1,746
Views
9
CrossRef citations to date
0
Altmetric
Research Article

Bendamustine with total body irradiation conditioning yields tolerant T-cells while preserving T-cell-dependent graft-versus-leukemia

, , , , ORCID Icon, & ORCID Icon show all
Article: 1758011 | Received 24 Feb 2020, Accepted 15 Apr 2020, Published online: 30 Apr 2020

References

  • van den Brink MRM, Porter DL, Giralt S, Lu SX, Jenq RR, Hanash A, Bishop MR. Relapse after allogeneic hematopoietic cell therapy. Biol Blood Marrow Trans. 2010;16(1):S138–S145. doi:10.1016/j.bbmt.2009.10.023.
  • Stokes J, Hoffman EA, Zeng Y, Larmonier N, Katsanis E. Post-transplant bendamustine reduces GvHD while preserving GvL in experimental haploidentical bone marrow transplantation. Br J Haematol. 2016;174:102–17. doi:10.1111/bjh.14034.
  • Luznik L, Fuchs EJ. High-dose, post-transplantation cyclophosphamide to promote graft-host tolerance after allogeneic hematopoietic stem cell transplantation. Immunol Res. 2010;47:65–77. doi:10.1007/s12026-009-8139-0.
  • Luznik L, O’Donnell PV, Fuchs EJ. Post-transplantation cyclophosphamide for tolerance induction in HLA-haploidentical bone marrow transplantation. Semin Oncol. 2012;39:683–693. doi:10.1053/j.seminoncol.2012.09.005.
  • Katsanis E, Sapp LN, Varner N, Koza S, Stea B, Zeng Y. Haploidentical bone marrow transplantation with post-transplant cyclophosphamide/bendamustine in pediatric and young adult patients with hematologic malignancies. Biol Blood Marrow Transplant. 2018;24:2034–2039. doi:10.1016/j.bbmt.2018.06.007.
  • Gilli S, Novak U, Taleghani BM, Baerlocher GM, Leibundgut K, Banz Y, Zander T, Betticher D, Egger T, Rauch D, et al. BeEAM conditioning with bendamustine-replacing BCNU before autologous transplantation is safe and effective in lymphoma patients. Ann Hematol. 2017;96(3):421–429. doi:10.1007/s00277-016-2900-y.
  • Martino M, Tripepi G, Messina G, Vincelli ID, Console G, Recchia AG, Gentile M, Molica S, Morabito F. A phase II, single-arm, prospective study of bendamustine plus melphalan conditioning for second autologous stem cell transplantation in de novo multiple myeloma patients through a tandem transplant strategy. Bone Marrow Transplant. 2016;51:1197–1203. doi:10.1038/bmt.2016.94.
  • Hueso T, Gastinne T, Garciaz S, Tchernonog E, Delette C, Casasnovas R-O, Durot E, Houot R, Tessoulin B, Tournilhac O, et al. Bendamustine-EAM versus BEAM regimen in patients with mantle cell lymphoma undergoing autologous stem cell transplantation in the frontline setting: a multicenter retrospective study from Lymphoma Study Association (LYSA) centers. Bone Marrow Transplant. 2020. doi:10.1038/s41409-020-0783-y.
  • Khouri IF, Wei W, Korbling M, Turturro F, Ahmed S, Alousi A, Anderlini P, Ciurea S, Jabbour E, Oran B, et al. BFR (bendamustine, fludarabine, and rituximab) allogeneic conditioning for chronic lymphocytic leukemia/lymphoma: reduced myelosuppression and GVHD. Blood. 2014;124(14):2306–2312. doi:10.1182/blood-2014-07-587519.
  • Khouri IF, Sui D, Jabbour EJ, Samuels BI, Turturro F, Alatrash G, Anderlini P, Ahmed S, Oran B, Ciurea SO, et al. Bendamustine added to allogeneic conditioning improves long-term outcomes in patients with CLL. Bone Marrow Transplant. 2017;52:28–33. doi:10.1038/bmt.2016.204.
  • Holter-Chakrabarty JL, Pierson N, Zhang M-J, Zhu X, Akpek G, Aljurf MD, Artz AS, Baron F, Bredeson CN, Dvorak CC, et al. The sequence of cyclophosphamide and myeloablative total body irradiation in hematopoietic cell transplant for patients with acute leukemia. Biol Blood Marrow Transplant. 2015;21:1251–1257. doi:10.1016/j.bbmt.2015.03.017.
