Fact-checked by Grok 2 weeks ago

Modified vaccinia Ankara

Modified Vaccinia Ankara (MVA) is a highly attenuated, replication-deficient strain of the developed as a safe and effective third-generation against and related orthopoxviruses, such as monkeypox, and widely used as a platform for recombinant vaccines targeting various infectious diseases and cancers. MVA originated from the chorioallantois virus Ankara (CVA) strain, which was subjected to over 570 serial passages in chicken embryo fibroblast cells during the and by Anton Mayr at of Medical Microbiology, Infectious and Tropical Diseases at the University of and the Bavarian State Institute for , resulting in approximately 30 kilobases of genomic deletions that eliminated factors and rendered it non-replicative in mammalian and human cells. The strain was renamed MVA after its 516th passage in , and these modifications led to the loss of six major immunomodulatory genes, enhancing its safety profile while preserving strong through efficient expression of viral and inserted recombinant genes. First administered to humans as a in 1971 and licensed in in 1977, MVA was given to over 120,000 individuals by 1980 with an excellent safety record, demonstrating no serious adverse events even in vulnerable populations. Further development by Bavarian Nordic A/S produced the MVA-BN variant, marketed under brand names including Imvanex in Europe, Imvamune in Canada, and Jynneos in the United States, which received European Medicines Agency approval in 2013 for active immunization against smallpox, Canadian approval that same year, and U.S. Food and Drug Administration approval in 2019 for prevention of smallpox and monkeypox in adults aged 18 and older. Unlike first- and second-generation vaccinia vaccines like Dryvax or ACAM2000, which can replicate in humans and cause complications such as myocarditis or skin lesions, MVA does not replicate in human cells, making it suitable for immunocompromised individuals, those with atopic dermatitis, HIV, or cancer, and earning it a Biosafety Level 1 classification. The World Health Organization's Strategic Advisory Group of Experts on Immunization recommends MVA-BN for high-risk groups during mpox outbreaks, such as close contacts of cases, and for laboratory personnel handling orthopoxviruses, with its inclusion in the U.S. Strategic National Stockpile underscoring its role in public health emergency preparedness. Beyond protection, MVA's genetic stability and ability to induce robust innate and adaptive immune responses— including type I production via the cGAS/ pathway—have made it a cornerstone for vaccine vector development over the past 25 years, with recombinant versions in clinical trials for pathogens like , , , , , and MERS-CoV, as well as cancer immunotherapies. Its versatility stems from efficient foreign gene insertion without interference from , enabling single-dose regimens that elicit long-lasting immunity, and ongoing research continues to explore its potential against emerging infectious threats.

History and Development

Origin of the Ankara Strain

The strain of , designated as Chorioallantois Vaccinia virus Ankara (CVA), was developed at the Turkish Vaccine Institute in , , through repeated serial passages alternating between donkeys and calves to produce smallpox vaccine material. Pustule harvests from these animal passages served as the primary source for vaccine propagation, reflecting standard practices in poxvirus vaccine manufacturing during the mid-20th century. This strain emerged as a variant of earlier vaccinia lineages, likely tracing back to the Board of Health strain from the early 1900s, and was maintained for routine vaccine production in . In 1953, the Ankara strain was transferred to the Institute for Infectious Diseases and Tropical Medicine in , , where it underwent purification via ultracentrifugation and two additional passages in via cutaneous . Following this, it was introduced for public smallpox in the of starting in the 1954/55 season, demonstrating typical virus properties such as robust replication in mammalian hosts and induction of protective immunity, though with occasional side effects including secondary pox formations in approximately 781 reported cases that year. Early characterization confirmed its standard poxvirus morphology, antigenicity, and pathogenicity profile akin to other strains used at the time. During the , Turkish and German researchers conducted animal experiments with the Ankara strain to evaluate its propagation efficiency and , alongside limited human trials as part of ongoing smallpox vaccination efforts, which by then had reached over 100,000 administrations globally with a favorable safety profile compared to less attenuated strains. These studies highlighted the strain's potential for broader application amid the World Health Organization's Intensified Smallpox Eradication Programme, launched in 1967 to accelerate global elimination through improved surveillance and safer vaccines.

Attenuation and Serial Passaging

Modified vaccinia Ankara (MVA) was developed by Anton Mayr and his colleagues at the Institute of Medical Microbiology, Infectious and Epidemic Diseases, , in the late and early as a safer alternative to conventional vaccinia virus strains for . The process began with the chorioallantois vaccinia virus Ankara (CVA) strain, which was subjected to extensive serial passaging to attenuate its while preserving . The was accomplished through more than 570 serial passages of CVA in primary chicken embryo fibroblast (CEF) cells, a non-permissive host for most poxviruses, which selected for mutants with restricted host range. This prolonged adaptation process favored the emergence of replication-deficient variants incapable of productive replication in mammalian cells, as the virus evolved to propagate efficiently only in avian CEF cultures. By the 516th passage in , the strain was renamed MVA, and passaging continued to refine its safety profile. These passages resulted in substantial genomic alterations, including the loss of approximately 31 —about 15% of the original —manifested as six major deletions that truncated or eliminated 31 open reading frames, primarily those involved in host immune evasion and . The deletions, designated I through VI, disrupted key genes essential for replication in human and other mammalian cells, rendering MVA highly host-restricted. was verified in preclinical animal models, where MVA demonstrated markedly reduced compared to wild-type strains. In rabbits, intradermal produced no lesions, unlike the pronounced pock formation seen with parental CVA. Intracerebral challenge in mice showed survival rates exceeding 90% at doses lethal to wild-type , indicating negligible neurovirulence. Similarly, in nonhuman primates such as macaques, MVA induced no systemic and provided protective immunity against challenges without evidence of dissemination or severe adverse effects. These findings confirmed MVA's safety margin, paving the way for its evaluation in human trials.

Role in Smallpox Eradication

In the mid-1970s, (MVA) was introduced in as a safer alternative to traditional vaccines for primary , particularly in children and immunocompromised individuals. Developed through extensive , MVA demonstrated reduced while eliciting protective immunity against orthopoxviruses, addressing concerns over the reactogenicity of earlier strains. Its deployment marked a shift toward third-generation vaccines during the waning phases of the global eradication effort, prioritizing safety in vulnerable populations without compromising efficacy. During the final years of the World Health Organization's (WHO) smallpox eradication campaign from 1976 to 1980, MVA was administered to over 120,000 individuals, primarily in , with an exemplary safety record. No serious adverse events were attributed to MVA, with recipients experiencing only mild local reactions and no systemic complications such as fever or progressive vaccinia. In comparison to first-generation vaccines like Dryvax, which carried risks of severe side effects including and in approximately 1 in 1,000 primary vaccinations, MVA exhibited significantly lower reactogenicity while preserving robust and rates. This profile made it ideal for the intensified surveillance and containment strategies employed in the campaign's endgame. Following the WHO's declaration of smallpox eradication in 1980, MVA production and use were discontinued, and remaining stocks were archived. Interest in MVA revived in the late amid growing concerns, as intelligence revealed potential state-sponsored variola virus programs and the risks of accidental release from retained laboratory stocks. This resurgence positioned MVA as a key candidate for modern stockpiles, leveraging its historical safety data to support renewed clinical evaluation and manufacturing scale-up.

Biological Characteristics

Genomic Modifications

Modified vaccinia Ankara (MVA) exhibits a reduced of approximately 178 kilobase pairs (kbp), compared to about 192 kbp in its parental chorioallantois virus Ankara (CVA) , primarily due to the accumulation of deletions during serial passaging in chicken embryo fibroblasts. These deletions total around 31 kbp and encompass six major genomic regions (deletions I through VI), affecting over 30 open reading frames (ORFs) involved in host interaction and . Specifically, the deletions remove or fragment genes encoding host-range factors and immunomodulatory proteins, while preserving essential viral genes necessary for production and presentation. Key deletions include region II in the left terminal inverted repeat, spanning about 4.5 kbp and eliminating the K1L gene, which encodes an repeat-containing protein that restricts host range by inhibiting cellular signaling pathways. Deletion I, the largest at around 12 kbp in the left terminus, disrupts the C12L gene (homologous to the cowpox SPI-1 serine protease inhibitor) and several immunomodulatory ORFs that modulate innate immune activation. Other notable losses in deletions III, IV, and VI involve genes like A52R and A53R ( signaling inhibitors) and C3L/C2L ( binding proteins), further impairing the virus's ability to evade host defenses. These genomic alterations contribute to MVA's avirulent by abolishing factors required for efficient and spread in mammalian hosts, rendering the replication-deficient in human cells. Notably, the deletion of K1L prevents inhibition of the pathway, as K1L normally blocks IκBα degradation and RelA acetylation, leading to unchecked activation of this and heightened innate immune stimulation upon infection. This enhanced immune signaling, combined with the loss of other immunomodulatory genes, underscores MVA's safety and as a platform.

Replication Cycle and Host Restriction

Modified vaccinia Ankara (MVA) exhibits a highly restricted replication cycle in most mammalian cells, including cells, where infection is abortive despite successful entry and uncoating. Upon entry via fusion, MVA initiates early , producing proteins necessary for replication, which proceeds to a limited extent. However, progression to late and virion assembly is severely impaired, resulting in the accumulation of aberrant, non-infectious particles rather than progeny virions. This is attributed to host restriction factors such as the zinc-finger antiviral protein (ZAP), which targets late-stage , and other innate defenses that prevent full dissemination. In contrast, MVA undergoes productive replication exclusively in avian cells, such as chicken embryo fibroblasts (CEF), where all stages of the poxvirus —early and late , , and virion assembly—occur efficiently, yielding high titers of infectious particles. This host range restriction, stemming from genomic deletions in host adaptation genes, enables large-scale production in CEF cultures while ensuring safety in mammalian hosts. For instance, in CEF, MVA can increase over 400-fold within 24 hours, supporting robust amplification without the need for mammalian cell lines. MVA infection potently activates host antiviral responses in mammalian cells, more efficiently than replicating poxviruses, due to its inability to fully evade innate immunity. It strongly induces type I interferons through the cGAS-STING pathway, leading to upregulation of interferon-stimulated genes, and triggers via pro-apoptotic factors like NOXA while downregulating anti-apoptotic MCL1. These responses, including activation and signaling, limit viral spread and enhance immunogenicity without productive infection. In vivo, MVA expresses antigens transiently in directly infected cells but fails to replicate or spread systemically in mammalian hosts, minimizing tissue damage and pathogenicity. This localized expression suffices to prime robust adaptive immunity, as seen in mouse models where low doses elicit strong CD8+ T-cell responses without dissemination to distant organs or induction of lesions. Such behavior underscores MVA's safety profile, with no observed virulence in primates or humans even at high doses.

Safety Profile and Immunogenicity

Modified vaccinia Ankara (MVA) exhibits an attenuated reactogenicity profile characterized by predominantly mild and transient local reactions, such as injection-site , induration, pain, and swelling, which occur in the majority of recipients but resolve within days without sequelae. Unlike replication-competent vaccinia vaccines like ACAM2000, MVA does not cause severe adverse events such as or at rates above background, with only rare cases reported across multiple clinical evaluations as of 2025, and no established . Systemic reactions, including , , and , are also mild to moderate and occur at rates comparable to , underscoring MVA's favorable safety margin due to its replication deficiency in human cells. MVA elicits broad , inducing robust + T-cell, + T-cell, and humoral immune responses, facilitated by its ability to support prolonged expression in non-permissive host cells despite abortive replication. This results in strong neutralizing production and T-cell mediated immunity, often superior to and noninferior to traditional strains in rates. MVA demonstrates suitability for vulnerable populations, including HIV-positive individuals with CD4 counts above 350 cells/mm³, where it maintains a benign safety profile without impacting or counts, and generates comparable immune responses to those in immunocompetent hosts. Limited data support its use in pregnant women at high risk of exposure, as a non-replicating with no observed fetal harm in preclinical models, though human evidence remains preliminary. As of 2025, phase 3 trials in children, adolescents, and pregnant women have further confirmed its safety profile, with mild reactogenicity similar to adults and no serious adverse events observed. Antibody responses to MVA wane over time, typically within 1–2 years post-, but T-cell memory persists for several years, providing durable cellular . Booster doses effectively restore and enhance both humoral and cellular immunity, supporting long-term vaccination strategies.

Applications as a Poxvirus Vaccine

Smallpox Vaccination

Modified vaccinia Ankara (MVA) serves as a third-generation smallpox , designed for enhanced safety in modern populations while providing robust immunogenicity against orthopoxviruses like variola virus. As a non-replicating live , MVA-BN (the strain) was developed under the U.S. Project BioShield initiative to address biodefense needs following the , which heightened concerns over potential involving . The U.S. (FDA) approved JYNNEOS (MVA-BN), the commercial formulation of MVA, on September 24, 2019, for the prevention of disease in adults 18 years and older at high risk for . In the , it is approved as Imvanex, with similar indications. This approval marked MVA as a safer alternative to first- and second-generation vaccines like ACAM2000, which replicate in human cells and carry risks of severe adverse events such as and progressive . A pivotal phase 3, randomized, published in 2019 demonstrated MVA-BN's efficacy through non-inferiority to ACAM2000 in inducing protective s. In the study involving 440 healthy adults, two subcutaneous doses of MVA-BN (28 days apart) achieved seroconversion rates of 95.6% by vaccinia-specific (PRNT) and 95.0% by (ELISA) for vaccinia-specific IgG, comparable to 94.3% and 94.8% with ACAM2000, respectively. The trial confirmed MVA-BN's ability to elicit take rates (vesicle formation indicating ) in 91.8% of recipients after the second dose, supporting its protective potential against . MVA-BN exhibits a superior safety profile compared to replicating vaccines, with significantly fewer systemic and local adverse events. In the phase 3 trial, grade 3 or higher adverse events occurred in 8.2% of MVA-BN recipients versus 35.0% in the ACAM2000 group, and cardiac events were rare (0.3% versus 1.5%). No cases of or were reported with MVA-BN, making it suitable for broader use, including in individuals with or where ACAM2000 is contraindicated. In response to bioterrorism threats, the U.S. and have prioritized stockpiling MVA-BN vaccines for emergency preparedness. The U.S. government, through the Biomedical Advanced Research and Development Authority (BARDA), has procured millions of doses of JYNNEOS for the , with ongoing contracts ensuring supply into 2026. Similarly, the has established framework agreements enabling member states to stockpile MVA-BN as a critical . The standard dosing regimen for MVA-BN involves two 0.5 mL doses, each containing 10^8 infectious units (measured as TCID50), administered subcutaneously or intradermally 4 weeks apart. This schedule elicits durable humoral and cellular immunity, with geometric mean titers peaking 2 weeks post-second dose and persisting for at least 2 years.

Mpox Prevention

In response to the 2022 global mpox outbreak, the U.S. Food and Drug Administration (FDA) issued an Emergency Use Authorization (EUA) on August 9, 2022, for JYNNEOS, allowing intradermal administration in adults aged 18 years and older and subcutaneous administration in at-risk adolescents aged 12-17 to prevent mpox disease, building on its 2019 approval for subcutaneous use in high-risk adults. Similarly, the European Medicines Agency (EMA) recommended approval of Imvanex for mpox prevention in at-risk adults on July 22, 2022, extending its prior authorization for smallpox based on the same cross-protective evidence. Observational studies during the 2022 clade IIb outbreak estimated JYNNEOS effectiveness at 66-85% against symptomatic following two doses administered 28 days apart, with lower protection (around 35-75%) after a single dose. By the end of 2023, over 1 million doses of MVA-based like JYNNEOS had been administered globally as part of outbreak response efforts, primarily targeting high-risk groups such as men who have sex with men and close contacts. In 2024-2025, amid the emergence and spread of the more virulent Ib mpox variant in and beyond, MVA-based vaccines were deployed in vaccination campaigns, with modeling analyses indicating that targeted averted thousands of potential cases by reducing in outbreak hotspots. In March 2025, the FDA approved a freeze-dried formulation of JYNNEOS, improving shelf-life and ease of distribution. As of November 2025, amid ongoing clade Ib in , the WHO has allocated over 5.85 million doses of MVA-based vaccines to affected countries, though access and deployment challenges persist. A longitudinal published in The Lancet Infectious Diseases in 2025 tracked responses up to 12 months post- or , finding that neutralizing antibodies waned significantly after JYNNEOS (declining by over 50% at 6-12 months) but remained stable in individuals recovering from natural , suggesting durable long-term protection from prior exposure compared to vaccine-induced immunity. JYNNEOS is recommended for both (PrEP) in high-risk individuals and (PEP) when administered within 4-14 days of to potentially mitigate severity, though PEP efficacy data remain limited by study biases. To address vaccine supply constraints during outbreaks, an FDA-authorized intradermal dosing regimen (0.1 mL per dose) was implemented in , allowing each vial to yield up to five doses while maintaining comparable to .

Use as a Viral Vector

Construction of Recombinant MVAs

The construction of recombinant Modified Vaccinia Ankara (MVA) viruses primarily relies on to insert foreign s into the viral genome, enabling its use as a versatile . This process involves transfecting permissive host cells, such as chicken embryo fibroblasts (CEF) or DF-1 chicken fibroblast cells, that are simultaneously infected with wild-type MVA. A shuttle or transfer carrying the target , flanked by sequences homologous to the desired insertion site in the MVA genome (typically 500–1,000 base pairs on each side), facilitates during . The foreign is placed under the control of poxvirus-specific promoters, such as the synthetic early/late promoter Psyn for robust late-phase expression or the modified H5 promoter (PmH5) for balanced early and late transcription, ensuring efficient production without interfering with viral fitness. Insertion sites are strategically chosen in non- regions of the MVA genome to preserve core replication and host restriction functions. Common loci include the deletion III (del III) region or intergenic regions (IGRs), such as those between genes like MVA069R and MVA070L, which correspond to areas already fragmented by the six major genomic deletions (I–VI) accumulated during MVA's . These sites, including del II and del III, accommodate foreign DNA without restoring pathogenicity, as they lie outside conserved poxvirus genes critical for intracellular lifecycle. For multi-gene constructs, sequential insertions or multi-plasmid approaches target multiple non-essential loci to avoid overcrowding a single site. To identify and isolate recombinants, selection markers like (GFP), , or (lacZ) are incorporated into the transfer plasmid, allowing visual screening of positive plaques under fluorescence or enzymatic assays. The recombination efficiency is low (approximately 0.1% of progeny viruses), necessitating multiple rounds of plaque purification on CEF monolayers to enrich for stable recombinants and excise transient markers via intragenomic recombination or Cre-loxP systems. Final clones are verified by , restriction enzyme digestion, and sequencing to confirm insert integrity and absence of unwanted mutations. Compared to other viral vectors, recombinant MVA offers a large foreign insert capacity of up to 30 kb, enabling the expression of complex multi-antigen cassettes while maintaining genetic stability over serial passaging. As a cytoplasmic replicating , MVA recombinants do not integrate into the host genome, minimizing risks of and ensuring transient, high-level expression in permissive cells. This combination of safety, capacity, and ease of engineering has made MVA a preferred platform for development.

Delivery of Heterologous Antigens

Modified vaccinia Ankara (MVA) serves as an effective for the delivery of heterologous antigens by incorporating foreign genes into its , enabling their expression in host cells following . These inserted genes, such as those encoding HIV-1 Env and proteins, virus (GP), or spike protein, are driven by poxvirus promoters to produce the antigens in infected cells, including antigen-presenting cells like dendritic cells. This process facilitates and presentation via both ( and class II pathways: endogenous antigens are degraded by proteasomes and presented on MHC I to activate + T cells, while and proteasomal pathways contribute to MHC II presentation for + T cell stimulation, enhancing both cellular and humoral . MVA vectors support multivalent designs, allowing the co-expression of multiple antigens within a single construct to broaden immune coverage against related pathogens. A prominent example is MVA-BN-Filo, which simultaneously expresses glycoproteins from , , Marburg marburgvirus, and Taï Forest ebolavirus, enabling a single to target multiple filoviruses responsible for hemorrhagic fevers; it was approved in the as Mvabea in 2020 for immunization against virus disease in individuals aged 1 year and older when given as a booster in a prime-boost regimen with Ad26.ZEBOV (Zabdeno). This approach induces robust, cross-reactive and T cell responses, with prime-boost regimens (e.g., with adenovirus vectors) further amplifying glycoprotein-specific neutralizing antibodies and cellular immunity. The non-replicating nature of MVA in cells confers key immunological advantages for delivery, as it promotes strong cytotoxic T-lymphocyte (CTL) responses without the immune evasion mechanisms employed by wild-type poxviruses, such as interference with host . By aborting replication late in the viral cycle, MVA avoids downregulation of MHC molecules and enhances availability for , leading to potent, polyfunctional + T cell activation that is critical for controlling intracellular pathogens. This safety profile minimizes anti-vector immunity in repeat dosing, allowing sustained focus on transgene-specific responses. In , MVA vectors expressing antigens like clade B gp120 Env have been tested in regimens, such as the HVTN 505 trial (DNA prime/MVA boost), which elicited + and CD8+ T cell responses but failed to prevent acquisition, highlighting challenges in achieving protective efficacy despite immunogenicity. Conversely, for , MVA-BN-Filo has shown success as a booster in schedules with rVSV-ZEBOV, enhancing durable GP-specific antibodies and T cell responses that contributed to protection during the 2014-2016 outbreak. For , MVA-based candidates like MVA-SARS-2-S, expressing the full-length , have demonstrated strong neutralizing antibodies and T cell immunity in preclinical models and early trials; as of 2025, related candidates such as MVA-SARS-2-ST are in clinical evaluation as potential boosters for variant containment.

