Inhibition of BTK and ITK with Ibrutinib Is Effective in the Prevention of Chronic Graft-versus-Host Disease in Mice

Steven D Schutt, Jianing Fu, Hung Nguyen, David Bastian, Jessica Heinrichs, Yongxia Wu, Chen Liu, Daniel G McDonald, Joseph Pidala, Xue-Zhong Yu, Steven D Schutt, Jianing Fu, Hung Nguyen, David Bastian, Jessica Heinrichs, Yongxia Wu, Chen Liu, Daniel G McDonald, Joseph Pidala, Xue-Zhong Yu

Abstract

Bruton's Tyrosine Kinase (BTK) and IL-2 Inducible T-cell Kinase (ITK) are enzymes responsible for the phosphorylation and activation of downstream effectors in the B-cell receptor (BCR) signaling and T cell receptor (TCR) signaling pathways, respectively. Ibrutinib is an FDA-approved potent inhibitor of both BTK and ITK that impairs B-cell and T-cell function. CD4 T cells and B cells are essential for the induction of chronic graft-versus-host disease (cGVHD). We evaluated these targets by testing the ability of Ibrutinib to prevent or ameliorate cGVHD, which is one of the major complications for patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT). We found that Ibrutinib significantly alleviated cGVHD across four different mouse models, accompanied by increased long-term survival and reduced clinical score. The clinical improvements in Ibrutinib-treated recipients were associated with decreased serum-autoantibodies, costimulatory molecule activation, B-cell proliferation, and glomerulonephritis compared to vehicle controls. Ibrutinib was also able to alleviate the clinical manifestations in acute GVHD (aGVHD), where the recipients were given grafts with or without B cells, suggesting that an inhibitory effect of Ibrutinib on T cells contributes to a reduction in both aGVHD and cGVHD pathogenesis. An effective prophylactic regimen is still lacking to both reduce the incidence and severity of human cGVHD following allo-HSCT. Our study shows that Ibrutinib is an effective prophylaxis against several mouse models of cGVHD with minimal toxicity and could be a promising strategy to combat human cGVHD clinically.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Ibrutinib improves survival and reduces…
Fig 1. Ibrutinib improves survival and reduces glomerulonephritis induced proteinuria.
Lethally irradiated BALB/c recipients were transplanted with TCD-BM (5 x 106 per mouse) from DBA/2 donors with (n = 26) or without (n = 16) CD25- splenocytes at 40 x 106 per mouse. Groups were either given no treatment, daily oral gavage of vehicle alone, or Ibrutinib 10 mg/kg beginning 2–4 h before BMT and continued for 4 weeks. Mice were monitored for 60 d for survival (A) and proteinuria twice/week (B). Data are shown from two replicate experiments combined. In separate experiments under the same conditions as in A and B, daily oral gavage of vehicle (n = 13) and Ibrutinib treatment (n = 26) was delayed by either 1 (n = 13) or 2 (n = 13) weeks post-BMT and continued for 4 weeks. Mice were monitored for 60 d for survival (C) proteinuria twice/week (D). Data are shown from three replicate experiments combined. Asterisk indicates statistical significance between vehicle treatment and Ibrutinib treatment groups: *p<0.05, **p<0.01, ***p<0.001.
Fig 2. Ibrutinib reduces B-cell proliferation, costimulatory…
Fig 2. Ibrutinib reduces B-cell proliferation, costimulatory molecules, and dsDNA autoantibodies in cGVHD.
Lethally irradiated BALB/c recipients were transplanted with or without CFSE labeled CD25- splenocytes from DBA/2 donors at a dose of 40 x 106 per mouse. Groups were either given no treatment (n = 6), daily oral gavage of vehicle alone (n = 12), or Ibrutinib 10 mg/kg (n = 12) beginning 2–4 hours before BMT and continued for 4 days. Recipient mice were euthanized 4 days after transplant and spleen was taken for FACS analysis. The percentage of CFSE dilution (A, B) represents the amount of proliferated donor B cells. The expression of CD40, CD80, and CD86 costimulatory molecules on B cells were analyzed by gating on B220+ cells and shown as mean florescence intensity (MFI) (C). D panel represents a separate experimental design where BMTs and the number of subjects were as described previously except recipients were sacrificed 28 days after BMT and the Ibrutinib treatment duration was 4 weeks. Serum from whole blood was taken on day 28 post-BMT for ELISA measuring autoantibodies IgG and IgG2a (D). Data are pooled from three replicate experiments. Asterisk indicates statistical significance: *p<0.05.
