Denosumab in Combination With Chemotherapy as First-line Treatment of Metastatic Non-small Cell Lung Cancer

Sponsor
Amgen (Industry)
Overall Status
Completed
CT.gov ID
NCT01951586
Collaborator
(none)
226
74
2
46.9
3.1
0.1

Study Details

Study Description

Brief Summary

This randomized phase 2 trial is studying the effect of adding denosumab to standard chemotherapy in the treatment of advanced lung cancer.

Condition or Disease Intervention/Treatment Phase
  • Drug: Denosumab
  • Drug: Zoledronic acid
  • Drug: Placebo to Denosumab
  • Drug: Standard Chemotherapy
  • Drug: Placebo to Zoledronic Acid
Phase 2

Detailed Description

This is a global randomized double-blind placebo-controlled study in patients with Stage IV untreated non-small cell lung cancer (NSCLC) with or without bone metastasis. Eligible participants are to receive 4 to 6 cycles of a standard of care platinum-doublet chemotherapy regimen. Participants will be randomized in a 2:1 ratio to receive denosumab or matching placebo with the first investigational product dose coinciding with participant's first cycle of chemotherapy and continuing until the primary analysis, unacceptable toxicity, withdrawal of consent, death, or lost to follow-up. Participants who discontinued the investigational product early (ie, before primary analysis) were followed for disease status and survival. The primary analysis took place when 149 events of death had been reported. All participants were followed for 2 years after the last dose of blinded investigational product.

Study Design

Study Type:
Interventional
Actual Enrollment :
226 participants
Allocation:
Randomized
Intervention Model:
Parallel Assignment
Masking:
Double (Participant, Investigator)
Primary Purpose:
Treatment
Official Title:
A Randomized, Double-blind, Multi-center Phase 2 Trial of Denosumab in Combination With Chemotherapy as First-line Treatment of Metastatic Non-small Cell Lung Cancer
Actual Study Start Date :
Dec 31, 2013
Actual Primary Completion Date :
Jul 29, 2016
Actual Study Completion Date :
Nov 28, 2017

Arms and Interventions

Arm Intervention/Treatment
Placebo Comparator: Placebo

Participants received placebo matching to denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W.

Drug: Zoledronic acid
Administered by intravenous infusion in participants with bone metastasis upon investigative site request for IV bone-targeted therapy.
Other Names:
  • Zometa
  • Drug: Placebo to Denosumab
    Administered by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8; could be administered as often as every 3 weeks (Q3W) to participants receiving Q3W chemotherapy.

    Drug: Standard Chemotherapy
    Standard of care chemotherapy consisting of pemetrexed or gemcitabine in combination with cisplatin or carboplatin administered according to local practice.

    Experimental: Denosumab

    Participants received 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.

    Drug: Denosumab
    Administered by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8; could be administered as often as every 3 weeks (Q3W) to participants receiving Q3W chemotherapy.
    Other Names:
  • XGEVA
  • Drug: Standard Chemotherapy
    Standard of care chemotherapy consisting of pemetrexed or gemcitabine in combination with cisplatin or carboplatin administered according to local practice.

    Drug: Placebo to Zoledronic Acid
    Administered by intravenous infusion in participants with bone metastasis upon investigative site request for IV bone-targeted therapy.

    Outcome Measures

    Primary Outcome Measures

    1. Overall Survival (OS) [From randomization until the end of study; median time on study was 9.64 months.]

      Overall survival was calculated as the time from the date of randomization to the date of death from any cause. Participants last known to be alive were censored at the last contact date.

    Secondary Outcome Measures

    1. Correlation of Tumor Tissue RANK Expression With Overall Survival [From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.]

      To assess whether the treatment effect on overall survival was correlated with receptor activator of nuclear factor (NF)-κB (RANK) protein expression in tumor cells, RANK expression in archival tumor samples was measured using immunohistochemistry. The intensity of stain in the cytoplasm, membrane, and total was categorized as 0 (negative), 1+ (weak), 2+ (moderate) or 3+ (strong); All intensity is the sum of levels +1, +2 and +3. In addition, an H-score was calculated using the following formula: H-score=(percentage of cells of weak×1)+(percentage of cells of moderate×2)+(percentage of cells of strong×3). The maximum H-score was 300, corresponding to 100% of cells with strong intensity. The correlation between RANK expression level and OS was evaluated using a Cox proportional hazard models that included RANK expression level stratified by the randomization stratification factors in the corresponding treatment group.

    2. Correlation of Tumor Tissue RANK Ligand Expression With Overall Survival [From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.]

      To assess whether the treatment effect on overall survival was correlated with RANK ligand (RANKL) protein expression in tumor cells, RANKL expression in archival tumor samples was measured using immunohistochemistry. The intensity of stain in the cytoplasm was categorized as 0 (negative), 1+ (weak), 2+ (moderate) or 3+ (strong); All intensity is the sum of levels +1, +2 and +3. In addition, an H-score was calculated using the following formula: H-score=(percentage of cells of weak×1)+(percentage of cells of moderate×2)+(percentage of cells of strong×3). The maximum H-score was 300, corresponding to 100% of cells with strong intensity. The correlation between RANKL expression level and OS was evaluated using a Cox proportional hazard models that included RANKL expression level stratified by the randomization stratification factors in the corresponding treatment group.

    3. Objective Response Rate [From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.]

      Objective response rate was defined as the percentage of participants with a complete response (CR) or partial response (PR) based on modified RECIST 1.1 achieved over the study duration. CR: Disappearance of all target and non target lesions, normalization of tumor marker levels and no new lesions. PR: At least a 30% decrease in the size of target lesions with no progression of non-target lesions and no new lesions, or, disappearance of target lesions with persistence of one or more non-target lesions and/or maintenance of tumor marker levels above normal limits and no new lesions. Participants who underwent surgical resection while on study were not evaluated for response after the surgery. Participants who did not meet the criteria for an objective response by the analysis cutoff date were considered non-responders.

    4. Correlation of Tumor Tissue RANK Expression With Objective Response Rate [From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.]

      To assess whether the treatment effect on objective response rate (ORR) based on RECIST 1.1 was correlated with RANK protein expression in tumor cells, RANK expression in archival tumor samples was measured using immunohistochemistry. The intensity of stain in the cytoplasm, membrane, and total was categorized as 0 (negative), 1+ (weak), 2+ (moderate) or 3+ (strong); All intensity is the sum of levels +1, +2 and +3. In addition, an H-score was calculated using the following formula: H-score=(percentage of cells of weak×1)+(percentage of cells of moderate×2)+(percentage of cells of strong×3). The maximum H-score was 300, corresponding to 100% of cells with strong intensity. The correlation between RANK expression level and ORR was evaluated within each treatment group using a logistical regression model that included RANK expression value as an independent variable and stratified by the randomization stratification factors. The odds ratio and 95% confidence interval are reported.

    5. Correlation of Tumor Tissue RANKL Expression With Objective Response Rate [From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.]

