search
Back to results

Evaluation of ETC-1907206 With Dasatinib in Advanced Haematologic Malignancies

Primary Purpose

Ph+ Acute Lymphoblastic Leukemia (Ph+ALL), Ph- Acute Lymphoblastic Leukemia (Ph-ALL), Chronic Myeloid Leukemia Accelerated Phase (CML-AP, Ph+)

Status
Withdrawn
Phase
Phase 1
Locations
International
Study Type
Interventional
Intervention
ETC-1907206
dasatinib
Sponsored by
EDDC (Experimental Drug Development Centre), A*STAR Research Entities
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Ph+ Acute Lymphoblastic Leukemia (Ph+ALL) focused on measuring Mnk kinase inhibitor, Mnk kinase

Eligibility Criteria

18 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

INCLUSION CRITERIA:

Each patient (male or female) must meet all of the following criteria to be enrolled in this study:

  1. Capable of understanding the written informed consent, provides signed and witnessed written informed consent, and agrees to comply with protocol requirements.
  2. Age 18 years or older (US sites) or 21 years or older (Singapore site) at Baseline.
  3. Bone marrow (BM) cytogenetic analysis with at least 20 metaphase cells, confirmed advanced haematologic malignancies in any of the 4 following disease populations at Screening:

    • CML-AP, Ph+
    • CML-BC, Ph+
    • Ph+ ALL
    • Ph- ALL with relapsed and refractory disease who have exhausted all available therapy (for patients who develop T315I mutation related resistance, the definition requires failure of ponatinib treatment if drug is accessible).
  4. Meets definition for one of the following study subgroups:

    CML-AP:

    • ≥ 15% and < 30% blast in peripheral blood or bone marrow, or
    • ≥ 20% basophils in peripheral blood or bone marrow or
    • ≥ 30% blasts + promyelocytes in peripheral blood or bone marrow (but < 30% blasts) or
    • < 100 x 10^9 platelets/L in peripheral blood unrelated to therapy or
    • Cytogenetic, genetic evidence of clonal evolution and
    • No extramedullary disease.

    CML-BC:

    • ≥ 30% blasts in peripheral blood or bone marrow, or
    • extramedullary disease other than hepatosplenomegaly.

    Ph+ ALL:

    • ≥ 30% blasts in blood or bone marrow and
    • no prior history of CML.

    Ph- ALL:

    • ≥ 10% blasts in bone marrow.
  5. ECOG performance status of 0 to 2 at Baseline.
  6. Life expectancy of at least 3 months at Baseline.
  7. Adequate organ function at Baseline, including the following (noting that repeated tests at Baseline should not be performed unless there are sufficient reasons to assume the patient would meet the inclusion criteria with re testing):

    1. Total bilirubin ≤ 1.5 x upper limit of normal (ULN), unless resulting from haemolysis or documented Gilbert syndrome.
    2. Transaminases [aspartate aminotransferase (AST) and/or alanine aminotransferase (ALT) ≤ 2.5 x ULN]. [< 5 x ULN if liver infiltration with tumour present]
    3. Prothrombin time (PT) < 1.5 ULN.
    4. Calculated creatinine clearance ≥ 60 mL/min (Cockcroft and Gault formula).
    5. No clinically relevant abnormalities in the urinalysis results.
    6. Haematology:

      • Haemoglobin > 10 g/dL (transfusion allowed to reach the level)
      • Neutrophils > 1,000/µL
      • Platelets > 75,000/µL.
    7. Pancreatic status:

      • Lipase ≤ 1.5 x ULN
      • Amylase ≤ 1.5 x ULN.
  8. Capable of taking oral medication and following direction regarding taking study drug (either by himself/herself or by caregiver).
  9. Negative serum pregnancy test at Baseline plus a negative urine pregnancy test on Day 1, Cycle 1 prior to treatment (applies to females of childbearing potential only).
  10. A minimum of 2 weeks (14 days), or 5 half-lives (whichever is shorter) since last receipt of any anti-cancer therapy (except dasatinib, hydroxyurea, anagrelide or steroids), or 4 weeks from radiation or major surgery to the first administration of the study drug.
  11. All non-haematological AEs of any prior anti-cancer therapy, surgery, or radiotherapy have to be resolved to NCI CTCAE Grade ≤ 1 (except alopecia) within 2 weeks prior to starting study drug.
  12. Willing to submit the blood samples and bone marrow samples for PK and pharmacodynamic (PD) analyses.

CML-AP Ph+, CML-BC Ph+, Ph- ALL, and Ph+ ALL patients with relapsed and refractory disease who have exhausted all available therapy.

