Advertisement
FULL-LENGTH ARTICLE | Clinical Research| Volume 24, ISSUE 3, P311-319, March 2022

Download started.

Ok

Response to extracorporeal photopheresis therapy of patients with steroid-refractory/-resistant GvHD is associated with up-regulation of Th22 cells and Tfh cells

Published:October 25, 2021DOI:https://doi.org/10.1016/j.jcyt.2021.09.008

Highlights

  • ECP was able to increase Tfh cells to ameliorate GvHD.
  • Upregulation of Th22 cells was observed in aGvHD patients with CR.
  • Tim-3 expression was downregulated on effector T cells by ECP.
  • ECP shows immunomodulatory effects in GvHD setting.

Abstract

Extracorporeal photopheresis (ECP), a personalized cellular immunotherapy, constitutes a promising treatment for steroid-refractory/-resistant graft-versus-host disease (SR-GvHD), with encouraging clinical response rates. To further investigate its mechanism of action, ECP's effects on T helper (Th) cells as well as on expression of immune checkpoint (PD-1 and Tim-3) and apoptotic (Fas receptor [FasR]) molecules were investigated in 27 patients with SR-GvHD. Our data show that GvHD patients had significantly higher levels of Th2, Th17, Th22 and granulocyte-macrophage colony-stimulating factor (GM-CSF)-positive Th (ThG) cells and clearly lower levels of T follicular helper (Tfh) cells, including Th1- and Th2-like cells, compared with healthy donors. ECP therapy for GvHD was effective through the modulation of different Th subsets: increases of Th22 (1.52-fold) and Tfh cells (1.48-fold) in acute GvHD (aGvHD) and increases of Th2-like Tfh cells (1.74-fold) in chronic GvHD (cGvHD) patients were associated with clinical response. Expression of FasR was further upregulated in CD4+CD8+ T cells. Additionally, Tim-3–expressing effector T cells associated with the severity of GvHD were reduced. Taken together, these data show that ECP therapy exerts immunomodulatory effects by promoting a balanced immune reconstitution and inducing immune tolerance. Therefore it represents an attractive option for the treatment of GvHD.

Keywords

To read this article in full you will need to make a payment

Purchase one-time access:

Academic & Personal: 24 hour online accessCorporate R&D Professionals: 24 hour online access
One-time access price info
  • For academic or personal research use, select 'Academic and Personal'
  • For corporate R&D use, select 'Corporate R&D Professionals'

Subscribe:

Subscribe to Cytotherapy
Already a print subscriber? Claim online access
Already an online subscriber? Sign in
Institutional Access: Sign in to ScienceDirect

References

    • Martin P.J.
    • et al.
    An endpoint associated with clinical benefit after initial treatment of chronic graft-versus-host disease.
    Blood. 2017; 130: 360-367
    • Wang L.
    • et al.
    Modulation of B Cells and Homing Marker on NK Cells Through Extracorporeal Photopheresis in Patients With Steroid-Refractory/Resistant Graft-Vs.-Host Disease Without Hampering Anti-viral/Anti-leukemic Effects.
    Front Immunol. 2018; 9
    • Knobler R.
    • et al.
    Guidelines on the use of extracorporeal photopheresis.
    Journal of the European Academy of Dermatology and Venereology: JEADV. 2014; 28: 1-37
    • Jagasia M.
    • et al.
    Randomized controlled study of ECP with methoxsalen as first-line treatment of patients with moderate to severe cGVHD.
    Blood Adv. 2019; 3: 2218-2229
    • Mehta R.S.
    • et al.
    Randomized phase II trial of extracorporeal phototherapy and steroids vs. steroids alone for newly diagnosed acute GVHD.
    Bone Marrow Transplant. 2021;
    • Sestili S.
    • et al.
    Extracorporeal photopheresis as first-line strategy in the treatment of acute graft-versus-host disease after hematopoietic stem cell transplantation: A single-center experience.
    Cytotherapy. 2020; 22: 445-449
    • Berger M.
    • Albiani R.
    • Sini B.
    • Fagioli F.
    Extracorporeal photopheresis for graft-versus-host disease: the role of patient, transplant, and classification criteria and hematologic values on outcome-results from a large single-center study.
    Transfusion. 2015; 55: 736-747
    • Flowers M.E.
    • et al.
    A multicenter prospective phase 2 randomized study of extracorporeal photopheresis for treatment of chronic graft-versus-host disease.
    Blood. 2008; 112: 2667-2674
    • Greinix H.T.
    • et al.
    Extracorporeal photochemotherapy in the treatment of severe steroid-refractory acute graft-versus-host disease: a pilot study.
    Blood. 2000; 96: 2426-2431
    • Martin P.J.
    • et al.
    National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: VI. Design of Clinical Trials Working Group report.
    Biol Blood Marrow Transplant. 2006; 12: 491-505
  1. Robert Zeiser, P. D., Jürgen Finke, Hildegard Greinix, Ernst Holler, Thomas Luft, Stephan Mielke, Christof Scheid, Daniel Wolff. GVHD-PROPHYLAXE UND THERAPIE. zur allogenen Stammzelltransplantation von der Deutschen Arbeitsgemeinschaft für Knochenmark- und Blutstammzelltransplantation (DAG-KBT) (2016).

    • Ni M.
    • et al.
    Shaping of CD56(bri) Natural Killer Cells in Patients With Steroid-Refractory/Resistant Acute Graft-vs.-Host Disease via Extracorporeal Photopheresis.
    Front Immunol. 2019; 10: 547
    • Glucksberg H.
    • et al.
    Clinical manifestations of graft-versus-host disease in human recipients of marrow from HL-A-matched sibling donors.
    Transplantation. 1974; 18: 295-304
    • Harris A.C.
    • et al.
    International, Multicenter Standardization of Acute Graft-versus-Host Disease Clinical Data Collection: A Report from the Mount Sinai Acute GVHD International Consortium.
    Biol Blood Marrow Transplant. 2016; 22: 4-10
    • Jagasia M.H.
    • et al.
    National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group report.
    Biol Blood Marrow Transplant. 2015; 21: e381
    • Wang L.
    • et al.
    Standardization of cryopreserved peripheral blood mononuclear cells through a resting process for clinical immunomonitoring–Development of an algorithm.
    Cytometry A. 2016; 89: 246-258
    • Nygaard M.
    • et al.
    Longitudinal follow-up of response status and concomitant immunosuppression in patients treated with extracorporeal photopheresis for chronic graft versus host disease.
    Bone Marrow Transplant. 2019; 54: 35-43
    • Oarbeascoa G.
    • et al.
    Retrospective Multicenter Study of Extracorporeal Photopheresis in Steroid-Refractory Acute and Chronic Graft-versus-Host Disease.
    Biol Blood Marrow Transplant. 2020; 26: 651-658
    • Florek M.
    • et al.
    Autologous apoptotic cells preceding transplantation enhance survival in lethal murine graft-versus-host models.
    Blood. 2014; 124: 1832-1842
    • Pilon C.
    • et al.
    Human Apoptotic Cells, Generated by Extracorporeal Photopheresis, Modulate Allogeneic Immune Response.
    Front Immunol. 2019; 10: 2908
    • Lorenz K.
    • et al.
    Modulation of lymphocyte subpopulations by extracorporeal photopheresis in patients with acute graft-versus-host disease or graft rejection.
    Leuk Lymphoma. 2015; 56: 671-675
    • Hill G.R.
    • Betts B.C.
    • Tkachev V.
    • Kean L.S.
    • Blazar B.R.
    Current Concepts and Advances in Graft-Versus-Host Disease Immunology.
    Annu Rev Immunol. 2021;
    • Zeiser R.
    • Blazar B.R.
    Pathophysiology of Chronic Graft-versus-Host Disease and Therapeutic Targets.
    The New England journal of medicine. 2017; 377: 2565-2579
    • Zeiser R.
    • Blazar B.R.
    Acute Graft-versus-Host Disease.
    The New England journal of medicine. 2018; 378: 586
    • Fu J.
    • Heinrichs J.
    • Yu X.Z.
    Helper T-cell differentiation in graft-versus-host disease after allogeneic hematopoietic stem cell transplantation.
    Arch Immunol Ther Exp (Warsz). 2014; 62: 277-301
    • Schewitz-Bowers L.P.
    • et al.
    Glucocorticoid-resistant Th17 cells are selectively attenuated by cyclosporine A.
    Proc Natl Acad Sci U S A. 2015; 112: 4080-4085
    • Gartlan K.H.
    • et al.
    Th17 plasticity and transition toward a pathogenic cytokine signature are regulated by cyclosporine after allogeneic SCT.
    Blood Adv. 2017; 1: 341-351
    • Ullrich E.
    • et al.
    BATF-dependent IL-7RhiGM-CSF+ T cells control intestinal graft-versus-host disease.
    J Clin Invest. 2018; 128: 916-930
    • Gartlan K.H.
    • et al.
    Donor T-cell-derived GM-CSF drives alloantigen presentation by dendritic cells in the gastrointestinal tract.
    Blood Adv. 2019; 3: 2859-2865
    • Piper C.
    • et al.
    Pathogenic Bhlhe40+ GM-CSF+ CD4+ T cells promote indirect alloantigen presentation in the GI tract during GVHD.
    Blood. 2020; 135: 568-581
    • Elenkov I.J.
    Glucocorticoids and the Th1/Th2 balance.
    Ann N Y Acad Sci. 2004; 1024: 138-146
    • O'Garra A.
    Cytokines induce the development of functionally heterogeneous T helper cell subsets.
    Immunity. 1998; 8: 275-283
    • Holtan S.G.
    • Pasquini M.
    • Weisdorf D.J.
    Acute graft-versus-host disease: a bench-to-bedside update.
    Blood. 2014; 124: 363-373
    • Gorgun G.
    • Miller K.B.
    • Foss F.M.
    Immunologic mechanisms of extracorporeal photochemotherapy in chronic graft-versus-host disease.
    Blood. 2002; 100: 941-947
    • Molldrem J.J.
    • Lu S.
    A GVHD kill switch helps immune reconstitution.
    Blood. 2014; 123: 3849-3850
    • Forcade E.
    • et al.
    Circulating T follicular helper cells with increased function during chronic graft-versus-host disease.
    Blood. 2016; 127: 2489-2497
    • Yang X.
    • et al.
    T follicular helper cells and regulatory B cells dynamics in systemic lupus erythematosus.
    PLoS One. 2014; 9: e88441
    • Duhen T.
    • Geiger R.
    • Jarrossay D.
    • Lanzavecchia A.
    • Sallusto F.
    Production of interleukin 22 but not interleukin 17 by a subset of human skin-homing memory T cells.
    Nat Immunol. 2009; 10: 857-863
    • Gartlan K.H.
    • et al.
    A critical role for donor-derived IL-22 in cutaneous chronic GVHD.
    Am J Transplant. 2018; 18: 810-820
    • Hanash A.M.
    • et al.
    Interleukin-22 protects intestinal stem cells from immune-mediated tissue damage and regulates sensitivity to graft versus host disease.
    Immunity. 2012; 37: 339-350
    • Zhao D.
    • et al.
    Survival signal REG3alpha prevents crypt apoptosis to control acute gastrointestinal graft-versus-host disease.
    J Clin Invest. 2018; 128: 4970-4979
    • Lamarthee B.
    • et al.
    Donor interleukin-22 and host type I interferon signaling pathway participate in intestinal graft-versus-host disease via STAT1 activation and CXCL10.
    Mucosal Immunol. 2016; 9: 309-321
    • Zhao K.
    • et al.
    The identification and characteristics of IL-22-producing T cells in acute graft-versus-host disease following allogeneic bone marrow transplantation.
    Immunobiology. 2013; 218: 1505-1513
    • Hansen J.A.
    • et al.
    A novel soluble form of Tim-3 associated with severe graft-versus-host disease.
    Biol Blood Marrow Transplant. 2013; 19: 1323-1330
    • Ni X.
    • et al.
    PD-L1 interacts with CD80 to regulate graft-versus-leukemia activity of donor CD8+ T cells.
    J Clin Invest. 2017; 127: 1960-1977
    • Shustov A.
    • Nguyen P.
    • Finkelman F.
    • Elkon K.B.
    • Via C.S.
    Differential expression of Fas and Fas ligand in acute and chronic graft-versus-host disease: up-regulation of Fas and Fas ligand requires CD8+ T cell activation and IFN-gamma production.
    J Immunol. 1998; 161: 2848-2855
    • Gallez-Hawkins G.M.
    • et al.
    Increased programmed death-1 molecule expression in cytomegalovirus disease and acute graft-versus-host disease after allogeneic hematopoietic cell transplantation.
    Biol Blood Marrow Transplant. 2009; 15: 872-880
    • Parel Y.
    • et al.
    Presence of CD4+CD8+ double-positive T cells with very high interleukin-4 production potential in lesional skin of patients with systemic sclerosis.
    Arthritis Rheum. 2007; 56: 3459-3467
    • Strasser A.
    • Jost P.J.
    • Nagata S.
    The many roles of FAS receptor signaling in the immune system.
    Immunity. 2009; 30: 180-192
    • Yamada A.
    • Arakaki R.
    • Saito M.
    • Kudo Y.
    • Ishimaru N.
    Dual Role of Fas/FasL-Mediated Signal in Peripheral Immune Tolerance.
    Front Immunol. 2017; 8: 403
View full text