Treatment of chronic GvHD with mesenchymal stromal cells induces durable responses: A phase II study

Erik Boberg, Lena von Bahr, Gabriel Afram, Carina Lindström, Per Ljungman, Nina Heldring, Peter Petzelbauer, Karin Garming Legert, Nadir Kadri, Katarina Le Blanc, Erik Boberg, Lena von Bahr, Gabriel Afram, Carina Lindström, Per Ljungman, Nina Heldring, Peter Petzelbauer, Karin Garming Legert, Nadir Kadri, Katarina Le Blanc

Abstract

Steroid-refractory chronic graft-vs-host disease (cGvHD) contributes to morbidity after allogeneic hematopoietic stem cell transplantation. Here, we report on 11 patients with severe, refractory cGvHD treated with repeated infusions of allogeneic bone marrow-derived mesenchymal stromal cells (MSC) over a 6- to 12-month period. Six patients responded to MSC treatment following National Institutes of Health response criteria, accompanied by improvement in GvHD-related symptoms and quality of life. This response was durable, with systemic immunosuppressive therapy withdrawn from two responders, and a further two free from steroids and tapering calcineurin inhibitors. All responders displayed a distinct immune phenotype characterized by higher levels of naïve T cells and B cells before treatment compared with the nonresponders, and a significantly higher fraction of CD31+ naïve CD4+ T cells. MSC treatment was associated with significant increases in naïve T cells, B cells, and Tregs 7 days after each infusion. Skin biopsies showed resolution of epidermal pathology. CXCL9 and CXCL10 showed differential responses in responder and nonresponder patients. Our data support the use of MSC infusions as treatment for steroid-refractory cGvHD with durable responses. We propose CXCL9 and CXCL10 as early biomarkers for responsiveness to MSC treatment. Our results highlight the importance of the MSC recipient immune phenotype in promoting treatment response. This trial was registered at www.ClinicalTrials.gov as #NCT01522716.

Keywords: cellular therapy; clinical trials; hematopoietic stem cell transplantation; mesenchymal stem cells; thymus.

Conflict of interest statement

The authors declared no potential conflicts of interest.

© 2020 The Authors. STEM CELLS TRANSLATIONAL MEDICINE published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.

Figures

FIGURE 1
FIGURE 1
Heatmap of individual responses: NIH organ score, National Institutes of Health (NIH) global score, range of motion (ROM), and body surface area (BSA) percentage involved with sclerosis. Time points are at study enrolment and at the end of MSC treatment. Black boxes denote organs with response and red boxes denote organs with progression. CNI, calcineurin inhibitors; ECP, extracorporeal photopheresis; F, fascia; J, joints; MMF, mycophenolate mofetil; M, muscles; P, prednisolone; Tac, tacrolimus
FIGURE 2
FIGURE 2
Responders have higher absolute and relative levels of naïve CD4+ T cells and naïve B cells compared with nonresponders. A and B, Representative plots of the flow cytometry analysis. CD4+ T cells were divided into naïve, CM, EM, and T‐EMRA based on CD27 and CD45RA. 32 Naïve B cells were defined as CD19+CD27‐IgD+CD21hi. CD21 was used to exclude CD27‐CD21low B cells, a population previously suggested to contain aberrant B cells in cGvHD. 33 C and D, Relative and absolute numbers of naïve CD4+ T cells were higher in R compared with NR throughout the study. There were no differences in CM CD4+ T‐cell numbers. E and F, Memory B cells were not different between R and NR. Absolute numbers of naïve B cells were higher in R compared with NR throughout the study. P‐values in the absolute number graphs represent the P‐values for the Responder factor in the linear mixed effects analysis. P‐values for relative numbers with Wilcoxon rank‐sum test. *P < .05. Error bars show mean ± SEM. CM, central memory T‐cell; EM, effector memory T‐cell; T‐EMRA, terminally differentiated effector memory T cells
FIGURE 3
FIGURE 3
Mesenchymal stromal cell (MSC) treatment induces improvements in inflammatory cytokine profile in responders and in epidermal skin pathology in both responders and nonresponders. A, Multiplex analysis of plasma samples taken before each MSC infusion is presented. Levels of several pro‐inflammatory chemokines, as well as TNFα and IL6 increased in NR and decreased in R after two and six MSC infusions (1 and 5 months after treatment start, respectively), compared to before the first infusion. P‐values for within group changes using repeated measures ANOVA. P‐values for comparing NR and R at different time points with t test. *P < .05, **P < .005, ***P < .0005. B, Blinded pathological review of skin biopsies taken before and after treatment revealed improvements with mainly epidermal changes in both R and NR after treatment. For details, see Table S5. Representative images of one R (patient 5) and one NR (patient 10) showing mononuclear infiltration (asterisks), extensive vacuolization of KC (black arrows) as well as apoptotic and Civatte bodies in the epithelium (white arrows) before treatment. C, The epidermal pathology regressed in both R and NR after treatment with little or no remaining vacuolization and few apoptotic cells, but in the NR dermal sclerosis progressed (X). CXCL, chemokine (C‐X‐C motif) ligand; IL, interleukin; KC, keratinocytes; LP, lamina propria; MCP, monocyte chemotactic protein; NR, nonresponder; R, responder; TNF, tumor necrosis factor
FIGURE 4
FIGURE 4
Short‐term increases in naïve miRNA, T cells, and Tregs after infusion. A, Four circulating miRNAs were significantly up regulated in R and NR plasma at 1 hour after mesenchymal stromal cell (MSC) infusion 1 and returned to baseline levels 24 hours after infusion. t Test with Benjamini‐Hochberg correction. B, Representative plots of the flow cytometry analysis. Tregs are defined as CD4+ FoxP3+ T cells. Naïve CD4+ T cells are defined as in Figure 2 (CD4+ CD45RA+ CD27+). C and D, Absolute number of naïve CD4+ T cells and Tregs increase in R 1 and 7 days after each MSC infusion compared with before infusion. These short‐term increases are not seen in NR. X‐axes display infusion number. The P‐values are derived from the mixed effects model and represent the significance of the factor Days since last infusion. *P < .05, **P < .005. Error bars show mean ± SEM. R, responder; NR, nonresponder; Treg, regulatory T cell
FIGURE 5
FIGURE 5
Thymic function and proliferation of naïve CD4+ T cells. A, Thymic function was assessed by measuring the proportion of CD31+ cells among naïve CD4+ T cells. We found a higher percentage of CD31+ naïve CD4+ T cells in R compared to NR before infusion, indicating better thymic function. B, Proliferation of naïve CD4+ T cells was assessed using levels of Ki67. The levels of Ki67 were not different between the responders and the nonresponders. Data from infusion 3. P‐values calculated using the Wilcoxon rank‐sum test. *P < .05. Error bars show mean ± SEM

