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   43874   clinical trials with a EudraCT protocol, of which   7293   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:2016-003957-14
    Sponsor's Protocol Code Number:KCP-330-023
    National Competent Authority:Spain - AEMPS
    Clinical Trial Type:EEA CTA
    Trial Status:Completed
    Date on which this record was first entered in the EudraCT database:2017-03-10
    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 number2016-003957-14
    A.3Full title of the trial
    A Phase 3 Randomized, Controlled, Open-label Study of Selinexor, Bortezomib, and Dexamethasone (SVd) versus Bortezomib and Dexamethasone (Vd) in Patients with Relapsed or Refractory Multiple Myeloma (RRMM)
    Estudio de fase III, aleatorizado, controlado y abierto sobre selinexor, bortezomib y dexametasona (SVd) frente a bortezomib y dexametasona (Vd) en pacientes con mieloma múltiple recidivado o resistente al tratamiento (MMRR)
    A.3.1Title of the trial for lay people, in easily understood, i.e. non-technical, language
    Bortezomib, Selinexor and Dexamethasone in Patients with Multiple Myeloma
    Bortezomib, selinexor y dexametasona en pacientes con mieloma múltiple
    A.3.2Name or abbreviated title of the trial where available
    BOSTON
    BOSTON
    A.4.1Sponsor's protocol code numberKCP-330-023
    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 SponsorKaryopharm Therapeutics Inc.
    B.1.3.4CountryUnited States
    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 supportKaryopharm Therapeutics Inc.
    B.4.2CountryUnited States
    B.5 Contact point designated by the sponsor for further information on the trial
    B.5.1Name of organisationKaryopharm Therapeutics Inc.
    B.5.2Functional name of contact pointClinical Trial Information Desk
    B.5.3 Address:
    B.5.3.1Street Address85 Wells Ave
    B.5.3.2Town/ cityNewton
    B.5.3.3Post codeMA 02459
    B.5.3.4CountryUnited States
    B.5.6E-mailclinicaltrials@karyopharm.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 Yes
    D.2.5.1Orphan drug designation numberEU/3/14/1355
    D.3 Description of the IMP
    D.3.1Product nameSelinexor
    D.3.2Product code KPT-330
    D.3.4Pharmaceutical form Film-coated tablet
    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 INNSelinexor
    D.3.9.1CAS number 1393477-72-9
    D.3.9.2Current sponsor codeKPT-330
    D.3.9.3Other descriptive nameSELINEXOR
    D.3.9.4EV Substance CodeSUB177942
    D.3.10 Strength
    D.3.10.1Concentration unit mg milligram(s)
    D.3.10.2Concentration typeequal
    D.3.10.3Concentration number20
    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.2.5The 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 nameBortezomib
    D.3.4Pharmaceutical form Powder for solution for injection
    D.3.4.1Specific paediatric formulation No
    D.3.7Routes of administration for this IMPSubcutaneous use
    D.3.8 to D.3.10 IMP Identification Details (Active Substances)
    D.3.8INN - Proposed INNBORTEZOMIB
    D.3.9.1CAS number 179324-69-7
    D.3.9.3Other descriptive namebortezomib
    D.3.9.4EV Substance CodeSUB20020
    D.3.10 Strength
    D.3.10.1Concentration unit mg milligram(s)
    D.3.10.2Concentration typeequal
    D.3.10.3Concentration number3.5
    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: 3
    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.2.5The 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 nameDexamethasone
    D.3.4Pharmaceutical form Tablet
    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 INNDEXAMETHASONE
    D.3.9.1CAS number 50-02-2
    D.3.9.3Other descriptive nameDexamethasone
    D.3.9.4EV Substance CodeSUB07017MIG
    D.3.10 Strength
    D.3.10.1Concentration unit mg milligram(s)
    D.3.10.2Concentration typeequal
    D.3.10.3Concentration number4
    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: 4
    D.1.2 and D.1.3IMP RoleComparator
    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.2.5The 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 nameBortezomib
    D.3.4Pharmaceutical form Powder for solution for injection
    D.3.4.1Specific paediatric formulation No
    D.3.7Routes of administration for this IMPSubcutaneous use
    D.3.8 to D.3.10 IMP Identification Details (Active Substances)
    D.3.8INN - Proposed INNBORTEZOMIB
    D.3.9.1CAS number 179324-69-7
    D.3.9.3Other descriptive namebortezomib
    D.3.9.4EV Substance CodeSUB20020
    D.3.10 Strength
    D.3.10.1Concentration unit mg milligram(s)
    D.3.10.2Concentration typeequal
    D.3.10.3Concentration number3.5
