SOLID ORAL DOSAGE FORMS OF 2R,6R-HYDROXYNORKETAMINE OR DERIVATIVES THEREOF

Information

  • Patent Application
  • 20190380978
  • Publication Number
    20190380978
  • Date Filed
    June 05, 2017
    7 years ago
  • Date Published
    December 19, 2019
    4 years ago
Abstract
This invention relates to solid oral dosage forms of 2R6R-hydroxynorketamine or prodrugs thereof having Formula Ib, including any pharmaceutically acceptable salt of the foregoing, for use in a therapeutic method for the treatment of a depressive disorder in a patient.
Description
FIELD OF THE INVENTION

This invention relates to solid oral dosage forms of metabolites of ketamine and prodrugs of metabolites of ketamine and analogues thereof, including any pharmaceutically acceptable salt of the foregoing, for use in a therapeutic method for the treatment of a depressive disorder in a patient. In particular the invention relates to solid oral dosage forms comprising the ketamine metabolite 2R,6R-hydroxynorketamine or prodrugs thereof for use in a therapeutic method for the treatment of a depressive disorder in a patient.


BACKGROUND TO THE INVENTION

Depressive disorders are a category of mood disorders that cause a persistent feeling of sadness and loss of interest in the sufferer. Symptoms of depressive disorders include feelings of helplessness and hopelessness, loss of interest in daily activities, appetite or weight changes, sleep changes, anger or irritability, loss of energy, self-loathing, reckless behaviour, concentration problems, and unexplained aches and pains.


Depressive disorders are generally treated with psychotherapy, medication, or both.


Psychotherapy can help people with depressive disorders. Effective psychotherapies include Cognitive Behavioural Therapy (CBT), Self-Help or Support Groups, and Stress-Management Techniques.


CBT is a type of psychotherapy that can help patients with depressive disorders. It teaches a patient different ways of thinking, behaving, and helps the patient exercise control on their mood. CBT can also help patients learn and practice social skills which can assist with depressive disorders.


CBT may be conducted individually or with a group of patients who have similar problems.


Group therapy is particularly effective for social depressive disorder. Often “homework” is assigned for participants to complete between sessions.


Meditation can help patients with depressive disorders calm themselves and may enhance the effects of therapy.


In some cases, medications are used as the initial treatment of a depressive disorder; more commonly medications are used if there is insufficient response to a course of psychotherapy.


The most common classes of medications used to combat depressive disorders, also referred to as antidepressant agents, are serotonin modulators.


Serotonin modulators can be effective in treating depressive disorders, but they take several weeks to start working and may cause side effects such as headache, nausea, or difficulty sleeping. In order to minimize the impact of serotonin modulator side effects, typical dosing regimens start a patient on a low dose of the medication and increase the dose slowly over time.


There is a need in the art to find new and improved therapies for the treatment of depressive disorders. The present invention provides novel therapies for the treatment of depressive disorders. These therapies have the potential to provide greater efficacy than current treatment regimens, achieve greater compliance and/or improve outcomes in patients without introducing unacceptable side-effect liability.


SUMMARY OF THE INVENTION

A first aspect of the present invention provides a solid oral dosage form comprising a compound selected from a metabolite of ketamine and a prodrug of a metabolite of ketamine, or a pharmaceutically acceptable salt thereof, for use in the treatment of a depressive disorder in a patient.


Intravenous infusions of 0.5 mg/kg of ketamine hydrochloride administered over a period of 40 mins are known to exhibit potent and rapid anti-depressant effects in treatment-resistant depression patients, reported, for example, in Murrough et al; Anti-depressant Efficacy of Ketamine in Treatment-Resistant Major Depression: A Two-Site Randomized Controlled Trial: Am J Pschiatry. 1 Oct. 2013: 170(10): 1134-1142.


The use of ketamine hydrochloride in the treatment of mental health is limited by the dosage form in which it is typically provided. Ketamine hydrochloride is approved in a form administrable only by intravenous infusion. This renders it inconvenient and/or inappropriate for use in depressed patients: it is preferable to be able to administer therapy in an out-patient setting. Accordingly, the present invention provides a solid oral dosage form comprising a compound selected from a metabolite of ketamine, and a prodrug of a metabolite of ketamine, or a pharmaceutically acceptable salt thereof, for use in the treatment of a depressive disorder in a patient, wherein the solid oral dosage form is suitable for use as a first-line therapy.


In preferred embodiments of the first aspect of the present invention, the metabolite of ketamine is a mono-hydroxylated metabolite, preferably one selected from the metabolites listed in Table 5.


In preferred embodiments of the first aspect of the present invention, the metabolite of ketamine is in the form of an acid-addition salt.


In preferred embodiments of the first aspect of the present invention the solid oral dosage form comprises 2R,6R-hydroxynorketamine or a prodrug thereof, or a pharmaceutically acceptable salt of 2R,6R-hydroxynorketamine or a prodrug thereof.


In preferred embodiments of the first aspect of the present invention, the solid oral dosage form comprises 2R,6R-hydroxynorketamine in the form of an acid-addition salt.


In the first aspect of the present invention, the pharmaceutically acceptable salt of 2R,6R-hydroxynorketamine is preferably not 2R,6R-hydroxynorketamine hydrochloride.


In preferred embodiments the first aspect of the present invention the solid oral dosage form comprises the active ingredient 2R,6R-hydroxynorketamine, or a prodrug thereof, or pharmaceutically acceptable salts thereof, wherein the active ingredient is present in an amount equivalent to between 10 mg and 100 mg of 2R,6R-hydroxynorketamine freebase.


In preferred embodiments of the first aspect of the invention, the metabolite of ketamine, or a prodrug thereof, or pharmaceutically acceptable salts thereof, are present in crystalline form.


In preferred embodiments of the first aspect of the present invention, the solid oral dosage form is selected from a tablet and a capsule.


In a second aspect of the present invention, the solid oral dosage forms of the first aspect comprise a blend of one or more diluent. Preferably the diluent blend comprises one or more, and preferably two or more, diluents selected from anhydrous lactose, D-mannitol, dicalcium phosphate, calcium carbonate, magnesium oxide, magnesium carbonate, glucose, sorbitol, sucrose, calcium sulphate, starch, dextrates, kaolinite, maltodextrin and lactitol. More preferred diluents are selected from microcrystalline cellulose, dicalcium phosphate, kaolinite, starch and calcium carbonate.


In preferred embodiments of the second aspect the solid oral dosage form is a tablet and comprises a diluent blend comprising microcrystalline cellulose. In further embodiments of the second aspect, the diluent blend comprises two or more diluents wherein one diluent is selected from microcrystalline cellulose and starch, and wherein a further diluent is an inorganic diluent.


In further embodiments of the second aspect, the diluent blend comprises dicalcium phosphate and microcrystalline cellulose.


In a preferred embodiment of the second aspect, the diluent blend excludes lactose monohydrate.


Preferred embodiments of the second aspect are wherein the solid oral dosage form is a capsule wherein the capsule shell comprises a constituent selected from gelatin and hydroxypropyl methylcellulose.


A third aspect of the present invention provides solid oral dosage forms according to either the first aspect or the second aspect, wherein the solid oral dosage form is for use in combination with a serotonin modulator. In preferred embodiments of the third aspect, the serotonin modulator is a selective serotonin reuptake inhibitor. Preferred serotonin modulators for use in the third aspect of the present invention are selected from vortioxetine, etoperidone, lorpiprazole, lubazodone, mepiprazole, nefazodone, trazodone, citalopram, escitalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, dapoxetine, venlafaxine, milnacipran, duloxetine, levomilnacipran, desvenlafaxine, sibutramine, aptazapine, esmirtazapine, mianserin, mirtazapine, and setiptiline. Particularly preferred serotonin modulators for use in the third aspect of the present invention are selected from citalopram, escitalopram, fluoxetine, fluvoxamine, paroxetine, sertraline and dapoxetine.


A fourth aspect of the present invention provides solid oral dosage forms according to the first, second or third aspect of the invention, in particular the third aspect, wherein the patient has experienced one or more manic episode or hypomanic episode, and/or is suffering or is at risk of suffering one or more manic episode or hypomanic episode.


In preferred embodiments of the first, second, third or fourth aspect of the present invention, and in particular the fourth aspect, said patient is suffering from a condition selected from major depression, bipolar disorder Type I, bipolar disorder Type II, bipolar depression, postpartum depression, dementia-related depression, obsessive-compulsive disorder (OCD) with co-morbid depression, and post-traumatic stress disorder (PTSD) with co-morbid depression, tics with comorbid depression, and social anxiety with comorbid depression.


A fifth aspect of the present invention provides a prodrug of a metabolite of ketamine, or analogue thereof, having the structure of Formula I, or a pharmaceutically acceptable salt thereof:




embedded image


wherein OR1 is bonded to the cyclohexanone ring at either the 3, 4, 5 or 6 position, wherein X is one, two, or three haloatoms each independently positioned ortho, meta or para (to the bond attaching the phenyl to the cyclohexanone ring), wherein each X independently selected from F, Cl, Br, and I, and wherein either:

    • (i) R1 is H, R3 is selected from H and C1-C4 alkyl, and R2 is selected from CH2OPO3H2, CH2OSO3H and R4, wherein R4 is selected from the proteinogenic alpha-amino acids (when taken to include the amine group in Formula I) provided in Table 1; or
    • (ii) R2 is H, R3 is selected from H and C1-C4 alkyl, and R1 is selected from PO3H2, SO3H CO2H, and R5, wherein R5 is selected from the proteinogenic alpha-amino acids (when taken to include the hydroxyl group in Formula I) provided in Table 2:









TABLE 1









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image


















TABLE 2









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image











Preferred embodiments of the fifth aspect of the present invention provide solid oral dosage forms according to any one of the first, second, third or fourth aspect of the invention, and in particular the first or second aspect, comprising a prodrug of a metabolite of ketamine, or analogue thereof, having the structure of Formula I, or a pharmaceutically acceptable salt thereof, wherein X, R1, R2, R3, R4 and R5 are as defined hereinabove.


In preferred embodiments of prodrugs of the fifth aspect of the present invention, OR1 is bonded to the cyclohexanone ring at either the 3, 4, 5 or 6 position, X is one, two, or three haloatoms each independently positioned ortho, meta or para, and each independently selected from F, Cl, Br, and I, and wherein

    • (i) R1 is H, R3 is selected from H and C1-C4 alkyl, and R2 is selected from CH2OPO3H2, CH2OSO3H and R4, wherein R4 is selected from the proteinogenic alpha-amino acids (when taken to include the amine group in Formula I) provided in Table 1; or
    • (ii) R2 is H, R3 is selected from H and C1-C4 alkyl, and R1 is selected from PO3H2, SO3H CO2H, and R5, wherein R5 is selected from




embedded image


preferably wherein R5 has the same stereochemistry as the corresponding L-amino acid.


