Human umbilical cord mesenchymal stem cells for psoriasis: a phase 1/2a, single-arm study

Lamei Cheng, Siqi Wang, Cong Peng, Xiao Zou, Chao Yang, Hua Mei, Chuang Li, Xian Su, Na Xiao, Qi Ouyang, Mi Zhang, Qiaolin Wang, Yan Luo, Minxue Shen, Qun Qin, Honglin Wang, Wu Zhu, Guangxiu Lu, Ge Lin, Yehong Kuang, Xiang Chen, Lamei Cheng, Siqi Wang, Cong Peng, Xiao Zou, Chao Yang, Hua Mei, Chuang Li, Xian Su, Na Xiao, Qi Ouyang, Mi Zhang, Qiaolin Wang, Yan Luo, Minxue Shen, Qun Qin, Honglin Wang, Wu Zhu, Guangxiu Lu, Ge Lin, Yehong Kuang, Xiang Chen

Abstract

Psoriasis is a common, chronic immune-mediated systemic disease that had no effective and durable treatment. Mesenchymal stem cells (MSCs) have immunomodulatory properties. Therefore, we performed a phase 1/2a, single-arm clinical trial to evaluate the safety and efficacy of human umbilical cord-derived MSCs (UMSCs) in the treatment of psoriasis and to preliminarily explore the possible mechanisms. Seventeen patients with psoriasis were enrolled and received UMSC infusions. Adverse events, laboratory parameters, PASI, and PGA were analyzed. We did not observe obvious side effects during the treatment and 6-month follow-up. A total of 47.1% (8/17) of the psoriasis patients had at least 40% improvement in the PASI score, and 17.6% (3/17) had no sign of disease or minimal disease based on the PGA score. And the efficiency was 25% (2/8) for males and 66.7% (6/9) for females. After UMSC transplantation (UMSCT), the frequencies of Tregs and CD4+ memory T cells were significantly increased, and the frequencies of T helper (Th) 17 and CD4+ naive T cells were significantly decreased in peripheral blood (PB) of psoriasis patients. And all responders showed significant increases in Tregs and CD4+ memory T cells, and significant decreases in Th17 cells and serum IL-17 level after UMSCT. And baseline level of Tregs in responders were significantly lower than those in nonresponders. In conclusion, allogeneic UMSCT is safe and partially effective in psoriasis patients, and level of Tregs may be used as a potent biomarker to predict the clinical efficacy of UMSCT. Trial registration Clinical Trials NCT03765957.

Conflict of interest statement

The authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Flowchart of the study
Fig. 2
Fig. 2
The figure showed three outcome measures of the 6 responders including PASI score, BSA and PGA. a PASI score; b BSA; c PGA
Fig. 3
Fig. 3
Representative skin images of 3 patients before and after treatment
Fig. 4
Fig. 4
Frequencies and numbers of T lymphocyte subsets in PB of HC and patients with psoriasis pre- and post-UMSCT. a Frequencies and numbers of CD4+ memory T cells (CD45RO+ in CD3+CD4+). b Frequencies and numbers of Tregs (CD25+CD127-/low in CD3+CD4+). c Frequencies and numbers of CD8+ TCM cells (CCR7+CD45RA- in CD3+CD8+). (HC, n = 15; Patients, n = 17; ns, no significant, VS HC; *P < 0.05, **P < 0.01, ***P < 0.001 VS Pre-UMSCT); d Frequencies of Th1, Th2, Th17 and the ratio of Th1/Th2. (HC, n = 15; Patients, n = 17; *P < 0.05, VS HC; ns, no significant, VS Pre-UMSCT
Fig. 5
Fig. 5
The inflammatory factors in the serum in HC and psoriasis patients pre- and post-UMSCT. a Serum TNF-α concentration. b Serum IL-6 concentration. c Serum IL-1β concentration. d Serum IL-17 concentration in HC, all psoriasis patients and responders pre- and post-UMSCT. (n = 17, **P < 0.01, ***P < 0.001, vs Pre-UMSCT)
Fig. 6
Fig. 6
Baseline immune indices in PB in response group and no-response group. a Baseline level of CD4+TCM cells. b Baseline level of Tregs. c Baseline level of the ratio of Treg/Th17 (response group: n = 6, no-response group: n = 11, *P < 0.05; **P < 0.01)
Fig. 7
Fig. 7
The changes in T lymphocyte subsets in responders pre- and post-UMSCT. ae Changes of frequencies and numbers of CD4 naive T, CD4+ TCM, CD4+ memory T, Treg and CD8+ TCM cells in PB in responders (n = 6, *P < 0.05, **P < 0.01, ***P < 0.001); fg Changes of frequencies of Th1, Th2, Th17 and Th1/Th2 and Treg/Th17 in PB in responders (n = 6, *P < 0.05, **P < 0.01)

