Novel catecholamine prodrugs for the treatment of parkinson's disease

文档序号:327753 发布日期:2021-11-30 浏览:25次 中文

阅读说明:本技术 用于治疗帕金森病的新儿茶酚胺前药 (Novel catecholamine prodrugs for the treatment of parkinson's disease ) 是由 M·约尔根森 E·阿希 M·祖尔 K.G.詹森 A·皮施尔 B·邦-安德森 于 2020-05-19 设计创作,主要内容包括:本发明提供了用于治疗神经退行性疾病和障碍的作为儿茶酚胺的前药的具有式(Id)的化合物。本发明还提供了包含本发明化合物的药物组合物,以及使用本发明化合物治疗神经退行性或神经精神性疾病和障碍,特别是帕金森病的方法。(The present invention provides compounds having formula (Id) as prodrugs of catecholamines for use in the treatment of neurodegenerative diseases and disorders. The invention also provides pharmaceutical compositions comprising the compounds of the invention and methods of using the compounds of the invention to treat neurodegenerative or neuropsychiatric diseases and disorders, particularly Parkinson's disease.)

1. A compound according to formula (Id)

Wherein R1, R2 and R3 are according to a) or b) below:

a) r1 and R2 are each independently selected from H, C1-C6Alkyl, benzyl and the following substituents (i), and R3 is absent,

wherein the points of attachment are indicated by the reference numerals,

provided that R1 and R2 cannot both be H;

or

b) R1 and R2 are both H, and R3 is substituent (ii), N is positively charged, and

wherein indicates the attachment point;

and wherein R4 and R5 are each independently selected from H, C1-C6Alkyl, phenyl, benzyl, C3-C6Cycloalkyl and quilt C3-C6Cycloalkyl-substituted methyl;

or a pharmaceutically acceptable salt thereof.

2. The compound according to claim 1, wherein the compound is of formula (Ie),

wherein

R1 and R2 are each independently selected from H, C1-C6Alkyl, benzyl and substituent (i);

provided that R1 and R2 cannot both be H;

wherein indicates the attachment point; and wherein

R4 and R5 are each independently selected from H, C1-C6Alkyl, phenyl, benzyl, C3-C6Cycloalkyl and quilt C3-C6Cycloalkyl-substituted methyl;

or a pharmaceutically acceptable salt thereof.

3. The compound according to claim 1, or a pharmaceutically acceptable salt thereof,

r1 is H and R2 is substituent (i) and R3 is absent.

4. The compound according to claim 1, or a pharmaceutically acceptable salt thereof,

r1 is substituent (i) and R2 is H and R3 is absent.

5. The compound according to claim 1, or a pharmaceutically acceptable salt thereof,

r1 and R2 are both represented by substituent (i), and R3 is absent.

6. The compound according to claim 1, or a pharmaceutically acceptable salt thereof,

r1 and R2 are both H; and is

R3 is substituent (ii).

7. The compound according to any one of claims 1-6, or a pharmaceutically acceptable salt thereof,

r4 and R5 are both H.

8. The compound according to any one of claims 1-6, or a pharmaceutically acceptable salt thereof,

at least one of R4 and R5 is benzyl.

9. The compound of claim 1, wherein the compound is selected from the group consisting of:

(4aR,10aR) -6, 7-bis (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinoline;

(4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

(4aR,10aR) -7-methoxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

benzyl ((4aR,10aR) -6- ((bis (benzyloxy) phosphoryl) oxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-7-yl) hydrogen phosphate;

benzyl ((4aR,10aR) -7- ((bis (benzyloxy) phosphoryl) oxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl) hydrogen phosphate;

(4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6, 7-diylbis (dihydrogen phosphate);

benzyl ((4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl) hydrogen phosphate;

(4aR,10aR) -7-hydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl dihydrogenphosphate; and

((1S,4aR,10aR) -6, 7-dihydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methylphosphonate;

or a pharmaceutically acceptable salt of any of these compounds.

10. The compound of claim 1, wherein the compound is selected from the group consisting of:

compound (2): (4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

compound (3): (4aR,10aR) -7-methoxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

compound (6): (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6, 7-diylbis (dihydrogen phosphate);

compound (8): (4aR,10aR) -7-hydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl dihydrogenphosphate; and

compound (9): ((1S,4aR,10aR) -6, 7-dihydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methylphosphonate;

or a pharmaceutically acceptable salt of any of these compounds.

11. The compound of claim 1, wherein the compound is selected from the group consisting of:

compound (2): (4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

compound (3): (4aR,10aR) -7-methoxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

or a pharmaceutically acceptable salt of any of these compounds.

12. The compound of claim 1, wherein the compound is selected from the group consisting of:

compound (6): (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6, 7-diylbis (dihydrogen phosphate);

compound (8): (4aR,10aR) -7-hydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl dihydrogenphosphate; and

compound (9): ((1S,4aR,10aR) -6, 7-dihydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methylphosphonate;

or a pharmaceutically acceptable salt of any of these compounds.

13. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of claims 1-12, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers or diluents.

14. A compound according to any one of claims 1-12, or a pharmaceutically acceptable salt thereof, for use as a medicament.

15. A compound according to any one of claims 1-12, or a pharmaceutically acceptable salt thereof, for use in the treatment of a neurodegenerative disease or disorder, such as parkinson's disease, huntington's disease, restless leg syndrome, or alzheimer's disease; or for the treatment of neuropsychiatric diseases or disorders such as schizophrenia, attention deficit hyperactivity disorder or drug addiction.

16. A method for treating a disease or disorder comprising: neurodegenerative diseases or disorders, such as parkinson's disease, huntington's disease, restless leg syndrome, or alzheimer's disease; or a neuropsychiatric disease or disorder, such as schizophrenia, attention deficit hyperactivity disorder or drug addiction; the method comprising administering to a patient in need thereof a therapeutically effective amount of a compound according to any one of claims 1-12, or a pharmaceutically acceptable salt thereof.

17. Use of a compound according to any one of claims 1-12, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a neurodegenerative disease or disorder, such as parkinson's disease, huntington's disease, restless leg syndrome, or alzheimer's disease; or for the treatment of neuropsychiatric diseases or disorders such as schizophrenia, attention deficit hyperactivity disorder or drug addiction.

Technical Field

The present invention provides compounds that are phosphate esters, phosphonooxymethyl derivatives and ether derivative prodrugs of dopamine agonists (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydro-benzo [ g ] quinoline-6, 7-diol, and their use in parkinson's disease and/or other conditions for which treatment with dopamine agonists is therapeutically beneficial, such as, but not limited to, restless leg syndrome, huntington's disease and alzheimer's disease; and also neuropsychiatric diseases and disorders such as, but not limited to, schizophrenia, attention deficit hyperactivity disorder and drug addiction. The invention also provides a pharmaceutical composition containing the compound.

Background

Parkinson's Disease (PD) is a common neurodegenerative disorder that becomes more common with age and affects an estimated seven to ten million people worldwide. Parkinson's disease is a multifaceted disease characterized by both motor symptoms and non-motor symptoms. Motor symptoms include resting tremor (tremor), bradykinesia/akinesia (slowness and difficulty in movement), muscle rigidity, postural instability and gait disturbance; while non-motor symptoms include neuropsychiatric disorders (e.g., depression, psychotic symptoms, anxiety, apathy, mild cognitive impairment and dementia) as well as autonomic dysfunction and sleep disorders (Poewe et al, Nature Review (2017) Vol. 3, items 17013: 1-21).

A key hallmark of the pathophysiology of parkinson's disease is the loss of the pigment dopaminergic neurons in the substantia nigra pars compacta, which supply dopaminergic innervation to the striatum and other brain regions. Such progressive neurodegeneration leads to a reduction in dopamine striatal levels, which ultimately leads to a series of changes in the basal nuclear circuit, ultimately leading to the development of the four major motor features of parkinson's disease. The primary target of dopamine in the striatum consists of medium spiny gabaergic neurons (MSNs) that selectively express either the D1 or D2 receptors awaiting localization projections. Gamma-aminobutyric acid (GABA) projecting to the lateral globus pallidus (also known as striatum-globus pallidus 'indirect pathway') expresses the D2 receptor (MSN-2) MSN; whereas gaba, which projects to the substantia nigra pars compacta and lateral globus pallidus (also known as the striatum-substantia nigra 'direct pathway'), expresses the D1 receptor (MSN-1) by MSN. Depletion of dopamine due to neuronal loss leads to unbalanced activity of both pathways, leading to a significant decrease in thalamic and cortical export activity, and ultimately dyskinesias (Gerfen et al, Science [ Science ] (1990)250: 1429-32; Delong, (1990) Trends in Neuroscience [ Neuroscience trend ]13: 281-5; Alexander et Crutcher, (1990) Trends in Neuroscience [ Neuroscience trend ]13: 266-71; and for reviews, Poewe et al, Nature Review [ Nature Review ] (2017) Vol.3, items 17013: 1-21).

The most effective therapeutic strategies available for patients with parkinson's disease and aimed at controlling motor symptoms are mainly indirect and direct dopamine agonists. Classical and gold standard treatment regimens include chronic oral ingestion of L-3, 4-dihydroxyphenylalanine (L-DOPA), which decarboxylates in the brain to form dopamine. Other methods consist in administering dopamine receptor agonists (such as apomorphine, which acts on both the D1 and D2 receptor subtypes, or pramipexole, ropinirole, etc., which is directed primarily against the D2 receptor subtype). The best motor relief obtained with L-DOPA and apomorphine is due to their activation of both the D1 and D2 receptor subtypes and global rebalancing of the indirect-direct pathway (i.e., while D2 agonists only reverse the indirect pathway barrier).

L-DOPA and apomorphine have the structures depicted below and are currently the most potent PD drugs in clinical applications.

L-DOPA is a prodrug of dopamine and remains the most effective drug in the treatment of motor parkinson's disease. However, after several years of treatment (i.e. the honey month period), complications arise due to the intrinsic progression of the disease (sustained loss of dopaminergic neurons) and the poor Pharmacokinetic (PK) profile of L-DOPA. Those complications include: 1) dyskinesias, which are abnormal involuntary movements that occur during the optimal "duration effect" of a drug; and 2) fluctuations during which the positive effect of L-DOPA disappears and symptoms reappear or worsen (Sprenger and Poewe, CNS Drugs [ CNS Drugs ] (2013),27: 259-272).

Direct dopamine receptor agonists activate dopamine autoreceptors as well as postsynaptic dopamine receptors located on medium spiny neurons MSN-1 and MSN-2. Apomorphine belongs to a class of dopamine agonists that have a1, 2-dihydroxybenzene (catechol) moiety. Catecholamines generally have low or no oral bioavailability when combined with phenethylamine motifs, as is the case with apomorphine. Apomorphine is used clinically in the treatment of PD, although in non-oral delivery (typically intermittent subcutaneous administration via a pump or continuous daily parenteral infusion). For apomorphine, animal studies have shown that transdermal delivery or implants may provide possible forms of administration. However, when studying apomorphine delivery from implants in monkeys (Bibbiani et al, Chase Experimental Neurology (2005),192:73-78), it was found that in most cases animals had to be treated with the immunosuppressant dexamethasone in order to prevent local irritation and other complications after implant surgery. Alternative delivery strategies for apomorphine therapy in PD have been extensively developed, such as inhalation and sublingual formulations (see, e.g., Grosset et al, Acta Neurol Scand. [ Scandinavian neurology bulletin ] (2013),128: 166-. However, these efforts have not yet been used in clinical applications for the treatment of PD.

