Intravenous allogeneic umbilical cord blood-derived mesenchymal stem cell therapy in recessive dystrophic epidermolysis bullosa patients

Sang Eun Lee, Seung-Ju Lee, Song-Ee Kim, Kinam Kim, Boyoung Cho, Kyounghwan Roh, Soo-Chan Kim, Sang Eun Lee, Seung-Ju Lee, Song-Ee Kim, Kinam Kim, Boyoung Cho, Kyounghwan Roh, Soo-Chan Kim

Abstract

BACKGROUNDRecessive dystrophic epidermolysis bullosa (RDEB) is an incurable disease that causes severe mucocutaneous fragility due to mutations in COL7A1 (encoding type VII collagen [C7]). In this phase I/IIa trial, we evaluated the safety and possible clinical efficacy of intravenous infusion of allogeneic human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) in patients with RDEB.METHODSFour adult and two pediatric patients with RDEB were treated with 3 intravenous injections of hUCB-MSCs (1 × 106 to 3 × 106 cells/kg) every 2 weeks and followed up for 8-24 months after treatment. The primary endpoint was safety. Secondary endpoints related to efficacy included clinical parameters, such as disease severity score, wound assessment, itch and pain score, and quality of life. C7 expression levels and inflammatory infiltrates in the skin, as well as serum levels of inflammatory markers and neuropeptides, were also assessed.RESULTSIntravenous hUCB-MSC infusions were well tolerated, without serious adverse events. Improvements in the Birmingham Epidermolysis Bullosa Severity Score, body surface area involvement, blister counts, pain, pruritus, and quality of life were observed with maximal effects at 56-112 days after treatment. hUCB-MSC administration induced M2 macrophage polarization and reduced mast cell infiltration in RDEB skin. Serum levels of substance P were decreased after therapy. Increased C7 expression was observed at the dermoepidermal junction in 1 of 6 patients at day 56.CONCLUSIONTo the best of our knowledge, this is the first clinical trial of systemic administration of allogeneic hUCB-MSCs in patients with RDEB, demonstrating safety and transient clinical benefits.TRIAL REGISTRATIONClinicalTrials.gov NCT04520022.FUNDINGThis work was supported by Daewoong Pharmaceutical Co. Ltd. and Kangstem Biotech Co. Ltd.

Keywords: Clinical Trials; Dermatology; Genetic diseases; Skin; Stem cell transplantation.

Conflict of interest statement

Conflict of interest: KR works in the Department of Clinical Development of Kangstem Biotech Co. Ltd., a biotechnology company focused on stem cell therapeutics. KK and BC work in the Cellular Therapeutics Team at Daewoong Pharmaceutical Co. Ltd.

