Elsevier

Leukemia Research

Volume 61, October 2017, Pages 108-116
Leukemia Research

Research paper
Randomized phase II trial of cytosine arabinoside with and without the CHK1 inhibitor MK-8776 in relapsed and refractory acute myeloid leukemia

https://doi.org/10.1016/j.leukres.2017.09.005Get rights and content

Highlights

  • First randomized trial of HiDAc ± Chk1 inhibition in relapsed/refractory AML.

  • Response rates and overall survival between the two arms were comparable.

  • MK-8776 led to an increase in DNA damage in vivo in circulating leukemic blasts.

  • Timed sequential therapy led to a 44% response rate in the control arm.

Abstract

Purpose

Cytosine arabinoside (AraC) remains the backbone of most treatment regimens for acute myeloid leukemia (AML). Incorporation of AraC into DNA activates checkpoint kinase 1 (Chk1), leading to cell-cycle arrest and diminished AraC cytotoxicity, which can be reversed by the selective Chk1 inhibitor MK-8776. Building on a Phase I trial, we conducted a phase II trial comparing timed sequential AraC with or without MK-8776.

Methods

Patients with relapsed or primary refractory AML were randomized 1:1 to receive either AraC with MK-8776 (Arm A); or AraC alone (Arm B).

Results

32 patients were treated: 14 assigned to Arm A and 18 to Arm B. There were 5 (36%) complete responses (CR/CRi) and 1 (7%) partial response (PR) in Arm A, and 8 (44%) CR/CRis and 1 (6%) PR in Arm B. Median survival did not differ significantly between the two groups (5.9 months in Arm A vs. 4.5 months in Arm B). MK-8776 led to a robust increase in DNA damage in circulating leukemic blasts as measured by increased γ-H2AX (16.9% ± 6.1% prior and 36.4% ± 6.8% at one hour after MK-8776 infusion, p = 0.016).

Conclusion

Response rates and survival were similar between the two groups in spite of evidence that MK-8776 augmented DNA damage in circulating leukemic blasts. Better than expected results in the control arm using timed sequential AraC and truncated patient enrollment may have limited the ability to detect clinical benefit from the combination.

Introduction

Cytosine arabinoside (AraC) is the backbone of both first-line induction and salvage regimens for acute myeloid leukemia (AML) [1]. In newly diagnosed AML, AraC-based regimens lead to complete remission (CR) in up to 80% of patients, yet more than half will ultimately relapse [2]. Outcomes of salvage therapy for relapsed and refractory AML patients are worse, with only 23–32% achieving remission with high-dose AraC (HiDAc) alone, and there is scant evidence that adding a second agent improves outcomes [3], [4], [5], [6]. The low cure rate among newly diagnosed patients and the limited effectiveness of AraC-based salvage regimens suggest that many patients harbor undetectable residual disease with either de novo or acquired AraC resistance. Thus treatments designed to overcome AraC resistance might help to eradicate minimal residual disease (MRD), thereby increasing progression free survival among newly diagnosed patients and improving the rates of remission for relapsed/refractory AML.

AraC exerts its cytotoxic effect on leukemic cells through its incorporation into DNA, which is dependent on the intracellular concentration of and duration of exposure to AraC triphosphate, an active metabolite of AraC [7]. Numerous studies have demonstrated that cellular retention of AraC triphosphate correlates with the likelihood of achieving CR [8], [9], [10]. Consistent with this view, changes that reduce AraC triphosphate formation and retention have been reported to cause AraC resistance [11]. Upon incorporation into cellular DNA, AraC activates ataxia telangiectasia and Rad3-related protein (ATR), and checkpoint kinase 1 (Chk1) [12], [13]. The ATR-Chk1 checkpoint signaling pathway causes cell cycle arrest and diminishes AraC cytotoxicity by stabilizing stalled replication forks, activating DNA repair, and suppressing apoptosis [12], [14], [15], [16], [17], [18]. Indeed, a recent study found that high levels of Chk1 are associated with AraC resistance during induction chemotherapy [19]. Conversely, disruption of Chk1 in vitro abrogates these protective effects and yields increased sensitivity to antimetabolites such as AraC [16], [20], [21], [22].

A number of clinical studies have investigated the safety and efficacy of Chk1 inhibition to overcome AraC resistance in AML. In a phase I study, the combination of AraC and tanespimycin, an inhibitor of heat shock protein 90 (Hsp90) that downregulates Chk1 ex vivo, led to modest Chk1 downregulation in vivo at clinically intolerable tanespimycin doses without any discernible impact on patient outcomes [23]. Similarly, in a trial of AraC with UCN-01, an inhibitor of Chk1 and other kinases, only 1/13 AML patients achieved a CR despite modest decreases in Chk1 activation [24], [25]. More recently, high content screening identified MK-8776 as a potent and selective Chk1 inhibitor that sensitized AML cells to AraC in vitro [13], [26]. Additionally, Chk1 was pulled out as a major hit in an unbiased AML RNAi screen of the human kinome with AraC, validating Chk1 as a high priority target in AML [27]. A phase I study of the AraC/MK-8776 combination in relapsed or refractory acute leukemias established a maximum tolerated dose (2 g/m2 of AraC over 72 h on days 1 and 10 with a 100-mg flat dose of MK-8776 on days 2, 3, 11, and 12) and suggested a possible dose response curve with a response rate of 39% at the highest dose levels [28]. Administration of MK-8776 to patients receiving AraC also led to increased γ  H2AX, an established biomarker of DNA damage [29], [30], [31], [32], in bone marrow blasts from 3 of 5 patients [28]. These observations provided the basis for the current randomized phase II trial of AraC versus AraC plus MK-8776 in relapsed or refractory AML.

The AraC dosing strategy chosen for this study differs from conventional re-induction approaches using HiDAc [3] and is based on the principles of timed sequential therapy (TST). In TST, an initial dose of chemotherapy stimulates humoral factors that increase cell cycling, thereby increasing the incorporation of a subsequent dose of cell-cycle specific agents, such as AraC, into DNA, a key event in cytotoxicity [33]. An in vivo model of TST using consecutive cycles of AraC has shown that the timing of the second cycle of AraC is key to extending survival [34]. Based on these findings, TST-based approaches have been used both as initial induction and salvage AML therapy with promising results [35], [36], [37]. Thus a salvage regimen based on TST was chosen in combination with MK-8776 for the initial phase I trial [3], [28].

Section snippets

Patient eligibility and selection

Between June 2013 and September 2014, patients aged 18–75 years with a pathologically confirmed diagnosis of relapsed or primary refractory AML were enrolled in a multi-institution study with a planned enrollment of 52 patients. Patients were eligible if they received ≤2 prior cytotoxic induction regimens and were >2 weeks beyond previous cytotoxic chemotherapy or radiation. The study was conducted in accordance with the Declaration of Helsinki after approval by the ethics committee of each

Patient characteristics

Thirty-two patients (Arm A: n = 14, Arm B: n = 18) from 4 institutions were randomized, treated, and included in the analysis. An interim analysis for futility after 20 patients showed the predicted probability of rejecting the null hypothesis was 0.27, which did not meet the stopping criteria. The second interim analysis was not performed and accrual was truncated after 32 patients due to the discontinued development of MK-8776.

Clinical demographics and disease biological features of all patients

Discussion

Increasing the cytotoxicity of AraC and improving responses seen with AraC-based salvage regimens for refractory and relapsed AML remains a challenge. Chk1 inhibition has been shown to augment AraC-induced DNA damage and cytotoxicity in AML ex vivo. Accordingly, the ability of the Chk1 inhibitor MK-8776 to improve the response rate or progression-free survival in relapsed/refractory AML was assessed in the present study. Accrual to this randomized study was stopped due to the termination of the

Conflict of interest statement

The authors declare no potential conflicts of interest.

Financial support

Larry Karnitz received support from R01 CA190473 for the correlative studies. The study also received support from UO1 CA70095, UM1 CA186691, and P30 CA006973.

Acknowledgements

We are grateful for the tireless work and dedication of the nursing staff, residents, fellows and faculty on the inpatient units at each center and most of all, the patients and their families for participating in this research. The study was sponsored by the Cancer Therapy Evaluation Program (CTEP) in the Division of Cancer Treatment and Diagnosis at the National Cancer Institute (NCI). Funding was provided through NCI grantsR01 CA190473, UO1 CA70095, UM1 CA186691, and P30 CA006973.

References (51)

  • P.J. Burke et al.

    A rationale for sequential high-dose chemotherapy of leukemia timed to coincide with induced tumor proliferation

    Blood

    (1980)
  • S. Castaigne et al.

    Randomized comparison of double induction and timed-sequential induction to a 3 + 7 induction in adults with AML: long-term analysis of the Acute Leukemia French Association (ALFA) 9000 study

    Blood

    (2004)
  • X. Thomas et al.

    Dose-escalation study of single dose mitoxantrone in combination with timed sequential chemotherapy in patients with refractory or relapsing acute myelogenous leukemia

    Leuk. Res.

    (2000)
  • J.E. Karp et al.

    Phase 1 and pharmacokinetic study of bolus-infusion flavopiridol followed by cytosine arabinoside and mitoxantrone for acute leukemias

    Blood

    (2011)
  • H. Dohner et al.

    Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet

    Blood

    (2010)
  • H. Techer et al.

    Signaling from mus81-Eme2-dependent DNA damage elicited by chk1 deficiency modulates replication fork speed and origin usage

    Cell Rep.

    (2016)
  • S. Faderl et al.

    Results of a phase 1–2 study of clofarabine in combination with cytarabine (ara-C) in relapsed and refractory acute leukemias

    Blood

    (2005)
  • S. Miyawaki et al.

    A randomized comparison of 4 courses of standard-dose multiagent chemotherapy versus 3 courses of high-dose cytarabine alone in postremission therapy for acute myeloid leukemia in adults: the JALSG AML201 Study

    Blood

    (2011)
  • Z. Zeng et al.

    Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML

    Blood

    (2009)
  • J.E. Lancet et al.

    Phase 2 trial of CPX-351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML

    Blood

    (2014)
  • B. Lowenberg et al.

    Cytarabine dose for acute myeloid leukemia

    N. Engl. J. Med.

    (2011)
  • W.R. Vogler et al.

    A phase III trial of high-dose cytosine arabinoside with or without etoposide in relapsed and refractory acute myelogenous leukemia. A Southeastern Cancer Study Group trial

    Leukemia

    (1994)
  • S. Faderl et al.

    Clofarabine plus cytarabine compared with cytarabine alone in older patients with relapsed or refractory acute myelogenous leukemia: results from the CLASSIC I Trial

    J. Clin. Oncol.

    (2012)
  • Y.M. Rustum et al.

    Correlation between leukemic cell retention of 1-beta-D-arabinofuranosylcytosine 5'-triphosphate and response to therapy

    Cancer Res.

    (1979)
  • E. Cros et al.

    Problems related to resistance to cytarabine in acute myeloid leukemia

    Leuk. Lymphoma

    (2004)
  • Cited by (0)

    View full text