An alternative to non-oral formulations of catecholamines involves the use of prodrugs that mask free catechol hydroxyl groups to enable oral administration. However, a known problem associated with the development of prodrugs for clinical use is the difficulty associated with predicting conversion to the parent compound in humans.

Different ester prodrugs of catecholamines have been reported in the literature, such as enteric coated N-propyl-Noraporphine (NPA) and the mono-pivaloyl ester of apomorphine (see e.g. WO 02/100377) for duodenal delivery and the D1-like agonist aclonide (diacetyl prodrug of A-86929) (Giardina and Williams; CNS Drug Reviews [ CNS Drug review ] (2001), Vol.7 (3): 305-316). In humans, upon oral administration, acloritide undergoes extensive hepatic first pass metabolism and, as a result, has low oral bioavailability (about 4%). Intravenous (IV) aldrin has an anti-Parkinson efficacy comparable to L-DOPA in PD patients (Giardina and Williams; CNS Drug Reviews [ CNS Drug review ] (2001), Vol.7 (3): 305-.

In addition to ester prodrugs of catecholamines, alternative prodrug approaches involve masking the two catechol hydroxyl groups to the corresponding methylenedioxy derivatives, or to diacetal derivatives. For example, it has been described in Campbell et al, Neuropharmacology (1982); 21(10) 953-961 and US 4543256, WO 2009/026934 and WO 2009/026935 describe the principle of this prodrug.

For catecholamine prodrugs, yet another proposed approach is the formation of enone derivatives, as proposed in, for example, WO 2001/078713 and Liu et al, Bioorganic med. For further examples of catecholamine prodrugs, see, e.g., Sozio et al, exp. 7(5):385-406.

The compound (4aR,10aR) -1-n-propyl-1, 2,3,4,4a,5,10,10 a-octahydro-benzo [ g ] quinoline-6, 7-diol described as compound (I) below is disclosed in WO 2009/026934. The trans isomer was previously disclosed in Liu et al, J.Med.chem. [ journal of medicinal chemistry ] (2006),49: 1494-. The racemate was first disclosed in Cannon et al, J.Heterocyclic Chem. [ J.Heterocycli Chem. (1980); 1633 and 1636.

Compound (I) is a dopamine agonist with mixed D1 and D2 activity. Three prodrug derivatives of compound (I) are known in the art.

Liu et al, J.Med.chem. [ J.Pharmacol. (2006),49:1494- & ltI & gt 1498 & Liu et al, Bioorganic Med.chem. [ bio-organic chemistry & medicinal chemistry ] (2008),16:3438- & ltJ.3444 & gt disclose the enone derivatives of formula (Ia) described below, showing that the enone derivatives are converted to the active compound (I) in rats.

WO 2009/026934 and WO 2009/026935 disclose two types of prodrug derivatives of compound (I) including (6aR,10aR) -7-propyl-6, 6a,7,8,9,10,10a, 11-octahydro- [1,3] dioxolo [4',5':5,6] benzo [1,2-g ] quinoline, Methylenedioxy (MDO) derivatives having the following formula (Ib):

it has been demonstrated in WO 2010/097092 that compound (Ib) is converted to compound (I) in rat and human hepatocytes. Furthermore, the in vivo pharmacology of compounds (Ia) and (Ib) and the active "parent compound" (I) has been tested in different animal models for parkinson's disease (WO 2010/097092). Compound (I) and both compounds (Ia) and (Ib) were found to be effective, indicating that compounds (Ia) and (Ib) are converted to compound (I) in vivo. All three compounds have been reported to have a longer duration of action than observed for L-dopa and apomorphine.

Other prodrugs of compound (I) disclosed in WO 2009/026934 and WO 2009/026935 are conventional ester prodrugs of formula (Ic) shown below:

despite the long-standing interest in the art, there is clearly an unmet need for the development of highly potent, well-tolerated and orally active drugs for the treatment of PD. Prodrug derivatives of mixed D1/D2 agonists that can provide continuous dopaminergic stimulation, giving stable PK profiles, can meet such unmet needs.

Phosphate derivatives have previously been proposed and explored as a prodrug principle for various compounds containing catechol moieties. EP 0167204 discloses that oral absorption of L-DOPA can be improved by phosphorylation of a hydroxyphenol group. This is evidenced by showing increased renal blood flow in anesthetized dogs. US 3132171 discloses that diphosphate derivatization of L-DOPA provides a water-soluble and stable composition which is converted to L-DOPA upon parenteral administration in rats. US 5073547 discloses monophosphorylated L-DOPA esters and shows that these provide high bioavailability by the oral route (based on intraperitoneal administration), which is believed to be caused by a reduction in peripheral decarboxylation of the compound.

More recently, phosphate and diphosphate derivatives of carbidopa and levodopa have been disclosed in WO 2016/065019, with the aim of increasing the water solubility of these compounds. Plasma concentration profiles of levodopa and carbidopa indicate the conversion of carbidopa and phosphate derivatives of levodopa following intravenous and subcutaneous administration.

Phosphonooxymethyl derivatization of amines is considered to be a prodrug principle for amine compounds, as opposed to catecholamines.

Masking the catechol hydroxyl group of carbidopa derivatives with benzyl or small alkyl groups to improve intestinal absorption has been proposed in WO 2004/052841, but the prodrug potential of such compounds has not been demonstrated.

Disclosure of Invention

The present invention relates to novel compounds useful in the treatment of parkinson's disease. More specifically, the present invention relates to novel prodrug derivatives of the compound (4aR,10aR) -1-n-propyl-1, 2,3,4,4a,5,10,10 a-octahydro-benzo [ g ] quinoline-6, 7-diol (compound (I)). The compounds of the present invention have proven to be particularly useful for the oral delivery of compound (I). In a first aspect, the invention provides a compound according to formula (Id)

Wherein R1, R2 and R3 are according to a) or b) below:

a) r1 and R2 are each independently selected from H, C1-C6Alkyl, benzyl and the following substituents (i), and R3 is absent,

wherein the points of attachment are indicated by the reference numerals,

provided that R1 and R2 cannot both be H;

or

b) R1 and R2 are both H, and R3 is the following substituent (ii),

wherein indicates the attachment point;

and wherein R4 and R5 are each independently selected from H, C1-C6Alkyl, phenyl, benzyl, C3-C6Cycloalkyl and quilt C3-C6Cycloalkyl-substituted methyl;

or a pharmaceutically acceptable salt thereof.

Another aspect of the invention relates to a pharmaceutical composition comprising a compound according to formula (Id), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients.

Another aspect of the invention relates to a compound according to formula (Id) for use as a medicament.

Another aspect of the invention relates to a compound according to formula (Id) or a pharmaceutically acceptable salt thereof, for use in the treatment of a neurodegenerative disease or disorder, such as parkinson's disease, huntington's disease, restless leg syndrome, or alzheimer's disease; or for the treatment of neuropsychiatric diseases or disorders such as schizophrenia, attention deficit hyperactivity disorder or drug addiction.

Another aspect of the invention relates to methods for treating the following diseases or disorders: neurodegenerative diseases or disorders, such as parkinson's disease, huntington's disease, restless leg syndrome, or alzheimer's disease; or for the treatment of a neuropsychiatric disease or disorder, such as schizophrenia, attention deficit hyperactivity disorder or drug addiction; the method comprises administering to a patient in need thereof a therapeutically effective amount of a compound having formula (Id) or a pharmaceutically acceptable salt thereof.

Another aspect of the invention relates to the use of a compound according to formula (Id) or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a neurodegenerative disease or disorder, such as parkinson's disease, huntington's disease, restless leg syndrome or alzheimer's disease; or for the treatment of a neuropsychiatric disease or disorder, such as schizophrenia, attention deficit hyperactivity disorder or drug addiction.

Definition of

Attachment point

In the context of the present invention, it is understood that the carbon atom at the point of attachment on substituent (i) is at the anomeric position of (i). Similarly, the carbon atom at the point of attachment on substituent (ii) is at the anomeric position of (ii).

Compounds of the invention

Reference to a compound encompassed by the invention includes the free material (e.g., free base or zwitterion) of the compound of the invention, pharmaceutically acceptable salts, such as acid addition salts or base addition salts, of the compound of the invention, as well as polymorphic and amorphous forms of the compound of the invention and pharmaceutically acceptable salts thereof. In addition, the compounds of the present invention and pharmaceutically acceptable salts thereof can potentially exist in unsolvated forms as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The present invention encompasses both solvated and unsolvated forms.

Pharmaceutically acceptable salts

In the context of the present invention, pharmaceutically acceptable salts are intended to indicate non-toxic, i.e. physiologically acceptable salts.

The term "pharmaceutically acceptable salts" includes pharmaceutically acceptable acid addition salts, which are salts of an inorganic and/or organic acid formed at the nitrogen atom in the parent molecule. The acid may be selected from, for example, hydrochloric acid, hydrobromic acid, phosphoric acid, nitrous acid, sulfuric acid, benzoic acid, citric acid, gluconic acid, lactic acid, maleic acid, succinic acid, tartaric acid, acetic acid, propionic acid, oxalic acid, malonic acid, fumaric acid, glutamic acid, pyroglutamic acid, salicylic acid, gentisic acid, saccharine, and sulfonic acids, such as methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, naphthalene-2-sulfonic acid, 2-hydroxyethanesulfonic acid, and benzenesulfonic acid.

The term pharmaceutically acceptable salts also includes pharmaceutically acceptable base addition salts which are formed with inorganic and/or organic bases on the acid group of the compound having formula (Id). The base may be selected, for example, from zinc hydroxide, as well as alkali metal bases, such as sodium hydroxide, lithium hydroxide, potassium hydroxide, and alkaline earth metal bases, such as calcium hydroxide and magnesium hydroxide, and organic bases, such as choline, diethylamine, trimethylamine, and triethylamine.

Further examples of useful acids and bases to form pharmaceutically acceptable salts can be found, for example, in Stahl and Wermuth (editions) "Handbook of Pharmaceutical salts. Characteristics, selection and use ] ", Wiley-VCH (Willey-VCH Press), 2008.

Prodrugs

In the context of the present invention, the term "prodrug" or "prodrug derivative" indicates a compound that is converted in vivo to a pharmacologically active moiety upon administration to a living subject, such as a mammal, preferably a human. The transformation preferably takes place in a mammal, such as in a mouse, rat, dog, mini-pig, rabbit, monkey and/or human. In the context of the present invention, a "prodrug of the compound (4aR,10aR) -1-n-propyl-1, 2,3,4,4a,5,10,10 a-octahydro-benzo [ g ] quinoline-6, 7-diol", or a "prodrug of a compound of formula (I)", or a "prodrug of a compound (I)" is understood to be a compound which, after administration, is converted in vivo into the compound (4aR,10aR) -1-n-propyl-1, 2,3,4,4a,5,10,10 a-octahydro-benzo [ g ] quinoline-6, 7-diol. The administration may be by conventional routes of administration of pharmaceutical compositions known in the art, preferably by oral administration.

In the context of the present invention, the terms "parent compound" and "parent molecule" indicate the pharmacologically active moiety obtained after conversion of the corresponding prodrug. For example, a "parent compound" of one of the compounds (Ia), (Ib), (Ic) or of any of the compounds of the invention is understood to be a compound having formula (I).

Substituent group

In the context of the present invention, the given range may be indicated interchangeably with "-" (dash) or "to", for example the term "C1-C6Alkyl is "equivalent to" C1To C6Alkyl groups ".

The term "alkyl" refers to a straight-chain (i.e., unbranched) or branched-chain saturated hydrocarbon having from 1 to 6 carbon atoms, inclusive. Examples of such groups include, but are not limited to, methyl, ethyl, 1-propyl, 2-propyl, 1-butyl, 2-methyl-2-propyl, 2-methyl-1-butyl, and n-hexyl.

The term "cycloalkyl" as used herein refers to a saturated ring system containing all carbon atoms as ring members and 0 double bonds. The cycloalkyl group can be a monocyclic cycloalkyl group (e.g., cyclopropyl). Representative examples of cycloalkyl groups include, but are not limited to: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.

Pharmacokinetic definitions and abbreviations

As used herein, a "PK profile" is an abbreviation for "pharmacokinetic profile". The pharmacokinetic profiles and pharmacokinetic parameters described herein are based on plasma concentration-time data obtained for compounds having formula (I) using a non-compartmental model following oral administration of a compound of the invention. Abbreviated PK parameters are: cmax(maximum concentration); t is tmax(to C)maxTime of (d); t is t1/2(half-life); AUC0-24(area under the curve from time of administration and up to 24 hours post-administration) and "24 hour exposure" are the plasma concentrations of the compound of formula (I) as measured 24 hours post-administration.

A therapeutically effective amount

In the context of the present invention, the term "therapeutically effective amount" of a compound of the present invention means an amount sufficient to alleviate, block, partially block, eliminate or delay the clinical manifestations of a given disease and its complications in a therapeutic intervention involving the administration of said compound. An amount sufficient to achieve this is defined as a "therapeutically effective amount". An effective amount for each purpose will depend, for example, on the severity of the disease or injury and the weight and general state of the subject. It will be appreciated that determining the appropriate dose can be achieved using routine experimentation by constructing a matrix of values and testing different points in the matrix, all within the ordinary skill of a trained physician.

In the context of the present invention, a "therapeutically effective amount" of a compound of the invention indicates an amount of said compound of the invention which, when administered (preferably by oral route) to a mammal (preferably a human), is capable of providing an amount of compound (I) sufficient to alleviate, block, partially block, remove or delay the clinical manifestations of a given disease and its complications.

Treatment (Treatment and healing)

In the context of the present invention, "treatment" is intended to indicate the management and care of a patient for the purpose of alleviating, arresting, partially arresting, removing the clinical manifestations of a disease or delaying its progression. The patient to be treated is preferably a mammal, in particular a human being.

For the treatment of disorders

The compounds of the invention are intended for use in the treatment of neurodegenerative diseases and disorders, such as parkinson's disease and/or other conditions for which treatment with dopamine agonists is therapeutically beneficial.

Therapeutic indications include various central nervous system disorders characterized by movement and/or non-movement disorders, and for which a part of the underlying pathophysiology is a disorder of the striatum-mediated circuit. Such dysfunctions may be found in neurodegenerative diseases such as, but not limited to, Parkinson's Disease (PD), restless leg syndrome, huntington's disease and alzheimer's disease, as well as neuropsychiatric diseases such as, but not limited to, schizophrenia, attention deficit hyperactivity disorder and drug addiction.

In addition to neurodegenerative diseases and disorders, other conditions in which an increase in dopaminergic turnover may be beneficial are improved in different aspects of mental function, including cognition. It also has positive effects in depressed patients, and it can also be used as an appetite suppressant for the treatment of obesity, and for the treatment of drug addiction. It can improve Mild Brain Dysfunction (MBD), narcolepsy, attention deficit hyperactivity disorder and, potentially, negative, positive and cognitive symptoms of schizophrenia.

Restless Leg Syndrome (RLS) and Periodic Limb Movement Disorder (PLMD) are alternative indications to clinical treatment with dopamine agonists. In addition, impotence, erectile dysfunction, SSRI-induced sexual dysfunction, ovarian hyperstimulation syndrome (OHSS) and certain pituitary tumors (prolactinomas) may also be ameliorated by treatment with dopamine agonists. Dopamine is involved in the regulation of the cardiovascular and renal systems, and therefore renal failure and hypertension can be considered as alternative indications for the compounds of the invention.

The present invention encompasses the use of the compounds of the present invention for the treatment of the diseases and disorders listed above.

Combination of

In one embodiment of the invention, the compound having formula (Id) is used as the sole active compound as a stand-alone treatment. In another embodiment of the invention, the compound having formula (Id) may be used in combination with other agents useful in the treatment of neurodegenerative diseases or disorders, such as parkinson's disease. The terms "combined use", "combination with … …", and "combination of … …" and the like as used herein in the context of the methods of the invention (including the combined administration of a therapeutically effective amount of a compound of formula (Id) and another compound that is useful in the treatment of a neurodegenerative disease or disorder) are intended to mean the administration of a compound of formula (Id) either simultaneously or sequentially (in any order) with the other compound.

The two compounds may be administered simultaneously or with a time interval between the administration of the two compounds. The two compounds may be administered as part of the same pharmaceutical formulation or composition, or in separate pharmaceutical formulations or compositions. The two compounds may be administered on the same day or on different days. They can be administered by the same route, such as, for example, by oral administration, subcutaneous injection, by transdermal administration, by depot (depot) type, by intramuscular injection or intravenous injection; or by different routes, wherein one compound is administered, e.g. orally or placed through a depot, and e.g. another compound is injected. The two compounds may be administered by the same dosing regimen or interval, such as once or twice daily, weekly, or monthly; or by different dosing regimens, e.g., wherein one compound is administered once daily and the other compound is administered daily or twice weekly or monthly.

In some cases, when treatment is initiated with a compound having formula (Id), the patient to be treated may have been treated with one or more other compounds useful for treating a neurodegenerative disease or disorder. In other cases, the patient may have been treated with a compound having formula (Id) when treatment is initiated with one or more other compounds useful for treating a neurodegenerative disease or disorder. In other cases, treatment with a compound having formula (Id) and treatment with one or more other compounds useful for treating a neurodegenerative disease or disorder begin simultaneously.

Compounds for combination therapy

In the context of the present invention, the compound to be used in combination with the compound having formula (Id) may be selected from, for example, L-DOPA, droxidopa, folioglurax, MAO-B inhibitors, such as selegiline or rasagiline, COMT inhibitors, such as entacapone or tolcapone, adenosine 2a antagonists, such as itraphylline, anti-glutamines, such as amantadine or memantine, acetylcholinesterase inhibitors, such as rivastigmine, donepezil or galantamine, and antipsychotics, such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or ipiprazole.

In addition to small molecules, compounds for combination may also be included in the treatment of neurodegenerative diseases or disorders, emerging biologics approaches such as, for example, antibodies targeting α -synuclein, tau protein or a- β protein.

Route of administration

Pharmaceutical compositions comprising a compound having formula (Id), either as the sole active compound or in combination with another active compound, may be specifically formulated for administration by any suitable route, such as the oral, rectal, nasal, buccal, sublingual, pulmonary, transdermal and parenteral (e.g., subcutaneous, intramuscular and intravenous) routes. In the context of the present invention, the oral route is the preferred route of administration.

It will be appreciated that the route will depend on the general condition and age of the subject to be treated, the nature of the condition to be treated and the active ingredient.

Pharmaceutical formulations and excipients

Hereinafter, the term "excipient" or "pharmaceutically acceptable excipient" refers to a pharmaceutical excipient including, but not limited to, carriers, fillers, diluents, antiadherents, binders, coatings, colorants, disintegrants, flavoring agents, glidants, lubricants, preservatives, sorbents, sweeteners, solvents, carriers, and adjuvants.

The invention also provides a pharmaceutical composition comprising a compound having formula (Id) (one of the compounds as disclosed in the experimental section herein). The invention also provides methods for making pharmaceutical compositions comprising compounds having formula (Id). The pharmaceutical compositions according to the invention may be formulated with pharmaceutically acceptable excipients according to conventional techniques such as those disclosed in: remington, The Science and Practice of Pharmacy 22 nd edition (2013), editors Allen, Loyd v., Jr.

Preferably, the pharmaceutical composition comprising a compound of the invention is a pharmaceutical composition for oral administration. Pharmaceutical compositions for oral administration include solid oral dosage forms such as tablets, capsules, powders and granules; and liquid oral dosage forms such as solutions, emulsions, suspensions and syrups, as well as powders and granules to be dissolved or suspended in a suitable liquid.

Solid oral dosage forms may be presented as discrete units (e.g., tablets or hard or soft capsules), each containing a predetermined amount of the active ingredient, and preferably one or more suitable excipients. Where appropriate, solid dosage forms may be prepared with coatings, such as enteric coatings, or they may be formulated to provide modified release, such as delayed or extended release, of the active ingredient, according to methods well known in the art. Where appropriate, the solid dosage form may be one which disintegrates in saliva, such as, for example, an orodispersible tablet.

Examples of excipients suitable for use in solid oral formulations include, but are not limited to: microcrystalline cellulose, corn starch, lactose, mannitol, povidone, croscarmellose sodium, sucrose, cyclodextrin, talc, gelatin, pectin, magnesium stearate, stearic acid, and lower alkyl ethers of cellulose. Similarly, solid formulations may contain excipients known in the art for delayed or extended release formulations, such as glyceryl monostearate or hypromellose. If solid materials are used for oral administration, the formulations may be prepared, for example, by mixing the active ingredient with solid excipients and subsequently compressing the mixture in a conventional tableting machine; or the formulation may be placed in a hard capsule, for example, in the form of a powder, pill or mini-tablet. The amount of solid excipient will vary widely, but will typically range from about 25mg to about 1g per dosage unit.

Liquid oral dosage forms may be presented as, for example, elixirs, syrups, oral drops or liquid filled capsules. Liquid oral dosage forms may also be presented as powders, for solution or suspension in aqueous or non-aqueous liquids. Examples of excipients suitable for liquid oral formulations include, but are not limited to, ethanol, propylene glycol, glycerol, polyethylene glycol, poloxamers, sorbitol, polysorbates, mono-and diglycerides, cyclodextrin, coconut oil, palm oil, and water. Liquid oral dosage forms can be prepared, for example, by dissolving or suspending the active ingredient in an aqueous or non-aqueous liquid or by incorporating the active ingredient into an oil-in-water or water-in-oil liquid emulsion.

Additional excipients (e.g., coloring agents, flavoring agents, preservatives, and the like) may be used in the solid and liquid oral formulations.

Pharmaceutical compositions for parenteral administration include: sterile aqueous and non-aqueous solutions, dispersions, suspensions or emulsions for injection or infusion, concentrates for injection or infusion and sterile powders to be reconstituted in a sterile solution or dispersion for injection or infusion prior to use. Examples of excipients suitable for parenteral formulation include, but are not limited to, water, coconut oil, palm oil, and cyclodextrin solutions. The aqueous formulation should be suitably buffered if necessary and made isotonic with sufficient saline or glucose.

Other types of pharmaceutical compositions include suppositories, inhalants, creams, gels, skin patches, implants and formulations for buccal or sublingual administration.

The essential requirement is that the excipients used in any pharmaceutical formulation are compatible with the intended route of administration and with the active ingredient.

Dosage form

In one embodiment, the compounds of the present invention are administered in an amount of from about 0.0001mg/kg body weight to about 5mg/kg body weight per day. In particular, the daily dose may be in the range of 0.001mg/kg body weight to about 2mg/kg body weight per day. The precise dosage will depend upon the frequency and mode of administration, the sex, age, weight and general condition of the subject to be treated, the condition to be treated, the nature and severity of any concomitant diseases to be treated, the desired therapeutic effect and other factors known to those skilled in the art.

Typical oral dosages for adults will be in the following ranges: 0.01-100 mg/day of a compound of the invention, e.g. 0.05-50 mg/day, e.g. 0.1-10 mg/day or 0.1-5 mg/day. Conveniently, the compounds of the invention are administered in a unit dosage form comprising the compound in the following amounts: about 0.01 to 50mg, such as 0.05mg, 0.1mg, 0.2mg, 0.5mg, 1mg, 5mg, 10mg, 15mg, 20mg or up to 50mg of a compound of the invention.

Drawings

FIG. 1:schematic representation of the conversion of a compound of the invention to compound (Id).

Solid arrows: demonstrated transformation was obtained in vitro and in vivo. Dotted arrow: demonstrated transformation was obtained in vitro.

FIG. 2:PK profile in Wistar rats obtained after oral administration according to example 3. The curves are based on the mean plasma concentrations from 3 subjects for each compound.

An X axis: time (hours); y-axis: plasma concentrations (pg/mL) of compound (I) obtained after administration of the following compounds: ■: compound (Ia); solid content: compound (Ib); +: compound (9); ●: compound (8); x: compound (3) and a: compound (2).

Detailed Description

The inventors of the present invention have identified new compounds which are prodrugs of (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydro-benzo [ g ] quinoline-6, 7-diol [ compound (I) ] of dual D1/D2 agonists (see e.g. WO 2009/026934).

The compounds of the present invention are phosphate esters and phosphonooxymethyl derivatives as well as ether derivatives of compound (I).

It was found that oral administration of representative compounds of the invention in Wistar rats provided systemic exposure of compound (I) in plasma, indicating the usefulness of said compounds as orally active prodrugs of compound (I).

For all compounds, the dose was corrected by molecular weight to a dose equal to 300. mu.g/kg of compound (Ib) (corresponding to 287. mu.g/kg of compound (I)).

It has been observed that oral administration of compounds (Ia) and (Ib) to Wistar rats results in early and high peak plasma concentrations of compound (I). In humans, such high peak concentrations may be associated with side effects of dopaminergic drugs (such as, for example, nausea, vomiting, and mild headaches). In contrast, for the compounds of the present invention, a slower absorption rate was observed, with continued exposure of compound (I), avoiding the fast peak concentrations. Thus, plasma exposure of compound (I) was maintained for 24 hours in Wistar rats, although the AUC obtained for compound (I) was generally lower compared to the AUC obtained after administration of compounds (Ia) and (Ib). However, because the peak concentration of compound (I) that is expected to drive side effects is lower, higher doses of the compounds of the invention can be administered, potentially achieving higher overall plasma concentrations of compound (I) than can be achieved from administration of compounds (Ia) and (Ib). When investigating the PK properties of compound (Ic), the inventors of the present invention found that the plasma concentrations of compound (I) were extremely low, rendering compound (Ic) unsuitable for oral administration as a prodrug of compound (I), and confirmed that the oral bioavailability of the compound of the present invention was highly unpredictable. PK parameters for PK studies in Wistar rats are listed in table 4 and PK curves are depicted in figure 2. All compounds evaluated in vivo showed conversion to compound (I).

Phosphate esters and phosphonooxymethyl derivatives are preferred embodiments of the invention.

The biotransformation of the compounds of the invention to compounds of formula (Id) was also assessed by incubation in human plasma and/or human hepatocytes, as described in example 1. For the parent compound (I) itself, a short half-life is observed in plasma assays, which may explain why the presence of compound (I) is in some cases difficult to detect or only in very small amounts, since compound (I) may be metabolized at the same time as it is formed. With respect to compound (6), the occurrence of compound (I) is not observed, but since compound (8) is formed, it can be expected that compound (6) is converted to compound (I) by compound (8).

For the compounds of the invention, the conversion was evaluated in vitro or both, see table 1 below and figure 1.

Table 1: in vitro and in vivo transformation of Compounds of the invention

Nt: not tested; nd: not detected

Thus, in summary, the compounds of the present invention are useful as orally active prodrugs of compound (I) and have been observed in rats to provide PK profiles that avoid the peak C observed for known prodrugs (Ia) and (Ib)maxAnd provides a significantly higher AUC for compound (I) compared to compound (Ic).

Finally, an important problem associated with compound (Ib) is that it is an agonist of the 5-HT2B receptor. Since 5-HT2B receptor agonists have been implicated in the pathogenesis of Valvular Heart Disease (VHD) after long-term exposure, such compounds are not suitable for the treatment of chronic diseases (Rothman et al, Circulation [ loop ] (2000),102: 2836-2841; and also Cavero and Guillon, J.Pharmacol.Toxicol.methods [ J.Pharmacol.methods ] (2014),69: 150-161). Thus, a further advantage obtained with the compounds of the present invention is that these compounds are not 5-HT2B agonists, see example 2 and table 3.

The compounds of the invention are useful in the treatment of neurodegenerative diseases and disorders, such as parkinson's disease and/or other conditions for which treatment with dopamine agonists is therapeutically beneficial. Compounds suitable for oral administration have the potential to provide a new therapeutic paradigm for parkinson's disease.

In one embodiment of the invention, these compounds are useful as stand-alone treatments for neurodegenerative diseases or disorders. In another embodiment of the invention, these compounds are used in combination with other agents for treating PD, such as a compound selected from the group consisting of: L-DOPA, droxidopa, folioglurax, MAO-B inhibitors, such as selegiline or rasagiline, COMT inhibitors, such as entacapone or tolcapone, adenosine 2a antagonists, such as eltanopine, anti-glutamines, such as amantadine or memantine, acetylcholinesterase inhibitors, such as rivastigmine, donepezil or galantamine, antipsychotics, such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or ipiprazole; or in combination with antibodies targeting alpha-synuclein, tau protein or A-beta protein.

Examples of the invention

Hereinafter, embodiments of the present invention are disclosed. The first embodiment is denoted as E1, the second embodiment as E2, and so on:

E1. a compound according to formula (Id)

Wherein R1, R2 and R3 are according to a) or b) below:

a) r1 and R2 are each independently selected from H, C1-C6Alkyl, benzyl and the following substituents (i), and R3 is absent,

and wherein the points of attachment are indicated,

provided that R1 and R2 cannot both be H;

or

b) R1 and R2 are both H, and R3 is substituent (ii) N is positively charged,

and wherein indicates the attachment point;

and wherein

R4 and R5 are each independently selected from H, C1-C6Alkyl, phenyl, benzyl, C3-C6Cycloalkyl and quilt C3-C6Cycloalkyl-substituted methyl;

or a pharmaceutically acceptable salt thereof.

E2. The compound according to embodiment E1, wherein the compound is a compound having formula (Ie),

wherein

R1 and R2 are each independently selected from H, C1-C6Alkyl, benzyl and substituent (i);

provided that R1 and R2 cannot both be H;

wherein indicates the attachment point; and wherein

R4 and R5 are each independently selected from H, C1-C6Alkyl, phenyl, benzyl, C3-C6Cycloalkyl and quilt C3-C6Cycloalkyl-substituted methyl;

or a pharmaceutically acceptable salt thereof.

E3. The compound of embodiment E1-E2 or a pharmaceutically acceptable salt thereof, wherein

R1 is a substituent (i).

E4. The compound of embodiment E1-E3 or a pharmaceutically acceptable salt thereof, wherein

R1 is substituent (i); and is

R2 is selected from H, C1-C6Alkyl, benzyl and substituent (i).

E5. The compound of embodiment E1-E4 or a pharmaceutically acceptable salt thereof, wherein

R1 is selected from H, C1-C6Alkyl, benzyl and substituent (i); and is

R2 is a substituent (i).

E6. A compound according to embodiment E1 or a pharmaceutically acceptable salt thereof, wherein

R1 is H and R2 is substituent (i) and R3 is absent.

E7. A compound according to embodiment E1 or a pharmaceutically acceptable salt thereof, wherein

R1 is substituent (i) and R2 is H and R3 is absent.

E8. A compound according to embodiment E1 or a pharmaceutically acceptable salt thereof, wherein

R1 and R2 are both represented by substituent (i), and R3 is absent.

E9. A compound according to embodiment E1 or a pharmaceutically acceptable salt thereof, wherein

R1 and R2 are both H; and is

R3 is substituent (ii).

E10. The compound according to any one of embodiments E1 and E5, or a pharmaceutically acceptable salt thereof, wherein

R1 is benzyl.

E11. A compound according to embodiment E1 or a pharmaceutically acceptable salt thereof, wherein

R1 is benzyl and R2 is selected from H, C1-C6Alkyl, benzyl and substituent (i).

E12. A compound according to embodiment E1 or a pharmaceutically acceptable salt thereof, wherein

R1 is benzyl; and R2 is selected from H, C1-C6Alkyl, benzyl and substituent (i); and R3 is absent.

E13. A compound according to embodiment E1 or a pharmaceutically acceptable salt thereof, wherein

R1 is benzyl; and R2 is selected from H, C1-C6Alkyl, benzyl and substituent (i); and R3 is absent.

E14. The compound of any one of embodiments E1-E13, or a pharmaceutically acceptable salt thereof, wherein

R4 and R5 are independently selected from H, C1-C6Alkyl, phenyl and benzyl.

E15. The compound of any one of embodiments E1-E14, or a pharmaceutically acceptable salt thereof, wherein

R4 and R5 are independently selected from H and benzyl.

E16. The compound of any one of embodiments E1-E15, or a pharmaceutically acceptable salt thereof, wherein

R4 and R5 are selected from H, C1-C6Alkyl, phenyl and benzyl;

and at least one of R4 and R5 is H.

E17. The compound of any one of embodiments E1-E16, or a pharmaceutically acceptable salt thereof, wherein

R4 and R5 are both H.

E18. The compound of any one of embodiments E1-E16, or a pharmaceutically acceptable salt thereof, wherein

At least one of R4 and R5 is benzyl.

E19. The compound according to embodiment E1, wherein the compound is selected from the group consisting of:

compound 1): (4aR,10aR) -6, 7-bis (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinoline;

compound (2): (4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

compound (3): (4aR,10aR) -7-methoxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

compound (4): benzyl ((4aR,10aR) -6- ((bis (benzyloxy) phosphoryl) oxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-7-yl) hydrogen phosphate;

compound (5): benzyl ((4aR,10aR) -7- ((bis (benzyloxy) phosphoryl) oxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl) hydrogen phosphate;

compound (6): (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6, 7-diylbis (dihydrogen phosphate);

compound (7): benzyl ((4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl) hydrogen phosphate;

compound (8): (4aR,10aR) -7-hydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl dihydrogenphosphate; and

compound (9): ((1S,4aR,10aR) -6, 7-dihydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methylphosphonate;

or a pharmaceutically acceptable salt of any of these compounds.

E20. The compound according to embodiment E1, wherein the compound is selected from the group consisting of:

compound (2): (4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

compound (3): (4aR,10aR) -7-methoxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

compound (6): (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6, 7-diylbis (dihydrogen phosphate);

compound (8): (4aR,10aR) -7-hydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl dihydrogenphosphate; and

compound (9): ((1S,4aR,10aR) -6, 7-dihydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methylphosphonate;

or a pharmaceutically acceptable salt of any of these compounds.

E21. The compound according to embodiment E1, wherein the compound is selected from the group consisting of:

compound (6): (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6, 7-diylbis (dihydrogen phosphate);

compound (8): (4aR,10aR) -7-hydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl dihydrogenphosphate; and

compound (9): ((1S,4aR,10aR) -6, 7-dihydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methylphosphonate;

or a pharmaceutically acceptable salt of any of these compounds.

E22. The compound according to embodiment E1, wherein the compound is selected from the group consisting of:

compound (6): (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6, 7-diylbis (dihydrogen phosphate); and

compound (8): (4aR,10aR) -7-hydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl dihydrogenphosphate;

or a sodium salt of any of these compounds.

E23. The compound according to embodiment E1, wherein the compound is selected from the group consisting of:

compound (2): (4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

compound (3): (4aR,10aR) -7-methoxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol;

or a pharmaceutically acceptable salt of any of these compounds.

E24. A compound having the formula

Or a pharmaceutically acceptable salt thereof.

E25. A compound having the formula

Or a pharmaceutically acceptable salt thereof.

E26. A compound having the formula

Or a pharmaceutically acceptable salt thereof.

E27. A compound having the formula

Or a pharmaceutically acceptable salt thereof.

E28. A compound having the formula

Or a pharmaceutically acceptable salt thereof.

E29. A compound having the formula

Or a pharmaceutically acceptable salt thereof.

E30. A compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, for use in therapy.

E31. A compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, for use as a medicament.

E32. The compound or pharmaceutically acceptable salt thereof for use as a medicament according to embodiment E31, wherein the medicament is an oral medicament, such as a tablet or capsule for oral administration.

E33. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers or diluents.

E34. The pharmaceutical composition of embodiment E33, wherein the pharmaceutical composition is for oral administration.

E35. The pharmaceutical composition of any one of embodiments E33-E34, wherein the pharmaceutical composition is an oral pharmaceutical composition.

E36. The pharmaceutical composition of any one of embodiments E33-E35, wherein the pharmaceutical composition is a solid oral dosage form.

E37. The pharmaceutical composition of any one of embodiments E33-E36, wherein the pharmaceutical composition is a tablet or capsule for oral administration.

E38. The pharmaceutical composition of any one of embodiments E33-E37, wherein the pharmaceutical composition further comprises another agent useful for treating a neurodegenerative disease or disorder, such as parkinson's disease.

E39. The pharmaceutical composition of any one of embodiments E33-E38, wherein the pharmaceutical composition further comprises a compound selected from the group consisting of: L-DOPA, MAO-B inhibitors such as selegiline or rasagiline, COMT inhibitors such as entacapone or tolcapone, adenosine 2a antagonists such as octreotide, anti-glutamates such as amantadine or memantine, acetylcholinesterase inhibitors such as rivastigmine, donepezil or galantamine, antipsychotics such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or ipiprazole; or antibodies targeting alpha-synuclein, tau protein, or A-beta protein.

E40. A compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, for use in the treatment of a neurodegenerative disease or disorder, such as parkinson's disease, huntington's disease, restless leg syndrome, or alzheimer's disease; or a neuropsychiatric disease or disorder, such as schizophrenia, attention deficit hyperactivity disorder or drug addiction.

E41. A compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, for use in the treatment of embodiment E40, wherein the neurodegenerative disease or disorder is parkinson's disease.

E42. A compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, for use in a treatment according to any one of embodiments E40-E41, wherein the compound is to be used in combination with another agent useful in the treatment of a neurodegenerative disease or disorder, such as parkinson's disease.

E43. A compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, for use in the treatment of any one of embodiments E40-E42, wherein the compound is to be used in combination with a compound selected from the group consisting of: L-DOPA, droxidopa, folioglurax, MAO-B inhibitors, such as selegiline or rasagiline, COMT inhibitors, such as entacapone or tolcapone, adenosine 2a antagonists, such as eltanopine, anti-glutamines, such as amantadine or memantine, acetylcholinesterase inhibitors, such as rivastigmine, donepezil or galantamine, antipsychotics, such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or ipiprazole; or in combination with antibodies targeting alpha-synuclein, tau protein or A-beta protein.

E44. A compound according to any one of embodiments E1-E29 or a pharmaceutically acceptable salt thereof, for use in a treatment according to any one of embodiments E40-E43, wherein the treatment is by oral administration of the compound.

E45. A compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, for use in a treatment according to any one of embodiments E40-E44, wherein the compound is comprised in an oral pharmaceutical composition, such as a tablet or capsule for oral administration.

E46. A method for treating a disease or disorder comprising: neurodegenerative diseases or disorders, such as parkinson's disease, huntington's disease, restless leg syndrome, or alzheimer's disease; or for the treatment of a neuropsychiatric disease or disorder, such as schizophrenia, attention deficit hyperactivity disorder or drug addiction; the method comprises administering to a patient in need thereof a therapeutically effective amount of a compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof.

E47. The method of embodiment E46, wherein the neurodegenerative disease or disorder is parkinson's disease.

E48. The method according to any one of embodiments E46-E47, wherein the compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, is used in combination with another agent useful in the treatment of a neurodegenerative disease or disorder, such as parkinson's disease.

E49. The method according to any one of embodiments E46-E48, wherein the compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, is used in combination with a compound selected from the group consisting of: L-DOPA, droxidopa, folioglurax, MAO-B inhibitors, such as selegiline or rasagiline, COMT inhibitors, such as entacapone or tolcapone, adenosine 2a antagonists, such as eltanopine, anti-glutamines, such as amantadine or memantine, acetylcholinesterase inhibitors, such as rivastigmine, donepezil or galantamine, antipsychotics, such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or ipiprazole; or in combination with antibodies targeting alpha-synuclein, tau protein or A-beta protein.

E50. The method of any one of embodiments E46-E49, wherein the administering is by oral route.

E51. The method according to any one of embodiments E46-E50, wherein the compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, is comprised in an oral pharmaceutical composition, such as a tablet or capsule for oral administration.

E52. Use of a compound according to any one of embodiments E1-E29, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a neurodegenerative disease or disorder, such as parkinson's disease, huntington's disease, restless leg syndrome, or alzheimer's disease; or for the treatment of neuropsychiatric diseases or disorders such as schizophrenia, attention deficit hyperactivity disorder or drug addiction.

E53. The use of embodiment E52, wherein the neurodegenerative disease or disorder is parkinson's disease.

E54. The use of any one of embodiments E52-E53, wherein the medicament is used in combination with another medicament useful in the treatment of a neurodegenerative disease or disorder, such as parkinson's disease.

E55. The use of any one of embodiments E52-E54, wherein the medicament is used in combination with a compound selected from the group consisting of: L-DOPA, droxidopa, folioglurax, MAO-B inhibitors, such as selegiline or rasagiline, COMT inhibitors, such as entacapone or tolcapone, adenosine 2a antagonists, such as eltanopine, anti-glutamines, such as amantadine or memantine, acetylcholinesterase inhibitors, such as rivastigmine, donepezil or galantamine, antipsychotics, such as quetiapine, clozapine, risperidone, pimavanserin, olanzapine, haloperidol, aripiprazole or ipiprazole; or in combination with antibodies targeting alpha-synuclein, tau protein or A-beta protein.

E56. The use of any one of embodiments E52-E55, wherein the medicament is an oral medicament, such as a tablet or capsule for oral administration.

In the context of the present invention, it is understood that the carbon atom at the point of attachment on the substituent (ii) (depicted in example E1) is at the anomeric position of (ii).

All documents, including publications, patent applications, and patents, cited herein are hereby incorporated by reference in their entirety and to the same extent as if each document were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein (to the maximum extent permitted by law).

Headings and sub-headings are used herein for convenience only and should not be construed as limiting the invention in any way.

Unless stated otherwise or clearly contradicted by context, use herein of terms relating to one or more elements such as "comprising," "having," "containing," or "containing" in the description of any one or more aspects of the invention is intended to provide support for a similar one or more aspects of the invention "consisting of," "consisting essentially of," or "consisting essentially of" that one or more particular elements (e.g., a composition described herein as comprising a particular element should be understood as also describing a composition consisting of that element unless stated otherwise or clearly contradicted by context).

The use of any and all examples, or exemplary language (including "for example" (for instance, for example, e.g ")," as "(as), and" as (as) used in this specification, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed.

It should be understood that various aspects, embodiments, implementations, and features of the invention mentioned herein may be claimed separately or in any combination.

This invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law.

Compounds of the invention

Table 2: exemplary Compounds of the invention

Experimental part

Preparation of the Compounds of the invention

The compounds of formula (Id) may be prepared by the methods described below, as well as synthetic methods known in the art of organic chemistry or modifications familiar to those of ordinary skill in the art. The starting materials used herein are commercially available or can be prepared by conventional Methods known in the art, such as those described in standard reference books such as "Compendium of Organic Synthetic Methods [ outline of Organic Synthetic Methods ], volume I-XII (published by Wiley-Interscience). Preferred methods include, but are not limited to, those described below.

These schemes are representative of methods that can be used to synthesize the compounds of the invention. They are not intended to limit the scope of the invention in any way.

LC-MS method

Analytical LC-MS data were obtained using the methods identified below.

The method 25 comprises the following steps:MS: an ion source: (APPI) temperature 450 ℃ OR/RNG 20/200VOR/RNG 5/100V

Quality: 100-1000amu

HPLC: column: dC-184.6X 30mm 3 μm Atlantis (Waters)

Column temperature: 40 ℃ gradient, with inverse phase of ion pair

Solvent A: 100% H2O 0.05% TFA

Solvent B: 95% ACN 5% H2O 0.035% TFA

Flow rate: 3.3ml/min, injection volume: 15 μ l

Gradient: 2% B to 100% B in 2.4min, 2% B0.4 min, total run time: 2.8min, UV: 254 nm.

ELSD: glass tube: 21 ℃, evaporation chamber: 40 ℃, pressure: 2.3 bar.

The method 550 comprises the following steps:running the LC-MS on a Waters liquidy UPLC-MS, consisting of: w including a column manageraers liquidy, binary solvent manager, sample organizer, PDA detector (operating at 254 nM), ELS detector, and TQ-MS equipped with APPI source operating in positive ion mode.

LC-conditions: the column is Acquity UPLC BEH C181.7 μm; 2.1X 50mm, operated at 60 ℃ with a binary gradient of 1.2ml/min consisting of water + 0.05% trifluoroacetic acid (A) and acetonitrile/water (95:5) + 0.05% trifluoroacetic acid.

Gradient:

total run time: 1.15 minutes

The method 551 comprises the following steps:running the LC-MS on a Waters liquidy UPLC-MS, consisting of: waters Aquity including column manager, binary solvent manager, sample organizer, PDA detector (operating at 254 nM), ELS detector, and TQ-MS equipped with APPI source operating in positive ion mode.

LC-conditions: the column is Acquity UPLC HSS T31.8 μm; 2.1X 50mm, operated at 60 ℃ with a binary gradient of 1.2ml/min consisting of water + 0.05% trifluoroacetic acid (A) and acetonitrile/water (95:5) + 0.05% trifluoroacetic acid.

Gradient:

0.00min 2%B

1.00min 100%B

1.15min 2%B

total run time: 1.15 minutes

Method 0-30HPLC-AB (Agilent XDB):

0-30 HPLC-AB-conditions: the column was an agilent XDB-C184.6 x 50mm column (1.8 μm particles) operated at 40 ℃ with a binary gradient of 0.6mL/min consisting of water + 0.037% trifluoroacetic acid (a) and acetonitrile + 0.018% trifluoroacetic acid (B).

Gradient:

0.01-2.00min 0-30%B

2.00-4.00min 30%B

4.00-4.01min 0%-0%B

total run time: 4.00 minutes

Method AB01(Agilent 1200& 6120):

LC-conditions: the column was Luna-C18(2)2.0 x 50mm, 5 μm, operated at 40 ℃ with a binary gradient of 0.8ml/min consisting of water + 0.037% trifluoroacetic acid (A) and acetonitrile + 0.018% trifluoroacetic acid (B).

Gradient:

total run time: 4.50 minutes

List of abbreviations for Compounds

BnCl: benzyl chloride

CCl4: carbon tetrachloride

DIPEA: diisopropylethylamine

DMAP: 4-dimethylaminopyridine

DMF: dimethyl formamide

EtOAc: ethyl acetate

NaH: sodium hydride

MeCN: acetonitrile

MeI: methyl iodide

MeOH: methanol

MOM-Cl: chloromethyl methyl ether

Pd/C: palladium on carbon

THF: tetrahydrofuran (THF)

TMSCH2N2: trimethylsilyl diazomethane

TMSI: trimethyliodisilane

General procedure for preparation of Compounds of the invention

(4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinoline-6, 7-diol hydrochloride [ compound (I) ] (which may be prepared, for example, as disclosed in WO 2009/026934) is a substrate for the synthesis of the compounds of the invention.

Prodrugs having two phosphate moieties on the hydroxyl group of catechol may be prepared from (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]Quinoline-6, 7-diol hydrochloride by reaction with dibenzyl phosphonate and a suitable base (like K)2CO3Or DIPEA, but not limited thereto) followed by global deprotection, e.g. against (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]Quinoline-6, 7-diylbis (phosphate). In a similar manner, two monobenzylated catechol derivatives may be converted into the corresponding monophosphates, such as for (4aR,10aR) -7-hydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]Quinoline-6-yl phosphate.

With BnCl and a base (e.g. triethylamine or K)2CO3But not limited to) treatment of Compound (I) will afford (4aR,10aR) -6- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]Quinolin-7-ol and (4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]A mixture of quinolin-6-ols; these regioisomers can be separated. Using MeI instead of BnCl will provide the corresponding methyl ether mixture, which can be isolated.

Provided herein are alternative pathways to (4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol and (4aR,10aR) -7-methoxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol.

The four compounds are used as substrates for reactions with dibenzyl phosphonates or di-tert-butyl phosphates and suitable bases (like K)2CO3Or DIPEA, but not limited thereto) followed by global deprotection, e.g. by hydrogenolysis or acid treatment, will give a mixed prodrug like (4aR,10aR) -7-methoxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] as shown above]Quinolin-6-yl phosphate and (4aR,10aR) -6- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]Quinolin-7-yl phosphate.

Prodrugs can be prepared wherein the phosphate is attached to the N atom via a linker as described for ((1S,4aR,10aR) -6, 7-dihydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methylphosphonate.

Mixed O-bonded and N-bonded prodrugs can be prepared using chemistry analogous to that described above to give compounds like ((1S,4aR,10aR) -6-hydroxy-7-methoxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methylphosphonate.

Mixed N-bonded phosphate and O-benzyl prodrugs like ((1S,4aR,10aR) -7- (benzyloxy) -6-hydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methylphosphonate can be prepared using an alternative protecting group strategy.

Exemplary Compounds of the invention

Compound (1): (4aR,10aR) -6, 7-bis (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinoline

Reacting (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g]Quinoline-6, 7-diol hydrochloride (10.75g) and K2CO3(17.5g) was added to the flask, which was degassed under vacuum and N2Purging, after which DMF (107mL) and benzyl chloride (8.55mL) were added and the mixture was stirred at room temperature for 18 hours, then at 100 ℃ for 5 hours, and at room temperature for 19 hours. Addition of K2CO3(7.48g) and benzyl chloride (6.29mL), and the mixture was stirred at 100 ℃ for 5 hours. After cooling to room temperature, the mixture was partitioned between water (500mL) and heptane (250 mL). The aqueous phase was washed with heptane (3 × 100mL) and the combined organic phases were washed with brine (100mL) over Na2SO4Dried, filtered and concentrated to give the title compound (14.6 g).

Compound (2): (4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol hydroiodide

Reacting (4aR,10aR) -6, 7-bis (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]Quinoline (11.9g) was added to the flask, which was evaporated and treated with N2Purge, then add MeCN (180 mL). The mixture was stirred until homogeneous, after which time trimethylsilyliodide (10.0mL) was added and the mixture was stirred under N2Stirred at room temperature for 2 hours. MeOH (5.5mL) was added and the mixture was stirred for 1 hour. Isopropyl acetate/heptane (10/150mL) was added and the mixture was cooled to 0 deg.C and stirred for 1 hour. The solid was collected, washed with isopropyl acetate/heptane (3/47mL), and dried to give the title compound (7.6 g).

1H NMR(600MHz,CDCl3)δ10.42(bs,1H),7.43-7.33(m,4H),7.26(d,J=1.0Hz,1H),6.78(d,J=8.3Hz,1H),6.58(d,J=8.3Hz,1H),5.72(s,1H),5.08(s,2H),3.71(dd,J=11.70,15.0Hz,1H),3.58(d,J=11.70,1H),3.25-3.11(m,4H),2.94-2.86(m,1H),2.77-2.57(m,2H),2.26(dd,J=11.70Hz,17.0Hz 1H),2.19(d,J=13.80,1H),2.01-1.92(m,2H),1.80-1.69(m,1H),1.56-1.53(m,1H),1.39(qd,J=3.60Hz,13.30Hz,1H),1.06(t,J=7.2Hz,3H)。

LCMS (method 550), retention time 0.55 min, [ M + H]+=352.5m/z。

Compound (3): (4aR,10aR) -7-methoxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol

Step 1

(A1) The method comprises the following steps (4aR,10aR) -7- (benzyloxy) -6- (methoxymethyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinoline

At 0 ℃ under N2Down (4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]A mixture of quinoline-6-ol hydroiodide (20g) in DMF (400mL) was added NaH (4.17g, 60% dispersion) slowly. The mixture was stirred at 0 ℃ for 30 minutes, after which time MOMCl (3.5mL) was added dropwise at 0 ℃. The mixture was stirred at room temperature for 1 hour, after which it was poured into water (400mL) and stirred for 20 minutes, and then extracted with EtOAc (300mL × 3). The combined organic layers were washed with brine (500mL) and Na2SO4Dried, filtered and concentrated to give the title compound (20 g).

Step 2

(A2) The method comprises the following steps (4aR,10aR) -6- (methoxymethyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-7-ol

In N2Down (4aR,10aR) -7- (benzyloxy) -6- (methoxymethyloxy) -1-propyl-1, 2,34,4a,5,10,10 a-octahydrobenzo [ g ]]Quinoline (20g) in MeOH (140mL) was added Pd/C (10%, 30 g). The suspension is degassed under vacuum and treated with H2And (5) purging. At room temperature in H2The mixture was stirred (50psi) for 12 hours, after which the catalyst was filtered off. The filtrate was concentrated to give the title compound (15.4 g).

Step 3

(A3) The method comprises the following steps (4aR,10aR) -7-methoxy-6- (methoxymethyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinoline

To a solution of (4aR,10aR) -6- (methoxymethyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-7-ol (15g) in MeOH (150mL) was added dropwise (trimethylsilyl) diazomethane (2M in ether, 246mL) at room temperature over 0.5 h. The mixture was concentrated to give the title compound (15 g).

Step 4

Compound (3): (4aR,10aR) -7-methoxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-ol

Reacting (4aR,10aR) -7-methoxy-6- (methoxymethyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]A solution of quinoline (15g) in 4M HCl in MeOH (150mL) was stirred at room temperature for 1 hour before it was concentrated. The residue was dissolved in water (100mL) and the aqueous layer was washed with NaHCO3Basifying to pH 7-8. The aqueous layer was extracted with EtOAc (100mL and 50 mL). The combined organic layers were washed with brine (100mL) and Na2SO4Dried, filtered and concentrated to give the title compound (7 g).

LCMS (method 25), retention time 0.95min, 94.0% purity, [ M + H [ ]]+=276.1m/z。

1H NMR(400MHz,CDCl3)δ6.70(d,J=8.4Hz,1H),6.62(d,J=8.0Hz,1H),5.71(br s,1H),3.86(s,3H),3.07-3.18(m,2H),3.01(dd,J=5.2,17.6Hz,1H),2.72-2.89(m,2H),2.58-2.68(m,1H),2.29-2.44(m,2H),2.24(dd,J=12.0,17.6Hz,1H),1.97(d,J=13.2Hz,1H),1.70-1.92(m,3H),1.54-1.63(m,2H),1.10-1.23(m,1H),0.93(t,J=7.2Hz,3H)。

Compound (4): benzyl ((4aR,10aR) -6- ((bis (benzyloxy) phosphoryl) oxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-7-yl) hydrogen phosphate and/or benzyl ((4aR,10aR) -7- ((bis (benzyloxy) phosphoryl) oxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl) hydrogen phosphate [ Compound (5) ]

Reacting (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g]Quinoline-6, 7-diol hydrochloride (1g) and DMAP (117mg) were added to the flask and degassed under vacuum and purged with Ar before CCl was added4(3 mL). The reaction mixture was cooled to 0 ℃ before adding MeCN (15mL), DIPEA (6.66mL), and dibenzyl phosphate (5.02 g). The mixture was stirred at 0 ℃ for 2 hours, after which it was kept at KH2PO4Partition between aqueous (0.5M, 50mL) and EtOAc (20mL, 30 mL). The combined organic layers were washed with brine (20mL) and Na2SO4Dried, filtered and concentrated. The residue was purified by preparative HPLC using an shimadzu LC20AP instrument (Phenomenex Luna C18250 x 50mm, 10 μm particle column with a gradient of 80mL/min water + 0.1% tfa (a) and mecn (B) from 0-20 min 20% B to 50% B at room temperature, 20.1-25 min 100% B, 25.1-30 min 20% B procedure). Crude tetrabenzyl ((4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] is obtained]Quinoline-6, 7-diyl) bis (phosphate) (1.6 g). The crude product was repurified by preparative HPLC using an shimadzu LC20AP instrument (Phenomenex Luna C18250 x 50mm, 10 μm particle column with a gradient of 80mL/min water + 0.1% tfa (a) and mecn (B) from 0-20 min 20% B to 50% B at room temperature, 20.1-25 min 100% B, 25.1-30 min 20% B run) to give the title compound(s) (0.4 g).

Compound (6): tetrasodium (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinoline-6, 7-diyl bis (phosphate)

In N2To a mixture of Pd/C (0.2g, 50% (w/w)) in THF (20mL) and water (5mL) was added benzyl ((4aR,10aR) -6- ((bis (benzyloxy) phosphoryl) oxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]Quinolin-7-yl) hydrogen phosphate and/or benzyl ((4aR,10aR) -7- ((bis (benzyloxy) phosphoryl) oxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g)]Quinolin-6-yl) hydrogen phosphate (0.4g) and NaHCO3(91 mg). The suspension is degassed under vacuum and treated with H2And (5) purging. At room temperature in H2The mixture was stirred (50psi) for 12 hours, after which the catalyst was filtered off. The filtrate was concentrated to give the title compound (0.22 g).

1H NMR(400MHz,D2O)δ7.17(d,J=8.4Hz,1H),6.84(d,J=8.8Hz,1H),3.55(d,J=12.0Hz,1H),3.39(d,J=15.2Hz,1H),3.23-3.32(m,3H),3.11-3.14(m,2H),2.83(m,1H),2.48(m,1H),1.81-2.08(m,5H),1.69-1.71(m,1H),1.40-1.43(m,1H),0.97(t,J=7.2Hz,3H)。

QC-LCMS (method 0-30HPLC-AB), retention time 2.74 min, [ M + H-]+=422.1m/z。

Compound (7): benzyl ((4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl) hydrogen phosphate

To an oven dried microwave flask was added (4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]Quinoline-6-ol hydroiodide (2g) and DMAP (174 mg). The flask was degassed under vacuum and purged with argon before addition of MeCN (30 mL). The reaction mixture was cooled to 0 ℃ before adding CCl4(6mL), DIPEA (9.91mL), and dibenzyl phosphate (7.46 g). The mixture was stirred at 0 ℃ for 2 hours, after which it was kept at KH2PO4Partition between aqueous solution (0.5M, 50mL) and EtOAc (20mL and 30 mL). The combined organic phases were washed with brine (20mL) and Na2SO4Dried, filtered and concentrated. The residue was subjected to preparative HPLC using a Shimadzu LC20AP instrument (run 1: Phenomenex Luna C18250 x 50mm, 10 μm particle column, operating at room temperature with a gradient of 80mL/min water + 0.1% TFA (A) and MeCN (B) from 0-20 min 25% B to 55% B, 20.1-25 min 100% B, 25.1-30 min 25% BLine 2: phenomenex Luna C18250 x 40mm, 10 μm particle column, with a gradient of 60mL/min water + 0.1% tfa (a) and mecn (b) at room temperature: 35% B to 55% B for 0-10 min; 100% B in 10.1-12 min; 12.1-15 min 35% B run) was purified twice to give the title compound (0.2 g).

Compound (8): disodium (4aR,10aR) -7-hydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-6-yl phosphate

In N2Down Pd/C (0.1g, 50% (w/w)) in H2To a mixture of O (2mL) and THF (10mL) was added benzyl ((4aR,10aR) -7- (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]Quinolin-6-yl) hydrogen phosphate (0.2g) and NaHCO3(59 mg). The suspension is degassed under vacuum and treated with H2And (5) purging. At room temperature in H2The mixture was stirred (50psi) for 36 hours. The reaction mixture was filtered through a celite pad, and the filtrate was concentrated to give the title compound (120 mg).

QC-LCMS (method AB01), retention time ═ 1.87min, [ M + H-]+=342.1m/z。

1H NMR(400MHz,DMSO-d6)δ6.53(d,J=8.8Hz,1H),6.42(d,J=8.0Hz,1H),2.93-3.06(m,2H),2.89(d,J=10.8Hz,1H),2.61-2.73(m,1H),2.22-2.40(m,2H),2.12(t,J=14.0Hz,2H),1.95-2.02(m,1H),1.80(d,J=12.8Hz,1H),1.31-1.60(m,4H),1.20-1.25(m,1H),0.99-1.08(m,1H),0.84(t,J=7.2Hz,3H)。

Compound (9) ((1S,4aR,10aR) -6, 7-dihydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methyl hydrogen phosphate

Step 1: (4aR,10aR) -6, 7-bis (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinoline

To a vacuum dried 100mL round bottom flask equipped with a magnetic stir bar was added (4aR,10aR) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ]]Quinoline-6, 7-diol hydrochloride (0.8 g). The flask was sealed with a septum and evacuated and returned with argonFill (this procedure was repeated 3 times). Degassed methanol (80mL) was added followed by benzyl bromide (1.60mL) and NaOH (1.07 g). The mixture was stirred at room temperature for 10 minutes under argon atmosphere, then heated to 60 ℃ and stirred for another 90 minutes. The mixture was cooled to room temperature and concentrated under reduced pressure, then diluted with aqueous sodium bicarbonate (100mL) and EtOAc (100 mL). The phases were separated and the EtOAc phase was washed with water and brine (2X 50mL) and the combined aqueous phases were extracted with EtOAc (2X 50 mL). The combined EtOAc phases were taken over Na2SO4Dried, filtered and purified by combiflash to give the title compound (623 mg).

LCMS (method 450): retention time 0.73 minutes; [ M + H ]]+=442.3m/z。

Step 2: benzyl (((1S,4aR,10aR) -6, 7-bis (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methyl) phosphate

(4aR,10aR) -6, 7-bis (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinoline (200mg), dibenzylchloromethyl phosphate (296mg), NaI (272mg, 1.812mmol) and cesium carbonate (443mg, 1.36mmol) were dissolved in anhydrous acetonitrile (5.0mL), and the resulting solution was stirred under argon atmosphere at 40 ℃ for 90 minutes. The reaction mixture was diluted with EtOAc (50mL), water (25mL) and brine (25mL) and the two phases were separated. The EtOAc phase was washed with water (25mL) and brine (25 mL). The aqueous phase was extracted with EtOAc (2 × 25mL) and the combined EtOAc phases were concentrated. The residue was triturated from ether to give a solid, which was collected and purified by preparative HPLC to give the title compound (80 mg).

LCMS (method 551): RT ═ 0.89 min; [ M + H ]]+=642.7m/z。

And step 3: ((1S,4aR,10aR) -6, 7-dihydroxy-1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methylphosphonate

A solution of benzyl (((1S,4aR,10aR) -6, 7-bis (benzyloxy) -1-propyl-1, 2,3,4,4a,5,10,10 a-octahydrobenzo [ g ] quinolin-1-ium-1-yl) methyl) phosphate (78mg) in 20mL of methanol and 2mL of water was passed through H-Cube (10% Pd/C) at room temperature, 5 bar and a flow rate of 1.0 mL/min. The title compound (32mg) was obtained.

1H NMR (600MHz, methanol-d)4)δ6.66(d,J=8.0Hz,1H),6.49(d,J=8.0Hz,1H),5.28(s,1H),5.03(s,1H),3.78(d,J=12.5Hz,1H),3.69-3.59(m,2H),3.51-3.41(m,1H),3.41-3.34(m,1H),3.16(dd,J=16.9,5.0Hz,2H),2.99(t,J=13.6Hz,1H),2.34(dd,J=17.4,10.5Hz,1H),2.30-2.15(m,2H),2.12(d,J=13.4Hz,1H),1.98-1.78(m,3H),1.52-1.41(m,1H),1.05(t,J=7.2Hz,3H)。

Diastereomeric chemistry was demonstrated by NMR ROESY correlation.

QC-LCMS (method 551): retention time 0.35 min; [ M + H ]]+=372.4m/z。

In vitro and in vivo characterization of the Compounds of the invention

Example 1: transformation of compounds in human plasma and hepatocytes

Example 1 a: conversion of the Compounds of the invention in human plasma

Frozen human plasma was thawed and then centrifuged at 3200x g for 5 minutes to remove debris. The pH of the supernatant was then measured and adjusted to 7.4 ± 0.1 by adding 1% phosphoric acid or 1N sodium hydroxide. mu.L of dosing solution (50. mu.M for test compound and 100. mu.M for positive control (propantheline bromide)) was mixed with 98. mu.L of blank plasma to obtain the final concentrations of 1. mu.M test compound and 2. mu.M positive control. The mixture was incubated at 37 ℃ in a water bath and samples (in duplicate) were taken from the incubations at predetermined time points of 0, 0.5, 1,2, 4 and 6 hours. At each respective time point, 10 μ L of inhibitor and 20 μ L ascorbic acid and 2 μ L formic acid (20%) were added, and then 400 μ L "stop solution" (200 ng/mL tolbutamide plus 200ng/mL labetalol in 50% ACN/MeOH) was added to precipitate the protein. The material was mixed well and thereafter centrifuged at 4,000rpm for 20 minutes. Aliquots of supernatant (50. mu.L) were then transferred from each well to a sample plate and mixed with 100. mu.L of ultrapure water. The plate was shaken at 800rpm for about 10 minutes before being subjected to LC-MS/MS analysis.

Example 1 b: transformation of the Compounds of the invention in human hepatocytes

Incubation at 1. mu.M Compound concentration in 96-well platesIncubations were performed in duplicate. The hepatocyte cell concentration was 0.5X 106 cells/mL for 5% CO2Final incubation at 37 ℃ in an incubator at 95% relative humidity. Media control samples were added at 0 and 60 minutes in the absence of cells. In the final incubation, the total organic concentration was ≦ 1% (DMSO ≦ 0.1%). Controls (7-ethoxycoumarin and 7-hydroxycoumarin) were incubated at 3 μ M in parallel. mu.L of dosing solution (50. mu.M for test compound and 100. mu.M for positive control) was mixed with 98. mu.L of 100mM PBS to obtain the final concentration of 1. mu.M test compound and 2. mu.M positive control. The mixture was incubated at 37 ℃ in a water bath and samples (in duplicate) were taken from the incubations at predetermined time points of 0, 0.5, 1,2, 4 and 6 hours. To each sample was added 10. mu.L of inhibitor and 20. mu.L of ascorbic acid and 2. mu.L of formic acid (20%), followed by 400. mu.L of stop solution (200 ng/mL tolbutamide plus 200ng/mL labetalol in 50% ACN/MeOH). The material was mixed well and thereafter centrifuged at 4,000rpm for 20 minutes. An aliquot of supernatant (50 μ L) from each well was transferred to a sample plate and mixed with 100 μ L of ultrapure water. The plate was shaken at 800rpm for about 10 minutes before being subjected to LC-MS/MS analysis.

Instrumentation for analysis of plasma and hepatocyte incubation samples:

mass spectrometer (LC-MS/MS) UHPLC API 4000 from LC 20-AD Shimadzu, Shimadzu. Analytical column ACQUITYBEH Phenyl 1.7 μm 2.1X 50 mm. Mobile phase A: 0.1% formic acid in water. Mobile phase B: 0.1% formic acid in acetonitrile. Over 2.0 minutes, the gradient ran from 95/5% to 5/95. The flow rate was 0.7 mL/min. MRM monitoring (multiple reaction monitoring) of test items and added analytical standards (labetalol or tolbutamide).

Example 2: 5-HT2B agonist Activity and binding assays

5-HT2B agonist Activity assay

Measurement of the para-muscles by the European/Western Hipport company (Eurofins/Cerep) (France) Using the HTRF detection methodEffect of alcohol-phosphoric acid (IP1) produced Compounds the evaluation of agonist activity at the human 5-HT2B receptor of Compound (I), (Ia), (Ib), (Ic), Compound (2), Compound (3), Compound (6), Compound (8), and Compound (9) was carried out. Briefly, human 5-HT2B receptor was expressed in transfected CHO cells. Cells were suspended in a suspension containing 10mM Hepes/NaOH (pH 7.4), 4.2mM KCl, 146mM NaCl, 1mM CaCl2、0.5mM MgCl25.5mM glucose and 50mM LiCl, then dispensed at a density of 4100 cells/well in microwell plates and incubated for 30 minutes at 37 ℃ in the presence of buffer (basal control), test compound or reference agonist. For control measurements of stimulation, the assay wells alone contained 1 μ M5-HT. After incubation, cells were lysed and a fluorescence acceptor (fluorobenzene D2-labeled IP1) and a fluorescence donor (anti-IP 1 antibody labeled with europium cryptate) were added. After 60 minutes at room temperature, fluorescence transfer was measured using a microplate reader (Rubystar, BMG) at λ (Ex)337nm and λ (Em)620 and 665 nm. The IP1 concentration was determined by dividing the signal measured at 665nm by the signal measured at 620nm (ratio). Results are expressed as a percentage of controls responding to 1 μ M5-HT. The standard reference agonist was 5-HT, which was tested at several concentrations in each experiment to generate a concentration-response curve from which its EC was calculated as described above for the dopamine functional assay50The value is obtained.

5-HT2B binding assay

In the European/Western Hipport company (Eurofins/Cerep) (France), the assessment of the affinity of a compound for the human 5-HT2B receptor was determined in a radioligand binding assay. At room temperature, in the presence of 50mM Tris-HCl (pH 7.4), 5mM MgCl2Membrane homogenates prepared from CHO cells expressing the human 5HT2B receptor and 0.2nM [125I ] in a buffer of 10. mu.M pargyline and 0.1% ascorbic acid in the absence or presence of test compound](±) DOI (1- (4-iodo-2, 5-dimethoxyphenyl) propan-2-amine) were incubated for 60 minutes. Nonspecific binding was determined in the presence of 1. mu.M (. + -.) DOI. After incubation, 96 sample cell collectors (Unifilter, Packard Co.) were used, under vacuum, by pre-soaking with 0.3% Polyethylenimine (PEI)The samples were rapidly filtered on impregnated glass fiber filters (GF/B, Packard Co.) and rinsed several times with ice cold 50mM Tris-HCl. The filters were dried and radioactivity was counted in a scintillation counter (Topcount, Packard) using scintillation cocktail (Microscint 0, Packard). Results were expressed as percent inhibition of control radioligand specific binding. The standard reference compound was (±) DOI, which was tested at several concentrations in each experiment to obtain a competition curve from which its IC was calculated50

TABLE 3 in vitro Activity of the Compounds of the invention obtained according to example 2

Indicates binding affinity (% inhibition of control, specific binding at the indicated concentration).

Example 3: PK experiments in rats

For all experiments, approximately 0.68mL of blood sample was withdrawn from the tail vein or sublingual vein and placed in K which had been precooled and prepared with a stable solution consisting of 80. mu.L ascorbic acid and 40. mu.L 100mM D-glucaric acid 1,4 lactone in water3EDTA tubes. The tubes were gently inverted 6-8 times to ensure thorough mixing and then placed in wet ice. The collection tube was placed in wet ice for up to 30 minutes until centrifugation. Centrifugation was started immediately upon removal from the wet ice. Immediately after centrifugation, the samples were returned to wet ice. Three subsamples of 130 μ Ι _ plasma were transferred to each of three appropriately labeled cryo tubes containing 6.5 μ Ι _ pre-cooled formic acid (20%) (the tubes were pre-blended and frozen for storage prior to use). The tube cap was immediately replaced and the plasma solution was mixed well by gently inverting 6-8 times. Within 60 minutes after sampling, at nominal-70The samples were stored frozen at C. Centrifugation was carried out at 3000G at 4 ℃ for 10 minutes. After collection, the plasma was placed on water-ice. Finally stored at about-70 ℃.

Plasma samples were analyzed by solid phase extraction or direct protein precipitation followed by UPLC-MS/MS. MS detection using electrospray in positive ion mode, where specific mass-nuclear transitions of compound (I) are monitored, the reaction is corrected using an internal standard. The concentration-time data was analyzed using standard software using appropriate non-compartmentalization techniques to obtain estimates of derived PK parameters.

Instrumentation for the analysis of compound (I) from the administration of compound (Ia):

mass spectrometer (LC-MS/MS) was obtained from Acquity-Sciex API 5000, Watcht corporation. BEH UPLC Phenyl 100X 2.1mm column from column Watts corporation, 1.7 μm particle size. Mobile phase A: 20mM ammonium formate (aq) + 0.5% formic acid. Mobile phase B: and (3) acetonitrile. Over 6.1 minutes, the gradient ran from 95%/5% to 2/98. The flow rate was 0.5 mL/min. MRM monitoring of test items and added analytical standards (multiple reaction monitoring).

Dosing and blood sampling:han Wistar rats were provided by the Soothfield Chals River laboratory, Germany (Charles River Laboratories, Sulzfeld, Germany). A 12 hour manual, automatic control, light-dark cycle was maintained. Rats received a standard laboratory diet from Brogaarden corporation (Altromin 1324 pellet). Rats have received this diet without restriction. During the study period (4-week toxicity study), rats received oral dosing of (Ia) once daily by gavage. From rats given 300 μ g/kg (ia), on day 29 after dosing: blood samples from 3 male satellites were collected at the following time points of 0.5, 1,2, 4, 6, 8, 12 and 24 hours.

Instrumentation for the analysis of compound (I) from the administration of compound (Ib):

mass spectrometer (LC-MS/MS) was obtained from Acquity-Sciex API 5000, Watcht corporation. BEH UPLC Phenyl 100X 2.1mm column from column Watts corporation, 1.7 μm particle size. Mobile phase A: 20mM ammonium formate (aq) + 0.5% formic acid. Mobile phase B: and (3) acetonitrile. Over 6.1 minutes, the gradient ran from 95%/5% to 2/98. The flow rate was 0.5 mL/min. MRM monitoring of test items and added analytical standards.

Dosing and blood sampling:han Wistar rats were provided by Charles River Laboratories, UK. A 12 hour manual, automatic control, light-dark cycle was maintained. Rats received a standard laboratory diet (Teklad2014C diet). Rats have received this diet without restriction. During the study period (26 week toxicity study), rats received (Ib) orally once daily by gavage. From rats given 300 μ g/kg (ib), on day 182 post-dose: blood samples from 3 male satellites were collected at the following time points of 0.5, 1,2, 4, 8 and 24 hours.

Instrument set for analysis of compound (I) from administration of compound (Ic), compound (2), compound (3), compound (8), and compound (9)

Mass spectrometer (LC-MS/MS) was acquired by Vortex, Acquity-Vortex, XevoTQ-S. Analytical column Acquity BEH C18100 x 2.1.1 mm, 1.7 μm. Mobile phase A: 20mM NH4Formate + 0.2% formic acid. Mobile phase B: acetonitrile + 0.2% formic acid. The gradient was run from 95%/5% to 5%/95% over 11.0 minutes. The flow rate was 0.3 mL/min. MRM monitoring of test items and added analytical standards.

Administration and blood drawing for Compound (Ic), Compound (2), Compound (3), Compound (8) and Compound (9) Sample preparation:han Wistar rats are provided by Envigo, UK. A 12 hour manual, automatic control, light-dark cycle was maintained. Rats received a standard laboratory diet Teklad 2014C. Rats have received this diet without restriction. Male Han Wistar rats were dosed orally by gavage with a single oral gavage administration of the test compound. Rats were given 494 μ g/kg of (ic), 392 μ g/kg of compound (8), 426 μ g/kg of compound (9), 359 μ g/kg of compound (3) and 551 μ g/kg of compound (2). The following time points on day 1 post-dose: blood samples from 3 males were collected at 0.25, 0.5, 1,2, 4, 8 and 24 hours.

TABLE 4 according to example 3 when 0.300mg/kg (Ia), 0300mg/kg (Ib), 551. mu.g/kg Compound (2), 359 After oral administration of μ g/kg of compound (3), 392 μ g/kg of compound (8) and 426 μ g/kg of compound (9) to Wistar rats, (4aR,10aR) -1-n-propyl-1, 2,3,4,4a,5,10,10 a-octahydro-benzo [ g]Quinoline-6, 7-diol (Compound (I)) PK parameter of

42页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:作为JAK抑制剂的被取代的吡咯并吡啶

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!