  • Deeg HJ, O’Donnell M, Tolar J, Agarwal R, Harris RE, Feig SA, Territo MC, Collins RH, McSweeney PA, Copelan EA, et al. Optimization of conditioning for marrow transplantation from unrelated donors for patients with aplastic anemia after failure of immunosuppressive therapy. Blood. 2006;108:1485–1491. doi:10.1182/blood-2006-03-005041.
  • Bacigalupo A, Socie’ G, Lanino E, Prete A, Locatelli F, Locasciulli A, Cesaro S, Shimoni A, Marsh J, Brune M, et al. Fludarabine, cyclophosphamide, antithymocyte globulin, with or without low dose total body irradiation, for alternative donor transplants, in acquired severe aplastic anemia: a retrospective study from the EBMT-SAA Working Party. Haematologica. 2010;95(6):976–982. doi:10.3324/haematol.2009.018267.
  • Svenberg P, Remberger M, Svennilson J, Mattsson J, Leblanc K, Gustafsson B, Aschan J, Barkholt L, Winiarski J, Ljungman P, et al. Allogenic stem cell transplantation for nonmalignant disorders using matched unrelated donors. Biol Blood Marrow Transplant. 2004;10(12):877–882. doi:10.1016/j.bbmt.2004.08.002.
  • Deeg HJ, Amylon MD, Harris RE, Collins R, Beatty PG, Feig S, Ramsay N, Territo M, Khan SP, Pamphilon D, et al. Marrow transplants from unrelated donors for patients with aplastic anemia: minimum effective dose of total body irradiation. Biol Blood Marrow Transplant. 2001;7(4):208–215. doi:10.1053/bbmt.2001.v7.pm11349807.
  • Brodsky RA, Luznik L, Bolaños-Meade J, Leffell MS, Jones RJ, Fuchs EJ. Reduced intensity HLA-haploidentical BMT with post transplantation cyclophosphamide in nonmalignant hematologic diseases. Bone Marrow Transplantation. 2008;42(8):523–527. doi:10.1038/bmt.2008.203.
  • Nagler A, Rocha V, Labopin M, Unal A, Ben Othman T, Campos A, Volin L, Poire X, Aljurf M, Masszi T, et al. Allogeneic hematopoietic stem-cell transplantation for acute myeloid leukemia in remission: comparison of intravenous busulfan plus cyclophosphamide (Cy) versus total-body irradiation plus Cy as conditioning regimen–a report from the acute leukemia working party of the European group for blood and marrow transplantation. J Clin Oncol. 2013;31:3549–3556. doi:10.1200/JCO.2013.48.8114.
  • Gyurkocza B, Sandmaier BM. Conditioning regimens for hematopoietic cell transplantation: one size does not fit all. Blood. 2014;124:344–353. doi:10.1182/blood-2014-02-514778.
  • Stokes J, Hoffman EA, Molina MS, Eremija J, Larmonier N, Zeng Y, Katsanis E. Bendamustine with total body irradiation limits murine graft-versus-host disease in part through effects on myeloid-derived suppressor cells. Biol Blood Marrow Transplant. 2019;25:405–416. doi:10.1016/j.bbmt.2018.10.009.
  • Betts BC, Bastian D, Iamsawat S, Nguyen H, Heinrichs JL, Wu Y, Daenthanasanmak A, Veerapathran A, O’Mahony A, Walton K, et al. Targeting JAK2 reduces GVHD and xenograft rejection through regulation of T cell differentiation. Proc Natl Acad Sci U S A. 2018;115(7):1582–1587. doi:10.1073/pnas.1712452115.
  • Nikolic B, Lee S, Bronson RT, Grusby MJ, Sykes M. Th1 and Th2 mediate acute graft-versus-host disease, each with distinct end-organ targets. J Clin Invest. 2000;105:1289–1298. doi:10.1172/JCI7894.
  • Ganguly S, Ross DB, Panoskaltsis-Mortari A, Kanakry CG, Blazar BR, Levy RB, Luznik L. Donor CD4(+) Foxp3(+) regulatory T cells are necessary for posttransplantation cyclophosphamide-mediated protection against GVHD in mice. Blood. 2014;124:2131–2141. doi:10.1182/blood-2013-10-525873.
