WO2021046179A1 - Novel compounds - Google Patents

Novel compounds Download PDF

Info

Publication number
WO2021046179A1
WO2021046179A1 PCT/US2020/049141 US2020049141W WO2021046179A1 WO 2021046179 A1 WO2021046179 A1 WO 2021046179A1 US 2020049141 W US2020049141 W US 2020049141W WO 2021046179 A1 WO2021046179 A1 WO 2021046179A1
Authority
WO
WIPO (PCT)
Prior art keywords
hydroxy
compound according
substituted
compound
pde1 inhibitor
Prior art date
Application number
PCT/US2020/049141
Other languages
French (fr)
Inventor
Peng Li
Robert Davis
Original Assignee
Intra-Cellular Therapies, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Intra-Cellular Therapies, Inc. filed Critical Intra-Cellular Therapies, Inc.
Priority to US17/753,472 priority Critical patent/US20220356187A1/en
Priority to JP2022514214A priority patent/JP2022546710A/en
Priority to EP20861369.5A priority patent/EP4025202A4/en
Publication of WO2021046179A1 publication Critical patent/WO2021046179A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/005Enzyme inhibitors

Definitions

  • the present invention relates to PDE1 inhibitory compounds of Formula la as described below, processes for their production, their use as pharmaceuticals and pharmaceutical compositions comprising them. These compounds are useful e.g., in the treatment of diseases involving disorders of the dopamine D1 receptor intracellular pathway, such as, among others, Parkinson’s disease, depression, narcolepsy, psychosis, damage to cognitive function, e.g., in schizophrenia, or disorders that may be ameliorated through enhanced progesterone-signaling pathway, e.g., female sexual dysfunction.
  • diseases involving disorders of the dopamine D1 receptor intracellular pathway such as, among others, Parkinson’s disease, depression, narcolepsy, psychosis, damage to cognitive function, e.g., in schizophrenia, or disorders that may be ameliorated through enhanced progesterone-signaling pathway, e.g., female sexual dysfunction.
  • PDEs phosphodiesterases
  • CaM-PDEs Ca 2+ -calmodulin-dependent phosphodiesterases
  • CaM-PDEs Ca 2+ -calmodulin-dependent phosphodiesterases
  • PDEs are therefore active in stimulated conditions when intra-cellular calcium levels rise, leading to increased hydrolysis of cyclic nucleotides.
  • the three known CaM-PDE genes, PDE1A, PDE1B, and PDE1C are all expressed in central nervous system tissue.
  • PDE1A In the brain, the predominant expression of PDE1A is in the cortex and neostriatum, PDE1B is expressed in the neostriatum, prefrontal cortex, hippocampus, and olfactory tubercle, and PDE1C is more ubiquitously expressed.
  • Cyclic nucleotide phosphodiesterases decrease intracellular cAMP and cGMP signaling by hydrolyzing these cyclic nucleotides to their respective inactive 5 '-monophosphates (5 'AMP and 5'GMP).
  • CaM-PDEs play a critical role in mediating signal transduction in brain cells, particularly within an area of the brain known as the basal ganglia or striatum.
  • NMDA-type glutamate receptor activation and/or dopamine D2 receptor activation result in increased intracellular calcium concentrations, leading to activation of effectors such as calmodulin-dependent kinase II (CaMKII) and calcineurin and to activation of CaM-PDEs, resulting in reduced cAMP and cGMP.
  • CaMKII calmodulin-dependent kinase II
  • calcineurin calmodulin-dependent kinase II
  • CaM-PDEs calmodulin-dependent kinase II
  • Dopamine D1 receptor activation leads to activation of nucleotide cyclases, resulting in increased cAMP and cGMP.
  • PKA protein kinase A
  • PKG protein kinase G
  • DARPP-32 dopamine and cAMP-regulated phosphoprotein
  • CREB cAMP responsive element binding protein
  • Phosphorylated DARPP-32 in turn inhibits the activity of protein phosphates-1 (PP-1), thereby increasing the state of phosphorylation of substrate proteins such as progesterone receptor (PR), leading to induction of physiologic responses.
  • PP-1 protein phosphates-1
  • PR progesterone receptor
  • CaM-PDEs can therefore affect dopamine-regulated and other intracellular signaling pathways in the basal ganglia (striatum), including but not limited to nitric oxide, noradrenergic, neurotensin, CCK, VIP, serotonin, glutamate (e.g., NMDA receptor, AMPA receptor), GABA, acetylcholine, adenosine (e.g., A2A receptor), cannabinoid receptor, natriuretic peptide (e.g., ANP, BNP, CNP), DARPP-32, and endorphin intracellular signaling pathways.
  • nitric oxide e.g., noradrenergic
  • neurotensin e.g., CCK, VIP
  • serotonin e.g., glutamate (e.g., NMDA receptor, AMPA receptor)
  • GABA e.g., NMDA receptor, AMPA receptor
  • acetylcholine e.g
  • PDE1 phosphodiesterase 1
  • PDE1 functions in brain tissue as a regulator of locomotor activity and learning and memory.
  • PDE1 is a therapeutic target for regulation of intracellular signaling pathways, preferably in the nervous system, including but not limited to a dopamine D1 receptor, dopamine D2 receptor, nitric oxide, noradrenergic, neurotensin, CCK, VIP, serotonin, glutamate (e.g., NMDA receptor, AMPA receptor), GABA, acetylcholine, adenosine (e.g., A2A receptor), cannabinoid receptor, natriuretic peptide (e.g., ANP, GNP, CNP), endorphin intracellular signaling pathway and progesterone signaling pathway.
  • a dopamine D1 receptor e.g., dopamine D2 receptor
  • nitric oxide noradrenergic
  • neurotensin CCK
  • VIP serotonin
  • glutamate
  • inhibition of PDE IB should act to potentiate the effect of a dopamine D1 agonist by protecting cGMP and cAMP from degradation, and should similarly inhibit dopamine D2 receptor signaling pathways, by inhibiting PDE1 activity.
  • Chronic elevation in intracellular calcium levels is linked to cell death in numerous disorders, particularly in neurodegenerative diseases such as Alzheimer's, Parkinson's and Huntington's Diseases and in disorders of the circulatory system leading to stroke and myocardial infarction.
  • PDE1 inhibitors are therefore potentially useful in diseases characterized by reduced dopamine D1 receptor signaling activity, such as Parkinson's disease, restless leg syndrome, depression, narcolepsy and cognitive impairment.
  • PDE1 inhibitors are also useful in diseases that may be alleviated by the enhancement of progesterone- signaling such as female sexual dysfunction.
  • the inventors have further found that the metabolite of Formula M-I accounted for 84% of the total circulating drug related materials in human plasma after oral administration of Compound 1.
  • the current invention provides compounds which specifically limit and/or prevent metabolism occurring by these pathways. Due to the very similar properties of deuterium (3 ⁇ 4) atoms compared to normal hydrogen atoms ( 'H), drug compounds in which deuterium is substituted for hydrogen are believed to generally have similar biological activity to the non-deuterated analog, but potentially with improved pharmacokinetic properties. The extent to which such a substitution will result in an improvement of pharmacokinetic properties without a too severe loss in pharmacologic activity is variable. Thus, in some circumstances, the resulting deuterated compound only a moderate increase in pharmacokinetic stability, while in other circumstances, the resulting deuterated compound may have significantly improved stability. Moreover, it may be difficult to predict with certainty the effects of simultaneous deuterium substitutions. These may or may not result in additive (synergistic) improvement in metabolic stability.
  • the invention provides various PDE1 inhibitory compounds for use in treatment of any one or more of the following conditions:
  • Neurodegenerative diseases including Parkinson’s disease, restless leg, tremors, dyskinesias, Huntington’s disease, Alzheimer’s disease, and drug- induced movement disorders;
  • Circulatory and cardiovascular disorders including cerebrovascular disease, stroke, congestive heart disease, hypertension, pulmonary hypertension, e.g., pulmonary arterial hypertension, and sexual dysfunction, including cardiovascular diseases and related disorders as described in International Application No. PCT/US2014/16741, the contents of which are incorporated herein by reference;
  • Respiratory and inflammatory disorders including asthma, chronic obstructive pulmonary disease, and allergic rhinitis, as well as autoimmune and inflammatory diseases;
  • Cancers or tumors e.g., brain tumors, a glioma (e.g., ependymoma, astrocytoma, oligodendrogliomas, brain stem glioma, optic nerve glioma, or mixed gliomas, e.g., oligoastrocytomas), an astrocytoma (e.g., glioblastoma multiforme), osteosarcoma, melanoma, leukemia, neuroblastoma or leukemia;
  • a glioma e.g., ependymoma, astrocytoma, oligodendrogliomas, brain stem glioma, optic nerve glioma, or mixed gliomas, e.g., oligoastrocytomas
  • an astrocytoma e.g., glioblastoma multiforme
  • osteosarcoma melanom
  • Renal disorders e.g., kidney fibrosis, chronic kidney disease, renal failure, glomerulosclerosis and nephritis;
  • any disease or condition characterized by reduced dopamine D1 receptor signaling activity comprising administering an effective amount of a Compound of the Invention, e.g., a compound according to any of Formula la or 1.1 - 1.26, in free or pharmaceutically acceptable salt or prodrug form, to a human or animal patient in need thereof.
  • a Compound of the Invention e.g., a compound according to any of Formula la or 1.1 - 1.26, in free or pharmaceutically acceptable salt or prodrug form
  • the invention provides a method of preventing the formation of metabolites of the following compound: the method comprising deuterating the PDE1 inhibitor to block the formation of one or more metabolites.
  • the present disclosure provides that the PDE1 inhibitors for use in the methods as described herein are Formula la: wherein
  • R 2 and R 5 are independently H, D or hydroxy and R 3 and R 4 together form a tri or tetra-methylene bridge [pref. with the carbons carrying R 3 and R 4 having the R and S configuration respectively]; or R 2 and R 3 are each methyl and R 4 and R 5 are each H; or R 2 , R 4 and R 5 are H and R 3 is isopropyl [pref. the carbon carrying R 3 having the R configuration] ;
  • R 6 is (optionally halo-substituted or hydroxy-substituted) phenylamino
  • benzylamino (optionally halo-substituted or hydroxy-substituted) benzylamino, Ci- 4 alkyl, or Ci- 4alkyl sulfide; for example, phenylamino or 4-fluorophenylamino;
  • (iii) Rio is Ci- 4 alkyl, methylcarbonyl, hydroxy ethyl, carboxylic acid, sulfonamide, (optionally halo- or hydroxy-substituted) phenyl, (optionally halo- or hydroxy- substituted) pyridyl (for example 6-fluoropyrid-2-yl), or thiadiazolyl (e.g., 1,2,3- thiadiazol-4-yl); and
  • X and Y are independently C or N, in free, pharmaceutically acceptable salt or prodrug form, including its enantiomers, diastereoisomers and racemates.
  • the invention further provides compounds of Formula la as follows:
  • R 6 is hydroxy-substituted phenylamino or hydroxy-substituted benzylamino.
  • a compound according to Formula la or 1.1-1.12, wherein Riois 6-fluoropyrid-2- yi- A compound according to Formula la or 1.1-1.18, wherein X and Y are both C.
  • a compound according to Formula la or 1.1-1.19, wherein the PDE1 inhibitor is a compound according to the following:
