Clinical trial of ABCB5+ mesenchymal stem cells for recessive dystrophic epidermolysis bullosa

Dimitra Kiritsi, Kathrin Dieter, Elke Niebergall-Roth, Silvia Fluhr, Cristina Daniele, Jasmina Esterlechner, Samar Sadeghi, Seda Ballikaya, Leoni Erdinger, Franziska Schauer, Stella Gewert, Martin Laimer, Johann W Bauer, Alain Hovnanian, Giovanna Zambruno, May El Hachem, Emmanuelle Bourrat, Maria Papanikolaou, Gabriela Petrof, Sophie Kitzmüller, Christen L Ebens, Markus H Frank, Natasha Y Frank, Christoph Ganss, Anna E Martinez, John A McGrath, Jakub Tolar, Mark A Kluth, Dimitra Kiritsi, Kathrin Dieter, Elke Niebergall-Roth, Silvia Fluhr, Cristina Daniele, Jasmina Esterlechner, Samar Sadeghi, Seda Ballikaya, Leoni Erdinger, Franziska Schauer, Stella Gewert, Martin Laimer, Johann W Bauer, Alain Hovnanian, Giovanna Zambruno, May El Hachem, Emmanuelle Bourrat, Maria Papanikolaou, Gabriela Petrof, Sophie Kitzmüller, Christen L Ebens, Markus H Frank, Natasha Y Frank, Christoph Ganss, Anna E Martinez, John A McGrath, Jakub Tolar, Mark A Kluth

Abstract

BACKGROUNDRecessive dystrophic epidermolysis bullosa (RDEB) is a rare, devastating, and life-threatening inherited skin fragility disorder that comes about due to a lack of functional type VII collagen, for which no effective therapy exists. ABCB5+ dermal mesenchymal stem cells (ABCB5+ MSCs) possess immunomodulatory, inflammation-dampening, and tissue-healing capacities. In a Col7a1-/- mouse model of RDEB, treatment with ABCB5+ MSCs markedly extended the animals' lifespans.METHODSIn this international, multicentric, single-arm, phase I/IIa clinical trial, 16 patients (aged 4-36 years) enrolled into 4 age cohorts received 3 i.v. infusions of 2 × 106 ABCB5+ MSCs/kg on days 0, 17, and 35. Patients were followed up for 12 weeks regarding efficacy and 12 months regarding safety.RESULTSAt 12 weeks, statistically significant median (IQR) reductions in the Epidermolysis Bullosa Disease Activity and Scarring Index activity (EBDASI activity) score of 13.0% (2.9%-30%; P = 0.049) and the Instrument for Scoring Clinical Outcome of Research for Epidermolysis Bullosa clinician (iscorEB‑c) score of 18.2% (1.9%-39.8%; P = 0.037) were observed. Reductions in itch and pain numerical rating scale scores were greatest on day 35, amounting to 37.5% (0.0%-42.9%; P = 0.033) and 25.0% (-8.4% to 46.4%; P = 0.168), respectively. Three adverse events were considered related to the cell product: 1 mild lymphadenopathy and 2 hypersensitivity reactions. The latter 2 were serious but resolved without sequelae shortly after withdrawal of treatment.CONCLUSIONThis trial demonstrates good tolerability, manageable safety, and potential efficacy of i.v. ABCB5+ MSCs as a readily available disease-modifying therapy for RDEB and provides a rationale for further clinical evaluation.TRIAL REGISTRATIONClinicaltrials.gov NCT03529877; EudraCT 2018-001009-98.FUNDINGThe trial was sponsored by RHEACELL GmbH & Co. KG. Contributions by NYF and MHF to this work were supported by the NIH/National Eye Institute (NEI) grants RO1EY025794 and R24EY028767.

Keywords: Adult stem cells; Clinical Trials; Stem cells.