    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: 5
    D.1.2 and D.1.3IMP RoleComparator
    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.2.5The 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 nameDexamethasone
    D.3.4Pharmaceutical form Tablet
    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 INNDEXAMETHASONE
    D.3.9.1CAS number 50-02-2
    D.3.9.3Other descriptive nameDexamethasone
    D.3.9.4EV Substance CodeSUB07017MIG
    D.3.10 Strength
    D.3.10.1Concentration unit mg milligram(s)
    D.3.10.2Concentration typeequal
    D.3.10.3Concentration number4
    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.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
    Relapsed or refractory multiple myeloma (RRMM)
    Mieloma múltiple recidivado o resistente al tratamiento (MMRR)
    E.1.1.1Medical condition in easily understood language
    Multiple myeloma
    Mieloma múltiple
    E.1.1.2Therapeutic area Diseases [C] - Blood and lymphatic diseases [C15]
    MedDRA Classification
    E.1.2 Medical condition or disease under investigation
    E.1.2Version 19.1
    E.1.2Level LLT
    E.1.2Classification code 10028228
    E.1.2Term Multiple myeloma
    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
    - To compare PFS based on the Independent Review Committee’s (IRC’s) disease outcome assessments in patients randomized to the SVd Arm versus the Vd Arm.
    - To compare the ORR (>= PR) based on the IRC’s response outcome assessments, in patients randomized to the SVd Arm versus the Vd Arm.
    - Comparar la supervivencia libre de progresión (SLP) basada en las evaluaciones del desenlace clínico de la enfermedad por parte del Comité de revisión independiente (CRI) en los pacientes asignados aleatoriamente al grupo de tratamiento con SVd frente al grupo de tratamiento con Vd.
    - Comparar la tasa de respuesta global (TRG) (>= respuesta parcial [RP]) basada en las evaluaciones de las respuestas por parte del CRI en los pacientes asignados aleatoriamente al grupo de tratamiento con SVd frente al grupo de tratamiento con Vd.
    E.2.2Secondary objectives of the trial
    -To compare the incidence of any >= Grade 2 neuropathy events (total >= Grade 2 and separately for Grades 2, 3, and 4) in patients randomized to the SVd Arm vs patients randomized to the Vd Arm
    -To compare the no of patients with response >= very good partial response (VGPR), >=complete response (CR), >= stringent complete response (sCR), or minimal residual disease (MRD) negative (for patients who achieve CR or sCR) in patients randomized to the SVd Arm versus the Vd Arm
    -To compare the DOR in patients randomized to the SVd Arm vs the Vd Arm
    -To compare OS in patients randomized to the SVd Arm vs patients randomized to the Vd Arm who do not cross over to SVdX (OS1)
    -To compare ORR, PFS, and DOR for patients with 1 prior anti-MM regimen versus > 1 prior anti-MM regimen in patients randomized to the SVd Arm vs the Vd Arm
    -To determine ORR1 (ORR during SVdX treatment)
    -To determine PFS1 (PFS during SVdX treatment)
    - Comparar la incidencia de todos los acontecimientos de neuropatía de grado >=2 (total de grado >=2 y por separado para los grados 2, 3 y 4) en los pacientes asignados aleatoriamente al grupo con SVd frente al grupo con Vd.
    - Comparar el número de pacientes con respuesta >= RPMB, >= RC, >= RCr o ERM negativa (para los pacientes que alcanzan RC o RCr) en los pacientes asignados aleatoriamente al grupo de tratamiento con SVd frente al grupo Vd.
    - Comparar DR en los pacientes asignados aleatoriamente al grupo con SVd frente al grupo Vd.
    - Comparar SG en los pacientes asignados aleatoriamente al grupo con SVd frente al grupo con Vd que no han cambiado de grupo para recibir SVdX (SG1).
    - Comparar la TRG, la SLP y la DR en pacientes con 1 tratamiento previo contra el MM frente a >1 tratamiento previo contra el MM en los pacientes asignados al grupo con SVd frente al grupo Vd
    - Determinar TRG1 (TRG durante el tratamiento con SVdX)
    - Determinar SLP1 (SLP durante el tratamiento con SVdX)
    E.2.3Trial contains a sub-study No
    E.3Principal inclusion criteria
    1. Histologically confirmed MM with measurable disease per IMWG guidelines as defined by at least 1 of the following:
    a. Serum M-protein >= 0.5 g/dL (> 5 g/L) by serum protein electrophoresis (SPEP) or for immunoglobulin (Ig) A myeloma, by quantitative serum IgA levels; or
    b. Urinary M-protein excretion at least 200 mg/24 hours; or
    c. Serum free light chain (FLC) >= 100 mg/L, provided that the serum FLC ratio is abnormal (normal FLC ratio: 0.26 to 1.65).
