Flag of the European Union EU Clinical Trials Register Help

Clinical trials

The European Union Clinical Trials Register   allows you to search for protocol and results information on:
  • interventional clinical trials that were approved in the European Union (EU)/European Economic Area (EEA) under the Clinical Trials Directive 2001/20/EC
  • clinical trials conducted outside the EU/EEA that are linked to European paediatric-medicine development

  • EU/EEA interventional clinical trials approved under or transitioned to the Clinical Trial Regulation 536/2014 are publicly accessible through the
    Clinical Trials Information System (CTIS).


    The EU Clinical Trials Register currently displays   43876   clinical trials with a EudraCT protocol, of which   7294   are clinical trials conducted with subjects less than 18 years old.   The register also displays information on   18700   older paediatric trials (in scope of Article 45 of the Paediatric Regulation (EC) No 1901/2006).

    Phase 1 trials conducted solely on adults and that are not part of an agreed paediatric investigation plan (PIP) are not publicly available (see Frequently Asked Questions ).  
     
    Examples: Cancer AND drug name. Pneumonia AND sponsor name.
    How to search [pdf]
    Search Tips: Under advanced search you can use filters for Country, Age Group, Gender, Trial Phase, Trial Status, Date Range, Rare Diseases and Orphan Designation. For these items you should use the filters and not add them to your search terms in the text field.
    Advanced Search: Search tools
     

    < Back to search results

    Print Download

    Summary
    EudraCT Number:2019-000359-15
    Sponsor's Protocol Code Number:DZB-CS-201
    National Competent Authority:Spain - AEMPS
    Clinical Trial Type:EEA CTA
    Trial Status:Ongoing
    Date on which this record was first entered in the EudraCT database:2019-11-11
    Trial results View results
    Index
    A. PROTOCOL INFORMATION
    B. SPONSOR INFORMATION
    C. APPLICANT IDENTIFICATION
    D. IMP IDENTIFICATION
    D.8 INFORMATION ON PLACEBO
    E. GENERAL INFORMATION ON THE TRIAL
    F. POPULATION OF TRIAL SUBJECTS
    G. INVESTIGATOR NETWORKS TO BE INVOLVED IN THE TRIAL
    N. REVIEW BY THE COMPETENT AUTHORITY OR ETHICS COMMITTEE IN THE COUNTRY CONCERNED
    P. END OF TRIAL
    Expand All   Collapse All
    A. Protocol Information
    A.1Member State ConcernedSpain - AEMPS
    A.2EudraCT number2019-000359-15
    A.3Full title of the trial
    An open-label multi-cohort Phase 1b/2 study of derazantinib and atezolizumab in patients with urothelial cancer expressing activating molecular FGFR aberrations (FIDES-02)
    Un estudio Fase 1b/2, abierto, de múltiples cohortes, de derazantinib y atezolizumab en pacientes con cáncer urotelial con aberraciones moleculares activadoras del receptor del factor de crecimiento fibroblástico (FGFR, por sus siglas en inglés) (FIDES-02)
    A.3.1Title of the trial for lay people, in easily understood, i.e. non-technical, language
    Phase 1b/2 study of derazantinib and atezolizumab in patients with urothelial cancer.
    Fase 1b/2 de derazantinib y atezolizumab en pacientes con cáncer urotelial
    A.4.1Sponsor's protocol code numberDZB-CS-201
    A.7Trial is part of a Paediatric Investigation Plan No
    A.8EMA Decision number of Paediatric Investigation Plan
    B. Sponsor Information
    B.Sponsor: 1
    B.1.1Name of SponsorBasilea Pharmaceutica International Ltd.
    B.1.3.4CountrySwitzerland
    B.3.1 and B.3.2Status of the sponsorCommercial
    B.4 Source(s) of Monetary or Material Support for the clinical trial:
    B.4.1Name of organisation providing supportBasilea Pharmaceutica International Ltd.
    B.4.2CountrySwitzerland
    B.5 Contact point designated by the sponsor for further information on the trial
    B.5.1Name of organisationBasilea Pharmaceutica International Ltd.
    B.5.2Functional name of contact pointMedical Information
    B.5.3 Address:
    B.5.3.1Street AddressGrenzacherstrasse 487
    B.5.3.2Town/ cityBasel
    B.5.3.3Post code4005
    B.5.3.4CountrySwitzerland
    B.5.4Telephone number+4161 606 1400
    B.5.5Fax number+4161 606 1216
    B.5.6E-mailmedical.information@basilea.com
    D. IMP Identification
    D.IMP: 1
    D.1.2 and D.1.3IMP RoleTest
    D.2 Status of the IMP to be used in the clinical trial
    D.2.1IMP to be used in the trial has a marketing authorisation No
    D.2.5The IMP has been designated in this indication as an orphan drug in the Community No
    D.2.5.1Orphan drug designation number
    D.3 Description of the IMP
    D.3.1Product namederazantinib
    D.3.2Product code ARQ 087
    D.3.4Pharmaceutical form Capsule, hard
    D.3.4.1Specific paediatric formulation No
    D.3.7Routes of administration for this IMPOral use
    D.3.8 to D.3.10 IMP Identification Details (Active Substances)
    D.3.8INN - Proposed INNDerazantinib
    D.3.9.1CAS number 1234356-88-7
    D.3.9.2Current sponsor codeDerazantinib
    D.3.9.3Other descriptive nameDERAZANTINIB•2HCL
    D.3.9.4EV Substance CodeSUB197162
    D.3.10 Strength
    D.3.10.1Concentration unit mg milligram(s)
    D.3.10.2Concentration typeequal
    D.3.10.3Concentration number100
    D.3.11 The IMP contains an:
    D.3.11.1Active substance of chemical origin Yes
    D.3.11.2Active substance of biological/ biotechnological origin (other than Advanced Therapy IMP (ATIMP) No
    The IMP is a:
    D.3.11.3Advanced Therapy IMP (ATIMP) No
    D.3.11.3.1Somatic cell therapy medicinal product No
    D.3.11.3.2Gene therapy medical product No
    D.3.11.3.3Tissue Engineered Product No
    D.3.11.3.4Combination ATIMP (i.e. one involving a medical device) No
    D.3.11.3.5Committee on Advanced therapies (CAT) has issued a classification for this product No
    D.3.11.4Combination product that includes a device, but does not involve an Advanced Therapy No
    D.3.11.5Radiopharmaceutical medicinal product No
    D.3.11.6Immunological medicinal product (such as vaccine, allergen, immune serum) No
    D.3.11.7Plasma derived medicinal product No
    D.3.11.8Extractive medicinal product No
    D.3.11.9Recombinant medicinal product No
    D.3.11.10Medicinal product containing genetically modified organisms No
    D.3.11.11Herbal medicinal product No
    D.3.11.12Homeopathic medicinal product No
    D.3.11.13Another type of medicinal product No
