IXAZOMIB Plus Lenalidomide and Dexamethasone Versus Placebo Plus Lenalidomide and Dexamethasone in Adult Patients With Newly Diagnosed Multiple Myeloma

July 20, 2023 updated by: Millennium Pharmaceuticals, Inc.

A Phase 3, Randomized, Double-Blind, Multicenter Study Comparing Oral MLN9708 Plus Lenalidomide and Dexamethasone Versus Placebo Plus Lenalidomide and Dexamethasone in Adult Patients With Newly Diagnosed Multiple Myeloma

The purpose of this study is to provide continued access to ixazomib and/or lenalidomide to participants who are continuing to have clinical benefit and to continue collecting relevant safety data to monitor safety in participants with Newly Diagnosed Multiple Myeloma (NDMM) who are not eligible for stem cell transplant.

Study Overview

Study Type

Interventional

Enrollment (Actual)

705

Phase

  • Phase 3

Contacts and Locations

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

Study Locations

      • Bruges, Belgium
      • Liege, Belgium
    • Brussels
      • Brussel, Brussels, Belgium
      • Bruxelles, Brussels, Belgium
    • Namur
      • Yvoir, Namur, Belgium
    • Oost-Vlaanderen
      • Gent, Oost-Vlaanderen, Belgium
    • Vlaams Brabant
      • Leuven, Vlaams Brabant, Belgium
    • West-Vlaanderen
      • Roeselare, West-Vlaanderen, Belgium
    • Alberta
      • Calgary, Alberta, Canada
      • Edmonton, Alberta, Canada
    • British Columbia
      • Surrey, British Columbia, Canada
      • Victoria, British Columbia, Canada
    • New Brunswick
      • St. John, New Brunswick, Canada
    • Nova Scotia
      • Halifax, Nova Scotia, Canada
    • Ontario
      • Hamilton, Ontario, Canada
      • London, Ontario, Canada
      • Toronto, Ontario, Canada
    • Quebec
      • Fleurimont, Quebec, Canada
      • Levis, Quebec, Canada
      • Montreal, Quebec, Canada
      • Amiens, France
      • Bayonne, France
      • Bordeaux, France
      • Caen, France
      • Chalon sur Saone, France
      • Creteil, France
      • Dunkerque, France
      • La Roche sur Yon, France
      • Le Chesnay, France
      • Le Mans cedex 2, France
      • Lille, France
      • Montpellier cedex 5, France
      • Mulhouse, France
      • Paris, France
      • Perigueux, France
      • Pierre Benite, France
      • Pontoise, France
      • Rouen, France
      • TOULOUSE Cedex 9, France
      • Tours, France
    • Alpes-Maritimes
      • Nice, Alpes-Maritimes, France
    • Bas-Rhin
      • Strasbourg, Bas-Rhin, France
    • Finistere
      • Brest, Finistere, France
      • Saint-Brieuc, Finistere, France
    • Haute-Vienne
      • Limoges, Haute-Vienne, France
    • Herault
      • Castelnau-le-Lez, Herault, France
    • Isere
      • La Tronche, Isere, France
    • Loire-Atlantique
      • Nantes, Loire-Atlantique, France
    • Maine-et-Loire
      • Angers, Maine-et-Loire, France
    • Marne
      • Reims, Marne, France
    • Meurthe-et-Moselle
      • Vandoeuvre-les-nancy, Meurthe-et-Moselle, France
    • Morbihan
      • Vannes, Morbihan, France
    • Nord
      • Lille Cedex, Nord, France
    • Pas-de-Calais
      • Lens, Pas-de-Calais, France
    • Rhone
      • Saint-priest-en-jarez, Rhone, France
    • Seine-Maritime
      • Montivilliers, Seine-Maritime, France
    • Vienne
      • Poitiers, Vienne, France
      • Daegu, Korea, Republic of
      • Daejeon, Korea, Republic of
      • Incheon, Korea, Republic of
      • Jeongnam, Korea, Republic of
      • Seoul, Korea, Republic of
    • Gyeonggido
      • Goyang, Gyeonggido, Korea, Republic of
      • Seongnam-si, Gyeonggido, Korea, Republic of
      • Hamilton, New Zealand
    • North Island
      • Auckland, North Island, New Zealand
      • Wellington, North Island, New Zealand
    • South Island
      • Christchurch, South Island, New Zealand
      • Dunedin, South Island, New Zealand
      • Ryazan, Russian Federation
    • Alabama
      • Birmingham, Alabama, United States
    • Arizona
      • Chandler, Arizona, United States
      • Tucson, Arizona, United States
    • Arkansas
      • Little Rock, Arkansas, United States
    • California
      • Anaheim, California, United States
      • Corona, California, United States
