose and three-dose vaccination schedules with Sabin inactivated poliovirus vaccine in China: An open-label, randomized, controlled trial. This study offers evidence that two doses of sIPV administered at 4 and 8–11 months of age and three doses of sIPV administered at 2, 3, and 4 months of age both provide serological protection against poliomyelitis. 

February 19, 2021 - The twenty-seventh WHO meeting of the Emergency Committee under the International Health Regulations (2005) (IHR) on the international spread of poliovirus was convened by the WHO Director-General on 1 February 2021 with committee members and advisers attending via video conference, supported by the WHO Secretariat.  The Emergency Committee reviewed the data on wild poliovirus (WPV1) and circulating vaccine-derived polioviruses (cVDPV).

January 7, 2021 - South Korean-based biotechnology company announced that its Sabin-Inactivated Polio Vaccine (Sabin-IPV) Eupolio™ is the first to receive the World Health Organization prequalification be supplied globally.

September 1, 2019 - The sIPV is non-inferior compared with IPV. Both demonstrate a good safety profile.

April 8, 2019 - The results of this study were published in The Journal for Infectious Diseases. This analysis revealed that the studied sIPV demonstrated an immunogenicity profile non-inferior to that of the conventional IPV and a good safety profile.

Sabin IPV Vaccine Clinical Trials

The Sabin IPV Vaccine has been tested in over 38 clinical trials over the years.

 

0 min read
Generic: 
Inactivated Poliovirus Vaccine
Drug Class: 
Vaccine
Condition: 
Last Reviewed: 
Saturday, December 31, 2022 - 07:00
Brand: 
IPOL, Eupolio, Sinovac's sIPV
Abbreviation: 
IPV
Status: 
Rate Vaccine: 
QBsOtSMk

TAK-426 Zika Vaccine

TAK-426 Zika Vaccine

Takeda's TAK-426 (PIZV) is a purified, inactivated, alum-adjuvanted, whole Zika virus vaccine candidate. The absence of specific treatment makes a prophylactic Zika virus vaccine an unmet medical need, according to a recent study published by The Lancet. The vaccine was tested in a phase 1 study called PIZV or TAK-426 adjuvanted with aluminum hydroxide. TAK-426 was found well tolerated, with an acceptable safety profile, and was immunogenic in both flavivirus-naive and flavivirus-primed adults.

The peer-reviewed Journal of Infectious Diseases reported on December 9, 2022, on the two-year persistence of immune response to TAK-426 compared with that observed after natural infection. TAK-426 at 10-μg had an acceptable safety profile in FV-naive and FV-primed adults up to 24 months after dose 2. Seropositivity for neutralizing antibodies was 100% at one year and 93.8% and 76.2% at two years in FV-naive and FV-primed groups, respectively. TAK-426 responses were comparable in magnitude and kinetics with those elicited by natural Zika virus infection. These phase 1 study results support the further clinical development of TAK-426 for both FV-naive and FV-primed populations.

In February 2016, the World Health Organization (WHO) declared the Zika outbreak a Public Health Emergency of International Concern. On January 29, 2018, Takeda's Zika Vaccine candidate received U.S. FDA Fast Track Designation. On July 27, 2023, Takeda confirmed to support the expansion of our pipeline and the development of our programs, we have entered into partnerships with government organizations in Japan and the U.S. and leading global institutions.

Osaka, Japan-based Takeda is a patient-focused, values-based, R&D-driven global biopharmaceutical company committed to bringing Better Health and a Brighter Future to people worldwide. 

TAK-426 Vaccine Indication

The TAK-426 vaccine candidate prevents the Zika virus, which is caused by a virus transmitted by Aedes aegypti and Aedes albopictus mosquitoes. The Zika virus was first reported in continental South America in Brazil in May 2015. Zika virus is a flavivirus associated with congenital malformations and neurological complications. According to the World Health Organization (WHO), symptoms of Zika may include mild fever, skin rash, conjunctivitis, muscle and joint pain, malaise, or headache.

TAK-426 Vaccine Dosage

In a phase 1 study, TAK-426 was administered by intramuscular injection into the middle third of the deltoid muscle, preferably in the non-dominant arm on Days 1 (Visit 1) and 29 (Visit 4). Three different vaccine doses containing different protein concentrations (2, 5, or 10 micrograms [mcg]) will be given as a two-dose schedule to flavivirus-naive and primed healthy adults. Based on a recent study, the 10 μg TAK-426 dose was selected for further clinical development.

TAK-426 Vaccine News

December 8, 2022 - Researchers wrote: In conclusion, we confirm that PIZV has an acceptable safety profile in healthy adults aged 18–49 years, with no vaccine-related SAEs through 2 years after vaccination. Two vaccinations elicited immune responses that persisted at high titers (GMTs >100) comparable to those observed in convalescent ZIKV-infected patients up to 2 years after vaccination in FV-naive and FV-primed adults. These safety and immunogenicity profiles of the high dose (10-µg) PIZV confirm its suitability for further clinical development.