Fig 3. Ibrutinib reduces glomerulonephritis and dsDNA…
Fig 3. Ibrutinib reduces glomerulonephritis and dsDNA autoantibodies.
Unconditioned B6D2F1 recipients were transplanted with 80–95 x 106 splenocytes without BM from DBA/2 donors. Groups were either given no treatment (n = 6), daily oral gavage of vehicle (n = 15), or two different dosages of Ibrutinib, 10 mg/kg (n = 5) or 20mg/kg (n = 10) 2–4 hours before transplant and continued for 4 weeks. Animals were monitored for proteinuria (A). Serum from whole blood was taken bi-weekly to monitor levels of IgG (B) and IgG2a (C) autoantibodies. The data are pooled from three replicate experiments. Asterisk indicates statistical significance between vehicle treatment and Ibrutinib treatment groups: *p<0.05.
Fig 4. Ibrutinib delays onset and reduces…
Fig 4. Ibrutinib delays onset and reduces severity of scleroderma development.
Lethally irradiated BALB/c recipients were transplanted with TCD-BM (5 x 106 per mouse) from B10.D2 donors with or without whole splenocytes at a dose of 5 x 106 per mouse. Groups were either given no treatment (n = 4), daily oral gavage of vehicle alone (n = 9), or Ibrutinib at the dose of 10 mg/kg (n = 9) beginning 2–4 hours before BMT and continued for 4 weeks. Animals were monitored for survival and clinical score using a scoring system (A) and visual representations (C). At day 60, recipients were sacrificed and skin biopsies were taken for H&E staining (B) and scoring (D). Spleens were excised for FACS analysis (E-H). Percentage of B220loCD138+ plasma cells were reported for BM, vehicle, and Ibrutinib treated groups (E). Representative flow plot (F) as well as the combined data (G) for the percentage of Tfh cells were shown. Absolute numbers of B220+ cells were calculated and presented (H). Data shown is pooled from two replicate experiments. Asterisk indicates statistical significance between vehicle treatment and Ibrutinib treatment groups: *p<0.05, **p<0.01, ***p<0.001.
Fig 5. Ibrutinib improves survival and clinical…
Fig 5. Ibrutinib improves survival and clinical score.
Lethally irradiated BALB/c recipients were transplanted with TCD-BM (5 x 106 per mouse) from B6 donors with or without whole splenocytes at a dose of 1–2 x 106 per mouse. Groups either received no treatment (n = 4), daily oral gavage of vehicle (n = 10), or Ibrutinib 10 mg/kg (n = 11) 2–4 hours before BMT and continued for 4 weeks. Survival rate (A) and cGVHD clinical scores (B) were shown from two replicate experiments combined. Asterisk indicates statistical significance between vehicle treatment and Ibrutinib treatment groups: *p<0.05. A one-tailed student t test was used in (B).
Fig 6. Ibrutinib improves survival and clinical…
Fig 6. Ibrutinib improves survival and clinical manifestations of cGVHD independent of B cells.
Lethally irradiated BALB/c recipients were transplanted with TCD-BM (5 x 106 per mouse) plus purified T cells (0.5 x 106 per mouse) from B6.129S2-Ighmtm1Cgn/J (B-cell deficient; BKO) donors, and received daily oral gavage of vehicle (n = 10), or Ibrutinib at 10 mg/kg (n = 10), lasting 4 weeks. Bone marrow alone controls received TCD-BM (5 x 106 per mouse) from WT B6 mice without treatment (n = 4). Animals were monitored for survival (A) and aGVHD clinical score (B) for 80 days. Data are shown from two replicate experiments combined. Asterisk indicates statistical significance between vehicle treatment and Ibrutinib treatment groups: **p<0.01.

