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

Lei Wang, Ming Ni, Angela Hückelhoven-Krauss, Leopold Sellner, Jean-Marc Hoffmann, Brigitte Neuber, Thomas Luft, Ute Hegenbart, Stefan Schönland, Christian Kleist, Martin Sill, Bao-An Chen, Patrick Wuchter, Volker Eckstein, William Krüger, Inken Hilgendorf, Ronit Yerushalmi, Arnon Nagler, Carsten Müller-Tidow, Anthony D Ho, Peter Dreger, Michael Schmitt, Anita Schmitt, Lei Wang, Ming Ni, Angela Hückelhoven-Krauss, Leopold Sellner, Jean-Marc Hoffmann, Brigitte Neuber, Thomas Luft, Ute Hegenbart, Stefan Schönland, Christian Kleist, Martin Sill, Bao-An Chen, Patrick Wuchter, Volker Eckstein, William Krüger, Inken Hilgendorf, Ronit Yerushalmi, Arnon Nagler, Carsten Müller-Tidow, Anthony D Ho, Peter Dreger, Michael Schmitt, Anita Schmitt

Abstract

Graft-vs.-host disease (GvHD), a severe complication of allogeneic hematopoietic stem cell transplantation, significantly affects the post-transplant morbidity and mortality. Systemic steroids remain the gold standard for the initial management of GvHD. However, up to 60% of patients will not sufficiently respond to steroids. Extracorporeal photopheresis (ECP), a cell-based immunotherapy, has shown good clinical results in such steroid-refractory/resistant GvHD patients. Given its immunomodulatory, but not global immunosuppressive and steroid-sparing capacity, ECP constitutes an attractive option. In the case of GvHD, the balance of immune cells is destroyed: effector cells are not any longer efficiently controlled by regulatory cells. ECP therapy may restore this balance. However, the precise mechanism and the impact of ECP on anti-viral/anti-leukemic function remain unclear. In this study, 839 ECP treatments were performed on patients with acute GvHD (aGvHD) and chronic GvHD (cGvHD). A comprehensive analysis of effector and regulatory cells in patients under ECP therapy included multi-parametric flow cytometry and tetramer staining, LuminexTM-based cytokine, interferon-γ enzyme-linked immunospot, and chromium-51 release assays. Gene profiling of myeloid-derived suppressor cells (MDSCs) was performed by microarray analysis. Immunologically, modulations of effector and regulatory cells as well as proinflammatory cytokines were observed under ECP treatment: (1) GvHD-relevant cell subsets like CD62L+ NK cells and newly defined CD19hiCD20hi B cells were modulated, but (2) quantity and quality of anti-viral/anti-leukemic effector cells were preserved. (3) The development of MDSCs was promoted and switched from an inactivated subset (CD33-CD11b+) to an activated subset (CD33+CD11b+). (4) The frequency of Foxp3+CD4+ regulatory T cells (Tregs) and CD24+CD38hi regulatory B cells was considerably increased in aGvHD patients, and Foxp3+CD8+ Tregs in cGvHD patients. (5) Proinflammatory cytokines like IL-1β, IL-6, IL-8, and TNF-α were significantly reduced. In summary, ECP constitutes an effective immunomodulatory therapy for patients with steroid-refractory/resistant GvHD without impairment of anti-viral/leukemia effects.

Keywords: ECP; GvHD; anti-leukemic effect; anti-viral effect; effector cells; immunomodulation; proinflammatory cytokines; regulatory cells.

Figures

Figure 1
Figure 1
Differentiation and education of NK cell populations by ECP in aGvHD patients. The assessment of CD56briCD16− NK cells (A) and CD56dimCD16+ NK cells (C) before and after ECP therapy shows that ECP treatment can promote the development of NK cells from CD56briCD16− NK cells to CD56dimCD16+ NK cells as well as educate NK cells by decreasing expression of NKG2D and CD62L (B,D). *means p < 0.05.