References

    1. Gooley TA, Chien JW, Pergam SA, et al. Reduced mortality after allogeneic hematopoietic‐cell transplantation. N Engl J Med. 2010;363(22):2091‐2101.
    1. Duell T, van Lint MT, Ljungman P, et al. Health and functional status of long‐term survivors of bone marrow transplantation. EBMT working party on late effects and EULEP study group on late effects. European Group for Blood and Marrow Transplantation. Ann Intern Med. 1997;126(3):184‐192.
    1. Ruutu T, Gratwohl A, de Witte T, et al. Prophylaxis and treatment of GVHD: EBMT‐ELN working group recommendations for a standardized practice. Bone Marrow Transplant. 2014;49(2):168‐173.
    1. Koc S, Leisenring W, Flowers ME, et al. Therapy for chronic graft‐versus‐host disease: a randomized trial comparing cyclosporine plus prednisone versus prednisone alone. Blood. 2002;100(1):48‐51.
    1. Wolff D, Schleuning M, von Harsdorf S, et al. Consensus conference on clinical practice in chronic GVHD: second‐line treatment of chronic graft‐versus‐host disease. Biol Blood Marrow Transplant. 2011;17(1):1‐17.
    1. Arai S, Pidala J, Pusic I, et al. A randomized phase II crossover study of imatinib or rituximab for cutaneous sclerosis after hematopoietic cell transplantation. Clin Cancer Res. 2016;22(2):319‐327.
    1. Malik MI, Litzow M, Hogan W, et al. Extracorporeal photopheresis for chronic graft‐versus‐host disease: a systematic review and meta‐analysis. Blood Res. 2014;49(2):100‐106.
    1. Flowers ME, Apperley JF, van Besien K, et al. A multicenter prospective phase 2 randomized study of extracorporeal photopheresis for treatment of chronic graft‐versus‐host disease. Blood. 2008;112(7):2667‐2674.
    1. Miklos D, Cutler CS, Arora M, et al. Ibrutinib for chronic graft‐versus‐host disease after failure of prior therapy. Blood. 2017;130(21):2243‐2250.
    1. A Study of Ruxolitinib vs Best Available Therapy (BAT) in Patients With Steroid‐refractory Chronic Graft vs. Host Disease (GvHD) After Bone Marrow Transplantation (REACH3). Vol 2020 Wilmington: Incyte Corporation; 2017.
    1. Bernardo ME, Fibbe WE. Mesenchymal stromal cells: sensors and switchers of inflammation. Cell Stem Cell. 2013;13(4):392‐402.
    1. Le Blanc K, Rasmusson I, Sundberg B, et al. Treatment of severe acute graft‐versus‐host disease with third party haploidentical mesenchymal stem cells. Lancet. 2004;363(9419):1439‐1441.
    1. Lalu MM, McIntyre L, Pugliese C, et al.; Canadian Critical Care Trials Group. Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta‐analysis of clinical trials. PLoS One. 2012;7(10):e47559.
    1. Bernardo ME, Fibbe WE. Mesenchymal stromal cells and hematopoietic stem cell transplantation. Immunol Lett. 2015;168(2):215‐221.
    1. Le Blanc K, Davies LC. Mesenchymal stromal cells and the innate immune response. Immunol Lett. 2015;168(2):140‐146.
    1. Peng Y, Chen X, Liu Q, et al. Mesenchymal stromal cells infusions improve refractory chronic graft versus host disease through an increase of CD5+ regulatory B cells producing interleukin 10. Leukemia. 2015;29(3):636‐646.
    1. Weng JY, Du X, Geng SX, et al. Mesenchymal stem cell as salvage treatment for refractory chronic GVHD. Bone Marrow Transplant. 2010;45(12):1732‐1740.
    1. Zhou H, Guo M, Bian C, et al. Efficacy of bone marrow‐derived mesenchymal stem cells in the treatment of sclerodermatous chronic graft‐versus‐host disease: clinical report. Biol Blood Marrow Transplant. 2010;16(3):403‐412.
