Clinical Trials Logo

Clinical Trial Details — Status: Active, not recruiting

Administrative data

NCT number NCT02967692
Other study ID # CPDR001F2301
Secondary ID 2016-002794-35
Status Active, not recruiting
Phase Phase 3
First received
Last updated
Start date February 17, 2017
Est. completion date August 26, 2024

Study information

Verified date June 2024
Source Novartis
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

To evaluate the safety and efficacy of the combination of an anti-PD-1 antibody (Spartalizumab (PDR001)), a BRAF inhibitor (dabrafenib) and a MEK inhibitor (trametinib) in unresectable or metastatic BRAF V600 mutant melanoma


Description:

This study is designed as a Phase III, multi-center study consisting of three parts: - Part 1, known as the Safety Run-in, is an open-label part aimed at determining the recommended Phase III regimen (RP3R) of spartalizumab in combination with dabrafenib and trametinib for previously untreated subjects with BRAF V600 mutant unresectable or metastatic melanoma (Stage IIIC/IV per AJCC edition 7). In Part 1, spartalizumab was administered at a starting dose level (DL1) of 400 mg every 4 weeks (Q4W), along with fixed doses of dabrafenib (150 mg twice daily) and trametinib (2 mg once daily). The RP3R for Part 3 was determined using the Bayesian Logistic Regression Model (BLRM) with escalation with overdose control (EWOC) criteria. - Part 2, referred to as the Biomarker Cohort, is an open-label section focused on characterizing the kinetics of immune biomarkers and potential immune resistance mechanisms. Part 2 started when the fourth subject in dose level 1 (DL1) of Part 1 completed approximately 4 weeks of study treatment, and fewer than 3 dose-limiting toxicities (DLTs) were observed. Participants in Part 2 receive PDR001 (spartalizumab) at a dosage of 400 mg Q4W, in combination with dabrafenib (150 mg BID) and trametinib (2 mg QD). - Part 3 is a double-blind, randomized, placebo-controlled phase that compares the efficacy and safety of spartalizumab in combination with dabrafenib and trametinib to placebo in combination with dabrafenib and trametinib. Part 3 was initiated after determining the RP3R for the combination of spartalizumab with dabrafenib and trametinib in Part 1. Subjects were randomized in a 1:1 ratio to receive either the RP3R dose of spartalizumab identified in Part 1 or placebo, along with dabrafenib (150 mg BID) and trametinib (2 mg QD). For all parts of the study, the treatment is continued until the subject experiences any of the following events: disease progression according to RECIST 1.1 as determined by the Investigator, unacceptable toxicity, initiation of a new anti-neoplastic therapy, pregnancy, withdrawal of consent, physician's decision, loss to follow-up, death, or termination of the study by the Sponsor. Safety evaluations are conducted for all subjects for up to 150 days after the last dose of spartalizumab/placebo (safety follow-up period). Subjects who discontinue study treatment without disease progression as per RECIST 1.1 continue with tumor assessments according to the protocol until documented disease progression, withdrawal of consent, loss to follow-up, or death, regardless of the initiation of new anti-neoplastic therapy (efficacy follow-up period). Subjects enter the survival follow-up period after completing the safety follow-up period or experiencing disease progression as per RECIST 1.1 or response criteria for immunotherapy, whichever period is longer (survival follow-up period).


Recruitment information / eligibility

Status Active, not recruiting
Enrollment 569
Est. completion date August 26, 2024
Est. primary completion date August 11, 2020
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility Inclusion criteria Part 1: Safety run-in - Histologically confirmed, unresectable or metastatic melanoma with BRAF V600 mutation - Aspartate transaminase (AST) < 2.5× ULN and Alanine transaminase (ALT) < 2.5× ULN - Measurable disease according to RECIST 1.1 - ECOG performance status = 1 Part 2: Biomarker cohort - Histologically confirmed, unresectable or metastatic melanoma with BRAF V600 mutation - At least two cutaneous or subcutaneous or nodal lesions for tumor sample collection - Measurable disease according to RECIST 1.1 - ECOG performance status = 2 Part 3: Double-blind, randomized, placebo-controlled part - Histologically confirmed, unresectable or metastatic melanoma with BRAF V600 mutation - ECOG performance status = 2 - Measurable disease according to RECIST 1.1 Exclusion Criteria: Part 1: Safety run-in - Subjects with uveal or mucosal melanoma - Any history of CNS metastases - Prior systemic anti-cancer treatment for unresectable or metastatic melanoma - Neoadjuvant and/or adjuvant therapy for melanoma completed less than 6 months prior to enrollmen - Radiation therapy within 4 weeks prior to start of study treatment - Active autoimmune disease, and/or history of autoimmune disease(s) that required treatment Parts 2 & 3: Biomarker cohort & double-blind, randomized, placebo-controlled part - Subjects with uveal or mucosal melanoma - Prior systemic anti-cancer treatment for unresectable or metastatic melanoma - Neoadjuvant and/or adjuvant therapy for melanoma completed less than 6 months prior to enrollment - Radiation therapy within 4 weeks prior to start of study treatment - Clinically active cerebral melanoma metastasis. - Active autoimmune disease, and/or history of autoimmune disease(s) that required treatment Other protocol-defined Inclusion/Exclusion may apply.

Study Design


Related Conditions & MeSH terms


Intervention

Biological:
Spartalizumab
Spartalizumab powder for solution is used in Part 1 and Part 2, and as concentrate for solution for infusion for Part 3. Spartalizumab is administered via intravenous infusion over 30 minutes once every 4 weeks
Other:
Placebo
Placebo is administered via intravenous infusion over 30 minutes once every 4 weeks
Drug:
Dabrafenib
Dabrafenib 150 mg capsules BID is administered orally for Days 1-28 of a 28-day cycle, in fasting conditions.
Trametinib
Trametinib 2 mg tablets QD is administered orally for Days 1-28 of a 28-day cycle, in fasting conditions

Locations

Country Name City State
Argentina Novartis Investigative Site Buenos Aires
Argentina Novartis Investigative Site Caba Buenos Aires
Argentina Novartis Investigative Site Caba Buenos Aires
Argentina Novartis Investigative Site Rosario Santa Fe
Australia Novartis Investigative Site Gateshead New South Wales
Australia Novartis Investigative Site Greenslopes Queensland
Australia Novartis Investigative Site Nedlands Western Australia
Australia Novartis Investigative Site North Sydney New South Wales
Australia Novartis Investigative Site Prahran Victoria
Austria Novartis Investigative Site Graz
Austria Novartis Investigative Site Innsbruck Tyrol
Austria Novartis Investigative Site Linz
Austria Novartis Investigative Site Salzburg
Austria Novartis Investigative Site St Poelten
Belgium Novartis Investigative Site Bruxelles
Belgium Novartis Investigative Site Jette Brussel
Brazil Novartis Investigative Site Curitiba PR
Brazil Novartis Investigative Site Porto Alegre RS
Brazil Novartis Investigative Site Rio de Janeiro
Brazil Novartis Investigative Site Sao Paulo SP
Bulgaria Novartis Investigative Site Plovdiv
Bulgaria Novartis Investigative Site Sofia
Canada Novartis Investigative Site Montreal Quebec
Canada Novartis Investigative Site Montreal Quebec
Canada Novartis Investigative Site Sherbrooke Quebec
Canada Novartis Investigative Site Toronto Ontario
Canada Novartis Investigative Site Vancouver British Columbia
Chile Novartis Investigative Site Santiago
Chile Novartis Investigative Site Santiago
Chile Novartis Investigative Site Temuco Araucania
Czechia Novartis Investigative Site Brno
Czechia Novartis Investigative Site Hradec Kralove CZE
Czechia Novartis Investigative Site Olomouc CZE
Czechia Novartis Investigative Site Ostrava Poruba
Czechia Novartis Investigative Site Praha
Czechia Novartis Investigative Site Praha 10
Czechia Novartis Investigative Site Zlin Czech Republic
Denmark Novartis Investigative Site Aarhus
France Novartis Investigative Site Amiens
France Novartis Investigative Site Besancon Cedex
France Novartis Investigative Site Bobigny Cedex
France Novartis Investigative Site Bordeaux Cedex
France Novartis Investigative Site Boulogne Billancourt
France Novartis Investigative Site Caen
France Novartis Investigative Site Clermont Ferrand
France Novartis Investigative Site Dijon
France Novartis Investigative Site Grenoble
France Novartis Investigative Site Le Mans Cedex 09
France Novartis Investigative Site Lille
France Novartis Investigative Site Limoges Haute Vienne
France Novartis Investigative Site Lorient
France Novartis Investigative Site Lyon
France Novartis Investigative Site Marseille
France Novartis Investigative Site Mulhouse cedex
France Novartis Investigative Site Nice
France Novartis Investigative Site Paris 10
France Novartis Investigative Site Pierre Benite
France Novartis Investigative Site Poitiers
France Novartis Investigative Site Reims
France Novartis Investigative Site Rouen Cedex
France Novartis Investigative Site Strasbourg Cedex
France Novartis Investigative Site Toulouse
France Novartis Investigative Site Vandoeuvre-les-Nancy
France Novartis Investigative Site Villejuif
Germany Novartis Investigative Site Berlin
Germany Novartis Investigative Site Berlin
Germany Novartis Investigative Site Bonn
Germany Novartis Investigative Site Chemnitz
Germany Novartis Investigative Site Dresden
Germany Novartis Investigative Site Duesseldorf
Germany Novartis Investigative Site Erfurt
Germany Novartis Investigative Site Essen
Germany Novartis Investigative Site Freiburg
Germany Novartis Investigative Site Gera
Germany Novartis Investigative Site Halle Saale
Germany Novartis Investigative Site Hamburg
Germany Novartis Investigative Site Hannover
Germany Novartis Investigative Site Heidelberg
Germany Novartis Investigative Site Homburg
Germany Novartis Investigative Site Kiel
Germany Novartis Investigative Site Leipzig
Germany Novartis Investigative Site Mainz
Germany Novartis Investigative Site Mannheim Baden Wuerttemberg
Germany Novartis Investigative Site Marburg
Germany Novartis Investigative Site Minden
Germany Novartis Investigative Site Muenchen
Germany Novartis Investigative Site Muenster
Germany Novartis Investigative Site Regensburg Bavaria
Germany Novartis Investigative Site Stade
Germany Novartis Investigative Site Tuebingen
Greece Novartis Investigative Site Athens
Greece Novartis Investigative Site Athens
Hungary Novartis Investigative Site Budapest
Hungary Novartis Investigative Site Debrecen
Hungary Novartis Investigative Site Szeged
Israel Novartis Investigative Site Haifa
Israel Novartis Investigative Site Jerusalem
Israel Novartis Investigative Site Ramat Gan
Italy Novartis Investigative Site Bari BA
Italy Novartis Investigative Site Bergamo BG
Italy Novartis Investigative Site Bologna BO
Italy Novartis Investigative Site Brescia BS
Italy Novartis Investigative Site Candiolo TO
Italy Novartis Investigative Site Genova GE
Italy Novartis Investigative Site Meldola FC
Italy Novartis Investigative Site Milano MI
Italy Novartis Investigative Site Milano MI
Italy Novartis Investigative Site Modena MO
Italy Novartis Investigative Site Monza MB
Italy Novartis Investigative Site Napoli
Italy Novartis Investigative Site Padova PD
Italy Novartis Investigative Site Roma RM
Italy Novartis Investigative Site Siena SI
Italy Novartis Investigative Site Torino TO
Italy Novartis Investigative Site Verona VR
Japan Tokyo Metropolitan Komagome Hospital Bunkyo ku Tokyo
Japan National Cancer Hospital Chuo ku Tokyo
Japan Kyushu University Hospital Fukuoka city Fukuoka
Japan Osaka International Cancer Institute Osaka-city Osaka
Japan Kyoto University Hospital Sakyo Ku Kyoto
Mexico Novartis Investigative Site Guadalajara Jalisco
Mexico Novartis Investigative Site Leon Guanajuato
Mexico Novartis Investigative Site Mexico Distrito Federal
Netherlands Novartis Investigative Site Breda
Netherlands Novartis Investigative Site Leiden
Norway Novartis Investigative Site Oslo
Poland Novartis Investigative Site Gdansk
Poland Novartis Investigative Site Warszawa
Portugal Novartis Investigative Site Lisboa
Portugal Novartis Investigative Site Porto
Russian Federation Novartis Investigative Site Chelyabinsk
Russian Federation Novartis Investigative Site Moscow
Russian Federation Novartis Investigative Site Moscow
Russian Federation Novartis Investigative Site Nizhny Novgorod
Russian Federation Novartis Investigative Site Omsk
Russian Federation Novartis Investigative Site Saint Petersburg
Russian Federation Novartis Investigative Site Samara
Russian Federation Novartis Investigative Site St Petersburg
Spain Novartis Investigative Site Badalona Catalunya
Spain Novartis Investigative Site Barcelona Catalunya
Spain Novartis Investigative Site Jerez Cadiz
Spain Novartis Investigative Site La Coruna Galicia
Spain Novartis Investigative Site Las Palmas de Gran Canaria
Spain Novartis Investigative Site Madrid
Spain Novartis Investigative Site Madrid
Spain Novartis Investigative Site Madrid
Spain Novartis Investigative Site Madrid
Spain Novartis Investigative Site Malaga Andalucia
Spain Novartis Investigative Site Oviedo Asturias
Spain Novartis Investigative Site Sevilla Andalucia
Spain Novartis Investigative Site Valencia Comunidad Valenciana
Sweden Novartis Investigative Site Goteborg
Sweden Novartis Investigative Site Lund
Sweden Novartis Investigative Site Stockholm
Switzerland Novartis Investigative Site Aarau
Switzerland Novartis Investigative Site Zuerich
Thailand Novartis Investigative Site Bangkok
Thailand Novartis Investigative Site Songkhla Hat Yai
United Kingdom Novartis Investigative Site Leicester
United Kingdom Novartis Investigative Site London
United Kingdom Novartis Investigative Site Manchester
United Kingdom Novartis Investigative Site Middlesbrough
United Kingdom Novartis Investigative Site Northwood Middlesex
United Kingdom Novartis Investigative Site Preston
United Kingdom Novartis Investigative Site Surrey England
United Kingdom Novartis Investigative Site Sutton Surrey
United Kingdom Novartis Investigative Site Truro Cornwall
United States California Cancer Associates for Research and Excellence SC-2 Encinitas California
United States Univ of TX MD Anderson Cancer Cntr SC 2 Houston Texas
United States University of Tennessee Medical Center SC Knoxville Tennessee
United States Johns Hopkins U SC Lutherville Maryland
United States NYU Laura and Isaac Perlmutter Cancer Center SC New York New York
United States Nebraska Cancer Specialists Omaha Nebraska
United States UC Irvine Medical Center SC Orange California
United States University of Pittsburgh Med Center SC Pittsburgh Pennsylvania
United States Utah Cancer Specialists SC 2 Salt Lake City Utah
United States California Pacific Medical Center San Francisco California
United States Stanford Cancer Center SC-2 Stanford California
United States University of Kansas Cancer Center SC Westwood Kansas

Sponsors (1)

Lead Sponsor Collaborator
Novartis Pharmaceuticals

Countries where clinical trial is conducted

United States,  Argentina,  Australia,  Austria,  Belgium,  Brazil,  Bulgaria,  Canada,  Chile,  Czechia,  Denmark,  France,  Germany,  Greece,  Hungary,  Israel,  Italy,  Japan,  Mexico,  Netherlands,  Norway,  Poland,  Portugal,  Russian Federation,  Spain,  Sweden,  Switzerland,  Thailand,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary Safety Run-In (Part 1): Number of Participants With Dose Limiting Toxicities (DLTs) DLT was defined as an adverse event or abnormal laboratory value that was unrelated to disease, disease progression, inter-current illness, or concomitant medications and occured within 8 weeks of treatment with spartalizumab in combination with dabrafenib and trametinib. The DLT criteria included Grade 4 hematological adverse events, Grade 4 bilirubin elevation, specific gastrointestinal adverse events, symptomatic serum amylase or lipase elevation, Grade 3 or higher hypertension, Grade 3 or higher cardiac events, Grade 2 or higher pneumonitis, Grade 3 or higher immune-related toxicities, infusion-related reactions, other clinically significant adverse events, and toxicities leading to a dosing delay of over 12 weeks. NCI CTCAE v4.03 was used for grading DLTs Up to 8 weeks (Part 1)
Primary Biomarker Cohort (Part 2): Change From Baseline in Programmed Cell Death-ligand 1 (PD-L1) Expression Upon Treatment With Spartalizumab in Combination With Dabrafenib and Trametinib Change from baseline in PD-L1 expression (as determined by immunohistochemistry in tissue samples) upon treatment with spartalizumab in combination with dabrafenib and trametinib in participants from Part 2 Baseline, Cycle 1 Day 15 and Cycle 3 Day 1 (Part 2). Each cycle is 28 days
Primary Biomarker Cohort (Part 2): Change From Baseline in CD8+ Cells Upon Treatment With Spartalizumab in Combination With Dabrafenib and Trametinib Change from baseline in CD8+ cells (as determined by flow cytometry in blood samples) upon treatment with spartalizumab in combination with dabrafenib and trametinib in participants from Part 2 Baseline, Cycle 1 Day 15 and Cycle 3 Day 1 (Part 2). Each cycle is 28 days
Primary Randomized (Part 3): Progression-Free Survival (PFS) as Per Investigator's Assessment by RECIST 1.1 Progression-free survival was defined as the time from the date of first dose to the date of the first documented radiological progression per investigator's assessment according to RECIST 1.1 or death due to any cause. The distribution of PFS was estimated using the Kaplan-Meier (KM) method. If a patient had not had an event at the time of data cut-off, progression-free survival was censored at the date of last adequate tumor assessment. Up to disease progression or death due to any cause, whichever occurs first, assessed up to 2.8 years (Part 3)
Secondary Overall Survival (OS) Overall survival is defined as the time from date of randomization to date of death due to any cause Up to death due to any cause, assessed up to approximately 5 years
Secondary Overall Response Rate (ORR) as Per Investigator's Assessment by RECIST 1.1 ORR was defined as the percentage of subjects with confirmed best overall response of complete response (CR) or partial response (PR), as per investigator's assessment by RECIST 1.1.
CR: Disappearance of all non-nodal target lesions. In addition, any pathological lymph nodes assigned as target lesions must have a reduction in short axis to <10 mm PR: At least a 30% decrease in the sum of diameter of all target lesions, taking as reference the baseline sum of diameters
Part 1: Up to 3.3 years. Part 2: Up to 3 years. Part 3: Up to 2.8 years
Secondary Duration of Response (DOR) as Per Investigator's Assessment by RECIST 1.1 DOR was defined as the time from first documented response of CR or PR to date of first documented progression or death, according to RECIST 1.1 criteria. The distribution of DOR was estimated using the KM method.
CR: Disappearance of all non-nodal target lesions. In addition, any pathological lymph nodes assigned as target lesions must have a reduction in short axis to <10 mm PR: At least a 30% decrease in the sum of diameter of all target lesions, taking as reference the baseline sum of diameters
From first documented response to date of first documented progression or death, up to 3.3 years (Part 1), 3 years (Part 2) and 2.8 years (Part 3)
Secondary Disease Control Rate (DCR) as Per Investigator's Assessment by RECIST 1.1 DCR was defined as the percentage of participants with CR or PR or subjects with stable disease (SD) lasting for a duration of at least 24 weeks as per local review according to RECIST 1.1 criteria.
CR: Disappearance of all non-nodal target lesions. In addition, any pathological lymph nodes assigned as target lesions must have a reduction in short axis to <10 mm PR: At least a 30% decrease in the sum of diameter of all target lesions, taking as reference the baseline sum of diameters SD: Neither sufficient shrinkage to qualify for PR or CR nor an increase in lesions which would qualify for progressive disease.
Part 1: Up to 3.3 years. Part 2: Up to 3 years. Part 3: Up to 2.8 year
Secondary Randomized (Part 3): Change From Baseline in European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ) C30- Global Health Status Scores The EORTC QLQ-C30 was a 30-item questionnaire that patients complete, consisting of both multi-item scales and single-item measures. It included five functional scales, three symptom scales, six single items, and a Global Health Status/Quality of Life (GHS/QoL) scale.
The GHS/QoL scale had seven possible response scores ranging from 1 (very poor) to 7 (excellent), which were averaged and transformed to a 0-100 scale. A higher score on this scale indicated a better quality of life.
The change from baseline in GHS/QoL scores was calculated. A positive change from baseline indicated improvement in the patient's quality of life.
From baseline to 60 days post progression, assessed up to 2.8 years (Part 3)
Secondary Randomized (Part 3): Change From Baseline in European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ) C30- Physical Functioning Scale Scores The EORTC QLQ-C30 was a patient completed 30 item questionnaire that was composed of both multi-item scales and single-item measures. These included five functional scales, three symptom scales, six single items and a global health status/QoL scale.
The EORTC QLQ-C30 physical functioning scale measured a patient's ability to carry out daily activities and tasks requiring physical exertion. It consisted of five questions asking patients to rate their level of physical functioning, with response options ranging from 1="not at all" to 4="very much". The scores for each item were summed and transformed to a 0 to 100 scale, with higher scores indicating better physical functioning.
The change from baseline in physical functioning scale scores was calculated. A positive change from baseline indicated improvement in physical functioning.
From baseline to 60 days post progression, assessed up to 2.8 years (Part 3)
Secondary Randomized (Part 3): Change From Baseline in European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ) C30- Pain Symptom Scale Scores The EORTC QLQ-C30 was a patient completed 30 item questionnaire that was composed of both multi-item scales and single-item measures. These included five functional scales, three symptom scales, six single items and a global health status/QoL scale.
The EORTC QLQ-C30 pain symptom scale was one of the symptom scales in the questionnaire, which measured the severity of pain experienced by the patient. The pain symptom scale consisted of two items, one measuring the severity of pain and the other measuring the use of painkillers. The items were rated on a 4-point scale ranging from 1="not at all" to 4="very much". The scores for each item were summed and transformed to a 0 to 100 scale, with higher scores indicating more severe pain.
The change from baseline in pain symptom scale scores was calculated. A negative change from baseline indicated improvement.
From baseline to 60 days post progression, assessed up to 2.8 years (Part 3)
Secondary Randomized (Part 3): Time to 10 Point Definitive Deterioration in European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ) C30- Global Health Status The EORTC QLQ-C30 was a patient completed 30 item questionnaire that was composed of both multi-item scales and single-item measures. These included five functional scales, three symptom scales, six single items and a global health status/QoL scale.
The GHS/QoL scale had seven possible response scores ranging from 1 (very poor) to 7 (excellent), which were averaged and transformed to a 0-100 scale. A higher score on this scale indicated a better quality of life.
The time to definitive 10 point deterioration is defined as the time from the date of randomization to the date of event, which is defined as at least 10 points relative to baseline worsening of the GHS/QoL score or death due to any cause. If a subject had not had an event, the time to deterioration was censored at the date of the last adequate assessment. The distribution was estimated using KM method.
From baseline to date of at least 10 points relative to baseline worsening of the global health status score or death due to any cause, up to 2.8 years (Part 3)
Secondary Randomized (Part 3): Change From Baseline in Function Assessment Cancer Therapy-melanoma (FACT-M) Melanoma Subscale Score The Functional Assessment of Cancer Therapy-Melanoma (FACT-M) quality of life questionnaire was composed of the FACT-General (FACT-G) plus the Melanoma Subscale and the Melanoma Surgery Subscale, which complemented the general scale with items specific to quality of life (QoL) in melanoma.
The Melanoma Subscale of FACT-M included 16 questions, with response options of 0= "Not at all", 1= "a little bit", 2= "somewhat", 3= "quite a bit" and 4= "very much". The FACT-M melanoma subscale score ranged from 0 to 64, with higher scores indicating a higher quality of life in relation to melanoma.
The change from baseline in melanoma subscale scores was calculated. A positive change from baseline indicated improvement.
From baseline to 60 days post progression, assessed up to 2.8 years (Part 3)
Secondary Randomized (Part 3): Change From Baseline in EuroQoL 5-level Instrument (EQ-5D-5L)- Visual Analog Scale (VAS) Score The EQ-5D-5L is a standardized questionnaire used to assess health-related quality of life, and it includes a Visual Analog Scale (VAS). The VAS score is obtained by asking the individual to rate their current health status on a scale from 0 to 100, where 0 represents the worst possible health state and 100 represents the best possible health state.
The change from baseline in EQ-5D-5L VAS score was calculated. A positive change from baseline indicates improvement in the health status.
From baseline to 60 days post progression, assessed up to 2.8 years (Part 3)
Secondary Randomized (Part 3): PFS as Per Investigator's Assessment by RECIST 1.1 by PD-L1 Expression PFS was defined as the time from the date of first dose to the date of the first documented radiological progression as per investigator's assessment using RECIST 1.1 response criteria or death due to any cause. The distribution of PFS was estimated using the KM method. If a patient had not had an event at the time of data cut-off, progression-free survival was censored at the date of last adequate tumor assessment.
PFS analysis was performed by PD-L1 status (positive, negative) where a positive status was defined as having = 1% expression and a negative status was defined as having < 1% expression.
Up to disease progression or death due to any cause, up to 2.8 years (Part 3)
Secondary Randomized (Part 3): OS by PD-L1 Expression OS is defined as the time from date of randomization to date of death due to any cause.
OS analysis will be performed by PD-L1 subgroup (positive, negative) where a positive status is defined as having = 1% expression and a negative status is defined as having < 1% expression.
Up to death due to any cause, up to 5 years
Secondary Spartalizumab Anti-drug Antibody (ADA) Prevalence at Baseline Spartalizumab ADA prevalence at baseline was calculated as the percentage of participants who had an spartalizumab ADA positive result at baseline. Baseline
Secondary Spartalizumab ADA Incidence Spartalizumab ADA incidence was calculated as the percentage of participants who were treatment-induced spartalizumab ADA positive (post-baseline ADA positive with ADA-negative sample at baseline) and treatment-boosted spartalizumab ADA positive (post-baseline ADA positive with titer that is at least the fold titer change greater than the ADA-positive baseline titer) Throughout study until 150 days after the last dose of spartalizumab, up to 3.3 years (Part 1), 3 years (Part 2) and 2.8 years (Part 3).
Secondary Trough Concentration (Ctrough) for Spartalizumab Ctrough for spartalizumab refers to the serum concentration of spartalizumab immediately prior to the administration of a dose of spartalizumab on Day 1 of Cycle 2 and later cycles. Pre-infusion on Day 1 of each Cycle starting from Cycle 2, up to 3.3 years (Part 1), 3 years (Part 2) and 2.8 years (Part 3). Cycle=28 days
Secondary Pre-dose Plasma Concentration for Dabrafenib Plasma concentration of dabrafenib immediately prior to the administration of a dose of dabrafenib. Pre-infusion on Day 1 of every cycle from Cycle 2 to 12, and then every 6 cycles from Cycle 18 to 36, up to 3.3 years (Part 1), 3 years (Part 2) and 2.8 years (Part 3). Cycle=28 days
Secondary Pre-dose Plasma Concentration for Trametinib Plasma concentration of trametinib immediately prior to the administration of a dose of trametinib. Pre-infusion on Day 1 of every cycle from Cycle 2 to 12, and then every 6 cycles from Cycle 18 to 36, up to 3.3 years (Part 1), 3 years (Part 2) and 2.8 years (Part 3). Cycle=28 days
Secondary Number of Participants With Dose Interruptions Number of participants with dose interruptions for spartalizumab, dabrafenib and trametinib From baseline to end of treatment, up to 5 years
Secondary Number of Participants With Dose Reductions Number of patients with dose reductions for spartalizumab, dabrafenib and trametinib From baseline to end of treatment, up to 5 years
Secondary Relative Dose Intensity Relative dose intensity for spartalizumab, dabrafenib and trametinib computed as the ratio of dose intensity and planned dose intensity From baseline to end of treatment, up to 5 years
See also
  Status Clinical Trial Phase
Recruiting NCT05094804 - A Study of OR2805, a Monoclonal Antibody Targeting CD163, Alone and in Combination With Anticancer Agents Phase 1/Phase 2
Completed NCT03979872 - Risk Information and Skin-cancer Education for Undergraduate Prevention N/A
Recruiting NCT04986748 - Using QPOP to Predict Treatment for Sarcomas and Melanomas
Enrolling by invitation NCT00068003 - Harvesting Cells for Experimental Cancer Treatments
Recruiting NCT05707286 - Pilot Study to Determine Pro-Inflammatory Cytokine Kinetics During Immune Checkpoint Inhibitor Therapy
Active, not recruiting NCT05470283 - Phase I, Open-Label, Study of Tumor Infiltrating Lymphocytes Engineered With Membrane Bound IL15 Plus Acetazolamide in Adult Patients With Metastatic Melanoma Phase 1
Recruiting NCT05077137 - A Feasibility Study Utilizing Immune Recall to Increase Response to Checkpoint Therapy Phase 1
Active, not recruiting NCT02721459 - XL888 + Vemurafenib + Cobimetinib for Unresectable BRAF Mutated Stage III/IV Melanoma Phase 1
Completed NCT00341939 - Retrospective Analysis of a Drug-Metabolizing Genotype in Cancer Patients and Correlation With Pharmacokinetic and Pharmacodynamics Data
Recruiting NCT05839912 - Excision of Lymph Node Trial (EXCILYNT) (Mel69) N/A
Recruiting NCT04971499 - A Study of Dapansutrile Plus Pembrolizumab in Patients With PD-1 Refractory Advanced Melanoma Phase 1/Phase 2
Recruiting NCT05263453 - HL-085+Vemurafenib to Treat Advanced Melanoma Patients With BRAF V600E/K Mutation Phase 2
Active, not recruiting NCT05060432 - Study of EOS-448 With Standard of Care and/or Investigational Therapies in Participants With Advanced Solid Tumors Phase 1/Phase 2
Not yet recruiting NCT06413680 - A First-In Human (FIH) Trial to Find Out if REGN10597 is Safe and How Well it Works for Adult Participants With Advanced Solid Organ Malignancies Phase 1/Phase 2
Completed NCT03348891 - TNF in Melanoma Patients Treated With Immunotherapy N/A
Terminated NCT03399448 - NY-ESO-1-redirected CRISPR (TCRendo and PD1) Edited T Cells (NYCE T Cells) Phase 1
Completed NCT03171064 - Exercise as a Supportive Measure for Patients Undergoing Checkpoint-inhibitor Treatment Phase 2
Not yet recruiting NCT05539118 - Interferon-α1b Combined With Toripalimab and Anlotinib Hydrochloride in Advanced Unresectable Melanoma Phase 1/Phase 2
Recruiting NCT05171374 - pRospective Evaluation of Clinical Outcomes in Patients With metAsTatIс melanOma Treated With dabrafeNib and trAmetinib in reaL practicE
Withdrawn NCT02854488 - Yervoy Pregnancy Surveillance Study