Ex vivo-expanded highly pure ABCB5+ mesenchymal stromal cells as Good Manufacturing Practice-compliant autologous advanced therapy medicinal product for clinical use: process validation and first in-human data

Andreas Kerstan, Elke Niebergall-Roth, Jasmina Esterlechner, Hannes M Schröder, Martin Gasser, Ana M Waaga-Gasser, Matthias Goebeler, Katrin Rak, Philipp Schrüfer, Sabrina Endres, Petra Hagenbusch, Korinna Kraft, Kathrin Dieter, Seda Ballikaya, Nicole Stemler, Samar Sadeghi, Nils Tappenbeck, George F Murphy, Dennis P Orgill, Natasha Y Frank, Christoph Ganss, Karin Scharffetter-Kochanek, Markus H Frank, Mark A Kluth, Andreas Kerstan, Elke Niebergall-Roth, Jasmina Esterlechner, Hannes M Schröder, Martin Gasser, Ana M Waaga-Gasser, Matthias Goebeler, Katrin Rak, Philipp Schrüfer, Sabrina Endres, Petra Hagenbusch, Korinna Kraft, Kathrin Dieter, Seda Ballikaya, Nicole Stemler, Samar Sadeghi, Nils Tappenbeck, George F Murphy, Dennis P Orgill, Natasha Y Frank, Christoph Ganss, Karin Scharffetter-Kochanek, Markus H Frank, Mark A Kluth

Abstract

Background aim: Mesenchymal stromal cells (MSCs) hold promise for the treatment of tissue damage and injury. However, MSCs comprise multiple subpopulations with diverse properties, which could explain inconsistent therapeutic outcomes seen among therapeutic attempts. Recently, the adenosine triphosphate-binding cassette transporter ABCB5 has been shown to identify a novel dermal immunomodulatory MSC subpopulation.

Methods: The authors have established a validated Good Manufacturing Practice (GMP)-compliant expansion and manufacturing process by which ABCB5+ MSCs can be isolated from skin tissue and processed to generate a highly functional homogeneous cell population manufactured as an advanced therapy medicinal product (ATMP). This product has been approved by the German competent regulatory authority to be tested in a clinical trial to treat therapy-resistant chronic venous ulcers.

Results: As of now, 12 wounds in nine patients have been treated with 5 × 105 autologous ABCB5+ MSCs per cm2 wound area, eliciting a median wound size reduction of 63% (range, 32-100%) at 12 weeks and early relief of pain.

Conclusions: The authors describe here their GMP- and European Pharmacopoeia-compliant production and quality control process, report on a pre-clinical dose selection study and present the first in-human results. Together, these data substantiate the idea that ABCB5+ MSCs manufactured as ATMPs could deliver a clinically relevant wound closure strategy for patients with chronic therapy-resistant wounds.

Trial registration: ClinicalTrials.gov NCT03257098 NCT03267784 NCT03529877 NCT03860155.

Keywords: ABCB5; GMP manufacturing; advanced therapy medicinal product; chronic wound; mesenchymal stromal cells; venous ulcer.

Copyright © 2020 International Society for Cell & Gene Therapy. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.
Figure 1.
Morphology and growth behavior of cultured ABCB5+ MSCs. (A) Cultured MSCs at about 80% confluency, displaying a spindled-shaped appearance. Magnification ×200. (B) Live cell count measured over time (upper panel) in parallel with cell confluency as determined visually (lower panel) from representative donors. (C) Cdkn1α gene expression (means of relative quantification + SD) was determined at 30%, 70% and 99% confluency. (D) Cell cycle phase distribution expressed as mean (± SD) percentage of cells in G1, S and G2/M phases; n = 503 batches from 260 donors. SD, standard deviation. (Color version of figure is available online).
Figure 2.
Figure 2.
Validation of the expansion process of ABCB5+ MSCs. (A,B) Validation of passage number. Individual CPDs (n = 15 donors, represented by different colors) (A) and mean (± SD) division rates (n = 15 donors) (B), plotted against the respective passage number, demonstrating continuous cell expansion during up to 16 passages. (C) Comparability and homogeneity of batches between different passages of a given donor and between different donors as assessed by microarray-based gene expression analysis performed on samples from four various passages per donor for eight donors. Within each donor, the quantile-normalized data sets from the three higher passages were compared with the lowest passage. From each of these between-passage comparisons, the median gene ratio (of all genes that had shown an average signal >20 and at least 2-fold differential regulation) was determined. These values were used to calculate the median gene ratio (mean ± SD) for each delta passage value over all eight donors; n = number of between-passage comparisons with the delta passage value specified. Original microarray data were uploaded in Gene Expression Omnibus (accession no. GSE145589) and are available at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE145589. (D) Relative amount (mean percentage ± SD) of ABCB5+ cells in culture, plotted against the respective passage number (n = 17 donors) (left panel), and absolute yield of ABCB5+ MSCs shown as single values from 500 batches derived from 260 donors, giving a mean count of 26.27 × 106 ABCB5+ MSCs per batch (represented by red line, SD = 19.76 × 106) (right panel). SD, standard deviation. (Color version of figure is available online).
Figure 3.
Figure 3.
Effect of topical application of ABCB5+ MSCs on healing of experimental full-thickness skin wounds in NSG mice. (A–C) Wound sizes (means of both wounds of each animal) at 9 days (A), 11 days (B) and 13 days (C) after vehicle (control) or MSC application. (D) Total number of fully closed wounds at 13 days after vehicle (control) or MSC application, shown as percentage value for each group. (E–G) Immunohistochemical evaluation of the wounds for human CD31 (E), unspecific CD31 (antibody-detected mouse and human CD31) (F) and cytokeratin 14 (G). Expression was semi-quantitatively quantified as no (−), slight (1+), moderate (2+), strong (3+) and intense (4+) positivity. Shown are group means ± SD. Control: vehicle only (n = 12 wounds, six animals). Low dose: 1.875 × 105 ABCB5+ MSCs per wound (n = 18 wounds, nine animals; one animal was prematurely euthanized). Low to mid dose: 3.75 × 105 ABCB5+ MSCs per wound (n = 19 wounds, 10 animals; one wound was pathologically not evaluable). Mid to high: 7.5 × 105 ABCB5+ MSCs per wound (n = 20 wounds, 10 animals). High dose: 1.5 × 106 ABCB5+ MSCs per wound (n = 20 wounds, 10 animals). The high-dose group was excluded from wound size analysis (A–C) because of significantly smaller baseline wound size values (not shown). Non-parametric Kruskal-Wallis test followed by Dunn’s multiple comparisons. *P < 0.05, **P < 0.01, ***P < 0.01 versus control. d., dose; IHC, immunohistochemistry; SD, standard deviation. (Color version of figure is available online).
Figure 4.
Figure 4.
Effect of topical application of 5 × 105 autologous skin-derived ABCB5+ MSCs per cm2 wound area on healing of human chronic venous ulcers. (A) Photographs of the target wounds of three representative patients before (day 0, upper panel) and 12 weeks after (lower panel) cell application. For photographs of all patients who were eligible for efficacy evaluation, see supplementary Figure 4. (B) Wound closure kinetics before (during cell expansion) and throughout the 12-week period following cell application (efficacy follow-up). Shown are all patients who were eligible for efficacy evaluation. For patient 8, wound size data at week 4 are missing. Increase in the wound after week 6 coincided with two changes in routine wound care; namely, less frequent (once instead of twice weekly) dressing changes and a switch to less absorbent dressing (Mepilex instead of Biatain). For patient 9, wound size data at week 6 are missing; instead, week 7 measurement is included in the graph. Red shaded areas highlight the first 6 weeks of efficacy follow-up, during which wound healing was most pronounced. w6, week 6; w7, week 7; w12, week 12. (Color version of figure is available online).

References

    1. Menaa F, Shahrokhi S, Prasad Shastri V. Impact and challenges of mesenchymal stem cells in medicine: an overview of the current knowledge. Stem Cells Int 2018;2018:5023925.
    1. Yun CW, Lee SH. Enhancement of functionality and therapeutic efficacy of cell-based therapy using mesenchymal stem cells for cardiovascular disease. Int J Mol Sci 2019;20:982.
    1. Zhao L, Chen S, Yang P, Cao H, Li L. The role of mesenchymal stem cells in hematopoietic stem cell transplantation: prevention and treatment of graft-versus-host disease. Stem Cell Res Ther 2019;10:182.
    1. Wang M, Yuan Q, Xie L. Mesenchymal stem cell-based immunomodulation: properties and clinical application. Stem Cells Int 2018;2018:3057624.
    1. Mukai T, Tojo A, Nagamura-Inoue T. Mesenchymal stromal cells as a potential therapeutic for neurological disorders. Regen Ther 2018;9:32–7.
    1. Cho J, D’Antuono M, Glicksman M, Wang J, Jonklaas J. A review of clinical trials: mesenchymal stem cell transplant therapy in type 1 and type 2 diabetes mellitus. Am J Stem Cells 2018;7:82–93.
    1. de Miguel MP, Prieto I, Moratilla A, Arias J, Aller MA. Mesenchymal stem cells for liver regeneration in liver failure: from experimental models to clinical trials. Stem Cells Int 2019;2019:3945672.
    1. Bochon B, Kozubska M, Surygala G, Witkowska A, Kuzniewicz R, Grzeszczak W, et al. Mesenchymal stem cells—potential applications in kidney diseases. Int J Mol Sci 2019;20:2462.
    1. Fan XL, Zhang Z, Ma CY, Fu QL. Mesenchymal stem cells for inflammatory airway disorders: promises and challenges. Biosci Rep 2019;39:20182160.
    1. Rivera-Izquierdo M, Cabeza L, Lainez-Ramos-Bossini A, Quesada R, Perazzoli G, Alvarez P, et al. An updated review of adipose-derived mesenchymal stem cells and their applications in musculoskeletal disorders. Expert Opin Biol Ther 2019;19:233–48.
    1. Toyserkani NM, Jorgensen MG, Tabatabaeifar S, Jensen CH, Sheikh SP, Sorensen JA. Concise review: a safety assessment of adipose-derived cell therapy in clinical trials: a systematic review of reported adverse events. Stem Cells Transl Med 2017;6:1786–94.
    1. Wagner W, Horn P, Castoldi M, Diehlmann A, Bork S, Saffrich R, et al. Replicative senescence of mesenchymal stem cells: a continuous and organized process. PLoS One 2008;3:e2213.
    1. Liu S, de Castro LF, Jin P, Civini S, Ren J, Reems JA, et al. Manufacturing differences affect human bone marrow stromal cell characteristics and function: comparison of production methods and products from multiple centers. Sci Rep 2017;7:46731.
    1. Torre ML, Lucarelli E, Guidi S, Ferrari M, Alessandri G, De Girolamo L, et al. Ex vivo expanded mesenchymal stromal cell minimal quality requirements for clinical application. Stem Cells Dev 2015;24:677–85.
    1. Frank NY, Pendse SS, Lapchak PH, Margaryan A, Shlain D, Doeing C, et al. Regulation of progenitor cell fusion by ABCB5 P-glycoprotein, a novel human ATP-binding cassette transporter. J Biol Chem 2003;278:47156–65.
    1. Schatton T, Yang J, Kleffel S, Uehara M, Barthel SR, Schlapbach C, et al. ABCB5 identifies immunoregulatory dermal cells. Cell Rep 2015;12:1564–74.
    1. Jiang D, Muschhammer J, Qi Y, Kugler A, de Vries JC, Saffarzadeh M, et al. Suppression of neutrophil-mediated tissue damage—a novel skill of mesenchymal stem cells. Stem Cells 2016;34:2393–406.
    1. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006;8: 315–7.
    1. Vander Beken S, de Vries JC, Meier-Schiesser B, Meyer P, Jiang D, Sindrilaru A, et al. Newly defined ATP-binding cassette subfamily B member 5 positive dermal mesenchymal stem cells promote healing of chronic iron-overload wounds via secretion of interleukin-1 receptor antagonist. Stem Cells 2019;37:1057–74.
    1. Hu MS, Borrelli MR, Lorenz HP, Longaker MT, Wan DC. Mesenchymal stromal cells and cutaneous wound healing: a comprehensive review of the background, role, and therapeutic potential. Stem Cells Int 2018;2018:6901983.
    1. Ojeh N, Pastar I, Tomic-Canic M, Stojadinovic O. Stem cells in skin regeneration, wound healing, and their clinical applications. Int J Mol Sci 2015;16:25476–501.
    1. Tappenbeck N, Schröder HM, Niebergall-Roth E, Hassinger F, Dehio U, Dieter K, et al. In vivo safety profile and biodistribution of GMP-manufactured human skin-derived ABCB5-positive mesenchymal stromal cells for use in clinical trials. Cytotherapy 2019;21:546–60.
    1. International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use. In: ICH Harmonised Tripartite Guideline Q5A(R1): Viral safety evaluation of biotechnology products derived from cell lines of human or animal origin; 1999. [accessed 4 May, 2020].
    1. Parliament European and Council of the European Union. Directive 2010/63/EU of the European Parliament and of the Council of 22 September 2010 on the protection of animals used for scientific purposes. Official Journal of the European Union 2010;L276:33–79.
    1. Miura M, Miura Y, Padilla-Nash HM, Molinolo AA, Fu B, Patel V, et al. Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. Stem Cells 2006;24:1095–103.
    1. Rosland GV, Svendsen A, Torsvik A, Sobala E, McCormack E, Immervoll H, et al. Long-term cultures of bone marrow-derived human mesenchymal stem cells frequently undergo spontaneous malignant transformation. Cancer Res 2009;69: 5331–9.
    1. Wang Y, Zhang Z, Chi Y, Zhang Q, Xu F, Yang Z, et al. Long-term cultured mesenchymal stem cells frequently develop genomic mutations but do not undergo malignant transformation. Cell Death Dis 2013;4:e950.
    1. Ueyama H, Horibe T, Hinotsu S, Tanaka T, Inoue T, Urushihara H, et al. Chromosomal variability of human mesenchymal stem cells cultured under hypoxic conditions. Journal Cell Mol Med 2012;16:72–82.
    1. Tarte K, Gaillard J, Lataillade JJ, Fouillard L, Becker M, Mossafa H, et al. Clinical-grade production of human mesenchymal stromal cells: occurrence of aneuploidy without transformation. Blood 2010;115:1549–53.
    1. Binato R, de Souza Fernandez T, Lazzarotto-Silva C, Du Rocher B, Mencalha A, Pizzatti L, et al. Stability of human mesenchymal stem cells during in vitro culture: considerations for cell therapy. Cell Prolif 2013;46:10–22.
    1. Stultz BG, McGinnis K, Thompson EE, Lo Surdo JL, Bauer SR, Hursh DA. Chromosomal stability of mesenchymal stromal cells during in vitro culture. Cytotherapy 2016;18:336–43.
    1. Ben Menachem-Zidon O, Gropp M, Ben Shushan E, Reubinoff B, Shveiky D. Systemically transplanted mesenchymal stem cells induce vascular-like structure formation in a rat model of vaginal injury. PLoS One 2019;14:e0218081.
    1. Chen J, Park HC, Addabbo F, Ni J, Pelger E, Li H, et al. Kidney-derived mesenchymal stem cells contribute to vasculogenesis, angiogenesis and endothelial repair. Kidney Int 2008;74:879–89.
    1. Yue WM, Liu W, Bi YW, He XP, Sun WY, Pang XY, et al. Mesenchymal stem cells differentiate into an endothelial phenotype, reduce neointimal formation, and enhance endothelial function in a rat vein grafting model. Stem Cells Dev 2008;17:785–93.
    1. Hu X, Yu SP, Fraser JL, Lu Z, Ogle ME, Wang JA, et al. Transplantation of hypoxia-preconditioned mesenchymal stem cells improves infarcted heart function via enhanced survival of implanted cells and angiogenesis. J Thorac Cardiovasc Surg 2008;135:799–808.
    1. Song YS, Lee HJ, Park IH, Kim WK, KU JH, Kim SU. Potential differentiation of human mesenchymal stem cell transplanted in rat corpus cavernosum toward endothelial or smooth muscle cells. Int J Impot Res 2007;19:378–85.
    1. Fu X, Fang L, Li X, Cheng B, Sheng Z. Enhanced wound-healing quality with bone marrow mesenchymal stem cells autografting after skin injury. Wound Repair Regen 2006;14:325–35.
    1. U.S. Department of Health and Human Services. In: Food and Drug Administration. Guidance for Industry: Chronic Cutaneous Ulcer and Burn Wounds—Developing Products for Treatment; 2006. [accessed 31 July, 2020].
    1. O’Donnell TF Jr., Passman MA, Marston WA, Ennis WJ, Dalsing M, Kistner RL, et al. Management of venous leg ulcers: clinical practice guidelines of the Society for Vascular Surgery and the American Venous Forum. J Vasc Surg 2014;60(suppl 2):3S–59S.
    1. Franks PJ, Barker J, Collier M, Gethin G, Haesler E, Jawien A, et al. Management of patients with venous leg ulcers: challenges and current best practice. J Wound Care 2016;25(suppl 6):S1–67.
    1. European Dermatology Forum. Evidence-based (S3) guidelines for diagnostics and treatment of venous leg ulcers. J Eur Acad Dermatol Venereol 2016;30:1843–75.
    1. Marston W, Tang J, Kirsner RS, Ennis W. Wound Healing Society 2015 update on guidelines for venous ulcers. Wound Repair Regen 2016;24:136–44.
    1. Sindrilaru A, Peters T, Wieschalka S, Baican C, Baican A, Peter H, et al. An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice. J Clin Invest 2011;121:985–97.
    1. An Y, Liu WJ, Xue P, Ma Y, Zhang LQ, Zhu B, et al. Autophagy promotes MSC-mediated vascularization in cutaneous wound healing via regulation of VEGF secretion. Cell Death Dis 2018;9:58.
    1. Tao H, Han Z, Han ZC, Li Z. Proangiogenic features of mesenchymal stem cells and their therapeutic applications. Stem Cells Int 2016;2016:1314709.
    1. Ge Q, Zhang H, Hou J, Wan L, Cheng W, Wang X, et al. VEGF secreted by mesenchymal stem cells mediates the differentiation of endothelial progenitor cells into endothelial cells via paracrine mechanisms. Mol Med Rep 2018;17:1667–75.
    1. Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol 2003;31:890–6.
    1. Klyushnenkova E, Mosca JD, Zernetkina V, Majumdar MK, Beggs KJ, Simonetti DW, et al. T cell responses to allogeneic human mesenchymal stem cells: immunogenicity, tolerance, and suppression. J Biomed Sci 2005;12: 47–57.
    1. Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol 2014;32:252–60.
    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;8:288.
    1. Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP, Suncion VY, et al. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA 2012;308:2369–79.
    1. Premer C, Blum A, Bellio MA, Schulman IH, Hurwitz BE, Parker M, et al. Allogeneic mesenchymal stem cells restore endothelial function in heart failure by stimulating endothelial progenitor cells. EBioMedicine 2015;2:467–75.
    1. Webber BR, O’Connor KT, McElmurry RT, Durgin EN, Eide CR, Lees CJ, et al. Rapid generation of Col7a1−/− mouse model of recessive dystrophic epidermolysis bullosa and partial rescue via immunosuppressive dermal mesenchymal stem cells. Lab Invest 2017;97:1218–24.
    1. Hartwig V, Dewidar B, Lin T, Dropmann A, Ganss C, Kluth MA, et al. Human skin-derived ABCB5+ stem cell injection improves liver disease parameters in Mdr2KO mice. Arch Toxicol 2019;93:2645–60.

Source: PubMed

3
Se inscrever