    1. Perez‐Simon JA, Lopez‐Villar O, Andreu EJ, et al. Mesenchymal stem cells expanded in vitro with human serum for the treatment of acute and chronic graft‐versus‐host disease: results of a phase I/II clinical trial. Haematologica. 2011;96(7):1072‐1076.
    1. Flowers ME, Martin PJ. How we treat chronic graft‐versus‐host disease. Blood. 2015;125(4):606‐615.
    1. Martin PJ, Storer BE, Carpenter PA, et al. Comparison of short‐term response and long‐term outcomes after initial systemic treatment of chronic graft‐versus‐host disease. Biol Blood Marrow Transplant. 2011;17(1):124‐132.
    1. Schoemans HM, Lee SJ, Ferrara JL, et al. EBMT‐NIH‐CIBMTR task force position statement on standardized terminology & guidance for graft‐versus‐host disease assessment. Bone Marrow Transplant. 2018;53(11):1401‐1415.
    1. Lee SJ, Wolff D, Kitko C, et al. Measuring therapeutic response in chronic graft‐versus‐host disease. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft‐versus‐host disease: IV. The 2014 response criteria working group report. Biol Blood Marrow Transplant. 2015;21(6):984‐999.
    1. Greinix HT, van Besien K, Elmaagacli AH, et al.; UVADEX Chronic GVHD Study Group. Progressive improvement in cutaneous and extracutaneous chronic graft‐versus‐host disease after a 24‐week course of extracorporeal photopheresis–results of a crossover randomized study. Biol Blood Marrow Transplant. 2011;17(12):1775‐1782.
    1. Filipovich AH, Weisdorf D, Pavletic S, et al. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft‐versus‐host disease: I. diagnosis and staging working group report. Biol Blood Marrow Transplant. 2005;11(12):945‐956.
    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. Lancet. 2008;371(9624):1579‐1586.
    1. Ljungman P, Boeckh M, Hirsch HH, et al.; Disease Definitions Working Group of the Cytomegalovirus Drug Development Forum. Definitions of cytomegalovirus infection and disease in transplant patients for use in clinical trials. Clin Infect Dis. 2017;64(1):87‐91.
    1. Lee S, Cook EF, Soiffer R, Antin JH. Development and validation of a scale to measure symptoms of chronic graft‐versus‐host disease. Biol Blood Marrow Transplant. 2002;8(8):444‐452.
    1. McQuellon RP, Russell GB, Cella DF, et al. Quality of life measurement in bone marrow transplantation: development of the Functional Assessment of Cancer Therapy‐Bone Marrow Transplant (FACT‐BMT) scale. Bone Marrow Transplant. 1997;19(4):357‐368.
    1. FlowJo Software Team . FlowJo. Ashland, OR: Tree Star, Inc; 2015.
    1. Shulman HM, Cardona DM, Greenson JK, et al. NIH consensus development project on criteria for clinical trials in chronic graft‐versus‐host disease: II. The 2014 Pathology Working Group Report. Biol Blood Marrow Transplant. 2015;21(4):589‐603.
    1. Larbi A, Fulop T. From "truly naive" to "exhausted senescent" T cells: when markers predict functionality. Cytometry A. 2014;85(1):25‐35.
    1. Sarantopoulos S, Ritz J. Aberrant B‐cell homeostasis in chronic GVHD. Blood. 2015;125(11):1703‐1707.
    1. Di Mitri D, Azevedo RI, Henson SM, et al. Reversible senescence in human CD4+CD45RA+CD27‐ memory T cells. J Immunol. 2011;187(5):2093‐2100.
    1. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401(6754):708‐712.
    1. Peng Y, Chen X, Liu Q, et al. Alteration of naive and memory B‐cell subset in chronic graft‐versus‐host disease patients after treatment with mesenchymal stromal cells. Stem Cells Translational Med. 2014;3(9):1023‐1031.
    1. Maecker HT, McCoy JP, Nussenblatt R. Standardizing immunophenotyping for the human immunology project. Nat Rev Immunol. 2012;12(3):191‐200.