Figures

Figure 1. Study summary.
Figure 1. Study summary.
(A and B) Trial design and trial flow chart. ASigned the informed consent form. BFailed to attend the screening visit (due to poor general health, n = 1) or day 0 visit (due to travel restrictions associated with the COVID‑19 pandemic, n = 1). CPatient was prematurely withdrawn from treatment due to occurrence of a hypersensitivity reaction during the second cell infusion. FU, follow-up.
Figure 2. Representative photographs of patients at…
Figure 2. Representative photographs of patients at baseline (Day 0) and after 3 infusions of ABCB5+ MSCs (Week 12).
(A) Right lateral upper arm and right knee of a 24‑year-old Female patient (cohort 1). (B) Shoulder/neck area (back and front) and right hand of a 13‑year-old Female patient (cohort 2). (C) Right lateral upper arm and dorsum of the right foot of a 13‑year-old Female patient (cohort 2). (D) Back shoulder area of a 9‑year-old Male patient (cohort 3). All patients had consented to publication of their photographs.
Figure 3. Changes in EBDASI.
Figure 3. Changes in EBDASI.
(A) Percent changes in the EBDASI overall score and total activity and damage subscores at 12 weeks (with the last observation carried forward [LOCF] in cases of missing data), expressed as percentage of the baseline value, in the full analysis set (FAS) and the per-protocol set (PP). (B) Percent changes in the EBDASI activity score by visit, expressed as percentage of the baseline value, in the FAS (no LOCF). Data are shown as medians with IQR; P values (2-sided Wilcoxon signed rank test) indicate statistical significance of changes from baseline. Kruskal-Wallis tests followed by Dunn’s multiple comparison tests revealed no statistically significant differences between the 3 postbaseline visits (day 17, day 35, and week 12; P > 0.05). For EBDASI overall and damage score data, see Supplemental Table 1.
Figure 4. Changes in iscorEB.
Figure 4. Changes in iscorEB.
(A) Percent changes in the iscorEB overall score and iscorEB‑c and iscorEB‑p subscores at 12 weeks, expressed as percentage of the baseline value, in the full analysis set (FAS) and in the per-protocol set (PP). The lower number of data points for iscorEB overall and iscorEB‑c as compared with the iscorEB‑p is due to difficulties with blood sampling; for these patients, the lab values (anemia, albumin, inflammation) required for calculation of iscorEB overall and iscorEB‑c could not be obtained. (B) Percent changes in the iscorEB‑c score by visit, expressed as percentage of the baseline value, in the FAS. Data are shown as medians with IQR; P values (2‑sided Wilcoxon signed rank test) indicate statistical significance of changes from baseline. Kruskal-Wallis tests followed by Dunn’s multiple comparison tests revealed no statistically significant differences between the 3 postbaseline visits (day 17, day 35, and week 12; P > 0.05). For iscorEB overall and iscorEB‑p data, see Supplemental Table 2.
Figure 5. Changes in itch, pain, and…
Figure 5. Changes in itch, pain, and impact of RDEB on life quality in the full analysis set (FAS).
(AC) Changes in: itch score, pain score, and QOLEB score, expressed as percentage of the baseline value. The lower number of data points for the pain score as compared with itch and QOLEB scores at the postbaseline visits (day 17, day 35, week 12) is caused by 2 patients presenting with pain score = 0 at baseline; therefore, for these patients, percent changes from baseline could not be calculated at any postbaseline visit. Please note that the patient who presented with an extreme percent increase in QOLEB score on day 35 and at week 12 (with score category changing from mild [day 0] to very mild [day 17] to moderate [day 35 and week 12]) had received only an incomplete second cell dose (day 17) and no third cell dose (day 35). Data are shown as medians with IQR; P values (2‑sided Wilcoxon signed rank test) indicate statistical significance of changes from baseline. Kruskal-Wallis tests followed by Dunn’s multiple comparison tests revealed no statistically significant differences between the 3 postbaseline visits (day 17, day 35, and week 12; P > 0.05). For the data of the per-protocol set, see Supplemental Figure 2.
Figure 6. HMGB1 serum concentrations.
Figure 6. HMGB1 serum concentrations.
Each color represents an individual patient. Data are shown as medians with IQR. Kruskal-Wallis tests followed by Dunn’s multiple comparison tests revealed no statistically significant differences between visits (P > 0.05).

