WO2023147135A1 - Nk receptor antagonists for treatment of prostate cancer - Google Patents

Nk receptor antagonists for treatment of prostate cancer Download PDF

Info

Publication number
WO2023147135A1
WO2023147135A1 PCT/US2023/011892 US2023011892W WO2023147135A1 WO 2023147135 A1 WO2023147135 A1 WO 2023147135A1 US 2023011892 W US2023011892 W US 2023011892W WO 2023147135 A1 WO2023147135 A1 WO 2023147135A1
Authority
WO
WIPO (PCT)
Prior art keywords
patient
antagonist
administered
osanetant
pharmaceutically acceptable
Prior art date
Application number
PCT/US2023/011892
Other languages
French (fr)
Inventor
Jefferson E. Davis
William Patrick Parker
Elizabeth Marie WULFF-BURCHFIELD
Original Assignee
Acer Therapeutics, Inc.
University Of Kansas
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Acer Therapeutics, Inc., University Of Kansas filed Critical Acer Therapeutics, Inc.
Publication of WO2023147135A1 publication Critical patent/WO2023147135A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • NK neurokinin
  • Prostate cancer is a hormonally responsive tumor. Achieving and maintaining effective suppression of serum testosterone levels in men can assist with the management of prostate cancer. Historically, a serum testosterone below 50 ng/dL was considered to be the castrate level. Current data suggest that the new target for either surgical or chemical castration is a serum testosterone level of lower than 20 ng/dL. Currently used androgen deprivation therapies may cause testosterone levels to periodically rise, sometimes to non-castrate levels.
  • a method for prostate cancer or delaying progression of prostate cancer in a patient comprising administering a Neurokinin 3 receptor (NK3) antagonist to the patient in need thereof.
  • the NK3 antagonist is administered in an amount to reversibly lower the patient’s serum testosterone to lower than or equal to 50 ng/dL, 20 ng/dL, or 10 ng/dL, or the testosterone level is undetectable.
  • the NK3 antagonist is osanetant.
  • the patient is a low risk patient, an intermediate risk patient, or a high risk patient.
  • the patient has previously undergone or is currently undergoing androgen deprivation therapy (ADT).
  • ADT androgen deprivation therapy
  • the prostate cancer is hormone responsive. DETAILED DESCRIPTION
  • references to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se.
  • the term “about” includes the indicated amount ⁇ 10%.
  • the term “about” includes the indicated amount ⁇ 5%.
  • the term “about” includes the indicated amount ⁇ 1%.
  • to the term “about X” includes description of “X.”
  • Hormone responsive prostate cancer is any prostate cancer that may be treated, or delayed from progression, by a lowering of serum testosterone levels in a prostate cancer patient.
  • “Pharmaceutically acceptable” or “physiologically acceptable” refer to compounds, salts, compositions, dosage forms and other materials which are useful in preparing a pharmaceutical composition that is suitable for human or veterinary pharmaceutical use.
  • pharmaceutically acceptable salt of a given compound refers to salts that retain the biological effectiveness and properties of the given compound, and which are not biologically or otherwise undesirable.
  • “Pharmaceutically acceptable salts” or “physiologically acceptable salts” include, for example, salts with inorganic acids and salts with an organic acid.
  • the free base can be obtained by basifying a solution of the acid salt.
  • an addition salt, particularly a pharmaceutically acceptable addition salt may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds.
  • Pharmaceutically acceptable acid addition salts may be prepared from inorganic and organic acids. Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like.
  • pharmaceutically acceptable base addition salts can be prepared from inorganic and organic bases. Salts derived from inorganic bases include, by way of example only, sodium, potassium, lithium, ammonium, calcium and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines.
  • suitable amines include, by way of example only, isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2- dimethylaminoethanol, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like.
  • “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • a “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.
  • the present disclosure contemplates various stereoisomers and mixtures thereof and includes “enantiomers,” which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
  • a “prodrug” is any compound which releases an active parent drug according to a structure described herein in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of a compound described herein are prepared by modifying functional groups present in the compound described herein in such a way that the modifications may be cleaved in vivo to release the parent compound.
  • Prodrugs may be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds.
  • Prodrugs include compounds described herein wherein a hydroxy, amino, carboxyl, or sulfhydryl group in a compound described herein is bonded to any group that may be cleaved in vivo to regenerate the free hydroxy, amino, or sulfhydryl group, respectively.
  • Examples of prodrugs include, but are not limited to esters (e.g., acetate, formate and benzoate derivatives), amides, guanidines, carbamates (e.g. , N,N-dimethylaminocarbonyl) of hydroxy functional groups in compounds described herein and the like. Preparation, selection and use of prodrugs is discussed in T. Higuchi and V.
  • solvate refers to a complex formed by combining a compound and a solvent.
  • hydrate refers to a complex formed by combining a compound and water (i.e., a solvate when the solvent is water).
  • acid salt hydrate refers to a complex formed by combining an acid salt compound with water.
  • N-oxide refers to an oxidized tertiary or pyridinyl amine moiety.
  • isomorphic crystalline form refers to two or more crystalline forms that have the same space group, unit-cell dimensions, and types and positions of atoms, with the exception of a replacement of one or more atoms in one isomorphic crystalline form with a different atom in its counterpart isomorphic crystalline form.
  • a therapeutic amount can be administered to the patient, which can be determined by the treating physician, medical professional, or the like.
  • a therapeutic amount is administered orally.
  • a therapeutic amount is administered subcutaneously.
  • a therapeutic amount is administered transdermally.
  • a therapeutic amount is administered intravenously.
  • administering and “administration of,” when used in connection with a compound or tablet (and grammatical equivalents) refer both to direct administration, which may be administration to a patient by a medical professional or by self- administration by the patient, and/or to indirect administration, which may be the act of prescribing a drug. Administration entails delivery to the patient of the drug.
  • dose refers to the total amount of an active agent (e.g., osanetant or a pharmaceutically acceptable salt thereof) administered to a patient in a single day (24-hour period).
  • the desired dose can be administered once daily.
  • the desired dose may be administered in one, two, three, four or more sub-doses at appropriate intervals throughout the day, where the cumulative amount of the sub-doses equals the amount of the desired dose administered in a single day.
  • dose and “dosage” are used interchangeably herein.
  • an effective amount refers to an amount of a drug or an agent (e.g., osanetant or a pharmaceutically acceptable salt thereof) that when administered to a patient suffering from a condition, will have the intended therapeutic effect, e.g., alleviation, amelioration, palliation or elimination of one or more manifestations of the condition in the patient.
  • the full therapeutic effect does not necessarily occur by administration of one dose, and can occur only after administration of a series of doses and can be administered in one dose form or multiples thereof.
  • 500 mg of the drug can be administered in a single 500 mg strength tablet or two 250 mg strength tablets.
  • a therapeutically effective amount may be administered in one or more administrations.
  • the term “patient” or “subject” refers to a mammal, such as a human, bovine, rat, mouse, dog, monkey, ape, goat, sheep, cow, or deer.
  • a patient as described herein can be a human.
  • the patient can be a male or a female.
  • treatment covers the treatment of a human patient, and includes: (a) reducing the risk of occurrence of the condition in a patient determined to be predisposed to the disease but not yet diagnosed as having the condition, (b) impeding the development of the condition, and/or (c) relieving the condition, i.e., causing regression of the condition and/or relieving one or more symptoms of the condition.
  • prevention means any treatment of a disease or condition that causes the clinical symptoms of the disease or condition not to develop.
  • Compounds may, in some embodiments, be administered to a subject (including a human) who is at risk or has a family history of the disease or condition.
  • a method for treating prostate cancer or delaying progression of prostate cancer in a patient comprising administering a Neurokinin 3 receptor (NK3) antagonist to the patient in need thereof.
  • the prostate cancer is hormone responsive prostate cancer.
  • the NK3 antagonist is administered in an amount to reversibly lower the patient’s serum testosterone to castration levels.
  • the patient’s serum testosterone is reversibly reduced to lower than or equal to about 20 ng/dL.
  • the patient’s serum testosterone is reversibly reduced to lower than or equal to about 15 ng/dL.
  • the patient’s serum testosterone is reversibly reduced to lower than or equal to about 12 ng/dL.
  • the patient’s serum testosterone is reversibly reduced to lower than or equal to about 10 ng/dL. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to about 8 ng/dL. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to about 5 ng/dL.
  • the patient’s serum testosterone is reversibly reduced to lower than or equal to about 50 ng/dL. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to 40 ng/dL. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to 25 ng/dL.
  • the NK3 antagonist is a dual NK1/NK3 antagonist. In some embodiments, the NK3 antagonist is a selective NK3 antagonist.
  • the NK3 antagonist is selected from osanetant (SR-142,801), elinzanetant (NT-814), fezolinetant (ESN-364), talnetant (SB-223,412), pavinetant (AZD-4901/AZD- 2624), and SJX-653, or a pharmaceutically acceptable salt of each thereof.
  • the NK3 antagonist is selected from osanetant (SR-142,801), elinzanetant (NT-814), fezolinetant (ESN-364), talnetant (SB-223,412), and pavinetant (AZD-4901/AZD- 2624), or a pharmaceutically acceptable salt of each thereof.
  • the NK3 antagonist is administered to the patient once a day. In some embodiments, the NK3 antagonist is administered to the patient twice a day.
  • the NK3 antagonist is osanetant, or a pharmaceutically acceptable salt thereof.
  • the osanetant, or a pharmaceutically acceptable salt thereof is administered to the patient once or twice a day for a total daily dose ranging from about 10 mg to about 800 mg, about 25 to about 750 mg per day, about 50 to about 700 mg per day, about 50 to about 600 mg per day, about 50 to about 500 mg per day, about 50 to about 400 mg per day, about 75 to about 650 mg per day, about 70 to about 500 mg per day, about 75 to about 400 mg per day, about 100 to about 600 mg per day, about 100 to about 500 mg per day, or about 100 to about 400 mg per day.
  • the effective amount of osanetant, or a pharmaceutically acceptable salt thereof is less than about 400 mg per day. In some embodiments, the effective amount of osanetant, or a pharmaceutically acceptable salt thereof, is from about 10 to about 350 mg per day. In some embodiments, the effective amount of osanetant, or a pharmaceutically acceptable salt thereof, is less than about 200 mg per day. In some embodiments, the effective amount of osanetant, or a pharmaceutically acceptable salt thereof, is from about 10 to about 150 mg per day. In some embodiments, the effective amount of osanetant, or a pharmaceutically acceptable salt thereof, is about 300 mg per day. In some embodiments, the osanetant is administered once a day. In some embodiments, the osanetant, or a pharmaceutically acceptable salt thereof, is administered twice a day, each dose being about 150 mg.
  • the osanetant, or a pharmaceutically acceptable salt thereof is administered to the patient twice a day for a total daily dose ranging from about 10 mg to about 800 mg, about 25 to about 750 mg per day, about 50 to about 700 mg per day, about 50 to about 600 mg per day, about 50 to about 500 mg per day, about 50 to about 400 mg per day, about 75 to about 650 mg per day, about 70 to about 500 mg per day, about 75 to about 400 mg per day, about 100 to about 600 mg per day, about 100 to about 500 mg per day, or about 100 to about 400 mg per day.
  • the osanetant, or a pharmaceutically acceptable salt thereof is administered twice a day, each dose an equal fraction of the total daily dose and comprising from about 5 mg to about 400 mg of osanetant, or a pharmaceutically acceptable salt thereof.
  • hormone-dependent cancers such as prostate, and testicular cancers
  • the proliferation of cells is driven by hormone -receptor interactions on cell surfaces.
  • these sex hormones namely estrogen, progesterone, and testosterone
  • the hormone-dependent cells replicate more frequently, increasing the opportunity for genetic errors to occur and accumulate, potentially leading to cancer.
  • Pharmaceutical interventions e.g. hormone deprivation therapy such as androgen deprivation therapy, for the treatment or prevention of hormone-dependent cancers include compounds that inhibit the synthesis of these sex hormones, such as gonadotropin-releasing hormone (GnRH) agonists and antagonists, or nonsteroidal antiandrogens (NS A As).
  • GnRH gonadotropin-releasing hormone
  • NS A As nonsteroidal antiandrogens
  • the patient will be undergoing hormone deprivation therapy.
  • the patient may be undergoing androgen deprivation therapy (ADT).
  • ADT comprises administration of a gonadotropin releasing hormone (GnRH) agonist, a GnRH antagonist, a histamine H2 receptor antagonist, a nonsteroidal antiandrogen (NSAA), or an androgen receptor antagonist.
  • GnRH gonadotropin releasing hormone
  • NSAA nonsteroidal antiandrogen
  • the androgen deprivation therapy comprises surgical castration, luteinizing hormone -releasing hormone (LHRH) agonists (e.g.
  • leuprolide goserelin, triptorelin, or leuprolide mesylate
  • LHRH antagonists e.g., degarelix, relugolix, elagolix
  • abiraterone e.g., abiraterone, ketoconzale, etc.
  • GnRH agonist e.g., leuprolide (also known as leuprorelin)
  • GnRH antagonist e.g.
  • elagolix linzagolix, relugolix
  • goserelin triptorelin
  • buserelin deslorelin, fertirelin, gonadorelin, histrelin, lecirelin, nafarelin, peforelin, triptorelin, abarelix, cetrorelix, degarelix, or ganirelix.
  • an androgen receptor antagonist e.g., darolutamide
  • the patient is undergoing androgen deprivation therapy and the NK3 antagonist is administered in combination with the androgen deprivation therapy.
  • the androgen deprivation therapy is leuprolide, darulotamide, elagolix, linzagolix, or relugolix.
  • the androgen deprivation therapy is leuprolide, elagolix, linzagolix, relugolix, goserelin, triptorelin, buserelin, deslorelin, fertirelin, gonadorelin, histrelin, lecirelin, nafarelin, peforelin, triptorelin, abarelix, cetrorelix, degarelix, or ganirelix.
  • the hormone deprivation therapy is surgery e.g., orchiectomy.
  • the patient is co-administered estrogen deprivation therapy.
  • gynemastia and breast pain is reduced.
  • the estrogen deprivation therapy is selected from a selective estrogen modulator (SERM) or a selective estrogen receptor degrader (SERD).
  • the estrogen deprivation therapy is tamoxifen.
  • the SERM is raloxifene, toremifene, arzoxifene, or lasofoxifene, broparestrol, clomifene (E, or Z), cyclofenil, ormeloxifene.
  • the estrogen deprivation therapy is treatment with a selective estrogen receptor de
  • the osanetant is administered for a short period prior to onset of the androgen deprivation therapy, e.g., prior to prostatectomy or orchiectomy or prior to start of treatment with ADT (e.g., leuprolide, elagolix, linzagolix, or relugolix).
  • ADT e.g., leuprolide, elagolix, linzagolix, or relugolix.
  • the osanetant is administered for a period of 1 day to 1 month prior to onset of androgen deprivation therapy.
  • the osanetant is administered for a period of 1 day to 3 weeks prior to onset of androgen deprivation therapy.
  • the osanetant is administered for a period of 1 day to 2 weeks prior to onset of androgen deprivation therapy. In some embodiments, the osanetant is administered for a period of 1 day to 1 week prior to onset of androgen deprivation therapy. In some embodiments, the osanetant is administered for a period of 1 day to 5 days prior to onset of androgen deprivation therapy. In some embodiments, the osanetant is administered for a period of 1 day to 3 days prior to onset of androgen deprivation therapy.
  • the osanetant is administered prior to onset of androgen deprivation therapy, and during the androgen deprivation therapy. In some embodiments, the osanetant is administered after the androgen deprivation therapy (e.g., as maintenance therapy after cessation of primary tumor treatment (e.g., after prostatectomy or after orchiectomy).
  • the patient is a male patient with high levels of testosterone.
  • the patient is undergoing androgen deprivation therapy (ADT) and the
  • the ADT is leuprolide, elagolix, linzagolix, relugolix, goserelin, triptorelin, buserelin, deslorelin, fertirelin, gonadorelin, histrelin, lecirelin, nafarelin, peforelin, triptorelin, abarelix, cetrorelix, degarelix, ganirelix, flutamide, nilutamide, bicalutamide, topilutamide, apalutamide, enzalutamide, darolutamide, cimetidine, proxalutamide, seviteronel, cioteronel, inocoterone acetate, RU-58841 (CAS No.
  • the ADT is elagolix, linzagolix, or relugolix. In some embodiments, the ADT is leuprolide. In some embodiments, the ADT is darolutamide.
  • the patient treated with an NK3 antagonist is not undergoing ADT.
  • the NK3 antagonist is administered in combination with radiation.
  • the NK3 antagonist is administered in combination with ADT and radiation.
  • the levels of luteinizing hormone (LH) and/or testosterone (T) in the patient are each reduced by at least 70% compared to baseline levels of LH and T.
  • the patient has undergone prostatectomy. In some embodiments, the patient has undergone prostatectomy and is undergoing ADT and/or radiation therapy.
  • the methods described herein lower LH and/or testosterone levels in the patient and the reduction in levels of LH and/or T is reversible. In some embodiments, the methods described herein may reversibly lower LH, FSH, and estradiol levels.
  • the patient is suffering from non-metastatic prostate cancer. In some of such embodiments, the patient is not undergoing ADT. In some of such embodiments, administration of NK3 antagonists is useful in maintaining low levels of testosterone thereby slowing or delaying progression of prostate cancer.
  • the NK3 antagonist is administered for a period of at least 8 days. In some embodiments, the NK3 antagonist is administered for a period of 8 to 28 days. In some embodiments, the NK3 antagonist is administered for a period of at least 28 days. In some embodiments, the NK3 antagonist is administered for a period of at least 8 days at a first dose and thereafter administration is continued for a further period of at least 8 days at a second dose which is the same as, or different from, the first dose.
  • the androgen deprivation therapy is treatment with a gonadotropinreleasing hormone (GnRH) agonist or antagonist.
  • the patient is a male patient.
  • the GnRH agonist is leuprolide.
  • the GnRH antagonist is elagolix.
  • the GnRH antagonist is relugolix.
  • the GnRH antagonist is linzagolix.
  • the cancer is hormone receptor-positive cancer.
  • the hormone receptor-positive cancer is prostate cancer.
  • the neurokinin receptor antagonist is administered to a patient for a time period prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy as described herein.
  • the NK antagonist is administered concurrently with androgen deprivation therapy, a medical and/or surgical procedure and/or radiation therapy.
  • the patient continues to receive a NK antagonist after the androgen deprivation therapy, a medical and/or surgical procedure and/or radiation therapy.
  • the patient receives a NK antagonist after short-term (e.g., 1 to 6 months, 1 to 3 months, 1 month, 1-3 weeks, 1 week) androgen deprivation therapy. Any combination of these therapeutic regimens is contemplated within the scope of embodiments presented herein.
  • NK antagonists are useful in the methods described herein.
  • NK receptor As used herein, “NK receptor,” “neurokinin receptor,” or “tachykinin receptor” is a transmembrane G-protein coupled receptor. The three known tachykinin receptors are NK1, NK2, and NK3. These receptors act on a variety of human functions, which regulate numerous biological systems, including the reproductive system.
  • NK receptor antagonists are a class of drugs which interact with tachykinin receptors NK1, NK2, and NK3, and dampen the normal agonist-mediated biological responses.
  • the tachykinin receptors have been associated with the transmission of stress signals and pain, the contraction of smooth muscles, inflammation, and modulating the hypothalamus-pituitary-gonadal axis.
  • NK receptor antagonists are indicated for the treatment of migraine, emesis, gastrointestinal disorders, disorders of the reproductive system, and psychiatric disorders, including anxiety, addiction, depression, and schizophrenia.
  • the NK antagonist is a NK1, NK2, or NK3 antagonist or a combination thereof.
  • the NK antagonist is a NK1 receptor antagonist selected from aprepitant, casopitant, ezlopitant, fosaprepitant, lanepitant, maropitant, rolapitant, vestipitant, L-733,060, L-741,671, L-742,694, RP-67580, RPR-100,893, CP-96345, CP-99994, GR-205,171, TAK-637, T-2328, and combinations thereof.
  • the NK antagonist is a NK2 receptor antagonist selected from ibodutant, saredutant, GR-159,897, MEN-10376, and combinations thereof.
  • the NK antagonist is a NK3 receptor antagonist selected from fezolinetant, osanetant, pavinetant, talnetant, SB-222,200 (structure below), SB-218,795 (structure below), NT-814 (structure below), and combinations thereof.
  • the NK antagonist is a NK3 antagonist.
  • the NK3 receptor, and its associated tachykinin neuropeptide, neurokinin B act on a variety of human functions, affecting the hypothalamus-pituitary-gonadal axis, which regulates numerous biological systems, including the reproductive system.
  • the NK3 antagonist is osanetant.
  • the neurokinin-3 receptor antagonist is selected from osanetant, fezolinetant, pavinetant, talnetant, SB-222,200, SB-218,795, and NT-814.
  • the neurokinin-3 receptor antagonist is osanetant.
  • the neurokinin-3 receptor antagonist is fezolinetant. In certain embodiments, the neurokinin-3 receptor antagonist is pavinetant. In certain embodiments, the neurokinin-3 receptor antagonist is talnetant. In certain embodiments, the neurokinin-3 receptor antagonist is SB-222,200. In certain embodiments, the neurokinin-3 receptor antagonist is SB-218,795. In certain embodiments, the neurokinin-3 receptor antagonist is NT-814. In certain embodiments, the NK3 antagonist is osanetant.
  • Osanetant was originally developed for the treatment of schizophrenia and other central nervous system disorders.
  • the NK antagonist is osanetant or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof.
  • the chemical name of osanetant is (R)-N-(l-(3-(l-benzoyl-3-(3,4- dichlorophenyl)piperidin-3-yl)propyl)-4-phenylpiperidin-4-yl)-N-methylacetamide, and has the following structure:
  • Osanetant can also form pharmaceutically acceptable salts, such as osanetant hydrochloride, osanetant hydrobromide, osanetant sulfate, osanetant hydrogen sulfate, osanetant dihydrogen phosphate, osanetant methanesulfonate, osanetant methyl sulfate, osanetant maleate, osanetant fumarate, osanetant 2- naphthalenesulfonate, osanetant benzenesulfonate, osanetant glycolate, osanetant gluconate, and osanetant citrate, osanetant isethionate, osanetant p-toluenesulfonate, and the like.
  • osanetant is administered as a hydrochloride salt thereof.
  • Osanetant as well as pharmaceutically acceptable salts thereof, can be purchased from commercial sources or can synthesized using published procedures. Patient Selection
  • a patient selected for treatment using the methods described herein is a low risk patient, an intermediate risk patient, or a high risk patient.
  • a patient selected for treatment using the methods described herein is a low risk patient.
  • a low risk patient may be deemed to have “low risk prostate cancer”.
  • Prostate cancer is described as “low risk” if it is only found in the prostate (localized) and it is highly likely to grow only very slowly, or not at all (low risk of progression).
  • the medical criteria for low risk prostate cancer are: 1) the cancer is found in only one of the two sides (lobes) of the prostate; 2) the cancer takes up less than half of the affected prostate lobe; 3) the cancer cells have not mutated much and are not very aggressive; and/or 4) the cancer has not spread to any lymph nodes or led to the growth of tumors in other parts of the body.
  • Low risk patient is intended to encompass patients having low risk prostate cancer, e.g., a patient suffering from non-metastatic hormone responsive/sensitive cancer or a slow progression cancer. In some embodiments such a low risk patient does not elect to undergo androgen deprivation therapy.
  • a patient selected for treatment using the methods described herein is undergoing radiation therapy with or without radiation therapy.
  • a patient selected for treatment using methods described herein is being treated with lower doses of androgen deprivation therapy compared to the recommended dosing of androgen deprivation treatment.
  • a patient selected for treatment using the methods described herein is suffering from severe side effects of androgen deprivation therapy and is treated with lower doses of androgen deprivation therapy compared to the recommended dosing along with an NK receptor antagonist.
  • a “low risk patient” is also intended to include those patients who elect active surveillance over other therapies; who are older or have serious illnesses, where other therapies can be detrimental to their current conditions. Active surveillance can include regular PSA tests and/or biopsies.
  • a low risk patient in a low risk patient, the tumor is confined to the prostate, and the PSA is ⁇ 10 ng/mL or grade is group 1 (Gleason score is 6).
  • a low risk patient suffers from slow-growing tumors and may be a “very low risk” patient in which fewer than 3 biopsy tissue samples contain cancer cells and the cancer is not detectable by a digital rectal exam.
  • a patient selected for treatment using the methods described herein is an intermediate risk patient where the tumor is confined to the prostate, the PSA is between 10 and 20 ng/mL, or the grade is group 2 or 3 (Gleason score of 7).
  • the patient may have a “favorable” or an “unfavorable” intermediate risk.
  • a patient selected for treatment using the methods described herein is a high risk patient in whom the tumor extends outside the prostate, the PSA is >20 ng/mL, or grade group 4 or 5 (Gleason score 8 to 10).
  • a high risk patient has a subset of aggressive tumors and is a “very high risk” patient in whom the tumor has extended into the seminal vesicles (T3b) or the rectum or bladder (T4), or there are multiple biopsy samples with high grade cancer.
  • a patient selected for treatment using the methods described herein is a patient who will benefit from castration or lowering of testosterone levels to castration levels.
  • a patient selected for treatment using the methods described herein has been treated with higher than recommended doses or recommended doses of androgen deprivation therapy but has not had a lowering of testosterone to castration levels (e.g., lower than 20 ng/dL, or lower than 10 ng/dL).
  • the methods described herein provide for treatment with lower doses of the androgen deprivation therapeutic compared to the recommended dosing, and/or for a longer duration of time compared to the recommended duration, along with administration of an NK receptor antagonist, thereby reversibly lowering testosterone to castration levels in the patient.
  • “recommended dosing” or “recommended duration” refers to dosing and/or durations approved by health agencies and/or provided on product labels.
  • a method to reduce testosterone to non-detectable levels comprising administration of a lower dose of an ADT and driving testosterone to below castrate levels, e.g., less than 20 ng/dL, less than 10 ng/dL.
  • osanetant is administered in combination with the ADT.
  • the reduction in testosterone occurs faster than the reduction if a recommended dose of ADT would have been administered alone.
  • the lowest testosterone level achieved is lower than the level that would be achieved if a recommended dose of ADT would have been administered alone.
  • a patient selected for treatment using the methods described herein is not treated with NK3 antagonist MLE4901 (also known as AZD4901) at a dosage of 40 mg twice daily for 7 days.
  • a patient selected for treatment using the methods described herein is not treated with NK3 antagonist fezolinetant (ESN-364) at a dose of 3-180 mg as a single dose.
  • a patient selected for treatment using the methods described herein is not treated with NK3 antagonist fezolinetant (ESN-364) at a dose of 20, 60 or 180 mg once daily for 10 days.
  • a patient selected for treatment using the methods described herein is not treated with NK3 antagonist SJX-653 at a dose of 0.5 - 90 mg as a single dose.
  • the time period over which the neurokinin receptor antagonist is administered to a patient prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 12 weeks or 8 weeks or 4 weeks to the day of the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy. In some embodiments, the time period over which the neurokinin receptor antagonist is administered to a patient prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 3 weeks to the day of the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy.
  • the time period over which the neurokinin receptor antagonist is administered to a patient prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 2 weeks to the day of the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy. In some embodiments, the time period over which the neurokinin receptor antagonist is administered prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 2 weeks. In some embodiments, the time period over which the neurokinin receptor antagonist is administered prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 1 week.
  • the neurokinin receptor antagonist is administered to a patient concurrently with the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy.
  • the time period over which the neurokinin receptor antagonist is administered to a patient after the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 12 weeks. In some embodiments, the time period over which the neurokinin receptor antagonist is administered to a patient after the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 8 weeks. In some embodiments, the time period over which the neurokinin receptor antagonist is administered to a patient after the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 4 weeks.
  • the patient is administered osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, for about 1 week prior to, concurrently with, and for about 12 weeks after, an androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy.
  • the patient is administered osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, for about 2 weeks prior to, concurrently with, and for about 12 weeks or more after, an androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy.
  • the patient is administered osanetant or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, for about 1 week prior to, concurrently with, and for about 8 weeks after, an androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy.
  • the patient is administered osanetant for about 2 weeks prior to, concurrently with, and for about 8 weeks after, an androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy.
  • the neurokinin receptor antagonist is orally administered.
  • the neurokinin receptor antagonist is subcutaneously administered.
  • the neurokinin receptor antagonist is transdermally administered.
  • the neurokinin receptor antagonist is intravenously administered.
  • the neurokinin receptor antagonist is administered once daily. In some embodiments, the neurokinin receptor antagonist is administered as two, three, four or more sub-doses at appropriate intervals throughout the day, where the cumulative amount of the sub-doses equals the amount of the desired dose administered in a single day.
  • the therapeutically effective amount of the osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof is about 0.25 mg/day to about 1000 mg/day.
  • the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, osanetant is about 0.5 mg/day to about 500 mg/day.
  • the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof is about 0.75 mg/day to about 450 mg/day, is about 1 mg/day to about 400 mg/day or 10 mg/day to about 350 mg/day.
  • the osanetant is administered is less than about 400 mg/day. In some embodiments, the osanetant is administered is less than about 200 mg/day or about 10 mg/day to about 150 mg/day.
  • the therapeutically effective amount of the osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N- oxide or isomorphic crystalline form thereof is about 1 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 50 mg/day.
  • the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof is about 100 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 200 mg/day.
  • the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof is about 300 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 400 mg/day.
  • the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof is about 500 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 600 mg/day.
  • the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof is about 800 mg/day. In some embodiments, the therapeutically effective amount of osanetant is about 1000 mg/day.
  • osanetant or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is dosed at about 0.25 mg/day to about 1 mg/day, in order to treat bone loss while avoiding transient increases in liver transaminase (alanine aminotransferase) concentrations, and maintaining healthy liver function.
  • liver transaminase alanine aminotransferase
  • osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose ranging from about 5 mg per day to about 600 mg per day. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose ranging from about 25 mg per day to about 500 mg per day.
  • osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose ranging from about 50 mg per day to about 500 mg per day. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose ranging from about 100 mg per day to about 400 mg per day.
  • osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose ranging from about 150 mg per day to about 300 mg per day.
  • the total daily dose may be administered as a single dose, or split between multiple doses (e.g., daily dose of 300 mg QD or daily dose of 300 mg split into 150 mg BID, or daily dose of 300 mg split into 100 mg TID).
  • osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose of 300 mg, split into two doses of 150 mg each, i.e., 150 mg BID. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose of 200 mg, split into two doses of 100 mg each, i.e., 100 mg BID.
  • osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose of 100 mg, split into two doses of 50 mg each, i.e., 50 mg BID. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose of 50 mg, split into two doses of 25 mg each, i.e., 25 mg BID.
  • osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose of 25 mg, split into two doses of 12.5 mg each, i.e., 12.5 mg BID.
  • the dose of leuprolide, elagolix, relugolix, or linzagolix or any other ADT may be lower than the recommended dosing and may be for a longer duration of time compared to the recommended duration of therapy.
  • compositions that comprise osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, and one or more pharmaceutically acceptable vehicles selected from carrier, adjuvants, and excipients.
  • Suitable pharmaceutically acceptable vehicles may include, for example, inert solid diluents and fillers, diluents, including sterile aqueous solutions and various organic solvents, permeation enhancers, solubilizers, and adjuvants.
  • Such compositions are prepared in a manner well known in the pharmaceutical art. See, e.g., Remington’s Pharmaceutical Sciences, Mace Publishing Co., Philadelphia, Pa. 17th Ed. (1985); and Modern Pharmaceutics, Marcel Dekker, Inc. 3rd Ed. (G.S. Banker & C.T. Rhodes, Eds.).
  • the pharmaceutical compositions may be administered in either single or multiple doses.
  • the pharmaceutical composition may be administered by various methods including, for example, rectal, intravenous, subcutaneous, and transdermal routes.
  • the pharmaceutical composition may be administered by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.
  • the pharmaceutical composition may be in the form of a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be sterile injectable solution or suspension in a non-toxic parentally acceptable vehicle, for example as a solution in 1,3-butanediol.
  • a non-toxic parentally acceptable vehicle for example as a solution in 1,3-butanediol.
  • the acceptable vehicles that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid can be useful in the preparation of injectables.
  • Such solutions may be formulated as 0.01% -10% isotonic solutions, pH 5-7, with appropriate salts.
  • the compound described herein may be administered parenterally in a sterile medium.
  • Parenteral administration includes subcutaneous injections, intravenous, intramuscular, intrathecal injection or infusion techniques.
  • the compound described herein, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle.
  • adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle.
  • the carrier comprises at least 90% by weight of the total composition.
  • the carrier for parenteral administration is chosen from propylene glycol, ethyl oleate, pyrrolidone, ethanol, and sesame oil.
  • a pharmaceutical composition for example, for injection, may comprise a cyclodextrin.
  • the cyclodextrin may be, for example, a hydroxypropyl cyclodextrin or a sulfobutylether cyclodextrin.
  • the cyclodextrin may be, for example, an a-cyclodextrin, a P-cyclodextrin, or a y-cyclodextrin.
  • a compound described herein may also be administered via microspheres, liposomes, other microparticulate delivery systems or sustained release formulations placed in certain tissues including blood.
  • sustained release carriers include semi-permeable polymer matrices in the form of shared articles, e.g., suppositories or microcapsules. Examples can be found, e.g., in Remington's Pharmaceutical Sciences, 18th edition, Gennaro, A. R., Lippincott Williams & Wilkins; 20th edition (Dec. 15, 2000) ISBN 0-912734-04-3 and Pharmaceutical Dosage Forms and Drug Delivery Systems; Ansel, N. C. et al. 7th Edition ISBN 0-683305-72-7, the entire disclosures of which are herein incorporated by reference.
  • Oral administration may be another route for administration of the compounds described herein. Administration may be via, for example, capsule or enteric coated tablets.
  • the active ingredient is usually diluted by an excipient and/or enclosed within such a carrier that can be in the form of a capsule, sachet, paper or other container.
  • the excipient serves as a diluent, it can be in the form of a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, sterile injectable solutions, and sterile packaged powders.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose.
  • the formulations can additionally include lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl and propylhydroxybenzoates; sweetening agents; and flavoring agents.
  • compositions that include at least one compound described herein can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the subject by employing procedures known in the art.
  • Controlled release drug delivery systems for oral administration include osmotic pump systems and dissolutional systems containing polymer-coated reservoirs or drug-polymer matrix formulations. Examples of controlled release systems are given in U.S. Patent Nos. 3,845,770; 4,326,525; 4,902,514; and 5,616,345.
  • Another formulation for use in the methods disclosed herein employ transdermal delivery devices (“patches”). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds described herein in controlled amounts.
  • transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Patent Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • the principal active ingredient may be mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound described herein.
  • a pharmaceutical excipient When referring to these preformulation compositions as homogeneous, the active ingredient may be dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • the tablets or pills of the compounds described herein may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action, or to protect from the acid conditions of the stomach.
  • the tablet or pill can include an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer that serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • the compound described herein can be incorporated into oral liquid preparations such as aqueous or oily suspensions, solutions, emulsions, syrups, or elixirs, for example.
  • oral liquid preparations such as aqueous or oily suspensions, solutions, emulsions, syrups, or elixirs, for example.
  • pharmaceutical compositions containing the compound described herein can be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations can contain conventional additives, such as suspending agents (e.g., sorbitol syrup, methyl cellulose, glucose/sugar, syrup, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminum stearate gel, and hydrogenated edible fats), emulsifying agents (e.g., lecithin, sorbitan monooleate, or acacia), non-aqueous vehicles, which can include edible oils (e.g., almond oil, fractionated coconut oil, silyl esters, propylene glycol and ethyl alcohol), and preservatives (e.g., methyl or propyl p-hydroxybenzoate and sorbic acid).
  • suspending agents e.g., sorbitol syrup, methyl cellulose, glucose/sugar, syrup, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminum stearate gel, and hydrogenated edible fats
  • emulsifying agents e.g
  • compositions for inhalation or insufflation may include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described herein.
  • the compositions are administered by the oral respiratory route for local or systemic effect.
  • compositions in pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a facemask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, orally from devices that deliver the formulation in an appropriate manner.
  • compositions described herein may be administered, for example, as quick-dissolving tablets, lozenges, powders, sprays, ointments, gels, or liquid suspensions.
  • mucoadhesive agents include agarose, chitosan, trimethylated chitosan, chitosan-EDTA, gelatin, hyaluronic acid, guar gum, hakea gum, xanthan gum, gellan gum, carrageenan, pectin, sodium alginate, cellulose derivatives, CMC, thiolated CMC, sodium CMC, HEC, HPC, HPMC, MC, poly(acrylic acid)-based polymers, CP, PC, PAA, copolymers of acrylic acid and PEG, PVA, PVP, CP, aminodextran, dimethylaminoethyl-dextran, hydroxyethyl starch, poly(ethylene oxide), scleroglucan, cyanoacrylate, hydroxylated methacrylate, and poly(methacrylic acid).
  • Suitable penetration enhancers include sodium lauryl sulfate, cetyl pyridinium chloride, Poloxamer, Brij, Span, Myrj, Tween, sodium glycocholate, sodium tauro deoxycholate, sodium tauro cholate, oleic acid, caprylic acid, lauric acid, lyso phosphatidyl choline, phosphatidyl choline, a-, P-, and y-cyclodextrin, methylated P-cyclodextrin, EDTA, citric acid, sodium salicylate, methoxy salicylate, chitosan, trimethyl chitosan, poly-L-arginine, and L-lysine.
  • suitable enzyme inhibitors include aprotinin, bestatin, and puromycin.
  • the neurokinin receptor antagonist may be administered in combination with one or more additional active agents.
  • the neurokinin receptor antagonist may be administered in combination with a second active agent that the patient is being treated with.
  • the second active agent is an androgen deprivation agent such as a GnRH agonist, e.g. leuprolide.
  • the second active agent is an androgen deprivation agent such as a GnRH antagonist, e.g.
  • elagolix linzagolix, relugolix, goserelin, triptorelin, buserelin, deslorelin, fertirelin, gonadorelin, histrelin, lecirelin, nafarelin, peforelin, triptorelin, abarelix, cetrorelix, degarelix, or ganirelix.
  • the second active agent is an androgen deprivation agent such as a nonsteroidal antiandrogen (NSAA), including, but not limited to, flutamide, nilutamide, bicalutamide, topilutamide, apalutamide, enzalutamide, darolutamide, cimetidine, proxalutamide, seviteronel, cioteronel, inocoterone acetate, and RU-58841.
  • NSAA nonsteroidal antiandrogen
  • the second active agent is an androgen deprivation agent is a selective androgen receptor degrader (SARD), including, but not limited to, dimethylcurcumin, AZ3514, arv-110, MTX-23, ARD-61, and UT-34.
  • SARD selective androgen receptor degrader
  • the second active agent is an androgen deprivation agent is a selective androgen receptor modulator (SARM), including, but not limited to, FL442, MK-4541, LY2452473, GTx-024/Enobosarm, and GSK2881078.
  • SARM selective androgen receptor modulator
  • GnRH gonadotropin-releasing hormone agonists and antagonists
  • GnRH agonists and antagonists are classes of drugs which prevent the GnRH-mediated release of sex hormones.
  • GnRH is a peptide hormone produced by GnRH neurons in the hypothalamus, responsible for the release of follicle-stimulating hormone and luteinizing hormone from the pituitary gland, beginning the hypothalamic -pituitary-gonadal axis synthesis and release of sex hormones.
  • composition comprising osanetant and a second active agent.
  • This study will evaluate the use of osanetant to reduce testosterone in men with adenocarcinoma of the prostate.
  • the study is a single-arm pilot study. This study involves 10 patients with prostate cancer. All 10 will undergo prostatectomy and measurement of 4 hormones FSH, LH, Estradiol, and Testosterone) at baseline and days 2, 3, 7, 14, and 28.
  • Protocol synopsis Men will undergo baseline screening (serum PSA and testosterone and history and physical examination). Those men who met inclusion criteria and elect to participate will be scheduled for the study protocol, which begins 35 days prior to scheduled surgery. Osanetant will be administered by the investigational pharmacy at a dosing of 200 mg BID. On days 2, 3, 7, and 14, and 28, an AM (0800 +/- 30 min) testosterone, LH, FSH, and Estradiol levels will be obtained. On day 35 (1 week after completing protocol therapy), the patient will undergo standard of care surgery. On day 42 post-operatively (+/- 3 days) serum testosterone, LH, FSH, Estradiol, and PSA will be obtained.
  • Patient Sample Adult men with clinically localized prostate cancer (cTl-3bN0M0) who are scheduled for radical prostatectomy.
  • Inclusion criteria Age >18; Clinically localized disease: cTl-3bN0M0; Testosterone >150 ng/ml
  • Exclusion criteria Current or recent (within 6 months) use of testosterone/estrogen modulating agents (leuprolide, degarelix, bicalutamide, enzalutamide, apalutamide, darolutamide, abiraterone, systemic ketoconazole, tamoxifen, etc); Current use of CYP3A4 inhibitors; Cognitive impairment (defined as the presence of diagnosed dementia); Impaired renal function: Cr >1.8; Medical history of osteoporosis; Current systemic corticosteroid, long-term opioid, spironolactone, or eplerenone use.
  • testosterone/estrogen modulating agents leuprolide, degarelix, bicalutamide, enzalutamide, apalutamide, darolutamide, abiraterone, systemic ketoconazole, tamoxifen, etc
  • Current use of CYP3A4 inhibitors Cognitive impairment (defined as the presence of diagnosed dementia); Impaired renal function: Cr >1.8; Medical history
  • Endpoints Testosterone levels during treatment; LH, FSH, and Estradiol levels during treatment; PSA response to Osanetant; Rate of development of castrate testosterone levels (defined as testosterone ⁇ 50 ng/mL); Reversibility of hormonal changes.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure relates generally to a method of treating prostate cancer or delaying progression of prostate cancer comprising administration of a neurokinin (NK) receptor antagonist to a patient.

Description

NK RECEPTOR ANTAGONISTS FOR TREATMENT OF PROSTATE CANCER
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit under 35 U.S.C. § 119(e) of United States Provisional Application Serial Number 63/305,209, filed January 31, 2022, and United States Provisional Application Serial Number 63/306,215, filed February 3, 2022, the contents of which are hereby incorporated by reference in their entirety.
FIELD
[0002] Provided herein are methods for treating prostate cancer or delaying progression of prostate cancer by administering a neurokinin (NK) receptor antagonist.
BACKGROUND
[0003] Prostate cancer is a hormonally responsive tumor. Achieving and maintaining effective suppression of serum testosterone levels in men can assist with the management of prostate cancer. Historically, a serum testosterone below 50 ng/dL was considered to be the castrate level. Current data suggest that the new target for either surgical or chemical castration is a serum testosterone level of lower than 20 ng/dL. Currently used androgen deprivation therapies may cause testosterone levels to periodically rise, sometimes to non-castrate levels.
[0004] There is a need for therapies that can maximize therapeutic outcomes for prostate cancer by consistently achieving and/or maintaining the lowest testosterone levels possible.
SUMMARY
[0005] Provided herein is a method for prostate cancer or delaying progression of prostate cancer in a patient comprising administering a Neurokinin 3 receptor (NK3) antagonist to the patient in need thereof. In some embodiments, the NK3 antagonist is administered in an amount to reversibly lower the patient’s serum testosterone to lower than or equal to 50 ng/dL, 20 ng/dL, or 10 ng/dL, or the testosterone level is undetectable. In some embodiments, the NK3 antagonist is osanetant. In some embodiments, the patient is a low risk patient, an intermediate risk patient, or a high risk patient. In some embodiments, the patient has previously undergone or is currently undergoing androgen deprivation therapy (ADT). In some embodiments, the prostate cancer is hormone responsive. DETAILED DESCRIPTION
[0006] The following description sets forth exemplary embodiments of the present technology. It should be recognized, however, that such description is not intended as a limitation on the scope of the present disclosure but is instead provided as a description of exemplary embodiments.
[0007] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art. As used herein, the below terms have the following meanings unless specified otherwise. Any methods, devices and materials similar or equivalent to those described herein can be used in the practice of the compositions and methods described herein. The following definitions are provided to facilitate understanding of certain terms used frequently herein and are not meant to limit the scope of the present disclosure. All references referred to herein are incorporated by reference in their entirety.
[0008] The term “comprise” and variations thereof, such as, “comprises” and “comprising” are to be construed in an open, inclusive sense, that is, as “including, but not limited to.” Further, the singular forms “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. Thus, references to “the agent” includes a plurality of such agents.
[0009] Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. In certain embodiments, the term “about” includes the indicated amount ± 10%. In other embodiments, the term “about” includes the indicated amount ± 5%. In certain other embodiments, the term “about” includes the indicated amount ± 1%. Also, to the term “about X” includes description of “X.”
[0010] Hormone responsive prostate cancer is any prostate cancer that may be treated, or delayed from progression, by a lowering of serum testosterone levels in a prostate cancer patient.
[0011] “Pharmaceutically acceptable” or “physiologically acceptable” refer to compounds, salts, compositions, dosage forms and other materials which are useful in preparing a pharmaceutical composition that is suitable for human or veterinary pharmaceutical use.
[0012] The term “pharmaceutically acceptable salt” of a given compound refers to salts that retain the biological effectiveness and properties of the given compound, and which are not biologically or otherwise undesirable. “Pharmaceutically acceptable salts” or “physiologically acceptable salts” include, for example, salts with inorganic acids and salts with an organic acid. In addition, if the compounds described herein are obtained as an acid addition salt, the free base can be obtained by basifying a solution of the acid salt. Conversely, if the product is a free base, an addition salt, particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds. Those skilled in the art will recognize various synthetic methodologies that may be used to prepare nontoxic pharmaceutically acceptable addition salts. Pharmaceutically acceptable acid addition salts may be prepared from inorganic and organic acids. Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like. Likewise, pharmaceutically acceptable base addition salts can be prepared from inorganic and organic bases. Salts derived from inorganic bases include, by way of example only, sodium, potassium, lithium, ammonium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines. Specific examples of suitable amines include, by way of example only, isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2- dimethylaminoethanol, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like.
[0013] As used herein, “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
[0014] A “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present disclosure contemplates various stereoisomers and mixtures thereof and includes “enantiomers,” which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
[0015] A “prodrug” is any compound which releases an active parent drug according to a structure described herein in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound described herein are prepared by modifying functional groups present in the compound described herein in such a way that the modifications may be cleaved in vivo to release the parent compound. Prodrugs may be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds. Prodrugs include compounds described herein wherein a hydroxy, amino, carboxyl, or sulfhydryl group in a compound described herein is bonded to any group that may be cleaved in vivo to regenerate the free hydroxy, amino, or sulfhydryl group, respectively. Examples of prodrugs include, but are not limited to esters (e.g., acetate, formate and benzoate derivatives), amides, guanidines, carbamates (e.g. , N,N-dimethylaminocarbonyl) of hydroxy functional groups in compounds described herein and the like. Preparation, selection and use of prodrugs is discussed in T. Higuchi and V. Stella, “Pro-drugs as Novel Delivery Systems,” Vol. 14 of the A.C.S. Symposium Series; “Design of Prodrugs,” ed. H. Bundgaard, Elsevier, 1985; and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, each of which are hereby incorporated by reference in their entirety.
[0016] As used herein, the term “solvate” refers to a complex formed by combining a compound and a solvent.
[0017] As used herein, the term “hydrate” refers to a complex formed by combining a compound and water (i.e., a solvate when the solvent is water).
[0018] As used herein, the term “acid salt hydrate” refers to a complex formed by combining an acid salt compound with water.
[0019] As used herein, the term “N-oxide” refers to an oxidized tertiary or pyridinyl amine moiety.
[0020] As used herein, the term “isomorphic crystalline form” refers to two or more crystalline forms that have the same space group, unit-cell dimensions, and types and positions of atoms, with the exception of a replacement of one or more atoms in one isomorphic crystalline form with a different atom in its counterpart isomorphic crystalline form.
[0021] As used herein, the term “administration” refers to introducing an agent into a patient. For example, a therapeutic amount can be administered to the patient, which can be determined by the treating physician, medical professional, or the like. In some embodiments, a therapeutic amount is administered orally. In some embodiments, a therapeutic amount is administered subcutaneously. In some embodiments, a therapeutic amount is administered transdermally. In some embodiments, a therapeutic amount is administered intravenously. The related terms and phrases “administering” and “administration of,” when used in connection with a compound or tablet (and grammatical equivalents) refer both to direct administration, which may be administration to a patient by a medical professional or by self- administration by the patient, and/or to indirect administration, which may be the act of prescribing a drug. Administration entails delivery to the patient of the drug.
[0022] The term “dose” or “dosage” refers to the total amount of an active agent (e.g., osanetant or a pharmaceutically acceptable salt thereof) administered to a patient in a single day (24-hour period). The desired dose can be administered once daily. In some embodiments, the desired dose may be administered in one, two, three, four or more sub-doses at appropriate intervals throughout the day, where the cumulative amount of the sub-doses equals the amount of the desired dose administered in a single day. The terms “dose” and “dosage” are used interchangeably herein.
[0023] As used herein, “effective amount,” “therapeutically effective amount,” or “therapeutic amount” refers to an amount of a drug or an agent (e.g., osanetant or a pharmaceutically acceptable salt thereof) that when administered to a patient suffering from a condition, will have the intended therapeutic effect, e.g., alleviation, amelioration, palliation or elimination of one or more manifestations of the condition in the patient. The full therapeutic effect does not necessarily occur by administration of one dose, and can occur only after administration of a series of doses and can be administered in one dose form or multiples thereof. For example, 500 mg of the drug can be administered in a single 500 mg strength tablet or two 250 mg strength tablets. Thus, a therapeutically effective amount may be administered in one or more administrations.
[0024] As used herein, the term “patient” or “subject” refers to a mammal, such as a human, bovine, rat, mouse, dog, monkey, ape, goat, sheep, cow, or deer. A patient as described herein can be a human. The patient can be a male or a female.
[0025] As used herein, “treatment,” “treating,” and “treat” are defined as acting upon a disease, disorder, or condition with an agent to reduce or ameliorate the harmful or any other undesired effects of the disease, disorder, or condition and/or its symptoms. Treatment, as used herein, covers the treatment of a human patient, and includes: (a) reducing the risk of occurrence of the condition in a patient determined to be predisposed to the disease but not yet diagnosed as having the condition, (b) impeding the development of the condition, and/or (c) relieving the condition, i.e., causing regression of the condition and/or relieving one or more symptoms of the condition.
[0026] “Prevention” or “preventing” means any treatment of a disease or condition that causes the clinical symptoms of the disease or condition not to develop. Compounds may, in some embodiments, be administered to a subject (including a human) who is at risk or has a family history of the disease or condition. Methods
[0027] Provided herein is a method for treating prostate cancer or delaying progression of prostate cancer in a patient comprising administering a Neurokinin 3 receptor (NK3) antagonist to the patient in need thereof. In some embodiments, the prostate cancer is hormone responsive prostate cancer. In some embodiments, the NK3 antagonist is administered in an amount to reversibly lower the patient’s serum testosterone to castration levels. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to about 20 ng/dL. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to about 15 ng/dL. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to about 12 ng/dL. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to about 10 ng/dL. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to about 8 ng/dL. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to about 5 ng/dL.
[0028] In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to about 50 ng/dL. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to 40 ng/dL. In some embodiments, the patient’s serum testosterone is reversibly reduced to lower than or equal to 25 ng/dL.
[0029] In some embodiments, the NK3 antagonist is a dual NK1/NK3 antagonist. In some embodiments, the NK3 antagonist is a selective NK3 antagonist.
[0030] In some embodiments, the NK3 antagonist is selected from osanetant (SR-142,801), elinzanetant (NT-814), fezolinetant (ESN-364), talnetant (SB-223,412), pavinetant (AZD-4901/AZD- 2624), and SJX-653, or a pharmaceutically acceptable salt of each thereof.
[0031] In some embodiments, the NK3 antagonist is selected from osanetant (SR-142,801), elinzanetant (NT-814), fezolinetant (ESN-364), talnetant (SB-223,412), and pavinetant (AZD-4901/AZD- 2624), or a pharmaceutically acceptable salt of each thereof.
[0032] In some embodiments, the NK3 antagonist is administered to the patient once a day. In some embodiments, the NK3 antagonist is administered to the patient twice a day.
[0033] In some embodiments, the NK3 antagonist is osanetant, or a pharmaceutically acceptable salt thereof. In some embodiments, the osanetant, or a pharmaceutically acceptable salt thereof, is administered to the patient once or twice a day for a total daily dose ranging from about 10 mg to about 800 mg, about 25 to about 750 mg per day, about 50 to about 700 mg per day, about 50 to about 600 mg per day, about 50 to about 500 mg per day, about 50 to about 400 mg per day, about 75 to about 650 mg per day, about 70 to about 500 mg per day, about 75 to about 400 mg per day, about 100 to about 600 mg per day, about 100 to about 500 mg per day, or about 100 to about 400 mg per day.
[0034] In some embodiment the effective amount of osanetant, or a pharmaceutically acceptable salt thereof, is less than about 400 mg per day. In some embodiments, the effective amount of osanetant, or a pharmaceutically acceptable salt thereof, is from about 10 to about 350 mg per day. In some embodiments, the effective amount of osanetant, or a pharmaceutically acceptable salt thereof, is less than about 200 mg per day. In some embodiments, the effective amount of osanetant, or a pharmaceutically acceptable salt thereof, is from about 10 to about 150 mg per day. In some embodiments, the effective amount of osanetant, or a pharmaceutically acceptable salt thereof, is about 300 mg per day. In some embodiments, the osanetant is administered once a day. In some embodiments, the osanetant, or a pharmaceutically acceptable salt thereof, is administered twice a day, each dose being about 150 mg.
[0035] In some embodiments, the osanetant, or a pharmaceutically acceptable salt thereof, is administered to the patient twice a day for a total daily dose ranging from about 10 mg to about 800 mg, about 25 to about 750 mg per day, about 50 to about 700 mg per day, about 50 to about 600 mg per day, about 50 to about 500 mg per day, about 50 to about 400 mg per day, about 75 to about 650 mg per day, about 70 to about 500 mg per day, about 75 to about 400 mg per day, about 100 to about 600 mg per day, about 100 to about 500 mg per day, or about 100 to about 400 mg per day. In some embodiments, the osanetant, or a pharmaceutically acceptable salt thereof, is administered twice a day, each dose an equal fraction of the total daily dose and comprising from about 5 mg to about 400 mg of osanetant, or a pharmaceutically acceptable salt thereof.
[0036] For men suffering from prostate cancer, surgical resection of the prostate (prostatectomy), seminal vesicles, and neighboring lymph nodes, as well as androgen suppression or deprivation therapy (ADT) are common treatments for the disease. In situations of advanced prostate cancer, removal of the testes (orchiectomy), the primary source of endogenous testosterone in the male body, can be undertaken to further limit the growth and spread of the hormone-dependent prostate cancer.
[0037] For hormone-dependent cancers, such as prostate, and testicular cancers, the proliferation of cells is driven by hormone -receptor interactions on cell surfaces. In the presence of these sex hormones, namely estrogen, progesterone, and testosterone, the hormone-dependent cells replicate more frequently, increasing the opportunity for genetic errors to occur and accumulate, potentially leading to cancer. Pharmaceutical interventions, e.g. hormone deprivation therapy such as androgen deprivation therapy, for the treatment or prevention of hormone-dependent cancers include compounds that inhibit the synthesis of these sex hormones, such as gonadotropin-releasing hormone (GnRH) agonists and antagonists, or nonsteroidal antiandrogens (NS A As). In certain embodiments, the patient will be undergoing hormone deprivation therapy. For instance, in the case of prostate cancer, the patient may be undergoing androgen deprivation therapy (ADT). In some embodiments, the ADT comprises administration of a gonadotropin releasing hormone (GnRH) agonist, a GnRH antagonist, a histamine H2 receptor antagonist, a nonsteroidal antiandrogen (NSAA), or an androgen receptor antagonist. In some embodiments, the androgen deprivation therapy comprises surgical castration, luteinizing hormone -releasing hormone (LHRH) agonists (e.g. leuprolide, goserelin, triptorelin, or leuprolide mesylate), LHRH antagonists (e.g., degarelix, relugolix, elagolix), abiraterone, ketoconzale, etc.
[0038] Many patients opt for medical or surgical interventions to remove the cancer and surrounding tissues. Many patients also elect for the removal of sex hormone producing organs, namely the testes. In certain embodiments, the patient to be treated will be undergoing removal of testes. In certain embodiments, the patient will already be receiving a GnRH agonist e.g., leuprolide (also known as leuprorelin), or a GnRH antagonist, e.g. elagolix, linzagolix, relugolix, goserelin, triptorelin, buserelin, deslorelin, fertirelin, gonadorelin, histrelin, lecirelin, nafarelin, peforelin, triptorelin, abarelix, cetrorelix, degarelix, or ganirelix. In certain embodiments, the patient will already be receiving an androgen receptor antagonist (e.g., darolutamide).
[0039] In some embodiments, the patient is undergoing androgen deprivation therapy and the NK3 antagonist is administered in combination with the androgen deprivation therapy. In some embodiments, the androgen deprivation therapy is leuprolide, darulotamide, elagolix, linzagolix, or relugolix. In some embodiments, the androgen deprivation therapy is leuprolide, elagolix, linzagolix, relugolix, goserelin, triptorelin, buserelin, deslorelin, fertirelin, gonadorelin, histrelin, lecirelin, nafarelin, peforelin, triptorelin, abarelix, cetrorelix, degarelix, or ganirelix. In some embodiments, the hormone deprivation therapy is surgery e.g., orchiectomy.
[0040] In some embodiments, the patient is co-administered estrogen deprivation therapy. In some embodiments, gynemastia and breast pain is reduced. In some embodiments, the estrogen deprivation therapy is selected from a selective estrogen modulator (SERM) or a selective estrogen receptor degrader (SERD). In some embodiments, the estrogen deprivation therapy is tamoxifen. In some embodiments, the SERM is raloxifene, toremifene, arzoxifene, or lasofoxifene, broparestrol, clomifene (E, or Z), cyclofenil, ormeloxifene. ospemifene, acolbifene, afimoxifene (4-hydroxy tamoxifen), elacestrant, endoxifen (4-hydroxy-N-desmethyltamoxifen), clomiphene N-oxide, droloxifene (3-hydroxytamoxifen), etacstil, fispemifene, afimoxiphene, bazedoxifene, hydroxyetacstil, idoxifene (pyrrolidino-4- iodotamoxifen), levormeloxifene ((L)-ormeloxifene), miproxifene, nafoxidine, nitromifene (CI-628), NNC 45-0095, panomifene, pipendoxifene (ERA-923), trioxifene, or zindoxifene (D-16726). In some embodiments, the estrogen deprivation therapy is treatment with a selective estrogen receptor degrader (SERD). Examples of SERDs include and are not limited to
Figure imgf000010_0001
[0041] In some embodiments, the osanetant is administered for a short period prior to onset of the androgen deprivation therapy, e.g., prior to prostatectomy or orchiectomy or prior to start of treatment with ADT (e.g., leuprolide, elagolix, linzagolix, or relugolix). In some embodiments, the osanetant is administered for a period of 1 day to 1 month prior to onset of androgen deprivation therapy. In some embodiments, the osanetant is administered for a period of 1 day to 3 weeks prior to onset of androgen deprivation therapy. In some embodiments, the osanetant is administered for a period of 1 day to 2 weeks prior to onset of androgen deprivation therapy. In some embodiments, the osanetant is administered for a period of 1 day to 1 week prior to onset of androgen deprivation therapy. In some embodiments, the osanetant is administered for a period of 1 day to 5 days prior to onset of androgen deprivation therapy. In some embodiments, the osanetant is administered for a period of 1 day to 3 days prior to onset of androgen deprivation therapy. [0042] In some embodiments, the osanetant is administered prior to onset of androgen deprivation therapy, and during the androgen deprivation therapy. In some embodiments, the osanetant is administered after the androgen deprivation therapy (e.g., as maintenance therapy after cessation of primary tumor treatment (e.g., after prostatectomy or after orchiectomy).
[0043] In some embodiments, the patient is a male patient with high levels of testosterone.
[0044] In some embodiments, the patient is undergoing androgen deprivation therapy (ADT) and the
NK3 antagonist is administered in combination with the ADT. In some embodiments, the ADT is leuprolide, elagolix, linzagolix, relugolix, goserelin, triptorelin, buserelin, deslorelin, fertirelin, gonadorelin, histrelin, lecirelin, nafarelin, peforelin, triptorelin, abarelix, cetrorelix, degarelix, ganirelix, flutamide, nilutamide, bicalutamide, topilutamide, apalutamide, enzalutamide, darolutamide, cimetidine, proxalutamide, seviteronel, cioteronel, inocoterone acetate, RU-58841 (CAS No. 154992-24-2), dimethylcurcumin, AZ3514, arv-110 (CAS No. 2222112-77-6), MTX-23, ARD-61, UT-34 (2168525-92- 4), FL442, MK-4541 (CAS No. 796885-38-6), LY2452473 (Cas No. 1029692-15-6), GTx- 024/Enobosarm (CAS No. 841205-47-8), or GSK2881078 (CAS No. 1539314-06-1). In some embodiments, the ADT is elagolix, linzagolix, or relugolix. In some embodiments, the ADT is leuprolide. In some embodiments, the ADT is darolutamide. In some embodiments of the methods, the patient treated with an NK3 antagonist is not undergoing ADT. In some embodiments of the methods, the NK3 antagonist is administered in combination with radiation. In some embodiments of the methods, the NK3 antagonist is administered in combination with ADT and radiation.
[0045] In some embodiments of the methods, the levels of luteinizing hormone (LH) and/or testosterone (T) in the patient are each reduced by at least 70% compared to baseline levels of LH and T.
[0046] In some embodiments, the patient has undergone prostatectomy. In some embodiments, the patient has undergone prostatectomy and is undergoing ADT and/or radiation therapy.
[0047] In some embodiments, the methods described herein lower LH and/or testosterone levels in the patient and the reduction in levels of LH and/or T is reversible. In some embodiments, the methods described herein may reversibly lower LH, FSH, and estradiol levels.
[0048] In some embodiments, the patient is suffering from non-metastatic prostate cancer. In some of such embodiments, the patient is not undergoing ADT. In some of such embodiments, administration of NK3 antagonists is useful in maintaining low levels of testosterone thereby slowing or delaying progression of prostate cancer. [0049] In some embodiments, the NK3 antagonist is administered for a period of at least 8 days. In some embodiments, the NK3 antagonist is administered for a period of 8 to 28 days. In some embodiments, the NK3 antagonist is administered for a period of at least 28 days. In some embodiments, the NK3 antagonist is administered for a period of at least 8 days at a first dose and thereafter administration is continued for a further period of at least 8 days at a second dose which is the same as, or different from, the first dose.
[0050] In some embodiments, the androgen deprivation therapy is treatment with a gonadotropinreleasing hormone (GnRH) agonist or antagonist. In some of such embodiments, the patient is a male patient. In some embodiments, the GnRH agonist is leuprolide. In some embodiments, the GnRH antagonist is elagolix. In some embodiments, the GnRH antagonist is relugolix. In some embodiments, the GnRH antagonist is linzagolix.
[0051] In some embodiments, the cancer is hormone receptor-positive cancer. In some embodiments, the hormone receptor-positive cancer is prostate cancer.
[0052] In some embodiments, the neurokinin receptor antagonist is administered to a patient for a time period prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy as described herein. In some embodiments, the NK antagonist is administered concurrently with androgen deprivation therapy, a medical and/or surgical procedure and/or radiation therapy. In some embodiments, the patient continues to receive a NK antagonist after the androgen deprivation therapy, a medical and/or surgical procedure and/or radiation therapy. In some embodiments, the patient receives a NK antagonist after short-term (e.g., 1 to 6 months, 1 to 3 months, 1 month, 1-3 weeks, 1 week) androgen deprivation therapy. Any combination of these therapeutic regimens is contemplated within the scope of embodiments presented herein.
NK Antagonists
[0053] As discussed above, NK antagonists are useful in the methods described herein. As used herein, “NK receptor,” “neurokinin receptor,” or “tachykinin receptor” is a transmembrane G-protein coupled receptor. The three known tachykinin receptors are NK1, NK2, and NK3. These receptors act on a variety of human functions, which regulate numerous biological systems, including the reproductive system.
[0054] As used herein, “NK receptor antagonists,” “neurokinin receptor antagonists,” or “tachykinin receptor antagonists” are a class of drugs which interact with tachykinin receptors NK1, NK2, and NK3, and dampen the normal agonist-mediated biological responses. The tachykinin receptors have been associated with the transmission of stress signals and pain, the contraction of smooth muscles, inflammation, and modulating the hypothalamus-pituitary-gonadal axis. NK receptor antagonists are indicated for the treatment of migraine, emesis, gastrointestinal disorders, disorders of the reproductive system, and psychiatric disorders, including anxiety, addiction, depression, and schizophrenia. In certain embodiments, the NK antagonist is a NK1, NK2, or NK3 antagonist or a combination thereof.
[0055] In certain embodiments, the NK antagonist is a NK1 receptor antagonist selected from aprepitant, casopitant, ezlopitant, fosaprepitant, lanepitant, maropitant, rolapitant, vestipitant, L-733,060, L-741,671, L-742,694, RP-67580, RPR-100,893, CP-96345, CP-99994, GR-205,171, TAK-637, T-2328, and combinations thereof. In certain embodiments, the NK antagonist is a NK2 receptor antagonist selected from ibodutant, saredutant, GR-159,897, MEN-10376, and combinations thereof. In certain embodiments, the NK antagonist is a NK3 receptor antagonist selected from fezolinetant, osanetant, pavinetant, talnetant, SB-222,200 (structure below), SB-218,795 (structure below), NT-814 (structure below), and combinations thereof.
Figure imgf000013_0001
[0056] In certain embodiments, the NK antagonist is a NK3 antagonist. The NK3 receptor, and its associated tachykinin neuropeptide, neurokinin B, act on a variety of human functions, affecting the hypothalamus-pituitary-gonadal axis, which regulates numerous biological systems, including the reproductive system. In certain embodiments, the NK3 antagonist is osanetant. [0057] In certain embodiments, the neurokinin-3 receptor antagonist is selected from osanetant, fezolinetant, pavinetant, talnetant, SB-222,200, SB-218,795, and NT-814. In certain embodiments, the neurokinin-3 receptor antagonist is osanetant. In certain embodiments, the neurokinin-3 receptor antagonist is fezolinetant. In certain embodiments, the neurokinin-3 receptor antagonist is pavinetant. In certain embodiments, the neurokinin-3 receptor antagonist is talnetant. In certain embodiments, the neurokinin-3 receptor antagonist is SB-222,200. In certain embodiments, the neurokinin-3 receptor antagonist is SB-218,795. In certain embodiments, the neurokinin-3 receptor antagonist is NT-814. In certain embodiments, the NK3 antagonist is osanetant.
[0058] Osanetant was originally developed for the treatment of schizophrenia and other central nervous system disorders. In certain embodiments the NK antagonist is osanetant or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof. The chemical name of osanetant is (R)-N-(l-(3-(l-benzoyl-3-(3,4- dichlorophenyl)piperidin-3-yl)propyl)-4-phenylpiperidin-4-yl)-N-methylacetamide, and has the following structure:
Figure imgf000014_0001
[0059] Osanetant can also form pharmaceutically acceptable salts, such as osanetant hydrochloride, osanetant hydrobromide, osanetant sulfate, osanetant hydrogen sulfate, osanetant dihydrogen phosphate, osanetant methanesulfonate, osanetant methyl sulfate, osanetant maleate, osanetant fumarate, osanetant 2- naphthalenesulfonate, osanetant benzenesulfonate, osanetant glycolate, osanetant gluconate, and osanetant citrate, osanetant isethionate, osanetant p-toluenesulfonate, and the like. In some embodiments provided herein, osanetant is administered as a hydrochloride salt thereof.
[0060] Osanetant, as well as pharmaceutically acceptable salts thereof, can be purchased from commercial sources or can synthesized using published procedures. Patient Selection
[0061] In some embodiments, a patient selected for treatment using the methods described herein is a low risk patient, an intermediate risk patient, or a high risk patient.
[0062] In some embodiments, a patient selected for treatment using the methods described herein is a low risk patient. A low risk patient may be deemed to have “low risk prostate cancer”. Prostate cancer is described as “low risk” if it is only found in the prostate (localized) and it is highly likely to grow only very slowly, or not at all (low risk of progression). The medical criteria for low risk prostate cancer are: 1) the cancer is found in only one of the two sides (lobes) of the prostate; 2) the cancer takes up less than half of the affected prostate lobe; 3) the cancer cells have not mutated much and are not very aggressive; and/or 4) the cancer has not spread to any lymph nodes or led to the growth of tumors in other parts of the body.
[0063] “Low risk patient” is intended to encompass patients having low risk prostate cancer, e.g., a patient suffering from non-metastatic hormone responsive/sensitive cancer or a slow progression cancer. In some embodiments such a low risk patient does not elect to undergo androgen deprivation therapy. In some embodiments, a patient selected for treatment using the methods described herein is undergoing radiation therapy with or without radiation therapy. In some embodiments, a patient selected for treatment using methods described herein is being treated with lower doses of androgen deprivation therapy compared to the recommended dosing of androgen deprivation treatment. In some embodiments, a patient selected for treatment using the methods described herein is suffering from severe side effects of androgen deprivation therapy and is treated with lower doses of androgen deprivation therapy compared to the recommended dosing along with an NK receptor antagonist. Additionally, a “low risk patient” is also intended to include those patients who elect active surveillance over other therapies; who are older or have serious illnesses, where other therapies can be detrimental to their current conditions. Active surveillance can include regular PSA tests and/or biopsies.
[0064] In some embodiments, in a low risk patient, the tumor is confined to the prostate, and the PSA is <10 ng/mL or grade is group 1 (Gleason score is 6). In some embodiments, a low risk patient suffers from slow-growing tumors and may be a “very low risk” patient in which fewer than 3 biopsy tissue samples contain cancer cells and the cancer is not detectable by a digital rectal exam.
[0065] In some embodiments, a patient selected for treatment using the methods described herein is an intermediate risk patient where the tumor is confined to the prostate, the PSA is between 10 and 20 ng/mL, or the grade is group 2 or 3 (Gleason score of 7). In some of such embodiments, the patient may have a “favorable” or an “unfavorable” intermediate risk.
[0066] In some embodiments, a patient selected for treatment using the methods described herein is a high risk patient in whom the tumor extends outside the prostate, the PSA is >20 ng/mL, or grade group 4 or 5 (Gleason score 8 to 10). In some embodiments, a high risk patient has a subset of aggressive tumors and is a “very high risk” patient in whom the tumor has extended into the seminal vesicles (T3b) or the rectum or bladder (T4), or there are multiple biopsy samples with high grade cancer.
[0067] In some embodiments, a patient selected for treatment using the methods described herein is a patient who will benefit from castration or lowering of testosterone levels to castration levels.
[0068] In some embodiments, a patient selected for treatment using the methods described herein has been treated with higher than recommended doses or recommended doses of androgen deprivation therapy but has not had a lowering of testosterone to castration levels (e.g., lower than 20 ng/dL, or lower than 10 ng/dL). In some of such embodiments, the methods described herein provide for treatment with lower doses of the androgen deprivation therapeutic compared to the recommended dosing, and/or for a longer duration of time compared to the recommended duration, along with administration of an NK receptor antagonist, thereby reversibly lowering testosterone to castration levels in the patient. As used herein “recommended dosing” or “recommended duration” refers to dosing and/or durations approved by health agencies and/or provided on product labels.
[0069] In some embodiments, provided herein is a method to reduce testosterone to non-detectable levels comprising administration of a lower dose of an ADT and driving testosterone to below castrate levels, e.g., less than 20 ng/dL, less than 10 ng/dL. In some of such embodiments, osanetant is administered in combination with the ADT. In some of such embodiments, the reduction in testosterone occurs faster than the reduction if a recommended dose of ADT would have been administered alone. In some of such embodiments, the lowest testosterone level achieved is lower than the level that would be achieved if a recommended dose of ADT would have been administered alone.
[0070] In some embodiments, a patient selected for treatment using the methods described herein is not treated with NK3 antagonist MLE4901 (also known as AZD4901) at a dosage of 40 mg twice daily for 7 days. In some embodiments, a patient selected for treatment using the methods described herein is not treated with NK3 antagonist fezolinetant (ESN-364) at a dose of 3-180 mg as a single dose. In some embodiments, a patient selected for treatment using the methods described herein is not treated with NK3 antagonist fezolinetant (ESN-364) at a dose of 20, 60 or 180 mg once daily for 10 days. In some embodiments, a patient selected for treatment using the methods described herein is not treated with NK3 antagonist SJX-653 at a dose of 0.5 - 90 mg as a single dose.
Administration
[0071] In some embodiments, the time period over which the neurokinin receptor antagonist is administered to a patient prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 12 weeks or 8 weeks or 4 weeks to the day of the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy. In some embodiments, the time period over which the neurokinin receptor antagonist is administered to a patient prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 3 weeks to the day of the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy. In some embodiments, the time period over which the neurokinin receptor antagonist is administered to a patient prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 2 weeks to the day of the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy. In some embodiments, the time period over which the neurokinin receptor antagonist is administered prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 2 weeks. In some embodiments, the time period over which the neurokinin receptor antagonist is administered prior to the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 1 week.
[0072] In some embodiments, the neurokinin receptor antagonist is administered to a patient concurrently with the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy.
[0073] In some embodiments, the time period over which the neurokinin receptor antagonist is administered to a patient after the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 12 weeks. In some embodiments, the time period over which the neurokinin receptor antagonist is administered to a patient after the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 8 weeks. In some embodiments, the time period over which the neurokinin receptor antagonist is administered to a patient after the androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy is about 4 weeks.
[0074] In some embodiments, the patient is administered osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, for about 1 week prior to, concurrently with, and for about 12 weeks after, an androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy. In some embodiments, the patient is administered osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, for about 2 weeks prior to, concurrently with, and for about 12 weeks or more after, an androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy.
[0075] In some embodiments, the patient is administered osanetant or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, for about 1 week prior to, concurrently with, and for about 8 weeks after, an androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy. In some embodiments, the patient is administered osanetant for about 2 weeks prior to, concurrently with, and for about 8 weeks after, an androgen deprivation therapy, and/or medical or surgical procedure and/or radiation therapy.
[0076] In some embodiments, the neurokinin receptor antagonist is orally administered.
[0077] In some embodiments, the neurokinin receptor antagonist is subcutaneously administered.
[0078] In some embodiments, the neurokinin receptor antagonist is transdermally administered.
[0079] In some embodiments, the neurokinin receptor antagonist is intravenously administered.
[0080] In some embodiments, the neurokinin receptor antagonist is administered once daily. In some embodiments, the neurokinin receptor antagonist is administered as two, three, four or more sub-doses at appropriate intervals throughout the day, where the cumulative amount of the sub-doses equals the amount of the desired dose administered in a single day.
[0081] In some embodiments, when the NK antagonist is osanetant or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, the therapeutically effective amount of the osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 0.25 mg/day to about 1000 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, osanetant is about 0.5 mg/day to about 500 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 0.75 mg/day to about 450 mg/day, is about 1 mg/day to about 400 mg/day or 10 mg/day to about 350 mg/day. In some embodiments, the osanetant is administered is less than about 400 mg/day. In some embodiments, the osanetant is administered is less than about 200 mg/day or about 10 mg/day to about 150 mg/day.
[0082] In some embodiments, the therapeutically effective amount of the osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N- oxide or isomorphic crystalline form thereof, is about 1 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 50 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 100 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 200 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 300 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 400 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 500 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 600 mg/day. In some embodiments, the therapeutically effective amount of osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is about 800 mg/day. In some embodiments, the therapeutically effective amount of osanetant is about 1000 mg/day.
[0083] In certain embodiments, osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, is dosed at about 0.25 mg/day to about 1 mg/day, in order to treat bone loss while avoiding transient increases in liver transaminase (alanine aminotransferase) concentrations, and maintaining healthy liver function.
[0084] In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose ranging from about 5 mg per day to about 600 mg per day. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose ranging from about 25 mg per day to about 500 mg per day. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose ranging from about 50 mg per day to about 500 mg per day. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose ranging from about 100 mg per day to about 400 mg per day. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose ranging from about 150 mg per day to about 300 mg per day. In any of these embodiments, the total daily dose may be administered as a single dose, or split between multiple doses (e.g., daily dose of 300 mg QD or daily dose of 300 mg split into 150 mg BID, or daily dose of 300 mg split into 100 mg TID). In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose of 300 mg, split into two doses of 150 mg each, i.e., 150 mg BID. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose of 200 mg, split into two doses of 100 mg each, i.e., 100 mg BID. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose of 100 mg, split into two doses of 50 mg each, i.e., 50 mg BID. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose of 50 mg, split into two doses of 25 mg each, i.e., 25 mg BID. In some embodiments, osanetant is administered prior to, and/or concurrently with, and/or after administration of leuprolide, elagolix, relugolix, or linzagolix or any other ADT in a daily dose of 25 mg, split into two doses of 12.5 mg each, i.e., 12.5 mg BID. In any of such embodiments, the dose of leuprolide, elagolix, relugolix, or linzagolix or any other ADT may be lower than the recommended dosing and may be for a longer duration of time compared to the recommended duration of therapy. Pharmaceutical Compositions
[0085] Also provided herein, in some embodiments, are pharmaceutical compositions that comprise osanetant, or a stereoisomer, mixture of stereoisomers, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof, and one or more pharmaceutically acceptable vehicles selected from carrier, adjuvants, and excipients.
[0086] Suitable pharmaceutically acceptable vehicles may include, for example, inert solid diluents and fillers, diluents, including sterile aqueous solutions and various organic solvents, permeation enhancers, solubilizers, and adjuvants. Such compositions are prepared in a manner well known in the pharmaceutical art. See, e.g., Remington’s Pharmaceutical Sciences, Mace Publishing Co., Philadelphia, Pa. 17th Ed. (1985); and Modern Pharmaceutics, Marcel Dekker, Inc. 3rd Ed. (G.S. Banker & C.T. Rhodes, Eds.).
[0087] The pharmaceutical compositions may be administered in either single or multiple doses. The pharmaceutical composition may be administered by various methods including, for example, rectal, intravenous, subcutaneous, and transdermal routes. In certain embodiments, the pharmaceutical composition may be administered by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.
[0088] One mode for administration is parenteral, for example, by injection. The forms in which the pharmaceutical compositions described herein may be incorporated for administration by injection include, for example, aqueous or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles.
[0089] The pharmaceutical composition may be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be sterile injectable solution or suspension in a non-toxic parentally acceptable vehicle, for example as a solution in 1,3-butanediol. Among the acceptable vehicles that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid can be useful in the preparation of injectables. Such solutions may be formulated as 0.01% -10% isotonic solutions, pH 5-7, with appropriate salts. [0090] The compound described herein may be administered parenterally in a sterile medium. Parenteral administration includes subcutaneous injections, intravenous, intramuscular, intrathecal injection or infusion techniques. The compound described herein, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle. Advantageously, adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle. In many pharmaceutical compositions for parenteral administration the carrier comprises at least 90% by weight of the total composition. In some embodiments, the carrier for parenteral administration is chosen from propylene glycol, ethyl oleate, pyrrolidone, ethanol, and sesame oil.
[0091] A pharmaceutical composition, for example, for injection, may comprise a cyclodextrin. The cyclodextrin may be, for example, a hydroxypropyl cyclodextrin or a sulfobutylether cyclodextrin. The cyclodextrin may be, for example, an a-cyclodextrin, a P-cyclodextrin, or a y-cyclodextrin.
[0092] A compound described herein may also be administered via microspheres, liposomes, other microparticulate delivery systems or sustained release formulations placed in certain tissues including blood. Suitable examples of sustained release carriers include semi-permeable polymer matrices in the form of shared articles, e.g., suppositories or microcapsules. Examples can be found, e.g., in Remington's Pharmaceutical Sciences, 18th edition, Gennaro, A. R., Lippincott Williams & Wilkins; 20th edition (Dec. 15, 2000) ISBN 0-912734-04-3 and Pharmaceutical Dosage Forms and Drug Delivery Systems; Ansel, N. C. et al. 7th Edition ISBN 0-683305-72-7, the entire disclosures of which are herein incorporated by reference.
[0093] Oral administration may be another route for administration of the compounds described herein. Administration may be via, for example, capsule or enteric coated tablets. In making the pharmaceutical compositions that include at least one compound described herein, the active ingredient is usually diluted by an excipient and/or enclosed within such a carrier that can be in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be in the form of a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, sterile injectable solutions, and sterile packaged powders.
[0094] Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose. The formulations can additionally include lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl and propylhydroxybenzoates; sweetening agents; and flavoring agents.
[0095] The compositions that include at least one compound described herein can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the subject by employing procedures known in the art. Controlled release drug delivery systems for oral administration include osmotic pump systems and dissolutional systems containing polymer-coated reservoirs or drug-polymer matrix formulations. Examples of controlled release systems are given in U.S. Patent Nos. 3,845,770; 4,326,525; 4,902,514; and 5,616,345. Another formulation for use in the methods disclosed herein employ transdermal delivery devices (“patches”). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds described herein in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Patent Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
[0096] For preparing solid compositions such as tablets, the principal active ingredient may be mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound described herein. When referring to these preformulation compositions as homogeneous, the active ingredient may be dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
[0097] The tablets or pills of the compounds described herein may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action, or to protect from the acid conditions of the stomach. For example, the tablet or pill can include an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer that serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
[0098] The compound described herein can be incorporated into oral liquid preparations such as aqueous or oily suspensions, solutions, emulsions, syrups, or elixirs, for example. Furthermore, pharmaceutical compositions containing the compound described herein can be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations can contain conventional additives, such as suspending agents (e.g., sorbitol syrup, methyl cellulose, glucose/sugar, syrup, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminum stearate gel, and hydrogenated edible fats), emulsifying agents (e.g., lecithin, sorbitan monooleate, or acacia), non-aqueous vehicles, which can include edible oils (e.g., almond oil, fractionated coconut oil, silyl esters, propylene glycol and ethyl alcohol), and preservatives (e.g., methyl or propyl p-hydroxybenzoate and sorbic acid).
[0099] Compositions for inhalation or insufflation may include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described herein. In some embodiments, the compositions are administered by the oral respiratory route for local or systemic effect. In other embodiments, compositions in pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a facemask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, orally from devices that deliver the formulation in an appropriate manner.
[0100] The forms in which the pharmaceutical compositions described herein may be administered, for example, as quick-dissolving tablets, lozenges, powders, sprays, ointments, gels, or liquid suspensions.
[0101] Some examples of suitable mucoadhesive agents include agarose, chitosan, trimethylated chitosan, chitosan-EDTA, gelatin, hyaluronic acid, guar gum, hakea gum, xanthan gum, gellan gum, carrageenan, pectin, sodium alginate, cellulose derivatives, CMC, thiolated CMC, sodium CMC, HEC, HPC, HPMC, MC, poly(acrylic acid)-based polymers, CP, PC, PAA, copolymers of acrylic acid and PEG, PVA, PVP, CP, aminodextran, dimethylaminoethyl-dextran, hydroxyethyl starch, poly(ethylene oxide), scleroglucan, cyanoacrylate, hydroxylated methacrylate, and poly(methacrylic acid). Some examples of suitable penetration enhancers include sodium lauryl sulfate, cetyl pyridinium chloride, Poloxamer, Brij, Span, Myrj, Tween, sodium glycocholate, sodium tauro deoxycholate, sodium tauro cholate, oleic acid, caprylic acid, lauric acid, lyso phosphatidyl choline, phosphatidyl choline, a-, P-, and y-cyclodextrin, methylated P-cyclodextrin, EDTA, citric acid, sodium salicylate, methoxy salicylate, chitosan, trimethyl chitosan, poly-L-arginine, and L-lysine. Some examples of suitable enzyme inhibitors include aprotinin, bestatin, and puromycin. Combination Therapy
[0102] The neurokinin receptor antagonist may be administered in combination with one or more additional active agents. In some embodiments, the neurokinin receptor antagonist may be administered in combination with a second active agent that the patient is being treated with. In some embodiments, the second active agent is an androgen deprivation agent such as a GnRH agonist, e.g. leuprolide. In some embodiments, the second active agent is an androgen deprivation agent such as a GnRH antagonist, e.g. elagolix, linzagolix, relugolix, goserelin, triptorelin, buserelin, deslorelin, fertirelin, gonadorelin, histrelin, lecirelin, nafarelin, peforelin, triptorelin, abarelix, cetrorelix, degarelix, or ganirelix. In some embodiments, the second active agent is an androgen deprivation agent such as a nonsteroidal antiandrogen (NSAA), including, but not limited to, flutamide, nilutamide, bicalutamide, topilutamide, apalutamide, enzalutamide, darolutamide, cimetidine, proxalutamide, seviteronel, cioteronel, inocoterone acetate, and RU-58841. In some embodiments, the second active agent is an androgen deprivation agent is a selective androgen receptor degrader (SARD), including, but not limited to, dimethylcurcumin, AZ3514, arv-110, MTX-23, ARD-61, and UT-34.
[0103] In some embodiments, the second active agent is an androgen deprivation agent is a selective androgen receptor modulator (SARM), including, but not limited to, FL442, MK-4541, LY2452473, GTx-024/Enobosarm, and GSK2881078.
[0104] As used herein, “gonadotropin-releasing hormone agonists and antagonists” or “GnRH agonists and antagonists” are classes of drugs which prevent the GnRH-mediated release of sex hormones. GnRH is a peptide hormone produced by GnRH neurons in the hypothalamus, responsible for the release of follicle-stimulating hormone and luteinizing hormone from the pituitary gland, beginning the hypothalamic -pituitary-gonadal axis synthesis and release of sex hormones.
[0105] Also provided is a pharmaceutical composition comprising osanetant and a second active agent.
EXAMPLES
[0106] It is understood that modifications which do not substantially affect the activity of the various embodiments of this disclosure are also included within the definition of the disclosure provided herein. Accordingly, the following examples are intended to illustrate but not limit the present disclosure.
Example 1
[0107] This study will evaluate the use of osanetant to reduce testosterone in men with adenocarcinoma of the prostate. [0108] The study is a single-arm pilot study. This study involves 10 patients with prostate cancer. All 10 will undergo prostatectomy and measurement of 4 hormones FSH, LH, Estradiol, and Testosterone) at baseline and days 2, 3, 7, 14, and 28.
[0109] Protocol synopsis: Men will undergo baseline screening (serum PSA and testosterone and history and physical examination). Those men who met inclusion criteria and elect to participate will be scheduled for the study protocol, which begins 35 days prior to scheduled surgery. Osanetant will be administered by the investigational pharmacy at a dosing of 200 mg BID. On days 2, 3, 7, and 14, and 28, an AM (0800 +/- 30 min) testosterone, LH, FSH, and Estradiol levels will be obtained. On day 35 (1 week after completing protocol therapy), the patient will undergo standard of care surgery. On day 42 post-operatively (+/- 3 days) serum testosterone, LH, FSH, Estradiol, and PSA will be obtained.
[0110] Patient Sample: Adult men with clinically localized prostate cancer (cTl-3bN0M0) who are scheduled for radical prostatectomy.
[0111] Inclusion criteria: Age >18; Clinically localized disease: cTl-3bN0M0; Testosterone >150 ng/ml
[0112] Exclusion criteria: Current or recent (within 6 months) use of testosterone/estrogen modulating agents (leuprolide, degarelix, bicalutamide, enzalutamide, apalutamide, darolutamide, abiraterone, systemic ketoconazole, tamoxifen, etc); Current use of CYP3A4 inhibitors; Cognitive impairment (defined as the presence of diagnosed dementia); Impaired renal function: Cr >1.8; Medical history of osteoporosis; Current systemic corticosteroid, long-term opioid, spironolactone, or eplerenone use.
[0113] Endpoints: Testosterone levels during treatment; LH, FSH, and Estradiol levels during treatment; PSA response to Osanetant; Rate of development of castrate testosterone levels (defined as testosterone <50 ng/mL); Reversibility of hormonal changes.
[0114] Patients will serve as their own comparator and hormonal labs will be compared at all timepoints, as will the trajectory of hormone levels over the study period. PSA response will be evaluated using percent reduction in PSA compared to baseline will be used to quantify response. The proportion of patients achieving castrate testosterone levels during the study period will be evaluated.
* * * [0115] It is to be understood that while the disclosure has been described in conjunction with the above embodiments, that the foregoing description and examples are intended to illustrate and not limit the scope of the disclosure. Other aspects, advantages and modifications within the scope of the disclosure will be apparent to those skilled in the art to which the disclosure pertains.

Claims

What is claimed is:
1. A method for treating prostate cancer or delaying progression of prostate cancer in a patient comprising administering a Neurokinin 3 receptor (NK3) antagonist to the patient in need thereof.
2. The method of claim 1, wherein the prostate cancer is a hormone responsive prostate cancer.
3. The method of claim 1, wherein the NK3 antagonist is administered in an amount to reversibly lower the patient’s serum testosterone to lower than or equal to about 20 ng/dL.
4. The method of claim 1 , wherein the NK3 antagonist is administered in an amount to reversibly lower the patient’s serum testosterone to lower than or equal to about 10 ng/dL.
5. The method of claim 1, wherein the NK3 antagonist is a dual NK1/NK3 antagonist.
6. The method of claim 1, wherein the NK3 antagonist is a selective NK3 antagonist.
7. The method of any one of claims 1-6, wherein the NK3 antagonist is selected from osanetant
(SR-142,801), elinzanetant (NT-814), fezolinetant (ESN-364), talnetant (SB-223,412), pavinetant (AZD- 4901 / AZD-2624 / MLE4901), and SJX-653, or a pharmaceutically acceptable salt of each thereof.
8. The method of any one of claims 1-7, wherein the NK3 antagonist is selected from osanetant (SR-142,801), elinzanetant (NT-814), fezolinetant (ESN-364), talnetant (SB-223,412), and pavinetant (AZD-4901/AZD-2624), or a pharmaceutically acceptable salt of each thereof.
9. The method of any one of claims 1-8, wherein the NK3 antagonist is administered to the patient once a day.
10. The method of any one of claims 1-8, wherein the NK3 antagonist is administered to the patient twice a day.
11. The method of any preceding claim, wherein the NK3 antagonist is osanetant, or a pharmaceutically acceptable salt thereof.
12. The method of claim 11, wherein the osanetant, or a pharmaceutically acceptable salt thereof, is administered to the patient twice a day for a total daily dose ranging from about 50 mg to about 600 mg.
13. The method of claim 11, wherein the osanetant, or a pharmaceutically acceptable salt thereof, is administered to the patient twice a day for a total daily dose ranging from about 100 mg to about 400 mg.
14. The method of claim 11, wherein the osanetant, or a pharmaceutically acceptable salt thereof, is administered twice a day, each dose comprising from about 25 mg to about 300 mg of osanetant, or a pharmaceutically acceptable salt thereof.
15. The method of any preceding claim, wherein the patient is a low risk patient, an intermediate risk patient, or a high risk patient.
16. The method of any preceding claim, wherein the patient is not undergoing ADT.
17. The method of any preceding claim, wherein the patient is undergoing androgen deprivation therapy (ADT) and the NK3 antagonist is administered in combination with the ADT.
18. The method of claim 17 wherein the ADT comprises administration of a gonadotropin releasing hormone (GnRH) agonist, a GnRH antagonist, a histamine H2 receptor antagonist, a non-steroidal antiandrogen (NSAA), or an androgen receptor antagonist.
19. The method of claim 18, wherein the ADT is leuprolide, elagolix, linzagolix, relugolix, goserelin, triptorelin, buserelin, deslorelin, fertirelin, gonadorelin, histrelin, lecirelin, nafarelin, peforelin, triptorelin, abarelix, cetrorelix, degarelix, ganirelix, flutamide, nilutamide, bicalutamide, topilutamide, apalutamide, enzalutamide, darolutamide, cimetidine, proxalutamide, seviteronel, cioteronel, inocoterone acetate, RU-58841, dimethylcurcumin, AZ3514, arv-110, MTX-23, ARD-61, UT-34, FL442, MK-4541, LY2452473, GTx-024/Enobosarm, or GSK2881078.
20. The method of any preceding claim, wherein the NK3 antagonist is administered in combination with radiation treatment.
21. The method of any preceding claim, wherein the levels of lutenizing hormone (LH) and/or testosterone (T) in the patient are each reduced by at least 70% compared to baseline levels of LH and T.
22. The method of any preceding claim, wherein the patient has undergone prostatectomy.
23. The method of any preceding claim, wherein the reduction in levels of LH and/or T is reversible.
24. The method of any preceding claim, wherein the patient is suffering from non-metastatic prostate cancer.
25. The method of any preceding claim, wherein the NK3 antagonist is administered for a period of at least 8 days.
26. The method of any preceding claim, wherein the NK3 antagonist is administered for a period of 8 to 28 days.
27. The method of any preceding claim, wherein the NK3 antagonist is administered for a period of at least 28 days.
28. The method of any preceding claim, wherein the NK3 antagonist is administered for a period of at least 8 days at a first dose and thereafter administration is continued for a further period of at least 8 days at a second dose which is the same as, or different from, the first dose.
PCT/US2023/011892 2022-01-31 2023-01-30 Nk receptor antagonists for treatment of prostate cancer WO2023147135A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263305209P 2022-01-31 2022-01-31
US63/305,209 2022-01-31
US202263306215P 2022-02-03 2022-02-03
US63/306,215 2022-02-03

Publications (1)

Publication Number Publication Date
WO2023147135A1 true WO2023147135A1 (en) 2023-08-03

Family

ID=85382936

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/011892 WO2023147135A1 (en) 2022-01-31 2023-01-30 Nk receptor antagonists for treatment of prostate cancer

Country Status (1)

Country Link
WO (1) WO2023147135A1 (en)

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US4326525A (en) 1980-10-14 1982-04-27 Alza Corporation Osmotic device that improves delivery properties of agent in situ
US4902514A (en) 1988-07-21 1990-02-20 Alza Corporation Dosage form for administering nilvadipine for treating cardiovascular symptoms
US4992445A (en) 1987-06-12 1991-02-12 American Cyanamid Co. Transdermal delivery of pharmaceuticals
US5001139A (en) 1987-06-12 1991-03-19 American Cyanamid Company Enchancers for the transdermal flux of nivadipine
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US5616345A (en) 1983-12-22 1997-04-01 Elan Corporation Plc Controlled absorption diltiazen formulation for once-daily administration
US20030092602A1 (en) * 2001-05-08 2003-05-15 Schering Corporation Use of neurokinin receptor antagonists to treat androgen-dependent diseases
EP2604276A1 (en) * 2010-08-12 2013-06-19 Servicio Andaluz De Salud Use of antibodies against nk1, nk2 and/or nk3 receptors in cancer treatment
US20160339037A1 (en) * 2015-05-18 2016-11-24 Nerre Therapeutics Limited Novel Uses
US20200270254A1 (en) * 2017-07-12 2020-08-27 Ogeda Sa Deuterated fezolinetant
WO2023278843A2 (en) * 2021-07-02 2023-01-05 Acer Therapeutics, Inc. Solid forms of osanetant

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US4326525A (en) 1980-10-14 1982-04-27 Alza Corporation Osmotic device that improves delivery properties of agent in situ
US5616345A (en) 1983-12-22 1997-04-01 Elan Corporation Plc Controlled absorption diltiazen formulation for once-daily administration
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US4992445A (en) 1987-06-12 1991-02-12 American Cyanamid Co. Transdermal delivery of pharmaceuticals
US5001139A (en) 1987-06-12 1991-03-19 American Cyanamid Company Enchancers for the transdermal flux of nivadipine
US4902514A (en) 1988-07-21 1990-02-20 Alza Corporation Dosage form for administering nilvadipine for treating cardiovascular symptoms
US20030092602A1 (en) * 2001-05-08 2003-05-15 Schering Corporation Use of neurokinin receptor antagonists to treat androgen-dependent diseases
EP2604276A1 (en) * 2010-08-12 2013-06-19 Servicio Andaluz De Salud Use of antibodies against nk1, nk2 and/or nk3 receptors in cancer treatment
US20160339037A1 (en) * 2015-05-18 2016-11-24 Nerre Therapeutics Limited Novel Uses
US20200270254A1 (en) * 2017-07-12 2020-08-27 Ogeda Sa Deuterated fezolinetant
WO2023278843A2 (en) * 2021-07-02 2023-01-05 Acer Therapeutics, Inc. Solid forms of osanetant

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, PERGAMON PRESS
ANONYMOUS: "History of Changes for Study: NCT05607342 Pilot of Osanetant to Reduce Testosterone in Men With Adenocarcinoma of the Prostate (PORT-MAP)", 31 October 2022 (2022-10-31), XP093041714, Retrieved from the Internet <URL:https://clinicaltrials.gov/ct2/history/NCT05607342?A=1&B=1&C=merged#StudyPageTop> [retrieved on 20230424] *
ANSEL, N. C. ET AL.: "Pharmaceutical Dosage Forms and Drug Delivery Systems", MARCEL DEKKER, INC.
CAS , no. 1029692-15-6
CAS , no. 1539314-06-1
GENNARO, A. R.: "Remington's Pharmaceutical Sciences", 15 December 2000, LIPPINCOTT WILLIAMS & WILKINS
PARI MALHERBE+ ET AL: "Tachykinin neurokinin 3 receptor antagonists: a patent review (2005 -- 2010)", EXPERT OPINION ON THERAPEUTIC PATENTS, TAYLOR & FRANCIS, GB, vol. 21, no. 5, 1 January 2011 (2011-01-01), pages 637 - 655, XP007922679, ISSN: 1354-3776, DOI: 10.1517/13543776.2011.568482 *
T. HIGUCHIV. STELLA: "A.C.S. Symposium Series", vol. 14, article "Pro-drugs as Novel Delivery Systems"

Similar Documents

Publication Publication Date Title
US10004731B2 (en) Flibanserin for the treatment of urinary incontinence and related diseases
US6362202B1 (en) Methods and compositions using (−) norcisapride in combination with proton pump inhibitors or H2 receptor antagonists
US6552045B2 (en) Methods and compositions using (+) norcisapride in combination with proton pump inhibitors or H2 receptor antagonists
WO2022026901A2 (en) Nk antagonists for contraception
JP2021059579A5 (en)
IL255261B1 (en) A combination of an m-tor inhibitor and rad19010 for use in a method of inhibiting tumor growth or producing tumor regression in a subject having a deug-resistant andqor mutant estrogen receptor
HU221589B (en) Combinative pharmaceutical composition for treating mammal and uterial carcinomas and process for preparing of the same
JP4969016B2 (en) Combination of antiestrogens and farnesyl protein transferase inhibitors
US20230277518A1 (en) Nk receptor antagonists for cancer patients
JP2017522389A (en) Treatment of hot flush and bone loss caused by androgen deprivation therapy with cis-clomiphene
WO2013119910A1 (en) Combination of muscarinic receptor antagonists and beta- 3 adrenoceptor agonists for treating overactive bladder
WO2023147135A1 (en) Nk receptor antagonists for treatment of prostate cancer
US10369159B2 (en) Estrogen receptor β partial agonist having estrogen receptor α inhibitory effect, and gynecological disease therapeutic agent using same
JP2004501967A (en) Pharmaceutical combination of anti-androgen and tamoxifen to provide anti-androgen action and aromatase inhibition
JP2008513469A (en) Treatment of osteoporosis caused by androgen removal
EP3709997A1 (en) Pharmaceutical combination comprising lsz102 and ribociclib
JPWO2009013846A1 (en) Pharmaceutical composition for ameliorating lower urinary tract symptoms associated with prostatic hypertrophy
WO2023147134A2 (en) Methods for treating patients having low estrogen with nk3 antagonists
US20090317464A1 (en) Methods and compositions for producing anti-androgenic effects
WO2020205608A1 (en) Uses of androgen receptor antagonists and jnk pathway inhibitors, and pharmaceutical compositions related thereto
JP2009067737A (en) Therapeutic and/or prophylactic agent for lower urinary tract symptom after radiation therapy

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23707562

Country of ref document: EP

Kind code of ref document: A1