Trial Outcomes & Findings for A Study of Belimumab in Idiopathic Membranous Glomerulonephropathy (NCT NCT01610492)

NCT ID: NCT01610492

Last Updated: 2017-10-13

Results Overview

Proteinuria based on urinary protein creatinine ratio (PCR) was measured from 2 consecutive 24 hour (h) urine collection pre and post dosing at Baseline and Week 28 and the mean PCR was determined at each time point. Baseline is defined as the mean of the pre and post dosing Day 0 values. The ratio is defined as the Week 28 value divided by the Baseline value. Ratio to Baseline: Estimated value = 0.76, 2-sided 95% confidence interval (CI)=0.57 to 1.01. The geometric mean method was used to calculate the CI. The analysis was performed on Intent-to-treat (ITT) Population which comprised of all eligible participants who received at least one dose of investigational drug. Only those participants available at the indicated time point (Week 28) were analyzed.

Recruitment status

COMPLETED

Study phase

PHASE2

Target enrollment

14 participants

Primary outcome timeframe

Baseline and Week 28

Results posted on

2017-10-13

Participant Flow

Eligible participants were recruited from July 2012 until March 2014, into this 2 part study of a initial treatment phase, a long term treatment phase, and then followed up for a further 6 months. Results have previously been presented for the initial treatment phase, up to the Week 28 and are now presented for the completed study.

Screening occurred within 35 days and no less than 14 days before the first scheduled study treatment dose. Total 21 participants were screened; 14 were randomized and entered the initial treatment phase while 11 participants entered the long-term treatment phase. Common reasons for screen failures were insufficient, or improvements in proteinuria.

Participant milestones

Participant milestones
Measure
Belimumab 10 mg/kg IV
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Overall Study
STARTED
14
Overall Study
COMPLETED
8
Overall Study
NOT COMPLETED
6

Reasons for withdrawal

Reasons for withdrawal
Measure
Belimumab 10 mg/kg IV
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Overall Study
Adverse Event
1
Overall Study
Lack of Efficacy
3
Overall Study
Other: Reached stopping criteria
1
Overall Study
Other:Treatment stopped due to remission
1

Baseline Characteristics

A Study of Belimumab in Idiopathic Membranous Glomerulonephropathy

Baseline characteristics by cohort

Baseline characteristics by cohort
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Age, Continuous
46.1 Years
STANDARD_DEVIATION 13.30 • n=5 Participants
Sex: Female, Male
Female
3 Participants
n=5 Participants
Sex: Female, Male
Male
11 Participants
n=5 Participants
Race/Ethnicity, Customized
Asian - Central/South Asian Heritage
4 Participants
n=5 Participants
Race/Ethnicity, Customized
White- White/Caucasian/European Heritage
9 Participants
n=5 Participants
Race/Ethnicity, Customized
Unknown
1 Participants
n=5 Participants

PRIMARY outcome

Timeframe: Baseline and Week 28

Population: Intent-to-Treat (ITT) Population

Proteinuria based on urinary protein creatinine ratio (PCR) was measured from 2 consecutive 24 hour (h) urine collection pre and post dosing at Baseline and Week 28 and the mean PCR was determined at each time point. Baseline is defined as the mean of the pre and post dosing Day 0 values. The ratio is defined as the Week 28 value divided by the Baseline value. Ratio to Baseline: Estimated value = 0.76, 2-sided 95% confidence interval (CI)=0.57 to 1.01. The geometric mean method was used to calculate the CI. The analysis was performed on Intent-to-treat (ITT) Population which comprised of all eligible participants who received at least one dose of investigational drug. Only those participants available at the indicated time point (Week 28) were analyzed.

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=11 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Proteinuria Levels at Week 28
0.7552 Ratio
Geometric Coefficient of Variation 45.0

PRIMARY outcome

Timeframe: Baseline and Week 28

Population: ITT Population. Only those participants available at the indicated time point (Week 28) were analyzed.

PLA2R autoantibody titers in serum were analyzed at Baseline and Week 28 by means of a validated anti- PLA2R enzyme linked immunosorbent assay (ELISA) from EuroImmun. Baseline is defined as the Day 0 value and change from Baseline was calculated as ratio to Baseline by dividing the Week 28 values with the Baseline values. Ratio to Baseline: Estimated value = 0.27, 2-sided 95% CI=0.12 to 0.58. The geometric mean method was used to calculate the CI.

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=11 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Anti-phospholipase A2 Receptor (PLA2R) Autoantibody Titers at Week 28
0.2666 Ratio
Geometric Coefficient of Variation 171.0

SECONDARY outcome

Timeframe: Baseline and Week 12, 28, 52, 76, 104 and 128/6 month follow-up

Population: ITT Population

Proteinuria based on urinary protein creatinine ratio (PCR) measured from 2 consecutive 24h urine collections at Baseline and Week 28, from a pre-intervention spot urine sample and 24 hour urine collection after visit at Weeks 12, 52, 76 and 104, and from a spot urine sample at week 128. Mean PCR was calculated at each time point where there were 2 samples. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Proteinuria Levels at the Indicated Time Points
Baseline, n=14
724.3157 milligrams per millimole (mg/mmol)
Geometric Coefficient of Variation 40.2
Proteinuria Levels at the Indicated Time Points
Week 12, n=13
670.8655 milligrams per millimole (mg/mmol)
Geometric Coefficient of Variation 46.9
Proteinuria Levels at the Indicated Time Points
Week 28, n=11
498.1255 milligrams per millimole (mg/mmol)
Geometric Coefficient of Variation 40.9
Proteinuria Levels at the Indicated Time Points
Week 52, n=9
356.4209 milligrams per millimole (mg/mmol)
Geometric Coefficient of Variation 174.8
Proteinuria Levels at the Indicated Time Points
Week 76, n=8
274.9714 milligrams per millimole (mg/mmol)
Geometric Coefficient of Variation 70.4
Proteinuria Levels at the Indicated Time Points
Week 104, n=10
129.9761 milligrams per millimole (mg/mmol)
Geometric Coefficient of Variation 186.0
Proteinuria Levels at the Indicated Time Points
Week 128, n=9
75.2359 milligrams per millimole (mg/mmol)
Geometric Coefficient of Variation 136.4

SECONDARY outcome

Timeframe: Baseline and Week 12, 28, 52, 76, 104 and Week 128/6 month follow up

Population: ITT Population.

Proteinuria based on urinary protein creatinine ratio (PCR) measured from 2 consecutive 24h urine collections at Baseline and Week 28, from a pre-intervention spot urine sample and 24 hour urine collection after visit at Weeks 12, 52, 76 and 104, and from a spot urine sample at week 128. Mean PCR was calculated at each time point where there were 2 samples. Baseline is defined as Day 0 value and change from Baseline was calculated as ratio of post-Baseline value divided by the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Proteinuria Levels at the Indicated Time Points
Week 12; n= 13
0.9437 Ratio
Geometric Coefficient of Variation 39.3
Change From Baseline in Proteinuria Levels at the Indicated Time Points
Week 28; n= 11
0.7552 Ratio
Geometric Coefficient of Variation 45.0
Change From Baseline in Proteinuria Levels at the Indicated Time Points
Week 52; n= 9
0.5297 Ratio
Geometric Coefficient of Variation 206.7
Change From Baseline in Proteinuria Levels at the Indicated Time Points
Week 76; n= 8
0.4177 Ratio
Geometric Coefficient of Variation 54.3
Change From Baseline in Proteinuria Levels at the Indicated Time Points
Week 104; n= 10
0.1874 Ratio
Geometric Coefficient of Variation 189.3
Change From Baseline in Proteinuria Levels at the Indicated Time Points
Week 128; n= 9
0.1118 Ratio
Geometric Coefficient of Variation 139.1

SECONDARY outcome

Timeframe: Baseline and Week 12, 28, 52, 76, 104 and 128/6 month follow-up

Population: ITT Population.

Anti-PLA2R autoantibody titers in serum were analyzed by means of a validated anti-PLA2R enzyme linked immunosorbent assay (ELISA) assay from Euroimmun. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Anti-phospholipase A2 Receptor (PLA2R) Autoantibody Levels at Indicated Time Points
Baseline, n=14
168.3 relative units per milliliter (RU/mL)
Geometric Coefficient of Variation 138.9
Anti-phospholipase A2 Receptor (PLA2R) Autoantibody Levels at Indicated Time Points
Week 28, n=11
46.4 relative units per milliliter (RU/mL)
Geometric Coefficient of Variation 319.6
Anti-phospholipase A2 Receptor (PLA2R) Autoantibody Levels at Indicated Time Points
Week 52, n=9
12.9 relative units per milliliter (RU/mL)
Geometric Coefficient of Variation 358.4
Anti-phospholipase A2 Receptor (PLA2R) Autoantibody Levels at Indicated Time Points
Week 76, n=8
7.5 relative units per milliliter (RU/mL)
Geometric Coefficient of Variation 140.4
Anti-phospholipase A2 Receptor (PLA2R) Autoantibody Levels at Indicated Time Points
Week 104, n=10
3.7 relative units per milliliter (RU/mL)
Geometric Coefficient of Variation 72.7
Anti-phospholipase A2 Receptor (PLA2R) Autoantibody Levels at Indicated Time Points
Week 128, n=8
4.4 relative units per milliliter (RU/mL)
Geometric Coefficient of Variation 112.0
Anti-phospholipase A2 Receptor (PLA2R) Autoantibody Levels at Indicated Time Points
Week 12, n=13
91.0 relative units per milliliter (RU/mL)
Geometric Coefficient of Variation 200.4

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128.

