Next Article in Journal
Unseen Weapons: Bacterial Extracellular Vesicles and the Spread of Antibiotic Resistance in Aquatic Environments
Previous Article in Journal
Oxidative Stress-Mediated Repression of Virulence Gene Transcription and Biofilm Formation as Antibacterial Action of Cinnamomum burmannii Essential Oil on Staphylococcus aureus
Previous Article in Special Issue
Specific Activation of T Cells by an ACE2-Based CAR-Like Receptor upon Recognition of SARS-CoV-2 Spike Protein
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Advanced Therapy Medicinal Products as Potential Therapeutic Strategy against COVID-19 and Immune-Related Disorders

by
Panagiotis Mallis
1,2,*,
Efstathios Michalopoulos
1 and
Catherine Stavropoulos-Giokas
1
1
Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens (BRFAA), 115 27 Athens, Greece
2
Immunology Department & National Tissue Typing Center, General Hospital of Athens “Gennimatas”, 154 Mesogeion Ave., 115 27 Athens, Greece
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(5), 3079; https://doi.org/10.3390/ijms25053079
Submission received: 28 February 2024 / Accepted: 2 March 2024 / Published: 6 March 2024
Advanced Therapy Medicinal Products (ATMPs) comprise a heterogenous class of innovative medicinal products, which further require extensive preclinical and clinical assessments before their broader use in the general population [1]. ATMPs can be either recombinant nucleic acids or engineered cells and tissues [1,2]. Therefore, ATMPs can be classified into four subcategories, including (1) somatic cell therapy medicinal products (SCTMPs), tissue-engineered products (TEPs), gene therapy medicinal products (GTMPs), and combined ATMPs (cATMPs) [3]. The European Union (EU) has a specific legal framework, which, among others, also clearly indicates the differentiation between cell-based products and cell-based therapies. In this context, if human tissues/cells when minimally manipulated, including in vitro processes that do not alter their relevant biological characteristics, such as (a) centrifugation, (b) trimming, (c) flushing, (d) refrigeration, (e) freezing, (f) freeze drying, (g) the use of additives (e.g., cryopreservatives, anticoagulants, antimicrobial agents), (g) or irradiation, are not considered as ATMPs, the active substance cannot be commercialized or manufactured on an industrial scale [3,4]. Therefore, the proper classification of ATMPs defines the regulatory framework further, where the requirements of development and marketing are also included. The EU regulation 1394/2007/EC, in combination with the Directive 2009/120/EU, provides the overall framework and also the updated definitions and detailed scientific/technical requirements of ATMPs [1,2,3,4].
In the context of the authorization and release of ATMPs in the EU, the application of the clinical trial must be prepared and submitted to the national competent authority [3]. Additionally, the Committee for Advanced Therapies (CATs) and the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA) are responsible for the scientific evaluation and approval of ATMPs. These committees are dedicated to the assurance of the safety, quality, and efficacy of ATMPs, thus performing the draft opinion and the final decision for the release of the ATMPs.
The first released ATMP in the EU was performed in 2009 and constituted a tissue-engineered product known as ChondroCelect®, which is intended as a therapeutic treatment of cartilage and bone defects [2]. Accordingly, in the United States (US), PROVENGE®, a somatic cell therapy for prostate cancer treatment, was approved as ATMP and released in 2010 [2]. In the field of gene therapy, Glyberia® was the first ATMP of this category that was released in the EU in 2012, which was designed to reverse lipoprotein lipase deficiency (LPLD) in patients suffering from severe pancreatitis.
Providing the main framework where the ATMPs can act beneficially in human disorders, recent applications of the above can also be found in COVID-19 and immune-related diseases. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the responsible agent of COVID-19, which was initiated in December 2019 and further escalated into a global pandemic [5]. Based on the World Health Organization (WHO), more than 774 million cases and more than 8.6 million deaths have been reported worldwide. Currently, COVID-19 has spread to more than 220 countries, with the US acting as a leader for new SARS-CoV-2 cases (with more than 103,400,000 confirmed cases) [6]. Moreover, SARS-CoV-2, from its first appearance until today, has undergone a great number of genome mutations, leading to the identification of new variants and subvariants such as Beta, Alpha, and Omicron (BA.2.86, XBB.1.9.1, XBB.2.3, CH.1.1, etc.), accompanied by differences in disease transmission and pathogenesis [7]. Among the different variants, SARS-CoV-2 can induce severe immunopathogenic damage, especially in individuals with immune system defects, elderly populations, or individuals with underlying disorders [7]. It is well known that the infected immune cells, such as macrophages and monocytes by SARS-CoV-2, may initiate the cytokine release syndrome (CRS) [8]. Damaged or infected cells also release damage-associated molecular patterns (DAMPs) and inflammatory cytokines such as interleukin 1β (ΙL-1β), tumor necrosis factor-A (TNF-A), IL-3, and IL-6. This further leads to the overactivation of other immune cells such as dendric cells (DCSs), T helper (Th) 1, Th2, Th17, natural killer (NK) cells and B cells, which produce a great number of inflammatory molecules, e.g., IL-1B, IL-1RA, IL-7, IL-8, IL-9, TNF-A, the fibroblast growth factor (FGF), transforming growth factor-β1 (TGF-β1), granulocyte–macrophage colony-stimulating factor (GM-CSF), IFN-γ, G-CSF, IP10, MCP1, MIP1A, platelet-derived growth factor (PDGF), TNF-α, and vascular endothelial growth factor (VEGF) [8]. In turn, SARS-CoV-2 can spread to the lung parenchyma, infecting, and damaging the alveolar epithelial cells and constituting the primary cause of severe acute respiratory syndrome [8]. The above events lead to a very serious situation, which needs to be properly administrated in order to prevent loss of life.
Moreover, the comprehensive study of SARS-CoV-2 led the scientific society to conclude that common pathogenetic characteristics are also found in other immune-related human disorders, such as autoimmune diseases. Moreover, individual’s genetic background plays a significant role in the underlining resistance against infectious diseases. Specifically, the COVID Human Genetic Effort (http://www.COVIDhge.com, accessed on 25 February 2024) has already reported that 15% of critically ill COVID-19 patients are actively associated with inborn errors of immunity (IEIs) of IFN type I, Toll-like receptor 3 (TLR3) and interferon regulatory factor (IRF) 7 [8]. In addition, more than 10% of critically ill COVID-19 patients were characterized by neutralizing antibodies against interferons, cytokines, and signal-inducing molecules favoring immune responses [8].
Recently, besides the already established protocols, ATMPs have also been considered as candidates for a therapeutic approach, especially for severely ill COVID-19 patients admitted to ICUs. ATMPs can utilize the mesenchymal stromal cells (with known immunoregulatory/immunomodulatory properties) and chimeric antigen receptor (CAR) T and NK cells, for the better administration of COVID-19. To date, the US Food and Drug Administration (FDA) has granted clearance for the performance of phase I/II clinical trials for investigational COVID-19 therapy utilizing allogeneic MSCs [8].
To properly attenuate SARS-CoV-2’s immunopathogenic defects, Gonzalez-Garcia et al. [9] reported the development of CAR-like constructs, which can recognize the ACE2 viral receptor that is responsible for virus entry to the host cells. These CAR-T cells can be obtained after the specific manipulation of patients’ cells and, thus, can be used to prevent the entry of SARS-CoV-2 and also its associated variants despite the genome mutations that it could bear. In the same line, Nova et al. [10] showed the efficient large-scale production of SARS-CoV-2-specific CD4+ and CD8+ T cells. Specifically, antigenic T-cells prepared by the IFN-γ cytokine capture system (CCS) have been classified as ATMPs, authorized for intended use by the EMA, and are currently used in critically ill SARS-CoV-2 patients. Another study conducted by Piplani et al. [11] and Barakat et al. [12] showed promising evidence regarding the design and synthesis of modern compounds targeting either the ACE2 receptor or the replication machinery of SARS-CoV-2. Moreover, Lee et al. [13] reported the beneficial role of compounds that can downregulate specific signaling pathways, such as the sterile alpha motif, histidine-aspartate domain-containing protein 1 (SAMHD1) tetramerization and the cytosolic DNA sensor cyclic-GMP–AMP synthase (cGAS), as a stimulator of interferon gene (STING) signaling pathways. The above pathways and the involved molecules play crucial roles in SARS-CoV-2 pathogenesis by inducing CRS; therefore, their downregulation may act beneficially in critically ill patients. In the context of developing advanced ATMPs and mediated therapeutic strategies, Khan et al. [14] utilized Cheminformatics to discover potential probe inhibitors of the Omicron subvariant spike protein. The designed probes showed intriguing results in preventing viral entry; therefore, further in vitro and in vivo studies could be performed to more thoroughly assess the efficacy of these compounds against other SARS-CoV-2 subvariants.
In conclusion, ATMPs may represent valuable therapeutic strategies primarily for severely deceased patients. Currently, ATMPs are also applied to immune-related disorders and critically ill COVID-19 patients with beneficial effects. The accomplishment of efforts focused on the production of safe ATMPs may furnish new perspectives on the development of modern therapeutic strategies with a greater immunogenic impact against human disorders, thus providing more options to clinicians.

Author Contributions

Conceptualization, P.M.; writing—original draft, P.M.; writing—review and editing, P.M., E.M. and C.S.-G.; supervision, P.M. and E.M.; Project Administration, P.M. and E.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The guest editors of the current Special Issue want to express their gratitude to all contributors for their unique and outstanding articles. Additionally, special credit should be given to all reviewers for their comprehensive analysis and their overall effort in improving the quality of the published articles.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Gonçalves, E. Advanced Therapy Medicinal Products: Value Judgement and Ethical Evaluation in Health Technology Assessment. Eur. J. Health Econ. 2020, 21, 311–320. [Google Scholar] [CrossRef] [PubMed]
  2. Jönsson, B.; Grace, H.; Michaels, J.; Towse, A.; Graf Von Der Schulenburg, J.-M.; Olivier, W. Advanced Therapy Medicinal Products and Health Technology Assessment Principles and Practices for Value-Based and Sustainable Healthcare. Eur. J. Health Econ. 2019, 20, 427–438. [Google Scholar] [CrossRef] [PubMed]
  3. Iglesias-Lopez, C.; Agustí, A.; Obach, M.; Vallano, A. Regulatory Framework for Advanced Therapy Medicinal Products in Europe and United States. Front. Pharmacol. 2019, 10, 921. [Google Scholar] [CrossRef] [PubMed]
  4. FDA; CBER. Minimal Manipulation of Human Cells, Tissues, and Cellular and Tissue-Based Products Draft Guidance for Industry and Food and Drug Administration Staff Contains Nonbinding Recommendations Draft-Not for Implementation; Center for Biologics Evaluation and Research: Silver Spring, MD, USA, 2014. [Google Scholar]
  5. Kirtipal, N.; Bharadwaj, S.; Kang, S.G. From SARS to SARS-CoV-2, Insights on Structure, Pathogenicity and Immunity Aspects of Pandemic Human Coronaviruses. Infect. Genet. Evol. 2020, 85, 104502. [Google Scholar] [CrossRef] [PubMed]
  6. COVID-19 Cases|WHO COVID-19 Dashboard. Available online: https://data.who.int/dashboards/covid19/cases?n=c (accessed on 28 February 2024).
  7. Harvey, W.T.; Carabelli, A.M.; Jackson, B.; Gupta, R.K.; Thomson, E.C.; Harrison, E.M.; Ludden, C.; Reeve, R.; Rambaut, A.; Peacock, S.J.; et al. SARS-CoV-2 Variants, Spike Mutations and Immune Escape. Nat. Rev. Microbiol. 2021, 19, 409–424. [Google Scholar] [CrossRef] [PubMed]
  8. Mallis, P. Exploring the Immunomodulatory Properties of Stem Cells in Combating COVID-19: Can We Expect More? Bioengineering 2023, 10, 803. [Google Scholar] [CrossRef] [PubMed]
  9. Gonzalez-Garcia, P.; Muñoz-Miranda, J.P.; Fernandez-Cisnal, R.; Olvera, L.; Moares, N.; Gabucio, A.; Fernandez-Ponce, C.; Garcia-Cozar, F. Specific Activation of T Cells by an ACE2-Based CAR-Like Receptor upon Recognition of SARS-CoV-2 Spike Protein. Int. J. Mol. Sci. 2023, 24, 7641. [Google Scholar] [CrossRef] [PubMed]
  10. Nova, Z.; Zemanek, T.; Botek, N. Antigen-Specific T Cells and SARS-CoV-2 Infection: Current Approaches and Future Possibilities. Int. J. Mol. Sci. 2022, 23, 15122. [Google Scholar] [CrossRef] [PubMed]
  11. Piplani, S.; Singh, P.; Petrovsky, N.; Winkler, D.A. Identifying SARS-CoV-2 Drugs Binding to the Spike Fatty Acid Binding Pocket Using In Silico Docking and Molecular Dynamics. Int. J. Mol. Sci. 2023, 24, 4192. [Google Scholar] [CrossRef] [PubMed]
  12. Barakat, A.; Mostafa, A.; Ali, M.; Al-Majid, A.M.; Domingo, L.R.; Kutkat, O.; Moatasim, Y.; Zia, K.; Ul-Haq, Z.; Elshaier, Y.A.M.M. Design, Synthesis and In Vitro Evaluation of Spirooxindole-Based Phenylsulfonyl Moiety as a Candidate Anti-SAR-CoV-2 and MERS-CoV-2 with the Implementation of Combination Studies. Int. J. Mol. Sci. 2022, 23, 11861. [Google Scholar] [CrossRef] [PubMed]
  13. Lee, J.H.; Kanwar, B.; Khattak, A.; Balentine, J.; Nguyen, N.H.; Kast, R.E.; Lee, C.J.; Bourbeau, J.; Altschuler, E.L.; Sergi, C.M.; et al. COVID-19 Molecular Pathophysiology: Acetylation of Repurposing Drugs. Int. J. Mol. Sci. 2022, 23, 13260. [Google Scholar] [CrossRef] [PubMed]
  14. Khan, S.A.; Khan, A.; Zia, K.; Shawish, I.; Barakat, A.; Ul-Haq, Z. Cheminformatics-Based Discovery of Potential Chemical Probe Inhibitors of Omicron Spike Protein. Int. J. Mol. Sci. 2022, 23, 10315. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mallis, P.; Michalopoulos, E.; Stavropoulos-Giokas, C. Advanced Therapy Medicinal Products as Potential Therapeutic Strategy against COVID-19 and Immune-Related Disorders. Int. J. Mol. Sci. 2024, 25, 3079. https://doi.org/10.3390/ijms25053079

AMA Style

Mallis P, Michalopoulos E, Stavropoulos-Giokas C. Advanced Therapy Medicinal Products as Potential Therapeutic Strategy against COVID-19 and Immune-Related Disorders. International Journal of Molecular Sciences. 2024; 25(5):3079. https://doi.org/10.3390/ijms25053079

Chicago/Turabian Style

Mallis, Panagiotis, Efstathios Michalopoulos, and Catherine Stavropoulos-Giokas. 2024. "Advanced Therapy Medicinal Products as Potential Therapeutic Strategy against COVID-19 and Immune-Related Disorders" International Journal of Molecular Sciences 25, no. 5: 3079. https://doi.org/10.3390/ijms25053079

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop