Skip to main content

Clinical development of mRNA therapies against solid tumors

Abstract

The mRNA-based therapeutics have become the hot spot of biopharmaceutical industries in recent years. The landscape of this area is expanding from infectious disease to cancer, which needs to be summarized to provide data supports for industries and research institutions. Based on the Trialtrove database, a total of 108 clinical trials from 1999 to 2021 were retrospectively analyzed. We have demonstrated that the clinical development of mRNA therapies against solid tumors is still at an early stage. There are evolutions in delivery systems from the dendritic cell to the lipid-based platform and in encoding strategies from the fixed tumor antigens to the personalized neoantigens. The adjuvant or maintenance therapy and the combination treatment with checkpoint inhibitors are becoming the major clinical development orientation.

To the editor

Since the clinical application of COVID-19 vaccines, messenger RNA (mRNA)-based therapeutics have become the hot spot of biopharmaceutical industries in recent years [1]. With the advantages of personalized preparation, fast production and good immunogenicity [1,2,3], therapeutic areas of mRNA are expanding to cancer. Dozens of clinical trials have been launched with preliminary results [1,2,3]. However, there is no evidence of data on the panorama worldwide. Here, we summarize the current progress, put forward suggestions for future clinical development and provide data supports for industries and research institutions.

Based on the Trialtrove database [4], a total of 108 clinical trials for mRNA therapies against solid tumors were identified worldwide by the cut-off date December 31, 2021 (Additional file 1: Fig. S1). Most of the trials were at phase I (79, 73.1%) (Table 1). The exploratory investigator-initiated trials (IITs) accounted for a large proportion (49, 45.4%), and the sponsors were highly concentrated in a few countries (Additional file 1: Table S1), which suggested that the clinical development in this area was still at its early stage. As a result, many challenges and uncertainties have been raised, including how to optimize the delivery systems and encoded proteins of mRNA sequences and how to select clinical scenario.

Table 1 Distribution of cancer types by study phase of clinical trials for anticancer mRNA therapeutics

A total of 57 mRNA agents were further identified. Analyzing the delivery systems, the lipid-based platform, namely lipid nanoparticle (LNP) (13, 22.8%), lipoplex (LPX) (8, 14.0%) and lipopolyplex (LPP) (6, 10.5%), in aggregate accounted for the largest proportion (27, 47.4%) (Fig. 1, Additional file 1: Table S2). The dendritic cell (DC) was the main platform for drugs developed from 1999 to 2014 (15/25, 60.0%) (Fig. 1), but the ex vivo manipulation of DCs is laborious and time-consuming, with an unsatisfied mRNA transfection rate [1, 2, 5]. Compared with DCs, the lipid-based delivery systems are capable of rapid manufacture, with high plasticity, transfection rate and immunogenicity [6]. Thus, this technology has become the mainstream since 2015 (27/30, 90.0%) (Fig. 1). Given the complexity of the molecular structure, further optimizing the manufacturing process and reducing the unpredictable clinical effects will be the future direction of the lipid-based platform.

Fig. 1
figure 1

Annual numbers of newly tested mRNA therapeutics by delivery system

The tumor antigen is the most common coding category of mRNA therapies. There were 27 (47.4%), 8 (14.0%) and 5 (8.8%) agents encoding fixed tumor antigens, neoantigens and autologous tumor cell antigens, respectively (Additional file 1: Fig. S2a). MAGE-A1/A3 (6, 10.5%), survivin (6, 10.5%) and tyrosinase (5, 8.8%) were the most common specific encoded proteins (Additional file 1: Fig. S2b), which in line with the features of the most extensively studied cancer types, such as melanoma [7] (Table 1). However, due to the insufficient immunogenicity and the space–time heterogeneity of classic tumor antigens [8], mRNA agents encoding fixed, single antigens have not shown definite clinical benefit yet (Additional file 1: Table S3).

The personalized neoantigen-based therapies are expected to overcome the obstacles. These highly individualized and flexible products perfectly fit the fast manufacture of mRNA, with the development of bioinformatics technology [8]. Since 2009, the number of newly initiated trials for neoantigen-based agents has shown a trend to exceed that of fixed antigens (Additional file 1: Fig. S3). The representatives, mRNA-4157 and BNT-122, have already achieved promising short-term efficacy (Additional file 1: Table S3), though the reliability of the neoantigen prediction system and the encoding capacity of the mRNA sequences for multiple antigens still need to be investigated [9].

Among the 108 trials, there were 32 (29.6%) for adjuvant or maintenance therapy and 59 (54.6%) involving combination treatment, where the immunotherapy was the most prevalent combination strategies (44/59, 74.8%) (Additional file 1: Table S4). The above proportions are relatively high compared with the entire landscape of immuno-oncology [10, 11], which infer that mRNA agents may act as an “assistant” rather than the “backbone” in the cancer treatment. In addition, referring to the rationale of immune checkpoint inhibitors (ICIs), mRNA therapies encoding tumor antigens are more likely to play a greater role in the earlier stage of cancer or advanced stage with lower tumor burden and produce greater synergy with ICIs [12]. Therefore, adjuvant and combination therapies are the trends in the clinical scenarios for mRNA therapies.

In conclusion, the clinical development of mRNA therapies against solid tumors is still at an early stage. The notable shifts in delivery systems from the DC to the lipid-based platform and in encoding strategies from the fixed tumor antigens to the personalized neoantigens together mark a new era in this field. The adjuvant or maintenance therapy and the combination treatment with ICIs are becoming the important clinical development orientation.

Availability of data and materials

All the source data in this work are based on the Trialtrove database, with clinical trial details derived from clinical trial publicity platforms. The datasets used and analyzed during the study are available from the corresponding author on reasonable request.

Abbreviations

mRNA:

Messenger RNA

IIT:

Investigator-initiated trial

LNP:

Lipid nanoparticle

LPX:

Lipoplex

LPP:

Lipopolyplex

DC:

Dendritic cell

ICI:

Immune checkpoint inhibitor

References

  1. Beck JD, Reidenbach D, Salomon N, et al. mRNA therapeutics in cancer immunotherapy. Mol Cancer. 2021;20(1):69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Lorentzen CL, Haanen JB, Met Ö, et al. Clinical advances and ongoing trials on mRNA vaccines for cancer treatment. Lancet Oncol. 2022;23(10):e450–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Miao L, Zhang Y, Huang L. mRNA vaccine for cancer immunotherapy. Mol Cancer. 2021;20(1):41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. https://pharmaintelligence.informa.com/products-and-services/data-and-analysis/trialtrove. Accessed March 8, 2023

  5. Xu S, Yang K, Li R, et al. mRNA vaccine era-mechanisms, drug platform and clinical prospection. Int J Mol Sci. 2020;21(18):6582.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Igyártó BZ, Jacobsen S, Ndeupen S. Future considerations for the mRNA-lipid nanoparticle vaccine platform. Curr Opin Virol. 2021;48:65–72.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Li S, Shi X, Li J, et al. Pathogenicity of the MAGE family. Oncol Lett. 2021;22(6):844.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Blass E, Ott PA. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat Rev Clin Oncol. 2021;18(4):215–29.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Lang F, Schrörs B, Löwer M, et al. Identification of neoantigens for individualized therapeutic cancer vaccines. Nat Rev Drug Discov. 2022;21(4):261–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Xin YuJ, Hodge JP, Oliva C, et al. Trends in clinical development for PD-1/PD-L1 inhibitors. Nat Rev Drug Discov. 2020;19(3):163–4.

    Article  Google Scholar 

  11. Wu DW, Huang HY, Tang Y, et al. Clinical development of immuno-oncology in China. Lancet Oncol. 2020;21(8):1013–6.

    Article  PubMed  Google Scholar 

  12. Wu DW, Huang HY, Zhang MH, et al. The global landscape of neoadjuvant and adjuvant anti-PD-1/PD-L1 clinical trials. J Hematol Oncol. 2022;15(1):16.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the Chinese Academy of Medical Sciences (CAMS) Innovation Fund for Medical Sciences (CIFMS) (2021-I2M-1-012) and Digital Clinical Trial Training Program (76431397).

Author information

Authors and Affiliations

Authors

Contributions

WDW, HLF, WX and YY contributed to framework planning and draft writing, as well as data quality control, analysis and interpretation. JSP, HHY and LZW participated in data quality control and analysis. MJF, ZHB and TY participated in framework planning and contributed to data interpretation. NL led the overall framework planning and data interpretation. All the authors reviewed and approved the final manuscript.

Corresponding author

Correspondence to Ning Li.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

The content of the manuscript has not been previously published and is not under consideration for publication elsewhere.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1:

Data processing details and additional results.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, D., Hu, L., Wang, X. et al. Clinical development of mRNA therapies against solid tumors. J Hematol Oncol 16, 75 (2023). https://doi.org/10.1186/s13045-023-01457-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13045-023-01457-x

Keywords