Clinical Trials Logo

Clinical Trial Details — Status: Terminated

Administrative data

NCT number NCT04128696
Other study ID # 209229
Secondary ID 2019-002263-99
Status Terminated
Phase Phase 2/Phase 3
First received
Last updated
Start date November 21, 2019
Est. completion date June 20, 2023

Study information

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

Clinical Trial Summary

The purpose of study is to evaluate if the addition of GSK3359609 to pembrolizumab as first-line treatment improves the efficacy of pembrolizumab in participants with recurrent or metastatic (R/M) head and neck squamous cell carcinoma/cancer (HNSCC).This is a randomized, double-blind, adaptive Phase II/III study comparing a combination of GSK3359609 inducible T cell co-stimulatory receptor (ICOS) agonist and pembrolizumab to pembrolizumab plus placebo in participants with programmed death receptor 1-ligand 1 (PD-L1) combined positive score (CPS) >=1 R/M HNSCC.


Recruitment information / eligibility

Status Terminated
Enrollment 315
Est. completion date June 20, 2023
Est. primary completion date April 27, 2021
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility Inclusion Criteria: - Capable of giving signed informed consent - Male or female, age >=18 years - Histological or cytological documentation of Head and Neck Squamous Cell Carcinoma (HNSCC) that is considered incurable by local therapies - Primary tumor location of the oral cavity, oropharynx, hypopharynx or larynx - No prior systemic therapy administered in the recurrent or metastatic setting (except for systemic therapy given as part of multimodal treatment for locally advanced disease) - Measurable disease per RECIST version 1.1 guidelines - ECOG Performance PS score of 0 or 1 - Adequate organ function - Life expectancy of at least 12 weeks - Female participants: must not be pregnant, not breastfeeding, and at least one of the following conditions apply: 1. Not a woman of childbearing potential (WOCBP) 2. A WOCBP who agrees to use a method of birth control from 30 days prior to randomization and for at least 120 days after the last dose of study treatment - Male participants with female partners of child-bearing potential: must agree to use a highly effective contraception while receiving study treatment and for at least 120 days after the last dose of study treatment and refrain from donating sperm during this period - Provide tumor tissue from excisional or core biopsy (fine needle aspirates and bone biopsies are not acceptable) acquired within 2 years prior to randomization for PD-L1 immunohistochemistry (IHC) testing by central laboratory - Have PD-L1 Immunohistochemistry (IHC) CPS 1 status by central laboratory testing - Have results from testing of Human Papilloma Virus (HPV) status for oropharyngeal cancer Exclusion Criteria: - Prior therapy with an anti-PD-1/L1/L2 and/or anti-ICOS directed agent - Systemic approved or investigational anticancer therapy within 30 days or 5 half-lives of the drug, whichever is shorter - Major surgery 28 days prior to randomization - Has high risk of bleeding - Toxicity related to prior treatment that has not resolved to <=Grade 1 (except alopecia, hearing loss, endocrinopathy managed with replacement therapy, and peripheral neuropathy which must be<= Grade 2) - Received transfusion of blood products or administration of colony stimulating factors within 14 days prior to randomization - Central nervous system (CNS) metastases, with the following exception: Participants with asymptomatic CNS metastases who are clinically stable and have no requirement for steroids for at least 14 days prior to randomization - Invasive malignancy or history of invasive malignancy other than disease under study within the last 3 years, except as noted below: a. Any other invasive malignancy for which the participant was definitively treated, has been disease-free for 3 years and in the opinion of the principal investigator and GSK Medical Monitor will not affect the evaluation of the effects of the study treatment on the currently targeted malignancy, may be included in this clinical study - Autoimmune disease or syndrome that required systemic treatment within the past 2 years - Has a diagnosis of immunodeficiency or is receiving systemic steroids (=10 mg oral prednisone per day or equivalent) or other immunosuppressive agents within 7 days prior to randomization - Receipt of any live vaccine within 30 days prior randomization - Prior allogeneic/autologous bone marrow or solid organ transplantation - Has current pneumonitis or history of non-infectious pneumonitis that required steroids or other immunosuppressive agents - Recent history (within the past 6 months) of uncontrolled symptomatic ascites, pleural or pericardial effusions - Recent history (within the past 6 months) of gastrointestinal obstruction that required surgery, acute diverticulitis, inflammatory bowel disease, or intra-abdominal abscess - Recent history of allergen desensitization therapy within 4 weeks of randomization - History or evidence of cardiac abnormalities within the 6 months prior to randomization - Cirrhosis or current unstable liver or biliary disease per investigator assessment defined by the presence of ascites, encephalopathy, coagulopathy, hypoalbuminemia, esophageal or gastric varices, or persistent jaundice - Active infection requiring systemic therapy - Known HIV infection, or positive test for hepatitis B active infection (presence of hepatitis B surface antigen), or hepatitis C active infection - History of severe hypersensitivity to monoclonal antibodies or any ingredient used in the study treatment formulations - Known history of active tuberculosis - Any serious and/or unstable pre-existing medical (aside from malignancy), psychiatric disorder, or other condition that could interfere with participant's safety, obtaining informed consent, or compliance to the study procedures in the opinion of the investigator - Is currently participating in (unless in follow-up phase and 4 weeks have elapsed from last dose of prior investigational agent), or has participated in a study of an investigational agent or has used an investigational device within 4 weeks prior to date of randomization - Pregnant, breastfeeding, or expecting to conceive or father children within the projected duration of the study

