OA16400A - 4-(5-cyano-pyrazol-1-yl)-piperidine derivatives as GPR 119 modulators. - Google Patents

4-(5-cyano-pyrazol-1-yl)-piperidine derivatives as GPR 119 modulators. Download PDF

Info

Publication number
OA16400A
OA16400A OA1201300182 OA16400A OA 16400 A OA16400 A OA 16400A OA 1201300182 OA1201300182 OA 1201300182 OA 16400 A OA16400 A OA 16400A
Authority
OA
OAPI
Prior art keywords
methyl
cyano
carboxylate
pyrazol
piperidine
Prior art date
Application number
OA1201300182
Inventor
Vincent Mascitti
Kim Francis MC-CLURE
Michael John Munchhof
Ralph Pelton Robinson, Jr.
Original Assignee
Pfizer Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Inc. filed Critical Pfizer Inc.
Publication of OA16400A publication Critical patent/OA16400A/en

Links

Abstract

Compounds that modulate the activity of the G-protein-coupled receptor GPR119 and their uses in the treatment of diseases linked to the modulation of the G-protein- coupled receptor GPR119 in animals are described herein.

Description

FIELD OF THE INVENTION
The présent invention relates to a new class of cyanopyrazoles, pharmaceutical compositions containing these compounds, and their use to modulate the activity of the G-protein-coupled receptor, GPR119,
BACKGROUND
Diabètes mellitus are disorders in which hrgh levels of blood glucose occur as a conséquence of abnormal glucose homeostasis. The most common forms of diabètes mellitus are Type I (also referred to as insulin-dependent diabètes mellitus) and Type II diabètes (also referred to as non-insulin-dependent diabètes mellitus). Type II diabètes, accounting for roughly 90% of ail diabetic cases, is a serious progressive disease that results in microvascular complications (including retinopathy, neuropathy and nephropathy) as well as macrovascular complications (including accelerated atherosclerosis, coronary heart disease and stroke).
Currently, there is no cure for diabètes. Standard treatments for the disease are limited, and focus on controlling blood glucose levels to minimize or delay complications.
Current treatments target either insulin résistance (metformin, thiazolidinediones, or insulin) releasefrom beta cells (sulphonylureas, exanatide). Sulphonylureas and other compounds that act via depolarization of the beta cell promote hypoglycemia as they stimulate insulin sécrétion independent of circulating glucose concentrations. One approved drug, exanatide, stimulâtes insulin sécrétion only in the presence of high glucose, but must be injected due to a lack of oral bioavailablity. Sitagliptin, a dipeptidyl peptidase IV inhibitor, is a new drug that increases blood levels of incretin hormones, which can increase insulin sécrétion, reduce glucagon sécrétion and hâve other less well characterized effects. However, sitagliptin and other dipeptidyl peptidases IV inhibitors may also influence the tissue levels of other hormones and peptides, and the long-term conséquences of this broader effect hâve not been fully investigated. In Type II diabètes, muscle, fat and liver cells fail to respond normally to insulin.
This condition (insulin résistance) may be due to reduced numbers of cellular insulin receptors, disruption of cellular signaling pathways, or both. At first, the beta cells compensate for insulin résistance by increasing insulin output. Eventually, however, the
beta cells become unable to produce sufficient insulin to maintain normal glucose levels (euglycemia), indicating progression to Type II diabètes.
In Type II diabètes, fasting hyperglycemia occurs due to insulin résistance combined with beta cell dysfunction. There are two aspects of beta cell defect dysfunction: 1 ) increased basal insulin release (occurring at low, non-stimulatory glucose concentrations), which is observed in obese, insulin-resistant pre-diabetic stages as well as in Type II diabètes, and 2) in response to a hyperglycémie challenge, a failure to increase insulin release above the already elevated basal level, which does not occur in pre-diabetic stages and may signal the transition from normo-glycemic insulin-resistant states to Type II diabètes. Current thérapies to treat the latter aspect include inhibitors of the beta-cell ATP-sensitive potassium channel to trigger the release of endogenous insulin stores, and administration of exogenous insulin. Neither achieves accurate normalization of blood glucose levels and both carry the risk of eliciting hypoglycémie.
Thus, there has been great interest in the discovery of agents that fonction in a glucose-dependent manner. Physiological signaling pathways which fonction in this way are well known, including gut peptides GLP-1 and GIP. These hormones signal via cognate G-protein coupled receptors to stimulate production of cAMP in pancreatic beta-cells. Increased cAMP apparently does not resuit in stimulation of insulin release during the fasting or pre-prandial state. However, a number of biochemical targets of cAMP, including the ATP-sensitive potassium channel, voltage-sensitive potassium channels and the exocytotic machinery, are modulated such that insulin sécrétion due to postprandial glucose stimulation is significantly enhanced. Therefore, agonist modulators of novel, similarly functioning, beta-cell GPCRs, including GPR119, would also stimulate the release of endogenous insulin and promote normalization of glucose levels in Type II diabètes patients. It has also been shown that increased cAMP, for example as a resuit of GLP-1 stimulation, promotes beta-cell prolifération, inhibits betacell death and, thus, improves islet mass. This positive effect on beta-cell mass should be bénéficiai in Type II diabètes where insufficient insulin is produced.
It is well known that metabolic diseases hâve négative effects on other physiological Systems and there is often co-occurrence of multiple disease states (e.g., Type I diabètes, Type II diabètes, inadéquate glucose tolérance, insulin résistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, obesity or cardiovascular disease in Syndrome X) or secondary diseases which occur secondary to diabètes such as kidney disease, and peripheral
neuropathy. Thus, treatment of the diabetic condition should be of benefit to such interconnected disease states.
SUMMARY OF THE INVENTION
In accordance with the présent invention, a new class of GPR 119 modulators has been discovered. These compounds include:
Isopropyl 4-{5-cyano-4-[(2,4-difluorophenoxy)methyl]-1 H-pyrazol-1 -yljpiperidine-1 carboxylate;
Isopropyl 4-{5-cyano-4-[(2-methylphenoxy)methyl]-1 H-pyrazol-1 -ylfpi pend i ne-1 carboxylate;
-Methylcyclopropyl 4-{5-cyano-4-[(2,5-difluorophenoxy)methyl]-1 H-pyrazol-1 y l}pi perid i n e-1 -carboxylate;
-Methylcyclopropyl 4-{5-cyano-4-[(2,3-difluorophenoxy)methyl]-1 H-pyrazol-1 y l}pi pend i ne-1 -carboxylate;
-Methylcyclopropyl 4-{4-[(4-carbamoyl-2-fluorophenoxy)methyl]-5-cyano-1 H-pyrazol-1 y l}p iperid i ne-1 -carboxylate;
-Methylcyclopropyl 4-(4-(( 4-carbamoylphenoxy)methyl]-5-cyano-1 H-pyrazol-1 yl}p iperid i n e-1 -carboxylate;
1-Methylcyclopropyl 4-(5-cyano-4-((4-cyanophenoxy)methyl)-1 H-pyrazol-1 -yl)piperidine-
1-carboxylate;
Isopropyl 4-(4-((4-(1 H-pyrazol-1-yl)phenoxy)methyl)-5-cyano-1 H-pyrazol-1-yl)piperidine-
1-carboxylate;
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(1 H-tetrazol-5-yl)phenoxy)methyl)-1 H-pyrazol-1yl)piperidine-1 -carboxylate and Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2H-tetrazol-5yl)phenoxy)methyl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate;
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(1-methyl-1 H-tetrazol-5-yl)phenoxy)methyl)-1Hpyrazol-1 -y I )pi pe rid in e-1 -carboxylate;
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-methyl-2H-tetrazol-5-yl)phenoxy)methyl)-1Hpyrazol-1 -yl)piperidine-1 -carboxylate;
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-(2-hydroxyethyl)-2H-tetrazol-5yl)phenoxy)methyl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate;
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(1-(2-hydroxyethyl)-1 H-tetrazol-5yl)phenoxy)methyl)-1 H-pyrazol-1 -y I )piperid i ne-1 -carboxylate;
-Methylcyclopropyl 4-(5-cyano-4-([2-fluoro-4-( 1 -methyl-1 H-tetrazol-5yl)phenoxy]methyl}-1 H-pyrazol-1 -yl)piperidine-1 -carboxylate;
Λ !eje|Àxoqjeo-t
-9uipuadid([A-|,-|0ZBjXd-H i4|Aqisw(Axouaqd|Aqi9UJ-£-oiieAo-tz)]-t7-oueAo-g}-t7 |Adojdos| !9}B|Axoqjeo -L-ou!p!Jadid{|A-i.-|ozejXd-|-| L-[|Xq)9(ju(Axou9qdoueAo-£)]-ÿ-oueXo-g}-t7 |Xdojdos| ;a|!J]!uoqjeo-g-9|OzejXd 0£ -H Î-{|Xt|loœi[Axou9qd(|A- l-|ozbjj9]-|-| l )-v]}-k-[|^-V-u!P!J9did( iX-g-uipiLuijAdiAq^-g)- j,]-1, lo;e|Xxoqjeo-i.-9U[p!J8djd(|X -L-|ozejÂd-|-| |,“{|Aq)9Lu[Axou9qd(|X-|,-|ozej]9]-H |,)-t7]}-t7-oueAo-g)-t7 |Adojdo|oAo|Aqj9[/\H !9je|Axoqjeo-|,-e u i pi J9d id [|A- |.-|ozejAd
Ή Hl^MJ9uj{ou!we[|A-£-u!p!jAd(|Auo.||ns|Xqi9iu)-g-|Aq}9UJ-3]})-y-oueA9-g]-t> |Adojdos| ςζ !eie|Axoqjeo- |_-e ui pi J9d id[|A- ^iozejXd-μ [, -(|Xq)9w{ouiuje[|A-c-u!P!jAd(|A- [.-ιοζΒμιν^' (.-μ |J-g-|Aq|9w-z]})-|7-oueAibg]-ÿ |Xdojdos| !9}B|AxoqjBO- (,-9uipu9did[|A- |.-|ozejAd
-H t-(|Aqi9Lu{Axo[|A-£-U!puAd(|A“ ^lozeu^'g' μμ L)-9-|^w-z]})-y-ouBAo-gJ-ÿ |Adojdos| !9lB|XxoqiBO- |_-9U!P!J9did(|A- [,-|0ZBjAd Oc -H ^-{|Aqlθlu[Axouθqd(|A-g-|ozBp!lUl·H L-iAqiew-iJ-tz-ojoniy^-tz-ouBAo-g)--^ |Adojdos| !aiB|AxoqjBQ-1,-9 u i pi j 9d td ( |A- |,-|ozejAd
-H L-{|Xq)9iu[Xxou9qd(|A-s-|OZBp!LU!-H |,-|Aq}ew-|J-t7-ojonu-g]}-y-oueAo-g)-t7 |Adojdos| !9;e|AxoqjE9 -|,-9U!P!J9d!d{|A-|,-|OzejAd-|-|l.-[|Aqi9Uj(Axoueqdojonmj}-9'£,g)]-t7-ouBA9-g}-t7 |Adojdos| g| !9je|AxoqjBO- l-oui pi J9did{]A -l_-|ozBjAd-HHl^qi9w(Axou9qdojony!J)-9‘£'z)]-t7-ouBAo-g}-t7 |AdojdopAo|Aq}9|/\|-|.
!9]B|AxoqjB3- |,-a u I pu edi d( [A -l.-|ozBjAd-HL-{|Aqi9Lu[Axo(|A-£-U!P!jAd|Aqi9LU-3)]H-ouBA3-g)-t7 |Adojdo|oAo|Aq}9|/\|-L· !9}B|Axoqjeo-|,-9U!piJ9d!d(|A oi
-L-|ozBjXd-H|.-{|Xdojd[|Xu9qd(|Auoj|ns|Aq)9iu)-tz-ojon|j-s]-3}-ÿ-ouBAo-g)-y |Adojdos| !9)B|AxoqjB3
- t-9uIpiJ9dId(|A- [,-|ozBjXd-H |,-{|Aqj9[Xxo(|X-£-uipuAd|Aqj9iu-s)]- iJ-ÿ-oueAo-gJ-t? [Adoidos] !9]B|AxoqjB0- L-9U!p!J9did(|A -|.-|OzejAd-H [,-{|Xq)9[Axou9qd([Xuoj|ns|Aq]9Uj)-t7-OJOnu-s]-|.}-ÿ-ouBXo-g)-t7 |Adojdos| g !9]B|AxoqjBD- L-euipijedfd(|A
- L-|ozBjAd-|-| |,-oueAo-g-(|Aq}9iu(Axou9qdojon|y£-|Aoiueqjeo-ÿ))-tz)-t7 [Adojdo|oAo|Aqiey\|-1, !9)B|AxoqjB3- [.-a u ιρμ ed id ( |A-1. - [ozejAd -H v-(|Aqi9Lu[Axou9qd(|A-g-|OZBJi9]-Hl,-|Aq)9UJ-|,)-ÿ]}-ÿ-ouBAo-g)-ÿ |Adojdo|oAo|Aqi9i/\|- L t7
Isopropyl 4-{5-cyano-4-[(4-cyanophenoxy)methyl]-1 H-pyrazol-1-yl}piperidine-1carboxylate;
4-[(4-Cyano-2-fluorophenoxy)methyl]-1-[1-(5-ethylpyrimidin-2-yl)piperidln-4-yl]-1Hpyrazole-5-carbonitrile;
terf-Butyl 4-{5-cyano-4-[(4-cyano-2-fluorophenoxy)methyl]-1 H-pyrazol-1-yl}piperidine-1carboxylate;
Isopropyl 4-{5-cyano-4-[(2-cyano-4-fluorophenoxy)methyl]-1 H-pyrazol-1 -ylfpiperidine-1 carboxylate;
Isopropyl 4-(5-cyano-4-{[4-(dimethylcarbamoyl)-2-fluorophenoxy]methyl}-1 H-pyrazol-1 y I ) pipe rid in e-1 -carboxylate;
-Methylcyclopropyl 4-(5-cyano-4-{[4-(dimethylcarbamoyl)-2-fluorophenoxy]methyl}-1 Hpyrazol-1 -yl )p i perid i n e-1 -carboxylate;
1-Methylcyclopropyl 4-(5-cyano-4-{[2-fluoro-4-(methylcarbamoyl)phenoxy]methyl}-1Hpyrazol-1-yl)piperidine-1 -carboxylate;
4-({5-Cyano-1-[1 -(5-ethylpyrimidin-2-yl)piperidin-4-yl]-1H-pyrazol-4-yl)methoxy)-3-fluoroΝ,Ν-dimethylbenzamide;
-Methylcyclopropyl 4-{5-cyano-4-[(4-cyano-2-fluorophenoxy)methyl]-1 H-pyrazol-1 yl}p iperîd i n e-1 -carboxylate;
terf-Butyl (3S,4S)-4-(5-cyano-4-{[2-fluoro-4-(methylcarbamoyl)phenoxy]methyl}-1Hpyrazol-1 -yl)-3-fluoropiperidine-1 -carboxylate;
terf-Butyl (3R,4S)-4-(5-cyano-4-{[2-fluoro-4-(methylcarbamoyl)phenoxy]methyl}-1Hpyrazol-1 -yl)-3-fluoropiperidine-1 -carboxylate;
1-Methylcyclopropyl (3S,4S)-4-(5-cyano-4-{[2-fluoro-4(methylcarbamoyl)phenoxy]methyl}-1 H-pyrazol-1 -yl)-3-fluoropiperidine-1 -carboxylate;
-Methylcyclopropyl (3R,4R)-4-(5-cyano-4-{[2-fluoro-4(methylcarbamoyl)phenoxy]methyl}-1 H-pyrazol-1 -yl)-3-fluoropiperidine-1 -carboxylate; terf-Butyl (3S,4S)-4-(5-cyano-4-{[(2-methylpyridin-3-yl)oxy]methyl}-1 H-pyrazol-1 -yl)-3fluoropiperidine-1 -carboxylate;
terf-Butyl (3S,4R)-4-(5-cyano-4-{[(2-methylpyridin-3-yl)oxy]methyl}-1 H-pyrazol-1 -yl)-3flu oro pi perid i ne-1 -carboxylate;
1-Methylcyclopropyl (3S,4R)-4-(5-cyano-4-{[(2-methylpyridin-3-yl)oxy]methyl}-1 Hpyrazol-1 -yl )-3-fluoro piperid ine-1 -carboxylate;
1-Methylcyclopropyl (3S,4R)-4-(5-cyano-4-{[(2-methylpyridin-3-yl)oxy]methyl}-1 Hpyrazol-1 -yl)-3-fluoropiperidine-1 -carboxylate;
1-Methylcyclopropyl (3R,4S)-4-(5-cyano-4-{[(2-methylpyridin-3-yl)oxy]methyl}-1 H- pyrazol-1-yl)-3-fluoropiperidine-1-carboxylate;
ferf-Butyl 4-(5-cyano-4-{[4-(1 H-1,2,3-triazol-1-yl)phenoxy]methyl}-1 H-pyrazol-1 yl )p ip er id i ne-1 -carboxylate;
ferf-Butyl 4-(5-cyano-4-{[4-(2H-1,2,3-triazol-2-yl)phenoxy]methyl}-1 H-pyrazol-1yl)piperidine-1-carboxylate;
1-Methylcyclopropyl 4-(4-((4-(1 H-1,2,3-triazol-1-yl)phenoxy)methyl)-5-cyano-1 Hpyrazol-1-yl)piperidine-1-carboxylate;
-Methylcyclopropyl 4-(4-((4-(2H-1,2,3-triazol-2-yl)phenoxy)methyl)-5-cyano-1 Hpyrazol-1 -yl )pi pend i ne-1 -carboxylate;
ferf-Butyl 4-[5-cyano-4-({[1-(methylsulfonyl)piperidin-4-yl]oxy}methyl)-1 H-pyrazol-1 y I ]pi péri d ine-1 -carboxylate;
ferf-Butyl 4-[5-cyano-4-((2-fluoro-4-[(2hydroxyethyl)(methyl)carbamoyl]phenoxy}methyl)-1H-pyrazol-1-yl]piperidine-1carboxylate;
ferf-Butyl 4-[5-cyano-4-({2-fluoro-4-[(3-hydroxypyrrolidin-1-yl)carbonyl]phenoxy}methyl)1 H-pyrazol-1 -yljpiper id ine-1 -carboxylate;
ferf-Butyl 4-(4-{[4-(azetidin-1 -ylcarbonyl)-2-fluorophenoxy]methyl)-5-cyano-1 H-pyrazol1 -yl)piperidine-1 -carboxylate;
-Methylcyclopropyl 4-[5-cyano-4-({[1 -(methylsulfonyl)piperidin-4-yl]oxy}methyl)-1 Hpyrazol-1 -yl] piperid ine-1 -carboxylate;
1-methylcyclopropyl 4-(5-cyano-4-((2-fluoro-4-(1 H-1,2,3-triazol-1-yl)phenoxy)methyl)-
H-pyrazol-1-yl)piperidine-1-carboxylate;
isopropyl 4-(5-cyano-4-((2-fluoro-4-(1 H-1,2,3-triazol-1-yl)phenoxy)methyl)-1 H-pyrazol-1yl)piperidine-1 -carboxylate;
-( 1 -(5-ethylpyrimidin-2-yl)piperidin-4-yl)-4-((2-fluoro-4-( 1 H-1,2,3-triazol-1 yl)phenoxy)methyl)-1H-pyrazole-5-carbonitrile;
isopropyl 4-(4-((4-(1 H-1,2,3-triazol-1-yl)phenoxy)methyl)-5-cyano-1 H-pyrazol-1 yl)piperidine-1-carboxylate;
4-((4-(1 H-1,2,3-triazol-1-yl)phenoxy)methyl)-1-(1-(5-ethylpyrimidin-2-yl)piperidin-4-yl)1 H-pyrazole-5-carbonitrile;
isopropyl 4-(4-((4-(2H-1,2,3-triazol-2-yl)phenoxy)methyl)-5-cyano-1 H-pyrazol-1yl)piperidine-1 -carboxylate;
4-((4-(2H-1,2,3-triazol-2-yl)phenoxy)methyl)-1 -(1 -(5-ethylpyrimidin-2-yl)piperidin-4-yl)1 H-pyrazole-5-carbonitrile;
1-methylcyclopropyl 4-(5-cyano-4-((2-fluoro-4-(2H-1,2,3-triazol-2-yl)phenoxy)methyl)1H-pyrazol-1-yl)piperidine-1-carboxylate;
isopropyl 4-(5-cyano-4-((2-fluoro-4-(2H-1,2,3-triazol-2-yl)phenoxy)methyl)-1 H-pyrazol-1yl)piperidine-1-carboxylate;
1-(1-(5-ethylpyrimidin-2-yl)piperidin-4-yl)-4-((2-fluoro-4-(2H-1,2,3-triazol-2yl)phenoxy)methyl)-1H-pyrazole-5-carbonitrile;
1-methylcyclopropyl 4-(4-((5-(1 H-1,2,3-triazol-1-yl)pyridin-2-yloxy)methyl)-5-cyano-1Hpyrazol-1 -y I )pip erid i ne-1 -carboxylate;
isopropyl 4-(4-((5-( 1 H-1,2,3-triazoI-1 -yI)pyridin-2-yioxy)methyl)-5-cyano-1 H-pyrazol-1 yl)piperidine-1-carboxylate;
1-methylcyclopropyl 4-(5-cyano-4-((3-fluoro-4-(1 H-tetrazol-1-yl)phenoxy)methyl)-1 Hpyrazol-1 -yl) pipe rid i ne-1 -carboxylate;
isopropyl 4-(5-cyano-4-((3-fluoro-4-(1 H-tetrazol-1-yl)phenoxy)methyl)-1 H-pyrazol-1 yl)piperidine-1-carboxylate;
1-(1-(5-ethylpynmidin-2-yl)piperidin-4-yl)-4-((3-fluoro-4-(1H-tetrazol-1yl)phenoxy)methyl)-1H-pyrazole-5-carbonitrile;
4-((5-(1 H-1,2,3-triazol-1-yl)pyridin-2-yloxy)methyl )-1-(1 -(5-ethylpyrimidin-2-yl)piperidin4-yl)-1H-pyrazole-5-carbonitrile;
1-(1-(5-ethylpyrimidin-2-yl)piperidin-4-yl)-4-((2-methyl-6-(1H-1,2,3-triazol-1-yl)pyrïdin-3yloxy)methyl)-1H-pyrazole-5-carbonitrile;
isopropyl 4-(5-cyano-4-((2-methyl-6-(1H-1,213-triazol-1-yl)pyridin-3-yloxy)methyl)-1Hpyrazol-1 -yl )pi perid in e-1 -carboxylate;
1-methylcyclopropyl 4-(5-cyano-4-((2-methyl-6-(1 H-1,2,3-triazol-1 -yl)pyridin-3yloxy)methyl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate;
1-(1-(5-ethylpyrimidin-2-yl)piperidin-4-yl)-4-((2-fluoro-4-(1-methyl-1 H-tetrazol-5yl)phenoxy)methyl)-1H-pyrazole-5-carbonitrile;
-methylcyclopropyl 4-(4-((4-(azetidine-1 -carbonyl)-2-fluorophenoxy)methyl)-5-cyano1 H-pyrazol-1 -y I ) pi perid ine-1 -carboxylate;
isopropyl 4-(4-((4-(azetidine-1-carbonyl)-2-fluorophenoxy)methyl)-5-cyano-1 H-pyrazol1 -yl)piperidine-1 -carboxylate; and
4-((4-(azetidine-1-carbonyl)-2-fluorophenoxy)methyl)-1-(1-(5-ethylpyrimidin-2yl)piperïdin-4-yl)-1H-pyrazole-5-carbonitnle;
or a pharmaceutically acceptable sait thereof.