Subgroup-specific intolerance definition: [Intolerance to tyrosine kinase inhibitors (TKIs) or other approved treatments for CML-AP, CML-BC and Ph+ ALL; to approved treatments for Ph- ALL] defined as:

  • Non-haematologic intolerance:

Patients with Grade 3 or 4 toxicity while on therapy, or with persistent Grade 2 toxicity, unresponsive to optimal management, including dose adjustments (unless a dose reduction is not considered in the best interest of the patient if response is already suboptimal) in absence of: major haematologic response (MaHR) for accelerated phase (AP), blast crisis (BC) or Ph+ ALL patients; complete remission (CR) or complete haematological response with partial haematologic recovery of peripheral blood count (CRh) for Ph- ALL.

  • Haematologic intolerance:

Patients with Grade 3 or 4 toxicity [absolute neutrophil count (ANC) or platelets] while on therapy that is recurrent after dose reduction to the lowest dose recommended by drug manufacturers in the absence of: MaHR for AP, BC or Ph+ ALL patients; CR or CRh for Ph- ALL.

NOTE: For dasatinib, non-haematologic and haematologic intolerance is defined as: CTCAE Grade > 2 requiring discontinuation.

EXCLUSION CRITERIA:

Patients meeting any of the following criteria will be excluded from the study:

  1. Is a male patient with sexual partner(s) of childbearing potential who is unwilling to use a highly effective method of contraception, one of which includes a condom. Sexually active male patients must use a condom during intercourse throughout the study and for 12 weeks after the end of treatment and should not father a child in this period. A condom is required to be used also by vasectomised males in order to prevent potential delivery of the study drug via seminal fluid. Female partners of male patients must be advised to also use one of the following contraception methods:

    • intrauterine device or intrauterine system;
    • prior sterilisation; or
    • total abstinence from male/female intercourse.
  2. Is a female patient of childbearing potential, defined as a female physiologically capable of becoming pregnant (including a female whose career, lifestyle, or sexual orientation precludes intercourse with a male partner, and females whose partners have been sterilised by vasectomy or other means), unless they are using a highly effective method for birth control throughout the study and for 12 weeks after the end of treatment. Highly effective methods for birth control include the following:

    • Total abstinence: This is an acceptable method when this is consistent with the preferred and usual lifestyle of the patient. Periodic abstinence (e.g., calendar, ovulation, symptothermal, post-ovulation methods) and withdrawal are not acceptable methods of contraception.
    • Female sterilisation: The patient has had a surgical bilateral oophorectomy (with or without hysterectomy) or tubal ligation at least 6 weeks prior to taking study drug. In case of an oophorectomy alone, the reproductive status of the patient must have been confirmed by follow-up hormone level assessment.
    • Male partner sterilisation: The patient has the appropriate post-vasectomy documentation of the absence of sperm in the ejaculate. (For female patients on the study, the vasectomised male partner should be the sole partner for that patient.) These patients must also agree to the use an intrauterine device or intrauterine system AND a barrier method of contraception: condom or occlusive cap (diaphragm or cervical/vault caps) with spermicidal foam, gel, film, or cream, or vaginal suppository. Reliable contraception must be maintained throughout the study and for 12 weeks after study drug discontinuation.
    • Females considered post-menopausal and not of childbearing potential: The definition applies to females who have had 12 months of natural (spontaneous) amenorrhoea with an appropriate clinical profile (e.g., age appropriate, history of vasomotor symptoms) or 6 months of spontaneous amenorrhoea with serum follicle-stimulating hormone (FSH) levels > 40 million international units per milliliter (mIU/mL) (for US only: and estradiol < 20 pg/mL) or have had surgical bilateral oophorectomy (with or without hysterectomy) at least 6 weeks prior to starting treatment. In the case of oophorectomy alone, only when the reproductive status of the patient has been confirmed by follow-up hormone level assessment is she considered not of childbearing potential.
  3. Is a pregnant or nursing (lactating) female, where pregnancy is defined as the state of a female after conception and until the termination of gestation, confirmed by a positive human chorionic gonadotropin (hCG) laboratory test (> 5 mIU/mL).
  4. Has dasatinib intolerance (haematologic and non-haematologic). Defined as: CTCAE Grade >2
  5. Has received anti-cancer therapy within 2 weeks or 5 half-lives, whichever is shorter (except for hydroxyurea, steroids, allopurinol, febuxostat, rasburicase, and intravenous hydration), prior to starting study drug or the side effects of such therapy have not resolved to Grade ≤1 within 2 weeks prior to starting study drug.
  6. Is receiving concomitant anti-cancer therapy (except for hydroxyurea, steroids, anagrelide, allopurinol, febuxostat, rasburicase, and intravenous hydration during the first week of the study drug[s] administration, or corticosteroids when appropriate).
  7. Has used other investigational drugs within 2 weeks or 5 half-lives (whichever is shorter) prior to the first dose of study drug.
  8. Has undergone autologous or allogenic stem cell transplantation < 60 days prior to the first dose of study drug;
  9. Has any evidence of on-going graft-versus-host disease (GVHD).
  10. Has evidence of another malignancy not in remission or history of such a malignancy within the last 3 years (except for treated basal or squamous cell carcinoma of the skin, or in situ cancer of the cervix).
  11. Has central nervous system (CNS) metastases.
  12. Has significant bleeding disorder unrelated to the disease.
  13. Has a history of long QT syndrome or prolonged QT interval corrected based on Fridericia's method (QTcF) > 450 ms.
  14. Has ECG evidence of complete left bundle branch block, or ventricular pacing.
  15. Has abnormalities in the 12-lead ECG that in the opinion of the Investigator increase the risk of participating in the study (e.g., sinus rhythm with PR interval > 240 ms or second degree or higher atrioventricular (AV) block confirmed by a repeat ECG).
  16. Has blood pressure and heart rate (HR) higher than 160/100 mmHg and 100 beats per minute (bpm), respectively, or lower than 80/50 mm Hg and 45 bpm, respectively, confirmed by a repeat assessment.
  17. Is receiving treatment with drugs known to be associated with Torsade de Pointes.
  18. Has ophthalmic signs or symptoms, such as flashes and colour perception changes.
  19. Has evidence of electrolyte imbalance such as hypokalaemia, hypocalcaemia, and hypomagnesaemia of NCI-CTCAE Grade ≥ 2 (NCI CTCAE version 4.03).
  20. Has symptomatic chronic heart failure; unstable angina pectoris, cardiac arrhythmia.
  21. Has cardiac left ventricular ejection fraction (LVEF) < 40% (assessed by transthoracic echocardiography).
  22. Has a history of myocardial infarction and/or thromboembolism in the past 6 months.
  23. Has uncontrolled diabetes mellitus, neurologic or psychiatric condition, an ongoing systemic (including opportunistic) clinically significant infections or any other significant or unstable concurrent medical illness that in the opinion of the Investigator would preclude protocol therapy.
  24. Has a known history of human immunodeficiency virus (HIV) sero positivity and/or is receiving combination anti retroviral therapy.
  25. Has a history of gastric bypass surgery or with pre-existing gastrointestinal disorders that may interfere with proper absorption of the drug, as per Investigator's judgement.
  26. Has history of seizure disorders or CNS leukaemia.
  27. Is receiving cytochrome P450 3A4 (CYP3A4) inhibitors within 7 days prior to the first dose of ETC-1907206 or receiving CYP3A4 inducers within 14 days prior to the first dose of ETC-1907206 and dasatinib.
  28. Cannot start treatment with dasatinib 140 mg daily, oral.
  29. Is unwilling or unable to comply with the protocol

