throbber
CLINICAL—ALIMENTARY TRACT
`
`GASTROENTEROLOGY 2012;143:1473–1481
`
`CLINICALAT
`
`Teduglutide Reduces Need for Parenteral Support Among Patients With
`Short Bowel Syndrome With Intestinal Failure
`PALLE B. JEPPESEN,* MAREK PERTKIEWICZ,‡ BERNARD MESSING,§ KISHORE IYER,储 DOUGLAS L. SEIDNER,¶
`STEPHEN J. D. O’KEEFE,# ALASTAIR FORBES,** HARTMUT HEINZE,‡‡ and BO JOELSSON§§
`
`*Department of Medical Gastroenterology, Rigshospitalet, Copenhagen, Denmark; ‡Department of General Surgery and Clinical Nutrition, Warszawa, Poland; §Hopital
`Beaujon Service de Gastroenterologie et Assistance Nutritive, Clichy, France; 储Mount Sinai Medical Center, New York, New York; ¶Vanderbilt University Medical
`Center, Nashville, Tennessee; #University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; **University College London, London, UK; ‡‡Nycomed, a Takeda
`Company, Nycomed GmbH, Konstanz, Germany; and §§NPS Pharmaceuticals, Bedminster, New Jersey
`
`See Covering the Cover synopsis on page
`1403; see editorial on page 1416.
`
`BACKGROUND & AIMS: Teduglutide, a glucagon-like
`peptide 2 analogue, might restore intestinal structural
`and functional integrity by promoting growth of the mu-
`cosa and reducing gastric emptying and secretion. These
`factors could increase fluid and nutrient absorption in
`patients with short bowel syndrome with intestinal failure
`(SBS-IF). We performed a prospective study to determine
`whether teduglutide reduces parenteral support in pa-
`tients with SBS-IF. METHODS: We performed a 24-week
`study of patients with SBS-IF who were given subcutane-
`ous teduglutide (0.05 mg/kg/d; n ⫽ 43) or placebo (n ⫽
`43) once daily. Parenteral support was reduced if 48-hour
`urine volumes exceeded baseline values by ⱖ10%. The
`primary efficacy end point was number of responders
`(patients with ⬎20% reduction in parenteral support vol-
`ume from baseline at weeks 20 and 24). RESULTS: There
`were significantly more responders in the teduglutide
`group (27/43 [63%]) than the placebo group (13/43 [30%];
`P ⫽ .002). At week 24, the mean reduction in parenteral
`support volume in the teduglutide group was 4.4 ⫾ 3.8
`L/wk (baseline 12.9 ⫾ 7.8 L/wk) compared with 2.3 ⫾ 2.7
`L/wk (baseline 13.2 ⫾ 7.4 L/wk) in the placebo group
`(P ⬍ .001). The percentage of patients with a 1-day or
`more reduction in the weekly need for parenteral support
`was greater in the teduglutide group (21/39 [54%]) than in
`the placebo group (9/39 [23%]; P ⫽ .005). Teduglutide
`increased plasma concentrations of citrulline, a biomarker
`of mucosal mass. The distribution of treatment-emergent
`adverse events that led to study discontinuation was sim-
`ilar between patients given teduglutide (n ⫽ 2) and pla-
`cebo (n ⫽ 3). CONCLUSIONS: Twenty-four weeks of
`teduglutide treatment was generally well tolerated in
`patients with SBS-IF. Treatment with teduglutide re-
`duced volumes and numbers of days of parenteral
`support for patients with SBS-IF;
`
`Scan the quick response (QR) code to the left with
`your mobile device to watch this article’s video ab-
`stract and others. Don’t have a QR code reader? Get
`one by searching ‘QR Scanner’ in your mobile de-
`vice’s app store.
`
`Short bowel syndrome (SBS) results from surgical re-
`
`section, congenital defect, or disease associated loss of
`absorption. The concomitant malabsorptive spectrum of
`SBS is wide, and patients with SBS are heterogeneous
`because of large variations in remnant bowel anatomy and
`function. Patients with intestinal insufficiency are able to
`compensate for their malabsorption by physiologic or
`pharmacologic adaptation,1,2 whereas supplemental par-
`enteral support (PS; parenteral nutrition and/or intrave-
`nous [PN/IV] fluids) is required to maintain fluid, elec-
`trolytes, trace elements, vitamins, and nutrient balances in
`patients with SBS with intestinal failure (SBS-IF).3,4 Treat-
`ments aim to maximize remnant intestinal absorptive
`capacity; to minimize the symptoms of malabsorption;
`and to avoid, minimize, or eliminate the need for PS,
`thereby alleviating the daily burden of this debilitating
`condition. Hormonal therapies focusing on enhancing
`the structural and functional integrity of the remaining
`intestine are emerging. Glucagon-like peptide 2 (GLP-2), a
`peptide secreted from the intestinal L cells after food
`ingestion, ameliorates the pathophysiologic consequences
`of SBS. GLP-2 administration inhibits gastric acid secre-
`
`Abbreviations used in this paper: AE, adverse event; FCE, fluid com-
`posite effect; GLP-2, glucagon-like peptide 2;
`IF,
`intestinal failure;
`PN/IV, parenteral nutrition and/or intravenous; PS, parenteral support;
`SBS, short bowel syndrome; TEAE, treatment-emergent adverse event.
