This is the classic website, which will be retired eventually. Please visit the modernized ClinicalTrials.gov instead.
Working…
ClinicalTrials.gov
ClinicalTrials.gov Menu

Study of AG-120 in Previously Treated Advanced Cholangiocarcinoma With IDH1 Mutations (ClarIDHy) (ClarIDHy)

The safety and scientific validity of this study is the responsibility of the study sponsor and investigators. Listing a study does not mean it has been evaluated by the U.S. Federal Government. Read our disclaimer for details.
 
ClinicalTrials.gov Identifier: NCT02989857
Recruitment Status : Completed
First Posted : December 12, 2016
Results First Posted : April 13, 2022
Last Update Posted : January 17, 2023
Sponsor:
Information provided by (Responsible Party):
Servier ( Institut de Recherches Internationales Servier )

Brief Summary:
Study AG120-C-005 is a Phase 3, multicenter, randomized, double-blind, placebo-controlled study of orally administered AG-120. Participants, all personnel involved in the evaluation of participants' response to treatment (e.g., Investigators, study coordinators, study pharmacists), and designated Sponsor team members will be blinded to study treatment. Participants are required to have a histologically-confirmed diagnosis of isocitrate dehydrogenase-1 (IDH1) gene-mutated cholangiocarcinoma that is not eligible for curative resection, transplantation, or ablative therapies prior to enrollment. IDH1 mutation testing will be performed at participating investigative sites. Participants must have progression of disease and have received at least 1 but not more than 2 prior treatment regimens for advanced disease (nonresectable or metastatic). All participants must have received either a gemcitabine or a 5 fluorouracil (5-FU) based chemotherapy regimen.

Condition or disease Intervention/treatment Phase
Advanced Cholangiocarcinoma Metastatic Cholangiocarcinoma Drug: AG-120 Drug: Placebo Phase 3

Layout table for study information
Study Type : Interventional  (Clinical Trial)
Actual Enrollment : 187 participants
Allocation: Randomized
Intervention Model: Parallel Assignment
Intervention Model Description: Patients randomized in a 2:1 allocation (AG-120 vs Placebo)
Masking: Double (Participant, Investigator)
Primary Purpose: Treatment
Official Title: A Phase 3, Multicenter, Randomized, Double-Blind, Placebo-controlled Study of AG-120 in Previously-treated Subjects With Nonresectable or Metastatic Cholangiocarcinoma With an IDH1 Mutation
Actual Study Start Date : February 20, 2017
Actual Primary Completion Date : January 31, 2019
Actual Study Completion Date : May 17, 2021

Resource links provided by the National Library of Medicine

Drug Information available for: Ivosidenib

Arm Intervention/treatment
Active Comparator: AG-120
Participants received AG-120 500 mg, tablet, orally, once a day (QD) in each 28-day treatment cycle, until occurrence of disease progression, unacceptable toxicity, confirmed pregnancy, death, subject withdrawal, lost to follow-up, or the sponsor ended the study for up to approximately 45 months.
Drug: AG-120
Tablet administered orally
Other Name: Ivosidenib

Placebo Comparator: Placebo
Participants received AG-120 matched placebo, orally, QD in each 28-day treatment cycle, until occurrence of disease progression, unacceptable toxicity, confirmed pregnancy, death, subject withdrawal, lost to follow-up or the sponsor ended the study for up to approximately 7 months. Participants who experienced disease progression and received placebo were allowed to cross over and receive AG-120.
Drug: Placebo
Tablet administered orally

Experimental: After Cross over to AG-120
Participants who experienced disease progression and received placebo were allowed to cross over to receive AG-120 500 mg, tablet, orally, QD in each 28-day treatment cycle for up to approximately 32 months.
Drug: AG-120
Tablet administered orally
Other Name: Ivosidenib




Primary Outcome Measures :
  1. Progression Free Survival (PFS) as Determined by the Independent Radiology Committee (IRC) [ Time Frame: From the date of randomization to the date of first documentation of disease progression or death due to any cause (Up to approximately 2 years) ]
    PFS is defined as the time from date of randomization to the date of first documented disease progression as assessed by the IRC using Response Evaluation Criteria in Solid Tumors [RECIST] v1.1, or date of death due to any cause, whichever occurred first. Disease progression was defined as greater than or equal to (≥)20 percent (%) increase in sum of the diameter of target lesions, taking as reference the smallest sum diameter recorded since the treatment started. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 millimeters (mm) or the appearance of 1 or more new lesions.


Secondary Outcome Measures :
  1. Percentage of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs) [ Time Frame: From first dose of study drug up to 28 days after last dose for each intervention (Up to approximately 4 Years) ]
    An AE is any untoward medical occurrence associated with the use of a drug in participants, whether or not considered drug related. An AE or suspected adverse reaction is considered serious (an SAE) if it is fatal, life-threatening, causes in-patient hospitalization or prolongation of existing hospitalization, persistent or significant incapacity or substantial disruption of the ability to conduct normal life functions, congenital anomaly/birth defect in a neonate/infant born to a mother or father exposed to study treatment or is an important medical event. Treatment-emergent adverse events are reported.

  2. Percentage of Participants Who Experienced Laboratory Abnormalities Reported as Grade 3 or Higher Adverse Events [ Time Frame: From first dose of the study drug up to end of treatment visit for each intervention (Up to approximately 4 Years) ]
    The laboratory parameters evaluated by the investigator included hematology and chemistry. Laboratory abnormalities reported in this endpoint are Grade 3 or higher adverse events. Grading categories were determined by the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE), version 4.03.

  3. Percentage of Participants With Clinically Significant Grade 3 or Higher Vital Signs AEs [ Time Frame: From first dose of the study drug up to end of treatment visit for each intervention (Up to approximately 4 Years) ]
    Clinically significant vital signs were recorded as adverse events; there were some vital signs reported as Grade 3 or higher adverse events. Grading categories were determined by NCI CTCAE, version 4.03.

  4. Percentage of Participants With Eastern Cooperative Oncology Group (ECOG) Performance Status [ Time Frame: Baseline ]
    The Eastern Cooperative Oncology Group Performance Status (ECOG PS) score classified participants according to their functional impairment, with scores ranging from 0 to 4. ECOG PS: 0 = fully active, able to carry on all pre-disease performance without restriction; 1 = restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature, e.g., light housework, office work; 2 = ambulatory and capable of all self-care but unable to carry out any work activities, up and about more than 50% of waking hours; 3 = capable of only limited self-care, confined to bed or chair more than 50% of waking hours; 4 = completely disabled, cannot carry on any self-care, totally confined to bed or chair. A higher score means a worse functional status.

  5. Percentage of Participants Who Required At Least One Concomitant Medications During the Treatment [ Time Frame: From first dose of study drug up to 28 days after last dose (Up to approximately 4 Years) ]
    Concomitant medications were medications that were ongoing or initiated after the first dose of the study drug but before the last dose plus 28 days. Percentage of participants who required at least one concomitant medications during the study along with their prescribed study drug (AG-120 or placebo) were reported.

  6. Percentage of Participants With Abnormal Electrocardiogram (ECG) Changes Reported as Adverse Events [ Time Frame: Pre-dose C1D1, C2D1; Post-dose C1D1, C1D15, C2D1 and Day 1 of C3D1 and all cycles thereafter up to last dose plus 28 days (Up to approximately 4 years) ]
  7. Overall Survival (OS) [ Time Frame: From date of randomization until the date of death due to any cause (Up to approximately 2 years) ]
    Overall survival was defined as the time in months from date of randomization to the date of death due to any cause. Participants without documentation of death at the time of the final collection were censored at the date the participant was last known to be alive, or the final collection date, whichever is earlier.

  8. Objective Response Rate (ORR) as Assessed by the Investigator RECIST Version 1.1 [ Time Frame: From the date of randomization up to confirmed CR or PR (Up to approximately 2 years) ]
    ORR as assessed by the investigator was defined as the percentage of participants with a best overall response (BOR) defined as complete response (CR) or partial response (PR) per RECIST v1.1. CR: disappearance of all target and non-target lesions (TLs) and all pathological lymph nodes (LNs) (target and non target), with short axis <10mm. PR: ≥30% decrease in sum of diameters (SOD) from Baseline.

  9. ORR as Assessed by the IRC Per RECIST v1.1 [ Time Frame: From the date of randomization up to confirmed CR or PR (Up to approximately 2 years) ]
    ORR as assessed by the IRC was defined as the percentage of participants with a best overall response (BOR) defined as complete response (CR) or partial response (PR) per RECIST v1.1. CR: disappearance of all target and non-target lesions (TLs) and all pathological lymph nodes (LNs) (target and non target), with short axis <10mm. PR: ≥30% decrease in sum of diameters (SOD) from Baseline.

  10. Duration of Response (DOR) as Assessed by the Investigator [ Time Frame: From the date of first confirmed CR or PR to disease progression or death regardless of cause (Up to approximately 2 years) ]
    DOR was defined as the time in months from date of first documented CR or PR to date of first documented disease progression or death due to any cause, whichever is earlier, as assessed by the Investigator per RECIST v1.1. CR: disappearance of all target and non-target lesions (TLs) and all pathological lymph nodes (LNs) (target and non target), with short axis <10mm. PR: ≥30% decrease in sum of diameters (SOD) from Baseline. Participants with response and without progression were censored at the last observation.

  11. DOR as Assessed by the IRC Per RECIST v1.1 [ Time Frame: From the date of first confirmed CR or PR to disease progression or death regardless of cause (Up to approximately 2 years) ]
    DOR was defined as the time in months from date of first documented CR or PR to date of first documented disease progression or death due to any cause, whichever is earlier, as assessed by the IRC per RECIST v1.1. CR: disappearance of all target and non-target lesions (TLs) and all pathological lymph nodes (LNs) (target and non target), with short axis <10mm. PR: ≥30% decrease in sum of diameters (SOD) from Baseline. Participants with response and without progression were censored at the last observation.

  12. Time to Response (TTR) as Assessed by the Investigator [ Time Frame: From the date of randomization up to the date of first documented CR or PR (Up to approximately 2 years) ]
    TTR was defined as the time from date of randomization to date of first documented CR or PR for responders, as assessed by the Investigator per RECIST v1.1. CR: disappearance of all target and non-target lesions (TLs) and all pathological lymph nodes (LNs) (target and non target), with short axis <10mm. PR: ≥30% decrease in sum of diameters (SOD) from Baseline. Only responders were analyzed for this outcome measure.

  13. TTR as Assessed by the IRC Per RECIST v1.1 [ Time Frame: From the date of randomization up to the date of first documented CR or PR (Up to approximately 2 years) ]
    TTR was defined as the time from date of randomization to date of first documented CR or PR for responders, as assessed by the IRC per RECIST v1.1. CR: disappearance of all target and non-target lesions (TLs) and all pathological lymph nodes (LNs) (target and non target), with short axis <10mm. PR: ≥30% decrease in sum of diameters (SOD) from Baseline. Only responders were analyzed for this outcome measure.

  14. PFS as Determined by Investigator [ Time Frame: From the date of randomization to the date of first documentation of disease progression or death due to any cause (Up to approximately 2 years) ]
    PFS was defined as the time from date of randomization to the date of first documented disease progression as assessed by the investigator using RECIST v1.1, or date of death due to any cause, whichever occurred first. Disease progression was defined as ≥20% increase in sum of the diameter of target lesions, taking as reference the smallest sum diameter recorded since the treatment started. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm or the appearance of 1 or more new lesions. No progression or death by data cutoff date was censored at the last adequate assessment date.

  15. Change From Baseline in Health-Related Quality of Life (HRQOL) Based on European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire- Core 30 Subscales Scores [ Time Frame: Cycle 2 Day 1 and Cycle 3 Day 1 ]
    EORTC-QLQ-C30 is the European Organization for Research and Treatment of Cancer - Quality of Life Questionnaire - Core Questionnaire. For EORTC QLQ-C30, subscales of physical functioning, pain, and appetite loss were assessed. These had 4 response levels (not at all, a little, quite a bit, and very much). For functional scales, higher scores=better QOL (positive change from Baseline=improvement). For symptom scales, lower scores=better QOL (negative change from Baseline=improvement). Using linear transformation, raw scores were standardized, so that scores ranged from 0 to 100.

  16. Change From Baseline in HRQOL Based on: Quality of Life Questionnaire - Cholangiocarcinoma and Gallbladder Cancer Module (QLQ-BIL21) [ Time Frame: Cycle 2 Day 1 and Cycle 3 Day 1 ]
    For HRQOL based on QLQ-BIL21, subscales of eating symptoms and pain symptoms were assessed. Each item is a 4-point Likert scale. There are 4 response levels (not at all, a little, quite a bit, and very much). Raw scores are converted into scale scores ranging from 0 to 100. For symptom scales, lower scores=better QOL (negative change from Baseline=improvement).

  17. Percentage of Participants With Change Based on HRQOL: Patient Global Impression of Change (PGI-C) [ Time Frame: Cycle 2 Day 1 and Cycle 3 Day 1 ]
    The PGI-C is a self-rated evaluative instrument for assessment across 3 domains (physical function, appetite loss, and pain). The PGI-C is measured using a 7-point Likert scale, with 6= very much better, 5= moderately better, 4= a little better, 3= no change, 2= a little worse, 1= moderately worse, and 0= very much worse. A lower score indicates a worse outcome. Percentages are rounded off to whole number at the nearest decimal.

  18. Percentage of Participants With Severity Based on HRQOL: Patient Global Impression of Severity (PGI-S) [ Time Frame: Cycle 2 Day 1 and Cycle 3 Day 1 ]
    The anchor-based questionnaire PGI-S contains the following 3 items (the severity of the physical functioning decline over the past week, the severity of the appetite decrease over the past week, and the severity of the pain over the past week). The PGI-S was measured using the possible outcomes none, mild, moderate, severe, and very severe. Percentages are rounded off to whole number at the nearest decimal.

  19. Percentage of Participants With Each EuroQol 5 Dimensions 5 Levels (EQ-5D-5L) Dimension Response [ Time Frame: Cycle 3 Day 1 ]
    The EQ-5D-5L assesses general health-related quality of life. Health is defined in 5 dimensions: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension has 5 levels: no problems, slight problems, moderate problems, severe problems, and extreme problems. Percentages are rounded off to whole number at the nearest decimal.

  20. Change From Baseline in EQ-5D-5L Visual Analogue Scale (EQ-5D-5L VAS) Score [ Time Frame: Cycle 3 Day 1 ]
    The EQ visual analogue scale records the patient's self-rated health on a vertical visual analogue scale, where the endpoints are labelled "The best health you can imagine" and "The worst health you can imagine." Responses are marked on a 0-100 scale with higher scores indicating higher health-related quality of life (i.e., better outcome).

  21. Maximum Observed Plasma Concentration (Cmax) of AG-120 [ Time Frame: Post-dose Cycle 1 Day 1 and Cycle 2 Day 1 (each cycle = 28 days) ]
    Crossover C1D1 and crossover C2D1 visits were combined with C1D1 and C2D1 visits, respectively for the analysis of this endpoint.

  22. Time to Reach Maximal Plasma Concentration (Tmax) of AG-120 [ Time Frame: Post-dose Cycle 1 Day 1 and Cycle 2 Day 1 (each cycle = 28 days) ]
    Crossover C1D1 and crossover C2D1 visits were combined with C1D1 and C2D1 visits, respectively for the analysis of this endpoint.

  23. Area Under the Plasma Concentration-time Curve From Time Zero to 24 Hours (AUC0-24) [ Time Frame: Post-dose of Cycle 2 Day 1 (each cycle = 28 days) ]
    Crossover C2D1 visit was combined with C2D1 visit for the analysis of this endpoint.

  24. Area Under the Plasma Concentration-time Curve From Time Zero to 4 Hours (AUC0-4) [ Time Frame: Post-dose of Cycle 1 Day 1 and Cycle 2 Day 1 (each cycle = 28 days) ]
    Crossover C1D1 and crossover C2D1 visits were combined with C1D1 and C2D1 visits, respectively for the analysis of this endpoint.

  25. Accumulation Ratio Based on AUC0-4 (Racc AUC0-4) [ Time Frame: Post-dose Cycle 2 Day 1 (each cycle = 28 days) ]
    Crossover C2D1 visit was combined with C2D1 visit for the analysis of this endpoint.