Sites / Locations

  • Winship Cancer Institute, Emory University
  • The Center for Cancer and Blood Disorders
  • Memorial Sloan Kettering Cancer Center
  • Oregon Health & Science University
  • MD Anderson Cancer Center
  • Singapore General Hospital

Arms of the Study

Arm 1

Arm 2

Arm Type

Experimental

Experimental

Arm Label

Dose Escalation (Phase 1A)

Dose Expansion (Phase 1B)

Arm Description

An adaptive design using the ordinal Continual Reassessment Method (oCRM) will be used to determine the MTD and RD of ETC-1907206 in combination with dasatinib.

Once the MTD and/or RD has been determined in Phase 1A, an expansion cohort will be enrolled in order to characterize the safety, PK and preliminary clinical activity of ETC-1907206 in combination with dasatinib. Subjects may continue treatment in the study until disease progression, unacceptable toxicity, withdrawal of consent or it is judged not to be in the patient's interest to continue on the study.

Outcomes

Primary Outcome Measures

Maximum Tolerated Dose (MTD) (Phase 1A)
The MTD is defined as the highest possible dose with a predicted probability of having DLT not exceeding the target toxicity rate. The target toxicity rate (or the target predicted probability of DLT) for this study is set at 25%.
Phase 1B Safety: Incidence of Adverse Events (AEs) during Phase 1B
Incidence and Severity of AEs
Phase 1B PK: Area under the drug concentration-time curve (AUC) from time zero to infinite time (AUC0-inf)
Single-dose PK measurement of AUC0-inf after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1B PK: AUC from time zero to the last measureable concentration (AUC0-t)
Single-dose PK measurement of AUC0-t after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1B PK: First-order rate constant for elimination of drug (kel)
Single-dose PK measurement of kel after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1B PK: Time to reach maximum plasma concentration (Tmax)
Single-dose PK measurement of Tmax after dosing on Day 1 and Day 15 (Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1B PK: Time between drug administration and first observed concentration above lower limit if quantitation in plasma (Tlag)
Single-dose PK measurement of Tlag after dosing on Day 1 and Day 15 (Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1B PK: Total clearance (CL)
Single-dose PK measurement of CL after dosing on Day 1 and Day 15 (Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1B PK: Volume of distribution (Vd)
Single-dose PK measurement of Vd after dosing on Day 1 and Day 15 (Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1B PK: Half-life (T1/2)
Single-dose PK measurement of T1/2 after dosing on Day 1 and Day 15 (Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).