References

    1. Schroeder MA, DiPersio JF. Mouse models of graft-versus-host disease: advances and limitations. Disease models & mechanisms. 2011;4(3):318–33. 10.1242/dmm.006668
    1. Kansu E. Administration of cyclosporine for 24 months compared with 6 months for prevention of chronic graft-versus-host disease: a prospective randomized clinical trial. Blood. 2001;98(13):3868–70.
    1. Vogelsang GB. How I treat chronic graft-versus-host disease. Blood. 2001;97(5):1196–201.
    1. Zhang C, Todorov I, Zhang Z, Liu Y, Kandeel F, Forman S, et al. Donor CD4+ T and B cells in transplants induce chronic graft-versus-host disease with autoimmune manifestations. Blood. 2006;107(7):2993–3001.
    1. Wu T, Young JS, Johnston H, Ni X, Deng R, Racine J, et al. Thymic damage, impaired negative selection, and development of chronic graft-versus-host disease caused by donor CD4+ and CD8+ T cells. Journal of immunology (Baltimore, Md: 1950). 2013;191(1):488–99.
    1. Srinivasan M, Flynn R, Price A, Ranger A, Browning JL, Taylor PA, et al. Donor B-cell alloantibody deposition and germinal center formation are required for the development of murine chronic GVHD and bronchiolitis obliterans. Blood. 2012;119(6):1570–80. 10.1182/blood-2011-07-364414
    1. Kapur R, Ebeling S, Hagenbeek A. B-cell involvement in chronic graft-versus-host disease. Haematologica. 2008;93(11):1702–11. 10.3324/haematol.13311
    1. Allen JL, Tata PV, Fore MS, Wooten J, Rudra S, Deal AM, et al. Increased BCR responsiveness in B cells from patients with chronic GVHD. Blood. 2014;123(13):2108–15. 10.1182/blood-2013-10-533562
    1. Zhao D, Young JS, Chen YH, Shen E, Yi T, Todorov I, et al. Alloimmune response results in expansion of autoreactive donor CD4+ T cells in transplants that can mediate chronic graft-versus-host disease. Journal of immunology (Baltimore, Md: 1950). 2011;186(2):856–68.
    1. Satterthwaite AB, Witte ON. The role of Bruton's tyrosine kinase in B-cell development and function: a genetic perspective. Immunological reviews. 2000;175:120–7.
    1. Gibson S, August A, Kawakami Y, Kawakami T, Dupont B, Mills GB. The EMT/ITK/TSK (EMT) tyrosine kinase is activated during TCR signaling: LCK is required for optimal activation of EMT. Journal of immunology (Baltimore, Md: 1950). 1996;156(8):2716–22.
    1. August A, Gibson S, Kawakami Y, Kawakami T, Mills GB, Dupont B. CD28 is associated with and induces the immediate tyrosine phosphorylation and activation of the Tec family kinase ITK/EMT in the human Jurkat leukemic T-cell line. Proceedings of the National Academy of Sciences of the United States of America. 1994;91(20):9347–51.
    1. Lopez-Herrera G, Vargas-Hernandez A, Gonzalez-Serrano ME, Berron-Ruiz L, Rodriguez-Alba JC, Espinosa-Rosales F, et al. Bruton's tyrosine kinase—an integral protein of B cell development that also has an essential role in the innate immune system. Journal of leukocyte biology. 2014;95(2):243–50. 10.1189/jlb.0513307
    1. Mohamed AJ, Nore BF, Christensson B, Smith CI. Signalling of Bruton's tyrosine kinase, Btk. Scandinavian journal of immunology. 1999;49(2):113–8.
    1. Miller AT, Wilcox HM, Lai Z, Berg LJ. Signaling through Itk promotes T helper 2 differentiation via negative regulation of T-bet. Immunity. 2004;21(1):67–80.