    2. Had at least 1 prior anti-MM regimen and no more than 3 prior anti-MM regimens. Induction therapy followed by stem cell transplant and consolidation/maintenance therapy will be considered as 1 anti-MM regimen.
    3. Documented evidence of progressive MM (based on the Investigator's determination according to the modified IMWG response criteria) on or after their most recent regimen.
    4. Prior treatment with bortezomib or other PI is allowed, provided all of the following criteria are met:
    -Best response achieved with prior bortezomib at any time was >= PR and with the last PI therapy (alone or in combination) was >= PR, AND
    -Participant did not discontinue bortezomib due to >= Grade 3 related toxicity, AND
    -Must have had at least a 6-month PI-treatment-free interval prior to C1D1 of study treatment.
    5. Must have an ECOG Status score of 0, 1, or 2.
    6. Written informed consent in accordance with federal, local, and institutional guidelines.
    7. Age >= 18 years.
    8. Resolution of any clinically significant non-hematological toxicities (if any) from previous treatments to =< Grade 1 by C1D1. Patients with chronic, stable Grade 2 non-hematological toxicities may be included following approval from the Medical Monitor.
    9. Adequate hepatic function within 28 days prior to C1D1:
    a. Total bilirubin < 1.5 × upper limit of normal (ULN) (except patients with Gilbert’s syndrome who must have a total bilirubin of < 3 × ULN), and b. Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) normal to < 2 × ULN.
    10. Adequate renal function within 28 days prior to C1D1 (estimated creatinine clearance [CrCl] of >= 20 mL/min, calculated using the formula of Cockroft and Gault): (140-Age) × Mass (kg)/(72 × creatinine mg/dL) Multiply by 0.85 if the patient is female, or if CrCl is >= 20 mL/min as measured by 24-hour urine collection.
    11. Adequate hematopoietic function within 7 days prior to C1D1: total white blood cell (WBC) count >=1500/mm3, absolute neutrophil count >= 1000/mm3, hemoglobin >= 8.5 g/dL and platelet count >=75,000/mm3 (patients for whom < 50% of bone marrow nucleated cells are plasma cells) or >= 50,000/mm3 (patients for whom >= 50% of bone marrow nucleated cells are plasma cells).
    a. Patients receiving hematopoietic growth factor support, including erythropoietin, darbepoetin, granulocyte-colony stimulating factor (G-CSF), granulocyte macrophage-colony stimulating factor (GM-CSF), and platelet stimulators (e.g., eltrombopag, romiplostim, or interleukin-11) must have a 2-week interval between growth factor support and the Screening assessments, but they may receive growth factor support during the study.
    b. Patients must have:
    -At least a 2-week interval from the last red blood cell (RBC) transfusion prior to the Screening hemoglobin assessment, and
    -At least a 1-week interval from the last platelet transfusion prior to the Screening platelet assessment.
    However, patients may receive RBC and/or platelet transfusions as clinically indicated per institutional guidelines during the study.
    12. Female patients of childbearing potential must agree to use 2 methods of contraception (including 1 highly effective and 1 effective method of contraception) and have a negative serum pregnancy test at Screening. Male patients must use an effective barrier method of contraception if sexually active with a female of childbearing potential. For both male and female patients, effective methods of contraception must be used throughout the study and for 3 months following the last dose of study treatment.
    1. MM confirmada histológicamente, con enfermedad medible según las directrices del IMWG, definida por al menos uno de los criterios siguientes:
    a. Proteína M sérica >= 0,5 g/dl (>5 g/l) según electroforesis de proteínas en suero (EPS), o en caso del mieloma de inmunoglobulina (Ig) A, mediante los niveles de IgA séricos cuantitativos; o
    b. Excreción de la proteína M en orina de al menos 200 mg/24 horas; o
    c. Cadena ligera libre sérica (CLL) >= 100 mg/l si la relación de CLL sérica es anómala (relación de CLL normal: entre 0,26 y 1,65).
    2. Al menos 1 tratamiento previo contra el MM y no más de 3 tratamientos previos contra el MM. El tratamiento de inducción seguido por el trasplante de células progenitoras y el tratamiento de consolidación o mantenimiento se considerarán 1 tratamiento contra el MM.
