Study of eFT226 in Subjects With Selected Advanced Solid Tumor Malignancies (Zotatifin)

December 19, 2023 updated by: Effector Therapeutics

A Phase 1-2 Dose-Escalation and Cohort-Expansion Study of Intravenous Zotatifin (eFT226) in Subjects With Selected Advanced Solid Tumor Malignancies

This clinical trial is a Phase 1-2, open-label, sequential-group, dose-escalation and cohort-expansion study evaluating the safety, pharmacokinetics (PK), pharmacodynamics, and antitumor activity of Zotatifin (eFT226) in subjects with selected advanced solid tumor malignancies.

Study Overview

Detailed Description

Part 1 (Dose Escalation): Completed; Recommended Phase 2 Dose (RP2D) and Maximum Tolerated Dose (MTD) identified

Part 1a (Dose Escalation) This cohort will enroll patients with an advanced breast cancer that is refractory or intolerant to SOC therapy.

Part 1b (Dose Escalation) This cohort will enroll patients with an advanced breast cancer that is refractory or intolerant to SOC therapy.

Part 2 (Expansion Cohort) provides defined expansion cohorts to further explore the safety, pharmacology, and clinical activity of eFT226 monotherapy and in various combinations in subjects with previously treated advanced solid tumor malignancies.

Study Type

Interventional

Enrollment (Estimated)

30

Phase

  • Phase 2
  • Phase 1

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Contact

Study Locations

    • California
      • Los Angeles, California, United States, 90033
        • Recruiting
        • University of Southern California
        • Principal Investigator:
          • Anthony El-Khoueiry, MD
        • Contact:
      • Los Angeles, California, United States, 90067
      • Newport Beach, California, United States, 92663
        • Completed
        • Hoag Memorial Hospital Presbyterian
      • Palo Alto, California, United States, 94304
        • Recruiting
        • Stanford University
        • Principal Investigator:
          • Jennifer Caswell-Jin, MD
        • Contact:
    • Michigan
      • Grand Rapids, Michigan, United States, 49546
        • Recruiting
        • Start Midwest
        • Principal Investigator:
          • Manish Sharma, MD
        • Contact:
    • Nevada
      • Las Vegas, Nevada, United States, 89169
        • Completed
        • Comprehensive Cancer Centers of Nevada
    • New Jersey
      • Middletown, New Jersey, United States, 07748
        • Recruiting
        • Memorial Sloan Kettering Cancer Center- Monmouth
        • Contact:
        • Contact:
        • Principal Investigator:
          • Ezra Rosen, MD
    • New York
      • Commack, New York, United States, 11725
        • Recruiting
        • Memorial Sloan Kettering Cancer Center- Commack
        • Contact:
        • Principal Investigator:
          • Ezra Rosen, MD
        • Contact:
      • Harrison, New York, United States, 10604
        • Recruiting
        • Memorial Sloan Kettering Cancer Center- Westchester
        • Contact:
        • Contact:
        • Principal Investigator:
          • Ezra Rosen, MD
      • New York, New York, United States, 11101
        • Recruiting
        • Memorial Sloan Kettering Cancer Center- David H. Koch Center for Cancer Care
        • Contact:
        • Principal Investigator:
          • Ezra Rosen, MD
        • Contact:
    • Ohio
      • Toledo, Ohio, United States, 43614
        • Completed
        • University of Toledo Medical Center
    • Texas
      • Houston, Texas, United States, 77030
        • Recruiting
        • MD Anderson Cancer Center
        • Principal Investigator:
          • Funda Meric-Bernstam, MD
        • Contact:
      • San Antonio, Texas, United States, 78229
        • Recruiting
        • New Experimental Therapeutics of San Antonio - NEXT Oncology
        • Principal Investigator:
          • David Sommerhalder, MD
        • Contact:
    • Virginia
      • Fairfax, Virginia, United States, 22031
        • Recruiting
        • Virginia Cancer Specialists
        • Principal Investigator:
          • Alexander Spira, MD
        • Contact:

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

16 years and older (Adult, Older Adult)

Accepts Healthy Volunteers

No

Description

Key Criteria:

