Clinical Trials Logo

Clinical Trial Details — Status: Completed

Administrative data

NCT number NCT02005471
Other study ID # GO28667
Secondary ID 2013-002110-12
Status Completed
Phase Phase 3
First received
Last updated
Start date March 17, 2014
Est. completion date August 3, 2022

Study information

Verified date September 2023
Source Hoffmann-La Roche
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

The purpose of this open-label, multicenter, randomized, Phase III study is to evaluate the benefit of venetoclax in combination with rituximab compared with bendamustine in combination with rituximab in participants with relapsed or refractory CLL. Participants will be randomly assigned in 1:1 ratio to receive either venetoclax + rituximab (Arm A) or bendamustine + rituximab (Arm B).


Recruitment information / eligibility

Status Completed
Enrollment 389
Est. completion date August 3, 2022
Est. primary completion date May 8, 2017
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility Inclusion Criteria: - Diagnosis of CLL per diagnostic criteria for relapsed or refractory CLL per the international workshop on chronic lymphocytic leukemia (iwCLL) guidelines - Previously treated with 1-3 lines of therapy (example: completed greater than or equal to [>/=] 2 treatment cycles per therapy), including at least one standard chemotherapy-containing regimen - Participants previously treated with bendamustine only if their duration of response was >/= 24 months - Eastern Cooperative Oncology Group (ECOG) performance score of less than or equal to (</=) 1 - Adequate bone marrow function - Adequate renal and hepatic function - Participants must use effective birth control throughout study until at least 30 days after study treatment or 1 year after rituximab treatment, whichever is later; female participants must not be pregnant or breast-feeding - For participants with the 17p deletion, previously treated with 1-3 lines of therapy, including at least one prior standard chemotherapy-containing regimen or at least one prior alemtuzumab-containing therapy Inclusion Criteria R/C Substudy: - Participants randomized to Arm A or Arm B with a confirmed disease progression of CLL per iwCLL criteria - Participants who have not received new anti-CLL therapy following disease progression in Arm A or Arm B - Adequate renal and hepatic function per laboratory reference range Exclusion Criteria: - Transformation of CLL to aggressive non-Hodgkin lymphoma or central nervous system (CNS) involvement by CLL - Undergone an allogenic stem cell transplant - A history of significant renal, neurologic, psychiatric, endocrine, metabolic, immunologic, cardiovascular or hepatic disease - Hepatitis B or C or known human immunodeficiency virus (HIV) positive - Receiving warfarin treatment - Received an anti-CLL monoclonal antibody within 8 weeks prior to the first dose of study drug - Received any anti-cancer or investigational therapy within 28 days prior to the first dose of study drug or has not recovered to less than Grade 2 clinically significant adverse effect(s)/toxicity(ies) of any previous therapy - Received cytochrome P450 3A4 (CYP3A4) inhibitors (such as fluconazole, ketoconazole and clarithromycin) or inducers (such as rifampin, carbamazapine, phenytoin, St. John's Wort) within 7 days prior to the first dose of venetoclax - History of prior venetoclax treatment - Participants with another cancer, history of another cancer considered uncured on in complete remission for <5 years, or currently under treatment for another suspected cancer except non-melanoma skin cancer or carcinoma in situ of the cervix that has been treated or excised and is considered resolved - Malabsorption syndrome or other condition that precludes enteral route of administration - Other clinically significant uncontrolled condition(s) including, but not limited to, systemic infection (viral, bacterial or fungal) - Vaccination with a live vaccine within 28 days prior to randomization - Consumed grapefruit or grapefruit products, seville oranges (including marmalade containing seville oranges), or star fruit within 3 days prior to the first dose of study treatment - A cardiovascular disability status of New York Heart Association Class >/=3. Class 3 is defined as cardiac disease in which participants are comfortable at rest but have marked limitation of physical activity due to fatigue, palpitations, dyspnea, or anginal pain - Major surgery within 30 days prior to the first dose of study treatment - A participant who is pregnant or breastfeeding - Known allergy to both xanthine oxidase inhibitors and rasburicase Exclusion Criteria R/C Substudy: - Transformation of CLL to aggressive NHL (e.g., Richter's transformation, prolymphocytic leukemia, or DLBCL) or CNS involvement by CLL - Evidence of other clinically significant uncontrolled condition(s) including, but not limited to, uncontrolled systemic infection (viral, bacterial, or fungal) - Development of other malignancy since enrollment into the study, with the exception of curatively treated basal cell carcinoma or squamous cell carcinoma of the skin or carcinoma in situ of the cervix - Uncontrolled autoimmune hemolytic anemia or immune thrombocytopenia - History of severe (i.e., requiring permanent discontinuation of prior rituximab therapy) prior allergic or anaphylactic reactions to rituximab - Known HIV positivity - Positive test results for chronic hepatitis B infection (defined as positive hepatitis B surface antigen [HbsAg] serology) - Positive test results for hepatitis C virus (HCV; HCV antibody serology testing) - Requires the use of warfarin (due to potential drug interactions that may potentially increase the exposure of warfarin) - Has not recovered to less than Grade 2 clinically significant adverse effect(s)/toxicity(ies) of any previous therapy - Received potent CYP3A4 inhibitors (such as fluconazole, ketoconazole, and clarithromycin) within 7 days prior to the first dose of study treatment - Received potent CYP3A4 inducers (such as rifampin, carbamazepine, phenytoin, St. John's wort) within 7 days prior to the first dose of study treatment - Consumed grapefruit or grapefruit products, Seville oranges (including marmalade containing Seville oranges), or star fruit within 3 days prior to the first dose of study treatment - A cardiovascular disability status of New York Heart Association Class >/= 3 - A significant history of renal, neurologic, psychiatric, endocrine, metabolic, immunologic, cardiovascular, or hepatic disease that, in the opinion of the investigator, would adversely affect the participants's participation in this study or interpretation of study outcomes - Major surgery within 30 days prior to the first dose of study treatment - A participant who is pregnant or breastfeeding - Malabsorption syndrome or other condition that precludes enteral route of administration - Known allergy to both xanthine oxidase inhibitors and rasburicase - Vaccination with a live vaccine within 28 days prior to randomization

Study Design


Related Conditions & MeSH terms


Intervention

Drug:
Bendamustine
Bendamustine will be administered at a dose of 70 mg/m^2 via IV infusion on Days 1 and 2 of each 28-day cycle, for 6 cycles.
Venetoclax
Venetoclax will be administered at an initial dose of 20 mg via tablet orally QD, incremented weekly up to a maximum dose of 400 mg during a 5-week ramp-up period. Venetoclax will be continued at 400 mg QD from Week 6 (Day 1 of Cycle 1 of combination therapy) onwards up to disease progression (PD) or 2 years, whichever occurs first. R/C Substudy: venetoclax will be administered for 5-week dose ramp-up period to reach the target dose of 400 mg QD. Venetoclax will continue to be administered during the rituximab cycles until disease progression or for a maximum of 2 years from Cycle 1R/C Day 1 of the R/C Substudy.
Rituximab
Rituximab will be administered at a dose of 375 mg/m^2 via IV infusion on Day 1 of Cycle 1 and at a dose of 500 mg/m^2 on Day 1 of Cycles 2-6. R/C Substudy: Following the venetoclax ramp-up period, rituximab will be administered for 6 cycles consisting of a single infusion on the first day of each 28-day cycle.

Locations

Country Name City State
Australia Royal Adelaide Hospital Adelaide South Australia
Australia Flinders Medical Centre Bedford Park South Australia
Australia Concord Repatriation General Hospital Concord New South Wales
Australia Frankston Hospital Frankston Victoria
Australia The Canberra Hospital Garran Australian Capital Territory
Australia Royal Hobart Hospital Hobart Tasmania
Australia St George Hospital Kogarah, New South Wales New South Wales
Australia Monash Medical Centre; Haematology Melbourne Victoria
Australia Slade Health Pharmacy Mount Waverley Victoria
Australia The Perth Blood Institute Nedlands Western Australia
Australia Peter MacCallum Cancer Center North Melbourne Victoria
Australia Royal Melbourne Hospital Parkville Victoria
Australia Princess Alexandra Hospital Woolloongabba Queensland
Austria Medizinische Universität Innsbruck Innsbruck
Austria LKH - Universitätsklinikum der PMU Salzburg Salzburg
Austria Klinik Ottakring Wien
Austria Medizinische Universität Wien Wien
Belgium ZNA Antwerpen; Department Hematology Antwerpen
Belgium Cliniques Universitaires Saint-Luc; Hematology Bruxelles
Belgium AZ Groeninge Kortrijk
Belgium UZ Leuven; Department Hematology Leuven
Belgium CHU UCL Mont-Godinne Mont-godinne
Belgium AZ Delta (Campus Rumbeke) Roeselare
Canada Foothills Medical Centre; Centre Dept of Medical Clinical Neuroscience Calgary Alberta
Canada Juravinski Cancer Clinic Hamilton Ontario
Canada Jewish General Hospital Montreal Quebec
Canada Saskatoon City Hospital;Saskatchewan Cancer Centre Saskatoon Saskatchewan
Czechia Fakultni nemocnice Brno Brno
Czechia Fakultni nemocnice Hradec Kralove Hradec Kralove
Czechia Fakultní nemocnice Olomouc Olomouc
Czechia Fakultni nemocnice Ostrava Ostrava - Poruba
Czechia Fakultni nemocnice Kralovske Vinohrady Praha
Czechia Vseobecna fakultni nemocnice v Praze Praha 2
Denmark Herlev Hospital Herlev
Denmark Rigshospitalet København Ø
Denmark Odense Universitetshospital Odense C
Denmark Sjællands Universitetshospital, Roskilde Roskilde
Denmark Sygehus Lillebælt, Vejle Vejle
France Hôpital Morvan Brest
France Centre Hospitalier Départemental Les Oudairies La Roche sur Yon
France Hopital Claude Huriez - CHU Lille Lille
France Hopital Saint Eloi Montpellier
France CHU Nantes - Hôtel Dieu; Service Assistance Medicale à la Procreation Nantes
France Hopital Robert Debre Paris
France Centre Hospitalier Lyon Sud Pierre Benite
France CHU Poitiers - Hopital La Miletrie Poitiers
France CHU de Rennes - Hopital de Pontchaillo Rennes
France Centre Henri Becquerel Rouen
France Institut Universitaire du Cancer - Oncopole Toulouse (IUCT-O) Toulouse
France CHU Tours - Hôpital Bretonneau Tours
France Hôpital de Brabois Adultes Vandoeuvre-les-nancy
Germany Universitätsklinikum "Carl Gustav Carus" der Technischen Universität Dresden Dresden
Germany Universitaetsklinikum Freiburg Freiburg
Germany Universitätsklinikum Tübingen Tübingen
Hungary Orszagos Onkologiai Intezet Budapest
Hungary Semmelweis Egyetem Budapest
Hungary Debreceni Egyetem Klinikai Központ; B?rgyógyászati Klinika Debrecen
Hungary Somogy Megyei Kaposi Mor Oktato Korhaz Pecs
Hungary Szegedi Tud.Egyetem Szent-Gyorgyi Albert Klin.Kozp. Szeged
Italy Istituto Tumori Giovanni Paolo II IRCCS Ospedale Oncologico Bari Bari Puglia
Italy Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII (Presidio Papa Giovanni XXIII) Bergamo Lombardia
Italy Azienda Ospedaliera Universitaria Careggi Florence Toscana
Italy Azienda Ospedaliero Universitaria San Martino Genova Liguria
Italy Asst Grande Ospedale Metropolitano Niguarda; SC Farmacia Ospedale Milano Lombardia
Italy Ospedale San Raffaele Milano Lombardia
Italy Azienda Ospedaliera Di Padova Padova Veneto
Italy Azienda Ospedaliera Città della Salute e della Scienza di Torino; Radiology Torino Abruzzo
Italy Azienda Ospedaliero Universitaria Ospedali Riuniti Torrette Di Ancona Marche
Korea, Republic of Seoul National University Bundang Hospital Seongnam-si
Korea, Republic of Konkuk University Medical Center Seoul
Korea, Republic of Severance Hospital, Yonsei University Health System Seoul
Korea, Republic of The Catholic University of Korea Seoul St. Mary?s Hospital Seoul
Netherlands Amsterdam UMC Location AMC Amsterdam
Netherlands Amsterdam UMC, Locatie VUMC; Neurology Amsterdam
Netherlands Albert Schweitzer Ziekenhuis, Dordwijk; Internal Medicine, Hemato-Oncology Dordrecht
Netherlands Medisch Spectrum Twente Enschede
Netherlands Leids Universitair Medisch Centrum; Cardiology Leiden
Netherlands Erasmus Medisch Centrum Rotterdam
Netherlands UMC Utrecht Utrecht
New Zealand Middlemore Hospital Auckland
New Zealand North Shore Hospital; Haematolgy Auckland
New Zealand Christchurch Hospital NZ Christchurch
New Zealand Baxter Healthcare Mount Wellington
Poland SP ZOZ Zespol Szpitali Miejskich w Chorzowie Chorzow
Poland Uniwersyteckie Centrum Kliniczne Gdansk
Poland Wojewódzkie Wielospecjalistyczne Centrum Onkologii i Traumatologii im. M. Kopernika w Lodzi Lodz
Poland Szpital Wojewodzki w Opolu Opole
Poland MTZ Clinical Research Sp. z o.o. Warszawa
Poland Samodzielny Publiczny Szpital Kliniczny nr 1 Zabrze
Russian Federation Kemerovo Regional Clinical Hospital Kemerovo
Russian Federation FSBSI "Russian Oncological Scientific Center n.a. N.N. Blokhin" Moscow Moskovskaja Oblast
Russian Federation BHI of Omsk region Clinical Oncology Dispensary Omsk
Russian Federation SRI of Hematology and Transfusiology Sankt-peterburg Sankt Petersburg
Russian Federation North-West Federal Medical Research Center n.a. V.A. Almazov St. Petersburg Sankt Petersburg
Spain Hospital Clinic i Provincial de Barcelona; Hematology Barcelona
Spain Hospital Universitari Vall d'Hebron Barcelona
Spain Hospital Universitario 12 de Octubre Madrid
Spain Complejo Hospitalario de Navarra Pamplona Navarra
Spain Hospital Clinico Universitario de Salamanca Salamanca
Sweden Skånes Universitetssjukhus Lund
Sweden Akademiska Sjukhuset Uppsala
Taiwan National Taiwan University Hospital Taipei
United Kingdom Bristol Haematology and Oncology centre Bristol
United Kingdom The Christie Manchester
United Kingdom Southampton General Hospital Southampton
United Kingdom Singleton Hospital; Pharmacy Department Swansea
United States Henry Ford Health System Detroit Michigan
United States University of California San Diego Medical Center La Jolla California
United States Memorial Sloan Kettering Cancer Center; Clinical Trials Office New York New York
United States Perlmutter Cancer Center NYU Langone Health New York New York
United States Huntsman Cancer Institute; University of Utah Salt Lake City Utah

Sponsors (2)

Lead Sponsor Collaborator
Hoffmann-La Roche AbbVie

Countries where clinical trial is conducted

United States,  Australia,  Austria,  Belgium,  Canada,  Czechia,  Denmark,  France,  Germany,  Hungary,  Italy,  Korea, Republic of,  Netherlands,  New Zealand,  Poland,  Russian Federation,  Spain,  Sweden,  Taiwan,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Other Change From Baseline in Lymphocyte Subset Counts at Specified Time Points Baseline, C4D14-28, Study Treatment Completion/Early Withdrawal (STC/EW, up to C6D28), EoCTR visit (8 to 12 weeks after C6D1), and at FUVs (every 12 weeks after EoCTR up to 3 years); Cycle length = 28 days
Primary Percentage of Participants With PD as Assessed by the Investigator Using Standard International Workshop on Chronic Lymphocytic Leukemia (iwCLL) Guidelines or Death Assessment of response was performed by the investigator according to the iwCLL guidelines. PD was defined as occurrence of one of the following events: appearance of any new extra nodal lesion; new palpable lymph node (greater than [>] 1.5 centimeters [cm]); unequivocal progression of non-target lesion; an increase of greater than or equal to (>/=) 50 percent (%) compared to baseline in splenomegaly, hepatomegaly, number of blood lymphocytes with lymphocyte count >/=5000 per microliter (mcL), or in longest diameter of any extra nodal lesion; transformation to a more aggressive histology; decrease of >/=50% compared to baseline in platelet or neutrophil count; or decrease in hemoglobin level by >2 grams per deciliter (g/dL) or to less than [<] 10 g/dL. Percentages are rounded off. Baseline up to PD or death from any cause, whichever occurred first (up to approximately 8 years 5 months)
Primary Progression-Free Survival (PFS) as Assessed by the Investigator Using Standard iwCLL Guidelines PFS was defined as the time from randomization until first occurrence of PD/relapse as assessed by the investigator using iwCLL guidelines, or death from any cause, whichever occurred first. PD: occurrence of one of the following: new lesion; new palpable lymph node (>1.5 cm); unequivocal progression of non-target lesion; increase of >/=50% in splenomegaly, hepatomegaly, blood lymphocytes with count >/=5000/mcL, longest diameter of any lesion; transformation to more aggressive histology; decrease of >/=50% in platelet or neutrophil count, or hemoglobin level by >2 g/dL or to <10 g/dL. Participants who had not progressed, relapsed, or died at the time of analysis, were censored on the date of last assessment. In case of no disease assessment after baseline, PFS was censored at the time of randomization+1 day. The median PFS was estimated using Kaplan-Meier method and the 95% confidence interval (CI) was computed using method of Brookmeyer and Crowley. Baseline up to PD or death, whichever occurred first (up to approximately 8 years 5 months)
Secondary Percentage of Participants With PD or Death as Assessed by the Independent Review Committee (IRC) Using Standard iwCLL Guidelines Assessment of response was performed by the IRC according to the iwCLL guidelines. PD was defined as occurrence of one of the following events: appearance of any new extra nodal lesion; new palpable lymph node (>1.5 cm); unequivocal progression of non-target lesion; an increase of >/=50% compared to baseline in splenomegaly, hepatomegaly, number of blood lymphocytes with lymphocyte count >/=5000/mcL, or in longest diameter of any extra nodal lesion; transformation to a more aggressive histology; decrease of >/=50% compared to baseline in platelet or neutrophil count; or decrease in hemoglobin level by >2 g/dL or to <10 g/dL. No new IRC data was generated post the primary analysis. Baseline up to PD or death, whichever occurred first (up to approximately 3 years)
Secondary PFS as Assessed by the IRC Using Standard iwCLL Guidelines PFS was defined as the time from randomization until first occurrence of PD/relapse as assessed by the IRC using iwCLL guidelines, or death from any cause, whichever occurred first. PD: occurrence of one of the following: new lesion; new palpable lymph node (>1.5 cm); unequivocal progression of non-target lesion; increase of >/=50% in splenomegaly, hepatomegaly, blood lymphocytes with count >/=5000/mcL, longest diameter of any lesion; transformation to more aggressive histology; decrease of >/=50% in platelet or neutrophil count, or hemoglobin level by >2 g/dL or to <10 g/dL. Participants who had not progressed, relapsed, or died at the time of analysis, were censored on the date of last assessment. In case of no disease assessment after baseline, PFS was censored at the time of randomization+1 day. The median PFS was estimated using Kaplan-Meier method and the 95% CI was computed using method of Brookmeyer and Crowley. No new IRC data was generated post the primary analysis. Baseline up to PD or death, whichever occurred first (up to approximately 3 years)
Secondary Percentage of Participants With PD or Death as Assessed by the Investigator Using Standard iwCLL Guidelines in Participants With 17p Deletion as Identified by Fluorescence In-situ Hybridization (FISH) Test Assessment of response was performed by the investigator according to the iwCLL guidelines. PD was defined as occurrence of one of the following events: appearance of any new extra nodal lesion; new palpable lymph node (>1.5 cm); unequivocal progression of non-target lesion; an increase of >/=50% compared to baseline in splenomegaly, hepatomegaly, number of blood lymphocytes with lymphocyte count >/=5000/mcL, or in longest diameter of any extra nodal lesion; transformation to a more aggressive histology; decrease of >/=50% compared to baseline in platelet or neutrophil count; or decrease in hemoglobin level by >2 g/dL or to <10 g/dL. Percentages are rounded off. Baseline up to PD or death, whichever occurred first (up to approximately 8 years 5 months)
Secondary PFS as Assessed by the Investigator Using Standard iwCLL Guidelines in Participants With 17p Deletion as Identified by FISH Test PFS was defined as the time from randomization until first occurrence of PD/relapse as assessed by the investigator using iwCLL guidelines, or death from any cause, whichever occurred first. PD: occurrence of one of the following: new lesion; new palpable lymph node (>1.5 cm); unequivocal progression of non-target lesion; increase of >/=50% in splenomegaly, hepatomegaly, blood lymphocytes with count >/=5000/mcL, longest diameter of any lesion; transformation to more aggressive histology; decrease of >/=50% in platelet or neutrophil count, or hemoglobin level by >2 g/dL or to <10 g/dL. Participants who had not progressed, relapsed, or died at the time of analysis, were censored on the date of last assessment. In case of no disease assessment after baseline, PFS was censored at the time of randomization+1 day. The median PFS was estimated using Kaplan-Meier method and the 95% CI was computed using method of Brookmeyer and Crowley. Baseline up to PD or death, whichever occurred first (up to approximately 8 years 5 months)
Secondary Percentage of Participants With PD or Death as Assessed by the IRC Using Standard iwCLL Guidelines in Participants With 17p Deletion as Identified by FISH Test Assessment of response was performed by the IRC according to the iwCLL guidelines. PD was defined as occurrence of one of the following events: appearance of any new extra nodal lesion; new palpable lymph node (>1.5 cm); unequivocal progression of non-target lesion; an increase of >/=50% compared to baseline in splenomegaly, hepatomegaly, number of blood lymphocytes with lymphocyte count >/=5000/mcL, or in longest diameter of any extra nodal lesion; transformation to a more aggressive histology; decrease of >/=50% compared to baseline in platelet or neutrophil count; or decrease in hemoglobin level by >2 g/dL or to <10 g/dL. No new IRC data was generated post the primary analysis. Baseline up to PD or death, whichever occurred first (up to approximately 3 years)
Secondary PFS as Assessed by the IRC Using Standard iwCLL Guidelines in Participants With 17p Deletion as Identified by FISH Test PFS was defined as the time from randomization until first occurrence of PD/relapse as assessed by the IRC using iwCLL guidelines, or death from any cause, whichever occurred first. PD: occurrence of one of the following: new lesion; new palpable lymph node (>1.5 cm); unequivocal progression of non-target lesion; increase of >/=50% in splenomegaly, hepatomegaly, blood lymphocytes with count >/=5000/mcL, longest diameter of any lesion; transformation to more aggressive histology; decrease of >/=50% in platelet or neutrophil count, or hemoglobin level by >2 g/dL or to <10 g/dL. Participants who had not progressed, relapsed, or died at the time of analysis, were censored on the date of last assessment. In case of no disease assessment after baseline, PFS was censored at the time of randomization+1 day. The median PFS was estimated using Kaplan-Meier method and the 95% CI was computed using method of Brookmeyer and Crowley. No new IRC data was generated post the primary analysis. Baseline up to PD or death, whichever occurred first (up to approximately 3 years)
Secondary Percentage of Participants With Best Overall Response of Complete Response (CR), CR With Incomplete Bone Marrow Recovery (CRi), Nodular Partial Response (nPR), or Partial Response (PR) as Assessed by the Investigator Using iwCLL Guidelines Response was assessed by investigator according to iwCLL guidelines and was confirmed by repeat assessment >/=4 weeks after initial documentation. CR: peripheral blood lymphocytes <4000/mcL; absence of any new lesion, nodal disease, lymphadenopathy, hepatomegaly, splenomegaly, and constitutional symptoms; neutrophils >1500/mcL, platelets >100000/mcL, hemoglobin >11.0 g/dL without need for transfusion or exogenous growth factors; normocellular bone marrow with <30% lymphocytes; no lymphoid nodules. CRi: fulfilling all CR criteria but persistent cytopenia. PR: >/=50% reduction in two of following: peripheral blood lymphocytes, lymphadenopathy, spleen and/or liver enlargement; and 1 of following: neutrophils >1500/mcL, platelets >100000/mcL, hemoglobin >11.0 g/dL or >/=50% improvement without need for transfusion or exogenous growth factors. nPR: fulfilling all CR criteria but presence of lymphoid nodules. 95% CI was computed using Pearson-Clopper method.Percentages are rounded off. Baseline up to approximately 8 years 5 months
Secondary Percentage of Participants With Best Overall Response of CR, CRi, nPR, or PR as Assessed by the IRC Using iwCLL Guidelines Response was assessed by IRC according to iwCLL guidelines and was confirmed by repeat assessment >/=4 weeks after initial documentation. CR: peripheral blood lymphocytes <4000/mcL; absence of any new lesion, nodal disease, lymphadenopathy, hepatomegaly, splenomegaly, constitutional symptoms; neutrophils >1500/mcL, platelets >100000/mcL, hemoglobin >11.0 g/dL without need for transfusion or exogenous growth factors; normocellular bone marrow with <30% lymphocytes; no lymphoid nodules. CRi: fulfilling all CR criteria but persistent cytopenia. PR: >/=50% reduction in 2 of following: peripheral blood lymphocytes, lymphadenopathy, spleen and/or liver enlargement; 1 of following: neutrophils >1500/mcL, platelets >100000/mcL, hemoglobin >11.0 g/dL or >/=50% improvement without need for transfusion or exogenous growth factors. nPR: fulfilling all CR criteria but presence of lymphoid nodules. 95% CI was computed using Pearson-Clopper method.No new IRC data was generated post primary analysis. Baseline up to last FUV (up to approximately 3 years)
Secondary Percentage of Participants With Overall Response of CR, Cri, nPR, or PR at End of Combination Treatment Visit as Assessed by the Investigator Using iwCLL Guidelines Response was assessed by investigator according to iwCLL guidelines and was confirmed by repeat assessment >/=4 weeks after initial documentation. CR: peripheral blood lymphocytes <4000/mcL; absence of any new lesion, nodal disease, lymphadenopathy, hepatomegaly, splenomegaly, and constitutional symptoms; neutrophils >1500/mcL, platelets >100000/mcL, hemoglobin >11.0 g/dL without need for transfusion or exogenous growth factors; normocellular bone marrow with <30% lymphocytes; no lymphoid nodules. CRi: fulfilling all CR criteria but persistent cytopenia. PR: >/=50% reduction in 2 of following: peripheral blood lymphocytes, lymphadenopathy, spleen and/or liver enlargement; and 1 of following: neutrophils >1500/mcL, platelets >100000/mcL, hemoglobin >11.0 g/dL or >/=50% improvement without need for transfusion or exogenous growth factors. nPR: fulfilling all CR criteria but presence of lymphoid nodules. 95% CI was computed using Pearson-Clopper method. Percentages are rounded off. End of combination treatment response (EoCTR) visit (8 to 12 weeks after Cycle [C] 6 Day [1]); Cycle length = 28 days
Secondary Percentage of Participants With Overall Response of CR, Cri, nPR, or PR at End of Combination Treatment Visit as Assessed by the IRC Using iwCLL Guidelines Response was assessed by the IRC according to the iwCLL guidelines and was confirmed by repeat assessment >/=4 weeks after initial documentation. CR: peripheral blood lymphocytes <4000/mcL; absence of any new lesion, nodal disease, lymphadenopathy, hepatomegaly, splenomegaly, and constitutional symptoms; neutrophils >1500/mcL, platelets >100000/mcL, hemoglobin >11.0 g/dL without need for transfusion or exogenous growth factors; normocellular bone marrow with <30% lymphocytes; no lymphoid nodules. CRi: fulfilling all CR criteria but persistent cytopenia. PR: >/=50% reduction in two of the following: peripheral blood lymphocytes, lymphadenopathy, spleen and/or liver enlargement; and one of the following: neutrophils >1500/mcL, platelets >100000/mcL, hemoglobin >11.0 g/dL or >/=50% improvement without need for transfusion or exogenous growth factors. nPR: fulfilling all CR criteria but presence of lymphoid nodules. The 95% CI was computed using Pearson-Clopper method. EoCTR visit (8 to 12 weeks after C6D1); Cycle length = 28 days
Secondary Percentage of Participants Who Died Percentage of participants who died from any cause, during the study, was reported. Percentage is rounded off. Baseline up to approximately 8 years 5 months
Secondary Overall Survival (OS) OS was defined as the time from the date of randomization to the date of death from any cause. Participants alive at the time of the analysis were censored at the date when they were last known to be alive as documented by the investigator. The median OS was estimated using Kaplan-Meier method and the 95% CI was computed using method of Brookmeyer and Crowley. Baseline up to approximately 8 years 5 months
Secondary Percentage of Participants With PD/Relapse, Start of a New Anti-Chronic Lymphocytic Leukemia (CLL) Therapy, or Death as Assessed by the Investigator Using iwCLL Guidelines Percentage of participants with PD/relapse, death from any cause, or start of a new non-protocol-specified anti-CLL therapy as assessed by the investigator, during the study, was reported. PD was defined as occurrence of one of the following events: appearance of any new extra nodal lesion; new palpable lymph node (>1.5 cm); unequivocal progression of non-target lesion; an increase of >/=50% compared to baseline in splenomegaly, hepatomegaly, number of blood lymphocytes with lymphocyte count >/=5000/mcL, or in longest diameter of any extra nodal lesion; transformation to a more aggressive histology; decrease of >/=50% compared to baseline in platelet or neutrophil count; or decrease in hemoglobin level by >2 g/dL or to <10 g/dL. Percentages are rounded off. Baseline up to PD/relapse, start of a new anti-CLL therapy, or death from any cause, whichever occurred first (approximately 8 years 5 months)
Secondary Event-Free Survival (EFS) as Assessed by the Investigator Using iwCLL Guidelines EFS was defined as the time from date of randomization until the date of PD/relapse, start of a new non-protocol-specified anti-CLL therapy, or death from any cause, whichever occurred first, as assessed by the investigator. PD: occurrence of one of the following: new lesion; new palpable lymph node (>1.5 cm); unequivocal progression of non-target lesion; increase of >/=50% in splenomegaly, hepatomegaly, blood lymphocytes with count >/=5000/mcL, longest diameter of any lesion; transformation to more aggressive histology; decrease of >/=50% in platelet or neutrophil count, or hemoglobin level by >2 g/dL or to <10 g/dL. Participants without any of the specified event at the time of analysis were censored at the date of last adequate response assessment. In case of no post-baseline response assessment, participants were censored at the randomization date. The median EFS was estimated using Kaplan-Meier method and the 95% CI was computed using method of Brookmeyer and Crowley. Baseline up to PD/relapse, start of a new anti-CLL therapy, or death from any cause, whichever occurred first (approximately 8 years 5 months)
Secondary Percentage of Participants With PD or Death Among Participants With Best Overall Response of CR, CRi, nPR, or PR as Assessed by the Investigator Using iwCLL Guidelines Percentage of participants with PD as assessed by the investigator according to the iwCLL guidelines or death from any cause during the study was reported. PD was defined as occurrence of one of the following events: appearance of any new extra nodal lesion; new palpable lymph node (>1.5 cm); unequivocal progression of non-target lesion; an increase of >/=50% compared to baseline in splenomegaly, hepatomegaly, number of blood lymphocytes with lymphocyte count >/=5000/mcL, or in longest diameter of any extra nodal lesion; transformation to a more aggressive histology; decrease of >/=50% compared to baseline in platelet or neutrophil count; or decrease in hemoglobin level by >2 g/dL or to <10 g/dL. CR, CRi, nPR, and PR have been defined in previous outcomes, and are not repeated here due to space constraint. Percentage is rounded off. From time of achieving best overall response until PD or death from any cause, whichever occurred first (up to approximately 8 years 5 months)
Secondary Duration of Responses (DOR) as Assessed by the Investigator Using iwCLL Guidelines DOR was defined as the time from first occurrence of a documented response of CR, CRi, nPR, or PR until PD/relapse, as assessed by the investigator according to the iwCLL guidelines, or death from any cause. PD: occurrence of one of the following: new lesion; new palpable lymph node (>1.5 cm); unequivocal progression of non-target lesion; increase of >/=50% in splenomegaly, hepatomegaly, blood lymphocytes with count >/=5000/mcL, longest diameter of any lesion; transformation to more aggressive histology; decrease of >/=50% in platelet or neutrophil count, or hemoglobin level by >2 g/dL or to <10 g/dL. Participants without PD or death after response were censored at the last date of adequate response assessment. The median DOR was estimated using Kaplan-Meier method and the 95% CI was computed using method of Brookmeyer and Crowley. CR, CRi, nPR, and PR have been defined in previous outcomes, and are not repeated here due to space constraint. From time of achieving best overall response until PD or death from any cause, whichever occurred first (up to approximately 8 years 5 months)
Secondary Percentage of Participants With Start of New Anti-CLL Treatment or Death as Assessed by the Investigator Percentage of participants with start of new non-protocol-specified anti-CLL therapy, as assessed by the investigator, or death from any cause, during the study, was reported. Percentage is rounded off. Baseline up to start of new ani-CLL therapy or death, whichever occurred first (up to approximately 8 years 5 months)
Secondary Time to New Anti-CLL Treatment (TTNT) as Assessed by the Investigator TTNT was defined as the time from randomization until start of new non-protocol-specified anti-CLL treatment or death from any cause. Participants without the event at the time of analysis were censored at the last visit date for this outcome measure analysis. The median TTNT was estimated using Kaplan-Meier method and the 95% CI was computed using method of Brookmeyer and Crowley. Baseline up to start of new ani-CLL therapy or death, whichever occurred first (up to approximately 8 years 5 months)
Secondary Percentage of Participants With Minimal Residual Disease (MRD) Negativity in Peripheral Blood MRD-negativity was defined as the presence of <1 malignant B-cell per 10000 normal B-cells in a sample of at least 200000 B-cells, as assessed by the allele specific oligonucleotide polymerase chain reaction (ASO-PCR) and/or flow cytometry technique. Percentage of participants with MRD-negativity was reported. The 95% CI was computed using Pearson-Clopper method. Percentage is rounded off. EoCTR visit (8 to 12 weeks after C6D1); Cycle length = 28 days
Secondary Percentage of Participants With MRD Negativity in Bone Marrow MRD-negativity was defined as the presence of <1 malignant B-cell per 10000 normal B-cells in a sample of at least 200000 B-cells, as assessed flow cytometry technique. Percentage of participants with MRD-negativity was reported. The 95% CI was computed using Pearson-Clopper method. Percentages are rounded off. EoCTR visit (8 to 12 weeks after C6D1); Cycle length = 28 days
Secondary Plasma Venetoclax Concentrations Pre-dose (0 hour, anytime before venetoclax administration) and 4 hours post-dose on D1 of Cycles 1 and 4; Cycle length = 28 days
Secondary Change From Baseline in Monroe Dunaway (MD) Anderson Symptom Inventory (MDASI) Core Symptom Severity, Module Symptom Severity, and Interference Scores MDASI is a 25-item validated questionnaire consisting of 2 parts. Part 1: 19-items divided into 2 scales, Core Symptom Severity (average of Questions 1 to 13; total 13 items: pain, fatigue, nausea, disturbed sleep, distress, shortness of breath, remembering things, lack of appetite, drowsiness, dry mouth, sadness, vomiting, and numbness) and Module Symptom Severity (average of Questions 14 to 19; total 6 items: night sweats, fevers and chills, lymph node swelling, diarrhea, bruising easy or bleeding, and constipation). Part 2: 6-items to assess Interference (symptom distress) (average of Questions 20 to 25; total 6 items: general activity, walking, work, mood, relations with other people, and enjoyment of life). Each item was rated from 0 to 10, with lower scores indicating better outcome. Total score for Core Symptom Severity, Module Symptom Severity, and Interference are reported which range from 0 to 10, with lower scores indicating better health-related quality of life (HRQoL). Baseline, Days 1, 8, and 15 of Cycles 1, 2, and 3; Cycle length = 28 days
Secondary Change From Baseline in HRQoL as Measured by European Organization for Research and Treatment of Cancer Core Quality of Life Questionnaire (EORTC QLQ-C30) Functional Scales Score and Global Health Status/Global Quality-of-Life (QoL) Scale Score EORTC QLQ-C30 is a validated self-report measure consisting of 30 questions incorporated into 5 functional scales (Physical, Role, Cognitive, Emotional, and Social), 3 symptom scales (fatigue, pain, nausea, and vomiting), a global health status/global QoL scale, and single items (dyspnea, appetite loss, sleep disturbance, constipation, and diarrhea). Most questions used 4-point scale (1='Not at all' to 4='Very much'), while 2 questions used 7-point scale (1='very poor' to 7='Excellent'). Scores were averaged, transformed to 0-100 scale; where higher score for functional scales=poor level of functioning; higher score for global health status/global QoL=better HRQoL. Baseline, D1 of Cycles 1, 2, 3, 4, 5, 6, STC/EW visit (up to C6D28), EoCTR visit (8 to 12 weeks after C6D1), and FUVs (every 12 weeks after EoCTR up to 3 years); Cycle length = 28 days
Secondary Change From Baseline in HRQoL as Measured by Quality of Life Questionnaire Associated CLL Module (QLQ-CLL16) Multi-Item Scales Score The EORTC QLQ-CLL16 module is designed for participants with Stage 0 to Stage 4 CLL. It is composed of 16 questions and there are four multi-item scales on Fatigue (2 items), Treatment-related side effects (TRSE, 4 items), Disease-related symptoms (DRS, 4 items), and Infection (4 items); and two single-item scales on social activities and future health worries. Multi-item scales score are reported and the total score for each multi-item scale was transformed to result in a total score range of 0 to 100, where higher score = poor HRQoL. Baseline, D1 of Cycles 1, 2, 3, 4, 5, 6, STC/EW visit (up to C6D28), EoCTR visit (8 to 12 weeks after C6D1), and FUVs (every 12 weeks after EoCTR up to 3 years); Cycle length = 28 days
Secondary Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs) An AE was defined as any untoward medical occurrence in a participant administered with Mircera and which does not necessarily have a causal relationship with Mircera. A Serious Adverse Event (SAE) is any significant hazard, contraindication, side effect that is fatal or life threatening; requires in-patient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; is medically significant or requires intervention to prevent one or other of the outcomes listed above. AEs were assessed using National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.0 (NCI CTCAE, v4.0) From signing of informed consent form up to approximately 8 years 5 months
Secondary Number of Participants With Grade 3 or Higher Tumor Lysis Syndrome (TLS) and Infusion-related Reactions (IRRs) An AE was defined as any untoward medical occurrence in a participant administered with Mircera and which does not necessarily have a causal relationship with Mircera. A SAE is any significant hazard, contraindication, side effect that is fatal or life threatening; requires in-patient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; is medically significant or requires intervention to prevent one or other of the outcomes listed above. TLS and IRRs were graded using the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE v4.0). Grade 1 = Mild; Grade 2 = Moderate; Grade 3 = Severe or medically significant; Grade 4 = Life-threatening; Grade 5 = Death. A higher grade indicates a worse outcome. From signing of informed consent form up to approximately 8 years 5 months
See also
  Status Clinical Trial Phase
Recruiting NCT05400122 - Natural Killer (NK) Cells in Combination With Interleukin-2 (IL-2) and Transforming Growth Factor Beta (TGFbeta) Receptor I Inhibitor Vactosertib in Cancer Phase 1
Enrolling by invitation NCT01804686 - A Long-term Extension Study of PCI-32765 (Ibrutinib) Phase 3
Completed NCT02057185 - Occupational Status and Hematological Disease
Active, not recruiting NCT04240704 - Safety and Preliminary Efficacy of JBH492 Monotherapy in Patients With CLL and NHL Phase 1
Recruiting NCT03676504 - Treatment of Patients With Relapsed or Refractory CD19+ Lymphoid Disease With T Cells Expressing a Third-generation CAR Phase 1/Phase 2
Active, not recruiting NCT03280160 - Protocol GELLC-7: Ibrutinib Followed by Ibrutinib Consolidation in Combination With Ofatumumab Phase 2
Active, not recruiting NCT03844048 - An Extension Study of Venetoclax for Subjects Who Have Completed a Prior Venetoclax Clinical Trial Phase 3
Completed NCT00038025 - A Study Of Deoxycoformycin(DCF)/Pentostatin In Lymphoid Malignancies Phase 2
Recruiting NCT04904588 - HLA-Mismatched Unrelated Donor Hematopoietic Cell Transplantation With Post-Transplantation Cyclophosphamide Phase 2
Terminated NCT02231853 - Phase I/II Trial of Early Infusion of Rapidly-generated Multivirus Specific T Cells (MVST) to Prevent Post Transplant Viral Infections Phase 1
Recruiting NCT05417165 - Anti-pneumococcal Vaccine Strategy in Patients With Chronic Lymphocytic Leukemia Phase 2
Recruiting NCT04028531 - Understanding Chronic Lymphocytic Leukemia
Completed NCT00001637 - Immunosuppressive Preparation Followed by Blood Cell Transplant for the Treatment of Blood Cancers in Older Adults Phase 2
Completed NCT02910583 - Ibrutinib Plus Venetoclax in Subjects With Treatment-naive Chronic Lymphocytic Leukemia /Small Lymphocytic Lymphoma (CLL/SLL) Phase 2
Completed NCT01527045 - Donor Atorvastatin Treatment in Preventing Severe Acute GVHD After Nonmyeloablative Peripheral Blood Stem Cell Transplant in Patients With Hematological Malignancies Phase 2
Recruiting NCT04679012 - Polatuzumab Vedotin in Combination With Chemotherapy in Subjects With Richter's Transformation Phase 2
Recruiting NCT05405309 - RP-3500 and Olaparib in DNA Damage Repair Pathway Deficient Relapsed/Refractory Chronic Lymphocytic Leukemia Phase 1/Phase 2
Active, not recruiting NCT05023980 - A Study of Pirtobrutinib (LOXO-305) Versus Bendamustine Plus Rituximab (BR) in Untreated Patients With Chronic Lymphocytic Leukemia (CLL)/Small Lymphocytic Lymphoma (SLL) Phase 3
Recruiting NCT04553692 - Phase 1a/1b Study of Aplitabart (IGM-8444) Alone or in Combination in Participants With Relapsed, Refractory, or Newly Diagnosed Cancers Phase 1
Completed NCT04666025 - SARS-CoV-2 Donor-Recipient Immunity Transfer