  • the present disclosure further provides for a radiolabeled PDE1 inhibitor [Compound 2], e.g., for use in the methods as described herein, according to Formula la or 1.1-1.26.
  • a radiolabeled PDE1 inhibitor [Compound 2] e.g., for use in the methods as described herein, according to Formula la or 1.1-1.26.
  • radiolabeled PDE1 inhibitor is a compound according to the following:
  • selective PDE1 inhibitors of the preceding formula are compounds that inhibit phosphodiesterase-mediated (e.g., PDE1- mediated, especially PDE IB -mediated) hydrolysis of cGMP, e.g., the preferred compounds have an IC50 of less than ImM, preferably less than 500 nM, preferably less than 50 nM, and preferably less than 5nM in an immohili zed-metal affinity particle reagent PDE assay, in free or salt form.
  • Alkyl as used herein is a saturated or unsaturated hydrocarbon moiety, preferably saturated, preferably having one to six carbon atoms, which may be linear or branched, and may be optionally mono-, di- or tri- substituted, e.g., with halogen (e.g., chloro or fluoro), hydroxy, or carboxy.
  • halogen e.g., chloro or fluoro
  • Cycloalkyl as used herein is a saturated or unsaturated nonaromatic hydrocarbon moiety, preferably saturated, preferably comprising three to nine carbon atoms, at least some of which form a nonaromatic mono- or bicyclic, or bridged cyclic structure, and which may be optionally substituted, e.g., with halogen (e.g., chloro or fluoro), hydroxy, or carboxy.
  • halogen e.g., chloro or fluoro
  • the cycloalkyl optionally contains one or more atoms selected from N and O and/or S, said cycloalkyl may also be a heterocycloalkyl.
  • Heterocycloalkyl is, unless otherwise indicated, saturated or unsaturated nonaromatic hydrocarbon moiety, preferably saturated, preferably comprising three to nine carbon atoms, at least some of which form a nonaromatic mono- or bicyclic, or bridged cyclic structure, wherein at least one carbon atom is replaced with N, O or S, which heterocycloalkyl may be optionally substituted, e.g., with halogen (e.g., chloro or fluoro), hydroxy, or carboxy.
  • halogen e.g., chloro or fluoro
  • Aryl as used herein is a mono or bicyclic aromatic hydrocarbon, preferably phenyl, optionally substituted, e.g., with alkyl (e.g., methyl), halogen (e.g., chloro or fluoro), haloalkyl (e.g., trifluoromethyl), hydroxy, carboxy, or an additional aryl or heteroaryl (e.g., biphenyl or pyridylphenyl).
  • alkyl e.g., methyl
  • halogen e.g., chloro or fluoro
  • haloalkyl e.g., trifluoromethyl
  • hydroxy carboxy
  • an additional aryl or heteroaryl e.g., biphenyl or pyridylphenyl
  • Heteroaryl as used herein is an aromatic moiety wherein one or more of the atoms making up the aromatic ring is sulfur or nitrogen rather than carbon, e.g., pyridyl or thiadiazolyl, which may be optionally substituted, e.g., with alkyl, halogen, haloalkyl, hydroxy or carboxy.
  • substituents end in “ene”, for example, alkylene, phenylene or arylalkylene, said substituents are intended to bridge or be connected to two other substituents. Therefore, methylene is intended to be -CH2- and phenylene intended to be - C6H4- and arylalkylene is intended to be -C6H4-CH2- or - CH2-C6H4-.
  • Compounds of the Invention e.g., substituted 4,5,7,8-tetrahydro-2H-imidazo[l,2- a]pyrrolo[3,4-e]pyrimidine or 4,5,7,8,9-pentahydro-2H-pyrimido[l,2-a]pyrrolo[3,4-e]pyrimidine, e.g., Compounds of Formula la, may exist in free or salt form, e.g., as acid addition salts.
  • language such as “Compounds of the Invention” is to be understood as embracing the compounds in any form, for example free or acid addition salt form, or where the compounds contain acidic substituents, in base addition salt form.
  • compositions of the Invention are intended for use as pharmaceuticals, therefore pharmaceutically acceptable salts are preferred. Salts which are unsuitable for pharmaceutical uses may be useful, for example, for the isolation or purification of free Compounds of the Invention or their pharmaceutically acceptable salts, are therefore also included.
  • Compounds of the Invention may in some cases also exist in prodrug form.
  • a prodrug form is compound which converts in the body to a Compound of the Invention.
  • these substituents may form physiologically hydroly sable and acceptable esters.
  • physiologically hydrolysable and acceptable ester means esters of Compounds of the Invention which are hydrolysable under physiological conditions to yield acids (in the case of Compounds of the Invention which have hydroxy substituents) or alcohols (in the case of Compounds of the Invention which have carboxy substituents) which are themselves physiologically tolerable at doses to be administered.
  • the Compound of the Invention contains a hydroxy group, for example, Compound-OH
  • the acyl ester prodrug of such compound i.e., Compound- 0-C(0)-Ci- 4 alkyl
  • the Compound of the Invention contains a carboxylic acid, for example, Compound- C(0)0H
  • the acid ester prodrug of such compound Compound-C(0)0-Ci- 4 alkyl can hydrolyze to form Compound-C(0)OH and HO-Ci-4alkyl.
  • the term thus embraces conventional pharmaceutical prodrug forms.
  • the invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising a Compound of the Invention, in free or pharmaceutically acceptable salt form, in admixture with a pharmaceutically acceptable carrier, for use in the treatment of a disease or disorder mediated by PDE1.
  • Compounds of the Invention may in some cases also exist in prodrug form.
  • a prodrug form is compound which converts in the body to a Compound of the Invention.
  • these substituents may form physiologically hydrolysable and acceptable esters.
  • physiologically hydrolysable and acceptable ester means esters of Compounds of the Invention which are hydrolysable under physiological conditions to yield acids (in the case of Compounds of the Invention which have hydroxy substituents) or alcohols (in the case of Compounds of the Invention which have carboxy substituents) which are themselves physiologically tolerable at doses to be administered.
  • the Compound of the Invention contains a hydroxy group, for example, Compound-OH
  • the acyl ester prodrug of such compound i.e., Compound- 0-C(0)-Ci- 4 alkyl
  • the Compound of the Invention contains a carboxylic acid, for example, Compound- C(0)0H
  • the acid ester prodrug of such compound Compound-C(0)0-Ci- 4 alkyl can hydrolyze to form Compound-C(0)OH and HO-Ci-4alkyl.
  • the term thus embraces conventional pharmaceutical prodrug forms.
  • the invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising a Compound of the Invention, in free, pharmaceutically acceptable salt or prodrug form, in admixture with a pharmaceutically acceptable carrier, for use in the treatment of a disease or disorder mediated by PDE1.
  • the compounds of the Invention and their pharmaceutically acceptable salts may be made using the methods as described and exemplified herein and by methods similar thereto and by methods known in the chemical art. Such methods include, but not limited to, those described below. If not commercially available, starting materials for these processes may be made by procedures, which are selected from the chemical art using techniques which are similar or analogous to the synthesis of known compounds.
  • the Compounds of the Invention include their enantiomers, diastereoisomers and racemates, as well as their polymorphs, hydrates, solvates and complexes.
  • Some individual compounds within the scope of this invention may contain double bonds. Representations of double bonds in this invention are meant to include both the E and the Z isomer of the double bond.
  • some compounds within the scope of this invention may contain one or more asymmetric centers. This invention includes the use of any of the optically pure stereoisomers as well as any combination of stereoisomers.
  • the Compounds of the Invention encompass their stable and unstable isotopes.
  • Stable isotopes are nonradioactive isotopes which contain one additional neutron compared to the abundant nuclides of the same species (i.e., element). It is expected that the activity of compounds comprising such isotopes would be retained, and such compound would also have utility for measuring pharmacokinetics of the non-isotopic analogs.
  • the hydrogen atom at a certain position on the Compounds of the Invention may be replaced with deuterium (a stable isotope which is non-radioactive). Examples of known stable isotopes include, but not limited to, deuterium, 13 C, 15 N, 18 O.
  • unstable isotopes which are radioactive isotopes which contain additional neutrons compared to the abundant nuclides of the same species (i.e., element), e.g., 123 1, 131 I, 125 I, U C, 18 F, may replace the corresponding abundant species of I, C and F.
  • Another example of useful isotope of the compound of the invention is the U C isotope.
  • the Compounds of the Invention are useful in the treatment of diseases characterized by disruption of or damage to cAMP and cGMP mediated pathways, e.g., as a result of increased expression of PDE1 or decreased expression of cAMP and cGMP due to inhibition or reduced levels of inducers of cyclic nucleotide synthesis, such as dopamine and nitric oxide (NO).
  • inducers of cyclic nucleotide synthesis such as dopamine and nitric oxide (NO).
  • the invention provides methods of treatment of any one or more of the following conditions:
  • Neurodegenerative diseases including Parkinson’s disease, restless leg, tremors, dyskinesias, Huntington’s disease, Alzheimer’s disease, and drug-induced movement disorders;
  • Circulatory and cardiovascular disorders including cerebrovascular disease, stroke, congestive heart disease, hypertension, pulmonary hypertension, e.g., pulmonary arterial hypertension, and sexual dysfunction, including cardiovascular diseases and related disorders as described in International Application No. PCT/US2014/16741, the contents of which are incorporated herein by reference;
  • Respiratory and inflammatory disorders including asthma, chronic obstructive pulmonary disease, and allergic rhinitis, as well as autoimmune and inflammatory diseases;
  • Any disease or condition characterized by reduced dopamine D1 receptor signaling activity comprising administering an effective amount of a Compound of the Invention, e.g., a compound according to any of Formula la or 1.1-1.26, in free or pharmaceutically acceptable salt or prodrug form, to a human or animal patient in need thereof.
  • a Compound of the Invention e.g., a compound according to any of Formula la or 1.1-1.26, in free or pharmaceutically acceptable salt or prodrug form
  • the Compounds of the Invention are useful in the treatment of inflammatory diseases or conditions, particularly neuroinflammatory diseases or conditions. Therefore, administration or use of a preferred PDE1 inhibitor as described herein, e.g., a PDE1 inhibitor as hereinbefore described, e.g., a Compound of Formula la, provides a means to regulate inflammation (e.g., prevent, reduce, and/or reverse neuroinflammation, and diseases or disorders related to neuro inflammation), and in certain embodiments provide a treatment for various inflammatory diseases and disorders.
  • the inflammatory disease or condition is selected from: a.
  • neurodegenerative conditions such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and demyelinating conditions, e.g., multiple sclerosis (MS), and prion diseases
  • b stroke, cardiac arrest, hypoxia, intracerebral hemorrhage or traumatic brain injury
  • c conditions characterized by abnormal neurotransmitter production and/or response, including depression, schizophrenia, post-traumatic stress disorder, anxiety, attention deficit disorder, and bipolar disease; e.g., wherein any of the foregoing are associated with neuroinflammation; and d.
  • chronic CNS infections e.g., Lyme disease or CNS infection consequent to an immunosuppressive condition, e.g., HIV-dementia; e. neuroinflammation consequent to chemotherapy; comprising administering an effective amount of a PDE1 inhibitor of the current invention (e.g., a PDE1 inhibitor of Formula la as herein described), e.g., an amount effective to (i) reduce or inhibit activation of Ml microglia, and/or (ii) and amount effective to reduce levels of one or more pro-inflammatory cytokines (e.g., IEIb, TNF- a, and Ccl2, or combination thereof); to a patient in need thereof.
  • a PDE1 inhibitor of the current invention e.g., a PDE1 inhibitor of Formula la as herein described
  • an effective amount effective to (i) reduce or inhibit activation of Ml microglia, and/or (ii) and amount effective to reduce levels of one or more pro-inflammatory cytokines e.g.,
  • the Compounds of the Invention are useful in the treatment of cancers or tumors, e.g., in the inhibition of the proliferation of cancerous or tumorous cells. Therefore, administration or use of a preferred PDE1 inhibitor as described herein, e.g., a PDE1 inhibitor as hereinbefore described, e.g., a Compound of Formula la, in the treatment or prevention of a cancer or tumor.
  • the compounds of the present disclosure can be used in the treatment of a tumor or cancer selected from one or more of acoustic neuroma, astrocytoma, chordoma, lymphoma (e.g., CNS lymphoma, Hodgkin’s lymphoma or non-Hodgkin’s lymphoma), craniopharyngioma, gliomas (e.g., Brain stem glioma, ependymoma, mixed glioma, optic nerve glioma), subependymoma, medulloblastoma, meningioma, metastatic brain tumors, oligodendroglioma, pituitary tumors, primitive neuroectodermal (PNET), schwannoma, adenomas (e.g., basophilic adenoma, eosinophilic adenoma, chromophobe a
  • lymphoma e.
  • lung cancer pancreatic cancer, prostate cancer, urothelial cancer, cancers of the head and neck, or leukemia (e.g., a lymphocytic leukemia or a myelogenous leukemia), colon cancer (e.g., colorectal cancer) and cancers of the kidney, ureter, bladder or urethra.
  • leukemia e.g., a lymphocytic leukemia or a myelogenous leukemia
  • colon cancer e.g., colorectal cancer
  • cancers of the kidney, ureter, bladder or urethra e.g., colorectal cancer
  • the current invention also provides compounds which specifically limit and/or prevent metabolism of PDE1 inhibitors, as well as related methods. Therefore, in one embodiment the invention provides a method (Method 1) of inhibiting the metabolism of a PDE1 inhibitor, e.g. a PDE1 inhibitor according to Formula la or 1.1-1.26, the method comprising deuterating the PDE1 inhibitor to block the formation of one or more metabolites. .
  • Method 1 wherein the PDE1 inhibitor is a compound according to the following formula: in free or pharmaceutically acceptable salt form. . Any of the preceding Methods, wherein deuterating the PDE1 inhibitor comprises reacting:
  • deuterating the PDE1 inhibitor comprises reacting:
  • Method 1.2 wherein the reaction is carried out in benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate, THF and 1,8- Diazabicyclo[5.4.0]undec-7-ene.
  • Method 1.2 or 1.6 wherein the is reacted with thionyl chloride in THF to form the following product: and subjecting the product to chiral column separation to obtain:
  • Compounds of the Invention or “PDE 1 inhibitors of the Invention”, or like terms, encompasses any and all of the compounds disclosed herewith, e.g., a Compound of Formula la or 1.1-1.26.
  • treatment and “treating” are to be understood accordingly as embracing prophylaxis and treatment or amelioration of symptoms of disease as well as treatment of the cause of the disease.
  • the word “effective amount” is intended to encompass a therapeutically effective amount to treat or mitigate a specific disease or disorder, and/or a symptom thereof, and/or to inhibit PDE1 expression in a patient or subject.
  • patient includes a human or non-human (i.e., animal) patient.
  • the invention encompasses both humans and nonhuman animals.
  • the invention encompasses nonhuman animals.
  • the term encompasses humans.
  • Compounds of the Invention e.g., Formula la and 1.1-1.26 as hereinbefore described, in free or pharmaceutically acceptable salt form, may be used as a sole therapeutic agent, but may also be used in combination or for co-administration with other active agents.
  • the Compounds of the Invention e.g., Formula la or 1.1-1.26 as hereinbefore described, in free or pharmaceutically acceptable salt form, may be administered in combination (e.g.
  • SSRIs selective serotonin reuptake inhibitors
  • SNRIs serotonin-norepinephrine reuptake inhibitors
  • TCAs tricyclic antidepressants
  • Dosages employed in practicing the present invention will of course vary depending, e.g. on the particular disease or condition to be treated, the particular Compound of the Invention used, the mode of administration, and the therapy desired.
  • Compounds of the Invention may be administered by any suitable route, including orally, parenterally, transdermally, or by inhalation, but are preferably administered orally.
  • suitable routes including orally, parenterally, transdermally, or by inhalation, but are preferably administered orally.
  • satisfactory results e.g. for the treatment of diseases as hereinbefore set forth are indicated to be obtained on oral administration at dosages of the order from about 0.01 to 2.0 mg/kg.
  • an indicated daily dosage for oral administration will accordingly be in the range of from about 0.75 to 150 mg (depending on the drug to be administered and the condition to be treated, for example in the case of Compound 214, 0.5 to 25 mg, e.g., 1 to 10 mg, per diem, e.g., in monophosphate salt form, for treatment of PDE1- mediated conditions), conveniently administered once, or in divided doses 2 to 4 times, daily or in sustained release form.
  • Unit dosage forms for oral administration thus for example may comprise from about 0.2 to 75 or 150 mg, e.g. from about 0.2 or 2.0 to 50, 75 or 100 mg (e.g., 1, 2.5, 5, 10, or 20 mg) of a Compound of the Invention, e.g., together with a pharmaceutically acceptable diluent or carrier therefor.
  • compositions comprising Compounds of the Invention may be prepared using conventional diluents or excipients and techniques known in the galenic art.
  • oral dosage forms may include tablets, capsules, solutions, suspensions and the like.
  • Phosphodiesterase I B is a calcium/calmodulin dependent phosphodiesterase enzyme that converts cyclic guanosine monophosphate (cGMP) to 5'- guanosine monophosphate (5'-GMP). PDEIB can also convert a modified cGMP substrate, such as the fluorescent molecule cGMP-fluorescein, to the corresponding GMP-fluorescein. The generation of GMP-fluorescein from cGMP-fluorescein can be quantitated, using, for example, the IMAP (Molecular Devices, Sunnyvale, CA) immohili zed-metal affinity particle reagent.
  • IMAP Molecular Devices, Sunnyvale, CA
  • the IMAP reagent binds with high affinity to the free 5'- phosphate that is found in GMP-fluorescein and not in cGMP-fluorescein.
  • the resulting GMP-fluorescein — IMAP complex is large relative to cGMP-fluorescein.
  • Small fluorophores that are bound up in a large, slowly tumbling, complex can be distinguished from unbound fluorophores, because the photons emitted as they fluoresce retain the same polarity as the photons used to excite the fluorescence.
  • Assay The following phosphodiesterase enzymes may be used: 3',5'-cyclic- nucleo tide- specific bovine brain phosphodiesterase (Sigma, St. Louis, MO) (predominantly PDEIB) and recombinant full length human PDE1 A and PDE1B (r- hPDEl A and r-hPDElB respectively) which may be produced e.g., in HEK or SF9 cells by one skilled in the art.
  • the PDE1 enzyme is reconstituted with 50% glycerol to 2.5 U/ml. One unit of enzyme will hydrolyze 1.0 pm of 3',5'-cAMP to 5'-AMP per min at pH 7.5 at 30°C.
  • reaction buffer (30 pM CaCl 2 , 10 U/ml of calmodulin (Sigma P2277), lOmM Tris-HCl pH 7.2, lOmM MgCl 2 , 0.1% BSA, 0.05% NaN ) to yield a final concentration of 1.26mU/ml.
  • 99 pi of diluted enzyme solution is added into each well in a flat bottom 96-well polystyrene plate to which 1 pi of test compound dissolved in 100% DMSO is added. The compounds are mixed and pre-incubated with the enzyme for 10 min at room temperature.
  • the FL-GMP conversion reaction is initiated by combining 4 parts enzyme and inhibitor mix with 1 part substrate solution (0.225 pM) in a 384-well microtiter plate. The reaction is incubated in dark at room temperature for 15 min. The reaction is halted by addition of 60 pL of binding reagent (1:400 dilution of IMAP beads in binding buffer supplemented with 1:1800 dilution of antifoam) to each well of the 384-well plate. The plate is incubated at room temperature for 1 hour to allow IMAP binding to proceed to completion, and then placed in an Envision multimode microplate reader (PerkinElmer, Shelton, CT) to measure the fluorescence polarization (Amp).
  • Envision multimode microplate reader PerkinElmer, Shelton, CT
  • IC50 values are determined by measuring enzyme activity in the presence of 8 to 16 concentrations of compound ranging from 0.0037 nM to 80,000 nM and then plotting drug concentration versus AmP, which allows IC50 values to be estimated using nonlinear regression software (XLFit; IDBS, Cambridge, MA).
  • Compounds of the present disclosure are tested in an assay as described or similarly described herein for PDE1 inhibitory activity.
  • Compounds 1, 2 and 3 are identified as metabolites of a specific PDE1 inhibitor having the following structures:
  • the obtained product (332 mg) is further purified with a semipreparative HPLC system with a gradient of 0 to 30% acetonitrile in water containing 0.1% formic acid over 16 min.
  • the title compound is given as an off-white solid (60 mg).
  • MS (ESI) m/z 510.2071 [M + H] + .
  • a novel radiolabeled PDE1 inhibitor is synthesized according to the following Scheme:
  • This compound is added to a mixture of [ 14 C] aniline, potassium carbonate, Xantphos and tris(dibenzylideneacetone)dipalladium(0), dissolved in 2-methyl-2- butanol (21 mL), frozen and pumped under high vacuum. The mixture was purged with nitrogen, heated, filtered and washed with ethanol.
  • the filtrate was purified firstly by column chromatography on silica eluting with ethyl acetate :ethanol followed by reverse phase high pressure liquid chromatography on a C18 column, eluting with aqueous trifluoroacetic acid:acetonitrile.
  • the mixture was basified using potassium carbonate and was then partitioned between ethyl acetate and water.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

The present invention relates to PDE1 inhibitory compounds useful in the treatment of diseases involving disorders of the dopamine D1 receptor intracellular pathway, such as, among others, Parkinson's disease, depression, narcolepsy, psychosis, damage to cognitive function, e.g., in schizophrenia, or disorders that may be ameliorated through enhanced progesterone-signaling pathway, as well as their use as pharmaceuticals and pharmaceutical compositions comprising them. Methods of making and methods of use related to such compounds are further described.

Description

NOVEL COMPOUNDS
FIELD OF THE INVENTION
[0001] The present invention relates to PDE1 inhibitory compounds of Formula la as described below, processes for their production, their use as pharmaceuticals and pharmaceutical compositions comprising them. These compounds are useful e.g., in the treatment of diseases involving disorders of the dopamine D1 receptor intracellular pathway, such as, among others, Parkinson’s disease, depression, narcolepsy, psychosis, damage to cognitive function, e.g., in schizophrenia, or disorders that may be ameliorated through enhanced progesterone-signaling pathway, e.g., female sexual dysfunction.
BACKGROUND OF THE INVENTION
[0002] Eleven families of phosphodiesterases (PDEs) have been identified but only PDEs in Family I, the Ca2+-calmodulin-dependent phosphodiesterases (CaM-PDEs), are activated by the Ca2+-calmodulin and have been shown to mediate the calcium and cyclic nucleotide (e.g. cAMP and cGMP) signaling pathways. These PDEs are therefore active in stimulated conditions when intra-cellular calcium levels rise, leading to increased hydrolysis of cyclic nucleotides. The three known CaM-PDE genes, PDE1A, PDE1B, and PDE1C, are all expressed in central nervous system tissue. In the brain, the predominant expression of PDE1A is in the cortex and neostriatum, PDE1B is expressed in the neostriatum, prefrontal cortex, hippocampus, and olfactory tubercle, and PDE1C is more ubiquitously expressed.
[0003] Cyclic nucleotide phosphodiesterases decrease intracellular cAMP and cGMP signaling by hydrolyzing these cyclic nucleotides to their respective inactive 5 '-monophosphates (5 'AMP and 5'GMP). CaM-PDEs play a critical role in mediating signal transduction in brain cells, particularly within an area of the brain known as the basal ganglia or striatum. For example, NMDA-type glutamate receptor activation and/or dopamine D2 receptor activation result in increased intracellular calcium concentrations, leading to activation of effectors such as calmodulin-dependent kinase II (CaMKII) and calcineurin and to activation of CaM-PDEs, resulting in reduced cAMP and cGMP. Dopamine D1 receptor activation, on the other hand, leads to activation of nucleotide cyclases, resulting in increased cAMP and cGMP. These cyclic nucleotides in turn activate protein kinase A (PKA; cAMP-dependent protein kinase) and/or protein kinase G (PKG; cGMP-dependent protein kinase) that phosphorylate downstream signal transduction pathway elements such as DARPP-32 (dopamine and cAMP-regulated phosphoprotein) and cAMP responsive element binding protein (CREB). Phosphorylated DARPP-32 in turn inhibits the activity of protein phosphates-1 (PP-1), thereby increasing the state of phosphorylation of substrate proteins such as progesterone receptor (PR), leading to induction of physiologic responses. Studies in rodents have suggested that inducing cAMP and cGMP synthesis through activation of dopamine D1 or progesterone receptor enhances progesterone signaling associated with various physiological responses, including the lordosis response associated with receptivity to mating in some rodents. See Mani, et ah, Science (2000) 287: 1053, the contents of which are incorporated herein by reference.
[0004] CaM-PDEs can therefore affect dopamine-regulated and other intracellular signaling pathways in the basal ganglia (striatum), including but not limited to nitric oxide, noradrenergic, neurotensin, CCK, VIP, serotonin, glutamate (e.g., NMDA receptor, AMPA receptor), GABA, acetylcholine, adenosine (e.g., A2A receptor), cannabinoid receptor, natriuretic peptide (e.g., ANP, BNP, CNP), DARPP-32, and endorphin intracellular signaling pathways.
[0005] Phosphodiesterase (PDE) activity, in particular, phosphodiesterase 1 (PDE1) activity, functions in brain tissue as a regulator of locomotor activity and learning and memory. PDE1 is a therapeutic target for regulation of intracellular signaling pathways, preferably in the nervous system, including but not limited to a dopamine D1 receptor, dopamine D2 receptor, nitric oxide, noradrenergic, neurotensin, CCK, VIP, serotonin, glutamate (e.g., NMDA receptor, AMPA receptor), GABA, acetylcholine, adenosine (e.g., A2A receptor), cannabinoid receptor, natriuretic peptide (e.g., ANP, GNP, CNP), endorphin intracellular signaling pathway and progesterone signaling pathway. For example, inhibition of PDE IB should act to potentiate the effect of a dopamine D1 agonist by protecting cGMP and cAMP from degradation, and should similarly inhibit dopamine D2 receptor signaling pathways, by inhibiting PDE1 activity. Chronic elevation in intracellular calcium levels is linked to cell death in numerous disorders, particularly in neurodegenerative diseases such as Alzheimer's, Parkinson's and Huntington's Diseases and in disorders of the circulatory system leading to stroke and myocardial infarction. PDE1 inhibitors are therefore potentially useful in diseases characterized by reduced dopamine D1 receptor signaling activity, such as Parkinson's disease, restless leg syndrome, depression, narcolepsy and cognitive impairment. PDE1 inhibitors are also useful in diseases that may be alleviated by the enhancement of progesterone- signaling such as female sexual dysfunction.
SUMMARY OF THE INVENTION
[0006] The inventors have unexpectedly discovered that the major route of metabolism of substituted 4,5,7,8-tetrahydro-2H-imidazo[l,2-a]pyrrolo[3,4-e]pyrimidine or 4, 5, 7,8,9- pentahydro-2H-pyrimido[l,2-a]pyrrolo[3,4-e]pyrimidines of Formula la is by way of hydroxylation at the cyclopentyl ring and/or at the phenylamino ring. For example, Compound 1 metabolizes to yield the compounds of Formula M-I, M-II, and M-III, as shown below:
Figure imgf000004_0001
The inventors have further found that the metabolite of Formula M-I accounted for 84% of the total circulating drug related materials in human plasma after oral administration of Compound 1.
[0007] Without being bound by theory, the current invention provides compounds which specifically limit and/or prevent metabolism occurring by these pathways. Due to the very similar properties of deuterium (¾) atoms compared to normal hydrogen atoms ( 'H), drug compounds in which deuterium is substituted for hydrogen are believed to generally have similar biological activity to the non-deuterated analog, but potentially with improved pharmacokinetic properties. The extent to which such a substitution will result in an improvement of pharmacokinetic properties without a too severe loss in pharmacologic activity is variable. Thus, in some circumstances, the resulting deuterated compound only a moderate increase in pharmacokinetic stability, while in other circumstances, the resulting deuterated compound may have significantly improved stability. Moreover, it may be difficult to predict with certainty the effects of simultaneous deuterium substitutions. These may or may not result in additive (synergistic) improvement in metabolic stability.
[0008] In various embodiments, the invention provides various PDE1 inhibitory compounds for use in treatment of any one or more of the following conditions:
(i) Neurodegenerative diseases, including Parkinson’s disease, restless leg, tremors, dyskinesias, Huntington’s disease, Alzheimer’s disease, and drug- induced movement disorders;
(ii) Mental disorders, including depression, attention deficit disorder, attention deficit hyperactivity disorder, bipolar illness, anxiety, sleep disorders, e.g., narcolepsy, cognitive impairment, e.g., cognitive impairment of schizophrenia, dementia, Tourette’s syndrome, autism, fragile X syndrome, psychostimulant withdrawal, and drug addiction;
(iii) Circulatory and cardiovascular disorders, including cerebrovascular disease, stroke, congestive heart disease, hypertension, pulmonary hypertension, e.g., pulmonary arterial hypertension, and sexual dysfunction, including cardiovascular diseases and related disorders as described in International Application No. PCT/US2014/16741, the contents of which are incorporated herein by reference;
(iv) Respiratory and inflammatory disorders, including asthma, chronic obstructive pulmonary disease, and allergic rhinitis, as well as autoimmune and inflammatory diseases;
(v) Diseases that may be alleviated by the enhancement of progesterone- signaling such as female sexual dysfunction; (vi) A disease or disorder such as psychosis, glaucoma, or elevated intraocular pressure;
(vii) Traumatic brain injury;
(viii) Cancers or tumors, e.g., brain tumors, a glioma (e.g., ependymoma, astrocytoma, oligodendrogliomas, brain stem glioma, optic nerve glioma, or mixed gliomas, e.g., oligoastrocytomas), an astrocytoma (e.g., glioblastoma multiforme), osteosarcoma, melanoma, leukemia, neuroblastoma or leukemia;
(ix) Renal disorders, e.g., kidney fibrosis, chronic kidney disease, renal failure, glomerulosclerosis and nephritis;
(x) Any disease or condition characterized by low levels of cAMP and/or cGMP (or inhibition of cAMP and/or cGMP signaling pathways) in cells expressing PDE1; and/or
(xi) Any disease or condition characterized by reduced dopamine D1 receptor signaling activity, comprising administering an effective amount of a Compound of the Invention, e.g., a compound according to any of Formula la or 1.1 - 1.26, in free or pharmaceutically acceptable salt or prodrug form, to a human or animal patient in need thereof.
[0009] In various embodiments, the invention provides a method of preventing the formation of metabolites of the following compound:
Figure imgf000006_0001
the method comprising deuterating the PDE1 inhibitor to block the formation of one or more metabolites.
[0010] Further embodiments of the invention are set forth or evident from the detailed description below and the examples herein. DETAILED DESCRIPTION
Compounds of the Present Disclosure
[0011] In one embodiment, the present disclosure provides that the PDE1 inhibitors for use in the methods as described herein are Formula la:
Figure imgf000007_0001
wherein
(i) R2 and R5 are independently H, D or hydroxy and R3 and R4 together form a tri or tetra-methylene bridge [pref. with the carbons carrying R3 and R4 having the R and S configuration respectively]; or R2 and R3 are each methyl and R4 and R5 are each H; or R2, R4 and R5 are H and R3 is isopropyl [pref. the carbon carrying R3 having the R configuration] ;
(ii) R6 is (optionally halo-substituted or hydroxy-substituted) phenylamino,
(optionally halo-substituted or hydroxy-substituted) benzylamino, Ci-4alkyl, or Ci- 4alkyl sulfide; for example, phenylamino or 4-fluorophenylamino;
(iii) Rio is Ci-4alkyl, methylcarbonyl, hydroxy ethyl, carboxylic acid, sulfonamide, (optionally halo- or hydroxy-substituted) phenyl, (optionally halo- or hydroxy- substituted) pyridyl (for example 6-fluoropyrid-2-yl), or thiadiazolyl (e.g., 1,2,3- thiadiazol-4-yl); and
X and Y are independently C or N, in free, pharmaceutically acceptable salt or prodrug form, including its enantiomers, diastereoisomers and racemates. [0012] The invention further provides compounds of Formula la as follows:
1.1 A compound according to Formula la, wherein R2 and R5 are independently D or hydroxy; or
R6 is hydroxy-substituted phenylamino or hydroxy-substituted benzylamino.
1.2 A compound according to Formula la or 1.1, wherein R2 and R5 are independently D or hydroxy.
1.3 A compound according to Formula la or 1.1 -1.2, wherein R6 is hydroxy- substituted phenylamino or hydroxy-substituted benzylamino.
1.4 A compound according to Formula la or 1.1 -1.3, wherein R6 is hydroxy- substituted phenylamino.
1.5 A compound according to Formula la or 1.1 -1.3, wherein R6 is hydroxy- substituted benzylamino.
1.6 A compound according to Formula la or 1.1 -1.5, wherein R2 and R5 are independently H, D or hydroxy and R3 and R4 together form a tri- or tetra- methylene bridge [pref. with the carbons carrying R3 and R4 having the R and S configuration respectively] .
1.7 A compound according to Formula la or 1.1-1.6, wherein at least one of R2 and R5 are D.
1.8 A compound according to Formula la or 1.1-1.7, wherein R2 and R5 are both D.
1.9 A compound according to Formula la or 1.1-1.8, wherein R2 and R5 are both D and R3 and R4 together form a tri- or tetra-methylene bridge.
1.10 A compound according to Formula la or 1.1-1.5, wherein at least one of R2 and R5 are hydroxy.
1.11 A compound according to Formula la, 1.1- 1.5 or 1.10, wherein at least one of R2 and R5 are hydroxy and R3 and R4 together form a tri- or tetra-methylene bridge.
1.12 A compound according to Formula la, 1.1-1.5 or 1.10-1.11, wherein R2 is hydroxy.
1.13 A compound according to Formula la, 1.1-1.5 or 1.0-1.11, wherein R5 is hydroxy.
1.14 A compound according to Formula la or 1.1-1.11, wherein R 10 is pyridy 1.
1.15 A compound according to Formula la or 1.1-1.12, wherein Riois halo- or hydroxy-substituted pyridyl. A compound according to Formula la or 1.1-1.12, wherein Riois halo-substituted pyridyl. A compound according to Formula la or 1.1-1.12, wherein Riois hydroxy- substituted pyridyl. A compound according to Formula la or 1.1-1.12, wherein Riois 6-fluoropyrid-2- yi- A compound according to Formula la or 1.1-1.18, wherein X and Y are both C. A compound according to Formula la or 1.1-1.19, wherein the PDE1 inhibitor is a compound according to the following:
Figure imgf000009_0001
Figure imgf000010_0001
1.21 A compound according to Formula la or 1.1-1.20, wherein the PDE1 inhibitor is a compound according to the following:
Figure imgf000010_0002
Figure imgf000011_0001
A compound according to Formula la or 1.1-1.21, wherein the PDE1 inhibitor is a compound according to the following:
Figure imgf000011_0002
A compound according to Formula la or 1.1-1.22, wherein the PDE1 inhibitor is a compound according to the following:
Figure imgf000012_0001
A compound according to Formula la or 1.1-1.22, wherein the PDE1 inhibitor is a compound according to the following:
Figure imgf000012_0002
A compound according to Formula la or 1.1-1.22, wherein the PDE1 inhibitor is a compound according to the following:
Figure imgf000012_0003
A compound according to Formula la or 1.1-1.21, wherein the PDE1 inhibitor is a compound according to the following:
Figure imgf000013_0001
1.27 Any foregoing compound, wherein the compound is deuterated, e.g., wherein the deuteriurmprotium ratio at a specified position in the molecule is significantly higher, e.g., at least 2x, for example at least lOx higher, than the natural isotope ratios.
[0013] In another embodiment, the present disclosure further provides for a radiolabeled PDE1 inhibitor [Compound 2], e.g., for use in the methods as described herein, according to Formula la or 1.1-1.26.
2.1 Compound 2, wherein at least one carbon atom substituted with Carbon- 14 ([14C]).
2.2 Any of the preceding Compounds, wherein one carbon atom substituted with Carbon-14 ([14C]).
2.3 Any of the preceding Compounds, wherein the radiolabeled PDE1 inhibitor is a compound according to the following:
Figure imgf000014_0001
or a pharmaceutically acceptable salt thereof.
2.4 Compound 2.3, wherein the compound is in salt form.
2.5 Compound 2.4, wherein the compound is in phosphate salt form.
2.6 Compound 2.5, wherein the compound is in mono-phosphate salt form.
[0014] In one embodiment, selective PDE1 inhibitors of the preceding formula (e.g., Formula la or 1.1-1.26) are compounds that inhibit phosphodiesterase-mediated (e.g., PDE1- mediated, especially PDE IB -mediated) hydrolysis of cGMP, e.g., the preferred compounds have an IC50 of less than ImM, preferably less than 500 nM, preferably less than 50 nM, and preferably less than 5nM in an immohili zed-metal affinity particle reagent PDE assay, in free or salt form.
[0015] If not otherwise specified or clear from context, the following terms herein have the following meanings:
“Alkyl” as used herein is a saturated or unsaturated hydrocarbon moiety, preferably saturated, preferably having one to six carbon atoms, which may be linear or branched, and may be optionally mono-, di- or tri- substituted, e.g., with halogen (e.g., chloro or fluoro), hydroxy, or carboxy.
“Cycloalkyl” as used herein is a saturated or unsaturated nonaromatic hydrocarbon moiety, preferably saturated, preferably comprising three to nine carbon atoms, at least some of which form a nonaromatic mono- or bicyclic, or bridged cyclic structure, and which may be optionally substituted, e.g., with halogen (e.g., chloro or fluoro), hydroxy, or carboxy. Wherein the cycloalkyl optionally contains one or more atoms selected from N and O and/or S, said cycloalkyl may also be a heterocycloalkyl.
“Heterocycloalkyl” is, unless otherwise indicated, saturated or unsaturated nonaromatic hydrocarbon moiety, preferably saturated, preferably comprising three to nine carbon atoms, at least some of which form a nonaromatic mono- or bicyclic, or bridged cyclic structure, wherein at least one carbon atom is replaced with N, O or S, which heterocycloalkyl may be optionally substituted, e.g., with halogen (e.g., chloro or fluoro), hydroxy, or carboxy.
“Aryl” as used herein is a mono or bicyclic aromatic hydrocarbon, preferably phenyl, optionally substituted, e.g., with alkyl (e.g., methyl), halogen (e.g., chloro or fluoro), haloalkyl (e.g., trifluoromethyl), hydroxy, carboxy, or an additional aryl or heteroaryl (e.g., biphenyl or pyridylphenyl).
“Heteroaryl” as used herein is an aromatic moiety wherein one or more of the atoms making up the aromatic ring is sulfur or nitrogen rather than carbon, e.g., pyridyl or thiadiazolyl, which may be optionally substituted, e.g., with alkyl, halogen, haloalkyl, hydroxy or carboxy.
[0016] It is intended that wherein the substituents end in “ene”, for example, alkylene, phenylene or arylalkylene, said substituents are intended to bridge or be connected to two other substituents. Therefore, methylene is intended to be -CH2- and phenylene intended to be - C6H4- and arylalkylene is intended to be -C6H4-CH2- or - CH2-C6H4-.
[0017] Compounds of the Invention, e.g., substituted 4,5,7,8-tetrahydro-2H-imidazo[l,2- a]pyrrolo[3,4-e]pyrimidine or 4,5,7,8,9-pentahydro-2H-pyrimido[l,2-a]pyrrolo[3,4-e]pyrimidine, e.g., Compounds of Formula la, may exist in free or salt form, e.g., as acid addition salts. In this specification unless otherwise indicated, language such as “Compounds of the Invention” is to be understood as embracing the compounds in any form, for example free or acid addition salt form, or where the compounds contain acidic substituents, in base addition salt form. The Compounds of the Invention are intended for use as pharmaceuticals, therefore pharmaceutically acceptable salts are preferred. Salts which are unsuitable for pharmaceutical uses may be useful, for example, for the isolation or purification of free Compounds of the Invention or their pharmaceutically acceptable salts, are therefore also included.
[0018] Compounds of the Invention may in some cases also exist in prodrug form. A prodrug form is compound which converts in the body to a Compound of the Invention. For example when the Compounds of the Invention contain hydroxy or carboxy substituents, these substituents may form physiologically hydroly sable and acceptable esters. As used herein, “physiologically hydrolysable and acceptable ester” means esters of Compounds of the Invention which are hydrolysable under physiological conditions to yield acids (in the case of Compounds of the Invention which have hydroxy substituents) or alcohols (in the case of Compounds of the Invention which have carboxy substituents) which are themselves physiologically tolerable at doses to be administered. Therefore, wherein the Compound of the Invention contains a hydroxy group, for example, Compound-OH, the acyl ester prodrug of such compound, i.e., Compound- 0-C(0)-Ci-4alkyl, can hydrolyze in the body to form physiologically hydrolysable alcohol (Compound-OH) on the one hand and acid on the other (e.g., H0C(0)-Ci-4alkyl). Alternatively, wherein the Compound of the Invention contains a carboxylic acid, for example, Compound- C(0)0H, the acid ester prodrug of such compound, Compound-C(0)0-Ci-4alkyl can hydrolyze to form Compound-C(0)OH and HO-Ci-4alkyl. As will be appreciated the term thus embraces conventional pharmaceutical prodrug forms.
[0019] In another embodiment, the invention further provides a pharmaceutical composition comprising a Compound of the Invention, in free or pharmaceutically acceptable salt form, in admixture with a pharmaceutically acceptable carrier, for use in the treatment of a disease or disorder mediated by PDE1.
[0020] Compounds of the Invention may in some cases also exist in prodrug form. A prodrug form is compound which converts in the body to a Compound of the Invention. For example when the Compounds of the Invention contain hydroxy or carboxy substituents, these substituents may form physiologically hydrolysable and acceptable esters. As used herein, “physiologically hydrolysable and acceptable ester” means esters of Compounds of the Invention which are hydrolysable under physiological conditions to yield acids (in the case of Compounds of the Invention which have hydroxy substituents) or alcohols (in the case of Compounds of the Invention which have carboxy substituents) which are themselves physiologically tolerable at doses to be administered. Therefore, wherein the Compound of the Invention contains a hydroxy group, for example, Compound-OH, the acyl ester prodrug of such compound, i.e., Compound- 0-C(0)-Ci-4alkyl, can hydrolyze in the body to form physiologically hydroly sable alcohol (Compound-OH) on the one hand and acid on the other (e.g., H0C(0)-Ci-4alkyl). Alternatively, wherein the Compound of the Invention contains a carboxylic acid, for example, Compound- C(0)0H, the acid ester prodrug of such compound, Compound-C(0)0-Ci-4alkyl can hydrolyze to form Compound-C(0)OH and HO-Ci-4alkyl. As will be appreciated the term thus embraces conventional pharmaceutical prodrug forms.
[0021] In another embodiment, the invention further provides a pharmaceutical composition comprising a Compound of the Invention, in free, pharmaceutically acceptable salt or prodrug form, in admixture with a pharmaceutically acceptable carrier, for use in the treatment of a disease or disorder mediated by PDE1.
Methods of Making Compounds of the Invention
[0022] The compounds of the Invention and their pharmaceutically acceptable salts may be made using the methods as described and exemplified herein and by methods similar thereto and by methods known in the chemical art. Such methods include, but not limited to, those described below. If not commercially available, starting materials for these processes may be made by procedures, which are selected from the chemical art using techniques which are similar or analogous to the synthesis of known compounds.
[0023] Various starting materials and/or Compounds of the Invention may be prepared using methods described in US 2008-0188492 Al, US 2010-0173878 Al, US 2010-0273754 Al, US 2010-0273753 Al, WO 2010/065153, WO 2010/065151, WO 2010/065151, WO 2010/065149, WO 2010/065147, WO 2010/065152, WO 2011/153129, WO 2011/133224, WO 2011/153135, WO 2011/153136, WO 2011/153138, and US Pat. 9,073,936, the contents of each of which herein are hereby incorporated by reference in their entireties.
[0024] The Compounds of the Invention include their enantiomers, diastereoisomers and racemates, as well as their polymorphs, hydrates, solvates and complexes. Some individual compounds within the scope of this invention may contain double bonds. Representations of double bonds in this invention are meant to include both the E and the Z isomer of the double bond. In addition, some compounds within the scope of this invention may contain one or more asymmetric centers. This invention includes the use of any of the optically pure stereoisomers as well as any combination of stereoisomers.
[0025] It is also intended that the Compounds of the Invention encompass their stable and unstable isotopes. Stable isotopes are nonradioactive isotopes which contain one additional neutron compared to the abundant nuclides of the same species (i.e., element). It is expected that the activity of compounds comprising such isotopes would be retained, and such compound would also have utility for measuring pharmacokinetics of the non-isotopic analogs. For example, the hydrogen atom at a certain position on the Compounds of the Invention may be replaced with deuterium (a stable isotope which is non-radioactive). Examples of known stable isotopes include, but not limited to, deuterium, 13 C, 15 N, 18 O. Alternatively, unstable isotopes, which are radioactive isotopes which contain additional neutrons compared to the abundant nuclides of the same species (i.e., element), e.g., 1231, 131I, 125I, UC, 18F, may replace the corresponding abundant species of I, C and F. Another example of useful isotope of the compound of the invention is the UC isotope. These radio isotopes are useful for radio-imaging and/or pharmacokinetic studies of the compounds of the invention.
[0026] Melting points are uncorrected and (dec) indicates decomposition. Temperature are given in degrees Celsius (°C); unless otherwise stated, operations are carried out at room or ambient temperature, that is, at a temperature in the range of 18-25 °C. Chromatography means flash chromatography on silica gel; thin layer chromatography (TEC) is carried out on silica gel plates. NMR data is in the delta values of major diagnostic protons, given in parts per million (ppm) relative to tetramethylsilane (TMS) as an internal standard. Conventional abbreviations for signal shape are used. Coupling constants (J) are given in Hz. For mass spectra (MS), the lowest mass major ion is reported for molecules where isotope splitting results in multiple mass spectral peaks. Solvent mixture compositions are given as volume percentages or volume ratios. In cases where the NMR spectra are complex, only diagnostic signals are reported. Methods of using Compounds of the Invention
[0027] The Compounds of the Invention are useful in the treatment of diseases characterized by disruption of or damage to cAMP and cGMP mediated pathways, e.g., as a result of increased expression of PDE1 or decreased expression of cAMP and cGMP due to inhibition or reduced levels of inducers of cyclic nucleotide synthesis, such as dopamine and nitric oxide (NO). By preventing the degradation of cAMP and cGMP by PDE1, thereby increasing intracellular levels of cAMP and cGMP, the Compounds of the Invention potentiate the activity of cyclic nucleotide synthesis inducers.
[0028] The invention provides methods of treatment of any one or more of the following conditions:
(i) Neurodegenerative diseases, including Parkinson’s disease, restless leg, tremors, dyskinesias, Huntington’s disease, Alzheimer’s disease, and drug-induced movement disorders;
(ii) Mental disorders, including depression, attention deficit disorder, attention deficit hyperactivity disorder, bipolar illness, anxiety, sleep disorders, e.g., narcolepsy, cognitive impairment, e.g., cognitive impairment of schizophrenia, dementia, Tourette’s syndrome, autism, fragile X syndrome, psychostimulant withdrawal, and drug addiction;
(iii) Circulatory and cardiovascular disorders, including cerebrovascular disease, stroke, congestive heart disease, hypertension, pulmonary hypertension, e.g., pulmonary arterial hypertension, and sexual dysfunction, including cardiovascular diseases and related disorders as described in International Application No. PCT/US2014/16741, the contents of which are incorporated herein by reference;
(iv) Respiratory and inflammatory disorders, including asthma, chronic obstructive pulmonary disease, and allergic rhinitis, as well as autoimmune and inflammatory diseases;
(v) Diseases that may be alleviated by the enhancement of progesterone- signaling such as female sexual dysfunction; (vi) A disease or disorder such as psychosis, glaucoma, or elevated intraocular pressure;
(vii) Traumatic brain injury;
(viii) Any disease or condition characterized by low levels of cAMP and/or cGMP (or inhibition of cAMP and/or cGMP signaling pathways) in cells expressing PDE1; and/or
(ix) Any disease or condition characterized by reduced dopamine D1 receptor signaling activity, comprising administering an effective amount of a Compound of the Invention, e.g., a compound according to any of Formula la or 1.1-1.26, in free or pharmaceutically acceptable salt or prodrug form, to a human or animal patient in need thereof.
[0029] The Compounds of the Invention are useful in the treatment of inflammatory diseases or conditions, particularly neuroinflammatory diseases or conditions. Therefore, administration or use of a preferred PDE1 inhibitor as described herein, e.g., a PDE1 inhibitor as hereinbefore described, e.g., a Compound of Formula la, provides a means to regulate inflammation (e.g., prevent, reduce, and/or reverse neuroinflammation, and diseases or disorders related to neuro inflammation), and in certain embodiments provide a treatment for various inflammatory diseases and disorders. In certain embodiments, the inflammatory disease or condition is selected from: a. neurodegenerative conditions such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and demyelinating conditions, e.g., multiple sclerosis (MS), and prion diseases; b. stroke, cardiac arrest, hypoxia, intracerebral hemorrhage or traumatic brain injury; c. conditions characterized by abnormal neurotransmitter production and/or response, including depression, schizophrenia, post-traumatic stress disorder, anxiety, attention deficit disorder, and bipolar disease; e.g., wherein any of the foregoing are associated with neuroinflammation; and d. chronic CNS infections, e.g., Lyme disease or CNS infection consequent to an immunosuppressive condition, e.g., HIV-dementia; e. neuroinflammation consequent to chemotherapy; comprising administering an effective amount of a PDE1 inhibitor of the current invention (e.g., a PDE1 inhibitor of Formula la as herein described), e.g., an amount effective to (i) reduce or inhibit activation of Ml microglia, and/or (ii) and amount effective to reduce levels of one or more pro-inflammatory cytokines (e.g., IEIb, TNF- a, and Ccl2, or combination thereof); to a patient in need thereof.
[0030] The Compounds of the Invention are useful in the treatment of cancers or tumors, e.g., in the inhibition of the proliferation of cancerous or tumorous cells. Therefore, administration or use of a preferred PDE1 inhibitor as described herein, e.g., a PDE1 inhibitor as hereinbefore described, e.g., a Compound of Formula la, in the treatment or prevention of a cancer or tumor.
[0031] It is contemplated that the compounds of the present disclosure can be used in the treatment of a tumor or cancer selected from one or more of acoustic neuroma, astrocytoma, chordoma, lymphoma (e.g., CNS lymphoma, Hodgkin’s lymphoma or non-Hodgkin’s lymphoma), craniopharyngioma, gliomas (e.g., Brain stem glioma, ependymoma, mixed glioma, optic nerve glioma), subependymoma, medulloblastoma, meningioma, metastatic brain tumors, oligodendroglioma, pituitary tumors, primitive neuroectodermal (PNET), schwannoma, adenomas (e.g., basophilic adenoma, eosinophilic adenoma, chromophobe adenoma, parathyroid adenoma, islet adenoma, fibroadenoma), fibroids (fibrous histiocytoma), fibromas, hemangiomas, lipomas (e.g., angiolipoma, myelolipoma, fibrolipoma, spindle cell lipoma, hibernoma, atypical lipoma), myxoma, osteoma, preleukemias, rhabdomyoma, papilloma, seborrheic keratosis, skin adnexal tumors, hepatic adenomas, renal tubular adenoma, bile duct adenoma, transitional cell papilloma, hydatidiform moles, ganglioneuroma, meningioma, neurilemmoma, neurofibroma, C cell hyperplasia, pheochromocytoma, insulinoma, gastrinoma, carcinoids, chemodectoma, paraganglioma, nevus, actinic keratosis, cervical dysplasia, metaplasia (e.g., metaplasia of the lung), leukoplakia, hemangioma, lymphangioma, carcinoma (e.g., squamous cell carcinoma, epidermoid carcinoma, adenocarcinoma, hepatoma, hepatocellular carcinoma, renal cell carcinoma, cholangiocarcinoma, transitional cell carcinoma, embryonal cell carcinoma, parathyroid carcinoma, medullary carcinoma of thyroid, bronchial carcinoid, oat cell carcinoma, islet cell carcinoma, malignant carcinoid, Merkel cell carcinoma), sarcoma (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, malignant fibrous histiocytoma, hemangiosarcoma, angiosarcoma, lymphangiosarcoma, leiomyosarcoma, rhabdomyosarcoma, neurofibrosarcoma), blastoma (e.g., medulloblastoma and glioblastoma, types of brain tumor, retinoblastoma, a tumor in the retina of the eye, osteoblastoma, bone tumors, neuroblastoma), germ cell tumor, mesothelioma, malignant skin adnexal tumors, hypernephroma, seminoma, glioma, malignant meningioma, malignant schwannoma, malignant pheochromocytoma, malignant paraganglioma, melanoma, Merkel cell neoplasm, cystosarcoma phylloides, or Wilms tumor. Further treatments include lung cancer, pancreatic cancer, prostate cancer, urothelial cancer, cancers of the head and neck, or leukemia (e.g., a lymphocytic leukemia or a myelogenous leukemia), colon cancer (e.g., colorectal cancer) and cancers of the kidney, ureter, bladder or urethra.
[0032] The current invention also provides compounds which specifically limit and/or prevent metabolism of PDE1 inhibitors, as well as related methods. Therefore, in one embodiment the invention provides a method (Method 1) of inhibiting the metabolism of a PDE1 inhibitor, e.g. a PDE1 inhibitor according to Formula la or 1.1-1.26, the method comprising deuterating the PDE1 inhibitor to block the formation of one or more metabolites. . Method 1, wherein the PDE1 inhibitor is a compound according to the following formula:
Figure imgf000022_0001
in free or pharmaceutically acceptable salt form. . Any of the preceding Methods, wherein deuterating the PDE1 inhibitor comprises reacting:
Figure imgf000023_0001
1.3. The preceding Method, wherein deuterating the PDE1 inhibitor comprises reacting:
Figure imgf000023_0002
Figure imgf000024_0001
The preceding Method, wherein the reaction is carried out in benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate, THF and 1,8- Diazabicyclo[5.4.0]undec-7-ene.
The two preceding Methods, wherein the
Figure imgf000024_0002
is reacted with thionyl chloride in THF to form
Figure imgf000025_0001
Method 1.2, wherein the reaction is carried out in benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate, THF and 1,8- Diazabicyclo[5.4.0]undec-7-ene. Method 1.2 or 1.6, wherein the
Figure imgf000025_0002
is reacted with thionyl chloride in THF to form the following product:
Figure imgf000025_0003
and subjecting the product to chiral column separation to obtain:
Figure imgf000026_0001
[0033] The phrase “Compounds of the Invention” or “PDE 1 inhibitors of the Invention”, or like terms, encompasses any and all of the compounds disclosed herewith, e.g., a Compound of Formula la or 1.1-1.26.
[0034] The words "treatment" and "treating" are to be understood accordingly as embracing prophylaxis and treatment or amelioration of symptoms of disease as well as treatment of the cause of the disease.
[0035] For methods of treatment, the word “effective amount” is intended to encompass a therapeutically effective amount to treat or mitigate a specific disease or disorder, and/or a symptom thereof, and/or to inhibit PDE1 expression in a patient or subject.
[0036] The term “patient” includes a human or non-human (i.e., animal) patient. In a particular embodiment, the invention encompasses both humans and nonhuman animals. In another embodiment, the invention encompasses nonhuman animals. In other embodiments, the term encompasses humans.
[0037] The term “comprising” as used in this disclosure is intended to be open-ended and does not exclude additional, unrecited elements or method steps.
[0038] Compounds of the Invention, e.g., Formula la and 1.1-1.26 as hereinbefore described, in free or pharmaceutically acceptable salt form, may be used as a sole therapeutic agent, but may also be used in combination or for co-administration with other active agents. [0039] For example, in certain embodiments, the Compounds of the Invention, e.g., Formula la or 1.1-1.26 as hereinbefore described, in free or pharmaceutically acceptable salt form, may be administered in combination (e.g. administered sequentially or simultaneously or within a 24 hour period) with other active agents, e.g., with one or more antidepressant agents, e.g., with one or more compounds in free or pharmaceutically acceptable salt form, selected from selective serotonin reuptake inhibitors (SSRIs) serotonin-norepinephrine reuptake inhibitors (SNRIs), c) tricyclic antidepressants (TCAs), and atypical antipsychotics. [0040] Dosages employed in practicing the present invention will of course vary depending, e.g. on the particular disease or condition to be treated, the particular Compound of the Invention used, the mode of administration, and the therapy desired. Compounds of the Invention may be administered by any suitable route, including orally, parenterally, transdermally, or by inhalation, but are preferably administered orally. In general, satisfactory results, e.g. for the treatment of diseases as hereinbefore set forth are indicated to be obtained on oral administration at dosages of the order from about 0.01 to 2.0 mg/kg. In larger mammals, for example humans, an indicated daily dosage for oral administration will accordingly be in the range of from about 0.75 to 150 mg (depending on the drug to be administered and the condition to be treated, for example in the case of Compound 214, 0.5 to 25 mg, e.g., 1 to 10 mg, per diem, e.g., in monophosphate salt form, for treatment of PDE1- mediated conditions), conveniently administered once, or in divided doses 2 to 4 times, daily or in sustained release form. Unit dosage forms for oral administration thus for example may comprise from about 0.2 to 75 or 150 mg, e.g. from about 0.2 or 2.0 to 50, 75 or 100 mg (e.g., 1, 2.5, 5, 10, or 20 mg) of a Compound of the Invention, e.g., together with a pharmaceutically acceptable diluent or carrier therefor.
[0041] Pharmaceutical compositions comprising Compounds of the Invention may be prepared using conventional diluents or excipients and techniques known in the galenic art. Thus oral dosage forms may include tablets, capsules, solutions, suspensions and the like.
EXAMPLE 1
Measurement of PDEIB inhibition in vitro using IMAP Phosphodiesterase Assay Kit
[0042] Phosphodiesterase I B (PDEIB) is a calcium/calmodulin dependent phosphodiesterase enzyme that converts cyclic guanosine monophosphate (cGMP) to 5'- guanosine monophosphate (5'-GMP). PDEIB can also convert a modified cGMP substrate, such as the fluorescent molecule cGMP-fluorescein, to the corresponding GMP-fluorescein. The generation of GMP-fluorescein from cGMP-fluorescein can be quantitated, using, for example, the IMAP (Molecular Devices, Sunnyvale, CA) immohili zed-metal affinity particle reagent.
[0043] Briefly, the IMAP reagent binds with high affinity to the free 5'- phosphate that is found in GMP-fluorescein and not in cGMP-fluorescein. The resulting GMP-fluorescein — IMAP complex is large relative to cGMP-fluorescein. Small fluorophores that are bound up in a large, slowly tumbling, complex can be distinguished from unbound fluorophores, because the photons emitted as they fluoresce retain the same polarity as the photons used to excite the fluorescence.
[0044] In the phosphodiesterase assay, cGMP-fluorescein, which cannot be bound to IMAP, and therefore retains little fluorescence polarization, is converted to GMP-fluorescein, which, when bound to IMAP, yields a large increase in fluorescence polarization (Amp). Inhibition of phosphodiesterase, therefore, is detected as a decrease in Amp.
Enzyme assay
[0045] Materials: All chemicals are available from Sigma- Aldrich (St. Louis, MO) except for IMAP reagents (reaction buffer, binding buffer, FL-GMP and IMAP beads), which are available from Molecular Devices (Sunnyvale, CA).
[0046] Assay: The following phosphodiesterase enzymes may be used: 3',5'-cyclic- nucleo tide- specific bovine brain phosphodiesterase (Sigma, St. Louis, MO) (predominantly PDEIB) and recombinant full length human PDE1 A and PDE1B (r- hPDEl A and r-hPDElB respectively) which may be produced e.g., in HEK or SF9 cells by one skilled in the art. The PDE1 enzyme is reconstituted with 50% glycerol to 2.5 U/ml. One unit of enzyme will hydrolyze 1.0 pm of 3',5'-cAMP to 5'-AMP per min at pH 7.5 at 30°C. One part enzyme is added to 1999 parts reaction buffer (30 pM CaCl 2 , 10 U/ml of calmodulin (Sigma P2277), lOmM Tris-HCl pH 7.2, lOmM MgCl 2 , 0.1% BSA, 0.05% NaN ) to yield a final concentration of 1.26mU/ml. 99 pi of diluted enzyme solution is added into each well in a flat bottom 96-well polystyrene plate to which 1 pi of test compound dissolved in 100% DMSO is added. The compounds are mixed and pre-incubated with the enzyme for 10 min at room temperature.
[0047] The FL-GMP conversion reaction is initiated by combining 4 parts enzyme and inhibitor mix with 1 part substrate solution (0.225 pM) in a 384-well microtiter plate. The reaction is incubated in dark at room temperature for 15 min. The reaction is halted by addition of 60 pL of binding reagent (1:400 dilution of IMAP beads in binding buffer supplemented with 1:1800 dilution of antifoam) to each well of the 384-well plate. The plate is incubated at room temperature for 1 hour to allow IMAP binding to proceed to completion, and then placed in an Envision multimode microplate reader (PerkinElmer, Shelton, CT) to measure the fluorescence polarization (Amp).
[0048] A decrease in GMP concentration, measured as decreased Amp, is indicative of inhibition of PDE activity. IC50 values are determined by measuring enzyme activity in the presence of 8 to 16 concentrations of compound ranging from 0.0037 nM to 80,000 nM and then plotting drug concentration versus AmP, which allows IC50 values to be estimated using nonlinear regression software (XLFit; IDBS, Cambridge, MA).
[0049] Compounds of the present disclosure are tested in an assay as described or similarly described herein for PDE1 inhibitory activity. For example, Compounds 1, 2 and 3 are identified as metabolites of a specific PDE1 inhibitor having the following structures:
Figure imgf000029_0001
Compound 2
Figure imgf000030_0001
Compound 3
These compounds have efficacy at nanomolar or sub-nanomolar levels vs PDE1 and generally high selectivity over other PDE families, as depicted on the following table:
Figure imgf000030_0002
Figure imgf000031_0002
EXAMPLE 2
Synthesis of (6aS, 9aR)-6a, 9a-dideuterio-2-(4-(6-fluoropyridin-2-yl)benzyl)-5-methyl-3- (phenylamino)-5, 6a, 7, 8, 9, 9a-hexahydrocyclopenta[4, 5 ]imidazo[l,2-a ]pyrazolo[4, 3 -e Jpyrimidin- 4(2H)-one
[0050] The title compound is generally carried out according to Scheme 1.
Scheme 1
Figure imgf000031_0001
2-(hydroxyimino)cyclopentanone (2).To a solution of NaOH (2.816 g, 70.42 mmol) in water (30 mL) at 0°C is sequentially added ethyl 2-oxocyclopentanecarboxylate (10.0 g, 64.02mmol) and a solution of sodium nitrite (4.417 g, 64.02 mmol) in water (25 mL) dropwise over a period of 20 minutes. The reaction mixture is stirred at room temperature for 2 days and cooled to 0°C. The cooled mixture is acidified to pH= 5 with6 N H2SO4 (8 mL, 48.0 mmol). Diethyl ether (100 mL) is added and the organic layer is separated. The aqueous phase is extracted with diethyl ether (2 x 100 mL). The extracts are combined and condensed under reduced pressure. The residue is further purified by silica-gel column chromatography using a gradient of 0-100% ethyl acetate in hexane as eluent. The title compound 2 is given as a light brown solid (2.41 g, 33%).MS (ESI) ml 7 [M + H]+. NMR (500 MHz, Chloroform- ) d 2.83 (t, / = 7.5 Hz, 2H), 2.49 (t, / = 7.9 Hz, 2H), 2.12 - 2.03 (m, 2H).
2-amino-l,2-dideuteriocyclopentanol (3). To a solution of 2-(hydroxyimino)cyclopentanone 2 (1.16 g, 10.26 mmol) in CH30D(7 mL) at 0°C is added NaBD4 (646 mg, 15.38 mmol) in small portions. The resulting mixture is stirred at 0°C for 1 h and NiCh is added, followed by NaBD4 (646 mg, 15.38 mmol) in small portions. The mixture is stirred at 0°C for 0.5 h and at room temperature for 2h. The solvent is removed under reduced pressure and the residue is extracted with a mixture of CH2CI2 and CH3OH (10 : 1, 3 x 70 mL). The combined extracts are evaporated and further dried under high vacuum to give the title compound 3 as a light pink solid (0.88 g, crude).MS (ESI) ml 7. 104.04 [M + H]+. This crude product is directly used for the next reaction without further purification.
3-chloro-2-(4-(6-fluoropyridin-2-yl)benzyl)-7-(4-methoxybenzyl)-5-methyl-2H- pyrazolo[3,4-d]pyrimidine-4,6(5H,7H)-dione (5) A solution of 2-(4-(6-fluoropyridin-2- yl)benzyl)-7-(4-methoxybenzyl)-5-methyl-2H-pyrazolo[3,4-d]pyrimidine-4,6(5H,7H)-dione 4 (9.04 g, 38.2 mmol) in THF (lOOmL) is cooled to 0 °C under argon, and lithium bis(trimethylsilyl)amide in toluene (1M, 25.5 mL, 25.5 mmol) is added dropwise via a syringe. The reaction mixture is stirred at 0 °C for 4 h and quenched with water (30 mL). The mixture is stirred at room temperature for 5 min and the precipitate is filtered. The filtered cake is washed with water (lOOmL) and then suspended in ethyl acetate (lOOmL). The mixture is filtered, and the filtered cake is dried under high vacuum to give the title compound as a white solid (4.78g, 45% yield). MS (ESI) mlz 506.14 [M + H]+.
2-(4-(6-fluoropyridin-2-yl)benzyl)-7-(4-methoxybenzyl)-5-methyl-3-(phenylamino)-2H- pyrazolo[3,4-d]pyrimidine-4,6(5H,7H)-dione (6). A suspension of 3-chloro-2-(4-(6- fluoropyridin-2-yl)benzyl)-7-(4-methoxybenzyl)-5-methyl-2H-pyrazolo[3,4-d]pyrimidine- 4,6(5H,7H)-dione 5 (4.37 g, 8.65 mmol), aniline (1.18 mL, 13 mmol), cesium carbonate (5.63 g, 17.3 mmol), XPhos (1.6 g, 3.37 mmol), and tris(dibenzylideneacetone)dipalladium(0) (2.37 g, 2.6 mmol) in DMF (40mL) is bubbled with argon at room temperature for 5 mins, and heated up to 100 °C .The mixture is stirred at this temperature for 5 h until the reaction is completed. The solvent is evaporated and the residue is purified by silica-gel column chromatography with a gradient of 0-50%ethyl acetate in hexane. The title compound is given as an off-white solid (4.34g, 89% yield). MS (ESI) mlz 563.2019 [M + H]+.
2-(4-(6-fluoropyridin-2-yl)benzyl)-5-methyl-3-(phenylamino)-2H-pyrazolo[3,4- d]pyrimidine-4,6(5H,7H)-dione (7). To a solution of 2-(4-(6-fluoropyridin-2-yl)benzyl)-7-(4- methoxybenzyl)-5-methyl-3-(phenylamino)-2H-pyrazolo[3,4-d]pyrimidine-4,6(5H,7H)-dione 6 (3.44g, 6.1 lmmol) in DCM (6 mL) at room temperature under argon is added trifluoroacetic acid (6.09mL, 79.4mmol), followed by trifluoromethanesulfonic acid (1.96mL, 22mmol). The resulting solution is stirred at room temperature for 2 h, and the solvents are removed under reduced pressure. The residue is neutralized with 7 N ammonium in methanol, and the mixture is evaporated to dryness. To the residue is added ethyl acetate (30mL) and the resulting mixture is filtered. The filtered cake is washed with water (60mL) and dried under high vacuum to generate the title compound 1.21 g. The filtrates are combined and evaporated to dryness. The residue is suspended in methanol (60mL) and the suspension is filtered. The filtered cake is dried under vacuum to afford the title compound 0.94g. (total 2.15g, 79% yield). MS (ESI) m/z443.1474 [M + H]+. NMR (500 MHz, DMSO -d6) d 11.52 (s, 1H), 8.77 (s, 1H), 8.12 - 8.03 (m, 1H), 7.99 (d, 2H), 7.90 (dd, / = 7.6, 2.6 Hz, 1H), 7.25 (d, 2H), 7.18 (dd, / = 8.5, 7.2 Hz, 2H), 7.13 (dd, / = 8.1, 2.7 Hz, 1H), 6.91 - 6.75 (m, 3H), 5.27 (s, 2H), 3.34 (s, 2H), 3.08 (s, 3H), 2.63 - 2.41 (m, 2H). 6-((l,2-dideuterio-2-hydroxycyclopentyl)amino)-2-(4-(6-fluoropyridin-2-yl)benzyl)-5- methyl-3-(phenylamino)-2H-pyrazolo[3,4-d]pyrimidin-4(5H)-one (8). To a suspension of 2- (4-(6-fluoropyridin-2-yl)benzyl)-5-methyl-3-(phenylamino)-2H-pyrazolo[3,4-d]pyrimidine- 4,6(5H,7H)-dione 7 (0.8 g, crude) and BOP (1.04g, 2.35mmol) in anhydrous THF (4mL) at room temperature is added 8-diazabicyclo[5.4.0]undec-7-ene (0.83mL, 5.45 mmol)under argon atmosphere. After the reaction mixture is stirred at room temperature for 5 minutes, solid 2- amino-l,2-dideuteriocyclopentanol (372mg, crude) is added. The mixture is stirred at room temperature for 3 days. The solvent is removed, and the residue is purified by silica-gel column chromatography using a gradient of 0-100% a mixed solvent (CH2CI2 : CH3OH : 7 N NH3 in methanol = 10 : 1 : 0.1) in CH2CI2 as eluent. The obtained product (1.80 g, brown solid) contains other impurities, which is directly used for the next reaction without further purification.MS (ESI) m/z 528.2061 [M + H]+.
6a,9a-dideuterio-2-(4-(6-fluoropyridin-2-yl)benzyl)-5-methyl-3-(phenylamino)- 5,6a,7,8,9,9a-hexahydrocyclopenta[4,5]imidazo[l,2-a]pyrazolo[4,3-e]pyrimidin-4(2H)-one (9). Thionyl chloride (0.360 mL, 4.96 mmol) is added dropwise to a stirred solution of 6-((l,2- dideuterio-2-hydroxycyclopentyl)amino)-2-(4-(6-fluoropyridin-2-yl)benzyl)-5-methyl-3- (phenylamino)-2H-pyrazolo[3,4-d]pyrimidin-4(5H)-one 8 (1.80g, crude) in anhydrous DMF (lOmL) at room temperature under argon atmosphere. The reaction mixture is stirred for 2 h and the solvent is removed under reduced pressure. The residue is purified by silica-gel column chromatography with a gradient of 0-100% a mixed solvent (ethyl acetate : methanol : 7 N ammonia in methanol = 10 : 1 : 0.1) in ethyl acetate. The obtained product (332 mg) is further purified with a semipreparative HPLC system with a gradient of 0 to 30% acetonitrile in water containing 0.1% formic acid over 16 min. The title compound is given as an off-white solid (60 mg). MS (ESI) m/z 510.2071 [M + H]+.
(6aS,9aR)-6a,9a-dideuterio-2-(4-(6-fluoropyridin-2-yl)benzyl)-5-methyl-3-(phenylamino)-
5,6a,7,8,9,9a-hexahydrocyclopenta[4,5]imidazo[l,2-a]pyrazolo[4,3-e]pyrimidin-4(2H)-one.
6a,9a-dideuterio-2-(4-(6-fluoropyridin-2-yl)benzyl)-5-methyl-3-(phenylamino)-5,6a,7,8,9,9a- hexahydrocyclopenta[4,5]imidazo[l,2-a]pyrazolo[4,3-e]pyrimidin-4(2H)-one 9 (60 mg) is dissolved in a mixture of hexane, isopropanol and diethylamine (40 : 60 : 0.1 volume) (10 mL). The solution is loaded to a semipreparative HPLC system with a chiralpak AD-H column (20 x 250 mm) and eluted with a mixed solvent (of hexane, isopropanol and diethylamine (40 : 60 : 0.1 volume). The title compound is obtained as white solid (19 mg, 32% yield). MS (ESI) m/z 510.2743 [M + H]+. 1H NMR (500 MHz, Chloroform-d) d 7.92 - 7.86 (m, 2H), 7.86 - 7.80 (m, 1H), 7.58 (dd, J = 7.6, 2.4 Hz, 1H), 7.31 - 7.26 (m, 2H), 7.13 - 7.07 (m, 1H), 7.07 - 7.02 (m, 2H), 6.97 - 6.90 (m, 2H), 6.89 - 6.81 (m, 2H), 4.94 (s, 2H), 3.34 (s, 3H), 1.96 (s, 1H), 1.86 - 1.79 (m, 1H), 1.77 - 1.70 (m, 2H), 1.63 - 1.52 (m, 2H).
EXAMPLE 3: Synthesis of novel [14C] radiolabeled PDE1 inhibitor
[0051] A novel radiolabeled PDE1 inhibitor is synthesized according to the following Scheme:
Scheme 1:
Figure imgf000035_0001
[0052] (6aR,9aS)-3-chloro-2-(4-(6-fluoropyridin-2-yl)benzyl)-5-methyl-5,6a,7,8,9,9a- hexahydrocyclopenta[4,5]imidazo[l,2-a]pyrazolo[4,3-e]pyrimidin-4(2H)-one is synthesized according to known methods, e.g., as disclosed in International Publications W02009/075784A1 or WO2014/205354A1, both of which are incorporated herein by reference in their entireties. This compound is added to a mixture of [14C] aniline, potassium carbonate, Xantphos and tris(dibenzylideneacetone)dipalladium(0), dissolved in 2-methyl-2- butanol (21 mL), frozen and pumped under high vacuum. The mixture was purged with nitrogen, heated, filtered and washed with ethanol.
[0053] The filtrate was purified firstly by column chromatography on silica eluting with ethyl acetate :ethanol followed by reverse phase high pressure liquid chromatography on a C18 column, eluting with aqueous trifluoroacetic acid:acetonitrile. The mixture was basified using potassium carbonate and was then partitioned between ethyl acetate and water. The solvent was removed with vacuum from the organic layer to give the [14C] free base ((6aR,9aS)-2-(4-(6-fluoropyridin-2-yl)benzyl)-5-methyl-3-([U- 14C]-phenylamino)-5, 6a, 7,8,9, 9a-hexahydrocyclopenta[4,5]imidazo[l,2-a]pyrazolo [4,3- e]pyrimidin-4-(2H)-one).
[0054] The free base was dissolved in acetonitrile. The mixture was heated and a solution of phosphoric acid in acetonitrile was added under nitrogen. The mixture was then stirred under nitrogen. The mixture was filtered and washed with acetonitrile. The solid was dried over di-phosphorus pentoxide to give the phosphate salt ((6aR,9aS)-2- (4-(6-fluoropyridin-2-yl)benzyl)-5-methyl-3-([U-14C]-phenylamino)-5,6a,7,8,9,9a- hexahydrocyclopenta[4,5]imidazo[l,2-a]pyrazolo[4,3-e]pyrimidin-4-(2H)-one phosphate).

Claims

CLAIMS We claim:
1. A compound according to Formula la:
Figure imgf000037_0001
(i) R2 and R5 are independently H, D or hydroxy and R3 and R4 together form a tri or tetra-methylene bridge [pref. with the carbons carrying R3 and R4 having the R and S configuration respectively]; or R2 and R3 are each methyl and R4 and R5 are each H; or R2, R4 and R5 are H and R3 is isopropyl [pref. the carbon carrying R3 having the R configuration] ;
(ii) R6 is (optionally halo-substituted or hydroxy-substituted) phenylamino,
(optionally halo-substituted or hydroxy-substituted) benzylamino, Ci-4alkyl, or Ci- 4alkyl sulfide; for example, phenylamino or 4-fluorophenylamino;
(iii) Rio is Ci-4alkyl, methylcarbonyl, hydroxy ethyl, carboxylic acid, sulfonamide, (optionally halo- or hydroxy-substituted) phenyl, (optionally halo- or hydroxy- substituted) pyridyl (for example 6-fluoropyrid-2-yl), or thiadiazolyl (e.g., 1,2,3- thiadiazol-4-yl); and
X and Y are independently C or N, wherein R2 and R5 are independently D or hydroxy; or R6 is hydroxy-substituted phenylamino or hydroxy-substituted benzylamino, optionally wherein at least one carbon atom substituted with Carbon-14 ([14C]), in free, pharmaceutically acceptable salt or prodrug form, including its enantiomers, diastereoisomers and racemates.
2. A compound according to claim 1, wherein R2 and R5 are independently D or hydroxy.
3. A compound according to any preceding claim, wherein R6 is hydroxy-substituted phenylamino or hydroxy-substituted benzylamino.
4. A compound according to any preceding claim, wherein R6 is hydroxy-substituted phenylamino.
5. A compound according to any preceding claim, wherein R2 and R5 are independently H, D or hydroxy and R3 and R4 together form a tri- or tetra-methylene bridge
6. A compound according to any preceding claim, wherein R2 and R5 are both D.
7. A compound according to any preceding claim, wherein R2 and R5 are both D and R3 and R4 together form a tri- or tetra-methylene bridge.
8. A compound according to any of claims 1-5, wherein at least one of R2 and R5 are hydroxy.
9. A compound according to any of claims 1-5 or 8, wherein at least one of R2 and R5 are hydroxy and R3 and R4 together form a tri- or tetra-methylene bridge.
10. A compound according to any of claims 1-5 or 8-9, wherein R2 is hydroxy.
11. A compound according to any of claims 1-5 or 8-9, wherein R5 is hydroxy.
12. A compound according to any of the preceding claims, wherein the PDE1 inhibitor is a compound according to the following:
Figure imgf000039_0001
13. A compound according to any of the preceding claims, wherein the PDE1 inhibitor is a compound according to the following:
Figure imgf000040_0001
14. A compound according to any of the preceding claims, wherein one carbon atom substituted with Carbon- 14 ([14C]).
15. A compound according to claim 14, wherein the PDE1 inhibitor is a compound according to the following:
Figure imgf000040_0002
or a pharmaceutically acceptable salt thereof.
16. A method of inhibiting the metabolism of a PDE1 inhibitor, e.g. a PDE1 inhibitor according to claim 1, the method comprising deuterating the PDE1 inhibitor to block the formation of one or more metabolites.
17. The method of claim 16, wherein the PDE1 inhibitor is a compound according to the following formula:
Figure imgf000041_0001
in free or pharmaceutically acceptable salt form.
18. The method according to claim 17, wherein deuterating the PDE1 inhibitor comprises reacting:
Figure imgf000041_0002
with
Figure imgf000042_0002
19. The method according to claim 18, wherein the reaction is carried out in benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate, THF and 1,8- Diazabicyclo[5.4.0]undec-7-ene.
20. The method according to claims 18 or 19, wherein the
Figure imgf000042_0001
is reacted with thionyl chloride in THF to form
Figure imgf000043_0001
PCT/US2020/049141 2019-09-03 2020-09-03 Novel compounds WO2021046179A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US17/753,472 US20220356187A1 (en) 2019-09-03 2020-09-03 Novel compounds
JP2022514214A JP2022546710A (en) 2019-09-03 2020-09-03 New compound
EP20861369.5A EP4025202A4 (en) 2019-09-03 2020-09-03 Novel compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962895477P 2019-09-03 2019-09-03
US62/895,477 2019-09-03

Publications (1)

Publication Number Publication Date
WO2021046179A1 true WO2021046179A1 (en) 2021-03-11

Family

ID=74853008

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/049141 WO2021046179A1 (en) 2019-09-03 2020-09-03 Novel compounds

Country Status (4)

Country Link
US (1) US20220356187A1 (en)
EP (1) EP4025202A4 (en)
JP (1) JP2022546710A (en)
WO (1) WO2021046179A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010065151A1 (en) * 2008-12-06 2010-06-10 Intra-Cellular Therapies, Inc. Organic compounds
WO2010132127A1 (en) * 2009-05-13 2010-11-18 Intra-Cellular Therapies, Inc. Organic compounds
US9884872B2 (en) * 2014-06-20 2018-02-06 Intra-Cellular Therapies, Inc. Organic compounds

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2006255028B2 (en) * 2005-06-06 2012-04-19 Intra-Cellular Therapies, Inc. Organic compounds
ES2717688T3 (en) * 2013-03-15 2019-06-24 Intra Cellular Therapies Inc PDE1 inhibitors for use in the treatment and / or prevention of CNS lesions, and diseases, disorders or injuries of SNP
US9849132B2 (en) * 2014-01-08 2017-12-26 Intra-Cellular Therapies, Inc. Products and pharmaceutical compositions

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010065151A1 (en) * 2008-12-06 2010-06-10 Intra-Cellular Therapies, Inc. Organic compounds
WO2010132127A1 (en) * 2009-05-13 2010-11-18 Intra-Cellular Therapies, Inc. Organic compounds
US9884872B2 (en) * 2014-06-20 2018-02-06 Intra-Cellular Therapies, Inc. Organic compounds

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
GRAHAM S TIMMINS: "Deuterated drugs; where are we now?", EXPERT OPIN THER PAT., vol. 24, no. 10, 2014, pages 1067 - 1075, XP055132523 *
LI ET AL.: "Discovery of Potent and Selective Inhibitors of Phosphodiesterase 1 for the Treatment of Cognitive Impairment Associated with Neurodegenerative and Neuropsychiatric Diseases", J. MED. CHEM., vol. 59, pages 1149 - 1164, XP055443845, DOI: 10.1021/acs.jmedchem.5b01751 *
See also references of EP4025202A4 *

Also Published As

Publication number Publication date
JP2022546710A (en) 2022-11-07
US20220356187A1 (en) 2022-11-10
EP4025202A1 (en) 2022-07-13
EP4025202A4 (en) 2023-08-02

Similar Documents

Publication Publication Date Title
AU2006255028B2 (en) Organic compounds
JP6625616B2 (en) P2X7 regulated N-acyl-triazolopyrazine
EP2240028A1 (en) Organic compounds
KR20120012831A (en) Organic compounds
EP3479825A1 (en) Pde1 inhibitors for use in the treatment and/or prevention of cns or pns diseases or disorders
EP3725789B1 (en) 7,8-dihydro-[2h]-imidazo-[1,2-a]pyrazolo[4,3-e]pyrimidin-4(5h)-one derivatives as phosphodiesterase 1 (pde1) inhibitors for treating diseases, disorders or injuries of the central nervous system (cns)
SK11572003A3 (en) 4-(2-Butylamino)-2,7-dimethyl-8-(2-methyl-6-methoxypyrid-3-yl) pyrazolo-[1,5-a]-1,3,5-triazine, its enantiomers and pharmaceutically acceptable salts as corticotropin releasing factor receptor ligands
AU2015357496B2 (en) Organic compounds
WO2021046179A1 (en) Novel compounds
WO2020086650A2 (en) Novel compounds
WO2021127517A1 (en) Novel uses
EP4017858A1 (en) Organic compounds
MX2007015356A (en) Organic compounds

Legal Events

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

Ref document number: 20861369

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2022514214

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020861369

Country of ref document: EP

Effective date: 20220404