Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 307
Filter
1.
J Neuroendocrinol ; 36(5): e13384, 2024 May.
Article in English | MEDLINE | ID: mdl-38516965

ABSTRACT

Psychosocial stress negatively impacts reproductive function by inhibiting pulsatile luteinizing hormone (LH) secretion. The posterodorsal medial amygdala (MePD) is responsible in part for processing stress and modulating the reproductive axis. Activation of the neurokinin 3 receptor (NK3R) suppresses the gonadotropin-releasing hormone (GnRH) pulse generator, under hypoestrogenic conditions, and NK3R activity in the amygdala has been documented to play a role in stress and anxiety. We investigate whether NK3R activation in the MePD is involved in mediating the inhibitory effect of psychosocial stress on LH pulsatility in ovariectomised female mice. First, we administered senktide, an NK3R agonist, into the MePD and monitored the effect on pulsatile LH secretion. We then delivered SB222200, a selective NK3R antagonist, intra-MePD in the presence of predator odour, 2,4,5-trimethylthiazole (TMT) and examined the effect on LH pulses. Senktide administration into the MePD dose-dependently suppresses pulsatile LH secretion. Moreover, NK3R signalling in the MePD mediates TMT-induced suppression of the GnRH pulse generator, which we verified using a mathematical model. The model verifies our experimental findings: (i) predator odour exposure inhibits LH pulses, (ii) activation of NK3R in the MePD inhibits LH pulses and (iii) NK3R antagonism in the MePD blocks stressor-induced inhibition of LH pulse frequency in the absence of ovarian steroids. These results demonstrate for the first time that NK3R neurons in the MePD mediate psychosocial stress-induced suppression of the GnRH pulse generator.


Subject(s)
Luteinizing Hormone , Quinolines , Receptors, Neurokinin-3 , Signal Transduction , Stress, Psychological , Substance P/analogs & derivatives , Animals , Female , Receptors, Neurokinin-3/metabolism , Receptors, Neurokinin-3/antagonists & inhibitors , Receptors, Neurokinin-3/agonists , Luteinizing Hormone/metabolism , Stress, Psychological/metabolism , Mice , Signal Transduction/physiology , Signal Transduction/drug effects , Corticomedial Nuclear Complex/metabolism , Corticomedial Nuclear Complex/drug effects , Corticomedial Nuclear Complex/physiology , Peptide Fragments/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Mice, Inbred C57BL , Amygdala/metabolism , Amygdala/drug effects
2.
Menopause ; 31(4): 342-354, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38471077

ABSTRACT

IMPORTANCE: Vasomotor symptoms (VMS) affect many postmenopausal persons and impact sleep and quality of life. OBJECTIVE: This systematic review examines the literature describing the safety and efficacy of neurokinin-3 receptor antagonists approved and in development for postmenopausal persons with VMS. EVIDENCE REVIEW: A search of MEDLINE, EMBASE, and International Pharmaceutical Abstracts was conducted using the search terms and permutations of neurokinin-3 receptor antagonist, elinzanetant, fezolinetant, and osanetant. Inclusion criteria of reporting on efficacy or safety of fezolinetant, elinzanetant, or osanetant; studies in participants identifying as female; full record in English; and primary literature were applied. Abstract-only records were excluded. Extracted data were synthesized to allow comparison of reported study characteristics, efficacy outcomes, and safety events. Eligible records were evaluated for risk of bias via the Cochrane Risk of Bias 2 tool for randomized studies and the Grading of Recommendations Assessment, Development and Evaluation system was used. This study was neither funded nor registered. FINDINGS: The search returned 191 records; 186 were screened after deduplication. Inclusion criteria were met by six randomized controlled trials (RCT), four reported on fezolinetant, and two reported on elinzanetant. One record was a post hoc analysis of a fezolinetant RCT. An additional study was identified outside the database search. Three fezolinetant RCT demonstrated a reduction in VMS frequency/severity, improvement in Menopause-Specific Quality of Life scores, and improvement in sleep quality at weeks 4 and 12 compared with placebo without serious adverse events. The two RCT on elinzanetant also showed improvements in VMS frequency and severity. All eight records evaluated safety through treatment-emergent adverse events; the most common adverse events were COVID-19, headache, somnolence, and gastrointestinal. Each record evaluated had a low risk of bias. There is a strong certainty of evidence as per the Grading of Recommendations Assessment, Development and Evaluation system. CONCLUSIONS AND RELEVANCE: Because of the high-quality evidence supporting the efficacy of fezolinetant and elinzanetant, these agents may be an effective option with mild adverse events for women seeking nonhormone treatment of VMS.


