Skip to main content

EDITORIAL article

Front. Immunol., 08 August 2023
Sec. Molecular Innate Immunity
This article is part of the Research Topic Delivering Nucleic Acids to Immune and Non-Immune Cells View all 16 articles

Editorial: Delivering nucleic acids to immune and non-immune cells

  • 1Nanoscale Science Program, Department of Chemistry, University of North Carolina Charlotte, Charlotte, NC, United States
  • 2Department of Medicine and Surgery, University of Milano Bicocca, Milano, Italy
  • 3Shenzhen Institute of Advanced Technology (SIAT) of the Chinese Academy of Science (CAS), and China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen, China
  • 4Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche, Napoli, Italy
  • 5Stazione Zoologica Anton Dohrn, Napoli, Italy

For decades, there has been ongoing research focused on the targeted delivery of therapeutic DNA and/or RNA molecules designed to regulate the expression of specific genes. This field of study aims to advance new clinical approaches for therapies that target the genetic components of various diseases in a personalized manner. The technology had an impressively rapid advancement and gained global recognition, particularly in the new vaccination strategies adopted during the COVID-19 pandemic (1, 2). However, a major obstacle that still hinders the broader success of nucleic acid-based therapies and vaccines lies in the inefficient delivery of these biopolymers to human cells, as well as in ensuring their subsequent intracellular release and optimal performance. The challenges arise from the inherent limitations of naked, non-modified oligonucleotides, among which are their susceptibility to rapid degradation by nucleases and renal clearance, the inefficient crossing of biological membranes, and the potential to induce uncontrolled immunological responses (3, 4). To optimize the efficacy while minimizing the harmful side effects of nucleic acid cargos, recent research has focused on improving the design of delivery systems suitable for rationally designed DNA and RNA molecules (5). A special emphasis on targeting immune cells is warranted in the case of vaccination. The strategy aims to shuttle the antigen-coding nucleic acids to cells that will be able to synthesize, process, and present the antigens. By doing so, optimally protective immune responses will lead to the development of long-term protective immunological memory.

The current Research Topic features a collection of fifteen review and research articles curated by international leaders in the fields of nucleic acid therapies, immunology, and drug delivery. The manuscripts present a range of innovative technologies encompassing the design of various nucleic acid-based therapeutics, assessment of their biological activities, and optimization of administration conditions.

Antigen-encoding nucleic acids can be optimized for a particular application through several strategies. The review by Vishweshwaraiah and Dokholyan provides a comprehensive assessment of rational design and optimization strategies for mRNA vaccines, highlights different platforms available for vaccine delivery, and discusses the limitations and future challenges associated with this emerging technology.

By encoding appropriate target proteins, such as the Spike protein of SARS-CoV-2, and selecting between the B- and T-cell epitopes, it becomes possible to elicit either antibody or cellular responses. Also, the specification of CD4 and CD8 epitopes can assist in the stimulation of helper and cytotoxic responses, respectively. The research by Del Riego et al. provides an analysis of the levels of SARS-CoV-2 specific antibodies and T cells in intensive care unit workers. The aim is to gain additional insights into their immune protection following vaccination. Additionally, Becker et al. share longitudinal vaccination response data in dialysis patients and control subjects, as part of a mixed mRNA vaccination scheme, and Raptis et al. report differences in immune responses for patients with inflammatory rheumatic diseases, which depended on the type of administered mRNA vaccine. Garcia-Dominguez et al. discover that the dosing intervals in mRNA vaccination could improve the durability of immune responses, with longer intervals being preferable.

The inclusion of chemical modifications and sequence optimization plays a crucial role in defining the stability, cellular localization, and therapeutic efficacy of delivered mRNA (6, 7). These strategies enable fine-tuning mRNA physicochemical properties, enhancing its lifetime, and optimizing its performance as a therapeutic agent. In their research article, Bai et al. present a nucleoside-modified Rabies mRNA-lipid nanoparticle vaccine able to induce prolonged immune responses in animal models, while Rice at al. employ heterologous vaccination to elicit robust cellular immunogenicity with increased protection against the emerging variants of SARS-CoV-2 infection.

Nucleic acid technologies, combined with the innate capacity of the human immune system to detect nucleic acids and initiate efficient immune/inflammatory responses, have paved the way for their potential use as adjuvants and immunostimulants (8). These applications are crucial for enhancing vaccine efficacy. The review by Gu et al. provides a good overview on the relationship between circular RNA and immunity, highlighting the current applications and future directions of these technologies. Consequently, it is imperative to focus research efforts on the design of nucleic acids that can induce controlled inflammation in a limited manner, both temporally and anatomically. For instance, this can be achieved by targeting Toll-like receptors (TLRs) or NOD-like receptors (NLRs). A clinical trial reported by Daniel et al. reveals the first-in-human Phase 1 study of immunostimulatory spherical nucleic acids designed to target TLR9. The results make this compound promising as an immunotherapy agent.

Recent advancements in nucleic acid bioengineering have led to the emergence of nucleic nanoparticles (NANPs), multistranded nanoassemblies composed exclusively of RNA, DNA, and their chemical analogs, which offer an innovative approach for regulated personalized immunostimulation and drug delivery (9, 10). By changing the composition and architectural features of NANPs, it becomes possible to target the activation of specific pattern recognition receptors (e.g., TLR7 or RIG-I) and promote the delivery of scaffolded antigens and therapeutics to the diseased cells (11, 12). Panigaj et al. review recent developments and applications of NANPs, rationally designed for therapeutic immunomodulation, and identified current limitations and future directions of this innovative platform.

Efficient intracellular delivery of nucleic acid therapeutics relies on the use of various carriers, among which are viral vectors, highly efficient but with problems of immunogenicity and specificity of targeting, lipid nanoparticles (LNPs), currently the lead delivery systems for nucleic acid-based vaccines and therapeutics, exosomes, extracellular vesicles, a recent promising delivery and diagnostic technology that is still at early stages of its development, as well as various inorganic nanoparticles and polymers (13, 14). In their review, González-Rioja et al. revise the synthetic methods, physicochemical characterization, and pharmacokinetics of surfactant ionizable lipid nanoparticles loaded with RNA therapeutics. The review articles by Gao et al. and Zhang et al. discuss the application of extracellular vesicles and exosomes as new diagnostic tool and drug delivery carriers for cancer immunotherapies, respectively. Finally, Gusta et al. develop a panel of cationic gold nanoparticle-based nanovectors used for the safe and sustained internalization of mRNAs via endocytosis. The obtained results with cell culture experiments show promise for this technology to be used as a delivery platform for nucleic acids.

With further developments of nucleic acid-based technologies, it is essential to address the potential risks associated not only with therapeutic cargos but also with other formulation components that can lead to excessive inflammation, cytokine storms, and the development of autoimmune and inflammatory diseases (15, 16). The review by Dobrovolskaia shares the valuable insights obtained from the extensive experience of the Nanotechnology Characterization Laboratory regarding the interactions between different nanoparticles and the immune system, which significantly impact the safety and effectiveness of formulations.

In summary, while nucleic acid therapies hold immense promise and have achieved significant advancements, their broader applications still necessitate further improvements, among which are increasing delivery efficiencies, providing storage and handling of all formulations at ambient temperatures, design-driven regulation of immunorecognition and toxicities, and lowering production costs by addressing technological hurdles and logistical challenges (17).

Author contributions

KA, DB, and FR contributed to writing and revision of the manuscript. All authors contributed to the article and approved the submitted version.

Funding

Research reported in this publication was supported by the National Institute of General Medical Sciences of the National Institutes of Health under Award Number R35GM139587 (to KA). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. DB was supported by the EU H2020 project PANDORA (GA 671881) and by the Key Collaborative Research Program of the Alliance of International Science Organizations (ANSO-CR-KP-2022-01). FR was supported by the Centro Nazionale per lo sviluppo di terapia genica e farmaci con tecnologia a RNA, PNRR Ministero dell’Università e della Ricerca of Italy.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Tseng HF, Ackerson BK, Luo Y, Sy LS, Talarico CA, Tian Y, et al. Effectiveness of mRNA-1273 against SARS-CoV-2 Omicron and Delta variants. Nat Med (2022) 28(5):1063–71. doi: 10.1038/s41591-022-01753-y

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Jackson LA, Anderson EJ, Rouphael NG, Roberts PC, Makhene M, Coler RN, et al. An mRNA Vaccine against SARS-CoV-2 - Preliminary Report. N Engl J Med (2020) 383(20):1920–31. doi: 10.1056/NEJMoa2022483

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Kulkarni JA, Witzigmann D, Thomson SB, Chen S, Leavitt BR, Cullis PR, et al. The current landscape of nucleic acid therapeutics. Nat Nanotechnol (2021) 16(6):630–43. doi: 10.1038/s41565-021-00898-0

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Johnson MB, Chandler M, Afonin KA. Nucleic acid nanoparticles (NANPs) as molecular tools to direct desirable and avoid undesirable immunological effects. Adv Drug Deliver Rev (2021) 173:427–38. doi: 10.1016/j.addr.2021.04.011

CrossRef Full Text | Google Scholar

5. Afonin KA, Dobrovolskaia MA, Church G, Bathe M. Opportunities, barriers, and a strategy for overcoming translational challenges to therapeutic nucleic acid nanotechnology. ACS Nano (2020) 14(8):9221–7. doi: 10.1021/acsnano.0c04753

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Leppek K, Byeon GW, Kladwang W, Wayment-Steele HK, Kerr CH, Xu AF, et al. Combinatorial optimization of mRNA structure, stability, and translation for RNA-based therapeutics. Nat Commun (2022) 13(1):1536. doi: 10.1038/s41467-022-28776-w

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Zhang H, Zhang L, Lin A, Xu C, Li Z, Liu K, et al. Algorithm for optimized mRNA design improves stability and immunogenicity. Nature (2023). doi: 10.1038/s41586-023-06127-z

CrossRef Full Text | Google Scholar

8. Pulendran B, SA P, O'Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat Rev Drug Discovery (2021) 20(6):454–75. doi: 10.1038/s41573-021-00163-y

CrossRef Full Text | Google Scholar

9. Chandler M, Jain S, Halman J, Hong E, Dobrovolskaia MA, Zakharov AV, et al. Artificial immune cell, AI-cell, a new tool to predict interferon production by peripheral blood monocytes in response to nucleic acid nanoparticles. Small (2022) 18(46):e2204941. doi: 10.1002/smll.202204941

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Hong E, Halman JR, Shah AB, Khisamutdinov EF, Dobrovolskaia MA, Afonin KA. Structure and composition define immunorecognition of nucleic acid nanoparticles. Nano Lett (2018) 18(7):4309–21. doi: 10.1021/acs.nanolett.8b01283

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Chandler M, Rolband L, Johnson MB, Shi D, Avila YI, Cedrone E, et al. Expanding structural space for immunomodulatory nucleic acid nanoparticles (Nanps) via spatial arrangement of their therapeutic moieties. Adv Funct Mater (2022) 32(43):2205581. doi: 10.1002/adfm.202205581

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Johnson MB, Halman JR, Miller DK, Cooper JS, Khisamutdinov EF, Marriott I, et al. The immunorecognition, subcellular compartmentalization, and physicochemical properties of nucleic acid nanoparticles can be controlled by composition modification. Nucleic Acids Res (2020) 48(20):11785–98. doi: 10.1093/nar/gkaa908

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Ke WN, Afonin KA. Exosomes as natural delivery carriers for programmable therapeutic nucleic acid nanoparticles (NANPs). Adv Drug Deliver Rev (2021) 176:113835. doi: 10.1016/j.addr.2021.113835

CrossRef Full Text | Google Scholar

14. Mendes BB, Conniot J, Avital A, Yao D, Jiang X, Zhou X, et al. Nanodelivery of nucleic acids. Nat Rev Methods Primers (2022) 2:24. doi: 10.1038/s43586-022-00104-y

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Dobrovolskaia MA, Afonin KA. Use of human peripheral blood mononuclear cells to define immunological properties of nucleic acid nanoparticles. Nat Protoc (2020) 15(11):3678–98. doi: 10.1038/s41596-020-0393-6

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Dobrovolskaia MA, McNeil SE. Strategy for selecting nanotechnology carriers to overcome immunological and hematological toxicities challenging clinical translation of nucleic acid-based therapeutics. Expert Opin Drug Delivery (2015) 12(7):1163–75. doi: 10.1517/17425247.2015.1042857

CrossRef Full Text | Google Scholar

17. Afonin KA, Dobrovolskaia MA, Ke WN, Grodzinski P, Bathe M. Critical review of nucleic acid nanotechnology to identify gaps and inform a strategy for accelerated clinical translation. Adv Drug Deliver Rev (2022) 181:114081. doi: 10.1016/j.addr.2021.114081

CrossRef Full Text | Google Scholar

Keywords: delivery, nucleic acids, vaccination, antigen presentation, nanoparticles, NANPs

Citation: Afonin KA, Re F and Boraschi D (2023) Editorial: Delivering nucleic acids to immune and non-immune cells. Front. Immunol. 14:1237506. doi: 10.3389/fimmu.2023.1237506

Received: 09 June 2023; Accepted: 28 July 2023;
Published: 08 August 2023.

Edited and Reviewed by:

Bernd Lepenies, University of Veterinary Medicine Hannover, Germany

Copyright © 2023 Afonin, Re and Boraschi. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Kirill A. Afonin, kafonin@uncc.edu; Francesca Re, francesca.re1@unimib.it; Diana Boraschi, diana.boraschi@itb.cnr.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.