Safety, immunological effects and clinical response in a phase I trial of umbilical cord mesenchymal stromal cells in patients with treatment refractory SLE

Diane L Kamen, Caroline Wallace, Zihai Li, Megan Wyatt, Crystal Paulos, Chungwen Wei, Hongjun Wang, Bethany J Wolf, Paul J Nietert, Gary Gilkeson, Diane L Kamen, Caroline Wallace, Zihai Li, Megan Wyatt, Crystal Paulos, Chungwen Wei, Hongjun Wang, Bethany J Wolf, Paul J Nietert, Gary Gilkeson

Abstract

Background: Reports of clinical improvement following mesenchymal stromal cell (MSC) infusions in refractory lupus patients at a single centre in China led us to perform an explorative phase I trial of umbilical cord derived MSCs in patients refractory to 6 months of immunosuppressive therapy.

Methods: Six women with a SLEDAI >6, having failed standard of care therapy, received one intravenous infusion of 1×106 MSCs/kg of body weight. They maintained their current immunosuppressives, but their physician was allowed to adjust corticosteroids initially for symptom management. The clinical endpoint was an SRI of 4 with no new British Isles Lupus Activity Guide (BILAG) As and no increase in Physician Global Assessment score of >0.3 with tapering of prednisone to 10 mg or less by 20 weeks.

Results: Of six patients, five (83.3%; 95% CI 35.9% to 99.6%) achieved the clinical endpoint of an SRI of 4. Adverse events were minimal. Mechanistic studies revealed significant reductions in CD27IgD double negative B cells, switched memory B cells and activated naïve B cells, with increased transitional B cells in the five patients who met the endpoint. There was a trend towards decreased autoantibody levels in specific patients. Two patients had increases in their Helios+Treg cells, but no other significant T cell changes were noted. GARP-TGFβ complexes were significantly increased following the MSC infusions. The B cell changes and the GARP-TGFβ increases significantly correlated with changes in SLEDAI scores.

Conclusion: This phase 1 trial suggests that umbilical cord (UC) MSC infusions are very safe and may have efficacy in lupus. The B cell and GARP-TGFβ changes provide novel insight into mechanisms by which MSCs may impact disease.

Trial registration number: NCT03171194.

Keywords: Autoimmunity; B-Lymphocytes; Lupus Erythematosus, Systemic.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Clinical and patient-reported outcomes. (A) Line plot of the change in SELENA-SLEDAI scores over the course of the trial. This is the overall score combining lab and clinical criteria. The primary endpoint was the SLE Responder Index (SRI) 4 at 24 weeks). There was overall a significant decline in SELENA-SLEDAI scores of 5/6 patients meeting the SRI-4 endpoint (p

Figure 2

B cell subset changes over…

Figure 2

B cell subset changes over time by flow cytometry. (B) Cell subset changes…

Figure 2
B cell subset changes over time by flow cytometry. (B) Cell subset changes over time as determined by flow cytometry. Significant differences in B cell subsets occurred over the trial period as also noted in online supplemental table 2. The cell markers used are listed in the methods and are previously published. Most significant changes were in decreased double negative B cells including DN2, increased transitional B cells and decreased activated naive B cells. IgD−CD27+=SM+PB, IgD−CD27−=DN1+DN2+(DN3 +4), IgD+CD27=(T1 +T2)+(rN +T3)+aN. aN, activated naïve; DN, double negative; rN, resting naïve; SM, switched memory; T, transitional; PB, plasmablasts; USM, unswitched memory.