    1. Burr SP, Dazzi F, Garden OA. Mesenchymal stromal cells and regulatory T cells: the Yin and Yang of peripheral tolerance? Immunol Cell Biol. 2013;91(1):12‐18.
    1. Miyara M, Yoshioka Y, Kitoh A, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 2009;30(6):899‐911.
    1. Kohler S, Thiel A. Life after the thymus: CD31+ and CD31‐ human naive CD4+ T‐cell subsets. Blood. 2009;113(4):769‐774.
    1. Krenger W, Blazar BR, Hollander GA. Thymic T‐cell development in allogeneic stem cell transplantation. Blood. 2011;117(25):6768‐6776.
    1. Bohmann EM, Fehn U, Holler B, et al. Altered immune reconstitution of B and T cells precedes the onset of clinical symptoms of chronic graft‐versus‐host disease and is influenced by the type of onset. Ann Hematol. 2017;96(2):299‐310.
    1. Stenger EO, Krishnamurti L, Galipeau J. Mesenchymal stromal cells to modulate immune reconstitution early post‐hematopoietic cell transplantation. BMC Immunol. 2015;16:74.
    1. Batorov EV, Shevela EY, Tikhonova MA, et al. Mesenchymal stromal cells improve early lymphocyte recovery and T cell reconstitution after autologous hematopoietic stem cell transplantation in patients with malignant lymphomas. Cell Immunol. 2015;297(2):80‐86.
    1. Koç ON, Gerson SL, Cooper BW, et al. Rapid hematopoietic recovery after coinfusion of autologous‐blood stem cells and culture‐expanded marrow mesenchymal stem cells in advanced breast cancer patients receiving high‐dose chemotherapy. J Clin Oncol. 2000;18(2):307‐316.
    1. Liu G, Wang L, Pang T, et al. Umbilical cord‐derived mesenchymal stem cells regulate thymic epithelial cell development and function in Foxn1(−/−) mice. Cell Mol Immunol. 2014;11(3):275‐284.
    1. Hu KX, Wang MH, Fan C, Wang L, Guo M, Ai HS. CM‐DiI labeled mesenchymal stem cells homed to thymus inducing immune recovery of mice after haploidentical bone marrow transplantation. Int Immunopharmacol. 2011;11(9):1265‐1270.
    1. Wong N, Wang X. miRDB: an online resource for microRNA target prediction and functional annotations. Nucleic Acids Res. 2015;43(Database Issue):D146‐D152.
    1. Cooke KR, Luznik L, Sarantopoulos S, et al. The biology of chronic graft‐versus‐host disease: a task force report from the National Institutes of Health consensus development project on criteria for clinical trials in chronic graft‐versus‐host disease. Biol Blood Marrow Transplant. 2017;23(2):211‐234.
    1. Mougiakakos D, Jitschin R, Johansson CC, Okita R, Kiessling R, Le Blanc K. The impact of inflammatory licensing on heme oxygenase‐1‐mediated induction of regulatory T cells by human mesenchymal stem cells. Blood. 2011;117(18):4826‐4835.
    1. Smigiel KS, Srivastava S, Stolley JM, Campbell DJ. Regulatory T‐cell homeostasis: steady‐state maintenance and modulation during inflammation. Immunol Rev. 2014;259(1):40‐59.
    1. Burzyn D, Benoist C, Mathis D. Regulatory T cells in nonlymphoid tissues. Nat Immunol. 2013;14(10):1007‐1013.
    1. Kmieciak M, Gowda M, Graham L, et al. Human T cells express CD25 and Foxp3 upon activation and exhibit effector/memory phenotypes without any regulatory/suppressor function. J Transl Med. 2009;7:89.
    1. Ariel A, Serhan CN. New lives given by cell death: macrophage differentiation following their encounter with apoptotic leukocytes during the resolution of inflammation. Front Immunol. 2012;3:4.
    1. Galleu A, Riffo‐Vasquez Y, Trento C, et al. Apoptosis in mesenchymal stromal cells induces in vivo recipient‐mediated immunomodulation. Sci Transl Med. 2017;9(416).–.

Source: PubMed

3
Abonneren