`© 2012 by the AGA Institute
`0016-5085/$36.00
`
`Keywords: Clinical Trial; Glucagon-Like Peptide 2; Gastro-
`intestinal Disorder; Therapy.
`
`Watch this article’s video abstract and others at http://
`tiny.cc/j026c.
`
`Page 1
`
`

`
`1474 JEPPESEN ET AL
`
`GASTROENTEROLOGY Vol. 143, No. 6
`
`Table 1. Key Inclusion and Exclusion Criteria
`
`Inclusion criteria
`
`Exclusion criteria
`
`SBS resulting from intestinal failure caused by a major intestinal
`resection (eg, injury, cancer, Crohn’s disease, vascular disease,
`volvulus)
`At least 12 continuous months of PS dependency (PN and/or IV fluids)
`before signing informed consent
`PS required ⱖ3 times weekly to meet caloric, fluid, or electrolyte needs
`Patients with Crohn’s disease had to be in clinical remission for ⱖ12
`wk before dosing
`
`Cancer within last 5 y
`Body mass index ⬍15 kg/m2
`Inflammatory bowel disease on immunosuppressant therapy that
`has been introduced or changed within last 3 mo or treatment
`with biologics within last 6 mo
`Previous use of teduglutide
`Previous use of native GLP-2 or human growth hormone within 6
`mo before screening
`⬎4 SBS-related hospital admissions within 12 mo or hospital
`admission within 30 d before screening
`
`CLINICAL AT
`
`tion and motility,5,6 stimulates intestinal blood flow,7
`increases intestinal barrier function,8 and enhances nutri-
`ent and fluid absorption in preclinical and clinical mod-
`els.9-12
`Teduglutide, a dipeptidyl-peptidase degradation-resis-
`tant GLP-2 analogue, has been demonstrated to enhance
`structural and functional integrity of the remaining intes-
`tine in SBS. Open-label, uncontrolled studies in adult
`patients with SBS have suggested clinically meaningful
`reductions of fecal excretions of wet weight (⬃700 to
`1000 g/d) and energy (⬃1 MJ/d) after treatment with
`GLP-2 and teduglutide.10,11,13,14 A recently published, ran-
`domized, placebo-controlled phase 3 study investigated
`whether teduglutide, by increasing intestinal absorption,
`could facilitate PS reductions in patients with SBS-IF.12
`Contrary to the expectations of a dose response, a 0.10-
`mg/kg/d dosage did not meet the primary end point of PS
`reduction, but significant findings from the ad hoc anal-
`ysis of a 0.05-mg/kg/d dosage in that study suggested that
`these differences could be explained by the limitation of
`PS volume reductions to no more than 10% of baseline
`levels, beginning only at the fourth week of dosing, along
`with a trend toward larger baseline PS volume require-
`ments in the 0.10-mg/kg/d group. Therefore, the primary
`objective of this study, the largest double-blind, random-
`ized, placebo-controlled trial performed in patients with
`SBS-IF, was to evaluate whether teduglutide at the 0.05-
`mg/kg/d dosage and with a protocol allowing for earlier
`(ie, at second week of dosing) and more aggressive PS
`reductions of 10% to 30% of baseline levels of PN/IV fluid
`could reduce PS volume in these patients.
`
`Materials and Methods
`All authors had access to the study data and have re-
`viewed and approved the final manuscript.
`
`Patients
`After receiving approval from local institutional review
`boards or medical ethics committees, centers screened patients
`of both sexes who were 18 years of age or older and who had a
`history of SBS that resulted in a dependency on PS for a period
`of at least 12 months before the start of the study. PS depen-
`dency was defined as at least 12 continuous months of PS
`required at least 3 times weekly to meet their caloric, fluid, or
`electrolyte needs because of ongoing malabsorption. The proto-
`
`col did not specify whether previous attempts at weaning had to
`be made.
