search
Back to results

Personalized Medicine for Advanced Biliary Cancer Patients (SAFIR-ABC10)

Primary Purpose

Biliary Tract Neoplasms

Status
Not yet recruiting
Phase
Phase 3
Locations
International
Study Type
Interventional
Intervention
Futibatinib
Ivosidenib
Zanidatamab
Trastuzumab
Neratinib
Encorafenib
Binimetinib
Niraparib
Cisplatin
Gemcitabine
Sponsored by
UNICANCER
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Biliary Tract Neoplasms focused on measuring Biliary Tract Neoplasms, Targeted therapy, Personalised medicine

Eligibility Criteria

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

SCREENING PHASE Inclusion Criteria: Signed a written informed consent form prior to any trial specific procedures (Consent #1) Histologically-proven intrahepatic, perihilar or distal cholangiocarcinoma, or gallbladder carcinoma (ampullary carcinoma excluded) De novo or recurrent, locally advanced (non-resectable) or metastatic disease Availability of a suitable archived sample of primary or metastatic tumour tissue (frozen, or FFPE) or able to undergo a biopsy to obtain a suitable malignant tissue sample Aged ≥18 years Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 Estimated life expectancy >3 months Candidate for standard of care first line (1L-SoC) therapy, or has initiated first cycle of 1L-SoC therapy Affiliated to a social security system or in possession of equivalent private health insurance (according to local country health provision arrangements). Exclusion Criteria: Contraindication to 1L-SoC Patients who are candidates for locoregional therapy Contraindication to tumour biopsy in the absence of suitable archived sample of tumour tissue Prior anticancer therapy in the palliative setting. Adjuvant capecitabine allowed if completed ≥ 183 days prior to study entry Received more than 1 cycle of treatment with 1L-SoC Prior treatment with any of the molecular targeted therapies (MTT) under investigation in the SAFIR-ABC10 study Current malignancies (other than advanced biliary cancer), with the exception of adequately treated cone-biopsied in situ carcinoma of the cervix uteri and basal or squamous cell carcinoma of the skin. Cancer survivors, who have undergone potentially curative therapy for a prior malignancy, have no evidence of that disease for 5 years or more and are deemed at negligible risk for recurrence, are eligible for the trial Any condition which in the Investigator's opinion makes it undesirable for the subject to participate in the trial or which would jeopardize compliance with the protocol Patients unwilling or unable to comply with the medical follow-up required by the trial because of geographic, familial, social, or psychological reasons Individuals deprived of liberty or placed under protective custody or guardianship RANDOMISED TRIAL Inclusion Criteria: Signed a written informed consent form prior to any trial specific procedures (Consent #2) Molecular profile showing the tumour harbours at least one targetable molecular alteration with a MTT in the study portfolio (as determined by the trial MTB) Disease control (stable or responsive) after 4 cycles of 1L-SoC, compared to a pre-treatment disease evaluation, as assessed by the investigator ECOG performance status of 0 or 1 Presence of at least one evaluable lesion according to RECIST v1.1, or complete response to 12 weeks 1L-SoC Adequate bone marrow function: absolute neutrophil count (ANC) ≥1.5 × 10⁹/L, platelet count ≥100 × 10⁹/L, and haemoglobin ≥9 g/dL Adequate liver function: total bilirubin level ≤1.5 × the upper limit of normal (ULN) range unless the patient has documented Gilbert syndrome, and aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels ≤2.5 × ULN (AST and ALT ≤5 ULN when documented tumour liver involvement) Adequate renal function: estimated creatinine clearance ≥45 mL/min according to the Cockcroft-Gault formula Adequate cardiac function: left ventricular ejection fraction ≥50% at baseline as determined by either echocardiogram or multigated acquisition scan (MUGA) Adequate biliary drainage, with no evidence of ongoing infection Men, and women of childbearing potential (WOCBP) must agree to use adequate contraception for the duration of trial participation and as required after completing study treatment. Men must also agree to not donate sperm and women must agree to not donate oocytes during the specified period. Women of childbearing potential must have a negative serum pregnancy test performed within 3 days before the date of randomisation Willing and able to comply with the protocol for the duration of the study including scheduled visits, treatment plan, laboratory tests, and other study procedures Affiliated to a social security system or in possession of equivalent private health insurance (according to local country health provision arrangements) Exclusion Criteria: Disease progression occurring at any time prior randomisation, or toxicity that led to the discontinuation of the 1L-SoC before 4 full cycles have been delivered Toxicities from 1L-SoC not resolved to Grade ≤ 2 (according to version 5.0 the National Cancer Institute - Common terminology criteria for adverse events [NCI-CTCAE v5.0]) before randomisation, with the exception of alopecia Contraindication or known hypersensitivity to the MTT for the molecular alteration found in the patient, or any component in their formulation Note: For patients with multiple target alterations, contraindication to one MTT will not warrant exclusion if MTT to an alternative target is feasible. Microsatellite instability high (MSI-H) or mismatch repair deficient (dMMR) cancers Major surgery within 4 weeks of randomisation Untreated central nervous system (CNS) metastases, symptomatic CNS metastases, or radiation treatment for CNS metastases within 4 weeks of start of study treatment. Stable, treated brain