Figure 2
Figure 2
The role of CD19hiCD20hi B cells in cGvHD. (A) shows representative dot plots of CD19hiCD20hi B cells among HD, aGvHD and cGvHD groups. (B) displays the frequency of CD19hiCD20hi B cells in both aGvHD and cGvHD groups prior to ECP treatment. (C) Characterization of CD19hiCD20hi B cells showed significantly lower expression of BAFF-R and CD38 but slightly increased CD24 expression. (D,E) When compared to CD19+CD20+ B cells, CD19hiCD20hi B cells showed a different component pattern, a significantly higher BAFF-R+CD38− proportion and memory B cells. Dashed lines represent the corresponding median value of frequencies observed in 25 HDs. Differences in cell frequency between different groups were assessed by Independent T test. In all tests, a p-value < 0.05 was considered to be statistically significant. *means p < 0.05.
Figure 3
Figure 3
CD14+HLA-DR−/low MDSC subpopulations in the peripheral blood of GvHD patients with ECP treatment. The immunophenotype of MDSCs was assessed by flow cytometry. (A) Different components of inactivated, transitional and activated subsets were observed within CD14+HLA-DR−/low MDSCs among aGvHD patients, cGvHD patients and healthy donors (HDs) suggesting a development from inactivated into activated MDSCs. (B) A reduction of inactivated MDSCs was observed after ECP therapy in aGvHD patients. (C) The volcano plot shows the gene expression between activated MDSCs and inactivated MDSCs. The horizontal axis represents the fold change in intensity and the vertical axis represents statistical significance (Log Odds). The bar chart indicates the differential gene expression between activated and inactivated MDSCs. Gene expression was assessed after adjustment by the Benjamini-Hochberg procedure. Differences in cell frequency between different groups were assessed by paired-sample T test. In all tests, a p-value < 0.05 was considered to be statistically significant. *means p < 0.05.
Figure 4
Figure 4
Immunomodulation of regulatory T and B cells through ECP. The percentages of CD8+ Tregs (A), FoxP3+CD25+CD4+ Tregs (B), and CD24+CD38hi Bregs (C) were monitored in patients with aGvHD and cGvHD before and after ECP therapy. Foxp3+CD8+ Tregs significantly increased under ECP therapy in both aGvHD and cGvHD patients, along with significant up-regulation of Foxp3+CD4+ Tregs and Bregs in aGvHD patients, as assessed by paired-sample T test. * means p < 0.05.
Figure 5
Figure 5
A fast reduction of proinflammatory cytokines IL-1β (A), IL-6 (B), IL-8 (C), and TNF-α (D) was observed in all patients. Patient #7 showed a rebound of IL-1β and TNF-α after rapid steroid reduction. Eventually the level of both cytokines decreased when ECP was continued. Dashed lines represent the corresponding median value of cytokine levels observed in healthy donors. The frequency of ECP cycles is indicated on the x-axis. The black bars below the x-axis indicate a high frequency of ECP treatment during the first 12 weeks (twice per week) followed by a gray bar representing a reduced frequency (twice every second week) in weeks 13–28.
Figure 6
Figure 6
Impact of ECP therapy on anti-viral and anti-leukemic immune responses. (A) The representative dot plots with the frequency of CMV-specific CD8+ T cells are shown at different time (T) points before (T1) and after (T2 and T3) ECP treatment in aGvHD patient #5 and cGvHD patient #13. (B) The distribution of TCM, TN, TEM, and TE within the CMV specific CD8+ T cells in patients #5 and #13 is indicated. (C) The secretion of IFN-γ by virus specific T cells was measured by IFN-γ ELISpot assay. The bar chart shows the overview of the INF-γ secretion by CD8+ T cells in seven patients under ECP treatment. There is no significant difference among T1, T2, and T3 (p ≥ 0.05, one-way ANOVA test). Under ECP treatment, the frequency of CMV-specific CD8+ T cells was maintained. Most cells were TE cells followed by TEM cells. The function of these cells in terms of IFN-γ release kept stable. (D) The dynamic changes of CD4+CD8+ T cells, γδ T cells and NKT cells in aGvHD (upper panel) and cGvHD patients (lower panel) under the ECP treatment. Cell frequencies were not significantly different between before and under ECP treatment groups, which assessed by Paired sample T test. (E) A 4-h 51Cr release assay was performed to test the NK activity, which was calculated by the following formula: % specific lysis = [c.p.m. (experimental release)–mean c.p.m. (spontaneous release)]/[mean c.p.m. (maximal release)–mean c.p.m. (spontaneous release)] × 100. The box chart shows the NK activity against K562 cells at two different time points in aGvHD group and cGvHD group. There was no significant difference, as assessed by Paired sample T test. Each box represents three independent patients. The NK cell function was stable over the time of ECP treatment. The dashed lines represent the corresponding median value of frequencies observed in 25 healthy donors. In all tests, a p < 0.05 was considered to be statistically significant.
