Skip to main content

Maintenance therapy in acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation

Abstract

Relapse remains the main cause of treatment failure in acute myeloid leukemia (AML) undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT). Emerging evidence has demonstrated that AML patients might benefit from maintenance therapy post-transplantation, especially for high-risk AML patients. In this mini-review, we will summarize targeted drugs, such as hypomethylating agents, FLT3 inhibitors and isocitrate dehydrogenase inhibitors, as maintenance therapy post-transplantation in AML patients undergoing allo-HSCT.

Background

Acute myeloid leukemia (AML) is a heterogeneous group of clonal diseases. Conventional chemotherapy can result in the complete remission (CR) rate of approximately 70–80% in AML patients [1, 2]. Leukemia relapse remains the main cause of treatment failure, including the patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) [3]. Therefore, there is an urgent need to identify an effective and safe approach to improve post-remission survival. Conventional chemotherapy as maintenance therapy might have no benefit to most AML patients, as the efficacy is often offset by treatment-related mortality [4, 5]. Recently, novel targeted drugs have been used as maintenance therapy in patients with AML, including maintenance after allo-HSCT [6,7,8,9]. Some studies have shown that AML patients might benefit from maintenance with targeted drugs post-transplantation [6,7,8,9]. In this study, we do a mini-review about targeted drugs as maintenance post-transplantation in AML patients undergoing allo-HSCT.

The broad definition of maintenance therapy includes preemptive therapy based on measurable residual disease (MRD) and prophylactic therapy not based on MRD. The narrow definition of maintenance therapy refers only to prophylactic therapy. It remains under discussion whether preemptive or prophylactic therapy is superior in reducing relapse and improving survival for AML patients [10, 11]. Here, we mainly focus on prophylactic therapy post-transplantation.

Hypomethylating agents

Hypomethylating agents such as azacitidine and decitabine have shown favorable efficacy and tolerability as induction therapy in AML patients, especially elderly patients unable to tolerate intensive chemotherapy [12, 13]. Some retrospective and small-sample prospective studies demonstrated that maintenance therapy with hypomethylating agents post-transplantation was safe and could reduce relapse, thereby prolonging the survival of AML patients, especially high-risk AML patients [14,15,16,17]. On the contrary, other studies suggested that AML patients might not benefit from maintenance with hypomethylating agents after allo-HSCT [18, 19]. Recently, a prospective phase II trial revealed that preemptive therapy with azacitidine could prevent or delay hematological relapse in patients with high-risk AML or myelodysplastic syndrome (MDS) who achieved CR after chemotherapy or allo-HSCT [20]. Another prospective phase I/II study revealed that CC-486, an oral formulation of azacitidine, was well tolerated as maintenance post-transplantation in patients with AML or MDS undergoing allo-HSCT, with the 1-year relapse rate of 21% [21]. It was a pity that both studies were single-armed. A phase II randomized controlled trial (RCT) from China has demonstrated that minimal-dose decitabine maintenance combined with recombinant human granulocyte colony-stimulating factor after allo-HSCT could reduce relapse for high-risk AML patients undergoing allo-HSCT, with the 2-year relapse rate of 15.0% and 38.3% in the intervention and non-intervention groups [22]. Nowadays, there are few reports comparing the effect of preemptive or prophylactic use with hypomethylating agents post-transplantation on the outcomes of AML patients undergoing allo-HSCT. Key studies of hypomethylating agents as maintenance strategy in AML patients after allo-HSCT are summarized in Table 1.

Table 1 Key studies of hypomethylating agents as maintenance therapy in AML patients after allo-HSCT

FLT3 inhibitors

FLT3 internal tandem duplication (FLT3-ITD) mutations occur in approximately 25% of adults with AML. In contrast to AML with FLT3 wild-type, AML patients with FLT3-ITD mutations have shorter remissions and higher relapse rates [23, 24]. Allo-HSCT could improve the survival of patients with FLT3-ITD AML, but leukemia relapse remains high [23,24,25]. Currently, FLT3 inhibitors, including sorafenib, midostaurin, gilteritinib, quizartinib and crenolanib, have been applied to clinical practice [23, 24]. They have been explored in various settings for patients with FLT3-ITD AML, including induction, post-remission maintenance pre- and post-transplantation and salvage therapy for refractory relapsed patients [23,24,25,26,27,28,29,30,31,32,33]. Besides, FLT3 inhibitors have been also explored in the treatment of AML patients without FLT3 mutations [26, 34]. Growing evidence has suggested that patients with FLT3-ITD AML undergoing allo-HSCT might benefit from maintenance with FLT3 inhibitors post-transplantation [7,8,9, 23].