metastases are allowed (defined as subjects who are off steroids and anticonvulsants and are neurologically stable with no evidence of radiographic progression for at least 4 weeks at the time of screening). Known leptomeningeal disease. If leptomeningeal disease has been reported radiographically on baseline magnetic resonance imaging (MRI), but is not suspected clinically by the investigator, the subject must be free of neurological symptoms. Concurrent malignancy (other than ABC), with the exception of adequately treated cone-biopsied in situ carcinoma of the cervix uteri and basal or squamous cell carcinoma of the skin. Cancer survivors, who have undergone potentially curative therapy for a prior malignancy, have no evidence of that disease for 5 years or more and are deemed at negligible risk for recurrence, are eligible for the trial Known active hepatitis B virus or hepatitis C virus infection or human immunodeficiency virus (HIV) or known acquired immunodeficiency syndrome Any condition which in the Investigator's opinion makes it undesirable for the subject to participate in the trial or which would jeopardize compliance with the protocol Women who are pregnant or breast-feeding Participation in another therapeutic trial within the 30 days prior to entering the study. Participation in an observational trial would be acceptable Patients unwilling or unable to comply with the medical follow-up required by the trial because of geographic, familial, social, or psychological reasons Individuals deprived of liberty or placed under protective custody or guardianship ADDITIONAL EXCLUSION CRITERIA FOR SPECIFIC MTTs: Patients assigned to receive oral therapies: Inability or unwillingness to swallow pills History of malabsorption syndrome or other condition that would interfere with enteral absorption. For example, active intestine inflammation (e.g., Crohn's disease or ulcerative colitis) requiring immunosuppressive therapy Futibatinib: 1. History and/or current evidence of any of the following disorders: Non-tumour related alteration of the calcium-phosphorus homeostasis that is considered clinically significant in the opinion of the Investigator Ectopic mineralization/calcification, including but not limited to soft tissue, kidneys, intestine, or myocardia and lung, considered clinically significant in the opinion of the Investigator Retinal or corneal disorder confirmed by retinal/corneal examination and considered clinically significant in the opinion of the Investigator Ivosidenib: Patients with history of torsade de pointes Concomitant treatment with digoxin where this cannot be substituted for another therapy Patients with a heart-rate corrected QT interval (using Fridericia's formula) (QTcF) ≥ 450 msec or other factors that increased the risk of QT prolongation or arrhythmic events (e.g. heart failure, hypokalemia, family history of long QT interval syndrome) Zanidatamab: Treatment with anthracyclines within 90 days before first dose of zanidatamab and/or total lifetime load exceeding 360 mg/m2 Adriamycin® or equivalent Use of corticosteroids administered at doses equivalent to > 15 mg per day of prednisone within 2 weeks of first zanidatamab dosing unless otherwise approved by the coordinating investigator. Topical, ocular, intra-articular, intranasal, and/or inhalational corticosteroids are permitted QTcF > 470 ms History of myocardial infarction or unstable angina within 6 months prior to enrollment, troponin levels consistent with myocardial infarction, or clinically significant cardiac disease, such as ventricular arrhythmia requiring therapy, uncontrolled hypertension, or any history of symptomatic congestive heart failure Acute or chronic uncontrolled pancreatitis or Child-Pugh Class C liver disease Clinically significant infiltrative pulmonary disease not related to lung metastases A history of life-threatening hypersensitivity to monoclonal antibodies or recombinant proteins Neratinib & trastuzumab: Patients with severe hepatic impairment (Child-Pugh Class C) Co-administration with the following medical products that are strong inducers of the CYP3A4/P-gp isoform of cytochrome P450, such as carbamazepine, phenytoin (antiepileptics), St John's wort (Hypericum perforatum) or rifampicin (antimycobacterial) Patients who are experiencing dyspnoea at rest due to complications of advanced malignancy or co-morbidities Hypersensitivity to murine proteins Encorafenib & binimetinib: Patients with a history or current evidence of retinal vein occlusion or risk factors for retinal vein occlusion (e.g., uncontrolled glaucoma or history of hyperviscosity or hypercoagulability syndrome) Patients with concurrent neuromuscular disorders associated with elevated cytokinin Clinically significant cardiovascular disease (recent acute myocardial infarction, treated congestive heart failure [2 or above on the New York Heart Association functional classification scale], recent thromboembolic or cerebrovascular events [within 12 weeks, excepted if related to indwelling catheter], known prolonged QT syndrome) Current or expected use of a strong inhibitor of CYP3A4 Niraparib: Hypertension that is inadequately treated or controlled Participants receiving corticosteroids who have not been on a stable dose for at least 4 weeks prior to niraparib History of Myelodysplastic Syndrome/Acute Myeloid Leukemia History of Reversible Encephalopathy Syndrome Current active pneumonitis within 90 days of receiving niraparib or a known history of interstitial lung disease, drug-related pneumonitis or radiation pneumonitis requiring steroid treatment Increased bleeding risk due to concurrent conditions Receipt of a live vaccine within 30 days of planned start of therapy Radiation encompassing >20% of bone marrow within 2 weeks ; or any radiation therapy within 1 week prior to receiving niraparib

