Abstract

Context

The thermoregulatory center in the hypothalamus is stimulated by neurokinin 3 receptor (NK3R) activation and inhibited by estrogen-negative feedback. This balance is disrupted in menopause, producing vasomotor symptoms (VMSs).

Objective

To evaluate safety and efficacy of the NK3R antagonist fezolinetant in menopausal VMSs.

Design

Twelve-week, double-blind, randomized, placebo-controlled study.

Setting

Eight Belgian centers from September 2015 to October 2016.

Participants

Generally healthy menopausal women aged 40 to 65 years with moderate/severe VMSs.

Interventions

Subjects were randomized (1:1) to 90 mg of fezolinetant twice daily or placebo for 12 weeks.

Main Outcome Measures

Subjects captured VMS severity and frequency using an electronic diary. The primary outcome was change from baseline to week 12 in total VMS score with fezolinetant vs placebo. Secondary outcomes included timing of changes in frequency and severity of moderate/severe VMSs and quality-of-life assessments at weeks 4, 8, and 12. Pharmacodynamic and pharmacokinetic effects were assessed, as were safety and tolerability.

Results

Of 122 subjects screened, 87 were randomized and 80 (92%) completed the study. At week 12, fezolinetant significantly reduced total VMS score vs placebo (−26.5 vs −12.2, P < 0.001) and decreased mean frequency of moderate/severe VMSs by five episodes per day vs placebo. Severity and frequency of moderate/severe VMSs were reduced from the first day of treatment. Improvements were achieved in all quality-of-life measures. Fezolinetant was well tolerated. The most common fezolinetant-related adverse event was gastrointestinal disorder (n = 6).

Conclusions

Fezolinetant rapidly and significantly reduced moderate/severe VMSs, supporting its potential as an effective nonhormonal treatment option for menopausal women.

Vasomotor symptoms (VMSs) (e.g., hot flashes, night sweats) are reported by up to 80% of women in the United States during the menopausal transition (1) and persist for a median duration of 7.4 years (2). VMSs are the main menopause-related problem prompting women to seek medical treatment (3), and studies indicate that untreated VMSs impart a substantial, direct cost burden on society (4, 5). Menopausal hormone therapy (MHT; also known as hormone replacement therapy) is currently the standard treatment of VMSs, although use continues to decline (6), mainly because of perceived safety concerns, including the risks of venous thromboembolism and breast cancer (7, 8). Although the overall risk-benefit of MHT was recently reassessed (9, 10), concerns persist and women continue to seek alternatives for treatment of VMSs (11, 12).

VMSs are caused by a loss of thermoregulatory control. The thermoregulatory center in the hypothalamus is innervated by kisspeptin/neurokinin B/dynorphin (KNDy) neurons (1318) that are stimulated by neurokinin B (NKB) (1922) and inhibited by estrogen (2325) as part of the feedback mechanism that supports female reproductive endocrinology and fertility (1316, 1925). This balance is disrupted with the decline of estrogen levels in menopause, and it has been proposed that the consequent, unregulated KNDy neuron activation is a key, underlying component of VMSs (26, 27). Thus, negative modulation of KNDy neuron signaling via antagonism of NKB signaling at its cognate receptor [neurokinin 3 receptor (NK3R)] may offer an alternative mechanism to MHT for control of VMSs (16).

Fezolinetant (ESN364) (28) is an oral NK3R antagonist that is currently in clinical development for VMSs associated with menopause. Fezolinetant selectively and reversibly blocks NKB signaling, slowing GnRH pulse frequency consistent with a decrease in KNDy neuron activity (29, 30). In this report, we present the efficacy and safety profile of fezolinetant from the first pilot/phase 2a clinical trial of an NK3 antagonist conducted according to US Food and Drug Administration (FDA) guidelines for the clinical evaluation of treatments for VMSs (31, 32). The primary objective of the study was to evaluate the effect of fezolinetant on the severity and frequency of VMSs in menopausal women. Secondary objectives were to evaluate the effect of fezolinetant on (i) time course of effects on VMSs, (ii) quality of life, (iii) pharmacodynamic effects on reproductive hormones, and (iv) safety and tolerability. Assessment of fezolinetant pharmacokinetics was an exploratory objective.

Materials and Methods

Study design

This was a 12-week, double-blind, placebo-controlled study in menopausal women, designed in accordance with FDA guidance (31) and conducted from September 2015 to October 2016 at eight Belgian clinical centers. Following a ≤4-week screening period, subjects were randomized on day 1 to receive placebo or 90 mg of fezolinetant twice daily for 12 weeks. Subjects returned to the clinic for assessment at weeks 4, 8, and 12 and were followed for 2 to 3 weeks after the end of treatment.

Randomization was based on a computer-generated schedule, balanced using randomly permuted blocks across treatment groups without stratification, and prepared by SGS Life Science Services using SAS® software (SAS Institute, Cary, NC). Subjects, investigators, other employees of the clinical site, and the sponsor were blinded to treatment assignment. To ensure blinding, the placebo was identical to active treatment with regard to appearance, taste, and smell.

All subjects provided written informed consent prior to the study. The study protocol was approved by an Independent Ethics Committee at each center. This study has been registered with EudraCT (2015-002578-20).

Subject eligibility criteria

This study enrolled women aged 40 to 65 years in good general health who had reached menopause and were experiencing moderate or severe VMSs. Menopause was defined as spontaneous amenorrhea for ≥12 consecutive months; or spontaneous amenorrhea for ≥6 months with biochemical criteria of menopause (FSH >40 IU/L); or spontaneous amenorrhea for ≥3 months with FSH >40 IU/L and estradiol (E2) <0.21 nmol/L; or bilateral oophorectomy ≥6 weeks prior to screening. Only subjects who recorded ≥7 episodes of moderate/severe VMSs (hot flashes or night sweats) per day over a period of 7 consecutive days (i.e., ≥49 moderate/severe VMS episodes) during the screening period were included in the study.

Subjects were excluded from the study if they had any medical condition that could confound results (e.g., history of drug/alcohol abuse, suicide event in the last 3 years, active liver disease/jaundice, elevated liver function enzymes, impaired kidney function). Women with a recent history (within 1 year prior to screening) of a psychological disorder according to Diagnostic and Statistical Manual of Mental Disorders (4th edition) criteria were excluded; such disorders included current major depression.

Women were also excluded if they were taking prohibited medications. These medications were defined as drugs that could interfere with the occurrence of VMSs, including selective serotonin reuptake inhibitors, selective norepinephrine reuptake inhibitors, tricyclic antidepressants, sedatives, or hypnotics. Use of hormonal medications, such as MHT, hormonal contraception, or any treatment of hot flashes (prescription, over the counter, or herbal), was not allowed during the study; these treatments were to be discontinued at least 6 weeks prior to screening.

Study drug administration

Subjects were sequentially randomized (1:1 ratio according to the randomization schedule) to receive placebo or 90 mg of fezolinetant 90 twice daily, self-administered as a single gelatin capsule with water and a light meal in the morning and in the evening. On days when site visits where scheduled, treatment was administered under fasting condition at the clinical site. Compliance was assessed by counting returned dosage units in addition to electronic patient-reported outcome (ePRO) entries upon study medication intake.

Study assessments and related outcomes

VMS severity and frequency were recorded at least twice daily from screening through follow-up visits using an ePRO diary. Data collected during the screening period were used to determine the baseline values for VMS score and severity and frequency of moderate/severe VMSs. The electronic diaries were kept by the patients from screening onward through the end-of-study visit. Patients were instructed to fill out the diaries at least twice daily for the duration of the study. The data recorded for ≥7 days during the screening period were used to assess eligibility.

As defined in FDA guidelines (31), the severity of VMSs was classified as mild (sensation of heat without sweating; at night, the subject did not wake up but later noticed damp sheets or clothing), moderate (sensation of heat with sweating; at night, subject woke up), and severe [sensation of heat with sweating, causing cessation of activity; at night, subject woke up and took action (e.g., opening window)]. As a measure of severity, the mean daily total VMS score, a composite combining both severity and frequency data, was used. The mean daily total VMS score during a given period was calculated by multiplying the number of mild, moderate, or severe VMS episodes during the period by 1, 2, or 3, respectively, summing the values and dividing by the number of days in the period (33). A higher score indicates worse symptoms; there is no maximum score.

Frequency (number of episodes) and severity score (calculated as for total VMS score) of moderate/severe VMSs were assessed at weeks 4, 8, 12, and at follow-up, taking into account only moderate and severe symptoms, as per guidelines (31). Post hoc analysis evaluated the response each day during the first week of treatment.

The ePROs included quality-of-life questionnaires to assess perceived daily interference using the Hot Flash Related Daily Interference Scale (HFRDIS) (34), sleep quality using the Leeds Sleep Evaluation Questionnaire (LSEQ) (35), changes in climacteric symptoms using the Greene Climacteric Scale (GCS) (36), and daily functional impairment using the Sheehan Disability Scale (SDS) (37, 38) (all evaluated at baseline and at weeks 4, 8, and 12).

Blood samples for pharmacokinetic assessments were collected predose at baseline and weeks 4, 8, and 12, and 3 hours postdose at week 12 into vacuum tubes containing lithium heparin (Venoject green top or equivalent). Samples were centrifuged at 4°C for 10 minutes at ≈1500 × g and plasma was stored frozen at −20°C or below until analysis. Blood samples for the assessment of hormone biomarkers (including LH, E2, FSH, and SHBG levels) were collected at screening and predose at baseline and weeks 4, 8, and 12, and 3 hours postdose at week 12 [coincident with the previously determined tmax of fezolinetant (30)], and at follow-up. After appropriate labeling, plasma samples were stored frozen at −70°C or below until analysis.

Safety and tolerability were assessed by the occurrence of treatment-emergent adverse events (TEAEs), physical examination results [including suicidal behavior using Columbia-Suicide Severity Rating Scale (39)], clinical laboratory tests, vital signs, electrocardiograms, and plasma bone density markers (bone alkaline phosphatase and carboxyl-terminal telopeptide of type I collagen). Adverse events were monitored continuously from informed consent until the last study-related activity. At regular intervals during the study, participants were asked nonleading questions to determine the occurrence of any adverse events. All adverse events reported spontaneously during the course of the study were recorded as well.