    1. Gomez-Rodriguez J, Wohlfert EA, Handon R, Meylan F, Wu JZ, Anderson SM, et al. Itk-mediated integration of T cell receptor and cytokine signaling regulates the balance between Th17 and regulatory T cells. The Journal of experimental medicine. 2014;211(3):529–43. 10.1084/jem.20131459
    1. Dubovsky JA, Beckwith KA, Natarajan G, Woyach JA, Jaglowski S, Zhong Y, et al. Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1-selective pressure in T lymphocytes. Blood. 2013;122(15):2539–49. 10.1182/blood-2013-06-507947
    1. Byrd JC, Brown JR, O'Brien S, Barrientos JC, Kay NE, Reddy NM, et al. Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia. The New England journal of medicine. 2014;371(3):213–23. 10.1056/NEJMoa1400376
    1. Chu YW, Gress RE. Murine models of chronic graft-versus-host disease: insights and unresolved issues. Biology of blood and marrow transplantation: journal of the American Society for Blood and Marrow Transplantation. 2008;14(4):365–78.
    1. Tschetter JR, Mozes E, Shearer GM. Progression from acute to chronic disease in a murine parent-into-F1 model of graft-versus-host disease. Journal of immunology (Baltimore, Md: 1950). 2000;165(10):5987–94.
    1. Le Huu D, Matsushita T, Jin G, Hamaguchi Y, Hasegawa M, Takehara K, et al. Donor-derived regulatory B cells are important for suppression of murine sclerodermatous chronic graft-versus-host disease. Blood. 2013;121(16):3274–83. 10.1182/blood-2012-11-465658
    1. Yu Y, Wang D, Liu C, Kaosaard K, Semple K, Anasetti C, et al. Prevention of GVHD while sparing GVL effect by targeting Th1 and Th17 transcription factor T-bet and RORgammat in mice. Blood. 2011;118(18):5011–20. 10.1182/blood-2011-03-340315
    1. Cooke KR, Kobzik L, Martin TR, Brewer J, Delmonte J Jr., Crawford JM, et al. An experimental model of idiopathic pneumonia syndrome after bone marrow transplantation: I. The roles of minor H antigens and endotoxin. Blood. 1996;88(8):3230–9.
    1. Liang Y, Liu C, Djeu JY, Zhong B, Peters T, Scharffetter-Kochanek K, et al. Beta2 integrins separate graft-versus-host disease and graft-versus-leukemia effects. Blood. 2008;111(2):954–62.
    1. Anderson BE, McNiff J, Yan J, Doyle H, Mamula M, Shlomchik MJ, et al. Memory CD4+ T cells do not induce graft-versus-host disease. The Journal of clinical investigation. 2003;112(1):101–8.
    1. Valenzuela JO, Iclozan C, Hossain MS, Prlic M, Hopewell E, Bronk CC, et al. PKCtheta is required for alloreactivity and GVHD but not for immune responses toward leukemia and infection in mice. The Journal of clinical investigation. 2009;119(12):3774–86. 10.1172/JCI39692
    1. Singh RR, Kumar V, Ebling FM, Southwood S, Sette A, Sercarz EE, et al. T cell determinants from autoantibodies to DNA can upregulate autoimmunity in murine systemic lupus erythematosus. The Journal of experimental medicine. 1995;181(6):2017–27.
    1. Anderson BE, McNiff JM, Jain D, Blazar BR, Shlomchik WD, Shlomchik MJ. Distinct roles for donor- and host-derived antigen-presenting cells and costimulatory molecules in murine chronic graft-versus-host disease: requirements depend on target organ. Blood. 2005;105(5):2227–34.