Sites / Locations

  • Cliniques universitaires Saint-Luc
  • CHU Amiens Picardie
  • CHU d'Angers
  • Institut du Cancer Avignon Provence
  • CHU de Besançon
  • CHU de Bordeaux - Hôpital Haut-Leveque
  • Centre François Baclesse
  • Centre Jean Perrin
  • CHU Estaing de Clermont Ferrand
  • APHP - Hopital Henri Mondor
  • CHU de Dijon
  • CHU Grenoble Alpes
  • Centre Oscar Lambret
  • CHU Lille
  • CHU Dupuytren
  • Centre Leon Bérard
  • CHU de Lyon
  • Clinique Privée Jean Mermoz
  • APHM - CHU La Timone
  • Institut Paoli Calmettes
  • CHU Montpellier
  • Institut de Cancer de Montpellier
  • CHU Nantes - Hôtel Dieu
  • Centre Antoine Lacassagne
  • APHP - Hôpital Beaujon
  • APHP - Hôpital Cochin
  • APHP - Hôpital Saint Antoine
  • Groupe Hospitalier Diaconesses Croix Saint-Simon
  • Institute Mutualiste Montsouris
  • Hôpital Privé des Côtes d'Armor
  • CHU Poitiers
  • CHU de Reims
  • Institut Jean Godinot
  • Centre Eugène Marquis
  • CHU Charles Nicolle
  • Institut de Cancerologie de l'Ouest
  • Hôpital Foch
  • CHU Toulouse
  • CHRU de Nancy
  • APHP - Hôpital Paul Brousse
  • Gustave Roussy
  • Queen Elizabeth Hospital
  • Bristol
  • Addenbrooke's Hospital
  • Castle Hill Hospital
  • St James's Hospital
  • Clatterbridge Centre for Oncology
  • Guy's & St Thomas' Hospital
  • Hammersmith Hospital
  • Royal Marsden Hospital
  • University College London
  • Maidstone Hospital
  • The Christie Hospital
  • Nottingham University Hospital
  • Oxford
  • Sheffield
  • Southampton General Hospital

Arms of the Study

Arm 1

Arm 2

Arm Type

Experimental

Active Comparator

Arm Label

Experimental

Control

Arm Description

Molecular targeted therapy matched to genetic alteration carried by the tumour

Continued standard of care treatment for first-line biliary tract cancer

Outcomes

Primary Outcome Measures

Progression-free survival (PFS)
Time from randomisation to the first documented progression of disease (PD) as assessed by the investigator according to RECIST v1.1, or death from any cause, whichever occurs first.

Secondary Outcome Measures

Overall Survival (OS)
The overall survival is the length of time from randomization that patients enrolled in the study are still alive.
Objective response rate
Objective response rate is defined as the proportion of patients achieving complete response (CR) or partial response (PR) (according to RECIST v1.1). Objective response rate will be presented as the best response achieved compared to the disease assessment performed at randomisation.
Time to treatment failure
Time from patient starting their allocated treatment to the date at which a patient first experiences a treatment failure event. The following will be considered as treatment failure events: early treatment discontinuation (regardless of reason), disease progression, death, starting a new treatment after completing scheduled treatment, withdrawal from the study due to any reason or loss to follow-up.
Progression-free survival after next line of treatment (PFS2)
Time from randomisation to the date of second disease progression or death, whichever occurs first.
Duration of response
Duration of response is defined as the time from first documented response (compared to baseline measurement taken at randomisation) until the date of disease progression, as assessed by the investigator according to RECIST v1.1, or death from any cause whichever occurs first.
Disease control rate
Disease control rate is defined as the proportion of randomised patients achieving CR, PR, stable disease (SD)/no evidence of disease (NED) as assessed by the investigator according to RECIST v1.1.
Percentage change in tumour size
Taking the measurements at randomisation as the reference.

Full Information

First Posted
October 28, 2022
Last Updated
August 28, 2023
Sponsor
UNICANCER
Collaborators
Cancer Research UK & UCL Cancer Trials Centre, Belgian Group of Digestive Oncology, National Cancer Institute, France, Cancer Research UK, Taiho Oncology, Inc., Servier, Zymeworks Inc., Accord Healthcare, Inc., Pierre Fabre Medicament, GlaxoSmithKline Research & Development Limited
search

1. Study Identification

Unique Protocol Identification Number
NCT05615818
Brief Title
Personalized Medicine for Advanced Biliary Cancer Patients
Acronym
SAFIR-ABC10
Official Title
Molecular Targeted Maintenance Therapy Versus Standard of Care in Advanced Biliary Cancer: an International, Randomised, Controlled, Open-label, Platform Phase 3 Trial
Study Type
Interventional

2. Study Status

Record Verification Date
August 2023
Overall Recruitment Status
Not yet recruiting
Study Start Date
October 2023 (Anticipated)
Primary Completion Date
June 2027 (Anticipated)
Study Completion Date
June 2028 (Anticipated)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
UNICANCER
Collaborators
Cancer Research UK & UCL Cancer Trials Centre, Belgian Group of Digestive Oncology, National Cancer Institute, France, Cancer Research UK, Taiho Oncology, Inc., Servier, Zymeworks Inc., Accord Healthcare, Inc., Pierre Fabre Medicament, GlaxoSmithKline Research & Development Limited

4. Oversight

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

5. Study Description

Brief Summary
The object of this trial is to evaluate whether the introduction of a targeted therapy after 4 cycles of the current standard-of-care treatment for advanced biliary cancer is superior to continuing with the standard treatment. The trial is composed of two phases: (i) An initial screening phase to identify a suitable patient population, during which a molecular profile of the patient's tumour will be obtained, and (ii) a randomised comparative trial in which patients with disease control after 4 cycles of standard treatment, and whose tumour harbours a targetable molecular alteration, will be randomised (2:1) to receive either a matched targeted therapy or to continue with the standard treatment.
Detailed Description
This is a Phase 3, multicentre, randomised, open-label trial to evaluate whether the introduction of molecular targeted therapy (MTT) as maintenance after 4 cycles of standard-of-care first-line systemic therapy (1L SoC) is superior to continuation of 1L-SoC in the treatment of patients with ABC. The trial is composed of two phases: (i) An initial screening phase to identify a suitable patient population, and (ii) a randomised comparative trial. The aim of the screening phase is to identify a medically suitable population, to obtain a molecular profile of the patient's tumour, to collect baseline data concerning patient demographics and disease characteristics and to obtain pre-treatment blood and tumour samples for further translational research. A genetic profile will be obtained from tumour-derived DNA and RNA samples by next-generation sequencing and from circulating tumour DNA. The trial Molecular Tumour Board will determine whether each patient harbours a targetable molecular alteration for one or more of the trial MTTs. Patients with disease control after 4 cycles of 1L-SoC, who did not experience limiting toxicity, and whose tumour harbours at least one targetable molecular alteration, will be invited to participate in the randomised phase of the trial in which 159 eligible patients will be randomised (2:1) to receive either maintenance therapy with a matched MTT or to continue 1L-SoC treatment.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Biliary Tract Neoplasms
Keywords
Biliary Tract Neoplasms, Targeted therapy, Personalised medicine

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 3
Interventional Study Model
Parallel Assignment
Masking
None (Open Label)
Allocation
Randomized
Enrollment
800 (Anticipated)

8. Arms, Groups, and Interventions

Arm Title
Experimental
Arm Type
Experimental
Arm Description
Molecular targeted therapy matched to genetic alteration carried by the tumour
Arm Title
Control
Arm Type
Active Comparator
Arm Description
Continued standard of care treatment for first-line biliary tract cancer
Intervention Type
Drug
Intervention Name(s)
Futibatinib
Intervention Description
Dose 20 mg once a day (QD)
Intervention Type
Drug
Intervention Name(s)
Ivosidenib
Other Intervention Name(s)
Tibsovo
Intervention Description
Dose 500 mg QD
Intervention Type
Drug
Intervention Name(s)
Zanidatamab
Intervention Description
Dose: Patients < 70 kg: 1800 mg every 3 weeks (Q3W), Patients ≥ 70 kg: 2400 mg Q3W
Intervention Type
Drug
Intervention Name(s)
Trastuzumab
Other Intervention Name(s)
Zercepac
Intervention Description
Loading dose 8 mg/kg, then 6 mg/kg Q3W (Combination with neratinib)
Intervention Type
Drug
Intervention Name(s)
Neratinib
Other Intervention Name(s)
Nerlynx
Intervention Description
Dose: 240 mg QD (combination with trastuzumab)
Intervention Type
Drug
Intervention Name(s)
Encorafenib
Other Intervention Name(s)
Braftovi
Intervention Description
Dose: 450 mg QD (Combination with binimetinib)
Intervention Type
Drug
Intervention Name(s)
Binimetinib
Other Intervention Name(s)
Mektovi
Intervention Description
Dose: 45 mg twice a day (BID) (Combination with encorafenib)
Intervention Type
Drug
Intervention Name(s)
Niraparib
Other Intervention Name(s)
Zejula
Intervention Description
Dose: 200 mg QD or 300 mg QD
Intervention Type
Drug
Intervention Name(s)
Cisplatin
Intervention Description
Dose: 25 mg/m2 IV on days 1 and 8 Q3W (CISGEM)
Intervention Type
Drug
Intervention Name(s)
Gemcitabine
Intervention Description
Dose: 1000 mg/m2 IV on days 1 and 8 Q3W (CISGEM)
Primary Outcome Measure Information:
Title
Progression-free survival (PFS)
Description
Time from randomisation to the first documented progression of disease (PD) as assessed by the investigator according to RECIST v1.1, or death from any cause, whichever occurs first.
Time Frame
From randomisation to disease progression or death, up to 5 years.
Secondary Outcome Measure Information:
Title
Overall Survival (OS)
Description
The overall survival is the length of time from randomization that patients enrolled in the study are still alive.
Time Frame
From randomisation to death, up to 5 years.
Title
Objective response rate
Description
Objective response rate is defined as the proportion of patients achieving complete response (CR) or partial response (PR) (according to RECIST v1.1). Objective response rate will be presented as the best response achieved compared to the disease assessment performed at randomisation.
Time Frame
From randomisation, up to 5 years.
Title
Time to treatment failure
Description
Time from patient starting their allocated treatment to the date at which a patient first experiences a treatment failure event. The following will be considered as treatment failure events: early treatment discontinuation (regardless of reason), disease progression, death, starting a new treatment after completing scheduled treatment, withdrawal from the study due to any reason or loss to follow-up.
Time Frame
From randomisation to treatment failure event, up to 5 years.
Title
Progression-free survival after next line of treatment (PFS2)
Description
Time from randomisation to the date of second disease progression or death, whichever occurs first.
Time Frame
From randomisation to second disease progression or death, up to 5 years.
Title
Duration of response
Description
Duration of response is defined as the time from first documented response (compared to baseline measurement taken at randomisation) until the date of disease progression, as assessed by the investigator according to RECIST v1.1, or death from any cause whichever occurs first.
Time Frame
From response to disease progression or death, up to 5 years.
Title
Disease control rate
Description
Disease control rate is defined as the proportion of randomised patients achieving CR, PR, stable disease (SD)/no evidence of disease (NED) as assessed by the investigator according to RECIST v1.1.
Time Frame
From randomisation, up to 5 years.
Title
Percentage change in tumour size
Description
Taking the measurements at randomisation as the reference.
Time Frame
From randomisation, up to 5 years.
Other Pre-specified Outcome Measures:
Title
Feasibility of molecular screening
Description
The proportion of patients with an available MTB proposition at the time of the 3-month standard of care treatment evaluation.
Time Frame
Up to 3 months from start of treatment
Title
Quality of life questionnaire - Core 30 (QLQ-C30)
Description
Developed by the EORTC, this self-reported questionnaire assesses the health-related quality of life of cancer patients in clinical trials. The questionnaire includes five functional scales (physical, everyday activity, cognitive, emotional, and social), three symptom scales (fatigue, pain, nausea and vomiting), a health/quality of life overall scale, and a number of additional elements assessing common symptoms (including dyspnea, loss of appetite, insomnia, constipation, and diarrhea), as well as, the perceived financial impact of the disease. All of the scales and single-item measures range in score from 0 to 100. A high scale score represents a higher response level.
Time Frame
From baseline, every 3 cycles (every 9 weeks) until end of treatment, an average of 1 year
Title
Quality of life Questionnaire - Biliary tract cancer module (QLQ-BIL21)
Description
This EORTC cholangiocarcinoma and gallbladder cancer specific questionnaire is intended to supplement the QLQ-C30. The QLQ-BIL21 contains 21 items to assess symptoms. All items are rated on a four-point Likert-type scale (1 = "not at all", 2 = "a little", 3 = "quite a bit", and 4 = "very much"), and are linearly transformed to a 0-100 scale.
Time Frame
From baseline, every 3 cycles (every 9 weeks) until end of treatment, an average of 1 year
Title
EuroQOL EQ-5D-5L questionnaire
Description
Developed by the EuroQol group, the self-reported questionnaire assesses the health-related quality of life of cancer patients in clinical trials consists of a descriptive system and a visual analogue scale (VAS). The EQ-5D-5L descriptive system comprises five dimensions (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression), each dimension has 5 levels (1 = "no problems", 2 = "slight problems", 3 = "moderate problems", 4 = "severe problems", and 5 = "extreme problems"). This questionnaire provide a 5-digit score which generate a health state profile. The VAS records the patient's self-rated health on a vertical visual analogue scale where the score range from 0 (The best health you can imagine) to 100 (The worst health you can imagine). The VAS is used as a quantitative measure of health outcome that reflects the patient's own judgement.
Time Frame
From baseline, every 3 cycles (every 9 weeks) until end of treatment, an average of 1 year
Title
Incidence of Adverse Events
Description
Safety and tolerability of the treatment will be evaluated using the National Cancer Institute-Common Terminology Criteria for Adverse Events version 5 (NCI-CTCAE v5). NCI-CTCAE is widely accepted in the community of oncology research as the leading rating scale for adverse events. This scale, divided into 5 grades (1 = "mild", 2 = "moderate", 3 = "severe", 4 = "life-threatening", and 5 = "death") determined by the investigator, will make it possible to assess the severity of the disorders.
Time Frame
From randomisation, up to 5 years

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
SCREENING PHASE Inclusion Criteria: Signed a written informed consent form prior to any trial specific procedures (Consent #1) Histologically-proven intrahepatic, perihilar or distal cholangiocarcinoma, or gallbladder carcinoma (ampullary carcinoma excluded) De novo or recurrent, locally advanced (non-resectable) or metastatic disease Availability of a suitable archived sample of primary or metastatic tumour tissue (frozen, or FFPE) or able to undergo a biopsy to obtain a suitable malignant tissue sample Aged ≥18 years Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 Estimated life expectancy >3 months Candidate for standard of care first line (1L-SoC) therapy, or has initiated first cycle of 1L-SoC therapy Affiliated to a social security system or in possession of equivalent private health insurance (according to local country health provision arrangements). Exclusion Criteria: Contraindication to 1L-SoC Patients who are candidates for locoregional therapy Contraindication to tumour biopsy in the absence of suitable archived sample of tumour tissue Prior anticancer therapy in the palliative setting. Adjuvant capecitabine allowed if completed ≥ 183 days prior to study entry Received more than 1 cycle of treatment with 1L-SoC Prior treatment with any of the molecular targeted therapies (MTT) under investigation in the SAFIR-ABC10 study Current malignancies (other than advanced biliary cancer), with the exception of adequately treated cone-biopsied in situ carcinoma of the cervix uteri and basal or squamous cell carcinoma of the skin. Cancer survivors, who have undergone potentially curative therapy for a prior malignancy, have no evidence of that disease for 5 years or more and are deemed at negligible risk for recurrence, are eligible for the trial Any condition which in the Investigator's opinion makes it undesirable for the subject to participate in the trial or which would jeopardize compliance with the protocol Patients unwilling or unable to comply with the medical follow-up required by the trial because of geographic, familial, social, or psychological reasons Individuals deprived of liberty or placed under protective custody or guardianship RANDOMISED TRIAL Inclusion Criteria: Signed a written informed consent form prior to any trial specific procedures (Consent #2) Molecular profile showing the tumour harbours at least one targetable molecular alteration with a MTT in the study portfolio (as determined by the trial MTB) Disease control (stable or responsive) after 4 cycles of 1L-SoC, compared to a pre-treatment disease evaluation, as assessed by the investigator ECOG performance status of 0 or 1 Presence of at least one evaluable lesion according to RECIST v1.1, or complete response to 12 weeks 1L-SoC Adequate bone marrow function: absolute neutrophil count (ANC) ≥1.5 × 10⁹/L, platelet count ≥100 × 10⁹/L, and haemoglobin ≥9 g/dL Adequate liver function: total bilirubin level ≤1.5 × the upper limit of normal (ULN) range unless the patient has documented Gilbert syndrome, and aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels ≤2.5 × ULN (AST and ALT ≤5 ULN when documented tumour liver involvement) Adequate renal function: estimated creatinine clearance ≥45 mL/min according to the Cockcroft-Gault formula Adequate cardiac function: left ventricular ejection fraction ≥50% at baseline as determined by either echocardiogram or multigated acquisition scan (MUGA) Adequate biliary drainage, with no evidence of ongoing infection Men, and women of childbearing potential (WOCBP) must agree to use adequate contraception for the duration of trial participation and as required after completing study treatment. Men must also agree to not donate sperm and women must agree to not donate oocytes during the specified period. Women of childbearing potential must have a negative serum pregnancy test performed within 3 days before the date of randomisation Willing and able to comply with the protocol for the duration of the study including scheduled visits, treatment plan, laboratory tests, and other study procedures Affiliated to a social security system or in possession of equivalent private health insurance (according to local country health provision arrangements) Exclusion Criteria: Disease progression occurring at any time prior randomisation, or toxicity that led to the discontinuation of the 1L-SoC before 4 full cycles have been delivered Toxicities from 1L-SoC not resolved to Grade ≤ 2 (according to version 5.0 the National Cancer Institute - Common terminology criteria for adverse events [NCI-CTCAE v5.0]) before randomisation, with the exception of alopecia Contraindication or known hypersensitivity to the MTT for the molecular alteration found in the patient, or any component in their formulation Note: For patients with multiple target alterations, contraindication to one MTT will not warrant exclusion if MTT to an alternative target is feasible. Microsatellite instability high (MSI-H) or mismatch repair deficient (dMMR) cancers Major surgery within 4 weeks of randomisation Untreated central nervous system (CNS) metastases, symptomatic CNS metastases, or radiation treatment for CNS metastases within 4 weeks of start of study treatment. Stable, treated brain metastases are allowed (defined as subjects who are off steroids and anticonvulsants and are neurologically stable with no evidence of radiographic progression for at least 4 weeks at the time of screening). Known leptomeningeal disease. If leptomeningeal disease has been reported radiographically on baseline magnetic resonance imaging (MRI), but is not suspected clinically by the investigator, the subject must be free of neurological symptoms. Concurrent malignancy (other than ABC), with the exception of adequately treated cone-biopsied in situ carcinoma of the cervix uteri and basal or squamous cell carcinoma of the skin. Cancer survivors, who have undergone potentially curative therapy for a prior malignancy, have no evidence of that disease for 5 years or more and are deemed at negligible risk for recurrence, are eligible for the trial Known active hepatitis B virus or hepatitis C virus infection or human immunodeficiency virus (HIV) or known acquired immunodeficiency syndrome Any condition which in the Investigator's opinion makes it undesirable for the subject to participate in the trial or which would jeopardize compliance with the protocol Women who are pregnant or breast-feeding Participation in another therapeutic trial within the 30 days prior to entering the study. Participation in an observational trial would be acceptable Patients unwilling or unable to comply with the medical follow-up required by the trial because of geographic, familial, social, or psychological reasons Individuals deprived of liberty or placed under protective custody or guardianship ADDITIONAL EXCLUSION CRITERIA FOR SPECIFIC MTTs: Patients assigned to receive oral therapies: Inability or unwillingness to swallow pills History of malabsorption syndrome or other condition that would interfere with enteral absorption. For example, active intestine inflammation (e.g., Crohn's disease or ulcerative colitis) requiring immunosuppressive therapy Futibatinib: 1. History and/or current evidence of any of the following disorders: Non-tumour related alteration of the calcium-phosphorus homeostasis that is considered clinically significant in the opinion of the Investigator Ectopic mineralization/calcification, including but not limited to soft tissue, kidneys, intestine, or myocardia and lung, considered clinically significant in the opinion of the Investigator Retinal or corneal disorder confirmed by retinal/corneal examination and considered clinically significant in the opinion of the Investigator