  • Hoffmann P, Ermann J, Edinger M, Fathman CG, Strober S. Donor-type CD4+CD25+ regulatory T Cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med. 2002;196:389–399. doi:10.1084/jem.20020399.
  • Hotta M, Yoshimura H, Satake A, Tsubokura Y, Ito T, Nomura S. GM-CSF therapy inhibits chronic graft-versus-host disease via expansion of regulatory T cells. Eur J Immunol. 2019;49(1):179–191. doi:10.1002/eji.201847684.
  • Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity. 2016;44:989–1004. doi:10.1016/j.immuni.2016.05.001.
  • Blazar BR, Taylor PA, Panoskaltsis-Mortari A, Sharpe AH, Vallera DA. Opposing roles of CD28: b7and CTLA-4: b7Pathways in Regulating In Vivo alloresponses in murine recipients of MHC disparate T cells. J Immunol. 1999;162:6368.
  • Blazar BR, Carreno BM, Panoskaltsis-Mortari A, Carter L, Iwai Y, Yagita H, Nishimura H, Taylor PA. Blockade of programmed Death-1 engagement accelerates graft-versus-host disease lethality by an IFN-γ-dependent mechanism. J Immunol. 2003;171:1272. doi:10.4049/jimmunol.171.3.1272.
  • Bour-Jordan H, Esensten JH, Martinez-Llordella M, Penaranda C, Stumpf M, Bluestone JA. Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/ B7 family. Immunol Rev. 2011;241(1):180–205. doi:10.1111/j.1600-065X.2011.01011.x.
  • Taylor PA, Panoskaltsis-Mortari A, Freeman GJ, Sharpe AH, Noelle RJ, Rudensky AY, Mak TW, Serody JS, Blazar BR. Targeting of inducible costimulator (ICOS) expressed on alloreactive T cells down-regulates graft-versus-host disease (GVHD) and facilitates engraftment of allogeneic bone marrow (BM). Blood. 2005;105:3372–3380. doi:10.1182/blood-2004-10-3869.
  • Chandrashekar DV, Suresh P, Kumar R, Bhamidipati R, Mullangi R, Richter W, Srinivas N. Sensitive LC-MS/MS method for the simultaneous determination of bendamustine and its active metabolite, gamma-hydroxybendamustine in small volume mice and dog plasma and its application to a pharmacokinetic study in mice and dogs. Drug Res. 2017;67:497–508. doi:10.1055/s-0043-108124.
  • Sadagopan N, Cohen L, Roberts B, Collard W, Omer C. Liquid chromatography-tandem mass spectrometric quantitation of cyclophosphamide and its hydroxy metabolite in plasma and tissue for determination of tissue distribution. J Chromatogr B Biomed Sci Appl. 2001;759:277–284. doi:10.1016/S0378-4347(01)00243-2.
  • Srinivas NR, Richter W, Devaraj V, Suresh P, Bhamdipati R, Mullangi R. Infusion rate dependent pharmacokinetics of bendamustine with altered formation of gamma-hydroxybendamustine (M3) metabolite following 30- and 60-min infusion of bendamustine in rats. Drug Res. 2016;66:351–356. doi:10.1055/s-0042-105745.
  • Cooke KR, Kobzik L, Martin TR, Brewer J, Delmonte JJ, Crawford JM, Ferrara JL. An experimental model of idiopathic pneumonia syndrome after bone marrow transplantation: I. The roles of minor H antigens and endotoxin. Blood. 1996;88(8):3230–3239. doi:10.1182/blood.V88.8.3230.bloodjournal8883230.
  • Gatwood KS, Labopin M, Savani BN, Finke J, Socie G, Beelen D, Yakoub-Agha I, Chevallier P, Ganser A, Blaise D, et al. Transplant outcomes for patients with therapy-related acute myeloid leukemia with prior lymphoid malignancy: an ALWP of EBMT study. Bone Marrow Transplant. 2020;55(1):224–232. doi:10.1038/s41409-019-0673-3.
