Next Article in Journal
Silver Nanoparticles Synthesized from Enicostemma littorale Exhibit Gut Tight Junction Restoration and Hepatoprotective Activity via Regulation of the Inflammatory Pathway
Previous Article in Journal
Inhalable Nanotechnology-Based Drug Delivery Systems for the Treatment of Inflammatory Lung Diseases
Previous Article in Special Issue
Synergistic Effect of Conditioned Medium from Amniotic Membrane Mesenchymal Stromal Cells Combined with Paclitaxel on Ovarian Cancer Cell Viability and Migration in 2D and 3D In Vitro Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

An Editorial on the Special Issue “Where Are We Now and Where Is Cell Therapy Headed?”

1
Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
2
Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2025, 17(7), 894; https://doi.org/10.3390/pharmaceutics17070894
Submission received: 2 July 2025 / Accepted: 7 July 2025 / Published: 9 July 2025
(This article belongs to the Special Issue Where Are We Now and Where Is Cell Therapy Headed?)
Cell-based therapies have swiftly transitioned from experimental modalities to core components of modern translational medicine. Initially envisioned as a regenerative tool relying on engraftment and tissue integration, cell therapy has undergone a conceptual shift, and it is becoming increasingly evident that its therapeutic impact is rooted not in cellular persistence, but in the transient orchestration of complex biological responses.
A paradigmatic case is the evolution of mesenchymal stromal cells (MSCs), which have historically been employed for tissue regeneration but are now recognized for their immunomodulatory and paracrine effects [10.1002/sctm.17-0051]. The growing understanding of MSCs as secretory platforms has accelerated interest in cell-free therapeutic strategies, including extracellular vesicles (EVs) and complex secretomes [1,2,3]. These acellular approaches may mitigate issues related to immune rejection and engraftment failure while offering greater control and standardization.
Nonetheless, certain cell-based interventions remain clinically indispensable. Allogeneic hematopoietic cell transplantation (HCT), for example, is still a foundational therapy for hematological malignancies such as leukemia, lymphoma, and multiple myeloma [4,5]. At the same time, challenges such as graft versus host disease (GvHD) and donor availability have prompted the development of precision genome-editing approaches—most notably, CRISPR-Cas9—to reduce immunogenicity and correct autologous mutations [6].
In oncology, the success of engineered immune effectors—particularly chimeric antigen receptor (CAR) T-cells—has redefined expectations. Initially approved for specific forms of relapsed or refractory B cell malignancies, CAR T-cell therapies represent a new class of “living drugs” capable of sustained, antigen-driven immune activity [7,8]. Their efficacy has spurred efforts to extend the platform to solid tumors and integrate it with other immune-modulatory strategies.
Despite these advances, critical bottlenecks persist. Complex manufacturing pipelines, variability in product quality, nonuniform global regulatory frameworks, and cost-related access disparities all impede clinical scalability [9,10]. The articles in this Special Issue “Where Are We Now and Where Is Cell Therapy Headed?” reflect a translational field that confronts these challenges directly. Collectively, they map a landscape in which cellular and acellular approaches are no longer mutually exclusive but increasingly complementary.
Several contributions focus on the shift from autologous to allogeneic and off-the-shelf strategies. Jeannerat and colleagues demonstrate that FE002-derived progenitor cells retain chondrogenic potential and survive hypoxic and inflammatory stress in intervertebral disc models (Contribution 1). Extending their approach, they develop bioengineered neoligament constructs with FE002-derived tenocytes, achieving rapid fabrication and effective integration (Contribution 2). Philippe and colleagues compare autologous and allogeneic grafts for knee cartilage repair and find that allogeneic products reduce patient variability and enhance scalability without compromising clinical outcomes (Contribution 3). In their paper on burn care, Chen and colleagues describe a hybrid dermo-epidermal graft incorporating both autologous and banked fibroblasts, offering a more agile production timeline with sustained efficacy (Contribution 4).
In parallel, the therapeutic potential of secreted factors is gaining clinical traction. Wu and colleagues provide a comprehensive analysis of ESCs, iPSCs, MSCs, and retinal progenitors for treating degenerative retinal conditions, outlining both opportunities and translational barriers (Contribution 5). Within the cancer immunotherapy space, Li and colleagues. explore cellular strategies to modulate the tumor microenvironment (TME), including the reprogramming of tumor-associated macrophages (Contribution 6) and the enhancement of checkpoint inhibitor responses in non-small cell lung cancer (Contribution 7). Meanwhile, Chiodelli and colleagues demonstrate that conditioned medium from human amniotic MSCs suppresses ovarian cancer proliferation and migration, exhibiting enhanced efficacy in combination with paclitaxel (Contribution 8).
Together, these contributions illustrate a rapidly diversifying field. On the one hand, the development of standardized, bankable cell products reflects a drive toward consistency and broader accessibility. On the other hand, cell-derived EVs and secretomes are emerging as potent therapeutic agents, capable of modulating immune responses, promoting tissue repair, and engaging in cross-tissue signaling.
Regardless, considerable challenges lie ahead. Product heterogeneity remains a critical concern for both cellular and extracellular therapies [11,12], and the lack of universally accepted potency assays and markers of activity further complicates development [13]. Issues of manufacturing scalability [9], cost containment [14,15,16], and international regulatory alignment [10,17,18] continue to shape—and constrain—the pathway from bench to bedside.
Nevertheless, the field is clearly beginning to gravitate toward therapies that extend beyond the cells themselves. The next generation of interventions is likely to harness not the cell as a unit, but the molecular and vesicular cargo it deploys. This reframing positions cell therapy as a modular, signal-driven platform, which is defined less by cellular identity and more by the biological programs it initiates within the host.