Figure 7
Figure 7
Cell population dynamics are displayed with steroid dosing and clinical parameters in representative patients under ECP therapy.
Figure 8
Figure 8
Schematic overview of mechanisms of immunomodulation in aGvHD patients under ECP therapy. (A) ECP is a cell-based immunotherapy, involving (i) apheresis with ex vivo collection of peripheral mononuclear cells, (ii) photoactivation with exposure of leukocyte-enriched plasma to the photosensitizing agent 8-methoxypsoralen and ultraviolet A light which results in crosslinking of the pyrimidine bases in DNA leading to cell death through apoptosis, (iii) reinfusion of the ECP-treated cells to the patient. (B) Apoptosis of ECP-treated cells play a key role in vivo. Engulfing these apoptotic cells by immature dendritic cells results in a tolerogenic phenotype and promotes tolerance through the secretion of immunosuppressive cytokines such as IL-10 and TGF-β as well. Upregulation of activated MDSCs, Th2, vδ2+ T cells, FoxP3+ Tregs, double negative (DN) T cells and Bregs result in an overall increase in immune tolerance, accompanied by a decrease of immune effector cells like IL-17+ T cells and Th1 cells as well as education of TE/EM cells via decreasing CD62L expression. Besides these, ECP promotes the NK cell differentiation from CD56bri to CD56dim NK cells with loss of expression of NKG2D and CD62L.

References

    1. Harris AC, Young R, Devine S, Hogan WJ, Ayuk F, Bunworasate U, 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. 10.1016/j.bbmt.2015.09.001
    1. Socie G, Ritz J. Current issues in chronic graft-versus-host disease. Blood. (2014) 124:374–84. 10.1182/blood-2014-01-514752
    1. Shulman HM, Cardona DM, Greenson JK, Hingorani S, Horn T, Huber E, 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) 21:589–603. 10.1016/j.bbmt.2014.12.031
    1. MacMillan ML, DeFor TE, Weisdorf DJ. The best endpoint for acute GVHD treatment trials. Blood. (2010) 115:5412–7. 10.1182/blood-2009-12-258442
    1. Rubio MT, Labopin M, Blaise D, Socie G, Contreras RR, Chevallier P, et al. . The impact of graft-versus-host disease prophylaxis in reduced-intensity conditioning allogeneic stem cell transplant in acute myeloid leukemia: a study from the Acute Leukemia Working Party of the European Group for Blood and Marrow Transplantation. Haematologica (2015) 100:683–9. 10.3324/haematol.2014.119339
    1. Abu-Dalle I, Reljic T, Nishihori T, Antar A, Bazarbachi A, Djulbegovic B, et al. . Extracorporeal photopheresis in steroid-refractory acute or chronic graft-versus-host disease: results of a systematic review of prospective studies. Biol Blood Marrow Transplant (2014) 20:1677–86. 10.1016/j.bbmt.2014.05.017
    1. Alcindor T, Gorgun G, Miller KB, Roberts TF, Sprague K, Schenkein DP, et al. . Immunomodulatory effects of extracorporeal photochemotherapy in patients with extensive chronic graft-versus-host disease. Blood (2001) 98:1622–5. 10.1182/blood.V98.5.1622
    1. Gorgun G, Miller KB, Foss FM. Immunologic mechanisms of extracorporeal photochemotherapy in chronic graft-versus-host disease. Blood (2002) 100:941–7. 10.1182/blood-2002-01-0068
    1. Biagi E, Di Biaso I, Leoni V, Gaipa G, Rossi V, Bugarin C, et al. . Extracorporeal photochemotherapy is accompanied by increasing levels of circulating CD4+CD25+GITR+Foxp3+CD62L+ functional regulatory T-cells in patients with graft-versus-host disease. Transplantation (2007) 84:31–9. 10.1097/01.tp.0000267785.52567.9c