  • Rubio MT, D’Aveni-Piney, M., Labopin, M., Hamladji, R.M., Sanz, M.A., Blaise, D., Ozdogu, H., Daguindeau, E., Richard, C., Santarone, S, et al. Impact of in vivo T cell depletion in HLA-identical allogeneic stem cell transplantation for acute myeloid leukemia in first complete remission conditioned with a fludarabine iv-busulfan myeloablative regimen: a report from the EBMT Acute Leukemia Working Party. J Hematol Oncol. 2017;10:31.
  • Konuma T, Tsukada N, Kanda J, Uchida N, Ohno Y, Miyakoshi S, Kanamori H, Hidaka M, Sakura T, Onizuka M, et al. Comparison of transplant outcomes from matched sibling bone marrow or peripheral blood stem cell and unrelated cord blood in patients 50 years or older. Am J Hematol. 2016;91(5):E284–E292. doi:10.1002/ajh.24340.
  • Holtan SG, DeFor TE, Lazaryan A, Bejanyan N, Arora M, Brunstein CG, Blazar BR, MacMillan ML, Weisdorf DJ. Composite end point of graft-versus-host disease-free, relapse-free survival after allogeneic hematopoietic cell transplantation. Blood. 2015;125(8):1333–1338. doi:10.1182/blood-2014-10-609032.
  • Centuori SM, Trad M, LaCasse CJ, Alizadeh D, Larmonier CB, Hanke NT, Kartchner J, Janikashvili N, Bonnotte B, Larmonier N. Myeloid-derived suppressor cells from tumor-bearing mice impair TGF-β-induced differentiation of CD4+CD25+FoxP3+ Tregs from CD4+CD25-FoxP3- T cells. J Leukoc Biol. 2012;92:987–997. doi:10.1189/jlb.0911465.
  • Zeng Y, Stokes J, Hahn S, Hoffman E, Katsanis E. Activated MHC-mismatched T helper-1 lymphocyte infusion enhances GvL with limited GvHD. Bone Marrow Transplant. 2014;49:1076. doi:10.1038/bmt.2014.91.
  • Zeng Y, Hahn S, Stokes J, Hoffman EA, Schmelz M, Proytcheva M, Chernoff J, Katsanis E. Pak2 regulates myeloid-derived suppressor cell development in mice. Blood Adv. 2017;1:1923–1933. doi:10.1182/bloodadvances.2017007435.
  • Kruisbeek AM, Shevach E, Thornton AM. Proliferative assays for T cell function. Curr Protoc Immunol. 2004;60(1):3. 12.11–13.12.20. doi:10.1002/0471142735.im0312s60.
  • Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc. 1958;53:457–481. doi:10.1080/01621459.1958.10501452.
  • Peto R, Peto J. Asymptotically efficient rank invariant test procedures. J R Stat Soc Ser A (General). 1972;135:185–207. doi:10.2307/2344317.
  • Taylor PA, Lees CJ, Blazar BR. The infusion of ex vivo activated and expanded CD4(+)CD25(+) immune regulatory cells inhibits graft-versus-host disease lethality. Blood. 2002;99:3493–3499. doi:10.1182/blood.V99.10.3493.
  • Mutis T, van Rijn RS, Simonetti ER, Aarts-Riemens T, Emmelot ME, van Bloois L, Martens A, Verdonck LF, Ebeling SB. Human regulatory T cells control xenogeneic graft-versus-host disease induced by autologous T cells in RAG2−/−γc−/− Immunodeficient mice. Clin Cancer Res. 2006;12:5520. doi:10.1158/1078-0432.CCR-06-0035.
  • Pierini A, Colonna L, Alvarez M, Schneidawind D, Nishikii H, Baker J, Pan Y, Florek M, Kim B-S, Negrin RS, et al. Donor requirements for regulatory T cell suppression of murine graft-versus-host disease. J Immunol. 2015;195(1):347. doi:10.4049/jimmunol.1402861.
  • Blazar BR, Lindberg FP, Ingulli E, Panoskaltsis-Mortari A, Oldenborg P-A, Iizuka K, Yokoyama WM, Taylor PA. CD47 (integrin-associated protein) engagement of dendritic cell and macrophage counterreceptors is required to prevent the clearance of donor lymphohematopoietic cells. J Exp Med. 2001;194(4):541–549. doi:10.1084/jem.194.4.541.