`. Al-
`Inclusion and exclusion criteria are listed in
`though patients with neoplasms could be included in the study,
`patients with ongoing radiation enteritis or the presence of
`damaged enteral tissue due to radiation enteritis were excluded,
`along with any condition or circumstance that, in the investiga-
`tor’s opinion, put the patient at undue risk or jeopardized the
`integrity of the study results, including the presence of any of
`the excluded disease states described in the Supplementary Ma-
`terials (Supplementary Table 1).
`Patients were not categorized by whether they were receiving
`parenteral nutrition vs intravenous fluids alone. Throughout the
`study, patients were requested to maintain habitual diet and
`fluids, and no new medications were started or ongoing treat-
`ments changed during the stabilization period or throughout
`the 24-week treatment period unless deemed medically neces-
`sary. Patients who completed the 24-week treatment period were
`offered entry into an open-label extension study, the results of
`which will be the subject of a separate report.
`
`Study Design
`In this multinational, multicenter, randomized, double-
`blind, placebo-controlled, parallel-group, 2-stage, phase 3 study
`(Figure 1), patients were recruited from 27 sites in 10 countries
`across Europe and North America. Stage 1 consisted of a screen-
`ing visit and optimization and stabilization periods. After
`screening, eligible patients underwent a PS optimization period,
`if needed, of up to 8 weeks to achieve a stable target urine output
`of 1.0 to 2.0 L/d. This range, at the higher end of normal output
`(⬃1.5 L/d), was considered to minimize the risk for dehydration-
`related complications (eg, renal calculi) without provoking hy-
`perhydration in a population that is prone to diarrhea and
`
`Figure 1. Study design.
`
`Page 2
`
`

`
`CLINICALAT
`
`December 2012
`
`TEDUGLUTIDE AND SBS 1475
`
`Table 2. PN/IV Adjustments Based on 48-Hour Urinary Output
`
`Urine outputa
`
`PN/IV action
`
`⬍1.0 L/d or target based on stabilized urine output
`ⱖ1.0 L/d but less than baseline
`0% to ⬍10% increase over baseline
`ⱖ10% increase over baseline
`
`Increase PN/IV by ⱖ10% (wk 2) or to previous level
`If patient is dehydrated or inadequately nourished, increase PS; if not, maintain PS
`Maintain PS
`Reduce PS by ⱖ10% of stabilized baseline level up to a clinically appropriate
`amount (maximum of 30%)
`
`aBaseline urine output is the urine volume obtained during the stabilization period before initiating treatment.
`
`dehydration. All patients then underwent a 4- to 8-week stabi-
`lization period during which PS usage was to match prescribed
`PS, and oral fluid intake and urine volume could not deviate
`⬎25% from the optimized levels. Although the osmolarity and
`oral intake were not strictly controlled, patients were asked to
`keep intake as constant as possible. Patients who were not stable
`could repeat stage 1 once.
`Stage 2 began when the patients demonstrated PS volume
`stability. Eighty-six patients were randomized in a 1:1 ratio to
`placebo or teduglutide 0.05 mg/kg/d (administered once daily
`subcutaneously into the abdomen, thigh, or arm, at approxi-
`mately the same time each day) for 24 weeks (Figure 1). Ran-
`domization was performed according to a computer-generated
`interactive response system and was stratified at 2 levels of
`baseline PS volume (ⱕ6 or ⬎6 L/wk). The postrandomization
`study evaluations and visits were scheduled at weeks 1, 2, 4, 8,
`12, 16, 20, and 24.
`All patients were required to record PS volume, 48-hour oral
`fluid intake and urinary output, and study drug dosing infor-
`mation in an electronic diary. PS volume was recorded daily and
`48-hour oral fluid intake and urinary output was recorded dur-
`ing the optimization and stabilization periods and at weeks 2, 4,
`8, 12, 16, 20, and 24 during the treatment period. If there was a
`change in oral intake, the clinician considered whether to adjust
`PS volume. Attempts to reduce PS volume were made at every
`visit before week 24.
`
`Optimization and Stabilization
`During the optimization period, patients were assessed
`at planned intervals (weeks 2, 4, 6, and 8, ⫾3 days) for hydration
`and nutrition. PS was adjusted in targeted increments of ⱖ10%
`of the volume at the previous visit. Immediately before each
`scheduled visit, 48-hour oral fluid intake and urine output were
`measured. The measurement included 1 day on and 1 day off PS,
`unless the PS was infused daily. Blood and urine samples were
`collected at each visit to evaluate hydration and nutrition. A
`targeted urine output of 1.0 to 2.0 L/d was used to determine if
`patients required optimization or could enter the stabilization
`period.