Figure 3

Cell changes over time. (A)…

Figure 3

Cell changes over time. (A) Treg changes over time are presented. Participant 1…

Figure 3
Cell changes over time. (A) Treg changes over time are presented. Participant 1 had a significant increase over time in Tregs both Helios+and Helios− looking at percentage change and fold change over baseline. Participant 2 had a significant increase with time compared with baseline in both Treg subsets, though the percentage change was small given the low percentage of Tregs at baseline (B). A representative flow block is shown in figure part C demonstrating the increase in Tregs over time from 0% to 8% in participant 1. (D–F) GARP serum measures. Figure part D shows measures of circulating serum GARP-LAP (latency-associated peptide) complexes as measured by a sandwich ELISA assay as previously described. Random SLE patients in the MUSC cohort (n=30) had significantly less (p=0.0226) circulating GARP-LAP complex than age/sex/race matched controls (n=16) y-axis is od 450. (E) Plots GARP–LAP complexes via ELISA versus patient SLEDAI score at the time of blood draw (n=21) for the MUSC lupus cohort. The y-axis is OD 450 reading via ELISA. There is a significant correlation between SLEDAI score and GARP–LAP complexes in patients with SLEDAI scores >10. Figure part F is a sandwich ELISA measure of soluble serum GARP expressed as ng/mL of serum in the MSC treated patients over time demonstrating near 0 levels of serum GARP at baseline with significant increases at week 4, decreasing at week 8 prior to rebounding at week 24 in 3/5 patients. Mann-Whitney U test was used to determine significance (week 0–week4- p=0.003). GARP, glycoprotein A repetition predominant; LAP, latency-associated peptide; MUSC, Medical University of South Carolina; MSC, mesenchymal stromal cell; OD, optical density.
Figure 2
Figure 2
B cell subset changes over time by flow cytometry. (B) Cell subset changes over time as determined by flow cytometry. Significant differences in B cell subsets occurred over the trial period as also noted in online supplemental table 2. The cell markers used are listed in the methods and are previously published. Most significant changes were in decreased double negative B cells including DN2, increased transitional B cells and decreased activated naive B cells. IgD−CD27+=SM+PB, IgD−CD27−=DN1+DN2+(DN3 +4), IgD+CD27=(T1 +T2)+(rN +T3)+aN. aN, activated naïve; DN, double negative; rN, resting naïve; SM, switched memory; T, transitional; PB, plasmablasts; USM, unswitched memory.
Figure 3
Figure 3
Cell changes over time. (A) Treg changes over time are presented. Participant 1 had a significant increase over time in Tregs both Helios+and Helios− looking at percentage change and fold change over baseline. Participant 2 had a significant increase with time compared with baseline in both Treg subsets, though the percentage change was small given the low percentage of Tregs at baseline (B). A representative flow block is shown in figure part C demonstrating the increase in Tregs over time from 0% to 8% in participant 1. (D–F) GARP serum measures. Figure part D shows measures of circulating serum GARP-LAP (latency-associated peptide) complexes as measured by a sandwich ELISA assay as previously described. Random SLE patients in the MUSC cohort (n=30) had significantly less (p=0.0226) circulating GARP-LAP complex than age/sex/race matched controls (n=16) y-axis is od 450. (E) Plots GARP–LAP complexes via ELISA versus patient SLEDAI score at the time of blood draw (n=21) for the MUSC lupus cohort. The y-axis is OD 450 reading via ELISA. There is a significant correlation between SLEDAI score and GARP–LAP complexes in patients with SLEDAI scores >10. Figure part F is a sandwich ELISA measure of soluble serum GARP expressed as ng/mL of serum in the MSC treated patients over time demonstrating near 0 levels of serum GARP at baseline with significant increases at week 4, decreasing at week 8 prior to rebounding at week 24 in 3/5 patients. Mann-Whitney U test was used to determine significance (week 0–week4- p=0.003). GARP, glycoprotein A repetition predominant; LAP, latency-associated peptide; MUSC, Medical University of South Carolina; MSC, mesenchymal stromal cell; OD, optical density.

References

    1. Kaul A, Gordon C, Crow MK, et al. . Systemic lupus erythematosus. Nat Rev Dis Primers 2016;2:16039. 10.1038/nrdp.2016.39
    1. Ginzler EM, Dooley MA, Aranow C, et al. . Mycophenolate mofetil or intravenous cyclophosphamide for lupus nephritis. N Engl J Med 2005;353:2219–28. 10.1056/NEJMoa043731
    1. Sedhain A, Hada R, Agrawal RK, et al. . Low dose mycophenolate mofetil versus cyclophosphamide in the induction therapy of lupus nephritis in nepalese population: a randomized control trial. BMC Nephrol 2018;19:175. 10.1186/s12882-018-0973-7
    1. Maidhof W, Hilas O. Lupus: an overview of the disease and management options. P T 2012;37:240–9.
    1. Miura M, Gronthos S, Zhao M, et al. . Shed: stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci U S A 2003;100:5807–12. 10.1073/pnas.0937635100
    1. Dominici M, Le Blanc K, Mueller I, et al. . Minimal criteria for defining multipotent mesenchymal stromal cells. the international society for cellular therapy position statement. Cytotherapy 2006;8:315–7. 10.1080/14653240600855905
    1. Gronthos S, Mankani M, Brahim J, et al. . Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci U S A 2000;97:13625–30. 10.1073/pnas.240309797
    1. Lee DE, Ayoub N, Agrawal DK. Mesenchymal stem cells and cutaneous wound healing: novel methods to increase cell delivery and therapeutic efficacy. Stem Cell Res Ther 2016;7:37. 10.1186/s13287-016-0303-6
    1. Grada A, Falanga V. Novel stem cell therapies for applications to wound healing and tissue repair. Surg Technol Int 2016;29:29–37.
    1. Tse WT, Pendleton JD, Beyer WM, et al. . Suppression of allogeneic T-cell proliferation by human marrow stromal cells: implications in transplantation. Transplantation 2003;75:389–97. 10.1097/01.TP.0000045055.63901.A9
    1. Di Nicola M, Carlo-Stella C, Magni M, et al. . Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 2002;99:3838–43. 10.1182/blood.V99.10.3838
    1. Forbes GM, Sturm MJ, Leong RW, et al. . A Phase 2 study of allogeneic mesenchymal stromal cells for Luminal crohn’s disease refractory to biologic therapy. Clinical Gastroenterology and Hepatology 2014;12:64–71. 10.1016/j.cgh.2013.06.021
    1. Christopeit M, Schendel M, Föll J, et al. . Marked improvement of severe progressive systemic sclerosis after transplantation of mesenchymal stem cells from an allogeneic haploidentical-related donor mediated by ligation of CD137L. Leukemia 2008;22:1062–4. 10.1038/sj.leu.2404996
    1. Wang L, Wang L, Cong X, et al. . Human umbilical cord mesenchymal stem cell therapy for patients with active rheumatoid arthritis: safety and efficacy. Stem Cells Dev 2013;22:3192–202. 10.1089/scd.2013.0023
    1. Wang D, Zhang H, Liang J, et al. . Allogeneic mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus: 4 years of experience. Cell Transplant 2013;22:2267–77. 10.3727/096368911X582769c
    1. Liang J, Zhang H, Hua B, et al. . Allogenic mesenchymal stem cells transplantation in refractory systemic lupus erythematosus: a pilot clinical study. Ann Rheum Dis 2010;69:1423–9. 10.1136/ard.2009.123463
    1. Augello A, Tasso R, Negrini SM, et al. . Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis. Arthritis Rheum 2007;56:1175–86. 10.1002/art.22511
    1. Bonig H, Kuçi Z, Kuçi S, et al. . Children and adults with refractory acute graft-versus-host disease respond to treatment with the mesenchymal stromal cell preparation “MSC-FFM”—outcome report of 92 patients. Cells 2019;8:1577. 10.3390/cells8121577
    1. Molendijk I, Bonsing BA, Roelofs H, et al. . Allogeneic bone marrow–derived mesenchymal stromal cells promote healing of refractory perianal fistulas in patients with crohn’s disease. Gastroenterology 2015;149:918–27. 10.1053/j.gastro.2015.06.014
    1. Wang D, Zhang H, Liang J, et al. . A long-term follow-up study of allogeneic mesenchymal stem/stromal cell transplantation in patients with drug-resistant systemic lupus erythematosus. Stem Cell Reports 2018;10:933–41. 10.1016/j.stemcr.2018.01.029
    1. Sun L, Wang D, Liang J, et al. . Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus. Arthritis & Rheumatism 2010;62:2467–75. 10.1002/art.27548
    1. Sun L, Akiyama K, Zhang H, et al. . Mesenchymal stem cell transplantation reverses multiorgan dysfunction in systemic lupus erythematosus mice and humans. Stem Cells 2009;27:1421–32. 10.1002/stem.68
    1. Gu F, Wang D, Zhang H, et al. . Allogeneic mesenchymal stem cell transplantation for lupus nephritis patients refractory to conventional therapy. Clin Rheumatol 2014;33:1611–9. 10.1007/s10067-014-2754-4
    1. Deng D, Zhang P, Guo Y, et al. . A randomised double-blind, placebo-controlled trial of allogeneic umbilical cord-derived mesenchymal stem cell for lupus nephritis. Ann Rheum Dis 2017;76:1436–9. 10.1136/annrheumdis-2017-211073
    1. Musiał-Wysocka A, Kot M, Majka M. The pros and cons of mesenchymal stem cell-based therapies. Cell Transplant 2019;28:801–12. 10.1177/0963689719837897
    1. Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol 2014;32:252–60. 10.1038/nbt.2816
    1. Abdi R, Fiorina P, Adra CN, et al. . Immunomodulation by mesenchymal stem cells: a potential therapeutic strategy for type 1 diabetes. Diabetes 2008;57:1759–67. 10.2337/db08-0180
    1. Wang M, Yuan Q, Xie L. Mesenchymal stem cell-based immunomodulation: properties and clinical application. Stem Cells Int 2018;2018:1–12. 10.1155/2018/3057624
    1. Collins E, Gu F, Qi M, et al. . Differential efficacy of human mesenchymal stem cells based on source of origin. J Immunol 2014;193:4381–90. 10.4049/jimmunol.1401636
    1. Gao L, Bird AK, Meednu N, et al. . Bone marrow-derived mesenchymal stem cells from patients with systemic lupus erythematosus have a senescence-associated secretory phenotype mediated by a mitochondrial antiviral signaling protein-Interferon-β feedback loop. Arthritis & Rheumatology 2017;69:1623–35. 10.1002/art.40142
    1. Carrion F, Nova E, Ruiz C, et al. . Autologous mesenchymal stem cell treatment increased T regulatory cells with no effect on disease activity in two systemic lupus erythematosus patients. Lupus 2010;19:317–22. 10.1177/0961203309348983
    1. Hochberg MC. Updating the American college of rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis & Rheumatism 1997;40:1725. 10.1002/art.1780400928
    1. Jolly M, Kosinski M, Garris CP, et al. . Prospective validation of the lupus impact Tracker: a patient-completed tool for clinical practice to evaluate the impact of systemic lupus erythematosus. Arthritis & Rheumatology 2016;68:1422–31. 10.1002/art.39601
    1. Fitzmaurice GM, Laird NM, Ware JH. Applied longitudinal analysis. New York: John Wiley & Sons, Inc, 2004.
    1. Jenks SA, Cashman KS, Zumaquero E, et al. . Distinct effector B cells induced by unregulated Toll-like receptor 7 contribute to pathogenic responses in systemic lupus erythematosus. Immunity 2020;52:203. 10.1016/j.immuni.2019.12.005
    1. Wang D, Huang S, Yuan X, 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. 10.1038/cmi.2015.89
    1. Tran DQ, Andersson J, Wang R, et al. . GARP (LRRC32) is essential for the surface expression of latent TGF-β on platelets and activated FOXP3 + regulatory T cells. Proc Natl Acad Sci U S A 2009;106:13445–50. 10.1073/pnas.0901944106
    1. Carrillo-Galvez AB, Cobo M, Cuevas-Ocaña S, et al. . Mesenchymal stromal cells express GARP/LRRC32 on their surface: effects on their biology and immunomodulatory capacity. Stem Cells 2015;33:183–95. 10.1002/stem.1821
    1. Wallace CH, Wu BX, Salem M, et al. . B lymphocytes confer immune tolerance via cell surface GARP-TGF-β complex. JCI Insight 2018;3. 10.1172/jci.insight.99863
    1. Barbado J, Tabera S, Sánchez A, et al. . Therapeutic potential of allogeneic mesenchymal stromal cells transplantation for lupus nephritis. Lupus 2018;27:2161–5. 10.1177/0961203318804922
    1. Woodworth TG, Furst DE. Safety and feasibility of umbilical cord mesenchymal stem cells in treatment-refractory systemic lupus erythematosus nephritis: time for a double-blind placebo-controlled trial to determine efficacy. Arthritis Res Ther 2014;16:113. 10.1186/ar4677
    1. Zhou T, Li H-Y, Liao C, et al. . Clinical efficacy and safety of mesenchymal stem cells for systemic lupus erythematosus. Stem Cells Int 2020;2020:1–11. 10.1155/2020/6518508
    1. Wen L, Labopin M, Badoglio M, et al. . Prognostic factors for clinical response in systemic lupus erythematosus patients treated by allogeneic mesenchymal stem cells. Stem Cells Int 2019;2019:1–7. 10.1155/2019/7061408
    1. Galipeau J. The mesenchymal stromal cells dilemma—does a negative phase III trial of random donor mesenchymal stromal cells in steroid-resistant graft-versus-host disease represent a death knell or a bump in the road? Cytotherapy 2013;15:2–8. 10.1016/j.jcyt.2012.10.002
    1. Nie Y, Lau C, Lie A, et al. . Defective phenotype of mesenchymal stem cells in patients with systemic lupus erythematosus. Lupus 2010;19:850–9. 10.1177/0961203310361482
    1. Geng L, Li X, Feng X, et al. . Association of TNF- α with impaired migration capacity of mesenchymal stem cells in patients with systemic lupus erythematosus. J Immunol Res 2014;2014:1–14. 10.1155/2014/169082
    1. Zhu Y, Feng X. Genetic contribution to mesenchymal stem cell dysfunction in systemic lupus erythematosus. Stem Cell Res Ther 2018;9:149. 10.1186/s13287-018-0898-x
    1. Gao J, Dennis JE, Muzic RF, et al. . The dynamic in vivo distribution of bone marrow-derived mesenchymal stem cells after infusion. Cells Tissues Organs 2001;169:12–20. 10.1159/000047856
    1. Karp JM, Leng Teo GS. Mesenchymal stem cell homing: the devil is in the details. Cell Stem Cell 2009;4:206–16. 10.1016/j.stem.2009.02.001
    1. Devine SM, Cobbs C, Jennings M, et al. . Mesenchymal stem cells distribute to a wide range of tissues following systemic infusion into nonhuman primates. Blood 2003;101:2999–3001. 10.1182/blood-2002-06-1830
    1. Le Blanc K, Frassoni F, Ball L, et al. . Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. The Lancet 2008;371:1579–86. 10.1016/S0140-6736(08)60690-X
    1. Kean TJ, Lin P, Caplan AI, et al. . Mscs: delivery routes and engraftment, cell-targeting strategies, and immune modulation. Stem Cells Int 2013;2013:1–13. 10.1155/2013/732742
    1. Wang D, Feng X, Lu L, et al. . A CD8 T Cell/Indoleamine 2,3‐dioxygenase axis is required for mesenchymal stem cell suppression of human systemic lupus erythematosus. Arthritis Rheumatol 2014;66:2234–45. 10.1002/art.38674
    1. Meisel R, Zibert A, Laryea M, et al. . Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase–mediated tryptophan degradation. Blood 2004;103:4619–21. 10.1182/blood-2003-11-3909
    1. Singer NG, Caplan AI. Mesenchymal stem cells: mechanisms of inflammation. Annu Rev Pathol 2011;6:457–78. 10.1146/annurev-pathol-011110-130230
    1. Yuan X, Qin X, Wang D, et al. . Mesenchymal stem cell therapy induces FLT3L and CD1c+ dendritic cells in systemic lupus erythematosus patients. Nat Commun 2019;10:2498. 10.1038/s41467-019-10491-8
    1. de Witte SFH, Luk F, Sierra Parraga JM, et al. . Immunomodulation by therapeutic mesenchymal stromal cells (MSC) is triggered through phagocytosis of MSC by monocytic cells. Stem Cells 2018;36:602–15. 10.1002/stem.2779
    1. Carrión F, Nova E, Luz P, et al. . Opposing effect of mesenchymal stem cells on Th1 and Th17 cell polarization according to the state of CD4+ T cell activation. Immunol Lett 2011;135:10–16. 10.1016/j.imlet.2010.09.006
    1. Jenks PJ, Chevalier C, Ecobichon C, et al. . Identification of nonessential Helicobacter pylori genes using random mutagenesis and loop amplification. Res Microbiol 2001;152:725–34. 10.1016/S0923-2508(01)01253-0

Source: PubMed

3
Iratkozz fel