    1. Di Biaso I, Di Maio L, Bugarin C, Gaipa G, Dander E, Balduzzi A, et al. Regulatory T cells and extracorporeal photochemotherapy: correlation with clinical response and decreased frequency of proinflammatory T cells. Transplantation (2009) 87:1422–5. 10.1097/TP.0b013e3181a27a5d
    1. Lorenz K, Rommel K, Mani J, Jin N, Hilgendorf I, Ho AD, et al. Modulation of lymphocyte subpopulations by extracorporeal photopheresis in patients with acute graft-versus-host disease or graft rejection. Leuk Lymphoma (2015) 56:671–5. 10.3109/10428194.2014.931956
    1. Rieber N, Wecker I, Neri D, Fuchs K, Schafer I, Brand A, et al. . Extracorporeal photopheresis increases neutrophilic myeloid-derived suppressor cells in patients with GvHD. Bone Marrow Transplant (2014) 49:545–52. 10.1038/bmt.2013.236
    1. Schmitt S, Johnson TS, Karakhanova S, Naher H, Mahnke K, Enk AH. Extracorporeal photophoresis augments function of CD4+CD25+FoxP3+ regulatory T cells by triggering adenosine production. Transplantation (2009) 88:411–6. 10.1097/TP.0b013e3181aed927
    1. Jamil MO, Mineishi S. State-of-the-art acute and chronic GVHD treatment. Int J Hematol. (2015) 101:452–66. 10.1007/s12185-015-1785-1
    1. Ruutu T, Gratwohl A, de Witte T, Afanasyev B, Apperley J, Bacigalupo A, et al. . Prophylaxis and treatment of GVHD: EBMT-ELN working group recommendations for a standardized practice. Bone Marrow Transplant (2014) 49:168–73. 10.1038/bmt.2013.107
    1. Knobler R, Berlin G, Calzavara-Pinton P, Greinix H, Jaksch P, Laroche L, et al. . Guidelines on the use of extracorporeal photopheresis. J Eur Acad Dermatol Venereol. (2014) 28(Suppl. 1):1–37. 10.1111/jdv.12311
    1. Guidelines for Allogeneic Stem Cell Transplantation German Association of Bone Marrow and Blood Stem Cell Transplantation. (2016).
    1. Wang L, Huckelhoven A, Hong J, Jin N, Mani J, Chen BA, et al. . Standardization of cryopreserved peripheral blood mononuclear cells through a resting process for clinical immunomonitoring–Development of an algorithm. Cytometry A (2016) 89:246–58. 10.1002/cyto.a.22813
    1. McNeil LK, Price L, Britten CM, Jaimes M, Maecker H, Odunsi K, et al. . A harmonized approach to intracellular cytokine staining gating: results from an international multiconsortia proficiency panel conducted by the Cancer Immunotherapy Consortium (CIC/CRI). Cytometry A (2013) 83:728–38. 10.1002/cyto.a.22319
    1. Welters MJ, Gouttefangeas C, Ramwadhdoebe TH, Letsch A, Ottensmeier CH, Britten CM, et al. . Harmonization of the intracellular cytokine staining assay. Cancer Immunol Immunother. (2012) 61:967–78. 10.1007/s00262-012-1282-9
    1. Schmitt M, Schmitt A, Wiesneth M, Huckelhoven A, Wu Z, Kuball J, et al. . Peptide vaccination in the presence of adjuvants in patients after hematopoietic stem cell transplantation with CD4+ T cell reconstitution elicits consistent CD8+ T cell responses. Theranostics (2017) 7:1705–18. 10.7150/thno.18301
    1. Schmitt M, Schmitt A, Rojewski MT, Chen J, Giannopoulos K, Fei F, et al. . RHAMM-R3 peptide vaccination in patients with acute myeloid leukemia, myelodysplastic syndrome, and multiple myeloma elicits immunologic and clinical responses. Blood (2008) 111:1357–65. 10.1182/blood-2007-07-099366
