Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Obesity (Silver Spring). 2012 Feb; 20(2): 330–342.
Published online 2011 Nov 3. doi: 10.1038/oby.2011.330
PMCID: PMC3270297
PMID: 22051941

Controlled-Release Phentermine/Topiramate in Severely Obese Adults: A Randomized Controlled Trial (EQUIP)

Associated Data

Supplementary Materials

Abstract

A 56-week randomized controlled trial was conducted to evaluate safety and efficacy of a controlled-release combination of phentermine and topiramate (PHEN/TPM CR) for weight loss (WL) and metabolic improvements. Men and women with class II and III obesity (BMI ≥ 35 kg/m2) were randomized to placebo, PHEN/TPM CR 3.75/23 mg, or PHEN/TPM CR 15/92 mg, added to a reduced-energy diet. Primary end points were percent WL and proportions of patients achieving 5% WL. Secondary end points included waist circumference (WC), systolic and diastolic blood pressure (BP), fasting glucose, and lipid measures. In the primary analysis (randomized patients with at least one postbaseline weight measurement who took at least one dose of assigned drug or placebo), patients in the placebo, 3.75/23, and 15/92 groups lost 1.6%, 5.1%, and 10.9% of baseline body weight (BW), respectively, at 56 weeks (P < 0.0001). In categorical analysis, 17.3% of placebo patients, 44.9% of 3.75/23 patients, and 66.7% of 15/92 patients, lost at least 5% of baseline BW at 56 weeks (P < 0.0001). The 15/92 group had significantly greater changes relative to placebo for WC, systolic and diastolic BP, fasting glucose, triglycerides, total cholesterol, low-density lipoprotein (LDL), and high-density lipoprotein (HDL). The most common adverse events were paresthesia, dry mouth, constipation, dysgeusia, and insomnia. Dropout rate from the study was 47.1% for placebo patients, 39.0% for 3.75/23 patients, and 33.6% of 15/92 patients. PHEN/TPM CR demonstrated dose-dependent effects on weight and metabolic variables in the direction expected to be beneficial with no evidence of serious adverse events induced by treatment.

Introduction

Obesity, a highly prevalent major public health problem, is associated with increased mortality and morbidity, including an increased risk of type 2 diabetes mellitus and cardiovascular disease, physical disabilities, sleep apnea, and reduced quality of life (1). When achieved by medically recommended procedures, weight loss (WL) is associated with reduced morbidities in obese persons (2). Beyond surgery, long-term weight reductions much greater than 3–6 kg remain elusive (3). Hence, generating additional medical treatment options is a priority.

Phentermine hydrochloride is a sympathomimetic amine approved by the US Food and Drug Administration (FDA) in 1959 with a dose range of up to 37.5 mg/day for short-term obesity treatment. Phentermine stimulates increased hypothalamic release of norepinephrine with no detectable effect on serotonin (4). Topiramate, a fructose monosaccharide derivative with sulphamate functionality, was approved for the treatment of epilepsy in 1996 and the prevention of migraine in 2004. Randomized controlled trials (RCTs) show that topiramate monotherapy produces WL among obese individuals of ~6–8 kg at 24 weeks and improvements in lipids, glycemic control, and blood pressure (BP) (5,6,7). However, topiramate has been associated with adverse events (AEs) that may limit its use as a single agent at optimal doses for WL. With respect to possible mechanisms for the WL effects of topiramate, animal experiments suggest that topiramate-induced WL results from increased energy expenditure, decreased energetic efficiency, and decreased caloric intake (8,9,10). A significant factor associated with topiramate-induced WL in humans appears to be decreased caloric intake (11,12,13). However, consistent with animal findings, reduction in caloric intake does not appear to fully explain the observed WL (11,12); thus, as suggested by animal studies, topiramate-induced WL in humans may be also related to other mechanisms, such as increased energy expenditure or decreased energy efficiency.

Controlled-release phentermine/topiramate (PHEN/TPM CR) is an investigational WL therapy combining immediate-release phentermine and controlled-release topiramate given in a single daily morning dose. The top dose of PHEN/TPM CR contains phentermine 15 mg (expressed as free-base) and topiramate 92 mg. PHEN/TPM CR contains lower doses of these components than are currently marketed or have been studied for monotherapy in obesity (6,14). Each of the individual components has published dose-related efficacy, tolerability, and AE data (15,16,17). The combination's efficacy in WL exceeds the maximal response achieved with either individual agent alone at equivalent doses (18), which plausibly results from each component targeting multiple mechanisms that impact energy balance. The goal in developing this combination therapy was to use the dose of each respective agent that provided the greatest level of WL efficacy while minimizing tolerability concerns.

Methods and Procedures

Design overview

A double-blind, parallel-group design was used with three arms: placebo (n = 514), PHEN/TPM CR 3.75/23 mg (n = 241), and PHEN/TPM CR 15/92 mg (n = 512). After screening, all eligible patients underwent a blinded 4-week postrandomization titration period and 52 weeks at randomized dose. All interventions were added to a standardized lifestyle program. Total treatment duration was 56 weeks.

