Elsevier

Drug Discovery Today

Volume 25, Issue 3, March 2020, Pages 552-560
Drug Discovery Today

Review
Gene to screen
Comparison of DNA and mRNA vaccines against cancer

https://doi.org/10.1016/j.drudis.2019.12.003Get rights and content

Highlights

Nucleic acid vaccines (NAVs) have recently been tested as a cancer therapy. DNA and mRNA vaccines deliver genetic information encoding tumor antigens (TAs) to the host, which then produces immune responses against cancer cells that express the TAs. Although NAVs are easy, safe, and simple to manufacture, they have not so far been considered viable alternatives to peptide vaccines. Choosing the right TAs, insufficient immunogenicity, and the immunosuppressive nature of cancer are some challenges to this approach. In this review, we discuss approaches that been used to improve the efficiency of anticancer NAVs.

Introduction

Cancer remains a challenging medical problem affecting millions of people around the world. Treatment strategies, such as surgery, chemotherapy, radiotherapy, and hormonal therapies, are applied alone or in combinations. More recently, targeted drugs and immunotherapy have gained research attention. Nevertheless, innovative cancer treatments are still being investigated in ever-increasing numbers [1]. The immune system has the potential to fight cancer and, thus, immunotherapy is designed to educate the immune system to identify and eliminate tumors; as a result, it has fewer adverse effects compared with chemotherapy [2]. Cancer immunotherapy strategies, such as cancer vaccines, bispecific antibodies, chimeric antigen receptors (CAR) T cells, checkpoint inhibitors, and other cell-based therapies, are important platforms 2, 3. Conventional vaccines based on live attenuated and inactivated pathogens, synthetic peptides, and recombinant subunit vaccines are widely used to prevent many infectious diseases. However, the manufacturing procedures of vaccines are not completely safe, and have a high risk of contamination with living pathogens. Therefore, the development of alternative vaccines is necessary for both infectious diseases and for non-infectious diseases, such as cancer 4, 5. Cancer vaccines have been studied for decades with some sporadic success, but have yet to penetrate the oncological mainstream. They include peptide vaccines, cell-based vaccines, viral vector vaccines, and NAVs. All these vaccines are designed to trigger or augment an immune response toward antigens expressed more or less specifically on tumor cells [6]. Among the different types of cancer vaccine tested, NAVs, such as DNA or mRNA vaccines, have been considered attractive because of their safe, simple, and rapid manufacturing process [7]. Cancer vaccines are more often used as a therapeutic approach, compared with infectious diseases, where prophylactic vaccines are more common. Cancer vaccines are designed to induce an immune response against tumor-derived antigens or TAs [8]. TAs can have a central role in tumor initiation, progression, and metastasis 6, 8. They can include tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs). Oncofetal antigens, cancer-testis antigens (CTAs), and overexpressed self-antigens have been considered to be TAAs. CTAs and oncofetal antigens are considered to be good candidates for cancer immunotherapy because they display zero or low expression in normal adult somatic cells; moreover, they are shared by many cancerous tumors in different patients to various extents, and are also expressed in different pathological types of epithelial tumor [9]. By contrast, mutated self-antigens or TSAs/neoantigens require expensive and laborious identification in the tumors of individual patients, but have shown improved efficacy in clinical trials compared with TAAs. Generally speaking, TAAs have been more often studied than TSAs in NAVs used for cancer [10].

In this review, we discuss current approaches using NAVs for cancer, compare DNA and mRNA cancer vaccines (Box 1 provides an introduction to DNA and mRNA cancer vaccines), summarize the latest challenges and recent successes, and offer perspectives for the future application of NAVs in cancer therapy.

Section snippets

Cellular processing and delivery methods

The first crucial step for the success of NAVs is their internalization into the cytoplasm and nucleus (if necessary) of the host cells, especially dendritic cells (DCs), which are the most important type of antigen-presenting cell (APC). Improved delivery systems for nucleic acids have been designed to enhance the efficiency of gene therapy and also NAVs [11]. There are two general delivery approaches for NAVs: in vivo delivery and ex vivo delivery. The first approach involves administering

Recent clinical trials of anticancer NAVs

Over the past few years, there have been several clinical trials for cancer (mostly Phase I/II) using both DNA and mRNA vaccines. Cancer DNA vaccines have mostly been applied in cervical, prostate, and breast cancer in clinical studies. By contrast, melanoma, glioblastoma, and prostate cancer have been the most frequent cancers for which mRNA vaccines have been tested. The application of immunotherapy and endocrine therapy in combination with anticancer NAVs, as well as adjuvants and

Future directions and conclusions

Despite many ongoing efforts to optimize cancer NAVs, researchers still need to deal with many challenges to provide fully effective NAVs for cancer immunotherapy; however, with sufficient time, they might be able to solve all of them. Suggested reasons for the lack of convincing evidence of benefit gained by using current NAVs are as follows. First, unclear understanding of the biology of cancer cells makes it difficult to identify TAs that can engender a powerful immune response, and deeper

Concluding remarks

Taken together, more extensive studies need to be carried out that concentrate on molecular and cellular aspects, to translate preclinical research successes of NAVs into clinical trials that will provide efficient therapy. For instance, the use of small-molecule targeting of inflammatory signaling cascades (especially in the case of mRNA vaccines), better selection of immunogenic TAs, improvement of delivery systems, and choice of suitable combination therapies will be needed to ensure the

Conflicts of interest

M.R.H. declares the following potential conflicts of interest. Scientific Advisory Boards: Transdermal Cap Inc; BeWell Global Inc.; Hologenix Inc.; LumiThera Inc.; Vielight; Bright Photomedicine; Quantum Dynamics LLC; Global Photon Inc.; Medical Coherence; NeuroThera; JOOVV Inc.; AIRx Medical; FIR Industries, Inc.; UVLRx Therapeutics; Ultralux UV Inc.; Illumiheal & Petthera; MB Lasertherapy; ARRC LED; Varuna Biomedical Corp.; and Niraxx Light Therapeutics, Inc. Consulting: Lexington Int.; USHIO

Acknowledgments

M.R.H. was supported by US NIH Grants R01AI050875 and R21AI121700. The authors are thankful for the support of the Immunology Research Center, Tabriz University of Medical Science.

References (61)

  • J.B. Ulmer

    RNA-based vaccines

    Vaccine

    (2012)
  • C. Chakraborty

    The novel strategies for next-generation cancer treatment: miRNA combined with chemotherapeutic agents for the treatment of cancer

    Oncotarget

    (2018)
  • C.L. Ventola

    Cancer immunotherapy, part 1: current strategies and agents

    P. T.

    (2017)
  • A. Lopes

    Cancer DNA vaccines: current preclinical and clinical developments and future perspectives

    J. Exp. Clin. Cancer Res.

    (2019)
  • M.A. McNamara

    RNA-based vaccines in cancer immunotherapy

    J. Immunol. Res.

    (2015)
  • M. Tagliamonte

    Antigen-specific vaccines for cancer treatment

    Hum. Vaccines Immunother.

    (2014)
  • R. Thomas

    NY-ESO-1 based immunotherapy of cancer: current perspectives

    Front. Immunol.

    (2018)
  • S. Wagner

    Colorectal cancer vaccines: tumor-associated antigens vs neoantigens

    World J. Gastroenterol.

    (2018)
  • H. Zhang

    Enhanced gene transfection efficiency by low-dose 25 kDa polyethylenimine by the assistance of 1.8 kDa polyethylenimine

    Drug Deliv.

    (2018)
  • A. Terbuch et al.

    Next generation cancer vaccines–make it personal!

    Vaccines

    (2018)
  • P.M. Santos et al.

    Dendritic cell-based cancer vaccines

    J. Immunol.

    (2018)
  • K.E. Broderick et al.

    Electroporation-enhanced delivery of nucleic acid vaccines

    Expert Rev. Vaccines

    (2015)
  • N. Pardi

    mRNA vaccines—a new era in vaccinology

    Nat. Rev. Drug Discov.

    (2018)
  • A. Mokhtarzadeh

    P53-Derived peptides conjugation to PEI: an approach to producing versatile and highly efficient targeted gene delivery carriers into cancer cells

    Expert Opin. Drug Deliv.

    (2016)
  • F. Soltani

    Synthetic and biological vesicular nano-carriers designed for gene delivery

    Curr. Pharm. Des.

    (2015)
  • C. Grunwitz et al.

    mRNA cancer vaccines—messages that prevail

    Curr. Top. Microbiol. Immunol.

    (2017)
  • D.J. Irvine

    Synthetic nanoparticles for vaccines and immunotherapy

    Chem. Rev.

    (2015)
  • J.J. Suschak

    Advancements in DNA vaccine vectors, non-mechanical delivery methods, and molecular adjuvants to increase immunogenicity

    Hum. Vaccines Immunother.

    (2017)
  • S. Rauch

    New vaccine technologies to combat outbreak situations

    Front. Immunol.

    (2018)
  • M.A. Liu

    A comparison of plasmid DNA and mRNA as vaccine technologies

    Vaccines

    (2019)
  • Cited by (0)

    View full text