    D.IMP: 2
    D.1.2 and D.1.3IMP RoleTest
    D.2 Status of the IMP to be used in the clinical trial
    D.2.1IMP to be used in the trial has a marketing authorisation Yes
    D.2.1.1.1Trade name Tecentriq
    D.2.1.1.2Name of the Marketing Authorisation holderRoche Registration GmbH
    D.2.1.2Country which granted the Marketing AuthorisationEuropean Union
    D.2.5The IMP has been designated in this indication as an orphan drug in the Community No
    D.2.5.1Orphan drug designation number
    D.3 Description of the IMP
    D.3.4Pharmaceutical form Concentrate for solution for infusion
    D.3.4.1Specific paediatric formulation No
    D.3.7Routes of administration for this IMPIntravenous use
    D.3.8 to D.3.10 IMP Identification Details (Active Substances)
    D.3.8INN - Proposed INNATEZOLIZUMAB
    D.3.9.2Current sponsor codeatezolizumab
    D.3.9.4EV Substance CodeSUB178312
    D.3.10 Strength
    D.3.10.1Concentration unit mg milligram(s)
    D.3.10.2Concentration typeequal
    D.3.10.3Concentration number1200
    D.3.11 The IMP contains an:
    D.3.11.1Active substance of chemical origin No
    D.3.11.2Active substance of biological/ biotechnological origin (other than Advanced Therapy IMP (ATIMP) Yes
    The IMP is a:
    D.3.11.3Advanced Therapy IMP (ATIMP) No
    D.3.11.3.1Somatic cell therapy medicinal product No
    D.3.11.3.2Gene therapy medical product No
    D.3.11.3.3Tissue Engineered Product No
    D.3.11.3.4Combination ATIMP (i.e. one involving a medical device) No
    D.3.11.3.5Committee on Advanced therapies (CAT) has issued a classification for this product No
    D.3.11.4Combination product that includes a device, but does not involve an Advanced Therapy No
    D.3.11.5Radiopharmaceutical medicinal product No
    D.3.11.6Immunological medicinal product (such as vaccine, allergen, immune serum) No
    D.3.11.7Plasma derived medicinal product No
    D.3.11.8Extractive medicinal product No
    D.3.11.9Recombinant medicinal product Yes
    D.3.11.10Medicinal product containing genetically modified organisms No
    D.3.11.11Herbal medicinal product No
    D.3.11.12Homeopathic medicinal product No
    D.3.11.13Another type of medicinal product No
    D.8 Information on Placebo
    E. General Information on the Trial
    E.1 Medical condition or disease under investigation
    E.1.1Medical condition(s) being investigated
    urothelial cancer
    cáncer urotelial
    E.1.1.1Medical condition in easily understood language
    urothelial cancer
    cáncer urotelial
    E.1.1.2Therapeutic area Diseases [C] - Cancer [C04]
    MedDRA Classification
    E.1.2 Medical condition or disease under investigation
    E.1.2Version 21.1
    E.1.2Level LLT
    E.1.2Classification code 10077840
    E.1.2Term Urothelial cancer of renal pelvis
    E.1.2System Organ Class 100000004864
    E.1.3Condition being studied is a rare disease No
    E.2 Objective of the trial
    E.2.1Main objective of the trial
    Primary efficacy objective
    • To evaluate the objective response rate (ORR) of derazantinib
    monotherapy (in Substudies 1 and 5, and Cohort 4a) and of
    derazantinib-atezolizumab in combination (in Substudy 3 and Cohort 4b)
    in patients with mUC expressing FGFR1–3 GAs.
    Primary safety objectives
    • To confirm derazantinib 200 mg BID plus atezolizumab 1200 mg Q3W
    as a safe and tolerable regimen (Substudy 3).
    • To confirm derazantinib 200 mg BID as a safe and tolerable dose regimen of derazantinib monotherapy (Substudy 5).
    Objetivo principal de eficacia
    • Evaluar la tasa de respuesta objetiva (TRO) de derazantinib monoterapia (en los subestudios 1 y 5, y la cohorte 4a) y de derazantinib-atezolizumab en combinación (en el subestudio 3 y la cohorte 4b) en pacientes con mUC que expresan FGFR1–3 GA.
    Objetivos primarios de seguridad
    • Para confirmar derazantinib 200 mg BID más atezolizumab 1200 mg Q3W como un régimen seguro y tolerable (subestudio 3).
    • Confirmar derazantinib 200 mg dos veces al día como una dosis segura y tolerable régimen de monoterapia con derazantinib (subestudio 5
    E.2.2Secondary objectives of the trial
    Secondary objectives
    • To evaluate the efficacy of the study drugs as measured by disease
    control rate (DCR), duration of response (DOR), progression-free
    survival (PFS) and overall survival (OS)
    • To assess the safety and tolerability of the study drugs
    • To characterize the PK profile of derazantinib 200 mg BID (and, if
    applicable, derazantinib metabolites) as monotherapy (Substudy 5) and
    in combination with atezolizumab (Substudy 3).
    • To evaluate changes, and assess the minimally important difference, in
    health-related quality of life (HR-QoL) and symptom response from
    baseline by cohort, substudy and in the overall study population using
    the EORTC QLQ C30, FACT-Bl, EQ-5D (5L) visual analogue scale (VAS),
    and Health Transition Index/G-SET.
    Objetivos secundarios
    • Evaluar la eficacia de los fármacos del estudio según la enfermedad.
    tasa de control (DCR), duración de la respuesta (DOR), sin progresión
    supervivencia (SLP) y supervivencia global (SG)
    • Evaluar la seguridad y tolerabilidad de los fármacos del estudio.
    • Caracterizar el perfil farmacocinético de derazantinib 200 mg dos veces al día (y, si
    aplicable, metabolitos de derazantinib) como monoterapia (Subestudio 5) y
    en combinación con atezolizumab (subestudio 3).
    • Evaluar los cambios y valorar la diferencia mínimamente importante, en
    calidad de vida relacionada con la salud (HR-QoL) y respuesta a los síntomas de
    línea de base por cohorte, subestudio y en la población general del estudio utilizando
    la escala analógica visual (VAS) EORTC QLQ C30, FACT-Bl, EQ-5D (5L),
    e Índice de transición sanitaria / G-SET.
    E.2.3Trial contains a sub-study Yes
    E.2.3.1Full title, date and version of each sub-study and their related objectives
    1) Future Biomedical Research
    Future biomedical research specimens will be stored to provide a resource for future studies conducted by the Sponsor focused on the study of biomarkers responsible for how a drug enters and is removed by the human body, how a drug works, other pathways a drug may interact with, or other aspects of disease. The specimens may be used for future assay development and/or drug development.