Author Contributions

Conceptualization, A.P. and P.C.; writing—original draft preparation, A.P.; writing—review and editing, A.P. and P.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by intramural funds at Università Cattolica del Sacro Cuore, Linea D1 (A.P.).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
MSCMesenchymal Stromal Cells
HCTHematopoietic cell transplantation
ESCEmbryonic Stem Cells
GvHDGraft Versus Host Disease
iPSCInduced Pluripotent Stem Cells
TMETumor Microenvironment
EVExtracellular Vesicle
CARChimeric Antigen Receptor

List of Contributions

  • Jeannerat, A.; Peneveyre, C.; Jaccoud, S.; Philippe, V.; Scaletta, C.; Hirt-Burri, N.; Abdel-Sayed, P.; Martin, R.; Applegate, L.A.; Pioletti, D.P.; et al. Banked Primary Progenitor Cells for Allogeneic Intervertebral Disc (IVD) Therapy: Preclinical Qualification and Functional Optimization within a Cell Spheroid Formulation Process. Pharmaceutics 2024, 16, 1274. https://doi.org/10.3390/pharmaceutics16101274.
  • Jeannerat, A.; Meuli, J.; Peneveyre, C.; Jaccoud, S.; Chemali, M.; Thomas, A.; Liao, Z.; Abdel-Sayed, P.; Scaletta, C.; Hirt-Burri, N.; et al. Bio-Enhanced Neoligaments Graft Bearing FE002 Primary Progenitor Tenocytes: Allogeneic Tissue Engineering & Surgical Proofs-of-Concept for Hand Ligament Regenerative Medicine. Pharmaceutics 2023, 15, 1873. https://doi.org/10.3390/pharmaceutics15071873.
  • Philippe, V.; Jeannerat, A.; Peneveyre, C.; Jaccoud, S.; Scaletta, C.; Hirt-Burri, N.; Abdel-Sayed, P.; Raffoul, W.; Darwiche, S.; Applegate, L.A.; et al. Autologous and Allogeneic Cytotherapies for Large Knee (Osteo)Chondral Defects: Manufacturing Process Benchmarking and Parallel Functional Qualification. Pharmaceutics 2023, 15, 2333. https://doi.org/10.3390/pharmaceutics15092333.
  • Chen, X.; Laurent, A.; Liao, Z.; Jaccoud, S.; Abdel-Sayed, P.; Flahaut, M.; Scaletta, C.; Raffoul, W.; Applegate, L.A.; Hirt-Burri, N. Cutaneous Cell Therapy Manufacturing Timeframe Rationalization: Allogeneic Off-the-Freezer Fibroblasts for Dermo-Epidermal Combined Preparations (DE-FE002-SK2) in Burn Care. Pharmaceutics 2023, 15, 2334. https://doi.org/10.3390/pharmaceutics15092334.
  • Wu, K.Y.; Dhaliwal, J.K.; Sasitharan, A.; Kalevar, A. Cell Therapy for Retinal Degenerative Diseases: Progress and Prospects. Pharmaceutics 2024, 16, 1299. https://doi.org/10.3390/pharmaceutics16101299.
  • Li, M.-Y.; Ye, W.; Luo, K.-W. Immunotherapies Targeting Tumor-Associated Macrophages (TAMs) in Cancer. Pharmaceutics 2024, 16, 865. https://doi.org/10.3390/pharmaceutics16070865.
  • Ye, W.; Li, M.; Luo, K. Therapies Targeting Immune Cells in Tumor Microenvironment for Non-Small Cell Lung Cancer. Pharmaceutics 2023, 15, 1788. https://doi.org/10.3390/pharmaceutics15071788.
  • Chiodelli, P.; Bonassi Signoroni, P.; Scalvini, E.; Farigu, S.; Giuzzi, E.; Paini, A.; Papait, A.; Stefani, F.R.; Silini, A.R.; Parolini, O. Synergistic Effect of Conditioned Medium from Amniotic Membrane Mesenchymal Stromal Cells Combined with Paclitaxel on Ovarian Cancer Cell Viability and Migration in 2D and 3D In Vitro Models. Pharmaceutics 2025, 17, 420. https://doi.org/10.3390/pharmaceutics17040420.

References

  1. Silini, A.R.; Papait, A.; Cargnoni, A.; Vertua, E.; Romele, P.; Bonassi Signoroni, P.; Magatti, M.; De Munari, S.; Masserdotti, A.; Pasotti, A.; et al. CM from intact hAM: An easily obtained product with relevant implications for translation in regenerative medicine. Stem Cell Res. Ther. 2021, 12, 540. [Google Scholar] [CrossRef] [PubMed]
  2. Ragni, E.; Papait, A.; Taiana, M.M.; De Luca, P.; Grieco, G.; Vertua, E.; Romele, P.; Colombo, C.; Silini, A.R.; Parolini, O.; et al. Cell culture expansion media choice affects secretory, protective and immuno-modulatory features of adipose mesenchymal stromal cell-derived secretomes for orthopaedic applications. Regen. Ther. 2025, 28, 481–497. [Google Scholar] [CrossRef] [PubMed]
  3. Kumar, M.A.; Baba, S.K.; Sadida, H.Q.; Marzooqi, S.A.; Jerobin, J.; Altemani, F.H.; Algehainy, N.; Alanazi, M.A.; Abou-Samra, A.B.; Kumar, R.; et al. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct. Target. Ther. 2024, 9, 27. [Google Scholar] [CrossRef]
  4. Prockop, S.; Wachter, F. The current landscape: Allogeneic hematopoietic stem cell transplant for acute lymphoblastic leukemia. Best Pract. Res. Clin. Haematol. 2023, 36, 101485. [Google Scholar] [CrossRef] [PubMed]
  5. Vuelta, E.; Garcia-Tunon, I.; Hernandez-Carabias, P.; Mendez, L.; Sanchez-Martin, M. Future Approaches for Treating Chronic Myeloid Leukemia: CRISPR Therapy. Biology 2021, 10, 118. [Google Scholar] [CrossRef] [PubMed]
  6. Bashor, C.J.; Hilton, I.B.; Bandukwala, H.; Smith, D.M.; Veiseh, O. Engineering the next generation of cell-based therapeutics. Nat. Rev. Drug Discov. 2022, 21, 655–675. [Google Scholar] [CrossRef] [PubMed]
  7. Sterner, R.C.; Sterner, R.M. CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 2021, 11, 69. [Google Scholar] [CrossRef] [PubMed]
  8. Holzinger, A.; Abken, H. Treatment with Living Drugs: Pharmaceutical Aspects of CAR T Cells. Pharmacology 2022, 107, 446–463. [Google Scholar] [CrossRef] [PubMed]
  9. Gavan, S.P.; Wright, S.J.; Thistlethwaite, F.; Payne, K. Capturing the Impact of Constraints on the Cost-Effectiveness of Cell and Gene Therapies: A Systematic Review. Pharmacoeconomics 2023, 41, 675–692. [Google Scholar] [CrossRef] [PubMed]
  10. Joppi, R.; Bertele, V.; Vannini, T.; Garattini, S.; Banzi, R. Food and Drug Administration vs European Medicines Agency: Review times and clinical evidence on novel drugs at the time of approval. Br. J. Clin. Pharmacol. 2020, 86, 170–174. [Google Scholar] [CrossRef] [PubMed]
  11. Li, J.; Wu, Z.; Zhao, L.; Liu, Y.; Su, Y.; Gong, X.; Liu, F.; Zhang, L. The heterogeneity of mesenchymal stem cells: An important issue to be addressed in cell therapy. Stem Cell Res. Ther. 2023, 14, 381. [Google Scholar] [CrossRef] [PubMed]
  12. Almeria, C.; Kress, S.; Weber, V.; Egger, D.; Kasper, C. Heterogeneity of mesenchymal stem cell-derived extracellular vesicles is highly impacted by the tissue/cell source and culture conditions. Cell Biosci. 2022, 12, 51. [Google Scholar] [CrossRef] [PubMed]
  13. Capelli, C.; Cuofano, C.; Pavoni, C.; Frigerio, S.; Lisini, D.; Nava, S.; Quaroni, M.; Colombo, V.; Galli, F.; Bezukladova, S.; et al. Potency assays and biomarkers for cell-based advanced therapy medicinal products. Front. Immunol. 2023, 14, 1186224. [Google Scholar] [CrossRef] [PubMed]
  14. Majhail, N.S.; Mau, L.W.; Denzen, E.M.; Arneson, T.J. Costs of autologous and allogeneic hematopoietic cell transplantation in the United States: A study using a large national private claims database. Bone Marrow Transplant. 2013, 48, 294–300. [Google Scholar] [CrossRef] [PubMed]
  15. Ten Ham, R.M.T.; Hovels, A.M.; Hoekman, J.; Frederix, G.W.J.; Leufkens, H.G.M.; Klungel, O.H.; Jedema, I.; Veld, S.A.J.; Nikolic, T.; Van Pel, M.; et al. What does cell therapy manufacturing cost? A framework and methodology to facilitate academic and other small-scale cell therapy manufacturing costings. Cytotherapy 2020, 22, 388–397. [Google Scholar] [CrossRef] [PubMed]
  16. Khang, M.; Suryaprakash, S.; Kotrappa, M.; Mulyasasmita, W.; Topp, S.; Wu, J. Manufacturing innovation to drive down cell therapy costs. Trends Biotechnol. 2023, 41, 1216–1219. [Google Scholar] [CrossRef] [PubMed]
  17. Olaghere, J.; Williams, D.A.; Farrar, J.; Buning, H.; Calhoun, C.; Ho, T.; Inamdar, M.S.; Liu, D.; Makani, J.; Nyarko, K.; et al. Scientific Advancements in Gene Therapies: Opportunities for Global Regulatory Convergence. Biomedicines 2025, 13, 758. [Google Scholar] [CrossRef] [PubMed]
  18. Maryamchik, E.; Ikonomou, L.; Roxland, B.E.; Grignon, F.; Levine, B.L.; Grilley, B.J.; ISCT Committee on the Ethics of Cell and Gene Therapy Expanded Access Working Group. International Society for Cell & Gene Therapy Expanded Access Working Group position paper: Key considerations to support equitable and ethical expanded access to investigational cell- and gene-based interventions. Cytotherapy 2025, 27, 671–677. [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

Papait, A.; Chiodelli, P. An Editorial on the Special Issue “Where Are We Now and Where Is Cell Therapy Headed?”. Pharmaceutics 2025, 17, 894. https://doi.org/10.3390/pharmaceutics17070894

AMA Style

Papait A, Chiodelli P. An Editorial on the Special Issue “Where Are We Now and Where Is Cell Therapy Headed?”. Pharmaceutics. 2025; 17(7):894. https://doi.org/10.3390/pharmaceutics17070894

Chicago/Turabian Style

Papait, Andrea, and Paola Chiodelli. 2025. "An Editorial on the Special Issue “Where Are We Now and Where Is Cell Therapy Headed?”" Pharmaceutics 17, no. 7: 894. https://doi.org/10.3390/pharmaceutics17070894

APA Style

Papait, A., & Chiodelli, P. (2025). An Editorial on the Special Issue “Where Are We Now and Where Is Cell Therapy Headed?”. Pharmaceutics, 17(7), 894. https://doi.org/10.3390/pharmaceutics17070894

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