    3. Signos confirmados de progresión del MM (basados en la determinación del investigador según los criterios de respuesta del IMWG modificados) durante o después del tratamiento más reciente.
    4. Se permite el tratamiento previo con bortezomib u otro IP si se cumplen todos los criterios siguientes:
    - Mejor respuesta alcanzada con el tratamiento previo con bortezomib en cualquier momento >= RP y con el último tratamiento con un IP (en monoterapia o en combinación) >= RP, Y
    - El participante no suspendió el tratamiento con bortezomib por toxicidad relacionada de grado >=3, Y
    - Debe haber transcurrido un intervalo sin tratamiento con IP de al menos 6 meses antes del C1D1 del tratamiento del estudio.
    5. Puntuación del estado funcional del ECOG de 0, 1 o 2.
    6. Consentimiento informado por escrito de acuerdo con las normas estatales, regionales y del centro.
    7. Edad >=18 años.
    8. Resolución de todas las toxicidades no hematológicas clínicamente significativas (si las había) de tratamientos previos a un grado =<1 en el C1D1. Los pacientes con toxicidades no hematológicas de grado 2 estables y crónicas pueden incluirse después de la aprobación del monitor médico.
    9. Función hepática aceptable en los 28 días previos al C1D1:
    a. Bilirrubina total <1,5 × límite superior de la normalidad (LSN) (excepto los pacientes con síndrome de Gilbert que deben presentar una bilirrubina total de <3 × LSN), y
    b. Aspartato-transaminasa (AST) y alanina-transaminasa (ALT) normales o <2 × LSN.
    10. Función renal aceptable en los 28 días previos al C1D1 (aclaramiento de creatinina [CrCl] estimado de >=20 ml/min, calculado mediante la fórmula de Cockroft y Gault):
    (140-edad) × masa (kg)/(72 × creatinina mg/dl)
    Multiplicar por 0,85 en el caso de mujeres, o si el CrCl es >=20 ml/min según la medición en orina recogida durante 24 horas.
    11. Función hematopoyética aceptable en los 7 días previos al C1D1: número total de leucocitos (LEU) >=1500/mm3, recuento absoluto de neutrófilos >=1000/mm3, hemoglobina >=8,5 g/dl y número de plaquetas >=75 000/mm3 (pacientes en los cuales <50 % de las células nucleadas de la médula ósea son células plasmáticas) o >=50 000/mm3 (pacientes en los cuales >=50 % de las células nucleadas de la médula ósea son células plasmáticas).
    a. Los pacientes que reciben tratamiento complementario con factores de crecimiento hematopoyético, incluidos eritropoyetina, darbepoetina, factor estimulante de colonias de granulocitos (G-CSF), factor estimulante de colonias de granulocitos y macrófagos (GM-CSF) y estimuladores de plaquetas (p. ej., eltrombopag, romiplostim o interleucina 11) deben esperar un intervalo de 2 semanas entre el tratamiento con los factores de crecimiento y las evaluaciones de selección, pero pueden recibir tratamiento complementario con factores de crecimiento durante el estudio.
    b. Los pacientes deben esperar:
    - Un intervalo de al menos 2 semanas desde la última transfusión de eritrocitos antes de la evaluación de la hemoglobina de la selección, y
    - Un intervalo de al menos 1 semana desde la última transfusión de plaquetas antes de la evaluación de plaquetas de la selección.
    Sin embargo, los pacientes pueden recibir transfusiones de eritrocitos o de plaquetas según indicación clínica y conforme a las normas del centro durante el estudio.
    12. Las mujeres con capacidad de procrear deben aceptar la utilización de 2 métodos anticonceptivos (incluidos un método anticonceptivo muy eficaz y uno eficaz) y deben presentar una prueba de embarazo en suero negativa en la selección. Los pacientes varones deberán utilizar un método anticonceptivo de barrera si tienen actividad sexual con una mujer con capacidad de procrear. Tanto los varones como las mujeres deberán utilizar métodos anticonceptivos eficaces durante el estudio y hasta 3 meses después de la última dosis del tratamiento del estudio.
    E.4Principal exclusion criteria
    1. Has received selinexor or another XPO1 inhibitor previously.
    2. Prior malignancy that required treatment, or has shown evidence of recurrence (except for non-melanoma skin cancer or adequately treated cervical carcinoma in situ) during the 5 years prior to randomization. Cancer treated with curative intent for > 5 years
    previously and without evidence of recurrence will be allowed.
    3. Has any concurrent medical condition or disease (e.g., uncontrolled active hypertension, uncontrolled active diabetes, active systemic infection, etc.) that is likely to interfere with study procedures.
    4. Uncontrolled active infection requiring parenteral antibiotics, antivirals, or antifungals within 1 week prior to C1D1. Patients on prophylactic antibiotics or with a controlled infection within 1 week prior to C1D1 are acceptable.
    5. Active plasma cell leukemia.
    6. Documented systemic light chain amyloidosis.
    7. MM involving the central nervous system.
    8. Polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skinchanges (POEMS) syndrome.
    9. Spinal cord compression.
    10. Greater than Grade 2 neuropathy or >= Grade 2 neuropathy with pain at baseline, regardless of whether or not the patient is currently receiving medication.
    11. Intolerance, hypersensitivity, or contraindication to glucocorticoids.
    12. Radiation, chemotherapy, or immunotherapy or any other anticancer therapy =< 2 weeks prior to C1D1. Localized radiation to a single site at least 1 week before C1D1 is permitted. Glucocorticoids within 2 weeks of C1D1 are permitted. Patients on long-term glucocorticoids during Screening do not require a washout period but must be able to tolerate the specified dexamethasone dose in this study.
    13. Prior autologous stem cell transplantation < 1 month or allogeneic stem cell transplantation < 4 months prior to C1D1.
    14. Active graft versus host disease (after allogeneic stem cell transplantation) at C1D1.
    15. Pregnant or breastfeeding females.
    16. BSA < 1.4 m2 at baseline, calculated by the Dubois (Dubois 1916) or Mosteller (Mosteller 1987) method.
    17. Life expectancy of < 4 months.
    18. Major surgery within 4 weeks prior to C1D1.
    19. Active, unstable cardiovascular function:
    a. Symptomatic ischemia, or
    b. Uncontrolled clinically significant conduction abnormalities (e.g., patients with ventricular tachycardia on anti-arrhythmics are excluded; patients with first-degree atrioventricular block or asymptomatic left anterior fascicular block/right bundle branch block will not be excluded), or
    c. Congestive heart failure of New York Heart Association Class >= 3 or known left ventricular ejection fraction < 40%, or
    d. Myocardial infarction within 3 months prior to C1D1.
    20. Known active human immunodeficiency virus (HIV) infection or HIV seropositivity.
    21. Known active hepatitis A, B, or C infection; or known to be positive for hepatitis C virus ribonucleic acid (RNA) or hepatitis B virus surface antigen.
    22. Any active gastrointestinal dysfunction interfering with the patient’s ability to swallow tablets, or any active gastrointestinal dysfunction that could interfere with absorption of study treatment.
    23. Any active, serious psychiatric, medical, or other conditions/situations that, in the opinion of the Investigator, could interfere with treatment, compliance, or the ability to give informed consent.
    24. Contraindication to any of the required concomitant drugs or supportive treatments.
    25. Patients unwilling or unable to comply with the protocol, including providing 24-hour urine samples for urine protein electrophoresis at the required time points.
    1. Paciente que ha recibido selinexor u otro inhibidor XPO1 previamente.
    2. Paciente con neoplasia maligna previa que precisó tratamiento o que ha mostrado signos de recurrencia (excepto cáncer de piel no melanocítico o carcinoma cervical in situ tratado satisfactoriamente) durante los 5 años previos a la aleatorización. Se permitirá la participación de pacientes con cáncer tratado con intención curativa >5 años previamente y sin signos de recurrencia.
    3. Paciente con un estado clínico o enfermedad (p. ej., hipertensión activa no controlada, diabetes activa no controlada, infección sistémica activa, etc.) que pueda interferir en los procedimientos del estudio.
    4. Infección activa no controlada que necesite tratamiento parenteral con antibióticos, antivirales o antifúngicos en la semana previa al C1D1. Se aceptan los pacientes en tratamiento profiláctico con antibióticos o con una infección controlada en la semana previa al C1D1.
    5. Leucemia de células plasmáticas activa.
    6. Amiloidosis de cadena ligera sistémica confirmada.
    7. MM con afectación del sistema nervioso central.
    8. Síndrome POEMS (polineuropatía, organomegalia, endocrinopatía, gammapatía monoclonal y alteraciones cutáneas).
    9. Compresión medular.
    10. Neuropatía superior a grado 2 o de grado >=2 con dolor basal, independientemente de si el paciente recibe medicación actualmente.