      • Fountain Valley, California, United States
      • Fullerton, California, United States
      • Irvine, California, United States
      • La Jolla, California, United States
      • Mission Hills, California, United States
      • Oakland, California, United States
      • Riverside, California, United States
      • San Diego, California, United States
      • San Jose, California, United States
      • San Leandro, California, United States
      • Santa Clara, California, United States
      • South San Francisco, California, United States
      • Vallejo, California, United States
    • Colorado
      • Denver, Colorado, United States
      • Fort Collins, Colorado, United States
    • Connecticut
      • Hartford, Connecticut, United States
      • Plainville, Connecticut, United States
    • Florida
      • Fort Myers, Florida, United States
      • Jacksonville, Florida, United States
      • Miami, Florida, United States
      • Miami Beach, Florida, United States
      • West Palm Beach, Florida, United States
    • Georgia
      • Atlanta, Georgia, United States
      • Marietta, Georgia, United States
    • Hawaii
      • Honolulu, Hawaii, United States
    • Iowa
      • Iowa City, Iowa, United States
      • Sioux City, Iowa, United States
    • Kentucky
      • Louisville, Kentucky, United States
    • Louisiana
      • Shreveport, Louisiana, United States
    • Maryland
      • Baltimore, Maryland, United States
      • Towson, Maryland, United States
    • Massachusetts
      • Burlington, Massachusetts, United States
      • Worcester, Massachusetts, United States
    • Michigan
      • Ann Arbor, Michigan, United States
      • Lansing, Michigan, United States
      • Southfield, Michigan, United States
    • Minnesota
      • Duluth, Minnesota, United States
      • Minneapolis, Minnesota, United States
      • Rochester, Minnesota, United States
    • Missouri
      • Bridgeton, Missouri, United States
      • Kansas City, Missouri, United States
    • New Jersey
      • Little Silver, New Jersey, United States
    • New Mexico
      • Albuquerque, New Mexico, United States
      • Farmington, New Mexico, United States
      • Las Cruces, New Mexico, United States
    • New York
      • Buffalo, New York, United States
      • Lake Success, New York, United States
      • New York, New York, United States
      • Poughkeepsie, New York, United States
    • Ohio
      • Cincinnati, Ohio, United States
      • Cleveland, Ohio, United States
    • Oklahoma
      • Tulsa, Oklahoma, United States
    • Oregon
      • Eugene, Oregon, United States
      • Portland, Oregon, United States
    • Pennsylvania
      • Hershey, Pennsylvania, United States
      • Pittsburgh, Pennsylvania, United States
      • Scranton, Pennsylvania, United States
    • South Carolina
      • Charleston, South Carolina, United States
    • Tennessee
      • Cookeville, Tennessee, United States
      • Nashville, Tennessee, United States
    • Texas
      • Austin, Texas, United States
      • Galveston, Texas, United States
      • San Antonio, Texas, United States
      • Temple, Texas, United States
      • Tyler, Texas, United States
    • Virginia
      • Christiansburg, Virginia, United States
      • Richmond, Virginia, United States
    • Washington
      • Everett, Washington, United States
      • Seattle, Washington, United States
      • Spokane, Washington, United States
      • Vancouver, Washington, United States
      • Yakima, Washington, United States

Participation Criteria

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

Eligibility Criteria

Ages Eligible for Study

18 years and older (Adult, Older Adult)

Accepts Healthy Volunteers

No

Description

Inclusion Criteria:

  1. Male or female participants 18 years or older diagnosed with Multiple Myeloma according to standard criteria who have not received prior treatment for multiple myeloma.
  2. Participants for whom lenalidomide and dexamethasone treatment is appropriate and who are not eligible for high-dose therapy followed by stem-cell transplantation (HDT-SCT) for 1 or more of the following reasons:

    • The participant is 65 years of age or older.
    • The participant is less than 65 years of age but has significant comorbid condition(s) that are, in the opinion of the investigator, likely to have a negative impact on tolerability of HDT-SCT.
  3. Measurable disease as specified in study protocol.
  4. Eastern Cooperative Oncology Group (ECOG) performance status of 0 to 2.
  5. Meet the clinical laboratories criteria as specified in the protocol.
  6. Female participants who are post menopausal, surgically sterile, or agree to practice 2 effective methods of contraception or agree to practice true abstinence, and must also agree to ongoing pregnancy testing; must also adhere to the guidelines of the lenalidomide pregnancy prevention program.
  7. Male participants who agree to practice effective barrier contraception or agree to practice true abstinence AND must adhere to the guidelines of the lenalidomide pregnancy prevention program.
  8. Suitable venous access for the study-required blood sampling.
  9. Must be able to take concurrent aspirin 70 mg to 325 mg daily (or enoxaparin if aspirin allergic).
  10. Voluntary written consent.
  11. Participant is willing and able to adhere to the study visit schedule and other protocol requirements.

Exclusion Criteria:

  1. Prior treatment for multiple myeloma with either standard of care treatment or investigational regimen.
  2. Diagnosed and treated for another malignancy within 5 years before randomization or previously diagnosed with another malignancy and have any evidence of residual disease. Participants with nonmelanoma skin cancer or carcinoma in situ of any type are not excluded if they have undergone complete resection.
  3. Inability or unwillingness to receive antithrombotic therapy.
  4. Female participants who are lactating or pregnant.
  5. Major surgery or radiotherapy within 14 days before randomization.
  6. Infection requiring intravenous antibiotics within 14 days before the first dose of study drug.
  7. Central nervous system involvement.
  8. Diagnosis of Waldenstrom's macroglobulinemia, polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes (POEMS) syndrome, plasma cell leukemia, primary amyloidosis, myelodysplastic syndrome, or myeloproliferative syndrome.
  9. Evidence of current uncontrolled cardiovascular conditions within 6 months prior to randomization, including: Uncontrolled hypertension, cardiac arrhythmias, or congestive heart failure; Unstable angina, or Myocardial infarction.
  10. Systemic treatment with strong inhibitors of CYP1A2 (fluvoxamine, enoxacin, ciprofloxacin), strong inhibitors of CYP3A (clarithromycin, telithromycin,itraconazole, voriconazole, ketoconazole, nefazodone, posaconazole) or strong CYP3A inducers (rifampin, rifapentine, rifabutin, carbamazepine, phenytoin, phenobarbital), or use of Ginkgo biloba or St. John's wort within 14 days before randomization in the study.
  11. Active hepatitis B or C virus infection, or known human immunodeficiency virus (HIV) positive.
  12. Comorbid systemic illnesses or other severe concurrent disease which, in the judgment of the investigator, would make the participant inappropriate for entry into this study or interfere significantly with the proper assessment of safety and toxicity of the prescribed regimens (e.g., peripheral neuropathy that is Grade 1 with pain or Grade 2 or higher of any cause).
  13. Psychiatric illness/social situation that would limit compliance with study requirements.
  14. Known allergy to any of the study medications.
  15. Inability to swallow oral medication, inability or unwillingness to comply with the drug administration requirements, or gastrointestinal (GI) procedure that could interfere with the oral absorption or tolerance of treatment.
  16. Treatment with any investigational products within 60 days before randomization.

Study Plan

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

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Randomized
  • Interventional Model: Parallel Assignment
  • Masking: Quadruple

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Placebo Comparator: Placebo + LenDex
Participants who were randomly assigned to receive placebo matching capsule single oral dose on Days 1, 8 and 15 along with standard regimen of LenDex (lenalidomide 25 mg capsules orally on Days 1-21 and dexamethasone 40 mg tablets orally on Days 1, 8, 15 and 22) for the first 18 cycles (each cycle was of 28 days). Following Cycle 18, participants received 3.0 mg ixazomib matching placebo capsule as single oral dose on Days 1, 8 and 15 along with lenalidomide 10 mg capsules orally on Days 1-21 in each 28-day cycle until progressive disease or unacceptable toxicity, whichever comes first up to end of study (up to approximately 109 months).
Dexamethasone tablets.
Lenalidomide capsules.
IXAZOMIB matching-placebo capsules.
Active Comparator: Active Comparator: Ixazomib + LenDex
Participants who were randomly assigned to receive Ixazomib 4.0 mg capsule single oral dose on Days 1, 8 and 15 along with standard regimen of LenDex (lenalidomide 25 mg capsules orally on Days 1-21 and dexamethasone 40 mg tablets orally on Days 1, 8, 15 and 22) for the first 18 cycles (each cycle was of 28 days). Following Cycle 18, participants received 3.0 mg ixazomib capsule as single oral dose on Days 1, 8 and 15 along with lenalidomide 10 mg capsules orally on Days 1-21 in each 28-day cycle until progressive disease or unacceptable toxicity, whichever comes first up to end of study (up to approximately 109 months).
Dexamethasone tablets.
Lenalidomide capsules.
IXAZOMIB capsules.
Other Names:
  • MLN9708

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Progression Free Survival (PFS)
Time Frame: Up to approximately 79 months
PFS was defined as the time from the date of randomization to the date of first documentation of progressive disease (PD) or death due to any cause according to International Myeloma Working Group (IMWG) criteria whichever occurs first. PD required one of the following: Increase of >=25% from nadir in: Serum M-component and/or (the absolute increase must be >=0.5 g/dL); Urine M-component and/or (the absolute increase must be >=200 mg/24 hours); in participants without measurable serum and urine M-protein levels: the difference between involved and uninvolved free light chain (FLC) levels (absolute increase must be > 10 mg/dL); Bone marrow plasma cell percentage: the absolute % must be >10%; development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; hypercalcemia (corrected serum calcium > 11.5 mg/dL or 2.85 mmol/L).
Up to approximately 79 months

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Overall Survival (OS)
Time Frame: From the date of randomization to death due to any cause (Up to approximately 9 years)
OS was defined as the time from the date of randomization to the date of death. Participants without documented death at the time of analysis are censored at the date last known to be alive.
From the date of randomization to death due to any cause (Up to approximately 9 years)
Complete Response (CR) Rate
Time Frame: Up to approximately 9 years
CR rate was defined as the percentage of participants who achieve CR assessed by an IRC relative to the intent-to-treat (ITT) population during the treatment period. Percentage of participants with CR, as assessed by IMWG disease assessment criteria were reported. CR was defined as negative immunofixation of serum and urine along with the disappearance of any soft tissue plasmacytomas and <5 % plasma cells (PC's) in bone marrow.
Up to approximately 9 years
Pain Response Rate as Assessed by the Brief Pain Inventory- Short Form (BPI-SF) and Analgesic Use
Time Frame: Up to approximately 9 years
Pain response rate was defined as percentage of participants with pain response. Pain response was defined as the occurrence of at least a 30% reduction from baseline in BPI-SF worst pain score over the last 24 hours without an increase in analgesic use for 2 consecutive measurements > 28 days apart, were reported. Brief Pain Inventory - Short Form (m-BPI-SF) is a participant rated 11-point Likert rating scale ranged from 0 (no pain) to 10 (worst pain imaginable). Percentages are rounded off to the nearest single decimal.
Up to approximately 9 years
Overall Response Rate (ORR)
Time Frame: Up to approximately 9 years
ORR was defined as the percentage of participants who achieved CR + partial response (PR) + very good partial response (VGPR) (including sCR) or better relative to the ITT population during treatment period. CR was defined as negative immunofixation of serum and urine along with the disappearance of any soft tissue plasmacytomas and <5 % PC's in bone marrow. PR was defined as ≥50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥90% along with ≥50% reduction in the size of soft tissue plasmacytomas. VGPR was defined as ≥90% in serum M-component plus urine M-component <100 mg/24. sCR is defined as stringent complete response. Percentages are rounded off to nearest whole numbers.
Up to approximately 9 years
Time to Response
Time Frame: Up to approximately 9 years
Time to response was defined as the time from the date of randomization to the first documentation of PR or better, as measured by IMWG criteria.
Up to approximately 9 years
Duration of Response
Time Frame: Up to approximately 9 years
Duration of response was measured as the time from the date of first documentation of PR or better to the date of first documented progression (PD) for responders, as measured by IMWG criteria.
Up to approximately 9 years
Time to Progression (TTP)
Time Frame: Up to approximately 9 years
Time to progression was defined as the time from randomization to the date of first documented disease progression.
Up to approximately 9 years
Progression Free Survival (PFS)-2
Time Frame: Up to approximately 9 years
PFS2 was defined as the time from the date of randomization to the date of documentation of disease progression on the subsequent line of anticancer therapy, as assessed by the investigator in accordance with IMWG criteria, or death due to any cause, whichever occurs first.
Up to approximately 9 years
Number of Participants With Shifts From Baseline to Worst Value in Eastern Cooperative Oncology Group (ECOG) Performance Score
Time Frame: Up to approximately 9 years
Eastern Cooperative Oncology Group (ECOG) scale score ranged from 0 to 5, where 0 indicated normal activity and 5 indicated death. The data is reported for those categories where at least 1 participant had worst post-baseline value for each ECOG score.
Up to approximately 9 years
Number of Participants With Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)
Time Frame: From the date of randomization through 30 days after the last dose of study drug up to end of study (up to approximately 9 years)
An AE was any untoward medical occurrence in a participant administered a medicinal investigational drug. The untoward medical occurrence does not necessarily have to have a causal relationship with treatment. An SAE is any untoward medical occurrence that results in death; is life-threatening; requires inpatient hospitalization or prolongation of present hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect or is a medically important event that may not be immediately life-threatening or result in death or hospitalization, but may jeopardize the participant or may require intervention to prevent one of other outcomes listed in definition above, or involves suspected transmission via a medicinal product of an infectious agent.
From the date of randomization through 30 days after the last dose of study drug up to end of study (up to approximately 9 years)
Number of Participants With Abnormal Serum Chemistry and Hematology Laboratory Values Based on Treatment-emergent Adverse Events (TEAEs)
Time Frame: From the date of randomization through 30 days after the last dose of study drug up to end of study (up to approximately 9 years)
The laboratory values assessment included serum chemistry and hematology. The Serum chemistry assessment included blood urea nitrogen (BUN), creatinine, bilirubin (total), urate, lactate dehydrogenase, phosphate, albumin, alkaline phosphatase (ALP), aspartate aminotransferase (AST), alanine aminotransferase (ALT), glucose, sodium, potassium, calcium, chloride, carbon dioxide (CO2), magnesium, thyroid stimulating hormone (TSH). Hematology assessment included hemoglobin, hematocrit, platelet (count), leukocytes with differential neutrophils (ANC). Participants with abnormal serum chemistry laboratory values reported as TEAEs are reported. TEAEs were defined as events that occurred after administration of the first dose of any agent in the study drug regimen and through 30 days after the last dose of any agent in the study drug regimen.
From the date of randomization through 30 days after the last dose of study drug up to end of study (up to approximately 9 years)
Change From Baseline in Health-Related Quality of Life (HRQOL) Measured by European Organisation for Research and Treatment of Cancer-Quality of Life Questionnaire (EORTC-QLQ)-C30 Scale Total Score
Time Frame: Baseline to approximately 9 years
EORTC-QLQ-C30 scale was used to assess HRQOL in cancer participants and contains 30 items. Subscale with individual items include physical functioning items 1-5, role functioning items 6-7, emotional functioning items 21-24, cognitive functioning items 20, 25, social functioning items 26-27, quality of life items 29-30, fatigue items 10, 12, 18, nausea and vomiting items 14-15, pain items 9, 19, dyspnoea item 8, insomnia item 11, appetite loss item 13, constipation item 16, diarrhoea item 17, financial difficulties item 28. Raw scores were converted into scale scores ranging from 0 to 100. For the functional scales and the global health status scale, higher scores represent better HRQOL; whereas for the symptom scales lower scores represent better HRQOL. Positive change in functional and global health status scale indicated improvement; negative change for the symptom scales indicates improvement.
Baseline to approximately 9 years
Change From Baseline in HRQOL Measured by EORTC-QLQ-MY20 Scale
Time Frame: Baseline to approximately 9 years
EORTC QLQ-MY20 was a validated questionnaire to assess the overall quality of life in participants with multiple myeloma. The scale has 20 questions. Subscale and individual items include future perspective items 18-20, body image item 17, disease symptoms items 1-6, side effects of treatment items 7-16. Raw scores are averaged, and transformed to 0-100 scale, where higher score is better quality of life. Positive change indicates improvement.
Baseline to approximately 9 years
OS in High-risk Population Carrying Del(17p), t(4;14), or t(14;16) Mutations
Time Frame: From the date of randomization to death due to any cause (Up to approximately 9 years)
OS was defined as the time from the date of randomization to the date of death, as assessed in high-risk population carrying del(17p), t(4;14), or t(14;16) mutations. High risk category includes t(4;14), t(14;16), or del(17) abnormalities.
From the date of randomization to death due to any cause (Up to approximately 9 years)
PFS in High-risk Population Carrying Del(17p), t(4;14), or t(14;16) Mutations
Time Frame: Up to approximately 9 years
PFS was defined as the time from the date of randomization to the date of first documentation of progressive disease based on central laboratory results and IMWG criteria as evaluated by an independent review committee (IRC) or death due to any cause, whichever occurs first, as assessed in high-risk population carrying del(17p), t(4;14), or t(14;16) mutations.
Up to approximately 9 years
Percentage of Participants With MRD-Negative Status as Assessed by Flow Cytometry
Time Frame: Up to Cycle 18 (cycle length = 28 days)
The absence of minimal residual disease (MRD negativity) was tested in all participants who achieve a CR and maintained it until Cycle 18, using bone marrow aspirates.
Up to Cycle 18 (cycle length = 28 days)
Time to Pain Progression
Time Frame: Up to approximately 9 years
Time to pain progression was assessed as the time from randomization to the date of initial progression classification. Pain progression was defined as the occurrence of 1 of the following and confirmed by 2 consecutive evaluations (To qualify as progression, the participant must have a BPI-SF worst pain score > 4 during pain progression): 1) a ≥ 2 point and 30% increase from Baseline in BPI-SF worst pain score without an increase in analgesic use, or 2) a 25% or more increase in analgesic use from Baseline without a decrease in BPI-SF worst pain score from Baseline. Brief Pain Inventory - Short Form (m-BPI-SF) is a participant rated 11-point Likert rating scale ranged from 0 (no pain) to 10 (worst pain imaginable).
Up to approximately 9 years
Cmax: Maximum Plasma Concentration for Ixazomib
Time Frame: Cycle 1 Day 1: Post-dose at multiple timepoints up to 4 hours; Pre-dose at Cycle 1 Day 14, Cycles 2-3 Day 1 and Day 14, Cycles 4-11 Day 1 (Each cycle length = 28 days)
Cycle 1 Day 1: Post-dose at multiple timepoints up to 4 hours; Pre-dose at Cycle 1 Day 14, Cycles 2-3 Day 1 and Day 14, Cycles 4-11 Day 1 (Each cycle length = 28 days)
Percentage of Participants With New or Worsening of Existing Skeletal-related Events (SREs)
Time Frame: From the date of randomization through 30 days after the last dose of study drug up to end of study (up to approximately 9 years)
SRE is defined as new fractures [including vertebral compression fractures], irradiation of or surgery on bone, or spinal cord compression.
From the date of randomization through 30 days after the last dose of study drug up to end of study (up to approximately 9 years)

Collaborators and Investigators

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

Investigators

  • Study Director: Medical Director Clinical Science, Millennium Pharmaceuticals, Inc.

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

Helpful Links

Study record dates

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

Study Major Dates

Study Start (Actual)

April 29, 2013

Primary Completion (Actual)

December 2, 2019

Study Completion (Actual)

June 24, 2022

Study Registration Dates

First Submitted

April 26, 2013

First Submitted That Met QC Criteria

May 6, 2013

First Posted (Estimated)

May 9, 2013

Study Record Updates

Last Update Posted (Actual)

July 21, 2023

Last Update Submitted That Met QC Criteria

July 20, 2023

Last Verified

July 1, 2023

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

YES

IPD Plan Description

Takeda provides access to the de-identified individual participant data (IPD) for eligible studies to aid qualified researchers in addressing legitimate scientific objectives (Takeda's data sharing commitment is available on https://clinicaltrials.takeda.com/takedas-commitment?commitment=5). These IPDs will be provided in a secure research environment following approval of a data sharing request, and under the terms of a data sharing agreement.

IPD Sharing Access Criteria

IPD from eligible studies will be shared with qualified researchers according to the criteria and process described on https://vivli.org/ourmember/takeda/. For approved requests, the researchers will be provided access to anonymized data (to respect patient privacy in line with applicable laws and regulations) and with information necessary to address the research objectives under the terms of a data sharing agreement.

IPD Sharing Supporting Information Type

  • STUDY_PROTOCOL
  • SAP
  • ICF
  • CSR

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

Yes

Studies a U.S. FDA-regulated device product

No

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

Clinical Trials on Multiple Myeloma

Clinical Trials on Dexamethasone

3
Subscribe