Secondary Outcome Measures

Phase 1A Safety: Incidence of Adverse Events (AEs) during Phase 1A
Incidence and Severity of AEs
Phase 1A PK: Area under the drug concentration-time curve (AUC) from time zero to infinite time (AUC0-inf)
Single-dose PK measurement of AUC0-inf after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1A PK: AUC from time zero to the last measureable concentration (AUC0-t)
Single-dose PK measurement of AUC0-t after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1A PK: First-order rate constant for elimination of drug (kel)
Single-dose PK measurement of kel after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1A PK: Time to reach maximum plasma concentration (Tmax)
Single-dose PK measurement of Tmax after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1A PK: Time between drug administration and first observed concentration above lower limit if quantitation in plasma (Tlag)
Single-dose PK measurement of Tlag after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1A PK: Total clearance (CL)
Single-dose PK measurement of CL after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1A PK: Volume of distribution (Vd)
Single-dose PK measurement of Vd after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1A PK: Half-life (T1/2)
Single-dose PK measurement of T1/2 after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Phase 1B Clinical Activity: Best Overall Response (BOR)
The BOR for each patient is determined by the following hierarchical orders: For CML-AP Ph+, CML-BC Ph+ and Ph+ ALL: major molecular response, major cytogenetic response (complete response, partial response), major haematologic response (complete response, complete remission), minor haematologic response,cytogenetic response (minor response, minimal response, no response), progressive disease For Ph- ALL: complete haematologic response, complete response with partial haematologic recovery, progressive disease The response rate will be summarised and two-sided 95% confidence intervals (CIs) on the response rates will be calculated. The best overall response will be listed.
Phase 1B Clinical Activity: objective response rate (ORR)
Phase 1B Clinical Activity: duration of objective response (DOR)
Phase 1B Clinical Activity: duration of major molecular response (DOMMR)
Phase 1B Clinical Activity: duration of complete haematologic response (DOCHR)
Phase 1B Clinical Activity: duration of complete remission (DOCRe)
Phase 1B Clinical Activity: duration of complete cytogenetic response (DOCCyR)
Phase 1B Clinical Activity: time to objective response (TTR)
Phase 1B Clinical Activity: time to major molecular response (TTMMR)
Phase 1B Clinical Activity: time to complete haematologic response (TTCHR)
Phase 1B Clinical Activity: time to complete remission (TTCRe)
Phase 1B Clinical Activity: time to complete cytogenetic response (TTCCyR)
Phase 1B Clinical Activity: progression-free survival (PFS)
Phase 1B Clinical Activity: overall survival (OS)

Full Information

First Posted
January 14, 2018
Last Updated
October 23, 2018
Sponsor
EDDC (Experimental Drug Development Centre), A*STAR Research Entities
Collaborators
Chiltern International Inc.
search

1. Study Identification

Unique Protocol Identification Number
NCT03414450
Brief Title
Evaluation of ETC-1907206 With Dasatinib in Advanced Haematologic Malignancies
Official Title
A Phase 1A Dose Escalation and Phase 1B Expansion Study to Evaluate the Safety and Tolerability of ETC-1907206 in Combination With Dasatinib in Advanced Haematologic Malignancies
Study Type
Interventional

2. Study Status

Record Verification Date
October 2018
Overall Recruitment Status
Withdrawn
Why Stopped
Withdrawal requested by D3
Study Start Date
April 25, 2018 (Actual)
Primary Completion Date
February 2022 (Anticipated)
Study Completion Date
February 2023 (Anticipated)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
EDDC (Experimental Drug Development Centre), A*STAR Research Entities
Collaborators
Chiltern International Inc.

4. Oversight

Studies a U.S. FDA-regulated Drug Product
Yes
Studies a U.S. FDA-regulated Device Product
No
Data Monitoring Committee
No