References

    1. Parisi R, et al. National, regional, and worldwide epidemiology of psoriasis: systematic analysis and modelling study. BMJ. 2020;369:m1590. doi: 10.1136/bmj.m1590.
    1. Ding X, et al. Prevalence of psoriasis in China: a population-based study in six cities. Eur. J. Dermatol. 2012;22:663–667. doi: 10.1684/ejd.2012.1802.
    1. Chen Y, et al. Human umbilical cord-derived mesenchymal stem cells ameliorate psoriasis-like dermatitis by suppressing IL-17-producing gammadelta T cells. Cell Tissue Res. 2022;388:549–563. doi: 10.1007/s00441-022-03616-x.
    1. Griffiths CEM, Armstrong AW, Gudjonsson JE, Barker JNWN. Psoriasis. Lancet. 2021;397:1301–1315. doi: 10.1016/S0140-6736(20)32549-6.
    1. Papp KA, et al. Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis. N. Engl. J. Med. 2012;366:1181–1189. doi: 10.1056/NEJMoa1109017.
    1. Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immunomodulation: mechanisms and therapeutic potential. Trends Pharmacol. Sci. 2020;41:653–664. doi: 10.1016/j.tips.2020.06.009.
    1. DelaRosa O, et al. Requirement of IFN-gamma-mediated indoleamine 2,3-dioxygenase expression in the modulation of lymphocyte proliferation by human adipose-derived stem cells. Tissue Eng. Part A. 2009;15:2795–2806. doi: 10.1089/ten.tea.2008.0630.
    1. Nemeth K, et al. Bone marrow stromal cells use TGF-beta to suppress allergic responses in a mouse model of ragweed-induced asthma. Proc. Natl Acad. Sci. USA. 2010;107:5652–5657. doi: 10.1073/pnas.0910720107.
    1. Ortiz LA, et al. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc. Natl Acad. Sci. USA. 2007;104:11002–11007. doi: 10.1073/pnas.0704421104.
    1. Rozenberg A, et al. Human mesenchymal stem cells impact Th17 and Th1 responses through a prostaglandin E2 and myeloid-dependent mechanism. Stem Cells Transl. Med. 2016;5:1506–1514. doi: 10.5966/sctm.2015-0243.
    1. Wang D, et al. The regulation of the Treg/Th17 balance by mesenchymal stem cells in human systemic lupus erythematosus. Cell Mol. Immunol. 2017;14:423–431. doi: 10.1038/cmi.2015.89.
    1. Gonzalez MA, Gonzalez-Rey E, Rico L, Buscher D, Delgado M. Treatment of experimental arthritis by inducing immune tolerance with human adipose-derived mesenchymal stem cells. Arthritis Rheum. 2009;60:1006–1019. doi: 10.1002/art.24405.
    1. Xu J, et al. Allogeneic mesenchymal stem cell treatment alleviates experimental and clinical Sjogren syndrome. Blood. 2012;120:3142–3151. doi: 10.1182/blood-2011-11-391144.
    1. Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 2014;14:195–208. doi: 10.1038/nri3622.
    1. Chen M, et al. Mesenchymal stem cells alleviate moderate-to-severe psoriasis by reducing the production of type i interferon (IFN-I) by plasmacytoid dendritic cells (pDCs) Stem Cells Int. 2019;2019:6961052.
    1. Cai J, et al. Umbilical cord mesenchymal stromal cell with autologous bone marrow cell transplantation in established type 1 diabetes: a pilot randomized controlled open-label clinical study to assess safety and impact on insulin secretion. Diabetes Care. 2016;39:149–157. doi: 10.2337/dc15-0171.
    1. Park EH, et al. Intravenous infusion of umbilical cord blood-derived mesenchymal stem cells in rheumatoid arthritis: a phase ia clinical trial. Stem Cells Transl. Med. 2018;7:636–642. doi: 10.1002/sctm.18-0031.
    1. Wang D, et al. Umbilical cord mesenchymal stem cell transplantation in active and refractory systemic lupus erythematosus: a multicenter clinical study. Arthritis Res Ther. 2014;16:R79. doi: 10.1186/ar4520.