  26. Accumulation Ratio Based on Cmax (Racc Cmax) [ Time Frame: Post-dose Cycle 2 Day 1 (each cycle = 28 days) ]
    Crossover C2D1 visit was combined with C2D1 visit for the analysis of this endpoint.

  27. Plasma 2-hydroxyglutarate (2-HG) Levels of AG-120: B (Baseline Effect Value) [ Time Frame: Post-dose Cycle 1 Day 1 and Cycle 2 Day 1 (each cycle = 28 days) ]
    B is the Baseline Effect Value. Crossover C1D1 and crossover C2D1 visits were combined with C1D1 and C2D1 visits, respectively for the analysis of this endpoint.

  28. Plasma 2-hydroxyglutarate (2-HG) Levels of AG-120: AUEC0-4 [ Time Frame: Post-dose Cycle 1 Day 1 and Cycle 2 Day 1 (each cycle = 28 days) ]
    AUEC0-4 is the area of the response curve from time point zero (predose) up to 4 hr postdose. Crossover C1D1 and crossover C2D1 visits were combined with C1D1 and C2D1 visits, respectively for the analysis of this endpoint.

  29. Plasma 2-hydroxyglutarate (2-HG) Levels of AG-120: %BAUEC0-4 [ Time Frame: Post-dose Cycle 1 Day 1 and Cycle 2 Day 1 (each cycle = 28 days) ]
    %BAUEC0-4 is the percent inhibition for AUEC0-4. Crossover C1D1 and crossover C2D1 visits were combined with C1D1 and C2D1 visits, respectively for the analysis of this endpoint.

  30. Plasma 2-hydroxyglutarate (2-HG) Levels of AG-120: Rtrough [ Time Frame: Post-dose Cycle 2 Day 1 (each cycle = 28 days) ]
    Rtrough is the observed response value at the end of a dosing interval. Crossover C2D1 visit was combined with C2D1 visit for the analysis of this endpoint.

  31. Plasma 2-hydroxyglutarate (2-HG) Levels of AG-120: %BRtrough [ Time Frame: Post-dose Cycle 2 Day 1 (each cycle = 28 days) ]
    %BRtrough is the percent inhibition for Rtrough. Crossover C2D1 visit was combined with C2D1 visit for the analysis of this endpoint.



Information from the National Library of Medicine

Choosing to participate in a study is an important personal decision. Talk with your doctor and family members or friends about deciding to join a study. To learn more about this study, you or your doctor may contact the study research staff using the contacts provided below. For general information, Learn About Clinical Studies.


Layout table for eligibility information
Ages Eligible for Study:   18 Years and older   (Adult, Older Adult)
Sexes Eligible for Study:   All
Accepts Healthy Volunteers:   No
Criteria

Inclusion Criteria:

  1. Be ≥18 years of age.
  2. Have a histopathological diagnosis (fresh or banked tumor biopsy sample, preferably collected within the last 3 years) of nonresectable or metastatic cholangiocarcinoma and are not eligible for curative resection, transplantation, or ablative therapies.
  3. Have documented IDH1 gene-mutated disease (from a fresh tumor biopsy or the most recent banked tumor tissue available) based on central laboratory testing (R132C/L/G/H/S mutation variants tested).
  4. Have an Eastern Cooperative Oncology Group performance status (ECOG PS) score of 0 or 1
  5. Have an expected survival of ≥3 months.
  6. Have at least one evaluable and measurable lesion as defined by Response Evaluation Criteria in Solid Tumors (RECIST) v1.1. Participants who have received prior local therapy (including but not limited to embolization, chemoembolization, radiofrequency ablation, or radiation therapy) are eligible provided measurable disease falls outside of the treatment field or within the field and has shown ≥20% growth in size since post-treatment assessment.
  7. Have documented disease progression following at least 1 and no more than 2 prior systemic regimens for advanced disease (nonresectable or metastatic). Participants must have received at least 1 gemcitabine- or 5-FU-containing regimen for advanced cholangiocarcinoma. Participants who have received systemic adjuvant chemotherapy will be permitted provided there is documented disease progression during or within 6 months of completing the therapy.

Exclusion criteria:

  1. Received a prior IDH inhibitor.
  2. Received systemic anticancer therapy or an investigational agent <2 weeks prior to Day 1 (washout from prior immune based anticancer therapy is 4 weeks). In addition, the first dose of study treatment should not occur before a period ≥5 half-lives of the investigational agent has elapsed.
  3. Received radiotherapy to metastatic sites of disease <2 weeks prior to Day 1.
  4. Underwent hepatic radiation, chemoembolization, and radiofrequency ablation <4 weeks prior to Day 1.
  5. Have known symptomatic brain metastases requiring steroids. Participants with previously diagnosed brain metastases are eligible if they have completed their treatment and have recovered from the acute effects of radiation therapy or surgery prior to study entry, have discontinued corticosteroid treatment for these metastases for at least 4 weeks and have radiographically stable disease for at least 3 months prior to study entry. Note: up to 10 mg per day of prednisone equivalent will be allowed.

Information from the National Library of Medicine

To learn more about this study, you or your doctor may contact the study research staff using the contact information provided by the sponsor.

Please refer to this study by its ClinicalTrials.gov identifier (NCT number): NCT02989857


Locations
Show Show 49 study locations
Sponsors and Collaborators
Institut de Recherches Internationales Servier
Investigators
Layout table for investigator information
Study Chair: Medical Affairs Servier Pharmaceuticals LLC Servier Pharmaceuticals, LLC
  Study Documents (Full-Text)

Documents provided by Servier ( Institut de Recherches Internationales Servier ):
Study Protocol: Protocol v6  [PDF] March 5, 2019
Study Protocol: Protocol v7  [PDF] June 23, 2021
Statistical Analysis Plan  [PDF] April 1, 2019

Study Data/Documents: Individual Participant Data Set  This link exits the ClinicalTrials.gov site

Publications automatically indexed to this study by ClinicalTrials.gov Identifier (NCT Number):
Layout table for additonal information
Responsible Party: Institut de Recherches Internationales Servier
ClinicalTrials.gov Identifier: NCT02989857    
Other Study ID Numbers: AG120-C-005
First Posted: December 12, 2016    Key Record Dates
Results First Posted: April 13, 2022
Last Update Posted: January 17, 2023
Last Verified: December 2022
Individual Participant Data (IPD) Sharing Statement:
Plan to Share IPD: Yes
Plan Description:

Qualified scientific and medical researchers can request access to anonymized patient-level and study-level clinical trial data.

Access can be requested for all interventional clinical studies:

  • used for Marketing Authorization (MA) of medicines and new indications approved after 1 January 2014 in the European Economic Area (EEA) or the United States (US).
  • where Servier is the Marketing Authorization Holder (MAH). The date of the first MA of the new medicine (or the new indication) in one of the EEA Member States will be considered for this scope.

In addition, access can be requested for all interventional clinical studies in patients:

  • sponsored by Servier
  • with a first patient enrolled as of 1 January 2004 onwards
  • for New Chemical Entity or New Biological Entity (new pharmaceutical form excluded) for which development has been terminated before any Marketing authorization (MA) approval.
Supporting Materials: Study Protocol
Statistical Analysis Plan (SAP)
Informed Consent Form (ICF)
Time Frame: After Marketing Authorisation in EEA or US if the study is used for the approval.
Access Criteria: Researchers should register on Servier Data Portal and fill in the research proposal form. This form in four parts should be fully documented. The Research Proposal Form will not be reviewed until all mandatory fields are completed.
URL: https://clinicaltrials.servier.com/

Layout table for additional information
Studies a U.S. FDA-regulated Drug Product: Yes
Studies a U.S. FDA-regulated Device Product: No
Keywords provided by Servier ( Institut de Recherches Internationales Servier ):
IDH1
Additional relevant MeSH terms:
Layout table for MeSH terms
Cholangiocarcinoma
Adenocarcinoma
Carcinoma
Neoplasms, Glandular and Epithelial
Neoplasms by Histologic Type
Neoplasms
Ivosidenib
Antineoplastic Agents
Enzyme Inhibitors
Molecular Mechanisms of Pharmacological Action