Review Article

Advantages and Limitations of mRNA Cancer Vaccines

Nemat Khansari*

Tehran Medical Sciences University, School of Medicine, Department of Immunology, Iran

Received Date: 16/12/2023; Published Date: 01/05/2024

*Corresponding author: Nemat Khansari, Tehran Medical Sciences University, School of Medicine, Department of Immunology, Iran

DOI: 10.46998/IJCMCR.2023.36.000888

Abstract

While mRNA cancer vaccines hold immense promise, challenges remain in their development and optimization. Researchers are actively working to overcome tumor heterogeneity, enhance mRNA delivery efficiency, and optimize immune activation strategies. As research progresses, mRNA cancer vaccines are poised to revolutionize cancer therapy, offering a personalized, targeted, and practical approach to combating this complex disease. This article focuses on the advantages of traditional cancer treatments, such as chemotherapy and radiation therapy, as well as the limitations of the application of mRNA vaccines for the treatment and prevention of cancers.

Keywords: mRNA; Vaccine; Immunotherapy; Cancer; Checkpoint inhibitors

Cancer is a complex and multifaceted disease that affects millions of people worldwide. One of the most promising advances in cancer treatment and prevention is the development of mRNA cancer vaccines [1]. These vaccines harness the immune system’s power to target and eliminate cancer cells, offering a revolutionary approach to cancer therapy [2]. mRNA cancer vaccines utilize the unique properties of messenger RNA (mRNA) molecules to instruct the body's immune system to recognize and attack cancer cells. Unlike traditional vaccines that introduce a weakened or inactivated form of a virus or bacteria into the body, mRNA vaccines provide the genetic code for a specific cancer-associated protein or antigen [3]. When the mRNA is introduced into the body, cells take up it, which then uses the genetic instructions to produce the target antigen [4]. This antigen is presented on the cell surface, where the immune system recognizes it as foreign. Consequently, the immune system mounts a response against the antigen, priming it to identify and eliminate any cancer cells that display the same antigen in the future. This targeted approach enhances the body's ability to fight cancer and reduces the risk of side effects associated with conventional cancer treatments [5,6].

Advantages of mRNA Cancer Vaccines

mRNA cancer vaccines offer several advantages over traditional cancer treatments, such as chemotherapy and radiation therapy, including:

Personalized Treatment
One of the significant challenges in cancer treatment is the variability in cancer types and individual responses to therapy. mRNA cancer vaccines can be tailored to each patient's specific cancer, targeting the unique antigens present in their tumor. This personalized approach increases the chances of successful treatment, targeting particular proteins found in an individual's cancer cells [7].

Minimal Side Effects
Traditional cancer treatments, such as chemotherapy and radiation therapy, often cause severe side effects by damaging healthy cells along with cancer cells. In contrast, mRNA cancer vaccines specifically target cancer cells, minimizing damage to healthy tissue and reducing the risk of side effects [7].

Potential for Prevention
mRNA cancer vaccines may also promise to prevent cancer from developing in high-risk individuals. By introducing mRNA encoding tumor-specific antigens into the body, the immune system can be "primed" to recognize and eliminate cancer cells before they can form a tumor [8].

Potential Challenges and Limitations of mRNA Cancer Vaccines

Immune Tolerance
One potential challenge in developing effective mRNA cancer vaccines is overcoming immune tolerance. The immune system is designed to distinguish between self and non-self to prevent autoimmune reactions. However, some cancer cells can exploit this mechanism by expressing antigens that closely resemble normal cellular proteins, making it difficult for the immune system to recognize them as foreign. In such cases, an mRNA vaccine encoding these antigens may not elicit a potent enough immune response to target and eliminate cancer cells [9] effectively.

To address this challenge, researchers are exploring strategies to enhance the immunogenicity of mRNA-encoded antigens. These approaches include combining the mRNA vaccine with adjuvants or other immunostimulatory agents that can boost the immune response against tumor-specific antigens [10].

Escape Mechanisms
Cancer cells are known for their ability to evolve and adapt, which can lead to escape mechanisms that allow them to evade the immune response triggered by an mRNA cancer vaccine. For example, cancer cells could alter or down-regulate the expression of targeted antigens, rendering the vaccine less effective over time.

To counteract these escape mechanisms, scientists are investigating multi-targeted mRNA vaccines that encode multiple tumor-specific antigens simultaneously. By targeting several distinct antigens simultaneously, these vaccines increase the likelihood of successfully eliminating cancer cells, even if they undergo antigenic changes during treatment [11].

Another approach is to design personalized combination therapies that integrate mRNA cancer vaccines with other treatments, such as checkpoint inhibitors or targeted therapies. This strategy may help overcome resistance mechanisms by attacking cancer cells from multiple angles while focusing strongly on patient-specific targets. While challenges are associated with immune tolerance and escape mechanisms in developing successful mRNA cancer vaccines, ongoing research efforts aim to overcome these limitations through innovative strategies and combination therapies [12]. Overcoming these obstacles will be crucial in realizing the full potential of mRNA cancer vaccines in treating various types of cancer.

Enhancing Efficacy of mRNA Cancer Vaccines
Adjuvants play a crucial role in enhancing the efficacy of vaccines by boosting the immune response against target antigens. In the context of mRNA cancer vaccines, adjuvants can increase the immunogenicity and potency of the encoded antigens. Examples of adjuvants investigated for use with mRNA cancer vaccines include toll-like receptor (TLR) agonists, stimulators of interferon genes (STING) agonists, and cytokines such as interleukin-12 (IL-12). These adjuvants trigger innate immune pathways, leading to more robust and sustained adaptive immune responses against tumor-specific antigens [13].

Combination Therapies for Improved Outcomes
Combining mRNA cancer vaccines with other therapeutic modalities can enhance their effectiveness by addressing various aspects of cancer biology and overcoming potential limitations or resistance mechanisms. Some promising combination therapies include:

Checkpoint Inhibitors
As mentioned earlier, combining mRNA cancer vaccines with checkpoint inhibitors can create a synergistic effect that maximizes the immune system's ability to target and eliminate cancer cells. This combination approach may improve treatment outcomes across various types of cancers by priming the immune response against specific tumor antigens through an mRNA vaccine and then further unleashing T-cell activity using checkpoint inhibitors [14].

Targeted Therapies
Targeted therapies are designed to interfere with specific molecular pathways involved in cancer cell growth and survival. Combining targeted therapies with mRNA cancer vaccines may enhance treatment efficacy by simultaneously attacking cancer cells from multiple angles – both at the molecular level and through an immune-mediated response. This strategy may also help overcome resistance mechanisms that could arise during treatment [15].

Radiotherapy
Radiotherapy is a standard modality used in treating many cancers. Recent studies suggest that combining radiotherapy with mRNA cancer vaccines could have a synergistic effect. Radiotherapy can trigger the release of tumor-specific antigens, making them more accessible to the immune cells primed by an mRNA vaccine. Additionally, radiotherapy-induced inflammation may further enhance the immunogenicity of the target antigens and promote a more robust immune response against cancer cells [16].

Optimizing Vaccine Administration Schedules
Another approach to enhance the efficacy of mRNA cancer vaccines is optimizing their administration schedules. This involves determining the optimal timing, dosage, and sequence of vaccine doses for other treatments in a patient's therapy plan. By carefully designing these schedules, researchers aim to maximize the immune response against tumor-specific antigens while minimizing potential side effects or interference with other therapies. Furthermore, several strategies can be employed to enhance the efficacy of mRNA cancer vaccines, including adjuvants, combination therapies, and optimized administration schedules. By exploring these approaches in clinical trials and preclinical research, scientists aim to improve patient treatment outcomes and bring us closer to realizing the full potential of this promising new class of cancer therapeutics.

Ethical Considerations and Public Perceptions in Genetic Material-based Cancer Treatment Informed Consent and Privacy
The use of genetic material in cancer treatment raises ethical concerns regarding informed consent and privacy. Patients must be adequately informed about the nature of mRNA cancer vaccines, the potential risks, and the benefits before deciding to participate in clinical trials or receive treatment. Ensuring patients understand the implications of using their genetic information is crucial for maintaining trust and respecting autonomy.

Moreover, protecting patients' privacy is essential when handling sensitive genetic data. Strict measures should be in place to prevent unauthorized access or misuse of this information. Clear data storage, sharing, and usage guidelines are necessary to address potential privacy breaches.

Equitable Access to Treatment
Another ethical consideration is equitable access to mRNA cancer vaccines and other genetic material-based treatments. As these therapies often involve cutting-edge technology and personalized approaches, they may initially be expensive and available only to a limited number of patients. Establishing strategies that promote fair distribution of these treatments across various socio-economic groups and geographical locations is crucial to avoiding disparities in healthcare access.

Public Perceptions: Balancing Hope with Realistic Expectations
Public perceptions surrounding mRNA cancer vaccines play an essential role in shaping acceptance and adoption rates for these novel treatments. While it is vital to communicate the potential benefits of these therapies, it is equally important not to overstate their capabilities or promise immediate success.

Balancing hope with realistic expectations is crucial for maintaining public trust in scientific research and medical advancements. Providing accurate information about the progress of ongoing clinical trials, potential limitations, and uncertainties can help foster a better understanding among the public about the complexities involved in developing effective mRNA cancer vaccines.

In addition, ethical considerations such as informed consent, privacy protection, equitable access to treatment, and managing public perceptions through transparent communication will be critical factors in ensuring the responsible development and implementation of genetic material-based cancer treatments like mRNA cancer vaccines.

Economic Implications of Developing and Implementing Personalized mRNA Cancer Vaccines

Research and Development Costs
The development of personalized mRNA cancer vaccines requires significant investment in research and development. These costs stem from the need for advanced technologies, such as next-generation sequencing and bioinformatics tools, to identify tumor-specific antigens and design patient-specific vaccine candidates. Moreover, funding is necessary to support preclinical studies, clinical trials, and regulatory approval processes.

While these expenses can be substantial, successful mRNA cancer vaccine development can transform cancer treatment paradigms and provide long-term economic benefits by reducing cancer-related healthcare costs.

Manufacturing Challenges
Personalized mRNA cancer vaccines pose unique manufacturing challenges due to their individualized nature. Unlike traditional one-size-fits-all therapies, these vaccines must be custom-made for each patient based on their specific genetic information. This requirement requires flexible production facilities capable of swiftly adapting to different vaccine formulations.

Investment in modular manufacturing systems that rapidly produce personalized vaccines may be required to meet this demand. Although these systems may incur higher initial setup costs, they can streamline vaccine production while maintaining quality standards.

Pricing Strategies
Determining the appropriate pricing strategy for personalized mRNA cancer vaccines is a complex task that must balance multiple factors. These include research and development costs, manufacturing expenses, market competition, and equitable access to innovative treatments.

Pricing strategies should consider the value these novel therapies provide – such as improved survival rates or reduced side effects – and cost-saving benefits resulting from potentially reducing hospitalization or additional supportive care requirements.

Health Insurance Coverage and Reimbursement Policies
Health insurance coverage is critical in determining patients' access to new treatments, like personalized mRNA cancer vaccines. To ensure broad accessibility, insurance companies and public health systems will need to adapt reimbursement policies for these innovative therapies.

Collaboration between pharmaceutical companies, healthcare providers, policymakers, and insurers will be essential to develop reimbursement models that account for the unique nature of personalized mRNA cancer vaccines while promoting cost-effective healthcare solutions.

Economic Impact on Healthcare Systems
The widespread adoption of personalized mRNA cancer vaccines has the potential to impact healthcare systems significantly. By offering a more targeted approach to cancer treatment, these vaccines may improve patient outcomes, reducing healthcare costs associated with hospitalizations, supportive care, and long-term management of side effects from traditional therapies.

However, the initial investment required for the research, development, manufacturing, and implementation of these novel treatments must be weighed against their long-term economic benefits. Addressing the financial implications of developing and implementing personalized mRNA cancer vaccines is crucial for ensuring their successful integration into healthcare systems. Balancing costs with potential benefits will require collaborative efforts among stakeholders to develop sustainable pricing strategies and reimbursement policies that promote equitable access and efficient use of resources.

Collaborative Partnerships for Accelerating mRNA Cancer Vaccine Research
Fostering collaborative partnerships between academia, biotechnology companies, and pharmaceutical industries is crucial to expediting the development and implementation of mRNA cancer vaccines. These collaborations can harness each sector's unique expertise and resources to tackle vaccine design, manufacturing, and clinical testing challenges effectively.

Academic Institutions
Academic institutions drive cutting-edge research and innovation in mRNA cancer vaccines. By partnering with biotechnology companies and pharmaceutical industries, academic researchers can access funding, specialized equipment, and industry insights that can help translate their findings into viable treatment options.

Collaborations with academia also provide opportunities for training and education programs to nurture future generations of scientists who will contribute to the continued advancement of mRNA cancer vaccine research.

Biotechnology Companies
Biotechnology companies possess specialized knowledge in molecular biology, genetic engineering, and bioinformatics – all essential components in developing mRNA cancer vaccines. By collaborating with academic institutions and pharmaceutical industries, these companies can share their expertise while benefiting from complementary research resources provided by their partners.

These partnerships may lead to innovative solutions for overcoming challenges related to vaccine design, delivery systems optimization, or manufacturing processes – ultimately accelerating the development of effective patient treatments.

Pharmaceutical Industries
Pharmaceutical industries have extensive experience navigating regulatory landscapes and conducting large-scale clinical trials to market new therapies. In addition to providing financial support for research initiatives, these entities can offer guidance on navigating complex approval processes associated with novel therapeutics like mRNA cancer vaccines.

By partnering with academic institutions and biotechnology companies, pharmaceutical industries can help streamline the transition from preclinical studies to clinical trials while ensuring compliance with regulatory requirements during every stage of development.

Joint Research Initiatives & Consortia
Establishing joint research initiatives or consortia involving academia, biotechnology companies, and pharmaceutical industries is another approach to facilitate collaboration. Such consortia can serve as a platform for sharing knowledge, resources, and expertise while jointly addressing the challenges of mRNA cancer vaccine development.

These collaborative efforts can lead to standardized protocols, shared databases, or centralized bio-banks that enable more efficient research practices and accelerate the progress in mRNA cancer vaccines. It should be noted that fostering partnerships between academia, biotechnology companies, and pharmaceutical industries is essential for accelerating mRNA cancer vaccine research. By leveraging each sector's unique strengths and resources through collaborative efforts, these alliances can drive innovation and rapidly bring life-saving treatments to patients.

Long-term Safety and Potential Side Effects of mRNA Cancer Vaccines Duration of Immune Response
Understanding the duration of immune response elicited by mRNA cancer vaccines is crucial for evaluating their long-term safety. While these vaccines have shown promising results in generating potent immune responses, it is essential to determine how long-lasting these effects are and whether booster doses will be required over time. Further research should focus on monitoring patients treated with mRNA cancer vaccines to assess the persistence of immune response and modify vaccination strategies accordingly.

Late-Onset Side Effects
Although short-term side effects of mRNA cancer vaccines tend to be mild [17], it is necessary to investigate potential late-onset side effects that may emerge over an extended period. Long-term follow-up studies can help identify delayed adverse reactions by monitoring patients for several years after receiving the vaccine. This information will be crucial in refining the safety profile of these therapies and informing appropriate risk management strategies.

Autoimmunity Concerns
While mRNA cancer vaccines are designed to target tumor-specific antigens, there is a potential risk of triggering autoimmunity if the immune system mistakenly attacks healthy tissues expressing similar antigens. Long-term studies should evaluate the incidence of autoimmune disorders in patients who have received mRNA cancer vaccines compared to those who have not, providing valuable insights into potential risks associated with this treatment modality [18].

Integration into Host Genome
One concern related to genetic material-based therapies is the possible integration of exogenous genetic material into the host genome. This could lead to unintended consequences, such as gene disruption or activation of oncogenes. However, current evidence suggests that mRNA molecules used in cancer vaccines do not integrate into the host genome due to their transient nature and rapid degradation within cells. Nevertheless, ongoing research should continue to monitor this aspect closely to ensure long-term safety.

Impact on Fertility and Pregnancy
The potential impact of mRNA cancer vaccines on fertility and pregnancy outcomes remains an area that requires further investigation. Preclinical studies and clinical trials should include reproductive health assessments and evaluate potential adverse effects on fertility, embryonic development, or pregnancy outcomes. This information will be vital in informing appropriate patient counseling and treatment recommendations for individuals of reproductive age [19].

Furthermore, investigating mRNA cancer vaccines' long-term safety and potential side effects is essential for fully understanding their risk-benefit profile. By conducting comprehensive follow-up studies and closely monitoring patients over time, researchers can address concerns related to immune response duration, late-onset side effects, autoimmunity risks, genomic integration, and impact on fertility – ultimately contributing to the responsible development and implementation of these innovative cancer therapeutics [20].

Combining mRNA Cancer Vaccines with Traditional Cancer Treatments Chemotherapy
Exploring the combination of mRNA cancer vaccines with chemotherapy may provide synergistic effects in cancer treatment. While chemotherapy is effective at killing rapidly dividing cells, it can also cause damage to healthy tissues and lead to various side effects. By integrating mRNA cancer vaccines into a treatment regimen alongside chemotherapy, the immune system can be primed to target tumor-specific antigens more effectively, potentially reducing the required dosage of chemotherapeutic agents and minimizing side effects [21].

Moreover, combining these two therapies may also overcome some resistance mechanisms that cancer cells employ. While chemotherapy can induce cell death and release tumor-derived antigens, an mRNA vaccine can further boost the immune response against these antigens, enhancing overall treatment efficacy.

Radiation Therapy
Combining mRNA cancer vaccines with radiation therapy offers another promising approach for improving cancer treatment outcomes. As mentioned earlier, radiotherapy-induced inflammation can enhance the immunogenicity of tumor antigens and promote a more robust immune response against cancer cells. When used in conjunction with an mRNA vaccine that primes the immune system to recognize specific tumor antigens, this combination therapy may lead to improved anti-tumor activity.

Additionally, radiation therapy has been shown to increase the expression of specific molecules on the surface of cancer cells, which makes them more susceptible to immune-mediated attack. This effect could further amplify the benefits of combining radiation therapy with an mRNA cancer vaccine by increasing their ability to work together in targeting and eliminating malignant cells [22].

In conclusion, combining mRNA cancer vaccines with traditional cancer treatments, such as chemotherapy or radiation therapy, holds the potential for enhancing therapeutic outcomes while minimizing side effects. Further research is needed to optimize these combined approaches and determine their efficacy across different types of cancers.

Assessing the Potential of mRNA Cancer Vaccines for Rare or Difficult-to-Treat Cancers Broad

Applicability Across Cancer Types
One of the most promising aspects of mRNA cancer vaccines is their potential application across many cancer types. By targeting tumor-specific antigens, these vaccines can be tailored to suit individual patients' needs, regardless of the type or stage of their cancer. This flexibility makes them particularly attractive for treating rare or difficult-to-treat malignancies that may not respond well to traditional therapies.

Overcoming Heterogeneity in Tumors
Tumor heterogeneity poses a significant challenge in cancer treatment, as diverse populations of cancer cells within a single tumor may exhibit different characteristics and vulnerabilities. The adaptability of mRNA cancer vaccines allows them to target multiple antigens simultaneously [23]. This heterogeneity is addressed by inducing immune responses against various subpopulations of cancer cells. This multi-targeted approach could improve treatment outcomes for patients with complex or heterogeneous tumors.

Targeting Oncogenic Drivers and Neoantigens
In some rare or difficult-to-treat cancers, specific oncogenic drivers may play a crucial role in tumor development and progression. By identifying these drivers and designing mRNA vaccines that target their associated antigens, researchers can develop personalized treatments capable of combating unique aspects of these malignancies. Additionally, focusing on neoantigens – novel antigens generated by tumor-specific mutations – can further enhance the specificity and efficacy of mRNA cancer vaccines in challenging cases [24].

Addressing Treatment Resistance
Resistance to conventional therapies is often a significant barrier to successful treatment in rare or difficult-to-treat cancers. The combination approaches discussed earlier – such as combining mRNA cancer vaccines with chemotherapy or radiation therapy – can help overcome resistance mechanisms by attacking tumors from multiple angles simultaneously. This strategy may lead to improved outcomes for patients who have previously failed other forms of therapy.

Early Detection and Prophylactic Applications
Early detection and intervention are critical for achieving favorable outcomes in some rare or aggressive cancer types. The development of mRNA cancer vaccines targeting specific tumor antigens associated with these cancers could potentially be used as prophylactic measures in high-risk individuals or as adjuvant therapy following surgery to prevent recurrence. The potential for mRNA cancer vaccines to treat different types of cancers, including rare or difficult-to-treat malignancies, is an exciting prospect. By addressing challenges such as tumor heterogeneity, oncogenic drivers, treatment resistance, and early detection through tailored and combination approaches, these innovative therapeutics promise to revolutionize the way we combat even the most formidable cancers.

Effectiveness of mRNA Cancer Vaccines in Specific Patient Populations Elderly Individuals
Evaluating the effectiveness of mRNA cancer vaccines in elderly individuals is essential, as this population often experiences age-related changes in immune function that could impact vaccine response. Research should focus on understanding how factors such as immunosenescence, comorbidities, and polypharmacy influence the efficacy and safety of mRNA cancer vaccines in older adults. Tailoring vaccination strategies to account for these unique challenges may help optimize treatment outcomes for this vulnerable population [25].

Immunocompromised Patients
Another critical patient population to consider is those with compromised immune systems, such as individuals undergoing immunosuppressive therapy or those with autoimmune disorders. The efficacy of mRNA cancer vaccines in immunocompromised patients may be influenced by their underlying medical condition or medications that weaken the immune response. Investigating alternative dosing regimens or adjuvant interventions to enhance vaccine-induced immunity could be crucial for ensuring optimal therapeutic benefits for these patients.

Conclusion

Assessing the effectiveness of mRNA cancer vaccines in specific patient populations, like elderly individuals and those with compromised immune systems, is vital for developing tailored treatment approaches that consider each group's unique needs and challenges. By conducting focused research on these populations, we can work towards more inclusive and effective cancer therapies that cater to a diverse range of patients. In addition, investigating the efficacy and safety of mRNA cancer vaccines in pediatric patients is another important aspect of cancer research. Children's immune systems are still developing and may respond differently to vaccination compared to adults. Furthermore, the safety profile and potential side effects in this population must be carefully examined, as their long-term health and development must be considered. Studies focusing on pediatric cancer patients can help determine optimal dosing, administration schedules, and potential modifications to the vaccines to ensure both safety and effectiveness in treating childhood cancers.

References

  1. Vogel AB, Lambert L, Kinnear E, Busse D, Erbar S, Reuter KC, et al. Self-amplifying RNA vaccine give equivalent production against influenza to mRNA vaccines but at much lower doses. Mol. Ther, 2018; 26: 466-455.
  2. Wadhwa A, Aljabbara A, Lokras A, Foged C, Thakur A. Opportunities and challenges in the delivery of mRNA-based vaccines. Pharmaceutics, 2020; 12: 102-105.
  3. Rosa SS, Prazeres DMF, Azevedo AM, Marques MPC. mRNA vaccines manufacturing: Challenges and bottlenecks. Vaccine, 2020; 39: 2190-2200.
  4. Horie M, Honda T, Suzuki Y, Kobayashi Y, Daito T, et al. Endogenous non-retroviral RNA virus elements in mammalian RNA genomes. Nature, 2010; 463: 84-87.
  5. Green AK. Challenges in assessing the cost-effectiveness of cancer immunotherapy. JAMA, 2021; 4: e2034020.
  6. Lin MJ, Svensson-Arvelund J, Lubitz GS, Marabelle A, et al. Cancer vaccines: The next immunotherapy frontier. Nat. Cancer, 2022; 3: 911926.
  7. Ramirez F, Zambrano A, Hennis R, Holland N, Laksmanaswamy R, Chacon J. Sending a message using mRNA vaccines to target the tumor immune microenvironment. Vaccines, 2023; 11: 1465-1487.
  8. Melief CJ, vanHall T, Arens R, Ossendrop F, van der Burg SH. Therapeutic cancer vaccines. J. Clin. Investing, 2015; 125: 3401-3412.
  9. Baiersdorfer M, Boros G, Muramatsu H, Mahiny A, Vlatkovic I, Sahin U, et al. A facile method for the removal of dsRNA contaminant from in vitro-transcribed mRNA. Mol. Ther. Nucleic Acids, 2019; 15: 26-35.
  10. Weng Y, Li C, Yang T, Hu B, Zhang M, Guo S, et al. The challenge and proposal of mRNA therapeutics landscape. Biotechnol. Adv, 2020; 40: 107534-107538.
  11. Kreiter S, Vormebr M, van de Roemer N, Diken M, Lower M, Diekmann J, et al. Mutant MHC class II epitopes drive therapeutic immune response to cancer. Nature, 2015; 520: 692-696.
  12. Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob JJ, Cowey CL, et al. Overall survival with combined Nivolumab in advanced Melanoma. N. Engl. J. Med, 2017; 377: 1345-1356.
  13. Mittrucher HW, Visekruna A, Huber M. Heterogeneity in the differentiation and function of CD8+ T cell. Arch. Immunol. Ther. Exp, 2014; 62: 449-458.
  14. Gordon SR, Maute RL, Dulken BW, Hutter G. George BM, Mc Craken MN, et al. PD-1 expression by tumor-associated macrophages inhibits phagocytosis and immunity. Nature, 2017; 545: 495-499.
  15. Rusinewska B. Potential in anti-cancer therapy using RNA-overview therapies. J. Ed. Health and sport, 2022; 12: 817-827.
  16. Mei Y, Wang X. RNA modification in mRNA cancer vaccines. Clin. Exp. Med, 2023; 23: 1917-1931.
  17. Waissengrin B, Agbarya A, Safadi E, Padova H, Wolf I. Short-term safety of the BNT162b2 mRNA Covid-19 vaccine in patients with cancer treated with immune checkpoint inhibitors. Lancet Oncol, 2021; 22: 581-583.
  18. Duan LJ, Wang Q, Zhang C, Yang DX, Zhang XY. Potentialities and challenges of mRNA vaccine in cancer immunotherapy. Front. Immunol, 2022; 13: 3389-3396.
  19. Odeh-Natour R, Shapira, M, Estrada D, Freimann S, Tal Y, Atzman Y, et al. Does mRNA SARS-Cov-2 vaccine in the follicular fluid impact follicle and oocyte performance in IVF treatments? Am. J. Reprod. Immunol, 2022; 87: 1111-1117.
  20. Pardi N, Hogan MJ, Porter FW, Weisman D. mRNA vaccine a new era in vaccinology. Nat. Rev. Drug Discov, 2018; 17: 261-279.
  21. Hoeck LV, Saelens X. Therapeutic anti-tumor immunity directed against neoepitopes by intratumor delivery of mRNA encoding MLK. Cell Stress, 2018; 2: 279-286.
  22. Basler L, Kowalczyk A, Heidenreich R, Fontin-Melczeh M, et al. Abscopal effects of radiotherapy and combined and combined mRNA-based immunotherapy in syngeneic ova-expressing Thymoma mouse model. Cancer Immunol. Immunoth, 2018; 64: 653-662.
  23. Barbier AJ, Jiang AY, Zhang P, Wooster R, Anderson DG. The clinical progress of mRNA vaccines and immunotherapies. Nature Biotechnol, 2022; 40: 840-854.
  24. Cafri G, Gartner JJ, Zaks T, Hopson K, Levin N, Paria BC, et al. mRNA vaccine-induced neoantigen-specific T cell immunity in patients with gastrointestinal cancer. J. Clin. Invest, 2020; 130: 5976-5488.
  25. Khansari N. Potential benefits and challenges of mRNA cancer vaccines. J. Clin. Immunol. Microb, 2023; 4: 1-3.
logo

Subscribe to newsletter

© 2020. All rights reserved.

TOP