PONATINIB-AML: Ponatinib for FLT3-ITD Acute Myelogenous Leukemia

Sponsor
Versailles Hospital (Other)
Overall Status
Active, not recruiting
CT.gov ID
NCT02428543
Collaborator
(none)
49
37
1
124
1.3
0

Study Details

Study Description

Brief Summary

This project is part of a joint ALFA and GOELAM strategy aiming to improve the survival of patients with newly diagnosed Acute Myeloid Leukemia (AML) aged 18-70 years. The basis of this strategy is to evaluate intensified conventional chemotherapy and targeted drugs in selected disease-risk subgroups of adult patients with non promyelocytic AML. Participation will be proposed to almost all adult patients in France aged 18-70 years and diagnosed with AML.

FLT3 genetic alterations include FLT3 somatic point mutations within the second tyrosine kinase domain and internal duplications of the juxta-membrane domain. This alteration is refered to as FLT3-ITD. The FLT3-ITD mutation is found in around 30% of patients with cytogenetically normal AML. Patients with the FLT3-ITD genotype have been reported to have a poor outcome when treated with conventional chemotherapy with an estimated 4-year relapse-free survival of 25% (Schlenk et al. N Engl J Med 2008). More recently, the prognostic relevance of FLT3-ITD has been studied in the context of integrated genetic profiling. This confirmed the genetic complexity of AML and also that FLT3-ITD was associated with reduced overall survival in intermediate-risk AML. A multivariate analysis of several genetic alterations revealed that FLT3-ITD was the primary predictor of patient outcome. FLT3-ITD mutations were classified in 3 categories: 1) FLT3-ITD with +8, TET2, DNMT3A or MLL-PTD mutations (3-year OS 14.5%); 2) FLT3-ITD with wild type CEBPA, TET2, DNMT3 and MLL-PTD (3-year OS 35.2%) and 3) FLT3-ITD with CEBPA mutations (3-year OS 42%) (Patel JP et al. N Engl J Med 2012). However, FLT3-ITD was not a predictor of response to induction therapy, allowing the introduction of targeted therapies after the induction course.

Several FLT3 inhibitors have been evaluated or are currently being tested in the setting of relapsing AML. In most trials to date, patients were only eligible if the FLT3-ITD mutation was present. Disappointing results were reported with the first generation of FLT3 inhibitors, including lestaurtinib (CEP-701), midostaurin (PKC-412) and sorafenib. Second generation FLT3 inhibitors such as quizartinib (AC220) are currently under investigation with promising results. However, the hematologic toxicity of AC220 will likely present a major limitation in evaluating AC220 combined with standard or high-dose chemotherapy.

Ponatinib (AP24534) is a third generation tyrosine kinase inhibitor targeting the BCR-ABL tyrosine kinase domain. Ponatinib was rationally designed with an extensive network of optimized molecular contacts and triple bonds to accommodate the T315I mutation, a major cause of resistance to tyrosine kinase inhibitors in chronic and advanced phase chronic myelogenous leukemia (CML). Ponatinib also inhibits SRC (IC50: 5.4 nM) and members of the VEGFR, FGFR, and PDGFR families of receptor tyrosine kinases (O'Hare T, Cancer Cell 2009). Despite low activity against FLT3 based on the IC50 value (FLT3 IC50: 12.6 nM compared to BCR IC50: 0.37 nM), ponatinib has recently been reported to have significant cellular activity against the MV4-11 cell line which harbors an FLT3-ITD activating mutation. Ponatinib-induced apoptosis was maximal at 10 nM in vitro and a single dose of 5 and 10 mg/kg had a strong inhibitory effect in vivo in mice bearing MV4-11 xenografts. Primary blast cells from 4 FLT3-ITD AML patients were also tested and ponatinib reduced their viability (IC50: 4 nM) whereas no activity was shown on FLT3-ITD-negative blast cells (Gozgit JM et al. Mol Cancer Ther 2011).

Preliminary data from the phase I clinical trial showed that 15 mg ponatinib was associated with a Cmax of 51.1 nM. Cmax was increased to 111 nM and 149 nM in the 30 mg and 45 mg cohorts respectively. The trough concentrations were 55.3 nM and 61.9 nM for the 30 mg and 45 mg doses respectively (Ariad clinical investigator's brochure, version 3). Results from the ongoing phase II trial in CML patients suggest that the hematological toxicity profile of ponatinib is comparable with that of nilotinib or dasatinib, both of which have been successfully combined with conventional chemotherapy.