Study Design


Related Conditions & MeSH terms


Intervention

Drug:
feladilimab
feladilimab is available as an intravenous infusion.
Pembrolizumab
Pembrolizumab is available as an intravenous infusion.
Placebo
Placebo is available as an intravenous infusion.

Locations

Country Name City State
Argentina GSK Investigational Site Ciudad Autónoma de Buenos Aires
Argentina GSK Investigational Site Ciudad Autónoma de Buenos Aires Buenos Aires
Argentina GSK Investigational Site La Plata Buenos Aires
Argentina GSK Investigational Site San Juan
Australia GSK Investigational Site Blacktown New South Wales
Australia GSK Investigational Site Darlinghurst
Australia GSK Investigational Site Heidelberg Victoria
Australia GSK Investigational Site Herston Queensland
Australia GSK Investigational Site Melbourne Victoria
Australia GSK Investigational Site Nedlands Western Australia
Australia GSK Investigational Site St Leonards New South Wales
Brazil GSK Investigational Site Barretos São Paulo
Brazil GSK Investigational Site Belo Horizonte, Minas Gerais
Brazil GSK Investigational Site Florianopolis Santa Catarina
Brazil GSK Investigational Site Sao Jose do Rio Preto São Paulo
Brazil GSK Investigational Site São Paulo
Brazil GSK Investigational Site São Paulo
Brazil GSK Investigational Site Vitória Espírito Santo
Canada GSK Investigational Site Calgary Alberta
Canada GSK Investigational Site Edmonton Alberta
Canada GSK Investigational Site Hamilton Ontario
Canada GSK Investigational Site Montreal Quebec
Canada GSK Investigational Site Montreal Quebec
Canada GSK Investigational Site Quebec City Quebec
Canada GSK Investigational Site Rimouski Quebec
Canada GSK Investigational Site Toronto Ontario
Canada GSK Investigational Site Vancouver British Columbia
China GSK Investigational Site Bengbu
China GSK Investigational Site Chengdu Sichuan
China GSK Investigational Site Chengdu Sichuan
China GSK Investigational Site Guangzhou Guangdong
China GSK Investigational Site Guangzhou Guangdong
China GSK Investigational Site Guiyang Guizhou
China GSK Investigational Site Harbin
China GSK Investigational Site Hefei
China GSK Investigational Site Nanchang Jiangxi
China GSK Investigational Site Nanning Guangxi
China GSK Investigational Site Shnghai
China GSK Investigational Site Wuhan Hubei
Denmark GSK Investigational Site Copenhagen
France GSK Investigational Site Bordeaux
France GSK Investigational Site Epagny Metz-Tessy
France GSK Investigational Site Le Mans
France GSK Investigational Site Lille
France GSK Investigational Site Lyon cedex 08
France GSK Investigational Site Paris
France GSK Investigational Site Saint Herblain cedex
France GSK Investigational Site Strasbourg
France GSK Investigational Site Toulouse Cedex 9
France GSK Investigational Site Valenciennes Cedex
Germany GSK Investigational Site Aachen Nordrhein-Westfalen
Germany GSK Investigational Site Berlin
Germany GSK Investigational Site Hamburg
Germany GSK Investigational Site Hamburg
Germany GSK Investigational Site Leipzig Sachsen
Germany GSK Investigational Site Ulm Baden-Wuerttemberg
Greece GSK Investigational Site Heraklion,Crete
Greece GSK Investigational Site Thessaloniki
Greece GSK Investigational Site Thessaloniki
Ireland GSK Investigational Site Dublin
Ireland GSK Investigational Site Dublin
Israel GSK Investigational Site Jerusalem
Israel GSK Investigational Site Petah Tikva
Israel GSK Investigational Site Ramat Gan
Italy GSK Investigational Site Brescia Lombardia
Italy GSK Investigational Site Candiolo Piemonte
Italy GSK Investigational Site Legnago (VR) Veneto
Italy GSK Investigational Site Meldola (FC) Emilia-Romagna
Italy GSK Investigational Site Milano Lombardia
Italy GSK Investigational Site Milano Lombardia
Italy GSK Investigational Site Napoli Campania
Japan GSK Investigational Site Chiba
Japan GSK Investigational Site Chiba
Japan GSK Investigational Site Ehime
Japan GSK Investigational Site Fukuoka
Japan GSK Investigational Site Hokkaido
Japan GSK Investigational Site Hyogo
Japan GSK Investigational Site Ibaraki
Japan GSK Investigational Site Iwate
Japan GSK Investigational Site Kagawa
Japan GSK Investigational Site Kanagawa
Japan GSK Investigational Site Miyagi
Japan GSK Investigational Site Niigata
Japan GSK Investigational Site Osaka
Japan GSK Investigational Site Saitama
Japan GSK Investigational Site Shizuoka
Japan GSK Investigational Site Tokyo
Japan GSK Investigational Site Tokyo
Korea, Republic of GSK Investigational Site Busan
Korea, Republic of GSK Investigational Site Hwasun-gun, Jeollanam-do
Korea, Republic of GSK Investigational Site Incheon
Korea, Republic of GSK Investigational Site Seongnam-si, Gyeonggi-do
Korea, Republic of GSK Investigational Site Seoul
Korea, Republic of GSK Investigational Site Seoul
Korea, Republic of GSK Investigational Site Seoul
Korea, Republic of GSK Investigational Site Seoul,
Mexico GSK Investigational Site Guadalajara Jalisco
Mexico GSK Investigational Site Saltillo Coahuila
Netherlands GSK Investigational Site Maastricht
Netherlands GSK Investigational Site Rotterdam
Norway GSK Investigational Site Bergen
Norway GSK Investigational Site Oslo
Poland GSK Investigational Site Bydgoszcz
Poland GSK Investigational Site Gdynia
Poland GSK Investigational Site Gliwice
Poland GSK Investigational Site Krakow
Poland GSK Investigational Site Olsztyn
Poland GSK Investigational Site Tomaszow Mazowiecki
Poland GSK Investigational Site Warszawa
Portugal GSK Investigational Site Coimbra
Portugal GSK Investigational Site Lisboa
Portugal GSK Investigational Site Matosinhos
Portugal GSK Investigational Site Porto
Portugal GSK Investigational Site Porto
Romania GSK Investigational Site Brasov
Romania GSK Investigational Site Bucuresti
Romania GSK Investigational Site Cluj Napoca
Romania GSK Investigational Site Cluj-Napoca
Romania GSK Investigational Site Constanta
Romania GSK Investigational Site Craiova
Romania GSK Investigational Site Floresti
Romania GSK Investigational Site Iasi
Romania GSK Investigational Site Oradea
Romania GSK Investigational Site Otopeni
Romania GSK Investigational Site Satu Mare
Romania GSK Investigational Site Suceava
Russian Federation GSK Investigational Site Moscow
Russian Federation GSK Investigational Site Poselok Kuzmolovsky
Russian Federation GSK Investigational Site Pushkin
Russian Federation GSK Investigational Site Saint-Petersburg
Russian Federation GSK Investigational Site St. Petersburg
Russian Federation GSK Investigational Site Yaroslavl
Spain GSK Investigational Site Barcelona
Spain GSK Investigational Site Barcelona
Spain GSK Investigational Site Madrid
Spain GSK Investigational Site Málaga
Spain GSK Investigational Site Pozuelo De Alarcón/Madrid
Spain GSK Investigational Site Santiago de Compostela
Spain GSK Investigational Site Valencia
Spain GSK Investigational Site Zaragoza
Switzerland GSK Investigational Site St Gallen
Switzerland GSK Investigational Site Zuerich
Taiwan GSK Investigational Site Changhua
Taiwan GSK Investigational Site Kaohsiung City
Taiwan GSK Investigational Site Taipei
Taiwan GSK Investigational Site Taipei
Taiwan GSK Investigational Site Taipei
Taiwan GSK Investigational Site Taoyuan City
United Kingdom GSK Investigational Site London
United Kingdom GSK Investigational Site Manchester Lancashire
United Kingdom GSK Investigational Site Nottingham
United Kingdom GSK Investigational Site Sutton
United States GSK Investigational Site Boston Massachusetts
United States GSK Investigational Site Charleston South Carolina
United States GSK Investigational Site Charlotte North Carolina
United States GSK Investigational Site Chattanooga Tennessee
United States GSK Investigational Site Duarte California
United States GSK Investigational Site Greenville South Carolina
United States GSK Investigational Site Indianapolis Indiana
United States GSK Investigational Site Nashville Tennessee
United States GSK Investigational Site Philadelphia Pennsylvania
United States GSK Investigational Site Pittsburgh Pennsylvania
United States GSK Investigational Site Saint Petersburg Florida
United States GSK Investigational Site Seattle Washington
United States GSK Investigational Site Washington District of Columbia

Sponsors (2)

Lead Sponsor Collaborator
GlaxoSmithKline Merck Sharp & Dohme LLC

Countries where clinical trial is conducted

United States,  Argentina,  Australia,  Brazil,  Canada,  China,  Denmark,  France,  Germany,  Greece,  Ireland,  Israel,  Italy,  Japan,  Korea, Republic of,  Mexico,  Netherlands,  Norway,  Poland,  Portugal,  Romania,  Russian Federation,  Spain,  Switzerland,  Taiwan,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary Overall Survival (OS) in the Programmed Death Receptor-ligand 1 (PD-L1) Combined Positive Score (CPS) =1 Population OS was defined as the time from the date of randomization to the date of death due to any cause. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Kaplan-Meier estimate for the median OS is presented, along with associated 95% confidence interval, estimated using the Brookmeyer-Crowley method. Up to approximately 16 months
Primary OS in the PD-L1 Expression High (CPS =20) Population OS was defined as the time from the date of randomization to the date of death due to any cause. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Kaplan-Meier estimate for the median OS is presented, along with associated 95% confidence interval, estimated using the Brookmeyer-Crowley method. Up to approximately 16 months
Primary Progression-free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors (RECIST) in the PD-L1 CPS =1 Population PFS per RECIST version (v)1.1 was defined as the time from the date of randomization to the date of first documented disease progression or death due to any cause, whichever occurs first. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Kaplan-Meier estimate for the median PFS is presented, along with associated 95% confidence interval, estimated using the Brookmeyer-Crowley method. Up to approximately 16 months
Secondary PFS Per Immune-based RECIST (iRECIST) in the PD-L1 CPS =1 Population PFS per iRECIST was defined as the interval of time from the date of randomization to the date of the first documented disease progression confirmed consecutively per iRECIST based on investigator assessment, or death due to any cause, whichever occurs first. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Kaplan-Meier estimate for the median PFS is presented, along with associated 95% confidence interval, estimated using the Brookmeyer-Crowley method. Up to approximately 16 months
Secondary PFS Per RECIST in the PD-L1 CPS =20 Population PFS per RECIST v1.1 was defined as the time from the date of randomization to the date of first documented disease progression per RECIST v1.1. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Kaplan-Meier estimate for the median PFS is presented, along with associated 95% confidence interval, estimated using the Brookmeyer-Crowley method. Up to approximately 16 months
Secondary PFS Per iRECIST (iPFS) in the PD-L1 CPS =20 Population PFS per iRECIST was defined as the interval of time from the date of randomization to the date of the first documented disease progression confirmed consecutively per iRECIST based on investigator assessment, or death due to any cause, whichever occurs first. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Kaplan-Meier estimate for the median PFS is presented, along with associated 95% confidence interval, estimated using the Brookmeyer-Crowley method. Up to approximately 16 months
Secondary Milestone OS Rate at 12 Months in the PD-L1 CPS =1 Population Milestone OS rate at 12 months was estimated using the Kaplan-Meier method. Associated 95% confidence intervals are estimated using the Brookmeyer-Crowley method. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. 12 months
Secondary Milestone OS Rate at 24 Months in the PD-L1 CPS =1 Population Milestone OS rate at 24 months was not evaluated. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. 24 months
Secondary Milestone OS Rate at 12 Months in the PD-L1 CPS =20 Population Milestone OS rate at 12 months was estimated using the Kaplan-Meier method. Associated 95% confidence intervals are estimated using the Brookmeyer-Crowley method. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. 12 months
Secondary Milestone OS Rate at 24 Months in the PD-L1 CPS =20 Population Milestone OS rate at 24 months was not evaluated. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. 24 months
Secondary Overall Response Rate (ORR) Per RECIST v1.1 in the PD-L1 CPS =1 Population ORR per RECIST v1.1 was defined as the proportion of the participants who have a complete response (CR) or partial response (PR) as the best overall response per RECIST v1.1 based upon investigator assessment. As a randomized double-blind study in which primary endpoints are OS and PFS, the confirmation of CR and PR was not required. Rate and associated 2-sided 95 percent Exact (Clopper-Pearson) Confidence Intervals are provided for each treatment arm which are unadjusted. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Up to approximately 16 months
Secondary ORR Per RECIST v1.1 in the PD-L1 CPS =20 Population ORR per RECIST v1.1 was defined as the proportion of the participants who have a CR or PR as the best overall response per RECIST v1.1 based upon investigator assessment. As a randomized double-blind study in which primary endpoints are OS and PFS, the confirmation of CR and PR was not required. Rate and associated 2-sided 95 percent Exact (Clopper-Pearson) Confidence Intervals are provided for each treatment arm which are unadjusted. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Up to approximately 16 months
Secondary Disease Control Rate (DCR) Per RECIST v1.1 in the PD-L1 CPS =1 Population DCR per RECIST v1.1 based upon investigator assessment, was defined as the percentage of participants with a best overall response of CR or PR at any time plus stable disease (SD) meeting the minimum time of 15 weeks. A status of SD=15 weeks will be assigned if the follow-up disease assessment has met the SD criteria at least once after the date of randomization at a minimum of 14 weeks (98 days) considering a one-week visit window. Rate and associated 2-sided 95 percent Exact (Clopper-Pearson) Confidence Intervals are provided for each treatment arm which are unadjusted. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Up to approximately 16 months
Secondary DCR Per RECIST v1.1 in the PD-L1 CPS =20 Population DCR per RECIST v1.1 based upon investigator assessment, was defined as the percentage of participants with a best overall response of CR or PR at any time plus stable disease (SD) meeting the minimum time of 15 weeks. A status of SD=15 weeks will be assigned if the follow-up disease assessment has met the SD criteria at least once after the date of randomization at a minimum of 14 weeks (98 days) considering a one-week visit window. Rate and associated 2-sided 95 percent Exact (Clopper-Pearson) Confidence Intervals are provided for each treatment arm which are unadjusted. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Up to approximately 16 months
Secondary Duration of Response (DoR) Per RECIST v1.1 in the PD-L1 CPS =1 Population DoR per RECIST v1.1 is defined as the time from first documented evidence of CR or PR until first documented disease progression per RECIST v1.1 based upon investigator assessment or death due to any cause, whichever occurs first, among participants who demonstrated CR or PR as the best overall response per RECIST v1.1. Kaplan-Meier estimate for the median DoR is presented, along with associated 95% confidence interval, estimated using the Brookmeyer-Crowley method. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Up to approximately 16 months
Secondary DoR Per RECIST v1.1 in the PD-L1 CPS =20 Population DoR per RECIST v1.1 is defined as the time from first documented evidence of CR or PR until first documented disease progression per RECIST v1.1 based upon investigator assessment or death due to any cause, whichever occurs first, among participants who demonstrated CR or PR as the best overall response per RECIST v1.1. Kaplan-Meier estimate for the median DoR is presented, along with associated 95% confidence interval, estimated using the Brookmeyer-Crowley method. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Up to approximately 16 months
Secondary Number of Participants With Any Adverse Events (AEs) and Serious Adverse Events (SAEs) An AE was defined as any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. An SAE was defined as any untoward medical occurrence that, at any dose results in death, is life-threatening, requires in-patient hospitalization or prolongation of existing hospitalization, results in persistent disability/incapacity, is a congenital anomaly/birth defect, any other situation such as important medical events according to medical or scientific judgement. Up to approximately 16 months