References

    1. Has C, et al. Epidermolysis bullosa: molecular pathology of connective tissue components in the cutaneous basement membrane zone. Matrix Biol. 2018;71–72:313–329.
    1. Bardhan A, et al. Epidermolysis bullosa. Nat Rev Dis Primers. 2020;6(1):78. doi: 10.1038/s41572-020-0210-0.
    1. Adam MP, et al, eds. GeneReviews®. University of Washington; 2006.
    1. Li AW, et al. Inpatient management of children with recessive dystrophic epidermolysis bullosa: a review. Pediatr Dermatol. 2017;34(6):647–655. doi: 10.1111/pde.13276.
    1. Condorelli AG, et al. Epidermolysis bullosa-associated squamous cell carcinoma: from pathogenesis to therapeutic perspectives. Int J Mol Sci. 2019;20(22):5707. doi: 10.3390/ijms20225707.
    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. Pillay E, Clapham J. Development of best clinical practice guidelines for epidermolysis bullosa. Wounds International. 2018;9(4):20–27.
    1. Togo CCG, et al. Quality of life in people with epidermolysis bullosa: a systematic review. Qual Life Res. 2020;29(7):1731–1745. doi: 10.1007/s11136-020-02495-5.
    1. Rashidghamat E, McGrath JA. Novel and emerging therapies in the treatment of recessive dystrophic epidermolysis bullosa. Intractable Rare Dis Res. 2017;6(1):6–20. doi: 10.5582/irdr.2017.01005.
    1. Dourado Alcorte M, et al. Patent landscape of molecular and cellular targeted therapies for recessive dystrophic epidermolysis bullosa. Expert Opin Ther Pat. 2019;29(5):327–337. doi: 10.1080/13543776.2019.1608181.
    1. Wally V, et al. Small molecule drug development for rare genodermatoses — evaluation of the current status in epidermolysis bullosa. Orphanet J Rare Dis. 2020;15(1):292. doi: 10.1186/s13023-020-01467-9.
    1. Titeux M, et al. Emerging drugs for the treatment of epidermolysis bullosa. Expert Opin Emerg Drugs. 2020;25(4):467–489. doi: 10.1080/14728214.2020.1839049.
    1. Uitto J, et al. EB2017-progress in epidermolysis bullosa research toward treatment and cure. J Invest Dermatol. 2018;138(5):1010–1016. doi: 10.1016/j.jid.2017.12.016.
    1. Perdoni C, et al. Gene editing toward the use of autologous therapies in recessive dystrophic epidermolysis bullosa. Transl Res. 2016;168:50–58. doi: 10.1016/j.trsl.2015.05.008.
    1. Marinkovich MP, Tang JY. Gene therapy for epidermolysis bullosa. J Invest Dermatol. 2019;139(6):1221–1226. doi: 10.1016/j.jid.2018.11.036.
    1. Siprashvili Z, et al. Safety and wound outcomes following genetically corrected autologous epidermal grafts in patients with recessive dystrophic epidermolysis bullosa. JAMA. 2016;316(17):1808–1817. doi: 10.1001/jama.2016.15588.
    1. Lwin SM, et al. Safety and early efficacy outcomes for lentiviral fibroblast gene therapy in recessive dystrophic epidermolysis bullosa. JCI Insight. 2019;4(11):126243.
    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. Kiritsi D, Nyström A. Recent advances in understanding and managing epidermolysis bullosa. F1000Res. 2018;7:1097. doi: 10.12688/f1000research.14974.1.
    1. Bruckner-Tuderman L. Newer treatment modalities in epidermolysis bullosa. Indian Dermatol Online J. 2019;10(3):244–250. doi: 10.4103/idoj.IDOJ_287_18.
    1. Bruckner-Tuderman L. Skin fragility: perspectives on evidence-based therapies. Acta Derm Venereol. 2020;100(5):adv00053. doi: 10.2340/00015555-3398.
    1. Breitenbach JS, et al. Transcriptome and ultrastructural changes in dystrophic epidermolysis bullosa resemble skin aging. Aging (Albany NY) 2015;7(6):389–411.
    1. Cianfarani F, et al. Pathomechanisms of altered wound healing in recessive dystrophic epidermolysis bullosa. Am J Pathol. 2017;187(7):1445–1453. doi: 10.1016/j.ajpath.2017.03.003.
    1. Yamanaka K, et al. Persistent release of IL-1s from skin is associated with systemic cardio-vascular disease, emaciation and systemic amyloidosis: the potential of anti-IL-1 therapy for systemic inflammatory diseases. PLoS One. 2014;9(8):e104479. doi: 10.1371/journal.pone.0104479.
    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):1625. doi: 10.3390/ijms17101625.
    1. Annicchiarico G, et al. Canakinumab in recessive dystrophic epidermolysis bullosa: a novel unexpected weapon for non-healing wounds? Clin Exp Rheumatol. 2016;34(5):961–962.
    1. Ebens CL, et al. Bone marrow transplant with post-transplant cyclophosphamide for recessive dystrophic epidermolysis bullosa expands the related donor pool and permits tolerance of nonhaematopoietic cellular grafts. Br J Dermatol. 2019;181(6):1238–1246. doi: 10.1111/bjd.17858.
    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. 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. 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. Maseda R, et al. Beneficial effect of systemic allogeneic adipose derived mesenchymal cells on the clinical, inflammatory and immunologic status of a patient with recessive dystrophic epidermolysis bullosa: a case report. Front Med (Lausanne) 2020;7:576558.
    1. Lee SE, et al. Intravenous allogeneic umbilical cord blood-derived mesenchymal stem cell therapy in recessive dystrophic epidermolysis bullosa patients. JCI Insight. 2021;6(2):143606.
    1. Alexeev V, et al. Chemotaxis-driven disease-site targeting of therapeutic adult stem cells in dystrophic epidermolysis bullosa. Stem Cell Res Ther. 2016;7(1):124. doi: 10.1186/s13287-016-0388-y.
    1. Alexeev V, et al. Pro-inflammatory chemokines and cytokines dominate the blister fluid molecular signature in patients with epidermolysis bullosa and affect leukocyte and stem cell migration. J Invest Dermatol. 2017;137(11):2298–2308. doi: 10.1016/j.jid.2017.07.002.
    1. Jiang D, Scharffetter-Kochanek K. Mesenchymal stem cells adaptively respond to environmental cues thereby improving granulation tissue formation and wound healing. Front Cell Dev Biol. 2020;8:697. doi: 10.3389/fcell.2020.00697.
    1. Harrell CR, et al. The role of interleukin 1 receptor antagonist in mesenchymal stem cell-based tissue repair and regeneration. Biofactors. 2020;46(2):263–275. doi: 10.1002/biof.1587.
    1. Schatton T, et al. ABCB5 identifies immunoregulatory dermal cells. Cell Rep. 2015;12(10):1564–1574. doi: 10.1016/j.celrep.2015.08.010.
    1. Jiang D, et al. Suppression of neutrophil-mediated tissue damage-a novel skill of mesenchymal stem cells. Stem Cells. 2016;34(9):2393–2406. doi: 10.1002/stem.2417.
    1. Vander Beken S, 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(8):1057–1074. doi: 10.1002/stem.3022.
    1. Frank NY, et al. Regulation of progenitor cell fusion by ABCB5 P-glycoprotein, a novel human ATP-binding cassette transporter. J Biol Chem. 2003;278(47):47156–47165. doi: 10.1074/jbc.M308700200.
    1. Kerstan A, et al. 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. Cytotherapy. 2021;23(2):165–175. doi: 10.1016/j.jcyt.2020.08.012.
    1. Kerstan A et al. Allogeneic ABCB5+ mesenchymal stem cells for treatment-refractory chronic venous ulcers: a phase I/IIa clinical trial. JID Innov.
    1. Webber BR, 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(10):1218–1224. doi: 10.1038/labinvest.2017.85.
    1. Ballikaya S, et al. Process data of allogeneic ex vivo-expanded ABCB5+ mesenchymal stromal cells for human use: off-the-shelf GMP-manufactured donor-independent ATMP. Stem Cell Res Ther. 2020;11(1):482. doi: 10.1186/s13287-020-01987-y.
    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. Loh CC, et al. Development, reliability, and validity of a novel epidermolysis bullosa disease activity and scarring index (EBDASI) J Am Acad Dermatol. 2014;70(1):89–97. doi: 10.1016/j.jaad.2013.09.041.
    1. Jain SV, et al. The epidermolysis bullosa disease activity and scarring index (EBDASI): grading disease severity and assessing responsiveness to clinical change in epidermolysis bullosa. J Eur Acad Dermatol Venereol. 2017;31(4):692–698. doi: 10.1111/jdv.13953.
    1. Schwieger-Briel A, et al. Instrument for scoring clinical outcome of research for epidermolysis bullosa: a consensus-generated clinical research tool. Pediatr Dermatol. 2015;32(1):41–52. doi: 10.1111/pde.12317.
    1. Bruckner AL, et al. Reliability and validity of the instrument for scoring clinical outcomes of research for epidermolysis bullosa (iscorEB) Br J Dermatol. 2018;178(5):1128–1134. doi: 10.1111/bjd.16350.
    1. Frew JW, et al. Quality of life evaluation in epidermolysis bullosa (EB) through the development of the QOLEB questionnaire: an EB-specific quality of life instrument. Br J Dermatol. 2009;161(6):1323–1330. doi: 10.1111/j.1365-2133.2009.09347.x.
    1. Kot M, Baj-Krzyworzeka M, Szatanek R, Musiał-Wysocka A, Suda-Szczurek M, Majka M. The Importance of HLA Assessment in “Off-the-Shelf” Allogeneic Mesenchymal Stem Cells Based-Therapies. Int J Mol Sci. 2019;20(22):E5680.