    1. Paganelli A, Tarentini E, Benassi L, Kaleci S, Magnoni C. Mesenchymal stem cells for the treatment of psoriasis: a comprehensive review. Clin. Exp. Dermatol. 2020;45:824–830. doi: 10.1111/ced.14269.
    1. Lwin SM, Snowden JA, Griffiths CEM. The promise and challenges of cell therapy for psoriasis. Br. J. Dermatol. 2021;185:887–898. doi: 10.1111/bjd.20517.
    1. De Jesus MM, et al. Autologous adipose-derived mesenchymal stromal cells for the treatment of psoriasis vulgaris and psoriatic arthritis: a case report. Cell Transplant. 2016;25:2063–2069. doi: 10.3727/096368916X691998.
    1. Chen H, et al. Treatment of psoriasis with mesenchymal stem cells. Am. J. Med. 2016;129:e13–e14. doi: 10.1016/j.amjmed.2015.11.001.
    1. Saad A, et al. Autologous mesenchymal stem cells increase cortical perfusion in renovascular disease. J. Am. Soc. Nephrol. 2017;28:2777–2785. doi: 10.1681/ASN.2017020151.
    1. Ahn SY, Chang YS, Sung SI, Park WS. Mesenchymal stem cells for severe intraventricular hemorrhage in preterm infants: phase I dose-escalation clinical trial. Stem Cells Transl. Med. 2018;7:847–856. doi: 10.1002/sctm.17-0219.
    1. Nussbaum L, Chen YL, Ogg GS. Role of regulatory T cells in psoriasis pathogenesis and treatment. Br. J. Dermatol. 2021;184:14–24. doi: 10.1111/bjd.19380.
    1. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133:775–787. doi: 10.1016/j.cell.2008.05.009.
    1. Quaglino P, et al. Circulating CD4+CD25 bright FOXP3+ T cells are up-regulated by biological therapies and correlate with the clinical response in psoriasis patients. Dermatology. 2009;219:250–258. doi: 10.1159/000238305.
    1. Priyadarssini M, Chandrashekar L, Rajappa M. Effect of methotrexate monotherapy on T-cell subsets in the peripheral circulation in psoriasis. Clin. Exp. Dermatol. 2019;44:491–497. doi: 10.1111/ced.13795.
    1. Gregori S, et al. Regulatory T cells induced by 1 alpha,25-dihydroxyvitamin D3 and mycophenolate mofetil treatment mediate transplantation tolerance. J. Immunol. 2001;167:1945–1953. doi: 10.4049/jimmunol.167.4.1945.
    1. Zhang L, et al. Characterization of Th17 and FoxP3(+) Treg cells in paediatric psoriasis patients. Scand. J. Immunol. 2016;83:174–180. doi: 10.1111/sji.12404.
    1. Zhang L, Yang XQ, Cheng J, Hui RS, Gao TW. Increased Th17 cells are accompanied by FoxP3(+) Treg cell accumulation and correlated with psoriasis disease severity. Clin. Immunol. 2010;135:108–117. doi: 10.1016/j.clim.2009.11.008.
    1. Yang Y, et al. Serum IFN-gamma levels predict the therapeutic effect of mesenchymal stem cell transplantation in active rheumatoid arthritis. J. Transl. Med. 2018;16:165. doi: 10.1186/s12967-018-1541-4.
    1. Duffy MM, Ritter T, Ceredig R, Griffin MD. Mesenchymal stem cell effects on T-cell effector pathways. Stem Cell Res Ther. 2011;2:34. doi: 10.1186/scrt75.
    1. Bolandi Z, et al. Adipose derived mesenchymal stem cell exosomes loaded with miR-10a promote the differentiation of Th17 and Treg from naive CD4(+) T cell. Life Sci. 2020;259:118218. doi: 10.1016/j.lfs.2020.118218.
    1. Facciotti F, et al. Evidence for a pathogenic role of extrafollicular, IL-10-producing CCR6(+)B helper T cells in systemic lupus erythematosus. Proc. Natl Acad. Sci. USA. 2020;117:7305–7316. doi: 10.1073/pnas.1917834117.
    1. Li R, et al. Proinflammatory GM-CSF-producing B cells in multiple sclerosis and B cell depletion therapy. Sci. Transl. Med. 2015;7:310ra166.

Source: PubMed

3
Předplatit