New Vaccine for Melanoma: mRNA-4157 (V940) as a Personalized Cancer Vaccine

New Vaccine for Melanoma: mRNA-4157 (V940) as a Personalized Cancer Vaccine

The latest advancements in cancer immunotherapy have shown promising results toward personalized medicine. One such advancement is mRNA-4157 (V940), a novel mRNA-based individualized neoantigen therapy. This personalized cancer vaccine has the potential to revolutionize the treatment of high-risk stage III/IV melanoma when used in combination with pembrolizumab, a well-known immune checkpoint inhibitor.

mRNA-4157 (V940) is designed to encode up to 34 patient-specific tumor neoantigens, which can induce antigen-specific T-cell responses. These responses are crucial as they target the unique mutations present within an individual's tumor cells, making the treatment highly personalized and potentially more effective compared to traditional, one-size-fits-all therapies.

The open-label, randomized, phase 2 mRNA-4157-P201/KEYNOTE-942 trial has met its primary objective of improved recurrence-free survival (RFS) with the combination of mRNA-4157 (V940) and pembrolizumab versus pembrolizumab monotherapy. The study's results indicate a trend for improved RFS in patients who received the combination therapy, highlighting the potential of mRNA-4157 (V940) to enhance the clinical benefits of pembrolizumab.

The underlying mechanism of this synergistic effect may be attributed to the inflamed tumor microenvironment, which is known to be associated with a better clinical response to immune checkpoint inhibitors. Biomarkers such as tumor mutational burden (TMB), gene-expression profile (GEP) score, and programmed death ligand-1 (PD-L1) expression have been associated with responses to pembrolizumab. These biomarkers can promote endogenous responses to tumor neoantigens, with higher TMB providing additional opportunities for tumor-antigen recognition by T cells, and higher GEP score and PD-L1 expression indicative of T-cell infiltration and function.

In the mRNA-4157-P201/KEYNOTE-942 trial, the distribution of TMB, tumor inflammation signature (TIS), and PD-L1 expression in baseline tumors was balanced between the study arms, providing a robust basis for evaluating the prognostic impact of these biomarkers within the combination therapy of mRNA-4157 (V940) and pembrolizumab [1]. Biomarker assessments were conducted on formalin-fixed paraffin-embedded tumor biopsies, which were used for the design of mRNA-4157 (V940) and served as a foundation for exploring the prognostic impact of TMB, TIS, and PD-L1 expression.

The results of the trial suggest that the combination therapy demonstrates a trend for improved RFS compared to pembrolizumab alone in adjuvant melanoma patients across TMB subgroups, indicating that mRNA-4157 (V940) may deepen the clinical benefit in populations already responsive to pembrolizumab monotherapy and/or extend clinical benefit into new patient populations.

In summary, mRNA-4157 (V940) represents a significant step toward the personalization of cancer treatment, with the potential to tailor therapy to the individual's tumor profile and enhance the efficacy of existing treatments like pembrolizumab. The ongoing research and trial results are paving the way for a new era of precision medicine in oncology.

The Mechanism of Action of mRNA-4157 (V940)

mRNA-4157 (V940) is at the forefront of personalized cancer treatment, representing a novel approach in the realm of mRNA-based individualized neoantigen therapy. This cutting-edge therapeutic encodes up to 34 patient-specific tumor neoantigens, which are unique mutations present within an individual's cancer cells. By leveraging the body's own immune system, mRNA-4157 (V940) aims to train T cells to recognize and mount a targeted attack against these specific neoantigens.

The underlying principle of this vaccine lies in its ability to induce potent antigen-specific T-cell responses. Once administered, the mRNA within the vaccine is taken up by the patient's dendritic cells, which are key players in the immune response. These cells then use the mRNA blueprint to produce the encoded neoantigens, presenting them on their surface to T cells. The exposure of T cells to these neoantigens primes them to recognize and eliminate cancer cells harboring the same mutations.

An inflamed tumor microenvironment is generally considered a favorable condition for the effectiveness of immune checkpoint inhibitors. Factors such as tumor mutational burden (TMB), gene-expression profile (GEP) score, and programmed death ligand-1 (PD-L1) expression have been identified as biomarkers that may be associated with clinical responses to treatments like pembrolizumab. These biomarkers are believed to function by promoting endogenous immune responses to tumor neoantigens. A higher TMB may provide additional targets for T-cell recognition, suggesting a synergy with T-cell--based immunotherapies. Similarly, a higher GEP score and PD-L1 expression are indicative of an inflamed tumor microenvironment, characterized by T-cell infiltration and function, which are vital for the success of these therapies.

In essence, mRNA-4157 (V940) is designed to boost the body's immune response to cancer by providing a personalized set of targets for T cells to identify and attack. This strategy not only aims to enhance the efficacy of the immune response but also to minimize off-target effects by focusing on antigens that are unique to the tumor and not found on healthy cells. The ultimate goal is to achieve a more precise and potent anti-tumor effect with fewer side effects compared to conventional, non-specific treatments.

The Role of Pembrolizumab in Combination Therapy

Pembrolizumab, when used in combination with mRNA-4157 (V940), a personalized cancer vaccine, has shown to be a potent force in the fight against high-risk stage III/IV melanoma. Pembrolizumab acts as an immune checkpoint inhibitor, specifically targeting PD-1, a protein on T cells that normally helps keep these cells from attacking other cells in the body. By blocking PD-1, pembrolizumab boosts the immune response against cancer cells. This mechanism of action is particularly beneficial in a tumor microenvironment where the presence of biomarkers such as TMB, GEP score, and PD-L1 expression are associated with better clinical responses to treatment.

In the phase 2 mRNA-4157-P201/KEYNOTE-942 trial, the combination of pembrolizumab with mRNA-4157 (V940) has met its primary objective of improved recurrence-free survival (RFS) compared to pembrolizumab monotherapy. The study results demonstrated a hazard ratio (HR) of 0.561, indicating a trend towards reduced risk of recurrence or death in the combination therapy arm. The trial design involved patients receiving up to one year of pembrolizumab treatment alongside up to nine doses of mRNA-4157 (V940), reflecting a commitment to an intensive and personalized treatment approach.

The trial also explored the prognostic impact of TMB, TIS, and PD-L1 expression within each study arm, and assessed RFS in both biomarker-high and -low subgroups. Notably, the combination therapy showed a consistent RFS benefit across both high and non-high TMB subgroups, suggesting that the addition of mRNA-4157 (V940) could enhance the effects of pembrolizumab regardless of the patient's TMB status. This finding is of particular importance as it suggests that the vaccine could potentiate the efficacy of pembrolizumab even in patient populations that might not typically respond as well to checkpoint inhibitors alone.

Furthermore, the RFS benefit was observed in both TIS-high and TIS-low subgroups, indicating that the combination therapy could be effective across different levels of tumor inflammation. This observation is consistent with the known benefits of pembrolizumab across various indications and lines of therapy, where TIS-high patients typically show better responses. The data suggests that mRNA-4157 (V940) could possibly broaden the reach of pembrolizumab's efficacy to include TIS-low patients as well.

Overall, the combination of pembrolizumab with mRNA-4157 (V940) presents a promising therapeutic strategy, potentially deepening the clinical benefit for patients who are already responsive to pembrolizumab monotherapy and extending the benefits to new patient populations. The KEYNOTE-942 trial highlights the importance of personalized medicine and the role of biomarkers in optimizing cancer treatment, paving the way for future advances in immunotherapy.

Personalization of Cancer Treatment: mRNA-4157 (V940)

The advent of personalized medicine has heralded a new era in cancer treatment, with mRNA-4157 (V940) at the forefront as a personalized cancer vaccine. Developed as part of the pioneering efforts in the field of immunotherapy, mRNA-4157 (V940) leverages the power of mRNA technology to target and provoke an immune response against unique tumor neoantigens specific to each patien. This bespoke approach is indicative of the strides being made towards more individualized and effective cancer care.

The principle behind mRNA-4157 (V940) is the identification of mutations in a patient's tumor cells that are not present in normal cells. These mutations, known as neoantigens, are then used to create a personalized mRNA vaccine that encodes for up to 34 of these patient-specific tumor neoantigens. When administered, the vaccine prompts the immune system to recognize and launch an attack against these neoantigens, thereby targeting the tumor.

The concept of personalization in cancer treatment with mRNA-4157 (V940) is also reflected in the intricate process of vaccine development for each patient. The vaccine is designed based on the unique genetic makeup of an individual's tumor, collected through biopsy and analyzed using next-generation sequencing (NGS) to map out all the potential neoantigens. This highly tailored approach ensures that the therapeutic intervention is as precise and effective as possible, potentially increasing the chances of a successful outcome.

Moreover, the combination of mRNA-4157 (V940) with pembrolizumab, an immune checkpoint inhibitor, underscores the potential to further enhance the vaccine's efficacy. Pembrolizumab works by blocking the programmed death-1 (PD-1) pathway, which cancer cells often exploit to evade the immune system. By combining these two treatments, the immunogenicity of the vaccine can be augmented, potentially leading to improved clinical outcomes for patients with high-risk melanoma.

Personalized cancer vaccines like mRNA-4157 (V940) are a significant step forward in the battle against cancer. They not only represent a shift from a one-size-fits-all approach to treatment but also offer hope for increased efficacy and reduced toxicity by aligning therapy with the individual characteristics of each patient's tumor. As more data emerges, the role of personalized cancer vaccines in the broader landscape of cancer treatment will likely continue to expand, offering new avenues for combating this complex and diverse group of diseases.

Exploring the Clinical Trial Outcomes for mRNA-4157 (V940)

mRNA-4157 (V940) represents a pioneering approach in personalized cancer vaccines, specifically targeting individual tumor neoantigens. The phase 2 clinical trial outcomes for this novel mRNA-based therapy have been a point of interest within the oncology community, particularly in the context of adjuvant treatment for high-risk melanoma patients.

The phase 2 mRNA-4157-P201/KEYNOTE-942 trial explored the efficacy of combining mRNA-4157 (V940) with pembrolizumab, an immune checkpoint inhibitor, compared to pembrolizumab monotherapy. This trial demonstrated a trend towards improved recurrence-free survival (RFS) for the combination therapy in patients who had undergone complete surgical resection of high-risk stage III/IV melanoma.

The trial was meticulously designed to assess the primary endpoint of RFS and included additional secondary endpoints such as safety and tolerability. The patient demographics and trial design were carefully balanced to ensure robust results. With a median follow-up of 23 months for the combination arm and 24 months for the monotherapy arm, the study had sufficient power to detect a significant difference in RFS outcomes.

The safety profile and tolerability of the vaccine were also key considerations in the trial. By evaluating patients' reactions and potential adverse effects, the researchers aimed to understand the practical implications of mRNA-4157 (V940) for broader clinical use.

The trial results hold promise for the utilization of personalized cancer vaccines in conjunction with established therapies to enhance patient outcomes. As the field of oncology moves towards more tailored and patient-specific treatment modalities, the insights gained from the mRNA-4157-P201/KEYNOTE-942 trial represent a significant step forward in the fight against melanoma.

Phase 2 Trial Design and Patient Demographics

The Phase 2 mRNA-4157-P201/KEYNOTE-942 trial was designed as an open-label, randomized study primarily focused on evaluating the efficacy of the mRNA-based vaccine, mRNA-4157 (V940), in combination with pembrolizumab compared to pembrolizumab monotherapy. The trial included patients with resected high-risk stage III/IV melanoma and aimed to improve their recurrence-free survival (RFS).

Participants were stratified based on the disease stage and underwent a 2:1 randomization to receive either the combination treatment of mRNA-4157 (V940) plus pembrolizumab or pembrolizumab alone. The combination treatment arm included up to 9 doses of mRNA-4157 (V940) given intramuscularly every 3 weeks, along with pembrolizumab 200 mg administered intravenously every 3 weeks for up to 18 cycles. The control arm received up to 18 cycles of pembrolizumab monotherapy under the same dosing schedule. The study's primary endpoint was investigator-assessed RFS in the intention-to-treat population.

The demographics of the patient population were balanced between the two study arms, with a median follow-up of 23 months for the combination treatment arm and 24 months for the pembrolizumab monotherapy arm. Eligibility criteria required patients to have undergone complete surgical resection within 13 weeks prior to the first pembrolizumab dose and be disease-free at study entry. Additionally, an Eastern Cooperative Oncology Group performance status (ECOG PS) score between 0 and 1 was mandatory, ensuring participants were in good physical condition, and tissue availability for next-generation sequencing was necessary for personalized vaccine design.

Baseline tumor characteristics were evaluated through biomarker assessments, including tumor mutational burden (TMB), tumor inflammation signature (TIS), and programmed death ligand-1 (PD-L1) expression. These assessments provided insights into the tumor's genetic and immunological landscape, which could influence the response to the vaccine and pembrolizumab treatment.

The trial's design intended to detect a hazard ratio of 0.5 with at least 40 RFS events, providing 80% power with a one-sided alpha of 0.1. The results from the trial would contribute to the understanding of the safety and efficacy of combining a personalized cancer vaccine with an immune checkpoint inhibitor in the treatment of high-risk melanoma.

Efficacy: Recurrence-Free Survival (RFS) and Secondary Endpoints

The open-label, randomized phase 2 trial investigating mRNA-4157 (V940) in combination with pembrolizumab demonstrated a significant improvement in recurrence-free survival (RFS) when compared to pembrolizumab monotherapy in patients with resected high-risk stage III/IV melanoma. The primary objective was met with a hazard ratio (HR) of 0.561, indicating a 43.9% reduction in the risk of recurrence or death with the combination therapy (95% confidence interval [CI]: 0.309, 1.017; 1-sided P value = 0.0266). This outcome represents a promising advancement in the adjuvant treatment of melanoma.

The trial's key secondary endpoints included safety, tolerability, and distant metastasis-free survival (DMFS). The detailed efficacy analysis was performed after all patients completed a minimum of 12 months on study and at least 40 RFS events were observed. The median follow-up period was 23 months for the combination treatment arm and 24 months for the pembrolizumab-only arm, providing a solid basis for evaluating the long-term efficacy and safety profile of the treatments.

mRNA-4157 (V940) is a novel mRNA-based individualized neoantigen therapy designed to trigger antigen-specific T-cell responses to up to 34 patient-specific tumor neoantigens. The inflamed tumor microenvironment, which is characterized by markers such as tumor mutational burden (TMB), gene-expression profile (GEP) score, and programmed death ligand-1 (PD-L1) expression, has been associated with a favorable response to immune checkpoint inhibitors like pembrolizumab.

The study further evaluated the prognostic impact of TMB, tumor inflammation signature (TIS), and PD-L1 expression within the treatment arms. The RFS analyses in biomarker-high and -low subgroups aimed to assess whether the combination of mRNA-4157 (V940) with pembrolizumab not only deepened the clinical benefit in populations already responsive to pembrolizumab monotherapy but also extended benefit to new patient populations.

Results indicated that the increased RFS observed with the mRNA-4157 (V940) and pembrolizumab combination was consistent across both TMB-high and TMB-non-high subgroups. Notably, there was a larger subgroup of TMB-high patients in the combination arm, which might suggest a particular benefit in this population.

Additionally, the RFS benefit was observed in both TIS-high and TIS-low subgroups. This finding is in alignment with previous observations that pembrolizumab monotherapy provides improved RFS in TIS-high compared to TIS-low subgroups.

The analysis of baseline characteristics showed a balanced distribution of biomarkers across both study arms, supporting the validity of the comparisons made between the treatment groups. These biomarkers included TMB, TIS, and PD-L1 expression, which were evaluable in a significant majority of patients and used to evaluate their association with the primary study endpoint of RFS.

In conclusion, the combination of mRNA-4157 (V940) with pembrolizumab demonstrated a trend for improved RFS compared to pembrolizumab alone in adjuvant melanoma patients, with these benefits observed across various tumor mutational burden subgroups. This suggests that personalized cancer vaccines in combination with checkpoint inhibitors may represent a new frontier in the management of high-risk melanoma, with the potential to improve patient outcomes significantly.

new vaccine for melanoma

Safety Profile and Tolerability of the Vaccine

The safety and tolerability of mRNA-4157 (V940), an innovative mRNA-based personalized cancer vaccine, when used in conjunction with pembrolizumab, are critical considerations in its clinical evaluation. The mRNA-4157-P201/KEYNOTE-942 trial provided valuable insights into the vaccine's safety profile, which is paramount for patient care and treatment decision-making.

In the trial, the combination of mRNA-4157 (V940) with pembrolizumab was administered to a cohort of patients with resected high-risk stage III/IV melanoma. The safety and tolerability data generated from this trial are crucial for understanding the potential side effects and overall patient experience associated with this novel therapeutic approach.

Adverse events (AEs) associated with vaccine administration are a primary concern, as they can impact patient compliance and the overall success of the treatment regimen. The trial closely monitored patients for AEs, with a focus on identifying any that could be directly attributed to the vaccine therapy. This information is critical for clinicians to manage and mitigate potential risks to patients undergoing treatment.

Moreover, the tolerability of the vaccine, which refers to the degree to which observable AEs are bearable by patients, is another key outcome of interest. Tolerability measures help to ensure that the benefits of the vaccine outweigh the discomfort or complications experienced by the patient. It is essential for the maintenance of quality of life during treatment and for the long-term acceptance of the vaccine as a viable therapeutic option.

The comprehensive analysis of the safety profile and tolerability of mRNA-4157 (V940) in combination with pembrolizumab will inform future clinical practices and guide the development of protocols to enhance patient outcomes. By carefully examining and reporting these aspects, the trial contributes to the body of knowledge required to establish mRNA-4157 (V940) as a safe and patient-friendly treatment alternative for melanoma and potentially other cancers where neoantigen-targeting strategies are applicable.

It is important to note that detailed results regarding specific AEs, their severity, incidence rates, and the management strategies employed during the trial would further illuminate the vaccine's safety profile. Such data would be particularly valuable for healthcare providers in assessing the risk-benefit profile of incorporating mRNA-4157 (V940) into clinical practice for the treatment of high-risk melanoma patients.

Biomarker Analysis in mRNA-4157 (V940) Clinical Trials

Recent advances in immunotherapy have brought about the development of personalized cancer vaccines, one of which is mRNA-4157 (V940). The use of such vaccines in combination with immune checkpoint inhibitors like pembrolizumab has been under investigation in clinical trials to determine their efficacy in improving patient outcomes. In the mRNA-4157-P201/KEYNOTE-942 clinical trial, mRNA-4157 (V940) combined with pembrolizumab showed a trend for improved recurrence-free survival (RFS) compared to pembrolizumab alone in patients with resected high-risk stage III/IV melanoma.

The analysis of biomarkers such as Tumor Mutational Burden (TMB), Tumor Inflammation Signature (TIS), and Programmed Death Ligand-1 (PD-L1) expression in these trials is crucial for understanding the prognostic value of these factors and their associations with clinical outcomes. TMB is a measure of the number of mutations within a tumor and is thought to correlate with the likelihood of response to immunotherapies, as a higher TMB increases the potential for tumor antigens to be recognized by T cells . TIS is an exploratory gene-expression profile that reflects the level of inflammation within the tumor, which is indicative of T-cell infiltration and function. PD-L1 expression, evaluated on tumor cells and infiltrating immune cells, is another biomarker that has been associated with responses to pembrolizumab.

Objectives of biomarker analysis in these clinical trials included assessing the distribution of TMB, TIS, and PD-L1 expression in baseline tumors across both study arms, exploring the prognostic impact of these biomarkers within each arm, and assessing RFS in biomarker-high and -low subgroups.

Methods for biomarker analysis involved using formalin-fixed paraffin-embedded (FFPE) tumor biopsies and matched normal blood samples for whole-exome sequencing to determine TMB, and RNA sequencing for TIS. PD-L1 expression was examined through immunohistochemistry, with a combined positivity score (CPS) being used for evaluation.

Results of these analyses showed that baseline characteristics were generally balanced between the study arms across most biomarker subgroups. A larger subgroup of TMB-high patients was observed in the mRNA-4157 (V940) and pembrolizumab arm compared to the pembrolizumab monotherapy arm. The RFS benefit observed with the combination therapy was maintained across both high and non-high TMB subgroups, with a similar magnitude of treatment effect. Improved RFS was also seen in TMB-high compared to TMB--non-high patient subgroups in the pembrolizumab monotherapy arm. Additionally, an increased RFS benefit in the TMB-high subgroup was observed in the combination arm.

The study also reported RFS benefits in both TIS-high and TIS-low subgroups, with an increased benefit observed in the TIS-high subgroup. The distribution of TIS and PD-L1 expression was balanced between study arms, and the increased RFS benefit with the combination therapy was seen in all subgroups analyzed.

In conclusion, the mRNA-4157 (V940) clinical trials' biomarker analysis underscores the importance of TMB, TIS, and PD-L1 as potential prognostic tools in the context of personalized immunotherapy. The data suggest that patients with higher levels of these biomarkers may derive more significant benefits from the combination of mRNA-4157 (V940) with pembrolizumab, pointing towards a future where treatment can be more tailored to individual tumor profiles.

Tumor Mutational Burden (TMB) and Its Prognostic Value

Tumor Mutational Burden (TMB) is a measure of the total number of non-synonymous mutations per coding area of a tumor genome. High TMB is often associated with better responses to immunotherapies, such as checkpoint inhibitors, because a greater number of mutations increases the likelihood of producing neoantigens that are recognized by the immune system. This recognition is crucial for T-cell mediated anti-tumor responses, which are the basis of many immunotherapy strategies.

The prognostic value of TMB is well-documented in various cancers, with higher TMB predicting improved outcomes in patients treated with immunotherapies. In the context of mRNA-4157 (V940), a novel mRNA-based individualized neoantigen therapy, assessing TMB provides insights into its efficacy in combination with pembrolizumab, a widely used immune checkpoint inhibitor.

In clinical trials, TMB has been used to stratify patients and predict their response to treatment. For instance, in the mRNA-4157-P201/KEYNOTE-942 trial, TMB was evaluated using whole-exome sequencing (WES) to identify patient-specific somatic mutations. A threshold of 175 mutations per exome (equivalent to 10 mutations/megabase per F1CDx) was set to define TMB-high status, based on previously published WES TMB scores for pembrolizumab.

The trial's results showed that the combination of mRNA-4157 (V940) and pembrolizumab improved recurrence-free survival (RFS) when compared to pembrolizumab alone, and this benefit was observed across both high and non-high TMB subgroups. Notably, there was a larger proportion of TMB-high patients in the combination arm than in the pembrolizumab monotherapy arm.

Furthermore, Kaplan-Meier analyses and Cox proportional hazards models were used to assess the association between TMB and the primary study endpoint of RFS. These analyses indicated that patients with high TMB had better outcomes compared to those with non-high TMB in both study arms, aligning with previous findings that high TMB is a favorable prognostic factor for pembrolizumab therapy.

In summary, high TMB is a significant biomarker for predicting the prognosis of patients undergoing immunotherapy. The clinical trial data support the notion that patients with high TMB may derive greater benefit from the combination of mRNA-based individualized neoantigen therapies like mRNA-4157 (V940) and pembrolizumab. This underscores the importance of TMB as a biomarker in the development and application of personalized cancer immunotherapies.

Tumor Inflammation Signature (TIS) and PD-L1 Expression

The Tumor Inflammation Signature (TIS) is an exploratory gene-expression profile similar to the Gene Expression Profile (GEP) score, which provides a measure of the immune activity in the tumor microenvironment. The TIS has been recognized as a potential biomarker for predicting the response to immune checkpoint inhibitors, including pembrolizumab. TIS is calculated as the weighted average of 18 genes included in the GEP score and represents the level of tumor inflammation, which is indicative of T-cell infiltration and function. A high TIS score is generally associated with a more inflamed tumor microenvironment that may be more amenable to the effects of T-cell-based immunotherapies.

Programmed Death-Ligand 1 (PD-L1) expression is another biomarker that has been extensively studied in the context of immunotherapy. PD-L1 is a protein expressed on tumor cells and tumor-infiltrating immune cells that can inhibit T-cell function. The presence of PD-L1 on tumor cells can engage with PD-1 receptors on T-cells, leading to the suppression of anti-tumor immune responses. Immune checkpoint inhibitors, such as pembrolizumab, can block this interaction, thereby restoring the immune system's ability to attack cancer cells. In the mRNA-4157-P201/KEYNOTE-942 clinical trial, PD-L1 positivity was defined as a combined positivity score (CPS) of ≥1, as determined by immunohistochemistry staining.

In the context of the mRNA-4157-P201/KEYNOTE-942 study, the prognostic impact of TIS and PD-L1 expression within the study arms was explored. The study evaluated the recurrence-free survival (RFS) in biomarker-high and -low subgroups across the study arms. The goal was to determine whether the combination of mRNA-4157 (V940) and pembrolizumab could deepen the clinical benefit in populations already responsive to pembrolizumab monotherapy and/or extend clinical benefit into new patient populations.

The mRNA-4157 (V940) is a personalized cancer vaccine that encodes up to 34 patient-specific tumor neoantigens and has demonstrated the potential to induce antigen-specific T-cell responses. This personalized approach to cancer treatment, in combination with pembrolizumab, aims to harness the patient's immune system to target and eliminate tumor cells.

Results from the study showed that the distribution of TIS and PD-L1 expression in baseline tumor biopsies was balanced between the study arms, suggesting that any observed differences in treatment outcomes could be attributed to the treatment itself rather than baseline imbalances. Furthermore, the study found that the RFS benefit of the mRNA-4157 (V940) and pembrolizumab combination was observed in both TIS-high and TIS-low subgroups. This finding is consistent with other indications and lines of therapy for pembrolizumab, where improved RFS was observed in TIS-high compared to TIS-low subgroups.

In summary, the TIS and PD-L1 expression are valuable biomarkers for assessing the tumor microenvironment's readiness to respond to immunotherapy. The mRNA-4157-P201/KEYNOTE-942 trial's findings suggest that the combination of mRNA-4157 (V940) and pembrolizumab may offer a promising therapeutic strategy for patients with resected high-risk stage III/IV melanoma, irrespective of their baseline TIS and PD-L1 expression levels.

Associations Between Biomarkers and Clinical Outcomes

In the investigation of mRNA-4157 (V940) in conjunction with pembrolizumab for the treatment of high-risk resected melanoma, particular attention has been devoted to understanding how biomarkers such as Tumor Mutational Burden (TMB), Tumor Inflammation Signature (TIS), and PD-L1 expression correlate with clinical outcomes, notably recurrence-free survival (RFS). These biomarkers are critical as they offer a window into the tumor microenvironment and its potential response to immunotherapy.

The pivotal phase 2 mRNA-4157-P201/KEYNOTE-942 trial demonstrated that the combination therapy improved RFS compared to pembrolizumab monotherapy, with a noteworthy hazard ratio (HR) suggesting a trend toward benefit across different biomarker subgroups. It was observed that patients with a high TMB experienced a more pronounced RFS benefit when treated with the combination therapy compared to pembrolizumab alone, reflecting the assumption that a higher mutational load might increase the likelihood of tumor antigen recognition by T cells, thus enhancing the response to T-cell-based immunotherapies.

Similarly, patients with a high TIS, reflective of an inflamed tumor microenvironment, showed improved RFS. This finding aligns with prior evidence suggesting that an inflamed tumor milieu, characterized by T-cell infiltration and function, is conducive to the efficacy of T-cell-based immunotherapies. The beneficial impact of the mRNA-4157 (V940) and pembrolizumab combination was apparent in both TIS-high and TIS-low subgroups, indicating the vaccine's potential to elicit a favorable clinical response regardless of the initial inflammation status of the tumor.

Moreover, the analysis of PD-L1 expression, a biomarker indicative of immune evasion strategies employed by tumor cells, was included to assess its association with treatment outcomes. While the data on PD-L1's correlation with RFS in the context of this study have not been explicitly detailed, existing literature suggests that PD-L1 expression is a predictor of response to pembrolizumab, potentially providing a rationale for its combinatory use with mRNA-4157 (V940) in patients with PD-L1 positive tumors.

The Kaplan-Meier analyses and Cox proportional hazards model applied to the study data underscore the importance of these biomarkers in predicting and understanding the clinical outcomes of patients undergoing treatment with mRNA-4157 (V940) and pembrolizumab. These findings pave the way for a more nuanced approach to melanoma therapy, where individual patient biomarkers could guide treatment decisions and the use of personalized neoantigen vaccines in combination with checkpoint inhibitors to maximize clinical benefit.

Bibliography

  1. Shannon L. Oral presentation CT001. 2. J Cancer Res Clin Oncol. 2023;37(4):4685-4689. (https://s29.q4cdn.com/435878511/files/doc_presentations/2023/Apr/18/sullivan_mmapcv75571.pdf)

  2. Weber, Jeffrey S., et al. "Individualised neoantigen therapy mRNA-4157 (V940) plus pembrolizumab versus pembrolizumab monotherapy in resected melanoma (KEYNOTE-942): a randomised, phase 2b study." The Lancet (2024). (https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(23)02268-7/fulltext)

  3. Conroy, Russ. "FDA Grants Breakthrough Designation to mRNA Vaccine for Resected Melanoma." Cancer Network (2023): NA-NA. (https://go.gale.com/ps/i.do?p=HRCA&sw=w&issn=&v=2.1&it=r&id=GALE%7CA775268741&sid=googleScholar&linkaccess=abs)

  4. Conroy, Russ. "mRNA Vaccine Continues to Show Promise in Resected Melanoma." Oncology Nursing News (2023): NA-NA. (https://go.gale.com/ps/i.do?p=HRCA&sw=w&issn=&v=2.1&it=r&id=GALE%7CA777409821&sid=googleScholar&linkaccess=abs)

  5. Seymour, Caroline. "Cancer Vaccine mRNA-4157 Improves Recurrence-Free Survival for Patients With High-Risk Melanoma." (https://www.oncnursingnews.com/view/cancer-vaccine-mrna-4157-improves-recurrence-free-survival-for-patients-with-high-risk-melanoma)

  6. Froelich, Warren. "Cancer Vaccine Research Shows Promise as Melanoma Treatment." (2023): 25-30. (https://journals.lww.com/oncology-times/fulltext/2023/05200/cancer_vaccine_research_shows_promise_as_melanoma.13.aspx)

  7. Seymour, Caroline. "Combination of mRNA-4157 With Pembrolizumab Improves Survival in Melanoma." (https://www.targetedonc.com/view/combination-of-mrna-4157-with-pembrolizumab-improves-survival-in-melanoma)