WO2024026500A2 - Inhibitors of pde11a4 and methods of using same - Google Patents
Inhibitors of pde11a4 and methods of using same Download PDFInfo
- Publication number
- WO2024026500A2 WO2024026500A2 PCT/US2023/071289 US2023071289W WO2024026500A2 WO 2024026500 A2 WO2024026500 A2 WO 2024026500A2 US 2023071289 W US2023071289 W US 2023071289W WO 2024026500 A2 WO2024026500 A2 WO 2024026500A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- optionally substituted
- compound
- formula
- alkyl
- pde11a4
- Prior art date
- Legal status (The legal status 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 status listed.)
- Ceased
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
- C07D471/04—Ortho-condensed systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/28—Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
Definitions
- FIELD [0003] The disclosure relates generally to compounds that alter the subcellular location of PDE11A4 and/or inhibit the activity of PDE11A4, and methods of using such compounds as treatments for disease.
- BACKGROUND [0004] After the age of 60, nearly all individuals experience some form of cognitive decline—particularly memory deficits—and no drugs are able to prevent or reverse this loss. Indeed, advanced age is the strongest risk factor for dementia. Even in absence of dementia, age-related cognitive impairment increases health care costs and risk for disability. Age- related cognitive decline is not a uniform process, with variability in symptom severity observed across individuals and across cognitive domains.
- Associative long-term memories (aLTMs)—particularly those involving experiences with family and friends— are more susceptible to age-related cognitive decline than are recognition long-term memories (rLTMs) for reasons that are not well understood.
- rLTMs recognition long-term memories
- the disclosure provides a compound of formula (A), or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof: formula (A) wherein in formula (A): R 1 is selected from 5-membered heterocycle, 5-membered heteroaryl, 6-membered heterocycle, and 6-membered heteroaryl; or R 1 is R 3 is selected from 5-membered heterocycle, 5-membered heteroaryl, 6-membered heterocycle, and 6-membered heteroaryl; R 2 and R 4 are independently at each occurrence selected from hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkylheteroaryl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylaryl, optionally substituted aryl, optionally substituted aryl, optionally substituted
- the compound of formula (A) is a compound of formula (I), or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof: formula (I) wherein in formula (I): R 1 and R 3 are independently at each occurrence a 5-membered heterocycle, 5- membered heteroaryl, a 6-membered heterocycle, or 6-membered heteroaryl; R 2 and R 4 are independently at each occurrence selected from hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkylheteroaryl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylaryl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, hydroxy,
- R 1 is a 5-membered heteroaryl ring, optionally selected from thiazole, oxazole, imidazole, thiadiazole, oxadiazole, and triazole.
- the compound is a compound of formula (II): formula (II) wherein in formula (II): A is S, NR 5 , N, or O, optionally A is S or O; B 1 and B 2 are independently at each occurrence selected from N, O, or CR 5 , wherein B 1 is not O when either A or B 2 is O; and each R 5 is independently at each occurrence selected from hydrogen, C 1 -C 6 alkyl (e.g., C 1 -C 3 alkyl), and C 1 -C 3 haloalkyl (e.g., CH 2 F, CHF 2 , CF 3 ); R 3 is selected from 5-membered heterocycle, 5-membered heteroaryl, 6-membered heterocycle, and 6-member
- R 1 is selected from wherein: A a is O or S, and B a is N or CH; A b is N, and B b is O or S; A c is O or S, and B c is N; and R 5 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), and C 1 -C 3 haloalkyl (e.g., CH 2 F, CHF 2 , or CF 3 ).
- C 1 -C 3 alkyl e.g., methyl, ethyl, propyl, isopropyl
- C 1 -C 3 haloalkyl e.g., CH 2 F, CHF 2 , or CF 3
- R 1 is selected from , and ; and R 5 is selected from H, methyl, ethyl, propyl, isopropyl, CH 2 F, and CHF 2 .
- R 1 is selected from , ,. , , , , and .
- R 2 is selected from hydrogen, C 3 -C 6 cycloalkyl, C 1 -C 6 alkyl (e.g., C 1 -C 4 straight chain alkyl, C 3 -C 6 branched alkyl), -CF 3 , - CH 2 F, -CHF 2 , and - C(R 6 R 7 ) p X; wherein, p is an integer from 1-3; R 6 and R 7 are each independently selected from hydrogen, C 1 -C 3 alkyl, and C 3 -C 6 cycloalkyl; X is selected from -OH, -CN, -OR 8 , and NR 9 R 10 ; R 8 is selected from C 1 -C 3 alkyl (e.g., C 1 -C 3 straight or branched alkyl), and C 3 -C 6 cycloalkyl; and R 9 and R 10 are each independently selected from hydrogen, C 1 -C 3 alkyl, and C 3 -
- R 2 is selected from hydrogen, C 1 -C 4 alkyl (e.g., methyl, ethyl, propyl, isopropyl, butyl, tertbutyl), -CF 3 , -CH 2 F, and -CHF 2 .
- R 2 is selected from H, -CH 3 , -CF 3 , and -CHF 2 .
- R 3 is a 5-membered heteroaryl ring, optionally selected from pyrrole, thiophene, furan, pyrazole, thiazole, isothiazole, oxazole, isoxazole, and imidazole.
- R 3 is selected from , , , , , , , , wherein R 11 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), C 3 -C 6 cycloalkyl, CF 3 , CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F.
- R 3 is selected from , .
- R 11 is selected from hydrogen, methyl, ethyl, propyl, isopropyl, -CF 3 , and -CH 2 CF 3 .
- R 3 is selected from , [0012]
- R 4 is selected from hydrogen, C 1 -C 6 alkyl, and halo (e.g., F or Cl).
- n is 0, 1, or 2.
- n is 0 or 1.
- R 4 is selected from hydrogen, 4-fluoro, 3-fluoro, 2-fluoro, 2-chloro, and 4-chloro.
- R 4 is selected from hydrogen, 4-fluoro, 2-fluoro, and 2-chloro.
- n is 2 and each R 4 is 2-fluoro and 4-fluoro.
- the compound of formula (A), formula (I), or formula (II) is a compound having any one of formula 1001-1080, 1097-1176, 1193-1272, 1289-1368, 1385- 1729, 4001-4080, 4097-4176, 4193-4272, 4289-4368, 4385-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- the compound of formula (A), formula (I), or formula (II) is a compound having any one of formula 2001-2337, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- the compound of formula (A), formula (I), or formula (II) is a compound having any one of the formula of Compounds 1013, 1014, 1109, 1110, 1015, 1625, 1111, 1011, 1009, 1107, 1105, 2217, 2218, 2008, 2210, 2315, 2219, 1682, 1691, 2017, 2015, 2016, 2087, 2019, 2001, 2020, 2021, 2222, 2215, 1448, 1543, 2264, 1393, 1717, 2278, 2027, 2041, 1103, 1101, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- R 1 is ; and R 20 is selected from H, optionally substituted C 1 -C 6 alkyl, and optionally substituted C 1 -C 6 haloalkyl; R 21 is selected from optionally substituted C 1 -C 6 alkyl and optionally substituted C 1 -C 6 haloalkyl; or R 20 and R 21 are joined to form a 4-6-membered heterocycle with the nitrogen to which they are each bound.
- R 20 is selected from H, methyl, ethyl, propyl, isopropyl, butyl, tert-butyl, CF 3 , CHF 2 , CH 2 F, CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F; and R 21 is selected from methyl, ethyl, propyl, isopropyl, butyl, tert-butyl, CF 3 , CHF 2 , CH 2 F, CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F; or R 20 and R 21 are joined to form a 4-6-membered heterocycle with the nitrogen to which they are each bound.
- R 20 is selected from H, methyl, ethyl, propyl, tert-butyl, and CH 2 CH 2 F; and R 21 is selected from methyl, ethyl, propyl, tert- butyl, and CH 2 CH 2 F; or R 20 and R 21 are joined to form a 4-6-membered heterocycle with the nitrogen to which they are each bound.
- the compound of formula (A), pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof is a compound of formula (III): formula (III) wherein in formula (III): Z is selected from , , , , , and , ; R 3 is selected from 5-membered heterocycle, 5-membered heteroaryl, 6-membered heterocycle, and 6-membered heteroaryl; R 2 and R 4 are independently at each occurrence selected from hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkylheteroaryl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylaryl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroaryl, optionally substituted heteroaryl
- R 2 is selected from hydrogen, C 3 -C 6 cycloalkyl, C 1 -C 6 alkyl (e.g., C 1 -C 4 straight chain alkyl, C 3 -C 6 branched alkyl), -CF 3 , -CH 2 F, -CHF 2 , and -C(R 6 R 7 )pX; wherein, p is an integer from 1-3; R 6 and R 7 are each independently selected from hydrogen, C 1 -C 3 alkyl, and C 3 -C 6 cycloalkyl; X is selected from -OH, -CN, -OR 8 , and NR 9 R 10 ; R 8 is selected from C 1 -C 3 alkyl (e.g., C 1 -C 3 straight or branched alkyl), and C 3 -C 6 cycloalkyl; and R 9 and R 10 are each independently selected from hydrogen, C 1 -C 3 alkyl, and C 3 -C 6 cycloalkyl;
- R 2 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), -CF 3 , -CH 2 F, and -CHF 2 .
- R 2 is selected from hydrogen and -CH 3 .
- R 3 is a 5-membered heteroaryl ring, optionally selected from pyrrole, thiophene, furan, pyrazole, thiazole, isothiazole, oxazole, isoxazole, and imidazole.
- R 3 is selected from , , , , , wherein R 11 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), C 3 -C 6 cycloalkyl, CF 3 , CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F.
- R 3 is selected from , and
- R 11 is selected from hydrogen, methyl, ethyl, propyl, isopropyl, -CF 3 , and -CH 2 CF 3 .
- R 3 is selected from , , and .
- R 4 is selected from hydrogen, C 1 -C 6 alkyl, and halo (e.g., F or Cl). In embodiments, n is 0 or 1. In embodiments, R 4 is selected from hydrogen, 4-fluoro, 3- fluoro, 2-fluoro, 2-chloro, and 4-chloro. In embodiments, R 4 is selected from hydrogen, 4- fluoro, 3-fluoro, 2-fluoro, and 4-chloro. [0019] In embodiments, the compound of formula (A) or formula (III) is a compound having any one of formula 3001, 3006-3021, 3023-3031, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- a compound of the disclosure is a PDE11A4 inhibitor.
- the PDE11A4 inhibitor is a PDE11A4 selective inhibitor.
- the present disclosure provides pharmaceutical compositions comprising a compound of the disclosure, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof, and a physiologically compatible carrier medium.
- the present disclosure provides a pharmaceutical composition
- a pharmaceutical composition comprising a compound of the disclosure, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof, and a physiologically compatible carrier medium, wherein the amount of the compound in the composition is a therapeutically effective amount for the treatment or prevention of a disease or disorder alleviated by inhibiting PDE11A4 activity in a patient in need thereof.
- the disease or disorder is associated with cognitive decline.
- the disease or disorder is dementia, Alzheimer’s Disease (AD) including mild Alzheimer's disease and early-onset Alzheimer’s disease, Down’s syndrome, vascular dementia (cerebral amyloid angiopathy and stroke), dementia with Lewy bodies, HIV dementia, Mild Cognitive Impairment (MCI); Age-Associated Memory Impairment (AAMI); Age-Related Cognitive Decline (ARCD) (including age-related cognitive decline of associative long-term memories (aLTMs), dementia associated with traumatic brain injury, preclinical Alzheimer's Disease (PCAD); Cognitive Impairment No Dementia (CIND); or cognitive decline associated with spatial memory, other depression-related behaviors, additional anxiety-related behaviors, sensorimotor gating, or social behaviors.
- the present disclosure provides methods of treating or preventing a disease or disorder alleviated by inhibiting PDE11A4 activity in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of the disclosure.
- the present disclosure provides methods of treating or preventing a disease or disorder alleviated by inhibiting PDE11A4 activity in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition of the disclosure.
- the method includes administering the compound in a dosage unit form.
- the dosage unit comprises a physiologically compatible carrier medium.
- the method comprises treating or preventing a disease or disorder that is associated with cognitive decline.
- the disease or disorder is selected from dementia, Alzheimer’s Disease (AD) including mild Alzheimer's disease and early-onset Alzheimer’s disease, Down’s syndrome, vascular dementia (cerebral amyloid angiopathy and stroke), dementia with Lewy bodies, HIV dementia, Mild Cognitive Impairment (MCI); Age-Associated Memory Impairment (AAMI); Age-Related Cognitive Decline (ARCD) (including age-related cognitive decline of associative long-term memories (aLTMs), dementia associated with traumatic brain injury, preclinical Alzheimer's Disease (PCAD); Cognitive Impairment No Dementia (CIND), and cognitive decline associate with spatial memory, other depression-related behaviors, additional anxiety-related behaviors, sensorimotor gating, and social behaviors.
- Fig 1. is an image showing how PDE11A4 mRNA expression is restricted to the HIPP. Protein is expressed in neuronal cell bodies, dendrites, and axons.
- Figs.2A-2C illustrate increases in hippocampal PDE11A expression.
- Fig.2A is a graph of experimental data showing how hippocampal PDE11A expression is increased in old vs. young mice.
- Fig.2B. is a graph of experimental data showing how hippocampal PDE11A expression is increased in adult (18-40yrs) vs. prenatal humans.
- Fig.2C is a graph of experimental data showing how hippocampal PDE11A expression is increased in demented vs. non-demented aged humans (>75yrs) with a history of TBI. Post hoc *vs. Young, prenatal, or ‘No’ group, P ⁇ 0.05.
- A.U. arbitrary units.
- Figs.3A-3C illustrate aging preferentially impairs aLTMs in mice.
- Fig.4A is a graph of experimental data showing male and female old PDE11A WT mice (WT-O) having no memory for STFP 7 days after training; however old PDE11A KO mice (KO-O) and heterozygous mice show robust memory equivalent to that of young (Y) mice.
- Fig.5B shows western blots of DHIPP and VHIPP that illustrate titrating virus delivery allows overexpression of PDE11A4 in a dorsal ⁇ ventral gradient, as is seen in vivo.
- Fig.5D is a graph of experimental data illustrating mice trained on STFP and tested 7 days after training.
- Fig.5E is a graph of experimental data illustrating the ability of PDE11A4 overexpression to impair aLTM, as KO mice treated with either the GFP or PDE11A4 lentivirus show strong rLTM for NSOR.
- Fig.6 is a graph of experimental data demonstrating that compound 25b was more potent and more efficacious than the drug tadalafil in reversing aging-like PDE11A4 phenotype.
- Fig.7A is an image and graph of experimental data illustrating phosphomimic mutations of PDE11A4 at serines 117 and 124 (117D124D) synergize, increasing the accumulation of PDE11A4 in distinct structures. Phosphoresistant mutations S117AS124A have the opposite effect.
- Fig.7B is a graph and image showing how biochemical fractionation of S117DS124 shifts PDE11A4 from the cytosol to the membrane.
- Fig.7C is a graph of experimental data and image showing how S117 and S124 also synergize at the level of phosphorylation.
- Fig.7E is a graph of experimental data normalizing the hyperaccumulation that is seen with S117D124D.
- Fig.7F is a graph of experimental data and image showing expression of S117D124D mimics age-related decreases in cGMP levels.
- Fig.7G is a graph of experimental data and image showing how expression of the isolated GAF-B domain has the opposite effect as what is shown in Fig.7F.
- Fig 8. is an image showing immuno-fluorescence with a total PDE11A antibody (top—green) and pS117/pS124-PDE11A4 antibody (bottom—green) suggest that age-related increases in PDE11A expression occur in a compartment-specific manner. KOs virally overexpressing PDE11AWT show the same pattern as “old” .
- Fig 9 is a graph of experimental data and image showing age-related increases in VHIPP PDE11A4 protein expression occurs preferentially in membrane fractions, consistent with the in vitro studies showing S117D/S124D mutations shift PDE11A4 from the cytosol to the membrane.
- Fig 10 is an image showing how mPde11a4-mCherry reports transcriptional activity in ventral CA1 (vCA1) but not ventral dentate gyrus (vDG), consistent with endogenous expression pattern of mPde11a4 (see also Fig.1). In contrast, the ubiquitous Pgk-mCherry construct reports transcription in both subregions.
- Fig.12A is an image showing widefield fluorescence’s ability to label PDE11A4 mRNA (red) using RNAscope. A confocal microscope in the USC IRF are used to collect z- stack images.
- Fig.12B is an image showing an example of subcellular resolution provided by confocal microscope. Shown is PDE11A4 protein (green) in COS1 cells counterstained for the nuclear marker DAPI (blue). Optical sections through the cell (shown to the right and bottom) clarify whether labeling observed from above is in the nucleus or cytosol.
- Fig.13 is a flowchart illustrating a non-limiting example of a pathway for pharmacologic inhibition of PDE11A.
- Figs.14A-14C show increases in hippocampal PDE11A expression.
- Fig.14A is a graph of experimental data showing how hippocampal PDE11A expression is increased in Old vs. young mice.
- Fig.14B is a graph of experimental data showing how hippocampal PDE11A expression is increased in adult (18-40 yrs) vs prenatal humans.
- Fig.14C is a graph of experimental data showing how hippocampal PDE11A expression is increased in demented vs. non-demented aged humans (>75 years) with a history of TBI. Post hoc *vs. Young, prenatal, or “no” group, P ⁇ 0.05. A.U. – arbitrary units.
- Figs.15A-15C show aging preferentially impairs aLTMs in mice.
- Fig.16A is a graph of experimental data showing old PDE11A WT mice (WT-O) show no memory for STFP 7 days after training.; old PDE11A KO mice (KO-O) and heterogeneous mice (HT-O) show robust memory equivalent to that of young (Y) mice.
- Figs.17A-17C show disrupting PDE114A expression in old WT mice rescues age-related impairments in aLTM for STFP.
- Fig.17A is a graph of experimental data showing in vitro expression of an isolated GAF-B domain peptide (GB) disrupts PDE11A4 homodimerization and impairs PDE11A4 cGMP-hydrolytic activity relative to expression of an mCherry negative control (mCh).
- GB GAF-B domain peptide
- Fig.17B is a graph of experimental data showing viral expression of the GB peptide in dorsal and ventral hippocampus of old PDE11A wild-type mice reverses age-related impairments in aLTM for STFP.
- Fig.17C is a graph of experimental data showing GB had no effect on NSOR.
- Fig.18A is a graph of experimental data showing the results of COS1 monkey fibroblast cells transiently transfected with mouse PDE11A (95% homologous to human PDE11A4) used to confirm inhibition in a mammalian context and assess effects on subcellular compartmentalization.
- Fig.18B Is a graph showing cGMP relative to the negative control GFP.
- Fig.18D is a graph illustrating how age-related increases in PPDE11A expression lead to increased PPDE11A4 phosphorylation that causes formation of PPDE11A4 aggregates in the HIPP.
- Fig.18E is an image showing COS1 cells used to model the aggregation.
- Fig.18F is a graph of experimental data illustrating how manipulations that increase or decrease the aggregation.
- Fig.19 shows an image of chemical formulas of amine examples 1- 11.
- Fig.20 shows an image of chemical formulas amine examples 12-16.
- Fig.21 shows an image of chemical formulas amine examples 17-19.
- Fig.22 shows a non-limiting process of in vivo screening of young and old C57BL6 mice. Y, young; O, old, V, vehicle; D1-3, dose 1-3.
- Fig.23 is an image showing a non-limiting example of a scheme of a synthesis of compounds 5 (a-s).
- Fig.24 is an image showing a non-limiting example of a scheme to prepare compounds 10 (a-b).
- Fig.25 is an image showing a non-limiting example of a scheme to prepare compounds 12 (a-b).
- Fig.26A is an image of exemplary PDE11A inhibitors of the disclosure.
- Fig.26B is an image and a table illustrating PDE11A inhibition activity of exemplary compounds of the disclosure.
- Fig.27 is an image of exemplary PDE11A inhibitors of the disclosure.
- Fig.28 is an image of exemplary PDE11A inhibitors of the disclosure.
- Fig.29 is an image of HT22 cells overexpressing EmGFP-mPDE11A4.
- Figs.30A-31B is an image of HT-22 cells overexpressing EmGFP-mPDE11A4 treated with the vehicle DMSO (Fig.31A) or 1 uM 25b (Fig.31B).
- DEFINITIONS [0071] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this disclosure belongs. All patents and publications referred to herein are incorporated by reference in their entireties.
- administer refers to (1) providing, giving, dosing, and/or prescribing by either a health practitioner or his authorized agent or under his or her direction according to the disclosure; and/or (2) putting into, taking or consuming by the mammal, according to the disclosure.
- co-administration encompass administration of two or more active pharmaceutical ingredients to a subject so that both active pharmaceutical ingredients and/or their metabolites are present in the subject at the same time.
- Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which two or more active pharmaceutical ingredients are present. Simultaneous administration in separate compositions and administration in a composition in which both agents are present are preferred.
- active pharmaceutical ingredient and drug include the polypeptides, polynucleotides, and compositions described herein.
- active pharmaceutical ingredient and “drug” may also include those compounds described herein that bind PDE11A4 and thereby modulate (e.g. inhibit) PDE11A4 activity.
- isostere refers to a group or molecule whose chemical and/or physical properties are similar to those of another group or molecule.
- a “bioisostere” is a type of isostere and refers to a group or molecule whose biological properties are similar to those of another group or molecule.
- a carboxylic acid may be replaced by one of the following bioisosteres for carboxylic acids, including, without limitation, alkyl esters (COOR), acylsulfonamides (CONR-SO2R), hydroxamic acids (CONR-OH), hydroxamates (CONR-OR), tetrazoles, hydroxyisoxazoles, isoxazol-3-ones, and sulfonamides (SO2NR), where each R may independently represent hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
- in vivo refers to an event that takes place in a subject’s body.
- in vitro refers to an event that takes places outside of a subject’s body. In vitro assays encompass cell-based assays in which cells alive or dead are employed and may also encompass a cell-free assay in which no intact cells are employed.
- effective amount or “therapeutically effective amount” refers to that amount of a compound or combination of compounds as described herein that is sufficient to effect the intended application including, but not limited to, disease treatment.
- a therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated (e.g., the weight, age and gender of the subject), the severity of the disease condition, the manner of administration, etc., which can readily be determined by one of ordinary skill in the art.
- the term also applies to a dose that induces a particular response in target cells (e.g., the reduction of platelet adhesion and/or cell migration).
- the specific dose varies depending on the particular compounds chosen, the dosing regimen to be followed, whether the compound is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which the compound is carried.
- a prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
- the terms “treat,” “treatment,” and/or “treating” may refer to the management of a disease, disorder, or pathological condition, or symptom thereof with the intent to cure, ameliorate, stabilize, and/or control the disease, disorder, pathological condition or symptom thereof.
- control may include the absence of condition progression, as assessed by the response to the methods recited herein, where such response may be complete (e.g., placing the disease in remission) or partial (e.g., lessening or ameliorating any symptoms associated with the condition).
- control may include the absence of condition progression, as assessed by the response to the methods recited herein, where such response may be complete (e.g., placing the disease in remission) or partial (e.g., lessening or ameliorating any symptoms associated with the condition).
- prevent prevent
- prevention may refer to reducing the risk of developing a disease, disorder, or pathological condition.
- the terms “modulate” and “modulation” refer to a change in biological activity for a biological molecule (e.g., a protein, gene, peptide, antibody, and the like), where such change may relate to an increase in biological activity (e.g., increased activity, agonism, activation, expression, upregulation, and/or increased expression) or decrease in biological activity (e.g., decreased activity, antagonism, suppression, deactivation, downregulation, and/or decreased expression) for the biological molecule.
- the compounds described herein may modulate (e.g., inhibit) PDE11A4 protein.
- the compounds described herein may selectively modulate (e.g., selectively inhibit) PDE11A4 protein as compared to other PDE11A proteins. In some embodiments, the compounds described herein may selectively modulate (e.g., selectively inhibit) PDE11A4 protein as compared to other PDE or PDE11A proteins. “Modulate” and “modulation” also include changing the subcellular localization and/or location of PDE11A4. “Modulate” and “modulation” also include disrupting and/or preventing homodimerization of PDE11A4. “Modulate” and “modulation” also include direct modulation of PDE11A4 (e.g. modulation of catalytic activity of PDE11A4).
- Modulate” and “modulation” also include indirect modulation of PDE11A4 (e.g. disrupting and/or preventing homodimerization of PDE11A4).
- Inhibit” and “inhibiting” also include changing the subcellular localization and/or location of PDE11A4.
- “Inhibit” and “inhibiting” also include disrupting and/or preventing homodimerization of PDE11A4.
- “Inhibit” and “inhibiting” also include direct inhibition of PDE11A4 (e.g. inhibition of catalytic activity of PDE11A4).
- Inhibit” and “inhibiting” also include indirect inhibition of PDE11A4 (e.g. disrupting and/or preventing homodimerization of PDE11A4).
- the terms “QD,” “qd,” or “q.d.” mean quaque die, once a day, or once daily.
- the terms “BID,” “bid,” or “b.i.d.” mean bis in die, twice a day, or twice daily.
- the terms “TID,” “tid,” or “t.i.d.” mean ter in die, three times a day, or three times daily.
- the terms “QID,” “qid,” or “q.i.d.” mean quater in die, four times a day, or four times daily.
- pharmaceutically acceptable salt refers to salts derived from a variety of organic and inorganic counter ions known in the art.
- Pharmaceutically acceptable acid addition salts is formed with inorganic acids and organic acids.
- Preferred inorganic acids from which salts is derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid and phosphoric acid.
- Preferred organic acids from which salts is derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p- toluenesulfonic acid and salicylic acid.
- Pharmaceutically acceptable base addition salts is formed with inorganic and organic bases.
- Inorganic bases from which salts is derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese and aluminum.
- Organic bases from which salts is derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins. Specific examples include isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine.
- the pharmaceutically acceptable base addition salt is chosen from ammonium, potassium, sodium, calcium, and magnesium salts.
- cocrystal refers to a molecular complex derived from a number of cocrystal formers known in the art. Unlike a salt, a cocrystal typically does not involve hydrogen transfer between the cocrystal and the drug, and instead involves intermolecular interactions, such as hydrogen bonding, aromatic ring stacking, or dispersive forces, between the cocrystal former and the drug in the crystal structure.
- “Pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” or “physiologically compatible” carrier or carrier medium is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and inert ingredients.
- a “prodrug” refers to a derivative of a compound described herein, the pharmacologic action of which results from the conversion by chemical or metabolic processes in vivo to the active compound.
- Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues is covalently joined through an amide or ester bond to a free amino, hydroxyl or carboxylic acid group of a compound of any one of formula (I), formula (10), formula (II), formula (20), formula (21), formula (22), formula (23), formula (24), formula (25), formula (26), formula (27), formulas 1001-1144 and 2001-2032.
- the amino acid residues include but are not limited to the 20 naturally occurring amino acids commonly designated by one or three letter symbols but also include, for example, 4-hydroxyproline, hydroxylysine, desmosine, isodesmosine, 3- methylhistidine, beta-alanine, gamma-aminobutyric acid, citrulline, homocysteine, homoserine, ornithine and methionine sulfone. Additional types of prodrugs are also encompassed. For instance, free carboxyl groups is derivatized as amides or alkyl esters (e.g., methyl esters and acetoxy methyl esters).
- Prodrug esters as employed herein includes esters and carbonates formed by reacting one or more hydroxyls of compounds of the method of the disclosure with alkyl, alkoxy, or aryl substituted acylating agents employing procedures known to those skilled in the art to generate acetates, pivalates, methylcarbonates, benzoates and the like.
- free hydroxyl groups may be derivatized using groups including but not limited to hemisuccinates, phosphate esters, dimethylaminoacetates, and phosphoryloxymethyloxycarbonyls, as outlined in Advanced Drug Delivery Reviews, 1996, 19, 115.
- Carbamate prodrugs of hydroxyl and amino groups are also included, as are carbonate prodrugs, sulfonate prodrugs, sulfonate esters and sulfate esters of hydroxyl groups.
- Free amines can also be derivatized to amides, sulfonamides or phosphonamides. All of the stated prodrug moieties may incorporate groups including but not limited to ether, amine and carboxylic acid functionalities.
- any compound that is converted in vivo to provide the bioactive agent e.g., a compound of any one of formula (I), formula (10), formula (II), formula (20), formula (21), formula (22), formula (23), formula (24), formula (25), formula (26), formula (27), formulas 1001-1144 and 2001-2032
- a prodrug within the scope of the disclosure.
- Various forms of prodrugs are well known in the art. A comprehensive description of pro drugs and prodrug derivatives are described in: (a) The Practice of Medicinal Chemistry, Camille G. Wermuth et al., (Academic Press, 1996); (b) Design of Prodrugs, edited by H.
- prodrugs may be designed to improve the penetration of a drug across biological membranes in order to obtain improved drug absorption, to prolong duration of action of a drug (slow release of the parent drug from a prodrug, decreased first-pass metabolism of the drug), to target the drug action (e.g., organ or tumor-targeting, lymphocyte targeting), to modify or improve aqueous solubility of a drug (e.g., i.v.
- the chemical structures depicted herein are intended to include compounds which differ only in the presence of one or more isotopically enriched atoms.
- compounds where one or more hydrogen atoms is replaced by deuterium or tritium, or wherein one or more carbon atoms is replaced by 13 C- or 14 C-enriched carbons are within the scope of this disclosure.
- ranges are used herein to describe, for example, physical or chemical properties such as molecular weight or chemical formulae, all combinations and subcombinations of ranges and specific embodiments therein are intended to be included.
- Use of the term “about” when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary. The variation is typically from 0% to 15%, or from 0% to 10%, or from 0% to 5% of the stated number or numerical range.
- Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to ten carbon atoms (e.g., (C 1 - 10 )alkyl or C 1 - 10 alkyl).
- a numerical range such as “1 to 10” refers to each integer in the given range, e.g., “1 to 10 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms, although the definition is also intended to cover the occurrence of the term “alkyl” where no numerical range is specifically designated.
- Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl isobutyl, tertiary butyl, pentyl, isopentyl, neopentyl, hexyl, septyl, octyl, nonyl and decyl.
- the alkyl moiety may be attached to the rest of the molecule by a single bond, such as for example, methyl (Me), ethyl (Et), n-propyl (nPr), 1-methylethyl (isopropyl), n-butyl, n-pentyl, 1,1-dimethylethyl (t-butyl) and 3-methylhexyl.
- an alkyl group is optionally substituted by one or more of substituents which are independently heteroalkyl, acylsulfonamido, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , - S(O)tR a - (where t is 1 or 2), -OC(O)-R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , -OC(O)N(R a ) 2 , - C(O)N(R a ) 2 , -N(R a )C
- Alkylaryl refers to an -(alkyl)aryl radical where aryl and alkyl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for aryl and alkyl respectively.
- Alkylhetaryl refers to an -(alkyl)hetaryl radical where hetaryl and alkyl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for aryl and alkyl respectively.
- Alkylheterocycloalkyl refers to an -(alkyl) heterocyclic radical where alkyl and heterocycloalkyl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for heterocycloalkyl and alkyl respectively.
- An “alkene” moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon double bond
- an “alkyne” moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon triple bond.
- the alkyl moiety, whether saturated or unsaturated, may be branched, straight chain, or cyclic.
- Alkenyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, and having from two to ten carbon atoms (i.e., (C2-10)alkenyl or C2-10 alkenyl).
- C2-10)alkenyl or C2-10 alkenyl i.e., (C2-10)alkenyl or C2-10 alkenyl.
- a numerical range such as “2 to 10” refers to each integer in the given range - e.g., “2 to 10 carbon atoms” means that the alkenyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms.
- the alkenyl moiety may be attached to the rest of the molecule by a single bond, such as for example, ethenyl (i.e., vinyl), prop-1- enyl (i.e., allyl), but-1-enyl, pent-1-enyl and penta-1,4-dienyl.
- ethenyl i.e., vinyl
- prop-1- enyl i.e., allyl
- but-1-enyl i.e., pent-1-enyl and penta-1,4-dienyl.
- an alkenyl group is optionally substituted by one or more substituents which are independently alkyl, heteroalkyl, acylsulfonamido, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , - S(O) t R a - (where t is 1 or 2), -OC(O)-R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , -OC(O)N(R a ) 2 , - C(O)N(R a ) 2 , -N(R
- Alkenyl-cycloalkyl refers to an -(alkenyl)cycloalkyl radical where alkenyl and cycloalkyl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for alkenyl and cycloalkyl respectively.
- Alkynyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having from two to ten carbon atoms (i.e., (C 2 - 10 )alkynyl or C 2 - 10 alkynyl).
- a numerical range such as “2 to 10” refers to each integer in the given range - e.g., “2 to 10 carbon atoms” means that the alkynyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms.
- the alkynyl may be attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl and hexynyl.
- an alkynyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, acylsulfonamido, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , -S(O)tR a - (where t is 1 or 2), -OC(O)- R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , -OC(O)N(R a ) 2 , -C(O)N(R a ) 2 , -N(N(R a ) 2
- Alkynyl-cycloalkyl refers to an -(alkynyl)cycloalkyl radical where alkynyl and cycloalkyl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for alkynyl and cycloalkyl respectively.
- R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl, or heteroarylalkyl.
- Carbonyl groups may be substituted with the following exemplary substituents: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, acylsulfonamido, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , - S(O) t R a - (where t is 1 or 2), -OC(O)-R a , -N(R a ) 2 , -C(O)R a , -NR a -OR a -, -C(O)OR a , - OC(O)N(R a ) 2 , -C(O)N(R a ) 2 , -N
- Cycloalkyl refers to a monocyclic or polycyclic radical that contains only carbon and hydrogen, and may be saturated, or partially unsaturated. Cycloalkyl groups include groups having from 3 to 10 ring atoms (i.e., (C3-10)cycloalkyl or C3-10 cycloalkyl).
- a numerical range such as “3 to 10” refers to each integer in the given range - e.g., “3 to 10 carbon atoms” means that the cycloalkyl group may consist of 3 carbon atoms, etc., up to and including 10 carbon atoms.
- Illustrative examples of cycloalkyl groups include, but are not limited to the following moieties: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, norbornyl, and the like.
- a cycloalkyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, acylsulfonamido, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , -S(O) t R a - (where t is 1 or 2), -S(O)tR a - (where t is 1 or 2), -OC(O)-R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , - OC(O)N(R a
- Cycloalkyl-alkenyl refers to a -(cycloalkyl)alkenyl radical where cycloalkyl and alkenyl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for cycloalkyl and alkenyl, respectively.
- Cycloalkyl-heterocycloalkyl refers to a -(cycloalkyl)heterocycloalkyl radical where cycloalkyl and heterocycloalkyl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for cycloalkyl and heterocycloalkyl, respectively.
- Cycloalkyl-heteroaryl refers to a -(cycloalkyl)heteroaryl radical where cycloalkyl and heteroaryl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for cycloalkyl and heteroaryl, respectively.
- alkoxy refers to the group -O-alkyl, including from 1 to 8 carbon atoms of a straight, branched, cyclic configuration and combinations thereof attached to the parent structure through an oxygen. Examples include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy and cyclohexyloxy.
- “Lower alkoxy” refers to alkoxy groups containing one to six carbons.
- substituted alkoxy refers to alkoxy wherein the alkyl constituent is substituted (i.e., -O-(substituted alkyl)).
- alkyl moiety of an alkoxy group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, acylsulfonamido, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , - S(O) t R a - (where t is 1 or 2), -OC(O)-R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , -OC(O)N(R a ) 2 , - C(O)N(R a ) 2
- a (C1-6)alkoxycarbonyl group is an alkoxy group having from 1 to 6 carbon atoms attached through its oxygen to a carbonyl linker.
- Lower alkoxycarbonyl refers to an alkoxycarbonyl group wherein the alkoxy group is a lower alkoxy group.
- substituted alkoxycarbonyl refers to the group (substituted alkyl)-O- C(O)- wherein the group is attached to the parent structure through the carbonyl functionality.
- substituents which independently are: alkyl, heteroalkyl, acylsulfonamido, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , -S(O) t R a - (where t is 1 or 2), -OC(O)-R a , -
- Acyl refers to the groups (alkyl)-C(O)-, (aryl)-C(O)-, (heteroaryl)-C(O)-, (heteroalkyl)-C(O)- and (heterocycloalkyl)-C(O)-, wherein the group is attached to the parent structure through the carbonyl functionality. If the R radical is heteroaryl or heterocycloalkyl, the hetero ring or chain atoms contribute to the total number of chain or ring atoms.
- the alkyl, aryl or heteroaryl moiety of the acyl group is optionally substituted by one or more substituents which are independently alkyl, heteroalkyl, acylsulfonamido, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , -S(O) t R a - (where t is 1 or 2), -OC(O)-R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , -OC(O)N(R a ) 2 , -C(O)N, -C(O)N
- R of an acyloxy group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , -S(O)tR a - (where t is 1 or 2), -OC(O)-R a , - N(R a ) 2 , -C(O)R a , -C(O)OR a , -OC(O)N(R a ) 2 , -C(O)N(R a ) 2 , -N(R a )C(O)C(O)
- Amino or “amine” refers to a -N(R a ) 2 radical group, where each R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl, unless stated otherwise specifically in the specification.
- R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl, unless stated otherwise specifically in the specification.
- R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl
- -N(R a ) 2 is intended to include, but is not limited to, 1-pyrrolidinyl and 4- morpholinyl.
- an amino group is optionally substituted by one or more substituents which independently are: alkyl, acylsulfonamido, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , -S(O)tR a - (where t is 1 or 2), -OC(O)- R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a ,
- substituted amino also refers to N-oxides of the groups -NHR d , and NR d R d each as described above. N-oxides is prepared by treatment of the corresponding amino group with, for example, hydrogen peroxide or m-chloroperoxybenzoic acid.
- “Amide” or “amido” refers to a chemical moiety with formula -C(O)NR a R b or -NR a C(O)R b , where R a and R b are selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), each of which moiety may itself be optionally substituted.
- the R a and R b of -C(O)N R a R b amide may optionally be taken together with the nitrogen to which they are attached to form a 4-, 5-, 6- or 7-membered ring.
- an amido group is optionally substituted independently by one or more of the substituents as described herein for alkyl, amino, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl.
- An amide may be an amino acid or a peptide molecule attached to a compound disclosed herein, thereby forming a prodrug.
- the procedures and specific groups to make such amides are known to those of skill in the art and can readily be found in seminal stheces such as Greene and Wuts, Protective Groups in Organic Synthesis, 3 rd Ed., John Wiley & Sons, New York, N.Y., 1999, which is incorporated herein by reference in its entirety.
- “Aromatic” or “aryl” or “Ar” refers to an aromatic radical with six to ten ring atoms (e.g., C6-C10 aromatic or C6-C10 aryl) which has at least one ring having a conjugated pi electron system which is carbocyclic (e.g., phenyl, fluorenyl, and naphthyl).
- Bivalent radicals formed from substituted benzene derivatives and having the free valences at ring atoms are named as substituted phenylene radicals.
- Bivalent radicals derived from univalent polycyclic hydrocarbon radicals whose names end in “-yl” by removal of one hydrogen atom from the carbon atom with the free valence are named by adding “-idene” to the name of the corresponding univalent radical, e.g., a naphthyl group with two points of attachment is termed naphthylidene.
- a numerical range such as “6 to 10” refers to each integer in the given range; e.g., “6 to 10 ring atoms” means that the aryl group may consist of 6 ring atoms, 7 ring atoms, etc., up to and including 10 ring atoms.
- an aryl moiety is optionally substituted by one or more substituents which are independently alkyl, heteroalkyl, acylsulfonamido, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , -S(O)tR a - (where t is 1 or 2), -OC(O)-R a , - N(R a ) 2 , -C(O)R a , -C(O)OR a , -OC(
- “Aralkyl” or “arylalkyl” refers to an (aryl)alkyl-radical where aryl and alkyl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for aryl and alkyl respectively.
- “Ester” refers to a chemical radical of formula -COOR, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon).
- esters are known to those of skill in the art and can readily be found in seminal stheces such as Greene and Wuts, Protective Groups in Organic Synthesis, 3 rd Ed., John Wiley & Sons, New York, N.Y., 1999, which is incorporated herein by reference in its entirety.
- an ester group is optionally substituted by one or more substituents which independently are: alkyl, acylsulfonamido, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -SR a , -S(O)tR a - (where t is 1 or 2), -OC(O)- R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , -OC(O)N(R a ) 2 , -C(O)N(R a ) 2 , -N(R a
- Fluoroalkyl refers to an alkyl radical, as defined above, that is substituted by one or more fluoro radicals, as defined above, for example, trifluoromethyl, difluoromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, and the like.
- the alkyl part of the fluoroalkyl radical may be optionally substituted as defined above for an alkyl group.
- Halo “halide,” or, alternatively, “halogen” is intended to mean fluoro, chloro, bromo or iodo.
- haloalkyl examples include alkyl, alkenyl, alkynyl and alkoxy structures that are substituted with one or more halo groups or with combinations thereof.
- fluoroalkyl and “fluoroalkoxy” include haloalkyl and haloalkoxy groups, respectively, in which the halo is fluorine.
- Heteroalkyl refers to optionally substituted alkyl, alkenyl and alkynyl radicals and which have one or more skeletal chain atoms selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus or combinations thereof.
- a numerical range may be given - e.g., C 1 -C 4 heteroalkyl which refers to the chain length in total, which in this example is 4 atoms long.
- a heteroalkyl group may be substituted with one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, acylsulfonamido, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , -SR a , - S(O)tR a - (where t is 1 or 2), -OC(O)-R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , -OC(O)N(R a ) 2 , - C(O)N(R a ) 2 , -N(R a )C(O)OR a
- Heteroalkylaryl refers to an -(heteroalkyl)aryl radical where heteroalkyl and aryl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for heteroalkyl and aryl, respectively.
- Heteroalkylheteroaryl refers to an -(heteroalkyl)heteroaryl radical where heteroalkyl and heteroaryl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for heteroalkyl and heteroaryl, respectively.
- Heteroalkylheterocycloalkyl refers to an -(heteroalkyl)heterocycloalkyl radical where heteroalkyl and heterocycloalkyl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for heteroalkyl and heterocycloalkyl, respectively.
- Heteroalkylcycloalkyl refers to an -(heteroalkyl)cycloalkyl radical where heteroalkyl and cycloalkyl are as disclosed herein and which are optionally substituted by one or more of the substituents described as suitable substituents for heteroalkyl and cycloalkyl, respectively.
- Heteroaryl or “heteroaromatic” or “HetAr” refers to a 5- to 18-membered aromatic radical (e.g., C 5 -C 13 heteroaryl) that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur, and which may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system.
- a numerical range such as “5 to 18” refers to each integer in the given range - e.g., “5 to 18 ring atoms” means that the heteroaryl group may consist of 5 ring atoms, 6 ring atoms, etc., up to and including 18 ring atoms.
- Bivalent radicals derived from univalent heteroaryl radicals whose names end in “-yl” by removal of one hydrogen atom from the atom with the free valence are named by adding “- idene” to the name of the corresponding univalent radical - e.g., a pyridyl group with two points of attachment is a pyridylidene.
- a N-containing “heteroaromatic” or “heteroaryl” moiety refers to an aromatic group in which at least one of the skeletal atoms of the ring is a nitrogen atom.
- the polycyclic heteroaryl group may be fused or non-fused.
- the heteroatom(s) in the heteroaryl radical are optionally oxidized.
- heteroaryl may be attached to the rest of the molecule through any atom of the ring(s).
- heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1,3-benzodioxolyl, benzofuranyl, benzooxazolyl, benzo[d]thiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, benzo[b][1,4]oxazinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzoxazolyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benz
- a heteroaryl moiety is optionally substituted by one or more substituents which are independently: alkyl, acylsulfonamido, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , -SR a , -S(O) t R a - (where t is 1 or 2), -OC(O)-R a , - N(R a ) 2 , -C(O)R a , -C(O)OR a , -OC(O)N(R a ) 2 , -C(O)N(R a ) 2 , -C(O)N(R a )
- Substituted heteroaryl also includes ring systems substituted with one or more oxide (-O-) substituents, such as, for example, pyridinyl N-oxides.
- “Heteroarylalkyl” refers to a moiety having an aryl moiety, as described herein, connected to an alkylene moiety, as described herein, wherein the connection to the remainder of the molecule is through the alkylene group.
- “Heterocycloalkyl” refers to a stable 3- to 18-membered non-aromatic ring radical that comprises two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur.
- a numerical range such as “3 to 18” refers to each integer in the given range - e.g., “3 to 18 ring atoms” means that the heterocycloalkyl group may consist of 3 ring atoms, 4 ring atoms, etc., up to and including 18 ring atoms.
- the heterocycloalkyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems.
- the heteroatoms in the heterocycloalkyl radical may be optionally oxidized.
- One or more nitrogen atoms, if present, are optionally quaternized.
- the heterocycloalkyl radical is partially or fully saturated.
- the heterocycloalkyl may be attached to the rest of the molecule through any atom of the ring(s).
- heterocycloalkyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2- oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl,
- a heterocycloalkyl moiety is optionally substituted by one or more substituents which independently are: alkyl, acylsulfonamido, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, hydroxamate, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , -SR a , -S(O)tR a - (where t is 1 or 2), -OC(O)-R a , - N(R a ) 2 , -C(O)R a , -C(O)OR a , -OC(O)N(R a ) 2 , -C(O)N(R a ) 2 , -N
- Heterocycloalkyl also includes bicyclic ring systems wherein one non-aromatic ring, usually with 3 to 7 ring atoms, contains at least 2 carbon atoms in addition to 1-3 heteroatoms independently selected from oxygen, sulfur, and nitrogen, as well as combinations comprising at least one of the foregoing heteroatoms; and the other ring, usually with 3 to 7 ring atoms, optionally contains 1-3 heteroatoms independently selected from oxygen, sulfur, and nitrogen and is not aromatic.
- “Hydroxamate” refers to the –C(O)NR a OR a moiety, where each R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
- “Nitro” refers to the -NO2 radical.
- “Oxa” refers to the -O- radical.
- “Isomers” are different compounds that have the same molecular formula.
- “Stereoisomers” are isomers that differ only in the way the atoms are arranged in space - i.e., having a different stereochemical configuration.
- “Enantiomers” are a pair of stereoisomers that are non-superimposable mirror images of each other. A 1:1 mixture of a pair of enantiomers is a “racemic” mixture. The term “( ⁇ )” is used to designate a racemic mixture where appropriate.
- “Diastereoisomers” are stereoisomers that have at least two asymmetric atoms, but which are not mirror-images of each other. The absolute stereochemistry is specified according to the Cahn-Ingold-Prelog R-S system.
- Optically active (R)- and (S)-isomers is prepared using chiral synthons or chiral reagents, or resolved using conventional techniques.
- the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers.
- “Enantiomeric purity” as used herein refers to the relative amounts, expressed as a percentage, of the presence of a specific enantiomer relative to the other enantiomer.
- the enantiomeric purity is about 50% with respect to either the (R)- or (S)-isomer. If that compound has one isomeric form predominant over the other, for example, 80% (S)-isomer and 20% (R)-isomer, the enantiomeric purity of the compound with respect to the (S)-isomeric form is 80%.
- the enantiomeric purity of a compound is determined in a number of ways known in the art, including but not limited to chromatography using a chiral support, polarimetric measurement of the rotation of polarized light, nuclear magnetic resonance spectroscopy using chiral shift reagents which include but are not limited to lanthanide containing chiral complexes or Pirkle’s reagents, or derivatization of a compounds using a chiral compound such as Mosher’s acid followed by chromatography or nuclear magnetic resonance spectroscopy.
- the enantiomerically enriched composition has a higher potency with respect to therapeutic utility per unit mass than does the racemic mixture of that composition.
- Enantiomers is isolated from mixtures by methods known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts; or preferred enantiomers is prepared by asymmetric syntheses. See, for example, Jacques, et al., Enantiomers, Racemates and Resolutions, Wiley Interscience, New York (1981); E. L. Eliel, Stereochemistry of Carbon Compounds, McGraw-Hill, New York (1962); and E. L. Eliel and S. H. Wilen, Stereochemistry of Organic Compounds, Wiley-Interscience, New York (1994).
- an enantiomerically enriched preparation of the (S)-enantiomer means a preparation of the compound having greater than 50% by weight of the (S)-enantiomer relative to the (R)-enantiomer, such as at least 75% by weight, or such as at least 80% by weight.
- the enrichment is significantly greater than 80% by weight, providing a “substantially enantiomerically enriched” or a “substantially non-racemic” preparation, which refers to preparations of compositions which have at least 85% by weight of one enantiomer relative to other enantiomer, such as at least 90% by weight, or such as at least 95% by weight.
- enantiomerically pure or “substantially enantiomerically pure” refers to a composition that comprises at least 98% of a single enantiomer and less than 2% of the opposite enantiomer.
- “Moiety” refers to a specific segment or functional group of a molecule.
- “Tautomers” are structurally distinct isomers that interconvert by tautomerization. “Tautomerization” is a form of isomerization and includes prototropic or proton-shift tautomerization, which is considered a subset of acid-base chemistry. “Prototropic tautomerization” or “proton-shift tautomerization” involves the migration of a proton accompanied by changes in bond order, often the interchange of a single bond with an adjacent double bond. Where tautomerization is possible (e.g., in solution), a chemical equilibrium of tautomers is reached.
- keto-enol tautomerization An example of tautomerization is keto-enol tautomerization.
- keto-enol tautomerization is the interconversion of pentane-2,4-dione and 4-hydroxypent-3-en-2-one tautomers.
- phenol-keto tautomerization Another example of tautomerization is phenol-keto tautomerization.
- phenol-keto tautomerization is the interconversion of pyridin-4-ol and pyridin-4(1H)-one tautomers.
- a “leaving group or atom” is any group or atom that will, under selected reaction conditions, cleave from the starting material, thus promoting reaction at a specified site.
- Protecting group is intended to mean a group that selectively blocks one or more reactive sites in a multifunctional compound such that a chemical reaction is carried out selectively on another unprotected reactive site and the group can then be readily removed or deprotected after the selective reaction is complete.
- a variety of protecting groups are disclosed, for example, in T. H. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd Edition, John Wiley & Sons, New York (1999).
- Solvate refers to a compound in physical association with one or more molecules of a pharmaceutically acceptable solvent.
- “Substituted” means that the referenced group may have attached one or more additional groups, radicals or moieties individually and independently selected from, for example, acyl, alkyl, alkylaryl, cycloalkyl, aralkyl, aryl, carbohydrate, carbonate, heteroaryl, heterocycloalkyl, hydroxamate, hydroxy, alkoxy, aryloxy, mercapto, alkylthio, arylthio, cyano, halo, carbonyl, ester, thiocarbonyl, isocyanato, thiocyanato, isothiocyanato, nitro, oxo, perhaloalkyl, perfluoroalkyl, phosphate, silyl, sulfinyl, sulfonyl, sulfonamidyl,
- substituents themselves may be substituted, for example, a cycloalkyl substituent may itself have a halide substituent at one or more of its ring carbons.
- optionally substituted means optional substitution with the specified groups, radicals or moieties.
- “Sulfanyl” refers to groups that include -S-(optionally substituted alkyl), -S- (optionally substituted aryl), -S-(optionally substituted heteroaryl) and -S-(optionally substituted heterocycloalkyl).
- “Sulfinyl” refers to groups that include -S(O)-H, -S(O)-(optionally substituted alkyl), -S(O)-(optionally substituted amino), -S(O)-(optionally substituted aryl), -S(O)- (optionally substituted heteroaryl) and -S(O)-(optionally substituted heterocycloalkyl).
- “Sulfonyl” refers to groups that include -S(O2)-H, -S(O2)-(optionally substituted alkyl), -S(O 2 )-(optionally substituted amino), -S(O 2 )-(optionally substituted aryl), -S(O 2 )- (optionally substituted heteroaryl), and -S(O2)-(optionally substituted heterocycloalkyl).
- a sulfonamido group is optionally substituted by one or more of the substituents described for alkyl, cycloalkyl, aryl, heteroaryl, respectively.
- a sulfonate group is optionally substituted on R by one or more of the substituents described for alkyl, cycloalkyl, aryl, heteroaryl, respectively.
- Compounds of the disclosure also include crystalline and amorphous forms of those compounds, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, and amorphous forms of the compounds, as well as mixtures thereof.
- Crystal form and “polymorph” are intended to include all crystalline and amorphous forms of the compound, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, and amorphous forms, as well as mixtures thereof, unless a particular crystalline or amorphous form is referred to.
- DETAILED DESCRIPTION Inhibition of PDE11A4 An enzyme called phosphodiesterase 11A (PDE11A) and its role in the neurobiological substrates of memory and social behaviors has been examined. PDE11A is a member of the large phosphodiesterase enzyme family and was originally cloned in 2000.
- the enzyme is derived from a single gene product, hydrolyzes both cAMP and cGMP and exists in 4 isoforms.
- the enzyme is predominantly expressed in the brain.
- PDE11A is found in the anterior hippocampus and more specifically in neurons in the superficial layer of CA1, the subiculum and the amygdalohippocampal area of the hippocampus.
- PDE11A4 is the only PDE whose expression in brain emanates from the hippocampus, a region of the brain associated with associated long term memory (aLTM).
- PDE11A appears to regulate important signals for memory consolidation, including glutamatergic and calcium/calmodulin-dependent kinase II (CamKII) signaling, as well as protein synthesis. It has been shown that cAMP and cGMP signaling are decreased in aged and demented hippocampus in rats and humans. These age- related decreases in cyclic nucleotides are associated with increased expression of PDE11A4 in rodents and in humans with hippocampal dementia, versus non-demented aged subjects with a history of traumatic brain injury.
- CamKII calcium/calmodulin-dependent kinase II
- PDE11A4 is a dual acting cyclic nucleotide hydrolase expressed in neurons in the CA1, subiculum, amygdalostriatal transition area, and amygdalohippocampal area of the extended hippocampal formation.
- PDE11A4 is the only PDE enzyme to emanate solely from the hippocampal formation, a key brain region for the formation of long-term memory.
- PDE11A4 expression increases in the hippocampal formation of both humans and rodents as they age.
- PDE11A knockout mice do not show age-related deficits in associative memory and show no gross histopathology.
- PDE11A4 can be useful as a therapeutic option for age-related cognitive decline.
- the present disclosure discloses novel PDE11A4 inhibitors with improved potency, including more than ten-fold improvement in potency compared to tadalafil in cell based activities, and/or improvements in pharmaceutical properties including selectivity and cell-penetrant PDE11A4 inhibitors.
- the PDE11A family which breaks down cAMP and cGMP, is comprised of a single gene that is spliced into 4 isoforms, PDE11A1 through PDE11A4.
- PDE11A4 The longest isoform, PDE11A4, is the isoform that is expressed in brain and it is ⁇ 95% homologous across mouse, rat and human. This high degree of homology argues that the results obtained in rodent models translates across species.
- PDE11A single nucleotide polymorphisms SNPs
- MDD major depressive disorder
- BPD BPD
- Both MDD and BPD have been conceptualized as diseases of accelerated aging. It was established that PDE11A4 is expressed in the brain.
- PDE11A4 was found in brain because PDE11A knockout (KO) mice was phenotyped and what few phenotypes they had were found to be of relevance to ventral hippocampal function. Thus, the search for a PDE11A isoform in brain was directed to the ventral hippocampal formation (VHIPP) to detect expression restricted to this small brain region. Indeed, it was discovered that PDE11A4 was strongly expressed in neurons of the superficial layer of CA1, the subiculum, and the adjacently connected amygdalohippocampal area (AHi) of the VHIPP (Figs.1A), with little expression in dorsal HIPP (DHIPP) and no expression in other brain regions or over 20 peripheral organs.
- VHIPP ventral hippocampal formation
- PDE11A4 Only the nervous system shows a specific PDE11A4 signal. This makes PDE11A4 very unique because in brain it is the only PDE to emanate only from the HIPP, a structure critical for social aLTMs. This enrichment of PDE11A4 in the HIPP has now been independently confirmed by multiple investigators. This—along with the fact that PDE11A is a highly druggable enzyme—makes PDE11A a very attractive drug target because it stands to selectively restore aberrant cyclic nucleotide signaling in a brain region affected by age- related decline without directly affecting signaling in other brain regions or peripheral organs that might lead to unwanted side effects.
- PDE11A4 molecularly defines an exceptionally discrete circuit within a brain region key to learning and memory, making it ripe for the study of age-related cognitive decline.
- cAMP and cGMP signaling are decreased in the aged and demented hippocampus (rodents and humans), particularly when there is a history of traumatic brain injury (TBI).
- TBI traumatic brain injury
- PDE11A KO mice Consistent with PDE11A4’s restricted expression pattern, PDE11A KO mice appear normal on a wide range of sensory, motor and anxiety/depression-related behaviors, and show no gross peripheral pathology at least up to 1 year of age (later ages not assessed,). Instead, PDE11A KO mice exhibit select social phenotypes such as preferring to interact with other PDE11A KO mice vs wild-type (WT) mice and showing differences in the consolidation of social memories. PDE11A KO mice also have an increased sensitivity to the behavioral effects of lithium.
- PDE11A4 appears to regulate signals that are important for memory consolidation, including glutamatergic and calcium/calmodulin- dependent kinase II (CamKII) signaling as well as protein synthesis.
- CamKII calcium/calmodulin- dependent kinase II
- the longest isoform, PDE11A4 is ⁇ 95% homologous across mouse, rat and human. Tissue-specific distribution and function of other isoforms is discussed by others, and these isoforms are not present in the CNS. Given the paucity of PDE11A inhibitor discovery to date, no information on inhibition of these isoforms is in the public domain.
- PDE11A4 is strongly expressed in neurons of the ventral hippocampal formation (VHIPP; a.k.a. anterior hippocampus in primates), with much lower levels of expression in dorsal hippocampus as well as the adjacent amygdalohippocampal region and in some mice the nearby amygdalostriatal transition area.
- VHIPP ventral hippocampal formation
- PDE11A4 expression was reliably measured in the spinal cord and dorsal root ganglion (i.e., present in wild-type but not Pde11a knockout mice), with no reliable PDE11A4 expression observed in 20 peripheral organs.
- PDE11A4 unique because in brain it is the only PDE to be expressed preferentially in the VHIPP, a structure critical for associative long-term memories.
- PDE11A is a druggable enzyme makes PDE11A an attractive drug target because it stands to selectively restore aberrant cyclic nucleotide signaling in a brain region affected by various disease states without directly affecting signaling in other brain regions or peripheral organs.
- Pde11a KO mice appear normal on a wide range of sensory, motor and anxiety/depression-related behaviors, show no gross peripheral histopathology at least up to 1 year of age (later ages not assessed), and reproduce normally.
- PDE11A4 expression in the hippocampus increases across the lifespan of both humans and rodents.
- This age-related increase in PDE11A4 is consistent with literature showing decreases in cAMP and cGMP in the aged and demented hippocampus (rodents and humans), particularly when there is a history of traumatic brain injury (TBI).
- TBI traumatic brain injury
- In vitro and rodent studies have shown that age-related increases in PDE11A4 expression are driven by increased phosphorylation of the N-terminal regulatory domain at S117 and S124.
- Rodent studies also have shown these age-related increases in PDE11A4 expression drive age-related cognitive decline of social associative memories due to increased presence of the protein in the aged brain as opposed to a prolonged effect on the development of the brain.
- Fig.23 is an image showing a non-limiting example of a scheme of a synthesis of compounds 5 (a-s).
- Fig.24 is an image showing a non-limiting example of a scheme to prepare compounds 10 (a-b).
- Fig.25 is an image showing a non-limiting example of a scheme to prepare compounds 12 (a-b).
- Fig.26A is an image of exemplary PDE11A inhibitors of the disclosure.
- Fig.26B is an image and a table.
- Fig.27 is an image of exemplary PDE11A inhibitors of the disclosure.
- Fig.28 is an image of exemplary PDE11A inhibitors of the disclosure.
- Fig.29 is an image of HT22 cells overexpressing EmGFP- mPDE11A4.
- PDEs are discretely localized to specific subcellular domains. As a result, PDEs do not simply control the total cellular content of cyclic nucleotides, they generate individual pools or nanodomains of cyclic nucleotide signaling.
- cyclic nucleotide signaling allows a single cell to respond discretely to diverse intra- and extracellular signals.
- a PDE is localized is just as important to its overall function as is its catalytic activity.
- some cyclases and PDEs responsible for generating and breaking down cAMP/cGMP are expressed more in the cytosol than the membrane (like PDE11A4), others are enriched in the membrane (like the closely related PDE2A and PDE10A).
- cyclic nucleotide signaling deficits observed in bipolar disorder and Alzheimer’s disease appear to be more prominent in the cytosolic as opposed to membrane fractions.
- Associative long-term memories (aLTMs) —particularly those involving friends and family—are more susceptible to age-related cognitive decline than are recognition long- term memories (rLTMs) for reasons that are not well understood.
- the lack of knowledge of the molecular mechanisms that govern age-related decline slows the development of novel therapeutics.
- Age-related increases in phosphodiesterase 11A (PDE11A) an enzyme that breaks down cAMP/cGMP and regulates social behaviors, may be a fundamental mechanism underlying age-related cognitive decline of aLTMs for social experiences.
- PDE11A phosphodiesterase 11A
- the best controlled studies to date suggest that the longest isoform PDE11A4 is almost exclusively expressed in the ventral hippocampal formation (a.k.a.
- PDEs are expressed more in the cytosol than the membrane (e.g., PDE11A), while others are more highly expressed in the membrane versus cytosol (including PDE2A, PDE9A and PDE10A.
- PDEs are localized to specific subcellular domains, they are able to regulate individual pools or nanodomains of cyclic nucleotide signaling.
- Such subcellular compartmentalization of cyclic nucleotide signaling allows a single cell to respond specifically to simultaneous intra- and/or extracellular signals. Therefore, the subcellular localization of any PDE is equally important to its actual catalytic activity when considering its function.
- PDEs can become overexpressed and/or mislocalized with age and/or disease, which compromises the integrity of this physiological segregation of signals. Indeed, age-related diseases and neuropsychiatric diseases can show a loss of cyclic nucleotide signaling in one subcellular compartment but not another, suggesting therapeutic strategies should optimally target enzymes in a compartment-specific manner.
- phosphodiesterase 11A (PDE11A) has garnered particular interest in the context of altered cyclic nucleotide signaling related to ARCD and early-onset Alzheimer’s disease. PDE11A is encoded by a single gene and has four isoforms.
- the PDE11A catalytic domain is located within the C-terminal region, which is common to all isoforms, while the N-terminal region serves a regulatory function and is unique to each isoform.
- the regulatory N-terminus of PDE11A4, the longest PDE11A isoform, is unique in that it contains two full GAF (cGMP binding PDE, Anabaena adenylyl cyclase and E. coli FhlA) domains.
- the GAF-A domain binds cGMP as a potential allosteric regulatory site and the GAF-B domain regulates protein-protein interactions, including homodimerization.
- PDE11A4 is unique in that it is the only PDE whose expression in brain emanates solely from the extended hippocampal formation, a brain region critical to learning and memory and vulnerable to age-related deficits in cyclic nucleotide signaling. Possibly contributing to these hippocampal cyclic nucleotide signaling deficits are age-related increases in PDE11A4 expression that are conserved across mice, rats and humans.
- PDE11A4 protein expression is deleterious as 1) PDE11A KO mice are protected against age-related cognitive decline (ARCD) of remote social associative long- term memories (aLTMs) and 2) mimicking age-related overexpression of PDE11A4 in the CA1 field of hippocampus of either young or old PDE11A KO mice is sufficient to mimic ARCD of remote social aLTMs.
- ARCD age-related cognitive decline
- aLTMs remote social associative long- term memories
- membrane-associated PDE11A4 may be considered ectopic. That said, the membrane pool of PDE11A4—although lesser in relative quantity—continually shows itself to be critical in regulating social behaviors. For instance, it was found that social isolation selectively decreases expression of membrane-associated PDE11A4 in the VHIPP, and that these isolation-induced decreases in PDE11A4 are sufficient to cause changes in subsequent social preferences and social memory. Similarly, PDE11A4 protein expression differences in VHIPP of BALB/cJ versus C57BL/6J mice are restricted to the membrane pool.
- the disclosure provides a novel class of PDE11A4 modulators, which is a useful therapeutic target for treating social deficits associated with schizophrenia, bipolar disorder, or autism as well as for treating cognitive deficits/dementia associated with age-related cognitive decline, traumatic brain injury, or Alzheimer’s disease.
- the disclosure provides compounds that inhibit the activity of PDE11A4 and are capable of 1) altering social preferences/compatibility within the context of neuropsychiatric or neurodevelopmental disorders, 2) reversing cognitive decline associated with aging, dementia associated with traumatic brain injury, and/or Alzheimer’s disease, and/or 3) alleviating other disorders where PDE11A4 forms accumulated proteinopathies, particularly in the membrane fraction.
- PDE11A4 forms accumulated proteinopathies, particularly in the membrane fraction.
- the role of PDE11A4 in actual brain function is explored, and intramolecular mechanisms that control how PDE11A4 functions in terms of enzymatic activity and subcellular trafficking are examined.
- the disclosure includes molecules capable of inhibiting PDE11A.
- the compounds described herein are PDE11A inhibitors capable of inhibiting PDE11A1, PDE11A2, PDE11A3, and/or PDE11A4.
- the PDE11A inhibitor is a PDE11A1 inhibitor.
- the PDE11A inhibitor is a PDE11A2 inhibitor.
- the PDE11A inhibitor is a PDE11A3 inhibitor.
- the PDE11A inhibitor is a PDE11A4 inhibitor.
- the PDE11A4 inhibitor is a PDE11A4 selective inhibitor.
- the PDE11A4 inhibitor of the disclosure is about 1-fold, about 5-fold, about 10- fold, about 20-fold, about 50-fold, about 100-fold, about 500-fold, or about 1000-fold more selective for PDE11A4 over PDE11A1, PDE11A2, and/or PDE11A3.
- the disclosure provides a compound of formula (A), or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof: formula (A) wherein in formula (A): R 1 is selected from 5-membered heterocycle, 5-membered heteroaryl, 6-membered R heterocycle, and 6-membered heteroaryl; or R 1 is R 3 is selected from 5-membered heterocycle, 5-membered heteroaryl, 6-membered heterocycle, and 6-membered heteroaryl; R 2 and R 4 are independently at each occurrence selected from hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkylheteroaryl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylaryl, optionally substituted aryl, optionally substituted aryl
- the disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof: formula (I) wherein in formula (I): R 1 and R 3 are independently at each occurrence a 5-membered heterocycle, 5- membered heteroaryl, a 6-membered heterocycle, or 6-membered heteroaryl; R 2 and R 4 are independently at each occurrence selected from hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkylheteroaryl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylaryl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, hydroxy, halo,
- R 1 is a 5-membered heteroaryl ring. In some embodiments, R 1 is selected from thiazole, oxazole, imidazole, thiadiazole, oxadiazole, and triazole.
- the compound of formula (A) or formula (I) is a compound having formula (II), or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof: formula (II) wherein in formula (II): A is S, NR 5 , N, or O, optionally A is S or O; B 1 and B 2 are independently at each occurrence selected from N, O, or CR 5 , wherein B 1 is not O when either A or B 2 is O; and each R 5 is independently at each occurrence selected from hydrogen, C 1 -C 6 alkyl (e.g., C 1 -C 3 alkyl), and C 1 -C 3 haloalkyl (e.g., CH 2 F, CHF 2 , CF 3 ); R 3 is selected from 5-membered heterocycle, 5-membered heteroaryl, 6-membered heterocycle, and 6-membered heteroaryl; R 2 and R 4 are independently at each occurrence selected from hydrogen,
- R 1 is selected from wherein: A a is O or S, and B a is N or CH; A b is N, and B b is O or S; A c is O or S, and B c is N; and R 5 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), and C1- C 3 haloalkyl (e.g., CH 2 F, CHF 2 , or CF 3 ).
- C 1 -C 3 alkyl e.g., methyl, ethyl, propyl, isopropyl
- C1- C 3 haloalkyl e.g., CH 2 F, CHF 2 , or CF 3
- R 1 is selected from some embodiments
- R 5 is selected from H, methyl, ethyl, propyl, isopropyl, CH 2 F, and CHF 2 .
- R 5 is C 1 -C 3 alkyl.
- R 1 is selected from , , , , and [00176]
- R 2 is selected from hydrogen, C 3 -C 6 cycloalkyl, C 1 -C 6 alkyl (e.g., C 1 -C 4 straight chain alkyl, C 3 -C 6 branched alkyl), -CF 3 , -CH 2 F, -CHF 2 , and - C(R 6 R 7 ) p X; wherein, p is an integer from 1-3; R 6 and R 7 are each independently selected from hydrogen, C 1 -C 3 alkyl and C 3 -C 6 cycloalkyl; X is selected from -OH, -CN, -OR 8 , and NR 9 R 10 ; R 8 is selected from C 1 -C 3 alkyl, and C 3 -C 6 cycloalkyl; and R 9 and R 10 are each independently selected from hydrogen, C 1 -C 3 alkyl and C 3
- R 2 is C 1 -C 4 straight chain alkyl. In some embodiments, R 8 is C 1 -C 3 straight or branched alkyl. [00177] In some embodiments, R 2 is selected from hydrogen and C 1 -C 6 alkyl, optionally - CH 3 . [00178] In some embodiments, R 2 is selected from hydrogen, C 1 -C 4 alkyl (e.g., methyl, ethyl, propyl, isopropyl, butyl, tertbutyl), -CF 3 , -CH 2 F, and -CHF 2 . In some embodiments, R 2 is selected from H, -CH 3 , -CF 3 , and -CHF 2 .
- C 1 -C 4 alkyl e.g., methyl, ethyl, propyl, isopropyl, butyl, tertbutyl
- R 2 is selected from H, -CH 3 , -CF 3 , and
- R 3 is a 5-membered heteroaryl ring. In some embodiments, R 3 is selected from pyrrole, thiophene, furan, pyrazole, thiazole, isothiazole, oxazole, isoxazole, and imidazole. [00180] In some embodiments, R 3 is selected from , , , , wherein R 11 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), C 3 -C 6 cycloalkyl, CF 3 , CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F.
- C 1 -C 3 alkyl e.g., methyl, ethyl, propyl, isopropyl
- C 3 -C 6 cycloalkyl CF 3 , CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F.
- R 3 is C 1 -C 3 straight or branched alkyl. [00181] In some embodiments, R 3 is selected from [00182] In some embodiments, R 11 is selected from hydrogen, methyl, ethyl, propyl, isopropyl, -CF 3 , and -CH 2 CF 3 . [ [00184] In some embodiments, R 4 is selected from hydrogen, C 1 -C 6 alkyl, and halo (e.g., F or Cl).. In some embodiments, R 4 is F or Cl. In some embodiments, n is 0, 1, or 2. In some embodiments, n is 0 or 1.
- R 4 is selected from hydrogen, 4-fluoro, 3-fluoro, 2-fluoro, 2-chloro, and 4-chloro. In some embodiments, R 4 is selected from hydrogen, 4-fluoro, 2- fluoro, and 2-chloro. In some embodiments, n is 2 and each R 4 is 2-fluoro and 4-fluoro.
- the compound of formula (A), formula (I), or formula (II) is a compound having any one of formula 1001-1729 or 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof: Formula (Ia) formula (Ib)
- the compound of formula (A), formula (I), or formula (II) is a compound having any one of formula 2001-2337, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof:
- the compound of formula (A), formula (I), or formula (II) is a compound having any one of the following formula, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof:
- R 1 in formula (A) is R 20 [00190] In some embodiments, R 1 in formula (A) is , R 20 is selected from H, optionally substituted C 1 -C 6 alkyl, and optionally substituted C 1 -C 6 haloalkyl; R 21 is selected from optionally substituted C 1 -C 6 alkyl and optionally substituted C 1 -C 6 haloalkyl; or R 20 and R 21 are joined to form a 4-6-membered heterocycle with the nitrogen to which they are each bound, wherein the heterocycle may comprise 1 additional heteroatom selected from O and NH.
- R 20 is selected from H, methyl, ethyl, propyl, isopropyl, butyl, tert-butyl, CF 3 , CHF 2 , CH 2 F, CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F; and R 21 is selected from methyl, ethyl, propyl, isopropyl, butyl, tert-butyl, CF 3 , CHF 2 , CH 2 F, CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F; or R 20 and R 21 are joined to form a 4-6-membered heterocycle with the nitrogen to which they are each bound, wherein the heterocycle is selected from azetidine, pyrrolidine, morpholine, and piperazine.
- R 20 is selected from H, methyl, ethyl, propyl, tert-butyl, and CH 2 CH 2 F; and R 21 is selected from methyl, ethyl, propyl, tert-butyl, and CH 2 CH 2 F; or R 20 and R 21 are joined to form a 4-6-membered heterocycle with the nitrogen to which they are each bound, wherein the heterocycle is selected from azetidine, pyrrolidine, morpholine, and piperazine.
- the compound of formula (A) is a compound having formula (III), or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof:or pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof: formula (III) wherein in formula (III): R 3 is selected from 5-membered heterocycle, 5-membered heteroaryl, 6-membered heterocycle, and 6-membered heteroaryl; R 2 and R 4 are independently at each occurrence selected from hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkylheteroaryl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylaryl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl
- R 2 is selected from hydrogen, C 3 -C 6 cycloalkyl, C 1 -C 6 alkyl (e.g., C 1 -C 4 straight chain alkyl, C 3 -C 6 branched alkyl), -CF 3 , -CH 2 F, -CHF 2 , and - C(R 6 R 7 )pX; wherein, p is an integer from 1-3; R 6 and R 7 are each independently selected from hydrogen, C 1 -C 3 alkyl, and C 3 -C 6 cycloalkyl; X is selected from -OH, -CN, -OR 8 , and NR 9 R 10 ; R 8 is selected from C 1 -C 3 alkyl (e.g., C 1 -C 3 straight or branched alkyl), and C 3 -C 6 cycloalkyl; and R 9 and R 10 are each independently selected from hydrogen, C 1 -C 3 alkyl, and C 3 -C 6 alkyl; and R 9
- R 2 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), -CF 3 , -CH 2 F, and -CHF 2 .
- R 2 is selected from hydrogen and -CH 3 .
- R 3 is a 5-membered heteroaryl ring, optionally selected from pyrrole, thiophene, furan, pyrazole, thiazole, isothiazole, oxazole, isoxazole, and imidazole.
- R 3 is selected from , wherein R 11 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), C 3 -C 6 cycloalkyl, CF 3 , CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F. [00199] In some embodiments, R 3 is selected from , [00200] In some embodiments, R 11 is selected from hydrogen, methyl, ethyl, propyl, isopropyl, -CF 3 , and -CH 2 CF 3 .
- R 3 is selected from , [00202] In some embodiments, R 4 is selected from hydrogen, C 1 -C 6 alkyl, and halo (e.g., F or Cl). In some embodiments, n is 0 or 1. [00203] In some embodiments, R 4 is selected from hydrogen, 4-fluoro, 3-fluoro, 2-fluoro, 2-chloro, and 4-chloro. In some embodiments, R 4 is selected from hydrogen, 4-fluoro, 3- fluoro, 2-fluoro, and 4-chloro. [00204] In some embodiments, the compound of of formula (A) or formula (III) is a compound having any one of formula 3001-3031, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof:
- the compound having a formula of any one of formula (A), formula (I), formula (II), formula (III), formula 1001-1729, formula 2001-2337, formula 3001-3031, or formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof, is a PDE11A4 inhibitor.
- Methods of Treatment [00206] The compounds and compositions described herein can be used in methods for treating diseases and/or disorders. In some embodiments, the compounds and compositions described herein can be used in methods for treating diseases associated with PDE11A activity. In some embodiments, the compounds and compositions described herein can be used in methods for treating diseases associated with PDE11A4 activity.
- the disclosure relates to a method of treating a disease alleviated by inhibiting PDE11A activity in a patient in need thereof, including administering to the patient a therapeutically effective amount of a PDE11A inhibitor.
- the PDE11A inhibitor is a compound of formula (I), a compound of any one of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, or formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- the disclosure relates to a method of treating a disease alleviated by inhibiting PDE11A4 activity in a patient in need thereof, including administering to the patient a therapeutically effective amount of a PDE11A4 inhibitor.
- the PDE11A4 inhibitor is a compound of formula (I), a compound of any one of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, or formula 4001-4729,, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- the disease or disorder is associated with cognitive decline.
- the disease or disorder is selected from dementia, Alzheimer’s Disease (AD) including mild Alzheimer's disease and early-onset Alzheimer’s disease, Down’s syndrome, vascular dementia (cerebral amyloid angiopathy and stroke), dementia with Lewy bodies, HIV dementia, Mild Cognitive Impairment (MCI); Age- Associated Memory Impairment (AAMI); Age-Related Cognitive Decline (ARCD) (including age-related cognitive decline of associative long-term memories (aLTMs), dementia associated with traumatic brain injury, preclinical Alzheimer's Disease (PCAD); Cognitive Impairment No Dementia (CIND), and cognitive decline associate with spatial memory, other depression-related behaviors, additional anxiety-related behaviors, sensorimotor gating, and social behaviors.
- AD Alzheimer’s Disease
- AD Alzheimer's Disease
- MCI Mild Cognitive Impairment
- AAMI Age- Associated Memory Impairment
- ARCD Age-Related Cognitive Decline
- ALD Alzheimer-Related Cognitive Decline
- PCAD preclinical Alzheimer's
- “cognitive decline” includes be any negative change in an animal’s cognitive function.
- cognitive decline includes but is not limited to, memory loss (e.g. behavioral memory loss), failure to acquire new memories, confusion, impaired judgment, personality changes, disorientation, or any combination thereof.
- Pharmaceutical Compositions [00209]
- an active pharmaceutical ingredient or combination of active pharmaceutical ingredients such as any of the PDE11A inhibitors (e.g., PDE11A4 inhibitors) of the disclosure, is provided as a pharmaceutically acceptable composition.
- the PDE11A inhibitor is a compound of formula (I), a compound of any one of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001- 1729, formula 2001-2337, formula 3001-3031, or formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- the PDE11A inhibitor is a PDE11A4 inhibitor.
- the PDE11A4 inhibitor is a compound of formula (I), a compound of any one of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, or formula 4001-4729, , or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- the disclosure relates to a pharmaceutical composition including a therapeutically effective amount of a PDE11A inhibitor for the treatment of a disease alleviated by inhibiting PDE11A activity in a patient in need thereof, and a physiologically compatible carrier medium.
- the disclosure relates to a pharmaceutical composition including a therapeutically effective amount of a PDE11A4 inhibitor for the treatment of a disease alleviated by inhibiting PDE11A4 activity in a patient in need thereof, and a physiologically compatible carrier medium.
- the disease is associated with cognitive decline.
- the PDE11A inhibitor is a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- the PDE11A inhibitor is a PDE11A4 inhibitor.
- the PDE11A4 inhibitor is a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- the disease or disorder is selected from dementia, Alzheimer’s Disease (AD) including mild Alzheimer's disease and early-onset Alzheimer’s disease, Down’s syndrome, vascular dementia (cerebral amyloid angiopathy and stroke), dementia with Lewy bodies, HIV dementia, Mild Cognitive Impairment (MCI); Age- Associated Memory Impairment (AAMI); Age-Related Cognitive Decline (ARCD) (including age-related cognitive decline of associative long-term memories (aLTMs), dementia associated with traumatic brain injury, preclinical Alzheimer's Disease (PCAD); Cognitive Impairment No Dementia (CIND), and cognitive decline associate with spatial memory, other depression-related behaviors, additional anxiety-related behaviors, sensorimotor gating, and social behaviors.
- AD Alzheimer’s Disease
- AD Alzheimer's Disease
- MCI Mild Cognitive Impairment
- AAMI Age- Associated Memory Impairment
- ARCD Age-Related Cognitive Decline
- ALD Alzheimer-Related Cognitive Decline
- PCAD preclinical Alzheimer's
- the concentration of each of the active pharmaceutical ingredients provided in the pharmaceutical compositions of the disclosure is less than, for example, 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.00
- the concentration of each of the active pharmaceutical ingredients provided in the pharmaceutical compositions of the disclosure is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 11.75%, 11.50%, 11.25% 11%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.7
- the concentration of each of the active pharmaceutical ingredients provided in the pharmaceutical compositions of the disclosure is in the range from about 0.0001% to about 50%, about 0.001% to about 40%, about 0.01% to about 30%, about 0.02% to about 29%, about 0.03% to about 28%, about 0.04% to about 27%, about 0.05% to about 26%, about 0.06% to about 25%, about 0.07% to about 24%, about 0.08% to about 23%, about 0.09% to about 22%, about 0.1% to about 21%, about 0.2% to about 20%, about 0.3% to about 19%, about 0.4% to about 18%, about 0.5% to about 17%, about 0.6% to
- the concentration of each of the active pharmaceutical ingredients provided in the pharmaceutical compositions of the disclosure is in the range from about 0.001% to about 10%, about 0.01% to about 5%, about 0.02% to about 4.5%, about 0.03% to about 4%, about 0.04% to about 3.5%, about 0.05% to about 3%, about 0.06% to about 2.5%, about 0.07% to about 2%, about 0.08% to about 1.5%, about 0.09% to about 1%, about 0.1% to about 0.9% w/w, w/v, or v/v of the pharmaceutical composition.
- the amount of each of the active pharmaceutical ingredients provided in the pharmaceutical compositions of the disclosure is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25
- the amount of each of the active pharmaceutical ingredients provided in the pharmaceutical compositions of the disclosure is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015
- Each of the active pharmaceutical ingredients according to the disclosure is effective over a wide dosage range.
- dosages independently range from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that may be used.
- the exact dosage will depend upon the route of administration, the form in which the compound is administered, the gender and age of the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
- the clinically-established dosages of the PDE11A inhibitors (e.g., PDE11A4 inhibitors) of the disclosure may also be used if appropriate.
- the molar ratio of two active pharmaceutical ingredients in the pharmaceutical compositions is in the range from 10:1 to 1:10, preferably from 2.5:1 to 1:2.5, and more preferably about 1:1.
- the weight ratio of the molar ratio of two active pharmaceutical ingredients in the pharmaceutical compositions is selected from the group consisting of 20:1, 19:1, 18:1, 17:1, 16:1, 15:1, 14:1, 13:1, 12:1, 11:1, 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, and 1:20.
- the weight ratio of the molar ratio of two active pharmaceutical ingredients in the pharmaceutical compositions is selected from the group consisting of 20:1, 19:1, 18:1, 17:1, 16:1, 15:1, 14:1, 13:1, 12:1, 11:1, 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, and 1:20.
- the pharmaceutical compositions described herein such as any of the PDE11A inhibitors (e.g., PDE11A4 inhibitors) of the disclosure, for example a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, or formula 4001-4729, are for use in the treatment of a disease or disorder associated with cognitive decline.
- PDE11A inhibitors e.g., PDE11A4 inhibitors
- a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, or formula 4001-4729 are for use in the treatment of a disease or disorder associated with cognitive decline.
- the pharmaceutical compositions described herein are for use in the treatment of dementia, Alzheimer’s Disease (AD) including mild Alzheimer's disease and early-onset Alzheimer’s disease, Down’s syndrome, vascular dementia (cerebral amyloid angiopathy and stroke), dementia with Lewy bodies, HIV dementia, Mild Cognitive Impairment (MCI); Age- Associated Memory Impairment (AAMI); Age-Related Cognitive Decline (ARCD) (including age-related cognitive decline of associative long-term memories (aLTMs), dementia associated with traumatic brain injury, preclinical Alzheimer's Disease (PCAD); Cognitive Impairment No Dementia (CIND), or cognitive decline associate with spatial memory, other depression-related behaviors, additional anxiety-related behaviors, sensorimotor gating, and social behaviors.
- AD Alzheimer’s Disease
- MCI Mild Cognitive Impairment
- AAMI Age- Associated Memory Impairment
- ARCD Age-Related Cognitive Decline
- ALMs associative long-term memories
- PCAD preclinical Alzheimer's
- compositions for Oral Administration containing the active pharmaceutical ingredient or combination of active pharmaceutical ingredients, such as the PDE11A inhibitors (e.g., PDE11A4 inhibitors) described herein, and a pharmaceutical excipient suitable for oral administration.
- the disclosure provides a solid pharmaceutical composition for oral administration containing: (i) an effective amount of an active pharmaceutical ingredient or combination of active pharmaceutical ingredients, and (ii) a pharmaceutical excipient suitable for oral administration.
- the composition further contains (iii) an effective amount of a third active pharmaceutical ingredient, and optionally (iv) an effective amount of a fourth active pharmaceutical ingredient.
- the pharmaceutical composition may be a liquid pharmaceutical composition suitable for oral consumption.
- compositions of the disclosure suitable for oral administration can be presented as discrete dosage forms, such as capsules, sachets, or tablets, or liquids or aerosol sprays each containing a predetermined amount of an active ingredient as a powder or in granules, a solution, or a suspension in an aqueous or non-aqueous liquid, an oil-in-water emulsion, a water-in-oil liquid emulsion, powders for reconstitution, powders for oral consumptions, bottles (including powders or liquids in a bottle), orally dissolving films, lozenges, pastes, tubes, gums, and packs.
- discrete dosage forms such as capsules, sachets, or tablets, or liquids or aerosol sprays each containing a predetermined amount of an active ingredient as a powder or in granules, a solution, or a suspension in an aqueous or non-aqueous liquid, an oil-in-water emulsion, a water-in-oil liquid
- Such dosage forms can be prepared by any of the methods of pharmacy, but all methods include the step of bringing the active ingredient(s) into association with the carrier, which constitutes one or more necessary ingredients.
- the compositions are prepared by uniformly and intimately admixing the active ingredient(s) with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation.
- a tablet can be prepared by compression or molding, optionally with one or more accessory ingredients.
- Compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as powder or granules, optionally mixed with an excipient such as, but not limited to, a binder, a lubricant, an inert diluent, and/or a surface active or dispersing agent. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. [00225] The disclosure further encompasses anhydrous pharmaceutical compositions and dosage forms since water can facilitate the degradation of some compounds.
- water may be added (e.g., 5%) in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time.
- Anhydrous pharmaceutical compositions and dosage forms of the disclosure can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
- Pharmaceutical compositions and dosage forms of the disclosure which contain lactose can be made anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
- An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions may be packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits.
- Suitable packaging include, but are not limited to, hermetically sealed foils, plastic or the like, unit dose containers, blister packs, and strip packs.
- suitable packaging include, but are not limited to, hermetically sealed foils, plastic or the like, unit dose containers, blister packs, and strip packs.
- any of the usual pharmaceutical media can be employed as carriers, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like in the case of oral liquid preparations (such as suspensions, solutions, and elixirs) or aerosols; or carriers such as starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents can be used in the case of oral solid preparations, in some embodiments without employing the use of lactose.
- suitable carriers include powders, capsules, and tablets, with the solid oral preparations.
- Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, microcrystalline cellulose, and mixtures thereof.
- natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrol
- suitable fillers for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
- Disintegrants may be used in the compositions of the disclosure to provide tablets that disintegrate when exposed to an aqueous environment. Too much of a disintegrant may produce tablets which disintegrate in the bottle. Too little may be insufficient for disintegration to occur, thus altering the rate and extent of release of the active ingredients from the dosage form.
- a sufficient amount of disintegrant that is neither too little nor too much to detrimentally alter the release of the active ingredient(s) may be used to form the dosage forms of the compounds disclosed herein.
- the amount of disintegrant used may vary based upon the type of formulation and mode of administration, and may be readily discernible to those of ordinary skill in the art. About 0.5 to about 15 weight percent of disintegrant, or about 1 to about 5 weight percent of disintegrant, may be used in the pharmaceutical composition.
- Disintegrants that can be used to form pharmaceutical compositions and dosage forms of the disclosure include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums or mixtures thereof.
- Lubricants which can be used to form pharmaceutical compositions and dosage forms of the disclosure include, but are not limited to, calcium stearate, magnesium stearate, sodium stearyl fumarate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethylaureate, agar, or mixtures thereof.
- Additional lubricants include, for example, a syloid silica gel, a coagulated aerosol of synthetic silica, silicified microcrystalline cellulose, or mixtures thereof.
- a lubricant can optionally be added in an amount of less than about 0.5% or less than about 1% (by weight) of the pharmaceutical composition.
- the active pharmaceutical ingredient(s) may be combined with various sweetening or flavoring agents, coloring matter or dyes and, if so desired, emulsifying and/or suspending agents, together with such diluents as water, ethanol, propylene glycol, glycerin and various combinations thereof.
- the tablets can be uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
- a time delay material such as glyceryl monostearate or glyceryl distearate can be employed.
- Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
- Surfactants which can be used to form pharmaceutical compositions and dosage forms of the disclosure include, but are not limited to, hydrophilic surfactants, lipophilic surfactants, and mixtures thereof. That is, a mixture of hydrophilic surfactants may be employed, a mixture of lipophilic surfactants may be employed, or a mixture of at least one hydrophilic surfactant and at least one lipophilic surfactant may be employed.
- a suitable hydrophilic surfactant may generally have an HLB value of at least 10, while suitable lipophilic surfactants may generally have an HLB value of or less than about 10.
- HLB hydrophilic-lipophilic balance
- Surfactants with lower HLB values are more lipophilic or hydrophobic, and have greater solubility in oils, while surfactants with higher HLB values are more hydrophilic, and have greater solubility in aqueous solutions.
- Hydrophilic surfactants are generally considered to be those compounds having an HLB value greater than about 10, as well as anionic, cationic, or zwitterionic compounds for which the HLB scale is not generally applicable.
- lipophilic (i.e., hydrophobic) surfactants are compounds having an HLB value equal to or less than about 10.
- Hydrophilic surfactants may be either ionic or non-ionic. Suitable ionic surfactants include, but are not limited to, alkylammonium salts; fusidic acid salts; fatty acid derivatives of amino acids, oligopeptides, and polypeptides; glyceride derivatives of amino acids, oligopeptides, and polypeptides; lecithins and hydrogenated lecithins; lysolecithins and hydrogenated lysolecithins; phospholipids and derivatives thereof; lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acyl-lactylates; mono- and di-acetylated tartaric acid esters of mono- and di
- ionic surfactants include, by way of example: lecithins, lysolecithin, phospholipids, lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acylactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
- Ionic surfactants may be the ionized forms of lecithin, lysolecithin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatidic acid, phosphatidylserine, lysophosphatidylcholine, lysophosphatidylethanolamine, lysophosphatidylglycerol, lysophosphatidic acid, lysophosphatidylserine, PEG- phosphatidylethanolamine, PVP-phosphatidylethanolamine, lactylic esters of fatty acids, stearoyl-2-lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/diglycerides, cholylsarcosine, caproate, capry
- Hydrophilic non-ionic surfactants may include, but not limited to, alkylglucosides; alkylmaltosides; alkylthioglucosides; lauryl macrogolglycerides; polyoxyalkylene alkyl ethers such as polyethylene glycol alkyl ethers; polyoxyalkylene alkylphenols such as polyethylene glycol alkyl phenols; polyoxyalkylene alkyl phenol fatty acid esters such as polyethylene glycol fatty acids monoesters and polyethylene glycol fatty acids diesters; polyethylene glycol glycerol fatty acid esters; polyglycerol fatty acid esters; polyoxyalkylene sorbitan fatty acid esters such as polyethylene glycol sorbitan fatty acid esters; hydrophilic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids, and sterols; polyoxyethylene steas; poly
- the polyol may be glycerol, ethylene glycol, polyethylene glycol, sorbitol, propylene glycol, pentaerythritol, or a saccharide.
- Other hydrophilic-non-ionic surfactants include, without limitation, PEG-10 laurate, PEG-12 laurate, PEG-20 laurate, PEG-32 laurate, PEG-32 dilaurate, PEG-12 oleate, PEG-15 oleate, PEG-20 oleate, PEG-20 dioleate, PEG-32 oleate, PEG-200 oleate, PEG-400 oleate, PEG-15 stearate, PEG-32 distearate, PEG-40 stearate, PEG-100 stearate, PEG-20 dilaurate, PEG-25 glyceryl trioleate, PEG-32 dioleate, PEG-20 glyceryl laurate, PEG-30 glycerol,
- Suitable lipophilic surfactants include, by way of example only: fatty alcohols; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids esters; propylene glycol fatty acid esters; sorbitan fatty acid esters; polyethylene glycol sorbitan fatty acid esters; sterols and sterol derivatives; polyoxyethylated sterols and sterol derivatives; polyethylene glycol alkyl ethers; sugar esters; sugar ethers; lactic acid derivatives of mono- and di-glycerides; hydrophobic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids and sterols; oil-soluble vitamins/vitamin derivatives; and mixtures thereof.
- preferred lipophilic surfactants include glycerol fatty acid esters, propylene glycol fatty acid esters, and mixtures thereof, or are hydrophobic transesterification products of a polyol with at least one member of the group consisting of vegetable oils, hydrogenated vegetable oils, and triglycerides.
- the composition may include a solubilizer to ensure good solubilization and/or dissolution of the compound of the present disclosure and to minimize precipitation of the compound of the present disclosure. This can be especially important for compositions for non-oral use - e.g., compositions for injection.
- a solubilizer may also be added to increase the solubility of the hydrophilic drug and/or other components, such as surfactants, or to maintain the composition as a stable or homogeneous solution or dispersion.
- suitable solubilizers include, but are not limited to, the following: alcohols and polyols, such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol, hydroxypropyl methylcellulose and other cellulose derivatives, cyclodextrins and cyclodextrin derivatives; ethers of polyethylene glycols having an average molecular weight of about 200 to about 6000, such as t
- solubilizers may also be used. Examples include, but not limited to, triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, dimethylacetamide, N- methylpyrrolidone, N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropyl methylcellulose, hydroxypropyl cyclodextrins, ethanol, polyethylene glycol 200-100, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide.
- solubilizers include sorbitol, glycerol, triacetin, ethyl alcohol, PEG-400, glycofurol and propylene glycol.
- the amount of solubilizer that can be included is not particularly limited.
- the amount of a given solubilizer may be limited to a bioacceptable amount, which may be readily determined by one of skill in the art.
- the solubilizer can be in a weight ratio of 10%, 25%, 50%, 100%, or up to about 200% by weight, based on the combined weight of the drug, and other excipients. If desired, very small amounts of solubilizer may also be used, such as 5%, 2%, 1% or even less. Typically, the solubilizer may be present in an amount of about 1% to about 100%, more typically about 5% to about 25% by weight.
- the composition can further include one or more pharmaceutically acceptable additives and excipients.
- additives and excipients include, without limitation, detackifiers, anti-foaming agents, buffering agents, polymers, antioxidants, preservatives, chelating agents, viscomodulators, tonicifiers, flavorants, colorants, odorants, opacifiers, suspending agents, binders, fillers, plasticizers, lubricants, and mixtures thereof.
- an acid or a base may be incorporated into the composition to facilitate processing, to enhance stability, or for other reasons.
- Examples of pharmaceutically acceptable bases include amino acids, amino acid esters, ammonium hydroxide, potassium hydroxide, sodium hydroxide, sodium hydrogen carbonate, aluminum hydroxide, calcium carbonate, magnesium hydroxide, magnesium aluminum silicate, synthetic aluminum silicate, synthetic hydrocalcite, magnesium aluminum hydroxide, diisopropylethylamine, ethanolamine, ethylenediamine, triethanolamine, triethylamine, triisopropanolamine, trimethylamine, tris(hydroxymethyl)aminomethane (TRIS) and the like.
- bases that are salts of a pharmaceutically acceptable acid, such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, oxalic acid, para- bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid, and the like.
- a pharmaceutically acceptable acid such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids
- Salts of polyprotic acids such as sodium phosphate, disodium hydrogen phosphate, and sodium dihydrogen phosphate can also be used.
- the cation can be any convenient and pharmaceutically acceptable cation, such as ammonium, alkali metals and alkaline earth metals.
- Example may include, but not limited to, sodium, potassium, lithium, magnesium, calcium and ammonium.
- Suitable acids are pharmaceutically acceptable organic or inorganic acids. Examples of suitable inorganic acids include hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, nitric acid, boric acid, phosphoric acid, and the like.
- suitable organic acids include acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acids, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, methanesulfonic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid and uric acid.
- compositions for Injection containing an active pharmaceutical ingredient or combination of active pharmaceutical ingredients, such as a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, or formula 4001-4729, and a pharmaceutical excipient suitable for injection.
- active pharmaceutical ingredient or combination of active pharmaceutical ingredients such as a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, or formula 4001-4729, and a pharmaceutical excipient suitable for injection.
- Aqueous solutions in saline are also conventionally used for injection. Ethanol, glycerol, propylene glycol and liquid polyethylene glycol (and suitable mixtures thereof), cyclodextrin derivatives, and vegetable oils may also be employed.
- the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, for the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
- a coating such as lecithin
- the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and thimerosal.
- Sterile injectable solutions are prepared by incorporating an active pharmaceutical ingredient or combination of active pharmaceutical ingredients in the required amounts in the appropriate solvent with various other ingredients as enumerated above, as required, followed by filtered sterilization.
- dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
- a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
- certain desirable methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
- compositions for Topical Delivery containing an active pharmaceutical ingredient or combination of active pharmaceutical ingredients, such as compounds of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, and a pharmaceutical excipient suitable for transdermal delivery.
- active pharmaceutical ingredients such as compounds of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, and a pharmaceutical excipient suitable for transdermal delivery.
- compositions of the present disclosure can be formulated into preparations in solid, semi-solid, or liquid forms suitable for local or topical administration, such as gels, water soluble jellies, creams, lotions, suspensions, foams, powders, slurries, ointments, solutions, oils, pastes, suppositories, sprays, emulsions, saline solutions, dimethylsulfoxide (DMSO)-based solutions.
- DMSO dimethylsulfoxide
- carriers with higher densities are capable of providing an area with a prolonged exposure to the active ingredients.
- a solution formulation may provide more immediate exposure of the active ingredient to the chosen area.
- compositions also may comprise suitable solid or gel phase carriers or excipients, which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin.
- suitable solid or gel phase carriers or excipients which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin.
- humectants e.g., urea
- glycols e.g., propylene glycol
- alcohols e.g., ethanol
- fatty acids e.g., oleic acid
- surfactants e.g., isopropyl myristate and sodium lauryl sulfate
- pyrrolidones e.g., isopropyl myristate and sodium lauryl sulfate
- pyrrolidones e.glycerol monolaurate, sulfoxides, terpenes (e.g., menthol)
- amines amides, alkanes, alkanols, water, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
- transdermal delivery devices Such transdermal patches may be used to provide continuous or discontinuous infusion of an active pharmaceutical ingredient or combination of active pharmaceutical ingredients in controlled amounts, either with or without another active pharmaceutical ingredient.
- transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Patent Nos.5,023,252; 4,992,445; and 5,001,139, the entirety of which are incorporated herein by reference. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
- compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
- the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra and a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729 formula 2001-2337, formula 3001-3031, formula 4001-4729.
- the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
- Compositions in preferably pharmaceutically acceptable solvents may be nebulized by use of inert gases.
- Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a face mask tent, or intermittent positive pressure breathing machine.
- Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner. Dry powder inhalers may also be used to provide inhaled delivery of the compositions.
- compositions of a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, or a may also be prepared from compositions described herein and one or more pharmaceutically acceptable excipients suitable for sublingual, buccal, rectal, intraosseous, intraocular, intranasal, epidural, or intraspinal administration. Preparations for such pharmaceutical compositions are well-known in the art.
- Administration of an active pharmaceutical ingredient or combination of active pharmaceutical ingredients or a pharmaceutical composition thereof can be effected by any method that enables delivery of the compounds to the site of action.
- kits include oral routes, intraduodenal routes, parenteral injection (including intravenous, intraarterial, subcutaneous, intramuscular, intravascular, intraperitoneal or infusion), topical (e.g., transdermal application), rectal administration, via local delivery by catheter or stent or through inhalation.
- the active pharmaceutical ingredient or combination of active pharmaceutical ingredients can also be administered intraadiposally or intrathecally.
- Exemplary parenteral administration forms include solutions or suspensions of active compound in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
- Kits [00266] The disclosure also provides kits.
- kits include an active pharmaceutical ingredient or combination of active pharmaceutical ingredients, either alone or in combination in suitable packaging, and written material that can include instructions for use, discussion of clinical studies and listing of side effects.
- Such kits may also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider. Such information may be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials.
- the kit may further contain another active pharmaceutical ingredient.
- an active pharmaceutical ingredient or combination of active pharmaceutical ingredients are provided as separate compositions in separate containers within the kit.
- an active pharmaceutical ingredient or combination of active pharmaceutical ingredients are provided as a single composition within a container in the kit.
- Suitable packaging and additional articles for use e.g., measuring cup for liquid preparations, foil wrapping to minimize exposure to air, and the like
- Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits may also, in selected embodiments, be marketed directly to the consumer.
- the disclosure provides a kit comprising a composition comprising a therapeutically effective amount of an active pharmaceutical ingredient (e.g., a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729) or combination of active pharmaceutical ingredients or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- active pharmaceutical ingredient e.g., a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729
- active pharmaceutical ingredients e.g., a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729
- compositions are typically pharmaceutical compositions.
- the kit is for co-administration of
- the disclosure provides a kit comprising (1) a composition comprising a therapeutically effective amount of an active pharmaceutical ingredient (e.g., a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof), or combination of active pharmaceutical ingredients, and (2) a diagnostic test for determining whether a patient’s disease or disorder associated with cognitive decline is a particular subtype of a disease or disorder associated with cognitive decline. Any of the foregoing diagnostic methods may be utilized in the kit.
- an active pharmaceutical ingredient e.g., a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or pro
- kits described above are preferably for use in the treatment of the diseases and conditions described herein.
- the kits are for use in the treatment of a disease or disorder associated with cognitive decline .
- the kits are for use in the treatment of dementia, Alzheimer’s Disease (AD) including mild Alzheimer's disease and early-onset Alzheimer’s disease, Down’s syndrome, vascular dementia (cerebral amyloid angiopathy and stroke), dementia with Lewy bodies, HIV dementia, Mild Cognitive Impairment (MCI); Age-Associated Memory Impairment (AAMI); Age-Related Cognitive Decline (ARCD) (including age-related cognitive decline of associative long-term memories (aLTMs), dementia associated with traumatic brain injury, preclinical Alzheimer's Disease (PCAD); Cognitive Impairment No Dementia (CIND), or cognitive decline associate with spatial memory, other depression-related behaviors, additional anxiety-related behaviors, sensorimotor gating, and social behaviors.
- AD Alzheimer’s Disease
- MCI Mild Cognitive Impairment
- AAMI Age-Associated Memory Imp
- Dosages and Dosing Regimens [00270]
- the amounts of the pharmaceutical compositions administered using the methods herein, such as the dosages of a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof will be dependent on the human or mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the active pharmaceutical ingredients and the discretion of the prescribing physician.
- an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, such as about 1 to about 35 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.05 to 7 g/day, such as about 0.05 to about 2.5 g/day. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect - e.g., by dividing such larger doses into several small doses for administration throughout the day.
- the dosage of the pharmaceutical compositions and active pharmaceutical ingredients may be provided in units of mg/kg of body mass or in mg/m 2 of body surface area.
- the disclosure includes methods of treating a disease or disorder associated with cognitive decline in human subject suffering from the disease or disorder, the method comprising the steps of administering a therapeutically effective dose of an active pharmaceutical ingredient that is a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof to the human subject.
- a therapeutically effective dose of an active pharmaceutical ingredient that is a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof to the human subject.
- the disclosure includes methods of treating a disease or disorder associated with cognitive decline in a human subject suffering from the disease or disorder, the method comprising the steps of administering a therapeutically effective dose of an active pharmaceutical ingredient that is a compound of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof to the human subject to inhibit or decrease the activity of PDE11A protein.
- a pharmaceutical composition or active pharmaceutical ingredient is administered in a single dose.
- a pharmaceutical composition or active pharmaceutical ingredient is administered in multiple doses.
- a pharmaceutical composition is administered in multiple doses. Dosing may be once, twice, three times, four times, five times, six times, or more than six times per day. Dosing may be once a month, once every two weeks, once a week, or once every other day. In other embodiments, a pharmaceutical composition is administered about once per day to about 6 times per day.
- a pharmaceutical composition is administered once daily, while in other embodiments, a pharmaceutical composition is administered twice daily, and in other embodiments a pharmaceutical composition is administered three times daily. [00275] Administration of the active pharmaceutical ingredients may continue as long as necessary. In selected embodiments, a pharmaceutical composition is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 day(s). In some embodiments, a pharmaceutical composition is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day(s). In some embodiments, a pharmaceutical composition is administered chronically on an ongoing basis - e.g., for the treatment of chronic effects. In some embodiments, the administration of a pharmaceutical composition continues for less than about 7 days.
- an effective dosage of an active pharmaceutical ingredient disclosed herein for example any of the compounds of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof, is in the range of about 1 mg to about 500 mg, about 10 mg to about 300 mg, about 20 mg to about 250 mg, about 25 mg to about 200 mg, about 10 mg to about 200 mg, about 20 mg to about 150 mg, about 30 mg to about 120 mg, about 10 mg to about 90 mg, about 20 mg to about 80 mg, about 30 mg to about 70 mg, about 40 mg to about 60 mg, about 45 mg to about 55 mg, about 48 mg to about 52 mg,
- an effective dosage of an active pharmaceutical ingredient disclosed herein is less than about 25 mg, less than about 50 mg, less than about 75 mg, less than about 100 mg, less than about 125 mg, less than about 150 mg, less than about 175 mg, less than about 200 mg, less than about 225 mg, or less than about 250 mg. In some embodiments, an effective dosage of an active pharmaceutical ingredient disclosed herein is greater than about 25 mg, greater than about 50 mg, greater than about 75 mg, greater than about 100 mg, greater than about 125 mg, greater than about 150 mg, greater than about 175 mg, greater than about 200 mg, greater than about 225 mg, or greater than about 250 mg.
- an effective dosage of an active pharmaceutical ingredient disclosed herein for example any of the compounds of formula (A), formula (I), formula (II), formula (III), a compound of any one of formula 1001-1729, formula 2001-2337, formula 3001-3031, formula 4001-4729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof is in the range of about 0.01 mg/kg to about 200 mg/kg, or about 0.1 to 100 mg/kg, or about 1 to 50 mg/kg.
- an active pharmaceutical ingredient is administered at a dosage of 10 to 200 mg BID, including 50, 60, 70, 80, 90, 100, 150, or 200 mg BID.
- an active pharmaceutical ingredient is administered at a dosage of 10 to 500 mg BID, including 1, 5, 10, 15, 25, 50, 75, 100, 150, 200, 300, 400, or 500 mg BID.
- dosage levels below the lower limit of the aforesaid ranges may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g., by dividing such larger doses into several small doses for administration throughout the day.
- the dosage actually administered will depend upon the condition being treated, the age, health and weight of the recipient, the type of concurrent treatment, if any, and the frequency of treatment.
- the effective dosage amount may be determined by one skilled in the art on the basis of routine empirical activity testing to measure the bioactivity of the compound(s) in a bioassay, and thus establish the appropriate dosage to be administered.
- An effective amount of the combination of the active pharmaceutical ingredient may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.
- compositions described herein further include controlled-release, sustained release, or extended-release therapeutic dosage forms for administration of the compounds described herein, which involves incorporation of the compounds into a suitable delivery system in the formation of certain compositions.
- This dosage form controls release of the compound(s) in such a manner that an effective concentration of the compound(s) in the bloodstream may be maintained over an extended period of time, with the concentration in the blood remaining relatively constant, to improve therapeutic results and/or minimize side effects.
- a controlled-release system would provide minimum peak to trough fluctuations in blood plasma levels of the compound.
- Embodiment 2 The compound of Embodiment 1, wherein the compound of formula (A) is a compound of formula (I), or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof: formula (I) wherein in formula (I): R 1 and R 3 are independently at each occurrence a 5-membered heterocycle, 5- membered heteroaryl, a 6-membered heterocycle, or 6-membered heteroaryl; R 2 and R 4 are independently at each occurrence selected from hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkylheteroaryl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylaryl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally
- Embodiment 3 The compound of Embodiment 1 or Embodiment 2, wherein is R 1 is a 5-membered heteroaryl ring, optionally selected from thiazole, oxazole, imidazole, thiadiazole, oxadiazole, and triazole.
- Embodiment 4 The compound of any one of Embodiments 1 to 3, or pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof, wherein the compound is a compound of formula (II):
- A is S, NR 5 , N, or O, optionally A is S or O;
- B 1 and B 2 are independently at each occurrence selected from N, O, or CR 5 , wherein B 1 is not O when either A or B 2 is O; and each R 5 is independently at each occurrence selected from hydrogen, C 1 -C 6 alkyl (e.g., C 1 -C 3 alkyl), and C 1 -C 3 haloalkyl (e.g., CH 2 F, CHF 2 , CF 3 );
- R 3 is selected from 5-membered heterocycle, 5-membered heteroaryl, 6-membered heterocycle, and 6-membered heteroaryl;
- R 2 and R 4 are independently at each occurrence selected from hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkylheteroaryl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkyn
- Embodiment 5 The compound of Embodiment 4, wherein R 1 is selected from , , and , wherein: A a is O or S, and B a is N or CH; A b is N, and B b is O or S; A c is O or S, and B c is N; and R 5 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), and C 1 -C 3 haloalkyl (e.g., CH 2 F, CHF 2 , or CF 3 ).
- C 1 -C 3 alkyl e.g., methyl, ethyl, propyl, isopropyl
- C 1 -C 3 haloalkyl e.g., CH 2 F, CHF 2 , or CF 3 .
- Embodiment 7 The compound of any one of Embodiments 4-6, wherein R 1 is selected from , , , , , and [00290] Embodiment 8.
- R 2 is selected from hydrogen, C 3 -C 6 cycloalkyl, C 1 -C 6 alkyl (e.g., C 1 -C 4 straight chain alkyl, C 3 -C 6 branched alkyl), -CF 3 , -CH 2 F, -CHF 2 , and -C(R 6 R 7 )pX; wherein, p is an integer from 1-3; R 6 and R 7 are each independently selected from hydrogen, C 1 -C 3 alkyl, and C 3 -C 6 cycloalkyl; X is selected from -OH, -CN, -OR 8 , and NR 9 R 10 ; R 8 is selected from C 1 -C 3 alkyl (e.g., C 1 -C 3 straight or branched alkyl), and C 3 -C 6 cycloalkyl; and R 9 and R 10 are each independently selected from hydrogen, C 1 -C 3 alkyl,
- Embodiment 9 The compound of Embodiment 8, wherein R 2 is selected from hydrogen, C 1 -C 4 alkyl (e.g., methyl, ethyl, propyl, isopropyl, butyl, tertbutyl), -CF 3 , -CH 2 F, and -CHF 2 .
- Embodiment 10 The compound of Embodiment 8 or Embodiment 9, wherein R 2 is selected from H, -CH 3 , -CF 3 , and -CHF 2 .
- Embodiment 11 Embodiment 11.
- R 3 is a 5-membered heteroaryl ring, optionally selected from pyrrole, thiophene, furan, pyrazole, thiazole, isothiazole, oxazole, isoxazole, and imidazole.
- Embodiment 11 wherein R 3 is selected from , , , , , , , and , wherein R 11 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), C 3 -C 6 cycloalkyl, CF 3 , CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F.
- C 1 -C 3 alkyl e.g., methyl, ethyl, propyl, isopropyl
- C 3 -C 6 cycloalkyl CF 3 , CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F.
- Embodiment 12 The compound of Embodiment 12 or Embodiment 13, wherein R 11 is selected from hydrogen, methyl, ethyl, propyl, isopropyl, -CF 3 , and -CH 2 CF 3 .
- Embodiment 15 The compound of any one of Embodiments 11-14, wherein R 3 is selected from and [00298] Embodiment 16.
- R 4 is selected from hydrogen, C 1 -C 6 alkyl, and halo (e.g., F or Cl).
- Embodiment 19 The compound of Embodiment 18, wherein n is 0 or 1.
- Embodiment 20 The compound of any one of Embodiments 1-19, wherein R 4 is selected from hydrogen, 4-fluoro, 3-fluoro, 2-fluoro, 2-chloro, and 4-chloro.
- Embodiment 21 The compound of any one of Embodiments 1-20, wherein R 4 is selected from hydrogen, 4-fluoro, 2-fluoro, and 2-chloro.
- Embodiment 22 Embodiment 22.
- Embodiment 23 The compound of any one of Embodiments 1-4, wherein the compound of formula (A), formula (I), or formula (II) is a compound having any one of formula 1001-1080, 1097-1176, 1193-1272, 1289-1368, or 1385-1729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- Embodiment 24 is a compound having any one of formula 1001-1080, 1097-1176, 1193-1272, 1289-1368, or 1385-1729, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof.
- Embodiment 1 is ; and R 20 is selected from H, optionally substituted C 1 -C 6 alkyl, and optionally substituted C 1 -C 6 haloalkyl; R 21 is selected from optionally substituted C 1 -C 6 alkyl and optionally substituted C 1 -C 6 haloalkyl; or R 20 and R 21 are joined to form a 4-6-membered heterocycle with the nitrogen to which they are each bound.
- Embodiment 27 is ; and R 20 is selected from H, optionally substituted C 1 -C 6 alkyl, and optionally substituted C 1 -C 6 haloalkyl; or R 21 are joined to form a 4-6-membered heterocycle with the nitrogen to which they are each bound.
- R 20 is selected from H, methyl, ethyl, propyl, isopropyl, butyl, tert-butyl, CF 3 , CHF 2 , CH 2 F, CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F; and R 21 is selected from methyl, ethyl, propyl, isopropyl, butyl, tert-butyl, CF 3 , CHF 2 , CH 2 F, CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F; or R 20 and R 21 are joined to form a 4-6-membered heterocycle with the nitrogen to which they are each bound.
- Embodiment 28 The compound of Embodiment 1, 26, or Embodiment 27, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof, wherein R 20 is selected from H, methyl, ethyl, propyl, tert-butyl, and CH 2 CH 2 F; and R 21 is selected from methyl, ethyl, propyl, tert-butyl, and CH 2 CH 2 F; or R 20 and R 21 are joined to form a 4-6-membered heterocycle with the nitrogen to which they are each bound.
- Embodiment 29 Embodiment 29.
- Embodiment 30 The compound of any one of Embodiments 1 or 26-29, wherein R 2 is selected from hydrogen, C 3 -C 6 cycloalkyl, C 1 -C 6 alkyl (e.g., C 1 -C 4 straight chain alkyl, C 3 -C 6 branched alkyl), -CF 3 , -CH 2 F, -CHF 2 , and -C(R 6 R 7 )pX; wherein, p is an integer from 1-3; R 6 and R 7 are each independently selected from hydrogen, C 1 -C 3 alkyl, and C 3 -C 6 cycloalkyl; X is selected from -OH, -CN, -OR 8 , and NR 9 R 10 ; R 8 is selected from C 1 -C 3 alkyl (e.g., C 1 -C 3 straight or branched alkyl), and C 3 -C 6 cycloalkyl; and R 9 and R 10 are each independently
- Embodiment 31 The compound of Embodiment 30, wherein R 2 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), -CF 3 , -CH 2 F, and -CHF 2 .
- Embodiment 32 The compound of Embodiment 30 or Embodiment 31, wherein R 2 is selected from hydrogen and -CH 3 .
- Embodiment 33 Embodiment 33.
- Embodiment 34 The compound of any one of Embodiments 1 or 26-32, wherein is R 3 is a 5-membered heteroaryl ring, optionally selected from pyrrole, thiophene, furan, pyrazole, thiazole, isothiazole, oxazole, isoxazole, and imidazole. [00316] Embodiment 34.
- Embodiment 33 wherein R 3 is selected from , and , wherein R 11 is selected from hydrogen, C 1 -C 3 alkyl (e.g., methyl, ethyl, propyl, isopropyl), C 3 -C 6 cycloalkyl, CF 3 , CH 2 CF 3 , CH 2 CHF 2 , and CH 2 CH 2 F.
- R 3 is selected from , and [00318] Embodiment 36.
- Embodiment 34 or Embodiment 35 wherein R 11 is selected from hydrogen, methyl, ethyl, propyl, isopropyl, -CF 3 , and -CH 2 CF 3 .
- Embodiment 37 The compound of any one of Embodiments 1 or 26-36, wherein R 3 is selected from and [00320] Embodiment 38.
- R 4 is selected from hydrogen, C 1 -C 6 alkyl, and halo (e.g., F or Cl).
- Embodiment 39 The compound of any one of Embodiments 1 or 26-38, wherein n is 0 or 1.
- Embodiment 40 The compound of any one of Embodiments 1 or 26-39, wherein R 4 is selected from hydrogen, 4-fluoro, 3-fluoro, 2-fluoro, 2-chloro, and 4-chloro.
- Embodiment 41 The compound of any one of Embodiments 1 or 26-40, wherein R 4 is selected from hydrogen, 4-fluoro, 3-fluoro, 2-fluoro, and 4-chloro.
- Embodiment 43 The compound of any one of Embodiments 1 to 42, wherein the compound is a PDE11A4 inhibitor.
- Embodiment 44 The compound of Embodiment 43, wherein the PDE11A4 inhibitor is a PDE11A4 selective inhibitor.
- Embodiment 45 The compound of Embodiment 45, wherein the PDE11A4 inhibitor is a PDE11A4 selective inhibitor.
- a pharmaceutical composition comprising a compound of any one of Embodiments 1 to 43, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof, and a physiologically compatible carrier medium.
- Embodiment 46 A pharmaceutical composition comprising a compound of any one of Embodiments 1 to 43, or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof, and a physiologically compatible carrier medium, wherein the amount of the compound in the composition is a therapeutically effective amount for the treatment or prevention of a disease or disorder alleviated by inhibiting PDE11A4 activity in a patient in need thereof.
- Embodiment 47 Embodiment 47.
- Embodiment 45 or Embodiment 46 wherein the disease or disorder is associated with cognitive decline.
- Embodiment 48 The pharmaceutical composition of Embodiment 47, wherein the disease or disorder is selected from dementia, Alzheimer’s Disease (AD) including mild Alzheimer's disease and early-onset Alzheimer’s disease, Down’s syndrome, vascular dementia (cerebral amyloid angiopathy and stroke), dementia with Lewy bodies, HIV dementia, Mild Cognitive Impairment (MCI); Age-Associated Memory Impairment (AAMI); Age-Related Cognitive Decline (ARCD) (including age-related cognitive decline of associative long-term memories (aLTMs), dementia associated with traumatic brain injury, preclinical Alzheimer's Disease (PCAD); Cognitive Impairment No Dementia (CIND), and cognitive decline associate with spatial memory, other depression-related behaviors, additional anxiety-related behaviors, sensorimotor gating, and social behaviors.
- AD Alzheimer’s Disease
- MCI Mild Cognitive Impairment
- AAMI Age-Associated Memory Impairment
- Embodiment 49 A method of treating or preventing a disease or disorder alleviated by inhibiting PDE11A4 activity in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of any one of Embodiments 1 to 43.
- Embodiment 50 A method of treating or preventing a disease or disorder alleviated by inhibiting PDE11A4 activity in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition of any one of Embodiments 45-48.
- Embodiment 51 The method of any one of Embodiments 49 or 50, wherein the isolated fragment is administered in a dosage unit form.
- Embodiment 52 The method of Embodiment 51, wherein the dosage unit comprises a physiologically compatible carrier medium.
- Embodiment 53 The method of any one of Embodiments 49-52, wherein the disease or disorder is associated with cognitive decline.
- Embodiment 54 The method of any one of Embodiments 49-52, wherein the disease or disorder is associated with cognitive decline.
- the disease or disorder is selected from dementia, Alzheimer’s Disease (AD) including mild Alzheimer's disease and early-onset Alzheimer’s disease, Down’s syndrome, vascular dementia (cerebral amyloid angiopathy and stroke), dementia with Lewy bodies, HIV dementia, Mild Cognitive Impairment (MCI); Age-Associated Memory Impairment (AAMI); Age-Related Cognitive Decline (ARCD) (including age-related cognitive decline of associative long-term memories (aLTMs), dementia associated with traumatic brain injury, preclinical Alzheimer's Disease (PCAD); Cognitive Impairment No Dementia (CIND), and cognitive decline associate with spatial memory, other depression-related behaviors, additional anxiety-related behaviors, sensorimotor gating, and social behaviors.
- Example 1 Phosphodiesterase 11A (PDE11A) and its Role in the Neurobiological Substrates of Memory and Social Behaviors
- PDE11A Phosphodiesterase 11A
- This Example describes data testing the hypothesis that age-related increases in HIPP PDE11A4 occur in a compartmentalized manner and impair social aLTMs.
- a novel conditional transgenic system that controls the expression of PDE11A4 in a time- and brain region-specific manner by combining overexpression lentiviruses with PDE11A WT and KO mice is used.
- RNAscope probes are used to delineate nuclear vs. cytosolic localization of PDE11A4 mRNA.
- In vivo/ex vivo techniques are used to study of PDE compartmentalization—and its functional consequences— rather than the study of artificial FRET-based constructs in in vitro assays.
- the studies Given PDE11A4’s uniquely restricted expression pattern, the studies not only uncover the neurobiological role of a particular enzyme, they also define the function of an anatomically restricted, molecularly defined-circuit that appears to be uniquely specialized for processing social information. The focus on LTMs for social experiences is relatively unique in the field of learning memory.
- STM short-term memory
- Age-related cognitive decline is not a uniform process, with variability in symptom severity observed across cognitive domains. Human studies have demonstrated that associative long-term memories (aLTMs)—particularly those involving experiences with family and friends— are more susceptible to age-related cognitive decline than are recognition long-term memories (rLTMs). This differential sensitivity of social aLTMs vs rLTMs in mice (Fig.3) was recapitulated.
- preventing or minimizing age-related increases in PDE11A4 is sufficient to prevent the onset of age-related cognitive decline of social aLTMs (Figs.4A-4B). Further, the age-related cognitive decline observed in PDE11A WTs is due to the excessive PDE11A4 that is acutely present in the aged adult hippocampus. Identifying when and where PDE11A4 affects age-related cognitive decline allowed for the discovery of more sophisticated therapeutic approaches to target PDE11A4 for the reversal of aLTM deficits in aged adults or only as a prophylactic agent that prevents the onset of deficits.
- “young” is defined as 2-6 months of age and “Old” is defined as 18-22 months of age since 1) protective effects are observed in PDE11A KO mice as early as 14 months old and 2) it is expected 50% of the colony dies by the age of 24 months (i.e., do not want to introduce a selection bias by preferentially studying older mice with exceptional longevity).
- PDE11A WT or KO mice are administered a lentivirus containing a control fluorescent protein (EmGFP, as appropriate) or an N-terminal EmGFP-tagged PDE11A4 construct.
- EmGFP control fluorescent protein
- Coordinates were selected to target CA1 and subiculum—the portions of the hippocampus that naturally express PDE11A4 (DHIPP coordinates: AP -1.7 mm, ML +/- 1.6 mm, DV -1.4 mm; VHIPP coordinates: AP -3.3 mm, ML +/- 3.5 mm, DV -4.4 mm).
- DHIPP coordinates AP -1.7 mm, ML +/- 1.6 mm, DV -1.4 mm
- VHIPP coordinates AP -3.3 mm, ML +/- 3.5 mm, DV -4.4 mm.
- the ability to virally manipulate PDE11A4 expression has been measured and function in vivo and to measure aLTM and rLTM in virally-treated mice (Figs.5A-5E).
- transient amnesia is not required to see the protective effects of PDE11A deletion on remote aLTMs in aged mice.
- young vs. old WT, HT, and KO PDE11A mice are compared at all 3 time points (STM, recent LTM, and remote LTM) to determine the effects of age-related increases in PDE11A4 at each memory phase.
- STM recent LTM
- remote LTM remote LTM
- the circuit was identified (including nodes of activity and functional connectivity amongst nodes) that is engaged by aged PDE11A KO mice during the successful retrieval of a social aLTM. It is then determined if the circuit engaged by an aged PDE11A KO shares similarities with or diverges from that of young PDE11A WT mice. This approach shows whether preventing age-related increases in PDE11A prevents/delays the onset of aging pathophysiology or if it simply enables the brain to adopt a compensatory strategy for achieving equivalent behavioral performance (i.e., engages cognitive reserve).
- the aged brain may mount an immune response to the lentiviral constructs that is not typically seen in young adult mice.
- the construct in the case of the former (as per elevated IBA-1, GFAP, or IL-6 staining), the construct is moved to an adeno-associated virus.
- a conditional transgenic is developed and backcrossed onto the PDE11A line to allow reversible overexpression of PDE11A4 in WTs and KOs using doxycycline .
- a conditional knockout approach is pursued using an inducible FLEX-Cre recombinase system. Identify the circuit, cell-type and subcellular domain where PDE11A4 is upregulated with age and the signal driving this compartmentalized effect. [00350] Intramolecular signals have been identified that alter the trafficking of PDE11A4, including homodimerization and N-terminal phosphorylation (Figs.7A-7G).
- disrupting PDE11A4 homodimerization reduces PDE11A4 accumulation and shifts PDE11A4 from the membrane back to the cytosol.
- disrupting PDE11A4 homodimerization also reduces PDE11A4 cGMP hydrolytic (Fig 7G), consistent with the fact that it reduces pS117/pS124-PDE11A4 (Fig 7D).
- mice To establish the conservation of effects across species, these experiments utilize brains from young vs old 1) C57BL/6 and BALB/cBy mice, 2) Fischer 344 and Brown Norway rats, and 3) rhesus monkeys (all obtained from the NIA Rodent Colonies or Primate Tissue Bank). Mice and rats are available as live animals and, thus, are tested for remote LTM of STFP, SOR, and NSOR to confirm a selective age-related cognitive decline of social aLTMs. Monkey tissue is available only from post mortem stock and, thus, subjects are not cognitively phenotyped.
- the first hemisphere (rodents) or block of tissue (primate) are kept intact for processing by in situ hybridization and IF in order to determine in which hippocampal subfields age-related increases in PDE11A4 mRNA, protein expression, and phosphorylation occur (e.g., CA1 vs. subiculum or stratum radiatum vs. stratum pyramidale).
- hippocampal subfields age-related increases in PDE11A4 mRNA, protein expression, and phosphorylation occur e.g., CA1 vs. subiculum or stratum radiatum vs. stratum pyramidale.
- changes in the subcellular location of a PDE would impact function—but so would changes at the level of circuit or cell type. For example, changes in subiculum would indicate an effect on retrieval mechanisms; whereas, changes in CA1 would indicate an effect on input integration.
- changes in CA1 dendrites of stratum radiatum proximal to the cell body indicates modulation of CA3 input signals
- changes in distal CA1 dendrites indicates modulation of entorhinal cortex input signals.
- the age-related increases in PDE11A4 expression most strikingly occur in the filamentous structures; however, the preliminary study also showed that a subset of sporadically distributed neurons in the VHIPP stratum pyramidale exhibit increased PDE11A4 expression around the cell body. The sporadic nature of these cell bodies raises the possibility that these neurons reflect either a specific subtype of inhibitory interneuron or neurons that send projections to a discrete brain region.
- PDE11A4 continues to only be expressed in excitatory neurons during aging, then retrograde tracer studies using stereotaxically-delivered fluorogold are conducted to determine if this neuronal subpopulation segregates based on their projections.
- anterior cingulate cortex, entorhinal cortex, and retrosplenial cortex as these brain regions show heightened activation in the PDE11A KO during retrieval of enhanced remote social aLTM.
- Nucleus accumbens are also of interest given that a subset of PDE11A4-expressing neurons project to NAcc and ventral CA1-NAcc projections are required for social aLTM.
- NAcc Nucleus accumbens
- the lentiviruses express for at least 3 months, allowing for chronic manipulations.
- the GAB-B lentivirus have been obtained and it has been confirmed that it expresses in vivo.
- the effects of virally overexpressing S117A/S124A vs PDE11A4 WT are compared in HIPP of old PDE11A KO mice. In so doing, it is determined if preventing phosphorylation of S117/S124 is sufficient to block the accumulation of PDE11A4 that is seen with high levels of endogenous PDE11A4 expression (Fig.8) or viral overexpression of PDE11A4 WT .
- tissue is labeled from EmGFP-PDE11A4 infected PDE11A KOs since overexpressed EmGFP- PDE11A4 also accumulates in filamentous structures in KOs and has been previously validated a GFP antibody in EM using EmGFP-PDE11A4 transfected COS-1 cells.
- Westerns blots have been conducted and in situ hybridization in human tissue, so it is not anticipated unsurmountable difficulties in adapting the techniques for monkey tissue.
- the NIA Primate Tissue bank tracks only age and general health of the monkeys, not their cognitive abilities.
- PDE11A mRNA expression is also increased in the aged rat brain and more recently discovered that PDE11A mRNA increases with age in the human hippocampus (Fig.2B).
- age-related increases in PDE11A4 protein expression appear to be driven, at least in part, by increases in PDE11A4 transcription and/or transcript stability.
- transcription of a given gene is controlled by a core promoter, promoter-proximal elements, as well as enhancers or silencers.
- the core promoter falls within 30 base pairs (bp) of the transcription initiation site (TIS) and the promoter- proximal elements fall within 200 bp of the TIS, enhancers and silencers can fall anywhere within 50 kB of the TIS.
- TIS transcription initiation site
- enhancers and silencers can fall anywhere within 50 kB of the TIS.
- a number of studies have shown that a gene promoter is sufficient to drive transcription of a gene in a tissue-specific manner. Indeed transgenic technology makes great use of this fact by ligating the promoter of a tissue-specific gene to the coding sequence of a transgene in order to spatially restrict expression of that transgene to a desired tissue.
- Putative promoters have been described for hPDE11A4, hPDE11A3, and hPDE11A1 in the 1200 bps upstream of their respective TISs.
- a lentivirus construct that uses the 1200 bps upstream of the mPde11a4 TIS to drive expression of the mCherry reporter (mPde11a4-mCherry) was developed.
- the decision to take a lentiviral approach is based on a prototype study by Chhatwal and colleagues.
- in situ hybridization and Western blots are conducted on young vs. old HIPP tissue collected as described above to determine if age-related decreases in p54 nrb /NONO and XRN2 are observed in rodents and rhesus monkeys as they are in humans (Fig.11). Immunoprecipitation are also conducted using total homogenates using methods described above. Antibodies are used against p54 nrb /NONO and XRN2 to perform pull downs and then RT-PCR is conducted for PDE11A4 mRNA. It is determined if old animals show less binding of p54 nrb /NONO and XRN2 to the PDE11A4 transcript than do young animals.
- nuclear and cytoplasmic RNAs are isolated from young and aged rodents using the Ambion Paris system according to manufacturer’s instructions (Life Technologies) and, again, RT-PCR for PDE11A4 mRNA are conducted. This allows for determination if aging is associated with heightened accumulation of PDE11A4 mRNA in the nucleus vs. the cytosol, which indicates impaired degradation by the p54 nrb /NONO-XRN2 complex.
- brain sections collected as described above are used to conduct confirmatory in situ hybridization experiments using RNAscope probes (ACD).
- RNAscope probes enable single molecule-level resolution (see Fig.12A) and when combined with confocal imaging (Fig 12B), enable the qualitative assessment of PDE11A mRNA expression in the nucleus vs. the cytosol. Results [00367] It is expected to find increased mCherry expression in old vs young rodents only when they are injected with the mPde11a4-mCherry into hippocampal subfields that show increased PDE11A4 mRNA expression.
- the constitutive Pgk- mCherry reporter also shows increased expression in the aged hippocampus, then this result suggests a more global upregulation of transcriptional activity, or a potential indirect effect of the mCherry itself, as opposed to a selective upregulation of PDE11A4 transcriptional activity.
- the finding that aging is associated with increased PDE11A4 transcription or transcript stability does not rule out the possibility that aging is also associated with increased rates of translation or protein stability, and the possibility of age-related reductions in sumoylation or ubiquitination of PDE11A4 is further examined.
- the pilot data suggests the 1200 bps upstream of the PDE11A4 TIS is sufficient to control the transcription of mPde11a4 in terms of its spatial distribution, but it is possible that aging also/alternatively could influence rates of transcription by differentially engaging enhancer or silencing motifs within 50 kB of the TIS.
- a BAC transgenic approach is adopted as previously described by Koppel and colleagues. BAC transgenic mice are generated that express a fluorescent protein under the control of a minimal promoter coupled with various combinations of PDE11A4 exons, introns, 5’ upstream sequences, and 3’ downstream sequences.
- exoribonucleases are not the only mechanism regulating transcript stability; microRNAs, along with other noncoding RNAs, play an important role as well.
- the focus is to identify non-coding RNAs that regulate PDE11A4 expression.
- miR-375 is one of only 4 microRNAs that is predicted by TargetScan to target the PDE11A4 transcript.
- miR-375 expression decreases with age in the mouse brain, which is consistent with the observed age- related increases in PDE11A4.
- data are analyzed by multifactorial ANOVAs or by repeated measure ANOVAs where appropriate to account for multiple comparisons.
- in vivo data are analyzed for effects of sex, genotype, lentiviral treatment (in addition to assay-specific factors, such as food type).
- Statistical outliers are dropped from analyses, as previously described.
- Significant ANOVAs are followed by Student Newman–Keuls post hoc tests, with significance determined as P ⁇ 0.05.
- Data in figures are plotted as means ⁇ SEMs. Data rigor The rationale is based on both mouse and human data with follow-up with studies in mice, rats and monkeys, which strengthens the rigor of this proposal.
- mice are genotyped a priori to enable proper counterbalancing of experimental run lists; however, experimenters are blind to genotype at the time of data collection. Genotypes are then reconfirmed post death by Western blot or in situ hybridization. Physical parameters are counterbalanced across subjects (e.g., which is the “trained” spice and which is “novel”). Biological Variables: both males and females were tested for effect of sex. [00371] Since cohorts of mice are aged up, these experiments largely conducted in parallel across years 1-5. One exception is that some experiments slightly lag behind as species selection depends on the outcome of in situ hybridization studies described herein.
- PDE11A4 is problematic simply by virtue of increased steady state levels that are ubiquitously distributed or by virtue of a discretely localized upregulation or even an ectopic expression of PDE11A4.
- the effect of modulating PDE11A4 function within specific cell-types (e.g., excitatory vs. inhibitory) or sub-region (e.g., subiculum vs. CA1) is examined.
- the upstream signaling events are delineated, including the specific kinases that lead to age-related increases in the phosphorylation of PDE11A4.
- PDE11A is a highly druggable enzyme and it is positioned to selectively control cyclic nucleotide signaling in a molecularly-defined circuit that specifically regulates social LTMs, without affecting signaling elsewhere. This may relieve age-related impairments in aLTM without causing unwanted side effects.
- PDE11A exhibits all properties the pharmaceutical industry believes an ‘ideal’ drug target should have. The fact that PDE11A is a realistic candidate for drug development increases the value of understanding its biological function.
- Example 2 Role of Cyclic Nucleotide Signaling in Age-related Decline of Social Memories [00373] This Example describes studies where it is hypothesized that age-related increases in HIPP PDE11A4 occur in a compartmentalized manner and impair social aLTM. [00374] Determine if blocking age-related increases in PDE11A4 can prevent and/or rescue age-related impairments in social aLTM. It is not known if age-related increases in hippocampal PDE11A4 expression reflect a physiological breakdown or an attempt at protective compensation. In studies, PDE11A WT mice show age-related impairments in social transmission of food preference (STFP) aLTM but PDE11A KO mice do not.
- STFP food preference
- PDE11A represents a therapeutic target not only for preventing age-related decline of social aLTMs but also for reversing deficits.
- PDE11A4 protein expression increases with age in HIPP lysates. It is possible that this excess PDE11A4 may not be globally distributed, but rather discretely localized or even ectopically expressed in cell types or subcellular domains that are normally void of PDE11A4.
- biochemical fractionation shows PDE11A4 increases with age preferentially in membrane vs cytosol and nucleus.
- Immunofluorescence shows PDE11A4 ectopically accumulates in filamentous structures in the aged HIPP, which are rarely seen in young HIPP. Further, this pool of accumulated PDE11A4 is uniquely phosphorylated at serines 117 and 124. In vitro, phosphomimic mutation of S117 and S124 drives accumulation of PDE11A4, while phosphoresistant mutations or blocking homodimerization reduces this accumulation.
- the novel PDE11A4-promoter reporter is used to measure transcription in HIPP of aged vs. young mice.
- rates of PDE11A4 mRNA degradation are assessed by its known exoribonuclease (XRN2).
- XRN2 exoribonuclease
- Example 3 Biologic That Disrupts PDE11A4 Homodimerization in Hippocampus CA1 Reverses Age-Related Proteinopathies in PDE11A4 and Age-Related Decline of Social Memories [00379]
- PDE11A phosphodiesterase 11A
- VHIPP ventral hippocampal formation
- ARCD drive age-related cognitive decline
- age-related increases in PDE11A4 occur specifically within the membrane compartment and ectopically accumulate in filamentous structures termed ghost axons.
- the Pde11a mouse line obtained from Deltagen (San Mateo, CA) was maintained on a mixed C57BL6 background (99.8% multiple C57BL/6 substrains, 0.2% 129P2/OlaHsd).
- Pde11a mice were bred at the University of South Carolina in heterozygous (HT) x HT trio-matings. Same-sex wild-type (WT), heterozygous (HT), and knockout (KO) littermates were weaned and caged together to total 3-5 mice/cage. It is not believed litter effects are driving findings here due to each dataset reflecting multiple cohorts born and tested at different times. Further, a litter of mice normally contributes only 1-2 mice/genotype and parents contribute two litters at most to a cohort.
- mice While both males and females were used in experiments, analyze for sex effects (see figure legends for specific n’s/sex/group/experiment). In these studies, young mice were defined as 2-6 months and old mice were defined as 18-22 months. “Young” mice included both young Pde11a WT mice surgerized alongside old Pde11a WT mice (i.e., receiving bilateral injections of mCherry lentivirus to the dorsal and ventral hippocampi) and unsurgerized young C57BL6/J mice that were used as an internal control for the assays (Figs. 14A-14B).
- mice Since no obvious differences were found between groups of young surgerized Pde11a WT mice and young unsurgerized C57BL6/J mice, the data from these 2 subgroups were subsequently combined into a singular “young” (Figs.14C-14G). All mice used in experiments were generally healthy throughout the duration of testing. Gross pathology was not conducted but mice were routinely assessed by husbandry, veterinary, and laboratory staff. Mice with lethargy, altered gait, signs of malnutrition or dehydration, noticeable tumors >1 cm, were removed from study and euthanized.
- Tissue Collection Mice were euthanized (during light cycle) via rapid cervical dislocation and brains were immediately collected and flash frozen on 2-methylbutane sitting on dry ice. Brain tissue was then stored at -80 °C until cryosectioning at 20 ⁇ m to verify viral expression.
- Subjects access to food was restricted the two days prior to testing to one hour per day. The day prior to testing, all mice were placed in a clean home cage and given access to plain powdered chow packed into a glass jar. The following day, the designated “demonstrator mouse” was individually placed in a clean home cage and fed powdered chow flavored with a household spice (e.g. basil vs.
- a household spice e.g. basil vs.
- One flavored chow contained a novel spice and the other contained the spice that the demonstrator was given.
- the amount of food eaten was measured by an experimenter blind to treatment. All mice had to meet minimum inclusion criteria including eating at least 0.25 grams of food. Cohorts were able to be trained/tested at multiple time points using different spice combination to reduce the total number of mice used and we have shown that this does not confound interpretation of the data. Observer mice eating more food containing the familiar spice (i.e., the spice on their demonstrator’s breath) versus the novel spiced food constituted memory (preference ratio: familiar- novel/familiar+novel). [00385] Odor Recognition.
- Subjects were allowed to habituate to 1” round wooden beads (Woodworks) for at least seven days prior to testing by placing several beads in the subjects’ home cages.
- social odor recognition SOR
- the wooden beads take up the scent of the cage of each strain used (e.g., C57BL/6J Jax #000664, BALB/cJ Jax #000651, 129S6/ SvEv Taconic #129SVE) and are used as the social odor in testing.
- NOR non-social odor recognition
- the wooden beads were placed in a bag containing bedding saturated with a household spice (e.g., marjoram, cumin, etc.) for at least 7 days.
- Training for SOR and NSOR consisted of a habituation trial with 3 beads from the subject’s home cage, followed by two training trials that included 2 home-cage beads and 1 novel-scented bead. Recent and remote long-term memory were assessed 24 hours or 7 days after training, respectively.
- mice were tested with one home cage bead, one bead from the trained donor strain (familiar), and one bead from a second donor strain (novel).
- mice were tested with only two beads, one scented with the training spice and one a novel spice. The designation of which scent was “novel” within a given testing trial and the location of the novel scent (i.e., left versus right) was counterbalanced across subjects.
- mice were given two minutes to investigate the beads and the amount of time spent on each was manually scored by an experimenter blind to treatment and bead. It was previously determined that infusion of even a negative control lentivirus into the hippocampus reverses the recent long-term memory impairment observed in Pde11a KO mice 24 hours after training. Therefore, 24-hour memory following injection of the isolated GAF-B domain was not tested as the results would not be interpretable. All mice met minimum inclusion criteria of spending a minimum of 3 seconds in total sniffing the beads. Spending more time investigating the novel vs familiar scent constituted memory (preference ratio: novel-familiar/novel+familiar). [00386] Stereotaxic Surgeries.
- Stereotaxic surgeries and injections were performed using a NeuroStar motorized stereotaxic, drill, and injection robot (Tubingen, Germany). Mice were anesthetized with a steady flow of oxygen and isoflurane. The mice were induced at 3% isoflurane and maintained at 1-1.5%. Lack of reflexes was verified and the scalp was then shaved and cleaned with betadine. A small incision was made in the scalp and the skull was cleared with sterile saline. Cotton swabs were again used to visualize the skull and locate Bregma.
- a Hamilton syringe custom needle #7804-04: 26s gauge, 1” length, 25 degree bevel was then then placed to the following depths relative to Bregma: dCA1 DV, - 1.3, vCA1 DV, -4.4.
- the injection robot was used to inject 2 ⁇ l of lentivirus at 0.167 ⁇ l/minute. Following injection completion, the experimenter waited two minutes to allow the lentivirus to diffuse away from the needle and the needle was raised at the same speed. After all injections were complete, pronged tweezers were used to close the scalp and secured using glutures. Buprenorphine in sterile saline at a dose of 0.1 mg/kg was injected IP for pain management. For recovery, the mouse was placed on a warm Deltaphase pad and allowed to recover until moving normally and posturing upright. Mice were allowed at least 2 weeks of recovery in grouped home cages prior to behavioral testing.
- a lentivirus carrying an mCherry-tagged PDE11A4-GAFB served to disrupt PDE11A4 homodimerization, while an mCherry-only virus was used as a negative control.
- a lentiviral construct was used here in order to compare to previous studies examining the effects of overexpressing PDE11A4 in vivo, which required the use of a lentiviral cassette due to the large size of PDE11A4.
- the viruses were made on an “SPW” backbone that drives expression using the phosphoglycerate kinase 1 (PGK) promoter, which in theory is a ubiquitous promoter and yet is taken up preferentially by neurons.
- PGK phosphoglycerate kinase 1
- the isolated GAF-B construct disrupts PDE11A4 homodimerization by binding to PDE11A4 and triggering proteolytic degradation. For reasons that are not well understood, the GAF-B construct degrades PDE11A4 more significantly in the membrane versus cytosolic fractions.
- the lentiviruses were prepared and diluted in 0.2 M sucrose/42 mM NaCl/0.84 mM KCl/2.5 mM Na2HPO4/0.46 mM KH2PO4/0.35 mM EDTA and the original titers were as follows: mCherry, 7.37X10E10/mL; GAF-B, 1.82X10E10/ml.
- the slides were fixed in 4% paraformaldehyde in phosphate-buffered saline (PBS) for 20 minutes. After fixation, 3x10 minute washes with PBS and 3x10 minute washes with PBT (phosphate- buffered saline/0.4% BSA/0.3% Triton- X 100) were performed to reduce background.
- PBS phosphate-buffered saline
- PBT phosphate- buffered saline/0.4% BSA/0.3% Triton- X 100
- PDE11A4 Aves custom PDE11A4 #1 at 1:10,000; Fabgennix PPD11A-140AP at 1:1000; Fabgennix PPD11A-150AP at 1:500
- mCherry ThermoFisher #PA534974 at 1:1000; Invitrogen #M11217 at 1:500; PhosphoSolutions #1203-mCherry at 1:10,000.
- Multiple PDE11A antibodies were utilized to discern diffuse expression versus accumulations of PDE11A4.
- the PDE11A4#1 antibody labels all PDE11A4 and, thus, detects both diffusely localized and accumulated PDE11A
- the PDE11A4-140 and 150 antibodies specifically labels PDE11A4 phosphorylated at serines 117 and/or 124 and, thus, only detects PDE11A4 accumulated in ghost axons.
- mCherry antibodies hosted in a rodent species were pretreated with anti-Mouse FabFragments (0.15mg/ml; Jackson Immunoresearch # 715-007-003) in PBS for 2 hours, followed by 3x10 minute washes in PBT prior to adding primary antibody.
- COS-1 male line
- HEK293T female line
- HT-22 sex undefined cell culture and transfection were performed as previously described [1]. While kept in t-75 flasks, cells were grown in Dulbecco’s Modified Eagle Medium (DMEM) (GIBCO; Gaithersburg, MD, USA), 10% fetal bovine serum (FBS) (GE Healthcare Life Sciences; Logan, UT, USA), and 1% Penicillin/Streptomycin (P/S) (Mediatech, a Corning subsidary; Manassas, VA, USA).
- DMEM Modified Eagle Medium
- FBS fetal bovine serum
- P/S Penicillin/Streptomycin
- blot was normalized to a control condition (e.g., WT) to account for any technical variables (film exposure, antibody signal-noise, variance in chemiluminescence, etc.).
- a control condition e.g., WT
- PDE assay cAMP- and cGMP-PDE catalytic activity were measured. The assay was validated in vitro using HT-22 cells (a mouse hippocampal cell line). Buffer containing 20 mM Tris-HCl and 10 mM MgCl2 was used to harvest cells and kept on ice until ready to use.
- PDE activity was measured using 50 ⁇ l of sample and 50 uL of [3H] cAMP (Perkin Elmer, NET275) or cGMP (Perkin Elmer, NET337) and incubated for 10 minutes. After incubation, 0.1M HCl was added to quench the reaction, followed by 0.1M Tris to neutralize the reaction.3.75 mg/mL snake venom (Crotalus atrox, Sigma V-7000) was then added to complete the reaction and the mixture was incubated at 37 °C for 10 minutes. Samples were put into 5’polystyrene chromatography columns with coarse filters (Evergreen, 208-3383- 060) containing DEAE Sephadex A-25 resin (VWR, 95055-928).
- lentiviruses were used (Fig.13A) containing either mCherry alone (i.e., negative control) or an mCherry-tagged isolated GAF-B domain that disrupts PDE11A4 homodimerization (Fig.13B).
- mice demonstrated mCherry signal in dorsal and ventral CA1, with a subset of mice exhibiting expression in neighboring hippocampal sub-regions (e.g., dentate gyrus, CA3, CA2) that do not express PDE11A4 (Fig. 13D).
- mCherry signal in dorsal and ventral CA1
- neighboring hippocampal sub-regions e.g., dentate gyrus, CA3, CA2
- disrupting PDE11A4 homodimerization did significantly improve remote LTM for SOR memory (Fig.14G; Table 1), despite having no effect on SOR learning (Fig.14F) or total time sniffing (Table 2).
- these data suggest that disrupting PDE11A4 homodimerization is sufficient to reverse ARCD of remote social memory.
- PDE11A4 homodimerization is an independent intramolecular mechanism that regulates PDE11A4 trafficking and functioning. As noted above, disrupting PDE11A4 homodimerization significantly changes the subcellular compartmentalization of the enzyme.
- disrupting homodimerization not only reduces the accumulation of WT PDE11A4, it also reduces back to WT levels the potentiated accumulation caused by the aging-related phosphomimic mutant S117D/S124D .
- disruption of PDE11A4 homodimerization alters cyclic nucleotide signaling not by altering PDE11A4 catalytic activity directly, but rather by changing the subcellular localization of PDE11A4 protein (e.g., shifting from a cGMP-rich pool to a cAMP-rich pool).
- the isolated GAF-B domain could reduce accumulation not only of WT PDE11A4 but also PDE11A4-S117D/S124D, which mimics the age-related increase in PDE11A4-pS117/pS124 that drives the punctate accumulation of PDE11A4 in the aged brain.
- the isolated GAF-B domain reduced the punctate accumulation of both the WT and PDE11A4- S117D/S124D compared to the mCherry control (Fig.15E-15F).
- PDE11A4 homodimerization does not require phosphorylation of S162. As described above, disrupting PDE11A4 homodimerization reduces the accumulation of PDE11A4 in punctate structures both in vivo (Fig.13F) and in vitro (Figs.15E-15F).
- S162D changed the subcellular compartmentalization of PDE11A4 in a manner similar to that observed with the isolated GAF-B domain —namely, shifting PDE11A4 from the membrane to the cytosolic fraction (Fig.15G).
- the isolated GAF-B domain was able to effectively reduce the accumulation of both WT PDE11A4 as well as the phosphoresistant PDE11A4-S162A , suggesting phosphorylation of S162 is not needed for the dispersing effect of the isolated GAF-B domain (Fig.15H-15I). While not wishing be bound by any particular theory, this result suggests that disrupting homodimerization does not achieve effects by promoting phosphorylation of S162.
- S162D differed substantially from GAF-B in terms of regulating cyclic nucleotide levels. It was found that S162D elicited quite different effects on cyclic nucleotide levels than were described above for the isolated GAF-B domain (Figs.15C-15D). Specifically, S162D did not alter PDE11A4 hydrolysis of cGMP (Fig.15J) and appeared to reduce PDE11A4 hydrolysis of cAMP (Fig. 15K). Although not wishing to be bound by any particular theory, together these data suggest that homodimerization and pS162 are independent intramolecular mechanism that regulate PDE11A4 trafficking and function.
- Example 4 Pharmacologic Inhibition of PDE11A For Age-related Memory Disorders
- PDE11A phosphodiesterase 11A
- LTM long-term memory
- PDE11A expression increases with aging and dementia in rodents and humans.
- Viral disruption of PDE11A4 protein in old mice reverses age-related cognitive decline of LTMs, pointing to PDE11A inhibition as a novel therapeutic approach.
- Screening of a small molecule compound library using a novel yeast-based system identified multiple tractable hits, each with varying degrees of potency and selectivity. Based on existing SAR, these hits are examined to provide CNS-penetrant candidates for evaluation.
- a secondary cell-based assay validates candidates in a mammalian context and assess the potential for functional selectivity (e.g., the ability to preferentially inhibit cGMP-hydrolytic activity vs. cAMP-hydrolytic activity).
- Acute disruption of PDE11A4 in the hippocampus of old mice is able to reverse age-related LTM deficits, and genetic deletion of PDE11A reduces age-related decline of LTMs.
- age-related cognitive decline is not a uniform process, with variability in symptom severity observed across individuals and cognitive domains. For example, associative memories are more susceptible to age-related cognitive decline than are recognition memories for individual items for reasons that are not clear but may be related to deficient activation of anterior hippocampus. This lack of knowledge slows therapeutic development.
- Pde11a KO mice Consistent with PDE11A4’s restricted expression pattern, Pde11a KO mice appear normal on a wide range of sensory, motor and anxiety/depression-related behaviors, show no gross peripheral pathology at least up to 1 year of age (later ages not assessed), and reproduce normally. Instead, young adult Pde11a KO mice exhibit select social phenotypes such as preferring to interact with other Pde11a KO mice vs wild-type (WT) mice and showing differences in the consolidation of social memories. Pde11a KO mice also have an increased sensitivity to the behavioral effects of lithium.
- the initial focus on social aLTM stems from the fact that in young adult mice, PDE11A4 regulates social behaviors and social memory, but not non-social memory. While aging severely impairs PDE11A WT mice, old KO mice show robust aLTM for STFP on par with that of young PDE11A WT mice.
- Assays for PDE11A inhibition are carried out with both 200nM cAMP and 100 nM cGMP. Substrate concentrations for the remaining PDEs are 100 nM cGMP (PDE1C), 1 ⁇ M cGMP (PDE2A), 30 nM cGMP (PDE3B), 120 nM cAMP (PDE4D), 500 nM cGMP (PDE5A), 1.7 ⁇ M cGMP (PDE6C), 15 nM cAMP (PDE7A), 10 nM cAMP (PDE8A), 70nM cGMP (PDE9A), and 30 nM cAMP (PDE10A).
- PDE11A assays are carried out in an 11- point 1:2 titration from 50 ⁇ M to ⁇ 1nM to determine IC50 values from at least three independent experiments. Compounds displaying IC50 values of ⁇ 50nM against PDE11A are tested at 1 ⁇ M against the remaining 10 PDEs to assure at least a 20-fold level of selectivity. Those compounds for which the IC50 against PDE11A is ⁇ 50nM and demonstrate at least 20- fold selectivity versus other PDEs advance to ADME property evaluation In vitro ADME evaluation and mammalian cell based assay [00416] In vitro ADME properties of suitable PDE11A inhibitors are assessed and compounds meeting criteria move onto a secondary mammalian cell-based assay.
- This secondary screen confirms compounds are taken up by mammalian cells and inhibit mouse PDE11A (species used for in vivo screen). Suitable compounds advance to in vivo pharmacokinetic evaluation.
- Metabolic stability Samples at 37°C in the presence of human or mouse liver microsomes and NADPH according to standard methods. Aliquots are removed at 5 time points, quenched and analyzed (LCMS/MS and MS/MS as needed) for remaining test compound, along with a positive control. Microsomal protein content is adjusted to give consistent results. Data are reported as half-life and clearance. Assay acceptance criteria is 20% for all standards and 25% for the LLOQ.
- Aqueous solubility Kinetic aqueous solubility is measured by adding ⁇ 2 mg samples to pH 7.4 buffered aqueous solutions at 25°C. The mixture is agitated at 25°C for 1 hour , filtered and evaluated by UV and/or LCMSMS analysis. Data are reported as mg/mL and ⁇ M concentration.
- CYP inhibition Compounds are assessed for their ability to inhibit the three major human cytochrome P450 enzymes, 3A4, 2D6 and 2C9. Expressed enzymes are used to minimize non-specific binding and membrane partitioning issues.
- the 3A4 assay uses testosterone as a substrate and is analyzed by LC/MS/MS on a Waters TQ instrument using positive or negative electrospray ionization.
- the 2D6 and 2C9 assays use fluorescent substrates and are analyzed on an Envision Plate Reader.
- MDCK permeability MDCK cell monolayers (Absorption Systems, Malvern, PA) are grown to confluence on collagen-coated microporous membranes in 12-well assay plates.
- Assay buffer consists of Hanks’ balanced salt solution containing 10 mM HEPES and 15 mM glucose at pH – 7.4.
- the buffer in the receiver chamber also contains 1% bovine serum albumin. Compounds are tested at a final concentration of 5 ⁇ M in the assay buffer.
- Cell monolayers are dosed on the apical side (A-B) or basolateral side (B-A) and incubated at 37°C with 5% CO2 in a humidified incubator. Samples are taken from the donor and receiver chambers at 120 minutes. Each determination is performed in duplicate. The flux of Lucifer yellow is also measured post-experimentally for each monolayer to ensure no damage is inflicted to the monolayer during flux period. Samples are assayed on a Waters TQ LC/MS/MS using positive or negative electrospray ionization.
- Mammalian cell-based functional assay COS1 monkey fibroblast cells transiently transfected with mouse PDE11A (95% homologous to human PDE11A4) are used to confirm inhibition in a mammalian context and assess effects on subcellular compartmentalization (Figs.18A-18F).
- Cells are imaged to quantify changes in mPDE11A subcellular compartmentalization (Figs.18A-18F) and harvested to measure both cAMP and cGMP (as previously reported) to assess potential functional selectivity of enzymatic inhibition (e.g., preferential inhibition of cGMP vs.
- C57BL6 as the mouse strain of choice is driven by the fact that 1) mice from the NIA aging rodent colony are used for as many studies as possible in order to limit husbandry costs, 2) only C57BL6 and Balb/cBy mice are available through the NIA aging rodent colony, and 3) the C57BL6 strain is the closer of the two to the genetic background of the Pde11a mutant mouse line (87% C57BL/6J/12.4% C57BL6/N/1%129S6,).
- NIA limits orders to only 20 mice per month, a breeding colony of C57BL6 mice is established at USC using founders from the NIA stock (i.e., as opposed to the C57BL/J mice on hand).
- Pretreatment interval and the specific doses tested for each compound are empirically determined.
- the pretreatment interval is dictated by the tmax established in the in vivo PK study.
- the logarithmic dose range for behavioral testing is based on the active moiety and selected based on brain exposures from the PK study in comparison to the mammalian cell-based assay EC 50 .
- In vivo dose ranges for compounds with a nanomolar in vitro EC50 typically fall within the range of 0.1-3 mg/kg; whereas, in vivo dose ranges for compounds with a micromolar in vitro EC 50 typically range from 1-30 mg/kg.
- behaviorally profile compounds following both acute and chronic dosing is not performed. Compounds are not tested with na ⁇ ve cohorts for each behavioral assay tested.
- chronic dosing is utilized over acute dosing because a chronic dosing schedule better enables repeated behavioral testing as it is likely to yield steady-state levels of compound and avoid sensitization associated with a dose-withdrawal-dose schedule.
- a chronic dosing regime is used because 1) chronic dosing better mirrors the time course of PDE11A lentivirus (Fig.16) mice habituates over time to any stress that may be associated with dosing, 3) having drug on board during all phases of a memory assay (i.e., acquisition, consolidation and retrieval) mitigates concerns about potential state-dependent effects on memory performance drugs are typically dosed chronically in a clinical setting.
- Compounds are delivered orally via peanut butter pellets.
- mice The integrity of the blood brain barrier is known to deteriorate with age; thus, old male and female C57BL6 mice from the NIA aging rodent colony are used in these studies. Studies are designed in such a way that the same cohort of mice is used to establish oral bioavailability (%F; i.e., oral vs. i.v. [plasma]), target engagement (i.e., drug exposure in the brain), and target modulation (i.e., increased phosphorylation of established downstream target pS6-235/236 in the VHIPP. Mice are fasted and then administered a 3 mg/kg dose either orally via peanut butter pellet or by i.v. injection.
- %F oral bioavailability
- target engagement i.e., drug exposure in the brain
- target modulation i.e., increased phosphorylation of established downstream target pS6-235/236 in the VHIPP.
- Mice are fasted and then administered a 3 mg/kg dose either orally via peanut butter pellet or
- blood samples are placed into chilled, EDTA-fortified tubes, centrifuged for 10 minutes at 3000 rpm (4 °C), and resulting plasma aliquoted into 96-well plates for Liquid Chromatography-Tandem Mass Spectrometry (LC/MS/MS).
- VHIPP for processing via Western blot to measure pS6
- brain samples leftover following dissection of the VHIPP for processing by LC/MS/MS to measure drug exposure
- PBS snap frozen and separately stored at -80 °C.
- vehicle-treated young mice e.g., 10/sex/group; Fig.22
- a pharmacological “positive control” group is not included. That said, inclusion of a young vehicle-treated group ensures the assay itself is performing as expected on the test day (e.g., that it was possible to read out strong memory in the STFP assay).
- Phase I focuses on compound efficacy in the 7d STFP assay; however, to interpret effects in STFP SOR and NSOR are also assessed since an effect on STFP could manifest as a result of stronger stimulus detection or encoding as opposed to a true enhancement of the association between the 2 stimuli. Since STFP requires food restriction, SOR is tested first, then NSOR, then STFP, as has been done for the genetic/viral cohorts (Fig.22). Previously, it was shown that stronger 7d STFP memory retrieval caused by PDE11A deletion correlates with stronger neuronal activation and changes in functional connectivity of brain regions related to the consolidation of remote aLTMs.
- mice are killed immediately at the end of their 7d STFP test and brains harvested and hemisected so that changes in neuronal activity/functional connectivity are assessed via expression of the activity-regulated immediate early gene Arc in one hemisphere as well as protein expression of PDE11A4, other closely related PDEs (e.g., PDE2, PDE10) and pS6- 235/236 using the VHIPP of the other hemisphere. It is determined if PDE11i’s downregulate PDE11A4 protein since PDE11A HTs express 50% of mRNA, but only 20% of protein, suggesting PDE11A is self-regulating.
- PDE11i other closely related PDEs
- Locomotor activity is assessed first using the open field as per the previous methods so that potential sedatory/stimulatory effects (e.g., changes in total distance travelled or movement speed) are detected as well as potential anxiolytic/anxiogenic-like effects (i.e., changes in distance travelled/time spent in center:periphery).
- General health e.g., body weight and temperature, reflexes, muscle and skin tone, etc
- is then assessed using the “Frailty” battery which is highly similar to the SHIRPA observation screen used previously in adult Pde11a KO mice and saw no effects. These same mice are then tested to determine if the PDE11i alters social preference.
- mice act as the ‘mouse psychiatrist’ and be given an opportunity to interact with a vehicle-treated old C57BL6 vs. a drug-treated old C57BL6.
- mice are tested in a 2-day rotarod protocol, as has been previously published. This protocol allows for assessment of both motor coordination on day 1 as well as the phenomenon of “spontaneous improvement” that occurs during the consolidation of motor memory between days 1 and 2. Deficits in spontaneous improvement have been noted in patients with depression and animal models thereof.
- Recent STFP aLTM as well as Arc expression as a surrogate marker of neural activity/connectivity is assessed.
- SC is the process by which a hippocampal memory is relocated to the cortex and subsequently erased/silenced from the hippocampus.
- the origin and destination of retrieval vectors must also be updated. If PDE11A deletion relocates and erases the hippocampus memory ahead of schedule without updating the retrieval vector, this effectively creates a “blackout” period by temporarily misplacing the memory.
- active candidate(s) are profiled using the Pde11a mutant mouse line to establish effects of the compound is due to inhibition of PDE11A.
- PDE11A inhibitors improve memory in STFP in PDE11A WT mice.
- PDE11A inhibitors have no effect in PDE11A KO mice.
- PDE11i’s to produce favorable side effect profiles since 1) expression of the enzyme is relatively restricted and Pde11a KO mice appear normal on a wide range of sensory, motor and anxiety/depression-related behaviors, show no gross changes in general health or peripheral pathology, and reproduce normally. Such a profile is interpreted as proof in favor of developing and pursuing PDE11A4 inhibitors as potential therapeutics for age- related decline of aLTMs.
- alternative methods and starting materials for synthesis of target compounds are utilized.
- a bolus administration once/day may not provide a window of PDE11A inhibition that is sufficiently long to impact memory consolidation, given consolidation requires multiple waves of intracellular signaling across the first 24 hours if not for several days.
- the drug is delivered 2-3 times per day (depending on 1 ⁇ 2 lives) or via implanted Alzet osmotic mini-pumps that enable continuous drug delivery.
- mice In another non-limiting example, two additional groups of mice are included —1 group treated with the GAF-B lentivirus that reverses age-related STFP deficits (see Fig.22) and a 2 nd group with the respective negative viral control. Although including these additional groups reduce the total number of compounds screened in vivo, the GAF-B infused group could then serve as a “positive control”, albeit a genetic one.
- samples In Western blot experiments, samples generally span multiple blots. To account for non-specific technical differences across blots (e.g., transfer or antibody binding efficiency, etc.), all biochemical data are normalized as a fold change of the control group on a given blot. All datasets meeting normality and equal variance assumptions are tested by parametric statistics; those not meeting assumptions are tested by nonparametric statistics. In general, data are analyzed by multifactorial ANOVAs or by repeated measure ANOVAs where appropriate to account for multiple comparisons. For example, in vivo data are analyzed for effects of sex, genotype, and drug treatment in addition to assay-specific factors, such as food type. Statistical outliers (>2 standard deviations from the mean) are dropped from analyses.
- mice [00433] The rationale for the disclosure is based on both mouse and human data, which strengthens rigor. Further, the primary behavioral findings upon which this disclosure is based have been replicated across multiple cohorts of mice. [00434] Genetically modified mice are genotyped a priori to enable proper counterbalancing of experimental run lists (and genotypes reconfirmed post death by Western blot or in situ hybridization); however, experimenters are blind to genotype or drug treatment at the time of data collection/analysis.
- blinded drug pellets are prepared by an experimenter different than the one conducting the behavioral experiments.
- each cage of old mice include 1 subject from each treatment group to counterbalance any slight environmental differences that may arise between cages and to avoid litter effects (since most mice within cage tend to be from the same litter).
- Physical parameters are counterbalanced across subjects (e.g., which is the “trained” vs. “novel” spice).
- Biological Variables Equal numbers of males and females are always tested to test for effect of sex. SUMMARY [00436]
- PDE11A is a member of a highly druggable enzyme family that is positioned to selectively control cyclic nucleotide signaling in a molecularly-defined population of neurons, without affecting signaling elsewhere. This can diminish age-related deficits in at least social aLTMs without causing unwanted side effects.
- PDE11A is a realistic candidate for drug development increases the value of developing small molecule inhibitors and understanding its function.
- the characterization of lead candidates are broadened. Effects on social conditioned place preference, a different type of social aLTM, are examined as effects on nonsocial types of aLTMs. It is also of interest to test the lead PDE11Ai’s in other cognitive domains including spatial memory, other depression-related behaviors, additional anxiety-related behaviors, and sensorimotor gating.
- PDE11i can modulate lithium responsivity.
- the mechanism by which a PDE11i may improve age-related cognitive decline and determine therapeutic windows for lead candidates i.e., establish maximum tolerated dose, tissue toxicity, etc.
- PDE11A Inhibitors for CNS Disorders [00437] To test the hypothesis that age-related cognitive decline of aLTMs is driven by the age-related increases in PDE11A4 expression, young vs. old Pde11a WT and KO mice were compared in social transmission of food preference (STFP; Figs.16A-16B).
- Fig.16A shows aged male and female PDE11A WT mice show no STFP memory 7 days after training.
- Aged PDE11A KO mice show robust memory equivalent to young (Y) mice. This protective effect was replicated across both males and females in two large cohorts (26-29/group).
- the protective effect of PDE11A is specific for memory as evidenced by Fig.16B that illustrates that there is no difference in recognition long term memory (rLTM) among WT and PDE11A KO young mice, as measured by non-social order recognition (NSOR).
- Figs.15A-15C show a comparison of young and old mice that display reduced aLTMs based on STFP, and no reduction in rLTMs based on NSOR, or social order recognition assays.
- Fig.27A shows an exemplary group of compounds of the disclosure.
- Fig.27B provides data demonstrating the comparative potency of the listed compounds as PDE11A4 inhibitors.
- percent (%) values refer to percent inhibition, and non-percent values refer to IC 50 values.
- the mixture was heated to reflux under nitrogen overnight.
- the cooled reaction was diluted with ice water and neutralized to pH at least 7 with saturated sodium bicarbonate solution, then extracted with three portions of ethyl acetate.
- the collected organic extracts were dried and concentrated and the crude product purified by silica gel chromatography eluting with ethyl acetate/hexanes to furnish the product aminopyrazole.
- This aminopyrazole was dissolved in acetic acid and reacted with 1.1 equivalent of sodium diethyl oxaloacetate at reflux overnight.
- the cooled reaction mixture was poured over ice and the precipitated product was collected by filtration and washed with water then dried.
- Esters were hydrolyzed to the corresponding carboxylic acids as follows: Ester was dissolved in aqueous tetrahydrofuran at room temperature. Three equivalents of lithium hydroxide were added and the mixture stirred at room temperature overnight. The reaction was poured into ice water and pH was adjusted to 1 with 1N HCl. The resulting precipitate was collected by filtration and dried to furnish the desired carboxylic acids. [00451] Synthesis of oxa- and thio-(3,4-diazoles): [00452] Carboxylic acid (0.1 g) was dissolved in 3 ml POCl 3 at room temperature.
- Tricycle 6 (Figure 27) has good water solubility and generally favorable metabolic stability in human and mouse microsomes with a favorable CYP profile. However, it has fewer obvious vectors and a screen of commercially available analogs furnished flat structure-activity data.
- Pyrrolopyrimidine 5 (Figure 27) is highly lipophilic with poor water solubility, a clean CYP profile and good metabolic stability with three sites for optimization.
- Thienopyrimidinone 7 ( Figure 27) is metabolically unstable and analysis of commercially available propylthio analogs were significantly less potent (> 1 ⁇ M). While not wishing to be bound by any particular theory, this result suggests that this site would have limited value.
- tadalafil (1) as a positive control, compounds 14b and 23b were selected because 14b is comparable to 1 in enzymatic potency and difluoromethyl analog 23b is measurably more potent in the biochemical assay compared to 14b. Concentration-dependent effects of all three compounds on both cyclic nucleotides were observed to furnish EC 50 values shown in Table 6. Tadalafil (1) and 14b showed similar efficacy, and compound 23b showed a significant improvement in efficacy compared to 1 for both cAMP and cGMP.
- reaction mixture Upon completion of reaction, the reaction mixture was poured into ice cold water and extracted with three portions of ethyl acetate, washed with water, brine, dried and concentrated to afford crude product. The crude product was purified by flash chromatography eluting with hexanes/ethyl acetate.
- 24b To a solution of 24a (0.13 mmol, 1eq.) in DMF was added HATU (0.15 mmol, 1.2 eq.) and NH4Cl (0.26 mmol, 1.2 eq.) respectively. Triethylamine (0.65 mmol, 5 eq.) was added drop-wise and stirred at room temperature for overnight under N2. The reaction mixture was poured into ice water and the yellow precipitate that formed was collected by filtration, washed with water, and dried to furnish the desired amide in 78% yield.
- PDE3A using recombinant human PDE3A, PDE4D3, PDE5A, PDE6C, PDE10A1, and PDE11A4 (BPS Bioscience)).
- Substrate concentrations used were 15 nM cGMP (PDE3A), 18nM cAMP (PDE3A) 200 nM cAMP (PDE4D3), 500 nM cGMP (PDE5A), 1.7 ⁇ M cGMP (PDE6C), 30 nM cAMP (PDE10A), 1.3 ⁇ M cGMP (PDE10A), 100 nM cGMP (PDE11A4) and 240 nM cAMP (PDE11A4).
- Inhibitor concentrations using a 10 point curve that reduce enzyme activity by 50% are presented as calculated using an online IC50 Calculator. Inhibitor concentrations of 4, 13.7, 40, 123, 370, 1110, 3330 and 10,000 nM were used. The values reported are means of at least three independent experiments. Substrate concentrations were ⁇ 0.1 ⁇ K M for each enzyme; thus, IC 50 values approximate the Ki values.
- Cell-based assay [00485] HT-22 cells (sex undefined) were cultured and transfected as previously described.
- DMEM Modified Eagle Medium
- P/S Penicillin/Streptomycin
- FBS fetal bovine serum
- Example 7 Effects of PDE11A inhibitors in an in vitro model of aging [00486] In vivo, expression of PDE11A4 increases in the hippocampus across the lifespan in humans and rodents, resulting in age-related punctate accumulations of PDE11A4 protein in “ghost axons.” These age-related PDE11A4 accumulations were mimicked by overexpressing mouse PDE11A4 in mouse HT-22 hippocampal cells. An image of HT22 cells overexpressing EmGFP-mPDE11A4 is shown in FIG.30.
- Figs.31A-31B is an image of HT-22 cells overexpressing EmGFP-mPDE11A4 treated with the vehicle DMSO (Fig.30A) or 1 uM 25b (Fig.30B).
- PDE11A inhibitor compounds 9a, 14k, 14b and 25b were able to reverse these aging-like accumulations of mPDE11A4 in HT-22 cells, along with the drug Tadalafil, a PDE5 inhibitor that also potently inhibits PDE11A4, along with the commercially-available PDE11A4 inhibitor BC11-38.
- FIGS.35A and 35B show images of HT-22 cells overexpressing EmGFP-mPDE11A4 treated with the vehicle DMSO (35A) or 1 uM 25b (35B). Further, the compound 25b was found to be more potent and more efficacious than the drug tadalafil in reversing this aging-like PDE11A4 phenotype (Fig.6).
- Example 8 PDE11A4 Inhibition [00488] Compounds of the disclosure were evaluated in vitro for PDE11A4 inhibition: [00489] Compound 1001: 62% inhibition @ 500 nM. [00490] Compound 1003: IC 50 : 140 nM.
- Phosphodiesterase 11A (PDE11A), Enriched in Ventral Hippocampus Neurons, is Required for Consolidation of Social but not Nonsocial Memories in Mice.
- Neuropsychopharmacology official publication of the American College of Neuropsychopharmacology. 2016;41(12):2920-31. doi: 10.1038/npp.2016.106. PubMed PMID: 27339393; PubMed Central PMCID: PMC5061884. 3. Abbott A. Cognition: The brain's decline. Nature. 2012;492(7427):S4-5. doi: 10.1038/492S4a. PubMed PMID: 23222672. 4.
- Guerreiro R Bras J. The age factor in Alzheimer's disease. Genome medicine.
- PubMed PMID 26482651; PubMed Central PMCID: PMC4617238. 5. Plassman BL, Langa KM, Fisher GG, Heeringa SG, Weir DR, Ofstedal MB, Burke JR, Hurd MD, Potter GG, Rodgers WL, Steffens DC, McArdle JJ, Willis RJ, Wallace RB. Prevalence of cognitive impairment without dementia in the United States. Annals of internal medicine. 2008;148(6):427-34. PubMed PMID: 18347351; PubMed Central PMCID: PMC2670458. 6. Bender AR, Naveh-Benjamin M, Raz N.
- Kelsoe J inventor; THE REGENTS OF THE UNIVERSITY OF CALIFORNIA (Oakland, CA), assignee. METHOD TO PREDICT RESPONSE TO TREATMENT FOR PSYCHIATRIC ILLNESSES. USA2010. 16.
- Rizzo LB Costa LG, Mansur RB, Swardfager W, Belangero SI, Grassi-Oliveira R, McIntyre RS, Bauer ME, Brietzke E. The theory of bipolar disorder as an illness of accelerated aging: implications for clinical care and research. Neuroscience and biobehavioral reviews. 2014;42:157-69. doi: 10.1016/j.neubiorev.2014.02.004. PubMed PMID: 24548785. 18.
- Kinser PA Lyon DE. Major depressive disorder and measures of cellular aging: an integrative review. Nursing research and practice. 2013;2013:469070. doi: 10.1155/2013/469070.
- PubMed PMID 23691300; PubMed Central PMCID: PMC3649747. 19.
- Kelly MP Logue SF, Brennan J, Day JP, Lakkaraju S, Jiang L, Zhong X, Tam M, Sukoff Rizzo SJ, Platt BJ, Dwyer JM, Neal S, Pulito VL, Agostino MJ, Grauer SM, Navarra RL, Kelley C, Comery TA, Murrills RJ, Houslay MD, Brandon NJ.
- Phosphodiesterase 11A in brain is enriched in ventral hippocampus and deletion causes psychiatric disease-related phenotypes. Proceedings of the National Academy of Sciences of the United States of America.
- PubMed PMID 20822256; PubMed Central PMCID: PMC3011045. 29. Old SR, Naveh-Benjamin M. Differential effects of age on item and associative measures of memory: a meta-analysis. Psychology and aging. 2008;23(1):104-18. doi: 10.1037/0882-7974.23.1.104. PubMed PMID: 18361660. 30. Bridger EK, Kursawe AL, Bader R, Tibon R, Gronau N, Levy DA, Mecklinger A. Age effects on associative memory for novel picture pairings. Brain research. 2017;1664:102-15. doi: 10.1016/j.brainres.2017.03.031. PubMed PMID: 28377157. 31.
- PubMed PMID 27713000. 36. Dalton MA, Tu S, Hornberger M, Hodges JR, Piguet O. Medial temporal lobe contributions to intra-item associative recognition memory in the aging brain. Frontiers in behavioral neuroscience. 2013;7:222. doi: 10.3389/fnbeh.2013.00222. PubMed PMID: 24427127; PubMed Central PMCID: PMC3878719. 37. Yuasa K, Ohgaru T, Asahina M, Omori K. Identification of rat cyclic nucleotide phosphodiesterase 11A (PDE11A): comparison of rat and human PDE11A splicing variants.
- PDE11A rat cyclic nucleotide phosphodiesterase 11A
- Rizzo LB Costa LG
- Mansur RB Swardfager W
- Belangero SI Grassi-Oliveira R
- McIntyre RS Bauer ME
- Brietzke E The theory of bipolar disorder as an illness of accelerated aging: implications for clinical care and research. Neuroscience and biobehavioral reviews. 2014;42:157-69. doi: 10.1016/j.neubiorev.2014.02.004. PubMed PMID: 24548785.
- Kinser PA Lyon DE. Major depressive disorder and measures of cellular aging: an integrative review. Nursing research and practice. 2013;2013:469070.
- PubMed PMID 23691300; PubMed Central PMCID: PMC3649747. 48.
- Kelly MP Logue SF, Brennan J, Day JP, Lakkaraju S, Jiang L, Zhong X, Tam M, Sukoff Rizzo SJ, Platt BJ, Dwyer JM, Neal S, Pulito VL, Agostino MJ, Grauer SM, Navarra RL, Kelley C, Comery TA, Murrills RJ, Houslay MD, Brandon NJ.
- Phosphodiesterase 11A in brain is enriched in ventral hippocampus and deletion causes psychiatric disease-related phenotypes.
- PubMed PMID 22105073; PubMed Central PMCID: PMC3256904.
- Pilarzyk K Klett J, Pena EA, Porcher L, Smith AJ, Kelly MP. Loss of Function of Phosphodiesterase 11A4 Shows that Recent and Remote Long-Term Memories Is Uncoupled. Current biology: CB.2019. doi: 10.1016/j.cub.2019.06.018. PubMed PMID: 31303492.
- PDE11A regulates social behaviors and is a key mechanism by which social experience sculpts the brain. Neuroscience.
- PubMed PMID 20228279; PubMed Central PMCID: PMC2854023.
- de Medeiros AS Wyman AR, Alaamery MA, Allain C, Ivey FD, Wang L, Le H, Morken JP, Habara A, Le C, Cui S, Lerner A, Hoffman CS. Identification and characterization of a potent and biologically-active PDE4/7 inhibitor via fission yeast-based assays. Cellular signalling. 2017;40:73-80. doi: 10.1016/j.cellsig.2017.08.011. PubMed PMID: 28867658; PubMed Central PMCID: PMC5651194. 63.
- EGF-R epidermal growth factor receptor
- Her2(p185(erbB) Her2(p185(erbB)
- Schizosaccharomyces pombe Git7p a member of the Saccharomyces cerevisiae Sgtlp family, is required for glucose and cyclic AMP signaling, cell wall integrity, and septation.
- Stiefel J Wang L, Kelly DA, Janoo RTK, Seitz J, Whitehall SK, Hoffman CS. Suppressors of an adenylate cyclase deletion in the fission yeast Schizosaccharomyces pombe.
- Phosphodiesterase 11A (PDE11A), Enriched in Ventral Hippocampus Neurons, is Required for Consolidation of Social but not Nonsocial Memories in Mice. Neuropsychopharmacology, 2016, 41, 2920-2931. 130. Kelly, M.P.; Logue, S.F.; Brennan, J.E.; Brandon, N.J. Phosphodiesterase 11A in brain is enriched in ventral hippocampus and deletion causes psychiatric disease-related phenotypes. Proc. Natl. Acad. Sci. USA, 2010, 107, 8457-8462. 131.
- PDE10A inhibitors-clinical failure or a window into antipsychotic drug action Front. Neurosci.2021, 14, 600178. 154. Weeks, J. L.; Blount, M. A.; Beasley, A.; Zoraghi, R.; Thomas, M. K.; Sekhar, K. R.; Corbin, J. D.; Francis, S. H. Radiolabeled ligand binding to the catalytic or allosteric sites of PDE5 and PDE11. Meth. Mol. Biol.2005, 307, 239-262. 155. Pilarzyk, K.; Porcher, L.; Capell, W. R.; Burbano, S.D.; Davis, J.; Fisher, J.
Landscapes
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Neurosurgery (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biomedical Technology (AREA)
- Neurology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Life Sciences & Earth Sciences (AREA)
- Hospice & Palliative Care (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Psychiatry (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
Abstract
Description
Claims
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP23847619.6A EP4561567A2 (en) | 2022-07-28 | 2023-07-28 | Inhibitors of pde11a4 and methods of using same |
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202263393187P | 2022-07-28 | 2022-07-28 | |
| US63/393,187 | 2022-07-28 | ||
| US202263370546P | 2022-08-05 | 2022-08-05 | |
| US63/370,546 | 2022-08-05 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| WO2024026500A2 true WO2024026500A2 (en) | 2024-02-01 |
| WO2024026500A3 WO2024026500A3 (en) | 2024-03-07 |
Family
ID=89707415
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2023/071289 Ceased WO2024026500A2 (en) | 2022-07-28 | 2023-07-28 | Inhibitors of pde11a4 and methods of using same |
Country Status (2)
| Country | Link |
|---|---|
| EP (1) | EP4561567A2 (en) |
| WO (1) | WO2024026500A2 (en) |
Family Cites Families (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| GB0115109D0 (en) * | 2001-06-21 | 2001-08-15 | Aventis Pharma Ltd | Chemical compounds |
-
2023
- 2023-07-28 EP EP23847619.6A patent/EP4561567A2/en active Pending
- 2023-07-28 WO PCT/US2023/071289 patent/WO2024026500A2/en not_active Ceased
Also Published As
| Publication number | Publication date |
|---|---|
| EP4561567A2 (en) | 2025-06-04 |
| WO2024026500A3 (en) | 2024-03-07 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US12202810B2 (en) | Non-ATP/catalytic site p38 mitogen activated protein kinase inhibitors | |
| Lim et al. | 14-3-3ζ coordinates adipogenesis of visceral fat | |
| JP6022442B2 (en) | Male contraceptive compositions and methods of use | |
| CA3123897C (en) | Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment | |
| AU2022218616B2 (en) | Non-catalytic substrate-selective P38α-specific MAPK inhibitors with endothelial-stabilizing and anti-inflammatory activity, and methods of use thereof | |
| Rabal et al. | Multitarget approach for the treatment of Alzheimer’s disease: Inhibition of Phosphodiesterase 9 (PDE9) and Histone Deacetylases (HDACs) covering diverse selectivity profiles | |
| US20190192521A1 (en) | Methods of Treating Arid1A-Mutated Cancers With HDAC6 Inhibitors and EZH2 Inhibitors | |
| ES2770047T3 (en) | Azetidinyloxyphenylpyrrolidine compounds | |
| KR20110111495A (en) | Heteroaryl substituted pyridazinone derivatives | |
| CN103068800A (en) | Piperidinyl compound as a modulator of chemokine receptor activity | |
| Chi et al. | Discovery and synthesis of a pyrimidine-based aurora kinase inhibitor to reduce levels of MYC oncoproteins | |
| JP2016528266A (en) | V1a antagonists for treating phase shift sleep disorders | |
| US11453640B2 (en) | Small molecules for disrupting the super elongation complex and inhibiting transcription elongation for cancer therapy | |
| Dey et al. | Selective phosphodiesterase 1 inhibitor BTTQ reduces blood pressure in spontaneously hypertensive and dahl salt sensitive rats: role of peripheral vasodilation | |
| Ma et al. | Discovery of Novel 4, 5, 6, 7-Tetrahydro-7 H-pyrazolo [3, 4-c] pyridin-7-one Derivatives as Orally Efficacious ATX Allosteric Inhibitors for the Treatment of Pulmonary Fibrosis | |
| EP4561567A2 (en) | Inhibitors of pde11a4 and methods of using same | |
| US20240150315A1 (en) | Beta-2 adrenoreceptor modulators and methods of using same | |
| Francis | Target identification and validation of a novel family of anti-inflammatory 1, 4-benzodiazepine-2, 5-diones |
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: 23847619 Country of ref document: EP Kind code of ref document: A2 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2023847619 Country of ref document: EP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| ENP | Entry into the national phase |
Ref document number: 2023847619 Country of ref document: EP Effective date: 20250228 |
|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 23847619 Country of ref document: EP Kind code of ref document: A2 |
|
| WWP | Wipo information: published in national office |
Ref document number: 2023847619 Country of ref document: EP |