    1. Yu Y, Wang D, Kaosaard K, Liu C, Fu J, Haarberg K, et al. c-Rel is an essential transcription factor for the development of acute graft-versus-host disease in mice. European journal of immunology. 2013;43(9):2327–37. 10.1002/eji.201243282
    1. Fu J, Wang D, Yu Y, Heinrichs J, Wu Y, Schutt S, et al. T-bet Is Critical for the Development of Acute Graft-versus-Host Disease through Controlling T Cell Differentiation and Function. Journal of immunology (Baltimore, Md: 1950). 2014; 10.4049/jimmunol.1401618
    1. Dubovsky JA, Flynn R, Du J, Harrington BK, Zhong Y, Kaffenberger B, et al. Ibrutinib treatment ameliorates murine chronic graft-versus-host disease. The Journal of clinical investigation. 2014;124(11):4867–76. 10.1172/JCI75328
    1. Hata D, Kitaura J, Hartman SE, Kawakami Y, Yokota T, Kawakami T. Bruton's tyrosine kinase-mediated interleukin-2 gene activation in mast cells. Dependence on the c-Jun N-terminal kinase activation pathway. The Journal of biological chemistry. 1998;273(18):10979–87.
    1. Weil D, Power MA, Smith SI, Li CL. Predominant expression of murine Bmx tyrosine kinase in the granulo-monocytic lineage. Blood. 1997;90(11):4332–40.
    1. Yuko Kawakami LY, Miura Toru, Tsukada Satoshi, Witte Owen N., and Kawakami Toshiaki. Tyrosine Phosphorylation and Activation of BrutonTyrosine Kinase upon FcFRI Cross-Linking. Molecular and Cellular Biology. 1994;14(8):5108–13.
    1. Young JS, Wu T, Chen Y, Zhao D, Liu H, Yi T, et al. Donor B cells in transplants augment clonal expansion and survival of pathogenic CD4+ T cells that mediate autoimmune-like chronic graft-versus-host disease. Journal of immunology (Baltimore, Md: 1950). 2012;189(1):222–33.
    1. Ogawa Y. Periductal Area as the Primary Site for T-Cell Activation in Lacrimal Gland Chronic Graft-Versus-Host Disease. Investigative Ophthalmology & Visual Science. 2003;44(5):1888–96.
    1. DeNucci CC, Pagan AJ, Mitchell JS, Shimizu Y. Control of alpha4beta7 integrin expression and CD4 T cell homing by the beta1 integrin subunit. Journal of immunology (Baltimore, Md: 1950). 2010;184(5):2458–67.
    1. Woods ML, Kivens WJ, Adelsman MA, Qiu Y, August A, Shimizu Y. A novel function for the Tec family tyrosine kinase Itk in activation of beta 1 integrins by the T-cell receptor. The EMBO journal. 2001;20(6):1232–44.
    1. Ala A, Dhillon AP, Hodgson HJ. Role of cell adhesion molecules in leukocyte recruitment in the liver and gut. International Journal of Experimental Pathology. 2003;84(1):1–16.
    1. Westermann J, Söllner S, Ehlers E-M, Nohroudi K, Blessenohl M, Kalies K. Analyzing the Migration of Labeled T Cells In Vivo: An Essential Approach with Challenging Features. Laboratory Investigation. 2003;83(4):459–69.
    1. Levade M, David E, Garcia C, Laurent PA, Cadot S, Michallet AS, et al. Ibrutinib treatment affects collagen and von Willebrand factor-dependent platelet functions. Blood. 2014;124(26):3991–5. 10.1182/blood-2014-06-583294
    1. Jain N, Miu B, Jiang JK, McKinstry KK, Prince A, Swain SL, et al. CD28 and ITK signals regulate autoreactive T cell trafficking. Nature medicine. 2013;19(12):1632–7. 10.1038/nm.3393
    1. Flynn R, Du J, Veenstra RG, Reichenbach DK, Panoskaltsis-Mortari A, Taylor PA, et al. Increased T follicular helper cells and germinal center B cells are required for cGVHD and bronchiolitis obliterans. Blood. 2014;123(25):3988–98. 10.1182/blood-2014-03-562231
    1. Socie G, Ritz J. Current issues in chronic graft-versus-host disease. Blood. 2014;124(3):374–84. 10.1182/blood-2014-01-514752
    1. <HIGHLIGHTS OF PRESCRIBING INFORMATION [Internet]. U.S. Food and Drug Administration; 2013. [cited 2015]. Available from: .

Source: PubMed

3
購読する