Parts 1a and 1b (Dose Escalation + Fulvestrant):

  • Patient has histological or cytological confirmation of breast cancer.
  • Patient has metastatic disease or locoregionally recurrent disease which is refractory or intolerant to existing therapy(ies) known to provide clinical benefit.
  • Patient has had prior chemotherapy, endocrine therapy, or other therapy as follows:

    • Minimum of one prior line of therapy for advanced/metastatic disease.
    • Maximum of five prior lines of therapy for advanced/metastatic disease.
    • Recurrence or progression on at least one line of endocrine therapy in the advanced/metastatic disease setting.
    • Prior treatment has included a CDK4/6 inhibitor.
  • Tumor is ER+ (defined as ER IHC staining > 0%).

Cohort EMNK:

  • Patient has undergone treatment with platinum-based chemotherapy and an anti-PD-1/L1 agent, if appropriate.
  • Tumor has a known KRAS-activating mutation; Patients with KRAS G12C mutations are excluded.

Cohort EMBF:

  • Patient has had prior chemotherapy, endocrine therapy, or other therapy as follows:

    • Minimum of one prior line of therapy for advanced/metastatic disease.
    • Maximum of five prior lines of therapy for advanced/metastatic disease.
    • Recurrence or progression on at least one line of endocrine therapy in the advanced/metastatic disease setting, which may include combination therapy (eg, with a CDK4/6 inhibitor).
  • Tumor is ER+ (defined as ER IHC staining > 0%) and has FGFR amplification.

Cohort EMBH:

  • Patient has had prior chemotherapy, endocrine therapy, or other therapy as follows:

    • Minimum of one prior line of therapy for advanced/metastatic disease.
    • Minimum of one line of HER2-directed therapy Note: Prior treatment with CDK4/6 inhibitors is permitted.
  • Tumor is ER+ (defined as ER IHC staining > 0%) and HER2+ (defined as HER2 3+ IHC staining or HER2 2+ and FISH+).

Cohort ECNS:

  • Patient has histologically or cytologically confirmed stage IIIB (pleural or pericardial effusion) or stage IV NSCLC.
  • Patient has undergone treatment with platinum-based chemotherapy and an anti-PD-1/L1 agent, if appropriate. Note: Patients who have declined approved therapy(ies) or who per treating physician are not eligible for approved therapy(ies) (eg, due to intolerance) may be eligible following discussion with the Medical Monitor.
  • Tumor has a known G12C KRAS-activating mutation. Note: Patients who have been previously treated with KRAS-specific therapy are excluded.

Cohort ECBF:

  • Patient has had prior chemotherapy, endocrine therapy, or other therapy as follows:

    • Minimum of one prior line of therapy for advanced/metastatic disease.
    • Maximum of five prior lines of therapy for advanced/metastatic disease.
    • Recurrence or progression on at least one line of endocrine therapy in the advanced/metastatic disease setting.
    • Prior treatment has included a CDK4/6 inhibitor.
  • Tumor is ER+ (defined as ER IHC staining > 0%).

Cohort ECBF+A:

  • Patient has had prior chemotherapy, endocrine therapy, or other therapy as follows:

    • Minimum of one prior line of therapy for advanced/metastatic disease.
    • Maximum of five prior lines of therapy for advanced/metastatic disease.
    • Recurrence or progression on at least one line of endocrine therapy in the advanced/metastatic disease setting.
  • Tumor is ER+ (defined as ER IHC staining > 0%) and HER2- (defined as absence of HER2 3+ IHC staining and/or absence of FISH+).

Cohort ECBT:

  • Patient has progressed after treatment with at least one approved anti-HER2 agent and has been administered at least one line of chemotherapy.
  • Tumor is HER2+ (defined as HER2 3+ IHC staining or HER2 2+ and FISH+). Cohorts EMBF, EMBH, ECBF, ECBF+A: There is no limit on the number of lines of prior endocrine therapies.