Population: ITT Population

Anti-PLA2R autoantibody titers in serum were analyzed by means of a validated anti- PLA2R enzyme linked immunosorbent assay (ELISA) from Euroimmun. Baseline is defined as the Day 0 value and change from Baseline was calculated as ratio of post-Baseline value divided by the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Anti-PLA2R Autoantibody Titers at the Indicated Time Points
Week 12; n= 13
0.5362 Ratio
Geometric Coefficient of Variation 58.1
Change From Baseline in Anti-PLA2R Autoantibody Titers at the Indicated Time Points
Week 28; n= 11
0.2666 Ratio
Geometric Coefficient of Variation 171.0
Change From Baseline in Anti-PLA2R Autoantibody Titers at the Indicated Time Points
Week 52; n= 9
0.0737 Ratio
Geometric Coefficient of Variation 130.3
Change From Baseline in Anti-PLA2R Autoantibody Titers at the Indicated Time Points
Week 76; n= 8
0.0436 Ratio
Geometric Coefficient of Variation 102.8
Change From Baseline in Anti-PLA2R Autoantibody Titers at the Indicated Time Points
Week 104; n= 10
0.0212 Ratio
Geometric Coefficient of Variation 169.1
Change From Baseline in Anti-PLA2R Autoantibody Titers at the Indicated Time Points
Week 128; n= 8
0.0284 Ratio
Geometric Coefficient of Variation 243.7

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128

Population: ITT Population

Complete remission is defined as PCR \<30 mg/mmol (proteinuria \<0.3grams \[g\]/24 h) with no worsening in renal function (estimated glomerular filtration rate \[eGFR\] reduction from Baseline \<15 percent). Partial remission is defined as PCR \<350 mg/mmol (proteinuria \<3.5 g/24 h) but \>= 30 mg/mmol (proteinuria \>= 0.3g/24h) and decrease of \>50 percent from Day 0 Baseline, together with no consistent worsening in renal function (eGFR reduction from Baseline \<15percent). Only those participants available at the specified time points were analyzed (represented by n=X in category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Number of Participants With Complete or Partial Remission
Week 12; Partial remission; n= 13
0 Participants
Number of Participants With Complete or Partial Remission
Week 12; Complete remission; n= 13
0 Participants
Number of Participants With Complete or Partial Remission
Week 28; Partial remission; n= 11
1 Participants
Number of Participants With Complete or Partial Remission
Week 28; Complete remission; n= 11
0 Participants
Number of Participants With Complete or Partial Remission
Week 52; Partial remission; n= 9
2 Participants
Number of Participants With Complete or Partial Remission
Week 52; Complete remission; n= 9
1 Participants
Number of Participants With Complete or Partial Remission
Week 76; Partial remission; n= 8
3 Participants
Number of Participants With Complete or Partial Remission
Week 76; Complete remission; n= 8
0 Participants
Number of Participants With Complete or Partial Remission
Week 104; Partial remission; n= 10
6 Participants
Number of Participants With Complete or Partial Remission
Week 104; Complete remission; n= 10
1 Participants
Number of Participants With Complete or Partial Remission
Week 128; Partial remission; n= 9
6 Participants
Number of Participants With Complete or Partial Remission
Week 128; Complete remission; n= 9
1 Participants

SECONDARY outcome

Timeframe: Baseline and up to Week 128/6 month follow up

Population: ITT Population

Time to complete or partial remission was estimated using the Kaplan-Meier method. Complete remission is defined as PCR \<30 mg/mmol (proteinuria \<0.3g/24 h) with no worsening in renal function (eGFR reduction from Baseline \<15 percent ). Partial remission is defined as PCR \<350 mg/mmol (proteinuria \<3.5 g/24 h) but \>= 30 mg/mmol (proteinuria \>= 0.3g/24h) and decrease of \>50 percent from Day 0 Baseline, together with no consistent worsening in renal function (eGFR reduction from Baseline \<15 percent). Only 1 participant reached complete remission. Hence, statistical analysis for complete remission was not performed.

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Time to Complete or Partial Remission
68.20 Weeks
Interval 28.0 to 93.6

SECONDARY outcome

Timeframe: Baseline and up to Week 128/6 month follow up

Population: ITT Population

Complete remission is defined as PCR \<30 mg/mmol (proteinuria \<0.3g/24 h) with no worsening in renal function (eGFR reduction from Baseline \<15percent). Partial remission is defined as PCR \<350 mg/mmol (proteinuria \<3.5 g/24 h) but \>= 30 mg/mmol (proteinuria \>= 0.3g/24h) and decrease of \>50% from Day 0 Baseline, together with no consistent worsening in renal function (eGFR reduction from Baseline \<15percent). Only those participants available at the specified time points were analyzed (represented by n=X in category titles). NA indicates that data was not available as only 1 participant reached complete remission. Hence, standard deviation for complete remission was not calculated.

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Duration of Complete or Partial Remission
Complete remission; n= 1
365.0 Days
Standard Deviation NA
NA indicates that data was not available as only 1 participant reached complete remission. Hence, standard deviation for complete remission was not calculated.
Duration of Complete or Partial Remission
Partial remission; n= 9
378.6 Days
Standard Deviation 186.15

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128

Population: ITT Population

Incidence of anti-PLA2R autoantibody remission were evaluated by full response and partial response. Full response is defined as antibody undetectable, partial response is defined as reduction in titers by 50 percent. For anti PLA2R autoantibody data, log transformation was applied before the formal analyses. Anti-PLA2R autoantibody blood samples were evaluated at Week 12, 28, 52, 76, 104 and 128/6 week post last-dose. Only those participants available at the specified time points were analyzed (represented by n=X in category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Number of Participants With PLA2R Autoantibody Remission
Week 12; full response; n= 13
1 Participants
Number of Participants With PLA2R Autoantibody Remission
Week 12; partial response; n= 13
3 Participants
Number of Participants With PLA2R Autoantibody Remission
Week 28; full response; n= 11
3 Participants
Number of Participants With PLA2R Autoantibody Remission
Week 28; partial response; n= 11
6 Participants
Number of Participants With PLA2R Autoantibody Remission
Week 52; full response; n= 9
4 Participants
Number of Participants With PLA2R Autoantibody Remission
Week 52; partial response; n= 9
5 Participants
Number of Participants With PLA2R Autoantibody Remission
Week 76; full response; n= 8
4 Participants
Number of Participants With PLA2R Autoantibody Remission
Week 76; partial response; n= 8
4 Participants
Number of Participants With PLA2R Autoantibody Remission
Week 104; full response; n= 10
10 Participants
Number of Participants With PLA2R Autoantibody Remission
Week 104; partial response; n= 10
0 Participants
Number of Participants With PLA2R Autoantibody Remission
Week 128; Full response; n= 8
8 Participants
Number of Participants With PLA2R Autoantibody Remission
Week 128; partial response; n= 8
0 Participants

SECONDARY outcome

Timeframe: Baseline and up to Week 128/6 month follow up

Population: ITT Population

Time to anti-PLA2R autoantibody remission was estimated using Kaplan-Meier method for full response and partial response full response with antibody undetectable and partial response with reduction in titers by 50 percent.

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Time to Anti-PLA2R Autoantibody Remission
Partial response
16.20 Weeks
Interval 8.0 to 24.0
Time to Anti-PLA2R Autoantibody Remission
Complete response
82.00 Weeks
Interval 16.0 to 92.0

SECONDARY outcome

Timeframe: Baseline and up to Week 128/6 month follow up

Population: ITT Population

Incidence of anti-PLA2R autoantibody relapse defined as antibody detectable after previously undetectable. Anti-PLA2R autoantibody blood samples were evaluated at Week 12, 28, 52, 76, 104/4 week post last dose, Only those participants available at the specified time points were analyzed (represented by n=X in category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Number of Participants With Anti-PLA2R Autoantibody Relapse
Week 12; n= 13
0 Participants
Number of Participants With Anti-PLA2R Autoantibody Relapse
Week 28; n= 11
0 Participants
Number of Participants With Anti-PLA2R Autoantibody Relapse
Week 52; n= 9
0 Participants
Number of Participants With Anti-PLA2R Autoantibody Relapse
Week 76; n= 8
0 Participants
Number of Participants With Anti-PLA2R Autoantibody Relapse
Week 104; n= 10
0 Participants
Number of Participants With Anti-PLA2R Autoantibody Relapse
Week 128; n= 8
0 Participants

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128/6 month follow-up.

Population: ITT Population

eGFR was assessed from levels of creatinine using the 4 variable version of the modification of diet in renal disease (MDRD) equation as recommended by National Kidney Foundation-Chronic Kidney Disease (NKF-CKD) guidelines. Baseline for eGFR is defined as the mean of the screening and Day 0 values. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
eGFR Levels at the Indicated Time Points
Baseline; n= 14
69.8170 milliliter/minute (mL/min/1.73meter^2)
Geometric Coefficient of Variation 32.9
eGFR Levels at the Indicated Time Points
Week 12; n= 13
66.2639 milliliter/minute (mL/min/1.73meter^2)
Geometric Coefficient of Variation 35.4
eGFR Levels at the Indicated Time Points
Week 28; n= 11
65.0866 milliliter/minute (mL/min/1.73meter^2)
Geometric Coefficient of Variation 36.1
eGFR Levels at the Indicated Time Points
Week 52; n= 7
65.1308 milliliter/minute (mL/min/1.73meter^2)
Geometric Coefficient of Variation 45.0
eGFR Levels at the Indicated Time Points
Week 76; n= 8
61.6732 milliliter/minute (mL/min/1.73meter^2)
Geometric Coefficient of Variation 47.1
eGFR Levels at the Indicated Time Points
Week 104; n= 10
69.7692 milliliter/minute (mL/min/1.73meter^2)
Geometric Coefficient of Variation 39.1
eGFR Levels at the Indicated Time Points
Week 128; n= 9
64.7984 milliliter/minute (mL/min/1.73meter^2)
Geometric Coefficient of Variation 39.2

