Skip to main content

EDITORIAL article

Front. Cell Dev. Biol., 18 July 2022
Sec. Stem Cell Research
This article is part of the Research Topic Mesenchymal Stromal Cells: Preclinical and Clinical Challenges View all 23 articles

Editorial: Mesenchymal Stromal Cells: Preclinical and Clinical Challenges

  • 1Department of Clinical Research, Emmaus Life Sciences, Torrance, CA, United States
  • 2University of Pisa, Pisa, Italy
  • 3Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Creatio- Production and Validation Center of Advanced Therapies, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
  • 4Fujifilm Irvine Scientific, Inc, Santa Ana, CA, United States

For the past 20 years, mesenchymal stromal cells (MSCs) has become the most studied cell population in the development of adult cell therapies for disease treatment (Keshtkar et al., 2018; Han et al., 2019). MSCs can differentiate into many types of cells (neurons, hepatocytes, myoblast … ) and this is one reason why MSCs hold great promise to the treatment of many immune diseases, cardiac and neurological injuries, and tissue regenerative applications (Hwang et al., 2009). Although MSC research discoveries brought new information, the road for cell therapy approval is still at its dawn. Due to the insurgence of MSC therapies, federal agencies with regulatory oversight to healthcare such as the Food and Drug Administration, European Medical Agency, Pharmaceuticals and Medical Devices Agency, Federal Service for Surveillance in Healthcare, etc. have adapted and continue to update their guidelines as needed (Mendicino et al., 2014; Corbett et al., 2017; Pigeau et al., 2018; Stroncek et al., 2022). Similarly, cell therapy manufacturers and suppliers had to adapt quickly in establishing and adopting best practices that ensure safety, quality, and reproducibility of products and raw materials destined to be used in cell therapy manufacturing. Even so, many questions have surfaced around preclinical tests, scalability of MSC production and clinical application, reproducibility of the results, better characterization of the MSCs, the need for development of defined culture media and GMP compliant animal-free components, ancillary materials, and the development of 3D structures mimicking the tissue organization.

Friedenstein reported the first fibroblastic-like and spindle-shaped cells to differentiate into other type of cells: chondrocyte and osteoblast (Friedenstein et al., 1968; Friedenstein et al., 1974). MSCs have been isolated from different tissues: adipose tissue, dental pulp, periosteum, Wharton’s jelly, umbilical cord (Zuk et al., 2001; Nagamura-Inoue and He, 2014). Due to terminology discrepancies, one of the first task will be to use the exact terminology of MSCs depending on their functional attributes (Bhartiya, 2018). In addition of the Dominici minimal criteria, additional criteria were added for the MSC characterization like MSC responsive to INF-g, TNF-a, indoleamine 2,3 dioxygenase etc (Dominici et al., 2006; Bhartiya, 2018), thanks to the formation of international consortium among expert in MSCs. As mentioned by Najar et al, Wright et al. and Jayaraman et al., the identification of additional MSC markers will strongly support consistency of data obtained from studies. However, in vitro, and in vivo results obtained in clinical trials are still not consistent due to lot-to-lot variations, quality of the cells, variability among the donors. To decrease the variability among studies and to increase the reproducibility, standardization of the isolation, identification of additional surface markers, methodology of MSC expansion must be established (Stroncek et al., 2020). A recent study showed that, even if the laboratories use MSCs from the same material source, MSCs showed different behavior in terms of viability after thawing and different transcriptome due to different methodologies used in the lab and donor source (Stroncek et al., 2020). Standardization of MSC isolation, expansion, and characterization, like harmonizing the guidelines for clinicals trials is a major goal for the scientific community. Publications in this Research Topic hopefully will be part of the discussion to help understanding the challenges due to the failure to establish standardized protocols. After the characterization of the MSCs, one of the most important task and challenge encountered is to establish protocols to expand MSCs, that are in harmony and compliance with federal agencies.

There is still a debate around effective delivery and use of MSCs in treating diseases, by injection, by transplantation or by using the secreted extracellular vesicles. The cheapest and fastest methodology is the injection of isolated cells. Injection of cells showed encouraging results, but the long-term effects of MSCs are unknown in terms of treating the diseases, or impairing organ function due to random anchoring of the MSCs. In addition, the survival of injected MSCs is very low (Gyöngyösi et al., 2008), which could explain why treatment of patients is more complicated and not very efficient. Different approaches have been developed to overcome the low efficacy of injected MSCs: increasing the number of injected cells or increasing the number of injections, but it will require a larger scale manufacturing of MSCs, priming the cells, finding the optimal route of administration (Kurtz, 2008; Noronha et al., 2019). The development of cell sheets is an option to control the targeting of the cells in the organs. Myoblast cell sheets were transplanted on heart damaged areas, after heart failure, increasing the period of free events, increasing of survival, and decreasing of death rate. Characterization and the establishment of release criteria before transplantation is a major concern. Actually, only visual observation is used to determine when a cell sheet is ready, and the high variability in outcome is largely due to human dependency based on their experience and knowledge. A more rigorous approach as reported in by Ochiai et al., is to utilize physical characteristics (strength, optical) of the cell sheets to standardize the cell sheets release criteria in GMP facilities.

In addition, the formation of cell sheets modified the production of cytokines by the MSC (Bou-Ghannam et al., 2021). Also, it was noticed that the effect of MSCs in vivo studies cannot be explained by the number of cells that reach their target (Bou-Ghannam et al., 2021) indicating that paracrine factors, released by the MSCs, could be the major factor. It is well known that MSCs can modulate the immune system by secreting paracrine factors (Ferreira et al., 2018). MSCs influence inflammation through paracrine factors, which lead to the study of mechanism of action. Many publications, in this research topic, mentioned the importance of EV characterization and variability produced by MSCs, for a beneficial effect on treating different diseases such as spinal cord injury, amyotrophic lateral sclerosis, wound healing, pancreatic cancer, heart failure. As reported by Sykova et al., Johnson et al., Najar et al. and Fernandez-Gonzalez et al, the content of these EV can be altered due to genetic engineering, by priming the MSCs or by using them as a drug transporter, which allow the manipulator to “guide” the EV in a way to have an optimal curative property.

MSCs are used in clinical trials, reported by Garcia-Bernal et al., Sykova et al., Harman and Wiese et al., but as mentioned by Najara, MSCs are used to treat diseases and widely used in clinical trials, but the effect of MSCs on one of the most frequent diseases is still controversial. It is still unclear why MSCs can promote or repress tumors growth/survival. To better understand such opposite effects, retrospective analysis of hundreds of clinical trials is necessary but because reported data are incomplete, the data analysis will be challenging to explain MSC influence on tumors, as mentioned in this research topic by Zhao et al.

Due to the lack of knowledge and experience in MSC clinical applications, federal agencies had to update their guidelines, and keep improving them in parallel to increased pre-clinical and clinical experiences. Food and Drug Administration is a perfect example of how the federal agencies are updating and adding complementary guidelines in the translational field of Cellular and Gene Therapies (Food and Drug Administration, 2022). From 1998 to 2014, FDA released 11 guidelines, but from 2015 to 2022, FDA released 22 guidelines, underlining the importance for agencies to improve their guidelines due to the increase of pre-clinical and clinical studies (Couto et al., 2017; Kabat et al., 2020). However, challenges are still around the corner because many clinical trials published on clinicaltrials. gov are still failing to provide detailed information about the patient’s population, the manufacturing of the MSCs (that require a large-scale manufacturing). In addition to the topics mentioned, other crucial subjects about MSCs are presented and discussed in the research topic like priming by Berglund et al., alleviation of ischemia injuries by Arjmand et al. and Chua et al., development of tendinopathy animal model for MSC treatment by Meeremans et al., discovery of new drugs to accelerate MSC differentiation into osteoblast by Wang et al., use of immortalized MSC model for hormonal by Kulebyakin et al., tissue specificity of mesangiogenic progenitors cells by Barachini et al., transcriptional characterization of MSCs by Fan et al. and the impact of MSC senescence state used in clinical trials by Alves-Paiva et al. In order to continue progressing MSC field in clinical applications, academics, clinicians, and industry partners need to continue collaborating, sharing both knowledge and best practices that help to advance this field.

Author Contributions

JO wrote the first draft of the editorial. ML, SP and JMC edited and revised the editorial.

Conflict of Interest

Author ML is employed by FujiFilm Irvine Scientific, Inc. Author JO is employed by Emmaus Lifes Sciences. Inc.

The remaining 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

Food and Drug Administration (2022). Cellular & Gene Therapy Guidances.

Google Scholar

Bhartiya, D. (2018). The Need to Revisit the Definition of Mesenchymal and Adult Stem Cells Based on Their Functional Attributes. Stem Cell Res. Ther. 9 (1), 78. doi:10.1186/s13287-018-0833-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Bou-Ghannam, S., Kim, K., Grainger, D. W., and Okano, T. (2021). 3D Cell Sheet Structure Augments Mesenchymal Stem Cell Cytokine Production. Sci. Rep. 11 (1), 8170. doi:10.1038/s41598-021-87571-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Corbett, M. S., Webster, A., Hawkins, R., and Woolacott, N. (2017). Innovative Regenerative Medicines in the EU: a Better Future in Evidence? BMC Med. 15 (1), 49. doi:10.1186/s12916-017-0818-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Couto, P. S., Bersenev, A., and Verter, F. (2017). The First Decade of Advanced Cell Therapy Clinical Trials Using Perinatal Cells (2005-2015). Regen. Med. 12 (8), 953–968. doi:10.2217/rme-2017-0066

PubMed Abstract | CrossRef Full Text | Google Scholar

Dominici, M., Le Blanc, K., Mueller, I., Slaper-Cortenbach, I., Marini, F. C., Krause, D. S., et al. (2006). Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8 (4), 315–317. doi:10.1080/14653240600855905

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferreira, J. R., Teixeira, G. Q., Santos, S. G., Barbosa, M. A., Almeida-Porada, G., and Gonçalves, R. M. (2018). Mesenchymal Stromal Cell Secretome: Influencing Therapeutic Potential by Cellular Pre-conditioning. Front. Immunol. 9, 2837. doi:10.3389/fimmu.2018.02837

PubMed Abstract | CrossRef Full Text | Google Scholar

Friedenstein, A. J., Chailakhyan, R. K., Latsinik, N. V., Panasyuk, A. F., and Keiliss-Borok, I. V. (1974). Stromal Cells Responsible for Transferring the Microenvironment of the Hemopoietic Tissues. Transplantation 17 (4), 331–340. doi:10.1097/00007890-197404000-00001

PubMed Abstract | CrossRef Full Text | Google Scholar

Friedenstein, A. J., Petrakova, K. V., Kurolesova, A. I., and Frolova, G. P. (1968). Heterotopic Transplants of Bone Marrow. Transplantation 6 (2), 230–247. doi:10.1097/00007890-196803000-00009

PubMed Abstract | CrossRef Full Text | Google Scholar

Gyöngyösi, M., Blanco, J., Marian, T., Trón, L., Petneházy, O., Petrasi, Z., et al. (2008). Serial noninvasive In Vivo positron emission tomographic tracking of percutaneously intramyocardially injected autologous porcine mesenchymal stem cells modified for transgene reporter gene expression. Circ. Cardiovasc Imaging 1 (2), 94–103. doi:10.1161/CIRCIMAGING.108.797449

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Y., Li, X., Zhang, Y., Han, Y., Chang, F., and Ding, J. (2019). Mesenchymal Stem Cells for Regenerative Medicine. Cells 8 (8). doi:10.3390/cells8080886

PubMed Abstract | CrossRef Full Text | Google Scholar

Hwang, N. S., Zhang, C., Hwang, Y. S., and Varghese, S. (2009). Mesenchymal stem cell differentiation and roles in regenerative medicine. WIREs Mech. Dis. 1 (1), 97–106. doi:10.1002/wsbm.26

PubMed Abstract | CrossRef Full Text | Google Scholar

Kabat, M., Bobkov, I., Kumar, S., and Grumet, M. (2020). Trends in mesenchymal stem cell clinical trials 2004-2018: Is efficacy optimal in a narrow dose range? Stem Cells Transl. Med. 9 (1), 17–27. doi:10.1002/sctm.19-0202

PubMed Abstract | CrossRef Full Text | Google Scholar

Keshtkar, S., Azarpira, N., and Ghahremani, M. H. (2018). Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res. Ther. 9 (1), 63. doi:10.1186/s13287-018-0791-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Kurtz, A. (2008). Mesenchymal stem cell delivery routes and fate. Int. J. Stem Cells 1 (1), 1–7. doi:10.15283/ijsc.2008.1.1.1

PubMed Abstract | CrossRef Full Text | Google Scholar

Mendicino, M., Bailey, A. M., Wonnacott, K., Puri, R. K., and Bauer, S. R. (2014). MSC-based product characterization for clinical trials: an FDA perspective. Cell Stem Cell 14 (2), 141–145. doi:10.1016/j.stem.2014.01.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Nagamura-Inoue, T., and He, H. (2014). Umbilical cord-derived mesenchymal stem cells: Their advantages and potential clinical utility. Wjsc 6 (2), 195–202. doi:10.4252/wjsc.v6.i2.195

PubMed Abstract | CrossRef Full Text | Google Scholar

Noronha, N. d. C., Mizukami, A., Caliári-Oliveira, C., Cominal, J. G., Rocha, J. L. M., Covas, D. T., et al. (2019). Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res. Ther. 10 (1), 131. doi:10.1186/s13287-019-1224-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Pigeau, G. M., Csaszar, E., and Dulgar-Tulloch, A. (2018). Commercial Scale Manufacturing of Allogeneic Cell Therapy. Front. Med. 5, 233. doi:10.3389/fmed.2018.00233

PubMed Abstract | CrossRef Full Text | Google Scholar

Stroncek, D. F., Jin, P., McKenna, D. H., Takanashi, M., Fontaine, M. J., Pati, S., et al. (2020). Human Mesenchymal Stromal Cell (MSC) Characteristics Vary Among Laboratories When Manufactured From the Same Source Material: A Report by the Cellular Therapy Team of the Biomedical Excellence for Safer Transfusion (BEST) Collaborative. Front. Cell Dev. Biol. 8, 458. doi:10.3389/fcell.2020.00458

PubMed Abstract | CrossRef Full Text | Google Scholar

Stroncek, D. F., Somerville, R. P. T., and Highfill, S. L. (2022). Point-of-care cell therapy manufacturing; it's Not for everyone. J. Transl. Med. 20 (1), 34. doi:10.1186/s12967-022-03238-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Zuk, P. A., Zhu, M., Mizuno, H., Huang, J., Futrell, J. W., Katz, A. J., et al. (2001). Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 7 (2), 211–228. doi:10.1089/107632701300062859

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: MSC, translational medicine, GMP, regulations, cell/gene therapies

Citation: Oliva J, Pacini S, Canals JM and Lim M (2022) Editorial: Mesenchymal Stromal Cells: Preclinical and Clinical Challenges. Front. Cell Dev. Biol. 10:969178. doi: 10.3389/fcell.2022.969178

Received: 14 June 2022; Accepted: 22 June 2022;
Published: 18 July 2022.

Edited and reviewed by:

Valerie Kouskoff, The University of Manchester, United Kingdom

Copyright © 2022 Oliva, Pacini, Canals and Lim. 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: Joan Oliva, joliva@emmauslifesciences.com

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.