Clinical Trials Logo

Clinical Trial Details — Status: Active, not recruiting

Administrative data

NCT number NCT03301220
Other study ID # CR108172
Secondary ID 54767414SMM30012
Status Active, not recruiting
Phase Phase 3
First received
Last updated
Start date November 7, 2017
Est. completion date July 11, 2025

Study information

Verified date April 2024
Source Janssen Research & Development, LLC
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

The primary objective of this study is to determine whether treatment with daratumumab administered subcutaneously (SC) prolongs progression-free survival (PFS) compared with active monitoring in participants with high-risk smoldering multiple myeloma (SMM).


Description:

This study consists of 3 phases: Screening Phase (up to 35 days), an Active Monitoring Phase or a Treatment Phase of 39 cycles or 36 months (whichever occurs first), and a Follow-up Phase which will continue until death, lost to follow-up, consent withdrawal, or study end (approximately 8 years after the first participant is randomized), whichever occurs first. Active monitoring cycles and treatment cycles are 4 weeks in length. For all participants, disease evaluations will be performed every 12 weeks until confirmed progressive disease (PD). After PD, survival is to be followed at least every 6 months, until the end of the study. Participants will undergo tumor assessments, pharmacokinetics, biomarkers and safety evaluations (adverse events, laboratory tests, vital sign measurements, physical examinations, Eastern Cooperative Oncology Group [ECOG] performance status) over the time.


Recruitment information / eligibility

Status Active, not recruiting
Enrollment 390
Est. completion date July 11, 2025
Est. primary completion date May 1, 2024
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility Inclusion Criteria: - Diagnosis of high risk smoldering multiple myeloma (SMM) (per International Myeloma Working Group [IMWG] criteria) for less than or equal to (<=) 5 years with measurable disease at the time of randomization, defined as serum M protein greater than or equal to (>=) 10 gram per liter (g/L) or urine M protein >= 200 milligram per 24 hours (mg/24 hours) or involved serum free light chain (FLC) >=100 milligram per liter (mg/L) and abnormal serum FLC ratio - Clonal bone marrow plasma cells (BMPCs) >= 10 percentage (%); and at least 1 of the following risk factors; Serum M protein >= 30 g/L, immunoglobulin (Ig)A SMM, immunoparesis with reduction of 2 uninvolved immunoglobulin isotypes (only IgA, IgM, and IgG should be considered in determination for immunoparesis; IgD and IgE are not considered in this assessment), serum involved: uninvolved FLC ratio >= 8 and less than (<) 100, or clonal BMPCs greater than (>) 50% to <60% with measurable disease - Eastern Cooperative Oncology Group (ECOG) performance status score of 0 or 1 - Women of childbearing potential must commit to either abstain continuously from heterosexual sexual intercourse or to use highly effective method of contraception - A woman of childbearing potential must have a negative serum or urine pregnancy test at screening within 14 days prior to randomization - During the study and for 3 months after receiving the last dose of daratumumab, a woman must agree not to donate eggs (ova, oocytes) for the purposes of assisted reproduction Exclusion Criteria: - Multiple myeloma (MM), requiring treatment, defined by any of the following: 1. Bone lesions (1 or more osteolytic lesions on low-dose whole body computed tomography [LDCT], positron-emission tomography with computed tomography [PET-CT] or CT). Participants who have benign/post-traumatic bone lesions visible on screening images as well as previous imaging, may be considered for inclusion. Details (diagnosis, location, duration) on benign/post-traumatic pre-existing bone lesions that can be seen on the screening images (example [eg.], old fractures) and were also present on previous imaging are to be reported in the case report form (CRF) 2. Hypercalcemia (serum calcium greater than [>]0.25 millimoles per liter [mmol/L] [>1 milligram per deciliter {mg/dL}] higher than upper limit of normal [ULN] or >2.75 mmol/L [>11 mg/dL]). Participants who have clinically stable hypercalcemia attributable to a disease other than multiple myeloma (eg, hyperparathyroidism) may be considered for inclusion after a case by case review by the medical monitor 3. Renal insufficiency, preferably determined by creatinine clearance less than (<)40 milliliter per minute (mL/min) measured or estimated using the Modification of Diet in Renal Disease (MDRD), or serum creatinine >177 micromole per liter (µmol/L). Participants who have clinically stable renal insufficiency attributable to a disease other than multiple myeloma (eg, glomerulonephritis) may be considered for inclusion after a case by case review by the medical monitor 4. Anemia, defined as hemoglobin <10 gram per deciliter (g/dL) or >2 g/dL below lower limit of normal or both; transfusion support or concurrent treatment with erythropoietin stimulating agents is not permitted. Participants who have clinically stable anemia attributable to a disease other than multiple myeloma (eg, thalassemia, vitamin B12 deficiency, iron deficiency) may be considered for inclusion after a case by case review by the medical monitor 5. Clonal BMPC percentage >=60% 6. Serum FLC ratio (involved:uninvolved) >=100 (the involved FLC must be >=100 mg/L) 7. More than 1 focal lesion >=5 millimeter (mm) in diameter by magnetic resonance imaging (MRI) - Primary systemic amyloid light-chain (AL) (immunoglobulin light chain) amyloidosis - Exposure to any of the following: 1. Prior exposure to daratumumab or prior exposure to other anti-Cluster of Differentiation 38 (anti-CD38) therapies 2. Prior exposure to approved or investigational treatments for SMM or MM (including but not limited to conventional chemotherapies, immunomodulatory agent [IMiDs], or proteasome inhibitor [PIs]). Stable standard dosing of bisphosphonate and denosumab as indicated for osteoporosis is acceptable 3. Exposure to investigational drug (including investigational vaccines) or invasive investigational medical device for any indication within 4 weeks or 5 half-lives, whichever is longer, before Cycle 1, Day 1 4. Ongoing treatment with corticosteroids with a dose >10 milligram (mg) prednisone or equivalent per day at the time of randomization; or >280 mg cumulative prednisone dose or equivalent for any 4-week period in the year prior to randomization 5. Ongoing treatment with other monoclonal antibodies (eg, infliximab, rituximab), immunomodulators (eg, abatacept, methotrexate, azathioprine, cyclosporine) or other treatments that are likely to interfere with the study procedures or results - Received treatment (chemotherapy, surgery, et cetera [etc]) for a malignancy (other than SMM) within 3 years before the date of randomization (exceptions are squamous and basal cell carcinomas of the skin, carcinoma in situ of the cervix or breast, or other non-invasive lesion), which is considered cured with minimal risk of recurrence within 3 years - Medical or psychiatric condition or disease (for example, active systemic disease [including presence of auto-antibodies], uncontrolled diabetes) that is likely to interfere with the study procedures or results, or that in the opinion of the investigator, would constitute a hazard for participating in this study - Known allergies, hypersensitivity, or intolerance to corticosteroids, monoclonal antibodies, hyaluronidase, or other human proteins, or their excipients, or known sensitivity to mammalian-derived products (including dairy allergy)

Study Design


Intervention

Drug:
Daratumumab SC: daratumumab + rHuPH20
Participants will receive daratumumab SC injection (daratumumab 1800 mg + rHuPH20 [2000 U/mL]) once weekly for Cycles 1 and 2 (Days 1, 8, 15, and 22 of each week), every 2 weeks for Cycle 3 to Cycle 6 (Days 1 and 15), and thereafter every 4 weeks (Day 1) until 39 cycles or up to 36 months or until confirmed disease progression, unacceptable toxicity or withdrawal from the study treatment, study termination or study completion. Each cycle is 28 days in duration.

Locations

Country Name City State
Argentina Hospital Aleman Buenos Aires
Argentina Hospital Italiano de Buenos Aires Buenos Aires
Argentina CEMIC Saavedra Ciudad de Buenos Aires
Argentina Hospital Privado - Centro Medico de Cordoba Cordoba
Argentina Hospital Italiano de La Plata La Plata
Argentina Sanatorio Britanico de Rosario Rosario
Australia Austin Hospital Heidelberg
Australia Calvary Mater Newcastle Hospital Waratah
Australia The Perth Blood Institute West Perth
Australia Queen Elizabeth Hospital Woodville
Belgium ZNA Antwerpen
Belgium AZ St.-Jan Brugge-Oostende AV Brugge
Belgium UZBrussel Brussel
Belgium UZ Gent Gent
Belgium Virga Jessa Ziekenhuis Hasselt
Belgium Az Groeninge Kortrijk
Brazil Centro de Pesquisa e Ensino em Oncologia de Santa Catarina - CEPEN Florianopolis
Brazil Universidade Federal de Goias - Hospital das Clinicas da UFG Goiania
Brazil Instituto Joinvilense de Hematologia e Oncologia Ltda-Centro de Hematologia e Oncologia Joinville
Brazil Hospital das Clinicas de Porto Alegre Porto Alegre
Brazil Instituto de Educacao, Pesquisa e Gestao em Saude Instituto Americas (COI) Rio de Janeiro
Brazil Hospital Sao Rafael Salvador
Brazil Fundacao Faculdade Regional de Medicina de Sao Jose do Rio Preto - Hospital de Base Sao Jose do Rio Preto
Brazil Instituto de Ensino e Pesquisa São Lucas Sao Paulo
Brazil Clinica Sao Germano São Paulo
Brazil Hospital Santa Cruz São Paulo
Canada Tom Baker Cancer Center, University of Calgary Calgary Alberta
Canada Cross Cancer Institute Edmonton Alberta
Canada Lakeridge Health Oshawa Oshawa Ontario
Czechia Fakultni nemocnice Hradec Kralove Hradec Kralove
Czechia Fakultni nemocnice Ostrava Ostrava
Czechia Fakultni nemocnice Plzen, Hemato-onkologicke oddeleni Plzen
Czechia Vseobecna fakultni nemocnice v Praze - I. interni klinika - klinika hematologie Praha 2
Denmark Aarhus University Hospital Aarhus N.
Denmark Ålborg Universitetshospital Ålborg
Denmark Rigshospitalet Copenhagen
Denmark Odense Universitetshospital Odense C
France CHU de Limoges - Fédération Hépatologie Limoges
France Chu Hotel Dieu Nantes cedex 01
France CHU de Bordeaux - Hospital Haut-Leveque Pessac cedex
France Centre hospitalier Lyon-Sud Pierre Benite cedex
France CHU De Poitiers Poitiers
France l'Hôpital Pontchaillou Rennes
France CHU Bretonneau Tours Cedex 9
Germany Helios Kliniken Berlin Buch Gmbh Berlin
Germany St. Barbara-Klinik Hamm GmbH Hamm
Germany Universitaetsklinikum Heidelberg Medizinische Klinik V Heidelberg
Germany Medizinische Klinik A Muenster
Germany Universitaetsklinikum Tuebingen der Eberhard-Karls-Universitaet, Abteilung fuer Innere Medizin II, Tübingen
Germany Universitaetsklinikum Ulm Ulm
Greece Alexandra General Hospital of Athens Athens Attica
Hungary Del Pesti Centrumkorhaz Orszagos Hematologiai es Infektologiai Intezet Szent Laszlo Telephely Budapest
Hungary Semmelweis Egyetem I.Belgyogyaszati Klinika Budapest
Hungary Semmelweis Egyetem, I. Belgyogyaszati Klinika Budapest N/a
Hungary Debreceni Egyetem Klinikai Kozpont Debrecen
Israel Haemek Afula
Israel Barzilai Medical Center Ashkelon
Israel Bnai Zion Medical Center Haifa
Israel Carmel Medical Center Haifa
Israel Rambam Medical Center Haifa
Israel Hadassah Medical Center Jerusalem
Israel Galilee Medical Center Nahariya
Israel Rabin Medical Center Petah-Tiqva
Israel Sheba Medical Center Ramat Gan
Israel Sourasky Medical Center Tel-Aviv
Italy Policlinico Sant'Orsola Malpighi Bologna
Italy Businco Cancer Hospital Cagliari
Italy A.O. Santa Croce e Carle Cuneo
Italy Ospedale S. Eugenio Roma
Italy Università di Roma 'La Sapienza' - Ospedale Umberto 1° Roma
Italy A.O.U. Città della Salute e della Scienza di Torino- Divisione di Ematologia Torino
Italy ASST dei Sette Laghi, Ospedale di Circolo e Fonazione Macchi Varese
Japan Fukuoka University Hospital Fukuoka
Japan Chugoku Central Hospital Fukuyama
Japan Ogaki Municipal Hospital Gifu
Japan Kobe City Medical Center General Hospital Hyogo
Japan Kagoshima University Hospital Kagoshima
Japan Kanazawa University Hospital Kanazawa
Japan National Hospital Organization Osaka Minami Medical Center Kawachi-Nagano
Japan National Hospital Organization Kumamoto Medical Center Kumamoto-shi
Japan Kurume University Hospital Kurume
Japan Kyoto Kuramaguchi Medical Center Kyoto
Japan National Hospital Organization Matsumoto Medical Center Matsumoto
Japan Matsuyama Red Cross Hospital Matsuyama
Japan Nagoya City University Hospital Nagoya
Japan Niigata Cancer Center Hospital Niigata
Japan National Hospital Organization Okayama Medical Center Okayama
Japan National Hospital Organization Sendai Medical Center Sendai-City
Japan National Hospital Organization Shibukawa Medical Center Shibukawa
Japan Japanese Red Cross Medical Center Shibuya
Mexico iBiomed Research Unit Aguascalientes
Mexico JM Research, SC Cuernavaca
Mexico Centro de Investigación Farmacéutica Especializada Guadalajara
Mexico Centro de Atención e Investigación Clínica en Oncología Merida
Mexico Hospital Universitario de Nuevo León Monterrey
Netherlands Gelre Ziekenhuis Apeldoorn
Netherlands Haga ziekenhuis Den Haag
Netherlands Albert Schweitzer Ziekenhuis Dordrecht
Netherlands ETZ TweeSteden Tilburg
Norway Oslo University Hospital HF Ulleval sykehus Oslo
Poland Szpital Specjalistyczny w Brzozowie Podkarpacki Osrodek Onkologiczny im Ks B Markiewicza Brzozow
Poland Szpital Uniwersytecki nr 2 im. Jana Biziela w Bydgoszczy Bydgoszcz
Poland Wojewodzki Szpital Specjalistyczny w Legnicy Legnica
Poland Clinical Research Center sp z o o MEDIC R s k Poznan
Poland Instytut Hematologii i Transfuzjologii Warszawa
Russian Federation Emergency Hospital of Dzerzhinsk Dzerzhinsk
Russian Federation City Clinical Hospital # 40 Moscow
Russian Federation City clinical hospital n.a. S.P.Botkin Moscow
Russian Federation Nizhniy Novgorod Region Clinical Hospital Nizhny Novgorod
Russian Federation Perm Medical Sanitary Unit#1 Perm
Russian Federation Republican Hospital n.a.V.A.Baranov Petrozavodsk
Russian Federation Ryazan Regional Clinical Hospital Ryazan
Russian Federation Clinical Research Institute of Hematology and Transfusiology St-Petersburg
Russian Federation Oncology Dispensary of Komi Republic Syktyvkar
Spain Hosp. Univ. Germans Trias I Pujol Badalona
Spain Hosp. Clinic de Barcelona Barcelona
Spain Hosp. Univ. Vall D Hebron Barcelona
Spain Hosp. Gral. Univ. Gregorio Maranon Madrid
Spain Hosp. Univ. Infanta Leonor Madrid
Spain Hosp. Univ. Ramon Y Cajal Madrid
Spain Clinica Univ. de Navarra Pamplona
Spain Hosp. Quiron Madrid Pozuelo Pozuelo De Alarcon, Madrid
Spain Hosp. Clinico Univ. de Salamanca Salamanca
Spain Hosp. Univ. Dr. Peset Valencia
Sweden Falu Lasarett Falun
Sweden Sunderby Sjukhus Medicinkliniken Luelå
Sweden Karolinska Universitetssjukhuset Huddinge Stockholm
Turkey Ankara Numune Egitim ve Arastirma Hastanesi Ankara
Turkey Ankara University Medical Faculty Ankara
Turkey Trakya University Hospital Edirne
Turkey Istanbul University Istanbul Medical Faculty Istanbul
Turkey Erciyes University Medical Faculty Kayseri
Turkey On Dokuz Mayis University Medical Faculty Department of Hematology Samsun
United Kingdom Heart of England NHS Foundation Trust Birmingham
United Kingdom University Hospitals Bristol NHS Trust Bristol
United Kingdom Kent and Canterbury Hospital Canterbury
United Kingdom St Bartholomew's Hospital London
United Kingdom Christie Hospital NHS Trust Manchester
United Kingdom Nottingham University Hospitals NHS Trust Nottingham
United Kingdom Royal Stoke University Hospital Stoke-On-Trent
United States New York Oncology Hematology Albany New York
United States University of Michigan Comprehensive Cancer Center Ann Arbor Michigan
United States Emory University Atlanta Georgia
United States Texas Oncology P.A. Austin Texas
United States Dana Farber Cancer Institute Boston Massachusetts
United States University of North Carolina Chapel Hill North Carolina
United States Levine Cancer Institute, Carolinas HealthCare System Charlotte North Carolina
United States Cleveland Clinic Cleveland Ohio
United States The Ohio State University Columbus Ohio
United States VA North Texas Health Care System Dallas Texas
United States University of Iowa Hospitals & Clinics Iowa City Iowa
United States East Jefferson General Hospital Metairie Louisiana
United States Miami Cancer Institute Miami Florida
United States VA Southern Nevada Healthcare North Las Vegas Nevada
United States Fox Chase Cancer Center Philadelphia Pennsylvania
United States Arizona Oncology Associates, PC - HAL Phoenix Arizona
United States OHSU/CHM Portland Oregon
United States Mayo Clinic Rochester Minnesota
United States Washington University Saint Louis Missouri
United States University of Washington Seattle Washington
United States Stony Brook University Medical Center Stony Brook New York
United States Texas Oncology P.A. Tyler Texas
United States Innovative Clinical Research, Inc. Whittier California

Sponsors (1)

Lead Sponsor Collaborator
Janssen Research & Development, LLC

Countries where clinical trial is conducted

United States,  Argentina,  Australia,  Belgium,  Brazil,  Canada,  Czechia,  Denmark,  France,  Germany,  Greece,  Hungary,  Israel,  Italy,  Japan,  Mexico,  Netherlands,  Norway,  Poland,  Russian Federation,  Spain,  Sweden,  Turkey,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary Progression-free Survival (PFS) PFS is time from randomization to active MM according to International Myeloma Working Group (IMWG) criteria or death. Per IMWG criteria, active MM (SLiM-CRAB) is defined as: greater than or equal to (>=)60 percent (%) bone marrow plasma cells (BMPCs), free light chain (FLC) involved/uninvolved ratio >=100, greater than (>)1 focal bone lesions on magnetic resonance imaging (MRI), calcium elevation, renal insufficiency by creatinine clearance, anemia, or bone disease due to lytic bone lesions. From the date of randomization to active multiple myeloma (MM) or the date of death, whichever occurs first (up to approximately 8 years)
Secondary Time to Biochemical or Diagnostic (SLiM-CRAB) Progression Time to biochemical or diagnostic progression is defined as the earlier of time to biochemical progression or diagnostic (SLiM-CRAB) progression. SLiM-CRAB is defined as >=60% bone marrow plasma cells, free light chain involved/uninvolved ratio >=100, >1 focal bone lesions on magnetic resonance imaging (MRI), calcium elevation, renal insufficiency by creatinine clearance, anemia, or bone disease due to lytic bone lesions. Up to biochemical or diagnostic progression (up to approximately 8 years)
Secondary Overall Response Rate (ORR) ORR is defined as percentage of participants with partial response (PR) or better (very good partial response [VGPR], complete response [CR], and stringent complete response [sCR]) as defined by IMWG criteria. PR: >=50% reduction of serum M-protein and reduction in 24 hour urinary M-protein by >=90% or to less than (<)200 mg/24 hours; if serum and urine M-protein are not measurable, >=50% decrease in difference between involved and uninvolved free light chain (FLC) levels; if serum and urine M-protein and serum free light assay is not measurable, a >=50% reduction in BMPC, with baseline BMPC percentage >=30%. VGPR: serum and urine M-component detectable by immunofixation but not on electrophoresis or >=90% reduction in serum M-protein plus urine M-protein <100 mg/24 hours. CR: negative immunofixation on serum and urine, and <5% PCs in bone marrow. sCR: CR plus normal FLC ratio, and absence of clonal PCs by immunohistochemistry, immunofluorescence or 2- to 4- color flow cytometry. Up to approximately 8 years
Secondary Complete Response (CR) Rate CR rate was defined as the percentage of participants with a CR (or better [sCR]) as defined by the IMWG response criteria. IMWG criteria for CR: negative immunofixation on serum and urine, and <5% PCs in bone marrow. IMWG criteria for sCR: CR as defined above, plus normal FLC ratio, and absence of clonal PCs by immunohistochemistry, immunofluorescence or 2- to 4-color flow cytometry. Up to approximately 8 years
Secondary Time to First-Line Treatment for Multiple Myeloma (MM) Time to first-line treatment for MM was defined as the time from the date of randomization to the date of the first-line treatment given for MM (post disease progression). Post-progressive disease (PD) follow-up, every 6 months until end of study (up to approximately 8 years)
Secondary Progression-Free Survival on First-Line Treatment for MM (PFS2) PFS2 is time from date of randomization to date of documented PD on first line treatment given for MM or death, whichever comes first. IMWG criteria for PD: >=25% from lowest response level in serum M-component (the absolute increase must be >=0.5 gram per deciliter [g/dL]) and/or in urine M-component (the absolute increase must be >=200 mg/24 hour); only in participants without measurable serum and urine M-protein levels: increase of >=25% in the difference between involved and uninvolved free light chain levels and the absolute increase must be >10 mg/dL. BMPC%: the absolute % must be >=10%; definite increase in size of existing bone lesions or soft tissue plasmacytomas; definite development of new bone lesions or soft tissue plasmacytomas; development of hypercalcemia (corrected serum calcium >11.5 mg/dL or 2.65 millimoles per liter [mmol/L]) that can be attributed solely to PC proliferative disorder. Post-PD follow-up, every 6 months until end of study (up to approximately 8 years)
Secondary Overall Survival (OS) OS was defined as the time from the date of randomization to the date of the participant's death. Throughout study, and at least every 3 months until PD; post-PD, every 6 months until end of study (up to approximately 8 years)
Secondary Percentage of Participants who Progress to MM With Adverse Prognostic Features Adverse prognostic features includes International Staging System Stage III (based on beta2 [ß2]-microglobulin >=5.5 milligram per liter [mg/L] [median survival 29 months]) and adverse cytogenetic characteristics. At screening and PD (up to approximately 8 years)
Secondary Serum Daratumumab Pharmacokinetic (PK) Concentration PK concentration of Daratumumab will be measured. Cycles 1 and 3: Day 1 predose, and Day 4 postdose; Cycles 5, 7, 12, and 24: Day 1 predose; end of the treatment (EOT: 28 days after the last daratumumab dose); and 8 weeks after the last daratumumab dose (up to approximately 3 years and 8 weeks)
Secondary Maximum Observed Concentration (Cmax) of Daratumumab The Cmax is the maximum observed plasma concentration of Daratumumab. Cycles 1 and 3: Day 1 predose, and Day 4 postdose; Cycles 5, 7, 12, and 24: Day 1 predose; end of the treatment (EOT: 28 days after the last daratumumab dose); and 8 weeks after the last daratumumab dose (up to approximately 3 years and 8 weeks)
Secondary Minimum Observed Concentration (Cmin) of Daratumumab The Cmin is the minimum observed plasma concentration of Daratumumab. Cycles 1 and 3: Day 1 predose, and Day 4 postdose; Cycles 5, 7, 12, and 24: Day 1 predose; end of the treatment (EOT: 28 days after the last daratumumab dose); and 8 weeks after the last daratumumab dose (up to approximately 3 years and 8 weeks)
Secondary Number of Participants With Anti-daratumumab Antibodies Participant's serum samples will be collected and screened for antibodies binding to daratumumab using validated immunoassay methods for evaluation of potential immunogenicity. Cycles 1,3,5,7,12, and 24: Predose on Day 1; end of treatment (28 days after the last daratumumab dose), and 8 weeks after the last daratumumab dose (up to approximately 3 years and 8 weeks)
Secondary Number of Participants With Anti-Recombinant Human Hyaluronidase (rHuPH20) Antibodies Participant's plasma samples will be collected and screened for antibodies binding to rHuPH20 and will be assessed in confirmatory and titer assays as necessary for the rHuPH20 immunogenicity assessment. Cycles 1,3,5,7,12, and 24: Predose on Day 1; end of treatment (28 days after the last daratumumab dose), and 8 weeks after the last daratumumab dose (up to approximately 3 years and 8 weeks)
Secondary Change From Baseline in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30) Score EORTC QLQ-C30 is a questionnaire to assess quality of life of cancer participants. It is composed of 30 items, multiitem measure (28 items) and 2 single-item measures. For the multiple item measure, 4 point scale is used and the score for each item range from "1 = not at all" to "4 = very much". Higher scores indicate worsening. The 2 single-item measure involves question about the overall health and overall quality of life which will be rated on a 7 point scale ranging from "1 = very poor" to "7 = excellent". Lower scores indicate worsening. Scores are averaged, and transformed to 0-100 scale; higher score=better level of physical functioning Baseline, Weeks 12, 24, and 60, every year until end of treatment (EOT)/active monitoring, pre-PD follow-up (every year until end of study or PD), PD, and Months 3, 6, 12, and 18 post-PD (up to approximately 8 years)
Secondary Change From Baseline in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Future Perspective Scale EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in participants with multiple myeloma. EORTC QLQ-MY20 includes two scales: disease symptoms (6 questions) and future perspective (3 questions). Questions used a 4-point scale (from 1 'Not at All' to 4 'Very Much'). Scores were averaged, and transformed to a 0-100 scale; for the future perspective scale, a higher score indicates a better perspective of the future; and higher score for the disease symptoms scale indicates higher level of symptomatology. Baseline, Weeks 12, 24, and 60, every year until end of treatment (EOT)/active monitoring, pre-PD follow-up (every year until end of study or PD), PD, and Months 3, 6, 12, and 18 post-PD (up to approximately 8 years)
Secondary Change From Baseline in European Quality (EuroQoL) 5-Dimension 5-Level Health Status (EQ-5D-5L) Questionnaire The EQ-5D questionnaire is a brief, generic health-related quality of life assessment (HRQOL) that can also be used to incorporate participant preferences into health economic evaluations. The EQ-5D-5L descriptive system comprises the following 5 dimensions: mobility, self-care, usual activities, pain/discomfort and anxiety/depression and as overall health using a "thermometer" visual analog scale with response options ranging from 0 (worst imaginable health) to 100 (best imaginable health). Baseline, Weeks 12, 24, and 60, every year until end of treatment (EOT)/active monitoring, pre-PD follow-up (every year until end of study or PD), PD, and Months 3, 6, 12, and 18 post-PD (up to approximately 8 years)
Secondary Duration of Response Duration of response is defined as date of onset of first response (PR or better [VGPR, CR, sCR]) until date of disease progression or death. PR: >=50% reduction of serum M-protein and reduction in 24 hour urinary M-protein by >=90% or to <200 mg/24 hours; if the serum and urine M-protein are not measurable, >=50% decrease in difference between involved and uninvolved free light chain (FLC) levels; if serum and urine M-protein and serum free light assay is not measurable, a >=50% reduction in bone marrow PC, with baseline bone marrow PC percentage >=30%. VGPR: serum and urine M-component detectable by immunofixation but not on electrophoresis or >=90% reduction in serum M-protein plus urine M-protein <100 mg/24 hours. CR: negative immunofixation on serum and urine, and <5% PCs in bone marrow. sCR: CR plus normal FLC ratio, and absence of clonal PCs by immunohistochemistry, immunofluorescence or 2- to 4-color flow cytometry. From the date of initial documentation of a response to the date of first documented evidence of PD (up to approximately 8 years)
Secondary Time to Response Time to response is defined as the time from randomization until onset of first response (PR or better [VGPR, CR, sCR]). PR: >=50% reduction of serum M-protein and reduction in 24 hour urinary M-protein by >=90% or to <200 mg/24 hours; if the serum and urine M-protein are not measurable, >=50% decrease in difference between involved and uninvolved free light chain (FLC) levels; if serum and urine M-protein and serum free light assay is not measurable, a >=50% reduction in bone marrow PC, with baseline bone marrow PC percentage >=30%. VGPR: serum and urine M-component detectable by immunofixation but not on electrophoresis or >=90% reduction in serum M-protein plus urine M-protein <100 mg/24 hours. CR: negative immunofixation on serum and urine, and <5% PCs in bone marrow. sCR: CR plus normal FLC ratio, and absence of clonal PCs by immunohistochemistry, immunofluorescence or 2- to 4-color flow cytometry. Up to End of Treatment (up to approximately 39 cycles [each cycle of 28 days] or 36 months, whichever occurs first)
See also
  Status Clinical Trial Phase
Completed NCT01222286 - Study on the Anti-tumor Activity, Safety and Pharmacology of IPH2101 in Patients With Smoldering Multiple Myeloma Phase 2
Withdrawn NCT00503763 - Efficacy and Safety of Statin on the Course of Progressive Smoldering Multiple Myeloma Phase 2
Active, not recruiting NCT02916771 - Trial of Combination of Ixazomib and Lenalidomide and Dexamethasone in Smoldering Multiple Myeloma Phase 2
Completed NCT01955395 - Genomic and Psychosocial Effects of the 3RP on Patients With MGUS and Smoldering Multiple Myeloma N/A
Completed NCT01237054 - Imaging in MGUS, SMM and MM Phase 2
Completed NCT02903381 - A Phase II Trial If Nivolumab, Lenalidomide and Dexamethasone in High Risk Smoldering Myeloma Phase 2
Active, not recruiting NCT03236428 - Phase II Study of the CD38 Antibody Daratumumab in Patients With High-Risk MGUS and Low-Risk Smoldering Multiple Myeloma Phase 2
Completed NCT01718899 - Phase 1/2a Study of Cancer Vaccine to Treat Smoldering Multiple Myeloma Phase 1
Completed NCT00112827 - Melphalan and Radiation Therapy Followed By Lenalidomide in Treating Patients Who Are Undergoing Autologous Stem Cell Transplant for Stage I, Stage II, or Stage III Multiple Myeloma Phase 1/Phase 2
Completed NCT00099047 - Celecoxib in Preventing Multiple Myeloma in Patients With Monoclonal Gammopathy or Smoldering Myeloma Phase 2
Terminated NCT01248455 - A Phase II Trial of Anti-KIR in Smoldering Multiple Myeloma Phase 2
Active, not recruiting NCT03289299 - Aggressive Smoldering Curative Approach Evaluating Novel Therapies and Transplant Phase 2
Not yet recruiting NCT06365060 - Screening for AL Amyloidosis in Smoldering Multiple Myeloma
Active, not recruiting NCT02415413 - Carfilzomib in Treatment Patients Under 65 Years With High Risk Smoldering Multiple Myeloma Phase 2
Completed NCT01302886 - Study of BHQ880 in Patients With High Risk Smoldering Multiple Myeloma Phase 2
Recruiting NCT05841550 - The TG01 Study With TG01/QS-21 Vaccine in Patients With High-risk Smouldering Multiple Myeloma and Multiple Myeloma Phase 1/Phase 2
Recruiting NCT06383143 - Promoting Diagnosis and Management of AL in Italy (ProDigALIty)
Active, not recruiting NCT02886065 - A Study of PVX-410, a Cancer Vaccine, and Citarinostat +/- Lenalidomide for Smoldering MM Phase 1
Withdrawn NCT01571726 - Imaging Studies and the Development of Multiple Myeloma Phase 2
Terminated NCT01838369 - A Phase II Study of BI-505 in Smoldering Multiple Myeloma Phase 2