    1. Alho AC, Kim HT, Chammas MJ, Reynolds CG, Matos TR, Forcade E, et al. . Unbalanced recovery of regulatory and effector T cells after allogeneic stem cell transplantation contributes to chronic GVHD. Blood (2016) 127:646–57. 10.1182/blood-2015-10-672345
    1. Blazar BR, Murphy WJ, Abedi M. Advances in graft-versus-host disease biology and therapy. Nat Rev Immunol. (2012) 12:443–58. 10.1038/nri3212
    1. Zeiser R, Nguyen VH, Beilhack A, Buess M, Schulz S, Baker J, et al. . Inhibition of CD4+CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production. Blood (2006) 108:390–9. 10.1182/blood-2006-01-0329
    1. Malagola M, Cancelli V, Skert C, Leali PF, Ferrari E, Tiburzi A, et al. . Extracorporeal photopheresis for treatment of acute and chronic graft versus host disease: an italian multicentric retrospective analysis on 94 patients on behalf of the Gruppo Italiano Trapianto di Midollo Osseo. Transplantation (2016) 100:e147–55. 10.1097/TP.0000000000001466
    1. Das-Gupta E, Dignan F, Shaw B, Raj K, Malladi R, Gennery A, et al. . Extracorporeal photopheresis for treatment of adults and children with acute GVHD: UK consensus statement and review of published literature. Bone Marrow Transplant (2014) 49:1251–8. 10.1038/bmt.2014.106
    1. Greinix HT, Worel N, Just U, Knobler R. Extracorporeal photopheresis in acute and chronic graft-versus-host disease. Transfus Apher Sci. (2014) 50:349–57. 10.1016/j.transci.2014.04.005
    1. Holtan SG, Pasquini M, Weisdorf DJ. Acute graft-versus-host disease: a bench-to-bedside update. Blood (2014) 124:363–73. 10.1182/blood-2014-01-514786
    1. Moens L, Kane A, Tangye SG. Naive and memory B cells exhibit distinct biochemical responses following BCR engagement. Immunol Cell Biol. (2016) 94:774–86. 10.1038/icb.2016.41
    1. Buffa S, Pellicano M, Bulati M, Martorana A, Goldeck D, Caruso C, et al. . A novel B cell population revealed by a CD38/CD24 gating strategy: CD38(-)CD24 (-) B cells in centenarian offspring and elderly people. Age (2013) 35:2009–24. 10.1007/s11357-012-9488-5
    1. Sarantopoulos S, Ritz J. Aberrant B-cell homeostasis in chronic GVHD. Blood (2015) 125:1703–7. 10.1182/blood-2014-12-567834
    1. Zeiser R, Sarantopoulos S, Blazar BR. B-cell targeting in chronic graft-versus-host disease. Blood (2018) 131:1399–405. 10.1182/blood-2017-11-784017
    1. Dutt S, Ermann J, Tseng D, Liu YP, George TI, Fathman CG, et al. . L-selectin and beta7 integrin on donor CD4 T cells are required for the early migration to host mesenteric lymph nodes and acute colitis of graft-versus-host disease. Blood (2005) 106:4009–15. 10.1182/blood-2005-06-2339
    1. Juelke K, Killig M, Luetke-Eversloh M, Parente E, Gruen J, Morandi B, et al. . CD62L expression identifies a unique subset of polyfunctional CD56dim NK cells. Blood (2010) 116:1299–307. 10.1182/blood-2009-11-253286
    1. Zhang J, Barefoot BE, Mo W, Deoliveira D, Son J, Cui X, et al. . CD62L- memory T cells enhance T-cell regeneration after allogeneic stem cell transplantation by eliminating host resistance in mice. Blood (2012) 119:6344–53. 10.1182/blood-2011-03-342055
    1. Robb RJ, Lineburg KE, Kuns RD, Wilson YA, Raffelt NC, Olver SD, et al. . Identification and expansion of highly suppressive CD8(+)FoxP3(+) regulatory T cells after experimental allogeneic bone marrow transplantation. Blood (2012) 119:5898–908. 10.1182/blood-2011-12-396119