Subject(s)
Heterocyclic Compounds, 2-Ring , Hot Flashes , Menopause , Piperidines , Receptors, Neurokinin-3 , Sweating , Thiadiazoles , Vasomotor System , Female , Humans , Heterocyclic Compounds, 2-Ring/pharmacology , Heterocyclic Compounds, 2-Ring/therapeutic use , Menopause/drug effects , Menopause/physiology , Receptors, Neurokinin-3/antagonists & inhibitors , Thiadiazoles/chemistry , Thiadiazoles/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Hot Flashes/drug therapy , Sweating/drug effects , Vasomotor System/drug effects , Vasomotor System/physiopathology
3.
Curr Protein Pept Sci ; 25(4): 339-352, 2024.
Article in English | MEDLINE | ID: mdl-38243941

ABSTRACT

BACKGROUND: Neurokinin B; an endogenous decapeptide, mediates its reproductive physiological actions through gonadotropin releasing hormone. Despite the potential role of Neurokinin B on seminal vesicles, its effects on seminal vesicles in adult male mammals remain elusive. We aimed to investigate the potentials of variable doses of Neurokinin B, its agonist and antagonist on histomorphology and expression of NK3R on seminal vesicles, and secretory activity of seminal vesicles in adult male rats. METHODS: Adult male Sprague Dawley rats (n=10 in each group) were administered intraperitoneally with Neurokinin B in three variable doses: 1 µg, 1 ηg and 10 ρg while, Senktide (Neurokinin B agonist) and SB222200 (Neurokinin B antagonist) in 1 µg doses consecutively for 12 days. After 12 days of peptide treatment, half of the animals (n=05) in each group were sacrificed while remaining half (n=05) were kept for another 12 days without any treatment to investigate treatment reversal. Seminal vesicles were dissected and excised tissue was processed for light microscopy, immunohistochemistry and estimation of seminal fructose levels. RESULTS: Treatment with Neurokinin B and Senktide significantly increased while SB222200 slightly decrease the seminal vesicles weight, epithelial height and seminal fructose levels as compared to control. Light microscopy revealed increased epithelial height and epithelial folding as compared to control in all Neurokinin B and Senktide treated groups while decreased in SB222200. Effects of various doses of Neurokinin B, Senktide and SB222200 on seminal vesicles weight, epithelial height, seminal fructose levels and histomorphology were reversed when rats were maintained without treatments. Immuno-expression of Neurokinin B shows no change in treatment and reversal groups. CONCLUSION: Continuous administration of Neurokinin B and Senktide effect positively while SB222200 have detrimental effects on cellular morphology, epithelial height and seminal fructose levels in seminal vesicles. Effects of peptide treatments depicted a reversal towards control group when rats were kept without any treatment.


Subject(s)
Neurokinin B , Peptide Fragments , Rats, Sprague-Dawley , Receptors, Neurokinin-3 , Seminal Vesicles , Substance P , Animals , Male , Neurokinin B/metabolism , Seminal Vesicles/drug effects , Seminal Vesicles/metabolism , Rats , Receptors, Neurokinin-3/metabolism , Receptors, Neurokinin-3/antagonists & inhibitors , Peptide Fragments/pharmacology , Peptide Fragments/metabolism , Substance P/metabolism , Dose-Response Relationship, Drug , Cell Proliferation/drug effects
4.
Nature ; 624(7991): 425-432, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38057665

ABSTRACT

Maintenance of renal function and fluid transport are essential for vertebrates and invertebrates to adapt to physiological and pathological challenges. Human patients with malignant tumours frequently develop detrimental renal dysfunction and oliguria, and previous studies suggest the involvement of chemotherapeutic toxicity and tumour-associated inflammation1,2. However, how tumours might directly modulate renal functions remains largely unclear. Here, using conserved tumour models in Drosophila melanogaster3, we characterized isoform F of ion transport peptide (ITPF) as a fly antidiuretic hormone that is secreted by a subset of yki3SA gut tumour cells, impairs renal function and causes severe abdomen bloating and fluid accumulation. Mechanistically, tumour-derived ITPF targets the G-protein-coupled receptor TkR99D in stellate cells of Malpighian tubules-an excretory organ that is equivalent to renal tubules4-to activate nitric oxide synthase-cGMP signalling and inhibit fluid excretion. We further uncovered antidiuretic functions of mammalian neurokinin 3 receptor (NK3R), the homologue of fly TkR99D, as pharmaceutical blockade of NK3R efficiently alleviates renal tubular dysfunction in mice bearing different malignant tumours. Together, our results demonstrate a novel antidiuretic pathway mediating tumour-renal crosstalk across species and offer therapeutic opportunities for the treatment of cancer-associated renal dysfunction.


Subject(s)
Antidiuretic Agents , Kidney Diseases , Neoplasms , Neuropeptides , Receptors, Neurokinin-3 , Animals , Humans , Mice , Antidiuretic Agents/metabolism , Cyclic GMP/metabolism , Disease Models, Animal , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Kidney Diseases/complications , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Malpighian Tubules/cytology , Malpighian Tubules/metabolism , Neoplasms/complications , Neoplasms/metabolism , Nitric Oxide Synthase/metabolism , Receptors, Neurokinin-3/antagonists & inhibitors , Receptors, Neurokinin-3/metabolism , Xenograft Model Antitumor Assays , Arginine Vasopressin/metabolism , Drosophila Proteins/metabolism , Neuropeptides/metabolism
5.
Cell ; 186(16): 3332-3332.e1, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37541194

ABSTRACT

Menopausal hot flashes are common and debilitating. Menopausal Hormone Therapy (MHT) is effective for hot flashes but has risks and side effects that limit its use. NK3 receptor antagonism has emerged as a novel therapeutic strategy, leading to the recent FDA approval of fezolinetant, a first-in-class nonhormonal treatment for menopausal hot flashes. To view this Bench to Bedside, open or download the PDF.


Subject(s)
Hot Flashes , Menopause , Receptors, Neurokinin-3 , Thiadiazoles , Humans , Hot Flashes/drug therapy , Receptors, Neurokinin-3/antagonists & inhibitors , Thiadiazoles/therapeutic use
6.
Life Sci ; 310: 121078, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36252700

ABSTRACT

AIMS: Neurokinin-B (NKB)-Neurokinin-3-receptor (NK3R) pathway is remarkably sensitive to energy equilibrium; however, its role in metabolic regulation remains unexplored in polycystic ovary syndrome (PCOS). Therefore, this work aimed to investigate the role of NK3R antagonists (NK3Ra) on metabolic dysfunction and obesity in in vitro and in vivo PCOS models. MAIN METHODS: First, an observational study using serum samples collected from 19 PCOS patients was performed. Second, prospective case-control experimental studies where NK3Ra (SB222200) was used to treat PCOS-like mice (BALB/c mice), ovariectomized+estrogen implanted obese mice (C57BL/6J mice) and 3T3-L1 murine preadipocytes were carried out to investigate its effect on metabolism in vivo and in vitro. The fat volumes, serum biochemical indexes, adipokines and inflammatory cytokines, metabolism-related gene expression and the concentrations of ATP, NAD+, NADPH…etc. were studied. KEY FINDINGS: We found a positive correlation between serum NKB and lipid metabolism indicators in PCOS women. Using the mouse models, we demonstrated that administration of NK3Ra regulates serum adipokines, inhibits weight gain with a marked decrease in fat volume, adipocyte size, and inflammatory cytokines, and promotes oxidative metabolism and energy consumption. NK3Ra reduces lipid accumulation in mature murine adipocytes by inhibiting the expression of peroxisome proliferator- activated receptor gamma (PPAR-γ) and fatty acid binding protein 4 (FABP4) genes. NK3Ras also enhances oxidative metabolism and energy consumption by maintaining intracellular redox homeostasis. SIGNIFICANCE: This study backs the use of NK3Ras as a potential therapeutic for PCOS since it ameliorates both reproductive and metabolic aberrations.


Subject(s)
Obesity , Polycystic Ovary Syndrome , Receptors, Neurokinin-3 , Animals , Female , Humans , Mice , 3T3-L1 Cells , Adipocytes/metabolism , Adipokines/metabolism , Cytokines/metabolism , Lipid Metabolism , Mice, Inbred C57BL , Mice, Obese , Obesity/drug therapy , Obesity/metabolism , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/metabolism , PPAR gamma/metabolism , Receptors, Neurokinin-3/antagonists & inhibitors , Receptors, Neurokinin-3/metabolism
7.
Int J Mol Sci ; 23(9)2022 Apr 21.
Article in English | MEDLINE | ID: mdl-35562976

ABSTRACT

G protein-coupled receptors (GPCRs) facilitate the majority of signal transductions across cell membranes in humans, with numerous diseases attributed to inactivating GPCR mutations. Many of these mutations result in misfolding during nascent receptor synthesis in the endoplasmic reticulum (ER), resulting in intracellular retention and degradation. Pharmacological chaperones (PCs) are cell-permeant small molecules that can interact with misfolded receptors in the ER and stabilise/rescue their folding to promote ER exit and trafficking to the cell membrane. The neurokinin 3 receptor (NK3R) plays a pivotal role in the hypothalamic-pituitary-gonadal reproductive axis. We sought to determine whether NK3R missense mutations result in a loss of cell surface receptor expression and, if so, whether a cell-permeant small molecule NK3R antagonist could be repurposed as a PC to restore function to these mutants. Quantitation of cell surface expression levels of seven mutant NK3Rs identified in hypogonadal patients indicated that five had severely impaired cell surface expression. A small molecule NK3R antagonist, M8, increased cell surface expression in four of these five and resulted in post-translational receptor processing in a manner analogous to the wild type. Importantly, there was a significant improvement in receptor activation in response to neurokinin B (NKB) for all four receptors following their rescue with M8. This demonstrates that M8 may have potential for therapeutic development in the treatment of hypogonadal patients harbouring NK3R mutations. The repurposing of existing small molecule GPCR modulators as PCs represents a novel and therapeutically viable option for the treatment of disorders attributed to mutations in GPCRs that cause intracellular retention.


Subject(s)
Neurokinin B , Receptors, Neurokinin-3 , Cell Membrane/metabolism , Humans , Mutation , Neurokinin B/genetics , Neurokinin B/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neurokinin-3/antagonists & inhibitors , Receptors, Neurokinin-3/genetics , Receptors, Neurokinin-3/metabolism
8.
Eur J Pharmacol ; 905: 174207, 2021 Aug 15.
Article in English | MEDLINE | ID: mdl-34048742

ABSTRACT

The majority of women experience vasomotor symptoms (VMS), such as hot flashes and night sweats, during the menopausal transition. Recent evidence strongly suggests a connection between neurokinin 3 (NK3) receptor signaling and VMS associated with menopause. The NK3 receptor antagonist fezolinetant is currently in phase 3 development for treatment of moderate to severe VMS associated with menopause. We investigated the pharmacological effects of repeated administration of fezolinetant on levels of sex hormones and gonadotropins, neuronal activity in the hypothalamus, and skin temperature as an index of hot flash-like symptoms in ovariectomized rats as a model of menopause. Ovariectomized rats exhibited several typical menopausal symptoms: hyperphagia, increased body weight, significantly decreased plasma estradiol levels, increased luteinizing hormone (LH) and follicle-stimulating hormone (FSH) levels, and significantly increased skin temperature. Increased c-Fos expression (an indirect marker of neuronal activity) in median preoptic nucleus (MnPO) hypothalamic neurons was also observed in ovariectomized rats. Repeated oral administration of fezolinetant (1-10 mg/kg, twice daily) for 1 week dose-dependently reduced plasma LH levels without affecting estradiol or FSH levels, inhibited the activation of MnPO neurons, and attenuated hot flash-like symptoms. In addition, fezolinetant dose-dependently reduced hyperphagia and weight gain in ovariectomized rats. These preclinical findings suggest that fezolinetant attenuates hot flash-like symptoms via inhibition of neuronal activity in the MnPO of ovariectomized rats and provides further support for the ongoing clinical development of fezolinetant for the treatment of VMS associated with menopause.


Subject(s)
Heterocyclic Compounds, 2-Ring/pharmacology , Hot Flashes/drug therapy , Receptors, Neurokinin-3/antagonists & inhibitors , Thiadiazoles/pharmacology , Administration, Oral , Animals , Body Temperature/drug effects , Disease Models, Animal , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Heterocyclic Compounds, 2-Ring/administration & dosage , Hot Flashes/etiology , Injections, Subcutaneous , Luteinizing Hormone/blood , Menopause/drug effects , Ovariectomy/adverse effects , Preoptic Area/metabolism , Progesterone/blood , Rats, Wistar , Skin Temperature/drug effects , Testosterone/blood , Thiadiazoles/administration & dosage
9.
J Clin Endocrinol Metab ; 106(9): e3519-e3532, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34000049

ABSTRACT

CONTEXT: Polycystic ovary syndrome (PCOS), a highly prevalent endocrine disorder characterized by hyperandrogenism, is the leading cause of anovulatory infertility. OBJECTIVE: This proof-of-concept study evaluated clinical efficacy and safety of the neurokinin 3 (NK3) receptor antagonist fezolinetant in PCOS. METHODS: This was a phase 2a, randomized, double-blind, placebo-controlled, multicenter study (EudraCT 2014-004409-34). The study was conducted at 5 European clinical centers. Women with PCOS participated in the study. Interventions included fezolinetant 60 or 180 mg/day or placebo for 12 weeks. The primary efficacy end point was change in total testosterone. Gonadotropins, ovarian hormones, safety and tolerability were also assessed. RESULTS: Seventy-three women were randomly assigned, and 64 participants completed the study. Adjusted mean (SE) changes in total testosterone from baseline to week 12 for fezolinetant 180 and 60 mg/day were -0.80 (0.13) and -0.39 (0.12) nmol/L vs -0.05 (0.10) nmol/L with placebo (P < .001 and P < .05, respectively). Adjusted mean (SE) changes from baseline in luteinizing hormone (LH) for fezolinetant 180 and 60 mg/d were -10.17 (1.28) and -8.21 (1.18) vs -3.16 (1.04) IU/L with placebo (P < .001 and P = .002); corresponding changes in follicle-stimulating hormone (FSH) were -1.46 (0.32) and -0.92 (0.30) vs -0.57 (0.26) IU/L (P = .03 and P = .38), underpinning a dose-dependent decrease in the LH-to-FSH ratio vs placebo (P < .001). Circulating levels of progesterone and estradiol did not change significantly vs placebo (P > .10). Fezolinetant was well tolerated. CONCLUSION: Fezolinetant had a sustained effect to suppress hyperandrogenism and reduce the LH-to-FSH ratio in women with PCOS.


Subject(s)
Heterocyclic Compounds, 2-Ring/therapeutic use , Polycystic Ovary Syndrome/drug therapy , Receptors, Neurokinin-3/antagonists & inhibitors , Thiadiazoles/therapeutic use , Adolescent , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Female , Follicle Stimulating Hormone/blood , Gonadotropins/blood , Heterocyclic Compounds, 2-Ring/adverse effects , Humans , Hyperandrogenism/drug therapy , Luteinizing Hormone/blood , Middle Aged , Ovarian Function Tests , Testosterone/blood , Thiadiazoles/adverse effects , Treatment Outcome , Young Adult
10.
Expert Opin Investig Drugs ; 30(7): 681-694, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33724119

ABSTRACT

Introduction: Although international clinical practice guidelines recognize a continued role for menopausal hormone therapy (HT), particularly for symptomatic women <60 years of age or within 10 years of menopause, safety and tolerability concerns have discouraged HT use due to potential links with a perceived increased risk of hormone-dependent cancers, and an established risk of stroke and venous thromboembolism. There is therefore a need for safe, effective non-hormonal therapy for relief of menopausal vasomotor symptoms (VMS).Areas covered: This narrative review summarizes the dataset accrued for fezolinetant, a neurokinin-3 receptor (NK3R) antagonist in clinical development for menopause-associated VMS.Expert opinion: Altered signaling in neuroendocrine circuits at menopause leads to VMS wherein NK3R activity plays a key role to modulate the thermoregulatory center in a manner conducive to triggering the 'hot flash' response. Thus, a new generation of NK3R antagonists has entered clinical development to specifically target the mechanistic basis of VMS. Fezolinetant is the most advanced NK3R antagonist in terms of stage of clinical development. Results to date have demonstrated rapid and substantial reduction in VMS frequency and severity and associated improvements in health-related quality of life. NK3R antagonists offer a non-hormonal alternative to HT for the treatment of menopause-related VMS.


Subject(s)
Heterocyclic Compounds, 2-Ring/pharmacology , Menopause/physiology , Receptors, Neurokinin-3/antagonists & inhibitors , Thiadiazoles/pharmacology , Female , Hot Flashes/drug therapy , Humans , Middle Aged , Quality of Life , Receptors, Neurokinin-3/metabolism
11.
Endocrinology ; 162(5)2021 05 01.
Article in English | MEDLINE | ID: mdl-33522579

ABSTRACT

Polycystic ovary syndrome (PCOS) is a prevalent endocrine condition characterized by a range of endocrine, reproductive, and metabolic abnormalities. At present, management of women with PCOS is suboptimal as treatment is only symptomatic. Clinical and experimental advances in our understanding of PCOS etiology support a pivotal role for androgen neuroendocrine actions in PCOS pathogenesis. Hyperandrogenism is a key PCOS trait and androgen actions play a role in regulating the kisspeptin-/neurokinin B-/dynorphin (KNDy) system. This study aimed to investigate if targeted antagonism of neurokinin B signaling through the neurokinin 3 receptor (NK3R) would reverse PCOS traits in a dihydrotestosterone (DHT)-induced mouse model of PCOS. After 3 months, DHT exposure induced key reproductive PCOS traits of cycle irregularity and ovulatory dysfunction, and PCOS-like metabolic traits including increased body weight; white and brown fat pad weights; fasting serum triglyceride and glucose levels, and blood glucose incremental area under the curve. Treatment with a NK3R antagonist (MLE4901) did not impact the observed reproductive defects. In contrast, following NK3R antagonist treatment, PCOS-like females displayed decreased total body weight, adiposity, and adipocyte hypertrophy, but increased respiratory exchange ratio, suggesting NK3R antagonism altered the metabolic status of the PCOS-like females. NK3R antagonism did not improve circulating serum triglyceride or fasted glucose levels. Collectively, these findings demonstrate that NK3R antagonism may be beneficial in the treatment of adverse metabolic features associated with PCOS and support neuroendocrine targeting in the development of novel therapeutic strategies for PCOS.


Subject(s)
Lectins/administration & dosage , Membrane Proteins/administration & dosage , Polycystic Ovary Syndrome/drug therapy , Receptors, Neurokinin-3/antagonists & inhibitors , Androgens/blood , Animals , Blood Glucose/metabolism , Dihydrotestosterone/adverse effects , Disease Models, Animal , Female , Humans , Hyperandrogenism/genetics , Hyperandrogenism/metabolism , Mice , Mice, Inbred C57BL , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Receptors, Neurokinin-3/genetics , Receptors, Neurokinin-3/metabolism , Triglycerides/blood
12.
Climacteric ; 24(1): 25-31, 2021 02.
Article in English | MEDLINE | ID: mdl-33135940

ABSTRACT

Vasomotor symptoms (hot flushes, flashes, night sweats) occur in the majority of menopausal women, and are reported as being of the highest symptom priority as they often persist over many years and can be highly disruptive. Hormone therapy is the most effective available treatment but is not without risk if taken long term, and is sometimes contraindicated; for example, in women with a personal or family history of breast cancer, which is the most common female cancer worldwide. Other treatment alternatives are not as efficacious, can cause side effects, and/or are not widely available. A new, effective, targeted treatment could therefore benefit millions of women worldwide. This became possible to investigate after accumulated evidence from both animal and human models implicated heightened signaling of a hypothalamic neuropeptide together with its receptor (neurokinin B/NK3R) in the etiology of sex-steroid-deficient vasomotor symptoms. Four clinical trials of three chemically distinct oral NK3R antagonists for the treatment of menopausal flushes have since completed and published, which consistently demonstrate efficacy and tolerability of these agents. These suggest great promise to change practice in the future if ongoing further larger-scale studies of longer duration confirm the same; as, estrogen exposure will no longer be required to effectively and safely treat vasomotor symptoms.


Subject(s)
Hot Flashes/drug therapy , Menopause , Receptors, Neurokinin-3/antagonists & inhibitors , Female , Humans , Randomized Controlled Trials as Topic
13.
Bull Cancer ; 107(11): 1171-1185, 2020 Nov.
Article in French | MEDLINE | ID: mdl-32988609

ABSTRACT

Breast cancer is the most frequently diagnosed cancer in women and the first cause of cancer death in France. Among the different subtypes of breast cancer, the predominant form is characterized by positive hormone receptors (more than 70% of breast cancers). Hormone therapy thus plays a key role in the strategy of management of these cancers both in adjuvant and metastatic situations. The two types of adjuvant hormone therapy used are selective estrogen receptor modulators and aromatase inhibitors. Fulvestrant, an anti-estrogen, is used alone or in combination with other molecules in metastatic situations. Hot flashes are one of the symptoms most frequently reported by patients under hormone therapy. Hormone replacement therapy, which is currently the most effective treatment for hot flashes, is contraindicated in patients with a personal history of breast cancer. Various therapeutic classes of drugs have been tested in this indication but without real efficacy in the various studies carried out to date, and moreover associated with non-negligible side effects. The recent discovery of the implication of the kisspeptin system located at the hypothalamic level in the mechanism of genesis of hot flashes opens the way to possible new symptomatic treatments for hot flashes. Neurokinin 3 receptor antagonists have shown encouraging preliminary results in postmenopausal cancer-free patients and could be considered in patients in hormonal therapy for breast cancer. Broader additional studies are needed to confirm these initial results.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/chemistry , Breast Neoplasms/drug therapy , Hot Flashes/etiology , Selective Estrogen Receptor Modulators/therapeutic use , Contraindications, Drug , Estrogen Replacement Therapy/adverse effects , Female , Fulvestrant/therapeutic use , Humans , Kisspeptins/physiology , Ovary/drug effects , Ovary/surgery , Receptors, Neurokinin-3/antagonists & inhibitors , Selective Estrogen Receptor Modulators/administration & dosage , Tamoxifen/administration & dosage , Tamoxifen/therapeutic use
14.
J Clin Endocrinol Metab ; 105(12)2020 12 01.
Article in English | MEDLINE | ID: mdl-32946574

ABSTRACT

CONTEXT: SJX-653 is a novel neurokinin 3 receptor (NK3R) antagonist. The NK3 pathway is a central regulator of gonadotropin releasing hormone (GnRH) secretion and has also been implicated in the generation of hot flashes. Therefore, decreases of luteinizing hormone (LH) and testosterone in men serve as sensitive pharmacodynamic (PD) markers of central NK3 antagonism. OBJECTIVE: To characterize the safety, tolerability, pharmacokinetics, and pharmacodynamic activity of SJX-653 in healthy men. DESIGN: A randomized, placebo-controlled, double-blind, single ascending dose study. SETTING: Phase 1 unit. PATIENTS OR OTHER PARTICIPANTS: Seven cohorts of 6 healthy men 18-45 years of age (4:2 randomization to SJX-653/placebo per cohort). INTERVENTION(S): Single oral doses of 0.5-90 mg SJX-653. MAIN OUTCOME MEASURE(S): Safety assessments and serial pharmacokinetic (PK)/PD measurements. RESULTS: SJX-653 was well tolerated at all dose levels. Cmax and AUC0-24 increased in a dose-proportional manner. The terminal elimination half-life ranged between 9.8 and 12.5 hours independent of dose. A statistically significant, dose-dependent, reversible reduction of LH and testosterone was observed with near maximal effect after 15 mg and little to no effect at 4.5 mg. Maximal LH reduction was 70 ±â€…7% (mean ±â€…sd) at 6 hours after 30 mg SJX-653 versus 10 ±â€…43% for placebo (P = 0.0006); maximal T reduction was of 68 ±â€…5% at 8 hours after 60 mg SJX-653 versus 18 ±â€…11% for placebo (P < 0.0001). The plasma IC50 for LH reduction was 33 ng/mL. CONCLUSIONS: These data demonstrate clinical proof-of-mechanism for SJX-653 as a potent centrally-acting NK3R antagonist.