5. Study Description

Brief Summary
This study evaluates the use of ETC-1907206 in combination with dasatinib in certain types of blood cancers. The first phase of the study (1A) is designed to find the highest tolerated dose of ETC-1907206, while the second phase (1B) will assess the safety and tolerability of the recommended dose of ETC-1907206. ETC-1907206 has been designed to block the activity of an enzyme of the body known as Mnk kinase, which is thought to be involved in the development of a variety of cancers.
Detailed Description
This study consists of two parts: a Phase 1A dose escalation to identify the MTD and the RD of ETC-1907206 administered in combination with dasatinib and a Phase 1B expansion at the RD. Phase 1A: A dose escalation with an adaptive design model using ordinal continual reassessment method (oCRM) will be used to characterise the dose toxicity curve of ETC-1907206 when administered orally every other day (EOD) under fasted conditions in combination with oral once daily dasatinib (per locally approved product prescribing instructions) in order to identify the maximum tolerated dose (MTD) and recommended dose (RD) for Phase 1B. Phase 1B: Open-label, non-randomised, to assess preliminary clinical activity and safety of ETC-1907206 administered orally EOD under fasted conditions at the RD identified in Phase 1A, in combination with dasatinib (per locally approved product prescribing instructions). Patients will continue in the study until disease progression, the start of new anti-cancer therapy, unacceptable toxicity, death, or the completion of 12 separate 4-week treatment cycles, whichever occurs first. As long as the Sponsor agrees to continue treatment, patients who complete 12 cycles of treatment and have no evidence of disease progression are allowed to continue on treatment past the end of treatment (EOT) visit until there is disease progression, unacceptable toxicity, the patient decides to withdraw, or it is judged not to be in the patient's interest to continue on the study. Malignancy assessments of the underlying disease at enrolment (blood and bone marrow), Eastern Cooperative Oncology Group (ECOG) performance status, pharmacokinetic (PK) sampling for ETC-1907206 and dasatinib, sample collection for ETC-1907206 and dasatinib biomarker analysis, and safety and tolerability assessments will be performed during the study.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Ph+ Acute Lymphoblastic Leukemia (Ph+ALL), Ph- Acute Lymphoblastic Leukemia (Ph-ALL), Chronic Myeloid Leukemia Accelerated Phase (CML-AP, Ph+), Chronic Myeloid Leukemia Blast Crisis (CML-BC, Ph+)
Keywords
Mnk kinase inhibitor, Mnk kinase

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 1
Interventional Study Model
Sequential Assignment
Model Description
In Phase 1A, an adaptive design using the ordinal Continual Reassessment Method (oCRM) will be used to determine the MTD and RD of ETC-1907206 in combination with dasatinib.
Masking
None (Open Label)
Allocation
Non-Randomized
Enrollment
0 (Actual)

8. Arms, Groups, and Interventions

Arm Title
Dose Escalation (Phase 1A)
Arm Type
Experimental
Arm Description
An adaptive design using the ordinal Continual Reassessment Method (oCRM) will be used to determine the MTD and RD of ETC-1907206 in combination with dasatinib.
Arm Title
Dose Expansion (Phase 1B)
Arm Type
Experimental
Arm Description
Once the MTD and/or RD has been determined in Phase 1A, an expansion cohort will be enrolled in order to characterize the safety, PK and preliminary clinical activity of ETC-1907206 in combination with dasatinib. Subjects may continue treatment in the study until disease progression, unacceptable toxicity, withdrawal of consent or it is judged not to be in the patient's interest to continue on the study.
Intervention Type
Drug
Intervention Name(s)
ETC-1907206
Other Intervention Name(s)
ETC-206
Intervention Description
ETC-1907206 gelatin capsules will be dosed every other day (EOD) and contain 10 mg or 50 mg of ETC-1907206.
Intervention Type
Drug
Intervention Name(s)
dasatinib
Other Intervention Name(s)
Sprycel
Intervention Description
dasatinib tablets at 140 mg will be dosed every day
Primary Outcome Measure Information:
Title
Maximum Tolerated Dose (MTD) (Phase 1A)
Description
The MTD is defined as the highest possible dose with a predicted probability of having DLT not exceeding the target toxicity rate. The target toxicity rate (or the target predicted probability of DLT) for this study is set at 25%.
Time Frame
the initial 28 days of treatment
Title
Phase 1B Safety: Incidence of Adverse Events (AEs) during Phase 1B
Description
Incidence and Severity of AEs
Time Frame
up to 44 months
Title
Phase 1B PK: Area under the drug concentration-time curve (AUC) from time zero to infinite time (AUC0-inf)
Description
Single-dose PK measurement of AUC0-inf after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1B PK: AUC from time zero to the last measureable concentration (AUC0-t)
Description
Single-dose PK measurement of AUC0-t after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1B PK: First-order rate constant for elimination of drug (kel)
Description
Single-dose PK measurement of kel after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1B PK: Time to reach maximum plasma concentration (Tmax)
Description
Single-dose PK measurement of Tmax after dosing on Day 1 and Day 15 (Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1B PK: Time between drug administration and first observed concentration above lower limit if quantitation in plasma (Tlag)
Description
Single-dose PK measurement of Tlag after dosing on Day 1 and Day 15 (Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1B PK: Total clearance (CL)
Description
Single-dose PK measurement of CL after dosing on Day 1 and Day 15 (Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1B PK: Volume of distribution (Vd)
Description
Single-dose PK measurement of Vd after dosing on Day 1 and Day 15 (Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1B PK: Half-life (T1/2)
Description
Single-dose PK measurement of T1/2 after dosing on Day 1 and Day 15 (Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Secondary Outcome Measure Information:
Title
Phase 1A Safety: Incidence of Adverse Events (AEs) during Phase 1A
Description
Incidence and Severity of AEs
Time Frame
up to 24 months
Title
Phase 1A PK: Area under the drug concentration-time curve (AUC) from time zero to infinite time (AUC0-inf)
Description
Single-dose PK measurement of AUC0-inf after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1A PK: AUC from time zero to the last measureable concentration (AUC0-t)
Description
Single-dose PK measurement of AUC0-t after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1A PK: First-order rate constant for elimination of drug (kel)
Description
Single-dose PK measurement of kel after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1A PK: Time to reach maximum plasma concentration (Tmax)
Description
Single-dose PK measurement of Tmax after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1A PK: Time between drug administration and first observed concentration above lower limit if quantitation in plasma (Tlag)
Description
Single-dose PK measurement of Tlag after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1A PK: Total clearance (CL)
Description
Single-dose PK measurement of CL after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1A PK: Volume of distribution (Vd)
Description
Single-dose PK measurement of Vd after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1A PK: Half-life (T1/2)
Description
Single-dose PK measurement of T1/2 after dosing on Day 1 and Day 15 Cycle 1) and pre-dose on Day 1 of Cycle 2 and beyond (each Cycle is 28-days in length).
Time Frame
pre-dose, at 0.5, 1, 1.5, 2, 3, 4, 6, 8 hours (± 6 minutes), and at 24, 30, and 48 hours (± 2 hours) after dosing
Title
Phase 1B Clinical Activity: Best Overall Response (BOR)
Description
The BOR for each patient is determined by the following hierarchical orders: For CML-AP Ph+, CML-BC Ph+ and Ph+ ALL: major molecular response, major cytogenetic response (complete response, partial response), major haematologic response (complete response, complete remission), minor haematologic response,cytogenetic response (minor response, minimal response, no response), progressive disease For Ph- ALL: complete haematologic response, complete response with partial haematologic recovery, progressive disease The response rate will be summarised and two-sided 95% confidence intervals (CIs) on the response rates will be calculated. The best overall response will be listed.
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: objective response rate (ORR)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: duration of objective response (DOR)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: duration of major molecular response (DOMMR)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: duration of complete haematologic response (DOCHR)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: duration of complete remission (DOCRe)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: duration of complete cytogenetic response (DOCCyR)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: time to objective response (TTR)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: time to major molecular response (TTMMR)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: time to complete haematologic response (TTCHR)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: time to complete remission (TTCRe)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: time to complete cytogenetic response (TTCCyR)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: progression-free survival (PFS)
Time Frame
through study completion (44 months)
Title
Phase 1B Clinical Activity: overall survival (OS)
Time Frame
through study completion (44 months)