Figures

Figure 1. Study design.
Figure 1. Study design.
(A) Flow chart for clinical trial and treatment use (expanded access to investigational drugs for treatment use). (B) Study design for hUCB-MSC treatment and evaluation.
Figure 2. Marked reduction in erythema and…
Figure 2. Marked reduction in erythema and erosions after hUCB-MSC treatment.
Photographs of a pediatric (A, subject 4) and an adult patient with RDEB (B, subject 1) at baseline and after 3 repeated injections of hUCB-MSCs.
Figure 3. Systemic treatment with hUCB-MSCs improved…
Figure 3. Systemic treatment with hUCB-MSCs improved clinical symptoms in patients with RDEB.
The time course of changes in disease severity (assessed by Birmingham Epidermolysis Bullosa Severity Score [BEBSS]), blister count, visual analog scale (VAS) pain score, and VAS pruritus score was assessed throughout the trial. For each parameter, a graphical representation of mean score per visit with range per visit was added. Two-tailed Student’s t test was performed for all the comparisons (n = 6). *P < 0.05. S, subject. Values are shown as the mean ± SEM.
Figure 4. Systemic treatment with hUCB-MSCs does…
Figure 4. Systemic treatment with hUCB-MSCs does not significantly affect the expression levels of C7 at the DEJ in most patients, except for in 1 patient (subject 1), who showed an increase in C7 expression at day 56.
(A) Representative immunofluorescence staining for type VII collagen (C7) using LH7.2, a monoclonal antibody that recognizes the NC1 domain of C7, on skin biopsy samples obtained before treatment (baseline) and at day 56 from patients with RDEB (subjects 1 and 5) receiving hUCB-MSC treatment. Scale bars: 20 μm. White arrows indicate C7 expression at the dermoepidermal junction (DEJ). (B) The intensity of staining for C7 expression along the DEJ was morphometrically quantitated as MFI using ImageJ (NIH). Values are shown as the mean ± SEM.
Figure 5. hUCB-MSC treatment modulates macrophage phenotype…
Figure 5. hUCB-MSC treatment modulates macrophage phenotype and mast cell infiltration in skin from patients with RDEB.
(A) Representative immunofluorescence staining for total macrophages (CD68), CD206+ macrophages, and mast cells (c-kit) on skin biopsy samples before treatment (baseline) and at day 56 for 6 matched pairs of patients with RDEB receiving hUCB-MSC treatment. Scale bars: 50 μm. (B) Mean total numbers of skin-infiltrating cells in biopsies from healthy controls (HCs) and RDEB skin at day 0 and at day 56 following hUCB-MSC treatment. By day 56, hUCB-MSC treatment markedly increased CD206+ macrophage counts and reduced mast cell counts. Values are shown as the mean ± SEM. Wilcoxon’s signed-rank test was performed for all the comparisons (n = 6). ***P < 0.001. S, subject.
Figure 6. hUCB-MSC treatment reduces serum substance…
Figure 6. hUCB-MSC treatment reduces serum substance P levels in patients with RDEB.
Serum levels of inflammatory cytokines (IL-1β and IL-6) and neuropeptides (substance P and CGRP) were assessed in healthy controls (HCs) and patients with RDEB (n = 6) at baseline and at day 56 following hUCB-MSC treatment. Values are shown as the mean ± SEM. Wilcoxon’s signed-rank test was used to assess the statistical difference between the repeated measurements in the same patient. *P < 0.05. S, subject.

References

    1. Has C, et al. Consensus reclassification of inherited epidermolysis bullosa and other disorders with skin fragility. Br J Dermatol. 2020;183(4):614–627. doi: 10.1111/bjd.18921.
    1. Fine JD, Mellerio JE. Extracutaneous manifestations and complications of inherited epidermolysis bullosa: part I. Epithelial associated tissues. J Am Acad Dermatol. 2009;61(3):367–384. doi: 10.1016/j.jaad.2009.03.052.
    1. Fine JD, Mellerio JE. Extracutaneous manifestations and complications of inherited epidermolysis bullosa: part II. Other organs. J Am Acad Dermatol. 2009;61(3):387–402. doi: 10.1016/j.jaad.2009.03.053.
    1. Feinstein JA, et al. Assessment of the timing of milestone clinical events in patients with epidermolysis bullosa from North America. JAMA Dermatol. 2019;155(2):196–203. doi: 10.1001/jamadermatol.2018.4673.
    1. Wagner JE, et al. Bone marrow transplantation for recessive dystrophic epidermolysis bullosa. N Engl J Med. 2010;363(7):629–639. doi: 10.1056/NEJMoa0910501.
    1. Petrof G, et al. Fibroblast cell therapy enhances initial healing in recessive dystrophic epidermolysis bullosa wounds: results of a randomized, vehicle-controlled trial. Br J Dermatol. 2013;169(5):1025–1033. doi: 10.1111/bjd.12599.
    1. Venugopal SS, et al. A phase II randomized vehicle-controlled trial of intradermal allogeneic fibroblasts for recessive dystrophic epidermolysis bullosa. J Am Acad Dermatol. 2013;69(6):898–908. doi: 10.1016/j.jaad.2013.08.014.
    1. Petrof G, et al. Potential of systemic allogeneic mesenchymal stromal cell therapy for children with recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 2015;135(9):2319–2321. doi: 10.1038/jid.2015.158.
    1. El-Darouti M, et al. Treatment of dystrophic epidermolysis bullosa with bone marrow non-hematopoeitic stem cells: a randomized controlled trial. Dermatol Ther. 2016;29(2):96–100. doi: 10.1111/dth.12305.
    1. Ganier C, et al. Intradermal injection of bone marrow mesenchymal stromal cells corrects recessive dystrophic epidermolysis bullosa in a xenograft model. J Invest Dermatol. 2018;138(11):2483–2486. doi: 10.1016/j.jid.2018.04.028.
    1. Liao Y, et al. Cord blood-derived stem cells suppress fibrosis and may prevent malignant progression in recessive dystrophic epidermolysis bullosa. Stem Cells. 2018;36(12):1839–1850. doi: 10.1002/stem.2907.
    1. Mellerio JE, Uitto J. Meeting report: the first global congress on epidermolysis bullosa, EB2020 London: toward treatment and cure. J Invest Dermatol. 2020;140(9):1681–1687. doi: 10.1016/j.jid.2020.05.078.
    1. Eichstadt S, et al. Phase 1/2a clinical trial of gene-corrected autologous cell therapy for recessive dystrophic epidermolysis bullosa. JCI Insight. 2019;4(19):130554.
    1. Rashidghamat E, et al. Phase I/II open-label trial of intravenous allogeneic mesenchymal stromal cell therapy in adults with recessive dystrophic epidermolysis bullosa. J Am Acad Dermatol. 2020;83(2):447–454. doi: 10.1016/j.jaad.2019.11.038.
    1. Liao Y, et al. Human cord blood-derived unrestricted somatic stem cells promote wound healing and have therapeutic potential for patients with recessive dystrophic epidermolysis bullosa. Cell Transplant. 2014;23(3):303–317. doi: 10.3727/096368913X663569.
    1. Liao Y, et al. Rescue of the mucocutaneous manifestations by human cord blood derived nonhematopoietic stem cells in a mouse model of recessive dystrophic epidermolysis bullosa. Stem Cells. 2015;33(6):1807–1817. doi: 10.1002/stem.1966.
    1. Zhang X, et al. Isolation and characterization of mesenchymal stem cells from human umbilical cord blood: reevaluation of critical factors for successful isolation and high ability to proliferate and differentiate to chondrocytes as compared to mesenchymal stem cells from bone marrow and adipose tissue. J Cell Biochem. 2011;112(4):1206–1218. doi: 10.1002/jcb.23042.
    1. Heo JS, et al. Comparison of molecular profiles of human mesenchymal stem cells derived from bone marrow, umbilical cord blood, placenta and adipose tissue. Int J Mol Med. 2016;37(1):115–125. doi: 10.3892/ijmm.2015.2413.
    1. Olson AL, McNiece IK. Novel clinical uses for cord blood derived mesenchymal stromal cells. Cytotherapy. 2015;17(6):796–802. doi: 10.1016/j.jcyt.2015.03.612.
    1. Berglund S, et al. Advances in umbilical cord blood cell therapy: the present and the future. Expert Opin Biol Ther. 2017;17(6):691–699. doi: 10.1080/14712598.2017.1316713.
    1. Huang X, et al. Past, present, and future efforts to enhance the efficacy of cord blood hematopoietic cell transplantation. F1000Res. 2019;8:F1000 Faculty Rev-1833.
    1. Kern S, et al. 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. Jin HJ, et al. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int J Mol Sci. 2013;14(9):17986–18001. doi: 10.3390/ijms140917986.