The primary endpoint was change from baseline to week 12 in daily VMS score (averaged during the course of a week). Secondary endpoints included changes from baseline over time in weekly VMS frequency and severity scores, as well as scores on the HFRDIS, LSEQ, GCS, and SDS.

Analytical methods

Fezolinetant concentration was determined in lithium-heparinized human plasma using a validated liquid chromatography method with tandem mass spectrometric detection. Plasma samples for pharmacodynamic assessments were analyzed by Barc Laboratory (Gent, Belgium) using previously validated analytical methods: FSH, LH, and SHBG levels were determined using electrochemiluminescence immunoassay analyzed on a Roche Diagnostics cobas e immunoassay analyzer; E2 levels were determined using chemiluminescent microparticle immunoassay using an LKB gamma counter or PerkinElmer Wizard.

Statistical methods

A total of 80 subjects were planned to be randomized. Given the exploratory nature of this study and the lack of information on the effect size of NK3 antagonism, no formal power calculations were performed. The sample size determination was based on general recommendations for studies using VMS diary data, as well as approximate placebo and treatment effects from previous studies related to VMSs, while still allowing for some dropouts (33, 40). Additional statistical analyses were also conducted for further insight.

The safety population, used for biomarker, pharmacokinetic, and safety analyses, included all subjects who were randomized into the study and received at least one dose of the study drug. The modified intent-to-treat (mITT) population, used for all efficacy analyses, included all randomized subjects who received at least one dose of the study medication and who had postbaseline efficacy data. A per-protocol population, used only for sensitivity analyses on the primary endpoint, included those from the mITT population who had no major protocol violations.

Descriptive statistics were used to summarize changes in VMSs from baseline over time. Differences between treatment means were analyzed using an analysis of covariance model with treatment as fixed effect and baseline as covariate (observed cases approach). Quality-of-life questionnaire (HFRDIS, GCS, SDS, and LSEQ) results were evaluated using similar analysis of covariance models as well as descriptive statistics of actual values and changes from baseline. P values for secondary endpoints were interpreted descriptively, and no adjustment for multiplicity was made. Descriptive statistics were used to summarize safety data.

Baseline VMS data were calculated from the week preceding randomization. Safety and efficacy parameters were based on observed cases. Two sensitivity analyses were performed on the primary endpoint, one in which missing values were imputed using a last observation carried forward approach and one using the per-protocol population (based on observed cases without imputation).

Statistical analyses were performed by SGS Life Sciences using SAS® software (SAS Institute; version 9.2 or higher) for statistical computations and interpreted using a two-sided significance level of 0.05.

Results

Subject disposition and baseline characteristics

Of 122 subjects screened, 87 were randomized to receive fezolinetant (n = 43; 93% completed the study) or placebo (n = 44; 91% completed the study) (Fig. 1). All subjects were compliant throughout the study, except for one (later revealed as receiving placebo) who discontinued the study because of lack of improvement of her symptoms.

Patient disposition. *One subject randomized to receive fezolinetant treatment did not meet the inclusion criterion of ≥7 episodes of moderate or severe VMSs (hot flashes or night sweats) during a period of 7 consecutive days (i.e., ≥49 moderate/severe VMS episodes) during the screening period. This subject also did not meet the exclusion criteria relating to active liver disease/jaundice, elevated liver function enzymes, and impaired kidney function. This subject completed the trial. **Four subjects randomized to placebo did not meet inclusion or exclusion criteria. Two of these subjects did not meet inclusion criteria relating to definition of menopause, although they met criteria for VMSs. A different subject randomized to placebo did not meet the inclusion criterion of ≥7 episodes of moderate/severe VMSs (hot flashes or night sweats) during a period of 7 consecutive days (i.e., ≥49 moderate or severe VMS episodes) during the screening period. A further subject randomized to placebo did not meet the exclusion criteria relating to active liver disease/jaundice, elevated liver function enzymes, and impaired kidney function. Three of these subjects completed the trial. ***Considered possibly related to treatment (one subject with preexisting fibromyalgia experienced worsening of fibromyalgia, depression, dry mouth, headache, palpitations, diarrhea, and vomiting; one subject experienced headache and vertigo).
Figure 1.

Patient disposition. *One subject randomized to receive fezolinetant treatment did not meet the inclusion criterion of ≥7 episodes of moderate or severe VMSs (hot flashes or night sweats) during a period of 7 consecutive days (i.e., ≥49 moderate/severe VMS episodes) during the screening period. This subject also did not meet the exclusion criteria relating to active liver disease/jaundice, elevated liver function enzymes, and impaired kidney function. This subject completed the trial. **Four subjects randomized to placebo did not meet inclusion or exclusion criteria. Two of these subjects did not meet inclusion criteria relating to definition of menopause, although they met criteria for VMSs. A different subject randomized to placebo did not meet the inclusion criterion of ≥7 episodes of moderate/severe VMSs (hot flashes or night sweats) during a period of 7 consecutive days (i.e., ≥49 moderate or severe VMS episodes) during the screening period. A further subject randomized to placebo did not meet the exclusion criteria relating to active liver disease/jaundice, elevated liver function enzymes, and impaired kidney function. Three of these subjects completed the trial. ***Considered possibly related to treatment (one subject with preexisting fibromyalgia experienced worsening of fibromyalgia, depression, dry mouth, headache, palpitations, diarrhea, and vomiting; one subject experienced headache and vertigo).

Demographics and baseline characteristics were well matched between the placebo and fezolinetant groups (Table 1). All subjects were women, with a mean age of 53.5 years (range, 44 to 64 years) and mean (SD) body mass index of 25.8 (5.49) kg/m2. Mean daily total VMS score at baseline was 25.8 (95% CI, 22.6, 28.9) in the placebo group and 28.8 (95% CI, 24.6, 32.9) in the fezolinetant group. At baseline, mean frequency of moderate/severe VMSs was 72.0 episodes per week (95% CI, 63.9, 80.1) in the placebo group and 80.7 episodes per week (95% CI, 70.6, 90.8) in the fezolinetant group. Mean daily moderate/severe VMS score at baseline was 24.9 (95% CI, 21.7, 28.1) in the placebo group and 27.8 (95% CI, 23.7, 32.0) in the fezolinetant group.

Table 1.

Subject Demographics and Baseline Characteristics (Safety Population)

Placebo Twice Daily (n = 44)Fezolinetant (90 mg) Twice Daily (n = 43)
Race, n (%)
 White44 (100)42 (97.7)
 Multiple01 (2.3)
Age, y, mean (SD)53.7 (4.25)53.3 (4.03)
Height, cm, mean (SD)163.6 (6.04)165.7 (5.53)
Weight, kg, mean (SD)70.8 (16.27)69.0 (13.19)
Body mass index, mean kg/m2 (range)26.5 (6.15)25.1 (4.71)
Total VMS score, daily (measured during 1 wk)
 Mean25.828.8
 95% CI22.6, 28.924.6, 32.9
Frequency of moderate/severe VMSs, number per wk
 Mean72.080.7
 95% CI63.9, 80.170.6, 90.8
Moderate/severe VMS score, daily (measured during 1 wk)
 Mean24.927.8
 95% CI21.7, 28.123.7, 32.0
Placebo Twice Daily (n = 44)Fezolinetant (90 mg) Twice Daily (n = 43)
Race, n (%)
 White44 (100)42 (97.7)
 Multiple01 (2.3)
Age, y, mean (SD)53.7 (4.25)53.3 (4.03)
Height, cm, mean (SD)163.6 (6.04)165.7 (5.53)
Weight, kg, mean (SD)70.8 (16.27)69.0 (13.19)
Body mass index, mean kg/m2 (range)26.5 (6.15)25.1 (4.71)
Total VMS score, daily (measured during 1 wk)
 Mean25.828.8
 95% CI22.6, 28.924.6, 32.9
Frequency of moderate/severe VMSs, number per wk
 Mean72.080.7
 95% CI63.9, 80.170.6, 90.8
Moderate/severe VMS score, daily (measured during 1 wk)
 Mean24.927.8
 95% CI21.7, 28.123.7, 32.0
Table 1.

Subject Demographics and Baseline Characteristics (Safety Population)

Placebo Twice Daily (n = 44)Fezolinetant (90 mg) Twice Daily (n = 43)
Race, n (%)
 White44 (100)42 (97.7)
 Multiple01 (2.3)
Age, y, mean (SD)53.7 (4.25)53.3 (4.03)
Height, cm, mean (SD)163.6 (6.04)165.7 (5.53)
Weight, kg, mean (SD)70.8 (16.27)69.0 (13.19)
Body mass index, mean kg/m2 (range)26.5 (6.15)25.1 (4.71)
Total VMS score, daily (measured during 1 wk)
 Mean25.828.8
 95% CI22.6, 28.924.6, 32.9
Frequency of moderate/severe VMSs, number per wk
 Mean72.080.7
 95% CI63.9, 80.170.6, 90.8
Moderate/severe VMS score, daily (measured during 1 wk)
 Mean24.927.8
 95% CI21.7, 28.123.7, 32.0
Placebo Twice Daily (n = 44)Fezolinetant (90 mg) Twice Daily (n = 43)
Race, n (%)
 White44 (100)42 (97.7)
 Multiple01 (2.3)
Age, y, mean (SD)53.7 (4.25)53.3 (4.03)
Height, cm, mean (SD)163.6 (6.04)165.7 (5.53)
Weight, kg, mean (SD)70.8 (16.27)69.0 (13.19)
Body mass index, mean kg/m2 (range)26.5 (6.15)25.1 (4.71)
Total VMS score, daily (measured during 1 wk)
 Mean25.828.8
 95% CI22.6, 28.924.6, 32.9
Frequency of moderate/severe VMSs, number per wk
 Mean72.080.7
 95% CI63.9, 80.170.6, 90.8
Moderate/severe VMS score, daily (measured during 1 wk)
 Mean24.927.8
 95% CI21.7, 28.123.7, 32.0

Efficacy

At week 12, mean daily total VMS score was 14.4 (95% CI, 9.8, 19.0) with placebo and 2.7 (95% CI, 1.4, 4.0) with fezolinetant. On the primary endpoint, fezolinetant resulted in a significantly greater reduction in daily total VMS score from baseline to week 12 (−26.5; 95% CI, −30.8, −22.2) than placebo [−12.2; 95% CI, −16.5, −7.8; least squares mean difference (LSMD) −12.3; 95% CI, −16.9, −7.8; P < 0.001]. Mean daily total VMS score was also reduced with fezolinetant compared with placebo at week 4 and week 8 (Fig. 2A). Sensitivity analyses performed on the per protocol population (observed cases) and on the mITT population using a last observation carried forward imputation method were consistent with the primary analysis and confirmed the statistically significant difference (P < 0.001) in VMS score between fezolinetant and placebo at week 12.

Effect of fezolinetant on VMSs over time. (A) Daily total VMS score during week 4 and week 12. (B) Frequency of moderate/severe VMSs during the duration of the study. (C) Moderate/severe VMS score during the duration of the study. Data are presented as mean ± 95% CI (mITT population, observed data).
Figure 2.

Effect of fezolinetant on VMSs over time. (A) Daily total VMS score during week 4 and week 12. (B) Frequency of moderate/severe VMSs during the duration of the study. (C) Moderate/severe VMS score during the duration of the study. Data are presented as mean ± 95% CI (mITT population, observed data).

At week 12, mean frequency of moderate/severe VMSs was 39.0 episodes per week (95% CI, 26.6, 51.5) with placebo and 5.7 episodes per week (95% CI, 2.4, 9.1) with fezolinetant. Relative to baseline, VMS frequency was reduced by 93% with fezolinetant compared with 46% with placebo. Fezolinetant resulted in a greater reduction from baseline in frequency of moderate/severe VMSs (−76.1 episodes per week; 95% CI, −87.2, −65.0) than placebo (−35.3 episodes per week; 95% CI, −46.9, −23.6; LSMD, −35.2; 95% CI, −47.6, −22.8; P < 0.001). Fezolinetant also reduced the frequency of moderate/severe VMSs more than placebo at week 4 and at all other time periods from the first day of treatment (Fig. 2B).

At week 12, the mean daily moderate/severe VMS score was 13.5 (95% CI, 8.9, 18.2) with placebo and 1.7 (95% CI, 0.7, 2.7) with fezolinetant. Fezolinetant resulted in a greater reduction from baseline in daily moderate/severe VMS score (−26.6; 95% CI, −31.1, −22.2) than placebo (−12.1; 95% CI, −16.6, −7.7; LSMD, −12.4; 95% CI, −17.0, −7.8; P < 0.001), equivalent to five fewer moderate/severe VMS episodes each day. Fezolinetant also reduced the mean daily moderate/severe VMS score more than placebo at week 4 and at all other time periods from the first day of treatment (Fig. 2C).

Fezolinetant treatment resulted in improvement from baseline in sleep quality, overall daily interference, climacteric symptoms, and function at weeks 4, 8, and 12 (Fig. 3). Of the 22 additional subdomains in the questionnaires used, 18 showed improvement from baseline at week 12 of fezolinetant treatment, with most (n = 16) improved at the first time assessed (Table 2). There was no significant impact of fezolinetant at any time point on physical symptoms and loss of interest in sex, as assessed by the GCS.

Effect of fezolinetant on quality-of-life measures. (A) LSEQ (does not include a total mean score that incorporates all four dimensions assessed). (B) HFRDIS. (C) GCS. (D) SDS. Data are presented as mean ± 95% CI (mITT population).
Figure 3.

Effect of fezolinetant on quality-of-life measures. (A) LSEQ (does not include a total mean score that incorporates all four dimensions assessed). (B) HFRDIS. (C) GCS. (D) SDS. Data are presented as mean ± 95% CI (mITT population).

Table 2.

Effect of Fezolinetant on Subdomains of Quality-of-Life Measures (mITT Population)

SubdomainWeek 4Week 8Week 12
LSMD for Fezolinetant vs Placebo95% CIP ValueLSMD for Fezolinetant vs Placebo95% CIP ValueLSMD for Fezolinetant vs Placebo95% CIP Value
LSEQ
 Quality of sleep2.4231.308, 3.539<0.0012.2911.331, 3.251<0.0012.4331.334, 3.532<0.001
 Getting to sleep1.3750.651, 2.100<0.0011.1660.505, 1.827<0.0010.8950.190, 1.5990.014
 Awake following sleep0.877−0.034, 1.7890.0591.4570.579, 2.3350.0011.1130.107, 2.1200.031
 Behavior following wakening1.2030.317, 2.0880.0081.5970.639, 2.5560.0010.842−0.116, 1.8000.084
HFRDIS
 Overall mean score−1.98−2.73, −1.23<0.001−2.21−3.02, −1.40<0.001−1.98−2.83, −1.13<0.001
 Work−2.53−3.45, −1.60<0.001−2.29−3.15, −1.42<0.001−2.11−3.03, −1.20<0.001
 Social activities−2.33−3.19, −1.46<0.001−2.34−3.21, −1.47<0.001−2.19−3.09, −1.29<0.001
 Leisure activities−2.15−2.99, −1.31<0.001−2.37−3.19, −1.55<0.001−1.64−2.66, −0.620.002
 Sleep−2.93−4.08, −1.78<0.001−2.71−3.86, −1.57<0.001−2.61−3.67, −1.54<0.001
 Mood−1.74−2.67, −0.81<0.001−2.12−3.09, −1.16<0.001−1.79−2.69, −0.88<0.001
 Concentration−1.92−2.86, −0.97<0.001−2.23−3.21, −1.25<0.001−1.90−2.91, −0.88<0.001
 Relations with others−1.33−2.20, −0.460.003−1.85−2.80, −0.90<0.001−1.72−2.66, −0.77<0.001
 Sexuality−1.06−2.25, 0.130.081−1.40−2.80, 0.000.050−1.51−2.84, −0.190.026
 Enjoyment of life−1.65−2.51, −0.79<0.001−2.07−2.97, −1.18<0.001−1.83−2.88, −0.78<0.001
 Overall quality of life−1.92−2.86, −0.97<0.001−2.51−3.41, −1.61<0.001−2.36−3.31, −1.41<0.001
GCS
 Total score−4.6−8.2, −1.00.013−6.9−10.7, −3.1<0.001−6.3−9.9, −2.8<0.001
 Loss of interest in sex0.0−0.3, 0.30.881−0.2−0.6, 0.20.383−0.2−0.6, 0.20.301
 Psychological−2.5−4.5, −0.40.020−3.3−5.4, −1.20.003−3.1−5.2, −1.00.005
 Physical−0.2−1.2, 0.90.732−0.7−1.9, 0.60.282−0.6−1.7, 0.50.254
 Vasomotor−2.0−2.7, −1.4<0.001−1.8−2.4, −1.1<0.001−2.0−2.6, −1.3<0.001
SDS
 Global functional impairment−4.4−6.9, −2.0<0.001−5.8−8.0, −3.6<0.001−5.3−7.8, −2.8<0.001
 Work/school−1.7−2.7, −0.8<0.001−2.0−2.8, −1.3<0.001−1.6−2.5, −0.8<0.001
 Social life−1.4−2.2, −0.6<0.001−1.9−2.7, −1.1<0.001−1.6−2.4, −0.7<0.001
 Family life/home responsibilities−1.3−2.2, −0.40.005−1.7−2.5, −0.9<0.001−1.6−2.4, −0.7<0.001
 Days lost0.0−0.5, 0.60.9360.0−0.1, 0.10.884−0.2−0.4, 0.10.124
 Days unproductive−0.9−1.8, 0.00.052−0.9−1.7, 0.00.060−0.8−1.7, 0.00.049
SubdomainWeek 4Week 8Week 12
LSMD for Fezolinetant vs Placebo95% CIP ValueLSMD for Fezolinetant vs Placebo95% CIP ValueLSMD for Fezolinetant vs Placebo95% CIP Value
LSEQ
 Quality of sleep2.4231.308, 3.539<0.0012.2911.331, 3.251<0.0012.4331.334, 3.532<0.001
 Getting to sleep1.3750.651, 2.100<0.0011.1660.505, 1.827<0.0010.8950.190, 1.5990.014
 Awake following sleep0.877−0.034, 1.7890.0591.4570.579, 2.3350.0011.1130.107, 2.1200.031
 Behavior following wakening1.2030.317, 2.0880.0081.5970.639, 2.5560.0010.842−0.116, 1.8000.084
HFRDIS
 Overall mean score−1.98−2.73, −1.23<0.001−2.21−3.02, −1.40<0.001−1.98−2.83, −1.13<0.001
 Work−2.53−3.45, −1.60<0.001−2.29−3.15, −1.42<0.001−2.11−3.03, −1.20<0.001
 Social activities−2.33−3.19, −1.46<0.001−2.34−3.21, −1.47<0.001−2.19−3.09, −1.29<0.001
 Leisure activities−2.15−2.99, −1.31<0.001−2.37−3.19, −1.55<0.001−1.64−2.66, −0.620.002
 Sleep−2.93−4.08, −1.78<0.001−2.71−3.86, −1.57<0.001−2.61−3.67, −1.54<0.001
 Mood−1.74−2.67, −0.81<0.001−2.12−3.09, −1.16<0.001−1.79−2.69, −0.88<0.001
 Concentration−1.92−2.86, −0.97<0.001−2.23−3.21, −1.25<0.001−1.90−2.91, −0.88<0.001
 Relations with others−1.33−2.20, −0.460.003−1.85−2.80, −0.90<0.001−1.72−2.66, −0.77<0.001
 Sexuality−1.06−2.25, 0.130.081−1.40−2.80, 0.000.050−1.51−2.84, −0.190.026
 Enjoyment of life−1.65−2.51, −0.79<0.001−2.07−2.97, −1.18<0.001−1.83−2.88, −0.78<0.001
 Overall quality of life−1.92−2.86, −0.97<0.001−2.51−3.41, −1.61<0.001−2.36−3.31, −1.41<0.001
GCS
 Total score−4.6−8.2, −1.00.013−6.9−10.7, −3.1<0.001−6.3−9.9, −2.8<0.001
 Loss of interest in sex0.0−0.3, 0.30.881−0.2−0.6, 0.20.383−0.2−0.6, 0.20.301
 Psychological−2.5−4.5, −0.40.020−3.3−5.4, −1.20.003−3.1−5.2, −1.00.005
 Physical−0.2−1.2, 0.90.732−0.7−1.9, 0.60.282−0.6−1.7, 0.50.254
 Vasomotor−2.0−2.7, −1.4<0.001−1.8−2.4, −1.1<0.001−2.0−2.6, −1.3<0.001
SDS
 Global functional impairment−4.4−6.9, −2.0<0.001−5.8−8.0, −3.6<0.001−5.3−7.8, −2.8<0.001
 Work/school−1.7−2.7, −0.8<0.001−2.0−2.8, −1.3<0.001−1.6−2.5, −0.8<0.001
 Social life−1.4−2.2, −0.6<0.001−1.9−2.7, −1.1<0.001−1.6−2.4, −0.7<0.001
 Family life/home responsibilities−1.3−2.2, −0.40.005−1.7−2.5, −0.9<0.001−1.6−2.4, −0.7<0.001
 Days lost0.0−0.5, 0.60.9360.0−0.1, 0.10.884−0.2−0.4, 0.10.124
 Days unproductive−0.9−1.8, 0.00.052−0.9−1.7, 0.00.060−0.8−1.7, 0.00.049
Table 2.

Effect of Fezolinetant on Subdomains of Quality-of-Life Measures (mITT Population)

SubdomainWeek 4Week 8Week 12
LSMD for Fezolinetant vs Placebo95% CIP ValueLSMD for Fezolinetant vs Placebo95% CIP ValueLSMD for Fezolinetant vs Placebo95% CIP Value
LSEQ
 Quality of sleep2.4231.308, 3.539<0.0012.2911.331, 3.251<0.0012.4331.334, 3.532<0.001
 Getting to sleep1.3750.651, 2.100<0.0011.1660.505, 1.827<0.0010.8950.190, 1.5990.014
 Awake following sleep0.877−0.034, 1.7890.0591.4570.579, 2.3350.0011.1130.107, 2.1200.031
 Behavior following wakening1.2030.317, 2.0880.0081.5970.639, 2.5560.0010.842−0.116, 1.8000.084
HFRDIS
 Overall mean score−1.98−2.73, −1.23<0.001−2.21−3.02, −1.40<0.001−1.98−2.83, −1.13<0.001
 Work−2.53−3.45, −1.60<0.001−2.29−3.15, −1.42<0.001−2.11−3.03, −1.20<0.001
 Social activities−2.33−3.19, −1.46<0.001−2.34−3.21, −1.47<0.001−2.19−3.09, −1.29<0.001
 Leisure activities−2.15−2.99, −1.31<0.001−2.37−3.19, −1.55<0.001−1.64−2.66, −0.620.002
 Sleep−2.93−4.08, −1.78<0.001−2.71−3.86, −1.57<0.001−2.61−3.67, −1.54<0.001
 Mood−1.74−2.67, −0.81<0.001−2.12−3.09, −1.16<0.001−1.79−2.69, −0.88<0.001
 Concentration−1.92−2.86, −0.97<0.001−2.23−3.21, −1.25<0.001−1.90−2.91, −0.88<0.001
 Relations with others−1.33−2.20, −0.460.003−1.85−2.80, −0.90<0.001−1.72−2.66, −0.77<0.001
 Sexuality−1.06−2.25, 0.130.081−1.40−2.80, 0.000.050−1.51−2.84, −0.190.026
 Enjoyment of life−1.65−2.51, −0.79<0.001−2.07−2.97, −1.18<0.001−1.83−2.88, −0.78<0.001
 Overall quality of life−1.92−2.86, −0.97<0.001−2.51−3.41, −1.61<0.001−2.36−3.31, −1.41<0.001
GCS
 Total score−4.6−8.2, −1.00.013−6.9−10.7, −3.1<0.001−6.3−9.9, −2.8<0.001
 Loss of interest in sex0.0−0.3, 0.30.881−0.2−0.6, 0.20.383−0.2−0.6, 0.20.301
 Psychological−2.5−4.5, −0.40.020−3.3−5.4, −1.20.003−3.1−5.2, −1.00.005
 Physical−0.2−1.2, 0.90.732−0.7−1.9, 0.60.282−0.6−1.7, 0.50.254
 Vasomotor−2.0−2.7, −1.4<0.001−1.8−2.4, −1.1<0.001−2.0−2.6, −1.3<0.001
SDS
 Global functional impairment−4.4−6.9, −2.0<0.001−5.8−8.0, −3.6<0.001−5.3−7.8, −2.8<0.001
 Work/school−1.7−2.7, −0.8<0.001−2.0−2.8, −1.3<0.001−1.6−2.5, −0.8<0.001
 Social life−1.4−2.2, −0.6<0.001−1.9−2.7, −1.1<0.001−1.6−2.4, −0.7<0.001
 Family life/home responsibilities−1.3−2.2, −0.40.005−1.7−2.5, −0.9<0.001−1.6−2.4, −0.7<0.001
 Days lost0.0−0.5, 0.60.9360.0−0.1, 0.10.884−0.2−0.4, 0.10.124
 Days unproductive−0.9−1.8, 0.00.052−0.9−1.7, 0.00.060−0.8−1.7, 0.00.049
SubdomainWeek 4Week 8Week 12
LSMD for Fezolinetant vs Placebo95% CIP ValueLSMD for Fezolinetant vs Placebo95% CIP ValueLSMD for Fezolinetant vs Placebo95% CIP Value
LSEQ
 Quality of sleep2.4231.308, 3.539<0.0012.2911.331, 3.251<0.0012.4331.334, 3.532<0.001
 Getting to sleep1.3750.651, 2.100<0.0011.1660.505, 1.827<0.0010.8950.190, 1.5990.014
 Awake following sleep0.877−0.034, 1.7890.0591.4570.579, 2.3350.0011.1130.107, 2.1200.031
 Behavior following wakening1.2030.317, 2.0880.0081.5970.639, 2.5560.0010.842−0.116, 1.8000.084
HFRDIS
 Overall mean score−1.98−2.73, −1.23<0.001−2.21−3.02, −1.40<0.001−1.98−2.83, −1.13<0.001
 Work−2.53−3.45, −1.60<0.001−2.29−3.15, −1.42<0.001−2.11−3.03, −1.20<0.001
 Social activities−2.33−3.19, −1.46<0.001−2.34−3.21, −1.47<0.001−2.19−3.09, −1.29<0.001
 Leisure activities−2.15−2.99, −1.31<0.001−2.37−3.19, −1.55<0.001−1.64−2.66, −0.620.002
 Sleep−2.93−4.08, −1.78<0.001−2.71−3.86, −1.57<0.001−2.61−3.67, −1.54<0.001
 Mood−1.74−2.67, −0.81<0.001−2.12−3.09, −1.16<0.001−1.79−2.69, −0.88<0.001
 Concentration−1.92−2.86, −0.97<0.001−2.23−3.21, −1.25<0.001−1.90−2.91, −0.88<0.001
 Relations with others−1.33−2.20, −0.460.003−1.85−2.80, −0.90<0.001−1.72−2.66, −0.77<0.001
 Sexuality−1.06−2.25, 0.130.081−1.40−2.80, 0.000.050−1.51−2.84, −0.190.026
 Enjoyment of life−1.65−2.51, −0.79<0.001−2.07−2.97, −1.18<0.001−1.83−2.88, −0.78<0.001
 Overall quality of life−1.92−2.86, −0.97<0.001−2.51−3.41, −1.61<0.001−2.36−3.31, −1.41<0.001
GCS
 Total score−4.6−8.2, −1.00.013−6.9−10.7, −3.1<0.001−6.3−9.9, −2.8<0.001
 Loss of interest in sex0.0−0.3, 0.30.881−0.2−0.6, 0.20.383−0.2−0.6, 0.20.301
 Psychological−2.5−4.5, −0.40.020−3.3−5.4, −1.20.003−3.1−5.2, −1.00.005
 Physical−0.2−1.2, 0.90.732−0.7−1.9, 0.60.282−0.6−1.7, 0.50.254
 Vasomotor−2.0−2.7, −1.4<0.001−1.8−2.4, −1.1<0.001−2.0−2.6, −1.3<0.001
SDS
 Global functional impairment−4.4−6.9, −2.0<0.001−5.8−8.0, −3.6<0.001−5.3−7.8, −2.8<0.001
 Work/school−1.7−2.7, −0.8<0.001−2.0−2.8, −1.3<0.001−1.6−2.5, −0.8<0.001
 Social life−1.4−2.2, −0.6<0.001−1.9−2.7, −1.1<0.001−1.6−2.4, −0.7<0.001
 Family life/home responsibilities−1.3−2.2, −0.40.005−1.7−2.5, −0.9<0.001−1.6−2.4, −0.7<0.001
 Days lost0.0−0.5, 0.60.9360.0−0.1, 0.10.884−0.2−0.4, 0.10.124
 Days unproductive−0.9−1.8, 0.00.052−0.9−1.7, 0.00.060−0.8−1.7, 0.00.049

Pharmacokinetic/pharmacodynamic results

Fezolinetant plasma concentrations are summarized in Table 3.

Table 3.

Plasma Fezolinetant Concentrations (Safety Population)

Mean (SEM) (ng/mL)n
Trough (12 h postdose)
 Week 4283.7 (37.05)31
 Week 8311.8 (40.19)35
 Week 12288.8 (31.45)38
Peak (3 h postdose)
 Week 12845.9 (55.81)38
Mean (SEM) (ng/mL)n
Trough (12 h postdose)
 Week 4283.7 (37.05)31
 Week 8311.8 (40.19)35
 Week 12288.8 (31.45)38
Peak (3 h postdose)
 Week 12845.9 (55.81)38
Table 3.

Plasma Fezolinetant Concentrations (Safety Population)

Mean (SEM) (ng/mL)n
Trough (12 h postdose)
 Week 4283.7 (37.05)31
 Week 8311.8 (40.19)35
 Week 12288.8 (31.45)38
Peak (3 h postdose)
 Week 12845.9 (55.81)38
Mean (SEM) (ng/mL)n
Trough (12 h postdose)
 Week 4283.7 (37.05)31
 Week 8311.8 (40.19)35
 Week 12288.8 (31.45)38
Peak (3 h postdose)
 Week 12845.9 (55.81)38

At peak drug levels (i.e., 3 hours postdose), fezolinetant decreased plasma LH by 49.8% relative to baseline, compared with 16.4% with placebo (Fig. 4A). Plasma levels of E2, FSH, and SHBG showed little impact of fezolinetant treatment (Fig. 4B–4D). Mean levels of E2 at baseline and all subsequent time points were consistently higher for the placebo group than the fezolinetant group, possibly owing to perimenopausal subjects in the placebo group who did not meet inclusion criteria for menopause, although they satisfied the VMS inclusion criterion. Consistent with this explanation, the difference was removed when perimenopausal subjects were excluded (data not shown).

Effect of fezolinetant on plasma concentration of (A) LH, (B) E2, (C) FSH, and (D) SHBG levels. Data are presented as mean ± SE (mITT population).
Figure 4.

Effect of fezolinetant on plasma concentration of (A) LH, (B) E2, (C) FSH, and (D) SHBG levels. Data are presented as mean ± SE (mITT population).

Safety

Both treatments were well tolerated. Seven subjects discontinued the study early, four in the placebo group and three in the fezolinetant group. Treatment compliance was high, and other than the discontinued subjects, no subjects missed >10 study medication intakes over the trial duration. TEAEs were reported by 35 (79.5%) subjects in the placebo group and 29 (67.4%) subjects in the fezolinetant group (Table 4). All TEAEs were “mild” or “moderate” in severity. A serious TEAE of upper limb fracture was reported by one subject in the placebo group and was considered moderate in severity and not related to the study drug. No deaths were reported during the study.

Table 4.

Adverse Events Occurring in Two or More Fezolinetant-Treated Subjects (Safety Population)

Placebo Twice Daily (n = 44)Fezolinetant (90 mg) Twice Daily (n = 43)
TEAE, n (%)
 At least one TEAE35 (79.5)29 (67.4)
 At least one SAEa1 (2.3)0
 At least one TEAE leading to death00
 At least one severe TEAE00
 At least one TEAE leading to discontinuation of study drug02 (4.7)
 At least one TEAE that was considered treatment related11 (25.0)13 (30.2)
System organ class
 Preferred term, n (%)
Any TEAE35 (79.5)29 (67.4)
Cardiac disorders2 (4.5)3 (7.0)
 Palpitations2 (4.5)3 (7.0)
Ear and labyrinth disorders02 (4.7)
 Vertigo02 (4.7)
Gastrointestinal disorders4 (9.1)10 (23.3)
 Abdominal discomfort1 (2.3)2 (4.7)
 Diarrhea03 (7.0)
 Paresthesia oral02 (4.7)
General disorders and administration site conditions2 (4.5)2 (4.7)
 Fatigue02 (4.7)
Infections and infestation13 (29.5)11 (25.6)
 Gastroenteritis2 (4.5)0
 Influenza1 (2.3)3 (7.0)
 Nasopharyngitis4 (9.1)1 (2.3)
Musculoskeletal and connective tissue disorders9 (20.5)7 (16.3)
 Arthralgia1 (2.3)2 (4.7)
 Arthritis3 (6.8)0
 Back pain2 (4.5)0
 Fibromyalgia02 (4.7)
Nervous system disorders8 (18.2)8 (18.6)
 Dizziness02 (4.7)
 Headache6 (13.6)7 (16.3)
Psychiatric disorders1 (2.3)4 (9.3)
 Depression02 (4.7)
Reproductive system and breast disorders6 (13.6)2 (4.7)
 Postmenopausal hemorrhage2 (4.5)0
 Vaginal hemorrhage2 (4.5)1 (2.3)
Placebo Twice Daily (n = 44)Fezolinetant (90 mg) Twice Daily (n = 43)
TEAE, n (%)
 At least one TEAE35 (79.5)29 (67.4)
 At least one SAEa1 (2.3)0
 At least one TEAE leading to death00
 At least one severe TEAE00
 At least one TEAE leading to discontinuation of study drug02 (4.7)
 At least one TEAE that was considered treatment related11 (25.0)13 (30.2)
System organ class
 Preferred term, n (%)
Any TEAE35 (79.5)29 (67.4)
Cardiac disorders2 (4.5)3 (7.0)
 Palpitations2 (4.5)3 (7.0)
Ear and labyrinth disorders02 (4.7)
 Vertigo02 (4.7)
Gastrointestinal disorders4 (9.1)10 (23.3)
 Abdominal discomfort1 (2.3)2 (4.7)
 Diarrhea03 (7.0)
 Paresthesia oral02 (4.7)
General disorders and administration site conditions2 (4.5)2 (4.7)
 Fatigue02 (4.7)
Infections and infestation13 (29.5)11 (25.6)
 Gastroenteritis2 (4.5)0
 Influenza1 (2.3)3 (7.0)
 Nasopharyngitis4 (9.1)1 (2.3)
Musculoskeletal and connective tissue disorders9 (20.5)7 (16.3)
 Arthralgia1 (2.3)2 (4.7)
 Arthritis3 (6.8)0
 Back pain2 (4.5)0
 Fibromyalgia02 (4.7)
Nervous system disorders8 (18.2)8 (18.6)
 Dizziness02 (4.7)
 Headache6 (13.6)7 (16.3)
Psychiatric disorders1 (2.3)4 (9.3)
 Depression02 (4.7)
Reproductive system and breast disorders6 (13.6)2 (4.7)
 Postmenopausal hemorrhage2 (4.5)0
 Vaginal hemorrhage2 (4.5)1 (2.3)

Abbreviation: SAE, serious adverse event.

a

Upper limb fracture.

Table 4.

Adverse Events Occurring in Two or More Fezolinetant-Treated Subjects (Safety Population)

Placebo Twice Daily (n = 44)Fezolinetant (90 mg) Twice Daily (n = 43)
TEAE, n (%)
 At least one TEAE35 (79.5)29 (67.4)
 At least one SAEa1 (2.3)0
 At least one TEAE leading to death00
 At least one severe TEAE00
 At least one TEAE leading to discontinuation of study drug02 (4.7)
 At least one TEAE that was considered treatment related11 (25.0)13 (30.2)
System organ class
 Preferred term, n (%)
Any TEAE35 (79.5)29 (67.4)
Cardiac disorders2 (4.5)3 (7.0)
 Palpitations2 (4.5)3 (7.0)
Ear and labyrinth disorders02 (4.7)
 Vertigo02 (4.7)
Gastrointestinal disorders4 (9.1)10 (23.3)
 Abdominal discomfort1 (2.3)2 (4.7)
 Diarrhea03 (7.0)
 Paresthesia oral02 (4.7)
General disorders and administration site conditions2 (4.5)2 (4.7)
 Fatigue02 (4.7)
Infections and infestation13 (29.5)11 (25.6)
 Gastroenteritis2 (4.5)0
 Influenza1 (2.3)3 (7.0)
 Nasopharyngitis4 (9.1)1 (2.3)
Musculoskeletal and connective tissue disorders9 (20.5)7 (16.3)
 Arthralgia1 (2.3)2 (4.7)
 Arthritis3 (6.8)0
 Back pain2 (4.5)0
 Fibromyalgia02 (4.7)
Nervous system disorders8 (18.2)8 (18.6)
 Dizziness02 (4.7)
 Headache6 (13.6)7 (16.3)
Psychiatric disorders1 (2.3)4 (9.3)
 Depression02 (4.7)
Reproductive system and breast disorders6 (13.6)2 (4.7)
 Postmenopausal hemorrhage2 (4.5)0
 Vaginal hemorrhage2 (4.5)1 (2.3)
Placebo Twice Daily (n = 44)Fezolinetant (90 mg) Twice Daily (n = 43)
TEAE, n (%)
 At least one TEAE35 (79.5)29 (67.4)
 At least one SAEa1 (2.3)0
 At least one TEAE leading to death00
 At least one severe TEAE00
 At least one TEAE leading to discontinuation of study drug02 (4.7)
 At least one TEAE that was considered treatment related11 (25.0)13 (30.2)
System organ class
 Preferred term, n (%)
Any TEAE35 (79.5)29 (67.4)
Cardiac disorders2 (4.5)3 (7.0)
 Palpitations2 (4.5)3 (7.0)
Ear and labyrinth disorders02 (4.7)
 Vertigo02 (4.7)
Gastrointestinal disorders4 (9.1)10 (23.3)
 Abdominal discomfort1 (2.3)2 (4.7)
 Diarrhea03 (7.0)
 Paresthesia oral02 (4.7)
General disorders and administration site conditions2 (4.5)2 (4.7)
 Fatigue02 (4.7)
Infections and infestation13 (29.5)11 (25.6)
 Gastroenteritis2 (4.5)0
 Influenza1 (2.3)3 (7.0)
 Nasopharyngitis4 (9.1)1 (2.3)
Musculoskeletal and connective tissue disorders9 (20.5)7 (16.3)
 Arthralgia1 (2.3)2 (4.7)
 Arthritis3 (6.8)0
 Back pain2 (4.5)0
 Fibromyalgia02 (4.7)
Nervous system disorders8 (18.2)8 (18.6)
 Dizziness02 (4.7)
 Headache6 (13.6)7 (16.3)
Psychiatric disorders1 (2.3)4 (9.3)
 Depression02 (4.7)
Reproductive system and breast disorders6 (13.6)2 (4.7)
 Postmenopausal hemorrhage2 (4.5)0
 Vaginal hemorrhage2 (4.5)1 (2.3)

Abbreviation: SAE, serious adverse event.

a

Upper limb fracture.

The number of subjects who reported at least one TEAE that was considered to be related to treatment was 11 (25.0%) in the placebo group and 13 (30.2%) in the fezolinetant group. The most common treatment-related TEAEs were gastrointestinal disorders, reported by six (14.0%) subjects in the fezolinetant vs none in the placebo group. Two subjects (4.7%) in the fezolinetant group discontinued study drug because of TEAEs that were considered possibly related to study treatment: one subject with preexisting fibromyalgia reported worsening of fibromyalgia, depression, dry mouth, headache, palpitations, diarrhea, and vomiting, and the other reported headache and vertigo. None of the subjects in the placebo group discontinued the study drug because of a TEAE.

Increased values for aspartate aminotransferase (AST) were more frequent with placebo (four subjects, 9.1%) than fezolinetant (two subjects, 4.8%); increased values for alanine aminotransferase (ALT) were more frequent with fezolinetant (five subjects, 11.9%) than placebo (one subject, 2.3%). In all cases, the observed increases in ALT and AST were mild and transient and did not exceed three times the upper limit of normal. Except for one occurrence of raised ALT and AST in the placebo group, none of these abnormalities was considered clinically significant.

No relevant or consistent changes in vital signs, electrocardiograms, or bone density markers were observed at any point during the study. None of the treatment-emergent vital signs or electrocardiographic abnormalities was considered by the investigator to be clinically significant.

Discussion

To our knowledge, this is the first clinical study designed according to the FDA guidelines (31) that demonstrates a significant beneficial effect of an NK3 antagonist for the treatment of VMSs. This 12-week, multicenter, double-blind, placebo-controlled study demonstrated that fezolinetant substantially reduced the frequency of moderate/severe VMSs and moderate/severe VMS score, with improvements achieved within the first day of treatment and sustained throughout the study. Fezolinetant treatment also improved sleep quality and other subject-reported outcomes that measure impact of VMSs on quality of life. The effect of fezolinetant to lower LH levels, in step with peak levels of drug exposure, is consistent with the proposed mechanism of action of KNDy neuron inhibition. The recurrence of VMSs after stopping fezolinetant, with similar severity and frequency in the follow-up phase as in the placebo group, further confirms the potent activity and reversibility of fezolinetant treatment. Fezolinetant was well tolerated and no drug-related serious adverse events were reported.

The FDA guidelines on the conduct of clinical trials to measure drug efficacy in VMSs (31) facilitate comparison of the current trial results with those of marketed hormonal (4143) and nonhormonal therapies (44). Fezolinetant produced a 93% reduction in VMS frequency from baseline to week 12. The incremental reduction in the number of moderate/severe VMSs with fezolinetant relative to placebo was 35.2 VMS episodes per week (95% CI, 22.8, 47.6); that is, fezolinetant-treated subjects had a reduction of approximately five episodes per day beyond that of placebo-treated subjects at week 12. In comparison, paroxetine, the only FDA-approved nonhormonal menopausal therapy, reduced VMS frequency by 6.2 to 9.2 episodes per week at week 12 relative to placebo, equivalent to an incremental benefit of <1 to 1.4 episodes per day (44). The FDA considers a reduction of two episodes per day to be a clinically meaningful difference. The reduction in VMS frequency in fezolinetant-treated women was also accompanied by improvements in subject-reported outcomes (e.g., HFRDIS, GCS, LSEQ, SDS), providing further evidence that the treatment effect was clinically meaningful. Fezolinetant demonstrated rapid improvement in symptoms, with decrease in frequency of moderate/severe VMSs from the first day of treatment, concurrent with the maximal drug concentration in plasma observed within a few hours of dosing. In comparison, studies of hormonal treatments conducted according to FDA guidelines and with a similar patient population found significant improvements in VMSs after only 2 to 4 weeks of treatment (42, 45, 46). The rapid relief of VMSs reported here with fezolinetant is consistent with findings with another NK3 antagonist, MLE4901, that was formerly in development and tested using alternative study designs (4749).

Following hot flashes and night sweats, difficulty sleeping is the next most impactful menopausal symptom for which women seek medical attention (3). Sleep disturbances may worsen adverse impact on quality of life arising from mood changes and symptoms of depression (5053). Additionally, the dysregulated KNDy neuron activity in menopause may have direct consequences on various autonomic functions affecting sleep, stress, and aspects of social behavior (54). In our study, fezolinetant improved sleep quality at all test intervals, and improved psychological, social, and productivity-related subject-reported outcomes. These improvements in quality of life may be relevant to decreasing the societal burden of VMSs, including overall health care resource utilization and negative effects on work productivity (5, 55).

Our clinical study found that peak fezolinetant levels were associated with a sharp decrease from baseline in plasma LH levels, but not FSH levels, consistent with the proposed mechanism of action on KNDy neurons (15, 16, 23). This direct action on the thermoregulatory pathway is independent of effects on ovarian hormones, as demonstrated by plasma levels of E2 remaining unchanged from baseline during fezolinetant treatment. This may be considered a therapeutically relevant outcome in the treatment of menopausal subjects at risk for endometrial hyperplasia with unopposed estrogen therapies (56).

Fezolinetant was well tolerated. The total number of TEAEs was higher in the placebo than in the fezolinetant treatment group, and no drug-related serious adverse events were reported. Adverse events related to vaginal bleeding, a common complaint with MHT (57), were less common with fezolinetant than with placebo. Gastrointestinal events (e.g., abdominal discomfort, diarrhea) were more common with fezolinetant than placebo, which may be attributable to the fact that NK3 receptors also are expressed in the gastrointestinal tract (58).

Strengths of this study include the design in accordance with FDA guidance for VMS treatments. A limitation is the restriction of study population to healthy menopausal women of largely common ethnicity with moderate/severe VMSs and exclusion of women receiving other treatments or with disorders that might have interfered with interpretation of study results; therefore, results may not be generalizable to all menopausal women. One further limitation is that inclusion of women with spontaneous amenorrhea for ≥3 months with FSH >40 IU/L and E2 <0.21 nmol/L may have allowed inclusion of some perimenopausal women with fluctuating hormone levels and symptoms; however, as this was a randomized trial, we expected these participants to be evenly represented in both treatment groups, balancing any impact on results across both groups. Additionally, the minimum washout for hormone therapies (6 weeks prior to screening) was less than the FDA guidance for certain hormone therapy formulations; however, this was unlikely to have affected the results because it would apply equally to both groups, and because the screening period added an additional 1 to 4 weeks of washout before study drug initiation. Although results of this study are promising, larger studies of longer duration are required to confirm efficacy and safety.

In summary, fezolinetant significantly reduced frequency and severity of moderate/severe VMSs, with response apparent from the first day of treatment. Fezolinetant was well tolerated and had no effect on E2 levels. The safety and efficacy demonstrated in this study support the potential use of fezolinetant as an effective nonhormonal treatment option for VMSs in menopausal women.

Acknowledgments

The authors thank Josephine Wolfram, Astellas Pharma Global Development, for statistical review of this manuscript.

Financial Support: This work was supported by Ogeda SA (Gosselies, Belgium). Astellas Pharma Inc. acquired 100% of the equity of Ogeda SA on 17 May 2017. Development of this manuscript, including editorial support provided by Mike Zbreski and Rosalba Satta of SuccinctChoice Medical Communications (Chicago, IL), was sponsored by Astellas Pharma Global Development.

Clinical Trial Information: EudraCT no. 2015-002578-20 (registered 29 July 2015).

Author Contributions: H.D., G.L.F., and S.R. were involved in the study design, study management, and board meetings concerning the follow-up of this study. H.D., D.T., G.D., and P.S. were involved in collection of study data. All authors had full access to the study data during development of this manuscript. G.L.F. prepared the first draft of the manuscript. All authors provided critical review and approval of the manuscript.

Additional Information

Disclosure Summary: H.D. reports personal fees from Ogeda SA during conduct of the study. D.T. received fees (paid to University Hospitals Leuven) from Astellas Pharma Inc. during the conduction of the study. S.R., J.C., H.R.H., and G.L.F. were employees of Ogeda SA when the study was conducted. G.L.F. reports grants from Region Wallon during the conduct of the study. Additionally, G.L.F. and H.R.H. have patents WO 2013/050424, WO 2014/154895, and EP15159296 licensed to Astellas Pharma Inc. The remaining authors have nothing to disclose.

Data Availability: The datasets generated during and/or analyzed during the current study are not publicly available but are available from the corresponding author on reasonable request. Studies conducted with product indications or formulations that remain in development are assessed after study completion to determine if individual participant data can be shared. The plan to share individual participant data is based on the status of product approval or termination of the compound, in addition to other study specific criteria as described on www.clinicalstudydatarequest.com under “Sponsor Specific Details for Astellas.”

Abbreviations:

    Abbreviations:
     
  • ALT

    alanine aminotransferase

  •  
  • AST

    aspartate aminotransferase

  •  
  • E2

    estradiol

  •  
  • ePRO

    electronic patient-reported outcome

  •  
  • FDA

    US Food and Drug Administration

  •  
  • GCS

    Greene Climacteric Scale

  •  
  • HFRDIS

    Hot Flash Related Daily Interference Scale

  •  
  • KNDy

    kisspeptin/neurokinin B/dynorphin

  •  
  • LSEQ

    Leeds Sleep Evaluation Questionnaire

  •  
  • LSMD

    least squares mean difference

  •  
  • MHT

    menopausal hormone therapy

  •  
  • mITT

    modified intent-to-treat

  •  
  • NK3R

    neurokinin 3 receptor

  •  
  • NKB

    neurokinin B

  •  
  • SDS

    Sheehan Disability Scale

  •  
  • TEAE

    treatment-emergent adverse event

  •  
  • VMS

    vasomotor symptom

References and Notes

1.

Gold
EB
,
Colvin
A
,
Avis
N
,
Bromberger
J
,
Greendale
GA
,
Powell
L
,
Sternfeld
B
,
Matthews
K
.
Longitudinal analysis of the association between vasomotor symptoms and race/ethnicity across the menopausal transition: Study of Women’s Health Across the Nation
.
Am J Public Health
.
2006
;
96
(
7
):
1226
1235
.

2.

Avis
NE
,
Crawford
SL
,
Greendale
G
,
Bromberger
JT
,
Everson-Rose
SA
,
Gold
EB
,
Hess
R
,
Joffe
H
,
Kravitz
HM
,
Tepper
PG
,
Thurston
RC
;
Study of Women’s Health Across the Nation
.
Duration of menopausal vasomotor symptoms over the menopause transition
.
JAMA Intern Med
.
2015
;
175
(
4
):
531
539
.

3.

Williams
RE
,
Kalilani
L
,
DiBenedetti
DB
,
Zhou
X
,
Fehnel
SE
,
Clark
RV
.
Healthcare seeking and treatment for menopausal symptoms in the United States
.
Maturitas
.
2007
;
58
(
4
):
348
358
.

4.

Assaf
AR
,
Bushmakin
AG
,
Joyce
N
,
Louie
MJ
,
Flores
M
,
Moffatt
M
.
The relative burden of menopausal and postmenopausal symptoms versus other major conditions: a retrospective analysis of the Medical Expenditure Panel Survey data
.
Am Health Drug Benefits
.
2017
;
10
(
6
):
311
321
.

5.

Sarrel
P
,
Portman
D
,
Lefebvre
P
,
Lafeuille
M-H
,
Grittner
AM
,
Fortier
J
,
Gravel
J
,
Duh
MS
,
Aupperle
PM
.
Incremental direct and indirect costs of untreated vasomotor symptoms
.
Menopause
.
2015
;
22
(
3
):
260
266
.

6.

Weissfeld
JL
,
Liu
W
,
Woods
C
,
Zhang
R
,
Li
J
,
van der Vlugt
TH
,
Slaughter
SR
.
Trends in oral and vaginally administered estrogen use among US women 50 years of age or older with commercial health insurance
.
Menopause
.
2018
;
25
(
6
):
611
614
.

7.

Chaplin
S
.
NICE guideline: diagnosis and management of the menopause
.
Prescriber
.
2016
;
27
(
1
):
27
32
.

8.

Stuenkel
CA
,
Davis
SR
,
Gompel
A
,
Lumsden
MA
,
Murad
MH
,
Pinkerton
JV
,
Santen
RJ
.
Treatment of symptoms of the menopause: an Endocrine Society clinical practice guideline
.
J Clin Endocrinol Metab
.
2015
;
100
(
11
):
3975
4011
.

9.

Lobo
RA
.
Hormone-replacement therapy: current thinking
.
Nat Rev Endocrinol
.
2017
;
13
(
4
):
220
231
.

10.

Rossouw
JE
,
Manson
JE
,
Kaunitz
AM
,
Anderson
GL
.
Lessons learned from the Women’s Health Initiative trials of menopausal hormone therapy
.
Obstet Gynecol
.
2013
;
121
(
1
):
172
176
.

11.

Pinkerton
JV
,
Santoro
N
.
Compounded bioidentical hormone therapy: identifying use trends and knowledge gaps among US women
.
Menopause
.
2015
;
22
(
9
):
926
936
.

12.

Tao
M
,
Teng
Y
,
Shao
H
,
Wu
P
,
Mills
EJ
.
Knowledge, perceptions and information about hormone therapy (HT) among menopausal women: a systematic review and meta-synthesis
.
PLoS One
.
2011
;
6
(
9
):
e24661
.

13.

Crandall
CJ
,
Manson
JE
,
Hohensee
C
,
Horvath
S
,
Wactawski-Wende
J
,
LeBlanc
ES
,
Vitolins
MZ
,
Nassir
R
,
Sinsheimer
JS
.
Association of genetic variation in the tachykinin receptor 3 locus with hot flashes and night sweats in the Women’s Health Initiative Study
.
Menopause
.
2017
;
24
(
3
):
252
261
.

14.

Jayasena
CN
,
Comninos
AN
,
Stefanopoulou
E
,
Buckley
A
,
Narayanaswamy
S
,
Izzi-Engbeaya
C
,
Abbara
A
,
Ratnasabapathy
R
,
Mogford
J
,
Ng
N
,
Sarang
Z
,
Ghatei
MA
,
Bloom
SR
,
Hunter
MS
,
Dhillo
WS
.
Neurokinin B administration induces hot flushes in women
.
Sci Rep
.
2015
;
5
(
1
):
8466
.

15.

Mittelman-Smith
MA
,
Williams
H
,
Krajewski-Hall
SJ
,
McMullen
NT
,
Rance
NE
.
Role for kisspeptin/neurokinin B/dynorphin (KNDy) neurons in cutaneous vasodilatation and the estrogen modulation of body temperature
.
Proc Natl Acad Sci USA
.
2012
;
109
(
48
):
19846
19851
.

16.

Rance
NE
,
Dacks
PA
,
Mittelman-Smith
MA
,
Romanovsky
AA
,
Krajewski-Hall
SJ
.
Modulation of body temperature and LH secretion by hypothalamic KNDy (kisspeptin, neurokinin B and dynorphin) neurons: a novel hypothesis on the mechanism of hot flushes
.
Front Neuroendocrinol
.
2013
;
34
(
3
):
211
227
.

17.

Padilla
SL
,
Johnson
CW
,
Barker
FD
,
Patterson
MA
,
Palmiter
RD
.
A neural circuit underlying the generation of hot flushes
.
Cell Reports
.
2018
;
24
(
2
):
271
277
.

18.

Krajewski-Hall
SJ
,
Miranda Dos Santos
F
,
McMullen
NT
,
Blackmore
EM
,
Rance
NE
.
Glutamatergic neurokinin 3 receptor neurons in the median preoptic nucleus modulate heat-defense pathways in female mice
.
Endocrinology
.
2019
;
160
(
4
):
803
816
.

19.

Topaloglu
AK
,
Reimann
F
,
Guclu
M
,
Yalin
AS
,
Kotan
LD
,
Porter
KM
,
Serin
A
,
Mungan
NO
,
Cook
JR
,
Imamoglu
S
,
Akalin
NS
,
Yuksel
B
,
O’Rahilly
S
,
Semple
RK
.
TAC3 and TACR3 mutations in familial hypogonadotropic hypogonadism reveal a key role for neurokinin B in the central control of reproduction
.
Nat Genet
.
2009
;
41
(
3
):
354
358
.

20.

Francou
B
,
Bouligand
J
,
Voican
A
,
Amazit
L
,
Trabado
S
,
Fagart
J
,
Meduri
G
,
Brailly-Tabard
S
,
Chanson
P
,
Lecomte
P
,
Guiochon-Mantel
A
,
Young
J
.
Normosmic congenital hypogonadotropic hypogonadism due to TAC3/TACR3 mutations: characterization of neuroendocrine phenotypes and novel mutations
.
PLoS One
.
2011
;
6
(
10
):
e25614
.

21.

Skrapits
K
,
Borsay
BA
,
Herczeg
L
,
Ciofi
P
,
Liposits
Z
,
Hrabovszky
E
.
Neuropeptide co-expression in hypothalamic kisspeptin neurons of laboratory animals and the human
.
Front Neurosci
.
2015
;
9
:
29
.

22.

Fergani
C
,
Navarro
VM
.
Expanding the role of tachykinins in the neuroendocrine control of reproduction
.
Reproduction
.
2016
;
153
(
1
):
R1
R14
.

23.

Lehman
MN
,
Coolen
LM
,
Goodman
RL
.
Minireview: kisspeptin/neurokinin B/dynorphin (KNDy) cells of the arcuate nucleus: a central node in the control of gonadotropin-releasing hormone secretion
.
Endocrinology
.
2010
;
151
(
8
):
3479
3489
.

24.

Skorupskaite
K
,
George
JT
,
Anderson
RA
.
The kisspeptin-GnRH pathway in human reproductive health and disease
.
Hum Reprod Update
.
2014
;
20
(
4
):
485
500
.

25.

Ruka
KA
,
Burger
LL
,
Moenter
SM
.
Both estrogen and androgen modify the response to activation of neurokinin-3 and κ-opioid receptors in arcuate kisspeptin neurons from male mice
.
Endocrinology
.
2016
;
157
(
2
):
752
763
.

26.

Rance
NE
,
Young
WS
III
.
Hypertrophy and increased gene expression of neurons containing neurokinin-B and substance-P messenger ribonucleic acids in the hypothalami of postmenopausal women
.
Endocrinology
.
1991
;
128
(
5
):
2239
2247
.

27.

Rometo
AM
,
Krajewski
SJ
,
Voytko
ML
,
Rance
NE
.
Hypertrophy and increased kisspeptin gene expression in the hypothalamic infundibular nucleus of postmenopausal women and ovariectomized monkeys
.
J Clin Endocrinol Metab
.
2007
;
92
(
7
):
2744
2750
.

28.

Hoveyda
HR
,
Fraser
GL
,
Dutheuil
G
,
El Bousmaqui
M
,
Korac
J
,
Lenoir
F
,
Lapin
A
,
Noël
S
.
Optimization of novel antagonists to the neurokinin-3 receptor for the treatment of sex-hormone disorders (part II)
.
ACS Med Chem Lett
.
2015
;
6
(
7
):
736
740
.

29.

Fraser
GL
,
Hoveyda
HR
,
Clarke
IJ
,
Ramaswamy
S
,
Plant
TM
,
Rose
C
,
Millar
RP
.
The NK3 receptor antagonist ESN364 interrupts pulsatile LH secretion and moderates levels of ovarian hormones throughout the menstrual cycle
.
Endocrinology
.
2015
;
156
(
11
):
4214
4225
.

30.

Fraser
GL
,
Ramael
S
,
Hoveyda
HR
,
Gheyle
L
,
Combalbert
J
.
The NK3 receptor antagonist ESN364 suppresses sex hormones in men and women
.
J Clin Endocrinol Metab
.
2016
;
101
(
2
):
417
426
.

31.

U.S. Food and Drug Administration
.
Guidance for industry: estrogen and estrogen/progestin drug products to treat vasomotor symptoms and vulvar and vaginal atrophy symptoms — recommendations for clinical evaluation. Available at: http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm071643.pdf. Accessed 19 February 2019
.

32.

Cully
M
.
Neurokinin 3 receptor antagonist revival heats up with Astellas acquisition
.
Nat Rev Drug Discov
.
2017
;
16
(
6
):
377
.

33.

Sloan
JA
,
Loprinzi
CL
,
Novotny
PJ
,
Barton
DL
,
Lavasseur
BI
,
Windschitl
H
.
Methodologic lessons learned from hot flash studies
.
J Clin Oncol
.
2001
;
19
(
23
):
4280
4290
.

34.

Carpenter
JS
.
The Hot Flash Related Daily Interference Scale: a tool for assessing the impact of hot flashes on quality of life following breast cancer
.
J Pain Symptom Manage
.
2001
;
22
(
6
):
979
989
.

35.

Parrott
AC
,
Hindmarch
I
.
The Leeds Sleep Evaluation Questionnaire in psychopharmacological investigations—a review
.
Psychopharmacology (Berl)
.
1980
;
71
(
2
):
173
179
.

36.

Greene
JG
.
A factor analytic study of climacteric symptoms
.
J Psychosom Res
.
1976
;
20
(
5
):
425
430
.

37.

Sheehan
DV
.
The Sheehan Disability Scales. The Anxiety Disease and How to Overcome It
.
New York, NY
:
Charles Scribner and Sons
;
1983
.

38.

Rush
JA
.
Handbook of Psychiatric Measures. Washington, DC: American Psychiatric Association; 2005
.

39.

Posner
K
,
Brown
GK
,
Stanley
B
,
Brent
DA
,
Yershova
KV
,
Oquendo
MA
,
Currier
GW
,
Melvin
GA
,
Greenhill
L
,
Shen
S
,
Mann
JJ
.
The Columbia-Suicide Severity Rating Scale: initial validity and internal consistency findings from three multisite studies with adolescents and adults
.
Am J Psychiatry
.
2011
;
168
(
12
):
1266
1277
.

40.

Grant
MD
,
Marbella
A
,
Wang
AT
,
Pines
E
,
Hoag
J
,
Bonnell
C
,
Ziegler
KM
,
Aronson
N
.
Menopausal Symptoms: Comparative Effectiveness of Therapies AHRQ Comparative Effectiveness Reviews
.
Rockville, MD
:
Agency for Healthcare Research and Quality
;
2015
.

41.

Notelovitz
M
,
Lenihan
JP
,
McDermott
M
,
Kerber
IJ
,
Nanavati
N
,
Arce
J
.
Initial 17β-estradiol dose for treating vasomotor symptoms
.
Obstet Gynecol
.
2000
;
95
(
5
):
726
731
.

42.

Archer
DF
,
Pinkerton
JV
,
Utian
WH
,
Menegoci
JC
,
de Villiers
TJ
,
Yuen
CK
,
Levine
AB
,
Chines
AA
,
Constantine
GD
.
Bazedoxifene, a selective estrogen receptor modulator: effects on the endometrium, ovaries, and breast from a randomized controlled trial in osteoporotic postmenopausal women
.
Menopause
.
2009
;
16
(
6
):
1109
1115
.

43.

Utian
WH
,
Shoupe
D
,
Bachmann
G
,
Pinkerton
JV
,
Pickar
JH
.
Relief of vasomotor symptoms and vaginal atrophy with lower doses of conjugated equine estrogens and medroxyprogesterone acetate
.
Fertil Steril
.
2001
;
75
(
6
):
1065
1079
.

44.

Simon
JA
,
Portman
DJ
,
Kaunitz
AM
,
Mekonnen
H
,
Kazempour
K
,
Bhaskar
S
,
Lippman
J
.
Low-dose paroxetine 7.5 mg for menopausal vasomotor symptoms: two randomized controlled trials
.
Menopause
.
2013
;
20
(
10
):
1027
1035
.

45.

Kagan
R
,
Komm
BS
,
Ryan
KA
,
Lavenberg
J
,
Yu
CR
,
Pinkerton
JV
.
Timing and persistence of effect of conjugated estrogens/bazedoxifene in postmenopausal women
.
Menopause
.
2016
;
23
(
11
):
1204
1213
.

46.

Lobo
RA
,
Archer
DF
,
Kagan
R
,
Kaunitz
AM
,
Constantine
GD
,
Pickar
JH
,
Graham
S
,
Bernick
B
,
Mirkin
S
.
A 17β-Estradiol‒progesterone oral capsule for vasomotor symptoms in postmenopausal women: a randomized controlled trial
.
Obstet Gynecol
.
2018
;
132
(
1
):
161
170
.

47.

Prague
JK
,
Roberts
RE
,
Comninos
AN
,
Clarke
S
,
Jayasena
CN
,
Nash
Z
,
Doyle
C
,
Papadopoulou
DA
,
Bloom
SR
,
Mohideen
P
,
Panay
N
,
Hunter
MS
,
Veldhuis
JD
,
Webber
LC
,
Huson
L
,
Dhillo
WS
.
Neurokinin 3 receptor antagonism as a novel treatment for menopausal hot flushes: a phase 2, randomised, double-blind, placebo-controlled trial
.
Lancet
.
2017
;
389
(
10081
):
1809
1820
.

48.

Prague
JK
,
Roberts
RE
,
Comninos
AN
,
Clarke
S
,
Jayasena
CN
,
Mohideen
P
,
Lin
VH
,
Stern
TP
,
Panay
N
,
Hunter
MS
,
Webber
LC
,
Dhillo
WS
.
Neurokinin 3 receptor antagonism rapidly improves vasomotor symptoms with sustained duration of action
.
Menopause
.
2018
;
25
(
8
):
862
869
.

49.

Skorupskaite
K
,
George
JT
,
Veldhuis
JD
,
Millar
RP
,
Anderson
RA
.
Neurokinin 3 receptor antagonism reveals roles for neurokinin B in the regulation of gonadotropin secretion and hot flashes in postmenopausal women
.
Neuroendocrinology
.
2018
;
106
(
2
):
148
157
.

50.

Chung
HF
,
Pandeya
N
,
Dobson
AJ
,
Kuh
D
,
Brunner
EJ
,
Crawford
SL
,
Avis
NE
,
Gold
EB
,
Mitchell
ES
,
Woods
NF
,
Bromberger
JT
,
Thurston
RC
,
Joffe
H
,
Yoshizawa
T
,
Anderson
D
,
Mishra
GD
.
The role of sleep difficulties in the vasomotor menopausal symptoms and depressed mood relationships: an international pooled analysis of eight studies in the InterLACE consortium
.
Psychol Med
.
2018
;
48
(
15
):
2550
2561
.

51.

Jehan
S
,
Masters-Isarilov
A
,
Salifu
I
,
Zizi
F
,
Jean-Louis
G
,
Pandi-Perumal
SR
,
Gupta
R
,
Brzezinski
A
,
McFarlane
SI
.
Sleep disorders in postmenopausal women
.
J Sleep Disord Ther
.
2015
;
4
(
5
):
1000212
.

52.

Vincent
AJ
,
Ranasinha
S
,
Sayakhot
P
,
Mansfield
D
,
Teede
HJ
.
Sleep difficulty mediates effects of vasomotor symptoms on mood in younger breast cancer survivors
.
Climacteric
.
2014
;
17
(
5
):
598
604
.

53.

de Kruif
M
,
Spijker
AT
,
Molendijk
ML
.
Depression during the perimenopause: A meta-analysis
.
J Affect Disord
.
2016
;
206
:
174
180
.

54.

Lehman
MN
,
Coolen
LM
,
Steiner
RA
,
Neal-Perry
G
,
Wang
L
,
Moenter
SM
,
Moore
AM
,
Goodman
RL
,
Hwa-Yeo
S
,
Padilla
SL
,
Kauffman
AS
,
Garcia
J
,
Kelly
MJ
,
Clarkson
J
,
Radovick
S
,
Babwah
AV
,
Leon
S
,
Tena-Sempere
M
,
Comninos
A
,
Seminara
S
,
Dhillo
WS
,
Levine
J
,
Terasawa
E
,
Negron
A
,
Herbison
AE
.
The 3rd World Conference on Kisspeptin, “Kisspeptin 2017: Brain and Beyond”: unresolved questions, challenges and future directions for the field
.
J Neuroendocrinol
.
2018
;
30
(
5
):
e12600
.

55.

Griffiths
A
,
MacLennan
SJ
,
Hassard
J
.
Menopause and work: an electronic survey of employees’ attitudes in the UK
.
Maturitas
.
2013
;
76
(
2
):
155
159
.

56.

Lethaby
A
,
Suckling
J
,
Barlow
D
,
Farquhar
CM
,
Jepson
RG
,
Roberts
H
.
Hormone replacement therapy in postmenopausal women: endometrial hyperplasia and irregular bleeding
.
Cochrane Database Syst Rev
.
2004
;
3
(
3
):
CD000402
.

57.

Barnabei
VM
,
Cochrane
BB
,
Aragaki
AK
,
Nygaard
I
,
Williams
RS
,
McGovern
PG
,
Young
RL
,
Wells
EC
,
O’Sullivan
MJ
,
Chen
B
,
Schenken
R
,
Johnson
SR
;
Women’s Health Initiative Investigators
.
Menopausal symptoms and treatment-related effects of estrogen and progestin in the Women’s Health Initiative
.
Obstet Gynecol
.
2005
;
105
(
5 Pt 1
):
1063
1073
.

58.

Jaafari
N
,
Hua
G
,
Adélaïde
J
,
Julé
Y
,
Imbert
J
.
Expression of the tachykinin receptor mRNAs in healthy human colon
.
Eur J Pharmacol
.
2008
;
599
(
1-3
):
121
125
.