Preemptive interferon-α treatment could protect against relapse and improve long-term survival of ALL patients after allo-HSCT

Sining Liu, Xueyi Luo, Xiaohui Zhang, Lanping Xu, Yu Wang, Chenhua Yan, Huan Chen, Yuhong Chen, Wei Han, Fengrong Wang, Jingzhi Wang, Kaiyan Liu, Xiaojun Huang, Xiaodong Mo, Sining Liu, Xueyi Luo, Xiaohui Zhang, Lanping Xu, Yu Wang, Chenhua Yan, Huan Chen, Yuhong Chen, Wei Han, Fengrong Wang, Jingzhi Wang, Kaiyan Liu, Xiaojun Huang, Xiaodong Mo

Abstract

Relapse was the major cause of treatment failure in patients with acute lymphoblastic leukemia (ALL) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). We aimed to identify the efficacy and safety of preemptive interferon-α (IFN-α) treatment in ALL patients who had minimal residual disease (MRD) after allo-HSCT. Multiparameter flow cytometry and polymerase chain reaction assays were applied for MRD monitoring. Recombinant human IFN-α-2b injections were administered subcutaneously twice weekly in every 4 weeks cycle. Twenty-four (35.3%), 5 (7.4%), 6 (8.8%), and 13 (19.1%) patients achieved MRD negativity at 1, 2, 3, and > 3 months, respectively, after treatment. Seven patients showed grade ≥ 3 toxicities after IFN-α treatment. The 4-year cumulative incidence of total acute graft-versus-host disease (aGVHD), severe aGVHD, total chronic GVHD (cGVHD), and severe cGVHD after treatment was 14.7%, 2.9%, 40.0%, and 7.5%, respectively. The 4-year cumulative incidences of relapse and non-relapse mortality after treatment was 31.9% and 6.0%, respectively. The 4-year probabilities of disease-free survival and overall survival after IFN-α treatment were 62.1% and 71.1%, respectively. Thus, preemptive IFN-α treatment could protect against relapse and improve long-term survival for ALL patients who had MRD after allo-HSCT. The study was registered at https://ichgcp.net/clinical-trials-registry/NCT02185261" title="See in ClinicalTrials.gov">#NCT02185261 (09/07/2014).

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Univariate analysis for prognostic factors of preemptive IFN-α treatment: (A) relapse; (B) disease-free survival, and (C) overall survival.
Figure 2
Figure 2
Cumulative incidence of survival after MRD positivity between the preemptive IFN-α treatment group in the present study and those who had MRD but did not receive interventions in the historical cohort: (A) relapse; (B) non-relapse mortality; (C) disease-free survival, and (D) overall survival.
Figure 3
Figure 3
Diagram of patients enrolled.