Settings and participants

Subjects were enrolled beginning in November 2007 at 91 US sites, consisting of clinical practices, clinical trial sites, and academic centers. The last subject completed all study visits in May 2009. At every site, institutional review board approval and written informed consent were obtained. Eligibility criteria included age 18–70 years, BMI ≥35 kg/m2 (no upper limit), triglycerides ≤200 mg/dl with treatment of 0–1 lipid-lowering medication, BP ≤140/90 mm Hg with treatment of 0–2 antihypertensive medications, and fasting serum glucose level ≤110 mg/dl. For detailed entry criteria, see Supplementary Appendix 3 online.

Randomization and interventions

Patients were randomized and stratified by gender via a pseudo-random number generator constrained to a 2:1:2 allocation ratio. This allocation ratio was designed to increase the power of the safety analysis by having more patients receive placebo and by having the highest dose used. All study participants, study physicians, site staff, and sponsor representatives involved in the study conduct were blinded to patient assignment until after the trial was complete. There was an independent, unblinded data and safety monitoring board. The study drug and placebo were visually indistinguishable. Treatment was initiated with a 4-week blinded titration period (typically recommended with clinical topiramate use to minimize AEs), starting with PHEN/TPM CR 3.75/23 or matched placebo and thereafter increased weekly by 3.75/23 mg increments to the assigned dose. Following titration to assigned dose, patients were evaluated for an additional 52 weeks with monthly clinic visits. All patients were provided with standardized lifestyle counseling, based on the LEARN Manual (19) and advised to follow a 500-kcal daily reduction in dietary intake, increased water consumption, and increased physical activity.

Outcomes and follow-up

Outcomes of interest were percent, absolute, and categorical body-WL as well as numerous metabolic and cardiovascular outcomes. Patients were weighed using a calibrated digital scale, and waist circumference (WC) was measured by trained study personnel at each visit. BP measurements were obtained using a calibrated sphygmomanometer with appropriately sized cuff. Fasting blood samples were drawn at baseline and then at weeks 4, 8, 16, 28, and 56 and analyzed in a single central reference laboratory (Medpace Reference Laboratories, Cincinnati, OH). AEs were monitored at each visit with occurrence date, severity, relationship to study drug, action taken with respect to study drug, and outcome. Depression and suicidality were also assessed in detail (see Supplementary Appendix 2 online). As recommended elsewhere (20), patients who discontinued treatment were encouraged to continue with data assessments through study completion, even though they were no longer taking the drug.

Statistical analyses

Power analysis. Based on standard deviations for percent WL observed in a previous PHEN/TPM CR study (21), this study provided >95% power at the two-tailed 0.05 α-level to reject the null hypothesis of no treatment-placebo difference if the population difference was ~2% to 3% of body weight (BW).

Primary efficacy analyses. The primary efficacy measure was percent WL at end of study, analyzed once as a continuous variable and then as a dichotomous variable with cut-off points at 5%, 10%, and 15% of BW. Secondary efficacy measures were change in WC, systolic BP, diastolic BP, fasting triglycerides, total cholesterol, low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, total cholesterol/HDL ratio, and fasting serum glucose. Gender was included as a covariate in all models since it was used as a stratification factor.

For analysis of percent WL as a dependent variable, three main analyses were performed (listed as analyses A–C in Table 1) and are reported in detail with additional sensitivity analyses (analyses D–F in Table 1) in Supplementary Appendixes 1–3 online. Analysis A, the analysis prespecified in the study protocol, is an intent-to-treat (ITT) analysis according to FDA standards (22). Note that this analysis included data from patients who discontinued the drug but remained in the study and continued with data assessments through study completion, even though they were no longer taking the drug.

Table 1

Efficacy analyses conducted for percent weight loss as a continuous outcome
An external file that holds a picture, illustration, etc.
Object name is oby2011330t1.jpg

Analysis B was a prespecified secondary per protocol–based analysis that was included to facilitate assessment of the experience in patients who reported using the study drug or placebo for the full intended treatment course. It included only patients for whom end-point data were available and who reported taking their study drug/placebo within 7 days before final end-point measurement. Again, the analysis of covariance model described in Table 1 was used.

Analysis C was an ITT analysis as defined by Lachin (23) that included all randomized patients. To accommodate missing data, multiple imputation was used as described in Table 1 (for further detail, refer to Elobeid et al., (24)).

For analyses with percent weight change as a dichotomous outcome, the three analyses described above were used except that analysis of covariance was replaced by a corresponding logistic regression with the same covariates.

Sensitivity analyses for primary efficacy end points. To test the robustness of findings to variations in data analytic procedures, several additional efficacy analyses (analyses D–F in Table 1) on percent WL were conducted. For the mixed model described in analysis E of Table 1, a random-effects regression model with treatment, gender, baseline, and time as covariates was used, where time was modeled as continuous and calculated from first dose date. Random intercepts and slopes were estimated and an unstructured residual covariance matrix was specified. Results of these analyses are described in Supplementary Table S2 online.