    2) Pharmacodynamic research (optional)
    Patients enrolled into Substudies 3, 4 and 5 have the option to
    participate in an exploratory Pharmacodynamics Research Group (PDR
    Group) to investigate the role of CSF1R inhibition by derazantinib,comprising a skin biopsy and blood sampling for monocyte subsets priorm to the first dose of study drug at the Screening visit and on C2D1. Up to
    20 patients will be enrolled in this PDR Group.
    1) Futura investigación biomédica
    Las futuras muestras de investigación biomédica se almacenarán para proporcionar un recurso para futuros estudios realizados por el Patrocinador que se centran en el estudio de biomarcadores responsables de cómo un medicamento ingresa y se elimina por el cuerpo humano, cómo funciona un medicamento, otras vías con las que un medicamento puede interactuar. , u otros aspectos de la enfermedad. Las muestras pueden usarse para el desarrollo de ensayos futuros y / o el desarrollo de fármacos.
    2) Investigación farmacodinámica (opcional)
    Los pacientes inscritos en los subestudios 3, 4 y 5 tienen la opción de
    participar en un Grupo de Investigación de Farmacodinámica exploratoria (PDR
    Group) para investigar el papel de la inhibición de CSF1R por derazantinib,
    que comprende una biopsia de piel y una muestra de sangre para subconjuntos de monocitos antes
    a la primera dosis del fármaco del estudio en la visita de selección y en C2D1.
    Hasta Se inscribirán 20 pacientes en este grupo de PDR
    E.3Principal inclusion criteria
    1.ICF signed by the patient indicating that they understand the purpose of, and procedures required for, the study and are willing to participate in the study, prior to any study-related procedure.
    2.Male or female aged ≥ 18 years.
    3.Histologically-confirmed transitional cell carcinoma of the urothelium
    of the upper or lower urinary tract. Note: Minor components (< 50% overall) of variant histology such as glandular or squamous differentiation, or evolution to more aggressive phenotypes such as sarcomatoid or micropapillary change, are acceptable.
    4.Recurrent or progressing stage IV disease, or surgically unresectable, recurrent or progressing disease, as specified for each substudy:
    •Substudy 1: Patients with mUC expressing specific FGFR1–3 GAs who have progressed on at least one standard chemotherapy and/or immune-checkpoint blockade and have not received prior FGFR inhibiting treatment.
    •Substudy 2: Patients with any advanced solid tumor and any prior
    treatment (including FGFR inhibitor treatment), who have no standard
    treatment alternative (per multidisciplinary tumor board endorsement).
    •Substudy 3: First-line cisplatin-ineligible patients with patients with mUC expressing specific FGFR1–3 GAs and PD-L1 expression < 5% ('PDL1-low').
    Cisplatin-ineligibility as defined by any one of the following criteria: 1) grade ≥ 2 peripheral neuropathy; 2) CLCR calculated by Cockcroft-Gault >30 mL/min but < 60 mL/min; 3) hearing impairment (measured by audiometry) of > 25 dB at two contiguous test frequencies in at least one ear; 4) comorbidity that precludes high-volume hydration.Note: For atezolizumab, a low PD-L1 expression level is less than 5% of immune cells staining positive for PD-L1 by immunohistochemistry. Information
    on FDA-approved tests for the determination of PD-L1 expression in locally advanced or metastatic urothelial carcinoma or triple-negative
    breast cancer are available at:
    http://www.fda.gov/CompanionDiagnostics.
    •Substudy 4: Patients with FGFR inhibitor-resistant, mUC expressing specific FGFR1–3 GAs who have progressed on at least one standard regimen each of chemotherapy and immune-checkpoint blockade and have received FGFR inhibiting treatment (excluding derazantinib).
    Patients assessed as having progressed upon prior treatment with FGFR inhibitors must have received FGFR inhibitor treatment for at least 12 weeks and have undergone at least one on treatment tumor imaging assessment.
    Prior treatment with immune-checkpoint blockade and FGFR inhibitor in combination are not allowed; prior treatment with either sequential immune-checkpoint blockade and FGFR inhibitor treatment or combinations of FGFR inhibitor and chemotherapy are allowed.
    •TCS Group: Patients enrolled in the TCS Group need to have disease that is safely amenable to repeated biopsies.
    • Substudy 5: Patients with mUC expressing FGFR1–3 GAs who have
    progressed on at least one standard chemotherapy and/or immunecheckpoint
    blockade and have not received prior FGFR inhibiting treatment.
    5. Eligible FGFR1–3 GA-positive test result5.For enrollment into Substudy 1 or 3, patients require an FGFR1–3 GApositive
    central test result (see Section 3.1.1)
    6.For enrollment into Substudy 4, patients require a documented FGFR1–
    3 GA status from prior FGFR inhibiting treatment, or an FGFR1–3 GApositive central test result.
    7.Measurable disease, as defined by the Investigator using RECIST 1.1 criteria, documented within the 28 days prior to study drug administration.
    8.ECOG PS of 0, 1 or 2
    9.Adequate organ functions as indicated by the following Screening visit local laboratory values:
    •Hemoglobin ≥ 9 g/dL
    •Absolute neutrophil count (ANC) ≥ 1.5 × 1000000000/L
    •Platelets ≥ 75 × 1000000000/L
    •International normalized ratio (INR) 0.8 to upper limit of normal (ULN) or  3 for patients receiving anticoagulant therapy
    •Total bilirubin ≤ 2 × ULN
    •Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) ≤ 3 × ULN, or ≤ 5 × ULN for study patients with liver metastasis
    •Albumin ≥ 2.5 g/dL
    •CLCR ≥ 30 mL/min (as calculated by the Cockcroft-Gault formula)
    •For women of childbearing potential only, negative serum human chorionic gonadotropin (hCG)
    10.Men and women of childbearing potential must agree to avoid impregnating a partner or becoming pregnant, respectively, during the study, and for at least 5 months after the last dose of either investigational drug.
    1. Consentimiento informado firmado por el paciente que indique que entiende su propósito y los procedimientos necesarios para el estudio y está dispuesto a participar en el estudio antes de cualquier procedimiento relacionado con este.
    2. Hombres y mujeres de 18 años o más.
    3. Carcinoma de células de transición del urotelio confirmado histológicamente
    del tracto urinario superior o inferior.
    Nota: componentes menores (<50% en general) de histología variante como glandular o escamosa diferenciación, o evolución a fenotipos más agresivos como cambio sarcomatoide o micropapilar, son aceptables
    4. Enfermedad en estadio IV recurrente o en progresión o enfermedad recurrente o progresiva no extirpable quirúrgicamente, como se indica para cada subestudio:
    • Subestudio 1: Pacientes con CUm que expresa AG activadoras de FGFR1–3 y que presenten progresión con al menos una quimioterapia de referencia y bloqueo del punto de control inmunitario y no hayan recibido tratamiento previo con inhibidores de FGFR.
    • Subestudio 2: pacientes con cualquier tumor sólido avanzado y
    tratamiento (incluido el tratamiento con inhibidor de FGFR), que no tienen una alternativa de tratamiento estándar (según el respaldo multidisciplinario de la junta tumoral).
    • Subestudio 3: pacientes no elegibles con cisplatino de primera línea con pacientes con mUC que expresa GA FGFR1–3 específicas y expresión PD-L1 <5% ('PDL1-bajo').
    Inhabilitación de cisplatino según lo definido por cualquiera de los siguientes criterios: 1) neuropatía periférica grado ≥ 2; 2) CLCR calculado por Cockcroft-Gault> 30 ml / min pero <60 ml / min; 3) discapacidad auditiva (medida por radiometría) de> 25 dB a dos frecuencias de prueba contiguas en al menos un oído; 4) comorbilidad que impide la hidratación de alto volume.
    Nota: Para atezolizumab, un nivel bajo de expresión de PD-L1 es menos del 5% de las células inmunitarias que se tiñen positivas para PD-L1 por inmunohistoquímica. Información sobre pruebas aprobadas por la FDA para la determinación de la expresión de PD-L1 en
    El carcinoma urotelial localmente avanzado o metastásico o el cáncer de mama triple negativo están disponibles en:
    http://www.fda.gov/CompanionDiagnostics.
    • Subestudio 4: Los pacientes con CUm resistente a los inhibidores de FGFR que exprese AG activadoras de FGFR1–3 y que hayan presentado progresión con al menos un régimen de quimioterapia y bloqueo del punto de control inmunitario2 y hayan recibido previamente tratamiento inhibidor de FGFR3 (excluido derazantinib)Los pacientes evaluados como que han progresado con el tratamiento previo con inhibidores de FGFR deben haber recibido tratamiento con inhibidor de FGFR durante al menos 12 semanas y haber recibido al menos uno en la evaluación de imágenes de tumor de tratamiento.
    No se permite el tratamiento previo con el bloqueo del punto de control inmunitario y el inhibidor de FGFR en combinación; Se permite el tratamiento previo con bloqueo secuencial del punto de control inmunitario y tratamiento con inhibidor de FGFR o combinaciones de inhibidor de FGFR y quimioterapia.
    • Grupo TCS: Los pacientes incluidos en el grupo de TCS deben poder someterse de forma segura a biopsias repetidas.
    • Subestudio 5: Pacientes con mUC que expresan GA de FGFR1–3 que tienen
    progresó en al menos una quimioterapia estándar y / o un punto de control inmunológico
    bloqueo y no han recibido inhibición previa de FGFR tratamiento.
    5. Resultado de prueba positivo para FGFR1–3 GA elegible
    5.Para la inscripción en el subestudio 1 o 3, los pacientes requieren un resultado de prueba central positivo para FGFR1-3 GA (consulte la Sección 3.1.1)
    6.Para la inscripción en el subestudio 4, los pacientes requieren un estado de FGFR1–3 GA documentado de un tratamiento inhibidor de FGFR anterior, o un resultado de prueba central positivo para FGFR1–3 GA.
    7. Enfermedad medible, según la define el investigador utilizando los criterios RECIST 1.1, documentada dentro de los 28 días anteriores a la administración del fármaco del estudio.
    8. Estado funcional según el Grupo Oncológico Cooperativo de la Costa Este (ECOG) de 0, 1 o 2
    9. Funciones orgánicas adecuadas indicadas por los valores del laboratorio local en la visita de selección.
    10. Los hombres y mujeres en edad fértil deben acceder a abstenerse de engendrar hijos o quedarse embarazadas, respectivamente, durante el estudio y durante al menos 5 meses después de la última dosis de cualquiera de los fármacos en fase de investigación
    E.4Principal exclusion criteria
    Prior cancer treatment
    1.Receipt of treatment before the 1st dose of study drug (Cycle 1 Day 1) within an interval shorter than the following, as applicable:
    •One chemotherapy or biological (e.g: antibody) cycle interval
    •5 half-lives of any small molecule investigational or licensed medicinal product
    •2 W, for any investigational medicinal product (IMP) with an unknown half-life
    •4 W of curative radiotherapy
    •7 days of palliative radiotherapy
    •12 months of neo-adjuvant or adjuvant chemotherapy or radiation (only applies to SbS 3)
    2.For SbS 1 and SbS 3 patients, prior FGFR inhibiting treatment.
    3.Prior FGFR inhibiting treatment (except Substudy 4).
    4.For Substudy 4 patients, prior treatment with FGFR inhibitor in combination with anti-PD-1 or anti-PD-L1 therapeutic antibody or PD-1/PD-L1 pathway-targeting agents.
    5.Concurrent evidence of clinically significant corneal or retinal disorder
    likely to increase the risk of eye toxicity, including but not limited to bullous/band keratopathy, keratoconjunctivitis (unless keratoconjunctivitis sicca), corneal abrasion (unless related to trauma),
    inflammation/ulceration, confirmed by ophthalmological examination.
    6.History of clinically significant cardiac disorders:
    •Myocardial infarction, or New York Heart Association Class II to IV congestive heart failure, within 6 months of the first dose of study drug
    •Concurrent and clinically significant abnormalities on ECG at Screening, including TcF > 450 ms for males or > 460 ms for females(mean values from triplicate ECGs; see Section 5.3.2.4 of the protocol)
    7.Serum electrolyte abnormalities defined as follows:
    •Hyperphosphatemia: serum phosphate > institutional ULN
    •Hyperkalemia: serum potassium > institutional ULN
    •Hypokalemia: serum potassium < institutional LLN
    •Hypercalcemia: corrected serum calcium > 3.1 mmol/L (> 12.5 mg/dL)
    •Hypocalcemia: corrected serum calcium < 1.75 mmol/L (< 7.0 mg/dL)
    •Hypomagnesemia: < 0.4 mmol/L (< 0.9 mg/dL)
    9.Uncontrolled tumor-related hypercalcemia
    Medical history
    10. Bellmunt score 3, or 2 if based on a combination of hemoglobin < 10
    g/dL and presence of liver metastasis
    11. Any unresolved (at the time of Screening) clinically significant CTCAE
    grade ≥ 2 toxicity (except for alopecia, grade 2 platinum-therapy related
    neuropathy, grade 2 anemia from previous anti-tumor treatment, grade
    2 renal impairment per reduced CLCR by Cockcroft-Gault of 30–60
    mL/min [which is generally accepted for this cancer population], and/or
    medical/surgical procedures/interventions).
    12. Known CNS metastases
    13.Lack of recovery from major (e.g., open abdominal) surgery after 4
    weeks, or major elective surgery is planned during the foreseeable
    duration of the study
    14. Concurrent uncontrolled or active infection with human
    immunodeficiency virus (known HIV 1/2 antibodies positive)
    15. Active hepatitis B or chronic hepatitis B without current antiviral
    therapy and an HBV DNA ≥ 100 IU/mL
    Note: Active hepatitis B is defined as a known positive hepatitis B
    surface antigen (HBsAg) result.
    16. Active hepatitis C
    Note: Active hepatitis C is defined by a known positive Hep C antibody
    result and known quantitative hepatitis C virus (HCV) RNA results
    greater than the lower limits of detection of the assay.
    17.Significant gastrointestinal disorders that could, in the opinion of Investigator, interfere with the absorption, metabolism, or excretion of derazantinib (e.g., Crohn’s disease, ulcerative colitis, diarrhea, extensive gastric resection, functionally relevant gastrointestinal obstruction, or vomiting)
    18.History of additional malignancy that is progressing or requires active treatment. Exceptions include basal cell carcinoma of the skin, squamous cell carcinoma of the skin that has undergone potentially curative therapy, or in situ cervical cancer.
    An incidental finding of prostate cancer (identified upon resection of the prostate) is acceptable, provided that the following criteria are met: Stage T2N0M0 or lower; Gleason score ≤ 6, and prostate-specific antigen below lower limit of normal by local laboratory.
    19.Chronic leg ulcers, decubitus ulcers, or unhealed incisions.
    General patient disposition
    20.Known hypersensitivity or allergy any component of the derazantinib formulation
    22.Any other uncontrolled intercurrent illness that would unduly increase the risk of toxicity or limit compliance with study requirements, including but not limited to ongoing or active symptomatic infection, uncontrolled diabetes mellitus, or hepatic, renal, respiratory, or psychiatric illness.
    24.Pregnant or breast feeding.
    For full list please refer to protocol
    Tto previo del cáncer
    1.Recibimiento del tto antes de la primera dosis del fármaco del estudio (C1D1) en un intervalo más corto que el siguiente, según corresponda:
    • Intervalo ciclo de quimioterapia o biológico (ej: anticuerpo)
    • 5 v1/2 de cualquier mto de investigación de molécula pequeña o licenciado
    • 2 semanas, para cualquier mto en investigación (IMP) con 1 v1/2 desconocida
    • 4 semanas de radioterapia curativa.
    • 7 días de radioterapia paliativa.
    • 12 meses de quimioterapia o radiación neo-adyuvante o adyuvante (solo se aplica al Subest 3)
    2.Para los pacientes del Subestudio 1 y del Subestudio 3, tratamiento previo con inhibición de FGFR.
    3. Tratamiento inhibidor de FGFR previo (excepto Subestudio 4).
    4.Para pat Sbs 4, tratamiento previo con inhibidor de FGFR en combinación con anticuerpo terapéutico anti-PD-1 o anti-PD-L1 o agentes dirigidos a la vía PD-1 / PD-L1. Deficiencias de órganos críticos
    5.Evidencia concurrente de trastorno corneal o retiniano clínicamente significativo que probablemente aumente el riesgo de toxicidad ocular, que incluye, entre otros, queratopatía ampollosa / en banda, queratoconjuntivitis (a menos queratoconjuntivitis seca), abrasión corneal (a menos que esté relacionada con un traumatismo), inflamación / ulceración, confirmada por examen oftalmológico.
    6. Antecedentes de trastornos cardíacos clínicamente significativos, como infarto de miocardio o insuficiencia cardíaca congestiva de Clase II a IV de la Asociación de Cardiología de Nueva York (NYHA), en los 6 meses anteriores a la 1ªD del fco del estudio o alteraciones concomitantes y clínicamente significativas en ECG en la selección, incluido QTcF >450 ms en hombres o >460 ms en mujeres.(valores medios de ECG por triplicado; consulte la Sección 5.3.2.4 del protocol)
    7. Anomalías en los electrolitos séricos definidos:
    • Hiperfosfatemia: P sérico> ULN institucional
    • Hipercalemia: K sérico> ULN institucional
    • Hipocalemia: K sérico <LLN institucional
    • Hipercalcemia: Na sérico corregido> 3.1 mmol / L (> 12.5 mg / dL)
    • Hipocalcemia: Na sérico corregido <1.75 mmol / L (<7.0 mg / dL)
    • Hipomagnesemia: <0.4 mmol / L (<0.9 mg / dL)
    9. HiperCa relacionada con el tumor no controlado.
    Historial medico
    10. Puntaje de Bellmunt 3, o 2 si se basa en una combinación de hemoglobina <10 g / dL y presencia de metástasis hepática
    11. Cualquier CTCAE clínicamente significativo no resuelto (en el momento de la selección) toxicidad de grado ≥ 2 (excepto para alopecia, grado 2 relacionado con la terapia con platino neuropatía, anemia de grado 2 por tratamiento antitumoral previo, grado 2 insuficiencia renal por CLCR reducido por Cockcroft-Gault de 30 a 60 mL / min [que se acepta generalmente para esta población de cáncer], y / o intervenciones / procedimientos médicos / quirúrgicos).
    12. Metástasis del SNC conocidas
    13.Falta de recuperación de una cirugía mayor (p. Ej., Abdominal abierta) después de 4 semanas, o se planifica una cirugía mayor electiva durante el período previsible duración del estudio
    14. Infección concurrente no controlada o activa con humanos virus de inmunodeficiencia (positivo para anticuerpos VIH 1/2 conocido)
    15. Hepatitis B activa o hepatitis B crónica sin antiviral actual terapia y un ADN del VHB ≥ 100 UI / ml Nota: la hepatitis B activa se define como una hepatitis B positiva conocida resultado del antígeno de superficie (HBsAg).
    16. Hepatitis C activa Nota: la hepatitis C activa se define por un anticuerpo contra la hepatitis C positivo conocido resultado y resultados cuantitativos conocidos del ARN del virus de la hepatitis C (VHC) mayor que los límites inferiores de detección del ensayo.
    17. Trastornos GI significativos que, podrían interferir con la absorción, el metabolismo o la excreción de derazantinib
    18.Historia de malignidad adicional que está progresando o requiere tto activo. Excepciones incluyen carcinoma de cél basales de piel, carcinoma de células escamosas de la piel que se ha sometido a una terapia potencialmente curativa o el cáncer cervical in situ.