Sorafenib, a multi-kinase inhibitor, has shown promising efficacy in the treatment of FLT3-ITD AML, including maintenance post-transplantation [23,24,25,26,27,28,29,30,31,32,33]. Recently, two back-to-back RCTs including SORMAIN and our own have demonstrated that sorafenib maintenance can prevent relapse and improve survival for patients with FLT3-ITD AML following allo-​HSCT [35, 36]. SORMAIN, which was terminated early because of slow accrual, demonstrated that sorafenib maintenance could reduce the risk of relapse and death after allo-HSCT for patients with FLT3-ITD AML, with the 2-year overall survival (OS) of 90.5% and 66.2% in the sorafenib and placebo groups [36]. Our phase III RCT showed that sorafenib maintenance revealed a significant advantage over non-maintenance in relapse and OS, with the 2-year relapse rate and OS of 11.9% and 82.1% in the sorafenib group compared with 31.6% and 68.0% in the non-maintenance group [35]. Apart from the direct antileukemic effect of sorafenib, several studies including our own suggested the synergism between sorafenib and alloreactive donor T cells in promoting graft-versus-leukemia activity [35, 37, 38]. Therefore, the researchers of SORMAIN and we both proposed the prospective trials of sorafenib maintenance after allo­HSCT in AML patients without FLT3­ITD mutations [39].

Some retrospective and single-arm exploratory studies revealed that post-transplantation maintenance of other FLT3 inhibitors could also reduce relapse and improve survival for patients with FLT3-ITD AML [40, 41]. The preliminary result of a phase II RCT about midostaurin maintenance post-transplantation revealed that midostaurin maintenance could reduce the risk of relapse, with the 18-month leukemia-free survival (LFS) of 76% and 89% in the non-maintenance and midostaurin arms, respectively [42]. An ongoing phase III RCT is evaluating gilteritinib maintenance following allo-HSCT in patients with FLT3-ITD AML (NCT02997202) [43]. Further research is needed to ascertain which FLT3 inhibitors are most effective for post-transplantation maintenance in patients with FLT3-ITD AML. Key studies of FLT3 inhibitors as maintenance strategy in AML patients after allo-HSCT are summarized in Table 2.

Table 2 Key studies of FLT3 inhibitors as maintenance therapy in AML patients after allo-HSCT

Isocitrate dehydrogenase (IDH) inhibitors

Approximately 20% of AML genomes harbor mutations in one of two isoforms of IDH (IDH1 or IDH2). IDH mutation reduces α-ketoglutarate to 2-hydroxyglutarate, leading to histone hypermethylation and a block in myeloid differentiation [44]. AML patients with IDH mutation have a poor response to traditional chemotherapy and a higher relapse rate. Recently, ivosidenib and enasidenib, oral inhibitors of mutant IDH1 and IDH2, have shown good clinical response in relapsed/refractory or newly diagnosed IDH-mutated AML patients [45,46,47]. A phase I ongoing trial is evaluating the safety of enasidenib maintenance for IDH2-mutated myeloid neoplasms following allo-HSCT (NCT03515512).

Venetoclax

Venetoclax, a selective small-molecular inhibitor of B-cell lymphoma-2 (Bcl-2), has been demonstrated that single-agent or combined with other agents are effective and tolerable to AML patients including relapsed or refractory AML patients, especially in elderly patients unfit for intensive chemotherapy, with overall response rate of 19–72% [48,49,50]. A phase I study demonstrated the feasibility of venetoclax combined with chemotherapy followed by venetoclax maintenance in fit elderly AML patients, with the median OS of 11.2 months [49]. Recently, Kent et al. reported that venetoclax was safe and tolerable as post-transplant maintenance for AML patients at high risk of relapse, with the 6-month LFS of 87% [51]. Currently, two trials are under way to evaluate venetoclax combined with azacitidine as maintenance for AML patients undergoing allo-HSCT (NCT04161885, NCT04128501).

Histone deacetylase and hedgehog inhibitors

Histone deacetylase inhibitors (HDACi) are epigenetic modifiers that are shown to induce cell-cycle arrest, cell differentiation and apoptosis of AML cells. Panobinostat is an oral HDACi that has been reported to have moderate antileukemia activity in advanced AML and MDS patients [52]. A phase I/II study showed that panobinostat maintenance post-transplantation revealed favorable survival outcomes for high-risk MDS/AML patients compared with reports from similar patient groups [53].

The Hedgehog signaling pathway plays a critical role in embryonic development and aberrant Hedgehog signaling are deemed to contribute to the survival and expansion of leukemia stem cells [54]. Glasdegib, a Hedgehog pathway inhibitor, combined with low-dose cytarabine showed a favorable benefit-risk profile for AML patients unsuitable for intensive chemotherapy [55, 56]. A phase II study about glasdegib as maintenance for high-risk AML patients undergoing allo-HSCT is completed (NCT01841333), and the results have not been released.

Maintenance duration

To date, the duration of maintenance of targeted drugs post-transplantation is not firmly established. In some retrospective and prospective studies, targeted drugs are usually maintained for 1–2 years after allo-HSCT [7, 8, 23]. Currently, there is a lack of RCTs on different maintenance duration of targeted drugs post-transplantation. In our phase III RCT, sorafenib was administered at 30–60-day post-transplantation and continued until day 180, with the 2-year LFS and OS of 78.9% and 82.1% [35]. Sorafenib was administered for 24 months in SORMAIN, with the 2-year LFS and OS of 85.0% and 90.5% [36]. Although our duration of sorafenib maintenance was favorable in terms of tolerance, patient’s quality of life and cost-effectiveness, prolonging sorafenib maintenance for 1 year or more post-transplantation might yield greater benefits for these patients. Besides, receptor- and nonreceptor-related mutations, epigenetic changes and signaling pathway alterations might contribute to resistance to FLT3 inhibitors [57]. Whether long-term exposure to sorafenib induces secondary gene mutations and drug resistance remains unclear. Our results showed that only one patient with sorafenib maintenance acquired an FLT3 tyrosine kinase domain mutation at relapse, and the CR rate and OS after salvage therapy were similar between two groups, suggesting that sorafenib maintenance for six-month post-transplantation did not increase the risk of mutation and resistance [35].

Immunotherapies

Traditional immunotherapies such as donor lymphocyte infusion, interleukin-2 and interferon-α might play a role in preventing relapse for AML patients after allo-HSCT [11, 58]. However, their timing, dosing and co-administration with other agents require further study. Novel immunotherapeutic strategies including chimeric antigen receptor (CAR) T-cell therapy, antibody-directed therapy, immune checkpoint inhibitors and vaccines are under investigation [8, 59]. CAR T-cell therapy has yielded unprecedented efficacy in B cell malignancies, but there is limited data on its use in AML [59]. One of the major challenges in adopting CAR T-cell therapy in AML is the lack of an AML-specific antigen. Multiple targets for directed CAR T-cell therapy in AML include CD33, CD123, folate receptor β, NKG2D and Lewis Y [59]. Monoclonal antibodies such as anti-CD33, anti-CD123 and anti-CD45, bispecific antibodies and immune checkpoint inhibitors targeting programmed cell death protein-1 (PD-1) and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) are currently in early clinical trials [59]. The main categories of vaccines currently tested in AML are peptide vaccines and dendritic cell-based vaccines [59]. Wilms tumor-1 (WT-1) peptide vaccine was reported to be safe and potential as maintenance therapy in AML patients after allo-HSCT [60].

Conclusion

Over the past decade, some novel targeted drugs have been explored for post-transplant maintenance for AML patients, and have shown promising results. Two back-to-back RCTs both reveal that sorafenib maintenance after allo­HSCT prevents relapse in patients with FLT3­ITD AML, resulting in an OS benefit. However, most of the studies about maintenance therapy in AML patients after allo-HSCT are retrospective and non-randomized. Currently, there is no consensus on the optimal duration of targeted drugs to prevent relapse, and it remains unclear whether long-term exposure to targeted drugs induces drug resistance. And which specific subpopulations of AML patients will benefit post-transplant maintenance most remains to be further investigation. Therefore, well-designed, fully powered, prospective trials are needed to address the unsettled questions to further improve the outcomes post-transplantation. Except for targeted drugs, some immunotherapies including CAR T-cell therapy, antibody-directed therapy, immune checkpoint inhibitors and vaccines have also attracted board attention.

Availability of data and materials

Not applicable.

Abbreviations

AML:

Acute myeloid leukemia

CR:

Complete remission

allo-HSCT:

Allogeneic hematopoietic stem cell transplantation

MRD:

Measurable residual disease

MDS:

Myelodysplastic syndrome

RCT:

Randomized controlled trial

FLT3-ITD:

FLT3 internal tandem duplication

OS:

Overall survival

LFS:

Leukemia-free survival

IDH:

Isocitrate dehydrogenase

Bcl-2:

B-cell lymphoma-2

HDACi:

Histone deacetylase inhibitor

CAR:

Chimeric antigen receptor

PD-1:

Programmed cell death protein-1

CTLA-4:

Cytotoxic T lymphocyte-associated antigen-4

WT-1:

Wilms tumor-1

References

  1. Teuffel O, Leibundgut K, Lehrnbecher T, Alonzo TA, Beyene J, Sung L. Anthracyclines during induction therapy in acute myeloid leukaemia: a systematic review and meta-analysis. Br J Haematol. 2013;161(2):192–203.

    Article  CAS  PubMed  Google Scholar 

  2. Burnett AK, Russell NH, Hills RK, et al. A randomized comparison of daunorubicin 90 mg/m2 vs 60 mg/m2 in AML induction: results from the UK NCRI AML17 trial in 1206 patients. Blood. 2015;125(25):3878–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Piemontese S, Boumendil A, Labopin M, et al. Leukemia relapse following unmanipulated haploidentical transplantation: a risk factor analysis on behalf of the ALWP of the EBMT. J Hematol Oncol. 2019;12(1):68.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Löwenberg B, Suciu S, Archimbaud E, et al. Mitoxantrone versus daunorubicin in induction-consolidation chemotherapy—the value of low-dose cytarabine for maintenance of remission, and an assessment of prognostic factors in acute myeloid leukemia in the elderly: final report. European Organization for the Research and Treatment of Cancer and the Dutch-Belgian Hemato-Oncology Cooperative Hovon Group. J Clin Oncol. 1998;16(3):872–81.

    Article  PubMed  Google Scholar 

  5. Ferrero D, Crisà E, Marmont F, et al. Survival improvement of poor-prognosis AML/MDS patients by maintenance treatment with low-dose chemotherapy and differentiating agents. Ann Hematol. 2014;93(8):1391–400.

    Article  CAS  PubMed  Google Scholar 

  6. Saygin C, Carraway HE. Emerging therapies for acute myeloid leukemia. J Hematol Oncol. 2017;10(1):93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Molica M, Breccia M, Foa R, Jabbour E, Kadia TM. Maintenance therapy in AML: The past, the present and the future. Am J Hematol. 2019;94(11):1254–65.

    Article  PubMed  Google Scholar 

  8. Lee CJ, Savani BN, Mohty M, et al. Post-remission strategies for the prevention of relapse following allogeneic hematopoietic cell transplantation for high-risk acute myeloid leukemia: expert review from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Bone Marrow Transplant. 2019;54(4):519–30.

    Article  PubMed  Google Scholar 

  9. Lai C, Doucette K, Norsworthy K. Recent drug approvals for acute myeloid leukemia. J Hematol Oncol. 2019;12(1):100.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Schroeder T, Rautenberg C, Haas R, Germing U, Kobbe G. Hypomethylating agents for treatment and prevention of relapse after allogeneic blood stem cell transplantation. Int J Hematol. 2018;107(2):138–50.

    Article  CAS  PubMed  Google Scholar 

  11. Wang Y, Liu QF, Wu DP, et al. Impact of prophylactic/preemptive donor lymphocyte infusion and intensified conditioning for relapsed/refractory leukemia: a real-world study. Sci China Life Sci. 2020;63(10):1552–64.

    Article  CAS  PubMed  Google Scholar 

  12. Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol. 2012;30(21):2670–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Dombret H, Seymour JF, Butrym A, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with > 30% blasts. Blood. 2015;126(3):291–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Jabbour E, Giralt S, Kantarjian H, et al. Low-dose azacitidine after allogeneic stem cell transplantation for acute leukemia. Cancer. 2009;115(9):1899–905.

    Article  CAS  PubMed  Google Scholar 

  15. de Lima M, Giralt S, Thall PF, et al. Maintenance therapy with low-dose azacitidine after allogeneic hematopoietic stem cell transplantation for recurrent acute myelogenous leukemia or myelodysplastic syndrome: a dose and schedule finding study. Cancer. 2010;116(23):5420–31.

    Article  PubMed  CAS  Google Scholar 

  16. Pusic I, Choi J, Fiala MA, et al. Maintenance therapy with decitabine after allogeneic stem cell transplantation for acute myelogenous leukemia and myelodysplastic syndrome. Biol Blood Marrow Transplant. 2015;21(10):1761–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ma Y, Qu C, Dai H, et al. Maintenance therapy with decitabine after allogeneic hematopoietic stem cell transplantation to prevent relapse of high-risk acute myeloid leukemia. Bone Marrow Transplant. 2020;55(6):1206–8.

    Article  PubMed  Google Scholar 

  18. Maples KT, Sabo RT, McCarty JM, Toor AA, Hawks KG. Maintenance azacitidine after myeloablative allogeneic hematopoietic cell transplantation for myeloid malignancies. Leuk Lymphoma. 2018;59(12):2836–41.

    Article  CAS  PubMed  Google Scholar 

  19. Oshikawa G, Kakihana K, Saito M, et al. Post-transplant maintenance therapy with azacitidine and gemtuzumab ozogamicin for high-risk acute myeloid leukaemia. Br J Haematol. 2015;169(5):756–9.

    Article  CAS  PubMed  Google Scholar 

  20. Platzbecker U, Middeke JM, Sockel K, et al. Measurable residual disease-guided treatment with azacitidine to prevent haematological relapse in patients with myelodysplastic syndrome and acute myeloid leukaemia (RELAZA2): an open-label, multicentre, phase 2 trial. Lancet Oncol. 2018;19(12):1668–79.

    Article  CAS  PubMed  Google Scholar 

  21. de Lima M, Oran B, Champlin RE, et al. CC-486 Maintenance after stem cell transplantation in patients with acute myeloid leukemia or myelodysplastic syndromes. Biol Blood Marrow Transplant. 2018;24(10):2017–24.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  22. Gao L, Zhang Y, Wang S, et al. Effect of rhG-CSF combined with decitabine prophylaxis on relapse of patients with high-risk MRD-negative AML after HSCT: an open-label, multicenter. Randomized Controlled Trial J Clin Oncol. 2020. https://doi.org/10.1200/JCO.19.03277.

    Article  PubMed  Google Scholar 

  23. Bazarbachi A, Bug G, Baron F, et al. Clinical practice recommendation on hematopoietic stem cell transplantation for acute myeloid leukemia patients with FLT3-internal tandem duplication: a position statement from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Haematologica. 2020;105(6):1507–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Antar AI, Otrock ZK, Jabbour E, Mohty M, Bazarbachi A. FLT3 inhibitors in acute myeloid leukemia: ten frequently asked questions. Leukemia. 2020;34(3):682–96.

    Article  PubMed  Google Scholar 

  25. Xuan L, Wang Y, Huang F, et al. Effect of sorafenib on the outcomes of patients with FLT3-ITD acute myeloid leukemia undergoing allogeneic hematopoietic stem cell transplantation. Cancer. 2018;124(9):1954–63.

    Article  CAS  PubMed  Google Scholar 

  26. Rollig C, Serve H, Huttmann A, et al. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial. Lancet Oncol. 2015;16(16):1691–9.

    Article  PubMed  CAS  Google Scholar 

  27. Xuan L, Wang Y, Chen J, et al. Sorafenib therapy is associated with improved outcomes for FMS-like tyrosine kinase 3 internal tandem duplication acute myeloid leukemia relapsing after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2019;25(8):1674–81.

    Article  CAS  PubMed  Google Scholar 

  28. Yilmaz M, Alfayez M, DiNardo CD, et al. Outcomes with sequential FLT3-inhibitor-based therapies in patients with AML. J Hematol Oncol. 2020;13(1):132.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Chen YB, Li S, Lane AA, et al. Phase I trial of maintenance sorafenib after allogeneic hematopoietic stem cell transplantation for fms-like tyrosine kinase 3 internal tandem duplication acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20(12):2042–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Brunner AM, Li S, Fathi AT, et al. Haematopoietic cell transplantation with and without sorafenib maintenance for patients with FLT3-ITD acute myeloid leukaemia in first complete remission. Br J Haematol. 2016;175(3):496–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Battipaglia G, Ruggeri A, Massoud R, et al. Efficacy and feasibility of sorafenib as a maintenance agent after allogeneic hematopoietic stem cell transplantation for Fms-like tyrosine kinase 3-mutated acute myeloid leukemia. Cancer. 2017;123(15):2867–74.

    Article  CAS  PubMed  Google Scholar 

  32. Battipaglia G, Massoud R, Ahmed SO, et al. Efficacy and feasibility of sorafenib as a maintenance agent after allogeneic hematopoietic stem cell transplantation for FMS-like tyrosine kinase 3 mutated acute myeloid leukemia: an update. Clin Lymphoma Myeloma Leuk. 2019;19(8):506–8.

    Article  PubMed  Google Scholar 

  33. Bazarbachi A, Labopin M, Battipaglia G, et al. Allogeneic stem cell transplantation for FLT3-mutated acute myeloid leukemia: in vivo T-cell depletion and posttransplant sorafenib maintenance improve survival. a retrospective acute leukemia working party-European Society for Blood and Marrow Transplant Study. Clin Hematol Int. 2019;1(1):58–74.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Serve H, Krug U, Wagner R, et al. Sorafenib in combination with intensive chemotherapy in elderly patients with acute myeloid leukemia: results from a randomized, placebo-controlled trial. J Clin Oncol. 2013;31(25):3110–8.

    Article  CAS  PubMed  Google Scholar 

  35. Xuan L, Wang Y, Huang F, et al. Sorafenib maintenance in patients with FLT3-ITD acute myeloid leukaemia undergoing allogeneic haematopoietic stem-cell transplantation: an open-label, multicentre, randomised phase 3 trial. Lancet Oncol. 2020;21(9):1201–12.

    Article  CAS  PubMed  Google Scholar 

  36. Burchert A, Bug G, Fritz LV, et al. Sorafenib maintenance after allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia with FLT3-internal tandem duplication mutation (SORMAIN). J Clin Oncol. 2020;38(26):2993–3002.

    Article  PubMed  Google Scholar 

  37. Mathew NR, Baumgartner F, Braun L, et al. Sorafenib promotes graft-versus-leukemia activity in mice and humans through IL-15 production in FLT3-ITD-mutant leukemia cells. Nat Med. 2018;24(3):282–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Metzelder SK, Schroeder T, Finck A, et al. High activity of sorafenib in FLT3-ITD-positive acute myeloid leukemia synergizes with allo-immune effects to induce sustained responses. Leukemia. 2012;26(11):2353–9.

    Article  CAS  PubMed  Google Scholar 

  39. Romero D. Sorafenib prevents AML relapse after allo-HSCT. Nat Rev Clin Oncol. 2020;17(10):591.

    Article  PubMed  Google Scholar 

  40. Sandmaier BM, Khaled SK, Oran B, Gammon G, Trone D, Frankfurt O. Results of a phase 1 study of quizartinib (AC220) as maintenance therapy in subjects with acute myeloid leukemia in remission following allogeneic hematopoietic cell transplantation. Blood. 2014;124(21):428.

    Article  Google Scholar 

  41. Ganguly S, Cortes JE, Krämer A, et al. Clinical outcomes and characteristics of patients (pts) with FLT3-internal tandem duplication (FLT3-ITD)-mutated relapsed/refractory (R/R) acute myeloid leukemia (AML) undergoing hematopoietic stem cell transplant (HSCT) after quizartinib (Q) or salvage chemotherapy (SC) in the Quantum-R trial. Blood. 2019;134(Supplement_1):736.

    Article  Google Scholar 

  42. Maziarz RT, Patnaik MM, Scott BL, et al. Radius: a phase 2 randomized trial investigating standard of care ± midostaurin after allogeneic stem cell transplant in FLT3-ITD-mutated AML. Blood. 2018;132(Supplement_1):662.

    Article  Google Scholar 

  43. Levis MJ, Hamadani M, Logan BR, et al. BMT CTN protocol 1506: a phase 3 trial of gilteritinib as maintenance therapy after allogeneic hematopoietic stem cell transplantation in patients with FLT3-ITD+AML. Blood. 2019;134(Supplement_1):4602.

    Article  Google Scholar 

  44. Ward PS, Patel J, Wise DR, et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell. 2010;17(3):225–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. DiNardo CD, Stein EM, de Botton S, et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med. 2018;378(25):2386–98.

    Article  CAS  PubMed  Google Scholar 

  46. Yen K, Travins J, Wang F, et al. AG-221, a first-in-class therapy targeting acute myeloid leukemia harboring oncogenic IDH2 mutations. Cancer Discov. 2017;7(5):478–93.

    Article  CAS  PubMed  Google Scholar 

  47. Stein EM, DiNardo CD, Mims AS, et al. Ivosidenib or enasidenib combined with standard induction chemotherapy is well tolerated and active in patients with newly diagnosed AML with an IDH1 or IDH2 mutation: initial results from a phase 1 trial. Blood. 2017;130(Supplement 1):726.

    Google Scholar 

  48. Konopleva M, Pollyea DA, Potluri J, et al. Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discov. 2016;6(10):1106–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chua CC, Roberts AW, Reynolds J, et al. Chemotherapy and venetoclax in elderly acute myeloid leukemia trial (CAVEAT): a phase Ib dose-escalation study of venetoclax combined with modified intensive chemotherapy. J Clin Oncol. 2020;38(30):3506–17.

    Article  CAS  PubMed  Google Scholar 

  50. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383(7):617–29.

    Article  CAS  PubMed  Google Scholar 

  51. Kent A, Pollyea DA, Winters A, Jordan CT, Smith C, Gutman JA. Venetoclax is safe and tolerable as post-transplant maintenance therapy for AML patients at high risk for relapse. Blood. 2020;136(Supplement_1):11–2.

    Article  Google Scholar 

  52. DeAngelo DJ, Spencer A, Bhalla KN, et al. Phase Ia/II, two-arm, open-label, dose-escalation study of oral panobinostat administered via two dosing schedules in patients with advanced hematologic malignancies. Leukemia. 2013;27(8):1628–36.

    Article  CAS  PubMed  Google Scholar 

  53. Bug G, Burchert A, Wagner EM, et al. Phase I/II study of the deacetylase inhibitor panobinostat after allogeneic stem cell transplantation in patients with high-risk MDS or AML (PANOBEST trial). Leukemia. 2017;31(11):2523–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wellbrock J, Latuske E, Köhler J, et al. Expression of hedgehog pathway mediator GLI represents a negative prognostic marker in human acute myeloid leukemia and its inhibition exerts antileukemic effects. Clin Cancer Res. 2015;21(10):2388–98.

    Article  CAS  PubMed  Google Scholar 

  55. Cortes JE, Heidel FH, Hellmann A, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia. 2019;33(2):379–89.

    Article  CAS  PubMed  Google Scholar 

  56. Cortes JE, Heidel FH, Fiedler W, et al. Survival outcomes and clinical benefit in patients with acute myeloid leukemia treated with glasdegib and low-dose cytarabine according to response to therapy. J Hematol Oncol. 2020;13(1):92.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Gebru MT, Wang HG. Therapeutic targeting of FLT3 and associated drug resistance in acute myeloid leukemia. J Hematol Oncol. 2020;13(1):155.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Yan CH, Liu DH, Liu KY, 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(14):3256–62.

    Article  CAS  PubMed  Google Scholar 

  59. Liu Y, Bewersdorf JP, Stahl M, Zeidan AM. Immunotherapy in acute myeloid leukemia and myelodysplastic syndromes: The dawn of a new era? Blood Rev. 2019;34:67–83.

    Article  CAS  PubMed  Google Scholar 

  60. Maeda T, Hosen N, Fukushima K, et al. Maintenance of complete remission after allogeneic stem cell transplantation in leukemia patients treated with Wilms tumor 1 peptide vaccine. Blood Cancer J. 2013;3(8):e130.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by National Natural Science Foundation of China (Nos. 81770190, 81970161), National Key Research and Development Programme of China (Nos. 2017YFA105500, 2017YFA105504), Natural Science Foundation of Guangdong Province (No. 2019A1515011924), Project of the Zhujiang Science & Technology Star of Guangzhou City (No. 201806010029).

Author information

Authors and Affiliations

Authors

Contributions

QFL designed, directed, and revised the manuscript. LX drafted the manuscript. Both authors read and approved the final manuscript.

Corresponding author

Correspondence to Qifa Liu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xuan, L., Liu, Q. Maintenance therapy in acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. J Hematol Oncol 14, 4 (2021). https://doi.org/10.1186/s13045-020-01017-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13045-020-01017-7

Keywords