      To assess whether the treatment effect on objective response rate (ORR) based on RECIST 1.1 was correlated with RANKL protein expression in tumor cells, RANKL expression in archival tumor samples was measured using immunohistochemistry. The intensity of stain in the cytoplasm was categorized as 0 (negative), 1+ (weak), 2+ (moderate) or 3+ (strong); All intensity is the sum of levels +1, +2 and +3. In addition, an H-score was calculated using the following formula: H-score=(percentage of cells of weak×1)+(percentage of cells of moderate×2)+(percentage of cells of strong×3). The maximum H-score was 300, corresponding to 100% of cells with strong intensity. The correlation between RANKL expression level and ORR was evaluated within each treatment group using a logistical regression model that included RANKL expression value as an independent variable and stratified by the randomization stratification factors. The odds ratio and 95% confidence interval are reported.

    6. Clinical Benefit Rate [From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.]

      Clinical benefit rate was defined as the percentage of participants with an objective response (CR or PR) or stable disease (SD) or better for at least 16 weeks achieved over the study duration. If a participant underwent surgical resection while on study, the participant was not evaluated for response after the surgery. Participants who did not meet the criteria for clinical benefit by the analysis cutoff date were considered as non-responders.

    7. Progression-free Survival (PFS) [From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.]

      Progression-free survival was defined as the time from randomization to the first observed disease progression per modified RECIST 1.1 criteria or death from any cause. Participants last known to be alive who did not experience disease progression were censored at their last imaging assessment date, last contact date if they were in the survival follow up phase, end of the study date, or the primary analysis cut-off date, whichever was first. If a participant underwent surgical resection while on study, the participant was censored at the last evaluable imaging assessment prior to the surgery.

    8. Serum Denosumab Trough Levels in Participants Who Received Q3W Dosing [Prior to dosing at day 8 and weeks 3, 6, 9, 12, 15, 18, 21 and 24.]

      Serum samples were analyzed for denosumab using enzyme-linked immunosorbent assay (ELISA) following a validated procedure. The lower limit of quantification for the assay was 20 ng/mL.

    9. Serum Denosumab Trough Levels in Participants Who Received Q4W Dosing [Prior to dosing at day 8 and weeks 4, 8, 12, 16, 20 and 24]

      Serum samples were analyzed for denosumab using enzyme-linked immunosorbent assay (ELISA) following a validated procedure. The lower limit of quantification for the assay was 20 ng/mL.

    10. Number of Participants With Treatment-emergent Adverse Events [From first dose of study drug to the end of study date; the median (min, max) duration was 10.0 (0.2, 41.4) and 9.4 (0.2, 42.9) months for Placebo and Denosumab respectively.]

      An adverse event (AE) is defined as any untoward medical occurrence in a clinical trial participant. The event does not necessarily have a causal relationship with study treatment. A serious adverse event is defined as an AE that meets at least 1 of the following criteria fatal life threatening requires in-patient hospitalization or prolongation of existing hospitalization results in persistent or significant disability/incapacity congenital anomaly/birth defect other medically important serious event Treatment-related AEs include only TEAEs for which the investigator indicated there was a reasonable possibility they may have been caused by study drug. Fatal adverse events include only deaths reported on the Adverse Event Case Report Form.

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    18 Years to 100 Years
    Sexes Eligible for Study:
    All
    Accepts Healthy Volunteers:
    No
    Inclusion Criteria:
    • Histologically or cytologically confirmed stage IV non-small cell lung carcinoma (NSCLC), according to 7th Tumor/Node/Metastasis (TNM) classification (cytological specimens obtained by bronchial washing or brushing, or fine-needle aspiration are acceptable)

    • Subject has available and has provided consent to release to the sponsor (or designee) a tumor block with confirmed tumor content (or approximately 20 unstained charged slides [a minimum of 7 slides is mandatory]) and the corresponding pathology report

    • Planned to receive 4 to 6 cycles of pemetrexed or gemcitabine in combination with cisplatin or carboplatin

    • For subjects to receive pemetrexed, planned to receive vitamin B12 and folate per pemetrexed approved labeling

    • Radiographically evaluable (measurable or non-measurable) disease (according to modified Response Evaluation Criteria In Solid Tumors (RECIST) 1.1 criteria

    • Other inclusion criteria may apply

    Exclusion Criteria:
    • Known presence of documented sensitizing epidermal growth factor receptor (EGFR) activating mutation or echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase (EML4-ALK) translocation (screening following local standards, but strongly encouraged in non-squamous histology)

    • Known brain metastases (systematic screening of patients not mandatory)

    • Any prior systemic therapy (before randomization) for the treatment of NSCLC (including chemoradiation), except if for non-metastatic disease and was completed at least 6 months prior to randomization

    • Planned to receive bevacizumab

    • Significant dental/oral disease, including prior history or current evidence of osteonecrosis/ osteomyelitis of the jaw, or with the following:

    • Active dental or jaw condition which requires oral surgery

    • Non-healed dental/oral surgery

    • Planned invasive dental procedures for the course of the study.

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 Research Site Goodyear Arizona United States 85338
    2 Research Site Anaheim California United States 92801
    3 Research Site Long Beach California United States 90813
    4 Research Site Los Angeles California United States 90024
    5 Research Site Los Angeles California United States 90048
    6 Research Site Farmington Connecticut United States 06030-1628
    7 Research Site Alexandria Louisiana United States 71301
    8 Research Site Brewer Maine United States 04412
    9 Research Site Westminster Maryland United States 21157
    10 Research Site Fairhaven Massachusetts United States 02719
    11 Research Site Detroit Michigan United States 48202
    12 Research Site East Setauket New York United States 11733
    13 Research Site Durham North Carolina United States 27710
    14 Research Site Hickory North Carolina United States 28602
    15 Research Site Winston-Salem North Carolina United States 27157
    16 Research Site Bismarck North Dakota United States 58501
    17 Research Site Cincinnati Ohio United States 45267
    18 Research Site Nashville Tennessee United States 37203
    19 Research Site Kogarah New South Wales Australia 2217
    20 Research Site Wahroonga New South Wales Australia 2076
    21 Research Site Adelaide South Australia Australia 5000
    22 Research Site Footscray Victoria Australia 3011
    23 Research Site Parkville Victoria Australia 3050
    24 Research Site Wodonga Victoria Australia 3690
    25 Research Site Edmonton Alberta Canada T6G 1Z2
    26 Research Site Saint John New Brunswick Canada E2L 4L2
    27 Research Site Kitchener Ontario Canada N2G 1G3
    28 Research Site Sudbury Ontario Canada P3E 5J1
    29 Research Site Toronto Ontario Canada M5G 2M9
    30 Research Site Toronto Ontario Canada M9N 1N8
    31 Research Site Montreal Quebec Canada H2W 1S6
    32 Research Site Chomutov Czechia 430 12
    33 Research Site Ostrava-Poruba Czechia 708 52
    34 Research Site Pardubice Czechia 532 03
    35 Research Site Praha 8 Czechia 180 81
    36 Research Site Usti nad Labem Czechia 401 13
    37 Research Site Caen Cedex 5 France 14076
    38 Research Site Dijon cedex France 21079
    39 Research Site Nantes Cedex 2 France 44202
    40 Research Site Paris Cedex 10 France 75475
    41 Research Site Paris Cedex 14 France 75674
    42 Research Site Paris Cedex 20 France 75020
    43 Research Site Pessac Cedex France 33604
    44 Research Site Reims Cedex France 51056
    45 Research Site Saint Quentin France 02321
    46 Research Site Tours Cedex 9 France 37044
    47 Research Site Berlin Germany 14165
    48 Research Site Grosshansdorf Germany 22927
    49 Research Site Köln-Merheim Germany 51109
    50 Research Site Ulm Germany 89081
    51 Research Site Athens Greece 11522
    52 Research Site Athens Greece 12462
    53 Research Site Heraklion Greece 71110
    54 Research Site Patra Greece 26504
    55 Research Site Thessaloniki Greece 54622
    56 Research Site Thessaloniki Greece 57010
    57 Research Site Monza (MB) Italy 20900
    58 Research Site Orbassano (TO) Italy 10043
    59 Research Site Pavia Italy 27100
    60 Research Site Roma Italy 00128
    61 Research Site Saronno VA Italy 21047
    62 Research Site 's Hertogenbosch Netherlands 5223 GZ
    63 Research Site Arnhem Netherlands 6815 AD
    64 Research Site Harderwijk Netherlands 3844 DG
    65 Research Site Tilburg Netherlands 5022 GC
    66 Research Site Zutphen Netherlands 7207 AE
    67 Research Site Bristol United Kingdom BS2 8ED
    68 Research Site Exeter United Kingdom EX2 5DW
    69 Research Site Glasgow United Kingdom G42 9LF
    70 Research Site Guildford United Kingdom GU2 7XX
    71 Research Site London United Kingdom SE1 9RT
    72 Research Site London United Kingdom W6 8RF
    73 Research Site Plymouth United Kingdom PL6 8DH
    74 Research Site Preston United Kingdom PR2 9HT

    Sponsors and Collaborators

    • Amgen

    Investigators

    • Study Director: MD, Amgen

    Study Documents (Full-Text)

    None provided.

    More Information

    Additional Information:

    Publications

    Responsible Party:
    Amgen
    ClinicalTrials.gov Identifier:
    NCT01951586
    Other Study ID Numbers:
    • 20120249
    • 2013-001662-42
    First Posted:
    Sep 26, 2013
    Last Update Posted:
    Oct 25, 2021
    Last Verified:
    Oct 1, 2021
    Studies a U.S. FDA-regulated Drug Product:
    Yes
    Studies a U.S. FDA-regulated Device Product:
    No
    Additional relevant MeSH terms:

    Study Results

    Participant Flow

    Recruitment Details This study was conducted at 57 centers in 10 countries including the United Kingdom, Netherlands, France, Italy, Germany, Czech Republic, United States, Canada, Australia and Greece. Participants were enrolled from 31 December 2013 to 21 May 2015.
    Pre-assignment Detail Participants were randomized in a 2:1 ratio to receive denosumab or placebo. Randomization was stratified based on the presence of bone metastasis (yes or no), histology (squamous versus non-squamous), and geographic region (North America, Western Europe/Australia, and rest of world [ROW]).
    Arm/Group Title Placebo Denosumab
    Arm/Group Description Participants were randomized to receive placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Period Title: Overall Study
    STARTED 78 148
    Received Study Drug 76 145
    COMPLETED 0 0
    NOT COMPLETED 78 148

    Baseline Characteristics

    Arm/Group Title Placebo Denosumab Total
    Arm/Group Description Participants were randomized to receive placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W. Total of all reporting groups
    Overall Participants 78 148 226
    Age (years) [Mean (Standard Deviation) ]
    Mean (Standard Deviation) [years]
    64.0
    (10.1)
    63.8
    (9.7)
    63.8
    (9.8)
    Sex: Female, Male (Count of Participants)
    Female
    20
    25.6%
    54
    36.5%
    74
    32.7%
    Male
    58
    74.4%
    94
    63.5%
    152
    67.3%
    Race/Ethnicity, Customized (Count of Participants)
    Black or African American
    2
    2.6%
    7
    4.7%
    9
    4%
    White or Caucasian
    73
    93.6%
    133
    89.9%
    206
    91.2%
    Others
    3
    3.8%
    8
    5.4%
    11
    4.9%
    Presence of Bone Metastasis (Count of Participants)
    Yes
    35
    44.9%
    65
    43.9%
    100
    44.2%
    No
    43
    55.1%
    83
    56.1%
    126
    55.8%
    Histology (Count of Participants)
    Squamous
    18
    23.1%
    35
    23.6%
    53
    23.5%
    Non-squamous
    60
    76.9%
    113
    76.4%
    173
    76.5%
    Geographic region (Count of Participants)
    North America/Australia
    30
    38.5%
    63
    42.6%
    93
    41.2%
    Western Europe
    35
    44.9%
    67
    45.3%
    102
    45.1%
    Rest of World
    13
    16.7%
    18
    12.2%
    31
    13.7%
    Eastern Cooperative Oncology Group (ECOG) Performance Status (Count of Participants)
    0 (Fully active)
    33
    42.3%
    69
    46.6%
    102
    45.1%
    1 (Restrictive but ambulatory)
    45
    57.7%
    79
    53.4%
    124
    54.9%
    Time from Initial Diagnosis of NSCLC to Randomization (months) [Mean (Standard Deviation) ]
    Mean (Standard Deviation) [months]
    2.46
    (5.28)
    6.32
    (16.96)
    4.99
    (14.17)

    Outcome Measures

    1. Primary Outcome
    Title Overall Survival (OS)
    Description Overall survival was calculated as the time from the date of randomization to the date of death from any cause. Participants last known to be alive were censored at the last contact date.
    Time Frame From randomization until the end of study; median time on study was 9.64 months.

    Outcome Measure Data

    Analysis Population Description
    All randomized participants
    Arm/Group Title Placebo Denosumab
    Arm/Group Description Participants were randomized to receive placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Measure Participants 78 148
    Median (95% Confidence Interval) [months]
    10.9
    10.7
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.5157
    Comments
    Method Log Rank
    Comments Log rank test stratified by the randomization stratification factors.
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 1.11
    Confidence Interval (2-Sided) 95%
    0.81 to 1.53
    Parameter Dispersion Type:
    Value:
    Estimation Comments Based on a Cox proportional hazards model stratified by the randomized stratification factors. Hazard ratio < 1 favors denosumab.
    2. Secondary Outcome
    Title Correlation of Tumor Tissue RANK Expression With Overall Survival
    Description To assess whether the treatment effect on overall survival was correlated with receptor activator of nuclear factor (NF)-κB (RANK) protein expression in tumor cells, RANK expression in archival tumor samples was measured using immunohistochemistry. The intensity of stain in the cytoplasm, membrane, and total was categorized as 0 (negative), 1+ (weak), 2+ (moderate) or 3+ (strong); All intensity is the sum of levels +1, +2 and +3. In addition, an H-score was calculated using the following formula: H-score=(percentage of cells of weak×1)+(percentage of cells of moderate×2)+(percentage of cells of strong×3). The maximum H-score was 300, corresponding to 100% of cells with strong intensity. The correlation between RANK expression level and OS was evaluated using a Cox proportional hazard models that included RANK expression level stratified by the randomization stratification factors in the corresponding treatment group.
    Time Frame From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.

    Outcome Measure Data

    Analysis Population Description
    All randomized participants who had evaluable pre-treatment tumor RANK expression.
    Arm/Group Title Placebo Denosumab
    Arm/Group Description Participants were randomized to receive placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Measure Participants 64 123
    Cytoplasm all intensity
    0.84
    1.00
    Membrane all intensity
    0.72
    0.92
    Total all intensity
    0.80
    1.00
    Cytoplasm H-score
    0.83
    1.00
    Membrane H-score
    0.72
    0.93
    Total H-score
    0.80
    1.00
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level measured in the cytoplasm (all intensity) and treatment effect was evaluated using a Cox proportional hazard model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.3759
    Comments
    Method Cox propotional hazard model
    Comments
    Statistical Analysis 2
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level measured in membranes (all intensity) and treatment effect was evaluated using a Cox proportional hazard model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.3666
    Comments
    Method Cox propotional hazard model
    Comments
    Statistical Analysis 3
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level total (cytoplasm + membranes; all intensity) and treatment effect was evaluated using a Cox proportional hazard model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.1917
    Comments
    Method Cox propotional hazard model
    Comments
    Statistical Analysis 4
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level measured in the cytoplasm (H-score) and treatment effect was evaluated using a Cox proportional hazard model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.3353
    Comments
    Method Cox propotional hazard model
    Comments
    Statistical Analysis 5
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level measured in membranes (H-score) and treatment effect was evaluated using a Cox proportional hazard model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.3179
    Comments
    Method Cox propotional hazard model
    Comments
    Statistical Analysis 6
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level total (cytoplasm + membranes; H-score) and treatment effect was evaluated using a Cox proportional hazard model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.1583
    Comments
    Method Cox propotional hazard model
    Comments
    3. Secondary Outcome
    Title Correlation of Tumor Tissue RANK Ligand Expression With Overall Survival
    Description To assess whether the treatment effect on overall survival was correlated with RANK ligand (RANKL) protein expression in tumor cells, RANKL expression in archival tumor samples was measured using immunohistochemistry. The intensity of stain in the cytoplasm was categorized as 0 (negative), 1+ (weak), 2+ (moderate) or 3+ (strong); All intensity is the sum of levels +1, +2 and +3. In addition, an H-score was calculated using the following formula: H-score=(percentage of cells of weak×1)+(percentage of cells of moderate×2)+(percentage of cells of strong×3). The maximum H-score was 300, corresponding to 100% of cells with strong intensity. The correlation between RANKL expression level and OS was evaluated using a Cox proportional hazard models that included RANKL expression level stratified by the randomization stratification factors in the corresponding treatment group.
    Time Frame From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.

    Outcome Measure Data

    Analysis Population Description
    All randomized participants who had evaluable pre-treatment tumor RANKL expression.
    Arm/Group Title Placebo Denosumab
    Arm/Group Description Participants were randomized to receive placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Measure Participants 65 121
    Cytoplasm all intensity
    0.77
    0.93
    Cytoplasm H-score
    0.79
    0.93
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANKL expression level measured in the cytoplasm (all intensity) and treatment effect was evaluated using a Cox proportional hazard model that included treatment group, RANKL expression level and treatment-by-RANKL expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.3946
    Comments
    Method Cox propotional hazard model
    Comments
    Statistical Analysis 2
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANKL expression level measured in the cytoplasm (H-score) and treatment effect was evaluated using a Cox proportional hazard model that included treatment group, RANKL expression level and treatment-by-RANKL expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.3735
    Comments
    Method Cox propotional hazard model
    Comments
    4. Secondary Outcome
    Title Objective Response Rate
    Description Objective response rate was defined as the percentage of participants with a complete response (CR) or partial response (PR) based on modified RECIST 1.1 achieved over the study duration. CR: Disappearance of all target and non target lesions, normalization of tumor marker levels and no new lesions. PR: At least a 30% decrease in the size of target lesions with no progression of non-target lesions and no new lesions, or, disappearance of target lesions with persistence of one or more non-target lesions and/or maintenance of tumor marker levels above normal limits and no new lesions. Participants who underwent surgical resection while on study were not evaluated for response after the surgery. Participants who did not meet the criteria for an objective response by the analysis cutoff date were considered non-responders.
    Time Frame From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with at least one baseline measurable lesion per modified RECIST 1.1.
    Arm/Group Title Placebo Denosumab
    Arm/Group Description Participants were randomized to receive placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Measure Participants 76 144
    Number [percentage of participants]
    43.4
    55.6%
    36.8
    24.9%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.3491
    Comments
    Method Regression, Logistic
    Comments Logistic regression model adjusted for the randomization stratification factors.
    Method of Estimation Estimation Parameter Odds Ratio (OR)
    Estimated Value 0.76
    Confidence Interval (2-Sided) 95%
    0.43 to 1.35
    Parameter Dispersion Type:
    Value:
    Estimation Comments Based on a logistic regression model adjusted for the randomization stratification factors; an odds ratio ≥ 1 favors denosumab.
    5. Secondary Outcome
    Title Correlation of Tumor Tissue RANK Expression With Objective Response Rate
    Description To assess whether the treatment effect on objective response rate (ORR) based on RECIST 1.1 was correlated with RANK protein expression in tumor cells, RANK expression in archival tumor samples was measured using immunohistochemistry. The intensity of stain in the cytoplasm, membrane, and total was categorized as 0 (negative), 1+ (weak), 2+ (moderate) or 3+ (strong); All intensity is the sum of levels +1, +2 and +3. In addition, an H-score was calculated using the following formula: H-score=(percentage of cells of weak×1)+(percentage of cells of moderate×2)+(percentage of cells of strong×3). The maximum H-score was 300, corresponding to 100% of cells with strong intensity. The correlation between RANK expression level and ORR was evaluated within each treatment group using a logistical regression model that included RANK expression value as an independent variable and stratified by the randomization stratification factors. The odds ratio and 95% confidence interval are reported.
    Time Frame From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.

    Outcome Measure Data

    Analysis Population Description
    All randomized participants with at least one baseline measurable lesion per modified RECIST 1.1 who had evaluable pre-treatment tumor RANK expression.
    Arm/Group Title Placebo Denosumab
    Arm/Group Description Participants were randomized to receive placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Measure Participants 63 119
    Cytoplasm all intensity
    1.00
    0.88
    Membrane all intensity
    1.18
    0.82
    Total all intensity
    1.16
    0.88
    Cytoplasm H-score
    1.00
    0.87
    Membrane H-score
    1.14
    0.82
    Total H-score
    1.14
    0.87
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level measured in the cytoplasm (all intensity) and treatment effect was evaluated using a logistic regression model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.4760
    Comments
    Method Regression, Logistic
    Comments
    Statistical Analysis 2
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level measured in the membranes (all intensity) and treatment effect was evaluated using a logistic regression model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.2582
    Comments
    Method Regression, Logistic
    Comments
    Statistical Analysis 3
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level total (cytoplasm + membrane; all intensity) and treatment effect was evaluated using a logistic regression model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.2230
    Comments
    Method Regression, Logistic
    Comments
    Statistical Analysis 4
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level measured in the cytoplasm (H-score) and treatment effect was evaluated using a logistic regression model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.4329
    Comments
    Method Regression, Logistic
    Comments
    Statistical Analysis 5
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level measured in the membranes (H-score) and treatment effect was evaluated using a logistic regression model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.2620
    Comments
    Method Regression, Logistic
    Comments
    Statistical Analysis 6
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANK expression level total (cytoplasm + membrane; H-score) and treatment effect was evaluated using a logistic regression model that included treatment group, RANK expression level and treatment-by-RANK expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.2081
    Comments
    Method Regression, Logistic
    Comments
    6. Secondary Outcome
    Title Correlation of Tumor Tissue RANKL Expression With Objective Response Rate
    Description To assess whether the treatment effect on objective response rate (ORR) based on RECIST 1.1 was correlated with RANKL protein expression in tumor cells, RANKL expression in archival tumor samples was measured using immunohistochemistry. The intensity of stain in the cytoplasm was categorized as 0 (negative), 1+ (weak), 2+ (moderate) or 3+ (strong); All intensity is the sum of levels +1, +2 and +3. In addition, an H-score was calculated using the following formula: H-score=(percentage of cells of weak×1)+(percentage of cells of moderate×2)+(percentage of cells of strong×3). The maximum H-score was 300, corresponding to 100% of cells with strong intensity. The correlation between RANKL expression level and ORR was evaluated within each treatment group using a logistical regression model that included RANKL expression value as an independent variable and stratified by the randomization stratification factors. The odds ratio and 95% confidence interval are reported.
    Time Frame From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.

    Outcome Measure Data

    Analysis Population Description
    All randomized participants with at least one baseline measurable lesion per modified RECIST 1.1 who had evaluable pre-treatment tumor RANKL expression.
    Arm/Group Title Placebo Denosumab
    Arm/Group Description Participants were randomized to receive placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Measure Participants 64 117
    Cytoplasm all intensity
    1.27
    1.10
    Cytoplasm H-score
    1.25
    1.09
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANKL expression level measured in the cytoplasm (all intensity) and treatment effect was evaluated using a logistic regression model that included treatment group, RANKL expression level and treatment-by-RANKL expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.4671
    Comments
    Method Regression, Logistic
    Comments
    Statistical Analysis 2
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments The interaction of RANKL expression level measured in the cytoplasm (H-score) and treatment effect was evaluated using a logistic regression model that included treatment group, RANKL expression level and treatment-by-RANKL expression level interaction stratified by the randomization stratification factors.
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.4236
    Comments
    Method Regression, Logistic
    Comments
    7. Secondary Outcome
    Title Clinical Benefit Rate
    Description Clinical benefit rate was defined as the percentage of participants with an objective response (CR or PR) or stable disease (SD) or better for at least 16 weeks achieved over the study duration. If a participant underwent surgical resection while on study, the participant was not evaluated for response after the surgery. Participants who did not meet the criteria for clinical benefit by the analysis cutoff date were considered as non-responders.
    Time Frame From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with at least one baseline measurable lesion per modified RECIST 1.1.
    Arm/Group Title Placebo Denosumab
    Arm/Group Description Participants were randomized to receive placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Measure Participants 76 144
    Number [percentage of participants]
    53.9
    69.1%
    47.9
    32.4%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.4654
    Comments
    Method Regression, Logistic
    Comments Logistic regression model adjusted for the randomization stratification factors.
    Method of Estimation Estimation Parameter Odds Ratio (OR)
    Estimated Value 0.81
    Confidence Interval (2-Sided) 95%
    0.46 to 1.43
    Parameter Dispersion Type:
    Value:
    Estimation Comments Based on a logistic regression model adjusted for the randomization stratification factors; an odds ratio ≥ 1 favors denosumab.
    8. Secondary Outcome
    Title Progression-free Survival (PFS)
    Description Progression-free survival was defined as the time from randomization to the first observed disease progression per modified RECIST 1.1 criteria or death from any cause. Participants last known to be alive who did not experience disease progression were censored at their last imaging assessment date, last contact date if they were in the survival follow up phase, end of the study date, or the primary analysis cut-off date, whichever was first. If a participant underwent surgical resection while on study, the participant was censored at the last evaluable imaging assessment prior to the surgery.
    Time Frame From randomization until the data cut-off date of 29 July 2016; median time on study was 9.64 months.

    Outcome Measure Data

    Analysis Population Description
    All randomized participants
    Arm/Group Title Placebo Denosumab
    Arm/Group Description Participants were randomized to receive placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Measure Participants 78 148
    Median (95% Confidence Interval) [months]
    5.7
    5.2
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo, Denosumab
    Comments
    Type of Statistical Test Superiority or Other (legacy)
    Comments
    Statistical Test of Hypothesis p-Value 0.7363
    Comments
    Method Log Rank
    Comments Log rank test stratified by the randomized stratification factors.
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 1.05
    Confidence Interval (2-Sided) 95%
    0.78 to 1.43
    Parameter Dispersion Type:
    Value:
    Estimation Comments Based on a Cox proportional hazards model stratified by the randomized stratification factors. Hazard ratio < 1 favors denosumab.
    9. Secondary Outcome
    Title Serum Denosumab Trough Levels in Participants Who Received Q3W Dosing
    Description Serum samples were analyzed for denosumab using enzyme-linked immunosorbent assay (ELISA) following a validated procedure. The lower limit of quantification for the assay was 20 ng/mL.
    Time Frame Prior to dosing at day 8 and weeks 3, 6, 9, 12, 15, 18, 21 and 24.

    Outcome Measure Data

    Analysis Population Description
    Randomized participants who received denosumab every 3 weeks with at least one valid denosumab concentration measurement.
    Arm/Group Title Denosumab
    Arm/Group Description Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Measure Participants 104
    Dose 2 - Day 8
    8590
    (4080)
    Dose 3 - Week 3
    12200
    (5710)
    Dose 4 - Week 6
    19700
    (7350)
    Dose 5 - Week 9
    19600
    (8270)
    Dose 6 - Week 12
    22800
    (10400)
    Dose 7 - Week 15
    24300
    (11500)
    Dose 8 - Week 18
    22700
    (10600)
    Dose 9 - Week 21
    22100
    (11200)
    Dose 10 - Week 24
    23300
    (12700)
    10. Secondary Outcome
    Title Serum Denosumab Trough Levels in Participants Who Received Q4W Dosing
    Description Serum samples were analyzed for denosumab using enzyme-linked immunosorbent assay (ELISA) following a validated procedure. The lower limit of quantification for the assay was 20 ng/mL.
    Time Frame Prior to dosing at day 8 and weeks 4, 8, 12, 16, 20 and 24

    Outcome Measure Data

    Analysis Population Description
    Randomized participants who received denosumab every 4 weeks with at least one valid denosumab concentration measurement.
    Arm/Group Title Denosumab
    Arm/Group Description Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Measure Participants 8
    Dose 2 - Day 8
    8990
    (2470)
    Dose 3 - Week 4
    10900
    (3170)
    Dose 4 - Week 8
    15700
    (6710)
    Dose 5 - Week 12
    14500
    (4940)
    Dose 6 - Week 16
    13000
    (4380)
    Dose 7 - Week 20
    15400
    (5620)
    Dose 8 - Week 24
    15900
    (7810)
    11. Secondary Outcome
    Title Number of Participants With Treatment-emergent Adverse Events
    Description An adverse event (AE) is defined as any untoward medical occurrence in a clinical trial participant. The event does not necessarily have a causal relationship with study treatment. A serious adverse event is defined as an AE that meets at least 1 of the following criteria fatal life threatening requires in-patient hospitalization or prolongation of existing hospitalization results in persistent or significant disability/incapacity congenital anomaly/birth defect other medically important serious event Treatment-related AEs include only TEAEs for which the investigator indicated there was a reasonable possibility they may have been caused by study drug. Fatal adverse events include only deaths reported on the Adverse Event Case Report Form.
    Time Frame From first dose of study drug to the end of study date; the median (min, max) duration was 10.0 (0.2, 41.4) and 9.4 (0.2, 42.9) months for Placebo and Denosumab respectively.

    Outcome Measure Data

    Analysis Population Description
    All randomized participants who received at least one dose of study drug (denosumab or placebo)
    Arm/Group Title Placebo Denosumab
    Arm/Group Description Participants were randomized to receive placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants were randomized to receive 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    Measure Participants 76 145
    All adverse events (AEs)
    76
    97.4%
    144
    97.3%
    Serious adverse events
    68
    87.2%
    129
    87.2%
    AEs leading to discontinuation of study drug
    6
    7.7%
    20
    13.5%
    Fatal adverse events
    53
    67.9%
    113
    76.4%
    Treatment-related adverse events (TRAEs)
    24
    30.8%
    51
    34.5%
    Treatment-related serious adverse events
    0
    0%
    6
    4.1%
    TRAEs leading to discontinuation of study drug
    0
    0%
    7
    4.7%
    Treatment-related fatal adverse events
    0
    0%
    0
    0%

    Adverse Events

    Time Frame From first dose of study drug to the end of study date; the median (min, max) duration was 10.0 (0.2, 41.4) and 9.4 (0.2, 42.9) months for Placebo and Denosumab respectively.
    Adverse Event Reporting Description Data are included for all participants who received at least one dose of study drug. Other Adverse Events summarizes the non-serious occurrences of adverse events that exceed the indicated frequency threshold.
    Arm/Group Title Placebo Denosumab
    Arm/Group Description Participants received placebo matching to denosumab by subcutaneous injection once every 4 weeks (Q4W) plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received placebo as often as once every 3 weeks (Q3W) while receiving Q3W chemotherapy. Participants with bone metastases also received 4 mg zoledronic acid administered as an IV infusion Q4W or Q3W. Participants received 120 mg denosumab by subcutaneous injection once every 4 weeks plus one loading dose on study day 8 in addition to platinum-based standard chemotherapy. Participants may have received denosumab as often as Q3W while receiving Q3W chemotherapy. Participants with bone metastases also received placebo to zoledronic acid administered as an IV infusion Q4W or Q3W.
    All Cause Mortality
    Placebo Denosumab
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total / (NaN) / (NaN)
    Serious Adverse Events
    Placebo Denosumab
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 68/76 (89.5%) 129/145 (89%)
    Blood and lymphatic system disorders
    Anaemia 5/76 (6.6%) 5/145 (3.4%)
    Febrile neutropenia 5/76 (6.6%) 6/145 (4.1%)
    Neutropenia 2/76 (2.6%) 3/145 (2.1%)
    Pancytopenia 0/76 (0%) 2/145 (1.4%)
    Thrombocytopenia 5/76 (6.6%) 4/145 (2.8%)
    Cardiac disorders
    Acute myocardial infarction 1/76 (1.3%) 0/145 (0%)
    Atrial fibrillation 2/76 (2.6%) 1/145 (0.7%)
    Cardiac failure 2/76 (2.6%) 3/145 (2.1%)
    Cardiac failure congestive 0/76 (0%) 1/145 (0.7%)
    Cardiac tamponade 0/76 (0%) 1/145 (0.7%)
    Cardio-respiratory arrest 3/76 (3.9%) 4/145 (2.8%)
    Coronary artery insufficiency 1/76 (1.3%) 0/145 (0%)
    Myocardial infarction 0/76 (0%) 1/145 (0.7%)
    Palpitations 0/76 (0%) 1/145 (0.7%)
    Tachycardia 0/76 (0%) 1/145 (0.7%)
    Congenital, familial and genetic disorders
    Hereditary angioedema 0/76 (0%) 1/145 (0.7%)
    Endocrine disorders
    Inappropriate antidiuretic hormone secretion 0/76 (0%) 2/145 (1.4%)
    Gastrointestinal disorders
    Abdominal pain 0/76 (0%) 1/145 (0.7%)
    Abdominal wall haematoma 1/76 (1.3%) 0/145 (0%)
    Constipation 1/76 (1.3%) 0/145 (0%)
    Diarrhoea 0/76 (0%) 1/145 (0.7%)
    Intestinal obstruction 2/76 (2.6%) 1/145 (0.7%)
    Melaena 1/76 (1.3%) 0/145 (0%)
    Nausea 1/76 (1.3%) 6/145 (4.1%)
    Small intestinal obstruction 0/76 (0%) 2/145 (1.4%)
    Small intestinal perforation 0/76 (0%) 1/145 (0.7%)
    Upper gastrointestinal haemorrhage 0/76 (0%) 1/145 (0.7%)
    Vomiting 2/76 (2.6%) 4/145 (2.8%)
    General disorders
    Asthenia 0/76 (0%) 1/145 (0.7%)
    Chest pain 1/76 (1.3%) 4/145 (2.8%)
    Chills 0/76 (0%) 1/145 (0.7%)
    Condition aggravated 1/76 (1.3%) 1/145 (0.7%)
    Disease progression 2/76 (2.6%) 5/145 (3.4%)
    Fatigue 0/76 (0%) 2/145 (1.4%)
    General physical health deterioration 4/76 (5.3%) 7/145 (4.8%)
    Infusion site extravasation 1/76 (1.3%) 0/145 (0%)
    Malaise 1/76 (1.3%) 0/145 (0%)
    Oedema peripheral 1/76 (1.3%) 0/145 (0%)
    Pain 2/76 (2.6%) 0/145 (0%)
    Pyrexia 2/76 (2.6%) 12/145 (8.3%)
    Hepatobiliary disorders
    Hepatic failure 0/76 (0%) 1/145 (0.7%)
    Hyperbilirubinaemia 1/76 (1.3%) 0/145 (0%)
    Immune system disorders
    Anaphylactic reaction 0/76 (0%) 1/145 (0.7%)
    Infections and infestations
    Bronchitis 0/76 (0%) 1/145 (0.7%)
    Cellulitis 1/76 (1.3%) 2/145 (1.4%)
    Device related infection 1/76 (1.3%) 1/145 (0.7%)
    Fungal infection 1/76 (1.3%) 0/145 (0%)
    Gangrene 0/76 (0%) 1/145 (0.7%)
    Gastroenteritis shigella 0/76 (0%) 1/145 (0.7%)
    Lower respiratory tract infection 2/76 (2.6%) 1/145 (0.7%)
    Lung abscess 0/76 (0%) 1/145 (0.7%)
    Lung infection 0/76 (0%) 1/145 (0.7%)
    Oral herpes 0/76 (0%) 1/145 (0.7%)
    Pleural infection 1/76 (1.3%) 1/145 (0.7%)
    Pneumonia 7/76 (9.2%) 14/145 (9.7%)
    Respiratory tract infection 0/76 (0%) 3/145 (2.1%)
    Sepsis 1/76 (1.3%) 2/145 (1.4%)
    Septic shock 0/76 (0%) 3/145 (2.1%)
    Staphylococcal bacteraemia 1/76 (1.3%) 0/145 (0%)
    Injury, poisoning and procedural complications
    Femoral neck fracture 0/76 (0%) 1/145 (0.7%)
    Fracture 0/76 (0%) 1/145 (0.7%)
    Radiation oesophagitis 1/76 (1.3%) 0/145 (0%)
    Radius fracture 1/76 (1.3%) 0/145 (0%)
    Investigations
    C-reactive protein increased 1/76 (1.3%) 0/145 (0%)
    General physical condition abnormal 0/76 (0%) 1/145 (0.7%)
    Glomerular filtration rate decreased 0/76 (0%) 1/145 (0.7%)
    Nutritional condition abnormal 1/76 (1.3%) 0/145 (0%)
    Platelet count decreased 3/76 (3.9%) 1/145 (0.7%)
    Metabolism and nutrition disorders
    Cachexia 1/76 (1.3%) 1/145 (0.7%)
    Dehydration 2/76 (2.6%) 6/145 (4.1%)
    Hyperglycaemia 1/76 (1.3%) 2/145 (1.4%)
    Hypocalcaemia 0/76 (0%) 1/145 (0.7%)
    Hypoglycaemia 0/76 (0%) 1/145 (0.7%)
    Hypokalaemia 0/76 (0%) 1/145 (0.7%)
    Musculoskeletal and connective tissue disorders
    Back pain 1/76 (1.3%) 2/145 (1.4%)
    Musculoskeletal chest pain 1/76 (1.3%) 0/145 (0%)
    Musculoskeletal pain 2/76 (2.6%) 1/145 (0.7%)
    Osteonecrosis of jaw 0/76 (0%) 4/145 (2.8%)
    Spinal pain 0/76 (0%) 1/145 (0.7%)
    Vertebral lesion 0/76 (0%) 1/145 (0.7%)
    Neoplasms benign, malignant and unspecified (incl cysts and polyps)
    Adenocarcinoma 2/76 (2.6%) 4/145 (2.8%)
    Bronchial carcinoma 1/76 (1.3%) 0/145 (0%)
    Cancer pain 0/76 (0%) 1/145 (0.7%)
    Lung adenocarcinoma 2/76 (2.6%) 2/145 (1.4%)
    Lung adenocarcinoma stage IV 0/76 (0%) 1/145 (0.7%)
    Lung cancer metastatic 3/76 (3.9%) 9/145 (6.2%)
    Lung neoplasm malignant 5/76 (6.6%) 10/145 (6.9%)
    Lung squamous cell carcinoma metastatic 1/76 (1.3%) 0/145 (0%)
    Lymphangiosis carcinomatosa 0/76 (0%) 1/145 (0.7%)
    Malignant neoplasm progression 2/76 (2.6%) 2/145 (1.4%)
    Malignant pleural effusion 3/76 (3.9%) 1/145 (0.7%)
    Metastases to bone 2/76 (2.6%) 0/145 (0%)
    Metastases to bone marrow 0/76 (0%) 1/145 (0.7%)
    Metastases to central nervous system 1/76 (1.3%) 6/145 (4.1%)
    Metastases to lung 1/76 (1.3%) 0/145 (0%)
    Metastases to meninges 1/76 (1.3%) 2/145 (1.4%)
    Metastatic neoplasm 2/76 (2.6%) 0/145 (0%)
    Non-small cell lung cancer 9/76 (11.8%) 19/145 (13.1%)
    Non-small cell lung cancer metastatic 7/76 (9.2%) 14/145 (9.7%)
    Non-small cell lung cancer stage IV 5/76 (6.6%) 12/145 (8.3%)
    Nervous system disorders
    Altered state of consciousness 0/76 (0%) 2/145 (1.4%)
    Brain oedema 0/76 (0%) 1/145 (0.7%)
    Cerebrovascular accident 1/76 (1.3%) 2/145 (1.4%)
    Consciousness fluctuating 0/76 (0%) 1/145 (0.7%)
    Depressed level of consciousness 0/76 (0%) 1/145 (0.7%)
    Epilepsy 1/76 (1.3%) 0/145 (0%)
    Haemorrhage intracranial 0/76 (0%) 1/145 (0.7%)
    Headache 0/76 (0%) 1/145 (0.7%)
    Ischaemic stroke 0/76 (0%) 1/145 (0.7%)
    Lethargy 1/76 (1.3%) 0/145 (0%)
    Loss of consciousness 1/76 (1.3%) 0/145 (0%)
    Seizure 0/76 (0%) 2/145 (1.4%)
    Stroke in evolution 0/76 (0%) 1/145 (0.7%)
    Transient ischaemic attack 0/76 (0%) 1/145 (0.7%)
    Generalised tonic-clonic seizure 1/76 (1.3%) 0/145 (0%)
    Product Issues
    Device issue 1/76 (1.3%) 0/145 (0%)
    Psychiatric disorders
    Confusional state 0/76 (0%) 3/145 (2.1%)
    Delirium 0/76 (0%) 1/145 (0.7%)
    Mental status changes 0/76 (0%) 1/145 (0.7%)
    Suicide attempt 0/76 (0%) 1/145 (0.7%)
    Renal and urinary disorders
    Acute kidney injury 0/76 (0%) 1/145 (0.7%)
    Haematuria 1/76 (1.3%) 1/145 (0.7%)
    Renal haemorrhage 0/76 (0%) 1/145 (0.7%)
    Respiratory, thoracic and mediastinal disorders
    Acute respiratory distress syndrome 0/76 (0%) 1/145 (0.7%)
    Acute respiratory failure 1/76 (1.3%) 0/145 (0%)
    Chronic obstructive pulmonary disease 1/76 (1.3%) 5/145 (3.4%)
    Dyspnoea 2/76 (2.6%) 16/145 (11%)
    Epistaxis 1/76 (1.3%) 1/145 (0.7%)
    Haemoptysis 1/76 (1.3%) 2/145 (1.4%)
    Haemothorax 0/76 (0%) 1/145 (0.7%)
    Hypoxia 0/76 (0%) 2/145 (1.4%)
    Laryngeal mass 0/76 (0%) 1/145 (0.7%)
    Laryngeal obstruction 0/76 (0%) 1/145 (0.7%)
    Lung disorder 1/76 (1.3%) 0/145 (0%)
    Pleural effusion 2/76 (2.6%) 6/145 (4.1%)
    Pleuritic pain 0/76 (0%) 1/145 (0.7%)
    Pneumonitis 0/76 (0%) 1/145 (0.7%)
    Pneumothorax 1/76 (1.3%) 3/145 (2.1%)
    Pulmonary embolism 1/76 (1.3%) 6/145 (4.1%)
    Pulmonary haemorrhage 0/76 (0%) 1/145 (0.7%)
    Pulmonary hypertension 0/76 (0%) 1/145 (0.7%)
    Respiratory distress 0/76 (0%) 3/145 (2.1%)
    Respiratory failure 2/76 (2.6%) 4/145 (2.8%)
    Vascular disorders
    Aortic thrombosis 0/76 (0%) 1/145 (0.7%)
    Circulatory collapse 0/76 (0%) 1/145 (0.7%)
    Deep vein thrombosis 1/76 (1.3%) 1/145 (0.7%)
    Embolism 0/76 (0%) 1/145 (0.7%)
    Embolism venous 0/76 (0%) 1/145 (0.7%)
    Hypotension 0/76 (0%) 4/145 (2.8%)
    Jugular vein thrombosis 1/76 (1.3%) 0/145 (0%)
    Superior vena cava syndrome 0/76 (0%) 1/145 (0.7%)
    Thrombosis 0/76 (0%) 1/145 (0.7%)
    Other (Not Including Serious) Adverse Events
    Placebo Denosumab
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 71/76 (93.4%) 139/145 (95.9%)
    Blood and lymphatic system disorders
    Anaemia 38/76 (50%) 60/145 (41.4%)
    Leukopenia 4/76 (5.3%) 6/145 (4.1%)
    Neutropenia 15/76 (19.7%) 32/145 (22.1%)
    Thrombocytopenia 16/76 (21.1%) 16/145 (11%)
    Eye disorders
    Dry eye 4/76 (5.3%) 2/145 (1.4%)
    Lacrimation increased 3/76 (3.9%) 9/145 (6.2%)
    Gastrointestinal disorders
    Abdominal distension 4/76 (5.3%) 1/145 (0.7%)
    Abdominal pain 4/76 (5.3%) 13/145 (9%)
    Constipation 27/76 (35.5%) 50/145 (34.5%)
    Diarrhoea 14/76 (18.4%) 26/145 (17.9%)
    Dyspepsia 5/76 (6.6%) 8/145 (5.5%)
    Dysphagia 6/76 (7.9%) 5/145 (3.4%)
    Gastrooesophageal reflux disease 4/76 (5.3%) 6/145 (4.1%)
    Nausea 34/76 (44.7%) 64/145 (44.1%)
    Stomatitis 6/76 (7.9%) 7/145 (4.8%)
    Vomiting 14/76 (18.4%) 35/145 (24.1%)
    General disorders
    Asthenia 8/76 (10.5%) 24/145 (16.6%)
    Fatigue 33/76 (43.4%) 67/145 (46.2%)
    Mucosal inflammation 4/76 (5.3%) 8/145 (5.5%)
    Oedema peripheral 17/76 (22.4%) 26/145 (17.9%)
    Pain 7/76 (9.2%) 10/145 (6.9%)
    Pyrexia 9/76 (11.8%) 23/145 (15.9%)
    Infections and infestations
    Nasopharyngitis 5/76 (6.6%) 3/145 (2.1%)
    Oral candidiasis 5/76 (6.6%) 5/145 (3.4%)
    Pneumonia 2/76 (2.6%) 10/145 (6.9%)
    Urinary tract infection 4/76 (5.3%) 11/145 (7.6%)
    Herpes zoster 4/76 (5.3%) 0/145 (0%)
    Injury, poisoning and procedural complications
    Fall 5/76 (6.6%) 2/145 (1.4%)
    Investigations
    Aspartate aminotransferase increased 4/76 (5.3%) 3/145 (2.1%)
    Blood creatinine increased 3/76 (3.9%) 12/145 (8.3%)
    Platelet count decreased 8/76 (10.5%) 14/145 (9.7%)
    Weight decreased 10/76 (13.2%) 8/145 (5.5%)
    Metabolism and nutrition disorders
    Decreased appetite 23/76 (30.3%) 34/145 (23.4%)
    Hypercalcaemia 4/76 (5.3%) 5/145 (3.4%)
    Hyperglycaemia 4/76 (5.3%) 6/145 (4.1%)
    Hypocalcaemia 4/76 (5.3%) 24/145 (16.6%)
    Hypokalaemia 4/76 (5.3%) 8/145 (5.5%)
    Hypomagnesaemia 12/76 (15.8%) 17/145 (11.7%)
    Hyponatraemia 4/76 (5.3%) 3/145 (2.1%)
    Hypophosphataemia 5/76 (6.6%) 11/145 (7.6%)
    Musculoskeletal and connective tissue disorders
    Arthralgia 7/76 (9.2%) 11/145 (7.6%)
    Back pain 15/76 (19.7%) 20/145 (13.8%)
    Muscle spasms 4/76 (5.3%) 4/145 (2.8%)
    Musculoskeletal chest pain 5/76 (6.6%) 9/145 (6.2%)
    Musculoskeletal pain 5/76 (6.6%) 10/145 (6.9%)
    Pain in extremity 7/76 (9.2%) 14/145 (9.7%)
    Nervous system disorders
    Dizziness 7/76 (9.2%) 20/145 (13.8%)
    Dysgeusia 11/76 (14.5%) 8/145 (5.5%)
    Headache 8/76 (10.5%) 24/145 (16.6%)
    Neuropathy peripheral 9/76 (11.8%) 5/145 (3.4%)
    Paraesthesia 2/76 (2.6%) 8/145 (5.5%)
    Psychiatric disorders
    Anxiety 5/76 (6.6%) 14/145 (9.7%)
    Depression 6/76 (7.9%) 2/145 (1.4%)
    Insomnia 7/76 (9.2%) 12/145 (8.3%)
    Respiratory, thoracic and mediastinal disorders
    Cough 16/76 (21.1%) 26/145 (17.9%)
    Dyspnoea 18/76 (23.7%) 35/145 (24.1%)
    Epistaxis 5/76 (6.6%) 8/145 (5.5%)
    Haemoptysis 6/76 (7.9%) 10/145 (6.9%)
    Hiccups 4/76 (5.3%) 7/145 (4.8%)
    Productive cough 2/76 (2.6%) 9/145 (6.2%)
    Skin and subcutaneous tissue disorders
    Alopecia 6/76 (7.9%) 9/145 (6.2%)
    Dry skin 4/76 (5.3%) 6/145 (4.1%)
    Rash 8/76 (10.5%) 18/145 (12.4%)
    Vascular disorders
    Hypotension 5/76 (6.6%) 4/145 (2.8%)

    Limitations/Caveats

    [Not Specified]

    More Information

    Certain Agreements

    Principal Investigators are NOT employed by the organization sponsoring the study.

    The Clinical Trial Agreement generally does not restrict an investigator's discussion of trial results after completion. The Agreement permits Amgen a limited period of time to review material discussing trial results (typically up to 45 days and possible extension). Amgen may remove confidential information, but authors have final control and approval of publication content. For multicenter studies, the investigator agrees not to publish any results before the first multi-center publication.

    Results Point of Contact

    Name/Title Study Director
    Organization Amgen Inc.
    Phone 866-572-6436
    Email medinfo@amgen.com
    Responsible Party:
    Amgen
    ClinicalTrials.gov Identifier:
    NCT01951586
    Other Study ID Numbers:
    • 20120249
    • 2013-001662-42
    First Posted:
    Sep 26, 2013
    Last Update Posted:
    Oct 25, 2021
    Last Verified:
    Oct 1, 2021