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
INCLUSION CRITERIA: Each patient (male or female) must meet all of the following criteria to be enrolled in this study: Capable of understanding the written informed consent, provides signed and witnessed written informed consent, and agrees to comply with protocol requirements. Age 18 years or older (US sites) or 21 years or older (Singapore site) at Baseline. Bone marrow (BM) cytogenetic analysis with at least 20 metaphase cells, confirmed advanced haematologic malignancies in any of the 4 following disease populations at Screening: CML-AP, Ph+ CML-BC, Ph+ Ph+ ALL Ph- ALL with relapsed and refractory disease who have exhausted all available therapy (for patients who develop T315I mutation related resistance, the definition requires failure of ponatinib treatment if drug is accessible). Meets definition for one of the following study subgroups: CML-AP: ≥ 15% and < 30% blast in peripheral blood or bone marrow, or ≥ 20% basophils in peripheral blood or bone marrow or ≥ 30% blasts + promyelocytes in peripheral blood or bone marrow (but < 30% blasts) or < 100 x 10^9 platelets/L in peripheral blood unrelated to therapy or Cytogenetic, genetic evidence of clonal evolution and No extramedullary disease. CML-BC: ≥ 30% blasts in peripheral blood or bone marrow, or extramedullary disease other than hepatosplenomegaly. Ph+ ALL: ≥ 30% blasts in blood or bone marrow and no prior history of CML. Ph- ALL: ≥ 10% blasts in bone marrow. ECOG performance status of 0 to 2 at Baseline. Life expectancy of at least 3 months at Baseline. Adequate organ function at Baseline, including the following (noting that repeated tests at Baseline should not be performed unless there are sufficient reasons to assume the patient would meet the inclusion criteria with re testing): Total bilirubin ≤ 1.5 x upper limit of normal (ULN), unless resulting from haemolysis or documented Gilbert syndrome. Transaminases [aspartate aminotransferase (AST) and/or alanine aminotransferase (ALT) ≤ 2.5 x ULN]. [< 5 x ULN if liver infiltration with tumour present] Prothrombin time (PT) < 1.5 ULN. Calculated creatinine clearance ≥ 60 mL/min (Cockcroft and Gault formula). No clinically relevant abnormalities in the urinalysis results. Haematology: Haemoglobin > 10 g/dL (transfusion allowed to reach the level) Neutrophils > 1,000/µL Platelets > 75,000/µL. Pancreatic status: Lipase ≤ 1.5 x ULN Amylase ≤ 1.5 x ULN. Capable of taking oral medication and following direction regarding taking study drug (either by himself/herself or by caregiver). Negative serum pregnancy test at Baseline plus a negative urine pregnancy test on Day 1, Cycle 1 prior to treatment (applies to females of childbearing potential only). A minimum of 2 weeks (14 days), or 5 half-lives (whichever is shorter) since last receipt of any anti-cancer therapy (except dasatinib, hydroxyurea, anagrelide or steroids), or 4 weeks from radiation or major surgery to the first administration of the study drug. All non-haematological AEs of any prior anti-cancer therapy, surgery, or radiotherapy have to be resolved to NCI CTCAE Grade ≤ 1 (except alopecia) within 2 weeks prior to starting study drug. Willing to submit the blood samples and bone marrow samples for PK and pharmacodynamic (PD) analyses. CML-AP Ph+, CML-BC Ph+, Ph- ALL, and Ph+ ALL patients with relapsed and refractory disease who have exhausted all available therapy. Subgroup-specific intolerance definition: [Intolerance to tyrosine kinase inhibitors (TKIs) or other approved treatments for CML-AP, CML-BC and Ph+ ALL; to approved treatments for Ph- ALL] defined as: Non-haematologic intolerance: Patients with Grade 3 or 4 toxicity while on therapy, or with persistent Grade 2 toxicity, unresponsive to optimal management, including dose adjustments (unless a dose reduction is not considered in the best interest of the patient if response is already suboptimal) in absence of: major haematologic response (MaHR) for accelerated phase (AP), blast crisis (BC) or Ph+ ALL patients; complete remission (CR) or complete haematological response with partial haematologic recovery of peripheral blood count (CRh) for Ph- ALL. Haematologic intolerance: Patients with Grade 3 or 4 toxicity [absolute neutrophil count (ANC) or platelets] while on therapy that is recurrent after dose reduction to the lowest dose recommended by drug manufacturers in the absence of: MaHR for AP, BC or Ph+ ALL patients; CR or CRh for Ph- ALL. NOTE: For dasatinib, non-haematologic and haematologic intolerance is defined as: CTCAE Grade > 2 requiring discontinuation. EXCLUSION CRITERIA: Patients meeting any of the following criteria will be excluded from the study: Is a male patient with sexual partner(s) of childbearing potential who is unwilling to use a highly effective method of contraception, one of which includes a condom. Sexually active male patients must use a condom during intercourse throughout the study and for 12 weeks after the end of treatment and should not father a child in this period. A condom is required to be used also by vasectomised males in order to prevent potential delivery of the study drug via seminal fluid. Female partners of male patients must be advised to also use one of the following contraception methods: intrauterine device or intrauterine system; prior sterilisation; or total abstinence from male/female intercourse. Is a female patient of childbearing potential, defined as a female physiologically capable of becoming pregnant (including a female whose career, lifestyle, or sexual orientation precludes intercourse with a male partner, and females whose partners have been sterilised by vasectomy or other means), unless they are using a highly effective method for birth control throughout the study and for 12 weeks after the end of treatment. Highly effective methods for birth control include the following: Total abstinence: This is an acceptable method when this is consistent with the preferred and usual lifestyle of the patient. Periodic abstinence (e.g., calendar, ovulation, symptothermal, post-ovulation methods) and withdrawal are not acceptable methods of contraception. Female sterilisation: The patient has had a surgical bilateral oophorectomy (with or without hysterectomy) or tubal ligation at least 6 weeks prior to taking study drug. In