References

    1. Xu L, et al. The consensus on indications, conditioning regimen, and donor selection of allogeneic hematopoietic cell transplantation for hematological diseases in China-recommendations from the Chinese Society of Hematology. J. Hematol. Oncol. 2018;11:33–33. doi: 10.1186/s13045-018-0564-x.
    1. Yan CH, et al. Causes of mortality after haploidentical hematopoietic stem cell transplantation and the comparison with HLA-identical sibling hematopoietic stem cell transplantation. Bone Marrow Transplant. 2016;51:391–397. doi: 10.1038/bmt.2015.306.
    1. Campana D, Pui CH. Minimal residual disease-guided therapy in childhood acute lymphoblastic leukemia. Blood. 2017;129:1913–1918. doi: 10.1182/blood-2016-12-725804.
    1. Mo XD, et al. IFN-alpha is effective for treatment of minimal residual disease in patients with acute leukemia after allogeneic hematopoietic stem cell transplantation: Results of a registry study. Biol. Blood Marrow Transplant. 2017;23:1303–1310. doi: 10.1016/j.bbmt.2017.04.023.
    1. Wang Y, et al. The consensus on the monitoring, treatment, and prevention of leukemia relapse after allogeneic hematopoietic stem cell transplantation in China. Cancer Lett. 2018;438:63–75. doi: 10.1016/j.canlet.2018.08.030.
    1. Dominietto A, et al. Donor lymphocyte infusions for the treatment of minimal residual disease in acute leukemia. Blood. 2007;109:5063–5064. doi: 10.1182/blood-2007-02-072470.
    1. Yan CH, et al. Risk stratification-directed donor lymphocyte infusion could reduce relapse of standard-risk acute leukemia patients after allogeneic hematopoietic stem cell transplantation. Blood. 2012;119:3256–3262. doi: 10.1182/blood-2011-09-380386.
    1. Orti G, et al. Donor lymphocyte infusions in AML and MDS: Enhancing the graft-versus-leukemia effect. Exp. Hematol. 2017;48:1–11. doi: 10.1016/j.exphem.2016.12.004.
    1. Chen H, et al. Administration of imatinib after allogeneic hematopoietic stem cell transplantation may improve disease-free survival for patients with Philadelphia chromosome-positive acute lymphobla stic leukemia. J. Hematol. Oncol. 2012;5:29. doi: 10.1186/1756-8722-5-29.
    1. Wassmann B, et al. Early molecular response to posttransplantation imatinib determines outcome in MRD+ Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL) Blood. 2005;106:458–463. doi: 10.1182/blood-2004-05-1746.
    1. Cheng Y, et al. Donor-derived CD19-targeted T cell infusion eliminates B cell acute lymphoblastic leukemia minimal residual disease with no response to donor lymphocytes after allogeneic hematopoietic stem cell transplantation. Engineering. 2019;5:150–155. doi: 10.1016/j.eng.2018.12.006.
    1. Ogba N, et al. Chimeric antigen receptor T-cell therapy. J. Natl. Compr. Cancer Netw. 2018;16:1092–1106. doi: 10.6004/jnccn.2018.0073.
    1. Park JH, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 2018;378:449–459. doi: 10.1056/NEJMoa1709919.
    1. Chen H, et al. Management of cytokine release syndrome related to CAR-T cell therapy. Front. Med. 2019;13:610–617. doi: 10.1007/s11684-019-0714-8.
    1. Maude SL, Teachey DT, Porter DL, Grupp SA. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood. 2015;125:4017–4023. doi: 10.1182/blood-2014-12-580068.
    1. Shah NN, Fry TJ. Mechanisms of resistance to CAR T cell therapy. Nat. Rev. Clin. Oncol. 2019;16:372–385.
    1. Anguille S, et al. Interferon-α in acute myeloid leukemia: an old drug revisited. Leukemia. 2011;25:739–748. doi: 10.1038/leu.2010.324.
    1. Lin XJ, et al. Effects of preemptive interferon-alpha monotherapy in acute leukemia patients with relapse tendency after allogeneic hematopoietic stem cell transplantation: A case–control study. Ann. Hematol. 2018;97:2195–2204. doi: 10.1007/s00277-018-3429-z.
    1. Mo X, et al. Interferon alpha: The salvage therapy for patients with unsatisfactory response to minimal residual disease-directed modified donor lymphocyte infusion. Chin. Med. J. (Engl.) 2014;127:2583–2587.
    1. Mo X, et al. Minimal residual disease-directed immunotherapy for high-risk myelodysplastic syndrome after allogeneic hematopoietic stem cell transplantation. Front Med. 2019;13:354–364. doi: 10.1007/s11684-018-0665-5.
    1. Mo X-D, et al. Interferon-α is effective for treatment of minimal residual disease in patients with t(8;21) acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation: results of a prospective registry study. Oncologist. 2018;23:1349–1357. doi: 10.1634/theoncologist.2017-0692.
    1. Mo X-D, et al. Interferon-α: A potentially effective treatment for minimal residual disease in acute leukemia/myelodysplastic syndrome after allogeneic hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 2015;21:1939–1947. doi: 10.1016/j.bbmt.2015.06.014.
    1. Mo X, et al. Interferon-alpha salvage treatment is effective for patients with acute leukemia/myelodysplastic syndrome with unsatisfactory response to minimal residual disease-directed donor lymphocyte infusion after allogeneic hematopoietic stem cell transplantation. Front. Med. 2019;13:238–249. doi: 10.1007/s11684-017-0599-3.
    1. Weiden PL, et al. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N. Engl. J. Med. 1979;300:1068–1073. doi: 10.1056/NEJM197905103001902.
    1. Yeshurun M, et al. The impact of the graft-versus-leukemia effect on survival in acute lymphoblastic leukemia. Blood Adv. 2019;3:670–680. doi: 10.1182/bloodadvances.2018027003.
    1. Durrieu L, et al. Human interferon-alpha increases the cytotoxic effect of CD56(+) cord blood-derived cytokine-induced killer cells on human B-acute lymphoblastic leukemia cell lines. Cytotherapy. 2012;14:1245–1257. doi: 10.3109/14653249.2012.714864.
    1. Lee WH, Liu FH, Lee YL, Huang HM. Interferon-alpha induces the growth inhibition of human T-cell leukaemia line Jurkat through p38alpha and p38beta. J. Biochem. 2010;147:645–650. doi: 10.1093/jb/mvp213.
    1. Manabe A, Yi T, Kumagai M, Campana D. Use of stroma-supported cultures of leukemic cells to assess antileukemic drugs. I. Cytotoxicity of interferon alpha in acute lymphoblastic leukemia. Leukemia. 1993;7:1990–1995.
    1. Visani G, et al. Alpha-interferon improves survival and remission duration in P-190BCR-ABL positive adult acute lymphoblastic leukemia. Leukemia. 2000;14:22–27. doi: 10.1038/sj.leu.2401641.
    1. Visani G, Piccaluga P, Malagola M, Isidori A. Efficacy of dasatinib in conjunction with alpha-interferon for the treatment of imatinib-resistant and dasatinib-resistant Ph+ acute lymphoblastic leukemia. Leukemia. 2009;23:1687–1688. doi: 10.1038/leu.2009.96.
    1. Wassmann B, et al. Efficacy and safety of imatinib mesylate (Glivec) in combination with interferon-alpha (IFN-alpha) in Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ALL) Leukemia. 2003;17:1919–1924. doi: 10.1038/sj.leu.2403093.
    1. Kuang P, et al. Sustaining integrating imatinib and interferon-alpha into maintenance therapy improves survival of patients with Philadelphia positive acute lymphoblastic leukemia ineligible for allogeneic stem cell transplantation. Leuk. Lymphoma. 2016;57:2321–2329. doi: 10.3109/10428194.2016.1144882.
    1. Sumi M, et al. Second cord blood transplantation and interferon-alpha maintenance therapy for relapsed Ph(+) acute lymphoblastic leukemia with the T315I mutation. Leuk. Lymphoma. 2017;58:2005–2007. doi: 10.1080/10428194.2016.1266623.
    1. Nagafuji K, et al. Interferon-alpha treatment of acute lymphoblastic leukemia relapse after unrelated bone marrow transplantation. Int. J. Hematol. 1998;67:63–68. doi: 10.1016/S0925-5710(97)00081-9.
    1. Volodin L, et al. Successful treatment of first and second recurrence of acute lymphoblastic leukemia after related allogeneic bone marrow transplant at unusual sites using single-dose vincristine followed by interferon-alpha2b and granulocyte-macrophage colony-stimulating factor. Leuk. Lymphoma. 2013;54:1107–1109. doi: 10.3109/10428194.2012.734617.
    1. Bruggemann M, Kotrova M. Minimal residual disease in adult ALL: Technical aspects and implications for correct clinical interpretation. Blood Adv. 2017;1:2456–2466. doi: 10.1182/bloodadvances.2017009845.
    1. Modvig S, et al. Minimal residual disease quantification by flow cytometry provides reliable risk stratification in T-cell acute lymphoblastic leukemia. Leukemia. 2019;33:1324–1336. doi: 10.1038/s41375-018-0307-6.
    1. Pemmaraju N, et al. Significance of recurrence of minimal residual disease detected by multi-parameter flow cytometry in patients with acute lymphoblastic leukemia in morphological remission. Am. J. Hematol. 2017;92:279–285. doi: 10.1002/ajh.24629.
    1. Zhao XS, et al. Combined use of WT1 and flow cytometry monitoring can promote sensitivity of predicting relapse after allogeneic HSCT without affecting specificity. Ann. Hematol. 2013;92:1111–1119. doi: 10.1007/s00277-013-1733-1.
    1. Chiusa L, et al. Prognostic value of quantitative analysis of WT1 gene transcripts in adult acute lymphoblastic leukemia. Haematologica. 2006;91:270–271.
    1. Heesch S, et al. Prognostic implications of mutations and expression of the Wilms tumor 1 (WT1) gene in adult acute T-lymphoblastic leukemia. Haematologica. 2010;95:942–949. doi: 10.3324/haematol.2009.016386.
    1. Qin, Y. Z. et al. The prognostic significance of Wilms' tumor gene 1 (WT1) expression at diagnosis in adults with Ph-negative B cell precursor acute lymphoblastic leukemia. Ann. Hematol. (2019).
    1. Zhang B, et al. The relationship between WT1 expression level and prognosis in patients of acute T lymphoblastic leukemia following allogeneic hematopoietic stem cell transplantation. Zhonghua Xue Ye Xue Za Zhi. 2015;36:642–646.
    1. Boublikova L, et al. Wilms' tumor gene 1 (WT1) expression in childhood acute lymphoblastic leukemia: A wide range of WT1 expression levels, its impact on prognosis and minimal residual disease monitoring. Leukemia. 2006;20:254–263. doi: 10.1038/sj.leu.2404047.
    1. Schuurhuis GJ, et al. Minimal/measurable residual disease in AML: A consensus document from the European LeukemiaNet MRD Working Party. Blood. 2018;131:1275–1291. doi: 10.1182/blood-2017-09-801498.
    1. Klingemann HG, et al. Treatment with recombinant interferon (alpha-2b) early after bone marrow transplantation in patients at high risk for relapse [corrected] Blood. 1991;78:3306–3311. doi: 10.1182/blood.V78.12.3306.3306.
    1. Mo X-D, Lv M, Huang X-J. Preventing relapse after haematopoietic stem cell transplantation for acute leukaemia: The role of post-transplantation minimal residual disease (MRD) monitoring and MRD-directed intervention. Br. J. Haematol. 2017;179:184–197. doi: 10.1111/bjh.14778.
    1. Barsan V, Ramakrishna S, Davis KL. Immunotherapy for the treatment of acute lymphoblastic leukemia. Curr. Oncol. Rep. 2020;22:11. doi: 10.1007/s11912-020-0875-2.
    1. Ali S, et al. Blinatumomab for acute lymphoblastic leukemia: The first bispecific T-cell engager antibody to be approved by the EMA for minimal residual disease. Oncologist. 2020;25:e709–e715.
    1. Franquiz MJ, Short NJ. Blinatumomab for the treatment of adult B-cell acute lymphoblastic leukemia: Toward a new era of targeted immunotherapy. Biologics. 2020;14:23–34.
    1. Giebel S, et al. Hematopoietic stem cell transplantation for adults with Philadelphia chromosome-negative acute lymphoblastic leukemia in first remission: A position statement of the European Working Group for Adult Acute Lymphoblastic Leukemia (EWALL) and the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation (EBMT) Bone Marrow Transplant. 2019;54:798–809. doi: 10.1038/s41409-018-0373-4.
    1. Liu Y, et al. Comparison analysis between haplo identical stem cell transplantation and matched sibling donor stem cell transplantation for high-risk acute myeloid leukemia in first complete remission. Sci. China Life Sci. 2019;62:691–697. doi: 10.1007/s11427-018-9361-2.
    1. Tang, F. et al. Comparison of the clinical outcomes of hematologic malignancies after myeloablative haploidentical transplantation with G-CSF/ATG and posttransplant cyclophosphamide: Results from the Chinese Bone Marrow Transplantation Registry Group (CBMTRG). Sci China Life Sci. (2019).
    1. Wang Y, et al. Who is the best donor for a related HLA haplotype-mismatched transplant? Blood. 2014;124:843–850. doi: 10.1182/blood-2014-03-563130.
    1. Xiao-Jun H, et al. Partially matched related donor transplantation can achieve outcomes comparable with unrelated donor transplantation for patients with hematologic malignancies. Clin. Cancer Res. 2009;15:4777–4783. doi: 10.1158/1078-0432.CCR-09-0691.
    1. Wang Y, et al. Haploidentical vs identical-sibling transplant for AML in remission: A multicenter, prospective study. Blood. 2015;125:3956–3962. doi: 10.1182/blood-2015-02-627786.
    1. Mo X-D, et al. The role of collateral related donors in haploidentical hematopoietic stem cell transplantation. Sci. Bull. 2018;63:1376–1382. doi: 10.1016/j.scib.2018.08.008.
    1. Zhao XS, et al. Wilms' tumor gene 1 expression: An independent acute leukemia prognostic indicator following allogeneic hematopoietic SCT. Bone Marrow Transplant. 2012;47:499–507. doi: 10.1038/bmt.2011.121.
    1. Dignan FL, et al. Diagnosis and management of chronic graft-versus-host disease. Br. J. Haematol. 2012;158:46–61. doi: 10.1111/j.1365-2141.2012.09128.x.
    1. Dignan FL, et al. Diagnosis and management of acute graft-versus-host disease. Br. J. Haematol. 2012;158:30–45. doi: 10.1111/j.1365-2141.2012.09129.x.
    1. Liu, S. N. et al. Prognostic factors and long-term follow-up of basiliximab for steroid-refractory acute GVHD: Updated experienced from a large-scale study. Am. J. Hematol. (2020).
    1. Armand P, et al. Validation and refinement of the Disease Risk Index for allogeneic stem cell transplantation. Blood. 2014;123:3664–3671. doi: 10.1182/blood-2014-01-552984.
    1. Filipovich AH, 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;11:945–956. doi: 10.1016/j.bbmt.2005.09.004.
    1. Przepiorka D, et al. Consensus conference on acute GVHD grading. Bone Marrow Transplant. 1995;15:825–828.
    1. Gooley TA, Leisenring W, Crowley J, Storer BE. Estimation of failure probabilities in the presence of competing risks: New representations of old estimators. Stat. Med. 1999;18:695–706. doi: 10.1002/(SICI)1097-0258(19990330)18:6<695::AID-SIM60>;2-O.

Source: PubMed

3
Abonner