Cohort ECBF-D1:

  • Patient has metastatic disease or locoregionally recurrent disease which is refractory or intolerant to existing therapy(ies) known to provide clinical benefit.
  • Patient has had prior chemotherapy, endocrine therapy, or other therapy as follows:

    • Minimum of one prior line of therapy for advanced/metastatic disease.
    • Maximum of five prior lines of therapy for advanced/metastatic disease.
    • Recurrence or progression on at least one line of endocrine therapy in the advanced/metastatic disease setting.
    • Prior treatment has included a CDK4/6 inhibitor.
  • Tumor is ER+ (defined as ER IHC staining > 0%).
  • Tumor has amplification of Cyclin D1 as determined by next generation sequencing or in situ hybridization.

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Non-Randomized
  • Interventional Model: Sequential Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Part 1: Sequential escalation (Completed)
eFT226 administered IV weekly in 21-day cycles; dose escalated in sequential cohorts after subjects enrolled in a given cohort have completed DLT evaluation period.
eFT226 is a novel small-molecule, investigational drug being developed by eFFECTOR Therapeutics as an anticancer therapy. eFT226 is a potent and selective inhibitor of eIF4A1-mediated translation and selectively regulates the translation of a subset of mRNAs based on sequence specific recognition motifs in their 5'-UTR. eIF4A1 inhibition by eFT226 downregulates expression of receptor tyrosine kinases and KRAS, leading to decreased signaling through the PI3K/AKT and MAPK pathways. Preclinical efficacy testing of eFT226 demonstrates activity across models of solid tumor cancers with amplifications in HER2, FGFR1/2 and mutations in KRAS (including breast, NSCLC and CRC).
Other Names:
  • Selective translation inhibitor
Experimental: Part 2: Cohort Expansion, Monotherapy, NSCLC, KRAS (EMNK)
Cohort EMNK
eFT226 is a novel small-molecule, investigational drug being developed by eFFECTOR Therapeutics as an anticancer therapy. eFT226 is a potent and selective inhibitor of eIF4A1-mediated translation and selectively regulates the translation of a subset of mRNAs based on sequence specific recognition motifs in their 5'-UTR. eIF4A1 inhibition by eFT226 downregulates expression of receptor tyrosine kinases and KRAS, leading to decreased signaling through the PI3K/AKT and MAPK pathways. Preclinical efficacy testing of eFT226 demonstrates activity across models of solid tumor cancers with amplifications in HER2, FGFR1/2 and mutations in KRAS (including breast, NSCLC and CRC).
Other Names:
  • Selective translation inhibitor
Experimental: Part 2: Cohort Expansion, Monotherapy, Breast, FGFR (EMBF)
Cohort EMBF
eFT226 is a novel small-molecule, investigational drug being developed by eFFECTOR Therapeutics as an anticancer therapy. eFT226 is a potent and selective inhibitor of eIF4A1-mediated translation and selectively regulates the translation of a subset of mRNAs based on sequence specific recognition motifs in their 5'-UTR. eIF4A1 inhibition by eFT226 downregulates expression of receptor tyrosine kinases and KRAS, leading to decreased signaling through the PI3K/AKT and MAPK pathways. Preclinical efficacy testing of eFT226 demonstrates activity across models of solid tumor cancers with amplifications in HER2, FGFR1/2 and mutations in KRAS (including breast, NSCLC and CRC).
Other Names:
  • Selective translation inhibitor
Experimental: Part 2: Cohort Expansion, Monotherapy, Breast, HER2 (EMBH)
Cohort EMBH
eFT226 is a novel small-molecule, investigational drug being developed by eFFECTOR Therapeutics as an anticancer therapy. eFT226 is a potent and selective inhibitor of eIF4A1-mediated translation and selectively regulates the translation of a subset of mRNAs based on sequence specific recognition motifs in their 5'-UTR. eIF4A1 inhibition by eFT226 downregulates expression of receptor tyrosine kinases and KRAS, leading to decreased signaling through the PI3K/AKT and MAPK pathways. Preclinical efficacy testing of eFT226 demonstrates activity across models of solid tumor cancers with amplifications in HER2, FGFR1/2 and mutations in KRAS (including breast, NSCLC and CRC).
Other Names:
  • Selective translation inhibitor
Experimental: Part 2: Cohort Expansion, Combination, Breast, Fulvestrant (ECBF)
Cohort ECBF; Combination therapy partner administered per SOC at the approved dose.
500 mg administered intramuscularly on Days 1, 15, 29, and once monthly thereafter
Other Names:
  • Faslodex
Experimental: Part 2: Cohort Expansion, Combination, NSCLC, Sotorasib (ECNS)
Cohort ECNS; Combination therapy partner administered per SOC at the approved dose.
Recommended dosage: 960 mg orally once daily
Other Names:
  • Lumarkus
Experimental: Part 2: Cohort Expansion, Combination, Breast, Fulvestrant+Abemaciclib (ECBF+A)
Cohort ECBF+A; Combination therapy partner administered per SOC at the approved dose.
500 mg administered intramuscularly on Days 1, 15, 29, and once monthly thereafter
Other Names:
  • Faslodex
Dose in combination with fulvestrant: 150 mg twice daily
Other Names:
  • Verzenia
Experimental: Part 2: Cohort Expansion, Combination, Breast, Trastuzumab (ECBT)
Cohort ECBT; Combination therapy partner administered per SOC at the approved dose.
600 mg every 3 weeks
Other Names:
  • Herceptin
Experimental: Part 1a: Dose Escalation, Combination, Breast
eFT226 administered IV weekly in 21-day cycles. Fulvestrant will also be given. Dose escalations per protocol.
500 mg administered intramuscularly on Days 1, 15, 29, and once monthly thereafter
Other Names:
  • Faslodex
Experimental: Part 1b Dose Escalation, Combination, Breast
eFT226 administered IV every other week in 14-day cycles. Fulvestrant will also be given. Dose escalations per protocol.
500 mg administered intramuscularly on Days 1, 15, 29, and once monthly thereafter
Other Names:
  • Faslodex
Experimental: Part 2 Cohort Expansion, Combination, Breast, Fulvestrant, Cyclin D1
ECBF-D1; Combination therapy partner administered per SOC at the approved dose.
500 mg administered intramuscularly on Days 1, 15, 29, and once monthly thereafter
Other Names:
  • Faslodex

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Parts 1a and 1b: MTD
Time Frame: Through study completion, approximately 12 months
determined by occurrence of first cycle DLTs within a 3+3 or 3+3+3 clinical trial design
Through study completion, approximately 12 months
Parts 1a and 1b; incidence of AEs, serious adverse events (SAEs), and DLTs
Time Frame: Through study completion, approximately 12 months
according to National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE)
Through study completion, approximately 12 months
Parts 1a and 1b: RP2D
Time Frame: Through study completion, approximately 12 months
determined by Incidence and type of DLTs
Through study completion, approximately 12 months
Parts 1a and 1b: RP2D
Time Frame: Through study completion, approximately 12 months
determine by Incidence, type, and severity of AEs and SAEs graded as per NCI CTCAE
Through study completion, approximately 12 months
Part 2: Objective Response Rate- Efficacy
Time Frame: Through study completion, approximately 12 months
defined as confirmed Complete Response (CR) or Partial Response (PR)
Through study completion, approximately 12 months
Part 2: (Combination Cohorts) Determine MTD
Time Frame: Through study completion, approximately 12 months
determined by occurrence of first cycle Dose Limiting Toxicities (DLTs) within the study design
Through study completion, approximately 12 months
Part 2: (Combination Cohorts) Incidence, type, and severity of AEs and SAEs
Time Frame: Through study completion, approximately 12 months
via adverse event monitoring
Through study completion, approximately 12 months
Part 2: (Combination Cohorts) Determine RP2D
Time Frame: Through study completion, approximately 12 months
determined by incidence and type of DLTs, and incidence, type, and severity of Adverse Events (AEs) and Serious Adverse Events (SAEs)
Through study completion, approximately 12 months
Part 2: Percent change in tumor dimensions of target lesions- Efficacy
Time Frame: Through study completion, approximately 12 months
calculated by the percentage change from baseline in the sum of the LD of target lesions
Through study completion, approximately 12 months
Part 2: Time to Response (TTR)- Efficacy
Time Frame: Through study completion, approximately 12 months
defined as the interval from the start of study therapy to the first documentation of an objective response
Through study completion, approximately 12 months
Part 2: Duration of Response (DOR)- Efficacy
Time Frame: Through study completion, approximately 12 months
defined as the interval from the first documentation of objective response to the earlier of the first documentation of disease progression or death from any cause
Through study completion, approximately 12 months