Ivosidenib: Patients with history of torsade de pointes Concomitant treatment with digoxin where this cannot be substituted for another therapy Patients with a heart-rate corrected QT interval (using Fridericia's formula) (QTcF) ≥ 450 msec or other factors that increased the risk of QT prolongation or arrhythmic events (e.g. heart failure, hypokalemia, family history of long QT interval syndrome) Zanidatamab: Treatment with anthracyclines within 90 days before first dose of zanidatamab and/or total lifetime load exceeding 360 mg/m2 Adriamycin® or equivalent Use of corticosteroids administered at doses equivalent to > 15 mg per day of prednisone within 2 weeks of first zanidatamab dosing unless otherwise approved by the coordinating investigator. Topical, ocular, intra-articular, intranasal, and/or inhalational corticosteroids are permitted QTcF > 470 ms History of myocardial infarction or unstable angina within 6 months prior to enrollment, troponin levels consistent with myocardial infarction, or clinically significant cardiac disease, such as ventricular arrhythmia requiring therapy, uncontrolled hypertension, or any history of symptomatic congestive heart failure Acute or chronic uncontrolled pancreatitis or Child-Pugh Class C liver disease Clinically significant infiltrative pulmonary disease not related to lung metastases A history of life-threatening hypersensitivity to monoclonal antibodies or recombinant proteins Neratinib & trastuzumab: Patients with severe hepatic impairment (Child-Pugh Class C) Co-administration with the following medical products that are strong inducers of the CYP3A4/P-gp isoform of cytochrome P450, such as carbamazepine, phenytoin (antiepileptics), St John's wort (Hypericum perforatum) or rifampicin (antimycobacterial) Patients who are experiencing dyspnoea at rest due to complications of advanced malignancy or co-morbidities Hypersensitivity to murine proteins Encorafenib & binimetinib: Patients with a history or current evidence of retinal vein occlusion or risk factors for retinal vein occlusion (e.g., uncontrolled glaucoma or history of hyperviscosity or hypercoagulability syndrome) Patients with concurrent neuromuscular disorders associated with elevated cytokinin Clinically significant cardiovascular disease (recent acute myocardial infarction, treated congestive heart failure [2 or above on the New York Heart Association functional classification scale], recent thromboembolic or cerebrovascular events [within 12 weeks, excepted if related to indwelling catheter], known prolonged QT syndrome) Current or expected use of a strong inhibitor of CYP3A4 Niraparib: Hypertension that is inadequately treated or controlled Participants receiving corticosteroids who have not been on a stable dose for at least 4 weeks prior to niraparib History of Myelodysplastic Syndrome/Acute Myeloid Leukemia History of Reversible Encephalopathy Syndrome Current active pneumonitis within 90 days of receiving niraparib or a known history of interstitial lung disease, drug-related pneumonitis or radiation pneumonitis requiring steroid treatment Increased bleeding risk due to concurrent conditions Receipt of a live vaccine within 30 days of planned start of therapy Radiation encompassing >20% of bone marrow within 2 weeks ; or any radiation therapy within 1 week prior to receiving niraparib
Central Contact Person:
First Name & Middle Initial & Last Name or Official Title & Degree
Marta Jimenez
Phone
+33 (0) 1 44 23 55 58
Email
m-jimenez@unicancer.fr
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Malka David, MD
Organizational Affiliation
Institut Mutualiste Montsouris
Official's Role
Principal Investigator
First Name & Middle Initial & Last Name & Degree
Julien Edeline, MD
Organizational Affiliation
Centre Eugène Marquis
Official's Role
Principal Investigator
First Name & Middle Initial & Last Name & Degree
Ivan Borbath, MD
Organizational Affiliation
Cliniques universitaires Saint-Luc- Université Catholique de Louvain
Official's Role
Principal Investigator
First Name & Middle Initial & Last Name & Degree
John Bridgewater, MD
Organizational Affiliation
University College London Cancer Institute
Official's Role
Principal Investigator
First Name & Middle Initial & Last Name & Degree
Juan W Valle
Organizational Affiliation
University of Manchester and The Christie NHS Foundation Trust
Official's Role
Principal Investigator
Facility Information:
Facility Name
Cliniques universitaires Saint-Luc
City
Brussels
Country
Belgium
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Ivan BORBATH, MD
First Name & Middle Initial & Last Name & Degree
Ivan BORBATH, MD
Facility Name
CHU Amiens Picardie
City
Amiens
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Vincent HAUTEFEUILE, MD
First Name & Middle Initial & Last Name & Degree
Vincent HAUTEFEUILE, MD
Facility Name
CHU d'Angers
City
Angers
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Carole Vitellius
First Name & Middle Initial & Last Name & Degree
Carole Vitellius, MD
Facility Name
Institut du Cancer Avignon Provence
City
Avignon
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Clémence TOULLEC
First Name & Middle Initial & Last Name & Degree
Clémence TOULLEC, MD
Facility Name
CHU de Besançon
City
Besançon
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Christophe BORG
First Name & Middle Initial & Last Name & Degree
Christophe BORG, MD
Facility Name
CHU de Bordeaux - Hôpital Haut-Leveque
City
Bordeaux
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Jean-Frédéric BLANC
First Name & Middle Initial & Last Name & Degree
Jean-Frédéric BLANC, MD
Facility Name
Centre François Baclesse
City
Caen
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Stéphane CORBINAIS
First Name & Middle Initial & Last Name & Degree
Stéphane CORBINAIS, MD
Facility Name
Centre Jean Perrin
City
Clermont-Ferrand
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Florence OSAER-POLYCARPE
First Name & Middle Initial & Last Name & Degree
Florence OSAER-POLYCARPE, MD
Facility Name
CHU Estaing de Clermont Ferrand
City
Clermont-Ferrand
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Caroline PETORIN, MD
First Name & Middle Initial & Last Name & Degree
Caroline PETORIN, MD
Facility Name
APHP - Hopital Henri Mondor
City
Créteil
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Christophe TOURNIGAND, MD
First Name & Middle Initial & Last Name & Degree
Christophe TOURNIGAND, MD
Facility Name
CHU de Dijon
City
Dijon
Country
France
Facility Name
CHU Grenoble Alpes
City
Grenoble
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Gael ROTH, MD
First Name & Middle Initial & Last Name & Degree
Gael ROTH, MD
Facility Name
Centre Oscar Lambret
City
Lille
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Aurélien CARNOT, MD
First Name & Middle Initial & Last Name & Degree
Aurélien CARNOT, MD
Facility Name
CHU Lille
City
Lille
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Anthony TURPIN, MD
First Name & Middle Initial & Last Name & Degree
Anthony TURPIN, MD
Facility Name
CHU Dupuytren
City
Limoges
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Frédéric THUILLIER, MD
First Name & Middle Initial & Last Name & Degree
Frédéric THUILLIER, MD
Facility Name
Centre Leon Bérard
City
Lyon
Country
France
Facility Name
CHU de Lyon
City
Lyon
Country
France
Facility Name
Clinique Privée Jean Mermoz
City
Lyon
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Lea CLAVEL, MD
First Name & Middle Initial & Last Name & Degree
Lea CLAVEL, MD
Facility Name
APHM - CHU La Timone
City
Marseille
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Laétitia DAHAN, MD
First Name & Middle Initial & Last Name & Degree
Laétitia DAHAN, MD
Facility Name
Institut Paoli Calmettes
City
Marseille
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Simon LAUNAY, MD