Analyses for secondary efficacy end points. For all other efficacy measures, reported analyses correspond to analyses A–C described in Table 1, substituting change (or percent change for some variables as indicated in results section) in the dependent variable of interest for percent WL and baseline values of that dependent variable for baseline BW.

Safety analyses. All AE-preferred terms (defined by MedDRA coding dictionary version 10.1) with a frequency of 5% or more in any treatment group or those that occurred significantly more often with active treatment than placebo (at a two-tailed nominal 0.05 α-level) are listed in Table 4. Due to concerns with past WL drugs (25), we conducted extensive depression and suicidality assessments using the Patient Health Questionnaire (PHQ-9) and Columbia Suicidality Severity Rating Scale (C-SSRS) instruments (see Supplementary Appendix 2 online).

Table 4

Adverse events with a frequency of ≥5% in any treatment group or differing significantly in frequency between placebo and either treatment group
An external file that holds a picture, illustration, etc.
Object name is oby2011330t4.jpg

Results

Baseline characteristics

Baseline data are in Table 2. Groups were not significantly different on any baseline variable. Overall, mean age was 42.7 years, mean BMI was 42.0 kg/m2, and 83% were female, with substantial representation of black patients (16–18%). Means for BP, fasting glucose, triglycerides, and HDL cholesterol did not meet established thresholds indicative of increased cardiometabolic disease risk (26).

Table 2

Baseline data by treatment condition
An external file that holds a picture, illustration, etc.
Object name is oby2011330t2.jpg

Subject disposition

As shown in Figure 1, 59.9% of randomized patients completed the study regardless of whether they continued taking the assigned drug/placebo (52.9% placebo, 61.0% PHEN/TPM CR 3.75/23, 66.4% PHEN/TPM CR 15/92; P < 0.0001 for difference), and 53.7% reported taking the assigned study drug/placebo for the full intended treatment course (46.9% placebo, 57.3% PHEN/TPM CR 3.75/23, 58.8% PHEN/TPM CR 15/92; P = 0.0003 for difference). Most common reasons for discontinuation were lost to follow-up or withdrawal of consent (more common in placebo than active groups) or AEs (more common in active than placebo groups). Overall discontinuations were lower in patients receiving active treatments.

An external file that holds a picture, illustration, etc.
Object name is oby2011330f1.jpg

Patient disposition. After screening, all eligible patients underwent 56 weeks of treatment; 4-week blinded, postrandomization titration period, followed by 52 weeks at randomized dose. *Reason for discontinuation as reported while subject was receiving study drug; Reason for study discontinuation. Subjects were able to discontinue study drug and study for different reason, however, reason was often the same. When they differed, the final reason for discontinuation from the study was used. D/C, discontinued; Meds, medications; PHEN/TPM CR, controlled-release phentermine/topiramate.

Weight loss

Patients in the 15/92 group lost significantly more weight than patients in the 3.75/23 group who in turn lost significantly more weight than patients receiving placebo (P < 0.0001 for all comparisons; see Figure 2 and Table 3), regardless of analysis used. Using the prespecified ITT-last observation carried forward (LOCF) (Analysis A), patients receiving 15/92, 3.75/23, and placebo lost 10.9%, 5.1%, and 1.6% of BW, respectively. Moreover, completers (Table 3, Analysis B and Supplementary Table S2 and Figure S1 online) lost 14.4% of BW on 15/92 and 6.7% on 3.75/23 and only 2.1% on placebo. Statistical assumption testing suggested appropriateness of the models, with all sensitivity analyses (Analyses C–F) yielding confirmatory findings (Table 3 and Supplementary Appendix 1 and Table S2 online). We also conducted the baseline observation carried forward analyses (27), and although point estimates changed, conclusions about statistical significance and direction of effects remained unchanged (data not shown).

An external file that holds a picture, illustration, etc.
Object name is oby2011330f2.jpg

Plot of efficacy results with analysis A (prespecified ITT-LOCF). (a) Mean percent WL; (b) Patients achieving ≥5%, ≥10%, and ≥15% WL; (c) LS mean percent WL by baseline BMI category. Analysis A included data from patients who discontinued drug but remained in study and continued with data assessments through study completion even though no longer taking drug. The supplementary material presents mean percent WL for Analyses B and C. Error bars represent 95% confidence interval. ITT, intent-to-treat; LOCF, last observation carried forward; LS, least-squares; PHEN/TPM CR, controlled-release phentermine/topiramate; WL, weight loss.

Table 3

Weight loss and secondary outcomes at week 56 by treatment condition
An external file that holds a picture, illustration, etc.
Object name is oby2011330t3.jpg

When analyzed as dichotomous data, regardless of cut-off point (5%, 10%, or 15% of BW), a higher proportion of patients in the 15/92 group exceeded threshold than did patients in the 3.75/23 group, whose proportion exceeded that of placebo patients (P < 0.05 for all comparisons). In the prespecified-ITT-LOCF (Analysis A), percentages of patients losing ≥5%, ≥10%, and ≥15% of BW were, respectively, 66.7%, 47.2%, and 32.3% on 15/92; 44.9%, 18.8%, and 7.3% on 3.75/23; and 17.3%, 7.4%, and 3.4% on placebo (all comparisons P < 0.05). Among completers, percentages of patients losing ≥5%, ≥10%, and ≥15% of BW were, respectively, 83.5%, 67.7%, and 48.1% on 15/92; 59.1%, 27.7%, and 12.4% on 3.75/23; and 25.5%, 13.0%, and 5.9% on placebo (all comparisons P < 0.0001).