    1. Rogers CL, et al. A comparison study of outcome measures for epidermolysis bullosa: epidermolysis bullosa disease activity and scarring index (EBDASI) and the instrument for scoring clinical outcomes of research for epidermolysis bullosa (iscorEB) JAAD Int. 2021;2:134–152. doi: 10.1016/j.jdin.2020.12.007.
    1. Fine JD, et al. Assessment of mobility, activities and pain in different subtypes of epidermolysis bullosa. Clin Exp Dermatol. 2004;29(2):122–127. doi: 10.1111/j.1365-2230.2004.01428.x.
    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. 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. 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. Danial C, et al. Prevalence and characterization of pruritus in epidermolysis bullosa. Pediatr Dermatol. 2015;32(1):53–59. doi: 10.1111/pde.12391.
    1. Papanikolaou M, et al. Prevalence, pathophysiology and management of itch in epidermolysis bullosa. Br J Dermatol. 2021;184(5):816–825. doi: 10.1111/bjd.19496.
    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. Gorgulho CM, et al. Johnny on the spot-chronic inflammation is driven by HMGB1. Front Immunol. 2019;10:1561. doi: 10.3389/fimmu.2019.01561.
    1. Yang H, et al. Targeting inflammation driven by HMGB1. Front Immunol. 2020;11:484. doi: 10.3389/fimmu.2020.00484.
    1. Moss C, et al. The Birmingham epidermolysis bullosa severity score: development and validation. Br J Dermatol. 2009;160(5):1057–1065. doi: 10.1111/j.1365-2133.2009.09041.x.
    1. Caplan AI. Cell-based therapies: the nonresponder. Stem Cells Transl Med. 2018;7(11):762–766. doi: 10.1002/sctm.18-0074.
    1. Levy O, et al. Shattering barriers toward clinically meaningful MSC therapies. Sci Adv. 2020;6(30):eaba6884. doi: 10.1126/sciadv.aba6884.
    1. Nolta JA, et al. Improving mesenchymal stem/stromal cell potency and survival: proceedings from the international society of cell therapy (ISCT) MSC preconference held in May 2018, Palais des Congres de Montreal, organized by the ISCT MSC scientific committee. Cytotherapy. 2020;22(3):123–126. doi: 10.1016/j.jcyt.2020.01.004.
    1. Lalu MM, et al. Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials. PLoS One. 2012;7(10):e47559. doi: 10.1371/journal.pone.0047559.
    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. doi: 10.1016/j.eclinm.2019.100249.
    1. Griffin MD, et al. Anti-donor immune responses elicited by allogeneic mesenchymal stem cells: what have we learned so far? Immunol Cell Biol. 2013;91(1):40–51. doi: 10.1038/icb.2012.67.
    1. Ankrum JA, et al. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol. 2014;32(3):252–260. doi: 10.1038/nbt.2816.
    1. Packham DK, et al. Allogeneic mesenchymal precursor cells (MPC) in diabetic nephropathy: a randomized, placebo-controlled, dose escalation study. EBioMedicine. 2016;12:263–269. doi: 10.1016/j.ebiom.2016.09.011.
    1. Skyler JS, et al. Allogeneic mesenchymal precursor cells in type 2 diabetes: a randomized, placebo-controlled, dose-escalation safety and tolerability pilot study. Diabetes Care. 2015;38(9):1742–1749. doi: 10.2337/dc14-2830.
    1. Álvaro-Gracia JM, et al. Intravenous administration of expanded allogeneic adipose-derived mesenchymal stem cells in refractory rheumatoid arthritis (Cx611): results of a multicentre, dose escalation, randomised, single-blind, placebo-controlled phase Ib/IIa clinical trial. Ann Rheum Dis. 2017;76(1):196–202. doi: 10.1136/annrheumdis-2015-208918.
    1. Syme R, et al. The role of depletion of dimethyl sulfoxide before autografting: on hematologic recovery, side effects, and toxicity. Biol Blood Marrow Transplant. 2004;10(2):135–141. doi: 10.1016/j.bbmt.2003.09.016.
    1. Windrum P, et al. Variation in dimethyl sulfoxide use in stem cell transplantation: a survey of EBMT centres. Bone Marrow Transplant. 2005;36(7):601–603. doi: 10.1038/sj.bmt.1705100.
    1. Laxenaire MC, et al. [Anaphylactoid reactions to colloid plasma substitutes: incidence, risk factors, mechanisms. A French multicenter prospective study] Ann Fr Anesth Reanim. 1994;13(3):301–310. doi: 10.1016/S0750-7658(94)80038-3.
    1. Riedl J, et al. ABCB5+ dermal mesenchymal stromal cells with favorable skin homing and local immunomodulation for recessive dystrophic epidermolysis bullosa treatment. Stem Cells. 2021;39(7):897–903. doi: 10.1002/stem.3356.

Source: PubMed

3
購読する