November 19, 2021 - The company presented: Measurement Of Zika Virus-Specific Antibodies Using A Microsphere-Based Competitive Immunoassay. 'Collectively, our data suggest that the assay can differentiate ZIKV-specific antibodies from antibodies to other FVs elicited by natural infection or vaccination. Availability of a ZIKV-specific antibody-based immunoassay will improve differential diagnosis, serosurveillance, and support development and implementation of ZIKV vaccines in FV endemic regions.'

May 18, 2021 - Study Interpretation: TAK-426 was well tolerated,hadh an acceptable safety profile, and was immunogenic in both flavivirus-naive and flavivirus-primed adults. Based on the safety and immunogenicity profiles of all TAK-426 doses assessed, the 10 μg TAK-426 dose was selected for further clinical development. Study Funding: Takeda Vaccines and the US Biomedical Advanced Research and Development Authority.

August 26, 2019 – A recent Zika study focused on eliminating the mosquito and sexual transmission of the Zika virus with a preventive vaccine.

January 29, 2018 - Takeda Pharmaceutical Company Limited announced that the U.S. FDA had granted Fast Track designation to TAK-426, Takeda's purified, inactivated, alum-adjuvanted, whole Zika virus vaccine candidate.

September 2, 2016 - Takeda announced that BARDA, the Biomedical Advanced Research and Development Authority, has selected Takeda's Vaccine Business Unit to develop a vaccine to support the Zika response in the US and affected regions worldwide. Initial funding from BARDA, which is a division of the Office of the Assistant Secretary for Preparedness and Response within the US Department of Health and Human Services, is for $19.8 million to cover the vaccine development through Phase 1, with potential funding of up to $312 million if all options to take the vaccine through Phase 3 trials and filing of the Biologics License Application in the USA. 

TAK-426 Clinical Trials

The Journal of Infectious Diseases reported that a randomized, observer-blind, placebo-controlled, dose-selection, phase 1 trial was conducted in 18–49-year-old adults at nine centers (7 in the United States and 2 in Puerto Rico) from 13 November 2017 to 24 November 2020. The primary objectives were safety, tolerability, and immunogenicity of 3 increasing doses of TAK-426 administered as two doses 28 days apart to flavivirus (FV)–naive and FV-primed adults. Here, we report on the safety and persistence of immunity up to 2 years after primary vaccination with 10-μg TAK-426, the highest dose, and compare neutralizing antibody responses with those observed after natural infection. Results: TAK-426 at 10-μg had an acceptable safety profile in FV-naive and FV-primed adults up to 24 months after dose 2. Seropositivity for neutralizing antibodies was 100% at one year and 93.8% and 76.2% at two years in FV-naive and FV-primed groups, respectively. TAK-426 responses were comparable in magnitude and kinetics with those elicited by natural Zika virus infection.

On September 21, 2021, The Lancet reported the findings of this clinical trial. Between Nov 13, 2017, and Oct 24, 2018, 894 volunteers were screened, and 271 enrolled (125 flavivirus-naive and 146 flavivirus-primed participants). All TAK-426 doses were well tolerated with no deaths, no vaccine-related serious adverse events, and similar rates of mainly mild to moderate adverse events. Interpretation: The researchers concluded that TAK-426 was well tolerated,hadh an acceptable safety profile, and was immunogenic in both flavivirus-naive and flavivirus-primed adults. Based on these results, TAK-426 will continue to be tested in clinical settings.

0 min read
Availability: 
U.S. - pending
Generic: 
TAK-426
Drug Class: 
Vaccine
Condition: 
Last Reviewed: 
Thursday, January 4, 2024 - 05:50
Abbreviation: 
PIZV
Status: 
Manufacturer Country ID: 

TY014 Yellow Fever Monoclonal Antibody

TY014  Anti-Yellow Fever Monoclonal Antibody

TY014 will be the first therapeutic in the world explicitly targeting Yellow Fever Virus (YFV) to enter clinical trials. It is anticipated that a monoclonal antibody therapeutic could be administered to infected cases to reduce disease severity within the patient and their contacts. TY014 is a fully engineered human IgG1 monoclonal antibody against the yellow fever virus manufactured with ovary cells. It targets a conserved epitope on the yellow fever virus envelope (E) protein. 

TY014  Anti-Yellow Fever Monoclonal Antibody Indication

TY014 is indicated as a postinfection intravenously therapy for yellow fever virus infection.

TY014  Anti-Yellow Fever Monoclonal Antibody News

July 30, 2020 - Phase 1 Trial of a Therapeutic Anti–Yellow Fever Virus Human Antibody. This phase 1 trial of TY014 did not identify worrisome safety signals and suggested potential clinical benefit, which requires further assessment in a phase 2 trial. 

November 18, 2019 - "Our successful trials pave the way for a potential first-in-class treatment for Yellow Fever. This rapid development following the similar achievement for Zika affirms the exciting collaboration between Tychan and WuXi Biologics to enable a new paradigm to expedite biologics development initiatives to address emerging infectious disease outbreaks and eventually potentially expand to other therapeutics," said Teo Ming Kian, Chairman of the Board, Tychan.