    11. Intolerancia, hipersensibilidad o contraindicación al tratamiento con glucocorticoides.
    12. Radiación, quimioterapia, inmunoterapia o cualquier otro tratamiento antineoplásico =<2 semanas antes del C1D1. Se permite la radiación localizada en una localización única al menos 1 semana antes del C1D1. Se permite el tratamiento con glucocorticoides en las dos semanas previas al C1D1. Los pacientes en tratamiento a largo plazo con glucocorticoides durante la selección no necesitan un periodo de reposo farmacológico, pero deben ser capaces de tolerar la dosis de dexametasona especificada en este estudio.
    13. Autotrasplante de células progenitoras previo <1 mes o alotrasplante de células progenitoras <4 meses antes del C1D1.
    14. Enfermedad de injerto contra huésped activa (después de alotrasplante de células progenitoras) el C1D1.
    15. Mujeres embarazadas o en período de lactancia.
    16. SC <1,4 m2 en el periodo basal, calculada mediante el método de Dubois (Dubois 1916) o Mosteller (Mosteller 1987).
    17. Esperanza de vida <4 meses.
    18. Cirugía mayor en las 4 semanas previas al C1D1.
    19. Función cardiovascular inestable activa:
    a. Isquemia sintomática, o
    b. Anomalías de la conducción significativas no controladas clínicamente (p. ej., se excluyen los pacientes con taquicardia ventricular en tratamiento con antiarrítmicos; no se excluirán los pacientes con bloqueo auriculoventricular de primer grado o bloqueo fascicular anterior izquierdo/bloqueo de rama derecha del haz de His), o
    c. Insuficiencia cardíaca congestiva de la clase >= 3 de la New York Heart Association o fracción de expulsión del ventrículo izquierdo conocida <40 %, o
    d. Infarto de miocardio en los 3 meses anteriores al C1D1.
    20. Infección por el virus de la inmunodeficiencia humana (VIH) activa conocida o seropositividad para el VIH.
    21. Infección activa por el virus de la hepatitis A, B o C conocida; o positividad conocida para el ácido ribonucleico (ARN) del virus de la hepatitis C o antígeno de superficie del virus de la hepatitis B.
    22. Cualquier disfunción gastrointestinal activa que interfiera en la capacidad del paciente para tragar comprimidos o cualquier disfunción gastrointestinal activa que pudiera interferir en la absorción del tratamiento del estudio.
    23. Cualquier enfermedad o situación activa, psiquiátrica grave, médica u otras, que, según el investigador, podría interferir en el tratamiento, el cumplimiento o la capacidad para otorgar el consentimiento informado.
    24. Contraindicación de alguno de los fármacos concomitantes o tratamientos complementarios requeridos.
    25. Pacientes que no estén dispuestos o no sean capaces de cumplir con el protocolo, incluido proporcionar muestras de orina de 24 horas para electroforesis de proteínas en orina en los momentos requeridos.
    E.5 End points
    E.5.1Primary end point(s)
    - PFS, defined as time from date of randomization until the first date of PD, per IMWG response criteria, or death due to any cause, whichever occurs first. For the purposes of PFS determination, PD will be determined by the IRC.
    - ORR, defined as any response >= PR based on the IRC’s response outcome assessments, according to the IMWG response criteria.
    - SLP, definido como el tiempo transcurrido desde la fecha de aleatorización hasta la primera fecha de PE (según los criterios de respuesta del IMWG) o la muerte por cualquier causa (lo que suceda primero). El CRI evaluará centralmente la PE para el criterio principal de valoración de la SLP.
    - TRG definido como >= RP basada en las evaluaciones de la respuesta del CRI, en función de los criterios de respuesta del IMWG.
    E.5.1.1Timepoint(s) of evaluation of this end point
    ORR for the primary analysis will be assessed on the ITT population after the last patient randomized has had the opportunity to complete at least 2 post-C1D1 MM evaluations
    La TRG para el análisis principal se evaluará en la población IDT después de que el último paciente asignado aleatoriamente haya tenido la oportunidad de completar al menos dos evaluaciones del mieloma múltiple después del día 1 del ciclo 1.
    E.5.2Secondary end point(s)
    Secondary efficacy endpoints:
    - Response rates at any time prior to PD or death due to any cause, pooled and separately for the following responses: >= VGPR, >= CR >= sCR, or MRD negative (for patients who achieve CR or sCR)
    -DOR, defined as the duration of time from first occurrence of response >= PR until the first date of PD or death, whichever occurs first
    -OS1, defined as time to death, measured from date of randomization until death due to any cause, for patients randomized to the Vd Arm who do not cross over to SVdX
    -ORR, PFS, and DOR (for patients with 1 prior anti-MM regimen versus > 1 prior anti-MM regimen)
    -ORR1 (ORR for SVdX patients)
    -PFS1 (PFS for SVdX patients), defined as the duration of time from date of first dose of SVd treatment after crossover from the Vd Arm until the first date of PD, or death due to any cause.
    -TTNT, defined as duration of time from date of last dose of study treatment until the date of first dose of post-SVd/Vd/SVdX treatment.
    -TTR, defined as time from the date of first dose of study treatment until the date of first documented response (any response and best response) per IMWG response criteria.
    -PFS2 (PFS for patients who receive post-SVd/Vd/SVdX treatment), defined as the duration of time from the date of first dose of post-SVd/Vd/SVdX treatment until the first date of PD on post-SVd/Vd/SVdX treatment, or death due to any cause.
    Secondary Safety Endpoints:
    -Incidence of all grades and any >= Grade 2 neuropathy events (total >= Grade 2 and separately for Grades 2, 3, and 4) in patients randomized to the SVd Arm versus patients randomized to the Vd Arm. The incidence of any >= Grade 2 neuropathy events will be compared between the SVd Arm and the Vd Arm (using only events that occurred prior to crossover) as a secondary endpoint using the safety population.
    -Safety and tolerability of study treatment based on AE reports, physical examination results (including vital signs), Eastern Cooperative Oncology Group (ECOG) performance status score, 12-lead electrocardiogram (ECG) results, ophthalmic examination results, and clinical laboratory results.
    Secondary HR-QoL Endpoint
    -Patient-reported peripheral neuropathy, as measured by the EORTC-QLQ-CIPN20 instrument
    Exploratory Endpoints
    -OS, measured from date of randomization until death due to any cause, for all randomized patients
    -PFS (in patient subsets based on R-ISS criteria [Palumbo 2015], International Staging System (ISS) criteria, and single cytogenetic alterations included in the R-ISS plus 1q21 amplifications)
    -ORR (in patient subsets based on R-ISS criteria, ISS criteria, and single cytogenetic alterations included in the R-ISS plus 1q21 amplifications)
    -Treatment discontinuation rate
    -HR-QoL, as measured by the EORTC-QLQ-C30 and the EQ-5D-5L instruments
    Variables secundarias de eficacia:
    - Las tasas de respuesta en cualquier momento anterior a la PE o muerte por cualquier causa, combinados y por separado para las siguientes respuestas: >= RPMB, >= RC, >= RCr o ERM negativa (para los pacientes que alcanzan RC o RCr)
    -Duración de la respuesta, que se define como el tiempo transcurrido desde la primera aparición de la respuesta> = a la respuesta parcial hasta la primera fecha de la PE o la muerte, lo que ocurra primero.
    -SG1, definido como el tiempo hasta la muerte, medido desde la randomización hasta la muerte debido a cualquier causa, para los pacientes asignados al grupo de tratamiento Vd que no cruzan a SVdX.
    -TRG, SLP y DR (para pacientes con 1 tratamiento anti-MM previo versus > 1 tratamiento anti-MM previo)
    -TRG1 (TRG para pacientes del grupo SVdX)
    -SLP1 (SLP para pacientes del grupo SVdX), definida como la duración del tiempo desde la fecha de la primera dosis de tratamiento SVd después del cruce del grupo Vd hasta la primera fecha de Progresión de la Enfermedad o muerte por cualquier causa.
    - Tiempo transcurrido hasta el tratamiento siguiente (TTS) , definido como el tiempo desde la fecha de la última dosis de tratamiento del estudio hasta la fecha de la primera dosis de tratamiento post-SVd / Vd / SVdX.
    - Tiempo transcurrido hasta obtener respuesta (THR) , definido como el tiempo desde la fecha de la primera dosis del tratamiento del estudio hasta la fecha de la primera respuesta documentada (cualquier respuesta y mejor respuesta) según los criterios de respuesta del IMWG.
    - SLP2 (Supervivencia Libre de Progresión para los pacientes que reciben tratamiento post-SVd / Vd / SVdX), definida como el tiempo desde la fecha de la primera dosis de tratamiento post-SVd / Vd / SVdX hasta la primera fecha de PE con tratamiento post-SVd / Vd / SVdX o muerte por cualquier causa.
    Variables secundarias de seguridad:
    - Incidencia de episodios de neuropatía de todos los grados y de Grado >= 2 (total >= Grado 2 y por separado para Grados 2, 3 y 4) en pacientes asignados al grupo SVd versus los pacientes asignados al grupo Vd. La incidencia de cualquier episodio de neuropatía >= grado 2 se comparará entre el grupo SVd y el grupo Vd (utilizando sólo los episodios que ocurrieron antes del cruce) como un criterio de valoración secundario utilizando la población de seguridad.
    -Seguridad y tolerabilidad del tratamiento del estudio basado en los informes de Efectos Adversos, los resultados del examen físico (incluyendo signos vitales), puntuación del estado de rendimiento según el Eastern Cooperative Oncology Group (ECOG), resultados del electrocardiograma de 12 derivaciones (ECG), resultados de exploración oftalmológica y resultados de laboratorio clínico.
    Variable secundaria relativa a la calidad de vida:
    - Neuropatía periférica reportada por un paciente, medida según el EORTC-QLQ-CIPN20
    Variables de exploración:
    -Supervivencia Global (SG), medida desde la fecha de randomización hasta la muerte por cualquier causa, para todos los pacientes randomizados
    -SLP (en subgrupos de pacientes basados en criterios R-ISS [Palumbo 2015], criterios del Sistema Internacional de Estadificación (ISS) y alteraciones citogenéticas únicas incluidas en las amplificaciones R-ISS más 1q21)
    - TRG (en subgrupos de pacientes basados en criterios R-ISS, criterios ISS y alteraciones citogenéticas únicas incluidas en las amplificaciones R-ISS más 1q21)
    -Tasa de discontinuidad del tratamiento
    -Valoración de la calidad de vida medida con los cuestionarios EORTC-QLQ-C30 y EQ-5D-5L
    E.5.2.1Timepoint(s) of evaluation of this end point
    as per protocol
    Según protocolo
    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 Yes
    E.6.4Safety Yes
    E.6.5Efficacy Yes
    E.6.6Pharmacokinetic No
    E.6.7Pharmacodynamic No
    E.6.8Bioequivalence No
    E.6.9Dose response No
    E.6.10Pharmacogenetic No
    E.6.11Pharmacogenomic No
    E.6.12Pharmacoeconomic No
    E.6.13Others No
    E.7Trial type and phase
    E.7.1Human pharmacology (Phase I) No
    E.7.1.1First administration to humans No
    E.7.1.2Bioequivalence study No
    E.7.1.3Other No
    E.7.1.3.1Other trial type description
    E.7.2Therapeutic exploratory (Phase II) No
    E.7.3Therapeutic confirmatory (Phase III) Yes
    E.7.4Therapeutic use (Phase IV) No
    E.8 Design of the trial
    E.8.1Controlled Yes
    E.8.1.1Randomised Yes
    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 Yes
    E.8.1.7Other No
    E.8.2 Comparator of controlled trial
    E.8.2.1Other medicinal product(s) No
    E.8.2.2Placebo No
    E.8.2.3Other Yes
    E.8.2.3.1Comparator description
    sVd versus Vd
    sVd versus Vd
    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 EEA100
    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
    Austria
    Belgium
    Bulgaria
    Canada
    Czech Republic
    France
    Germany
    Greece
    Hungary
    Israel
    Italy
    Netherlands
    Poland
    Romania
    Russian Federation
    Serbia
    Spain
    Ukraine
    United Kingdom
    United States
    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
    LVLS
    Última visita del último paciente
    E.8.9 Initial estimate of the duration of the trial
    E.8.9.1In the Member State concerned years
    E.8.9.1In the Member State concerned months33
    E.8.9.1In the Member State concerned days
    E.8.9.2In all countries concerned by the trial months33
    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: 124
    F.1.3Elderly (>=65 years) Yes
    F.1.3.1Number of subjects for this age range: 240
    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 state44
    F.4.2 For a multinational trial
    F.4.2.1In the EEA 284
    F.4.2.2In the whole clinical trial 364
    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)
    After a patient has ceased his/her participation in the study, his/her medical doctor will offer the most appropriate treatment currently available.
    Después de que un paciente haya cesado su participación en el estudio, su médico le ofrecerá el tratamiento disponible más apropiado.
    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 Decision2017-04-12
    N.Ethics Committee Opinion of the trial applicationFavourable
    N.Ethics Committee Opinion: Reason(s) for unfavourable opinion
    N.Date of Ethics Committee Opinion2017-04-07
    P. End of Trial
    P.End of Trial StatusCompleted
    P.Date of the global end of the trial2022-05-12
    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-Thu May 09 02:05:37 CEST 2024 | Domenico Scarlattilaan 6, 1083 HS Amsterdam, The Netherlands
    EMA HMA