We tested whether results differed by baseline BMI by repeating Analysis A after categorizing patients by baseline BMI and then testing for interaction between baseline BMI category and treatment. The interaction was not significant (P = 0.8056), indicating that results did not significantly differ by baseline BMI (Figure 2).

Secondary end point analysis

The 15/92 group had significantly greater least-squares mean changes, relative to placebo, in WC, systolic BP, diastolic BP, glucose, triglycerides, total cholesterol/HDL ratio, total cholesterol, LDL cholesterol, and HDL cholesterol. The 3.75/23 group had numerically, but not always statistically significant, greater least-squares mean changes compared with placebo, in all these variables (Table 3).

Safety analyses

Table 4 lists AEs that occurred at a frequency of 5% or higher in any treatment group or occurred at a significantly higher frequency in the 15/92 group than placebo. The most common AEs occurring more often with 15/92 treatment than placebo included paresthesia, dry mouth, constipation, dysgeusia, and insomnia. Other less frequent events occurring more commonly with 15/92 treatment were depression, irritability, alopecia, anxiety, disturbance in attention, and hypoesthesia. Serious AE (SAE) rates were the same across treatment groups: 13 (2.5%) with placebo, 6 (2.5%) with 3.75/23, and 13 (2.5%) with 15/92.

Most AEs reported were mild in severity (Supplementary Table S3 online). AEs most often resulting in treatment discontinuation in the 15/92 group were insomnia, irritability, anxiety, headache, disturbance in attention, depression, dry mouth, and nephrolithiasis. The exact numbers and percentages of AE-related dropouts were 43 (8.4%) for placebo, 27 (11.3%) for 3.75/23, and 82 (16.0%) for 15/92.

Drug-related SAEs occurred in 2 (0.4%) patients treated with placebo, 1 (0.4%) patient treated with 3.75/23, and 1 (0.2%) patient treated with 15/92. The SAEs reported as drug-related in the placebo group included chest pain and pulmonary embolism, both of which resulted in discontinuation of study drug. The SAE reported in the 3.75/23 group was cholelithiasis that resolved following a 1-week interruption of treatment, and one patient treated with 15/92 was diagnosed with myelogenous leukemia ~6 months after starting treatment and discontinued the study due to this event. These data should be viewed with the understanding that very few SAEs have been reported as drug-related and that investigators' assessments of causality are speculative.

Change from baseline in heart rate was also assessed. By week 56, mean heart rate had decreased 0.2 bpm in placebo, 0.3 bpm on 3.75/23 (P = 0.9552 vs. placebo), and increased 1.2 bpm on 15/92 (P = 0.0830 vs. placebo). Both doses of PHEN/TPM CR resulted in a decrease in serum bicarbonate (mean changes in mEq/l: −0.3 for placebo, −1.6 for 3.75/23, and −1.7 for 15/92); significant reductions (<17 mEq/l at two consecutive visits) occurred in only 3 (1.3%) patients receiving 3.75/23, 4 (0.8%) patients receiving 15/92, and none receiving placebo.

In addition, depressive symptoms were assessed using the PHQ-9 questionnaire, and suicidality (ideation and behavior) was assessed using the C-SSRS (Supplementary Appendix 2 and Table S1 online). Change in mean PHQ-9 total score indicated improvement in depressive symptoms over time in all treatment groups, but no significant differences among groups. There was no significantly increased suicide risk as defined and assessed by the C-SSRS in patients treated with PHEN/TPM CR compared with placebo. Neither suicidal behavior nor suicidal ideation with an intent to act were reported at any time after treatment initiation, and rates of suicidal ideation without intent to act were comparable to rates seen in placebo-treated patients.

There were 15 pregnancies in women exposed to PHEN/TPM CR. Among these, there were three spontaneous abortions, three elective abortions, and nine healthy live births. No congenital malformations were observed.

Discussion

Three salient points emerge from this RCT. First, PHEN/TPM CR caused WL in obese patients. Compared with placebo, both doses of PHEN/TPM CR yielded significantly greater 1-year WL, with a greater proportion of patients losing more than 5%, 10%, or 15% of baseline BW. Results were consistent regardless of analytic method employed. Patients treated with PHEN/TPM CR 15/92 and 3.75/23 lost 10.9% and 5.1% of BW, respectively, when analyzed as ITT-LOCF, compared with 1.6% WL on placebo and 14.4% and 6.7% WL in completers-only analyses compared with 2.1% WL with placebo. Among patients who completed the course of 15/92, 48.1% lost ≥15% of BW, 67.7% lost ≥10%, and 83.5% lost ≥5%. Significant WL also occurred early; patients treated with 15/92 lost between 8% and 10% of BW after 3 months of treatment.

Second, WL induced by PHEN/TPM CR was accompanied by improvements in many cardiovascular and metabolic risk factors, such as WC, systolic BP, and total cholesterol/HDL cholesterol ratio in both doses. PHEN/TPM CR 15/92 treatment was also associated with significant improvements in diastolic BP, fasting glucose, LDL cholesterol, HDL cholesterol, and total cholesterol.

Third, certain AEs (paresthesia, dry mouth, constipation, and dysgeusia) occurred at a higher frequency with 15/92 treatment, with twice (16.2% vs. 8.4%) as many patients treated with 15/92 compared with placebo withdrawing due to one or more AE. An indication of tolerability can be obtained from the AE data. The most common AEs associated with PHEN/TPM CR treatment were paresthesia, dry mouth, constipation, dysgeusia, and insomnia; however, none of these events caused study discontinuation in more than 1% of patients. Mood-related (depression, anxiety, and irritability) and cognition-related (disturbance in attention) AEs occurred at higher frequencies among patients receiving 15/92. Mood assessments with standardized rating scales (PHQ-9) showed an improvement in mood-related symptoms overall (see Supplementary Table S1 online). The low incidence of treatment-emergent depression in this trial should be viewed in the context of exclusion of patients with substantial depressive symptoms at study entry (PHQ-9 score of >10 was an exclusion criterion, and the mean (s.d.) PHQ-9 score of 2.8 (2.8) was low, suggesting minimal depressive symptoms). However, the study allowed participation of patients with a self-reported history of depression, patients with adequately controlled depression based on PHQ-9 scores, and those on stable doses of antidepressants. Neither suicidal behavior nor suicidal ideation with intent to act was reported at any time after treatment initiation. There was no significantly increased suicide risk, as defined and assessed by the C-SSRS, in patients treated with PHEN/TPM CR compared with placebo.

In this study, there were no birth defects among infants whose mothers were exposed to study treatment. However, given the FDA's recent reclassification of topiramate use during pregnancy (FDA has recently added a warning about increased risk of cleft lip and cleft palate with topiramate and its pregnancy category has now been changed to category D) and the general belief that WL during pregnancy is not wise, the use of PHEN/TPM during pregnancy is not recommended. For more and recent information on this topic, refer to Mølgaard-Nielsen et al. (28).

Phentermine has been associated with elevations in BP (29). In this study, PHEN/TPM CR led to reduced BP. Phentermine has also been associated with elevations in heart rate (29). Here, there was an increase in resting heart rate seen in the 15/92 subgroup (1.2 bpm above the baseline value of 73.2 bpm; P = 0.083 compared with placebo), whereas heart rate was unaltered in the 3.75/23 subgroup (a decrease of 0.3 bpm from baseline; P = 0.955 compared with placebo).

This heart rate increase in the 15/92 group is plausibly attributable to the sympathomimetic action of phentermine, given that increased heart rates have been observed in patients treated with the higher doses of phentermine monotherapy approved for short-term therapy of obesity (29). In the 15/92 treatment group, the increased heart rate was accompanied by a reduction of 2.9 mm Hg in systolic BP (ITT-LOCF; P < 0.0001 compared with placebo) and a 1.5 mm Hg decrease in diastolic BP (P = 0.0002 compared with placebo). By comparison, in the SCOUT Trial, the sibutramine treatment group exhibited a significantly increased heart rate combined with a significant elevation in BP compared with placebo (30). In terms of overall cardiac risk, larger future studies can test the hypothesis that the beneficial effects of PHEN/TPM CR on weight, BP, lipids, and glycemic measures mitigate any AEs on heart rate.

Although most pharmaceutical obesity RCTs include BMIs ranging from 27–40, or 45 at most, this study included patients with class II and III obesity (BMI ≥35 kg/m2) without any upper BMI limit. WL magnitude resulting from PHEN/TPM CR administration was estimated to be consistent across this broader BMI spectrum (ranging from 35.0–78.7). This finding refutes a common notion that nonsurgical treatments are not efficacious among extremely obese persons. For example, the National Heart, Lung, and Blood Institute guidelines on obesity treatment state, “Extremely obese persons often do not benefit from the more conservative treatments for WL and weight maintenance” (31). Our results show that this is not necessarily true. This finding is germane to the 14% of the US adult population classified as “extremely obese” (32).

A study limitation was the 40% dropout rate (47.1%, 39.0%, and 33.6% for patients receiving placebo, 3.75/23, and 15/92, respectively). This dropout rate is consistent with what one would expect at 56 weeks in obesity trials based on a published meta-analytic prediction equation (24) and lower than the 50% dropout recently reported with two other phase 3 obesity RCTs (33,34). Although our approach to retaining subjects, even if they discontinued the drug, and our use of ITT analyses with established missing data management procedures may have mitigated these high dropout rates, retention of patients in WL trials of 1 year or longer remains challenging. Nevertheless, we used several different ways of handling the missing data, all of which obtained confirmatory results. These statistical methods rest on fewer and less restrictive assumptions than do complete case analyses (35). Moreover, retention rate was significantly higher with active treatment compared with placebo. Although speculative, this may have been due in part to greater WL efficacy in the treatment groups. Greater early WL may provide tangible encouragement for patients to pursue lifestyle guidance and has been shown to be associated with greater success in WL programs (36). There were other limitations, including the overrepresentation of women (83%). Further studies with larger groups of men may be informative. The trial included mostly white persons and many participants did not have significant obesity-associated comorbid diseases. A publication of another RCT of PHEN/TPM CR covers this broader population (37). Furthermore, the study report herein excluded patients with recurrent depression, although patients on stable antidepressants were allowed. In future studies, determining the extent to which WL effects of this combination therapy are maintained for 2 years and beyond will also be informative. It will also be valuable to monitor potential adverse outcomes for periods beyond 1 year. Gathering information regarding efficacy and safety, including rate-pressure product of this drug therapy in patients with these and other significant comorbidities, in future studies may be valuable.

Currently available pharmacologic agents generally offer efficacy of ≤9% body-WL (3,38), and although modest WL is associated with improvements in weight-related comorbidities (39,40), greater benefits are seen with increased WL (39,41). Some bariatric surgery procedures are associated with mean WL of ~33% before some weight gain tends to reoccur (42). Data suggest that lap-banding results in ~22% WL after 24 months (43). PHEN/TPM CR 15/92 was associated with 14.4% WL in study completers (Analysis B) at 1 year, arguably the relevant comparison to surgical treatment. This appears to represent a result between that of other available pharmacologic agents and that of lap-banding, although randomized head-to-head comparison studies would be required before any definitive statements about relative efficacy of treatments can be made. In this RCT of PHEN/TPM CR, both 3.75/23 and 15/92 in conjunction with lifestyle modification produced statistically significant WL compared with placebo. PHEN/TPM CR 15/92 provided WL and concomitant improvement in comorbidities that exceeds weight losses reported at 1 year for other currently available pharmaceutical treatments. PHEN/TPM CR demonstrated dose-dependent beneficial effects on weight and metabolic variables with no evidence of SAEs induced by treatment.

Acknowledgments

We gratefully acknowledge the assistance of consultants and advisory board members for the input into the initial design of the study: Nancy J. Bohannon (California Pacific Medical Center, St Luke's Campus); Arthur Frank (George Washington University, Washington, DC); David G. Orloff (Medpace, Cincinnati, OH); F. Xavier Pi-Sunyer (Columbia University College of Physicians and Surgeons, New York, NY); Donna H. Ryan (Pennington Biomedical Research Center, Baton Rouge, LA); Thomas A. Wadden (University of Pennsylvania School of Medicine, Philadelphia, PA). We are also grateful to the patients, site investigators, and study staff who participated. The Lockwood Group provided editorial and graphic support for this manuscript but drafted no text; their participation was funded by Vivus, Inc. Finally, we thank Dr Kyle Grimes of the University of Alabama at Birmingham who offered additional editorial comments and Dr Lloyd Edwards of the University of North Carolina, Chapel Hill who checked and confirmed all of our statistical results. Blinded treatments and funding for the EQUIP study (sponsored study name, not an acronym) were provided by VIVUS, Inc. External authors as well as the funding sponsor participated in initial study design, data analysis, and reporting. Study conduct was overseen by a designee of the sponsor.

Notes

D.B.A. has received grants, honoraria, donations, and consulting fees from numerous food, beverage, pharmaceutical companies, and other commercial and nonprofit entities with interests in obesity, including but not limited to GlaxoSmithKline, Merck, Pfizer, Eli Lilly and Company, Abbott Laboratories, Slim-Fast Foods, VIVUS, Orexigen, and Arena. K.M.G. has received research support from Bristol-Myers Squibb, Elan, Eli Lilly and Company, Forest Labs, GlaxoSmithKline, Johnson & Johnson, NIDDK, Pfizer, and VIVUS. He owns stock in Orexigen Therapeutics and has also served as a consultant for Arena Pharmaceuticals, Merck, and VIVUS prior to 2008. He has been awarded patents for inventions related to obesity and weight-loss interventions that are unrelated to the interventions tested in the current investigation. W.W.D., T.N., B.T., C.A.P., and P.Y.T. are employees of VIVUS. T.N. is also the inventor of the combination treatment using phentermine with topiramate. W.T.G. has received research support from Merck, VIVUS, Abbott Nutrition, and Isis/Genzyme. He has served as an advisor, consultant, and/or speaker for Merck, Abbott Nutrition, Daiichi-Sankyo, Tethys, VIVUS, and Kraft Pharmaceuticals. He is a scientific advisory board member at Daiichi-Sankyo and Tethys, and he holds stock in Merck, Isis/Genzyme, Pfizer, Eli Lilly and Company, Schering-Plough, Novartis Pharmaceuticals, VIVUS, and Bristol-Myers Squibb.

Footnotes

Supplementary material is linked to the online version of the paper at http://www.nature.com/oby

Supplementary Material

Supplementary Appendix 1.

Supplementary Appendix 2.

Supplementary Appendix 3.

Supplementary Table S1.

Supplementary Table S2.

Supplementary Table S3.

Supplementary Figure S1.

REFERENCES

  • Allison DB, Downey M, Atkinson RL.et al. Obesity as a disease: a white paper on evidence and arguments commissioned by the Council of the Obesity Society Obesity (Silver Spring) 2008161161–1177. [PubMed] [Google Scholar]
  • Pi-Sunyer FX. A review of long-term studies evaluating the efficacy of weight loss in ameliorating disorders associated with obesity. Clin Ther. 1996;18:1006–35; discussion 1005. [PubMed] [Google Scholar]
  • Franz MJ, VanWormer JJ, Crain AL.et al. Weight-loss outcomes: a systematic review and meta-analysis of weight-loss clinical trials with a minimum 1-year follow-up J Am Diet Assoc 20071071755–1767. [PubMed] [Google Scholar]
  • Rothman RB, Baumann MH. Appetite suppressants, cardiac valve disease and combination pharmacotherapy. Am J Ther. 2009;16:354–364. [PMC free article] [PubMed] [Google Scholar]
  • Wilding J, Van Gaal L, Rissanen A, Vercruysse F, Fitchet M, OBES-002 Study Group A randomized double-blind placebo-controlled study of the long-term efficacy and safety of topiramate in the treatment of obese subjects. Int J Obes Relat Metab Disord. 2004;28:1399–1410. [PubMed] [Google Scholar]
  • Bray GA, Hollander P, Klein S.et al. A 6-month randomized, placebo-controlled, dose-ranging trial of topiramate for weight loss in obesity Obes Res 200311722–733. [PubMed] [Google Scholar]
  • Tonstad S, Tykarski A, Weissgarten J.et al. Efficacy and safety of topiramate in the treatment of obese subjects with essential hypertension Am J Cardiol 200596243–251. [PubMed] [Google Scholar]
  • Richard D, Ferland J, Lalonde J, Samson P, Deshaies Y. Influence of topiramate in the regulation of energy balance. Nutrition. 2000;16:961–966. [PubMed] [Google Scholar]
  • Richard D, Picard F, Lemieux C.et al. The effects of topiramate and sex hormones on energy balance of male and female rats Int J Obes Relat Metab Disord 200226344–353. [PubMed] [Google Scholar]
  • Picard F, Deshaies Y, Lalonde J, Samson P, Richard D. Topiramate reduces energy and fat gains in lean (Fa/?) and obese (fa/fa) Zucker rats. Obes Res. 2000;8:656–663. [PubMed] [Google Scholar]
  • Ben-Menachem E, Axelsen M, Johanson EH, Stagge A, Smith U. Predictors of weight loss in adults with topiramate-treated epilepsy. Obes Res. 2003;11:556–562. [PubMed] [Google Scholar]
  • Astrup A, Caterson I, Zelissen P.et al. Topiramate: long-term maintenance of weight loss induced by a low-calorie diet in obese subjects Obes Res 2004121658–1669. [PubMed] [Google Scholar]
  • Tremblay A, Chaput JP, Bérubé-Parent S.et al. The effect of topiramate on energy balance in obese men: a 6-month double-blind randomized placebo-controlled study with a 6-month open-label extension Eur J Clin Pharmacol 200763123–134. [PubMed] [Google Scholar]
  • Toplak H, Hamann A, Moore R.et al. Efficacy and safety of topiramate in combination with metformin in the treatment of obese subjects with type 2 diabetes: a randomized, double-blind, placebo-controlled study Int J Obes (Lond) 200731138–146. [PubMed] [Google Scholar]
  • Brauer LH, Johanson CE, Schuster CR, Rothman RB, de Wit H. Evaluation of phentermine and fenfluramine, alone and in combination, in normal, healthy volunteers. Neuropsychopharmacology. 1996;14:233–241. [PubMed] [Google Scholar]
  • Magill RA, Waters WF, Bray GA.et al. Effects of tyrosine, phentermine, caffeine D-amphetamine, and placebo on cognitive and motor performance deficits during sleep deprivation Nutr Neurosci 20036237–246. [PubMed] [Google Scholar]
  • Topamax [product information] Ortho-McNeil-Janssen Pharmaceuticals, Inc.: Titusville, NJ; 2009. [Google Scholar]
  • Ryan D, Peterson C, Troupin B.et al. Weight loss at 6 months with VI-0521 (PHEN/TPM combination) treatment Obes Facts 20103146 [Google Scholar]
  • Brownell KD.The LEARN Program for Weight Management10th edn. American Health Publishing Company: Dallas; 2004 [Google Scholar]
  • Simons-Morton DG, Obarzanek E, Cutler JA. Obesity research–limitations of methods, measurements, and medications. JAMA. 2006;295:826–828. [PubMed] [Google Scholar]
  • Gadde KM, Yonish GM, Foust MS, Tam PY, Najarian T. A 24-week randomized controlled trial of VI-0521, a combination weight loss therapy, in obese adults [abstract] Obesity. 2006;14 Suppl:A17–A18. [Google Scholar]
  • US Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research . Guidance for Industry: Developing Products for Weight Management. Food and Drug Administration: Rockville; 2007. [Google Scholar]
  • Lachin JM. Statistical considerations in the intent-to-treat principle. Control Clin Trials. 2000;21:167–189. [PubMed] [Google Scholar]
  • Elobeid MA, Padilla MA, McVie T.et al. Missing data in randomized clinical trials for weight loss: scope of the problem, state of the field, and performance of statistical methods PLoS ONE 20094e6624. [PMC free article] [PubMed] [Google Scholar]
  • Robertson HT, Allison DB. Drugs associated with more suicidal ideations are also associated with more suicide attempts. PLoS ONE. 2009;4:e7312. [PMC free article] [PubMed] [Google Scholar]
  • Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults Executive summary of the third report of the National Cholesterol Education Program (NCEP) expert panel on detection, evaluation, and treatment of high blood cholesterol in adults (adult treatment panel III) JAMA. 2001;285:2486–2497. [PubMed] [Google Scholar]
  • Shao J, Jordan DC, Pritchett YL. Baseline observation carry forward: reasoning, properties, and practical issues. J Biopharm Stat. 2009;19:672–684. [PubMed] [Google Scholar]
  • Mølgaard-Nielsen D, Hviid A. Newer-generation antiepileptic drugs and the risk of major birth defects. JAMA. 2011;305:1996–2002. [PubMed] [Google Scholar]
  • Adipex-P [product information] Teva Pharmaceuticals USA: Sellersville, PA; 2005. [Google Scholar]
  • James WP, Caterson ID, Coutinho W, SCOUT Investigators et al. Effect of sibutramine on cardiovascular outcomes in overweight and obese subjects. N Engl J Med. 2010;363:905–917. [PubMed] [Google Scholar]
  • National Heart, Lung, and Blood Institute The Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults: The Evidence Report National Institutes of Health: Bethesda; 1998. NIH Publication No. 98-4083. [Google Scholar]
  • Flegal KM, Carroll MD, Ogden CL, Curtin LR. Prevalence and trends in obesity among US adults, 1999-2008. JAMA. 2010;303:235–241. [PubMed] [Google Scholar]
  • Smith SR, Weissman NJ, Anderson CM, Behavioral Modification and Lorcaserin for Overweight and Obesity Management (BLOOM) Study Group et al. Multicenter, placebo-controlled trial of lorcaserin for weight management. N Engl J Med. 2010;363:245–256. [PubMed] [Google Scholar]
  • Greenway FL, Fujioka K, Plodkowski RA, COR-I Study Group et al. Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (COR-I): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2010;376:595–605. [PubMed] [Google Scholar]
  • Gadbury GL, Coffey CS, Allison DB. Modern statistical methods for handling missing repeated measurements in obesity trial data: beyond LOCF. Obes Rev. 2003;4:175–184. [PubMed] [Google Scholar]
  • Astrup A, Rössner S. Lessons from obesity management programmes: greater initial weight loss improves long-term maintenance. Obes Rev. 2000;1:17–19. [PubMed] [Google Scholar]
  • Gadde KM, Allison DB, Ryan DH.et al. Effects of low-dose, controlled-release, phentermine plus topiramate combination on weight and associated comorbidities in overweight and obese adults (CONQUER): a randomised, placebo-controlled, phase 3 trial Lancet 20113771341–1352. [PubMed] [Google Scholar]
  • Rucker D, Padwal R, Li SK, Curioni C, Lau DC. Long term pharmacotherapy for obesity and overweight: updated meta-analysis. BMJ. 2007;335:1194–1199. [PMC free article] [PubMed] [Google Scholar]
  • Case CC, Jones PH, Nelson K, O'Brian Smith E, Ballantyne CM. Impact of weight loss on the metabolic syndrome. Diabetes Obes Metab. 2002;4:407–414. [PubMed] [Google Scholar]
  • Goldstein DJ. Beneficial health effects of modest weight loss. Int J Obes Relat Metab Disord. 1992;16:397–415. [PubMed] [Google Scholar]
  • UK Prospective Diabetes Study 7: response of fasting plasma glucose to diet therapy in newly presenting type II diabetic patients, UKPDS group. Metabolism. 1990;39:905–912. [PubMed] [Google Scholar]
  • Buchwald H, Avidor Y, Braunwald E.et al. Bariatric surgery: a systematic review and meta-analysis JAMA 20042921724–1737. [PubMed] [Google Scholar]
  • O'Brien PE, Dixon JB, Laurie C.et al. Treatment of mild to moderate obesity with laparoscopic adjustable gastric banding or an intensive medical program: a randomized trial Ann Intern Med 2006144625–633. [PubMed] [Google Scholar]
  • Rubin DB. Multiple Imputation for Non-Response in Surveys. Wiley Series in Probability and Mathematical Statistics. John Wiley & Sons: New York; 1987. [Google Scholar]
  • Schafer JL. Chapman & Hall/CRC: London; 1997. Analysis of Incomplete Multivariate Data. (Monographs on Statistics and Applied Probability 72). [Google Scholar]

Articles from Obesity (Silver Spring, Md.) are provided here courtesy of Nature Publishing Group

-