`Stability was defined as actual PS usage matching the pre-
`scribed PS, baseline 48-hour oral fluid intake and urine output
`volumes within ⫾25% of the respective 48-hour volumes, and
`urine output volume of 2 to 4 L per 48 hours. No further PS
`adjustments were permitted during the stabilization period.
`The purpose of the PS optimization period was to ensure that
`all patients received and tolerated a stable minimal level of PS
`before treatment, with adequate hydration as indicated by urine
`output. Patients who failed to remain stable for at least 4
`consecutive weeks immediately before randomization were to
`start the optimization period again. Those patients who failed to
`stabilize after 2 attempts could not proceed and were not ran-
`domized.
`
`Efficacy and Safety
`During the treatment period, PS adjustments were
`targeted to be ⱖ10% but ⬍30% of stabilized PS level. Patients
`were required to remain compliant with the prescribed PS
`throughout the study, with all adjustments based on the
`actual PS volume infused. Patients were assessed at planned
`intervals (baseline and weeks 2, 4, 8, 12, 16, and 20) for
`hydration and nutrition. Before all scheduled visits, 48-hour
`oral fluid intake and urinary output measurements were
`taken and included 1 day on and 1 day off PS, unless the PS
`was infused daily.
`Reductions in PS volumes by 10% to 30% of baseline PS levels
`were allowed if the 48-hour urinary volumes exceeded the base-
`line values by ⬎10%. Oral intake during these 48-hour balances
`was to be constant. Determination of the amount of PS volume
`reduction was based on 48-hour urinary output, according to
`the algorithm described in
`. The decision of whether to
`stop a day of PS, reduce the percentage volume of all days that
`PS was administered, or change the relative PN/IV constituents
`of the PS or whether total PS weaning was possible was based on
`the investigator’s clinical judgment and the personal preference
`of the patient.
`Interim safety evaluations 1 week after PS reductions ensured
`that PS reductions were well tolerated. This assessment of nour-
`ishment and hydrational status was based on repeated 48-hour
`urine collections and a clinical evaluation that included clinical
`signs and symptoms of dehydration, change in body weight,
`reviews of the recorded oral fluid intake, blood samples (hemat-
`ocrit, creatinine, blood urea nitrogen), and urine sodium. Be-
`cause the injection site reactions or stomal changes that are
`known to occur with GLP-2 and teduglutide might have un-
`blinded the observer, the clinician assessing and adjusting PS
`volume was required to be different from the one conducting
`the physical examination and assessing safety. If the reduced PS
`volume was well tolerated, the new weekly PS volume was main-
`tained until the next visit; if not, the previously tolerated PS
`volume was resumed. Patients could be rechallenged at the next
`visit if adequate hydration and nutrition requirements were met.
`The primary efficacy end point was the percentage of patients
`who demonstrated a response at week 20 and maintained that
`response at week 24 (responder). A response at a given visit was
`defined as the achievement of a 20% to 100% reduction from
`baseline in weekly PS volume. The secondary efficacy end points
`included the percentage and absolute change in PS and the
`number of patients who stopped PS and their time of discon-
`tinuation.12
`Exploratory end points included response by visit, reduction
`in days on PS, change from baseline in plasma concentrations of
`citrulline (an amino acid produced by enterocytes and used here
`as a biomarker of remnant enterocyte mass),15 and change in the
`fluid composite effect (FCE).12 The FCE is a putative measure of
`
`Page 3
`
`

`
`1476 JEPPESEN ET AL
`
`GASTROENTEROLOGY Vol. 143, No. 6
`
`the combined effects of teduglutide on intestinal fluid absorp-
`tion, that is, not only on PS volume reduction, but also on the
`ability to reduce oral fluid intake and increase urine output
`volume. The FCE was a summation of the increase in urine
`production, reduction in PN/IV volume, and reduction in oral
`fluid intake (L/wk), calculated as a baseline measurement of the
`individual components. The FCE was also calculated for each
`scheduled postbaseline visit using the following equation: reduc-
`tion in PS volume (L/wk) ⫹ reduction in oral fluid intake
`volume (L/wk) ⫹ increase in urine output volume (L/wk).
`Clinical evaluations (vital signs, physical examinations, and
`electrocardiograms), adverse event (AE) monitoring, and labora-
`tory tests (hematology, serum chemistries, and urinalysis) were
`assessed. Safety assessments also included body weight, 48-hour
`urine output, antibodies to teduglutide, and any required endo-
`scopic evaluations. In patients with colon, a baseline colonos-
`copy was required for inclusion to rule out the presence of
`polyps or active intestinal disease.
`Statistical Analysis
`Eighty-six patients were randomized in a 1:1 ratio to
`detect differences in responder rates between teduglutide 0.05
`mg/kg/d and placebo groups of 35% vs 6%, respectively, based on
`the response rates reported in the earlier phase 3 study12 (␣ ⫽
`.05, 2-sided test and power ⫽ 90%). Grounded on these assump-
`tions, nQuery Advisor (version 6.0, Statistical Solutions, Saugus,
`MS) based on Fisher exact test was used to calculate the power.
`The number and percentage of responders are presented here
`by treatment group. The intent-to-treat analysis compared the
`event rates for the 2 treatment groups using the Cochran-
`Mantel-Haenszel test statistics adjusted for the randomization
`stratification variable (ⱕ6 or ⬎6 L/wk of PS volume at baseline).
`The percentage and absolute change in PS volume from baseline
`to the last dosing visit as well as all scheduled visits starting at
`week 4 are presented by treatment group using descriptive sta-
`tistics. Treatment group differences were compared using an
`analysis of covariance model with effects for treatment and
`baseline PS volume, with the potential for the interaction of the
`2 variables also included as an effect. Safety analyses were de-
`scriptive.
`
`Results
`Patients
`From November 2008 to January 2011, one hun-
`dred and thirty-two patients who signed informed con-
`sent forms were screened, 86 were randomized, and 78
`completed the dosing period (Supplementary Figure 1).
`There were no significant differences between treatment
`groups regarding demographic characteristics and medi-
`cations at baseline (Table 3). A total of 39 patients (17 in
`the teduglutide group and 22 in the placebo group) re-
`quired optimization of PN/IV volume before entering the
`stabilization period; 26 patients in the teduglutide group
`and 21 in the placebo group went directly from screening
`to stabilization. However, 12 of these (6 in each group)
`failed to remain stable for a full 4-week period and re-
`quired a return to the optimization period. Overall, mean
`time spent in the optimization stage was 19 ⫾ 23 days for
`the teduglutide group and 23 ⫾ 24 days for the placebo
`group. No patients were weaned off PS during the opti-
`mization period.
`
`Efficacy
`Primary efficacy end point. The primary efficacy
`end point was the responder rate. There were 27/43 (63%)
`responders in the teduglutide group and 13/43 (30%) in
`the placebo group (P ⫽ .002). Small bowel length did not
`appear to be a predictor of response. Responder rate was
`higher for patients without colon in continuity (com-
`pared with patients with colon in continuity); however,
`findings did not achieve statistical significance.
`Secondary end points. At all visits, change from
`baseline in actual PS volume was greater in the teduglu-
`tide group than in the placebo group (Figure 2). At Week
`24, the mean ⫾ SD PS volume reduction in the teduglu-
`tide group was 4.4 ⫾ 3.8 L/wk from a baseline of 12.9 ⫾
`7.8 L/wk vs 2.3 ⫾ 2.7 L/wk from a baseline of 13.2 ⫾ 7.4
`L/wk in the placebo group. The difference in absolute
`change between the treatment groups was statistically
`significant at week 8 (P ⫽ .011) and remained significant
`through week 24 (P ⬍ .001). The percentage reduction in
`actual PS volume at week 24 was 32% ⫾ 19% in the
`teduglutide group vs 21% ⫾ 25% in the placebo group.
`The difference in percentage change between the treat-
`ment groups was significant at week 12 (P ⫽ .028) and
`remained significant through week 24 (P ⫽ .030). No
`patients were completely weaned from PS at week 24.
`Selected exploratory end points. The percentage
`of patients with response (20%⫺100% PS reduction vs
`baseline) was higher in the teduglutide group than in the
`placebo group at all visits. At week 24, 30/39 teduglutide
`patients (77%) demonstrated response vs 18/39 placebo
`patients (46%; P ⫽ .01). The percentage of patients with a
`1-day or more reduction in weekly actual PS use at week
`24 was higher in the teduglutide group (54%, n ⫽ 21/39
`[13 with 1 day off; 8 with ⱖ2 days off]) than in the
`placebo group (23%, n ⫽ 9/39; [6 with 1 day off; 3 with ⱖ2
`days off]; P ⫽ .005).
`Oral fluid intake was significantly higher in patients
`receiving placebo compared with those receiving teduglu-
`tide at weeks 12, 20, and 24 (Figure 3). At all visits, greater
`reduction in FCE was seen in the teduglutide group than
`in the placebo group. At week 24, the mean ⫾ SD reduc-
`tion in the teduglutide group was 5.4 ⫾ 6.0 L/wk vs 1.1 ⫾
`4.3 L/wk in the placebo group (P ⬍ .0006).
`Teduglutide resulted in a significant increase in plasma
`citrulline concentration from baseline levels. At baseline,
`mean ⫾ SD plasma citrulline concentration values were
`18.4 ⫾ 9.5 0 ␮mol/L and 17.5 ⫾ 9.0 ␮mol/L in the
`teduglutide and placebo groups, respectively. At 24 weeks,
`the mean ⫾ SD increase over baseline in plasma citrulline
`concentration was 20.6 ⫾ 17.5 ␮mol/L in the teduglutide
`group vs 0.7 ⫾ 6.3 ␮mol/L in the placebo group (P ⱕ
`.0001). Over 24 weeks, patients receiving teduglutide had
`a nonsignificant increase in body weight of 1.0 ⫾ 3.7 kg
`compared with baseline (P ⫽ .10), whereas patients receiv-
`ing placebo had a decrease in body weight of ⫺0.6 ⫾ 2.8
`kg (P ⫽ .20).
`
`CLINICAL AT
`
`Page 4
`
`

`
`December 2012
`
`TEDUGLUTIDE AND SBS 1477
`
`CLINICALAT
`
`P value
`
`.694a
`
`.759a
`
`.829b
`
`.694b
`
`.100b
`
`.277a
`
`.608a
`
`.332a
`
`.019b
`
`.504a
`
`.707a
`
`.388a
`
`1.000c
`
`Table 3. Demographic Characteristics and Medication at Baseline
`
`Placebo (n ⫽ 43)
`
`Teduglutide, 0.05
`mg/kg/d (n ⫽ 43)
`
`Overall (N ⫽ 86)
`
`Age, mean (SD), y
`Range
`BMI, mean (SD), kg/m2
`n
`Range
`Women, n (%)
`Cause of major intestinal resection, n (%)
`Vascular disease
`Crohn’s disease
`Volvulus
`Injury
`Cancer
`Other
`Intestinal anatomy or remnant small bowel
`length unknown, n
`Patients with stoma, n
`Types of stoma, n (%)
`Jejunostomy
`Ileostomy
`Colostomy
`Other (duodenostomy; jejunostomy ⫹
`ileostomy)
`Colon in continuity, n (%)
`Overall remnant small bowel length, mean
`(SD), cm
`
`n
`Median
`Range
`Remnant small bowel length in patients with
`jejunostomy/ileostomy, mean (SD), cm
`
`n
`Median
`Range
`Remnant small bowel length in patients with
`colon in continuity, mean (SD), cm
`
`n
`Median
`Range
`Remnant colon, n (%)
`⬎25%⫺50%
`⬎50%⫺75%
`⬎75%⫺100%
`Time since last small bowel resection, mean, y
`n
`⬍1
`ⱖ1 to ⬍2
`ⱖ2 to ⬍5
`ⱖ5
`Time receiving PS, mean (SD), y
`Median
`Range
`Parenteral volume, mean (SD), mL/d
`Median
`Range
`Time receiving PS, mean (SD) d/wk
`Median
`Range
`Parenteral volume stratification
`Parenteral volume ⱕ6 L/wk, n (%)
`Receiving PS 3/4/5/6/7 d/wk, n
`Parenteral volume ⬎6 L/wk, n (%)
`Receiving PS 3/4/5/6/7 d/wk, n
`
`49.7 (15.6)
`18–82
`22.3 (3.1)
`43
`17.5–28.6
`24 (56)
`
`16 (37)
`8 (19)
`6 (14)
`4 (9)
`2 (5)
`7 (16)
`3
`
`17
`
`5 (29)
`9 (53)
`1 (6)
`2 (12)
`
`23 (54)
`68.7 (63.9)
`
`40
`48.0
`5–343
`122.8 (81.6)
`
`13
`130
`40–343
`43.3 (31.5)
`
`22
`32.5
`5–100
`
`5 (12)
`8 (19)
`10 (23)
`7.9
`43
`0
`6
`17
`20
`5.9 (5.7)
`3.9
`1.0–25.8
`1929 (1026)
`1771
`514–5000
`5.9 (1.5)
`7.0
`3.0–7.0
`
`7 (16)
`4,1,0,1,1
`36 (84)
`2,2,2,6,24
`
`50.9 (12.6)
`22–78
`22.5 (3.2)
`42
`17.6–9.8
`22 (51)
`
`13 (30)
`10 (23)
`3 (7)
`4 (9)
`1 (2)
`12 (28)
`3
`
`21
`
`11 (52)
`6 (29)
`4 (19)
`0 (0)
`
`26 (61)
`84.4 (64.6)
`
`40
`70.0
`15–250
`137.7 (70.9)
`
`15
`120
`45–250
`52.4 (31.8)
`
`25
`50
`15–140
`
`14 (33)
`6 (14)
`3 (7)
`6.9
`42
`1
`7
`15
`19
`6.8 (6.3)
`3.6
`1.0–24.7
`1844 (1057)
`1714
`124–4714
`5.6 (1.7)
`7.0
`3.0–7.0
`
`8 (19)
`7,0,1,0,0
`35 (81)
`3,4,2,4,22
`
`50.3 (14.1)
`18–82
`22.4 (3.1)
`85
`17.5–29.8
`46 (54)
`
`29 (34)
`18 (21)
`9 (11)
`8 (9)
`3 (4)
`19 (22)
`6
`
`38
`
`16 (42)
`15 (40)
`5 (13)
`2 (5)
`
`49 (57)
`76.5 (64.4)
`
`80
`57.5
`5–343
`130.8 (75.0)
`
`28
`125
`40–343
`48.1 (31.6)
`
`47
`38
`5–140
`
`19 (22)
`14 (16)
`13 (15)
`7.4
`85
`1
`13
`32
`39
`6.3 (6.0)
`3.9
`1.0–25.8
`1887 (1036)
`1764
`124–5000
`5.8 (1.6)
`7.0
`3.0–7.0
`
`15
`11,1,1,1,1
`71
`5,6,4,10,46
`
`Page 5
`
`

`
`1478 JEPPESEN ET AL
`
`Table 3. Continued
`
`Concomitant medication
`Antidiarrheals, n (%)
`Antisecretory agents, n (%)
`
`GASTROENTEROLOGY Vol. 143, No. 6
`
`Placebo (n ⫽ 43)
`
`Teduglutide, 0.05
`mg/kg/d (n ⫽ 43)
`
`Overall (N ⫽ 86)
`
`P value
`
`16 (37)
`22 (51)
`
`22 (51)
`25 (58)
`
`38 (44)
`47 (55)
`
`BMI, body mass index.
`aP value is calculated using the general linear model method, with corresponding variables as dependent variables and treatment group as the
`independent variable.
`bP value is based on the ␹2 contingency table using the exact method.
`cP values for overall treatment comparisons are base on the Fisher exact test for categorical variables and on a 1-way analysis of variance with
`effect for treatment for continuous variable.
`
`CLINICAL AT
`
`Safety and Tolerability
`The safety population consisted of 85 patients who
`received ⱖ1 doses of study drug. Total exposure to study
`drug was similar in the 2 study groups. There were no
`deaths during the study. The number of patients with
`AEs, serious AEs, treatment-emergent AEs (TEAEs), or
`discontinuations due to treatment-emergent serious AEs
`was comparable between treatment groups. Treatment-
`emergent serious AEs were reported by 27 patients (15/42
`patients [36%] vs 12/43 patients [28%] in the teduglutide
`group vs placebo group, respectively). Sixty-nine patients
`were reported to have ⱖ1 TEAEs during the study (35/42
`[83%] vs 34/43 [79%]). Five patients were reported to have
`experienced TEAEs leading to discontinuation during the
`study (2/42 [5%] vs 3/43 [7%]); none were serious.
`The most frequently reported TEAEs in the teduglutide
`group were of gastrointestinal origin, such as abdominal
`pain, nausea, gastrointestinal stoma complication, or ab-
`dominal distension (Supplementary Table 2). Two treat-
`ment-emergent serious AEs reported in the teduglutide
`group were deemed related to the study treatment (acute
`cholecystitis and small intestinal stenosis); both resolved.
`No major findings were reported in the laboratory/
`chemistry or hematology tests of the teduglutide-treated
`vs placebo patients. Six patients from the teduglutide
`group and none from the placebo group developed anti-
`teduglutide antibodies after the start of study drug. An-
`
`Figure 2. Mean (SE) absolute reduction in parenteral support.
`
`tibodies were non-neutralizing and without evidence of
`decreased effect on PS volume reduction or evidence of
`systemic hypersensitivity or other clinically significant se-
`quelae.
`
`Discussion
`In this phase 3 study, teduglutide met the primary
`efficacy end point, defined as the percentage of patients
`who achieved a 20% to 100% reduction in weekly PS
`volume at weeks 20 and 24 compared with baseline. There
`were 27/43 (63%) responders in the teduglutide group and
`13/43 (30%) in the placebo group (P ⫽ .002). When
`addressing the proabsorptive effects of teduglutide, im-
`provements in intestinal fluid absorption under ideal cir-
`cumstances should be counterbalanced by equivalent
`PN/IV volume reductions.
