Clinical Trials Logo

Clinical Trial Details — Status: Terminated

Administrative data

NCT number NCT04428333
Other study ID # 209227
Secondary ID 2019-003981-42
Status Terminated
Phase Phase 2/Phase 3
First received
Last updated
Start date August 13, 2020
Est. completion date September 19, 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 this study is to evaluate if the addition of GSK3359609 to pembrolizumab in combination with 5FU-platinum based chemotherapy improves the efficacy of the pembrolizumab combination with 5FU-platinum based chemotherapy in participants with recurrent or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC). This randomized, double-blinded, Phase II/III study will compare the combination of GSK3359609 with pembrolizumab and 5FU-platinum chemotherapy to placebo in combination with pembrolizumab and 5FU-platinum chemotherapy in participants with recurrent or metastatic HNSCC of the oral cavity, oropharynx, hypopharynx or larynx.


Recruitment information / eligibility

Status Terminated
Enrollment 118
Est. completion date September 19, 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 - HNSCC that was diagnosed as recurrent or metastatic and 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 (with the exception of systemic therapy completed >6 months prior if given as part of multimodal treatment for locally advanced disease and no disease progression/recurrence within 6 months of the completion of curatively intended systemic treatment). - Measurable disease per RECIST version 1.1 guidelines - Eastern Cooperative Oncology Group (ECOG) Performance Status (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 be either not a woman of childbearing potential (WOCBP); or be a WOCBP who agrees to use a highly effective 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 periods. - 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 IHC CPS 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, anti-Inducible T Cell Co-Stimulatory Receptor (ICOS) directed agent. - Systemic approved or investigational anticancer therapy within 30 days or 5 half lives of the drug, whichever is shorter. At least 14 days must have elapsed between the last dose of prior anticancer agent and the date of randomization. - Has high risk of bleeding (examples include but not limited to tumors encasing or infiltrating a major vessel [i.e. carotid, jugular, bronchial artery] and/or exhibits other high-risk features such as an arteriovenous fistula) - Active tumor bleeding - Grade 3 or Grade 4 hypercalcemia. - Major surgery less than or equal to (<=) 28 days prior to randomization. - Participants must have also fully recovered from any surgery (major or minor) and/or its complications before randomization - Toxicity from previous anticancer treatment that includes: a. Grade 3/Grade 4 toxicity considered related to prior immunotherapy and that led to treatment discontinuation and b. 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 with the exception of: a. any other invasive malignancy for which the participant was definitively treated, has been disease-free for <=3 years. b. curatively treated non-melanoma skin cancer or successfully treated in situ carcinoma and/or. c. low-risk early stage prostate cancer defined as: Stage T1c or T2a with a Gleason score <=6 and prostatic-specific antigen less than (<)10 nanograms per milliliter (ng/mL) either treated with definitive intent or untreated in active surveillance that has been stable for the past year prior to randomization. - Autoimmune disease or syndrome that required systemic treatment within the past 2 years. - Has a diagnosis of immunodeficiency or is receiving systemic steroids (>10 milligram [mg] oral prednisone 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 which include: a. Serious, uncontrolled cardiac arrhythmia or clinically significant electrocardiogram abnormalities including second degree (Type II) or third-degree atrioventricular block. b. Cardiomyopathy, myocardial infarction, acute coronary syndromes(including unstable angina pectoris), coronary angioplasty, stenting or bypass grafting. c. Congestive heart failure (Class II, III, or IV) as defined by the New York Heart Association functional classification system. d. Symptomatic pericarditis. - Current unstable liver or biliary disease per investigator assessment defined by the presence of ascites, encephalopathy, coagulopathy, hypoalbuminemia, esophageal or gastric varices, persistent jaundice, or cirrhosis. - Active infection requiring systemic therapy. - Known human immunodeficiency virus (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 to the chemotherapies under investigation including any ingredient used in the formulation. - Known history of active tuberculosis. - Any serious (>=Grade 3) and/or unstable pre-existing medical condition (aside from malignancy). - Any 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 or has participated in a study of an investigational agent or has used an investigational device within 4 weeks prior to the date of randomization.

Study Design


Related Conditions & MeSH terms


Intervention

Drug:
Feladilimab
Humanized anti-inducible T cell co-stimulatory receptor (ICOS) immunoglobulin G4 (IgG4) monoclonal antibody (mAb)
Pembrolizumab
Humanized anti- programmed cell death receptor1 (anti-PD-1) IgG4 mAb
Placebo
Sterile normal saline
Platinum based chemotherapy
Cisplatin/carboplatin
Fluorouracil (5FU)
5-fluorouracil