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Parts 1a and 1b: Objective response
Time Frame: Through study completion, approximately 12 months
determined by confirmed CR or PR
Through study completion, approximately 12 months
Parts 1a and 1b: Percent change in tumor dimensions of target lesions
Time Frame: Through study completion, approximately 12 months
calculated by the percentage change from baseline in the sum of the LD of target lesions
Through study completion, approximately 12 months
Parts 1a and 1b: TTR
Time Frame: Through study completion, approximately 12 months
defined as the interval from the start of study therapy to the first documentation of an objective response
Through study completion, approximately 12 months
Parts 1a and 1b: DOR
Time Frame: Through study completion, approximately 12 months
defined as the interval from the first documentation of objective response to the earlier of the first documentation of disease progression or death from any cause
Through study completion, approximately 12 months
Parts 1a and 1b: PFS
Time Frame: Through study completion, approximately 12 months
defined as the interval from the start of study therapy to the earlier of the first documentation of disease progression or death from any cause
Through study completion, approximately 12 months
Part 2: (Monotherapy and Combination Cohorts) Incidence and severity of AEs, SAEs, and additional safety parameters
Time Frame: Through study completion, approximately 12 months
via adverse event monitoring
Through study completion, approximately 12 months
Part 2: Progression Free Survival
Time Frame: Through study completion, approximately 12 months
defined as the interval from the start of study therapy to the earlier of the first documentation of disease progression or death from any cause
Through study completion, approximately 12 months
Evaluate plasma Pharmacokinetic (PK) parameters of eFT226
Time Frame: Through study completion, approximately 12 months
including area under the plasma concentration-time curve
Through study completion, approximately 12 months
Evaluate plasma Pharmacokinetic (PK) parameters of eFT226
Time Frame: Through study completion, approximately 12 months
including maximum concentration
Through study completion, approximately 12 months
Evaluate plasma Pharmacokinetic (PK) parameters of eFT226
Time Frame: Through study completion, approximately 12 months
including terminal phase rate constant
Through study completion, approximately 12 months
Evaluate plasma Pharmacokinetic (PK) parameters of eFT226
Time Frame: Through study completion, approximately 12 months
including estimated steady-state volume of distribution [Vss]
Through study completion, approximately 12 months
Evaluate plasma Pharmacokinetic (PK) parameters of eFT226
Time Frame: Through study completion, approximately 12 months
including half-life (t½)
Through study completion, approximately 12 months
Evaluate plasma Pharmacokinetic (PK) parameters of eFT226
Time Frame: Through study completion, approximately 12 months
including total body clearance
Through study completion, approximately 12 months
Evaluate plasma Pharmacokinetic (PK) parameters of eFT226including terminal state volume of distribution
Time Frame: Through study completion, approximately 12 months
including terminal state volume of distribution
Through study completion, approximately 12 months
Evaluate plasma Pharmacokinetic (PK) parameters of eFT226 including terminal phase rate constant
Time Frame: Through study completion, approximately 12 months
including terminal phase rate constant
Through study completion, approximately 12 months

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Investigators

  • Study Director: Douglas Warner, MD, EFFECTOR Therapeutics, Inc.

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (Actual)

October 25, 2019

Primary Completion (Estimated)

December 31, 2024

Study Completion (Estimated)

March 31, 2025

Study Registration Dates

First Submitted

September 10, 2019

First Submitted That Met QC Criteria

September 13, 2019

First Posted (Actual)

September 17, 2019

Study Record Updates

Last Update Posted (Estimated)

December 20, 2023

Last Update Submitted That Met QC Criteria

December 19, 2023

Last Verified

December 1, 2023

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

NO

IPD Plan Description

There is not a plan to make IPD available

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

Yes

Studies a U.S. FDA-regulated device product

No

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Solid Tumor, Adult

Clinical Trials on Trastuzumab

3
Subscribe