Secondary Number of Participants With AEs by Severity An AE was defined as any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study intervention, whether or not considered related to the study intervention. Severity of each AE was reported during the study and was assigned a grade according to the National Cancer Institute- Common Toxicity Criteria for Adverse Events (NCI-CTCAE). AEs severity were graded on a 5-point scale as: 1 = mild; discomfort noticed, but no disruption to daily activity, 2 = moderate; discomfort sufficient to reduce or affect normal daily activity, 3 = severe; inability to work or perform normal daily activity, 4 = life-threatening consequences and 5 = death related to AE. Up to approximately 16 months
Secondary Number of Participants With SAEs by Severity An SAE was defined as any untoward medical occurrence that, at any dose results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent disability/incapacity, is a congenital anomaly/birth defect, any other situation such as important medical events according to medical or scientific judgement. Severity of each SAE was reported during the study and was assigned a grade according to the NCI-CTCAE. SAEs severity were graded on a 5-point scale as: 1 = mild; discomfort noticed, but no disruption to daily activity, 2 = moderate; discomfort sufficient to reduce or affect normal daily activity, 3 = severe; inability to work or perform normal daily activity, 4 = life-threatening consequences and 5 = death related to AE. Data of participants experiencing SAEs of Grade =3 have been presented. Up to approximately 16 months
Secondary Number of Participants With Adverse Events of Special Interest (AESI) AESI were defined as events of potential immunologic etiology, including immune-related AEs (irAEs). Such events recently reported after treatment with other immune modulatory therapy include colitis, uveitis, hepatitis, pneumonitis, diarrhea, endocrine disorders, and specific cutaneous toxicities, as well as other events that may be immune mediated. Up to approximately 16 months
Secondary Number of Participants With AESI by Severity AESI were defined as events of potential immunologic etiology, including immune-related AEs (irAEs). Such events recently reported after treatment with other immune modulatory therapy include colitis, uveitis, hepatitis, pneumonitis, diarrhea, endocrine disorders, and specific cutaneous toxicities, as well as other events that may be immune mediated. Severity of each AESI was reported during the study and was assigned a grade according to the NCI-CTCAE. AESIs severity were graded on a 5-point scale as: 1 = mild; discomfort noticed, but no disruption to daily activity, 2 = moderate; discomfort sufficient to reduce or affect normal daily activity, 3 = severe; inability to work or perform normal daily activity, 4 = life-threatening consequences and 5 = death related to AE. Data of participants experiencing AESIs of Grade =3 have been presented. Up to approximately 16 months
Secondary Number of Participants With Dose Modifications Number of participants with dose modifications (including dose interruptions, dose delays and treatment discontinuations) were reported by each interventional component. Up to approximately 16 months
Secondary Time to Deterioration (TTD) in Pain in the PD-L1 CPS =1 Population TTD in pain is defined as the time from randomization to the first definitive meaningful deterioration from baseline in the European Organization for Research and Treatment of Cancer Item Library(EORTC IL51) pain domain, i.e. an increase from baseline of at least 8.33 observed at all subsequent non-missing visits. The EORTC Quality of Life Questionnaire 35-Item Head and Neck Module (QLQ-H&N35) is a head and neck specific module with multi-item scales. The questionnaire scores for each scale and single-item measure are averaged and transformed linearly to present a score ranging from 0-100. A high score represents a high/healthy level of functioning. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Up to approximately 16 months
Secondary TTD in Pain in the PD-L1 CPS =20 Population TTD in pain is defined as the time from randomization to the first definitive meaningful deterioration from baseline in the EORTC IL51 pain domain, i.e. an increase from baseline of at least 8.33 observed at all subsequent non-missing visits. The EORTC QLQ-H&N35 is a head and neck specific module with multi-item scales. The mouth pain, swallowing, speech problems, opening mouth, coughing, feeding tube, and trouble with social eating domains were administered and referred to as the EORTC IL51. The questionnaire scores for each scale and single-item measure are averaged and transformed linearly to present a score ranging from 0-100. A high score represents a high/healthy level of functioning. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Up to approximately 16 months
Secondary TTD in Physical Function in the PD-L1 CPS =1 Population TTD in physical function is defined as the time from randomization to the first definitive meaningful deterioration from baseline in the PF T-score, i.e. a decrease from baseline of at least 2.4 observed at all subsequent non-missing visits, as measured by the PROMIS PF 8c. The PROMIS PF 8c is an 8-item fixed length short form derived from the PROMIS Physical Function item bank. It includes a 5-point scale with three sets of response options. Scores on the PROMIS PF 8c are reported on a T score metric (mean = 50 and SD = 10), with higher scores reflecting better physical functioning. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Up to approximately 16 months
Secondary TTD in Physical Function in the PD-L1 CPS =20 Population TTD in physical function is defined as the time from randomization to the first definitive meaningful deterioration from baseline in the PF T-score, i.e. a decrease from baseline of at least 2.4 observed at all subsequent non-missing visits, as measured by the PROMIS PF 8c. The PROMIS PF 8c is an 8-item fixed length short form derived from the PROMIS Physical Function item bank. It includes a 5-point scale with three sets of response options. Scores on the PROMIS PF 8c are reported on a T score metric (mean = 50 and SD = 10), with higher scores reflecting better physical functioning. Data are presented for the PD-L1 CPS >=1 participants from mITT population. CPS was defined as the ratio of the combined number of PD-L1 expressing tumor cells and immune cells (lymphocytes and macrophages) to the total number of viable tumor cells. Up to approximately 16 months
See also
  Status Clinical Trial Phase
Terminated NCT01370876 - Efficacy and Safety Study of Oxaliplatin/5-FU in Patients With Recurrent or Metastatic Head and Neck Cancer Phase 2
Completed NCT03178110 - Manual Therapy and Use of the Dynasplint for Trismus in Patients With Head and Neck Cancer N/A
Completed NCT00798655 - Trial of Postoperative Radiation, Cisplatin, and Panitumumab in Locally Advanced Head and Neck Cancer Phase 2
Completed NCT00387127 - Lapatinib Versus Placebo Given Concurrently With Cisplatin And Radiotherapy In Patients With Unresected Head And Neck Cancer Phase 2
Recruiting NCT06062420 - Phase 2 Platform Study of Novel Immunotherapy Combinations as First-Line Treatment in Participants With PD-L1 Positive Recurrent/Metastatic Squamous Cell Carcinoma of the Head and Neck Phase 2
Not yet recruiting NCT06256588 - A Study of Dostarlimab vs Placebo After Chemoradiation in Adult Participants With Locally Advanced Unresected Head and Neck Squamous Cell Carcinoma Phase 3
Active, not recruiting NCT04260126 - Study of PDS0101 and Pembrolizumab Combination I/O in Subjects With HPV16 + Recurrent and/or Metastatic HNSCC Phase 2
Active, not recruiting NCT02296684 - Immunotherapy With MK-3475 in Surgically Resectable Head and Neck Squamous Cell Carcinoma Phase 2
Completed NCT01110980 - Normalcy of Food Intake in Head and Neck Cancer Patients N/A
Completed NCT01116336 - Phase I Chemoprevention Trial With Green Tea Polyphenon E & Erlotinib in Patients With Premalignant Lesions of the Head & Neck Phase 1
Recruiting NCT02557048 - Head and Neck Cancer in Children: A Retrospective Study
Completed NCT03356093 - Change in Symptom Clusters in HNC Patients N/A
Terminated NCT02376699 - Safety Study of SEA-CD40 in Cancer Patients Phase 1
Completed NCT00424255 - Study Of Adjuvant Lapatinib In High-Risk Head And Neck Cancer Subjects After Surgery Phase 3
Completed NCT00725764 - Phase 2 Study of GSK1363089 (Formerly XL880) in Adults With Squamous Cell Cancer of the Head and Neck Phase 2
Terminated NCT04428333 - Study of GSK3359609 With Pembrolizumab and 5-fluorouracil (5-FU)-Platinum Chemotherapy in Participants With Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma Phase 2/Phase 3
Active, not recruiting NCT02573493 - Nab-Paclitaxel and Cisplatin or Nab-paclitaxel as Induction Therapy for Locally Advanced Squamous Cell Carcinoma of the Head and Neck (HNSCC) Phase 2