Locations

Country Name City State
Argentina GSK Investigational Site Ciudad Autonoma de Buenos Aires
Argentina GSK Investigational Site Ciudad Autónoma de Buenos Aires
Argentina GSK Investigational Site Oro Verde Entre Ríos
Argentina GSK Investigational Site Rosario Santa Fe
Argentina GSK Investigational Site San Miguel de Tucumán Tucumán
Australia GSK Investigational Site Blacktown New South Wales
Australia GSK Investigational Site Heidelberg Victoria
Australia GSK Investigational Site Melbourne Victoria
Australia GSK Investigational Site Woolloongabba Queensland
Belgium GSK Investigational Site Bruxelles
Belgium GSK Investigational Site Edegem
Brazil GSK Investigational Site Curitiba Paraná
Brazil GSK Investigational Site São Paulo
Brazil GSK Investigational Site Vitória Espírito Santo
Canada GSK Investigational Site Edmonton Alberta
Canada GSK Investigational Site Hamilton Ontario
Canada GSK Investigational Site London Ontario
Canada GSK Investigational Site Montreal Quebec
Canada GSK Investigational Site Toronto Ontario
Canada GSK Investigational Site Toronto Ontario
Denmark GSK Investigational Site Koebenhavn Oe
France GSK Investigational Site Bordeaux
France GSK Investigational Site Lyon cedex 08
France GSK Investigational Site Paris
France GSK Investigational Site Poitiers cedex
France GSK Investigational Site Strasbourg
Germany GSK Investigational Site Berlin
Germany GSK Investigational Site Hannover Niedersachsen
Germany GSK Investigational Site Leipzig Sachsen
Germany GSK Investigational Site Ulm Baden-Wuerttemberg
Hungary GSK Investigational Site Nyíregyháza
Ireland GSK Investigational Site Dublin
Italy GSK Investigational Site Brescia Lombardia
Italy GSK Investigational Site Milano Lombardia
Italy GSK Investigational Site Milano Lombardia
Italy GSK Investigational Site Napoli Campania
Italy GSK Investigational Site Prato Toscana
Italy GSK Investigational Site Roma Lazio
Italy GSK Investigational Site Savona Liguria
Japan GSK Investigational Site Osaka
Japan GSK Investigational Site Osaka
Japan GSK Investigational Site Tokyo
Japan GSK Investigational Site Tokyo
Korea, Republic of GSK Investigational Site Daegu-si
Korea, Republic of GSK Investigational Site Gyeonggi-do
Korea, Republic of GSK Investigational Site Hwasun-gun, Jeollanam-do
Korea, Republic of GSK Investigational Site Seoul
Korea, Republic of GSK Investigational Site Seoul
Poland GSK Investigational Site Bydgoszcz
Poland GSK Investigational Site Gdynia
Poland GSK Investigational Site Gliwice
Poland GSK Investigational Site Lublin
Poland GSK Investigational Site Tomaszow Mazowiecki
Poland GSK Investigational Site Warszawa
Romania GSK Investigational Site Bucuresti
Romania GSK Investigational Site Cluj Napoca
Romania GSK Investigational Site Craiova
Romania GSK Investigational Site Floresti
Romania GSK Investigational Site Otopeni
Romania GSK Investigational Site Suceava
Russian Federation GSK Investigational Site Pushkin
Spain GSK Investigational Site Barcelona
Spain GSK Investigational Site Madrid
Spain GSK Investigational Site Madrid
Spain GSK Investigational Site Madrid
Spain GSK Investigational Site Madrid
Spain GSK Investigational Site Valencia
Sweden GSK Investigational Site Stockholm
United Kingdom GSK Investigational Site London
United Kingdom GSK Investigational Site Nottingham
United Kingdom GSK Investigational Site Sutton
United States GSK Investigational Site Duarte California

Sponsors (2)

Lead Sponsor Collaborator
GlaxoSmithKline Merck Sharp & Dohme LLC

Countries where clinical trial is conducted

United States,  Argentina,  Australia,  Belgium,  Brazil,  Canada,  Denmark,  France,  Germany,  Hungary,  Ireland,  Italy,  Japan,  Korea, Republic of,  Poland,  Romania,  Russian Federation,  Spain,  Sweden,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary Overall Survival (OS) in mITT Population OS was defined as the time from the date of randomization to the date of death due to any cause. Kaplan-Meier estimate for the median OS is presented, along with associated 95% confidence interval, estimated using the Brookmeyer-Crowley method. Up to approximately 7 months
Primary OS in Programmed Death Receptor-ligand 1 (PD-L1) Combined Positive Score (CPS) =1 Population OS was defined as the time from the date of randomization until 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 7 months
Primary Progression-free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors (RECIST) Version (v)1.1 in mITT Population PFS per Response Evaluation Criteria in Solid Tumors (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. Kaplan-Meier estimate for the median PFS is presented, along with associated 95% confidence interval, estimated using the Brookmeyer-Crowley method. Up to approximately 7 months
Secondary PFS Per RECIST v1.1 in the PD-L1 CPS =1 Population PFS per RECIST v1.1 was defined as the time from the date of randomization until the date of 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 7 months
Secondary Milestone OS Rate at 12, 24 and 36 Months in mITT Population Milestone OS rate at 12, 24, and 36 months was not evaluated. Months 12, 24 and 36
Secondary Milestone OS Rate at 12, 24 and 36 Months in PD-L1 CPS =1 Population Milestone OS rate at 12, 24, and 36 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. Months 12, 24 and 36
Secondary Overall Response Rate (ORR) Per RECIST v1.1 in mITT 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. Up to approximately 7 months
Secondary ORR Per RECIST v1.1 in 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 7 months
Secondary Disease Control Rate (DCR) Per RECIST v1.1 in mITT 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. Up to approximately 7 months
Secondary DCR Per RECIST v1.1 in 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 7 months
Secondary Duration of Response (DoR) Per RECIST v1.1 in mITT 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. Up to approximately 7 months
Secondary DoR Per RECIST v1.1 in 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 7 months
Secondary Number of Participants With Adverse Events (AEs) in Safety Population An AE is 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 Up to approximately 7 months
Secondary Number of Participants With Serious Adverse Events (SAEs) in Safety Population SAE was defined as any untoward medical occurrence that, at any dose, results in death, was life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, was a congenital anomaly/birth defect and other situations according to medical or scientific judgement. Up to approximately 7 months
Secondary Number of Participants With Adverse Events of Special Interest (AESI) in Safety Population 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 7 months
Secondary Number of Participants With AEs in PD-L1 CPS =1 Population Any untoward or unfavorable medical occurrence in a participant, including any abnormal sign (for example, abnormal physical exam or laboratory finding), symptom, or disease, temporally associated with the participant's participation in the research, whether or not considered related to the participant's participation in the research. 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 7 months
Secondary Number of Participants With SAEs in PD-L1 CPS =1 Population SAE was defined as any untoward medical occurrence that, at any dose, results in death, was life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, was a congenital anomaly/birth defect and other situations according to medical or scientific judgement. 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 7 months
Secondary Number of Participants With AESIs in PD-L1 CPS =1 Population 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. 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 7 months
Secondary Severity of AEs in Safety Population 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 for each AE was reported during the study and assigned a grade according to the NCI-CTCAE v5.0. from Grade 1 through Grade 5. Grade 1= mild toxicity; Grade 2 = moderate toxicity; Grade 3 = severe toxicity; Grade 4 = life-threatening or disabling toxicity, Grade 5 = death related to toxicity. Up to approximately 7 months
Secondary Severity of SAEs in Safety Population A 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 for each SAE was reported during the study and assigned a grade according to the NCI-CTCAE v5.0. from Grade 1 through Grade 5. Grade 1= mild toxicity; Grade 2 = moderate toxicity; Grade 3 = severe toxicity; Grade 4 = life-threatening or disabling toxicity, Grade 5 = death related to toxicity. Data of participants experiencing SAEs of Grade >= 3 have been presented. Up to approximately 7 months
Secondary Severity of AESIs in Safety Population 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 7 months
Secondary Severity of AEs in PD-L1 CPS =1 Population 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 for each AE was reported during the study and assigned a grade according to the NCI-CTCAE v5.0. from Grade 1 through Grade 5. Grade 1= mild toxicity; Grade 2 = moderate toxicity; Grade 3 = severe toxicity; Grade 4 = life-threatening or disabling toxicity, Grade 5 = death related to toxicity. Data of participants experiencing AEs of Grade >= 3 have been presented. 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 7 months
Secondary Severity of SAEs in PD-L1 CPS =1 Population A 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 for each SAE was reported during the study and assigned a grade according to the NCI-CTCAE v5.0. from Grade 1 through Grade 5. Grade 1= mild toxicity; Grade 2 = moderate toxicity; Grade 3 = severe toxicity; Grade 4 = life-threatening or disabling toxicity, Grade 5 = death related to toxicity. Data of participants experiencing SAEs of Grade >= 3 have been presented. 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 7 months
Secondary Severity of AESI in PD-L1 CPS =1 Population 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 7 months
Secondary Number of Participants With Dose Modifications in Safety Population Number of participants with dose modifications (including dose interruptions, dose delays, dose reductions and treatment discontinuations) were reported by each interventional component. Up to approximately 7 months
Secondary Number of Participants With Dose Modifications in PD-L1 CPS =1 Population Number of participants with dose modifications (including dose interruptions, dose delays, dose reductions and treatment discontinuations) were reported by each interventional component. 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 7 months
Secondary Time to Deterioration (TTD) in Pain in mITT 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) Questionnaire 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 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. Kaplan-Meier estimate for the median TTD is presented, along with associated 95% confidence interval, estimated using the Brookmeyer-Crowley method. Up to approximately 7 months
Secondary TTD in Pain in 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 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. Kaplan-Meier estimate for the median TTD 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 to the total number of viable tumor cells. Up to approximately 7 months
Secondary TTD in Physical Function in mITT Population TTD in physical function (PF) 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 Patient-Reported Outcomes Measurement Information System - Physical Function (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. Up to approximately 7 months
Secondary TTD in Physical Function in PD-L1 CPS =1 Population TTD in PF 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 7 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
Terminated NCT04128696 - Study of GSK3359609 and Pembrolizumab in Programmed Death Receptor 1-ligand 1 (PD-L1) Positive Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma Phase 2/Phase 3
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 NCT01116336 - Phase I Chemoprevention Trial With Green Tea Polyphenon E & Erlotinib in Patients With Premalignant Lesions of the Head & Neck Phase 1
Completed NCT01110980 - Normalcy of Food Intake in Head and Neck Cancer Patients N/A
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
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