TY014  Anti-Yellow Fever Monoclonal Antibody Clinical Trials

Clinical Trial NCT03776786: Phase 1 Safety and Tolerability of an Antibody Against Yellow Fever Virus (TY014) in Humans

0 min read
Generic: 
Anti-yellow fever antibody
Drug Class: 
Antibody
Condition: 
Last Reviewed: 
Tuesday, June 18, 2024 - 02:30
Brand: 
TY014
Status: 
Manufacturer Country ID: 

QDENGA Dengue Vaccine

QDENGA® (TAK-003) Dengue Vaccine Clinical Trials, Dosage, Indication, Side Effects

Takeda GmbH's QDENGA® (TAK-003) (Dengue Tetravalent Vaccine [Live, Attenuated]) is an approved two-dose vaccine preventing dengue fever and/or Severe Dengue caused by any of the four serotypes of the dengue virus. QDENGA is based on dengue serotype 2, which provides the genetic backbone for all four dengue virus serotypes. The active substance of QDENGA (ATC code: J07BX04) contains live attenuated dengue viruses, which replicate locally and elicit humoral and cellular immune responses against dengue serotypes.

Takeda's clinical development program for QDENGA consists of Phase 3, Phase 2, and Phase 1 studies. Data in children and adolescents showed that QDENGA  induced immune responses against all four dengue serotypes in seropositive and seronegative participants. In addition, QDENGA was generally safe and well-tolerated. In a Phase 2 study published on October 17, 2024, TAK-003 elicited durable T cell responses against all four DENV serotypes, regardless of baseline serostatus, in individuals aged 4–16 years residing in dengue-endemic countries. Furthermore, TAK-003-elicited CD4+ and CD8+ T cells were multifunctional and persisted for up to 3 years post-vaccination.

On December 8, 2022, QDENGA became the first dengue vaccine approved in Europe for use regardless of previous exposure and without the need for pre-vaccination testing. Europe's Committee for Medicinal Products for Human Use adopted a positive opinion on October 13, 2022, recommending that Takeda GmbH be granted marketing authorization for QDENGA EMEA/H/C/005155. In 2023, Brazil was the first country to integrate QDENGA into its public health system. The World Health Organization (WHO) added QDENGA® (TAK-003) to its List of Prequalified Vaccines, effective May 9, 2024. On May 3, 2024, the WHO published a paper on dengue vaccines, including final guidance on the use of QDENGA in public vaccination programs based on the SAGE recommendation. The WHO-SAGE recommends that TAK-003 be considered for public programs in high-transmission areas for individuals aged 6-16. 

On November 3, 2025, Takeda announced data, including an exploratory analysis of a booster dose, confirming the favorable benefit-risk profile of QDENGA and that the two-dose regimen provides sustained protection against Dengue over 7 years. As of November 2025, QDENGA is authorized in the United Kingdom and a total of 41 countries.

Derek Wallace, President, Global Vaccine Business Unit, Takeda Pharmaceuticals, informed Vax-Before-Travel on November 6, 2025, "Dengue is spreading to areas once considered low-risk, with year-round peak seasons in many regions. These long-term data show that QDENGA's two-dose schedule provides sustained protection without a booster, simplifying vaccination efforts and improving our ability to combat Dengue globally."

Osaka-based Takeda Pharmaceutical Company Limited (TSE: 4502, NYSE: TAK) is a global, values-based, R&D-driven biopharmaceutical leader headquartered in Japan, committed to discovering and delivering life-transforming treatments, guided by our commitment to patients, our people, and the planet in approximately 80 countries and regions. For more information about QDENGA (TAK-003), visit www.TakedaVaccines.com. QDENGA - Trademark Details: 2019-06-04.

QDENGA (TAK-003) Vaccine Availability 2025

The WHO recommends that countries consider introducing TAK-003 into their routine immunization programs in areas with high Dengue transmission intensity, where it poses a significant public health problem. Since 2022, QDENGA has been authorized in 41 countries, and 18.6 million doses have been distributed in 11 endemic countries including Indonesia (approved Aug 2022, available April 2023), Brazil (approved Mar 2023, available June 2023, Thailand (approved May 2023, available Aug 2023), Argentina (approved Apr 2023, available Oct 2023), Malaysia (approved Feb 2024, available June 2024), Colombia (approved Sept 2023, available July 2024), Germany (approved Dec 2022, available Feb 2023), Finland (approved Dec 2022, available Feb 2023), Sweden (approved Dec 2022, available Feb 2023), Norway (approved Dec 2022, available Feb 2023), Denmark (approved Dec 2022, available Mar 2023), The Netherlands (approved Dec 2022, available Mar 2023), Luxembourg (approved Dec 2022, available March 2023), Czech Republic (approved Dec 2022, available April 2023), Austria (approved Dec 2022, available April 2023), Belgium (approved Dec 2022, available April 2023), Ireland (approved Dec 2022, available April 2023), Portugal (approved Dec 2022, available May 2023), Spain (approved Dec 2022, available May 2023), U.K. (approved Jan 2023, available 2024), Slovakia (approved Dec 2022, available Aug 2023), Italy (approved Dec 2022, available Oct 2023), Poland (approved Dec 2022, available Dec 2023), Israel (approved May 2024, available July 2024), Switzerland (approved Aug. 2024), Malasyia, Thailand, and Vietnam. As of 2025, availability in the Philippines and India was [ending.