Subject(s)
Hormone Antagonists/pharmacokinetics , Organic Chemicals/pharmacokinetics , Receptors, Neurokinin-3/antagonists & inhibitors , Adolescent , Adult , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Healthy Volunteers , Hormone Antagonists/administration & dosage , Hormone Antagonists/adverse effects , Humans , Male , Middle Aged , Organic Chemicals/administration & dosage , Organic Chemicals/adverse effects , Young Adult
15.
Expert Opin Ther Pat ; 30(7): 527-539, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32401556

ABSTRACT

INTRODUCTION: The tachykinin family of peptides (substance P, neurokinin A) via the neurokinin-1 (NK-1), NK-2, and NK-3 receptors is involved in many physiological/physiopathological actions. Antagonists of these receptors may be used to treat many human pathologies. AREAS COVERED: This review offers an overview (from 2014 to present) of the actions exerted by NK receptor (NK-R) antagonists on emesis, pruritus, cardiomyopathy, respiratory tract diseases, bacterial infection, cancer, ocular pain, corneal neovascularization, excess of body fat/weight, conditioned fear, social isolation stress, hot flush, melanogenesis, follicle development, fish reproduction, and sex-hormone-dependent diseases. EXPERT OPINION: From 2014, no invention has been published using NK-2R antagonists. Although the tachykinin/NK receptor system is involved in a great number of mechanisms, to date, the use of only five NK-1R antagonists have been approved in humans but no NK-2R or NK-3R antagonist. NK receptor antagonists are safe in human trials and are potential therapeutic agents, but this potential is currently minimized. In humans, more studies on molecules acting as NK receptor antagonists and exerting a potential therapeutic action must be carried out. The antipruritic or antitumor action of NK-1R antagonists must be explored in greater depth: the highest safe dose and the time of administration (for a long period of time) of these antagonists must be well established.


Subject(s)
Neurokinin-1 Receptor Antagonists/pharmacology , Receptors, Neurokinin-2/antagonists & inhibitors , Receptors, Neurokinin-3/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Humans , Patents as Topic , Receptors, Neurokinin-1/drug effects , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-2/metabolism , Receptors, Neurokinin-3/metabolism
16.
J Reprod Dev ; 66(4): 351-357, 2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32281549

ABSTRACT

Accumulating evidence suggests that KNDy neurons located in the hypothalamic arcuate nucleus (ARC), which are reported to express kisspeptin, neurokinin B, and dynorphin A, are indispensable for the gonadotropin-releasing hormone (GnRH) pulse generation that results in rhythmic GnRH secretion. The aims of the present study were to investigate the effects of peripheral administration of the neurokinin 3 receptor (NK3R/TACR3, a receptor for neurokinin B) antagonist, SB223412, on GnRH pulse-generating activity and pulsatile luteinizing hormone (LH) secretion in ovariectomized Shiba goats treated with luteal phase levels of estrogen. The NK3R antagonist was infused intravenously for 4 h {0.16 or 1.6 mg/(kg body weight [BW]·4 h)} during which multiple unit activity (MUA) in the ARC was recorded, an electrophysiological technique commonly employed to monitor GnRH pulse generator activity. In a separate experiment, the NK3R antagonist (40 or 200 mg/[kg BW·day]) was administered orally for 7 days to determine whether the NK3R antagonist could modulate pulsatile LH secretion when administered via the oral route. Intravenous infusion of the NK3R antagonist significantly increased the interval of episodic bursts of MUA compared with that of the controls. Oral administration of the antagonist for 7 days also significantly prolonged the interpulse interval of LH pulses. The results of this study demonstrate that peripheral administration of an NK3R antagonist suppresses pulsatile LH secretion by acting on the GnRH pulse generator, suggesting that NK3R antagonist administration could be used to modulate reproductive functions in ruminants.


Subject(s)
Estradiol/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Luteinizing Hormone/blood , Neurons/drug effects , Quinolines/pharmacology , Receptors, Neurokinin-3/antagonists & inhibitors , Administration, Oral , Animals , Female , Goats , Injections, Intravenous , Neurons/metabolism , Ovariectomy
17.
Brain Res ; 1734: 146729, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32067964

ABSTRACT

The neurokinins are a class of peptide signaling molecules that mediate a range of central and peripheral functions including pain processing, gastrointestinal function, stress responses, and anxiety. Recent data have linked these neuropeptides with drug-related behaviors. Specifically, substance P (SP) and neurokinin B (NKB), have been shown to influence responses to alcohol, cocaine, and/or opiate drugs. SP and NKB preferentially bind to the neurokinin-1 receptor (NK1R) and neurokinin-3 receptor (NK3R), respectively, but do have some affinity for all classes of neurokinin receptor at high concentrations. NK1R activity has been shown to influence reward and reinforcement for opiate drugs, stimulatory and neurochemical responses to cocaine, and escalated and stress-induced alcohol seeking. In reinstatement models of relapse-like behavior, NK1R antagonism attenuates stress-induced reinstatement for all classes of drugs tested to date. The NK3R also influences alcohol intake and behavioral/neurochemical responses to cocaine, but less research has been performed in regard to this particular receptor in preclinical models of addiction. Clinically, agents targeting these receptors have shown some promise, but have produced mixed results. Here, the preclinical findings for the NK1R and NK3R are reviewed, and discussion is provided to interpret clinical findings. Additionally, important factors to consider in regards to future clinical work are suggested.


Subject(s)
Behavior, Addictive/metabolism , Neurokinin-1 Receptor Antagonists/therapeutic use , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-3/metabolism , Substance-Related Disorders/metabolism , Alcoholism/drug therapy , Alcoholism/metabolism , Animals , Behavior, Addictive/drug therapy , Humans , Neurokinin-1 Receptor Antagonists/pharmacology , Receptors, Neurokinin-3/antagonists & inhibitors , Reward , Substance-Related Disorders/drug therapy
18.
J Clin Endocrinol Metab ; 104(12): 5893-5905, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31415087

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.


Subject(s)
Autonomic Nervous System Diseases/drug therapy , Heterocyclic Compounds, 2-Ring/therapeutic use , Menopause/physiology , Receptors, Neurokinin-3/antagonists & inhibitors , Thiadiazoles/therapeutic use , Vasomotor System/drug effects , Adult , Aged , Autonomic Nervous System Diseases/etiology , Belgium , Double-Blind Method , Female , Hot Flashes/drug therapy , Hot Flashes/etiology , Humans , Middle Aged , Quality of Life , Treatment Outcome
19.
Bioorg Med Chem ; 27(10): 2019-2026, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30975505

ABSTRACT

Neurokinin-3 receptor (NK3R) plays a pivotal role in the release of gonadotropin-releasing hormone in the hypothalamus-pituitary-gonadal (HPG) axis. To develop novel NK3R antagonists with less environmental toxicity, a series of heterocyclic scaffolds for the triazolopiperazine substructure in an NK3R antagonist fezolinetant were designed and synthesized. An isoxazolo[3,4-c]piperidine derivative exhibited moderate NK3R antagonistic activity and favorable properties that were decomposable under environmental conditions.


Subject(s)
Piperidines/chemistry , Receptors, Neurokinin-3/antagonists & inhibitors , Crystallography, X-Ray , Heterocyclic Compounds, 2-Ring/chemical synthesis , Heterocyclic Compounds, 2-Ring/chemistry , Inhibitory Concentration 50 , Molecular Conformation , Photolysis , Piperidines/metabolism , Receptors, Neurokinin-3/metabolism , Structure-Activity Relationship , Sunlight , Thiadiazoles/chemical synthesis , Thiadiazoles/chemistry
20.
Neuroendocrinology ; 109(3): 242-248, 2019.
Article in English | MEDLINE | ID: mdl-30504731

ABSTRACT

Menopause is associated with significant symptomatic burden, with approximately two-thirds of postmenopausal women suffering from vasomotor symptoms, hot flushes, and night sweats. The mainstay of treatment for hot flushes continues to be hormone replacement therapy. However, as hormone replacement therapy is contraindicated in some cases, alternative, efficacious treatment options are also required. Hot flushes are thought to arise as a result of significant changes in the neuroendocrine circuitry underpinning the reproductive axis during menopause. This includes reduced circulating ovarian oestrogens, hypersecretion of gonadotropins, and increased expression of kisspeptin and neurokinin B (NKB) within the infundibular nucleus of the hypothalamus. In recent years, NKB, predominantly acting via the neurokinin 3 receptor (NK3R), has emerged as an important player in the development of menopausal hot flushes. Antagonism of NK3R has garnered much interest as a novel therapeutic target to help ameliorate hot flush symptoms. Improvements in hot flush frequency, severity, and quality of life have been demonstrated in a number of clinical trials using novel NK3R antagonists in postmenopausal women. Within this review, we will explore the growing body of evidence supporting antagonism of NK3R as a potentially promising treatment for menopausal hot flushes.


Subject(s)
Hot Flashes/drug therapy , Menopause/drug effects , Receptors, Neurokinin-3/antagonists & inhibitors , Animals , Female , Hot Flashes/etiology , Hot Flashes/metabolism , Humans , Menopause/metabolism , Receptors, Neurokinin-3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...