First Name & Middle Initial & Last Name & Degree
Simon LAUNAY, MD
Facility Name
CHU Montpellier
City
Montpellier
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Eric ASSENAT, MD
First Name & Middle Initial & Last Name & Degree
Eric ASSENAT, MD
Facility Name
Institut de Cancer de Montpellier
City
Montpellier
Country
France
Facility Name
CHU Nantes - Hôtel Dieu
City
Nantes
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Yann TOUCHEFEU, MD
First Name & Middle Initial & Last Name & Degree
Yann TOUCHEFEU, MD
Facility Name
Centre Antoine Lacassagne
City
Nice
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Ludovic EVESQUE, MD
First Name & Middle Initial & Last Name & Degree
Ludovic EVESQUE, MD
Facility Name
APHP - Hôpital Beaujon
City
Paris
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Mohamed BOUATTOUR, MD
First Name & Middle Initial & Last Name & Degree
Mohamed BOUATTOUR, MD
Facility Name
APHP - Hôpital Cochin
City
Paris
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Romain CORIAT, MD
First Name & Middle Initial & Last Name & Degree
Romain CORIAT, MD
Facility Name
APHP - Hôpital Saint Antoine
City
Paris
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Helene BOUSSION DESLOGES, MD
First Name & Middle Initial & Last Name & Degree
Helene BOUSSION DESLOGES, MD
Facility Name
Groupe Hospitalier Diaconesses Croix Saint-Simon
City
Paris
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Olivier DUBREUIL, MD
First Name & Middle Initial & Last Name & Degree
Olivier DUBREUIL, MD
Facility Name
Institute Mutualiste Montsouris
City
Paris
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
David MALKA, MD
First Name & Middle Initial & Last Name & Degree
David MALKA, MD
Facility Name
Hôpital Privé des Côtes d'Armor
City
Plérin
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Jérôme MARTIN-BABAU, MD
First Name & Middle Initial & Last Name & Degree
Jérôme MARTIN-BABAU, MD
Facility Name
CHU Poitiers
City
Poitiers
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
David TOUGERON, MD
First Name & Middle Initial & Last Name & Degree
David TOUGERON, MD
Facility Name
CHU de Reims
City
Reims
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Alexandra HEURGUE, MD
First Name & Middle Initial & Last Name & Degree
Alexandra HEURGUE, MD
Facility Name
Institut Jean Godinot
City
Reims
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Damien BOTSEN, MD
First Name & Middle Initial & Last Name & Degree
Damien BOTSEN, MD
Facility Name
Centre Eugène Marquis
City
Rennes
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Julien Edeline
First Name & Middle Initial & Last Name & Degree
Julien Edeline, MD
Facility Name
CHU Charles Nicolle
City
Rouen
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Frederic DI FIORE, MD
First Name & Middle Initial & Last Name & Degree
Frederic DI FIORE, MD
Facility Name
Institut de Cancerologie de l'Ouest
City
Saint-Herblain
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Ludovic DOUCET, MD
First Name & Middle Initial & Last Name & Degree
Ludovic DOUCET, MD
Facility Name
Hôpital Foch
City
Suresnes
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Jaafar BENNOUNA, MD
First Name & Middle Initial & Last Name & Degree
Jaafar BENNOUNA, MD
Facility Name
CHU Toulouse
City
Toulouse
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Nadim FARES, MD
First Name & Middle Initial & Last Name & Degree
Nadim FARES, MD
Facility Name
CHRU de Nancy
City
Vandœuvre-lès-Nancy
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Marie MULLER, MD
First Name & Middle Initial & Last Name & Degree
Marie MULLER, MD
Facility Name
APHP - Hôpital Paul Brousse
City
Villejuif
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Olivier ROSMORDUC, MD
First Name & Middle Initial & Last Name & Degree
Olivier ROSMORDUC, MD
Facility Name
Gustave Roussy
City
Villejuif
Country
France
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Antoine Hollebecque
First Name & Middle Initial & Last Name & Degree
Antoine Hollebecque, MD
Facility Name
Queen Elizabeth Hospital
City
Birmingham
Country
United Kingdom
Facility Name
Bristol
City
Bristol
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Stephen FALK, MD
First Name & Middle Initial & Last Name & Degree
Stephen FALK, MD
Facility Name
Addenbrooke's Hospital
City
Cambridge
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Pippa CORRIE, MD
First Name & Middle Initial & Last Name & Degree
Pippa CORRIE, MD
Facility Name
Castle Hill Hospital
City
Cottingham
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Anthony MARAVEYAS, MD
First Name & Middle Initial & Last Name & Degree
Anthony MARAVEYAS, MD
Facility Name
St James's Hospital
City
Leeds
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Alan ANTHONEY, MD
First Name & Middle Initial & Last Name & Degree
Alan ANTHONEY, MD
Facility Name
Clatterbridge Centre for Oncology
City
Liverpool
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Daniel PALMER, MD
First Name & Middle Initial & Last Name & Degree
Daniel PALMER, MD
Facility Name
Guy's & St Thomas' Hospital
City
London
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Paul ROSS, MD
First Name & Middle Initial & Last Name & Degree
Paul ROSS, MD
Facility Name
Hammersmith Hospital
City
London
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Harpreet WASAN, MD
First Name & Middle Initial & Last Name & Degree
Harpreet WASAN, MD
Facility Name
Royal Marsden Hospital
City
London
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Sheela RAO, MD
First Name & Middle Initial & Last Name & Degree
Sheela RAO, MD
Facility Name
University College London
City
London
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
John BRIDGEWATER, MD
First Name & Middle Initial & Last Name & Degree
John BRIDGEWATER, MD
Facility Name
Maidstone Hospital
City
Maidstone
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Justin WATERS, MD
First Name & Middle Initial & Last Name & Degree
Justin WATERS, MD
Facility Name
The Christie Hospital
City
Manchester
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Juan W VALLE, MD
First Name & Middle Initial & Last Name & Degree
Juan W VALLE, MD
Facility Name
Nottingham University Hospital
City
Nottingham
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Aurora ARVIND, MD
First Name & Middle Initial & Last Name & Degree
Aurora ARVIND, MD
Facility Name
Oxford
City
Oxford
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Kinnari PATEL, MD
First Name & Middle Initial & Last Name & Degree
Kinnari PATEL, MD
Facility Name
Sheffield
City
Sheffield
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Jonathan WADSLEY, MD
First Name & Middle Initial & Last Name & Degree
Jonathan WADSLEY, MD
Facility Name
Southampton General Hospital
City
Southampton
Country
United Kingdom
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Tim IVESON, MD
First Name & Middle Initial & Last Name & Degree
Tim IVESON, MD

12. IPD Sharing Statement

Plan to Share IPD
Yes
IPD Sharing Plan Description
Unicancer will share de-identified individual data that underlie the results reported. A decision concerning the sharing of other study documents, including protocol and statistical analysis plan will be examined upon request.
IPD Sharing Time Frame
Unicancer will consider access to study data upon written detailed request sent to Unicancer, from 6 months until 5 years after publication of summary data.
IPD Sharing Access Criteria
The data shared will be limit to that required for independent mandated verification of the published results, the applicant will need authorization from Unicancer for personal access, and data will only be transferred after signing of a data access agreement.

Learn more about this trial

Personalized Medicine for Advanced Biliary Cancer Patients

We'll reach out to this number within 24 hrs