Clinical Research and Trials

Preclinical and Early Human Studies

Preclinical studies of Modified vaccinia Ankara (MVA) demonstrated robust protection in various animal models against challenges, establishing its potential as a safe platform. In murine models of infection, high-dose MVA (10^8 PFU) provided full protection against lethal intranasal WR challenge (25 LD50), with survival rates comparable to the licensed Dryvax , while lower doses (10^6 PFU) offered partial protection (60-80% survival). In rabbits, a model for aerosolized , MVA (IMVAMUNE, 10^8 TCID50) administered as one or two doses achieved 100% survival following challenge with ~500 LD50 rabbitpox , reducing clinical signs and lesions compared to unvaccinated controls that succumbed by day 7. Nonhuman (NHP) studies further validated MVA's efficacy; for instance, MVA vaccination in cynomolgus macaques resulted in 100% survival against lethal (MPXV) clade I challenge (5 × 10^7 PFU), with significantly reduced blood viremia and throat infectious loads versus controls (1/6 survival). Similarly, in challenge models, an MVA-BN-Filo boost following Ad26.ZEBOV priming protected 100% of cynomolgus macaques (25/25) from intramuscular EBOV Kikwit (100 PFU), correlating with high EBOV glycoprotein-binding antibody levels. Early human studies in the focused on Phase I dose-escalation trials assessing MVA's and in healthy volunteers. A 2002-2003 Phase I trial of MVA as a (10^7-10^8 TCID50) in vaccinia-naive adults showed no serious adverse events, with mild local reactions and transient systemic symptoms, alongside induction of vaccinia-specific neutralizing antibodies and T-cell responses in most participants. For HIV applications, a Phase I trial of DNA-prime/MVA-boost regimens (MVA- at 10^8 PFU) in healthy volunteers confirmed across doses, eliciting strong HIV-specific + and + T-cell responses without vector-specific interference. Proof-of-concept for MVA as a emerged in early Phase I/II studies targeting heterologous antigens, highlighting its capacity for Th1-biased immunity. In a 2008 Phase I/IIa trial of MVA-H5 (expressing A H5N1 ) in healthy adults, doses of 10^7-10^8 TCID50 were safe and induced H5-specific and T-cell responses, with profiles indicating a Th1-dominant profile (elevated IFN-γ). For cancer, a 2003 Phase I trial of recombinant MVA expressing (MVA-CEA) in advanced patients demonstrated safety and CEA-specific T-cell proliferation, with assays showing Th1-skewed responses (IFN-γ over IL-4) in responders. These foundational studies identified limitations, such as variable responses in low-responders, where approximately 10-20% of participants showed suboptimal T-cell induction, prompting exploration of adjuvants like TLR agonists to enhance without compromising .

Approved Vaccines and Efficacy Data

The JYNNEOS vaccine (also known as Imvamune or Imvanex), based on the modified vaccinia Ankara-Bavarian Nordic (MVA-BN) strain, received approval from the U.S. (FDA) in September 2019 for the prevention of and in adults aged 18 years and older at high risk of exposure. In response to the 2022 mpox outbreak, the vaccine's indication was expanded to include broader use for prevention, with the (EMA) granting similar approvals for in 2013 and in 2022. In September 2024, the EMA extended the indication of Imvanex to adolescents aged 12-17 years. Pivotal trials demonstrated through significant fold increases in vaccinia-specific neutralizing titers (e.g., over 20-fold) following the two-dose regimen, establishing a correlate of protection against orthopoxviruses. Another MVA-based product, MVA-BN-Filo, was approved by the in July 2020 as a component of a heterologous prime-boost regimen (with Ad26.ZEBOV) for against virus disease caused by in individuals aged one year and older. This targets filovirus glycoproteins and has shown durable humoral immune responses, with rates exceeding 95% after the full regimen in clinical studies. The LC16m8 , an attenuated strain developed in and distinct from MVA though sharing third-generation safety features, was licensed in in 1975 for prevention and received emergency use listing in November 2024 for in individuals aged one year and older. Pooled efficacy data from meta-analyses of clinical and real-world studies indicate that MVA-BN vaccines provide 70-90% protection against orthopoxvirus infections, with two doses conferring higher effectiveness than one. For mpox specifically, real-world evidence from 2022-2024 outbreaks in the United States and Europe reported vaccine effectiveness (VE) of 78% against symptomatic infection after a single dose and up to 89% after two doses, based on case-control studies involving over 10,000 participants. These findings underscore the vaccines' role in outbreak control, particularly among high-risk groups such as men who have sex with men. Post-licensure surveillance through systems like the (VAERS) and v-safe has confirmed the favorable safety profile of JYNNEOS, with no unexpected serious adverse events identified despite administration of over 5 million doses globally by mid-2025, predominantly in adults during outbreaks. Mild to moderate reactogenicity, such as injection-site pain and fatigue, remains the most common , occurring in 50-70% of recipients after the first dose.

Ongoing and Future Developments

Research into Modified vaccinia Ankara (MVA) continues to expand its applications beyond established vaccines, with active clinical pipelines targeting emerging infectious diseases and . For , MVA-based candidates such as COH04S1, which encodes the , are advancing in Phase 2 trials to evaluate their potential as boosters, demonstrating robust cross-reactive immunity against variants in preclinical models. In , MVA-BN-PRO, a recombinant MVA expressing , was investigated in a Phase 1 trial for men with biochemically recurrent , aiming to elicit tumor-specific T-cell responses to delay disease progression. Additionally, ongoing Phase 3 and Phase 2 trials are assessing MVA-BN for prevention in special populations, including pregnant women and children aged 2-11 years, to address gaps in current immunization strategies. Key challenges in MVA vaccine development include optimizing multi-epitope constructs to broaden immune coverage without inducing tolerance, particularly for rapidly evolving pathogens. For , the emergence of Ib variants poses risks of immune escape from existing MVA-BN formulations, necessitating updated antigens to maintain efficacy against diverse strains. Furthermore, waning responses post-MVA-BN , observed within 6-12 months, highlight the need for strategies to enhance long-term immunity duration. Innovations in MVA platforms are addressing production and efficacy hurdles. Synthetic MVA constructs, developed in 2024 studies, enable faster and higher yields, facilitating rapid adaptation for outbreak responses while preserving . Combining MVA vectors with mRNA vaccines in regimens has shown synergistic effects, boosting T-cell responses and protection against and in animal models. Looking ahead, MVA's role in pandemic preparedness is strengthening, bolstered by the World Health Organization's prequalification of MVA-BN in 2024, which facilitates equitable global distribution and procurement for up to 13 million doses by the end of 2025. Efforts are underway to integrate MVA platforms into broader vaccine alliances, targeting enhanced access in low-resource settings by 2026.

References

  1. [1]
    Modified Vaccinia Virus Ankara: History, Value in Basic Research ...
    Safety tested Modified Vaccinia virus Ankara (MVA) is licensed as third-generation vaccine against smallpox and serves as a potent vector system for development ...
  2. [2]
    MVA-BN (‎Modified Vaccinia Ankara – Bavarian Nordic)‎ smallpox ...
    Nov 27, 2024 · The MVA-BN (live Modified Vaccinia Ankara) vaccine is a third-generation smallpox vaccine, which contains a virus that cannot replicate in humans.
  3. [3]
    Second-Generation Smallpox Vaccine: Modified Vaccinia Ankara ...
    Nov 19, 2024 · The MVA vaccine was developed by Bavarian Nordic. NIAID supported early advanced development work for this important vaccine.
  4. [4]
    Abstammung, Eigenschaften und Verwendung des attenuierten ...
    Das MVA-Virus repräsentiert ein Labor-Virus, das sich durch zahlreiche biologische Marker von den bekannten Vaccinia-Stämmen wie auch von den anderen Viren ...Missing: al. | Show results with:al.
  5. [5]
    Smallpox Eradication Programme - SEP (1966-1980)
    May 1, 2010 · Before 1967, the smallpox eradication strategy relied on mass vaccination. However, this strategy was ineffective in densely populated regions ...
  6. [6]
  7. [7]
    Introduction of the Six Major Genomic Deletions of Modified Vaccinia ...
    The six major genomic deletions of modified Vaccinia virus Ankara (MVA) into the parental Vaccinia virus is not sufficient to reproduce an MVA-like phenotype.
  8. [8]
    Highly attenuated modified vaccinia virus Ankara (MVA) as an ...
    In summary, MVA has now been compared, to replication competent vaccinia strains, in three animal models, including influenza virus and SIV. In each ...<|control11|><|separator|>
  9. [9]
    Smallpox and Bioterrorism - PMC - PubMed Central - NIH
    This chapter will focus on information regarding smallpox and smallpox vaccination since 2001, notably, the persisting threat of smallpox as a bioterrorist ...
  10. [10]
    The Complete Genomic Sequence of the Modified Vaccinia Ankara ...
    The complete genomic DNA sequence of the highly attenuated vaccinia strain modified vaccinia Ankara (MVA) was determined. The genome of MVA is 178 kb in ...
  11. [11]
    Expanding the Repertoire of Modified Vaccinia Ankara-Based ...
    During this process of attenuation, MVA underwent deletion of 31 ... Roman numerals (I-VI) represent the sites of the major genomic deletions in MVA as compared ...
  12. [12]
    Repair of a previously uncharacterized second host-range gene ...
    Feb 4, 2020 · Repair of a previously uncharacterized second host-range gene contributes to full replication of modified vaccinia virus Ankara (MVA) in human ...Missing: modifications | Show results with:modifications
  13. [13]
    Deletion of the K1L Gene Results in a Vaccinia Virus That Is Less ...
    The vaccinia virus K1 ankyrin repeat protein inhibits NF-kB activation by preventing RelA acetylation. J Gen Virol97:2691–2702. View · PubMed · Web of Science.
  14. [14]
    Zinc-finger antiviral protein (ZAP) is a restriction factor for replication ...
    Aug 31, 2020 · Zinc-finger antiviral protein (ZAP) is a restriction factor for replication of modified vaccinia virus Ankara (MVA) in human cells · Chen Peng.
  15. [15]
    Quantitative proteomics defines mechanisms of antiviral defence ...
    Dec 8, 2023 · Through serial passage in chicken embryo fibroblasts, the MVA genome has accumulated six major deletions and many small insertion and ...
  16. [16]
    Differences and Similarities in Viral Life Cycle Progression and Host ...
    As a result of the deletion of genes involved in host-range restriction, replication of MVA in most nonavian cell types aborts at a late stage of the virus life ...
  17. [17]
  18. [18]
    Safety and Immunogenicity of Modified Vaccinia Ankara-Bavarian ...
    May 5, 2015 · Conclusions. Modified vaccinia Ankara was safe and immunogenic in subjects infected with HIV and represents a promising smallpox vaccine ...Missing: papers | Show results with:papers
  19. [19]
    Immunogenicity and Safety of Modified Vaccinia Ankara (MVA ...
    Aug 24, 2023 · We aimed to evaluate the safety and immunogenicity of the Modified Vaccinia Ankara (MVA) vaccine. We conducted a systematic review and meta- ...Missing: papers | Show results with:papers
  20. [20]
    Modified vaccinia virus Ankara as antigen delivery system
    After more than 570 passages in tissue culture MVA had lost the broad cellular host range of VV, being unable to productively grow in many cells of mammalian ...
  21. [21]
    Vaccines and Antiviral Therapies for Mpox Virus in Pregnant and ...
    Mar 22, 2025 · MVA-BN is recommended in pregnancy if the mother is at risk of infection. As it is a non-replicating vaccine, it is safer to use during ...
  22. [22]
    Prioritizing Vulnerable Populations in Clinical Research - PMC
    Oct 10, 2025 · Overall, the interim analysis found that the MVA-BN vaccine was safe and well tolerated in adolescents, with similar adverse events except for ...
  23. [23]
    Immune Response to MVA-BN Vaccination for Mpox - PubMed Central
    MVA-BN induces both humoral and cellular responses, including the production of binding and neutralising antibodies and pathogen-specific memory B cells and T ...
  24. [24]
    JYNNEOS | FDA
    May 14, 2025 · Indicated for prevention of smallpox and monkeypox disease in adults 18 years of age and older determined to be at high risk for smallpox or ...
  25. [25]
    JYNNEOS (MVA-BN) Mpox Smallpox Vaccine - Vax-Before-Travel
    Approved by the U.S. Food and Drug Administration (FDA) in 2019, JYNNEOS was the first smallpox vaccine successfully developed under the Project BioShield ...<|separator|>
  26. [26]
    News | Bavarian Nordic
    Oct 31, 2025 · The framework agreement also allowed the European Commission and European Member States to stockpile MVA-BN as a critical medical countermeasure ...
  27. [27]
    Phase 3 Efficacy Trial of Modified Vaccinia Ankara as a Vaccine ...
    Nov 13, 2019 · Immune responses and attenuation of the major cutaneous reaction suggest that this MVA vaccine protected against variola infection.Missing: virulence | Show results with:virulence
  28. [28]
    US Government Orders Another 2.5 Million Doses of Monkeypox ...
    Jul 15, 2022 · Bavarian Nordic to supply an additional 2.5 million doses of JYNNEOS® vaccine, bringing deliveries in 2022 and 2023 to a total of nearly 7 ...Missing: size | Show results with:size
  29. [29]
    [PDF] Package Insert - JYNNEOS (Refrigerator) - FDA
    Each 0.5 mL dose of JYNNEOS is formulated to contain 0.5 x 108 to 3.95 x 108 infectious units of. MVA-BN live virus. Each 0.5 mL dose contains 0.61 mg of Tris ( ...Missing: regimen | Show results with:regimen
  30. [30]
    [PDF] EMERGENCY USE AUTHORIZATION OF JYNNEOS (SMALLPOX ...
    Aug 9, 2022 · The FDA has granted an EUA for the emergency use of JYNNEOS for active immunization by subcutaneous injection for prevention of mpox disease in ...
  31. [31]
    EMA recommends approval of Imvanex for the prevention of ...
    Jul 22, 2022 · The medicine has been approved in the EU since 2013 for the prevention of smallpox. It contains an attenuated (weakened) form of the vaccinia ...
  32. [32]
    Global perspectives on smallpox vaccine against monkeypox
    However, recent research on the 2022 US mpox outbreak determined that the JYNNEOS vaccine had an estimated efficacy of 35%–75% after one dose and 66%–85 ...
  33. [33]
    Mpox vaccines – frequently asked questions (FAQs) | NCIRS
    JYNNEOS vaccine has been proven to be a safe vaccine, and more than a million doses have been given worldwide since the 2022 mpox outbreak. To date, most ...
  34. [34]
    Combining mpox vaccination and behavioural changes to control ...
    Jul 31, 2025 · The introduction of five mpox cases on 1 May 2025 resulted in 82% more cases when the newly introduced cases were infected with a subclade with ...
  35. [35]
    Interim Clinical Considerations for Use of Vaccine for Monkeypox ...
    Sep 5, 2025 · JYNNEOS® vaccine is licensed to prevent smallpox and monkeypox and ... On August 9, 2022, FDA issued an EUA for JYNNEOS monkeypox vaccine.
  36. [36]
  37. [37]
    Construction and Isolation of Recombinant Vaccinia Virus by ...
    The efficiency of homologous recombination event for vaccinia virus is relatively low, and recombinant viruses only account for 0.1% of progeny viruses.
  38. [38]
    Intergenic region 3 of modified vaccinia ankara is a functional site for ...
    Aug 1, 2010 · In this report, we evaluate an alternative insertion site known as intergenic region 3 (IGR3). Recombinant MVA carrying the cytomegalovirus pp65 ...
  39. [39]
    Modified Vaccinia Virus Ankara-Infected Dendritic Cells Present ...
    Here we characterized the MHCII antigen presentation pathways that are possibly involved in the immune response upon vaccination with modified vaccinia virus ...
  40. [40]
    Safety and immunogenicity of 2-dose heterologous Ad26.ZEBOV ...
    Jan 11, 2022 · ZEBOV, MVA-BN-Filo Ebola vaccination was safe and immunogenic in healthy African children. What did the researchers do and find? In this ...
  41. [41]
    Modified Vaccinia Virus Ankara (MVA) as Production Platform for ...
    Modified Vaccinia Virus Ankara (MVA) is a replication-deficient viral vector that holds great promise as a vaccine platform.Missing: avoiding | Show results with:avoiding
  42. [42]
    An MVA Vector Expressing HIV-1 Envelope under the Control of a ...
    We have generated and characterized the biology and immunogenicity of an optimized MVA-based vaccine candidate against HIV/AIDS expressing HIV-1 clade B gp120 ...
  43. [43]
    Failure Of Latest HIV Vaccine Test: A 'Huge Disappointment' - NPR
    Apr 26, 2013 · Leading AIDS researchers were disappointed by the failure of HVTN-505 – but not necessarily surprised, given the history of previous attempts.<|control11|><|separator|>
  44. [44]
    Immunogenicity and efficacy of the COVID-19 candidate vector ...
    Jun 23, 2021 · MVA-SARS-2-S expresses and safely delivers the full-length SARS-CoV-2 S protein, inducing balanced SARS-CoV-2–specific cellular and humoral immunity, and ...
  45. [45]
    Enhanced immunogenicity and protective effect conferred by ...
    In the present study, a variety of immune responses elicited by MVA and the licensed smallpox vaccine Dryvax in a murine model were compared.
  46. [46]
    Evaluation of the efficacy of modified vaccinia Ankara (MVA ... - NIH
    In this study, we tested the efficacy of IMVAMUNE® [modified vaccinia Ankara-Bavarian Nordic (MVA-BN®)] for protecting rabbits against aerosolized RPXV.
  47. [47]
  48. [48]
    Nonhuman primate to human immunobridging to infer the protective ...
    Dec 17, 2020 · The 100% survival observed in these studies for the Ad26.ZEBOV, MVA-BN-Filo regimen with an 8-week interval between doses precluded conducting ...
  49. [49]
    Study Details | NCT00046397 | Phase I Trial of Smallpox Vaccine
    This study will test the safety of an experimental vaccine called Modified Vaccinia Virus Ankara (MVA) for use against the smallpox virus.Missing: 1990s 2000s
  50. [50]
    Phase I clinical trial safety of DNA- and modified virus Ankara ...
    Phase I clinical trial safety of DNA- and modified virus Ankara-vectored human immunodeficiency virus type 1 (HIV-1) vaccines administered alone and in a prime- ...Missing: smallpox 1990s 2000s
  51. [51]
    Phase I immunotherapy with a modified vaccinia virus (MVA ...
    Mar 17, 2003 · Results of a phase I trial of a recombinant vaccinia virus that expresses carcinoembryonic antigen in patients with advanced colorectal cancer.<|control11|><|separator|>
  52. [52]
    Efficacy and safety of a modified vaccinia Ankara (MVA) vectored ...
    Aug 16, 2010 · ... vaccines have several disadvantages. They generally require strong adjuvants, elicit poor CD8+ T cell responses and have short shelf lives [60].
  53. [53]
    Mvabea | European Medicines Agency (EMA)
    Mvabea is a vaccine to protect adults and children aged one year and older against Ebola virus disease caused by Zaire ebolavirus. It is used with another Ebola ...Overview · Product information · Product details · Authorisation details
  54. [54]
    WHO adds LC16m8 mpox vaccine to Emergency Use Listing
    Nov 19, 2024 · On 13 September 2024, WHO prequalified the Modified Vaccinia Ankara-Bavarian Nordic (MVA-BN) vaccine and expanded its indication to include use ...
  55. [55]
    Predicting vaccine effectiveness for mpox | Nature Communications
    May 8, 2024 · This study provides a quantitative evidence-based approach for using antibody measurements to predict the effectiveness of mpox vaccination.
  56. [56]
    Effectiveness of a single dose of JYNNEOS vaccine in real world
    Sep 24, 2024 · Some studies have suggested that a single dose of JYNNEOS provides robust protection against mpox infection during this outbreak. , Sagy et al.
  57. [57]
    Evidence to Recommendations (EtR) Framework for Use of ... - CDC
    Jun 16, 2025 · During May 22, 2022 to January 13, 2023, a total of 1,125,168 JYNNEOS vaccine doses were administered. CDC monitored JYNNEOS safety using VAERS, ...
  58. [58]
    Safety Monitoring of JYNNEOS Vaccine During the 2022 Mpox ... - NIH
    Dec 9, 2022 · During May 22–October 21, 2022, nearly 1 million JYNNEOS doses were administered in the United States. The vaccine safety profile was consistent ...
  59. [59]
    Synthetic modified vaccinia Ankara vaccines confer cross-reactive ...
    Feb 16, 2024 · This is the first challenge study in a relevant animal model showing protection by an MVA-based vaccine against the newly emerged MPXV clade IIb ...
  60. [60]
    Phase I dose escalation trial of MVA-BN-PRO in men with ... - ASCO
    Background: MVA-BN-PRO is an investigational prostate cancer immunotherapy comprising of a highly attenuated non-replicating vaccinia virus engineered to ...
  61. [61]
    NCT06844500 | Phase 3 Maternal Safety & Immunogenicity Trial of ...
    This Phase 3 open-label study aims to assess the safety and immune response of the MVA-BN mpox vaccine when administered subcutaneously to pregnant and ...
  62. [62]
    Bavarian Nordic Announces the Initiation of Clinical Trials of Mpox ...
    Jun 26, 2025 · Topline results from this trial (NCT06549530) are anticipated in the third quarter of 2025. ... MVA-BN or Modified Vaccinia Ankara-Bavarian Nordic ...
  63. [63]
    Clade Ib Mpox Virus: How Can We Respond? - PMC
    Jul 18, 2025 · This review aims to provide an overview of the current understanding of mpox virus Clade Ib, including its genetic characteristics, ...
  64. [64]
    Rapid Decline of Mpox Antibody Responses Following MVA-BN ...
    Jul 25, 2025 · In this study, we show that the MVA-BN vaccine generated mpox serum antibody responses that largely waned after 6-12 months. Last updated on 07/ ...
  65. [65]
    Heterologous mRNA/MVA delivering trimeric-RBD as effective ...
    This study explores a heterologous mRNA/Modified Vaccinia virus Ankara (MVA) prime/boost regimen employing a trimeric form of the receptor binding domain (RBD) ...
  66. [66]
    Comparison of protection against mpox following mRNA or modified ...
    Sep 4, 2024 · MVA-immunized animals exhibited reduced viral loads in blood viremia and infectious virus in the throat compared with PBS-treated animals; ...
  67. [67]
    Search - Bavarian Nordic
    Bavarian Nordic Receives WHO Prequalification for Mpox Vaccine First mpox vaccine to obtain prequalification by the WHO Approval will help accelerate access ...
  68. [68]
    Gavi Signs Agreement with Bavarian Nordic to Rapidly Secure ...
    Sep 18, 2024 · The doses will be for delivery in 2024. The vaccines will be funded by Gavi's First Response Fund, a new financial mechanism created in June ...
  69. [69]
    WHO prequalifies first vaccine against mpox - BioSpectrum Asia
    Sep 16, 2024 · The World Health Organization (WHO) has announced MVA-BN vaccine as the first vaccine against mpox to be added to its prequalification list.