SECONDARY outcome

Timeframe: Baseline and up to Week 128/6 month follow up

Population: ITT Population

eGFR was assessed from levels of creatinine using the 4 variable version of the modification of diet in renal disease (MDRD) equation as recommended by national kidney foundation-chronic kidney disease (NKF-CKD) guidelines. Baseline for eGFR is defined as the mean of the Screening and Day 0 values and change from Baseline was calculated as ratio of post-Baseline value divided by the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 2, n=14
0.9954 Ratio
Geometric Coefficient of Variation 10.8
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 4, n= 13
0.9190 Ratio
Geometric Coefficient of Variation 16.4
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 8, n= 13
0.9035 Ratio
Geometric Coefficient of Variation 16.2
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 12, n= 13
0.9339 Ratio
Geometric Coefficient of Variation 17.1
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 16, n= 12
0.9521 Ratio
Geometric Coefficient of Variation 12.5
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 20, n= 8
0.9819 Ratio
Geometric Coefficient of Variation 11.8
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 24, n= 11
0.9274 Ratio
Geometric Coefficient of Variation 17.1
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 28, n= 11
0.9694 Ratio
Geometric Coefficient of Variation 21.1
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 32, n= 9
1.0099 Ratio
Geometric Coefficient of Variation 19.5
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 36, n= 11
0.9692 Ratio
Geometric Coefficient of Variation 17.4
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 40, n= 11
0.9425 Ratio
Geometric Coefficient of Variation 20.8
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 44, n= 10
0.9531 Ratio
Geometric Coefficient of Variation 22.6
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 48, n= 8
0.9929 Ratio
Geometric Coefficient of Variation 23.8
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 52, n= 7
0.9290 Ratio
Geometric Coefficient of Variation 27.7
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 56, n= 9
1.0181 Ratio
Geometric Coefficient of Variation 21.9
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 60, n= 9
0.9805 Ratio
Geometric Coefficient of Variation 26.8
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 64, n=9
1.0217 Ratio
Geometric Coefficient of Variation 23.5
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 68, n=8
1.0233 Ratio
Geometric Coefficient of Variation 24.6
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 72, n=8
1.0168 Ratio
Geometric Coefficient of Variation 28.3
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 76, n=8
1.0055 Ratio
Geometric Coefficient of Variation 33.3
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 80, n=8
1.0431 Ratio
Geometric Coefficient of Variation 21.6
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 84, n=8
0.9959 Ratio
Geometric Coefficient of Variation 25.3
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 88, n=8
1.0529 Ratio
Geometric Coefficient of Variation 16.3
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 92, n=8
1.0052 Ratio
Geometric Coefficient of Variation 26.3
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 96, n=8
1.1204 Ratio
Geometric Coefficient of Variation 22.1
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 100, n=8
1.1122 Ratio
Geometric Coefficient of Variation 24.7
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 104, n=10
1.0480 Ratio
Geometric Coefficient of Variation 23.3
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 116; n= 8
1.0373 Ratio
Geometric Coefficient of Variation 19.8
Change From Baseline in eGFR Levels at the Indicated Time Points
Week 128, n=9
0.9971 Ratio
Geometric Coefficient of Variation 35.2

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128/6 month follow-up

Population: ITT Population

Serum creatinine was assessed as a clinical chemistry laboratory parameter from Baseline up to Week 128/6 month follow-up visit. Baseline for serum creatinine is defined as the mean of the Screening and Day 0 values. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Serum Creatinine Levels at the Indicated Time Points
Baseline; n= 14
97.1658 micromoles/liter (µmol/L)
Geometric Coefficient of Variation 22.8
Serum Creatinine Levels at the Indicated Time Points
Week 12; n= 13
100.6421 micromoles/liter (µmol/L)
Geometric Coefficient of Variation 26.8
Serum Creatinine Levels at the Indicated Time Points
Week 28; n= 11
99.9535 micromoles/liter (µmol/L)
Geometric Coefficient of Variation 24.8
Serum Creatinine Levels at the Indicated Time Points
Week 52; n= 7
96.6583 micromoles/liter (µmol/L)
Geometric Coefficient of Variation 30.2
Serum Creatinine Levels at the Indicated Time Points
Week 76; n= 8
103.0265 micromoles/liter (µmol/L)
Geometric Coefficient of Variation 32.8
Serum Creatinine Levels at the Indicated Time Points
Week 104; n= 10
92.4547 micromoles/liter (µmol/L)
Geometric Coefficient of Variation 29.6
Serum Creatinine Levels at the Indicated Time Points
Week 128; n= 9
97.7260 micromoles/liter (µmol/L)
Geometric Coefficient of Variation 29.7

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128/6 month follow-up

Population: ITT Population

Serum creatinine was assessed as a clinical chemistry laboratory parameter from Baseline up to Week 128/6 month follow-up visit. Baseline for serum creatinine is defined as the mean of the Screening and Day 0 values and change from Baseline was calculated as ratio of post-Baseline value divided by the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Serum Creatinine Levels at the Indicated Time Points
Week 12; n= 13
1.0530 Ratio
Geometric Coefficient of Variation 14.8
Change From Baseline in Serum Creatinine Levels at the Indicated Time Points
Week 28; n= 11
1.0202 Ratio
Geometric Coefficient of Variation 18.6
Change From Baseline in Serum Creatinine Levels at the Indicated Time Points
Week 52; n= 7
1.0581 Ratio
Geometric Coefficient of Variation 23.7
Change From Baseline in Serum Creatinine Levels at the Indicated Time Points
Week 76; n= 8
0.9818 Ratio
Geometric Coefficient of Variation 28.7
Change From Baseline in Serum Creatinine Levels at the Indicated Time Points
Week 104; n= 10
0.9467 Ratio
Geometric Coefficient of Variation 20.1
Change From Baseline in Serum Creatinine Levels at the Indicated Time Points
Week 128; n= 9
0.9874 Ratio
Geometric Coefficient of Variation 30.4

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128/6 month follow-up.

Population: ITT Population

Serum albumin was assessed as a clinical chemistry laboratory parameter from Baseline up to Week 128/6 month follow-up visit. Baseline for serum albumin is defined as the mean of the Screening and Day 0 values. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Serum Albumin Levels at Indicated Time Points
Baseline; n= 14
23.3306 grams per liter (g/L)
Geometric Coefficient of Variation 22.7
Serum Albumin Levels at Indicated Time Points
Week 12; n= 13
24.4204 grams per liter (g/L)
Geometric Coefficient of Variation 27.5
Serum Albumin Levels at Indicated Time Points
Week 28; n= 11
27.8397 grams per liter (g/L)
Geometric Coefficient of Variation 23.4
Serum Albumin Levels at Indicated Time Points
Week 52; n= 7
31.9927 grams per liter (g/L)
Geometric Coefficient of Variation 18.3
Serum Albumin Levels at Indicated Time Points
Week 76; n= 8
33.9484 grams per liter (g/L)
Geometric Coefficient of Variation 10.9
Serum Albumin Levels at Indicated Time Points
Week 104; n= 10
39.1015 grams per liter (g/L)
Geometric Coefficient of Variation 7.5
Serum Albumin Levels at Indicated Time Points
Week 128; n= 9
39.2009 grams per liter (g/L)
Geometric Coefficient of Variation 8.8

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128/6 month follow-up

Population: ITT Population

Serum albumin was assessed as a clinical chemistry laboratory parameter from Baseline up to Week 128/6 month follow-up visit. Baseline for serum albumin is defined as the mean of the Screening and Day 0 values and change from Baseline was calculated as ratio of post-Baseline value divided by the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Levels of Serum Albumin at the Indicated Time Points
Week 28; n= 11
1.1211 ratio
Geometric Coefficient of Variation 16.9
Change From Baseline in Levels of Serum Albumin at the Indicated Time Points
Week 12; n= 13
1.0324 ratio
Geometric Coefficient of Variation 15.0
Change From Baseline in Levels of Serum Albumin at the Indicated Time Points
Week 52; n= 7
1.2828 ratio
Geometric Coefficient of Variation 28.1
Change From Baseline in Levels of Serum Albumin at the Indicated Time Points
Week 76; n= 8
1.3695 ratio
Geometric Coefficient of Variation 23.8
Change From Baseline in Levels of Serum Albumin at the Indicated Time Points
Week 104; n= 10
1.5879 ratio
Geometric Coefficient of Variation 19.6
Change From Baseline in Levels of Serum Albumin at the Indicated Time Points
Week 128; n= 9
1.5799 ratio
Geometric Coefficient of Variation 24.7

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128/6 month follow-up

Population: ITT Population

Serum cholesterol was assessed as a clinical chemistry laboratory parameter from Baseline up to Week 128/6 month follow-up visit. Baseline for serum cholesterol is defined as the mean of the Screening and Day 0 values. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Serum Cholesterol Levels at Indicated Time Points
Baseline; n= 14
7.6423 millimoles per liter (mmol/L)
Geometric Coefficient of Variation 36.0
Serum Cholesterol Levels at Indicated Time Points
Week 12; n= 13
7.2336 millimoles per liter (mmol/L)
Geometric Coefficient of Variation 31.5
Serum Cholesterol Levels at Indicated Time Points
Week 28; n= 11
6.2740 millimoles per liter (mmol/L)
Geometric Coefficient of Variation 23.1
Serum Cholesterol Levels at Indicated Time Points
Week 52; n= 7
5.8724 millimoles per liter (mmol/L)
Geometric Coefficient of Variation 18.6
Serum Cholesterol Levels at Indicated Time Points
Week 76; n= 8
5.0211 millimoles per liter (mmol/L)
Geometric Coefficient of Variation 18.4
Serum Cholesterol Levels at Indicated Time Points
Week 104; n= 10
4.8001 millimoles per liter (mmol/L)
Geometric Coefficient of Variation 24.5
Serum Cholesterol Levels at Indicated Time Points
Week 128; n= 9
4.2407 millimoles per liter (mmol/L)
Geometric Coefficient of Variation 12.7

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128/6 month follow-up

Population: ITT Population

Serum cholesterol was assessed as a clinical chemistry laboratory parameter from Baseline up to Week 128/6 month follow-up visit. Baseline for serum cholesterol is defined as the mean of the Screening and Day 0 values and change from Baseline was calculated as ratio of post-Baseline value divided by the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Serum Cholesterol at the Indicated Time Points
Week 12; n= 13
0.9238 mmol/L
Geometric Coefficient of Variation 16.8
Change From Baseline in Serum Cholesterol at the Indicated Time Points
Week 28; n= 11
0.8896 mmol/L
Geometric Coefficient of Variation 14.3
Change From Baseline in Serum Cholesterol at the Indicated Time Points
Week 52; n= 7
0.8571 mmol/L
Geometric Coefficient of Variation 16.7
Change From Baseline in Serum Cholesterol at the Indicated Time Points
Week 76; n= 8
0.7111 mmol/L
Geometric Coefficient of Variation 15.0
Change From Baseline in Serum Cholesterol at the Indicated Time Points
Week 104; n= 10
0.6851 mmol/L
Geometric Coefficient of Variation 17.3
Change From Baseline in Serum Cholesterol at the Indicated Time Points
Week 128; n= 9
0.6140 mmol/L
Geometric Coefficient of Variation 15.7

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128/6 month follow-up

Population: ITT Population

Serum IgG was assessed as a clinical chemistry laboratory parameter from Baseline up to Week 128/6 month follow-up visit. Baseline for serum IgG is defined as the pre-dose Day 0 value. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Serum Immunoglobulin G (IgG) Levels at Indicated Time Points
Baseline; n= 14
4.026 g/L
Standard Deviation 1.6810
Serum Immunoglobulin G (IgG) Levels at Indicated Time Points
Week 12; n= 13
3.871 g/L
Standard Deviation 1.5266
Serum Immunoglobulin G (IgG) Levels at Indicated Time Points
Week 28; n= 11
4.405 g/L
Standard Deviation 1.7833
Serum Immunoglobulin G (IgG) Levels at Indicated Time Points
Week 52; n= 8
5.823 g/L
Standard Deviation 2.2572
Serum Immunoglobulin G (IgG) Levels at Indicated Time Points
Week 76; n= 8
6.050 g/L
Standard Deviation 2.1103
Serum Immunoglobulin G (IgG) Levels at Indicated Time Points
Week 104; n= 10
7.611 g/L
Standard Deviation 2.8301
Serum Immunoglobulin G (IgG) Levels at Indicated Time Points
Week 128; n= 9
8.609 g/L
Standard Deviation 2.2597

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, 104 and 128/6 month follow up

Population: ITT Population

Serum IgG was assessed as a clinical chemistry laboratory parameter from Baseline up to Week 128/6 month follow-up visit. Baseline for serum IgG is defined as the pre-dose Day 0 value and change from Baseline was calculated as ratio of post-Baseline value divided by the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Serum IgG at the Indicated Time Points
Week 12; n= 13
0.055 Ratio
Standard Deviation 0.6625
Change From Baseline in Serum IgG at the Indicated Time Points
Week 28; n= 11
0.469 Ratio
Standard Deviation 1.4287
Change From Baseline in Serum IgG at the Indicated Time Points
Week 52; n= 8
1.525 Ratio
Standard Deviation 2.4411
Change From Baseline in Serum IgG at the Indicated Time Points
Week 76; n= 8
1.619 Ratio
Standard Deviation 1.6681
Change From Baseline in Serum IgG at the Indicated Time Points
Week 104; n= 10
3.613 Ratio
Standard Deviation 2.5488
Change From Baseline in Serum IgG at the Indicated Time Points
Week 128; n= 9
4.334 Ratio
Standard Deviation 2.0289

SECONDARY outcome

Timeframe: Baseline and Weeks 12, 28, 52, 76, and 104

Population: ITT Population

Reduction of proteinuria lessens the risk of thromboembolic and cardiovascular effects and reduces the edema in participants. Investigators physically reviewed participants for clinical manifestations of idiopathic membranous glomerulonephropathy (IMGN) (e.g. edema extending beyond calf) during study and analysis was performed at Week 12, 28, 52, 76 Week 104. Only those participants available at the specified time points were analyzed (represented by n=X in category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Number of Participants With Edema and Edema Extending Beyond Calf
4 Week PFD; Oedema extending beyond calf; n= 10
1 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 0; oedema; n= 14
13 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 0; oedema extending beyond calf; n= 14
5 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 12; oedema; n= 12
9 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 12; Oedema extending beyond calf; n= 12
1 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 28;Oedema; n= 11
7 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 28; Oedema extending beyond calf; n= 11
1 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 52; oedema; n= 9
4 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 52;Oedema extending beyond calf; n= 9
1 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 76;Oedema; n=8
4 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 76;Oedema extending beyond calf; n= 8
0 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
4 Week post final dose (PFD); Oedema; n=10
6 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 104 withdrawn (WD); Oedema; n= 1
0 Participants
Number of Participants With Edema and Edema Extending Beyond Calf
Week 104 WD;Oedema extending beyond calf; n= 1
0 Participants

SECONDARY outcome

Timeframe: Baseline and up to 4 week post last dose

Population: PK Population: all participants in the ITT Population for whom a PK sample was obtained and analyzed.

The first occurrence of Cmax was determined directly from the serum concentration-time data. The pharmacokinetic (PK) parameters were calculated by standard non-compartmental analysis and all calculations of non-compartmental parameters are being based on actual sampling times.

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Summary of Maximum Observed Serum Concentration (Cmax) of Belimumab at the Indicated Time Points
Day 0, 5 minutes; n= 13
267996.0 nanograms per milliliter (ng/mL)
Standard Deviation 70598.02
Summary of Maximum Observed Serum Concentration (Cmax) of Belimumab at the Indicated Time Points
Week 28, 5 minutes; n= 11
312462.2 nanograms per milliliter (ng/mL)
Standard Deviation 95171.16

SECONDARY outcome

Timeframe: Baseline and up to 4 week post last dose

Population: PK Population

Trough concentration (Cmin) samples collected on the specified days are being used to assess attainment whether there was sufficient belimumab despite it being lost in the urine from the proteinuria and to check if it improved as proteinuria resolved. Analysis was performed on pre-infusion samples at weeks 2,4,8,12,28,40,52,76 and the 4 week post last-dose.

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Summary of Minimum Observed Concentration (Cmin) of Belimumab at the Indicated Time Points
Pre-infusion; Week 2; n= 14
29940.8 ng/mL
Standard Deviation 20918.28
Summary of Minimum Observed Concentration (Cmin) of Belimumab at the Indicated Time Points
Pre-infusion; Week 4; n= 12
41220.9 ng/mL
Standard Deviation 35720.14
Summary of Minimum Observed Concentration (Cmin) of Belimumab at the Indicated Time Points
Pre-infusion; Week 8; n= 13
30350.9 ng/mL
Standard Deviation 25505.70
Summary of Minimum Observed Concentration (Cmin) of Belimumab at the Indicated Time Points
Pre-infusion; Week 12; n= 11
30913.7 ng/mL
Standard Deviation 30681.42
Summary of Minimum Observed Concentration (Cmin) of Belimumab at the Indicated Time Points
Pre-infusion; Week 28; n= 11
34904.7 ng/mL
Standard Deviation 26388.60
Summary of Minimum Observed Concentration (Cmin) of Belimumab at the Indicated Time Points
Pre-infusion; Week 40; n= 10
40800.6 ng/mL
Standard Deviation 28847.86
Summary of Minimum Observed Concentration (Cmin) of Belimumab at the Indicated Time Points
Pre-infusion; Week 52; n= 9
38375.9 ng/mL
Standard Deviation 14136.23
Summary of Minimum Observed Concentration (Cmin) of Belimumab at the Indicated Time Points
Pre-infusion; Week 76; n= 8
65655.8 ng/mL
Standard Deviation 32873.33
Summary of Minimum Observed Concentration (Cmin) of Belimumab at the Indicated Time Points
4 Weeks post last-dose; n= 9
60497.3 ng/mL
Standard Deviation 28074.37

SECONDARY outcome

Timeframe: Baseline and up to 4 week post last dose

Population: PK Population

The AUC(0-2) was determined using the linear trapezoidal rule for increasing concentrations and the logarithmic trapezoidal rule for decreasing concentrations. Blood samples for PK analysis were collected at the following time points: pre-dose (on dosing days): Days 0, 1, 4, 7, 14 and Week 4, 8, 12, 28, 40, 52, 76 and 4 week post last dose. Post-dose (5 minutes after dosing complete): Days 0 and 28. The results will be posted at later date following post hoc analysis.

Outcome measures

Outcome data not reported

SECONDARY outcome

Timeframe: Baseline and Up to 4 week post last dose

Population: PK Population

PK parameters from the urine concentration data: urine Ae(0-24) were assessed. 24 h urine collections for PK analysis were collected after the Day 0 and Weeks 12, 28, 52, 76 doses and at the 4 week post last dose visit. A population approach was undertaken to characterize the population PK parameters and associated variability of belimumab in nephrotic participants. The population approach could have provided derived clearance of belimumab for each participant after the first dose. The population PK analysis was conducted using nonlinear mixed-effect modeling (NONMEM) or appropriate nonlinear mixed-effect analysis software. Several samples were taken pre-dose at Day 0 and some at week 12 incorrectly which affects interpretation.

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Summary of Total Amount of Urine Excreted Ae(0-24)
Day 0; n= 14
105826.23 ng/hour
Standard Deviation 178943.857
Summary of Total Amount of Urine Excreted Ae(0-24)
Week 12; n= 12
95188.14 ng/hour
Standard Deviation 130217.976
Summary of Total Amount of Urine Excreted Ae(0-24)
Week 28; n= 11
92997.94 ng/hour
Standard Deviation 110142.599
Summary of Total Amount of Urine Excreted Ae(0-24)
Week 52; n= 9
219367.96 ng/hour
Standard Deviation 391752.377
Summary of Total Amount of Urine Excreted Ae(0-24)
Week 76; n= 8
145645.34 ng/hour
Standard Deviation 267292.225
Summary of Total Amount of Urine Excreted Ae(0-24)
4 Week post last dose; n= 6
7909.34 ng/hour
Standard Deviation 16771.559

SECONDARY outcome

Timeframe: Baseline and up to Week 104/4 week post last dose

Population: ITT Population

Health-related quality of life was assessed through participant self-completion of the short form health survey (SF-36 version \[v2\]), a general health related quality of life metrics. Norm-based Scores (NBS) for physical functioning, role emotional, role physical were assessed. The remaining SF-36 component scores require re-scaling and therefore will be added at a later date. SF-36 was administered prior to any procedures at Weeks 12, 28, 52, 76 and 104/4 week post last dose. Item score were recorded and higher score represented better health status. Baseline is defined as Day 0 pre dose value and change from Baseline was calculated by subtracting the Baseline values from the individual post-randomization values. Only those participants available at the specified time points were analyzed (represented by n=X in category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Physical functioning, Week 28; n= 11
-0.765 Scores on a scale
Standard Deviation 3.6780
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Physical functioning, Week 52; n= 9
0.468 Scores on a scale
Standard Deviation 7.3489
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Physical functioning, Week 76; n= 7
0.601 Scores on a scale
Standard Deviation 7.9409
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Physical functioning 4 Week post final dose;n=11
0.765 Scores on a scale
Standard Deviation 11.7275
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Role emotional, Week 12; n= 12
-0.000 Scores on a scale
Standard Deviation 10.4831
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Physical functioning, Week 12; n= 12
-4.034 Scores on a scale
Standard Deviation 5.1907
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Role emotional, Week 28; n= 11
1.413 Scores on a scale
Standard Deviation 15.1824
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Role emotional, Week 52; n= 9
3.023 Scores on a scale
Standard Deviation 11.4621
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Role emotional, Week 76; n= 7
3.332 Scores on a scale
Standard Deviation 7.2475
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Role emotional, 4 Week post final dose; n= 11
6.007 Scores on a scale
Standard Deviation 10.0413
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Role physical, Week 12; n= 12
0.816 Scores on a scale
Standard Deviation 11.0681
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Role physical, Week 28; n= 11
3.340 Scores on a scale
Standard Deviation 8.4324
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Role physical, Week 52; n= 9
3.537 Scores on a scale
Standard Deviation 9.1726
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Role physical, Week 76; n= 7
7.697 Scores on a scale
Standard Deviation 12.2803
Change From Baseline in Short Form (SF)-36 v2 Quality of Life (QoL) Questionnaire Score
Role physical 4 Week post final dose; n= 11
5.789 Scores on a scale
Standard Deviation 11.4489

SECONDARY outcome

Timeframe: Baseline and up to Week 128/6 month follow up

Population: ITT Population

An AE is any untoward medical occurrence in a clinical investigation participant, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. The analysis was performed on Safety Population which comprised of all participants who were randomized into the study. SAE is any untoward medical occurrence that at any dose results in death, is life-threatening, Requires hospitalization or prolongation of existing hospitalization, Results in disability/incapacity, is a congenital anomaly/birth defect, may require medical or surgical intervention, is associated with liver injury and impaired liver function.

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
AE
14 Participants
Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
SAE
3 Participants

SECONDARY outcome

Timeframe: Baseline and up to Week 116/16 week follow-up visit

Population: ITT Population

Blood samples were collected from participants for evaluation of clinical chemistry and hematology parameters. The clinical chemistry parameters included albumin, alkaline phosphatase (alk.phosph.), alanine amino transferase (ALT), aspartate amino transferase (AST), total and direct bilirubin, calcium, cholesterol, chloride, carbon dioxide, creatinine, gamma glutamyl transferase (GGT), glucose, potassium, lactate dehydrogenase (LD), magnesium, sodium, phosphorus, total protein, blood urea nitrogen (BUN) and uric acid. The hematology parameters included basophils, eosinophil, hemoglobin, hematocrit, lymphocytes, monocytes, total neutrophils, platelets, red blood cells (RBC) count and white blood cells (WBC) count. Participants were counted in the category that their value changes to (low or high) for the specific time points. Only those participants with data available at the specified data points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; Week 12; to high; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Albumin; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; Week 12; to high; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Alk.phosph.; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; Week 12; to high; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
ALT; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; Week 12; to high; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; Week 28; to high; n= 11
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
AST; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; Week 12; to high; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; 16 Week follow up; to high; n=8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Direct bilirubin; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; Week 12; to high; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total bilirubin; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; Week 12; to high; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; Week 104; to high; n= 10
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; Week 104; to low; n= 10
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Calcium; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; Week 12; to high; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Cholesterol; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; Week 12; to high; n= 13
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; Week 28; to high; n= 11
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Chloride; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; Week 12; to high; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; Week 12; to low; n= 13
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; Week 28; to low; n= 11
3 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; Week 104; to low; n= 10
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Carbon dioxide; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; Week 12; to high; n= 13
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; Week 76; to high; n= 8
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; Week 76; to low; n= 8
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Creatinine; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; Week 12; to high; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; 16 Week follow up; to high; n=8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
GGT; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; Week 12; to high; n= 13
3 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; Week 28; to high; n= 11
4 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; Week 52; to high; n= 7
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; Week 76; to high; n= 8
3 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; Week 104; to high; n= 10
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; 16 Week follow up; to high; n= 8
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Glucose; 16 Week follow up; to low; n= 8
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; Week 12; to high; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; Week 28; to high; n= 11
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; Week 28; to low; n= 11
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Potassium; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD; Week 12; to high; n= 13
3 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD; Week 28; to high; n= 11
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD; Week 52; to high; n= 7
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD; Week 76; to high; n= 8
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD;Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD;Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD; 16 week follow-up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
LD; 16 week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; Week 12; to high; n= 13
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; 16 week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Magnesium; 16 week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; Week 12; to high; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; Week 28; to low; n= 11
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; Week 52; to low; n= 7
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; Week 76; to low; n= 8
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; 16 week follow p; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Sodium; 16 week follow up; to low; n= 8
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus; Week 12; to high; n= 13
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus; Week 28; to high; n= 11
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus;Week 104; to high; n= 10
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus;Week 104; to low; n= 10
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus; 16 week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Phosphorus; 16 week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; Week 12; to high; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; Week 52; to high; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; Week 52; to low; n= 7
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; 16 Week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Total protein; 16 Week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; Week 12; to high; n= 13
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; Week 12; to low; n= 13
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; Week 52; to high; n= 7
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; Week 76; to high; n= 8
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; Week 104; to high; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; 16 week follow up; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
BUN; 16 week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; Week 12; to high; n= 13
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; Week 12; to low; n= 13
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; Week 28; to high; n= 11
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; Week 52; to high; n= 7
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; Week 52; to low; n= 7
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; Week 76; to high; n= 8
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; Week 104; to high; n= 10
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; Week 104; to low; n= 10
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; 16 week follow up; to high; n= 8
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Uric acid; 16 week follow up; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; Week 12; to high; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; Week 52; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; Week 52; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; Week 104; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; Week 104; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; 16 week follow up; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Basophils; 16 week follow up; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; Week 12; to high; n= 12
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; Week 28; to high; n= 11
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; Week 52; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; Week 52; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; Week 76; to high; n= 8
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; Week 104; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; Week 104; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; 16 week follow up; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Eosinophils; 16 week follow up; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; Week 12; to high; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; Week 52; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; Week 52; to low; n= 9
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; Week 76; to low; n= 8
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; Week 104; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; Week 104; to low; n= 9
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; 16 week follow up; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hemoglobin; 16 week follow up; to low; n= 9
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; Week 12; to high; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; Week 28; to high; n= 11
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; Week 52; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; Week 52; to low; n= 9
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; Week 104; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; Week 104; to low; n= 9
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; 16 week follow up; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Hematocrit; 16 week follow up; to low; n= 9
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; Week 12; to high; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; Week 52; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; Week 52; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; Week 104; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; Week 104; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; 16 week follow up; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Lymphocytes; 16 week follow up; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; Week 12; to high; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; Week 52; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; Week 52; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; Week 104; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; Week 104; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; 16 week follow up; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Monocytes; 16 week follow up; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; Week 12; to high; n= 12
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; Week 52; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; Week 52; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; Week 104; to high; n= 9
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; Week 104; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; 16 week follow up; to high; n=9
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Neutrophils; 16 week follow up; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; Week 12; to high; n= 12
3 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; Week 28; to high; n= 11
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; Week 52; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; Week 52; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; Week 104; to high; n= 9
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; Week 104; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; 16 week follow up; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
Platelet count; 16 week follow up; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; Week 12; to high; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; Week 12; to low; n= 12
3 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; Week 28; to low; n= 11
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; Week 52; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; Week 52; to low; n= 9
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; Week 104; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; Week 104; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; 16 week follow up; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
RBC; 16 week follow up; to low; n= 9
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; Week 12; to high; n= 12
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; Week 12; to low; n= 12
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; Week 28; to high; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; Week 28; to low; n= 11
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; Week 52; to high; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; Week 52; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; Week 76; to high; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; Week 76; to low; n= 8
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; Week 104; to high; n= 9
2 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; Week 104; to low; n= 9
0 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; 16 week follow up; to high; n= 9
1 Participants
Number of Participants With Abnormal Clinical Chemistry and Hematology Values
WBC; 16 week follow up; to low; n= 9
0 Participants

SECONDARY outcome

Timeframe: Baseline and up to Week 116/16 Week follow up

Population: ITT Population

Urine samples were collected for urinalysis by dipstick method from Baseline up to Week 116/16 months follow up and number of participants with findings were presented for Baseline, Week 12, 28, 52, 76, 104/4 weeks post last-dose and Week 116/16 week follow up (WF). The urinalysis parameters included occult blood, glucose, ketones, protein. The findings were presented as trace or 1/10 g/100 milliliter (dL), trace, negative, 4+, 3+, 3+ or 1 g/dL, 2+ or 1/2 g/dL, 2+, 1+ or 1/4 g/dL and 1+. Only participants present at the specific time points were presented (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Number of Participants With Urinalysis Dipstick Findings
Week 12; occult blood; Trace or 1/10 g/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12;Occult Blood; Trace; n= 12
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Occult Blood; negative; n= 12
2 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Occult Blood; 4+; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Occult Blood; 3+ OR 1 G/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Occult Blood; 3+; n= 12
2 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Occult Blood; 2+ OR 1/2 G/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Occult Blood; 2+ ; n= 12
6 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Occult Blood;1+ OR 1/4 G/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Occult Blood; 1+; n= 12
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; glucose; Trace or 1/10 g/DL; n= 12
3 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12;glucose; Trace; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; glucose; negative; n= 12
8 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; glucose; 4+; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; glucose; 3+ OR 1 G/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; glucose; 3+; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; glucose; 2+ OR 1/2 G/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; glucose; 2+ ; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; glucose;1+ OR 1/4 G/DL; n= 12
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; glucose; 1+; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; ketones; Trace or 1/10 g/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; ketones; Trace; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; ketones; negative; n= 12
12 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; ketones; 4+; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; ketones; 3+ OR 1 G/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; ketones; 3+; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; ketones; 2+ OR 1/2 G/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; ketones; 2+ ; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; ketones;1+ OR 1/4 G/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; ketones; 1+; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; protein; Trace or 1/10 g/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Protein; Trace; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Protein; negative; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Protein; 4+; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Protein; 3+ OR 1 G/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Protein; 3+; n= 12
8 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Protein; 2+ OR 1/2 G/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Protein; 2+ ; n= 12
3 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Protein;1+ OR 1/4 G/DL; n= 12
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 12; Protein; 1+; n= 12
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; occult blood; Trace or 1/10 g/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28;Occult Blood; Trace; n= 11
2 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Occult Blood; negative; n= 11
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Occult Blood; 4+; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Occult Blood; 3+ OR 1 G/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Occult Blood; 3+; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Occult Blood; 2+ OR 1/2 G/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Occult Blood; 2+ ; n= 11
4 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Occult Blood;1+ OR 1/4 G/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Occult Blood; 1+; n= 11
4 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; glucose; Trace or 1/10 g/DL; n= 11
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28;glucose; Trace; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; glucose; negative; n= 11
9 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; glucose; 4+; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; glucose; 3+ OR 1 G/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; glucose; 3+; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; glucose; 2+ OR 1/2 G/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; glucose; 2+ ; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; glucose;1+ OR 1/4 G/DL; n= 11
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; glucose; 1+; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; ketones; Trace or 1/10 g/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; ketones; Trace; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; ketones; negative; n= 11
11 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; ketones; 4+; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; ketones; 3+ OR 1 G/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; ketones; 3+; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; ketones; 2+ OR 1/2 G/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; ketones; 2+ ; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; ketones;1+ OR 1/4 G/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; ketones; 1+; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; protein; Trace or 1/10 g/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Protein; Trace; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Protein; negative; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; protein; 4+; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Protein; 3+ OR 1 G/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Protein; 3+; n= 11
10 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Protein; 2+ OR 1/2 G/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Protein; 2+ ; n= 11
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Protein;1+ OR 1/4 G/DL; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 28; Protein; 1+; n= 11
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; occult blood; Trace or 1/10 g/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52;Occult Blood; Trace; n= 9
2 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Occult Blood; negative; n= 9
3 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Occult Blood; 4+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Occult Blood; 3+ OR 1 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Occult Blood; 3+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Occult Blood; 2+ OR 1/2 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Occult Blood; 2+ ; n= 9
4 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Occult Blood;1+ OR 1/4 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Occult Blood; 1+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; glucose; Trace or 1/10 g/DL; n= 9
2 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52;glucose; Trace; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; glucose; negative; n= 9
7 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; glucose; 4+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; glucose; 3+ OR 1 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; glucose; 3+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; glucose; 2+ OR 1/2 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; glucose; 2+ ; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; ketones; 2+ ; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; glucose;1+ OR 1/4 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; glucose; 1+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; ketones; Trace or 1/10 g/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; ketones; Trace; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; ketones; negative; n= 9
9 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; ketones; 4+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; ketones; 3+ OR 1 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; ketones; 3+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; ketones; 2+ OR 1/2 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; ketones; 2+ ; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; ketones;1+ OR 1/4 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; ketones; 1+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; ketones;1+ OR 1/4 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; ketones; 1+; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; protein; Trace or 1/10 g/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; protein; Trace or 1/10 g/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Protein; Trace; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Protein; Trace; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Protein; negative; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; protein; 4+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Protein; 3+ OR 1 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Protein; 3+; n= 9
4 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Protein; 2+ OR 1/2 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Protein; 2+ ; n= 9
3 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Protein; negative; n= 7
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Protein;1+ OR 1/4 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; protein; 4+; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 52; Protein; 1+; n= 9
2 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; occult blood; Trace or 1/10 g/DL;n=7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76;Occult Blood; Trace; n= 7
3 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Occult Blood; negative; n= 7
3 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Occult Blood; 4+; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Occult Blood; 3+ OR 1 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Occult Blood; 3+; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Occult Blood; 2+ OR 1/2 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Occult Blood; 2+ ; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Occult Blood;1+ OR 1/4 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Protein; 3+ OR 1 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Occult Blood; 1+; n= 7
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; glucose; Trace or 1/10 g/DL; n= 7
2 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76;glucose; Trace; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; glucose; negative; n= 7
5 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; glucose; 4+; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; glucose; 3+ OR 1 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; glucose; 3+; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; glucose; 2+ OR 1/2 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; glucose; 2+ ; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; glucose;1+ OR 1/4 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; glucose; 1+; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; ketones; Trace or 1/10 g/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; ketones; Trace; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; ketones; negative; n= 7
7 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; ketones; 4+; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; ketones; 3+ OR 1 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; ketones; 3+; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; ketones; 2+ OR 1/2 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Protein; 3+; n= 7
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Protein; 2+ OR 1/2 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Protein; 2+ ; n= 7
4 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Protein;1+ OR 1/4 G/DL; n= 7
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 76; Protein; 1+; n= 7
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; occult blood; Trace or 1/10 g/DL;n=10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104;Occult Blood; Trace; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Occult Blood; negative; n= 10
8 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Occult Blood; 4+; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Occult Blood; 3+ OR 1 G/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Occult Blood; 3+; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Occult Blood; 2+ OR 1/2 G/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Occult Blood; 2+ ; n=10
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Occult Blood;1+ OR 1/4 G/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Occult Blood; 1+; n= 10
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; glucose; Trace or 1/10 g/DL; n= 10
2 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104 ;glucose; Trace; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; glucose; negative; n= 10
7 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; glucose; 4+; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; glucose; 3+ OR 1 G/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; glucose; 3+; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; glucose; 2+ OR 1/2 G/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; glucose; 2+ ; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; glucose;1+ OR 1/4 G/DL; n= 10
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; glucose; 1+; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; ketones; Trace or 1/10 g/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; ketones; Trace; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; ketones; negative; n= 10
10 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; ketones; 4+; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; ketones; 3+ OR 1 G/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; ketones; 3+; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; ketones; 2+ OR 1/2 G/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; ketones; 2+ ; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; ketones;1+ OR 1/4 G/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; ketones; 1+; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; protein; Trace or 1/10 g/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Protein; Trace; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Protein; negative; n= 10
1 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; protein; 4+; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Protein; 3+ OR 1 G/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Protein; 3+; n= 10
3 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Protein; 2+ OR 1/2 G/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Protein; 2+ ; n= 10
6 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Protein;1+ OR 1/4 G/DL; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
Week 104; Protein; 1+; n= 10
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Occult blood; TRACE OR 1/10 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Occult blood; TRACE; n= 9
2 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Occult blood; NEGATIVE; n= 9
6 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Occult blood; 4+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Occult blood; 3+ OR 1 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Occult blood; 3+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Occult blood; 2+ OR 1/2 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Occult blood; 2+; n= 9
1 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Occult blood; 1+ OR 1/4 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Occult blood; 1+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Glucose; TRACE OR 1/10 G/DL; n= 9
1 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Glucose; TRACE; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Glucose; negative; n= 9
7 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Glucose; 4+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Glucose; 3+ OR 1 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Glucose; 3+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Glucose; 2+ OR 1/2 G/DL; n= 9
1 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Glucose; 2+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Glucose; 1+ OR 1/4 G/DL; n=9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Glucose; 1+; n=9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Ketones; TRACE OR 1/10 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Ketones; TRACE; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Ketones; negative; n= 9
9 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Ketones; 4+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Ketones; 3+ OR 1 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Ketones; 3+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Ketones; 2+ OR 1/2 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Ketones; 2+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Ketones; 1+ OR 1/4 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Ketones; 1+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Protein; TRACE OR 1/10 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Protein; TRACE; n= 9
1 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Protein; negative; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Protein; 4+; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Protein; 3+ OR 1 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Protein; 3+; n= 9
3 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Protein; 2+ OR 1/2 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Protein; 2+; n= 9
4 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Protein; 1+ OR 1/4 G/DL; n= 9
0 Participants
Number of Participants With Urinalysis Dipstick Findings
16 WF; Protein; 1+; n= 9
1 Participants

SECONDARY outcome

Timeframe: Baseline and up to week 116/16 week follow-up visit

Population: ITT Population

SBP and DBP were measured from Baseline throughout the treatment period up to Week 116/ 16 week follow-up visit. The Baseline value was taken at Day 0 pre dose and change from Baseline was defined as post dose visit value minus Baseline value. Mean and standard deviation (SD) were measured and presented for Week 12, 28, 52, 76, 104 withdrawn visit, 4 Week post last dose and 16 Week follow-up visit. Only those participants with data available at the specified data points were analyzed (represented by n= X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
Week 12; SBP; n= 12
-4.2 millimeter of mercury (mmHg)
Standard Deviation 17.23
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
Week 28; SBP; n= 11
-5.8 millimeter of mercury (mmHg)
Standard Deviation 18.14
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
Week 52; SBP; n= 9
-1.9 millimeter of mercury (mmHg)
Standard Deviation 18.66
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
Week 76; SBP; n= 8
-1.3 millimeter of mercury (mmHg)
Standard Deviation 11.30
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
4 Week post last dose; SBP; n= 10
-12.2 millimeter of mercury (mmHg)
Standard Deviation 11.78
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
16 Week follow up; SBP; n= 9
-7.4 millimeter of mercury (mmHg)
Standard Deviation 15.72
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
Week 104 withdrawn visit; SBP; n= 2
-1.0 millimeter of mercury (mmHg)
Standard Deviation 5.66
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
Week 12; DBP; n= 12
3.8 millimeter of mercury (mmHg)
Standard Deviation 12.94
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
Week 28; DBP; n= 11
0.8 millimeter of mercury (mmHg)
Standard Deviation 10.75
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
Week 52; DBP; n= 9
1.2 millimeter of mercury (mmHg)
Standard Deviation 11.87
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
Week 76; DBP; n= 8
2.8 millimeter of mercury (mmHg)
Standard Deviation 9.68
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
4 Week post last dose; DBP; n= 10
-3.3 millimeter of mercury (mmHg)
Standard Deviation 9.87
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
16 Week follow up; DBP; n= 9
2.4 millimeter of mercury (mmHg)
Standard Deviation 14.98
Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
Week 104 withdrawn visit; DBP; n= 2
5.5 millimeter of mercury (mmHg)
Standard Deviation 2.12

SECONDARY outcome

Timeframe: Baseline and up to Week 116/16 week follow-up visit

Population: ITT Population

Pulse rate was measured from Baseline throughout the treatment period up to Week 116/ 16 week follow-up visit. The Baseline value was taken at Day 0 pre dose and change from Baseline was defined as post dose visit value minus Baseline value. Mean and standard deviation (SD) were measured and presented for Week 12, 28, 52, 76, 104 withdrawn visit, 4 Week post last dose and 16 Week post last dose visits. Only those participants with data available at the specified data points were analyzed (represented by n= X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Pulse Rate
Week 12; n= 12
-0.7 Beats per minute
Standard Deviation 11.40
Change From Baseline in Pulse Rate
Week 28; n= 11
1.0 Beats per minute
Standard Deviation 7.90
Change From Baseline in Pulse Rate
Week 52; n= 9
-1.7 Beats per minute
Standard Deviation 13.11
Change From Baseline in Pulse Rate
Week 76; n= 8
-2.3 Beats per minute
Standard Deviation 15.77
Change From Baseline in Pulse Rate
4 Week post last dose; n= 10
-2.8 Beats per minute
Standard Deviation 15.33
Change From Baseline in Pulse Rate
16 Week follow up; n= 9
-0.9 Beats per minute
Standard Deviation 16.72
Change From Baseline in Pulse Rate
Week 104 withdrawn visit; n= 2
5.0 Beats per minute
Standard Deviation 7.07

SECONDARY outcome

Timeframe: Baseline and up to Week 116/16 week follow-up visit

Population: ITT Population

Temperature was measured from Baseline throughout the treatment period up to Week 116/ 16 week follow-up visit. The Baseline value was taken at Day 0 pre dose and change from Baseline was defined as post dose visit value minus Baseline value. Mean and standard deviation (SD) were measured and presented for Week 12, 28, 52, 76, 104 withdrawn visit, 4 Week post last dose and 16 week post last dose visits. Only those participants with data available at the specified data points were analyzed (represented by n= X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in Temperature
Week 12; n= 12
-0.09 Celsius
Standard Deviation 0.591
Change From Baseline in Temperature
Week 28; n= 11
-0.00 Celsius
Standard Deviation 0.453
Change From Baseline in Temperature
Week 52; n= 9
-0.11 Celsius
Standard Deviation 0.639
Change From Baseline in Temperature
Week 76; n= 8
-0.17 Celsius
Standard Deviation 0.486
Change From Baseline in Temperature
4 Week post last dose; n= 8
-0.06 Celsius
Standard Deviation 0.604
Change From Baseline in Temperature
16 Week follow up; n= 6
0.30 Celsius
Standard Deviation 0.424
Change From Baseline in Temperature
Week 104 withdrawn visit; n= 2
0.15 Celsius
Standard Deviation 0.353

SECONDARY outcome

Timeframe: Baseline and up to Week 116/16 week follow-up visit

Population: ITT Population

Blood samples of participants were collected pre-dose on Weeks 0, 12, 28, 40, 52, 76, 4 week post last dose and 16 week post last dose visit for belimumab immunogenicity assay. No participants showed positive immunogenicity findings.

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Number of Participants With Positive Immunogenicity Findings
0 Participants

SECONDARY outcome

Timeframe: Baseline and up to 4 week post last dose

Population: ITT Population

Urine membrane attack complex was assayed quantitatively by ELISA method. Urine MAC samples were collected at Day 0 and Weeks 8, 28, 52, 76 and 4 week post last dose. Results were normalized using urine creatinine concentration to adjust for urine dilution. Endpoint was moved to 'Exploratory' in Protocol amendment 5 as risk of availability of functioning assay for urine membrane attack complex was noted. No assay was subsequently found and samples were not analyzed

Outcome measures

Outcome data not reported

SECONDARY outcome

Timeframe: Baseline and up to 4 week post last dose

Population: ITT Population

Urine membrane attack complex will be assayed quantitatively by ELISA method. Urine MAC samples are being collected at Day 0 and Weeks 8, 28, 52, 76 and 4 week post last dose. Results will be normalized using urine creatinine concentration to adjust for urine dilution, before calculation of the ratio as value at time point divided by value at Baseline (Day 0). Endpoint was changed to 'exploratory' as risk of availability of functioning assay for urine membrane attack complex was noted. No assay was subsequently found and samples were not analyzed.

Outcome measures

Outcome data not reported

SECONDARY outcome

Timeframe: Baseline and up to Week 128/6 month post last dose

Population: ITT Population

B cell Facs panels were used to measure changes over the course of therapy in B cell subsets such as transitional, naïve, memory and plasma B cell compartments by percent of the B cell compartments and absolute numbers. T cell Facs panel were used to measure changes in T cell subsets, such as T regs and CD4+ and CD8+ T cells, in terms of numbers and expression of activation markers to establish if B cell targeting with belimumab affects the T cell compartment perhaps through limiting B cell antigen presentation or cytokine release. Baseline is defined as Day 0 value and change from Baseline was calculated as ratio of post-Baseline value divided by the Baseline value. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles).

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+; Week 8; n= 12
0.7221 Ratio
Geometric Coefficient of Variation 82.1
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+; Week 16; n= 11
0.9209 Ratio
Geometric Coefficient of Variation 63.6
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+; Week 28; n= 10
1.0049 Ratio
Geometric Coefficient of Variation 71.6
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+; Week 104; n= 10
0.5193 Ratio
Geometric Coefficient of Variation 156.2
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+; 6 month follow up; n= 7
0.3828 Ratio
Geometric Coefficient of Variation 185.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD24b+ CD38b+ CD27-; Week 8; n= 12
0.2352 Ratio
Geometric Coefficient of Variation 340.0
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD24b+ CD38b+ CD27-; Week 16; n= 11
0.6228 Ratio
Geometric Coefficient of Variation 316.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD24b+ CD38b+ CD27-; Week 28; n= 10
0.6119 Ratio
Geometric Coefficient of Variation 656.0
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD24b+ CD38b+ CD27-; Week 104; n= 10
0.3582 Ratio
Geometric Coefficient of Variation 405.1
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD24b+ CD38b+ CD27-; 6 month follow up; n= 7
1.6471 Ratio
Geometric Coefficient of Variation 534.0
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD27- IgD+; Week 8; n= 12
0.3490 Ratio
Geometric Coefficient of Variation 131.6
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD27- IgD+; Week 16; n= 11
0.4486 Ratio
Geometric Coefficient of Variation 94.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD27- IgD+; Week 28; n= 10
0.4561 Ratio
Geometric Coefficient of Variation 82.5
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD27- IgD+; Week 104; n= 10
0.1808 Ratio
Geometric Coefficient of Variation 298.6
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD27- IgD+; 6 month follow up; n= 7
0.3278 Ratio
Geometric Coefficient of Variation 189.2
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD27-; Week 8; n= 12
0.4074 Ratio
Geometric Coefficient of Variation 109.2
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD27-; Week 16; n= 11
0.5079 Ratio
Geometric Coefficient of Variation 88.1
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD27-; Week 28; n= 10
0.5165 Ratio
Geometric Coefficient of Variation 76.5
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD27-; Week 104; n= 10
0.2602 Ratio
Geometric Coefficient of Variation 236.9
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD27-; 6 month follow up; n= 7
0.3756 Ratio
Geometric Coefficient of Variation 186.0
Change From Baseline in B Cell and T Cell Markers Concentration
CD19lo CD38hi CD27hi; Week 8; n= 12
0.5982 Ratio
Geometric Coefficient of Variation 555.4
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+; Week 28; n= 10
3.1352 Ratio
Geometric Coefficient of Variation 65.6
Change From Baseline in B Cell and T Cell Markers Concentration
CD19lo CD38hi CD27hi; Week 16; n= 11
0.4128 Ratio
Geometric Coefficient of Variation 1085.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD19lo CD38hi CD27hi; Week 28; n= 10
0.4481 Ratio
Geometric Coefficient of Variation 651.6
Change From Baseline in B Cell and T Cell Markers Concentration
CD19lo CD38hi CD27hi; Week 104; n= 10
0.2615 Ratio
Geometric Coefficient of Variation 238.8
Change From Baseline in B Cell and T Cell Markers Concentration
CD19lo CD38hi CD27hi; 6 month follow up; n= 7
0.0647 Ratio
Geometric Coefficient of Variation 1269.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD24+ CD27+ ; Week 8; n= 12
2.0747 Ratio
Geometric Coefficient of Variation 65.3
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD24+ CD27+ ; Week 16; n= 11
2.4161 Ratio
Geometric Coefficient of Variation 61.2
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD24+ CD27+ ; Week 28; n= 10
3.1294 Ratio
Geometric Coefficient of Variation 64.2
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD24+ CD27+ ; Week 104; n= 10
0.6089 Ratio
Geometric Coefficient of Variation 104014.8
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+ CD24+ CD27+ ; 6 month follow up; n= 7
0.7952 Ratio
Geometric Coefficient of Variation 328.6
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+; Week 8; n= 12
1.9195 Ratio
Geometric Coefficient of Variation 69.9
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+; Week 16; n= 11
2.3132 Ratio
Geometric Coefficient of Variation 62.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+; Week 104; n= 10
1.3273 Ratio
Geometric Coefficient of Variation 127.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+; 6 month follow up; n= 7
0.3338 Ratio
Geometric Coefficient of Variation 158.2
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+IgD; Week 8; n= 12
1.9365 Ratio
Geometric Coefficient of Variation 69.6
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+IgD; Week 16; n= 11
2.3147 Ratio
Geometric Coefficient of Variation 75.1
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+IgD; Week 28; n= 10
3.2313 Ratio
Geometric Coefficient of Variation 51.1
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+IgD; Week 104; n= 10
1.2814 Ratio
Geometric Coefficient of Variation 102.3
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+IgD; 6 month follow up; n= 7
0.2413 Ratio
Geometric Coefficient of Variation 163.9
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+IgD-; Week 8; n= 12
1.8846 Ratio
Geometric Coefficient of Variation 67.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+IgD-; Week 16; n= 11
2.2113 Ratio
Geometric Coefficient of Variation 70.4
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+IgD-; Week 28; n= 10
3.0478 Ratio
Geometric Coefficient of Variation 73.2
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+IgD-; Week 104; n= 10
1.3051 Ratio
Geometric Coefficient of Variation 147.1
Change From Baseline in B Cell and T Cell Markers Concentration
CD19+CD27+IgD-; 6 month follow up; n= 7
0.3440 Ratio
Geometric Coefficient of Variation 163.8
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi CD45RA- IL7Rhi; Week 8; n= 12
0.7010 Ratio
Geometric Coefficient of Variation 86.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi CD45RA- IL7Rhi; Week 16; n= 10
0.5340 Ratio
Geometric Coefficient of Variation 221.3
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi CD45RA- IL7Rhi; Week 28; n= 9
0.5718 Ratio
Geometric Coefficient of Variation 669.2
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi CD45RA- IL7Rhi; Week 104; n= 10
4.1865 Ratio
Geometric Coefficient of Variation 509.4
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi CD45RA- IL7Rh; 6 month follow up; n= 7
1.8067 Ratio
Geometric Coefficient of Variation 1456.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD45RA- IL-7Rhi; Week 8; n= 12
1.4174 Ratio
Geometric Coefficient of Variation 19513.4
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD45RA- IL-7Rhi; Week 16; n= 10
1.8613 Ratio
Geometric Coefficient of Variation 99527.6
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD45RA- IL-7Rhi; Week 28; n=9
2.8378 Ratio
Geometric Coefficient of Variation 49940.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD45RA- IL-7Rhi; Week 104; n= 10
0.1517 Ratio
Geometric Coefficient of Variation 33534.3
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD45RA- IL-7Rhi; 6 month follow-up; n= 7
0.3532 Ratio
Geometric Coefficient of Variation 966.4
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi xIL-7Rlo; Week 8; n= 12
0.7285 Ratio
Geometric Coefficient of Variation 85.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi xIL-7Rlo; Week 16; n= 11
0.6232 Ratio
Geometric Coefficient of Variation 317.6
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi xIL-7Rlo; Week 28; n= 10
0.7010 Ratio
Geometric Coefficient of Variation 961.5
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi xIL-7Rlo; Week 104; n= 10
3.1025 Ratio
Geometric Coefficient of Variation 932.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi xIL-7Rlo; 6 month follow-up; n= 7
2.0827 Ratio
Geometric Coefficient of Variation 832.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi IL-7Rlo; Week 8; 12
2.3001 Ratio
Geometric Coefficient of Variation 29702.5
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi IL-7Rlo; Week 16; n= 10
2.9023 Ratio
Geometric Coefficient of Variation 136702.6
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi IL-7Rlo; Week 28; n= 9
3.3453 Ratio
Geometric Coefficient of Variation 35372.4
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi IL-7Rlo; Week 104; n= 10
0.3410 Ratio
Geometric Coefficient of Variation 72506.2
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD25hi IL-7Rlo; 6 month follow up; n= 7
0.9978 Ratio
Geometric Coefficient of Variation 32.7
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD45RA-; Week 8; n= 12
0.7532 Ratio
Geometric Coefficient of Variation 44.5
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD45RA-; Week 16; n= 11
0.7638 Ratio
Geometric Coefficient of Variation 55.4
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD45RA-; Week 28; n= 10
0.9032 Ratio
Geometric Coefficient of Variation 71.1
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD45RA-; 104; n= 10
0.9750 Ratio
Geometric Coefficient of Variation 95.8
Change From Baseline in B Cell and T Cell Markers Concentration
CD4+ CD45RA-; 6 month follow up; n= 7
1.0792 Ratio
Geometric Coefficient of Variation 90.0

SECONDARY outcome

Timeframe: Baseline and up to Week 104/4 week post last dose

Population: ITT Population

Cytokine/chemokine associated with T helper skewing or autoimmune pathology will be analyzed using Luminex, ELISA. Serum analyte quantification were used to confirm altered protein levels of any gene expression increases or decreases identified by transcriptomic analysis. Endpoint was moved to 'Exploratory' in Protocol amendment 5 as benefits of assessing cytokines was deemed low. Samples were not analyzed.

Outcome measures

Outcome data not reported

SECONDARY outcome

Timeframe: Baseline and Week 116/16 week follow-up visit

Population: ITT Population

Free BLyS protein were analyzed using an ELISA. Serum samples were collected before treatment and after belimumab washout at Week 0 and Week 116/16 week follow-up visit.

Outcome measures

Outcome measures
Measure
Belimumab 10 mg/kg IV
n=14 Participants
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Serum BLys Levels
Day 0; n= 14
969.9595 pg/mL
Geometric Coefficient of Variation 16.8
Serum BLys Levels
16 week follow up; n= 8
12357.5558 pg/mL
Geometric Coefficient of Variation 123.3

SECONDARY outcome

Timeframe: Baseline and Week 116/16 week follow-up visit

Population: ITT Population

B lymphocyte stimulator (BLyS) normalized by creatinine as a ratio of BLyS: creatinine. Free BLyS protein is being analyzed using an ELISA. Urine samples are being collected before treatment and after belimumab washout at Week 0 and Week 116/16 week follow-up visit. Only raw BLyS values available and unable to be assessed due to lack of comparison to a creatinine as a urine concentration marker.

Outcome measures

Outcome data not reported

Adverse Events

Belimumab 10 mg/kg IV

Serious events: 3 serious events
Other events: 14 other events
Deaths: 0 deaths

Serious adverse events

Serious adverse events
Measure
Belimumab 10 mg/kg IV
n=14 participants at risk
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Infections and infestations
Cellulitis
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Investigations
Weight decreased
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Vascular disorders
Embolism
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.

Other adverse events

Other adverse events
Measure
Belimumab 10 mg/kg IV
n=14 participants at risk
Participants received 10 milligrams per kilogram (mg/kg) belimumab intravenous (IV) infusion at Week 0 Week 2 and Week 4, then every 4 weeks over 24 weeks. Participants then entered the long-term phase of the study and received belimumab 10 mg/kg every 4 weeks up to Week 100 or until complete remission had been achieved.
Infections and infestations
Upper respiratory tract infection
42.9%
6/14 • Number of events 10 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Infections and infestations
Cellulitis
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Infections and infestations
Lower respiratory tract infection
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Infections and infestations
Oral candidiasis
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Infections and infestations
Respiratory tract infection
7.1%
1/14 • Number of events 4 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Infections and infestations
Rhinitis
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Infections and infestations
Urinary tract infection
7.1%
1/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Infections and infestations
Viral upper respiratory tract infection
35.7%
5/14 • Number of events 5 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Diarrhoea
14.3%
2/14 • Number of events 3 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Abdominal pain
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Abdominal pain upper
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Ascites
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Dyspepsia
14.3%
2/14 • Number of events 3 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Irritable bowel syndrome
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Nausea
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Musculoskeletal and connective tissue disorders
Muscle spasms
28.6%
4/14 • Number of events 12 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Musculoskeletal and connective tissue disorders
Arthralgia
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Musculoskeletal and connective tissue disorders
Flank pain
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Musculoskeletal and connective tissue disorders
Joint swelling
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Musculoskeletal and connective tissue disorders
Myalgia
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Musculoskeletal and connective tissue disorders
Pain in extremity
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Nervous system disorders
Headache
14.3%
2/14 • Number of events 7 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Nervous system disorders
Dizziness
14.3%
2/14 • Number of events 3 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Nervous system disorders
Migraine
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Nervous system disorders
Presyncope
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Skin and subcutaneous tissue disorders
Rash
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Skin and subcutaneous tissue disorders
Rash pruritic
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Skin and subcutaneous tissue disorders
Skin lesion
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Skin and subcutaneous tissue disorders
Swelling face
7.1%
1/14 • Number of events 3 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
General disorders
Chest pain
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
General disorders
Malaise
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
General disorders
Pyrexia
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Respiratory, thoracic and mediastinal disorders
Cough
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Respiratory, thoracic and mediastinal disorders
Oropharyngeal pain
14.3%
2/14 • Number of events 3 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Ear and labyrinth disorders
Tinnitus
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Eye disorders
Periorbital oedema
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Investigations
Eosinophil count increased
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Metabolism and nutrition disorders
Decreased appetite
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Psychiatric disorders
Depression
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Renal and urinary disorders
Tubulointerstitial nephritis
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Infections and infestations
Viral infection
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Infections and infestations
Influenza
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Infections and infestations
Labyrinthitis
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Infections and infestations
Lower respiratory tract infection viral
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Rectal haemorrhage
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Vomiting
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Gingival bleeding
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Haemorrhoids
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Melaena
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Gastrointestinal disorders
Toothache
7.1%
1/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Nervous system disorders
Hypoaesthesia
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Nervous system disorders
Peripheral sensory neuropathy
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Nervous system disorders
Sciatica
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Musculoskeletal and connective tissue disorders
Back pain
14.3%
2/14 • Number of events 4 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Musculoskeletal and connective tissue disorders
Musculoskeletal discomfort
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Musculoskeletal and connective tissue disorders
Neck pain
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Musculoskeletal and connective tissue disorders
Tendonitis
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Skin and subcutaneous tissue disorders
Angioedema
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
General disorders
Chest discomfort
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Respiratory, thoracic and mediastinal disorders
Epistaxis
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Respiratory, thoracic and mediastinal disorders
Productive cough
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Psychiatric disorders
Insomnia
14.3%
2/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Cardiac disorders
Angina pectoris
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Cardiac disorders
Palpitations
7.1%
1/14 • Number of events 2 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Investigations
Blood parathyroid hormone increased
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Renal and urinary disorders
Costovertebral angle tenderness
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Immune system disorders
Seasonal allergy
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Injury, poisoning and procedural complications
Limb injury
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Neoplasms benign, malignant and unspecified (incl cysts and polyps)
Basal cell carcinoma
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Neoplasms benign, malignant and unspecified (incl cysts and polyps)
Skin papilloma
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.
Vascular disorders
Hypertension
7.1%
1/14 • Number of events 1 • On-therapy serious adverse events (SAEs) and non-serious adverse events (AEs) are presented from the start of study treatment up to end of study.
On-therapy SAEs and non-serious AEs are reported for members of the ITT Population, comprised of all participants who received at least one dose of investigational drug.

Additional Information

GSK Response Center

GlaxoSmithKline

Phone: 866-435-7343

Results disclosure agreements

  • Principal investigator is a sponsor employee GSK agreements may vary with individual investigators, but will not prohibit any investigator from publishing. GSK supports the publication of results from all centers of a multi-center trial but requests that reports based on single-site data not precede the primary publication of the entire clinical trial.
  • Publication restrictions are in place

Restriction type: OTHER