`One of the observations in the previous phase 3 study
`was that, in addition to significant reductions in PS vol-
`ume in patients assigned to teduglutide 0.05 mg/kg/d
`(and nonsignificant reductions in those receiving the 0.10-
`mg/kg/d dosage), there were also small decreases in PS
`volume in patients randomized to placebo.12 This sug-
`gests that appropriate day-to-day fluid management with
`strict adherence to the weaning algorithm can have an
`intrinsic benefit and that treatment with teduglutide 0.05
`mg/kg/d provides an important additional benefit.
`Compared with the previous phase 3 study, this study
`protocol allowed earlier (after 2 weeks vs 4 weeks) and
`more aggressive (10%⫺30% vs 10%) PS volume reduc-
`tions.12 This was intended to minimize fluid accumula-
`tion (edema), reductions in oral intake, and increases in
`urine production encountered in the previous phase 3
`study in relation to treatment with teduglutide 0.10 and
`0.05 mg/kg/d, which negatively affected the primary study
`end point.12
`At week 24, after these protocol adjustments, a larger
`mean PS volume reduction, of 4.4 ⫾ 3.8 L/wk (32%
`change from baseline mean of 12.5 L/wk) was observed
`compared with a reduction of 2.5 ⫾ 2.3 L/wk (27% change
`from baseline mean of 9.6 L/wk) in the teduglutide
`0.05-mg treatment group in the previous study, which
`might explain this study’s higher responder rate. In this
`study, the effects of teduglutide on PS volume reduction
`more closely reflected the FCE (5.4 ⫾ 6.0 L/wk). The FCE
`
`Page 6
`
`

`
`CLINICALAT
`
`December 2012
`
`TEDUGLUTIDE AND SBS 1479
`
`Figure 3. Summary of changes
`in (upper left panel) oral fluid in-
`take, from baseline mean, 1888
`(SD 883) mL/d for teduglutide,
`1637 (603) mL/d for placebo; (up-
`per right panel) urine volume, from
`baseline mean 1350 (216) mL/d
`for teduglutide, 1372 (269) mL/d
`for placebo; (lower left panel) par-
`enteral volume,
`from baseline
`mean 1777 (1096) mL/d for te-
`duglutide, 1899 (1077) mL/d for
`placebo; and (lower right panel)
`fluid composite effect, from base-
`line mean 2317 (1651) mL/d for te-
`duglutide, 2176 (1270) mL/d for
`placebo. Mean (SE) values for
`changes are shown.
`
`at week 24 was in the same magnitude in both studies
`(4.4 ⫾ 7.3 L/wk).
`The high placebo response in this study (30% vs 5% in
`the previous study) might be explained by addressing the
`components of the FCE (Figure 3). In the previous study,
`at week 24, patients receiving placebo had no significant
`changes in the oral fluid intake or urine output volume
`during 48-hour measurements. Therefore, the PS volume
`reduction (0.9 ⫾ 1.4 L/wk) was in the same magnitude as
`the FCE (0.8 ⫾ 6.2 L/wk) in the previous study. In the
`current study, where protocol modifications encouraged
`earlier and more aggressive PS reductions, significantly
`larger PS reductions were also achieved in patients receiv-
`ing placebo (2.3 ⫾ 2.7 L/wk), but subsequently these
`patients had to increase their oral fluid intake signifi-
`cantly (1.6 ⫾ 3.6 L/wk; P ⫽ .009) to maintain urine
`production and hydration constant (Figure 3).
`At week 24, the FCE was 1.1 ⫾ 4.3 L/wk in this study in
`the patients receiving placebo, which closely reflected
`findings in the first phase 3 study. In addition, because
`the oral fluid intake and urine output were not constant
`over time, the observed effects of teduglutide on absolute
`and relative PS volume reduction might imprecisely esti-
`mate the true effect of teduglutide on intestinal fluid
`absorption. Therefore, the FCE might better illustrate the
`summarized effects of teduglutide as also demonstrated
`
`in the 3-week, phase 2, metabolic balance study in pa-
`tients with SBS, in which fecal wet weight excretion was
`reduced from baseline by 711 ⫾ 734 g/d (P ⫽ .001) and
`intestinal wet weight absorption was increased by 743 ⫾
`477 g/d (P ⬍ .001).11 Although FCE is not a standard
`measure, it can be of value in the clinical assessment of
`patients with SBS as another potential indicator, along
`with urine output alone or determination of enteral bal-
`ance (ie, oral fluid intake minus stool volume) when
`making weaning decisions.
`The PS volume reductions from baseline of 32% ⫾ 19%
`(4.4 ⫾ 3.8 L/wk) demonstrated in this study provide
`correspondingly less time connected to the central line, if
`the infusion rate is the same. Because patients with SBS
`can spend up to 16 hours per day connected to the central
`line, this will liberate considerable time for unhindered
`daytime activities or undisturbed sleep.16 It is appeal

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket