Development of a human umbilical cord-derived mesenchymal stromal cell-based advanced therapy medicinal product to treat immune and/or inflammatory diseases

Miryam Mebarki, Nathan Iglicki, Céline Marigny, Camille Abadie, Claire Nicolet, Guillaume Churlaud, Camille Maheux, Hélène Boucher, Antoine Monsel, Philippe Menasché, Jérôme Larghero, Lionel Faivre, Audrey Cras, Miryam Mebarki, Nathan Iglicki, Céline Marigny, Camille Abadie, Claire Nicolet, Guillaume Churlaud, Camille Maheux, Hélène Boucher, Antoine Monsel, Philippe Menasché, Jérôme Larghero, Lionel Faivre, Audrey Cras

Abstract

Background: Umbilical cord-derived mesenchymal stromal cells (UC-MSCs) revealed their key role in immune regulation, offering promising therapeutic perspectives for immune and inflammatory diseases. We aimed to develop a production process of an UC-MSC-based product and then to characterize UC-MSC properties and immunomodulatory activities in vitro, related to their clinical use and finally, to transfer this technology to a good manufacturing practice (GMP) compliant facility, to manufacture an advanced therapy medicinal product (ATMP).

Methods: Fifteen human umbilical cords (UCs) were collected to develop the production process. Three batches of UC-MSCs from a single donor were characterized at basal state and after in vitro pro-inflammatory stimulation by interferon-γ (IFNγ) and tumor necrosis factor-α (TNFα). Proliferation, immunophenotype, activation markers' expression and the inhibition of T cell proliferation were assessed. Finally, this technology was transferred to a GMP-compliant facility to manufacture an UC-MSC-based ATMP, from a single donor, using the explant method followed by the establishment of master and work cell stocks.

Results: Twelve UCs were processed successfully allowing to isolate UC-MSCs with doubling time and population doubling remaining stable until passage 4. CD90, CD105, CD73, CD44, CD29, CD166 expression was positive; CD14, CD45, CD31, HLA-DR, CD40, CD80 and CD86 expression was negative, while CD146 and HLA-ABC expression was heterogeneous. Cell morphology, proliferation and immunophenotype were not modified by inflammatory treatment. Indoleamine 2,3-dioxygenase (IDO) expression was significantly induced by IFNγ and IFNγ + TNFα versus non-treated cells. Intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) expression was induced significantly after priming. T cell proliferation was significantly decreased in the presence of UC-MSCs in a dose-dependent manner. This inhibitory effect was improved by IFNγ or IFNγ + TNFα, at UC-MSCs:PBMC ratio 1:10 and 1:30, whereas only IFNγ allowed to decrease significantly T cell proliferation at ratio 1:100. The manufacturing process of the UC-MSC-based ATMP was qualified and authorized by the French regulatory agency for clinical use (NCT04333368).

Conclusion: This work allowed to develop an investigational UC-MSC-based ATMP authorized for clinical use. Our results showed that an inflammatory environment preserves the biological properties of UC-MSCs with an improvement of their immunomodulatory functions.

Keywords: Advanced therapy medicinal product; Good manufacturing practice; Human umbilical cord; Immunomodulation; Inflammation; Mesenchymal stromal cells.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Development of UC-MSCs’ production process. A Characteristics of collected and processed UCs (n = 15 UCs). B UC-MSCs morphology at passage 1 (P1) at 4X (left) and 10X (right) magnifications. C UC-MSCs quantity at P0 and P1 (left) and DT (h) and PD calculated from P0 to P1 (right) for each UC (n = 12 UCs). D DT (h) and PD from P1 to P7 (n = 4 UCs) (left) and UC-MSCs morphology at P7 (right) representing senescent morphology (head arrows). E Comparison of DT (h) and PD between culture media composed of Nutristem® + PL5% and MEM-α + PL 5% at P3 and P4 (n = 4 UCs)
Fig. 2
Fig. 2
UC-MSCs immunophenotype at basal state. A Expression of mesenchymal markers (CD90, CD105, CD73), adhesion molecules (CD44, CD29, CD166, CD146), hematopoietic (CD14, CD45) and endothelial (CD31) markers. B Expression of immunogenic (HLA-ABC, HLA-DR) and co-stimulatory (CD40, CD80, CD86) markers. C Summary of markers’ expression represented as mean ± standard deviation (%) (n = 3 batches from 1 UC)
Fig. 3
Fig. 3
UC-MSCs’ characteristics after pro-inflammatory priming. A UC-MSCs’ morphology; B UC-MSCs’ size (µm); C DT (h) (left) and PD (right); and D UC-MSCs’ immunophenotype at basal state (NT) and after pro-inflammatory treatment by IFNγ, TNFα, IFNγ + TNFα, IL6, IL1β, GM-CSF and Mix. NA: Not applicable. n = 3 batches from 1 UC
Fig. 4
Fig. 4
UC-MSCs’ biological activity in vitro after pro-inflammatory priming. A IDO expression (%) at basal state (NT) and after pro-inflammatory priming by IFNγ, TNFα, IFNγ + TNFα, IL1β, IL6, GM-CSF and Mix. B ICAM-1/CD54, PD-L1/CD274, VCAM-1/CD106, CD200, IFNγ-R/CD119, TNFα-RII/CD120b expression (%) at basal state (NT) and after pro-inflammatory priming by IFNγ, TNFα and IFNγ + TNFα. C T-lymphocyte proliferation (%) at ratio UC-MSCs:PBMC 0:1, 1:10, 1:30, 1:100, 1:300 and 1:1000 at basal state (NT) and after pro-inflammatory priming by IFNγ, TNFα and IFNγ + TNFα. */$p < 0.05, **/$$p < 0.01, ***/$$$p < 0.001, ****/$$$$p < 0.0001. n = 3 batches from 1 UC
Fig. 5
Fig. 5
Qualification of the UC-MSC-based ATMP manufacturing process. A Quantities and proliferation data of UC-MSCs from P0 to P3. B Quality controls performed during MCS (n = 2 batches) and WCS steps (n = 3 batches)

References

    1. Lazarus HM, Haynesworth SE, Gerson SL, Rosenthal NS, Caplan AI. Ex vivo expansion and subsequent infusion of human bone marrow-derived stromal progenitor cells (mesenchymal progenitor cells): implications for therapeutic use. Bone Marrow Transplant. 1995;16(4):557–564.
    1. Gómez-Barrena E, Padilla-Eguiluz NG, Rosset P, Hernigou P, Baldini N, Ciapetti G, et al. Osteonecrosis of the femoral head safely healed with autologous, expanded, bone marrow-derived mesenchymal stromal cells in a multicentric trial with minimum 5 years follow-up. J Clin Med. 2021;10(3):508. doi: 10.3390/jcm10030508.
    1. Li X, Bai J, Ji X, Li R, Xuan Y, Wang Y. Comprehensive characterization of four different populations of human mesenchymal stem cells as regards their immune properties, proliferation and differentiation. Int J Mol Med. 2014;34(3):695–704. doi: 10.3892/ijmm.2014.1821.
    1. Wu K-H, Chan C-K, Tsai C, Chang Y-H, Sieber M, Chiu T-H, et al. Effective treatment of severe steroid-resistant acute graft-versus-host disease with umbilical cord-derived mesenchymal stem cells. Transplantation. 2011;91(12):1412–1416. doi: 10.1097/TP.0b013e31821aba18.
    1. Wang D, Li J, Zhang Y, Zhang M, Chen J, Li X, et al. Umbilical cord mesenchymal stem cell transplantation in active and refractory systemic lupus erythematosus: a multicenter clinical study. Arthritis Res Ther. 2014;16(2):R79. doi: 10.1186/ar4520.
    1. Thompson M, Mei SHJ, Wolfe D, Champagne J, Fergusson D, Stewart DJ, et al. Cell therapy with intravascular administration of mesenchymal stromal cells continues to appear safe: an updated systematic review and meta-analysis. EClinicalMedicine. 2020;19:100249. doi: 10.1016/j.eclinm.2019.100249.
    1. Mebarki M, Abadie C, Larghero J, Cras A. Human umbilical cord-derived mesenchymal stem/stromal cells: a promising candidate for the development of advanced therapy medicinal products. Stem Cell Res Ther. 2021;12(1):152. doi: 10.1186/s13287-021-02222-y.
    1. EMA/140033/2021. Scientific recommendations on classification of advanced therapy medicinal products [Internet]. European Medicines Agency. 2021 [cited 2021 Jul 7]. Available from:
    1. Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, et al. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008;2(2):141–150. doi: 10.1016/j.stem.2007.11.014.
    1. de Witte SFH, Merino AM, Franquesa M, Strini T, van Zoggel JAA, Korevaar SS, et al. Cytokine treatment optimises the immunotherapeutic effects of umbilical cord-derived MSC for treatment of inflammatory liver disease. Stem Cell Res Ther. 2017;8(1):140. doi: 10.1186/s13287-017-0590-6.
    1. Uciechowski P, Dempke WCM. Interleukin-6: a masterplayer in the cytokine network. Oncology. 2020;98(3):131–137. doi: 10.1159/000505099.
    1. Becher B, Tugues S, Greter M. GM-CSF: from growth factor to central mediator of tissue inflammation. Immunity. 2016;45(5):963–973. doi: 10.1016/j.immuni.2016.10.026.
    1. Assistance Publique - Hôpitaux de Paris. Cell Therapy Using Umbilical Cord-derived Mesenchymal Stromal Cells in SARS-CoV-2-related ARDS [Internet]. ; 2020 Dec [cited 2021 Jul 5]. Report No.: NCT04333368. Available from:
    1. Marmotti A, Mattia S, Bruzzone M, Buttiglieri S, Risso A, Bonasia DE, et al. Minced umbilical cord fragments as a source of cells for orthopaedic tissue engineering: an in vitro study. Stem Cells Int. 2012;2012:326813. doi: 10.1155/2012/326813.
    1. Krampera M, Galipeau J, Shi Y, Tarte K, Sensebe L. MSC Committee of the International Society for Cellular Therapy (ISCT): Immunological characterization of multipotent mesenchymal stromal cells—The International Society for Cellular Therapy (ISCT) working proposal. Cytotherapy. 2013;15(9):1054–1061. doi: 10.1016/j.jcyt.2013.02.010.
    1. Nicotra T, Desnos A, Halimi J, Antonot H, Reppel L, Belmas T, et al. Mesenchymal stem/stromal cell quality control: validation of mixed lymphocyte reaction assay using flow cytometry according to ICH Q2(R1) Stem Cell Res Ther. 2020;11(1):426. doi: 10.1186/s13287-020-01947-6.
    1. Guidelines on Good Manufacturing Practice specific to Advanced Therapy Medicinal Products [Internet]. Available from:
    1. Ren G, Su J, Zhang L, Zhao X, Ling W, L’huillie A, et al. Species variation in the mechanisms of mesenchymal stem cell-mediated immunosuppression. Stem Cells. 2009;27(8):1954–1962. doi: 10.1002/stem.118.
    1. Can A, Celikkan FT, Cinar O. Umbilical cord mesenchymal stromal cell transplantations: a systemic analysis of clinical trials. Cytotherapy. 2017;19(12):1351–1382. doi: 10.1016/j.jcyt.2017.08.004.
    1. Nagamura-Inoue T, He H. Umbilical cord-derived mesenchymal stem cells: their advantages and potential clinical utility. World J Stem Cells. 2014;6(2):195–202. doi: 10.4252/wjsc.v6.i2.195.
    1. Bai L, Li D, Li J, Luo Z, Yu S, Cao S, et al. Bioactive molecules derived from umbilical cord mesenchymal stem cells. Acta Histochem. 2016;118(8):761–769. doi: 10.1016/j.acthis.2016.09.006.
    1. Viswanathan S, Shi Y, Galipeau J, Krampera M, Leblanc K, Martin I, et al. Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy (ISCT®) mesenchymal stromal cell committee position statement on nomenclature. Cytotherapy. 2019;21(10):1019–1024. doi: 10.1016/j.jcyt.2019.08.002.
    1. Kern S, Eichler H, Stoeve J, Klüter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 2006;24(5):1294–1301. doi: 10.1634/stemcells.2005-0342.
    1. Menard C, Pacelli L, Bassi G, Dulong J, Bifari F, Bezier I, et al. Clinical-grade mesenchymal stromal cells produced under various good manufacturing practice processes differ in their immunomodulatory properties: standardization of immune quality controls. Stem Cells Dev. 2013;22(12):1789–1801. doi: 10.1089/scd.2012.0594.
    1. Berglund AK, Fortier LA, Antczak DF, Schnabel LV. Immunoprivileged no more: measuring the immunogenicity of allogeneic adult mesenchymal stem cells. Stem Cell Res Ther. 2017;8(1):288. doi: 10.1186/s13287-017-0742-8.
    1. Bowles AC, Kouroupis D, Willman MA, Perucca Orfei C, Agarwal A, Correa D. Signature quality attributes of CD146+ mesenchymal stem/stromal cells correlate with high therapeutic and secretory potency. Stem Cells. 2020;38(8):1034–1049. doi: 10.1002/stem.3196.
    1. Ren G, Zhao X, Zhang L, Zhang J, L’Huillier A, Ling W, et al. Inflammatory cytokine-induced intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in mesenchymal stem cells are critical for immunosuppression. J Immunol. 2010;184(5):2321–2328. doi: 10.4049/jimmunol.0902023.
    1. Guan Q, Li Y, Shpiruk T, Bhagwat S, Wall DA. Inducible indoleamine 2,3-dioxygenase 1 and programmed death ligand 1 expression as the potency marker for mesenchymal stromal cells. Cytotherapy. 2018;20(5):639–649. doi: 10.1016/j.jcyt.2018.02.003.
    1. de Wolf C, van de Bovenkamp M, Hoefnagel M. Regulatory perspective on in vitro potency assays for human mesenchymal stromal cells used in immunotherapy. Cytotherapy. 2017;19(7):784–797. doi: 10.1016/j.jcyt.2017.03.076.
    1. Zhang C, Zhou L, Wang Z, Gao W, Chen W, Zhang H, et al. Eradication of specific donor-dependent variations of mesenchymal stem cells in immunomodulation to enhance therapeutic values. Cell Death Dis. 2021;12(4):1–11.

Source: PubMed

3
Subscribe