    1. Thompson M, 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.
    1. Mantovani A, et al. Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol. 2013;229(2):176–185. doi: 10.1002/path.4133.
    1. Wynn TA, Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity. 2016;44(3):450–462. doi: 10.1016/j.immuni.2016.02.015.
    1. Vasandan AB, et al. Human mesenchymal stem cells program macrophage plasticity by altering their metabolic status via a PGE2-dependent mechanism. Sci Rep. 2016;6:38308.
    1. He X, et al. MSC-derived exosome promotes M2 polarization and enhances cutaneous wound healing. Stem Cells Int. 2019;2019:7132708.
    1. Jin L, et al. Mesenchymal stem cells promote type 2 macrophage polarization to ameliorate the myocardial injury caused by diabetic cardiomyopathy. J Transl Med. 2019;17(1):251. doi: 10.1186/s12967-019-1999-8.
    1. Gupta K, Harvima IT. Mast cell-neural interactions contribute to pain and itch. Immunol Rev. 2018;282(1):168–187. doi: 10.1111/imr.12622.
    1. Siiskonen H, Harvima I. Mast cells and sensory nerves contribute to neurogenic inflammation and pruritus in chronic skin inflammation. Front Cell Neurosci. 2019;13:422.
    1. Petrof G, et al. Serum levels of high mobility group box 1 correlate with disease severity in recessive dystrophic epidermolysis bullosa. Exp Dermatol. 2013;22(6):433–435. doi: 10.1111/exd.12152.
    1. Annicchiarico G, et al. Proinflammatory cytokines and antiskin autoantibodies in patients with inherited epidermolysis bullosa. Medicine (Baltimore) 2015;94(42):e1528. doi: 10.1097/MD.0000000000001528.
    1. Esposito S, et al. Autoimmunity and cytokine imbalance in inherited epidermolysis bullosa. Int J Mol Sci. 2016;17(10):E1625.
    1. van Scheppingen C, et al. Main problems experienced by children with epidermolysis bullosa: a qualitative study with semi-structured interviews. Acta Derm Venereol. 2008;88(2):143–150. doi: 10.2340/00015555-0376.
    1. Goldschneider KR, Lucky AW. Pain management in epidermolysis bullosa. Dermatol Clin. 2010;28(2):273–282. doi: 10.1016/j.det.2010.01.008.
    1. Snauwaert JJ, et al. Burden of itch in epidermolysis bullosa. Br J Dermatol. 2014;171(1):73–78. doi: 10.1111/bjd.12885.
    1. doi: 10.1111/bjd.19496. Papanikolaou M, et al. Prevalence, pathophysiology and management of itch in epidermolysis bullosa [published online August 18, 2020]. Br J Dermatol .
    1. Jeon IK, et al. Quality of life and economic burden in recessive dystrophic epidermolysis bullosa. Ann Dermatol. 2016;28(1):6–14. doi: 10.5021/ad.2016.28.1.6.
    1. Lee JS, et al. Clinically important change in the visual analog scale after adequate pain control. Acad Emerg Med. 2003;10(10):1128–1130. doi: 10.1197/S1069-6563(03)00372-5.
    1. Reich A, et al. Itch assessment with visual analogue scale and numerical rating scale: determination of minimal clinically important difference in chronic itch. Acta Derm Venereol. 2016;96(7):978–980. doi: 10.2340/00015555-2433.
    1. Mack MR, et al. Peripheral neuro-immune pathology in recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 2015;135(4):1193–1197. doi: 10.1038/jid.2014.500.
    1. von Bischhoffshausen S, et al. Recessive dystrophic epidermolysis bullosa results in painful small fibre neuropathy. Brain. 2017;140(5):1238–1251. doi: 10.1093/brain/awx069.
    1. Agelopoulos K, et al. Neurokinin 1 receptor antagonists exhibit peripheral effects in prurigo nodularis including reduced ERK1/2 activation. J Eur Acad Dermatol Venereol. 2019;33(12):2371–2379. doi: 10.1111/jdv.15905.
    1. Kwatra SG, et al. Effects of neuroimmune axis modulation by aprepitant on antipruritic and global disease severity in patients with cutaneous T-cell lymphoma. Br J Dermatol. 2018;178(5):1221–1222. doi: 10.1111/bjd.16363.
    1. Chiou AS, et al. Phase 2 trial of a neurokinin-1 receptor antagonist for the treatment of chronic itch in patients with epidermolysis bullosa: a randomized clinical trial. J Am Acad Dermatol. 2020;82(6):1415–1421. doi: 10.1016/j.jaad.2019.09.014.
    1. Tappenbeck N, 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(5):546–560. doi: 10.1016/j.jcyt.2018.12.005.
    1. Park K-S, et al. Enhancement of therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res Ther. 2019;10(1):288. doi: 10.1186/s13287-019-1398-3.
    1. Qiu G, et al. Functional proteins of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res Ther. 2019;10(1):359. doi: 10.1186/s13287-019-1484-6.
    1. McBride JD, et al. Dual mechanism of type VII collagen transfer by bone marrow mesenchymal stem cell extracellular vesicles to recessive dystrophic epidermolysis bullosa fibroblasts. Biochimie. 2018;155:50–58. doi: 10.1016/j.biochi.2018.04.007.

Source: PubMed

3
Iratkozz fel