Preferred embodiments of the fifth aspect comprise compounds wherein X represents one haloatom selected from F, Cl, Br, and I, which is ortho to the C—C bond linking the aryl group to the cyclohexanone. In preferred embodiments of the fifth aspect X represents two haloatoms selected from F, Cl, Br, and I, preferably one or both of which are ortho to the C—C bond linking the aryl group to the cyclohexanone. In preferred embodiments of the fifth aspect one or two haloatoms are ortho to the C—C bond linking the aryl group to the cyclohexanone. In preferred embodiments of the fifth aspect, X represents one haloatom selected from F, Cl, Br, and I. In preferred embodiments of the fifth aspect, X represents two separate haloatoms independently selected from F, Cl, Br, and I, preferably one or both of which are ortho to the C—C bond linking the aryl group to the cyclohexanone. In preferred embodiments of the fifth aspect, X represents three separate haloatoms independently selected from F, Cl, Br, and I, preferably one or more of which are ortho to the C—C bond linking the aryl group to the cyclohexanone. In preferred embodiments of the fifth aspect, X represents three separate haloatoms independently selected from F, Cl, Br, and I, wherein two haloatoms are ortho and one is para to the C—C bond linking the aryl group to the cyclohexanone. In preferred embodiments of the fifth aspect X represents the same haloatom selected from F, Cl, Br, and I. In preferred embodiments of the fifth aspect, X represents Cl. In preferred embodiments of the fifth aspect, X represents one Cl. In preferred embodiments of the fifth aspect, X represents two separate Cl, preferably one or more of which are ortho to the C—C bond linking the aryl group to the cyclohexanone. In preferred embodiments of the fifth aspect, X represents three separate Cl, preferably one or more of which are ortho to the C—C bond linking the aryl group to the cyclohexanone. In preferred embodiments of the fifth aspect at least one haloatom represented by X is Cl. In preferred embodiments of the fifth aspect, all haloatoms represented by X are Cl.


Preferred embodiments of the fifth aspect of the present invention provide a prodrug of a metabolite of ketamine, or analogue thereof, or a pharmaceutically acceptable salt thereof, wherein R4 or R5 have the same stereochemistry as the corresponding L-amino acid.


Preferred embodiments of the fifth aspect of the present invention provide a prodrug of a metabolite of ketamine, or analogue thereof, or a pharmaceutically acceptable salt thereof, wherein the promoiety is R1. In this embodiment, preferred compounds of the fifth aspect are selected from Formulae II-XXV, listed in Table 3, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from H and C1-C4 alkyl, and R1 is selected from PO3H2, SO3H and CO2H. In preferred embodiments of compounds of Formulae II-XXV, X is Cl. In preferred embodiments of compounds of Formulae II-XXV, X is a single Cl. In preferred embodiments of compounds of Formulae II-XXV, X is a single ortho-Cl. In preferred embodiments of compounds of Formulae II-V, X-XIII, and XVIII-XXI, R3 is Me. In particularly preferred embodiments of compounds of Formulae II-V, X-XIII, and XVIII-XXI, X is ortho-Cl and R3 is Me.


Alternative preferred embodiments of the fifth aspect of the present invention provide a prodrug of a metabolite of ketamine, or analogue thereof, or a pharmaceutically acceptable salt thereof, wherein the promoiety is R2. In this embodiment, preferred compounds of the fifth aspect are selected from Formulae XXVI-XLIX, listed in Table 4, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from CH2OPO3H2, CH2OSO3H, and R4.


In preferred embodiments, the prodrug according to the fifth aspect of the present invention is a prodrug of a metabolite of ketamine selected from the metabolites listed in Table 5, or a pharmaceutically acceptable salt thereof.


In particularly preferred embodiments, the prodrug according to the fifth aspect of the present invention is a compound selected from Formulae L-LXXIII listed in Table 6, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from PO3H2, SO3H and CO2H. Most preferably, R1 is PO3H2.


In alternative preferred embodiments, the prodrug according to the fifth aspect of the present invention is a compound selected from Formulae LXXIV-XCVII, listed in Table 7, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from CH2OPO3H2, CH2OSO3H, and R4 as defined above. Most preferably. R2 is selected from CH2OPO3H2 and R4, wherein R4 is selected from




embedded image


preferably wherein R4 has the same stereochemistry as the corresponding L-amino acid.


Particularly preferred prodrugs according to the fifth aspect of the present invention are compounds 1-48 disclosed in Tables 8A and 8B.












TABLE 3











embedded image


II









embedded image


III









embedded image


IV









embedded image


V









embedded image


VI









embedded image


VII









embedded image


VIII









embedded image


IX









embedded image


X









embedded image


XI









embedded image


XII









embedded image


XIII









embedded image


XIV









embedded image


XV









embedded image


XVI









embedded image


XVII









embedded image


XVIII









embedded image


XIX









embedded image


XX









embedded image


XXI









embedded image


XXII









embedded image


XXIII









embedded image


XXIV









embedded image


XXV




















TABLE 4











embedded image


XXVI









embedded image


XXVII









embedded image


XXVIII









embedded image


XXIX









embedded image


XXX









embedded image


XXXI









embedded image


XXXII









embedded image


XXXIII









embedded image


XXXIV









embedded image


XXXV









embedded image


XXXVI









embedded image


XXXVII









embedded image


XXXVIII









embedded image


XXXIX









embedded image


XL









embedded image


XLI









embedded image


XLII









embedded image


XLIII









embedded image


XLIV









embedded image


XLV









embedded image


XLVI









embedded image


XLVII









embedded image


XLVIII









embedded image


XLIX

















TABLE 5









embedded image







2R,6R-hydroxyketamine,







embedded image







2R,6S-hydroxyketamine,







embedded image







2S,6R-hydroxyketamine,







embedded image







2S,6S-hydroxyketamine







embedded image







2R,6R-hydroxynorketamine,







embedded image







2R,6S-hydroxynorketamine,







embedded image







2S,6R-hydroxynorketamine,







embedded image







2S,6S-hydroxynorketamine







embedded image







2R,5R-hydroxyketamine,







embedded image







2R,5S-hydroxyketamine,







embedded image







2S,5R-hydroxyketamine,







embedded image







2S,5S-hydroxyketamine







embedded image







2R,5R-hydroxynorketamine,







embedded image







2R,5S-hydroxynorketamine,







embedded image







2S,5R-hydroxynorketamine,







embedded image







2S,5S-hydroxynorketamine







embedded image







2R,4R-hydroxyketamine,







embedded image







2R,4S-hydroxyketamine,







embedded image







2S,4R-hydroxyketamine,







embedded image







2S,4S-hydroxyketamine







embedded image







2R,4R-hydroxynorketamine,







embedded image







2R,4S-hydroxynorketamine,







embedded image







2S,4R-hydroxynorketamine,







embedded image







2S,4S-hydroxynorketamine



















TABLE 6











embedded image


L









embedded image


LI









embedded image


LII









embedded image


LIII









embedded image


LIV









embedded image


LV









embedded image


LVI









embedded image


LVII









embedded image


LVII









embedded image


LIX









embedded image


LX









embedded image


LXI









embedded image


LXII









embedded image


LXII









embedded image


LXIV









embedded image


LXV









embedded image


LXVI









embedded image


LXVII









embedded image


LXVIII









embedded image


LXIX









embedded image


LXX









embedded image


LXXI









embedded image


LXXII









embedded image


LXXIII




















TABLE 7











embedded image


LXXIV









embedded image


LXXV









embedded image


LXXVI









embedded image


LXXVII









embedded image


LXXVIII









embedded image


LXXIX









embedded image


LXXX









embedded image


LXXXI









embedded image


LXXXII









embedded image


LXXXIII









embedded image


LXXXIV









embedded image


LXXXV









embedded image


LXXXVI









embedded image


LXXXVII









embedded image


LXXXVIII









embedded image


LXXXIX









embedded image


XC









embedded image


XCI









embedded image


XCII









embedded image


XCIII









embedded image


XCIV









embedded image


XCV









embedded image


XCVI









embedded image


XCVII




















TABLE 8A











embedded image


1









embedded image


2









embedded image


3









embedded image


4









embedded image


5









embedded image


6









embedded image


7









embedded image


8









embedded image


9









embedded image


10









embedded image


11









embedded image


12









embedded image


13









embedded image


14









embedded image


15









embedded image


16









embedded image


17









embedded image


18









embedded image


19









embedded image


20









embedded image


21









embedded image


22









embedded image


23









embedded image


24




















TABLE 8B











embedded image


25









embedded image


26









embedded image


27









embedded image


28









embedded image


29









embedded image


30









embedded image


31









embedded image


32









embedded image


33









embedded image


34









embedded image


35









embedded image


36









embedded image


37









embedded image


38









embedded image


39









embedded image


40









embedded image


41









embedded image


42









embedded image


43









embedded image


44









embedded image


45









embedded image


46









embedded image


47









embedded image


48










A sixth aspect of the present invention provides solid oral dosage forms of metabolites of ketamine and prodrugs and analogues thereof, or pharmaceutically acceptable salts thereof, according to any one of aspects I to 5, wherein the solid oral dosage form is for use as a first-line therapy.


In a preferred embodiment of the sixth aspect of the present invention, the solid oral dosage form is for use in a patient who has failed to achieve adequate control of depression symptoms using psychotherapy alone.


In a preferred embodiment of the sixth aspect of the present invention, the solid oral dosage form is for use in a patient who has not previously been treated with an antidepressant agent.


In a seventh aspect of the present invention the pharmaceutically acceptable salt of the compounds described herein is a tartrate salt. Accordingly, a preferred embodiment of the seventh aspect of the present invention is a solid oral dosage form comprising a tartrate salt of a metabolite of ketamine or a prodrug of a metabolite of ketamine, for use in the treatment of a depressive disorder in a patient. In a particularly preferred embodiment of the seventh aspect of the present invention, the salt of the metabolite of ketamine is 2R,6R-hydroxynorketamine L-(+)-tartrate.


In preferred embodiments of the seventh aspect of the present invention, the metabolite of ketamine is a mono-hydroxylated metabolite selected from the metabolites listed in Table 5.


In preferred embodiments of the seventh aspect of the present invention the pharmaceutically acceptable salt comprises 2R,6R-hydroxynorketamine wherein the tartrate salt comprises L-(+)-tartrate, preferably wherein 80% or more of the metabolite of ketamine is 2R,6R-hydroxynorketamine. In an alternative embodiment of the seventh aspect of the present invention the pharmaceutically acceptable salt comprises 2S,6S-hydroxynorketamine wherein the tartrate salt comprises D-(−)-tartrate, preferably wherein 80% or more of the metabolite of ketamine is 2S,6S-hydroxynorketamine.


In preferred embodiments of the seventh aspect of the invention, the tartrate salt of the metabolite of ketamine is crystalline.


In an eighth aspect of the present invention, the solid oral dosage form according to the first, second, third, fourth or sixth aspect of the present invention comprises a tartrate salt of the seventh aspect of the present invention. Preferably the solid oral dosage form is selected from a tablet and a capsule. Preferably the tartrate salt is 2R,6R-hydroxynorketamine L(+)-tartrate.


In preferred embodiments of the eighth aspect of the present invention the solid oral dosage form comprises the active ingredient 2R,6R-hydroxynorketamine L-(+)-tartrate, wherein the active ingredient is present in an amount equivalent to between 10 mg and 100 mg of 2R,6R-hydroxynorketamine freebase.