    Un hallazgo incidental de cáncer de próstata (identificado en la resección de la próstata) es aceptable, siempre que se cumplan: Estadio T2N0M0 o inferior; Puntuación de Gleason ≤ 6, y Ag prostático específico por debajo del límite inferior de lo normal por el laboratorio local.
    19. Úlceras crónicas en las piernas, úlceras de decúbito o incisiones no curadas.Disposición general del paciente.
    20. Hipersensibilidad o alergia conocida a componentes de la formulación de derazantinib.
    22. Cualquier otra enfermedad intercurrente no controlada que aumentaría indebidamente el riesgo de toxicidad o limitaría el cumplimiento de los requisitos del estudio, que incluyen, entre otros, una infección sintomática activa o en curso, diabetes mellitus no controlada o enfermedad hepática, renal, respiratoria o psiquiátrica.
    21. Mujeres embarazadas o en período de lactancia.
    Para obtener una lista completa, consulte el protocolo
    E.5 End points
    E.5.1Primary end point(s)
    Primary endpoint in Substudies 1, 3 and 4
    •ORR, as measured by the proportion of patients with confirmed CR or PR by BICR.
    Criterio de valoración principal de seguridad (subestudios 3 y 5)
    Primary safety endpoint (Substudies 3 and 5)
    • For the safety interim analysis in Substudy 3, the primary endpoint is
    the proportion of patients with DLTs during treatment with derazantinib
    200 mg BID and atezolizumab 1200 mg Q3W in combination.
    • For the safety interim analysis in Substudy 5, the primary endpoint is
    the proportion of patients with DLTs during treatment with derazantinib
    200 mg BID monotherapy.
    Efficacy endpoint:
    The primary endpoint in all substudies will be ORR, defined as the
    achievement of confirmed CR or PR using RECIST 1.1, as assessed by
    BICR.
    -TRO, medida como la proporción de pacientes con respuesta completa (RC)1 o respuesta parcial (RP)2 confirmada por un revisor central independiente con enmascaramiento (BICR).
    Criterio de valoración principal de seguridad (subestudios 3 y 5)
    • Para el análisis intermedio de seguridad en el subestudio 3, el criterio de valoración principal es la proporción de pacientes con TLD durante el tratamiento con derazantinib 200 mg dos veces al día y atezolizumab 1200 mg cada 3 semanas en combinación.
    • Para el análisis intermedio de seguridad en el subestudio 5, el criterio de valoración principal es la proporción de pacientes con TLD durante el tratamiento con derazantinib 200 mg dos veces al día en monoterapia.
    Criterio de valoración de eficacia:
    El criterio de valoración principal en todos los subestudios será la TRO, definida como el logro de una RC o RP confirmada utilizando RECIST 1.1, según la evaluación de BICR.
    E.5.1.1Timepoint(s) of evaluation of this end point
    Safety and tolerability of both the monotherapy and the combination
    dose regimens will be concluded by joint decisions taken by the IDMC,
    Investigators, and the Sponsor in reviewing the aggregate of DLT, AE
    and PK data from the first 10 patients enrolled in Substudy 3 and 5.
    The final analysis for ORR will be performed once all patients have been
    enrolled per cohort/substudy and have been followed for approximately
    up to 27 weeks of treatment (i.e., each patient had up to four postbaseline
    study imaging assessments).
    Seguridad y tolerabilidad tanto de la monoterapia como de la combinación los regímenes de dosis se concluirán mediante decisiones conjuntas tomadas por el IDMC,
    Los investigadores y el patrocinador al revisar el conjunto de DLT, AE y datos farmacocinéticos de los primeros 10 pacientes inscritos en los subestudios 3 y 5.
    El análisis final de la TRO se realizará una vez que todos los pacientes hayan sido inscritos por cohorte / subestudio y han sido seguidos durante aproximadamente hasta 27 semanas de tratamiento (es decir, cada paciente tuvo hasta cuatro estudio de evaluaciones por imágenes).
    E.5.2Secondary end point(s)
    Secondary endpoints
    •DCR, as measured by the proportion of patients with confirmed CR, PR or stable disease (SD) by BICR
    •DOR, as calculated from the first date of documented tumor response to disease progression by BICR
    •Median PFS and PFS at 6 months, as determined by BICR and measured from time of first dose to time of objective tumor progression or death, and the proportion of patients alive and free of objective tumor progression 6 months after cohort assignment, respectively
    •Median OS and OS at 6 months, as measured from time of first dose until time of death and the proportion of patients alive 6 months after cohort assignment, respectively
    • Safety and tolerability of study drugs as measured by the frequency
    and severity of AEs, clinical laboratory parameters, vital signs, ECOG PS,
    physical examinations (including eye examinations), and ECG
    parameters over time, and graded by NCI CTCAE 5.0
    • Derazantinib (and – if applicable –derazantinib metabolites) plasma
    concentrations, Cmax, tmax, AUC0-12, AUC0-24, AUClast assessed by
    measurements in blood samples during treatment as monotherapy
    (Substudy 5) or in combination (Substudy 3)
    • Changes in HR-QoL and symptom response, measured by global,
    functional and symptom scores obtained from patient reported outcome
    instruments at baseline and over time (EORTC QLQ C30, FACT-Bl, EQ-5D
    [5L] VAS), and Health Transition Index/G-SET.
    Exploratory endpoints specific to efficacy-estimating substudies:
    • Responding and non-responding patients are to be analyzed by type of
    FGFR1–3 GA.
    • The proportion of patients with concordant molecular assessment from
    plasma-based and tissue-based testing measuring FGFR GAs in matched
    tumor-liquid biopsy samples obtained at baseline and during treatment
    will be described.
    • The DCR, DOR, PFS and OS of patients treated with derazantinibatezolizumab
    in combination will be evaluated for patients crossing over
    after documented disease progression on derazantinib monotherapy
    (Substudy 4).
    • The DCR, DOR and PFS of patients treated with derazantinibatezolizumab
    in combination will be evaluated by iRECIST (applicable to
    Substudies 3 and 4).
    • In Substudy 4, the DCR, DOR, PFS and OS of patients treated with
    derazantinib will be compared to that of patients treated with
    derazantinib-atezolizumab in combination to generate potential
    hypotheses for future comparative studies.
    • The DCR, DOR, PFS and OS of patients treated with derazantinib and
    derazantinib-atezolizumab in combination, respectively, will be
    evaluated by molecular profile, gene expression profile, and biomarkers.
    • The proportion of patients with a specific reduction of
    immunosuppressive macrophages in skin biopsies or peripheral blood
    monocytes on derazantinib monotherapy and derazantinib-atezolizumab
    combination treatment.
    • EQ-5D (5L) health status data will be used for obtaining utilitymeasures for economic modeling. For the EQ-5D (5L), further scoring
    and analysis may be reported in a separate document.
    Exploratory PK endpoints
    • Derazantinib plasma concentrations, assessed by measurements in
    blood samples.
    • Plasma concentrations of derazantinib metabolites (if applicable),
    assessed by measurements from blood samples.
    • Atezolizumab serum concentrations, assessed by measurements from
    blood samples (Substudy 3, Cohort 4b, and Crossover).
    • Serum ADA assayed from blood samples (Substudy 3, Cohort 4b and
    Crossover).
    • Derazantinib plasma concentrations (Cmax, tmax, AUC0-24, AUClast),
    assessed by measurements from blood samples (applicable to
    Substudies with 300 mg QD).
    Análisis de los criterios de valoración secundarios de los subestudios 1, 3, 4
    • TCE, medida por la proporción de pacientes con RC confirmada, RP o enfermedad estable (EE) por BICR
    • DdR, calculado a partir de la primera fecha de respuesta tumoral documentada a la progresión de la enfermedad por BICR
    • Medias de SSP y SSP a los 6 meses, según lo determinado por BICR y medido desde el momento de la primera dosis hasta el momento de la progresión o muerte objetiva del tumor, y la proporción de pacientes vivos y libres de progresión objetiva del tumor 6 meses después de la asignación de la cohorte, respectivamente
    • Mediana SGy SG a los 6 meses, medida desde el momento de la primera dosis hasta el momento de la muerte y la proporción de pacientes vivos 6 meses después de la asignación de la cohorte, respectivamente
    • Seguridad y tolerabilidad de los fármacos del estudio medida por la frecuencia y gravedad de los EA, parámetros de laboratorio clínico, signos vitales, ECOG PS, exámenes físicos (incluidos exámenes oculares) y ECG parámetros a lo largo del tiempo y calificados por NCI CTCAE 5.0
    • Plasma de derazantinib (y, si corresponde, metabolitos de derazantinib) concentraciones, Cmax, tmax, AUC0-12, AUC0-24, AUC last evaluado por mediciones en muestras de sangre durante el tratamiento como monoterapia (Subestudio 5) o en combinación (Subestudio 3) • Cambios en la HR-QoL y la respuesta a los síntomas, medidos por global, puntuaciones funcionales y de síntomas obtenidas del resultado informado por el paciente instrumentos al inicio y a lo largo del tiempo (EORTC QLQ C30, FACT-Bl, EQ-5D [5L] VAS) e Índice de transición de salud / G-SET. Criterios de valoración exploratorios específicos de los subestudios de estimación de la eficacia:
    • Los pacientes que responden y no responden deben ser analizados por tipo de FGFR1–3 GA.
    • La proporción de pacientes con evaluación molecular concordante de pruebas basadas en plasma y en tejidos que miden GA de FGFR en Muestras de biopsia de líquido tumoral obtenidas al inicio del estudio y durante el tratamiento. será descrito.
    • La DCR, DOR, PFS y SG de los pacientes tratados con derazantinibatezolizumab en combinación se evaluará para pacientes que cruzan después de la progresión documentada de la enfermedad con derazantinib en monoterapia (Subestudio 4).
    • La DCR, DOR y PFS de los pacientes tratados con derazantinibatezolizumab en combinación será evaluado por iRECIST (aplicable a Subestudios 3 y 4).
    • En el subestudio 4, la DCR, DOR, PFS y SG de los pacientes tratados con derazantinib se comparará con el de los pacientes tratados con derazantinib-atezolizumab en combinación para generar potencial hipótesis para futuros estudios comparativos.
    • La DCR, DOR, PFS y SG de los pacientes tratados con derazantinib y derazantinib-atezolizumab en combinación, respectivamente, serán evaluado por perfil molecular, perfil de expresión génica y biomarcadores.
    • La proporción de pacientes con una reducción específica de macrófagos inmunosupresores en biopsias de piel o sangre periférica monocitos en monoterapia con derazantinib y derazantinib-atezolizumab tratamiento combinado.
    • Los datos del estado de salud de EQ-5D (5L) se utilizarán para obtener servicios medidas para la modelización económica.
    Para el EQ-5D (5L), mayor puntuación
    y el análisis se puede informar en un documento separado.
    Puntos finales exploratorios de PK
    • Concentraciones plasmáticas de derazantinib, evaluadas mediante mediciones en
    muestras de sangre.
    • Concentraciones plasmáticas de metabolitos de derazantinib (si corresponde),
    evaluado mediante mediciones de muestras de sangre.
    • Concentraciones séricas de atezolizumab, evaluadas mediante mediciones de
    muestras de sangre (Subestudio 3, Cohorte 4b y Crossover).
    • ADA sérico analizado a partir de muestras de sangre (subestudio 3, cohorte 4b y
    Transversal).
    • Concentraciones plasmáticas de derazantinib (Cmax, tmax, AUC0-24, AUClast),
    evaluado mediante mediciones de muestras de sangre (aplicable a
    Subestudios con 300 mg QD)
    E.5.2.1Timepoint(s) of evaluation of this end point
    DCR, as measured by the proportion of patients with confirmed CR, PR or
    stable disease (SD) by BICR
    DoR will be calculated from the first date of documented tumor response
    (CR or PR) to the date of disease progression by BICR.
    PFS will be calculated as the time from cohort assignment by IWRS until
    disease progression per RECIST 1.1, or death from any cause.
    OS time will be calculated from the date of cohort assignment by IWRS
    until death from any cause. Any patient without a date of death in the
    database at the time the survival analyses are performed will be
    censored at the time of their last study contact.
    DCR, medida por la proporción de pacientes con RC, PR o
    enfermedad estable (SD) por BICR
    La DoR se calculará a partir de la primera fecha de respuesta tumoral documentada
    (CR o PR) hasta la fecha de progresión de la enfermedad por BICR.
    La SLP se calculará como el tiempo transcurrido desde la asignación de cohorte por el IWRS hasta
    progresión de la enfermedad según RECIST 1.1, o muerte por cualquier causa.
    El tiempo de SG se calculará a partir de la fecha de asignación de cohorte por IWRS
    hasta la muerte por cualquier causa.
    Cualquier paciente sin fecha de fallecimiento en el base de datos en el momento en que se realizan los análisis de supervivencia será
    censurados en el momento de su último contacto con el estudio.
    E.6 and E.7 Scope of the trial
    E.6Scope of the trial
    E.6.1Diagnosis No
    E.6.2Prophylaxis No
    E.6.3Therapy No
    E.6.4Safety Yes
    E.6.5Efficacy Yes
    E.6.6Pharmacokinetic Yes
    E.6.7Pharmacodynamic Yes
    E.6.8Bioequivalence No
    E.6.9Dose response No
    E.6.10Pharmacogenetic Yes
    E.6.11Pharmacogenomic Yes
    E.6.12Pharmacoeconomic No
    E.6.13Others Yes
    E.6.13.1Other scope of the trial description
    -Tolerability,
    -To describe the type of FGFR1–3 GAs in responders and non-responders
    -To explore the concordance between molecular FGFR assessments from
    plasma-based and tissue-based NGS testing
    -Tolerabilidad
    -Describir el tipo de Ag de FGFR!-3 en respuestas y no respuestas
    -Explorar la concordancia entre las evaluaciones moleculares de FGFR de pruebas de NGS basadas en plasma y en tejidos
    E.7Trial type and phase
    E.7.1Human pharmacology (Phase I) Yes
    E.7.1.1First administration to humans No
    E.7.1.2Bioequivalence study No
    E.7.1.3Other Yes
    E.7.1.3.1Other trial type description
    phase 1b/2
    Fase 1b/2
    E.7.2Therapeutic exploratory (Phase II) Yes
    E.7.3Therapeutic confirmatory (Phase III) No
    E.7.4Therapeutic use (Phase IV) No
    E.8 Design of the trial
    E.8.1Controlled No
    E.8.1.1Randomised No
    E.8.1.2Open Yes
    E.8.1.3Single blind No
    E.8.1.4Double blind No
    E.8.1.5Parallel group No
    E.8.1.6Cross over No
    E.8.1.7Other Yes
    E.8.1.7.1Other trial design description
    5 subestudios, abierto,en el subestudio 3 y 4 se ofrece diseño cruzado.
    5 sub-studies, open label, in sub-study 3 and 4 cross over design offered
    E.8.2 Comparator of controlled trial
    E.8.2.1Other medicinal product(s) No
    E.8.2.2Placebo No
    E.8.2.3Other No
    E.8.2.4Number of treatment arms in the trial2
    E.8.3 The trial involves single site in the Member State concerned No
    E.8.4 The trial involves multiple sites in the Member State concerned Yes
    E.8.4.1Number of sites anticipated in Member State concerned8
    E.8.5The trial involves multiple Member States Yes
    E.8.5.1Number of sites anticipated in the EEA63
    E.8.6 Trial involving sites outside the EEA
    E.8.6.1Trial being conducted both within and outside the EEA Yes
    E.8.6.2Trial being conducted completely outside of the EEA No
    E.8.6.3If E.8.6.1 or E.8.6.2 are Yes, specify the regions in which trial sites are planned
    Australia
    Canada
    Korea, Republic of
    United States
    Austria
    France
    Poland
    Spain
    Switzerland
    Czechia
    Germany
    Italy
    Belgium
    Hungary
    United Kingdom
    E.8.7Trial has a data monitoring committee Yes
    E.8.8 Definition of the end of the trial and justification where it is not the last visit of the last subject undergoing the trial
    The study ends when the last patient completes the last study-related phone-call or visit, discontinues from the study or is lost to follow-up (i.e. the patient is unable to be contacted by the investigator).
    El estudio finaliza cuando el último paciente completa la última llamada telefónica o visita relacionada con el estudio, se retira del estudio o se pierde durante el seguimiento (es decir, el investigador no puede contactar al paciente).
    E.8.9 Initial estimate of the duration of the trial
    E.8.9.1In the Member State concerned years3
    E.8.9.1In the Member State concerned months0
    E.8.9.1In the Member State concerned days0
    E.8.9.2In all countries concerned by the trial years3
    E.8.9.2In all countries concerned by the trial months0
    E.8.9.2In all countries concerned by the trial days0
    F. Population of Trial Subjects
    F.1 Age Range
    F.1.1Trial has subjects under 18 No
    F.1.1.1In Utero No
    F.1.1.2Preterm newborn infants (up to gestational age < 37 weeks) No
    F.1.1.3Newborns (0-27 days) No
    F.1.1.4Infants and toddlers (28 days-23 months) No
    F.1.1.5Children (2-11years) No
    F.1.1.6Adolescents (12-17 years) No
    F.1.2Adults (18-64 years) Yes
    F.1.2.1Number of subjects for this age range: 177
    F.1.3Elderly (>=65 years) Yes
    F.1.3.1Number of subjects for this age range: 95
    F.2 Gender
    F.2.1Female Yes
    F.2.2Male Yes
    F.3 Group of trial subjects
    F.3.1Healthy volunteers No
    F.3.2Patients Yes
    F.3.3Specific vulnerable populations Yes
    F.3.3.1Women of childbearing potential not using contraception No
    F.3.3.2Women of child-bearing potential using contraception Yes
    F.3.3.3Pregnant women No
    F.3.3.4Nursing women No
    F.3.3.5Emergency situation No
    F.3.3.6Subjects incapable of giving consent personally No
    F.3.3.7Others No
    F.4 Planned number of subjects to be included
    F.4.1In the member state25
    F.4.2 For a multinational trial
    F.4.2.1In the EEA 204
    F.4.2.2In the whole clinical trial 272
    F.5 Plans for treatment or care after the subject has ended the participation in the trial (if it is different from the expected normal treatment of that condition)
    If the patient continue to benefit from receiving derazantinib according to the study doctor, the Sponsor may assess the possibility of providing continued individual patients access to derazantinib (eg, in the context of compassionate use).
    Si el paciente continúa beneficiándose de recibir derazantinib de acuerdo con el médico del estudio, el Patrocinador puede evaluar la posibilidad de proporcionar a los pacientes individuales acceso a derazantinib (por ejemplo, en el contexto del uso compasivo).
    G. Investigator Networks to be involved in the Trial
    N. Review by the Competent Authority or Ethics Committee in the country concerned
    N.Competent Authority Decision Authorised
    N.Date of Competent Authority Decision2020-02-11
    N.Ethics Committee Opinion of the trial applicationFavourable
    N.Ethics Committee Opinion: Reason(s) for unfavourable opinion
    N.Date of Ethics Committee Opinion2019-11-08
    P. End of Trial
    P.End of Trial StatusOngoing
    For support, Contact us.
    The status and protocol content of GB trials is no longer updated since 1 January 2021. For the UK, as of 31 January 2021, EU Law applies only to the territory of Northern Ireland (NI) to the extent foreseen in the Protocol on Ireland/NI. Legal notice
    As of 31 January 2023, all EU/EEA initial clinical trial applications must be submitted through CTIS . Updated EudraCT trials information and information on PIP/Art 46 trials conducted exclusively in third countries continues to be submitted through EudraCT and published on this website.

    European Medicines Agency © 1995-Mon May 13 15:44:56 CEST 2024 | Domenico Scarlattilaan 6, 1083 HS Amsterdam, The Netherlands
    EMA HMA