    1. Blair PA, Norena LY, Flores-Borja F, Rawlings DJ, Isenberg DA, Ehrenstein MR, et al. . CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients. Immunity (2010) 32:129–40. 10.1016/j.immuni.2009.11.009
    1. de Masson A, Bouaziz JD, Le Buanec H, Robin M, O'Meara A, Parquet N, et al. CD24(hi)CD27(+) and plasmablast-like regulatory B cells in human chronic graft-versus-host disease. Blood (2015) 125:1830–9. 10.1182/blood-2014-09-599159
    1. Flores-Borja F, Bosma A, Ng D, Reddy V, Ehrenstein MR, Isenberg DA, et al. . CD19+CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation. Sci Transl Med. (2013) 5:173ra23. 10.1126/scitranslmed.3005407
    1. Hu Y, He GL, Zhao XY, Zhao XS, Wang Y, Xu LP, et al. . Regulatory B cells promote graft-versus-host disease prevention and maintain graft-versus-leukemia activity following allogeneic bone marrow transplantation. Oncoimmunology (2017) 6:e1284721. 10.1080/2162402X.2017.1284721
    1. Rowe V, Banovic T, MacDonald KP, Kuns R, Don AL, Morris ES, et al. . Host B cells produce IL-10 following TBI and attenuate acute GVHD after allogeneic bone marrow transplantation. Blood (2006) 108:2485–92. 10.1182/blood-2006-04-016063
    1. Ford MS, Young KJ, Zhang Z, Ohashi PS, Zhang L. The immune regulatory function of lymphoproliferative double negative T cells in vitro and in vivo. J Exp Med. (2002) 196:261–7. 10.1084/jem.20020029
    1. Gao JF, McIntyre MS, Juvet SC, Diao J, Li X, Vanama RB, et al. . Regulation of antigen-expressing dendritic cells by double negative regulatory T cells. Eur J Immunol. (2011) 41:2699–708. 10.1002/eji.201141428
    1. He KM, Ma Y, Wang S, Min WP, Zhong R, Jevnikar A, et al. . Donor double-negative Treg promote allogeneic mixed chimerism and tolerance. Eur J Immunol. (2007) 37:3455–66. 10.1002/eji.200737408
    1. Hillhouse EE, Delisle JS, Lesage S. Immunoregulatory CD4(-)CD8(-) T cells as a potential therapeutic tool for transplantation, autoimmunity, and cancer. Front Immunol. (2013) 4:6. 10.3389/fimmu.2013.00006
    1. Ma Y, He KM, Garcia B, Min W, Jevnikar A, Zhang ZX. Adoptive transfer of double negative T regulatory cells induces B-cell death in vivo and alters rejection pattern of rat-to-mouse heart transplantation. Xenotransplantation (2008) 15:56–63. 10.1111/j.1399-3089.2008.00444.x
    1. Levine JE, Logan BR, Wu J, Alousi AM, Bolanos-Meade J, Ferrara JL, et al. . Acute graft-versus-host disease biomarkers measured during therapy can predict treatment outcomes: a Blood and Marrow Transplant Clinical Trials Network study. Blood (2012) 119:3854–60. 10.1182/blood-2012-01-403063
    1. Chen YB, Cutler CS. Biomarkers for acute GVHD: can we predict the unpredictable? Bone Marrow Transplant (2013) 48:755–60. 10.1038/bmt.2012.143
    1. Pidala J, Sarwal M, Roedder S, Lee SJ. Biologic markers of chronic GVHD. Bone Marrow Transplant (2014) 49:324–31. 10.1038/bmt.2013.97
    1. de Mooij CEM, Netea MG, van der Velden W, Blijlevens NMA. Targeting the interleukin-1 pathway in patients with hematological disorders. Blood (2017) 129:3155–64. 10.1182/blood-2016-12-754994
    1. Cominelli F, Nast CC, Llerena R, Dinarello CA, Zipser RD. Interleukin 1 suppresses inflammation in rabbit colitis. Mediation by endogenous prostaglandins. J Clin Invest. (1990) 85:582–6. 10.1172/JCI114476
    1. Antin JH, Ferrara JL. Cytokine dysregulation and acute graft-versus-host disease. Blood (1992) 80:2964–8.
    1. Logan RM, Stringer AM, Bowen JM, Gibson RJ, Sonis ST, Keefe DM. Serum levels of NFkappaB and pro-inflammatory cytokines following administration of mucotoxic drugs. Cancer Biol Ther. (2008) 7:1139–45. 10.4161/cbt.7.7.6207
    1. Sartor RB. Cytokines in intestinal inflammation: pathophysiological and clinical considerations. Gastroenterology (1994) 106:533–9. 10.1016/0016-5085(94)90614-9
    1. Wu ZQ, Han XD, Wang Y, Yuan KL, Jin ZM, Di JZ, et al. . Interleukin-1 receptor antagonist reduced apoptosis and attenuated intestinal mucositis in a 5-fluorouracil chemotherapy model in mice. Cancer Chemother Pharmacol. (2011) 68:87–96. 10.1007/s00280-010-1451-5
    1. Toubai T, Mathewson ND, Magenau J, Reddy P. Danger signals and graft-versus-host disease: current understanding and future perspectives. Front Immunol. (2016) 7:539. 10.3389/fimmu.2016.00539
    1. Gartlan KH, Markey KA, Varelias A, Bunting MD, Koyama M, Kuns RD, et al. . Tc17 cells are a proinflammatory, plastic lineage of pathogenic CD8+ T cells that induce GVHD without antileukemic effects. Blood (2015) 126:1609–20. 10.1182/blood-2015-01-622662
    1. Tvedt THA, Ersvaer E, Tveita AA, Bruserud O. Interleukin-6 in allogeneic stem cell transplantation: its possible importance for immunoregulation and as a therapeutic target. Front Immunol. (2017) 8:667. 10.3389/fimmu.2017.00667
    1. Mancusi A, Piccinelli S, Velardi A, Pierini A. The effect of TNF-alpha on regulatory T cell function in graft-versus-host disease. Front Immunol. (2018) 9:356. 10.3389/fimmu.2018.00356
    1. Wang W, Fujii H, Kim HJ, Hermans K, Usenko T, Xie S, et al. . Enhanced human hematopoietic stem and progenitor cell engraftment by blocking donor T cell-mediated TNFalpha signaling. Sci Transl Med. (2017) 9:421. 10.1126/scitranslmed.aag3214
    1. Catchpoole EM, Thirunavukarasu CE, Varelias A, Schlebusch S, Olver S, Zomerdijk N, et al. Early Blood stream infection after BMT is associated with cytokine dysregulation and poor overall survival. Biol Blood Marrow Transplant (2018) 24:1360–6. 10.1016/j.bbmt.2018.02.025
    1. Paczesny S, Krijanovski OI, Braun TM, Choi SW, Clouthier SG, Kuick R, et al. . A biomarker panel for acute graft-versus-host disease. Blood (2009) 113:273–8. 10.1182/blood-2008-07-167098
    1. Greinix HT, Knobler RM, Worel N, Schneider B, Schneeberger A, Hoecker P, et al. . The effect of intensified extracorporeal photochemotherapy on long-term survival in patients with severe acute graft-versus-host disease. Haematologica (2006) 91:405–8.
    1. Kitko CL, Braun T, Couriel DR, Choi SW, Connelly J, Hoffmann S, et al. . Combination therapy for graft-versus-host disease prophylaxis with etanercept and extracorporeal photopheresis: results of a phase II clinical trial. Biol Blood Marrow Transplant (2016) 22:862–8. 10.1016/j.bbmt.2015.11.002
    1. Heshmati F. Updating ECP action mechanisms. Transfus Apher Sci. (2014) 50:330–9. 10.1016/j.transci.2014.04.003
    1. Im A, Pavletic SZ. Deciphering the mystery: extracorporeal photopheresis in graft-versus-host disease. Biol Blood Marrow Transplant (2015) 21:1861–2. 10.1016/j.bbmt.2015.09.011
    1. Jagasia MH, Greinix HT, Arora M, Williams KM, Wolff D, Cowen EW, 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) 21:389–401e1. 10.1016/j.bbmt.2014.12.001

Source: PubMed

3
S'abonner