In preferred embodiments of the eighth aspect of the present invention, the solid oral dosage forms comprise a blend of one or more diluent. Preferably the diluent blend comprises one or more, and preferably two or more, diluents selected from anhydrous lactose, D-mannitol, dicalcium phosphate, calcium carbonate, magnesium oxide, magnesium carbonate, glucose, sorbitol, sucrose, calcium sulphate, starch, dextrates, kaolinite, maltodextrin and lactitol.


More preferred diluents are selected from microcrystalline cellulose, dicalcium phosphate, kaolinite, starch and calcium carbonate.


In preferred embodiments of the eighth aspect the solid oral dosage form is a tablet and comprises a diluent blend comprising microcrystalline cellulose. In further embodiments of the eighth aspect, the diluent blend comprises two or more diluents wherein one diluent is selected from microcrystalline cellulose and starch, and wherein a further diluent is an inorganic diluent.


In further embodiments of the eighth aspect, the diluent blend comprises dicalcium phosphate and microcrystalline cellulose.


In a preferred embodiment of the eighth aspect, the diluent blend excludes lactose monohydrate.


Preferred embodiments of the eighth aspect are wherein the solid oral dosage form is a capsule wherein the capsule shell comprises a constituent selected from gelatin and hydroxypropyl methylcellulose.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1: Shows differential scanning calorimetry (DSC) analysis of a binary mix of 2R,6R-hydroxynorketamine and lactose monohydrate, in a 1:2 ratio of API:Excipient.



FIG. 2: Mean unbound fraction of 2R,6R-hydroxynorketamine in mouse, rat and human plasma.



FIG. 3: Shows the degree of UGT inhibition by 2R,6R-hydroxynorketamine in in vitro assays.



FIG. 4: Shows the rate of hepatic clearance in human, rat and dog hepatocyte assays.





DETAILED DESCRIPTION OF THE INVENTION

The present invention provides a solid oral dosage form comprising a compound selected from a metabolite of ketamine and a prodrug of a metabolite of ketamine, or a pharmaceutically acceptable salt thereof, collectively referred to as ‘compounds of the present invention’, for use in the treatment of a depressive disorder in a patient.


Throughout this specification, one or more aspect of the invention may be combined with one or more features described in the specification to define distinct embodiments of the invention.


References herein to a singular of a noun encompass the plural of the noun, and vice-versa, unless the context implies otherwise.


As used herein, the term ‘patient’ preferably refers to a human patient, but may also refer to a domestic mammal. The term does not encompass laboratory mammals.


As used herein, the term ‘first-line therapy’ is defined as the first course of pharmaceutical treatment administered in response to an episode of a disorder. The term ‘episode’ refers to a single noteworthy happening in the course of a longer series of events, such as one critical period of several during a prolonged disorder.


As used herein, the term ‘depressive disorder’ is defined as a category of mental disorders characterized by at least one of: (a) depressed mood, such as feeling sad, empty or tearful, and (b) significantly reduced interest or feeling no pleasure in all or most activities over a two-week period, combined with at least one further symptom selected from: (c) significant weight loss when not dieting, weight gain, or decrease or increase in appetite, (d) insomnia or increased desire to sleep (e) either restlessness or slowed behaviour that can be observed by others (f) fatigue or loss of energy, (g) feelings of worthlessness, or excessive or inappropriate guilt, (h) trouble making decisions, or trouble thinking or concentrating, and (i) recurrent thoughts of death or suicide, or a suicide attempt. Depressive disorders include: major depression, persistent depressive disorder, bipolar disorder, psychotic depression, postpartum depression, premenstrual dysphoric disorder (PMDD) and atypical depression.


Major depression, also referred to as clinical depression, is defined as the presence of five or more of the following symptoms over a period of two-weeks or more (also referred to herein as a ‘major depressive episode’), most of the day, nearly every day.

    • depressed mood, such as feeling sad, empty or tearful (in children and teens, depressed mood can appear as constant irritability);
    • significantly reduced interest or feeling no pleasure in all or most activities.
    • significant weight loss when not dieting, weight gain, or decrease or increase in appetite (in children, failure to gain weight as expected);
    • insomnia or increased desire to sleep;
    • either restlessness or slowed behaviour that can be observed by others;
    • fatigue or loss of energy;
    • feelings of worthlessness, or excessive or inappropriate guilt;
    • trouble making decisions, or trouble thinking or concentrating;
    • recurrent thoughts of death or suicide, or a suicide attempt.


At least one of the symptoms must be either a depressed mood or a loss of interest or pleasure.


Persistent depressive disorder, also known as dysthymia, is defined as a patient exhibiting the following two features:


A. has depressed mood for most the time almost every day for at least two years. Children and adolescents may have irritable mood, and the time frame is at least one year.


B. While depressed, a person experiences at least two of the following symptoms:

    • Either overeating or lack of appetite.
    • Sleeping too much or having difficulty sleeping.
    • Fatigue, lack of energy.
    • Poor self-esteem.
    • Difficulty with concentration or decision making.
    • Feeling hopeless.


Bipolar disorder, also known as manic-depressive illness, is a disorder that causes unusual shifts in mood, energy, activity levels, and the ability to carry out day-to-day tasks.


There are three defined sub-categories of bipolar disorder: all of them involve clear changes in mood, energy, and activity levels. These moods range from periods of extremely “up,” elated, and energized behaviour (known as manic episodes, and defined further below) to very sad, “down,” or hopeless periods (known as depressive episodes). Less severe manic periods are known as hypomanic episodes.


Bipolar I Disorder—defined by manic episodes that last at least 7 days, or by manic symptoms that are so severe that the person needs immediate hospital care. Usually, depressive episodes occur as well, typically lasting at least 2 weeks. Episodes of depression with mixed features (having depression and manic symptoms at the same time) are also possible.


Bipolar II Disorder—defined by a pattern of depressive episodes and hypomanic episodes, but not the full-blown manic episodes described above.


Bipolar depression is defined as an individual who is experiencing depressive symptoms with a previous or coexisting episode of manic symptoms, but does not fit the clinical criteria for bipolar disorder.


Cyclothymic Disorder (also called cyclothymia)—defined by numerous periods of hypomanic symptoms as well as numerous periods of depressive symptoms lasting for at least 2 years (1 year in children and adolescents). However, the symptoms do not meet the diagnostic requirements for a hypomanic episode and a depressive episode.


Psychotic depression, also known as depressive psychosis, is defined as a major depressive episode that is accompanied by psychotic symptoms including hallucinations and delusions.


Postpartum depression, also referred to as postnatal depression, as referred to herein refers to a diagnosis that meets the criteria for major depression when occurring in a mother within the first year from giving birth.


Premenstrual dysphoric disorder as referred to herein, is present in a patient who meets the following criteria A-C.


Criterion A is that in most menstrual cycles during the past year, at least 5 of the following 11 symptoms (including at least 1 of the 4 listed) must be present in the final week before the onset of menses, start to improve within a few days after the onset of menses, and become minimal or absent in the week postmenses.

    • 1. Marked lability (e.g., mood swings)
    • 2. Marked irritability or anger
    • 3. Markedly depressed mood
    • 4. Marked anxiety and tension
    • 5. Decreased interest in usual activities
    • 6. Difficulty in concentration
    • 7. Lethargy and marked lack of energy
    • 8. Marked change in appetite (e.g., overeating or specific food cravings)
    • 9. Hypersomnia or insomnia
    • 10. Feeling overwhelmed or out of control
    • 11. Physical symptoms (e.g., breast tenderness or swelling, joint or muscle pain, a sensation of ‘bloating’ and weight gain)


Criterion B one (or more) of the following symptoms must be present:

    • 1. Marked affective lability (e.g., mood swings; feeling suddenly sad or tearful, or increased sensitivity to rejection).
    • 2. Marked irritability or anger or increased interpersonal conflicts.
    • 3. Marked depressed mood, feelings of hopelessness, or self-deprecating thoughts.
    • 4. Marked anxiety, tension, and/or feelings of being keyed up or on edge.


Criterion C one (or more) of the following symptoms must be additionally be present, to reach a total of five symptoms when combined with symptoms from Criterion B above.

    • 1. Decreased interest in usual activities (e.g., work, school, friends, hobbies).
    • 2. Subjective difficulty in concentration.
    • 3. Lethargy, easy fatigability, or marked lack of energy.
    • 4. Marked change in appetite; overeating; or specific food cravings.
    • 5. Hypersomnia or insomnia
    • 6. A sense of being overwhelmed or out of control.
    • 7. Physical symptoms such as breast tenderness or swelling, joint or muscle pain, a sensation of “bloating,” or weight gain.


The symptoms in Criteria A-C must have been met for most menstrual cycles that occurred in the preceding year.


Atypical depression is a subtype of major depression or dysthymic disorder that involves several specific symptoms, including increased appetite or weight gain, sleepiness or excessive sleep, marked fatigue or weakness, moods that are strongly reactive to environmental circumstances, and feeling extremely sensitive to rejection.


Post-traumatic stress disorder (PTSD) is a condition which develops following a stressful event or situation of an exceptionally threatening or catastrophic nature, which is likely to cause pervasive distress in almost anyone. PTSD does not therefore develop following those upsetting situations that are described as ‘traumatic’ in everyday language, for example, divorce. PTSD symptoms include re-experiencing, such as flashbacks, avoidance of reminders of the trauma or rumination and hyperarousal or emotional numbing. PTSD with co-morbid depression is defined as PTSD in an individual who also meets the criteria for one or more depressive disorder.


Obsessive-compulsive disorder (OCD) is defined by the presence of either obsessions or compulsions, but commonly both. The symptoms can cause significant functional impairment and/or distress. An obsession is defined as an unwanted intrusive thought, image or urge that repeatedly enters the person's mind. Compulsions are repetitive behaviours or mental acts that the person feels driven to perform. A compulsion can either be overt and observable by others, such as checking that a door is locked, or a covert mental act that cannot be observed, such as repeating a certain phrase in one's mind. OCD with co-morbid depression is defined as OCD in an individual who also meets the criteria for one or more depressive disorder.


Social anxiety disorder is defined as the persistent fear and anxiety about one or more social or performance situations. The two main forms are generalised social anxiety w performance social anxiety. Generalised social anxiety affects most, if not all areas of life. This is the more common type and affects around 70% of those affected by the disorder. Performance social anxiety is where these feelings only occur in a few specific situations such as public speaking, eating in public or dealing with figures of authority. Social anxiety disorder with co-morbid depression is defined as social anxiety disorder in an individual who also meets the criteria for one or more depressive disorder.


Tic disorders are defined as neuropsychiatric syndrome, characterised by motor and vocal tics, which runs a fluctuating course. Tics can be defined as sudden, purposeless, repetitive, non-rhythmic, stereotyped movements or vocalisations for example eye twitching or blinking. Examples of vocal tics are throat clearing, grunting and barking. Tic disorder with co-morbid depression is defined as social anxiety disorder in an individual who also meets the criteria for one or more depressive disorder.


As used herein, the term ‘ketamine’ refers to 2-(2-Chlorophenyl)-2-(methylamino)-cyclohexanone. As used herein, the term ‘2R,6R-hydroxynorketamine’ refers to 2R,6R-2-(2-Chlorophenyl)-2-(amino)-6-hydroxycyclohexanone.


As used herein, the term ‘metabolite of ketamine’ includes R-norketamine, S-norketamine, 2R,6R-hydroxyketamine, 2R,6S-hydroxyketamine, 2S,6R-hydroxyketamine, 2S,6S-hydroxyketamine, R-(5,6)dehydroketamine, S-(5,6)dehydroketamine, R-(3,4)dehydroketamine, S-(3.4)dehydroketamine, R-(5,6)dehdronorketamine, S-(5,6)dehydronorketamine, R-(3,4)dehydronorketamine, S-(3,4)dehydronorketamine, 2R,6R-hydroxynorketamine, 2R,6S-hydroxynorketamine, 2S,6R-hydroxynorketamine, 2S,6S-hydroxynorketamine, 2R,5R-hydroxynorketamine, 2R,5S-hydroxynorketamine, 2S,5R-hydroxynorketamine, 2S,5S-hydroxynorketamine, 2R,6S-hydroxynorketamine, 2R,4S-hydroxynorketamine, 2S,4R-hydroxynorketamine, 2S,4S-hydroxynorketamine or any mixture thereof, including any racemic mixture thereof, or any pharmaceutically acceptable salt thereof.


In preferred embodiments, the compound for use in the present invention is selected from R-(5,6)dehydronorketamine, S-(5,6)dehydronorketamine, 2R,6R-hydroxynorketamine, 2R,6S-hydroxynorketamine, 2S,6R-hydroxynorketamine, 2S,6S-hydroxynorketamine, or any pharmaceutical salt thereof. In more preferred embodiments the compound for use in the present invention is a pharmaceutically acceptable salt of 2R,6R-hydroxynorketamine.


As used herein, the term ‘prodrug of a metabolite of ketamine’ is defined as a compound formed by the covalent bonding of the amine group or hydroxyl group of a metabolite of ketamine with a biolabile moiety, or ‘promoiety’, as defined by Formula I.


As used herein, the term ‘pharmaceutically acceptable salt’ is defined as the product of a reaction with an acid or base to form a non-toxic salt. The term pharmaceutically acceptable salt includes solvates thereof. Wherever a compound is referred to by its generic or systematic name, the term is taken to include all pharmaceutically acceptable salts.


In a preferred embodiment, the solid oral dosage forms of the present invention comprise a pharmaceutically acceptable salt of the compound of the present invention wherein said salt is not the hydrochloride salt, in particular wherein the solid oral dosage form comprises a metabolite of ketamine, rather than a prodrug thereof. The hydrochloride salt of metabolites of ketamine of the present invention are non-optimal for the preparation of solid oral dosage forms of the present invention. Without wishing to be bound by theory, it is believed that this is caused, in part, by the common-ion effect owing to the presence of high concentrations of chloride ions in the gastric juice produced in the stomach.


In a preferred embodiment, the compound for use according to the invention is for treating a depressive disorder selected from the list comprising major depression, persistent depressive disorder, bipolar disorder (type 1 or type 2), psychotic depression, postpartum depression, premenstrual dysphoric disorder (PMDD), atypical depression, post-traumatic stress disorder (PTSD) with comorbid depression, obsessive compulsive disorder (OCD) with comorbid depression, social anxiety with co-morbid depression, and tic disorders (eg. Tourette's Syndrome) with co-morbid depression.


In a preferred embodiment, the depressive disorder is major depression.


In an alternative preferred embodiment, the depressive disorder is persistent depressive disorder. In an alternative preferred embodiment the depressive disorder is bipolar disorder.


In a preferred embodiment, the depressive disorder is selected from bipolar depression, postpartum depression, premenstrual dysphoric disorder (PMDD), atypical depression, post-traumatic stress disorder (PTSD) with comorbid depression, obsessive compulsive disorder (OCD) with comorbid depression, social anxiety with co-morbid depression, and tic disorders (eg. Tourette's Syndrome) with co-morbid depression.


Prodrugs

An aspect of the present invention provides a prodrug of a metabolite of ketamine, or analogue thereof, having the structure of Formula I, or a pharmaceutically acceptable salt thereof:




embedded image


wherein OR1 is bonded to the cyclohexanone ring at either the 3, 4, 5 or 6 position, wherein X is one, two, or three haloatoms each independently positioned ortho, meta or para, and each independently selected from selected from F, Cl, Br, and I, and wherein

    • (i) wherein R1 is H, R3 is selected from H and C1-C4 alkyl, and R2 is selected from CH2OPO3H2, CH2OSO3H, and R4, wherein R4 is selected from the proteinogenic alpha-amino acids (when taken to include the amine group in Formula I) provided in Table 1; or
    • (ii) R2 is H, R3 is selected from H and C1-C4 alkyl, and R1 is selected from PO3H2, SO3H and CO2H, and R5, wherein R5 is selected from the proteinogenic alpha-amino acids (when taken to include the hydroxyl group in Formula I) provided in Table 2.


In preferred embodiments of prodrugs of Formula I, OR1 is bonded to the cyclohexanone ring at either the 3, 4, 5 or 6 position. X is one, two, or three haloatoms each independently positioned ortho, meta or para, and each independently selected from F, Cl, Br, and I, and wherein

    • (i) wherein R1 is H, R3 is selected from H and C1-C4 alkyl, and R2 is selected from CH2OPO3H2, CH2OSO3H and R4, wherein R4 is selected from the proteinogenic alpha-amino acids (when taken to include the amine group in Formula I) provided in Table 1: or
    • (ii) R1 is H, R3 is selected from H and C1-C4 alkyl, and R1 is selected from PO3H2, SO3H CO2H, and R5, wherein R5 is selected from




embedded image


preferably wherein R5 has the same stereochemistry as the corresponding L-amino acid.


A particular advantage of such prodrugs is that they are acidic, and capable of forming salts with cationic counterions. This is particularly advantageous because acids generally have higher oral bioavailability than bases, which is thought to be a result of better solubility and lower clearance. Bases tend to be protonated in the gastrointestinal tract and have reduced lipophilicity, which limits passive absorption across membranes. Accordingly, an aspect of the present invention is to provide pharmaceutically acceptable salts of a compound of Formula I with a cationic counterion. An embodiment of the invention provides a pharmaceutical formulation comprising a pharmaceutically acceptable salt of a compound of Formula I with a cationic counterion. A further embodiment of the invention provides a solid oral dosage form comprising a pharmaceutically acceptable salt of a compound of Formula I with a cationic counterion.


Preferred cationic counterions for use in pharmaceutically acceptable salts of a compound of Formula (IV) are selected from sodium, potassium, magnesium, calcium, aluminium, meglumine, benzathine, procaine, ethylene diamine, lysine, tromethamine and zinc.


Preferably the prodrug of the present invention is a prodrug of a metabolite of ketamine selected from 2R,6R-hydroxyketamine, 2R,6S-hydroxyketamine, 2S,6R-hydroxyketamine, 2S,6S-hydroxyketamine, 2R,6R-hydroxynorketamine, 2R,6S-hydroxynorketamine, 2S,6R-hydroxynorketamine, 2S,6S-hydroxynorketamine, 2R,5R-hydroxyketamine, 2R,5S-hydroxyketamine, 2S,5R-hydroxyketamine, 2S,5S-hydroxyketamine, 2R,5R-hydroxynorketamine, 2R,5S-hydroxynorketamine, 2S,5R-hydroxynorketamine, 2S,5S-hydroxynorketamine, 2R,4R-hydroxyketamine, 2R,4S-hydroxyketamine, 2S,4R-hydroxyketamine, 2S,4S-hydroxyketamine, 2R,4R-hydroxynorketamine, 2R,4S-hydroxynorketamine, 2S,4R-hydroxynorketamine, and 2S,4S-hydroxynorketamine.


Prodrugs with hydroxyl-bonded promoieties can be synthesized from the corresponding metabolite of ketamine or analogue thereof via the synthesis described in Scheme 1:




embedded image


In particular, O-phosphate prodrugs of ketamine metabolites may be synthesized from their corresponding ketamine metabolite via the synthesis provided in Scheme 2:




embedded image


Prodrugs with amine-bonded promoieties can be synthesized from the corresponding metabolite of ketamine or analogue thereof via the synthesis described in Scheme 3:




embedded image


In particular, N-methylenephosphate prodrugs of ketamine metabolites may be synthesized from their corresponding ketamine metabolite via the synthesis provided in Scheme 4:




embedded image


Preferred prodrugs of the present invention are prodrugs having a structure of Formula Ia, or a pharmaceutically acceptable salt thereof




embedded image




    • wherein:
      • (i) R1 is selected from PO3H2, SOH, CO2H, and R5, wherein R5 is selected from







embedded image




    • R2 is H;

    • R3 is selected from H and C1-C4 alkyl;

    • and wherein X is ortho-Cl; or
      • (ii) R1 is H:

    • R2 is selected from CH2OPO3H2, CH2OSO3H and R4, wherein R4 is selected from the groups listed in Table 1:












TABLE 1









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image











In preferable prodrugs, R3 is H or Me.


In preferable prodrugs, R3 is H. In preferable prodrugs, R1 is selected from PO3H2, SO3H, CO2H, and R5, wherein R5 is selected from




embedded image


In preferable prodrugs, X is Cl. In preferable prodrugs. X is a single ortho-Cl. In preferable prodrugs, R4 or R5 has the same stereochemistry as the corresponding L-amino acid. In preferable prodrugs, R1 is PO3H2 and R3 is Me or H.


In particularly preferable prodrugs, R3 is H and X is a single ortho-Cl having Formula Ib:




embedded image


wherein:

    • (i) R1 is selected from PO3H2, SO3H, CO2H, and R5, wherein R5 is selected from




embedded image




    • R2 is H; or

    • (ii) R1 is H;

    • R2 is selected from CH2OPO3H2, CH2OSO3H and R4, wherein R4 is selected from the groups listed in Table 1.





Tartrate Salt of Metabolites of Ketamine


An aspect of the present invention provides tartrate salts of metabolites of ketamine as described herein. It has been discovered that tartrate salts of metabolites of the present invention are suitable for use in oral formulations of the present invention. A further advantage of using tartrate salts in the solid dosage forms of the present invention is that either L-(+)-tartaric acid or D-(−)-tartaric acid may be employed in chiral recrystallization to aid separation of one metabolite of ketamine from its enantiomer. Thus, the appropriate tartrate salt of metabolites of ketamine described herein may be manufactured into solid oral dosage forms directly following chiral separation from its enantiomers and/or diastereomers.


Accordingly, in an aspect of the present invention the metabolites of ketamine described herein are in the form of their tartrate salt. A preferred embodiment of the present invention is a solid oral dosage form comprising a tartrate salt of a metabolite of ketamine or a prodrug of a metabolite of ketamine, for use in the treatment of a depressive disorder in a patient. A particularly preferred embodiment of the present invention is 2R,6R-hydroxynorketamine L-(+)-tartrate.


In preferred embodiments of the present invention, the metabolite of ketamine is a mono-hydroxylated metabolite selected from the metabolites listed in Table 5.


In preferred embodiments of the present invention the pharmaceutically acceptable salt comprises 2R,6R-hydroxynorketamine wherein the tartrate salt comprises L-(+)-tartrate, preferably wherein 80% or more of the metabolite of ketamine is 2R,6R-hydroxynorketamine. In an alternative preferred embodiment of the present invention the pharmaceutically acceptable salt comprises 2S,6S-hydroxynorketamine wherein the tartrate salt comprises D-(−)-tartrate, preferably wherein 80% or more of the metabolite of ketamine is 2S,6S-hydroxynorketamine.


In preferred embodiments of the invention, the tartrate salt of the metabolite of ketamine is crystalline.


In embodiments, the solid oral dosage form of the present invention comprises a tartrate salt. Preferably the solid oral dosage form is selected from tablet and capsule. Preferably the tartrate salt is 2R,6R-hydroxynorketamine L(+)-tartrate.


In preferred embodiments the solid oral dosage form comprises the active ingredient 2R,6R-hydroxynorketamine L-(+)-tartrate, wherein the active ingredient is present in an amount equivalent to between 10 mg and 100 mg of 2R,6R-hydroxynorketamine freebase.


In preferred embodiments of solid oral dosage forms comprising tartrate salts of the present invention, the solid oral dosage forms comprise a blend of one or more diluents. Preferably the diluent blend comprises one or more, and preferably two or more, diluents selected from anhydrous lactose, D-mannitol, dicalcium phosphate, calcium carbonate, magnesium oxide, magnesium carbonate, glucose, sorbitol, sucrose, calcium sulphate, starch, dextrates, kaolinite, maltodextrin and lactitol. More preferred diluents are selected from microcrystalline cellulose, dicalcium phosphate, kaolinite, starch and calcium carbonate.


In preferred embodiments of solid oral dosage forms comprising tartrate salts of the present invention, the solid oral dosage form is a tablet and comprises a diluent blend comprising microcrystalline cellulose. In further embodiments the diluent blend comprises two or more diluents wherein one diluent is selected from microcrystalline cellulose and starch, and wherein a further diluent is an inorganic diluent.


Preferred embodiments of solid oral dosage forms comprising tartrate salts of the present invention comprise dicalcium phosphate and microcrystalline cellulose.


In preferred embodiments of solid oral dosage forms comprising tartrate salts of the present invention, solid oral dosage form comprises a diluent blend which excludes lactose monohydrate.


Preferred embodiments of solid oral dosage forms comprising tartrate salts of the present invention, the solid oral dosage form is a capsule wherein the capsule shell comprises a constituent selected from gelatin and hydroxypropyl methylcellulose.


Combination with Serotonin Modulators


An aspect of the present invention provides a metabolite of ketamine or a pharmaceutically acceptable salt thereof, or a prodrug thereof, for co-administration with a serotonin modulator. As used herein, the term ‘serotonin modulator’ is defined as any compound which affects the serotonin neurotransmitter (serotonergic system). Serotonin modulators include serotonin stimulators (e.g. vortioxetine), serotonin antagonist and reuptake inhibitors (SARIs; e.g. etoperidone, lorpiprazole, lubazodone, mepiprazole, nefazodone, and trazodone), selective serotonin reuptake inhibitors (SSRIs; e.g. citalopram, escitalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, dapoxetine), serotonin-norepinephrine reuptake inhibitors (SNRIs; e.g. venlafaxine, milnacipran, duloxetine, levomilnacipran, desvenlafaxine, sibutramine), and noradrenergic and specific serotonergic antidepressants (NaSSAs; e.g. aptazapine, esmirtazapine, mianserin, mirtazapine, setiptiline).


As used herein, the term ‘coadministration’ is defined as either the administration of a formulation which contains both the dosage form of the present invention and the serotonin modulator, or the simultaneous, essentially simultaneous, sequential or separate administration within a given dosing period of separate formulations containing the solid oral dosage form of the present invention and the serotonin modulator, respectively. A problem common to all combination therapies is the risk of drug-drug interactions (DDIs) which can lead to unwanted side effects which are not present in one or other members of the combination. Specifically, the potential for adverse DDIs increases if one member of the combination affects the rate of metabolism of another member of the combination. Furthermore, when combined with a serotonin modulator, a drug which interferes with the serotonergic system may result in adverse DDIs. It has been discovered that ketamine metabolites according to the present invention do not adversely affect the rate of metabolism of serotonin modulators, nor do they interfere with the serotonergic system, and combinations of compounds of the present invention with serotonin modulators exhibit limited adverse DDIs.


A preferred class of serotonin modulators for use according to the present invention are SSRIs. Preferred SSRIs are citalopram, escitalopram, fluoxetine, fluvoxamine, paroxetine, sertraline and dapoxetine. Most preferred SSRIs are citalopram or escitalopram.


Accordingly, an aspect of the present invention provides a combination of an SSRI and the solid oral dosage form as described herein for use in the treatment of a depressive disorder in a patient. In a preferred embodiment, said combination is selected from the following list:


R-(5,6)dehydronorketamine and citalopram, S-(5,6)dehydronorketamine and citalopram, 2R,6R-hydroxynorketamine and citalopram, 2R,6S-hydroxynorketamine and citalopram, 2S,6R-hydroxynorketamine and citalopram, 2S,6S-hydroxynorketamine and citalopram. R-(5,6)dehydronorketamine and escitalopram, S-(5,6)dehydronorketamine and escitalopram, 2R,6R-hydroxynorketamine and escitalopram, 2R,6S-hydroxynorketamine and escitalopram, 2S,6R-hydroxynorketamine and escitalopram, 2S,6S-hydroxynorketamine and escitalopram, R-(5,6)dehydronorketamine and fluoxetine, S-(5,6)dehydronorketamine and fluoxetine, 2R,6R-hydroxynorketamine and fluoxetine, 2R,6S-hydroxynorketamine and fluoxetine, 2S,6R-hydroxynorketamine and fluoxetine, 2S,6S-hydroxynorketamine and fluoxetine, R-(5,6)dehydronorketamine and fluvoxamine, S-(5,6)dehydronorketamine and fluvoxamine, 2R,6R-hydroxynorketamine and fluvoxamine, 2R,6S-hydroxynorketamine and fluvoxamine, 2S,6R-hydroxynorketamine and fluvoxamine, 2S,6S-hydroxynorketamine and fluvoxamine, R-(5,6)dehydronorketamine and paroxetine, S-(5,6)dehydronorketamine and paroxetine, 2R,6R-hydroxynorketamine and paroxetine, 2R,6S-hydroxynorketamine and paroxetine, 2S,6R-hydroxynorketamine and paroxetine, 2S,6S-hydroxynorketamine and paroxetine, R-(5,6)dehydronorketamine and sertraline, S-(5,6)dehydronorketamine and sertraline, 2R,6R-hydroxynorketamine and sertraline, 2R,6S-hydroxynorketamine and sertraline, 2S,6R-hydroxynorketamine and sertraline, 2S,6S-hydroxynorketamine and sertraline, R-(5,6)dehydronorketamine and dapoxetine, S-(5,6)dehydronorketamine and dapoxetine, 2R,6R-hydroxynorketamine and dapoxetine, 2R,6S-hydroxynorketamine and dapoxetine, 2S,6R-hydroxynorketamine and dapoxetine, 2S,6S-hydroxynorketamine and dapoxetine,


More preferred combinations according to the invention are selected from the following: R-(5,6)dehydronorketamine and citalopram, S-(5,6)dehydronorketamine and citalopram, 2R,6R-hydroxynorketamine and citalopram, 2S,6S-hydroxynorketamine and citalopram, R-dehydroketamine and escitalopram, S-dehydroketamine and escitalopram, 2R,6R-hydroxynorketamine and escitalopram, 2S,6S-hydroxynorketamine and escitalopram. A particularly preferred combination according to the present invention is 2R,6R-hydroxynorketamine and escitalopram.


In a preferred embodiment of the invention, the compound is for use in a patient who has not previously been treated with an antidepressant agent. As used herein, the term ‘antidepressant agent’ is defined as any pharmaceutical product that is administered to combat or reduce the symptoms of a depressive disorder.


In a preferred embodiment of the invention, the patient is a patient who has failed to achieve adequate control of depression symptoms using psychotherapy alone. As used herein, the term ‘depression symptoms’ include:

    • depressed mood, such as feeling sad, empty or tearful (in children and teens, depressed mood can appear as constant irritability);
    • significantly reduced interest or feeling no pleasure in all or most activities;
    • significant weight loss when not dieting, weight gain, or decrease or increase in appetite (in children, failure to gain weight as expected);
    • insomnia or increased desire to sleep;
    • either restlessness or slowed behaviour that can be observed by others;
    • fatigue or loss of energy;
    • feelings of worthlessness, or excessive or inappropriate guilt;
    • trouble making decisions, or trouble thinking or concentrating;
    • recurrent thoughts of death or suicide, or a suicide attempt:


As used herein, the term ‘psychotherapy’ is defined as the use of psychological methods to help a person change and overcome problems in desired ways.


Depression symptoms may be quantified using a Patient Health Questionnaire-9 (PHQ-9).


The PHQ-9 is a self-administered patient questionnaire which asks the patient the following question:

    • ‘Over the last two weeks, how often have you been bothered by any of the following problems?’
      • Little interest or pleasure in doing things?
      • Feeling down, depressed, or hopeless?
      • Trouble falling or staying asleep, or sleeping too much?
      • Feeling tired or having little energy?
      • Poor appetite or overeating?
      • Feeling bad about yourself—or that you are a failure or have let yourself or your family down?
      • Trouble concentrating on things, such as reading the newspaper or watching television?
      • Moving or speaking so slowly that other people could have noticed?
      • Or the opposite—being so fidgety or restless that you have been moving around a lot more than usual?
      • Thoughts that you would be better off dead, or of hurting yourself in some way?


The PHQ-9 score is calculated by assigning scores of 0, 1, 2, and 3, respectively, to the response categories of ‘not at all’, ‘several days’, ‘more than half the days’, and ‘nearly every day’, and adding together the scores for the nine questions.


In embodiments of the invention, a reduction in PHQ-9 score is achieved of 2 points or more, or 3 points or more, or 5 points or more, or 10 points or more, or 15 points or more, or 20 points or more over the period of a course. As used herein, the term ‘course’ or ‘course of treatment’ is the period of time in which the patient is treated in accordance with the present invention. In embodiments of the invention, a course is between 2 weeks and 12 months, or between 3 weeks and 6 months, or between 4 weeks and 5 months or between 6 weeks and 4 months, or between 2 months and 3 months. In a preferred embodiment of the invention, a course is 6 months or less. In a preferred embodiment of the invention, a course is three months or less.


Preferably, when used in combination with a serotonin modulator, the metabolite of the present invention may be administered in any formulation suitable for pharmaceutical administration. Preferably the metabolite of the present invention is in a dosage form selected from a tablet, a pill, a capsule, a caplet, a powder, granules, orodispersible tablet, sterile parenteral solution or suspension, powder for injection, metered aerosol or liquid spray, drops, ampoule, autoinjector, suppository, sublingual spray, sublingual patch, sublingual film, buccal patch, buccal spray, buccal film, intranasal sprayable solution, intranasal sprayable suspension, and intranasal powder. In preferred embodiments of the invention, the dosage form of the compound of the present invention is selected from orodispersible tablet, sublingual spray, sublingual patch, sublingual film, buccal patch, buccal spray, buccal film, intranasal sprayable solution, intranasal sprayable suspension, and intranasal powder, and is preferably a dosage form suitable for intranasal administration.


Solid Oral Dosage Forms

An aspect of the present invention is a solid oral dosage form comprising a compound selected from a metabolite of ketamine and a prodrug of a metabolite of ketamine, or a pharmaceutically acceptable salt thereof. Preferably the solid oral dosage form is for use in the treatment of a depressive disorder in a patient.


Parenteral administration of metabolites of ketamine, in particular 2R,6R-hydroxynorketamine, have been found to be effective in treating depressive disorders in Zanos et al, Nature, (2016), 533, 481-486. However, these known parenteral formulations suffer some of the same drawbacks as are evident for the use of intravenous ketamine for treating depression, and are inappropriate for treating the majority of depressed patients, for whom treatment in an outpatient setting without the need of a medical professional would be preferable. There are, however, challenges in the development of alternative formulations which overcome the drawbacks of prior art formulations. For example, freebase metabolites of ketamine such as 2R,6R-hydroxynorketamine exist as a viscous oil under ambient conditions and are particularly difficult to process into a pharmaceutical formulation unless in the liquid state.


As used herein, the term ‘solid oral dosage form’ is defined as a solid pharmaceutical formulation which can be swallowed whole, chewed and swallowed, or dissolved, dispersed or absorbed via the oral cavity. Solid oral dosage forms include tablets, pills, capsules, caplets, orodispersible tablets, powders, granules and gums. Solid oral dosage forms are not taken to include liquid or aerosol formulations, powders for inhalation, or powders for injection.


In preferred embodiments of the first aspect of the present invention, the metabolite of ketamine is in the form of an acid-addition salt.


In a preferred embodiment, the solid oral dosage form for use according to the invention is for treating a depressive disorder selected from the list comprising major depression, persistent depressive disorder, bipolar disorder, psychotic depression, postpartum depression, premenstrual dysphoric disorder (PMDD) and atypical depression.


In a preferred embodiment, the solid oral dosage form is for use in treating major depression.


In a preferred embodiment, the solid oral dosage form is for use in treating persistent depressive disorder.


In a preferred embodiment, the solid oral dosage form is for use in treating a bipolar disorder.


In a preferred embodiment, the solid oral dosage form is for use in treating bipolar depression.


In a preferred embodiment, the solid oral dosage form is for use in treating postpartum depression.


In a preferred embodiment, the solid oral dosage form is for use in treating OCD with co-morbid depression.


In a preferred embodiment, the solid oral dosage form is for use in treating PTSD with co-morbid depression.


In a preferred embodiment, the solid oral dosage form is for use in treating a social anxiety disorder with co-morbid depression.


In a preferred embodiment of the invention, the solid oral dosage form further comprises a serotonin modulator. A preferred class of serotonin modulators comprised in the solid oral dosage form of the present invention are SSRIs.


In a preferred embodiment, the solid oral dosage form of the present invention is for use in a patient who has not previously been treated with an antidepressant agent. In a preferred embodiment of the invention, the solid oral dosage form is for use in a patient who has failed to achieve adequate control of anxiety symptoms using psychotherapy alone.


A preferred aspect of the present invention is a solid oral dosage form comprising (2R,6R)-hydroxynorketamine, or a pharmaceutically acceptable salt thereof, for use in the treatment of a depressive disorder. The solid oral dosage form may be for use in any method of treatment as described herein.


In preferred embodiments of the first aspect of the present invention, the solid oral dosage form comprises 2R,6R-hydroxynorketamine in the form of an acid-addition salt. It has been found that metabolites of ketamine of the present invention, and 2R,6R-hydroxynorketamine, present challenges for solid oral dosage formulation as a freebase, because they are viscous oils at ambient conditions. Accordingly, the formulation of solid oral dosage forms metabolites of ketamine according to the present invention are preferably prepared using an acid addition salt of said metabolite.


Solid oral dosage forms according to the present invention may be prepared by mixing the principle active agent(s) with a pharmaceutical carrier, e.g. corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, or dicalcium phosphate, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogenous mixture of the active agent(s). When referring to these preformulation compositions as homogenous, it is meant that the active agent is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective dosage forms such as tablets, pills and capsules. The solid preformulation composition is then subdivided into unit dosage forms of the type described above.


Suitable unit doses of the solid oral dosage form according to the present invention contain between 0.5 mg and 2000 mg of the metabolite of ketamine, or pharmaceutically acceptable salt thereof, or prodrug thereof, or between 1 mg and 900 mg, or between 2 mg and 800 mg, or between 3 mg and 700 mg, or between 4 mg and 600 mg, or between 5 mg and 500 mg, or between 6 mg and 400 mg, or between 7 mg and 300 mg, or between 8 mg and 200 mg, or between 9 mg and 150 mg, or between 10 mg and 100 mg, or between 20 mg and 90 mg, or between 30 mg and 80 mg, or between 40 mg and 70 mg, or between 50 mg and 60 mg. In preferred embodiments of the present invention, the solid oral dosage form is provided in a unit dose containing between 10 mg and 100 mg of the metabolite of ketamine, or pharmaceutically acceptable salt thereof, or prodrug thereof. In preferred embodiments of the present invention, the solid oral dosage form is provided in a unit dose containing between 10 mg and 100 mg of 2R,6R-hydroxynorketamine, or pharmaceutically acceptable salt thereof, or prodrug thereof.


Quantities of weight provided herein refer to the free-form equivalent of a compound of the present invention. For example a unit dose of 50 mg 2R,6R-hydroxynorketamine hydrochloride, contains the mass equivalent of 50 mg freebase 2R,6R-hydroxynorketamine, and has an actual mass of 57.6 mg.


Suitable unit doses of the serotonin modulators for use in aspects of the present invention include the unit doses for these compounds as described in Martindale: The Complete Drug Reference (London, the Pharmaceutical Press, 38th Edition) and US Pharmacopoeia and National Formulary 2016 Edition.


Suitable unit doses for citalopram include 10 mg, 20 mg and 40 mg. Suitable unit doses for escitalopram include 5 mg, 10 mg and 20 mg. Suitable unit doses for fluoxetine include 20 mg and 60 mg. Suitable unit doses for fluvoxamine include 50 mg and 100 mg. Suitable unit doses for paroxetine include 10 mg, 20 mg and 30 mg. Suitable unit doses for sertraline include 50 mg and 100 mg. Suitable unit doses for dapoxetine include 30 mg and 60 mg.


Solid oral dosage forms according to the present invention are preferably formulated in unit doses for administration between once a day and once a week, or between once every two days and twice a week. In a preferred embodiment the unit dose is for administration once a day.


Preferred embodiments of the invention are solid oral dosage forms comprising 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof. Preferably the solid oral dosage form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof is for use in treating a depressive disorder in a patient.


Preferably the solid oral dosage form comprises between 10 mg and 100 mg of 2R,6R-hydroxynorketamine, or the equivalent thereof.


Preferably the solid oral dosage form comprises a crystalline form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof.


Preferably the solid oral dosage form comprises a pharmaceutically acceptable salt of 2R,6R-hydroxynorketamine with the proviso that the solid oral dosage form does not comprise 2R,6R-hydroxynorketamine hydrochloride.


Preferably the solid oral dosage form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof is a capsule or a tablet.


Preferably the solid oral dosage form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof comprises a blend of one or more diluent.


Preferably the solid oral dosage form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof is a tablet wherein the blend of one or more diluent comprises microcrystalline cellulose.


Preferably the solid oral dosage form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof comprises a blend of one or more diluent with the proviso that the dosage form does not contain lactose monohydrate.


Preferably the solid oral dosage form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof is a capsule and the capsule shell comprises a constituent selected from gelatin and hydroxypropyl methylcellulose.


Preferably the solid oral dosage form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof is for administration in combination with a serotonin modulator, wherein the serotonin modulator is preferably a selective serotonin reuptake inhibitor (SSRI), more preferably selected from Citalopram, Escitalopram, Paroxetine, Fluoxetine, Fluvoxamine, Sertraline, Desvenlafaxine, Duloxetine, Levomilnacipran, Milnacipran. Tofenacin, Venlafaxine, Vilazodone, Vortioxetine, Etoperidone, Nefazodone, and Trazodone, or a pharmaceutically acceptable salt thereof, and most preferably selected from Citalopram, Escitalopram. Paroxetine, Sertraline, Duloxetine, and Venlafaxine, or a pharmaceutically acceptable salt thereof.


Preferably the solid oral dosage form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof and the serotonin modulator are for administration simultaneously, sequentially or separately.


In preferred embodiments of the present invention the solid oral dosage form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof is for treating a patient wherein the patient has experienced one or more manic episode or hypomanic episode, or is suffering a manic episode or hypomanic episode, or is at risk of suffering one or more manic episode or hypomanic episode.


In preferred embodiments of the present invention the solid oral dosage form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof is for use in treating a patient suffering from bipolar depression type 1, bipolar depression type 2, bipolar depression, or obsessive-compulsive disorder comorbid with depression.


In preferred embodiments of the present invention the solid oral dosage form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof is for use as a first-line therapy.


In preferred embodiments of the present invention the solid oral dosage form comprises 2R,6R-hydroxynorketamine L-(+)-tartrate.


Patients at Risk of Mania

In a particularly preferred embodiment, the solid oral dosage forms of the present invention are useful in the treatment of a depressive disorder in a patient who has experienced one or more manic episode or hypomanic episode. In an alternative embodiment, the solid oral dosage forms of the present invention are for use in the treatment of a depressive disorder in a patient who is suffering or is at risk of suffering one or more manic episode or hypomanic episode. Such patients include, but are not limited to, patients suffering from major depressive disorder or bipolar disorder.


As used herein, the term ‘manic episode’ is defined as a distinct period of abnormally and persistently elevated, expansive, or irritable mood and abnormally and persistently increased activity or energy, lasting at least 1 week and present most of the day, nearly every day, or any duration if hospitalization is necessary. As used herein the term ‘hypomanic episode’ is defined as a mood state characterized by persistent disinhibition and pervasive elevated euphoria with or without irritable mood for a period of four days or more.


Ketamine induced mania has been reported (Ricke A K, Snook R J, Anand A. Induction of prolonged mania during ketamine therapy for reflex sympathetic dystrophy. Biol Psychiatry, 2011; 70(4):p13-14). It has now been discovered that compounds according to the present invention, in particular 2R,6R-hydroxynorketamine, exhibit neither dopamine receptor agonism, nor activity at dopamine transporters. Accordingly, an aspect of the present invention provides a metabolite of ketamine, or a pharmaceutically acceptable salt thereof, alone or in combination with serotonin modulators described herein, for use in a patient who has experienced one or more manic episode or hypomanic episode, or in a patient who is suffering or at risk of suffering one or more manic episode or hypomanic episode. In a preferred embodiment, the metabolite of ketamine is 2R,6R-hydroxynorketamine, or a pharmaceutically acceptable salt thereof, or a prodrug thereof.


First-Line Therapy

The present invention provides a metabolite of ketamine, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, for use in the treatment of a depressive disorder in a patient, wherein the compound is for use as a first-line therapy.


Intravenous infusions of 0.5 mg/kg of ketamine hydrochloride administered over a period of 40 mins are known to exhibit potent and rapid anti-depressant effects in treatment-resistant depression patients, reported, for example, in Murrough et al; Antidepressant Efficacy of Ketamine in Treatment-Resistant Major Depression: A Two-Site Randomized Controlled Trial; Am J Psychiatry. 1 Oct. 2013; 170(10): 1134-1142.


However neither metabolites of ketamine, nor prodrugs of ketamine or its metabolites have been investigated as a first-line therapy for the treatment of a depressive disorder in a patient. The present invention provides, for the first time, a first-line therapy for use in the treatment of a patient suffering from a depressive disorder which overcomes drawbacks associated with existing first-line therapies.


Moreover, the use of ketamine hydrochloride in the treatment of mental health is limited by the dosage form in which it is typically provided. Ketamine hydrochloride is approved in a form administrable only by intravenous infusion. This renders it inconvenient and/or inappropriate for use as a first-line therapy: it is preferable to be able to administer first-line therapy in an out-patient setting. Accordingly, the present invention provides a solid oral dosage form comprising a compound selected from ketamine, a metabolite of ketamine, a prodrug of ketamine, a prodrug of a metabolite of ketamine, or a pharmaceutically acceptable salt thereof for use in the treatment of a depressive disorder in a patient, wherein the solid oral dosage form is for use as a first-line therapy.


EXAMPLES
Example 1: Formation of a Crystalline Solid Form of 2R,6R-Hydroxynorketamine

Racemic cis-hydroxynorketamine was synthesized according to the synthesis provided in Scheme 5.




embedded image


Crude obtained from Scheme 5 was purified by flash chromatography to produce cis-6-hydroxynorketamine freebase (racemic mixture of 2R,6R-hydroxynorketamine and 2S,6S-hydroxynorketamine). The racemic form of cis-6-hydroxynorketamine forms an amorphous oil at ambient conditions.


Racemic cis-hydroxynorketamine was dissolved to 25 mg/mL in methanol/IPA (1:1) and was then purified by SFC. Combined fractions of each of 2R,6R-hydroxynorketamine and 2S,6S-hydroxynorketamine were then evaporated to near dryness using a rotary evaporator, transferred into final vessels with DCM, which was removed under a stream of compressed air at 40 C before being stored in a vacuum oven at 40 C and 5 mbar for 16 hours to afford each of 2R,6R-hydroxynorketamine and 2S,6S-hydroxynorketamine. Both 2R,6R-hydroxynorketamine and 2S,6S-hydroxynorketamine form an amorphous oil at ambient conditions.


In an alternative separation method, a crystalline form of 2R,6R-hydroxynorketamine L-(+)-tartrate was obtained by treating racemic cis-hydroxynorketamine freebase with L-(+)-tartaric acid in methanol followed by recrystallization from acetone.


Example 2: Preparation of O-Phosphate Prodrug of 2R,6R-Hydroxynorketamine

(1R,3R)-3-amino-3-(2-chlorophenyl)-2-oxocyclohexyl dihydrogen phosphate (CI) was prepared following the synthesis of Scheme 6.




embedded image


The free acid form of CI was isolated as a crystalline salt, and the potassium salt was obtained, also as a crystalline salt, following neutralisation for 1 hour with aqueous KHCO3, followed by lyophilisation.


Structure of CI confirmed by 1H and 31P NMR: 1H-NMR (301 MHz, METHANOL-D4) δ 7.69-7.72 (m, 1H), 7.29-7.43 (m, 3H), 4.69-4.77 (m, 1H), 2.91-2.97 (m, 1H), 2.52-2.61 (m, 1H), 1.57-1.82 (m, 4H), (NH2 appears to be under H2O signal ˜4.85 ppm); 31P-NMR (162 MHz. METHANOL-D4) δ 1.87, 0.50.


Example 3: Solid Oral Dosage Formulations of 2R,6R-Hydroxynorketamine












Formulation (a)


2R,6R-HNK Capsule









Ingredient/Component
Unit Quantity
Reference





2R,6R-hydroxynorketamine (+)-tartrate
65 mg (40 mg
In-house



as free base)










Dicalcium phosphate
648
mg
Ph. Eur


Sodium lauryl sulphate
15
mg
Ph. Eur


Colloidal silica
7
mg
Ph. Eur









Coni-Snap ® Size 00 hard gelatin capsule
One
In-house


or Vcap ® Size 00 HPMC capsule











Total
735
mg



















Formulation (b)


2R,6R-HNK Capsule










Unit



Ingredient/Component
Quantity
Reference





2R,6R-hydroxynorketamine (+)-tartrate
65 mg (40 mg
In-house



as free base)










Dicalcium phosphate
577
mg
Ph. Eur


Starch
58
mg
Ph. Eur


Talc
7
mg
Ph. Eur


Hypromellose
28
mg
Ph. Eur









Coni-Snap ® Size 00 hard gelatin capsule
One
In-house


or Vcap ® Size 00 HPMC capsule











Total
735
mg



















Formulation (c)


2R,6R-HNK Capsule










Unit



Ingredient/Component
Quantity
Reference





2R,6R-hydroxynorketamine as free base
65 mg (40 mg
In-house



as free base)










Dicalcium phosphate
407
mg
Ph. Eur


Microcrystalline cellulose
190
mg
Ph. Eur


Crospovidone
59
mg
Ph. Eur


Colloidal silica
7
mg
Ph. Eur


Sodium lauryl sulphate
7
mg
Ph. Eur









Coni-Snap ® Size 00 hard gelatin capsule
One
In-house


or Vcap ® Size 00 HPMC capsule











Total
735
mg



















Formulation (d)


2R,6R-HNK Capsule










Unit



Ingredient/Component
Quantity
Reference





2R,6R-hydroxynorketamine as free base
65 mg (40 mg
In-house



as free base)










Starch
670
mg
Ph. Eur









Com-Snap ® Size 00 hard gelatin capsule
One
In-house


or Vcap ® Size 00 HPMC capsule











Total
735
mg









Example 4: Analysis of Composition of Solid Oral Dosage Forms

Differential Scanning Calorimetry (DSC) analysis was performed to determine the preferred diluent blend for formulating solid oral dosage forms of the present invention. The results indicate that, despite its widespread use in preparation of solid oral dosage forms, lactose monohydrate is incompatible with 2R,6R-hydroxynorketamine (see FIG. 1). These data demonstrated compatibility of 2R,6R-hydroxynorketamine with dicalcium phosphate and microcrystalline cellulose.


Analysis also demonstrated compatibility of 2R,6R-hydroxynorketamine with gelatin and hydroxypropyl methylcellulose capsule shells.


Example 5: Lack of Drug-Drug Interactions (DDIs) Between Ketamine Metabolites and Serotonin Modulators Such as SSRIs
Example 5a

Potential interactions between ketamine metabolites of the present invention with the serotonergic system were investigated to determine whether co-administration with serotonin modulators, especially selective serotonin-reuptake inhibitors (SSRIs) affects the risk of serotonin syndrome.


Serotonin syndrome describes a specific set of symptoms resulting from an excess of serotonin within the central and peripheral nervous systems. Symptoms can vary largely, and range in severity, including possible increases in temperature, agitation and tremor, through to arrhythmias and seizure.


SSRIs work by inhibiting the reuptake of neuronal serotonin, thus increasing the synaptic concentration of the neurotransmitter and therefore activation of 5HT receptors. No one 5HT receptor is thought to be responsible for the development of serotonin syndrome, although much focus has highlighted the importance of 5HT2A receptors in the development of this serotonin related toxicity. The risk of serotonin syndrome is increased if SSRIs are taken in combination with another drug that increases extracellular serotonin, or that additively potentiates the 5HT receptors involved in its development.


2R,6R-hydroxynorketamine was investigated for binding to monoamine oxidases A and B, monoamine transporters, as well as a variety of 5HT receptors. It was found that 2R,6R-hydroxynorketamine does not interact with transporters or enzymes that directly affect synaptic monoamine concentration. In addition, studies revealed that 2R,6R-hydroxynorketamine does not bind 5HT1A, 5HT1B, 5HT2A, 5HT2B or 5HT2C receptor subtypes, indicating no risk of additive, or indeed competitive, effects if given in combination with SSRIs.











TABLE 9






% Inhibition of Control
% of Control


Assay
Specific Binding
Specific Binding

















5-HT1A
5
97.9


5-HT1B
−2
102.7


5-HT2A
0
103.8


5-HT2B
5
100.9


5-HT2C
10
91.1


norepinephrine transporter
−16
117.6


dopamine transporter
11
87.2


5-HT transporter
2
102.0


5-HT1, Non-Selective
−9
105.4


MAO-A
−5
97.7


MAO-B
−1
100.4









An absence of interaction between 2R,6R-hydroxynorketamine and 5HT1 receptors is also of note. Rare postmarketing reports have described patients with weakness, hyperreflexia, and incoordination following the use of an SSRI and the 5HT1 agonist, sumatriptan. The possibility of such interactions should also be considered if other 5HT1 agonists are to be used in combination with SSRIs. As 2R,6R-hydroxynorketamine does not bind 5HT1 receptors, this potential drug-drug interaction (DDI) is avoided.


Example 5b

The p-gycloprotein-1 (p-gp) transporter mediates the efflux of drugs from cells. It is widely expressed throughout the body, including the luminal membrane of the small intestine, apical membranes of hepatocytes and kidney proximal tube epithelia, as well as the blood brain barrier (BBB). It is through its role at the BBB that p-gp plays a key role in limiting drug entry to the central nervous system. Many of the most commonly used antidepressants have been shown to be substrates of p-gp; including: amitriptyline, citalopram, desipramine, doxepine, fluoxetine, fluvoxamine, imipramine, nortriptyline, paroxetine, trimipramine, venlafaxine.


2R,6R-hydroxynorketamine was tested using a bidirectional transcellular transport assay (Caco-2). No active efflux of 2R,6R-hydroxynorketamine was observed (efflux ratio <2, Table 10) indicating that 2R,6R-hydroxynorketamine is not a substrate of either p-gp or Breast Cancer Resistance Protein (BCRP) efflux transporters. In contrast to the effect on p-gp substrate, talinolol, the transporter inhibitor, elacridar, did not affect the efflux ratio of 2R,6R-hydroxynorketamine (2R,6R-hydroxynorketamine efflux ratio=1.11, +elacridar=1.15; talinolol efflux ratio=37, +elacridar=0.882).


This finding eliminates the possibility that combination therapy of 2R,6R-hydroxynorketamine and an SSRI described above, would induce competition for the p-gp transporter. Such a finding is relevant for the prevention of DDIs involving increased concentrations of either compound within the brain.


Many SSRIs also act as inhibitors of p-gp transporters, with sertraline and paroxetine displaying an IC50 similar to that of established p-gp inhibitor quinidine. As 2R,6R-hydroxynorketamine is not a substrate of p-gp, combination therapy with these SSRIs will not induce DDIs based on a reduction in 2R,6R-hydroxynorketamine efflux from the brain.













TABLE 10











Efflux





Ratio



Direction = A2B
Direction = B2A
(Mean



















Papp




Papp




Papp



(10−6 cms−1)




(10−6 cms−1)




B2A/Mean




















Test
Replicate
Replicate
Mean Papp


Mean %
Replicate
Replicate
Mean Papp


Mean %
Papp


compound
1
2
(10−6 cms−1)
SD
n
Recovery
1
2
(10−6 cms−1)
SD
n
Recovery
A2B)























2R,6R-
49.1
42.3
45.7
4.8
2
98.9
52.8
48.5
50.7
3
2
103
1.11


hydroxy-


norketamine


talinolol
0.28
0.32
0.3
0.03
2
86.5
10.7
11.5
11.1
0.5
2
91.7
37.0









Example 5c

While the risk of DDIs due to displacement from plasma binding proteins (PPBs) is relatively low, it should be considered for drugs which display a high proportion PPB (fraction unbound (fu)<1%).


Many commonly used SSRIs are highly protein bound (fluoxetine: 94%; paroxetine: 95%; sertraline: 98%), with advice that co-administration with another drug that also displays high protein binding may result in adverse effects due to an increase in plasma levels of either unbound drug.


The fu was investigated for 2R,6R-hydroxynorketamine using a 100% plasma from 4 test species. 2R,6R-hydroxynorketamine was found to display low PPB in all species tested (mouse, rat, dog and human; FIG. 2).


As a small molecule with low levels of PPB, 2R,6R-hydroxynorketamine does not risk displacement of SSRIs.


Example 5d

Cytochrome P450s (CYPs) are a family of enzymes that play a primary role in the metabolism of a wide variety of drugs. 2R,6R-hydroxynorketamine was investigated for its potential to inhibit the action of 7 key CYP enzymes using an inhibition assay measuring reduction in the formation of metabolites compared to control. These data were then used to calculate an IC50 value.


2R,6R-hydroxynorketamine did not inhibit cytochrome P450 isoforms: CYP2B6, CYP2C8, CYP2C9, CYP2C19 and CYP2D6 (Table 11). 2R,6R-hydroxynorketamine displayed inhibition of CYP1A2, with an IC50 of ˜100M (Table 11). 2R,6R-hydroxynorketamine was found to inhibit the action of CYP3A4 on both substrate groups, midazolam and testosterone, with an IC50 of 69 μM and 81 μM respectively (Table 11).











TABLE 11









% Inhibition














Cytochrome P450
0 μM
0.4 μM
1 μM
4 μM
10 μM
40 μM
100 μM

















CYP2D6
0
3.16
6.65
11.5
9.41
11.2
5.73


CYP2C19
0
−2.01
−1.66
1.85
6.69
8.25
16.1


CYP2C8
0
5.63
4.91
12.5
8.33
8.35
2.08


CYP2C9
0
4.59
5.92
5.80
11.9
−0.113
−1.33


CYP2B6
0
6.03
10.1
10.6
14.4
14.8
16.7


CYP1A2
0
7.55
8.80
11.7
19.3
24.8
48.6


CYP3A4,
0
11.5
8.44
15.7
25.7
43.7
57.5


Substrate = midazolam


CYP3A4,
0
1.62
−0.509
7.04
12.4
35.2
53.4


Substrate = testosterone









CYP2D6 is the major enzyme involved in the metabolism of the majority of commonly used antidepressants, including fluoxetine, paroxetine and venlafaxine. 2R,6R-hydroxynorketamine does not cause inhibition of this CYP enzyme, evading potential DDIs due to decreased metabolism of such SSRIs.


The free (unbound) concentration of 2R,6R-hydroxynorketamine producing pharmacodynamics activity in the brain is estimated to be approximately 10 μM. This concentration has been demonstrated to be sufficient in causing an increase in field excitatory postsynaptic potentials (fEPSPs) in in vitro electrophysiological experiments, a finding is dependent on the potentiation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)-mediated currents. A free concentration 10 μM 2R,6R-hydroxynorketamine is sufficiently remote from the IC50 values for CYP1A2 and CYP3A4 to avoid DDI risk.


Example 5e

Following phase I metabolism by CYP enzymes, many) drugs undergo phase II metabolism, including glucuronidation, a process completed by the uridine glucuronyl transferase (UGT) family.


The SSRI sertraline is glucuronidated to sertraline N-carbamoyl glucuronide by a variety of UGT enzymes, with the greatest activity observed with UGT2B7. 2R,6R-hydroxynorketamine was therefore investigated for its potential to inhibit this isozyme, along with that of the major UGT isozyme, UGT1A1, using a UGT inhibition assay.


2R,6R-hydroxynorketamine does not inhibit UGT2B7 or UGT1A1 (Table 12, FIG. 3), supporting the possibility of its use in combination with sertraline.









TABLE 12







UGT Inhibition: Summary of IC50 Values (μM)










Inhibitor
Test Concentration
UGT1A1
UGT2B7













2R-6R-
0.1 μM-100 μM
>100
>100


Hydroxynorketamine


Hydrochloride


Atazanavir
0.006 μM-6 μM  
0.153


Diclofenac
 2.7 μM-2000 μM

16.4









Example 6: Demonstration that 2R,6R-Hydroxynorketamine is Orally Bioavailable

The solubility and permeability of 2R,6R-hydroxynorketamine was investigated using turbidimetric aqueous solubility and Caco-2 permeability assays respectively. 2R,6R-hydroxynorketamine was found to display high solubility (estimated precipitation range lower bound: 100 μM upper bound: >100 μM) and permeability (Table 13).













TABLE 13











Efflux





Ratio





(Mean



Direction = A2B
Direction = B2A
Papp



















Papp




Papp




B2A/



(10−6 cms−1)




(10−6 cms−1)




Mean




















Test
Replicate
Replicate
Mean Papp


Mean %
Replicate
Replicate
Mean Papp


Mean %
Papp


compound
1
2
(10−6 cms−1)
SD
n
Recovery
1
2
(10−6 cms−1)
SD
n
Recovery
A2B)























2R,6R-
49.1
42.3
45.7
4.8
2
98.9
52.8
48.5
50.7
3
2
103
1.11


hydroxy-


norketamine


atenolol
0.38
0.33
0.35
0.04
2
97.7
0.58
0.59
0.58
0.008
2
97.3
1.66


propranolol
26.0
27.4
26.7
1.01
2
71.1
27.0
26.2
26.6
0.55
2
82.8
0.997









Propranolol has a known human absorption of 90%. Mean apparent permeability coefficient (Papp) values for 2R,6R-hydroxynorketamine are greater than those seen for propranolol controls, suggestive of high human absorption.


Both the solubility and permeability of a compound play a major role in its suitability for oral administration. The data reported presently support 2R,6R-hydroxynorketamine as a suitable candidate for oral administration.


Following absorption, orally administered drugs undergo presystemic metabolism. This first-pass effect contributes one of largest factors affecting the oral bioavailability of a drug.


2R,6R-hydroxynorketamine was investigated using an in vitro hepatocyte stability assay, during which 2R,6R-hydroxynorketamine was incubated with cryopreserved hepatocytes from 3 test species. 2R,6R-hydroxynorketamine displayed a negative intrinsic clearance value (Clint) in a human hepatocyte stability assay, indicating it does not undergo hepatic clearance in humans (Table 14, FIG. 4). These data, along with metabolite profiling revealed that 2R,6R-hydroxynorketamine does not undergo phase I metabolism by CYP enzymes, but is glucuronidated and excreted via the renal system.













TABLE 14





Species
CLint (gL/min/106 cells)
SE CLint
T1/2 (min)
n



















Rat
55.2
4.24
25.1
5


Dog
4.72
1.15
294
6


Human
−1.07
0.293
−1300
6









The absence of phase I metabolism is supportive of 2R,6R-hydroxynorketamine being suitable for oral administration.

Claims
  • 1. A method of treating a depressive disorder in a patient comprising: administering to said patient a solid oral dosage form comprising 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof, or a prodrug of 2R,6R-hydroxynorketamine having a structure of Formula Ib, or a pharmaceutically acceptable salt thereof
  • 2. The method of claim 1 wherein R1 is selected from PO3H2, SO3H, CO2H, and R5, wherein R5 is selected from
  • 3. The method of claim 1 wherein R4 or R5 has the same stereochemistry as the corresponding L-amino acid.
  • 4. The method of claim 1 wherein R1 is PO3H2.
  • 5. The method of claim 1 wherein the solid oral dosage form comprises between 10 mg and 100 mg of 2R,6R-hydroxynorketamine, or the equivalent thereof.
  • 6. The method of claim 1 wherein the solid oral dosage form comprises a crystalline form of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof, or a crystalline form of a prodrug of 2R,6R-hydroxynorketamine or a pharmaceutically acceptable salt thereof.
  • 7. (canceled)
  • 8. The method of claim 1 wherein the solid oral dosage form is a capsule or a tablet.
  • 9. The method of claim 1 wherein the solid oral dosage form comprise a blend of one or more diluent.
  • 10. The method of claim 9 wherein the solid oral dosage form is a tablet and wherein the blend of one or more diluent comprises microcrystalline cellulose.
  • 11. The method of claim 10 wherein the blend of one or more diluent does not contain lactose monohydrate.
  • 12. The method of claim 1 wherein the dosage form is a capsule and the capsule shell comprises a constituent selected from gelatin and hydroxypropyl methylcellulose.
  • 13.-14. (canceled)
  • 15. The wherein said compound and said serotonin modulator are administered simultaneously, sequentially, or separately.
  • 16. The method of claim 1 wherein said serotonin modulator is selected from Citalopram, Escitalopram, Paroxetine, Fluoxetine, Fluvoxamine, Sertraline, Desvenlafaxine, Duloxetine, Levomilnacipran, Milnacipran, Tofenacin, Venlafaxine, Vilazodone, Vortioxetine, Etoperidone, Nefazodone, and Trazodone, or a pharmaceutically acceptable salt thereof.
  • 17. The method of claim 16 wherein the serotonin modulator is selected from Citalopram, Escitalopram, Paroxetine, Sertraline, Duloxetine, and Venlafaxine, or a pharmaceutically acceptable salt thereof.
  • 18. The method of claim 1 wherein the patient has experienced one or more manic episode or hypomanic episode, is suffering a manic episode or hypomanic episode, or is at risk of suffering one or more manic episode or hypomanic episode.
  • 19. The method of claim 1 wherein the patient is suffering from bipolar depression type 1, bipolar depression type 2, bipolar depression, or obsessive-compulsive disorder comorbid with depression.
  • 20. The method of claim 1 wherein the solid oral dosage form is for use as a first-line therapy.
  • 21. The method of claim 1 wherein the solid oral dosage form comprises 2R,6R-hydroxynorketamine L-(+)-tartrate.
  • 22. The method of claim 1 wherein said pharmaceutically acceptable salt is a tartrate salt.
  • 23. The method of claim 22 wherein the solid oral dosage form comprise 2R,6R-hydroxynorketamine L-(+)-tartrate.
Priority Claims (2)
Number Date Country Kind
1609790.9 Jun 2016 GB national
1620690.6 Dec 2016 GB national
PCT Information
Filing Document Filing Date Country Kind
PCT/GB2017/051618 6/5/2017 WO 00