  • Zeiser R, Youssef S, Baker J, Kambham N, Steinman L, Negrin RS. Preemptive HMG-CoA reductase inhibition provides graft-versus-host disease protection by Th-2 polarization while sparing graft-versus-leukemia activity. Blood. 2007;110(13):4588–4598. doi:10.1182/blood-2007-08-106005.
  • Malard F, Gaugler B, Lamarthee B, Mohty M. Translational opportunities for targeting the Th17 axis in acute graft-vs.-host disease. Mucosal Immunol. 2016;9:299–308. doi:10.1038/mi.2015.143.
  • Blazar BR, Sharpe AH, Chen AI, Panoskaltsis-Mortari A, Lees C, Akiba H, Yagita H, Killeen N, Taylor PA. Ligation of OX40 (CD134) regulates graft-versus-host disease (GVHD) and graft rejection in allogeneic bone marrow transplant recipients. Blood. 2003;101(9):3741–3748. doi:10.1182/blood-2002-10-3048.
  • Cassady K, Martin PJ, Zeng D. Regulation of GVHD and GVL activity via PD-L1 interaction with PD-1 and CD80. Front Immunol. 2018;9:3061. doi:10.3389/fimmu.2018.03061.
  • Albring JC, Sandau MM, Rapaport AS, Edelson BT, Satpathy A, Mashayekhi M, Lathrop SK, Hsieh C-S, Stelljes M, Colonna M, et al. Targeting of B and T lymphocyte associated (BTLA) prevents graft-versus-host disease without global immunosuppression. J Exp Med. 2010;207(12):2551–2559. doi:10.1084/jem.20102017.
  • Sabatos CA, Chakravarti S, Cha E, Schubart A, Sánchez-Fueyo A, Zheng XX, Coyle AJ, Strom TB, Freeman GJ, Kuchroo VK, et al. Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol. 2003;4(11):1102–1110. doi:10.1038/ni988.
  • Zhang P, Hill GR. Interleukin-10 mediated immune regulation after stem cell transplantation: mechanisms and implications for therapeutic intervention. Semin Immunol. 2019;44:101322. doi:10.1016/j.smim.2019.101322.
  • Henden AS, Hill GR. Cytokines in graft-versus-host disease. J Immunol. 2015;194(10):4604. doi:10.4049/jimmunol.1500117.
  • Wilkinson AN, Chang K, Kuns RD, Henden AS, Minnie SA, Ensbey KS, Clouston AD, Zhang P, Koyama M, Hidalgo J, et al. IL-6 dysregulation originates in dendritic cells and mediates graft-versus-host disease via classical signaling. Blood. 2019;134(23):2092–2106. doi:10.1182/blood.2019000396.
  • Yu H, Tian Y, Wang Y, Mineishi S, Zhang Y. Dendritic Cell Regulation of Graft-Vs.-Host Disease: immunostimulation and Tolerance. Front Immunol. 2019;10:93. doi:10.3389/fimmu.2019.00093.
  • MacDonald KP, Shlomchik WD, Reddy P. Biology of graft-versus-host responses: recent insights. Biol Blood Marrow Trans. 2013;19(1):S10–S14. doi:10.1016/j.bbmt.2012.11.005.
  • Priyadharshini B, Greiner DL, Brehm MA. T-cell activation and transplantation tolerance. Transplantation Reviews. 2012;26(3):212–222. doi:10.1016/j.trre.2011.09.002.
  • Thangavelu G, Blazar BR. Achievement of tolerance induction to prevent acute graft-vs.-host disease. Front Immunol. 2019;10:309. doi:10.3389/fimmu.2019.00309.
  • Bluestone JA. Mechanisms of tolerance. Immunol Rev. 2011;241(1):5–19. doi:10.1111/j.1600-065X.2011.01019.x.
  • Brennan TV, Yang Y. PD-L1 serves as a double agent in separating GVL from GVHD. J Clin Invest. 2017;127(5):1627–1630. doi:10.1172/JCI94196.
  • Burlion A, Brunel S, Petit NY, Olive D, Marodon G. Targeting the human T-cell inducible costimulator molecule with a monoclonal antibody prevents graft-vs-host disease and preserves graft vs leukemia in a xenograft murine model. Front Immunol. 2017;8:756. doi:10.3389/fimmu.2017.00756.
  • Yi T, Chen Y, Wang L, Du G, Huang D, Zhao D, Johnston H, Young J, Todorov I, Umetsu DT, et al. Reciprocal differentiation and tissue-specific pathogenesis of Th1, Th2, and Th17 cells in graft-versus-host disease. Blood. 2009;114(14):3101–3112. doi:10.1182/blood-2009-05-219402.
  • Murphy WJ, Welniak LA, Taub DD, Wiltrout RH, Taylor PA, Vallera DA, Kopf M, Young H, Longo DL, Blazar BR, et al. Differential effects of the absence of interferon-gamma and IL-4 in acute graft-versus-host disease after allogeneic bone marrow transplantation in mice. J Clin Invest. 1998;102(9):1742–1748. doi:10.1172/JCI3906.
  • Nagaraj S, Gupta K, Pisarev V, Kinarsky L, Sherman S, Kang L, Herber DL, Schneck J, Gabrilovich DI. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med. 2007;13(7):828. doi:10.1038/nm1609.
  • Yang F, Li Y, Wu T, Na N, Zhao Y, Li W, Han C, Zhang L, Lu J, Zhao Y, et al. TNFα-induced M-MDSCs promote transplant immune tolerance via nitric oxide. J Mol Med (Berl). 2016;94(8):911–920. doi:10.1007/s00109-016-1398-z.
  • Veenstra RG, Flynn R, Kreymborg K, McDonald-Hyman C, Saha A, Taylor PA, Osborn MJ, Panoskaltsis-Mortari A, Schmitt-Graeff A, Lieberknecht E, et al. B7-H3 expression in donor T cells and host cells negatively regulates acute graft-versus-host disease lethality. Blood. 2015;125(21):3335–3346. doi:10.1182/blood-2014-09-603357.
  • Levine JE. Implications of TNF-α in the pathogenesis and management of GVHD. Int J Hematol. 2011;93:571–577. doi:10.1007/s12185-011-0803-1.
  • D’andrea A, Aste-Amezaga M, Valiante NM, Ma X, Kubin M, Trinchieri G. Interleukin 10 (IL-10) Inhibits human lymphocyte interferon γ-production by suppressing natural killer cell stimulatory factor/IL-12 synthesis in accessory cells. J Exp Med. 1993;178:1041–1048. doi:10.1084/jem.178.3.1041.
  • Chaudhry A, Samstein R, Treuting P, Liang Y, Pils M, Heinrich J-M, Jack R, Wunderlich F, Brüning J, Müller W, et al. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity. 2011;34(4):566–578. doi:10.1016/j.immuni.2011.03.018.
  • Park MJ, Lee SH, Lee S-H, Lee E-J, Kim E-K, Choi JY, Cho M-L. IL-1 receptor blockade alleviates graft-versus-host disease through downregulation of an interleukin-1beta-dependent glycolytic pathway in Th17 cells. Mediators Inflamm. 2015;2015:631384. doi:10.1155/2015/631384.
  • Lu L, Yoshimoto K, Morita A, Kameda H, Takeuchi T. Bendamustine increases interleukin-10 secretion from B cells via p38 MAP kinase activation. Int Immunopharmacol. 2016;39:273–279. doi:10.1016/j.intimp.2016.07.033.
  • Reshef R, Luger SM, Hexner EO, Loren AW, Frey NV, Nasta SD, Goldstein SC, Stadtmauer EA, Smith J, Bailey S, et al. Blockade of lymphocyte chemotaxis in visceral graft-versus-host disease. N Engl J Med. 2012;367:135–145. doi:10.1056/NEJMoa1201248.
  • Reshef R, Ganetsky A, Acosta EP, Blauser R, Crisalli L, McGraw J, Frey NV, Hexner EO, Hoxie JA, Loren AW, et al. Extended CCR5 blockade for graft-versus-host disease prophylaxis improves outcomes of reduced-intensity unrelated donor hematopoietic cell transplantation: a phase II clinical trial. Biol Blood Marrow Transplant. 2019;25:515–521. doi:10.1016/j.bbmt.2018.09.034.
  • Munchel AT, Kasamon YL, Fuchs EJ. Treatment of hematological malignancies with nonmyeloablative, HLA-haploidentical bone marrow transplantation and high dose, post-transplantation cyclophosphamide. Best Pract Res Clin Haematol. 2011;24(3):359–368. doi:10.1016/j.beha.2011.05.001.