Investigators thus aim to combine ponatinib with cytarabine in FLT3-ITD AML patients in first complete remission.

Condition or Disease Intervention/Treatment Phase
  • Drug: Ponatinib and Cytarabine
Phase 1/Phase 2

Study Design

Study Type:
Interventional
Actual Enrollment :
49 participants
Allocation:
N/A
Intervention Model:
Single Group Assignment
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
A Phase I - II Study to Assess Safety and Efficacy of the Combination of Ponatinib With High or Intermediate-Dose Cytarabine as Consolidation Therapy for Patients With Intermediate-Risk Cytogenetic FLT3-ITD AML iIn First Complete Remission
Actual Study Start Date :
Jul 1, 2013
Anticipated Primary Completion Date :
Nov 1, 2020
Anticipated Study Completion Date :
Nov 1, 2023

Arms and Interventions

Arm Intervention/Treatment
Experimental: Ponatinib arm

dose-escalation Arm _ 15, 30, 45mg Ponatinib per day. Each cohort will consist of 3 evaluable patients

Drug: Ponatinib and Cytarabine
Prospective, non-randomized, open-label, multicenter, dose-escalation phase I-II trial; an adaptive Bayesian logistic regression dose-escalation model incorporating escalation with overdose control will be used (Babb 1998, Tighiouart 2005). Each cohort will consist of 3 evaluable patients

Outcome Measures

Primary Outcome Measures

  1. dose-limiting toxicity (DLT) of ponatinib during consolidation 1 with HDAC or IDAC [12 months]

    assess the safety of increased doses of ponatinib in combination with high or intermediate -dose cytarabine in AML FLT3-ITD patients in first complete remission

Secondary Outcome Measures

  1. Overall survival [5 years]

    To determine disease-free survival from achievement of first complete remission

  2. Relapse-free survival [5 years]

    To determine overall survival from achievement of first complete remission

  3. Event-free survival [5 years]

    To determine overall survival from diagnosis

  4. Minimal residual disease based on FLT3-ITD quantification, WT1 expression and/or NPM1 mutation quantification [18 months]

    To study minimal residual disease after induction and consolidation courses based on the quantification of the FLT3-ITD signal and /or WT1, NPM if available

  5. relationship between minimal residual disease and outcome [18 months]

    To study the relationship between minimal residual disease and outcome

  6. To study ponatinib resistance mechanisms [18 months]

    To assess FLT3-ITD mutant before and after ponatinib treatment

Eligibility Criteria

Criteria

Ages Eligible for Study:
18 Years to 70 Years
Sexes Eligible for Study:
All
Accepts Healthy Volunteers:
No
Inclusion Criteria:
    1. Patients aged 18 to 55-60 years: Cohort A b. Patients aged 55-60 to 70 years: Cohort B
  1. Signed informed consent

  2. Acute myeloid leukemia in first complete remission

  3. Platelets ≥ 100 Giga/l; Neutrophils ≥ 1 Giga/l

  4. Intermediate risk karyotype with FLT3-ITD activating mutant detected at diagnosis (mutant FLT3/wild-type allelic ratio higher than 10%) (appendix 16)

  5. Induction with intensive chemotherapy, dose dense sequential induction or 3 + 7 like regimen (daunorubicin or idarubicin) for Cohort A and inclusion in the ALFA backbone for cohort B.

  6. Pancreatic functions within the normal range

  7. AST or ALT less or equal to 2.5 fold upper normal range, bilirubin less or equal to 1.5 fold upper normal range

  8. Serum creatinine less or equal to 1.5 fold upper normal range

  9. Two planned consolidation courses with high-dose cytarabine (HDAC, Cohort A) or intermediate dose cytarabine (IDAC, Cohort B).

Exclusion Criteria:
  1. Acute promyelocytic leukemia

  2. Transformation of myeloproliferative or myelodysplastic syndromes

  3. Known central nervous system involvement

  4. Uncontrolled bacterial, viral or fungal infection

  5. Other active malignancy

  6. Previous episode of pancreatitis

  7. Hypertriglyceridemia > 4.5 g/L

  8. Lipase > 1.5 × ULN, amylase > 1.5 x ULN not related to leukemia

  9. QTc > 470 ms (Bazett formula, see Appendix 1)

  10. Patients at high or very high risk of cardiovascular disease with any of the following

  1. Established cardiovascular disease
  • Cardiac disease:

  • Congestive heart failure greater than class II NYHA or

  • Left ventricular ejection fraction (LVEF) < 50% or

  • Unstable angina (anginal symptoms at rest) or

  • New onset angina (began within the last 3 months) or

  • Myocardial infarction, coronary/peripheral artery disease, congestive heart failure, cerebrovascular accident including transient ischemic attack within the past 12 months or

  • History of thrombolic or embolic events

  • Arrhythmias

  • Any history of clinically significant cardiac arrhythmias requiring anti-arrhythmic therapy.

  1. Diabetes Mellitus untreated or not equilibrated with therapy h) Arterial Hypertension,
    • Any history of hypertension with
  • Hypertensive encephalopathy

  • Posterior leucoencephalopathy

  • Aortic or artery dissection i) Familial dysplipidemia. j) Taking medications that are known to be associated with Torsades de Pointes (see Appendix 11)

Contacts and Locations

Locations

Site City State Country Postal Code
1 Dr Abdelaziz CHAIB Aix-en-Provence France 13600
2 Chu Amiens Amiens France 80054
3 CHU d'Angers Angers France 49033
4 Hôpital VICTOR DUPOUY Argenteuil France 95107
5 Dr Edouard RANDIAMALALA Bayonne France 64100
6 CHU de Besançon Besançon France 25030
7 Dr Thorsten BRAUN Bobigny France 93000
8 CHU Boulogne Sur Mer Boulogne Sur Mer cedex France 62321
9 Chr Clemenceau Caen Cedex France 14033
10 Hôpital d'Instruction des Armées PERCY Clamart France 92141
11 Dr Stéphanie HAÏAT Corbeil-essonnes France 91100
12 Hôpital Henri Mondor Créteil France 94010
13 CHU de Dijon Dijon France 21079
14 Centre hospitalier de Versailles Le Chesnay cedex France 78157
15 Hôpital Claude Huriez Lille cedex France 59037
16 CHRU Dupuytren Limoges cedex France 87042
17 Hôpital Edouard Herriot Lyon cedex 03 France 69437
18 Dr Regis COSTELLO Marseille France 13000
19 Centre Hospitalier de Meaux Meaux France 77104
20 Dr Mario OJEDA-URIBE Mulhouse France 68000
21 Dr Jacques DELAUNAY Nantes France 44000
22 CHU Nice, Hôpital Archet 1 Nice cedex 3 France 06202
23 CHU de Nîmes Nîmes France 30029
24 Hôpital Saint Antoine Paris cedex 12 France 75751
25 Hôpital Necker Enfants Malades Paris cedex 15 France 75743
26 Hôpital Saint Louis Paris France 75010
27 Hôpital La Pitié Salpêtrière Paris France 75013
28 Dr Laurence SANHES Perpignan France 66000
29 Dr Arnaud PIGNEUX Pessac France 33604
30 Centre Hospitalier René Dubos Pontoise Cedex France 95303
31 Marc BERNARD Rennes France 35000
32 Dr Emilie LEMASLE Rouen France 76000
33 Centre Hospitalier René Huguenin Saint Cloud France 92210
34 Institut de Cancérologie de la Loire Saint-Priest-en-Jarez France 42270
35 Dr Réda GARIDI Saint-Quentin France 02100
36 Dr Christian RECHER Toulouse France 31000
37 Centre Hospitalier de Valenciennes Valenciennes France 59322

Sponsors and Collaborators

  • Versailles Hospital

Investigators

  • Principal Investigator: Rousselot Philippe, Pr, CH Versailles

Study Documents (Full-Text)

None provided.

More Information

Publications

None provided.
Responsible Party:
Philippe ROUSSELOT, Clinical coordinator, Versailles Hospital
ClinicalTrials.gov Identifier:
NCT02428543
Other Study ID Numbers:
  • 2013-000268-27
First Posted:
Apr 29, 2015
Last Update Posted:
Aug 10, 2020
Last Verified:
Aug 1, 2020
Keywords provided by Philippe ROUSSELOT, Clinical coordinator, Versailles Hospital
Additional relevant MeSH terms:

Study Results

No Results Posted as of Aug 10, 2020