Qdenga (TAK-003) Vaccine Efficacy

A Takeda-funded study, based in Thailand, concluded on June 17, 2025, that the pragmatic introduction of TAK-003 into the immunization program would prevent 44% of cases and 53% of hospitalizations, resulting in $ 1.346 billion in savings over 20 years.

On October 17, 2024, the journal NPJ Vaccines published results from a study that concluded that TAK-003 was well tolerated and elicited durable T cell responses against all four dengue virus (DENV) serotypes, irrespective of baseline serostatus, in children and adolescents aged 4–16 years living in dengue-endemic countries. TAK-003-elicited CD4+ and CD8+ T cells were multifunctional and persisted for up to 3 years post-vaccination.

An exploratory analysis published on September 11, 2024 (December 2, 2024) demonstrated a vaccine efficacy (VE) of 82.1% for the onset of protection with TAK-003 in the safety population, measured over the three months between the first and second doses. The journal Vaccine published a Short Communication on September 7, 2024, reporting that QDENGA's vaccine efficacy (VE) against disease was 82.1% up to 3 months after the first dose. On October 2, 2023, the WHO Strategic Advisory Group of Experts on Immunization (SAGE) confirmed that QDENGA demonstrated efficacy against all four serotypes of the virus in baseline seropositive children (4-16 years old) in endemic countries, as well as against serotypes 1 and 2 in baseline seronegative children. The SAGE recommended that the Vaccine be considered for introduction to children aged 6 to 16 years in settings with high dengue disease burden and high transmission intensity. Long-term efficacy results announced on June 9, 2022, supplemented previously published TIDES clinical study data, demonstrating QDENGA met its primary endpoint of overall VE against VCD, with 80.2% efficacy at 12-month follow-up, as well as all secondary endpoints for which there were a sufficient number of dengue cases at 18-month follow-up, including 90.4% VE against hospitalized Dengue. On May 20, 2021, an audio review of the Three-Year Efficacy of Takeda's Tetravalent Dengue Vaccine Candidate was published.

Qdenga (TAK-003) Time to Onset of Vaccine Efficacy

A study published in Vaccine on September 7, 2024, in the DEN-301 clinical trial estimated the Vaccine's efficacy onset to be approximately 2 weeks. Results from a study published in the Journal of Infectious Diseases on March 18, 2025, found that TAK-003 had a modest impact on asymptomatic dengue infections in the first months post-vaccination, particularly among baseline seropositive participants. 

QDENGA and U.S. FDA

Takeda announced on July 11, 2023, that the Company voluntarily withdrew the U.S. Biologics License Application (BLA) for TAK-003 following discussions with the U.S. Food and Drug Administration (FDA) regarding data-collection aspects that could not be addressed within the current BLA review cycle. The plan for TAK-003 in the U.S. will be further evaluated, given the need for travelers and those living in dengue-endemic areas, such as Puerto Rico, Mexico, and Florida. On November 22, 2022, Takeda announced that the FDA had accepted and granted priority review of the Biologics License Application (BLA) for QDENGA to prevent dengue disease caused by any dengue virus serotype in individuals aged four years through 60 years.

QDENGA and U.S. CDC

During the U.S. Centers for Disease Control and Prevention Advisory Committee on Immunization Practices (ACIPmeeting on October 26, 2023, Dr. Gabriela Paz-Bailey (CDC/NCEZID) led a QDENGA update. Wilbur Chen, MD, presented Updates on Dengue Vaccines. On June 22, 2023, I reviewed the following presentations: ACIP Dengue Vaccines Work Group introduction,   Wilbur Chen, MD, MSc; Economic analysis and health impacts of routine vaccination with TAK-003 dengue vaccine in San Juan, Puerto Rico; Summary of two economic models for dengue vaccine TAK-003 use in Puerto Rico; Partial Evidence to Recommendations Framework for Dengue Vaccine TAK-003, presented by Joshua Wong, MD. On February 23, 2023, the ACIP met to discuss the following presentations: Introduction, Dr. W Chen; Takeda dengue vaccine (TAK-003) safety and efficacy, Dr. Shibidas Biswal; Workgroup considerations, Dr. Gabriela Paz-Bailey. Previously, Laura Adams, DVM, MPH, DACVPM, led the U.S. CDC's Dengue Epidemiology vaccine overview on October 20, 2022, during the CDC/ACIP Dengue Vaccine Workgroup presentation.

QDENGA (TAK-003) Indication

Dengue is now detected in 129 countries and set a record for cases in 2024. As more travelers return to the U.S. with dengue virus infections, the risk of local mosquito-borne transmission increases. In European Union Member States, QDENGA is indicated for the prevention of dengue disease in individuals aged 4 years and above. It prevents Dengue, which is caused by the four Dengue serotypes. On May 15, 2024, the WHO confirmed that Qdenga is approved for use in children aged 6–16 in settings with a high dengue burden and transmission intensity. The WHO says TAK-003 does not prevent all cases of Dengue.

QDENGA (TAK-003) Dosage

Under the approved dosing regimen, QDENGA should be administered subcutaneously in a two-dose schedule (0 and 3 months) at a dose of 0.5 mL. The WHO SAGE recommends administering QDENGA in a 2-dose schedule with a 3-month interval between doses. QDENGA should be administered preferably in the upper arm in the deltoid region. QDENGA must not be injected intravascularly, intradermally, or intramuscularly. In Europe, the EMA approved after reconstitution, one dose (0.5 mL) contains Dengue virus serotype 1 (live, attenuated)*: ≥ 3.3 log10 PFU/dose Dengue virus serotype 2 (live, attenuated): ≥ 2.7 log10 PFU/dose Dengue virus serotype 3 (live, attenuated): ≥ 4.0 log10 PFU/dose Dengue virus serotype 4 (live, attenuated): ≥ 4.5 log10 PFU/dose.

On September 2, 2024, Vaccine published an analysis that concluded that one dose of TAK-003 was as effective as two doses.

QDENGA (TAK-003) Coadministraion

Co-administration with a hepatitis A vaccine has been studied without increased side effects or impaired antibody response. When co-administered with the yellow fever vaccine, a lower level of neutralizing antibodies against DENV1 was seen, the clinical significance of which is unclear

QDENGA Co-Administration With HPV9 Vaccine

According to a recent phase 3 clinical study published in the journal Vaccines (Volume 62, August 30, 2025, 127558), dengue-endemic countries, such as the Kingdom of Thailand, have already initiated national Human Papillomavirus (HPV) school-based vaccination programs. Researchers wrote on July 30, 2025, 'Due to the overlapping age range targeted by many vaccines, including 9vHPV (Merck GARDASIL 9®) and TAK-003 (Takeda's QDENGA®), integration of Dengue vaccination into existing vaccination programs could be a beneficial approach to increase vaccine coverage and reduce operational costs.'

QDENGA (TAK-003) Side Effects

A Phase 4 study published by ScienceDirect (Volume 42, Issue 26) on October 4, 2024, identified anaphylaxis as a safety signal for TAK-003 vaccines in adolescents. Most cases of anaphylaxis occurred 15 minutes after vaccination. In response, Brazil's Ministry of Health published recommendations to intensify efforts to ensure safe vaccination, including training for healthcare professionals and post-vaccination observation.

The ongoing Phase 3 study of TAK-003 demonstrated that rates of serious adverse events were 2.9% in the TAK-003 group and 3.5% in the placebo group during the continuing long-term follow-up (i.e., the second half of the three years following vaccination). Still, none of the cases were related to the study drug. The European Medicines Agency states that there is no clear evidence of an increased risk of severe dengue disease in individuals who have been previously infected with the disease.

QDENGA (TAK-003) Adverse Reactions

The most frequently reported adverse reactions post TAK-003 vaccination in subjects 4 to 60 years of age were injection site pain (50%), headache (35%), myalgia (31%), injection site erythema (27%), malaise (24%), asthenia (20%), and fever (11%). Very common (≥1/10 of subjects): upper respiratory tract infection, decreased appetite, irritability, headache, drowsiness, myalgia, injection site pain, injection site erythema, malaise, asthenia, and fever.

In February 2025, a study reported that Vaccine-related reactions were frequently reported, predominantly after the first dose in dengue-naïve participants.

QDENGA (TAK-003) Contraindications

Contraindications include hypersensitivity to the active substances or excipients listed, as well as a history of hypersensitivity to QDENGA. Individuals with congenital or acquired immune deficiency, including immunosuppressive therapies such as chemotherapy or high doses of systemic corticosteroids (e.g., 20 mg/day or 2 mg/kg body weight/day of prednisone for two weeks or more) within four weeks before vaccination. Individuals with symptomatic HIV infection or asymptomatic HIV infection, with impaired immune function, and pregnant and breastfeeding women.

QDENGA (TAK-003) Vaccination of Pregnant Women

A post-hoc analysis published on March 18, 2025, found no evidence of increased adverse pregnancy outcomes following unintentional TAK-003 vaccination within the exposure window, compared with placebo.

QDENGA Format

Qdenga is available as a powder and as an injection solution. 

QDENGA (TAK-003) Vaccination Breakthrough Case

The immune response to dengue infection is complex and multifaceted, and the infection can progress to severe disease with limited clinical signs. This study, published in October 2024, examined a case involving a child who developed dengue symptoms after receiving the Qdenga vaccination. Despite initial negative diagnostic results, molecular analysis confirmed DENV4 infection.

QDENGA (TAK-003) Price

Takeda indicated in a March 16, 2023, presentation that the price of QDENGA in Indonesia would be USD 40 per dose, USD 26 ex-factory. For the travel market segment, vaccine pricing will be similar to that of other innovative travel vaccines in their respective countries. For example, the retail price in Germany would be USD 115 per dose. In Brazil, reports indicate QDENGA's price is about $171.

Qdenga (TAK-003) Vaccine Revenues

QDENGA Q4 FY2024 (April 2024-March 2025) Revenue was JPY 35.6B (+259%), QDENGA's FY2024 H1 Revenue was JPY 19.9B (+863% growth); FY2024 Q1 Revenue was JPY 9.5B (+1,099% growth). 

QDENGA Vaccine Production

Biological E. Limited is committed to manufacturing up to 50 million QDENGA vaccine doses annually, accelerating Takeda's ability to deliver 100 million doses annually by 2030. 

QDENGA Vaccine News

November 3, 2025 - "QDENGA is the most comprehensively studied dengue vaccine, with more than 60,000 participants globally in the clinical program, and these long-term data highlight the durability of its safety and efficacy profile, across diverse populations worldwide," said Derek Wallace, M.D., president of the Global Vaccine Business Unit at Takeda.

August 25, 2025 - A study reported: We find some evidence of a risk of vaccine-induced disease enhancement in seronegative vaccine recipients for dengue serotypes 3 and 4, especially for children under 6 years of age. Because of this, the benefits of vaccination in lower-transmission settings are less specific, and more data on the long-term efficacy of Qdenga against serotypes 3 and 4 are needed.

June 9, 2025 - Thailand's Public Health Minister Somsak Thepsuthin unveiled a new national strategy during the 2025 ASEAN Dengue Day event, which includes deploying QDENGA vaccinations in 2025.

February 18, 2025 - The Philippine Medical Association requested access to new-generation dengue vaccines.

September 7, 2024: The journal Vaccine published a Short communication titled "Early onset of protection of the TAK-003 dengue vaccine: Data from the DEN-301 clinical trial." The Estimated time to the onset of Qdenga vaccine efficacy was approximately two weeks.

May 10, 2024 - The World Health Organization prequalified Takeda's QDENGA vaccine. "The prequalification of TAK-003 is an important step in the expansion of global access to dengue vaccines, as it is now eligible for procurement by UN agencies, including UNICEF and PAHO," said Dr Rogerio Gaspar, WHO Director for Regulation and Prequalification.

May 9, 2024 - Takeda announced financial results for fiscal year 2023 (period ended March 31, 2024).

October 3, 2023 – Takeda announced that the WHO Strategic Advisory Group of Experts on Immunization issued recommendations for the use of QDENGA.

July 11, 2023 - "Our clinical program was designed to account for the complex global nature of dengue, and data from our 4.5-year trial has built confidence in TAK-003's ability to help provide long-term protection against dengue, with a positive benefit and risk profile regardless of baseline serostatus," said Gary Dubin, M.D., president of Takeda's Vaccines Business Unit.

June 22, 2023 - The U.S. CDC's Advisory Committee on Immunization Practices reviewed various dengue vaccine presentations.

May 23, 2023 - Filomeno Fortes, Director of Portugal's Institute of Hygiene and Tropical Medicine, confirmed the authorization of the Qdenga vaccine.

May 11, 2023 - Takeda announced its collaboration with Brazil's National Health Surveillance Agency. In March 2023, QDENGA received the first approval in Latin America.

March 13, 2023 - José Manuel Caamaño, president of Takeda in Brazil, stated in a press release, "We are proud to make our vaccine available to the Brazilian government and health care providers with the hope that it may become an important tool to help combat dengue as part of an integrated dengue management program along with vector control."

February 6, 2023 - Takeda UK Ltd. announced that the U.K.'s Medicines and Healthcare products Regulatory Agency had granted marketing authorization for Qdenga. 

October 20, 2022: The U.S. CDC vaccine committee presented Dengue Epidemiology: Globally and in the U.S., including ACIP dengue vaccine recommendations.

October 14, 2022 - The EMA's human medicines committee recommended granting marketing authorization for the QDENGA Dengue Tetravalent Vaccine (Live, Attenuated) to prevent dengue virus serotypes 1, 2, 3, and 4 in individuals aged four years and above.

August 22, 2022: Gary Dubin, president of Takeda's Vaccine Business Unit, announced, "We're proud to introduce QDENGA as a new dengue prevention tool to the people of Indonesia. We will continue to work with additional regulatory agencies to make QDENGA available globally."

March 17, 2020 - The Lancet published the 'Safety and immunogenicity of a tetravalent dengue vaccine in children aged 2–17.

November 6, 2019: Takeda's Dengue Vaccine Candidate Demonstrates Protection in Children Aged Four to 16, Regardless of Previous Dengue Exposure. 

November 5, 2019 – Takeda Pharmaceutical Company announced plans for a new manufacturing plant in Singen, Germany, for TAK-003. 

January 29, 2019 - Takeda announced that TAK-003, which is in a pivotal Phase 3 trial, met the primary efficacy endpoint.

September 7, 2016 - Takeda Initiated the Global Phase 3 Clinical Trial (TIDES) of the Dengue Vaccine Candidate (TAK-003).

QDENGA TAK-003 Clinical Trials

The approval of QDENGA is based on results from 19 Phase 1, 2, and 3 trials involving more than 28,000 children and adults, including 4.5 years of follow-up data from the global, pivotal Phase 3 Tetravalent Immunization against Dengue Efficacy Study (TIDES) trial. The study was designed in collaboration with the World Health Organization (WHO) for a second-generation dengue vaccine. A study published in the journal Vaccine on August 22, 2023, reported an efficacy of approximately 80.2% (95% CI, 73.3–85.3) against any serotype in the primary analysis. QDENGA's effectiveness declined over time, with 62% (95% CI, 56.6–66.7) efficacy at 3 years and 61.2% (95% CI, 56.0–65.8) at 4.5 years.

As of June 9, 2022, through four and a half years, TAK-003 demonstrated 84.1% vaccine efficacy (VE) (95% CI: 77.8, 88.6) against hospitalized Dengue, with 85.9% VE (78.7, 90.7) in seropositive individuals and 79.3% VE (63.5, 88.2) in seronegative individuals. TAK-003 also demonstrated an overall vaccine efficacy (VE) of 61.2% (95% CI: 56.0-65.8) against virologically confirmed Dengue (VCD), with 64.2% VE (58.4-69.2) in seropositive individuals and 53.5% VE (41.6-62.9) in seronegative individuals. Observations of VE varied by serotype and remained consistent with previously reported results. TAK-003 was generally well tolerated, and no critical safety risks were identified. No evidence of disease enhancement was observed during the 54-month exploratory follow-up analysis.

0 min read
Availability: 
WHO Prequalified - Indonesia, Europe, UK, Brazil, Thailand, Argentina, Switzerland
Generic: 
TAK-003
Clinical Trial: 
https://www.takedavaccines.com/siteassets/vaccines/tak-003-clinical-development-program-infographic_final.pdf
Drug Class: 
Live-attenuated tetravalent vaccine
Condition: 
Last Reviewed: 
Monday, December 8, 2025 - 08:35
Brand: 
Qdenga
Abbreviation: 
TDV
Status: 
Manufacturer Country ID: 
FDA First In Class: 
Yes
Rate Vaccine: 
quEuHcLB

VGX-3100 HPV Cancer Vaccine

INOVIO VGX-3100 HPV Cancer Vaccine 2023

INOVIO's VGX-3100 is an investigational immunotherapy vaccine that includes DNA plasmids targeting the E6 and E7 proteins of human papillomavirus (HPV) types 16 and 18. VGX-3100 is HPV-specific immunotherapy developed as a non-surgical treatment for high-grade cervical dysplasia and related underlying persistent HPV infection. VGX-3100 works in vivo to activate functional, antigen-specific CD-8 T-cells to clear persistent HPV 16/18 infection and cause regression of precancerous cervical dysplasia. 

The Company announced on May 10, 2022, based on feedback from the U.S. Food and Drug Administration (FDA), INOVIO has changed its development plans for VGX-3100 for HPV-16/18-associated cervical HSIL to a biomarker-selected population. In a recent preliminary letter, the FDA advised INOVIO that the REVEAL2 Phase 3 study would not be sufficient to support approving a potential marketing application for VGX-3100 in that population.

INOVIO's Phase 3 program in cervical HSIL is assessing the efficacy of VGX-3100 to regress cervical HSIL, a direct precursor to cervical cancer, and to eliminate the HPV-16 and/or HPV-18 infection that causes these lesions. The REVEAL studies are prospective, randomized, double-blind, placebo-controlled trials evaluating adult women with HPV-16 and/or HPV-18 positive biopsy-proven cervical HSIL. REVEAL1 provided one-year post-endpoint safety data for a minimum of 198 participants randomized, while REVEAL2 will provide efficacy and one-month safety data for a minimum of 198 participants.

On May 11, 2023, the Company announced topline results for REVEAL2, the second Phase 3 trial for VGX-3100 as a treatment for cervical high-grade squamous intraepithelial lesions; Analysis of clinical characteristics of biomarker population is ongoing; findings to be shared in third quarter 2023; Trial results achieved statistical significance in all-participants population; Trial results did not meet the primary endpoint in biomarker-selected population.

INOVIO's first-of-their-kind DNA medicines are precisely designed DNA plasmids delivered through INOVIO's unique smart device directly into the body's cells to produce an immune response robust enough to potentially treat and prevent disease. Administration with the CELLECTRA device ensures that the DNA medicine is delivered directly into the body's cells, where it can go to work immediately, mounting an immune response. In addition, Inovio's DNA medicines are not interfering with or change in any way an individual's DNA, which is the case with gene therapy or gene editing.

Inovio is an innovative biotechnology company based in Pennsylvania focused on discovering, developing, and commercializing its synthetic nucleic technology targeted against cancers and infectious diseases.

VGX-3100 History

On December 9, 2020, Prakash Bhuyan, M.D., Ph.D., Senior Vice President and Head of HPV Therapeutic Clinical Development at INOVIO, said, "Anal dysplasia is a rare disease that is typically treated via surgical excision, electro-cautery, or laser therapy, with up to 50% of patients experiencing disease recurrence within one year of surgical treatment. We are encouraged by these positive results from our Phase 2 trial and look forward to continuing our work in Phase 3 trials to develop systemic DNA-based immunotherapy that leverages our DNA medicines platform to improve the current standard of care."

On January 6, 2021, Robert Edwards, M.D., Milton Lawrence McCall, Professor and Chair, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, and Principal Coordinating Investigator for the Phase 2 clinical trial, said, "These Phase 2 efficacy results are a very promising non-surgical advance for a recalcitrant disease that normally requires repetitive ablation and excision procedures to achieve disease and risk reduction, and may offer a more safe, tolerable and efficacious treatment option for patients."

VGX-3100 Indication

VGX-3100 is designed to treat precancers and cancers caused by human papillomavirus (HPV). HPV-16/18 causes more than 90% of all anal cancer, now considered one of the most rapidly rising causes of cancer incidence and mortality. According to the American Cancer Society, anal cancer will claim the lives of more than 1,300 people in the U.S., and 8,590 news cases (5,900 in women and 2,690 in men) will be diagnosed in 2020.

VGX-3100 Dosage

VGX-3100 is delivered intramuscularly, followed by electroporation with a CELLECTRA-5PSP delivery device.

VGX-3100 News

May 11, 2023 - The Company ended the first quarter of 2023 with $223.8 million in cash, cash equivalents, and short-term investments.

May 10, 2022 - The Company announced changes for VGX-3100 following input from the FDA on the need for additional trials for a marketing application for the treatment of HPV-associated cervical high-grade squamous intraepithelial lesions.

December 14, 2021 - INOVIO announced updates on the Phase 3 program for VGX-3100 for HPV-associated cervical high-grade squamous intraepithelial lesions, including a one-year follow-up of efficacy and safety data in participants from REVEAL1, completing enrollment in REVEAL2, and advancing its pre-treatment biomarker candidate for VGX-3100 to be further developed with QIAGEN. In addition, INOVIO's development partner within Greater China, ApolloBio Corp., dosed the first participant in a separate Phase 3 trial in China.

March 2, 2021 - INOVIO announced its phase 3 trial, REVEAL 1 met primary and secondary endpoints. INOVIO expects to present REVEAL 1 findings at a scientific meeting this year.

January 6, 2021 - INOVIO announced positive efficacy results for an open-label Phase 2 trial of VGX-3100 to treat HPV-16 and HPV-18-associated vulvar dysplasia. 

December 9, 2020 - INOVIO announced positive Phase 2 efficacy results demonstrating that DNA medicine VGX-3100, the Company's lead immunotherapy asset, showed resolution of HPV-16/18-associated precancerous anal lesions (HSIL) in 50% (11 of 22) of subjects six months following the start of treatment. The open-label, single-arm trial also showed VGX-3100 to be safe and well-tolerated in treating men and women with HPV-16-/18-associated anal dysplasia. INOVIO plans to pursue a registrational Phase 3 clinical trial for HPV-16-/18-associated anal dysplasia and apply rare and orphan disease designation for this indication in 2021.

March 26, 2020Inovio announced positive interim results from an open-label, Phase 2 study showing its lead DNA medicine candidate VGX-3100 to be safe and effective in treating men and women with anal dysplasia, also known as a high-grade squamous intraepithelial lesion (HSIL), a precancerous condition caused by high-risk human papillomavirus (HPV) types 16/18.

July 8, 2019Inovio announced that it had completed enrollment in its Phase 2 trial with VGX-3100 in patients with precancerous lesions of the vulva or vulvar intraepithelial neoplasia (VIN). VGX-3100 is an immunotherapy that targets human papillomavirus (HPV) 16 and 18 and is being studied to treat HPV-related precancerous lesions and the HPV infection that causes these lesions.

June 26, 2019 - Inovio has announced the completion of the target enrollment of 198 participants for its pivotal Phase 3 REVEAL 1 registration trial of VGX-3100, a novel DNA-based immunotherapy being tested to treat cervical dysplasia caused by human papillomavirus (HPV).

March 20, 2018 - Inovio will immediately receive $23 million in an upfront payment, an additional $20 million in future regulatory milestone payments, and double-digit tiered royalties on future sales. "With this license and collaboration agreement, we are now on the path to introduce late-stage innovative new drugs to meet severely unmet medical needs within the Greater China region," said Dr. Weiping Yang, Chief Executive Officer of ApolloBio. "We are excited at the potential for VGX-3100 to address multiple indications within HPV-associated precancer, and we are very pleased to launch this strategic collaboration with Inovio, an innovative global biotechnology partner."

VGX-3100 Clinical Trials

Clinical Trial NCT03185013: Phase 3 Clinical Trial REVEAL 1 Evaluation of VGX-3100 and Electroporation for the Treatment of Cervical HSIL. REVEAL 1 is designed to provide one-year safety data for at least 198 patients on VGX-3100—the last Update Posted: February 3, 2021.

Clinical Trial NCT03721978: Phase 3 Clinical Trial REVEAL 2 Evaluation of VGX-3100 and Electroporation for the Treatment of Cervical HSIL. A confirmatory Phase 3 trial (REVEAL 2) is currently enrolling and is designed to provide one-month safety data for a minimum of 198 patients. Last Update Posted: December 4, 2020.

Clinical Trial NCT03180684:  Phase 2 Evaluation of VGX-3100 and Electroporation Alone or in Combination With Imiquimod for the Treatment of HPV-16 and/or HPV-18 Related Vulvar HSIL

Clinical Trial  NCT03499795Phase 2 VGX-3100 Delivered Intramuscularly (IM) Followed by Electroporation (EP) for the Treatment of HPV-16 and/or HPV-18 Related Anal or Anal/Peri-Anal, High-Grade Squamous Intraepithelial Lesion (HSIL) in Individuals Seronegative for Human Immunodeficiency Virus (HIV)-1/2.

0 min read
Availability: 
Pending
Drug Class: 
Vaccine
Condition: 
Last Reviewed: 
Thursday, May 11, 2023 - 14:20
Brand: 
VGX-3100
Status: 
Manufacturer Country ID: