mRNA-1608 is an investigational therapeutic messenger RNA (mRNA) vaccine candidate currently under development by Moderna, Inc..[1] This vaccine is primarily engineered to target Herpes Simplex Virus type 2 (HSV-2), the principal etiological agent responsible for genital herpes, while also holding the potential for cross-protective immunity against Herpes Simplex Virus type 1 (HSV-1).[1] The fundamental therapeutic objective of mRNA-1608 is to alleviate the clinical burden associated with recurrent HSV-2 infections. This is anticipated to be achieved through the induction of a robust and multifaceted immune response, encompassing the generation of neutralizing and effector antibodies, complemented by the stimulation of potent cell-mediated immunity.[1]
Currently, mRNA-1608 is being evaluated in a fully enrolled Phase 1/2 clinical trial (NCT06033261, also identified as mRNA-1608-P101). This study is specifically designed to assess the vaccine's safety, immunogenicity, and preliminary clinical benefit in adult participants with a documented history of recurrent genital herpes.[1] The development of an effective therapeutic vaccine for HSV represents a significant unmet medical need, given the high global prevalence of genital herpes, the chronic nature of the infection characterized by recurrent painful lesions, and the limitations of existing antiviral therapies which only manage symptoms rather than prevent recurrences or eradicate the virus.[2]
The strategic focus on a therapeutic indication—targeting individuals already suffering from recurrent disease—may offer a more direct route to demonstrating clinical improvements in lesion frequency and viral shedding, key factors impacting patient quality of life.[1] The mRNA technology underpinning mRNA-1608 offers potential advantages in terms of rapid development and the capacity to elicit broad immune responses, including the critical cell-mediated arm necessary for controlling latent viral infections.[1] Initial data from the ongoing Phase 1/2 trial are anticipated around mid-2025, which will be crucial in determining the future trajectory of this candidate.[1] A key aspect that remains proprietary is the precise antigenic composition of mRNA-1608; this information is vital for a complete understanding of its immunogenic potential and mechanism of action. The success of mRNA-1608 would not only provide a much-needed therapeutic option for millions affected by HSV but also further validate the versatility of the mRNA platform for complex viral diseases.
mRNA-1608 is an investigational vaccine candidate developed by Moderna, Inc., a U.S.-based biotechnology company that has been a significant pioneer in the field of messenger RNA (mRNA) therapeutics and vaccines.[1] The company is leveraging its mRNA platform, notably utilized for its COVID-19 vaccine (Spikevax®), across a diverse pipeline that includes several vaccine candidates against latent viruses, such as HSV.[2]
The vaccine is based on mRNA technology, which involves the administration of synthetic mRNA molecules. These molecules carry the genetic instructions for host cells to produce specific viral antigens. Upon cellular uptake and translation of the mRNA, the host cells transiently express these antigens, thereby stimulating a targeted immune response without exposing the recipient to the live or inactivated virus.[1] This approach offers potential advantages in terms of speed of development, capacity to elicit broad and potent immune responses, and manufacturing scalability.[1]
The primary pathogen targeted by mRNA-1608 is Herpes Simplex Virus type 2 (HSV-2), which is the serotype predominantly responsible for causing genital herpes.[1] Beyond its primary focus on HSV-2, Moderna anticipates that mRNA-1608 may elicit immune responses capable of providing cross-protection against Herpes Simplex Virus type 1 (HSV-1).[1] This potential for cross-protection is biologically plausible due to the significant antigenic homology shared between the two serotypes, particularly within key surface glycoproteins.[10] Such cross-protection would be clinically valuable, as HSV-1 is increasingly recognized as a cause of anogenital herpes infections, particularly in younger individuals and in certain geographical regions.[12] A single vaccine addressing both major causative agents of genital herpes would therefore offer a more comprehensive public health solution.
HSV-1 and HSV-2 are neurotropic DNA viruses characterized by their ability to establish lifelong latent infections within sensory neurons following an initial (primary) infection at mucosal or cutaneous sites.[1] During latency, the viral genome persists in a quiescent state within the neuronal nucleus. Periodically, the virus can reactivate from this latent state, leading to the production of new viral particles that travel anterogradely along axons to the initial site of infection, resulting in recurrent viral shedding and, often, clinical lesions (e.g., blisters, ulcers).[1] A hallmark of herpesviruses, including HSV-1 and HSV-2, is their deployment of sophisticated immune evasion mechanisms that allow them to persist in the host despite ongoing immune surveillance.[1] These mechanisms include, for example, the expression of glycoprotein C (gC), which binds to complement component C3b thereby inhibiting complement-mediated lysis, and the glycoprotein E/I (gE/gI) complex, which functions as an IgG Fc receptor, potentially interfering with antibody-dependent effector functions.[14] The ability to establish latency and evade immune clearance presents a formidable challenge for vaccine development.
mRNA-1608 is being developed specifically as a therapeutic vaccine. Its intended use is for adults who are already infected with HSV-2 and suffer from recurrent episodes of genital herpes.[1] The primary therapeutic goals are to reduce the frequency, duration, and/or severity of these recurrent outbreaks, and potentially to decrease the extent and duration of viral shedding, which is responsible for transmission.[1]
Genital herpes represents a significant global public health concern. HSV-2 is highly prevalent, with estimates suggesting that approximately 5% of adults aged 18-49 worldwide are seropositive, translating to millions of individuals affected; in the United States alone, an estimated 18.6 million adults live with HSV-2.[1] The economic burden associated with genital herpes is substantial, encompassing direct healthcare costs and indirect costs due to lost productivity, estimated globally at $35 billion per year.[1]
The clinical manifestations of recurrent genital herpes, characterized by painful blisters and sores, significantly impair the quality of life of affected individuals, often causing physical discomfort, psychological distress, and social stigma.[2] Current standard-of-care antiviral medications, such as acyclovir, valacyclovir, and famciclovir, can effectively manage acute outbreaks (episodic therapy) or reduce the frequency of recurrences when taken continuously (suppressive therapy).[2] However, these treatments do not eradicate the latent virus, do not completely prevent viral shedding (and thus transmission), and only partially restore the quality of life for many patients.[2]
Critically, there is currently no FDA-approved vaccine available to either prevent HSV infection (prophylactic vaccine) or to treat established infection by reducing recurrences (therapeutic vaccine).[1] The development of an effective HSV vaccine has been a longstanding challenge in medical research, with decades of effort yielding numerous setbacks and no licensed products.[1] This history of difficulty underscores the complexity of inducing protective immunity against HSV. Furthermore, HSV-2 infection is a well-established epidemiological cofactor that increases an individual's risk of acquiring and transmitting HIV.[16] An effective HSV therapeutic vaccine could, therefore, have indirect benefits in HIV control efforts.
The lifelong persistence of HSV in neuronal ganglia, coupled with its array of immune evasion tactics, forms the core biological challenge that has historically hindered successful vaccine development.[1] Traditional vaccine approaches, often focusing on inducing neutralizing antibodies via subunit protein antigens, have generally proven insufficient to control latent infection or prevent reactivation effectively.[10] The mRNA vaccine platform, with its demonstrated ability to elicit not only potent and broad antibody responses but also robust cell-mediated immunity, offers a novel immunological strategy. This comprehensive immune stimulation is particularly relevant for therapeutic interventions against established latent viral infections, where cytotoxic T-lymphocytes (CTLs) are crucial for recognizing and eliminating virus-infected cells during reactivation.[1]
Messenger RNA (mRNA) vaccine technology represents a paradigm shift in vaccinology. Instead of administering a whole pathogen (live-attenuated or inactivated) or specific protein subunits, mRNA vaccines deliver synthetic mRNA molecules that encode the genetic information for one or more target antigens of a pathogen.[6] Once these mRNA molecules are delivered into host cells, typically via a lipid nanoparticle (LNP) carrier, the cell's own translational machinery (ribosomes) reads the mRNA sequence and synthesizes the encoded viral protein(s).[6] These endogenously produced antigens are then processed by the cell and presented on its surface in the context of Major Histocompatibility Complex (MHC) class I and class II molecules, or secreted, effectively mimicking aspects of a natural viral infection to the immune system.[6] This process triggers a comprehensive immune response, involving both the humoral arm (B-cell activation leading to antibody production) and the cellular arm (activation of CD4+ helper T-cells and CD8+ cytotoxic T-lymphocytes).[6]
Key advantages of mRNA technology include the speed with which vaccine candidates can be designed and manufactured once the target antigen sequence is known. Production does not require culturing live pathogens, simplifying manufacturing and enhancing safety.[1] Furthermore, mRNA vaccines have demonstrated high potency and the capacity to elicit broad immune responses. Unlike DNA vaccines, mRNA does not need to enter the cell nucleus to be functional, and it does not integrate into the host genome, thereby mitigating concerns about insertional mutagenesis.[6]
Moderna has explicitly stated that the intended immunological outcome for mRNA-1608 is the induction of a "strong antibody response that combines neutralizing and effector functionality with cell-mediated immunity".[1] This multi-pronged immune strategy is deemed essential for effectively combating a persistent viral infection like HSV-2.
Naked mRNA is highly susceptible to degradation by extracellular ribonucleases (RNases) and, due to its size and negative charge, cannot efficiently cross cell membranes to reach the cytoplasm where translation occurs. Consequently, mRNA vaccines like mRNA-1608 require a protective delivery system. Lipid Nanoparticles (LNPs) are the most clinically advanced delivery vehicles for mRNA.[1]
LNPs are typically spherical structures, tens to hundreds of nanometers in diameter, composed of several lipid components that self-assemble around the mRNA payload:
The exact LNP composition for mRNA-1608 is proprietary to Moderna. However, the company has amassed considerable expertise in LNP formulation through the development and global deployment of its COVID-19 vaccine, Spikevax®, and other pipeline candidates.[24] This platform knowledge is undoubtedly applied to optimize the LNP delivery system for mRNA-1608 to ensure efficient mRNA delivery, potent antigen expression, and an acceptable safety profile. The LNP itself is not merely a passive carrier; its composition can influence the immunogenicity of the vaccine, potentially possessing adjuvant-like properties by activating innate immune pathways. The careful balance between LNP components is critical for achieving desired efficacy while managing reactogenicity, which is often associated with both the mRNA and the LNP.[22]
Based on general knowledge of intramuscularly (IM) administered mRNA-LNP vaccines, such as mRNA-1608 [1], the vaccine is expected to remain largely localized at the injection site and drain to regional lymph nodes.[1] Muscle cells (myocytes) and resident antigen-presenting cells (APCs) like dendritic cells and macrophages at the injection site can take up the LNPs. APCs that have captured the LNPs or processed antigen can then migrate to draining lymph nodes, which are primary sites for the initiation of adaptive immune responses.[1]
A key characteristic of mRNA is its transient nature within the cell. The exogenous mRNA delivered by the vaccine does not replicate and is degraded by normal cellular RNase activity over a period of hours to days. Moderna has stated that for their IM vaccines, mRNA typically becomes undetectable in the body within five days of injection, with minimal levels present after only three days.[1] This limited persistence ensures that antigen production is temporary, which is considered a favorable safety attribute, reducing the potential for long-term off-target effects or induction of immune tolerance to the encoded antigen.
The dual immunological strategy of mRNA-1608, aiming for both robust antibody responses and potent cell-mediated immunity, is particularly well-suited for a therapeutic vaccine against a latent virus like HSV. While antibodies are effective against extracellular virions released during reactivation episodes, thereby limiting cell-to-cell spread and preventing new infections, T-lymphocytes are indispensable for recognizing and eliminating already infected cells. CD8+ cytotoxic T-lymphocytes (CTLs) are especially crucial for controlling viral replication within infected tissues and potentially for influencing the state of viral latency within neurons or limiting the efficiency of reactivation events.[13] The ability of mRNA vaccines to efficiently prime CTL responses is a significant advantage over many traditional vaccine platforms, particularly subunit protein vaccines which often require strong adjuvants to achieve comparable T-cell activation.
A critical aspect for understanding the precise immunological approach of mRNA-1608 is its antigenic composition. However, Moderna has not publicly disclosed the specific Herpes Simplex Virus type 2 (HSV-2) antigens that are encoded by the mRNA sequences within the mRNA-1608 vaccine candidate.[1] This information is considered proprietary and is fundamental to the vaccine's design and expected efficacy.
While direct information is lacking, some contextual clues can be drawn. A presentation slide from the World Health Organization (WHO) in April 2023, discussing mRNA vaccines against HSV, mentioned "Moderna's herpes simplex virus (HSV) vaccine candidate (mRNA-1608)".[14] In a related section on the same slide, it was noted that "BNT163 candidate vaccine encodes three HSV-2 glycoproteins, gC, gD and gE".[14] This juxtaposition places mRNA-1608 within the broader landscape of mRNA vaccines targeting HSV glycoproteins but does not constitute a direct statement about mRNA-1608's specific antigenic makeup.
Given that mRNA-1608 is a therapeutic vaccine intended to elicit both strong antibody and cell-mediated immunity to control recurrent HSV-2, its antigen selection strategy would likely be guided by principles different from, or additive to, purely prophylactic approaches.
Glycoproteins (e.g., gB, gC, gD, gE):
HSV glycoproteins are major structural components of the viral envelope and are essential for viral attachment, fusion, and entry into host cells. They are also primary targets for neutralizing antibodies.11
Tegument Proteins and Other Non-Structural Proteins (Key for T-cell Immunity):
For a therapeutic vaccine aimed at controlling an established latent infection and reducing recurrences, the induction of potent and durable T-cell mediated immunity is considered paramount.13 T-cells, particularly CD8+ CTLs, are essential for recognizing and eliminating virus-infected cells during reactivation.
The selection of antigens for mRNA-1608 would likely prioritize those capable of eliciting robust and broad T-cell responses, particularly CD8+ CTLs, which are crucial for clearing infected cells during reactivation, and CD4+ T-cells, which provide help for both CTL and B-cell responses. Given Moderna's stated aim to induce strong cell-mediated immunity alongside antibody responses for mRNA-1608 [1], it is plausible that the vaccine encodes a combination of antigens, potentially including both surface glycoproteins (for antibody targets) and internal or regulatory proteins (for T-cell targets). The success of the University of Pennsylvania's trivalent glycoprotein mRNA vaccine in preclinical prophylactic models [11] might inform Moderna's thinking, but a therapeutic vaccine may necessitate a different or expanded set of antigens to effectively target latently infected cells or control reactivation.
The strategy of targeting HSV-2 with an expectation of HSV-1 cross-protection [1] is immunologically sound due to the substantial amino acid sequence homology between the two viruses, especially within their glycoprotein envelopes.[10] Preclinical studies with the trivalent HSV-2 (gC2, gD2, gE2) mRNA vaccine indeed demonstrated excellent protection against HSV-1 challenge in mice.[11] If mRNA-1608 achieves similar cross-serotype efficacy in humans, it would represent a significant advantage, simplifying vaccination strategies for genital herpes, which can be caused by either serotype.
The lead clinical trial for mRNA-1608 is a Phase 1/2 study identified by the ClinicalTrials.gov identifier NCT06033261 and Moderna's internal study ID mRNA-1608-P101.[1] Key details of this trial are summarized in Table 1.
Table 1: Key Details of Clinical Trial NCT06033261 (mRNA-1608-P101)
Parameter | Details | Citations |
---|---|---|
Trial Identifier | NCT06033261; mRNA-1608-P101 | 1 |
Phase | Phase 1/2 | 1 |
Official Title | A Phase 1/2, Randomized, Observer-Blind, Controlled, Dose-Ranging Study of mRNA-1608, an HSV-2 Therapeutic Candidate Vaccine, in Healthy Adults 18 to 55 Years of Age With Recurrent HSV-2 Genital Herpes | 1 |
Study Objectives | Primary: To evaluate the safety and immunogenicity of mRNA-1608. Secondary: To establish proof-of-concept of clinical benefit. | 1 |
Study Design | Randomized, observer-blind (some sources suggest triple-blind 28), active-controlled, dose-ranging. | 1 |
Participant Population | Healthy adults aged 18 to 55 years with a documented history of recurrent HSV-2 genital herpes (defined as ≥3 to ≤9 recurrences in the 12 months prior to screening, or before initiation of suppressive therapy). | 1 |
Key Inclusion/Exclusion | Willing to stop suppressive antiviral therapy and refrain from episodic therapy during specific swabbing periods. Excludes prior HSV vaccination, ocular HSV, HSV-1 genital infection, HIV, Hepatitis B/C, anaphylaxis to mRNA vaccines or BEXSERO components. | 15 |
Intervention Arms | Participants randomized 1:1:1:1 to receive: mRNA-1608 Dose Level A; mRNA-1608 Dose Level B; mRNA-1608 Dose Level C; or Control (BEXSERO®, Meningococcal Group B Vaccine). | 1 |
Dosing Schedule | Two intramuscular (IM) injections administered at Day 1 (0 months) and Day 57 (2 months) for all arms. | 1 |
Enrollment Status | Fully enrolled. Approximately 300 participants per Moderna.1 Some trial aggregators list 365.28 Conducted in the USA. | 3 |
Key Timelines | Actual Study Start: September 6, 2023. Enrollment Completion: Confirmed by Moderna March 27, 2024; fully enrolled announced May 2, 2024. Estimated Primary Completion Date: April 11, 2025 (some sources cite June 2025). | 1 (April 2025); 6 (June 2025) |
Primary/Secondary Outcome Measures (General) | Safety: Incidence of adverse events. Immunogenicity: Antibody responses (e.g., neutralizing titers) and cell-mediated immune responses. Clinical Benefit: Reduction in frequency/severity of genital herpes recurrences, reduction in viral shedding (assessed via anogenital swabs). | Inferred from 1 |
The choice of BEXSERO®, a multicomponent meningococcal B vaccine, as an active control in NCT06033261 is noteworthy.[1] mRNA-LNP vaccines are known for their potential to cause local and systemic reactogenicity (e.g., injection site pain, fever, fatigue).[27] Using a saline placebo might easily unblind participants and investigators due to the noticeable difference in post-injection reactions. BEXSERO® itself is an immunogenic vaccine associated with a certain level of reactogenicity. Its use as a control likely aims to maintain the study blind by ensuring that both active and control groups experience some degree of post-vaccination symptoms. This allows for a more objective assessment of mRNA-1608's specific safety profile, distinguishing adverse events attributable to the HSV antigens or the specific mRNA-1608 formulation from common, non-specific vaccine reactions.
The dose-ranging component of this Phase 1/2 trial, evaluating three distinct dose levels of mRNA-1608, is a critical step in early clinical development.[1] The amount of mRNA administered can directly influence the level of antigen expression and, consequently, the magnitude and quality of the ensuing immune response. However, higher doses may also be associated with increased reactogenicity.[27] This dose-escalation/evaluation design allows Moderna to identify an optimal biological dose that achieves the desired immunogenicity and potential clinical benefit while maintaining an acceptable safety and tolerability profile. This "therapeutic window" is essential for guiding dose selection for subsequent, larger Phase 3 efficacy trials.
Furthermore, the study protocol requires participants to collect daily anogenital swabs for three separate 28-day periods.[15] These swabs are typically analyzed using sensitive molecular methods like PCR to detect and quantify HSV DNA, providing an objective measure of viral shedding. Viral shedding, which can occur both during symptomatic outbreaks and asymptomatically, is a key factor in HSV transmission and a marker of viral activity. A reduction in the frequency, duration, or quantity of viral shedding would be a strong, objective indicator of therapeutic efficacy, demonstrating improved immune control over the virus. This endpoint complements patient-reported outcomes like lesion recurrence rates, which can be more subjective.
mRNA-1608 is a significant component of Moderna's broader strategy to develop vaccines against latent viruses. This portfolio includes candidates for cytomegalovirus (CMV; mRNA-1647, currently in Phase 3), Epstein-Barr virus (EBV; mRNA-1189 and mRNA-1195, in Phase 2 and Phase 1/2 respectively), varicella-zoster virus (VZV; mRNA-1468, in Phase 1/2), and HIV (mRNA-1644 and mRNA-1574, in Phase 1).[1] This cohesive approach leverages the adaptability and potential of the mRNA platform to address the unique challenges posed by viruses that establish lifelong latency and cause chronic or recurrent disease.
The safety and tolerability of mRNA-1608 are primary endpoints of the ongoing NCT06033261 trial.[1] While specific data from this trial are not yet publicly available, expectations regarding its safety profile can be informed by the general characteristics of mRNA-LNP vaccine technology and the trial design.
Expected Safety Considerations based on mRNA Vaccine Technology:
mRNA-LNP vaccines, including Moderna's Spikevax®, are commonly associated with transient reactogenicity. These are typically mild to moderate and resolve within a few days.27
Safety Assessment in NCT06033261:
The Phase 1/2 trial is designed to meticulously evaluate the safety and tolerability of mRNA-1608 across three different dose levels compared to the BEXSERO® control.1 This involves:
The safety profile of a therapeutic vaccine for recurrent genital herpes will be subject to considerable scrutiny. Since genital herpes, while burdensome, is not typically a life-threatening condition for immunocompetent adults, the tolerance for vaccine-related adverse events is generally lower than for vaccines against acute, severe, or fatal diseases. Therefore, mRNA-1608 must demonstrate not only efficacy but also a highly favorable benefit-risk profile to gain acceptance by regulatory authorities, clinicians, and patients. The forthcoming safety data from NCT06033261 will be pivotal in this regard.
The development of an effective therapeutic vaccine for HSV-2, such as mRNA-1608, holds the promise of substantially improving the lives of millions of individuals worldwide who suffer from recurrent genital herpes.[2] A successful vaccine could lead to a significant reduction in the frequency, duration, and severity of painful genital lesions, thereby enhancing quality of life and reducing psychological distress. If the vaccine also markedly decreases viral shedding, it could play a role in reducing HSV transmission. Furthermore, an effective therapeutic vaccine might offer a more convenient and potentially more compliant alternative or adjunct to long-term suppressive antiviral drug therapy.[2]
However, the path to a licensed HSV vaccine is fraught with challenges, as evidenced by a long history of clinical trial failures over several decades.[1] GSK, for example, recently discontinued its therapeutic HSV vaccine candidate (GSK3943104) after it failed to meet its primary efficacy objective in a Phase II study.[5] The complex biology of HSV, including its ability to establish lifelong latency in neurons and its sophisticated immune evasion mechanisms, makes it a difficult target for vaccine-induced immune control.[1] Establishing robust and reliable correlates of protection for a therapeutic effect against a latent, periodically reactivating virus also remains a complex scientific endeavor.
The next critical milestone for mRNA-1608 will be the data readout from the ongoing Phase 1/2 trial (NCT06033261). With an estimated primary completion date around April-June 2025 [1], initial top-line results on safety and immunogenicity, and potentially preliminary signals of clinical benefit, could become available in mid to late 2025. These results will be eagerly anticipated by the scientific community, patients, and investors, as they will provide the first human data on this specific candidate.
If the Phase 1/2 results are positive, demonstrating an acceptable safety profile and encouraging immunogenicity and/or clinical activity, Moderna would likely proceed to larger, more definitive Phase 3 trials. These trials would need to confirm efficacy in a broader patient population and further characterize the long-term safety and durability of the immune response. It is noteworthy that, according to GlobalData, Phase II drugs for Genital Herpes have historically had a 30% phase transition success rate to Phase III, underscoring the high attrition rate in this therapeutic area.[1] While this benchmark reflects past difficulties, the novel mechanism of action of mRNA vaccines—particularly their potential to induce strong and broad T-cell responses crucial for controlling latent infections—may offer an improved probability of success compared to previous vaccine modalities that primarily focused on antibody generation.
The success or failure of mRNA-1608 will also have broader implications for the mRNA vaccine field. A positive outcome would further validate the versatility of mRNA technology beyond its proven efficacy against acute respiratory viruses like SARS-CoV-2 and RSV, extending its potential to tackle complex latent viral infections. This could encourage further investment and research into mRNA-based therapeutic vaccines for other chronic viral diseases.
mRNA-1608, Moderna's therapeutic vaccine candidate for HSV-2, represents a significant effort to address a long-standing unmet medical need using advanced mRNA technology. By aiming to induce both robust antibody and cell-mediated immune responses, mRNA-1608 seeks to control recurrent genital herpes, a condition that affects millions globally and significantly impacts quality of life. The ongoing Phase 1/2 clinical trial (NCT06033261) is a crucial step in evaluating its safety, immunogenicity, and potential clinical benefit, with initial results anticipated in 2025.
While the specific antigenic composition of mRNA-1608 remains undisclosed, the scientific rationale for using mRNA technology against HSV is compelling, given the limitations of past vaccine approaches and current antiviral therapies. The challenges in developing an effective HSV vaccine are substantial, highlighted by a history of clinical trial failures. However, the unique capabilities of the mRNA platform to elicit comprehensive immune responses offer new hope. The forthcoming data from the NCT06033261 trial will be pivotal in determining whether mRNA-1608 can overcome these historical hurdles and emerge as a viable therapeutic option for individuals suffering from recurrent genital herpes. Success in this endeavor would not only be a major clinical breakthrough but also a significant milestone for the field of mRNA-based therapeutics.
Based on the current understanding of mRNA-1608 and the broader field of HSV vaccine development, several areas warrant consideration for future research and development:
The development of mRNA-1608 is a complex but potentially highly rewarding endeavor. Addressing the multifaceted challenges of HSV latency and immune evasion requires innovative approaches, and mRNA technology offers a promising platform to advance the field.
Published at: May 22, 2025
This report is continuously updated as new research emerges.