case of an oophorectomy alone, the reproductive status of the patient must have been confirmed by follow-up hormone level assessment. Male partner sterilisation: The patient has the appropriate post-vasectomy documentation of the absence of sperm in the ejaculate. (For female patients on the study, the vasectomised male partner should be the sole partner for that patient.) These patients must also agree to the use an intrauterine device or intrauterine system AND a barrier method of contraception: condom or occlusive cap (diaphragm or cervical/vault caps) with spermicidal foam, gel, film, or cream, or vaginal suppository. Reliable contraception must be maintained throughout the study and for 12 weeks after study drug discontinuation. Females considered post-menopausal and not of childbearing potential: The definition applies to females who have had 12 months of natural (spontaneous) amenorrhoea with an appropriate clinical profile (e.g., age appropriate, history of vasomotor symptoms) or 6 months of spontaneous amenorrhoea with serum follicle-stimulating hormone (FSH) levels > 40 million international units per milliliter (mIU/mL) (for US only: and estradiol < 20 pg/mL) or have had surgical bilateral oophorectomy (with or without hysterectomy) at least 6 weeks prior to starting treatment. In the case of oophorectomy alone, only when the reproductive status of the patient has been confirmed by follow-up hormone level assessment is she considered not of childbearing potential. Is a pregnant or nursing (lactating) female, where pregnancy is defined as the state of a female after conception and until the termination of gestation, confirmed by a positive human chorionic gonadotropin (hCG) laboratory test (> 5 mIU/mL). Has dasatinib intolerance (haematologic and non-haematologic). Defined as: CTCAE Grade >2 Has received anti-cancer therapy within 2 weeks or 5 half-lives, whichever is shorter (except for hydroxyurea, steroids, allopurinol, febuxostat, rasburicase, and intravenous hydration), prior to starting study drug or the side effects of such therapy have not resolved to Grade ≤1 within 2 weeks prior to starting study drug. Is receiving concomitant anti-cancer therapy (except for hydroxyurea, steroids, anagrelide, allopurinol, febuxostat, rasburicase, and intravenous hydration during the first week of the study drug[s] administration, or corticosteroids when appropriate). Has used other investigational drugs within 2 weeks or 5 half-lives (whichever is shorter) prior to the first dose of study drug. Has undergone autologous or allogenic stem cell transplantation < 60 days prior to the first dose of study drug; Has any evidence of on-going graft-versus-host disease (GVHD). Has evidence of another malignancy not in remission or history of such a malignancy within the last 3 years (except for treated basal or squamous cell carcinoma of the skin, or in situ cancer of the cervix). Has central nervous system (CNS) metastases. Has significant bleeding disorder unrelated to the disease. Has a history of long QT syndrome or prolonged QT interval corrected based on Fridericia's method (QTcF) > 450 ms. Has ECG evidence of complete left bundle branch block, or ventricular pacing. Has abnormalities in the 12-lead ECG that in the opinion of the Investigator increase the risk of participating in the study (e.g., sinus rhythm with PR interval > 240 ms or second degree or higher atrioventricular (AV) block confirmed by a repeat ECG). Has blood pressure and heart rate (HR) higher than 160/100 mmHg and 100 beats per minute (bpm), respectively, or lower than 80/50 mm Hg and 45 bpm, respectively, confirmed by a repeat assessment. Is receiving treatment with drugs known to be associated with Torsade de Pointes. Has ophthalmic signs or symptoms, such as flashes and colour perception changes. Has evidence of electrolyte imbalance such as hypokalaemia, hypocalcaemia, and hypomagnesaemia of NCI-CTCAE Grade ≥ 2 (NCI CTCAE version 4.03). Has symptomatic chronic heart failure; unstable angina pectoris, cardiac arrhythmia. Has cardiac left ventricular ejection fraction (LVEF) < 40% (assessed by transthoracic echocardiography). Has a history of myocardial infarction and/or thromboembolism in the past 6 months. Has uncontrolled diabetes mellitus, neurologic or psychiatric condition, an ongoing systemic (including opportunistic) clinically significant infections or any other significant or unstable concurrent medical illness that in the opinion of the Investigator would preclude protocol therapy. Has a known history of human immunodeficiency virus (HIV) sero positivity and/or is receiving combination anti retroviral therapy. Has a history of gastric bypass surgery or with pre-existing gastrointestinal disorders that may interfere with proper absorption of the drug, as per Investigator's judgement. Has history of seizure disorders or CNS leukaemia. Is receiving cytochrome P450 3A4 (CYP3A4) inhibitors within 7 days prior to the first dose of ETC-1907206 or receiving CYP3A4 inducers within 14 days prior to the first dose of ETC-1907206 and dasatinib. Cannot start treatment with dasatinib 140 mg daily, oral. Is unwilling or unable to comply with the protocol
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Joel Leong, MD, PhD
Organizational Affiliation
D3 (Drug Discovery and Development)
Official's Role
Study Chair
Facility Information:
Facility Name
Winship Cancer Institute, Emory University
City
Atlanta
State/Province
Georgia
ZIP/Postal Code
30322
Country
United States
Facility Name
The Center for Cancer and Blood Disorders
City
Bethesda
State/Province
Maryland
ZIP/Postal Code
20874
Country
United States
Facility Name
Memorial Sloan Kettering Cancer Center
City
New York
State/Province
New York
ZIP/Postal Code
10065
Country
United States
Facility Name
Oregon Health & Science University
City
Portland
State/Province
Oregon
ZIP/Postal Code
97239
Country
United States
Facility Name
MD Anderson Cancer Center
City
Houston
State/Province
Texas
ZIP/Postal Code
77030
Country
United States
Facility Name
Singapore General Hospital
City
Singapore
ZIP/Postal Code
169608
Country
Singapore

12. IPD Sharing Statement

Plan to Share IPD
Undecided

Learn more about this trial

Evaluation of ETC-1907206 With Dasatinib in Advanced Haematologic Malignancies

We'll reach out to this number within 24 hrs