These compounds modulate the activity of the G-protein-coupled receptor. More specifically the compounds modulate GPR119. As such, said compounds are useful for the treatment of diseases, such as diabètes, in which the activity of GPR119 contributes to the pathology or symptoms of the disease. Examples of such conditions include hyperlipidemia, Type I diabètes mellitus, Type II diabètes mellitus, idiopathic Type I diabètes (Type Ib), latent autoimmune diabètes in adults (LADA), early-onset Type 2 diabètes (EOD), youth-onset atypical diabètes (YOAD), maturity onset diabètes of the young (MODY), malnutrition-related diabètes, gestational diabètes, coronary heart disease, ischémie stroke, restenosis afterangioplasty, peripheral vascular disease, intermittent claudication, myocardial infarction (e.g. necrosis and apoptosis), dyslipidemia, post-prandial lipemia, conditions of impaired glucose tolérance (IGT), conditions of impaired fasting plasma glucose, metabolic acidosis, ketosis, arthritis, obesity, osteoporosis, hypertension, congestive heart failure, left ventricular hypertrophy, peripheral arterial disease, diabetic retinopathy, macular degeneration, cataract, diabetic nephropathy, glomerulosclerosis, chronic rénal failure, diabetic neuropathy, metabolic syndrome, syndrome X, premenstrual syndrome, coronary heart disease, angina pectoris, thrombosis, atherosclerosis, transient ischémie attacks, stroke, vascular restenosis, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertrygliceridemia, insulin résistance, impaired glucose metabolism, conditions of impaired glucose tolérance, conditions of impaired fasting plasma glucose, obesity, erectile dysfunction, skin and connective tissue disorders, foot ulcérations and ulcerative colitis, endothélial dysfunction and impaired vascular compliance. The compounds may be used to treat neurological disorders such as Alzheimer's disease, schizophrenia, and impaired cognition. The compounds will also be bénéficiai in gastrointestinal illnesses such as inflammatory bowel disease, ulcerative colitis, Crohn's disease, irritable bowel syndrome, etc. As noted above, the compounds may also be used to stimulate weight loss in obese patients, especially those afflicted with diabètes.
A further embodiment of the invention is directed to pharmaceutical compositions containing a compound of this invention. Such formulations will typically contain a compound of this invention in admixture with at least one pharmaceutically acceptable excipient. Such formulations may also contain at least one additional pharmaceutical agent. Examples of such agents include anti-obesity agents and/or anti-diabetic agents. Additional aspects of the invention relate to the use of the compounds of this invention in the préparation of médicaments for the treatment of diabètes and related conditions as described herein.
It is to be understood that both the foregoing summary and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed.
DETAILED DESCRIPTION OF THE INVENTION
The présent invention may be understood even more readily by référencé to the following detailed description of exemplary embodiments of the invention and the examples included therein.
It is to be understood that this invention is not limited to spécifie synthetic methods of making that may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. The plural and singular should be treated as interchangeable, other than the indication of number;
a. ''halogen refers to a chlorine, fluorine, iodine, or bromine atom;
b. Ci - C4 alkyl” refers to a branched or straight chained alkyl group containing from 1 to 5 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, etc.;
c. C^ C4 alkoxy refers to a straight or branched chain alkoxy group containing from 1 to 4 carbon atoms, such as methoxy, ethoxy, n-propoxy, isopropoxy, nbutoxy, isobutoxy, etc.;
d. “C3-C6 cycloalkyl refers to a nonaromatic ring that is fully hydrogenated and exists as a single ring. Examples of such carbocyclic rings include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl;
e. C·!- C4 haloalkyl refers to a straight or branched chain alkyl group containing from 1 to 4 carbon atoms, substituted with one or more halogen atoms;
f. “C^ C4 haloalkoxy refers to a straight or branched chain alkoxy group containing from 1 to 4 carbon atoms, substituted with one or more halogen atoms;
g. 5 to 10 membered heteroaryl means a carbocyclic aromatic System having a total of 5 to 10 ring atoms and containing one, two, three or four heteroatoms selected independently from oxygen, nitrogen and sulfur and having one, two or three rings wherein such rings may be fused. The term fused means that a second ring is présent (ie, attached or formed) by having two adjacent atoms in common (ie, shared) with the first ring. The term fused is équivalent to the term condensed. The term heteroaryl embraces aromatic radicals such as pyridine,
pyridazine, pyrazine, pyrimidine, imidazo[1,2-a]pyndine, imidazo[1,5-a]pyridine, [1,2,4]triazolo[4,3-a]pyridine, [1,2,4]triazolo[4,3-b]pyridazine, [1,2,4]triazolo[4,3a]pyrimidine, and [1,2,4]triazolo[1,5-aJpyridine;
h. “therapeutically effective amount means an amount of a compound of the présent invention that (i) treats or prevents the particular disease, condition, or disorder, (ii) atténuâtes, améliorâtes, or éliminâtes one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein;
i. “patient refers to warm blooded animais such as, for example, guinea pigs, mice, rats, gerbils, cats, rabbits, dogs, monkeys, chimpanzees, and humans;
j. treat” embraces both preventative, i.e., prophylactic, and palliative treatment, i.e., relieve, alleviate, or slow the progression of the patient’s disease (or condition) or any tissue damage associated with the disease;
k. the terms modulated, “modulating”, or modulate(s), as used herein, unless otherwise indicated, refers to the activation of the G-protein-coupled receptor GPR119 with compounds of the présent invention;
l. “pharmaceutically acceptable indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingrédients comprising a formulation, and/or the mammal being treated therewith.
m. salts is intended to refer to pharmaceutically acceptable salts and to salts suitable for use in industrial processes, such as the préparation of the compound.
n. pharmaceutically acceptable salts” is intended to refer to either pharmaceutically acceptable acid addition salts or pharmaceutically acceptable basic addition salts depending upon the actual structure of the compound.
o. “pharmaceutically acceptable acid addition salts is intended to apply to any nontoxic organic or inorganic acid addition sait of the base compounds or any of its intermediates. Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulphuric, and phosphoric acid and acid métal salts such as sodium monohydrogen orthophosphate, and potassium hydrogen sulfate. Illustrative organic acids, which form suitable salts include the mono-, di-, and tricarboxylic acids. Illustrative of such acids are for example, acetic, glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, benzoic, hydroxy-benzoic, phenylacetic, cinnamic, salicylic, 2-phenoxybenzoic, p-toluenesulfonic acid, and sulfonic acids such as
methane sulfonic acid and 2-hydroxyethane sulfonic acid. Such salts can exist in eithera hydrated or substantially anhydrousform. In general, the acid addition salts of these compounds are soluble in water and various hydrophilic organic solvents.
p. “pharmaceutically acceptable basic addition salts is intended to apply to any non-toxic organic or inorganic basic addition salts of the compounds or any of its intermediates. Illustrative bases which form suitable salts include alkali métal or alkaline-earth métal hydroxides such as sodium, potassium, calcium, magnésium, or barium hydroxides; ammonia, and aliphatic, alicyclic, or aromatic organic amines such as methylamine, dimethylamine, trimethylamine, and picoline.
q. isomer’' means “stereoisomer and géométrie isomer as defined below. Stereoisomer refers to compounds that possess one or more chiral centers and each center may exist in the R or S configuration. Stereoisomers includes ail diastereomeric, enantiomeric and epimeric forms as well as racemates and mixtures thereof. Géométrie isomer’' refers to compounds that may exist in cis, trans, anti, syn, entgegen (E), and zusammen (Z) forms as well as mixtures thereof.
Certain of the compounds of this invention may exist as géométrie isomers. The compounds may possess one or more asymmetric centers, thus existing as two, or more, stereoisomeric forms. The présent invention includes ail the individual stereoisomers and géométrie isomers of the compounds of this invention and mixtures thereof. Individual enantiomers can be obtained by chiral séparation or using the relevant enantiomer in the synthesis. As noted above, some of the compounds exist as isomers. These isomeric mixtures can be separated into their individual isomers on the basis of their physical chemical différences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher’s acid chloride), separating the diastereoisomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Enantiomers can also be separated by use of a chiral HPLC column, Alternatively, the spécifie stereoisomers may be synthesized by using an optically active starting material, by asymmetric synthesis using optically active reagents, substrates,
catalysts or solvents, or by converting one stereoisomer into the other by asymmetric transformation.
The présent invention also embraces isotopically-labeled compounds of the présent invention which are identical to those recited herein, but for the fact that one or 5 more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine, and chlorine, such as ZH, 3H, 11C, 13C, 14C, 13N, 15N, 150,170, 180,31P, 32P, 35S, 10F, 123l, 125l and 36CI, respectively.
Certain isotopically-labeled compounds of the présent invention (e.g., those labeled with 3H and 14C) are useful in compound and/or substrate tissue distribution assays. Certain isotopically labeled ligands including tritium, 14C, 35S and 125l could be useful in radioligand binding assays. Tritiated (i.e., 3H) and carbon-14 (i.e., 14C) isotopes are particularly preferred for their ease of préparation and detectability.
Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Positron emitting isotopes such as 150,13N, 11C, and 18F are useful for positron émission tomography (PET) studies to examine receptor occupancy.
Isotopically labeled compounds of the présent invention can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an isotopically labeled reagent for a nonrsotopically labeled reagent.
Certain compounds of the présent invention may exist in more than one crystal form (generally referred to as “polymorphs). Polymorphs may be prepared by crystallization under various conditions, for example, using different solvents or different solvent mixtures for recrystallization; crystallization at different températures; and/or various modes of cooling, ranging from very fast to very slow cooling during crystallization. Polymorphs may also be obtained by heating or melting the compound of the présent invention followed by graduai or fast cooling. The presence of polymorphs may be determined by solid probe NMR spectroscopy, IR spectroscopy, differential scanning calorimetry, powder X-ray diffraction or such other techniques.
In addition, the compounds of the présent invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, éthanol, and the like. In general, the solvated forms are considered équivalent to the
unsolvated forms for the purposes of the présent invention. The compounds may also exist in one or more crystalline states, i.e. as co-crystals, polymorphs, or they may exist as amorphous solids. Ail such forms are encompassed by the invention and daims.
In an embodiment in the composition of this invention, the composition further includes at least one additional pharmaceutical agent selected from the group consisting of an anti-obesity agent and an anti-diabetic agent. Example anti-obesity agents include dirlotapide, mitratapide, implitapide, R56918 (CAS No. 403987), CAS No. 913541-47-6, lorcaserin, cetilistat, PYY3.36. naltrexone, oleoyl-estrone, obinepitide, pramlintide, tesofensine, leptin, liraglutide, bromocriptine, orlistat, exenatide, AOD-9604 (CAS No. 221231-10-3) and sibutramine. Example anti-diabetic agents include metformîn, acetohexamide, chlorpropamide, diabinese, glibenclamide, glipizide, glyburide, glimepiride, gliclazide, glipentide, gliquidone, gltsolamide, tolazamide, tolbutamide, tendamistat, trestatin, acarbose, adiposine, camiglibose, emiglitate, miglitol, voglibose, pradimicin-Q, salbostatin, balaglitazone, ciglitazone, darglitazone, englitazone, isaglitazone, pioglitazone, rosiglitazone, troglitazone, exendin-3, exendin-4, trodusquemîne, reservatrol, hyrtiosal extract, sitagliptin, vildagliptin, alogliptin and saxagliptin.
In another embodiment of a method of this invention, the compounds or compositions of this invention may be administered in an effective amount for treating a condition selected from the group consisting of hyperlipidemia, Type I diabètes, Type II diabètes mellitus, idiopathic Type I diabètes (Type lb), latent autoimmune diabètes in adults (LADA), early-onset Type 2 diabètes (EOD), youth-onset atypical diabètes (YOAD), maturity onset diabètes of the young (MODY), malnutrition-related diabètes, gestational diabètes, coronary heart disease, ischémie stroke, restenosis after angioplasty, peripheral vascular disease, intermittent claudication, myocardial infarction (e.g. necrosis and apoptosis), dyslipidemia, post-prandial lipemia, conditions of impaired glucose tolérance (IGT), conditions of impaired fasting plasma glucose, metabolic acidosis, ketosis, arthritis, obesity, osteoporosis, hypertension, congestive heart failure, left ventricular hypertrophy, peripheral arterial disease, diabetic retinopathy, macular degeneration, cataract, diabetic nephropathy, glomerulosclerosis, chronic rénal failure, diabetic neuropathy, metabolic syndrome, syndrome X, premenstrual syndrome, coronary heart disease, angina pectoris, thrombosis, atherosclerosis, myocardial infarction, transïent ischémie attacks, stroke, vascular restenosis, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertrygliceridemia, insulin résistance, impaired glucose metabolism, conditions of impaired glucose tolérance, conditions of impaired
fasting plasma glucose, obesity, erectile dysfunction, skin and connective tissue disorders, foot ulcérations and ulcerative colitis, endothélial dysfunction and impaired vascular compliance, hyper apo B lipoproteinemia, Alzheimer's disease, schizophrenia, impaired cognition, inflammatory bowel disease, ulcerative colitis, Crohn’s disease, and 5 irritable bowel syndrome.
In a further embodiment, the method further includes administering a second composition comprising at least one additional pharmaceutical agent selected from the group consisting of an anti-obesity agent and an anti-diabetic agent, and at least one pharmaceutically acceptable excipient. This method may be used for admistering the compositions simultaneously or sequentially and in any order.
In yet another embodiment, the compounds of this invention are useful in the manufacture of a médicament for treating a disease, condition or disorder that modulâtes the activity of G-protein-coupled receptor GPR119. Furthermore, the compounds are useful in the préparation of a médicament for the treatment of diabètes or a morbîdity associated with said diabètes.
Synthesis
For illustrative purposes, the reaction schemes depicted below provide potential routes for synthesizing the compounds of the présent invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled In the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although spécifie starting materials and reagents are depicted in the schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of dérivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
Compounds of the invention may be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the 30 description contained herein. The starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, Wl) or are readily prepared using methods known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reaqents for Organic Synthesis, v. 1-19, Wiley, New York (1967-1999 ed.), or Beilsteins Handbuch der organischen Chemie, 4,
Aufl. ed. Springer-Verlag, Berlin, including suppléments (also available via the Beilstein online database).
The compounds of this invention can be prepared using methods analogously known in the art for the production of ethers. The reader’s attention is directed to texts such as: 1) Hughes, D. L.; Organic Reactions 1992, 42 Hoboken, NJ, United States;
2) Tikad, A.; Routier, S.; Akssira, M.; Leger, J.-M.l; Jarry, C.; Guillaumet, G. Synlett 2006, 12, 1938-42; and 3) Loksha, Y. M.; Globisch, D.; Pedersen, E. B.; La Colla, P.; Collu, G.; Loddo, R. J. Het. Chem. 2008, 45, 1161-6 which describe such reactions in greater detail.
Scheme 1
Step 3
X-OH
Step 5
Scheme 1 may be used to préparé compounds of Formula N wherein
Z is -C(0)-0-R6 or pyrimidine substituted with CrC4 alkyl, CF3, halogen, cyano, C3-C6 cycloalkyl or C3-C6 cycloalkyl wherein one carbon atom of said cycloalkyl moiety may optionally be substituted with methyl or ethyl;
mis 1,2, or 3;
n is 0, 1 or 2;
R1 is hydrogen, C1-C4 alkyl, or C3-C6 cycloalkyl;
RZa is hydrogen, fluoro or C1-C4 alkyl;
each R3 is individually selected from the group consisting of: hydroxy, halogen, cyano, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, -SO2-R7, P(O)(OR8)(OR9), -C(O)-NR8R9, -N(CH3)-CO-O-(C1-C4) alkyl, -NH-C0-0-(Ci-C4) alkyl,NH-CO-(C1-C4)alky1, -N(CH3)-CO-(Ci-C4) alkyl, -NH-(CH2)2-OH and a 5 to 6-membered heteroaryl group containing 1,2, 3 or 4 heteroatoms each independently selected from oxygen, nitrogen and sulfur, wherein a carbon atom on said heteroaryl group is optionally substituted with R4aor a nitrogen atom on said heteroaryl group is optionally substituted with R4b;
R4a is hydrogen, CrC4 alkyl, C1-C4 alkoxy, C1-C4 haloalkyl, or halogen, wherein said alkyl is optionally substituted with hydroxyl or CrC4 alkoxy;
R4b is hydrogen, C1-C4 alkyl, -CH2-C1-C3 haloalkyl, -C2-C4 alkyl-OH or -CH2-Ci-C4 alkoxy;
R5 is hydrogen or when R1 is hydrogen then R5 is hydrogen or C1-C4 alkyl;
R6 is C1-C4 alkyl or C3-Cs cycloalkyl wherein one carbon atom of said cycloalkyl moiety may optionally be substituted with methyl or ethyl;
R7 is represented by C1-C4 alkyl, C3-C6 cycloalkyl, NH2, or -(CH2)2-OH;
R8 is represented by hydrogen or C1-C4 alkyl; and
R9 is represented by hydrogen, C1-C4 alkyl, C3-C6cycloalkyl, -(CH2)2-OH, (CH2)2-O-CH3, -(CH2)3-OH, -(CH2)3-O-CH3, 3-oxetanyl, or 3-hydroxycyclobutyl;
or when R3 is -C(O)-NR8R9, R8 and R9 can be taken together with the nitrogen atom to which they are attached to form an azetidine, a pyrrolidine, a piperidine or a 30 morpholine ring.
V
In Step 1, compounds of Formula C can be prepared via a condensation reaction of compounds of Formula A and the commercial compound B (Sigma-Aldrich) in a diverse array of solvents including but not limited to éthanol, toluene and acetonitrile at températures ranging from 22°C to 130°C depending upon the solvent utilized for a period of 1 to 72 hours. In cases where compounds of Formula A are hydrogen chloride or trifluoroacetic acid salts, base modifiers such as sodium acetate or sodium bicarbonate may be added in one to three équivalents to neutralize the salts. The reaction may be conducted in polar protic solvents such as methanol and éthanol at températures ranging from 22°C to 85°C. Typical conditions for this transformation include the use of 3 équivalents of sodium acetate in éthanol heated at 85°C for 3 hours.
Compounds of Formula A can be prepared via a four-step procedure starting with substituted or unsubstituted 4-piperidinone hydrochloride salts (J. Med. Chem. 2004, 47, 2180). First these salts are treated with an appropriate alkyl chloroformate or bis(alkyl) dicarbonate in the presence of excess base to form the corresponding alkyl carbamate. The ketone group is then condensed with tert-butoxycarbonyl hydrazide to form the corresponding W-(tert-butoxy)carbonyl (BOC) protected hydrazone dérivative. This is subsequently reduced to the corresponding BOC protected hydrazine dérivative using reducing agents such as sodium cyanoborohydride or sodium triacetoxyborohydride. Finally, the A/-(tert-butoxy)carbonyl group is cleaved under acidic conditions such as trifluoroacetic acid or hydrochloric acid to give compounds of Formula A, which are typically isolated and used as the corresponding salts (e.g., dihydrochloride sait).
In Step 2, compounds of Formula D may be prepared from compounds of Formula C via the formation of intermediate diazonium salts via the Sandmeyer reaction (Comp. Org. Synth., 1991, 6, 203) These salts may be prepared via diazotization of compounds of Formula C with sodium nitrite and aqueous acids such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric and acetic alone or in combinations. This reaction is typically carried out in water at 0°C to 100°C. Alternatively, anhydrous conditions using alkyl nitrites such as tert-butylnitrite with solvents such as acetonitrile may be utilized (J. Med. Chem. 2006, 49,1562) at températures ranging from 0°C to 95°C. These diazonium intermediates are then allowed to react with copper salts such as copper(ll) bromide, copper(l) bromide or with tribromomethane to form compounds of Formula D. Typical conditions for this transformation include the use of tert-butylnitrite, copper(ll) bromide in acetontrile at 65°C for 30 minutes.
In Step 3, compounds of Formula E may be prepared from compounds of Formula D via the use of reducing agents such as lithium aluminum hydride, sodium
borohydride, lithium borohydride, borane-dimethylsulfide, borane-tetrahydrofuran in polar aprotic solvents such as tetrahydrofuran, diethyl ether, 1,4-dioxane or 1,2dimethoxyethane at températures ranging from 0°C to 110°C for 1 to 24 hours. Typical conditions include the use of borane-dimethylsulfide in tetrahydrofuran at 70°C for 14 hours.
In order to préparé compounds of Formula F from compounds of Formula E, a cyano group must be introduced (Step 4) This may be achieved via a range of conditions. One method of cyano group introduction may be the use of a copper sait such as copper cyanide in a polar aprotic solvent such as N,A/-dimethylformamide (DMF), N-methylpyrrolidinone (NMP), /V,N-dimethylacetamide (DMA) at températures ranging from 22°C to 200°C for 1 to 24 hours. Copper cyanide in N,Ndimethylformamide heated at 165°C for 5 hours is a typical protocol for this transformation.
Alternatively in Step 4, alkali cyanide salts such as potassium or sodium cyanide may be used in conjunction with catalysts such as 18-crown-6 (US2005020564) and or tetrabutylammonium bromide (J. Med. Chem. 2003, 46,1144) in polar aprotic solvents such acetonitrile and dimethylsulfoxide at températures ranging from 22°C to 100°C for the addition of a cyano group to this template.
Finally, the use of métal catalysis is common for the transformation depicted in
Step 4. Common cyanide salts used in catalytic procedures include zinc cyanide, copper cyanide, sodium cyanide, and potassium hexacyanoferrate (II). The métal catalysts can be copper catalysts such as copper iodide and or palladium catalysts such as tris(dibenzylideneacetone)dipalladium (Pd2(dba)3), palladium tetrakistriphenylphosphine (Pd(PPh3)4), or dichloro(diphenyl-phosphinoferrocene)-palladium (Pd(dppf)Cl2). These catalysts may be used alone or in any combination with any of the above cyanide salts. To these reactions may be added ligands such as 1,Tbis(diphenylphosphino)-ferrocene (dppf) or métal additives such as zinc or copper métal. The reactions are carried out in polar aprotic solvents such as NMP, DMF, DMA with or without water as an additive. The reactions are carried out at températures ranging from
22°C to 150°C via conventional or microwave heating for 1 to 48 hours and may be conducted in a sealed or non-sealed reaction vessel. Typical conditions for Step 4 include the use of zinc cyanide, Pd2(dba)3t dppf, and zinc dustin DMA heated at 120°C in a microwave for 1 hour (J. Med. Chem. 2005, 48, 1132).
In Step 5, compounds of Formula G, can be synthesized from compounds of
Formula F via the Mitsunobu reaction. The Mitusunobu reaction has been reviewed in
the synthetic literature (e.g., Chem. Asian. J. 2007, 2,1340; Eur. J. Org. Chem. 2004,
2763; S. Chem. Eur. J. 2004, 10, 3130), and many of the synthetic protocols listed in these reviews may be used. The use of Mitsunobu reaction protocols utilizing azodicarboxylates such as diethyl azodicarboxylate (DEAD), di-tert-butyl azodicarboxylate (TBAD), diisopropyl azodicarboxylate (DIAD) and a phosphine reagent such as triphenylphosphine (PPh3), tributylphoshine (PBU3) and polymer supported triphenylphosphine (PS-PPh3) are combined with compounds of Formula F and a compound of general structure X-OH. Solvents utilized in this reaction may include aprotic solvents such as toluene, benzene, THF, 1,4-dioxane and acetonitrile at températures ranging from 0°C to 130°C depending on the solvent and azodicarboxylates utilized. Typical conditions for this transformation are the use of DEAD with PS-PPh3 in 1,4-dioxane at 22°C for 15 hours.
An alternative to the Mitsunobu reaction for preparing compounds of Formula G is to convert the compounds of Formula F to the corresponding methanesulfonate or para-toluenesulphonate dérivatives using methanesulfonyl chloride or paratoluenesulfonyl chloride, respectively, in the presence of a base such as triethylamine or pyridine. The intermediate sulfonate ester is then combined with a compound of general X-OH, in the presence of a base such as potassium carbonate, sodium hydride, or potassium terf-butoxide to yield compounds of Formula G.
Compounds of Formula K, wherein R1 is C1-C4 alkyl or C3-C6 cycloalkyl, may be prepared from compounds of Formula F in three Steps: 1 ) oxidation of the primary alcohol to the corresponding aldéhyde of Formula H (Step 6, Scheme 1), 2) reaction of the aldéhyde intermediate of Formula H with an organometallic reagent of the Formula R1M, wherein M is lithium (Li) or magnésium halide (MgCI, MgBr or Mgl) to provide a secondary alcohol of Formula J, wherein R1 is C1-C4 alkyl or C3-C6 cycloalkyl (Step 7), and 3) reaction of the secondary alcohol of Formula J with a phénol of the Formula ΧΟΗ under Mitsunobu reaction conditions (Step 8).
In Step 6 (Scheme 1), compounds of Formula H can are formed via oxidation procedures including the use of 1 to 20 équivalents of activated manganèse dioxide in solvents including but not limited to dichloromethane, acetonitrile, hexane or acetone alone or in combinations for 1 to 72 hours at 22°C to 80°C. Alternatively, this oxidation can be conducted with 1 to 3 équivalents of trichloroisocyanuric acid in the presence of 0.1 to 1 équivalents of 2,2,6,6-tetramethylpiperidine-1-oxyl (TEMPO) in dichloromethane or chloroform at températures ranging from 0°C to 22°C for 0.1 to 12 hours. Typical
conditions for this transformation are the use of trichloroisocyanuric acid in the presence of 0.1 équivalent of TEMPO in dichloromethane at 22°C for 1 hour.
The préparation of compounds of this invention wherein Y is NR5 is also shown in Scheme 1. Compounds of Formula L may be prepared from the intermediate compound of Formula H (Scheme 1) by reaction with an amino compound of the Formula X-NH-R5 under reductive amination conditions (Step 9) (J. Org. Chem., 1996, 67, 3849; Org. React. 2002, 59,1). Similarly compounds of Formula N, wherein R1 is CrC4 alkyl or C3-C6 cycloalkyl may be prepared in two steps from the intermediate of Formula J wherein R1 is C1-C4 alkyl or C3-C6 cycloalkyl, by 1) oxidation to the corresponding ketone of Formula M (Step 10), and 2) reaction of the ketone of Formula M with an amino compound of the Formula X-NH-R5 under reductive amination conditions (Step 11). Alternative^ compounds of Formula L and Formula N, wherein R5 is C1-C4 alkyl may be prepared from the corresponding compounds of Formula L, wherein Rs is H, or the corresponding compounds of Formula N, wherein R5 is H, by alkylation with an alkyl halide of Formula (Ci-C4)-CI, (C-i-C4)-Br or (Ci-C4)-I in the presence of a base.
Compounds of this invention may also be prepared as shown in Schemes 2 and 3, wherein X, Z, R1, R2a, R3, R4, R5, R6, R7, R8 and R9 are the same as described in Scheme 1. In particular, compounds of Formula R may be prepared as shown in Scheme 2.
OHC
Z
O
In Step 1 of Scheme 2, compounds of the Formula O can be formed from aldéhydes of Formula H (see also Scheme 1 ) via the use of either dimethyl (diazomethyl)phosphonate or dimethyl-1-diazo-2-oxopropylphosphonate and bases such as potassium carbonate or potassium tert-butoxide in solvents including methanol, éthanol or tetrahydrofuran at températures ranging from -78°C to 22°C for 0.1 to 24 hours. Typical conditions for this transformation include the use of dimethyl-1-diazo-2oxopropylphosphonate and 2 équivalents of potassium carbonate in methanol at 22°C for 0.75 hour.
In Step 2, compounds of Formula Q can be formed from compounds of Formula O via a metal-catalyzed Sonagashira coupling procedure with compounds of general structure X-P wherein P is a halide or trifluoromethsulfonate (triflate). The Sonogashira reaction has been extensively reviewed (Chem. Rev. 2007, 107, 874; Angew. Chem. Int.
Ed. 2007, 46, 834; Angew. Chem. Int. Ed. 2008, 47, 6954), and many of the synthetic protocols listed in these reviews may be used for the synthesis of compounds of Formula Q. Typically, the use of métal catalysts in this reaction can be copper catalysts such as copper iodide and or palladium catalysts such as Pd2(dba)3, Pd(PPh3)4,
Pd(dppf)Cl2 or Pd(PPh3)2CI2. These catalysts may be used alone or in any combination.
Base additives are typically used in this reaction and may include amine bases such as diethylamine, triethylamine, diisopropylethylamine or pyrrolidine or inorganic bases such as potassium carbonate or potassium fluoride. The reactions are carried out in solvents such as dichloromethane, chloroform, acetonitrile, DMF, toluene or 1,4-dioxane with or without water as an additive. The reactions are carried out at températures ranging from 0°C to 150°C depending on the solvent for times ranging from 0.1 to 48 hours. Typical conditions for this transformation include the use of Cul and Pd(PPh3)2Cl2 in DMF at 90°C for 2 hours.
Finally, in Step 3 compounds of Formula R can be formed from compounds of Formula Q via hydrogénation in the presence of transition métal catalysts. Common catalysts include the use of 5 - 20% palladium on carbon or 5 - 20% palladium hydroxide on carbon. These reactions can be conducted in a Parr shaker apparatus or in an H-Cube hydrogénation flow reactor (ThalesNano, U.K.) under pressures of hydrogen ranging from 1 to 50 psi in polar solvents such as tetrahydrofuran, ethyl acetate, methanol or éthanol at températures of 22°C to 50°C for times ranging from 0.1 to 24 hours. Typical conditions for Step 3 include the use compound of Formula Q in ethyl acetate at a flow rate of 1 mL/min through a 10% palladium on carbon cartridge on the H-Cube flow apparatus set at the full hydrogen setting.
Scheme 3 shows methods for the préparation of compounds of Formula W.
Scheme 3
In Step 1 of Scheme 3, compounds of Formula F (see also Scheme 2) can be treated with reagents such as phosphorus tribromide or carbon tetrabromide and triphenylphosphine to give compounds of Formula S. In Step 2, compounds of Formula S are then allowed to react with triphenylphosphine in solvents such as dichloromethane, chloroform, toluene, benzene, tetrahydrofuran (THF) or acetonitrile to give triphenylphosphonium salts of Formula T. The salts of Formula T, are then combined with carbonyl compounds of Formula U in the presence of bases such as nbutyllithium, sodium bis(trimethylsilyl)amide, lithium bis(trimethylsilyl)amide, potassium bis(trimethylsilyl)amide or lithium diisopropylamide in solvents such as THF, diethylether or 1,4-dioxane, to yield alkene compounds of Formula V, which are typically isolated as mixtures of E and Z géométrie isomers (Step 3). This reaction, commonly known as the Wittig olefination reaction, has been reviewed extensively in the literature (Chem. Rev. 1989, 89, 863; Modem Carbonyl Olefination 2004,1-17; Liebigs Ann.Chem. 1997, 1283).
In Step 4, compounds of Formula W are formed from compounds of Formula V via hydrogénation in the presence of transition métal catalysts. Common catalysts include the use of 5 - 20% palladium on carbon or 5 - 20% palladium hydroxide on
carbon. These reactions can be conducted in a similar manner as described for Step 3 of Scheme 2.
Alternatively compounds of Formula W may be prepared from aldéhydes of Formula H via Wittig reaction with triphenylphosphonium salts of Formula AA (Step 5, Scheme 3). As for Step 3, this reaction produces alkene compounds of Formula V, which again are typically isolated as mixtures of E and Z géométrie isomers, and may be converted to compounds of Formula W by hydrogénation. The salts of Formula AA are obtained in a similar manner to that used for preparing salts of Formula T via conversion of the corresponding alcohol to the bromide and subséquent reaction with triphenylphosphine.
Compounds of Formula BB shown below, wherein X, Z, R1 and R2a are as defined in Scheme 1 can be prepared from secondary alcohols of Formula J (see Scheme 2) or ketones of Formula M (see Scheme 2) through reaction sequences similar to those shown in Scheme 3. Conversion of compounds of Formula J to the corresponding bromides, followed by Wittig olefinatîon with aldéhydes of general formula X-CHO provides alkenes of Formula CC. Alkenes of Formula CC may also be obtained via Wittig reaction of ketones of Formula M with salts of the general structure X-CH2-PPh3+Br. The alkenes of Formula CC are then converted to compounds of Formula BB by hydrogénation.
NC
X
R
N BB i
Z
N CC i
Z
In certain instances it is possible to change the order of steps shown in Schemes
1, 2 and 3. For example, in Scheme 1, it is sometimes possible to introduce the cyano group on the pyrazole ring as the last step, i.e., inverting the order in which Steps 4 and 5 are carried out. Also, in certain cases, it is préférable to introduce or modify substituents R3 on the group X later in the synthesis, even as the last step. For example, when R3 is SO2R7, the SO2R7 group may be in formed in the last step by oxidation of the corresponding compound bearing a substituent of general formula S-R7.
Compounds of this invention may be prepared according to sequences analogous to those shown in Schemes 1,2 and 3 starting with 3,3-difluoro-4,4dihydroxy 1 -piperidine carboxylic acid 1,1 -dimethylethyl ester (WO 2008121687). In a manner similar to that described for the préparation of intermediates of formula A in Scheme 1, this material may be converted to hydrazine dervatives of formula DD, which are then used similarly to the intermediates of formula A in Scheme 1.
As is readily apparent to one skilled in the art, protection of remote functionality (e.g., primary or secondary amine) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the préparation methods. Suitable amino-protecting groups (NH-Pg) include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and 9fluorenylmethyleneoxycarbonyl (Fmoc). Similarly, a “hydroxy-protecting group” refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality. Suitable hydroxyl-protecting groups (O-Pg) include for example, allyl, acetyl, silyl, benzyl, para-methoxybenzyl, trityl, and the like. The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Orqanic Synthesis. John Wiley & Sons, New York, 1991.
As noted above, some of the compounds of this invention are acidic and they form salts with pharmaceutically acceptable cations. Some of the compounds of this invention are basic and form salts with pharmaceutically acceptable anions. Ail such salts are within the scope of this invention and they can be prepared by conventional methods such as combining the acidic and basic entities, usually in a stoichiometric ratio, in either an aqueous, non-aqueous or partially aqueous medium, as appropriate. The salts are recovered either by filtration, by précipitation with a non-solvent followed by filtration, by évaporation of the solvent, or, in the case of aqueous solutions, by lyophilization, as appropriate. The compounds are obtained in crystalline form according to procedures known in the art, such as by dissolution in an appropriate solvent(s) such as éthanol, hexanes or water/ethanol mixtures.
Medical Uses
Compounds of the présent invention modulate the activity of G-protein-coupled receptor GPR119. As such, said compounds are useful for the prophylaxis and treatment of diseases, such as diabètes, in which the activity of GPR119 contributes to the pathology or symptoms of the disease. Consequently, another aspect of the présent invention includes a method for the treatment of a metabolic disease and/or a metabolic-related disorder in an individual which comprises administering to the individual in need of such treatment a therapeutically effective amount of a compound of the invention, a sait of said compound or a pharmaceutical composition containing such compound. The metabolic diseases and metabolîsm-related disorders are selected from, but not limited to, hyperlipidemia, Type I diabètes, Type II diabètes mellitus, idiopathic Type I diabètes (Type Ib), latent autoimmune diabètes in adults (LADA), early-onset Type 2 diabètes (EOD), youth-onset atypical diabètes (YOAD), maturity onset diabètes of the young (MODY), malnutrition-related diabètes, gestational diabètes, coronary heart disease, ischémie stroke, restenosis after angioplasty, peripheral vascular disease, intermittent claudication, myocardial infarction (e.g. necrosis and apoptosis), dyslipidemîa, post-prandial lipemia, conditions of impaired glucose tolérance (IGT), conditions of impaired fasting plasma glucose, metabolic acidosis, ketosis, arthritis, obesity, osteoporosis, hypertension, congestive heart failure, left ventricular hypertrophy, peripheral arterial disease, diabetic retinopathy, maculardegeneration, cataract, diabetic nephropathy, glomerulosclerosis, chronic rénal failure, diabetic neuropathy, metabolic syndrome, syndrome X, premenstrual syndrome, coronary heart disease, angina pectoris, thrombosis, atherosclerosis, myocardial infarction, transient ischémie attacks, stroke, vascular restenosis, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertrygliceridemia, insulin résistance, impaired glucose metabolism, conditions of impaired glucose tolérance, conditions of impaired fasting plasma glucose, obesity, erectile dysfunction, skin and connective tissue disorders, foot ulcérations, endothélial dysfunction, hyper apo B lipoproteinemia and impaired vascular compliance. Additionally, the compounds may be used to treat neurological disorders such as Alzheimer's disease, schizophrenia, and impaired cognition. The compounds will also be bénéficiai in gastrointestinal illnesses such as inflammatory bowel disease, ulcerative colitis, Crohn’s disease, irritable bowel syndrome, etc. As noted above the compounds may also be used to stimulate weight loss in obese patients, especially those afflicted with diabètes.
In accordance with the foregoing, the présent invention further provides a method for preventing or ameliorating the symptoms of any of the diseases or disorders described above in a subject in need thereof, which method comprises administering to a subject a therapeutically effective amount of a compound of the présent invention. Further aspects of the invention include the préparation of médicaments for the treating diabètes and its related co-morbidities.
In order to exhibit the therapeutic properties described above, the compounds need to be administered in a quantity sufficient to modulate activation of the G-proteincoupled receptor GPR119. This amount can vary depending upon the particular disease/condition being treated, the severity of the patient's disease/condition, the patient, the particular compound being administered, the route of administration, and the presence of other underlying disease states within the patient, etc. When administered systemically, the compounds typically exhibit their effect at a dosage range of from about 0.1 mg/kg/day to about 100 mg/kg/day for any of the diseases or conditions listed above. Répétitive daily administration may be désirable and will vary according to the conditions outlined above.
The compounds of the présent invention may be administered by a variety of routes. They may be administered orally. The compounds may also be administered parenterally (i.e., subcutaneously, intravenously, intramuscularly, intraperitoneally, or intrathecally), rectally, or topically.
Co-Administration
The compounds of this invention may also be used in conjunction with other pharmaceutical agents for the treatment of the diseases, conditions and/or disorders described herein. Therefore, methods of treatment that include administering compounds of the présent invention in combination with other pharmaceutical agents are also provîded. Suitable pharmaceutical agents that may be used in combination with the compounds of the présent invention include anti-obesity agents (including appetite suppressants), anti-diabetic agents, anti-hyperglycemic agents, liprd lowering agents, and anti-hypertensive agents.
Suitable anti-diabetic agents include an acetyl-CoA carboxylase-2 (ACC-2) inhîbitor, a diacylglycérol O-acyltransferase 1 (DGAT-1) inhibitor, a phosphodiesterase (PDE)-10 inhibitor, a sulfonylurea (e.g., acetohexamide, chlorpropamide, diabinese, glibenclamide, glipizide, glyburide, glimepiride, gliclazide, glipentide, gliquidone, glisolamide, tolazamîde, and tolbutamide), a meglitinide, an a-amylase inhibitor (e.g., tendamistat, trestatin and AL-3688), an a-glucoside hydrolase inhibitor (e.g., acarbose), an a-glucosidase inhibitor (e.g., adiposine, camiglibose, emiglitate, miglitol, voglibose, pradimicin-Q, and salbostatin), a PPARy agonist (e.g., balaglitazone, ciglitazone, darglitazone, englitazone, isaglitazone, pioglitazone, rosiglitazone and troglitazone), a PPAR α/γ agonist (e.g., CLX-0940, GW-1536, GW-1929, GW-2433, KRP-297, L796449, LR-90, MK-0767 and SB-219994), a biguanide (e.g., metformin), a glucagonlike peptide 1 (GLP-1) agonist (e.g., exendin-3 and exendin-4), a protein tyrosine phosphatase-1 B (PTP-1B) inhibitor (e.g., trodusquemine, hyrtiosal extract, and compounds disclosed by Zhang, S., et al., Druq Discoverv Todav, 12(9/10), 373-381 (2007)), SIRT-1 inhibitor (e.g., reservatrol), a dipeptidyl peptidease IV (DPP-IV) inhibitor (e.g., sitagliptin, vildagliptin, alogliptin and saxagliptin), an insulin secreatagogue, a fatty acid oxidation inhibitor, an A2 antagonist, a c-jun amino-terminal kinase (JNK) inhibitor, insulin, an insulin mimetic, a glycogen phosphorylase inhibitor, a VPAC2 receptor agonist, and a SGLT2 inhibitor (sodium dépendent glucose transporter inhibitors such as dapagliflozin, etc). Preferred anti-diabetic agents are metformin and DPP-IV inhibitors (e.g., sitagliptin, vildagliptin, alogliptin and saxagliptin).
Suitable anti-obesity agents include 11β-hydroxy steroid dehydrogenase-1 (11 βHSD type 1) inhibitors, stearoyl-CoA desaturase-1 (SCD-1) inhibitor, MCR-4 agonists, cholecystokinin-A (CCK-A) agonists, monoamine reuptake inhibitors (such as sibutramine), sympathomimetic agents, β3 adrenergic agonists, dopamine agonists (such as bromocriptine), melanocyte-stimulating hormone analogs, 5HT2c agonists, melanin concentrating hormone antagonists, leptin (the OB protein), leptin analogs, leptin agonists, galanin antagonists, lipase inhibitors (such as tetrahydrolipstatin, i.e. orlistat), anorectic agents (such as a bombesin agonist), neuropeptide-Y antagonists (e.g., NPY Y5 antagonists), PYY3-36(including analogs thereof), thyromimetic agents, déhydroépiandrostérone or an analog thereof, glucocorticoid agonists or antagonists, orexin antagonists, glucagon-like peptide-1 agonists, ciliary neurotrophic factors (such as Axokine™ available from Regeneron Pharmaceuticals, Inc., Tarrytown, NY and Procter & Gamble Company, Cincinnati, OH), human agouti-related protein (AGRP) inhibitors, ghrelin antagonists, histamine 3 antagonists or inverse agonists, neuromedin U agonists, MTP/ApoB inhibitors (e.g., gut-selective MTP inhibitors, such as dirlotapide), opioid antagonist, orexin antagonist, and the like.
Preferred anti-obesity agents for use in the combination aspects of the présent invention include gut-selective MTP inhibitors (e.g., dirlotapide, mitratapide and implitapide, R56918 (CAS No. 403987) and CAS No. 913541-47-6), CCKa agonists (e.g., N-benzyl-2-[4-(1 H-indol-3-ylmethyl)-5-oxo-1 -phenyl-4,5-dihydro-2,3,6,10btetraaza-benzo[e]azulen-6-yl]-N-isopropyl-acetamide described in PCT Publication No. WO 2005/116034 or US Publication No. 2005-0267100 A1 ), 5HT2c agonists (e.g., lorcaserin), MCR4 agonist (e.g., compounds described in US 6,818,658), lipase inhibitor (e.g., Cetilistat), PYY3.36(as used herein PYY3.36 includes analogs, such as peglated PYY3-36 e.g., those described in US Publication 2006/0178501), opioid antagonists (e.g., naltrexone), oleoyl-estrone (CAS No. 180003-17-2), obinepitide (TM30338), pramlintide (Symlin®), tesofensine (NS2330), leptin, liraglutide, bromocriptine, orlistat, exenatide (Byetta®), AOD-9604 (CAS No. 221231-10-3) and sibutramine. Preferably, compounds of the présent invention and combination thérapies are administered in conjunction with exercise and a sensible diet.
Ail of the above recited U.S. patents and publications are incorporated herein by reference.
Pharmaceutical Formulations
The présent invention also provides pharmaceutical compositions which comprise a therapeutically effective amount of a compound, or a pharmaceutically acceptable sait thereof, in ad mixture with at least one pharmaceutically acceptable excipient. The compositions include those in a form adapted for oral, topical or parentéral use and can be used for the treatment of diabètes and related conditions as described above.
The composition can be formulated for administration by any route known in the art, such as subdermal, inhalation, oral, topical, parentéral, etc. The compositions may be in any form known in the art, including but not limited to tablets, capsules, powders, granules, lozenges, or liquid préparations, such as oral or stérile parentéral solutions or suspensions.
Tablets and capsules for oral administration may be in unit dose présentation form, and may contain conventional excipients such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrollidone; fillers, for example lactose, sugar, maize-starch, calcium phosphate, sorbitol or glycine; tabletting lubricants, for example magnésium stéarate, talc, polyethylene glycol or silica; disintegrants, for example potato starch; or acceptable wetting agents such as sodium lauryl sulphate.
The tablets may be coated according to methods well known in normal pharmaceutical practice.
Oral liquid préparations may be in the form of, for example, aqueous or oily suspensions, solutions, émulsions, syrups or élixirs, or may be presented as a dry product for reconstitution with water or other suitable vehicle before use. Such liquid préparations may contain conventional additives, such as suspending agents, for example sorbitol, methyl cellulose, glucose syrup, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminium stéarate gel or hydrogenated edible fats, emulsifying agents, for example lecithin, sorbitan monooleate, or acacia; non-aqueous vehicles (which may include edible oils), for example almond oil, oily esters such as glycerin, propylene glycol, or ethyl alcohol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid, and, if desired, conventional flavoring or coloring agents.
For parentéral administration, fluid unit dosage forms are prepared utilizing the compound and a stérile vehicle, water being preferred. The compound, depending on the vehicle and concentration used, can be either suspended or dissolved in the vehicle or other suitable solvent. In preparing solutions, the compound can be dissolved in water for injection and filter sterilized before filling into a suitable vial or ampoule and sealîng. Advantageously, agents such as local anesthetics, preservatives and buffering agents etc. can be dissolved in the vehicle. To enhance the stability, the composition can be frozen after filling into the vial and the water removed under vacuum. The dry lyophilized powder is then sealed in the vial and an accompanying vial of water for injection may be supplied to reconstitute the liquid prior to use. Parentéral suspensions are prepared in substantially the same manner except that the compound is suspended in the vehicle instead of being dissolved and sterilization cannot be accomplished by filtration. The compound can be sterilized by exposure to ethylene oxide before suspending in the stérile vehicle. Advantageously, a surfactant or wetting agent is included in the composition to facilitate uniform distribution of the compound.
The compositions may contain, for example, from about 0.1% to about 99 by weight, of the active material, depending on the method of administration. Where the compositions comprise dosage units, each unit will contain, for example, from about 0.1 to 900 mg of the active ingrédient, more typically from 1 mg to 250mg.
Compounds of the invention can be formulated for administration in any convenient way for use in human or veterinary medicine, by analogy with other antidiabetic agents. Such methods are known in the art and hâve been summarized above.
For a more detailed discussion regarding the préparation of such formulations; the reader's attention îs directed to Reminqtorï's Pharmaceutical Sciences, 21s Edition, by University of the Sciences in Philadelphia.
Embodiments of the présent invention are illustrated by the following Examples. It is to be understood, however, that the embodiments of the invention are not limited to the spécifie details of these Examples, as other variations thereof will be known, or apparent in light of the instant disclosure, to one of ordinary skill in the art.
EXAMPLES
Unless specified otherwise, starting materials are generally available from commercial sources such as Aldrich Chemicals Co. (Milwaukee, Wl), Lancaster Synthesis, Inc. (Windham, NH), Acros Organics (Fairlawn, NJ), Maybridge Chemical Company, Ltd. (Cornwall, England), Tyger Scientifîc (Princeton, NJ), and AstraZeneca Pharmaceuticals (London, England), Mallinckrodt Baker (Phillipsburg NJ); EMD (Gibbstown, NJ).
General Experimental Procedures
NMR spectra were recorded on a Varian Unity™ 400 (DG400-5 probe) or 500 (DG500-5 probe - both available from Varian Inc., Palo Alto, CA) at room température at 400 MHz or 500 MHz respectively for proton analysis. Chemical shifts are expressed in parts per million (delta) relative to residual solvent as an internai reference. The peak shapes are denoted as follows: s, singlet; d, doublet; dd, doublet of doublet; t, triplet; q, quartet; m, multiplet; bs, broad singlet; 2s, two singlets.
Atmospheric pressure chemical ionization mass spectra (APCI) were obtained on a Waters™ Spectrometer (Micromass ZMD, carrier gas: nitrogen) (available from Waters Corp., Milford, MA, USA) with a flow rate of 0.3 mL/minute and utilizing a 50:50 water/acetonitrile eluent System. Electrospray ionization mass spectra (ES) were obtained on a liquid chromatography mass spectrometer from Waters™ (Micromass ZQ or ZMD instrument (carrier gas: nitrogen) (Waters Corp., Milford, MA, USA) utilizing a gradient of 95:5 - 0:100 water in acetonitrile with 0.01% formic acid added to each solvent. These instruments utilized a Varian Polaris 5 C18-A20x2.0mm column (Varian Inc., Palo Alto, CA) at flow rates of 1 mL/minute for 3.75 minutes or 2 mL/minute for 1.95 minutes.
Column chromatography was performed using silica gel with either Flash 40 Biotage™ columns (ISC, Inc., Shelton, CT) or Biotage™ SNAP cartridge KPsil or
Redisep Rf silica (from Teledyne Isco Inc) under nitrogen pressure. Préparative HPLC was performed using a Waters FractionLynx System with photodiode array (Waters
2996) and mass spectrometer (Waters/Micromass ZQ) détection schemes. Analytical
HPLC work was conducted with a Waters 2795 Alliance HPLC or a Waters ACQUITY
UPLC with photodiode array, single quadrupole mass and evaporative light scattering détection schemes.
Concentration in vacuo refers to évaporation of solvent under reduced pressure using a rotary evaporator.
Unless otherwise noted, chemical reactions were performed at room température (about 23 degrees Celsius). Also, unless otherwise noted chemical reactions were run under an atmosphère of nitrogen.
PHARMACOLOGICAL DATA
The practice of the invention for the treatment of diseases modulated by the agonist activation of G-protein-coupled receptor GPR119 with compounds of the invention can be evidenced by activity in one or more of the functional assays described herein below, The source of supply is provided in parenthesis.
In-Vitro Functional Assays β-lactamase:
The assay for GPR119 agonists utilizes a cell-based (hGPR119 HEK293-CRE beta-lactamase) reporter construct where agonist activation of human GPR119 is coupled to beta-lactamase production via a cyclic AMP response element (CRE). GPR119 activity is then measured utilizing a FRET-enabled beta-lactamase substrate, CCF4-AM (Live Blazer FRET-B/G Loading kit, Invitrogen cat # K1027). Specifically, hGPR119-HEK-CRE- beta-lactamase cells (Invitrogen 2.5 x 107/ml_) were removed from liquid nitrogen storage, and diluted in plating medium (Dulbecco's modified Eagle medium high glucose (DMEM; Gibco Cat # 11995-065), 10% heat inactivated fêtai bovine sérum (HIFBS; Sigma Cat# F4135), 1X MEM Nonessential amino acids (Gibco Cat # 15630-080), 25 mM HEPES pH 7.0 (Gibco Cat # 15630-080), 200 nM potassium clavulanate (Sigma Cat # P3494). The cell concentration was adjusted using cell plating medium and 50 microL of this cell suspension (12.5 x 104 viable cells) was added into each well of a black, clear bottom, poly-d-lysine coated 384-well plate (Greiner Bio-One cat# 781946) and incubated at 37 degrees Celsius in a humidified environment containing 5% carbon dioxide. After 4 hours the plating medium was removed and replaced with 40 microL of assay medium (Assay medium is plating medium without potassium clavulanate and HIFBS). Varying concentrations of each compound to be tested was then added in a volume of 10 microL (final DMSO = 0.5%) and the cells were incubated for 16 hours at 37 degrees Celsius in a humidified environment containing 5% carbon dioxide. Plates were removed from the incubator and allowed to equilibrate to room température for approximately 15 minutes. 10 microL of 6 X CCF4/AM working dye solution (prepared according to instructions in the Live Blazer FRET-B/G Loading kit, Invitrogen cat # K1027) was added per well and incubated at room température for 2 hours in the dark. Fluorescence was measured on an EnVision fluorimetric plate reader, excitation 405 nm, émission 460 nm/535 nm. EC50 déterminations were made from agonist-response curves analyzed with a curve fitting program using a 4-parameter logistic dose-response équation.
cAMP:
GPR119 agonist activity was also determined with a cell-based assay utilizing an
HTRF (Homogeneous Time-Resolved Fluorescence) cAMP détection kit (cAMP dynamic 2 Assay Kit; Cis Bio cat # 62AM4PEC) that measures cAMP levels in the cell.
The method is a compétitive immunoassay between native cAMP produced by the cells and the cAMP labeled with the dye d2. The tracer binding is visualized by a Mab anticAMP labeled with Cryptate. The spécifie signal (i.e. energy transfer) is inversely proportional to the concentration of cAMP in either standard or sample.
Specifically, hGPR119 HEK-CRE beta-lactamase cells (Invitrogen 2.5 x 107/ml_; the same cell line used in the beta-lactamase assay described above) were removed from cryopreservation and diluted in growth medium (Dulbecco’s modified Eagle medium high glucose (DMEM; Gibco Cat# 11995-065), 1% charcoal dextran treated fêtai bovine sérum (CD sérum; HyClone Cat # SH30068.03), 1x MEM Nonessential amino acids (Gibco Cat# 15630-080) and 25 mM HEPES pH 7.0 (Gibco Cat # 15630080)). The cell concentration was adjusted to 1.5 x 105 cells/mL and 30 ml_s of this suspension was added to a T-175 flask and incubated at 37 degrees Celsius in a humidified environment in 5% carbon dioxide. After 16 hours (overnight), the cells were removed from the T-175 flask (by rapping the side of the flask), centrifuged at 800 x g and then re-suspended in assay medium (1x HBSS +CaCI2 + MgCI2 (Gibco Cat # 14025-092) and 25 mM HEPES pH 7.0 (Gibco Cat # 15630-080)). The cell concentration was adjusted to 6.25 x 105 cells/mL with assay medium and 8 pl of this cell suspension (5000 cells) was added to each well of a white Greiner 384-well, lowvolume assay plate (VWR cat # 82051-458).
Varying concentrations of each compound to be tested were diluted in assay buffer containing 3-isobutyl-1-methylxanthin (IBMX; Sigma cat # Ι5Θ79) and added to the assay plate wells in a volume of 2 microL (final IBMX concentration was 400 microM and final DMSO concentration was 0.58%). Following 30 minutes incubation at room température, 5 microL of labeled d2 cAMP and 5 microL of anti-cAMP antibody (both diluted 1:20 in cell lysis buffer; as described in the manufacturers assay protocol) were added to each well of the assay plate. The plates were then incubated at room température and after 60 minutes, changes in the HTRF signal were read with an Envision 2104 multilabel plate reader using excitation of 330 nm and émissions of 615 and 665 nm. Raw data were converted to nM cAMP by interpolation from a cAMP standard curve (as described in the manufacturées assay protocol) and EC50 déterminations were made from an agonist-response curves analyzed with a curve fitting program using a 4-paramter logistiedose response équation.
It is recognized that cAMP responses due to activation of GPR119 could be generated in cells other than the spécifie cell line used herein.
β-Arrestin:
GPR119 agonist actrvity was also determined with a cell-based assay utilizing DiscoverX PathHunter β-arrestin cell assay technology and their U20S hGPR119 β-arrestin cell line (DiscoverX Cat # 93-0356C3). In this assay, agonist activation is determined by measuring agonist-induced interaction of β-arrestin with activated GPR119. A small, 42 amino acid enzyme fragment, called ProLink was appended to the C-terminus of GPR119. Arrestin was fused to the larger enzyme fragment, termed EA (Enzyme Acceptor). Activation of GPR119 stimulâtes binding of arrestin and forces the complémentation of the two enzyme fragments, resulting in formation of a functional β-galactosidase enzyme capable of hydrolyzing substrate and generating a chemiluminescent signal.
Specifically, U2OS hGPR119 β-arrestin cells (DiscoverX 1 x 107/mL) were removed from cryopreservation and diluted in growth medium (Minimum essential medium (MEM; Gibco Cat# 11095-080), 10% heat inactivatedfêtai bovine sérum 15 (HIFBS; Sigma Cat # F4135-100), 100 mM sodium pyruvate (Sigma Cat # S8636), 500 microg/mL G418 (Sigma Cat #G8168) and 250 microg/mL Hygromycin B (Invitrogen Cat # 10687-010). The cell concentration was adjusted to 1.66 x 105 cells/mL and 30 ml_s of this suspension was added to a T-175 flask and incubated at 37 degrees Celsius in a humidified environment in 5% carbon dioxide. After 48 hours, the cells were removed from the T-175 flask with enzyme-free cell dissociation buffer (Gibco cat # 13151-014), centrifuged at 800 x g and then re-suspended in plating medium (OptiMEM I (Invitrogen/BRL Cat # 31985-070) and 2 % charcoal dextran treated fêtai bovine sérum (CD sérum; HyClone Cat # SH30068.03). The cell concentration was adjusted to
2.5 x 105 cells/mL with plating medium and 10 microL of this cell suspension (2500 cells) was added to each well of a white Greiner 384-well low volume assay plate (VWR cat # 82051-458) and the plates were incubated at 37 degrees Celsius in a humidified environment in 5% carbon dioxide.
After 16 hours (overnight) the assay plates were removed from the incubator and varying concentrations of each compound to be tested (diluted in assay buffer (1x
HBSS +CaCI2 + MgCI2 (Gibco Cat # 14025-092), 20 mM HEPES pH 7.0 (Gibco Cat # 15630-080) and 0.1% BSA (Sigma Cat # A9576)) were added to the assay plate wells in a volume of 2.5 microL (final DMSO concentration was 0.5 %). After a 90 minute incubation at 37degrees Celsius in a humidified environment in 5% carbon dioxide, 7.5 microL of Galacton Star β-galactosidase substrate (PathHunter Détection Kit (DiscoveRx Cat # 93-0001 ); prepared as described in the manufacturers assay protocol) was added to each well of the assay plate. The plates were incubated at room température and after 60 minutes, changes in the luminescence were read with an Envision 2104 multilabel plate reader at 0.1 seconds per well. EC50 déterminations were made from an agonist-response curves analyzed with a curve fitting program using a 4-parameter logistic dose response équation.
Expression of GPR119 Using BacMam and GPR119 Binding Assay
Wild-type human GPR119 (published in PCT patent publication no. 2010/106457) was amplified via polymerase chain reaction (PCR) (Pfu Turbo Mater Mix, Stratagene, La Jolla, CA) using plRES-puro-hGPR119 as a template and the following primers: hGPR119 BamH1, Upper
5-TAAATTGGATCCACCATGGAATCATCTTTCTCATTTGGAG-3’ (inserts a BamHI site at the 5’ end) hGPR119 EcoRI, Lower 5'-TAAATTGAATTCTTATCAGCCATCAAACTCTGAGC-3’ (inserts a EcoRI site at the 3' end)
The amplified product was purified (Qiaquîck Kit, Qiagen, Valencia, CA) and digested with BamHI and EcoRI (New England BioLabs, Ipswich, MA) according to the manufacturées protocols. The vector pFB-VSVG-CMV-poly (published in PCT patent publication no. 2010/106457) was digested with BamHI and EcoRI (New England BioLabs, Ipswich, MA). The digested DNA was separated by electrophoresis on a 1% agarose gel; the fragments were excised from the gel and purified (Qiaquîck Kit, Qiagen, Valencia, CA). The vector and gene fragments were ligated (Rapid Ligase Kit, Roche, Pleasanton, CA) and transformed into OneShot DH5alpha T1R cells (Invitrogen, Carlsbad, CA). Eight ampicillin-resistant colonies (clones 1-8) were grown for miniprep (Qiagen Miniprep Kit, Qiagen, Valencia, CA) and sequenced to confirm identity and correct insert orientation.
The pFB-VSVG-CMV-poly-hGPR119 construct (clone #1 ) was transformed into OneShot DHIOBac cells (Invitrogen, Carlsbad, CA) according to manufacturers’ protocols. Eight positive (i.e. white) colonies were re-streaked to confirm as ‘'positives and subsequently grown for bacmid isolation. The recombinant hGPR119 bacmid was isolated via a modified Alkaline Lysis procedure using the buffers from a Qiagen
Miniprep Kit (Qiagen, Valencia, CA). Briefly, pelleted cells were lysed in buffer P1, neutralized in buffer P2, and precipitated with buffer N3. Precipitate was pelleted via centrifugation (17,900xg for 10 minutes) and the supernatant was combined with isopropanol to precipitate the DNA. The DNA was pelleted via centrifugation (17,900xg for 30 minutes), washed once with 70% éthanol, and resuspended in 50 pL buffer EB (Tris-HCL, pH 8.5). Polymerase chain reaction (PCR) with commercially available primers (M13F, M13R, Invitrogen, Carlsbad, CA) was used to confirm the presence of the hGPR119 insert in the Bacmid.
Génération of hGPR119 Recombinant Baculovirus
Création of PO Virus Stock
Suspension adapted Sf9 cells grown in Sf900lI medium (Invitrogen, Carlsbad, CA) were transfected with 10 microL hGPR119 bacmid DNA according to the manufacturées protocol (Cellfectin, Invitrogen, Carlsbad, CA). After five days of incubation, the conditioned medium (i.e. “PO virus stock) was centrifuged and filtered through a 0.22 pm filter (Steriflip, Millipore, Billerica, MA).
Création of Frozen Virus (BIIC) Stocks
For long term virus storage and génération of working (i.e. “P1) viral stocks, frozen BIIC (Baculovirus Infected Insect Cells) stocks were created as follows: suspension adapted Sf9 cells were grown in Sf900ll medium (Invitrogen, Carlsbad, CA) and infected with hGPR119 PO virus stock. After 24 hours of growth, the infected cells were gently centrifuged (approximately 100 x g), resuspended in Freezing Medium (10% DMSO, 1% Albumin in Sf900lI medium) to a final density of 1 x 107 cells/mL and frozen according to standard freezing protocole in 1 mL aliquots.
Création of Workinq (PI) Virus Stock
Suspension adapted Sf9 cells grown in Sf900lI medium (Invitrogen, Carlsbad, CA) were infected with a 1:100 dilution of a thawed hGPR119 BIIC stock and incubated for several days (27 degrees Celsius with shaking). When the viability of the cells reached 70%, the conditioned medium was harvested by centrifugation and the virus titer determined by ELISA (BaculoElisa Kit, Clontech, Mountain View, CA)
Qver-expression of hGPR119 in Suspensîon-Adapted HEK 293FT Cells HEK 293FT cells (Invitrogen, Carlsbad, CA) were grown in a shake flask in 293Freestyle medium (Invitrogen) supplemented with 50 microg/mL neomycin and 10mM HEPES (37C, 8% carbon dioxide, shaking). The cells were centrifuged gently (approximately 500xg, 10 minutes) and the pellet resuspended in a mixture of Dulbecco’s PBS(minus Mg++/-Ca++) supplemented with 18% fêtai bovine sérum (Sigma Aldrich) and P1 virus such that the multiplicity of infection (MOI) was 10 and the final cell density was 1.3 x 10e/mL (total volume 2.5 liters). The cells were transferred to a 5 liter Wave Bioreactor Wavebag (Wave Technologies, MA) and incubated for 4 hours at 27 degrees Celsius (17 rocks/min, 7 degrees platform angle); at the end of the incubation period, an equal volume(2.5 liters) of 293Freestyle medium supplemented with 30mM sodium butyrate (Sigma Aldrich) was added (final concentration = 15 mM), and the cells were grown for 20 hours (37 degrees Celsius, 8% CO2 [0.2 liters/min}, 25 rocks/ minute, 7 degrees platform angle). Cells were harvested via centrifugation (3,000xg, 10 minutes), washed once on DPBS (minus Ca++/Mg++), resuspended in 0.25M sucrose, 25mM HEPES, 0.5mM EDTA, pH 7.4 and frozen at -80 degrees Celsius.
Membrane Préparation for Radioliqand Binding Assays
The frozen cells were thawed on ice and centrifuged at 700 x g (1400 rpm) for 10 minutes at 4 degrees Celsius. The cell pellet was resuspended in 20 mL phosphatebuffered saline, and centrifuged at 1400 rpm for 10 minutes. The cell pellet was then resuspended in homogenization buffer (10 mM HEPES (Gibco #15630), pH 7.5, 1 mM EDTA (BioSolutions, #BIO260-15), 1 mM EGTA (Sigma, #E-4378), 0.01 mg/mL benzamidine (Sigma #B 6506), 0.01 mg/mL bacitracin (Sigma #B 0125), 0.005 mg/mL leupeptin (Sigma #L 8511 ), 0,005 mg/mL aprotinin (Sigma #A 1153)) and incubated on ice for 10 minutes. Cells were then lysed with 15 gentle strokes of a tight-fitting glass Dounce homogenizer. The homogenate was centrifuged at 1000 x g (2200 rpm) for 10 minutes at 4 degrees Celsius. The supernatant was transferred into fresh centrifuge
tubes on ice. The cell pellet was resuspended in homogenization buffer, and centrifuged again at 1000 x g (2200 rpm)for 10 minutes at 4 degrees Celsius after which the supernatant was removed and the pellet resuspended in homogenization buffer. This process was repeated a third time, after which the supernatants were combined, Benzonase (Novagen # 71206) and MgCl2 (Fluka #63020) were added to final concentrations of 1 U/mL and 6 mM, respectively, and incubated on ice for one hour. The solution was then centrifuged at 25,000 x g (15000 rpm) for 20 minutes at 4 degrees Celsius, the supernatant was discarded, and the pellet was resuspended in fresh homogenization buffer (minus Benzonase and MgCh). After repeating the 25,000 x g centrifugation step, the final membrane pellet was resuspended in homogenization buffer and frozen at -80 degrees Celsius. The protein concentration was determined using the Pierce BCA protein assay kit (Pierce reagents A #23223 and B #23224).
Synthesis and Purification of [3H1-Compound A
CH2Cl2
Compound A ( isopropyl 4-(1-(4-(methylsulfonyl)phenyl)-3a,7a-dihydro-1H-pyrazolo[3,4d]pyrimidin-4-yloxy)piperidine-1-carboxylate, as shown above) (4 mg, 0.009 mmol) was dissolved in 0.5 mL of dichloromethane, and the resulting solution was treated with (1,5cyclooctadiene)(pyridine)(tricyclohexylphosphine)-iridium(l) hexaflurophosphate (J. Organometal. Chem. 1979, 168, 183) (5 mg, 0.006 mmol). The reaction vessel was sealed and the solution was stirred under an atmosphère of tritium gas for 17 hours. The reaction solvent was removed under reduced pressure and the resulting residue was dissolved in éthanol. Purification of crude [3H]-Compound A was performed by préparative HPLC using the following conditions.
Column: Atlantis, 4.6 x 150mm, 5pm
Mobil Phase A: water / acetonitrile / formic acid (98 / 2 / 0.1 )
Mobil Phase B: acetonitrile
5 Gradient: Time %B
0.00 30.0
1.00 30.0
13.00 80.0
Run time: 16 min
10 Post time: 5 min
Flow Rate: 1,5 mL/minute
Inj. Volume: 20-50 pl_
Inj. Solvent: DMSO
Détection: UV at 210 nm and 245 nm
The spécifie activity of purified [3H]-Compound A was determined by mass spectroscopy to be 70 Ci/mmol.
Alternatively the binding assay can be performed with [3H]-Compound B.
Synthesis and Purification of f3Hl-Compound B
tritium gas
(Crabtree's catalyst)
CH2Ct2
Compound B (tert-butyl 4-(1-(4-(methylsulfonyl)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-4yloxyjpiperîdine-l-carboxylate, as shown above)(5 mg, 10.6 pmol) was dissolved in 1.0 mL of dichloromethane and the resulting solution was treated with Crabtree's catalyst (5 mg, 6.2 μιηοΙ). The reaction vessel was sealed and the solution was stirred under an atmosphère of tritium gas for 17 hours. The reaction solvent was removed under reduced pressure and the resulting residue was dissolved in éthanol. Purification of crude [3H]-Compound B was performed by silica gel flash column chromatography eluting with 70% hexanes / 30% ethyl acetate, followed by silica gel flash column chromatography eluting with 60% petroleum ether / 40% ethyl acetate.
The spécifie activity of purified [3H]-Compound B was determined by mass spectroscopy 15 to be 57.8 Ci/mmol.
GPR119 Radioligand Binding Assay
Test compounds were serially diluted in 100% DMSO (J.T. Baker #922401). 2 20 microL of each dilution was added to appropriate wells of a 96-well plate (each concentration in triplicate). Unlabeled Compound A (or Compound B), at a final concentration of 10 microM, was used to détermine non-specific binding. [3H]-Compound A (or [3H]-Compound B) was diluted in binding buffer (50 mM Tris-HCI, pH 7.5, (Sigma #T7443), 10 mM MgCI2 (Fluka 63020), 1 mM EDTA (BioSolutions #BIO260-15), 0.15% bovine sérum albumin (Sigma #A7511 ), 0.01 mg/mL benzamidine (Sigma #B 6506), 0.01 mg/mL bacitracin (Sigma #B 0125), 0.005 mg/mL
V leupeptin (Sigma #L 8511), 0.005 mg/mL aprotinin (Sigma #A 1153)) to a concentration of 60 nM, and 100 microL added to ail wells of 96-well plate (Nalge Nunc # 267245). Membranes expressing GPR119 were thawed and diluted to a final concentration of 20 pg/100 microL per well in Binding Buffer, and 100 microL of diluted membranes were added to each well of 96-well plate.
The plate was incubated for 60 minutes w/shaking at room température (approximately 25 degrees Celsius). The assay was terminated by vacuum filtration onto GF/C filter plates (Packard # 6005174) presoaked in 0.3% polyethylenamine, using a Packard harvester. Filters were then washed six times using washing buffer (50 mM Tris-HCI, pH 7.5 kept at 4 degrees Celsius). The filter plates were then air-dyed at room température overnight. 30 μΙ of scintillation fluid (Ready Safe, Beckman Coulter #141349) was added to each well, plates were sealed, and radioactivity associated with each filter was measured using a Wallac Trilux MicroBeta, plate-based scintillation counter.
The Kd for [3H]-Compound A (or [3H]-Compound B) was determined by carrying out saturation binding, with data analysis by non-linear régression, fit to a one-site hyperbola (Graph Pad Prism). IC50 déterminations were made from compétition curves, analyzed with a proprietary curve fitting program (SIGHTS) and a 4-parameter logistic dose response équation. Ki values were calculated from IC50 values, using the ChengPrusoff équation.
The following results were obtained for the Beta-lactamase, Beta-arrestin, cAMP, and binding assays:
Example Number Human Blactamase Functional EC50 (nM) Intrinsic Activity* (%) Human cAMP Functional EC50 (nM) Intrinsic Activity* (%) Human Barrestin Functional EC50 (nM) Intrinsic Activity* (%) Human Binding Ki(nM)
11 782 98.6 135 65.7 171 51 530
12 5200 100 10000 1700
13 142 91.3 217 119 149
14 464 75.7 495 109 723
15 135 120 83 89.2 134
16 220 108 445 93.7
17 31.5 93 161 67.4
18 68 77.3 56.7 71.2 153
19 4390 100 4510 99.5 6100
20 89.4 104 72.9 115 478 63.4 76.5
21 372 98.1 43.2 32.6 86.4
22 154 29 278 52.8 159
23 1550 100 150
24 141 103 43.6 124 62.9 107 99
25 79.5 101 192 92.8 418
26 217 60.1 264 73.9 270
27 2520 101 394 27 1050
28 3.57 24.6 10000 6100
29 3010 100 2550 33.2 2290
30 151 94.6 821 64.1 556 54.7 2020
31 54.1 108 155 91.8 462
32 281 86.5 62.6 68 206 81.8 514
33 258 88.4 1330 100
34 270 102 62.3 86 85 96.5 87.4
35 97.3 91.7 24.9 66.4 50.6 49 29.8
36 848 100 268 63.2 240 57.1 870
37 514 109 1160 55.9 416
38 22.1 122 15 99.8 29.8
39 10.7 76.8 5.32 86.1 14.9
40 10000 10000 1210
41 262 24.1 250 44.1 349
42 120 84.3 147 86.1 223
43 2.78 75.5 5.36 82.5 9.25
44 2.13 79.8 45
45 47.1 51.8 102 58.2 352
46 30.2 102 13.5 91.6 44.8
47 52.7 130 32.8 106 150
48 51.4 113 71.7 111 200
49 24 66.3 4.11 113 18.5
50 19.3 120 30.2 97.6 36.5
51 40.7 56.1 142
52 10000 1440
53 194 83.8 668
54 233 115 594
55 456 22.5 361 46.5 939
56 245 36.7 164 53.4
57 2230 93.4 1520 84.5 4070
58 10000 6580
59 2070 96 1460 100 1380
60 7.4 77 18
61 156 47
62 152 104 393
63 226 31 509
64 6820 100** 4230
65 1050 88 187
66 2760 100** 226
67 52 70 53
68 10000
I Values are reported as the géométrie mean
*The intrinsic activity is the percent of maximal activity of the test compound, relative to the activity of a standard GPR119 agonist, 4-[[6-[(2-fluoro-4 methylsulfonylphenyl) amino]pyrimidin-4-yl]oxy]piperidine-1-carboxylic acid isopropyl ester (W02005121121 ), or (S)-l-methylcyclopropyl 4-(1-fluoro-2-(2-(2,3,6trifluorophenyl)acetamido)ethyl)piperidine-1-carboxylate (see Figure below), at a final concentration of 10 micromolar.
**the curve was extrapolated to 100% to calculate an EC50,
Please note blank entry spaces dénoté that the test was not performed for that 10 Example.
r
Structure of (SM-methylcyclopropyl 4-(1-fluoro-2-(2-(2,3,6-trifluorophenvl)acetamido)ethyl)piperidine-1-carboxvlate***
***Presented at the SMASH workshop NMR, It's Not Just For Structures: Détermination of Physicochemical Properties in Portland, Oregon on Tuesday September 28, 2010 ‘The Gauche effect: Using Conformational Restriction of a Ethyl Amide Sériés to Improve the Physical Properties of Analogues’ by Kathleen Farley.
In Vivo Data
Ail in vivo protocols were approved by the Pfizer's Animal Welfare Committee. Naïve male Wistar rats (200-250 g body weight on receipt) were obtained from Harlan Laboratories (Indianapolîs, IN), were pair housed in hanging plastic caging on Sanichips sawdust bedding, and fed ad libitum on Purina 5001 chow. The rats were housed under a controlled light cycle (light from 6 am to 6 pm) at controlled température and humidity conditions. Rats were acclimated to the facility for at least 1 week prior to study,
Compound préparation
Example 50 was formulated as a 10% SDD in the vehicle 20 mM Tris Buffer at pH 7.4 with 0.5% methylcellulose and 0.5% HPMCAS-HF. The dose (75 mg/kg) was formulated at 15 mg/mL for administration at 5 mL/kg, the required bulk was added to a mortar and ground with a small amount of vehicle to a smooth paste with a pestle, additional vehicle was added until the mixture flowed, when it was transferred to a stirred container, the mortar was rînsed several times with remaining quantity of vehicle and capped to prevent évaporation. The compound was formulated on the day of doing and was stirred continuously with a magnetic stir bar prior to, and during the dosing procedure.
Oral glucose tolérance test (OGTT) protocol
Rats were stratified (n=8/group) to one of four dose groups 90 min or 30 min preglucose vehicle (20 mM Tris Buffer at pH 7.4 with 0.5% methylcellulose and 0.5% Hydroxypropy! methylcellulose acetate succinate- high grade, fine particle (HPMCASHF), or 90 min or 30 min pre-glucose 75 mg/kg Example 50. Stratification was performed according to body weight on day -1 to ensure that each group had equal group mean body weight values. The rats were fasted overnight in clean cages (-15 hours) prior to the oral glucose tolérance test. Body weights were recorded on the morning of the study (post fasting) for dose volume calculation. Blood samples were collected via the tail vein from ail rats prior to dosing with vehicle or test compound via oral gavage (5 mL/kg). Ninety or thirty minutes later rats were bled and immediately dose with an oral dose of glucose (2 g/kg). The rats were re-bled at 15, 30, 60 and 120 minutes post-glucose load. Blood samples (—250 microliter/time point) were collected into EDTA tubes with aprotinin/DPPIVi (0.6 TIU/20 microliter per mL whole blood). Blood tubes were inverted several fîmes immediately foilowing collection and placed on ice, then spun at 14,000 rpm in a refrigerated centrifuge for 5 minutes. Plasma samples were analyzed for glucose levels using a Roche c311 clinical chemistry analyzer, plasma insulin concentrations were determined using the Alpco Ultra-Sensitive Insulin Rat ELISA, and total amide GLP-1 concentrations were determined using MSD ELISA kit.
The results are presented as mean +/- SEM (standard error of the mean) unless otherwise stated. Statistical évaluation of the data was carried out using one-way analysis of variance (ANOVA) with appropriate post-hoc analysis between timematched vehicle and treatment groups. Différences compared to vehicle with a P<0.05 were considered statistically signîficant using Unadjusted T-test.
Table 1: Effect of Example 50 during OGTT
Dose Time Dose (Example 50) Glucose 0-120 min AUC (percent vehicle response) Insulin 0-60 min AUC (percent vehicle response) Total Amide GLP-1 0-120 min AUC (percent vehicle response)
90 min pre 75 mg/kg ++
glucose 90 82 103
30 min pre 75 mg/kg ++
glucose 89 94 153
++ p < 0.01 compared to time-matched vehicle
Préparation of Starting Matériels
Préparation 1: Isopropyl 4-hydrazinopiperidine-1-carboxylate dihydrochloride sait
H2N—N—\
Isopropyl 4-{2-(tert-butoxycarbonyl)hydrazinyl}piperidine-1-carboxylate (obtained as described in W02008137436) (20.2 g, 67.02 mmol), was dissolved in absolute éthanol (250 mL), and the solution was stirred under nitrogen at room température.
Concentrated aqueous hydrochloric acid (27.9 mL, 335 mmol) was added slowly. The solution was stirred under nitrogen at room température for 4 hours. The reaction was concentrated to a white solid that contained some starting material. The solid was treated with a 4 M solution of hydrogen chloride in 1,4-dioxane (100 mL, 400 mmol) and the resulting mixture was stirred for 14 hours at room température. The reaction was then concentrated under reduced pressure to give a white solid, which was treated with heptane (100 mL) and concentrated again to yield the title compound as a white solid (15 g, 81%). 1H NMR (400MHz, methanol-d4) delta 4.9 (m, 1 H), 4.1 (m, 2 H), 3.2 (m, 1 H), 2.9 (m, 2 H), 2.0 (m, 2H), 1.4 (m, 2H), 1.2, (d, 6 H); LCMS (ES+): 202 (M+1 ).
Préparation 2: Isopropyl 4-[5-amino-4-(ethoxycarbonyl)-1 H-pyrazol-1-yll-piperidine-1carboxylate
A mixture of isopropyl 4-hydrazinopiperidine-1-carboxylate dihydrochloride sait (7.08 g,
25.8 mmol), ethyl 2-cyano-3-ethoxyacrylate (4.81 g, 28.4 mmol), sodium acetate (6.49 g,
77.5 mmol), and éthanol (80 mL) was stirred at 85 °C for 3 hours. The mixture was concentrated to about a third of the initial volume. Water (50 mL), saturated sodium bicarbonate (50 mL), and brine (50 mL) were added. The resulting mixture was extracted with ethyl acetate (2 x 50 mL). The combined organic extracts were washed with brine and dried over magnésium sulfate. The mixture was filtered, and the filtrate concentrated under vacuum to obtain the crude title compound as a light yellow solid (9.8 g), which was used in the next step without purification. An analytical sample was prepared by purification via chromatography on silica gel, eluting with a 30 % to 60 % solution of ethyl acetate in heptane. 1H NMR (500 MHz, deuterochloroform) delta 1.26 (d, 6 H) 1.35 (t, 3 H) 1.86 - 1.95 (m, 2 H) 2.04 - 2.17 (m, 2 H) 2.84 - 2.96 (m, 2 H) 3.89 -
3.98 (m, 1 H) 4.28 (q, 2 H) 4.25 - 4.40 (m, 2 H) 4.89 - 4.97 (m, 1 H) 5.06 (s, 2 H) 7.64 (s, 1 H); LCMS (ES+): 325.1 (M+1).
Préparation 3: Isopropyl 4-r5-bromo-4-(ethoxvcarbonvl)-1H-pyrazol-1-vllpiperidine-1carboxylate
O
Neat tert-butyl nitrite (4.8 mL, 39.3 mmol) was added slowly to a stirred mixture of isopropyl 4-[5-amino-4-(ethoxycarbonyl)-1H-pyrazol-1-yl]-piperidine-1-carboxylate (Préparation 2) (8.5 g, 26.2 mmol) and copper (II) bromide (3.7 g, 16 mmol) in acetonitrile (100 mL) at room température. A significant exothermic effect was observed with the mixture warming to about 50 °C. After continued heating at 65 °C for 30 minutes, the reaction was cooled to room température, and then concentrated under vacuum. An excess of 10 % aqueous ammonia was added, and the mixture was extracted with ethyl acetate. The organic phase was washed with water and brine, and concentrated under vacuum. The residue was purifïed by chromatography on silica gel eluting with 30 % to 70% ethyl acetate in heptane to provide the title compound as a yellow oil, which was about 70% pure by NMR and LCMS. The material was used in the next step without further purification. 1H NMR (400 MHz, deuterochloroform) delta
1.23 (d, 6 H) 1.34 (t, 3 H) 1.84 - 1.95 (m, 2 H) 2.01 - 2.15 (m, 2 H) 2.82 - 2.98 (m, 2 H) 4.25 - 4.36 (m, 2 H) 4.30 (q, 2 H) 4.45 - 4.56 (m, 1 H) 4.86 - 4.96 (m, 1 H) 7.95 (s, 1 H); LCMS (ES+): 387.9 (M+1).
Préparation 4: Isopropyl 4-[5-bromo-4-(hvdroxvmethyl)-1 H-pyrazol-1-vl]piperidine-1carboxylate
Br
HO
To a solution of isopropyl 4-[5-bromo-4-(ethoxycarbonyl)-1H-pyrazol-1-yl]piperidine-1carboxylate (3.59 g, 6.5 mmol) in tetrahydrofuran (32 mL) cooled to 0 °C was added a 2 M solution of borane-methyl sulfide complex in tetrahydrofuran (14.6 mL, 29.2 mmol). The reaction mixture was heated at reflux for 21 hours and then stirred for 4 hours at room température. The mixture was cooled to 0 °C, and methanol was added. The resulting solution was warmed to room température and stirred for 10 minutes. The solution was re-cooled to 0 °C and aqueous 2 M sodium hydroxide solution (10 mL) was added dropwise. The resulting mixture was diluted with ethyl acetate and stirred vigorously for 30 minutes. The layers were separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic layers were washed sequentially with water and brine and then dried over magnésium sulfate. The mixture was filtered, and the filtrate concentrated under vacuum. Chromatography over silica gel eluting with 55% to 70% ethyl acetate in heptane gave the title compound as an oil (1.89 g, 84 %). 1H NMR (400 MHz, deuterochloroform) delta 1.23 (d, 6 H), 1.87 -1.95 (br m, 3 H), 2.06 (qd, 2 H), 2.89 (br t, 2 H), 4.29 (br s, 2 H), 4.39 (tt, 1 H), 4.50 (d, 2 H),
4.90 (m, 1 H), 7.58 (s, 1 H); LCMS (ES+) 348.0 (M+1).
Préparation 5: Isopropyl 4-[5-cvano-4-(hvdroxvmethvl)-1H-pyrazol-1-vl1piperidine-1carboxylate
CN
Isopropyl 4-[5-bromo-4-(hydroxymethyl)-1 H-pyrazol-1 -yl]piperidine-1 -carboxylate (1.42 g, 4.10 mmol), tris-(dibenzylideneacetone) dipalladium (156 mg, 0.170 mmol), 1-1'-bis(diphenylphosphino) ferrocene (192 mg, 0.346 mmol), zinc dust (68.8 mg, 1.06 mmol), zinc cyanide (497 mg, 4.23 mmol) and A/,A/-dimethylacetamide (20 mL) were combined in a microwave vial. The vial was flushed with nitrogen, sealed and heated at 120 °C for 1 hour in a microwave reactor (Biotage Initiator 2.2). The reaction mixture was passed through a pad of Florisil™, diluted with ethyl acetate and then water was added. The aqueous phase was extracted 3 times with ethyl acetate and the combined organic layers were dried over magnésium sulfate. The mixture was filtered, and the filtrate evaporated under vacuum. Chromatography on silica gel eluting with 55% to 70% ethyl acetate in heptane gave the title compound as a green oïl that solidified upon standing (1.06 g, 88 %). 1H NMR (400 MHz, deuterochloroform) delta 1.24 (d, 6 H), 1.99 (br d, 2 H), 2.06 - 2.17 (m, 3 H), 2.93 (br t, 2 H), 4.31 (br s, 2 H), 4.48 (tt, 1 H), 4.71 (d, 2 H),
4.92 (m, 1 H), 7.60 (s, 1 H); LCMS (ES+): 293.1 (M+H).
Préparation 6: 2-Fluoro-4-i(2-hvdroxyethvl)thiolphenol
To a solution of 4-bromo-2-fluorophenol (0.75 mL, 6.8 mmol) and diisopropylethylamine (3.5 mL, 20.09 mmol) in 1,4-dioxane (35 mL) was added 9,9-dimethyl-4,5bis(diphenylphosphino)xanthene (415 mg, 0.717 mmol), bis(dibenzylideneacetone)palladium (322 mg, 0.351 mmol) and 2-mercaptoethanol (0.46 mL, 6.86 mmol), and the dark brown reaction solution was heated at 110 °C for 16 hours. The reaction was allowed to cool to room température, diluted with water and extracted with ethyl acetate four times. The organic extracts were combined and dried over magnésium sulfate, The mixture was filtered, and the filtrate concentrated under reduced pressure to give a maroon oïl which was purified by chromatography on silicon gel to afford the title compound (985 mg, 76 %) as a maroon solid. 1H NMR (400 MHz,
deuterochloroform) delta 3.00 (t, 2H, J=5.95 Hz) 3.69 (d, 2 H, J=3.71 Hz) 6.89-6.95 (m,
H) 7.11 (ddd, 1 H, J=8.39, 2.15, 1.17 Hz) 7.17 (dd, 1 H, J=10.54, 2.15 Hz). Préparation 7: 4-K2-{[tert-Butvl(dimethyl)silylloxv)ethyl)thiol-2-fluorophenol
F
/ \
To a solution of 2-fluoro-4-[(2-hydroxyethyl)thio]phenol (985 mg, 5.24 mmol) and imidazole (371 mg, 5.30 mmol) in /V,/\/-dimethylformamide (5 mL) was added tertbutyldimethylsilyl chloride (814 mg, 5.24 mmol) portion-wise, and the reaction was stirred at room température for 4 hours. The reaction was concentrated under reduced pressure, and the residue diluted with water followed by extraction with ethyl acetate three times. The combined organic extracts were washed with brine and dried over magnésium sulfate. The mixture was filtered, and the filtrate concentrated under reduced pressure to give the title compound as an orange oil (1.43 g, 90 %) which was used without further purification. LCMS (ES+): 301.1 (M-1 ).
Préparation 8:1-r4-(BenzvÎoxv)-3-fluorophenyll-1H-tetrazole
To a suspension of 4-(benzyloxy)-3-fluoroaniline (1.04 g, 4.8 mmol) (WO 2005030140) under a nitrogen atmosphère was added acetic acid (2.3 mL, 38.3 mmol), triethy lorthoformate (2.44 mL, 14.4 mmol) and sodium azide (0.34 g, 5.3 mmol), and the reaction mixture heated at 95 °C for 2.5 hours. The solution was then allowed to cool to room température, and water was added followed by extraction with ethyl acetate three times. The extracts were combined and washed with brine and dried over magnésium sulfate. The mixture was filtered and concentrated under reduced pressure, and the crude material purified by chromatography on silicon gel (20 - 40 % ethyl acetate in heptane) to give the title compound as a white solid (1.12 g, 86 %). 1H NMR (400 MHz,
deuteromethanol) delta 9.65 (s, 1 H), 7.73 - 7.68 (dd, 1 H, J=11, 2.5 Hz), 7.60 - 7.57 (m,
H) 7.47 - 7.45 (m, 2H), 7.40 - 7.30 (m, 5H), 5.24 (s, 2H); LCMS (ES+): 271.1 (M+1 ). Préparation 9: 2-Fluoro-4-(1H-tetrazol-1-yl)phenol
To 1-[4-(benzyloxy)-3-fluorophenyl]-1 H-tetrazole (1.12 g, 4.14 mmol) in a Parr shaker flask was added éthanol (40 mL), and the solution purged with nitrogen gas. 10% palladium on carbon (0.30 g) was added, and the reaction hydrogenated on a Parr shaker apparatus at 40 psi of hydrogen for 30 minutes, The mixture was then filtered through a micro pore filter, and the filtrate was concentrated under reduced pressure to yield the title compound as a white solid (0.67 g, 90 %) which was use without purification. 1H NMR (400 MHz, deuteromethanol) delta 9.62 (s, 1H), 7,65 - 7.62 (dd, 1 H, J=11,2.5 Hz), 7.50 - 7.46 (m, 1 H) 7.47 - 7.45 (dd, 1 H, J=9.0, 9.0 Hz); LCMS (ES+): 181.1 (M+1).
Préparation 10: Isopropyl 4-(5-cyano-4-((methvlsulfonvloxv)methvl)-1H-pyrazol-1yl)piperidine-1 -carboxvlate
N ///
Isopropyl 4-(5-cyano-4-(hydroxymethyl)-1 H-pyrazol-1 -yI)pipertdine-1 -carboxylate (Préparation 5) (75 mg, 0.24 mmol) was dissolved in 1 mL of anhydrous dichloromethane and triethylamine (0.1 mL, 0.74 mmol) was added. The reaction mixture was cooled in an ice bath and methanesulfonic anhydride (62 mg, 0.34 mmol) was then added. The solution was removed from the ice bath and stirred for 30 minutes. The reaction was quenched by addition of saturated aqueous sodium bicarbonate and the layers were separated. The aqueous layer was extracted three more times with dichloromethane. The organic extracts were combined and washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated to give an oil (75 mg,
100% yield). The crude material was used in subséquent steps without further purification.
Préparation 11 ; tert-Butyl 4-hydrazinopiperidine-1-carboxylate hydrochloride sait
Into a solution of tert-butyl 4-oxopiperidine-1-carboxylate (50 g, 0.25 mmol) in methanol (500 mL) in an autoclave was added hydrazine mono-hydrochloride (17.2 g, 0.25 mmol) in water (100 mL). The white mixture was stirred under argon followed by the addition of 5% platinum on carbon (750 mg) as a slurry in water. The autoclave was sealed and charged to 60 atmosphères with hydrogen, and the reaction was stirred for 15 hours. Upon completion, the reaction was filtered through Celite®, and the pad washed with methanol. This préparation was carried out six times. The combined filtrâtes were concentrated under reduced pressure, and the resulting white precipitate (di-tert-butyl4,4'-hydrazine-1,2-diyldipiperidine-1 -carboxylate) by-product was collected by filtration and washed several times with water. The aqueous filtrate was then concentrated under reduced pressure to give the desired product (221 g, 59%) as a colorless solid. 1H NMR (400MHz, deuterochloroform) delta 4.13 (br s, 2H), 3.32 (br t, 1H), 2.77 (brt, 2H), 2.16 (m, 2H), 1.66 (m,2H), 1.43 (s, 9H).
Préparation 12: tert-Butyl 4-[5-amino-4-(ethoxvcarbonvl)-1H-pyrazol-1-yllpiperidine-1carboxylate
O
A mixture of tert-butyl 4-hydrazinopiperidine-1-carboxylate hydrochloride sait (221 g, 880 mmol), ethyl 2-cyano-3-ethoxyacrylate (153 g, 880 mmol), sodium acetate trihydrate (477 g, 352 mmol) and éthanol (2000 mL) was stirred at 85 degrees Celsius for 8 hours. The mixture was concentrated under reduced pressure, and the residue dissolved in ethyl acetate and water. The layers were separated, and the aqueous layer
extracted with ethyl acetate. The combined organic extracts were then dried over magnésium sulfate. The mixture was filtered, and the filtrate concentrated under reduced pressure. The crude material was purified by filtration through a short plug of silica gel eluting with 40% ethyl acetate in heptane to produce the product as a pale yellow solid (214 g, 72%). 1H NMR (500 MHz, deuterochloroform) delta 7.60 (s, 1 H),
5.27 (br s, 2H), 4.23 (br q, 4H), 3.91 (m, 1 H), 2.81 (br s, 2H), 2.04 (m, 2H), 1.86 (m, 2H),
1.44 (s, 9H), 1.29 (t, 3H).
Préparation 13: terf-Butyl 4-r5-bromo-4-(ethoxvcarbonyl)-1 H-pyrazo!-1-vHpiDeridine-110 carboxylate
To a solution of copper (II) bromide (1.69 g, 770 mmol) in acetonitrile (1000 mL) was slowly added fert-butyl nitrite (112 mL, 960 mmol), and the solution was heated to 65 degrees Celsius. To this was added a solution of fert-butyl 4-[5-amino-4(ethoxycarbonyl)-1H-pyrazol-1-yl]piperidine-1-carboxylate (215 g, 640 mmol) in acetonitrile (650 mL) drop-wise over 30 minutes. After 4 hours, the reaction was allowed to cool to room température and was then poured into 2 M hydrochloric acid (1500 mL) in ice. The mixture was extracted with ethyl acetate three times, and the combined organic extracts were washed with saturated aqueous sodium bicarbonate and then dried over magnésium sulfate. The mixture was filtered, and the filtrate concentrated under reduced pressure. The resulting residue was purified by filtration through a short plug of silica gel eluting initially with 10% heptane in dichloromethane followed by dichloromethane to give the title compound (137 g, 53%) as a yellow oil which solidified on standing. 1H NMR (400 MHz, deuterochloroform) delta 7.95 (s, 1H), 4.48 (m, 1 H), 4.28 (br q, 4H), 2.86 (br s, 2H), 2.06 (m, 2H), 1.90 (m, 2H), 1.44 (s, 9H),
1.34 (t, 3H).
Préparation 14: tert-Butvl 4-[5-bromo-4-(hvdroxvmethvl)-1H-pvrazol-1-vllpiperidine-1carboxylate
Br
HO
To a solution of tert-butyl 4-[5-bromo-4-(ethoxycarbonyl)-1 H-pyrazol-1 -yl]piperidine-T carboxylate (137 g, 0.34 mol) in tetrahydrofuran (1300 mL) cooled to 0 degrees Celsius was slowly added borane-methyl sulfide (97 mL, 1.02 mol). The solution was allowed to warm to room température and then heated at reflux for 15 hours. The reaction was then cooled in an ice bath, and methanol (40 mL) added drop-wise. The solution was then stirred at room température for 20 minutes. Aqueous 2 M sodium hydroxide (1200 mL) was added, and the layers were separated. The aqueous layer was extracted with ethyl acetate, and the combined organics layers were washed with brine, dried over magnésium sulfate, and the solvent removed under reduced pressure. The resulting residue was purified by filtration through a short plug of silica gel eluting with 30% ethyl acetate in heptane to reveal the title compound as an colorless solid (61.4 g, 50%). Impure material from this purification was further purified via the above chromatographie procedure to provide a second batch of the title compound (22 g, 18%) as a colorless solid. 1H NMR (400 MHz, deuterochloroform) delta 7.59 (s, 1H), 4.52 (s, 2H), 4.37 (m, 1H), 4.25 (br s, 2H), 2.86 (br s, 2H), 2.06 (m br s, 2H), 1.89 (m, 2H), 1.45 (s, 9H).
Préparation 15: terf-Butyl 4-r5-cyano-4-(hvdroxvmethvl)-1H-pvrazol-1-yllpiperidine-1carboxylate
N
Copper (I) cyanide (2.97 g, 33.3 mmol) was added to a stirred solution of teri-butyl 4-[5bromo-4-(hydroxymethyl)-1 H-pyrazol-1-yl]piperidine-1-carboxylate (10 g, 27.8 mmol) in degassed dimethylformamide (100 mL). The reaction was then heated at 165 degrees Celsius for 4 hours and allowed to cool to room température. It was further cooled in an ice-bath, and a solution of ethylenediamine (5.5 mL) in water (20 mL) was added followed by dilution with more water (70 mL). The mixture was then extracted with ethyl acetate, and the layers separated. The organic layer was washed sequentially with water and brine and then dried over magnésium sulfate. The mixture was filtered and the filtrate concentrated under reduced pressure. This procedure was carried out in 8 batches. The final crude residues were combined and purified by repeated silica gel
column chromatography eluting with 40 % ethyl acetate in heptane to give the title compound (11.6 g, 17%) as a colorless solid. 1H NMR (400MHz, deuterochloroform) 7.59 (s, 1H), 4.71 (s, 2H), 4.45 (m, 1H), 4.26 (br s, 2H), 2.88 (br t, 2H). 2.08 (m, 2H),
1.98 (m, 2H), 1.48 (s, 9H); LCMS (ES+): 207.1 (M-Boc+H).
For an alternative synthesis of tert-butyl 4-[5-cyano-4-(hydroxymethyl)-1H-pyrazol-1yl]piperidine-1 -carboxylate please see Example 50.
Préparation 16: tert-Butyl 4-(5-cyano-4-((methyÎsulfonvloxv)methvl)-1H-pyrazol-110 yl)piperidine-1-carboxylate
N
To a stirred solution of tert-butyl 4-(5-cyano-4-(hydroxymethyl)-1H-pyrazol-1yl)piperidine-1-carboxylate (202 mg, 0.659 mmol) in dichloromethane (6.6 mL) was added triethylamine (0.18 mL, 1.32 mmol) followed by methanesulfonic anhydride (189 mg, 1.1 mmol) at room température. The mixture was stirred for 4.5 hours before it was diluted with dichloromethane and saturated aqueous bicarbonate. The layers were separated and the aqueous layer was extracted with dichloromethane. The combined organic extracts were washed with brine, dried over magnésium sulfate, filtered and the filtrate was concentrated in vacuo to give tert-butyl 4-(5-cyano-420 ((methylsulfonyloxy)methyl)-1H-pyrazol-1-yl)piperidine-1-carboxylate as an oil which was used without further purification.
Préparation 17:2-Fluoro-4-(1-((2-(trimethylsilyl)ethoxv)methyl)-1 H-tetrazol-5-yl)phenol and 2-Fluoro-4-(2-((2-(trimethylsilvl)ethoxv)methyl)-2H-tetrazol-5-vl)phenol
A) 4-(Benzyloxy)-3-fluorobenzonitrile
To a stirred solution of 3-fluoro-4-hydroxybenzonitrile (1.00 g, 7.30 mmol) in 20 mL of acetonitrile was added portion-wise potassium carbonate (2.02 g, 14.6 mmol). The resulting mixture was stirred for 10 minutes before benzyl bromide (1.33 mL, 10.9 mmol) was added, The mixture was stirred at room température for 70 hours before it was diluted with ethyl acetate and water. The organic phase was separated and washed with water, brine, dried over magnésium sulfate, filtered and the filtrate was concentrated in vacuo. The residue was purified by flash chromatography, eluting with a gradient of 5 to 20% of ethyl acetate in heptane to give 4-(benzyloxy)-3fluorobenzonitrile as a white solid (1.33 g).
B) 5-(4-(Benzyloxv)-3-fluorophenyl)-1 H-tetrazole and 5-(4-(Benzyloxv)-3-fÎuorophenyl)2H-tetrazole
A vial charged with 4-(benzyloxy)-3-fluorobenzonitrile (250 mg, 1.10 mmol), sodium azide (214 mg, 3.30 mmol), ammonium chloride (176 mg, 3.30 mmol) and 3 mL of N,Ndimethylformamide was heated at 110 degrees Celsius for 18 hours. The reaction mixture was cooled to room température, diluted with water and ethyl acetate and the pH was adjusted to 3 using aqueous 1 N hydrochloric acid. The organic phase was separated and washed with brine, dried over magnésium sulfate, filtered and the filtrate was concentrated in vacuo to give the title compounds as a white solid (270 mg). This material was used in subséquent steps without purification.
C) 5-(4-(Benzyloxv)-3-fluorophenyl)-1 -((2-(trimethvlsilvl)ethoxv)rnethyl)-1 H-tetrazole and 5-(4-(Benzvloxv)-3-fluorophenyl)-2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazole
To a solution of 5-(4-(benzyloxy)-3-fluorophenyl)-1 H-tetrazole and 5-(4-(benzyloxy)-3fluorophenyl)-2H-tetrazole (270 mg, 1 mmol) dissolved in tetrahydrofuran was added sodium hydride (44 mg, 1.1 mmol) in four portions and the resulting mixture was stirred at room température for 15 minutes. (2-(Chloromethoxy)ethyl)trimethylsilane (0.19 mL, 1.0 mmol) was then added and the reaction mixture was stirred at room température for 16 hours. The reaction was quenched by the addition of water and ethyl acetate was added. The organic phase was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over magnésium sulfate, filtered and the filtrate was concentrated under reduced pressure. Purification by flash chromatography, eluting with a gradient of ethyl acetate and heptane (5 to 20% ethyl acetate) gave the desired product as a white solid (270 mg, 67% yield).
D) 2-Fluoro-4-( 1 -((2-(trimethylsilvl)ethoxv)methyl)-1 H-tetrazol-5-yl)phenol and 2-Fluoro4-(2-((2-(trimethylsilyl)ethoxv)methvl)-2H-tetrazol-5-vl)phenol
To a solution of 5-(4-(benzyloxy)-3-fluorophenyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H5 tetrazole and 5-(4-(benzyloxy)-3-fluorophenyl)-2-((2-(trimethylsilyl)ethoxy)methyl)-2Htetrazole (140 mg, 0.35 mmol) dissolved in a mixture of 2 mL of éthanol and 2 mL of tetrahydrofuran was added palladium black (56 mg, 0.53 mmol) and formic acid (0.14 mL, 3.5 mmol). The resulting mixture was stirred at room température for 4 hours before being filtered though a pad of Celite®. The filtrate was concentrated under reduced pressure and the resulting crude material was used in the subséquent step without further purification.
Préparation 18: 5-(4-(Benzyloxv)-3-fluorophenvlM-(2-(trimethylsilvloxv)ethyl)-1Htetrazole and 5-(4-(Benzvloxv)-3-fluorophenvl)-2-(2-(trimethylsilvloxv)ethyl)-2l-l-tetrazole
To a solution of 5-(4-(benzyloxy)-3-fluorophenyl)-1H-tetrazole and 5-(4-(Benzyloxy)-3fluorophenyl)-2H-tetrazole (Préparation 17, Step B) (550 mg, 2 mmol) dissolved in N,Ndimethylformamide (8 mL) was added sodium hydride (163 mg, 4 mmol) in two portions and the resulting mixture was stirred at room température for 5 minutes. (2Bromoethoxy)trimethylsilane (1.3 mL, 6 mmol) was then added and the reaction mixture was stirred at 70 degrees Celsius for 16 hours before being cooled to room température. The reaction was quenched by addition of water and ethyl acetate was added. The organic phase was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over magnésium sulfate, filtered and concentrated under reduced pressure. The residue was purified by flash chromatography, eluting with a gradient of ethyl acetate and heptane (5 to 30% ethyl acetate) to give 5-(4-(benzyloxy)-3-fluorophenyl)-1-(2(trimethylsilyloxy)ethyl)-1 H-tetrazole (100 mg, 12% yield) and 5-(4-( benzyloxy)-3fluorophenyl)-2-(2-(trimethylsilyloxy)ethyl)-2H-tetrazole (600 mg, 69% yield).
Préparation 19: 2-Fluoro-4-(2-(2-(trimethvlsilvloxv)ethvl)-2H-tetrazol-5-yl)phenol
To a solution of 5-(4-(benzyloxy)-3-fluorophenyl)-2-(2-(trimethylsilyloxy)ethyl)-2Htetrazole (Préparation 18)(230 mg, 0.54 mmol) dissolved in a mixture of 6 mL of éthanol and 6 mL of tetrahydrofuran was added palladium black (86 mg, 0.806 mmol) and formic acid (0.215 mL, 5.4 mmol). The resulting mixture was stirred at room température for 4 hours before being filtered through a pad of Celite®. The filtrate was concentrated under reduced pressure and the resulting crude material (180 mg) was used in the subséquent step without further purification.
6l
Préparation 20: 2-Fluoro-4-( 1 -(2-(trimethvlsilvloxv)ethyl)-1 H-tetrazol-5-yl)phenol
To a solution of 5-(4-(benzyloxy)-3-fluorophenyl)-1-(2-(trimethylsilyloxy)ethyl)-1Htetrazole (Préparation 18)(130 mg, 0.30 mmol) dissolved in a mixture of 2 mL of éthanol and 2 mL of tetrahydrofuran was added palladium black (48 mg, 0.45 mmol) and formic acid (0.12 mL, 3 mmol). The resulting mixture was stirred at room température for 4 hours before being filtered over a pad of Celite®. The filtrate was concentrated under reduced pressure and the resulting crude material (94 mg) was used in the subséquent step without further purification.
Préparation 21: 2-Fluoro-4-(1-methvl-1H-tetrazol-5-yl)phenol
A) 5-(4-(Benzvloxy)-3-fluorophenvl)-1-methvl-1H-tetrazole and 5-(4-(Benzvloxv)-3fluorophenyl)-2-methvl-2H-tetrazole
To a stirred solution of 5-(4-(benzyloxy)-3-fluorophenyl)-1H-tetrazole and 5-(4(benzyloxy)-3-fluorophenyl)-2H-tetrazole (Préparation 17, Step B) (1.50 g, 5.55 mmol) in 30 mL of tetrahydrofuran was added sodium hydride (444 mg, 11.1 mmol) in two portions at room température. After 5 minutes, iodomethane (1.04 mL, 16.6 mmol) was added and the reaction was stirred under a nitrogen atmosphère for 15 hours at room température. The mixture was diluted with water and extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried with magnésium sulfate, filtered and the filtrate was concentrated in vacuo. The residue was purified by flash chromatography, eluting with 10-40% ethyl acetate in heptane to give 5-(4-(benzyloxy)3-fluorophenyl)-2-methyl-2H-tetrazole as a white solid (1.1 g) and 5-(4-(benzyloxy)-3fluorophenyl)-1-methyl-1H-tetrazole as a white solid (450 mg).
5-(4-(Benzyloxy)“3-fluorophenyl)-1-methyl-1H-tetrazole. 1H NMR(400 MHz, deuterochloroform) delta 4.15 (s, 3 H) 5.23 (s, 2 H) 7.15 (t, J=8.39 Hz, 1 H) 7.31 - 7.48 (m, 6 H) 7.52 (dd, J=11.13, 2.15 Hz, 1 H). LCMS (M+1 ) 285.1.
B) 2-Fluoro-4-( 1 -methyl-1 H-tetrazol-5-yl)phenol
To a solution of 5-(4-(benzyloxy)-3-fluorophenyl)-1-methyl-1 H-tetrazole (500 mg, 1.76 mmol) in 6 mL of éthanol and 6 mL of tetrahydrofuran was added formic acid (0.7 mL,
17,6 mmol) followed by palladium black (281 mg, 2.64 mmol). The reaction mixture was stirred at room température for 4 hours. The reaction mixture was filtered through Celite® and the filtrate was concentrated in vacuo to give 2-fluoro-4-(1-methyl-1 Htetrazol-5-yl)phenol as a white solid (330 mg) which was used for in subséquent reactions without further purification.
Préparation 22: 4-(1-Methyl-1l-l-tetrazol-5-vl)phenol
HO
A) 4-(Benzyloxy)-N-methvlbenzamide
To a flask charged with thionyl chloride (0.35 mL, 4.82 mmol) was added a solution of commercially available 4-benzyloxybenzoic acid (1.00 g, 4.38 mmol) in 10 mL of dichloromethane and 0.01 mL of Ν,Μ-dimethylformamide at zéro degrees Celsius with stirring. The ice bath was removed and the solution was stirred for 4 hours at room température. The mixture was concentrated in vacuo to give a white solid. This solid was taken up in 10 mL of methyl amine (2 M in tetrahydrofuran) and the resulting solution was stirred at room température for 70 hours. The mixture was diluted with ethyl acetate and water and the organic layer was separated, dried over magnésium sulfate, filtered and the filtrate was concentrated in vacuo to give a white solid. This solid was recrystallized from ethyl acetate and heptane to give 4-(benzyloxy)-Nmethylbenzamide as white needles (850 mg).
B) 5-(4-(Benzyloxv)phenvl)-1-methyl-1 H-tetrazole
To a stirred solution of 4-{benzyloxy)-N-methylbenzamide (200 mg, 0.829 mmol) in 3 mL of acetonitrïle and one drop of Λ/,/V-dimethylformamide, in a flask topped with a reflux condenser, was added triethylamine (0.12 mL) under a nitrogen atmosphère. The reaction mixture was stirred for 10 minutes before thionyl chloride (0.078 mL, 1.08 mmol) was added drop-wise. The yellow reaction mixture was stirred for 1 hour at room température under a nitrogen atmosphère. Triethylamine (0.36 mL) was then added slowly, followed by tetrabutylammonium chloride (37.4 mg, 0.12 mmol) and sodium
azide (611 mg, 1.82 mmol). The resulting yellow suspension was vigorously stirred for hours at room température under a nitrogen atmosphère. The mixture was diluted with water and ethyl acetate. The organic layer was separated, washed with brine, dried over magnésium sulfate, filtered and the filtrate was concentrated in vacuo. The residue was purified by flash chromatography, eluting with a gradient of 10 to 40% ethyl acetate in heptane to give 5-(4-(benzyloxy)phenyl)-1-methyl-1 H-tetrazole as a white solid (180 mg).
C) 4-(1 -Methyl-1 H-tetrazol-5-yl)phenol
To a stirred solution 5-(4-(benzyloxy)phenyl)-1 -methyl-1 H-tetrazole (180 mg, 0.676 mmol) in 3 mL of éthanol and 3 mL of tetrahydrofuran was added formic acid (0.27 mL,
6.76 mmol) followed by palladium black (108 mg, 1,01 mmol). The mixture was stirred at room température for 4 hours. The reaction mixture was filtered through Celite® and the filtrate was concentrated to give 4-(1-methyl-1H-tetrazol-5-yl)phenol as a white solid (110 mg), which was used in subséquent reactions without further purification.
Préparation 23: 3-Fluoro-4-hvdroxvbenzamide
A mixture of commercially available 3-fluoro-4-hydroxybenzonitrile (500 mg, 3.65 mmol) and potassium hydroxide (1.02 g, 18.2 mmol) in 10 mL of 80% éthanol was heated at reflux for 16 hours. After cooling to room température the mixture was concentrated in vacuo and the residue was taken up into water, acidified with acetic acid and extracted with ethyl acetate. The combined organic extracts were dried over magnésium sulfate, filtered and the filtrate was concentrated in vacuo. The residue was purified by flash chromatography, eluting with a gradient of 20 to 60% ethyl acetate in heptane to give 3fluoro-4-hydroxybenzamide as a white solid (210 mg).
Alternative^, 3-fluoro-4-hydroxybenzamide can be prepared asfollows:
To a stirred solution of urea hydrogen peroxide (4.2 g, 43.8 mmol) in 12 mL of water was added solid sodium hydroxide (1.04 g, 25.5 mmol). The resulting solution was cooled in an ice bath before a solution of 3-fluoro-4-hydroxybenzonitrile (1.00 g, 7.29 mmol) in 5 mL of éthanol was added. The mixture was vigorously stirred for 2 hours at room température before it was diluted with water (100 mL) and ethyl acetate (100 mL).
The mixture was stirred for 5 minutes before 1 M hydrochloric acid was added until pH
4. The aqueous layer was separated and extracted with ethyl acetate (3X100 mL). The combined organic layers were dried over magnésium sulfate, filtered, and the filtrate was concentrated to give a white solid. This solid was triturated with diethyl ether and heptane (2:1, 90 mL) for 1 hour, before filtering to give 3-fluoro-4-hydroxybenzamide as a white solid (1.05 g). 1H NMR (400 MHz, deutero dimethyl sulfoxide ) delta 6.93 (t, 7=8.69 Hz, 1 H) 7.19 (br. s., 1 H)7.53(dd, 7=8.39, 1.95 Hz, 1 H) 7.62 (dd, 7=12.40, 2.05 Hz, 1 H) 7.77 (br. s., 1 H) 10.39 (s, 1 H). LCMS (ES) 156.0 (M+1 ).
Préparation 24: 2-Fluoro-4-hydroxvbenzamide
To a stirred solution of urea hydrogen peroxide (2.1 g, 21.9 mmol) in 6 mL of water was added solid sodium hydroxide (521 mg, 12.8 mmol). The resulting solution was cooled in an ice bath before a solution of 2-fluoro-4-hydroxybenzonitrile (500 mg, 3.65 mmol) in 2 mL of éthanol was added. The mixture was vigorously stirred for 2 hours at room température before it was diluted with water (100 mL) and ethyl acetate (100 mL). The mixture was stirred for 5 minutes before 1 M hydrochloric acid was added until pH=4. The aqueous layer was separated and extracted with ethyl acetate (3X50 mL). The combined organic layers were dried over magnésium sulfate, filtered, and the filtrate was concentrated to give 2-fluoro-4-hydroxybenzamide as a white solid.
Préparation 25: Isopropyl 4-(5-cyano-4-(1-hvdroxvethyl)-1 H-pvrazol-1-vl)piperidine-1carboxylate
Isopropyl 4-(5-cyano-4-formyl-1H-pyrazol-1-yl)piperidine-1’Carboxylate (Example 9, Step A) (51 mg, 0.18 mmol) was dissolved in 2 mL of anhydrous tetrahydrofuran and cooled to négative 78 degrees Celsius under a nitrogen atmosphère. Méthylmagnésium bromide (0.070 mL, 0.21 mmol, 3 M in diethyl ether) was then added drop-wise. The
cold bath was removed and the mixture was stirred for 1 hour at room température. The mixture was diluted with 1 M aqueous potassium bisulfate and extracted three times with ethyl acetate. The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The residue was purified by flash chromatography, eluting with a gradient of ethyl acetate in heptane (20 to 100% ethyl acetate) to give isopropyl 4-(5-cyano-4-(1-hydroxyethyl)-1H-pyrazol-1yl)piperidine-1-carboxylate as a white solid (33 mg) which was used in subséquent steps without purification.
Préparation 26: 1-Methylcyclopropyl 4-nitrophenyl carbonate
A) 1-Methvlcyclopropanol
A 1 Lflask was charged with titanium methoxide (100 g), cyclohexanol (232 g), and toluene (461 mL). The flask was equipped with a Dean-Stark trap and condenser. The mixture was heated at 140 degrees Celsius until the methanol was removed. The toluene was removed at 180 degrees Celsius. More toluene was added and this process was repeated twice. After ail the toluene was removed the flask was dried under high vacuum. Diethyl ether (580 mL) was added to the flask to préparé a 1 M solution in diethyl ether. A 5 L, 3-neck flask was equipped with an overhead stirrer, inert gas inlet and a pressure-equalizing addition funnel. The flask was flushed with nitrogen gas and charged with methyl acetate (60.1 mL, 756 mmol), titanium cyclohexyloxide (1 M solution in ether 75.6 mL), and diethyl ether (1500 mL). The solution was stined while keeping the reaction flask in a room température water bath. The addition funnel was charged with the 3 M ethylmagnesium bromide solution (554 mL, 1.66 moles). The Grignard reagent was added drop-wise over 3 hours at room température. The mixture became a light yellow solution, and then gradually a precipitate formed which eventually turned to a dark green/brown/black colored mixture. After stirring for an additional 15 minutes, following the addition of the Grignard, the mixture was carefully poured into a mixture of 10% concentrated sulfuric acid in 1 L of water. The resulting mixture was stirred until ail the solids dissolved. The aqueous layer was separated and extracted with diethyl ether 2 x 500 mL. The combined organic extracts were washed sequentially with water, brine, dried over potassium carbonate (500 g) for 30 minutes, filtered and
the filtrate was concentrated in vacuo to an oil. Sodium bicarbonate (200 mg) was added and the crude material was distilled, collecting fractions boiling around 100 degrees Celsius to gîve the title compound (23 grams) with methyl ethyl ketone and 2butanol as minor impurities. 1H NMR (500 MHz, deuterochloroform) delta 0.45 (app. t, J=6.59 Hz, 2 H), 0.77 (app. t, J=5.61 Hz, 2 H), 1.46 (s, 3 H). The préparation of the title compound is also described in W009105717.
B) 1-Methvlcyclopropyi 4-nitrophenvl carbonate
A solution of 1-methylcyclopropanol (10 g, 137 mmol), 4-nitrophehyl chloroformate (32 g, 152 mmol), and a few crystals of 4-dimethylaminopyridine (150 mg, 1.2 mmol) in dichloromethane (462 mL), was cooled to zéro degree Celsius. Triethylamine (36.5 g, 361 mmol) was added drop-wise. After 10 minutes, the ice bath was removed and the reaction was allowed to stir at room température for 14 hours. The reaction mixture was washed twice with saturated aqueous sodium carbonate. The aqueous phase was extracted with dichloromethane. The combined organic extracts were washed with water, dried over magnésium sulfate, filtered and the filtrate concentrated in vacuo. The residue was purified by flash silica gel chromatography, eluting with a gradient mixture of ethyl acetate in heptane (0 to 5% ethyl acetate over the first 10 minutes, then isocratic at 5% ethyl acetate to heptane) to gîve 20.8 g of the desired carbonate as a clear oil. This oil solidified upon standing.
1H NMR (500 MHz, deuterochloroform) delta 0.77 (app. t, J=6.59 Hz, 2 H), 1.09 (app. t, J=7.07 Hz, 2 H), 1.67 (s, 3 H), 7.40 (app. dt, J=9.27, 3.17 Hz, 2 H), 8.29 (app. dt, J=9.27, 3.17 Hz, 2 H).
Alternatively the 1-methylcyclopropanol can be prepared as follows:
-Methylcyclopropanol
A 2000 mL 4-neck flask was equipped with a mechanical stirrer, inert gas inlet, thermometer, and two pressure - equalizing addition funnels. The flask was flushed with nitrogen and charged with 490 mL of diethyl ether followed by 18.2 mL (30 mmol) of titanium tetra(2-ethylhexyloxîde). One addition funnel was charged with a solution prepared from 28.6 mL (360 mmol) of methyl acetate diluted to 120 mL with ether. The second addition funnel was charged with 200 mL of 3 M ethylmagnesium bromide in ether solution. The reaction flask was cooled in an ice water bath to keep the internai température at 10 degrees Celsius or below. Forty milliliters of the methyl acetate solution was added to the flask. The Grignard reagent was then added drop-wise from the addition funnel at a rate of about 2 drops every second, and no faster than 2 mL per
minute. After the first 40 mL of Grignard reagent had been added, another 20 mL portion of methyl acetate in ether solution was added. After the second 40 mL of Grignard reagent had been added, another 20 mL portion of methyl acetate in diethyl ether solution was added. After the third 40 mL of Grignard reagent had been added, another 20 mL portion of methyl acetate in ether solution was added. After the fourth 40 mL of Grignard reagent had been added, the last 20 mL portion of methyl acetate in ether solution was added.
The mixture was stirred for an additional 15 minutes following the completion of the addition of Grignard reagent. The mixture was then poured into a mixture of 660 g of ice and 60 mL of concentrated sulfuric acid with rapid stirring to dissolve ail solids. The phases were separated and the aqueous phase was extracted again with 50 mL of diethyl ether. The combined ether extracts were washed with 15 mL of 10% aqueous sodium carbonate, 15 mL of brine, and dried over 30 grams magnésium sulfate for 1 hour with stirring. The ether solution was then filtered. Tri-n-butylamine (14.3 mL, 60 mmol) and mesitylene (10 mL were added. Most of the diethyl ether was removed by distillation at atmospheric pressure using a 2.5 cm x 30 cm jacketed Vigreux column. The remainîng liquid was transferred to a smaller distillation flask using two 10 mL portions of hexane to facilitate the transfer. Distillation at atmospheric pressure was continued through a 2 cm x 20 cm jacketed Vigreux column. The liquid distilling at 98 105 °C was collected to provide 14 g of the title compound as a coloriess liquid. 1H NMR (400 MHz, deuterochloroform) delta 0.42 - 0.48 (m, 2 H), 0.74 - 0.80 (m, 2 H),
1.45 (s, 3 H), 1.86 (br. s., 1 H).
Préparation 27: 2-fluoro-4-(1-methvl-1H-imidazol-5-vl)phenol
HO
A) 5-(3-Fluoro-4-methoxvphenyl)-1-methyl-1 H Imidazole
2-Fluoro-4-bromo anisole (0.216 mL, 1.63 mmol), tri(2-furyl)phosphine (25.9 mg, 0.108 mmol), and potassium carbonate (300 mg, 2.17 mmol) were placed in a microwave vial and dissolved in anhydrous N,N-dimethylformamide (4.8 mL). The mixture was degassed with a stream of nitrogen gas for 10 minutes, 1-methylimidazole (0.087 mL,
1.1 mmol) and palladium(ll) acetate (12.4 mg, 0.054 mmol) were added, and the mixture was degassed for another 10 minutes. The vessel was placed in a microwave
reactor at 140 degrees Celsius for 2 hours. The mixture was diluted with ethyl acetate, filtered through Celite®, and the filtrate was concentrated under reduced pressure. The crude material was purified by chromatography eluting with a 25 to 100% ethyl acetate in heptane then 0 to10% methanol in dichloromethane gradient to give the title compound as a yellow oil (210 mg). 1H NMR (500 MHz, deuterochloroform) delta 3.57 (s, 3 H), 3.85 (s, 3 H), 6.95 - 6.98 (m, 2 H), 7.00 - 7.07 (m, 2 H), 7.42 (s, 1 H). Proton shift at 7.42 is indicative of desired imidazole isomer as compared to literature (Eur. J.
Org. chem., 2008, 5436 and Eur. J. Org., 2006, 1379).
B) 2-Fluoro-4-(1-methvl-1H-imidazol-5-yl)phenol
To a solution of 5-(3-fluoro-4-methoxyphenyl)-1-methyl-1H Imidazole (101.8 mg, 0.494 mmol) in dichloromethane (2.0 mL ) was added a solution of boron(lll) bromide (0.50 mL, 1.0 M solution in heptane) at -30 degrees Celsius. The mixture was stirred at room température for 20 hours. The mixture was then cooled to -30 degrees Celsius and methanol (2 mL) was added to the mixture. The mixture was concentrated in vacuo, and the residue was dissolved in water and neutralized with 1M sodium hydroxide. The solution was concentrated to give the title compound as a yellow solid (90 mg). This compound was used further without purification.
Préparation 28:2-Fluoro-4-(1-methvl-1H-imidazol-2-yl)phenol
A) 2T3-Fluoro-4-methoxyphenyl)-1-methyl-1H Imidazole
2-Fluoro-4-bromoanisole (0.256 mL, 1.93 mmol) and copper(l) iodide (375 mg, 1.93 mmol) were placed in a microwave vial and dissolved in N.N-dimethylformamide (4.8 mL), The mixture was degassed for 10 minutes with a stream of nitrogen gas, 1methylimidazole (0.078 mL, 0.96 mmol) and palladium(ll) acetate (11 mg, 0.048 mmol) were added, and the mixture was degassed for another 10 minutes. The vessel was placed in a microwave reactor at 140 degrees Celsius for 2 hours. The mixture was diluted with ethyl acetate (3 mL), poured into saturated aqueous ammonium chloride solution, stirred in the open air for 30 minutes, and extracted twice with ethyl acetate. The combined organic phases were washed with water, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The crude material was purified by chromatography, eluting with a gradient mixture of ethyl acetate to heptane (25 to 100% ethyl acetate/heptane then 0 to10% methanol in dichloromethane) to give 2-(3-fluoro-4methoxyphenyl)-1-methyl-1H Imidazole as a yellow oil (35.8 mg). 1H NMR (400 MHz, deuterochloroform) delta 3.66 (s, 3 H), 3.88 (s, 3 H), 6.90 (s, 1 H), 6.96 (m 1 H), 7.10 (s, 1 H), 7.24 - 7.33 (m, 2 H). Proton NMR indicates desired imidazole isomer as compared to the proton NMR of 5-(3-fluoro-4-methoxyphenyl)-1-methyl-1H Imidazole (préparation 27) and the literature Eur, J. Org. chem., 2008, 5436 and Eur. J. Org., 2006, 1379).
B) 2-Fluoro-4-(1-methyl-1H-imidazol-2-vl)phenol
2-Fluoro-4-(1-methyl-1 H-îmidazol-2-yl)phenol was prepared from 2-(3-fluoro-4 methoxyphenyl)-1-methyl-1H Imidazole following a procedure analogous to that in Préparation 27 (B) to give the title compound as a brown solid (33.4 mg). The crude material was used further without purification.
Préparation 29: 2-Fluoro-4-(methvlsulfonvl)-1-(prop-1-en-2-yl)benzene
To a solution of 1-bromo-2-fluoro-4-(methylsulfonyl)benzene (199 mg, 0.790 mmol) and potassium isopropenyltrifluoroborate (300 mg, 2.57 mmol) in 2-propanol (10 mL) was added the catalyst 1,T-bis-(diphenylphosphino)“ferrocene palladium dichloride (67 mg, 0.089 mmol) and triethylamine (0.17 mL, 1.20 mmol) sequentially. The reaction was heated at 90 degrees Celsius for 15 hours, and then the reaction was stirred at room température for 48 hours. Water and ethyl acetate were then added, and the layers were separated. The aqueous layer was extracted with ethyl acetate. The organic extracts were combined, washed with brine and dried over sodium sulfate. The mixture was filtered, and the filtrate concentrated under reduced pressure. The residue was purified by silica gel chromatography (10 to 100% ethyl acetate in heptane) to give the title compound as a white solid (130 mg, 80%). 1H NMR (500 MHz, deuterochloroform) delta 2.17 (s, 3 H), 3.08 (s, 3 H), 5.29 - 5.43 (m, 2 H), 7.51 (t, J=7.56 Hz, 1 H), 7.64 (dd, J=9.88, 1.59 Hz, 1 H), 7.70 (dd, J=8.05, 1.71 Hz, 1 H).
Préparation 30: 4-Hydroxy-2-methylbenzonitrile
OH
N
Boron trichloride in dichloromethane (61.2 mL, 1M) was added slowly to dichloromethane (93 mL) and cooled to -78 degrees Celsius. To this was added a solution of 4-methoxy-2-methylbenzonitrile (3.00 g, 20.4 mmol) and tetrabutylammonium iodide (7.17 g, 61.2 mmol) in dichloromethane (20 mL). The reaction mixture was allowed to stir at -78 degrees Celsius for 5 minutes. The reaction mixture was then gradually warmed to room température and stirred for 2 hours. An ice slurry was slowly added to quench the reaction. The reaction was allowed to stir for 30 minutes and the layers were separated. The aqueous layer was extracted with dichloromethane (2x) and the combined organic extracts were passed through a phase separated cartridge and concentrated in vacuo. The crude mixture was purified by flash chromatography eluting with 0% to 60% ethyl acetate in pentane to give the target compound as a yellow solid (1.85 g, 68 %). 1H NMR deuteromethanol delta ppm 7.40 (d, 1 H), 6.80 (s, 1H), 6.70 (d, 1H), 2.40 (s, 3H); GCMS (Cl method) ES+= 133 [Μ] AP+ = 133 [M].
Préparation 31A: 3-Fluoro-4-hydroxv-N-methylbenzamide
OH
NH
A) Benzyl 4-(benzvloxy)-3-fluorobenzoate
3-Fluoro-4-hydroxybenzoic acid (5.00 g, 32.06 mmol), benzyl bromide (8.22 mL, 67.3 mmol) and potassium carbonate (13.3 g, 96.24 mmol) were combined in acetone and heated at reflux for 18 hours. The solution was cooled down to room température, the solids were filtered and the filtrate was diluted with ethyl acetate. The organic phase was washed with saturated aqueous brine solution, dried over magnésium sulfate, filtered and the filtrate was concentrated under reduced pressure to give the desired product benzyl 4-(benzyloxy)-3-fluorobenzoate. 1H NMR (500 MHz, deuterochloroform) delta ppm 5.22 (s, 2 H) 5.36 (s, 2 H) 7.03 (t, J=8.42 Hz, 1 H) 7.29 - 7.52 (m, 10 H) 7.76 7.89 (m, 2 H); LCMS (ES+)= 381.2 (M+45)
B) 4-(Benzvloxy)-3-fluorobenzoic acid
Benzyl 4-(benzyloxy)-3-fluorobenzoate (11,6 g, 34.2 mmol) was dissolved in tetrahydrofuran (50 mL) and methanol (50 mL). Aqueous sodium hydroxide (70 mL, 1M) was added to the reaction mixture and stirred for 18 hours. The reaction was cooled in an ice bath and acidified to pH 3 by careful addition of aqueous 1 A4 solution of hydrochloric acid, A white solid precipitated out and was filtered and dried over night to give the desired product, 4-(benzyloxy)-3-fiuorobenzoic acid, as a white solid (7.6 g,
90%). 1H NMR (500 MHz, deuterodimethylsulfoxide) delta ppm 3.32 (br. s., 1 H) 5.27 (s, 2 H) 7.34 - 7.39 (m, 2 H) 7.42 (t, J=7.44 Hz, 2 H) 7.45 - 7.50 (m, 2 H) 7.68 (dd,
J=11.83, 2.07 Hz, 1 H) 7.75 (d, J=8.78 Hz, 1 H)
C) 4-(Benzyloxy)-3-fluoro-N-methylbenzamide
Thionyl chloride (2.7 mL, 37 mmol) was added to a solution of 4-(benzyloxy)-3fluorobenzoic acid in dimethylformamide (0.048 mL, 0.617 mmol) and dichloromethane (100 mL) at 0 degree Celsius and the resulting solution was stirred at room température for 20 hours. The reaction was concentrated under reduced pressure and dried under high vacuum for 2 hours. The resulting yellow solid was dissolved in tetrahydrofuran (60 mL) and a 2M solution of methylamine in tetrahydrofuran (35mL) was added and the reaction stirred at room température for 18 hours. The reaction mixture was concentrated under reduced pressure to half the original volume and a white solid precipitated out. The solid was filtered off, washed with water and dried in a vacuum oven overnight to give the desired product as a white solid (6.00 g, 70%). 1H NMR (500
MHz, deuterodimethylsulfoxide) delta ppm 2.76 (d, J=4.39 Hz, 3 H) 5.24 (s, 2 H) 7.30 7.38 (m, 2 H) 7.41 (t, J=7.32 Hz, 2 H) 7.45 - 7.50 (m, 2 H) 7.58 - 7.75 (m, 2 H) 8.37 (d, J=4.39 Hz, 1 H)
D) 3-Fluoro-4-hydroxv-N-methylbenzamide
4-(Benzyloxy)-3-fluoro-N-methylbenzamide (1.03 g, 3.97 mmol) was suspended in éthanol (20 mL) in a Parr bottle. 10% Palladium on carbon (80 mg) in about 1.5 mL of water was added under a steady stream of nitrogen. The reaction was shaken under a 50 psi atmosphère of hydrogen at room température for 64 hours. The reaction mixture was carefully filtered through a pad of Celite® washing with copious amounts of ethyl acetate. The filtrate was concentrated under reduced pressure to give the desired product (628 mg, 93%) as a light brownish yellow solid. 1H NMR (500 MHz, deuterochloroform) delta ppm 3.02 (d, J=4.88 Hz, 3 H) 7.05 (t, J=8.42 Hz, 1 H) 7,44 (d, J=9.76 Hz, 1 H) 7.60 (dd, J=11.10, 2.07 Hz, 1 H)
Préparation 31 B: 3-Fluoro-4-hydroxy-N,N-dimethvlbenzamide
OH
F
I
3-Fluoro-4-hydroxybenzoic acid (2.00 g, 12.8 mmol), dimethylamine hydrochloride (4.28 g, 20.5 mmol), 1-hydroxy benzotriazole monohydrate (1.96 g, 12.8 mmol), and diisopropylethyl amine (4.5 mL, 26 mmol) were combined in dichloromethane. 1-Ethyl-
3-(3-dimethylaminopropyl) carbodiimide hydrochloride (3.93 g, 20.5 mmol) was added, and the reaction vessel was flushed with nitrogen, capped, and stirred overnight at room température. The reaction was diluted with dichloromethane and 1M phosphoric acid. The precipitate that formed was filtered off and the dichloromethane layer was washed with dilute aqueous sodium bicarbonate and brine, dried over sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by flash chromatography eluting with 65% ethyl acetate in heptanes to give the desired product (218 mg, 9%). LC/MS (ES+): 184.1 (M+1)
Préparation 32: tert-Butyl 3-Hvdroxv-4,4-dimethoxypiperidine-1-carboxylate
tert-Butyl 4-oxo-1-piperidinecarboxylate (2.00 g, 10 mmol) was dissolved in methanol (20 mL) and cooled to 0 degrees Celsius. Powdered potassium hydroxide (1.26 g, 22.1 mmol) was added. lodine (2.8 g, 11 mmol) was dissolved in methanol (25 mL) and was added drop wise to the reaction over 45 minutes. The reaction was then slowty warmed up to room température and stirred for 16 hours. The reaction was concentrated and toluene (50 mL) was added. The resulting solids were filtered off and washed with toluene. The filtrate was concentrated under reduced pressure and the residue was purified by flash chromatography eluting with a gradient from 30% to 100% ethyl acetate in heptane to give terf-butyl 3-hydroxy-4,4-dimethoxypiperidine-1-carboxylate (1.89 g, 72%). 1H NMR (deuteromethanol, 400 MHz) delta ppm 4.06-4.00 (m, 1H), 3.99-3.91 (m, 1H), 3.80-3.73 (m, 1H), 3.29 (s, 3H>, 3.28 (s, 3H), 3.22-3.10 (br m, 1H), 2.95-2.80 (br m, 1H), 1.91-1.77 (m, 2H), 1.50 (s, 9 H).
Préparation 33: tert-Butyl 3-Hydroxv-4-oxopiperidine-1-carboxylate
tert-Butyl 3-hydroxy-4,4-dimethoxypiperidine-1 -carboxylate (6,70g, 26 mmol) was dissolved in acetone (135 mL), and p-toluene sulfonic acid (244 mg, 1,28 mmol) was added. The reaction was stirred at room température for 16 hours. The mixture was concentrated and the resulting residue was dissolved in tert-butyl methyl ether and washed with saturated aqueous sodium bicarbonate solution. The organic layer was dried over sodium sulfate, filtered and the filtrate was concentrated under reduced pressure to give tert-butyl 3-hydroxy-4-oxopiperidine-1-carboxylate as an oil (4.67 g, 69%). GC/MS (method 1): R, = 4.95 min; MS (ESIpos): m/z = 159 [M-tBu]+.
Préparation 34: tert-Butyl 4-Hvdrazino-3-hvdroxypiperidine-1-carboxylate
HO
H2N HN tert-Butyl 3-hydroxy-4-oxopiperidine-1-carboxylate (5.50 g, 26 mmol) was dissolved in methanol (120 mL) and degassed with a stream of nitrogen in a capped Parr Shaker bottle. Hydrazine-hydrochloride (1.44 mg, 21 mmol) was dissolved in water (20 mL) and added to the reaction. The flask was rinsed with 5 mL of water and it was also added to the reaction. 10% Platinum on carbon catalyst (500 mg) was slurried in water and added to the reaction mixture. The mixture was shaken under at 50 psi atmosphère of hydrogen at room température for 16 hours. The reaction was filtered through a pad of Celite® washing with methanol. The filtrate was concentrated under reduced pressure and then diluted with heptanesand concentrated under reduced pressure to give the desired product, tert-butyl 4-hydrazino-3-hydroxypiperidine-1 -carboxylate.
Préparation 35: tert-Butyl 4-r5-Amino-4-(ethoxycarbonvl)-1H-pyrazol-1-vll-3hvdroxypiperidine-1 -carboxylate
HO ô NH2
tert-Butyl 4-hydrazino-3-hydroxypiperidine-1-carboxylate (5.30 g, 20 mmol) and ethyl(ethoxymethlene)cyanoacetate (3.42 g, 19.8 mmol) were combined in absolute éthanol (170 mL). Sodium acetate trihydrate (10.90 g, 79.2 mmol) was added, and the reaction mixture was heated at reflux for 4 hours. The reaction was cooled to room température, concentrated under reduced pressure and the resulting residue was diluted with water and ethyl acetate. The organic layer was dried over sodium sulfate, filtered and the filtrate was concentrated under reduced pressure. The crude oil was purified by flash chromatography eluting with a gradient from 10% to 100% ethyl acetate in heptanes togive tert-butyl 4-[5-amino-4-(ethoxycarbonyl)-1 H-pyrazol-1-yl]-3hydroxypiperidine-1-carboxylate.
Préparation 36: tert-Butyl 4-i5-amino-4-(ethoxvcarbonvl)-1 H-pyrazol-1-vll-3-
tert-Butyl 4-[5-amino-4-(ethoxycarbonyl)-1 H-pyrazol-1 -yl]-3-hydroxypiperidine-1 carboxylate (1.71 g, 4.82 mmol) was dissolved in dichloromethane (50 mL) and cooled to -78 degrees Celsius. Diethylaminosulfur trifluoride (0.710 mL, 0.58 mmol) was added drop wise, and then warmed up to 0 degrees Celsius for 25 minutes. The reaction solution was cooled to -78 degrees Celsius and methanol (10 mL) carefully added. The reaction was concentrated under reduced pressure and the residue was purified by flash chromatography eluting with a gradient from 10% to 100% ethyl acetate in heptanes to give tert-butyl 4-[5-amino-4-(ethoxycarbonyl)-1H-pyrazol-1-yl]-3fluoropiperidine-1 -carboxylate.
Préparation 37: tert-Butvl 4-f5-bromo-4-(ethoxvcarbonyl)-1 H-pyrazol-1-yl]-3fluoropiperidine-1 -carboxylate
tert-Butyl 4-[5-amino-4-(ethoxycarbonyl)-1H-pyrazol-1-yl]-3-fluoropiperidine-1carboxylate (710 mg, 1.99 mmol) was dissolved in acetonitrile (25 mL). Copper (II) bromide (539 mg, 2.39 mmol) was added and the reaction was heated to 60 degrees
Celsius. fert-Butyl nitrile (0.315 mL, 2.9 mmol) was added drop wise and the mixture was heated at 65 degrees Celsius for 15 minutes. The reaction was cooled to room température and poured into cold 1N hydrochloric acid and extracted with ethyl acetate (2x). The combined organic extracts were washed with saturated aqueous sodium bicarbonate and brine and dried over sodium sulfate, filtered and the filtrate was concentrated under reduced pressure. The crude residue was purified by flash chromatography eluting with a gradient from 10% to 50% ethyl acetate in heptanes to give tert-butyl 4-[5-bromo-4-(ethoxycarbonyl)-1 H-pyrazol-1-yl]-3-fluoropiperidine-1carboxylate (320 mg, 38%). 1H NMR (500 MHz, deuterochloroform) delta ppm 1.37 (t, 3 H) 1.49 (s, 9 H) 1.98 (d, J=13.42 Hz, 1 H) 2.12 - 2.26 (m, 1 H) 2.90 (br. s., 2 H) 4.18 (br. s., 1 H) 4.33 (q, J=7.24 Hz, 2 H) 4.44 - 4.70 (m, 2 H) 4.85 - 5.05 (m, 1 H) 8.04 (s, 1 H)
Préparation 38: Ethyl 5-cvano-1-(3-fluoropiperidin-4-vl)-1H-pvrazole-4-carboxylate
tert-Butyl 4-[5-bromo-4-(ethoxycarbonyl)-1H-pyrazol-1-yl]-3-fluoropiperidine-1carboxylate (185 mg, 0.31 mmol), 1,l'bis (dîphenylphosphino)ferrocene (18 mg, 0.032 mmol), zinc dust (18 mg, 0.27 mmol), zinc cyanide (39.1 mg, 0.33 mmol) and 10% palladium black (19.2 mg, 0.021 mmol) were combined in dimethylacetamide (3 mL) in a microwave vial. The reaction mixture was degassed with nîtrogen and heated at 170
Y degrees Celsius for 4.5 hours. The reaction mixture was cooled to room température and diluted with ethyl acetate. The reaction was filtered through a pad of Celite® and the filtrate was diluted with water and extracted with ethyl acetate (2x). The combined organic extracts were washed with water then brine and dried over sodium sulfate, filtered and the filtrate was concentrated under reduced pressure. The crude residue was purified by flash chromatography eluting with a gradient from 10% to 100% ethyl acetate in heptanes to give ethyl 5-cyano-1-(3-fluoropiperidin-4-yl)-1H-pyrazole-4carboxylate (80 mg, 98%).
Préparation 39: tert-Butyl 4-r5-cvano-4-(ethoxvcarbonvl)-1H-pyrazol-1-vll-3fluoropiperidine-1 -carboxylate
Ethyl 5-cyano-1-(3-fluoropiperidin-4-yl)-1H-pyrazole-4-carboxylate (60 mg, 0.22 mmol) was dissolved in tetrahydrofuran (3 mL) and triethylamine (40 uL, 0.27 mmol) was added. Di-tert-butyl dicarbonate (50 mg, 0.225 mmol) was added and the reaction was stirred at room température under nitrogen for 3 hours. The reaction was concentrated under reduced pressure and the residue was purified by flash chromatography eluting with a gradient from 10% to 100% ethyl acetate in heptanes to give fert-butyl 4-[5cyano-4-(ethoxycarbonyl)-1H-pyrazol-1-yl]-3-fluoropiperidine-1-carboxylate as an oil (52 mg, 63%).
Préparation 40: 1-f1-(tert-Butoxycarbonvl)-3-fluoropiperidin-4-vl1-5-cvano-1H-pyrazole-
4-carboxylic acid
terf-Butyl 4-[5-cyano-4-(ethoxycarbonyl)-1H-pyrazol-1-yl]-3-fluoropiperidine-1carboxylate (80 mg, 0.22 mmol) was dissolved in tetrahydrofuran (2.5 mL), water (1.5 mL) and methanol (0.4 mL). The solution was cooled to 0 degrees Celsius and lithium
hydroxide monohydrate (19 mg, 0.436 mmol) was added. The reaction was slowly allowed to warm up to room température over 2.5 hours. The reaction mixture was concentrated; the residue was dissolved in water and extracted with ethyl acetate and methyl tert-butyl ether. The organic layer was extracted with water. The combined 5 aqueous extracts were acidified with 1N aqueous sodium bisulfate to pH 2. The acidic solution was extracted with ethyl acetate (3x) and the extracts were washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated under reduced pressure to give 1-[1-(tert-butoxycarbonyl)-3-fluoropiperidin-4-yl]-5-cyano-1H-pyrazole-
4-carboxylic acid as a white solid.
Préparation 41: tert-Butvl 4-[5-cyano-4-(hvdroxvmethyl)-1 H-pyrazol-1-vll-3fluoropiperidine-1 -carboxvlate
Freshly recrystallized (from heptanes) cyanuric chloride (78 mg, 0.414 mmol) was dissolved in dimethoxyethane (2 mL) and 4-methyl-morpholine (0.020 mL, 0.215 mmol) was added. To this gummy solution was added 1-[1-(tert-butoxycarbonyl)-3fluoropiperidin-4-yl]-5-cyano-1H-pyrazole-4-carboxylic acid (70 mg, 0.21 mmol) dissolved in dimethoxyethane (2 mL). The reaction was heated at 60 degrees Celsius for 3 hours. The reaction was cooled to room température and filtered through a pad of Celite® washing with dimethoxyethane. The filtrate was cooled to 0 degrees Celsius and a solution of sodium borohydride (17 mg, 0.474 mmol) dissolved in water (0.4 mL) was added very slowly (drop wise). Once addition was complété, the reaction was allowed to warm up to room température for 2.5 hours. The reaction solution was further diluted with water and acidified to pH 2.5 using 1 A4 sodium bisulfate. The aqueous layer was extracted with ethyl acetate (2x) and the combined organic layers were dried over sodium sulfate, filtered and the filtrate was concentrated under reduced pressure. The crude residue was purified by flash chromatography eluting with a gradient from 10% to 100% ethyl actetate in heptanes to give ferf-butyl 4-[5-cyano-4(hydroxymethyl)-1H-pyrazol-1-yl]-3-fluoropiperidine-1-carboxylate as an oil (28 mg, 42%).
Préparation 42: tert-Butyl 4-(5-cyano-4-([(methvlsulfonyl)oxv1methvlÎ-1H-pyrazol-1-vl)3-fluoropiperidine-1 -carboxylate
fert-Butyl 4-[5-cyano-4-(hydroxymethyl)-1 H-pyrazol-1 -yl]-3-fluoropiperidine-1 5 carboxylate (28 mg, 0.086 mmol) was dissolved in dichloromethane (3 mL). Triethylamine (0.036 mL, 0.258 mmol) was added and the mixture was cooled to 0 degrees Celsius. Methanesulfonic anhydride (20 mg, 0.112 mmol) was added drop wise and slowly allowed to warm up to room température over 2 hours. Dichloromethane and saturated aqueous sodium bicarbonate were added to the reaction solution and the biphasic solution was separated. The aqueous layer was extracted with dichloromethane (2x) and the combined organic extracts were passed through a plug of cotton. The filtrate was concentrated under reduced pressure to give fert-butyl 4-(5-cyano-4-{[(methylsulfonyl)oxy]methyl}-1 H-pyrazol-1-yl)-3-fluoropiperidine1 -carboxylate as an oil (33 mg, 95%). 1H NMR (500 MHz, deuterochloroform) delta ppm 1.25 -1.30 (m, 2 H) 1.50 (s, 9 H) 2.01 - 2.06 (m, 1 H) 2.75 - 2.85 (m, 2 H) 3.08 (s,
H) 4.64 - 4.74 (m, 1 H) 4.79 - 4.98 (m, 2 H) 5.26 (s, 2 H) 7.75 (s, 1 H)
Préparation 43: Isomers of tert-Butvl-3-fluoro-4-hvdroxvpiperidine-1-carboxylate (B and Ç)
The experimental details are described in detail in Scheme 4 below.
Scheme 4
Step C
OH
N i
Step D
Step A) tert-Butvl-4-r(trimethvlsilvl)oxvl-3,6-dihydropyridine-1 (2H)-carboxvlate
To a solution of /V-tert-butoxycarbonyl-4-piperidone (30.0 g, 0.15 mol) in dry N,Ndimethylformamide (300 mL) at room température was added trimethylsilyl chloride (22.9 mL, 0.18 mol) and triethylamine (50.4 mL, 0.36 mol) successively via addition funnels. The resulting solution was heated at 80 degrees Celsius overnight and then cooled to room température. The reaction mixture was diluted with water and heptane.
The layers were separated, and the aqueous layer was extracted with heptane. The combined heptane layers were washed sequentially with water and brine and then dried over magnésium sulfate. The mixture was filtered, and the filtrate concentrated under reduced pressure to give the crude product as a yellow oil. The oil was purified by passtng it through a plug of silica gel eluting with 9:1 heptane/ethyl acetate to give the title compound as a colorless oil (33.6 g, 82%). 1H NMR (400 MHz, deuterochloroform) delta 4.78 (br s, 1H), 3.86 (br s, 2H), 3.51 (t, 2H), 2.09 (br s. 2H), 1.45 (s, 9H), 0.18 (s, 9H).
Step B) tert-Butyl-S-fluoro-^oxopiperidine-l-carboxylate
O
N i
0^0^
To a stirred solution of fert-butyl-4-[(trimethylsilyl)oxy]-3,6-dihydropyridine-1(2H)carboxylate (28.8 g, 0.11 mol) in acetonitrile (300 mL) at room température was added Selectfluor™ (41.4 g, 0.12 mol). The resulting pale yellow suspension was stirred at room température for 1.5 hours. Saturated aqueous sodium bicarbonate (300 mL) and ethyl acetate (300 mL) were added, and the layers were separated. The aqueous layer was extracted twice with ethyl acetate, and ail the organic layers were combined and washed sequentially with saturated aqueous sodium bicarbonate and brine and then dried over magnésium sulfate. The mixture was filtered, and the filtrate was concentrated under reduced pressure to give the crude product as a pale yellow oil. Purification of this material by repeated column chromatography on silica gel with heptane/ethyl acetate gradient (2:1 to 1:1 ) gave the title compound as a white solid (15.5 g, 67%). 1H NMR (400 MHz, deuterochloroform): delta 4.88 (dd, 0.5 H), 4.77 (dd, 0.5H), 4.47 (br s, 1 H), 4.17 (ddd, 1H), 3.25 (brs, 1H), 3.23 (ddd, 1H), 2.58 (m, 1H), 2.51 (m, 1H), 1.49 (s, 9H).
Alternatively Step B can be performed as follows, isolating the hydrate of the ketone. To a stirred solution of tert-butyl-4-[(trimethylsilyl)oxy]-3,6-dihydropyridine-1(2H)carboxylate (41.3 g, 0.15 mol) in acetonitrile (500 mL) at room température was added Selectfluor™ (56.9 g, 0.16 mol). The resulting pale yellow suspension was stirred at room température for 4 hours 10 minutes. Saturated aqueous sodium bicarbonate and ethyl acetate were added, and the layers were separated. The aqueous layer was extracted twice with ethyl acetate, and ail the organic layers were combined and washed sequentially with saturated aqueous sodium bicarbonate and brine and then dried over magnésium sulfate. The mixture was filtered, and the filtrate was concentrated under reduced pressure to give the crude tert-butyl-3-fluoro-4oxopiperidine-1-carboxylate as white solid. The crude tert-butyl-3-fluoro-4oxopiperidine-1-carboxylate was suspended in tetrahydrofuran (120 mL) and water (120 mL) was added. The resulting solution was stirred at room température for 5.5 hours and then concentrated under reduced pressure. The residue was dried under high vacuum, transferred to an Erlenmeyer flask, and suspended in dichloromethane (250
mL). The resulting suspension was stirred for 5 minutes and the solids collected by filtration using a sintered glassfunnel. The resulting filter cake was thoroughly washed with dichloromethane (200 mL), a 1:1 mixture of dichloromethane (200 mL) and heptane (100 mL). The solid was then dried under high vacuum to provide tert-butyl 3-fluoro-4,4dihydroxypiperidine-1-carboxylate (26.4 g). 1H NMR (500 MHz, deutero dimethyl sulfoxide) delta 1.38 (s, 9 H), 1.49-1.52 (m, 1 H), 1.63-1.68 (m, 1 H), 2.82 -3.20 (m, 2 H)
3.75 (br, 1 H), 3.97 (br, 1 H), 4.12 (d, J — 45, 1 H), 5.92 (s, 1 H), 5.97 (s, 1 H).
Step C) Isomers of (R* )-ferf-Butvl-3-(S)-fluoro-4-(R)-hvdroxypiperidine-1-carboxylate (racemic)
OH
OH
To a solution of tert-butyl-3-fluoro-4-oxopipendine-1-carboxylate (15.5 g, 71.3 mmol) in methanol (150 mL) atO degrees Celsius was added sodium borohydride (3.51 g, 93.7 mmol). The resulting mixture was stirred at 0 degrees Celsius for 2 hours and then allowed to warm to room température. Saturated aqueous ammonium chloride (200 mL) was added, and the mixture was extracted three times with ethyl acetate. The combined extracts were washed with brine and dried over magnésium sulfate. The mixture was filtered, and the filtrate was concentrated under reduced pressure to give the crude product mixture which was purified by column chromatography on silica gel eluting with heptane-ethyl acetate (3:2 -1:1) to give the first eluting product, terf-butyl(3,4-trans)-3-fluoro-4-hydroxypiperidine-1-carboxylate (compound C, Scheme 4) (3.81 g, 24%), as a pale yellow oil which solidified on standing to a white solid. 1H NMR (400 MHz, deuterochloroform) delta 4.35 (ddd, 0.5 H), 4.18 (ddd, 0.5 H), 4.15 (br s, 1H), 3.89-3.74 (m, 2H), 2.97 (br s, 1H), 2.93 (ddd, 1 H), 2.47 (s, 1 H), 2.05-1.92 (m, 1 H), 1.58-
1.46 (m, 1H), 1.44 (s, 9H).
The second eluting compound, tert-butyl-(3,4-c/s)-3-fluoro-4-hydroxy-piperidine1-carboxylate (compound B, Scheme 4) (10.57 g, 68%) was then isolated as a white solid. 1H NMR (400 MHz, deuterochloroform) delta 4.69 - 4.65 (m, 0.5H), 4.53-4.49 (m, 0.5H), 3.92 - 3.86 (m, 2H), 3.69 (br s, 1 H), 3.39 (br s, 1 H), 3.16 (br s, 1 H), 2.13 (s, 1 H), 1.88- 1.73 (m, 2H), 1.44 (s, 9H).
Alternatively Step C can be performed starting with the hydrate tert-butyl 3-fluoro-
4,4-dihydroxypiperidine-1-carboxylate (Step 2) as follows.
To a stirred solution of tert-butyl 3-fluoro-4,4-dihydroxypiperidtne-1-carboxylate (20.0 g, 85 mmol) in tetrahydrofuran (500 mL) at -35 degrees Celsius was added a solution of L-Selectride® in tetrahydrofuran (170 mL, 1 M, 170 mmol) drop-wise over 30 minutes. The reaction mixture was warmed to 0 degree Celsius over 1,5 h. The reaction mixture was quenched with saturated aqueous ammonium chloride (150 mL) and vigorously stirred for 15 minutes. To this 0 degree Celsius mixture was added pH 7 phosphate buffer (150 mL), followed by drop-wise addition of a 35% aqueous hydrogen peroxide solution (150 mL). The resulting mixture was stirred for 30 minutes and diluted with ethyl acetate. The organic layer was separated and sequentially with water, saturated aqueous sodium thiosulfate and brine. The organic layer was then dried over anhydrous magnésium sulfate, filtered and the filtrate was concentrated under reduced pressure give the crude product mixture which was purified by column chromatography on silica gel [ combiflash ISCO330 g column] eluting with heptane-ethyl acetate (10 to 60% gradient) to give tert-butyl-(3,4-c/s)-3-fluoro-4-hydroxypiperidine-1-carboxylate (13.9 g).
Step D) Enantiomers of tert-butvl-(3.4-c/s)-3-fluoro-4-hvdroxv-piperidine-1-carboxylate
A 1 gram sample of racemic tert-butyl-(3,4-c/s)-3-fluoro-4-hydroxy-piperidine-1carboxylate was purified into its enantiomers via preparatory high pressure liquid chromatography utilizing a Chiralpak AD-H column (10 x 250 mm) with a mobile phase of 90:10 carbon dioxide and éthanol respectively at a flow rate of 10 mL/minute. The wavelength for monitoring the séparation was 210 nM. The analytical purity of each enantiomer was determined using analytical high pressure chromatography using a Chiralpak AD-H (4.6 mm x 25 cm) column with an isocratic mobile phase of 90:10 carbon dioxide and éthanol respectively at a flow rate of 2.5 mL/minute. The wavelength for monitoring the peaks was 210 nm. The following two isomers were obtained: Compound E, Scheme 4) (3S,4/?)-tert-Butvl 3-fluoro-4-hydroxypiperidine-1 -carboxylate, enantiomer 1 (363 mg): R, ~ 2.67 min (100% ee) (optical rotation in dichloromethane = +21.2 degrees) and
OH
Compound D, Scheme 4) (3R,4S)-tert-Butyl 3-fluoro-4-hydroxypiperidine-1 -carboxylate, enantiomer 2 (403 mg): R( = 2.99 min (88% ee).
OH
The absolute stereochemistry of the tert-buty1-(3,4-c/s)-3-fluoro-4-hydroxy-piperidine-1carboxylate isomers was determined by making a (1 S)-(+)-camphorsulfonic acid sait of
5-(6-((3S,4R)-3-fluoropiperidin-4-yloxy)-5-methyipyrimidin-4-yl)-1 -methyl-1,4,5,6tetrahydropyrrolo[3,4-c]pyrazole (see by analogy the préparation in racemic form below), prepared using enanantiomer 1 above.
Préparation of 5-(6-f[(3,4-c/s)-3-fluoropiperidin-4-vlloxvÎ-5-methvlpvrimidin-4-ylÎ-1 methyl-1,4,5,6-tetrahvdropvrrolo[3,4-c|pvrazole (racemic)
A. Préparation of 5-(6-Chloro-5-meÎhvlpvrimidin-4-yl)-1-methyl-1,4,5,6tetrahydropyrrolo[3,4-c1pyrazole
1-Methyl-1,4,5,6-tetrahydropyrrolo[3,4-c]pyrazole bis-hydrochloride sait (2.00 g,
10.2 mmol) and 4,6-dichloro-5-methylpynmidine (1.66 g, 10.2 mmol) were suspended in tetrahydrofuran (51 mL) at room température. To this was added triethylamine (4.41 mL, 31.6 mmol), which caused cloudiness in the mixture and led to a brown solid sticking to the flask walls. This mixture was stirred at room température for 4 hours and then heated 50 degrees Celsius for an additional 19 hours. The reaction mixture was cooled to room température and diluted with water (100 mL). This mixture was extracted with ethyl acetate (3 x 100 mL). The organic extracts were pooled, washed
with brine, dried over sodium sulfate, and filtered. The filtrate was reduced to dryness under vacuum to yield the title compound as a light brown solid (1.95 g, 78%), which was used in the next step without further purification.
1H NMR (500 MHz, deuterochloroform) delta 2.54 (s, 3 H) 3.88 (s, 3 H) 4.90 (app. d, J=3.66 Hz, 4 H) 7.28 (s, 1 H) 8.29 (s, 1 H).
B. Préparation of tert-Butyl (3,4-c/s)-3-fluoro-4-([5-methvl-6-(1-methyl-4,6dihvdropvrrolo[3,4-clpvrazol-5(1H)-yl)pyrimidin-4-vl1oxy}piperidine-1-carboxylate (racemic)
A mixture of tert-butyl (3,4-c/s)-3-fluoro-4-hydroxypiperidine-1-carboxylate (1.67 g, 7.62 mmol) and 5-(6-chloro-5-methylpyrimidin-4-yl)-1 -methyl-1,4,5,6-tetrahydropyrrolo[3,4cjpyrazole prepared above (900 mg, 3.60 mmol) was dissolved in 1,4-dioxane (20 mL) and was heated to 105 degrees Celsius. After heating for 10 minutes, ail the materials 15 had gone into solution, and sodium bis(trimethylsilyl)amide (4.3 mL, 4.3 mmol, 1M in toluene) was rapidly added to the mixture, resulting in a cloudy yellow mixture that was then stirred for 2 hours at 105 degrees Celsius. The reaction was then cooled to room température and quenched by adding an equal volume mixture of water and saturated aqueous sodium bicarbonate solution. The mixture was extracted with ethyl acetate (3 20 x 15 mL). The combined organic extracts were washed with brine, dried over sodium sulfate, and filtered. The filtrate was concentrated under vacuum to give a yellow residue that was purified by column chromatography on silica gel eluting with 60 to 100% ethyl acetate in heptane. A mixture of the title compound and the starting 5-(6chloro-5-methylpyrimidin-4-yl)-1-methyl-1,4,5,6-tetrahydropyrrolo[3,4-c]pyrazole was isolated as a white solid (1.20 g) and was used without further purification in subséquent reactions.
A batch of crude tert-butyl (3,4-c/s)-3-fluoro-4-{[5-methyl-6-(1-methyl-4,6dihydropyrrolo[3,4-c]pyrazol-5(1 H)-yl)pyrimidin-4-yl]oxy}piperidine-1-carboxylate from a separate reaction, run under the same conditions, was purified by HPLC. The crude 30 sample (9.5 mg) was dissolved in dimethyl sulfoxide (1 mL) and purified by préparative reverse phase HPLC on a Waters XBridge Ci819 x 100 mm, 0.005 mm column, eluting
with a linear gradient of 80% water/acetonitrile (0.03% ammonium hydroxide modifier) to 0% water/acetonitrile in 8.5 minutes, followed by a 1.5 minute period at 0% water/acetonitrile; flow rate: 25mL/minute. The title compound (5 mg) was thus obtained. Analytical LCMS: rétention time 2.81 minutes (Waters XBridge Cw 4.6 x 50 mm, 0.005 mm column; 90% water/acetonitrile linear gradient to 5% water/acetonitrile over4.0 minutes, followed by a 1 minute period at 5% water/acetonitrile; 0.03% ammonium hydroxide modifier; flow rate: 2.0 mL/minute); LCMS (ES+) 433.2 (M+1).
C. Préparation of 5-(6-ffî3,4-c/s)-3-fluoropiperidin-4-vlloxvÎ-5-methvlpvrimidin-4vl)-1-methvl-1,4,5,6-tetrahydropvrrolo[3,4-clpvrazole (racemic)
NH
Crude tert-butyl (3,4-c/s)-3-fluoro-4-([5-methyl-6-(1-methyl-4,6-dihydropyrrolo[3,4-
c]pyrazol-5(1H)-yl)pyrimidin-4-yl]oxy}piperidine-1-carboxylate (1.20 g) prepared above was dissolved in dichloromethane (12 mL) and to this solution was added trifluoroacetic acid (5 mL). The reaction was stirred at room température for 1 hour. The solvent was removed under vacuum, and the residue was dissolved in water (50 mL) and 1N aqueous hydrochloric acid solution (10 mL). The mixture was extracted with dichloromethane (10 x 30 mL). The aqueous layer was then brought to pH 12 by the addition of 1N aqueous sodium hydroxide solution (20 mL) and was extracted three times with dichloromethane (40 mL). The combined organic extracts were washed with brine, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to afford 5-(6-{[(3,4-c/s)-3-fluoropiperidin-4-yl]oxy}-5-methylpyrimidin4-yl)-1-methyl-1,4,5,6-tetrahydropyrrolo[314-c]pyrazole (0.72 g, 60% over two steps) as a white solid that was used without additional purification.
1H NMR (500 MHz, deuterochloroform) delta 1.84 - 2.08 (m, 2 H) 2.33 (s, 3 H) 2.69-2.84 (m, 1 H) 2.83-3.01 (m, 1 H) 3.16 (d, J=13.66 Hz, 1 H) 3.27 - 3.44 (m, 1 H)
3.86 (s, 3 H) 4.78-4.91 (m, 1 H) 4.86 (d, J=1.95 Hz, 2 H) 4.88 (d, J=1.95 Hz, 2 H) 5.21 5.32 (m, 1 H) 7.26 (s, 1 H) 8.18 (s, 1 H); LCMS (ES+) 333.4 (M+1).
Please note: Example number begins at 11.
Example 11: Isopropyl 4-{5-cvano-4-[(2!4-difluorophenoxy)methyil-1 H-pyrazol-1yl}piperidine-1-carboxylate
Isopropyl 4-(5-cyano-4-((methylsulfonyloxy)methyl)-1 H-pyrazol-1 -yl)piperidine-1 carboxylate (Préparation 10) (166.5 mg, 0.449 mmol), 2,4-difluorophenol (0.052 mL,
0.539 mmol), and césium carbonate (293 mg, 0.898 mmol ) were placed in microwave vial, dissolved in acetonitrile (3 mL), and heated in a microwave reactor at 110 degrees Celsius for 20 minutes. The mixture was cooled to room température and concentrated under vacuum, diluted with 1 N sodium hydroxide solution, and extracted three times with dichloromethane. The combined organic extracts were washed with brine, dried over sodium sulfate, filtered, and the filtrate was concentrated under vacuum. The crude material was purified by préparative reverse-phase HPLC on a Waters Atlantis Cie column 4.6 x 50 mm, 0.005 mm eluting with a gradient of water in acetonitrile (0.05 % trifluoroacetic acid modifier) to give isopropyl 4-{5-cyano-4-[(2,4difluorophenoxy)methyl]-1 H-pyrazol-1 -yl}piperidine-1 -carboxylate. Analytical LCMS:
rétention time: 3.62 minutes (Waters Atlantis Ci8 4.6 x 50 mm, 0.005 mm; 95% water/acetonitrile linear gradient to 5% water/acetonitrile over 4.0min; 0.05 % trifluoroacetic acid modifier; flow rate 2.0mL/minute); LCMS (ES +): 405.18 (M + H).
Example 12: Isopropyl 4-(5-cvano-4-[(2-methylphenoxv)methvn-1H-pyrazol-120 vl}piperidine-1 -carboxylate
To a stirred solution of ortho-cresol (21 mg, 0.19 mmol) and isopropyl 4-(5-cyano-425 ((methylsulfonyloxy)methyl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate (Préparation 10) (60 mg, 0.16 mmol) in acetonitrile (1.6 mL) was added césium carbonate (106 mg, 0.32 mmol). The mixture was heated at reflux for 15 hours. After cooling to room température the crude material was concentrated to dryness in vacuo, and the residue was ta ken up in water and extracted 3 times with ethyl acetate (20 mL each extraction). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated to dryness under vacuum to give a tan residue (0.065 g, 100%). The crude sample was dissolved in dimethyl sulfoxide (1 mL) and purified by préparative reverse phase HPLC on a Waters Sunfire C-ie 19 x 100 mm, 0.005 mm column, eluting with a linear gradient of 80% water/acetonitrile to 0% water/acetonitrile in 8.5 minutes, followed by a 1.5 minute period at 0% water/acetonitrile (0.05% trifluoroacetic acid modifier); flow rate: 25mL/minute. Analytical LCMS: rétention time 3.82 minutes (Waters Atlantis C18
4.6 x 50 mm, 0.005 mm column; 95% water/acetonitrile linear gradient to 5% water/acetonitrile over 4.0 minutes, followed by a 1 minute period at 5% water/acetonitrile; 0.05% trifluoroacetic acid modifier; flow rate: 2.0 mL/minute); LCMS (ES+) 383.2 (M+1).
Example 13: 1-Methylcvclopropvl 4-f5-cvano-4-[(2.5-difluorophenoxv)methylMHpyrazol-1 -vl}piperidine-1 -carboxylate
A) terf-butyl 4-(5-cvano-4-((2,5-difluorophenoxv)methyl)-1H-pyrazol-1-vl)piperidine-1carboxylate
To a stirred solution of 2,5-difluorophenol (54 mg, 0.39 mmol) and tert-butyl 4-(5-cyano4-((methylsulfonyloxy)methyl)-1H-pyrazol-1-yl)piperidine-1 -carboxylate (Préparation 16) (126 mg, 0.33 mmol) in 3 mL of acetonitrile was added césium carbonate (214 mg, 0.66 mmol). The mixture was heated at reflux for 15 hours. The mixture was cooled to room température and diluted with ethyl acetate and water. The layers were separated and the aqueous phase was extracted with ethyl acetate. The combined organic phases were washed with brine, dried over magnésium sulfate, filtered, and the filtrate was concentrated in vacuo to give terf-butyl 4-(5-cyano-4-((2,5-difluorophenoxy)methyl)-1Hpyrazol-1-yl)piperidine-1-carboxylate which was used in the next step without purification.
B) 4-((2,5-Difluorophenoxy)methvl)-1 -(piperidin-4-yl)-1 H-pyrazole-5-carbonitrile
To a solution of fert-butyl 4-(5-cyano-4-((2,5-difluorophenoxy)methyl)-1H-pyrazol-1yl)piperidine-1-carboxylate (137 mg, 0.33 mmol) in 5 mL of dichloromethane was added
0.82 mL of hydrochloric acid (4 M in 1,4-dioxane). The mixture was stirred at room température for 2 hours beforethe mixture was concentrated in vacuo to give 4-((2,5difluorophenoxy)methyl)-1-(piperidin-4-yl)-1H-pyrazole-5-carbonitrile which was used in the next step without purification.
C) 1-Methylcyclopropyl 4-{5-cvano-4-f(2.5-difluorophenoxv)methyl1-1 H-pyrazol-1yl}piperidine-1 -carboxylate
To a stirred solution of 4-((2,5-difluorophenoxy)methyl)-1-(piperidin-4-yl)-1H-pyrazole-5carbonitrile (104 mg, 0.33 mmol) in 3.3 mL of dichloromethane was added triethylamine (0.18 mL, 1.3 mmol) followed by 1-methylcyclopropyl 4-nitrophenyl carbonate (see Préparation 26 and W009105717) (171 mg, 0.72 mmol) at room température. The resulting bright yellow mixture was stirred for 15 hours under a nitrogen atmosphère. The reaction mixture was diluted with dichloromethane and water. The layers were separated and the aqueous phase was extracted with dichloromethane. The combined organic phases were washed with saturated aqueous sodium bicarbonate, brine, dried over magnésium sulfate, filtered and the filtrate was concentrated in vacuo to give 225 mg of crude material. Part (45 mg) of this material was dissolved in dimethyl sulfoxide (0.9 mL) and purified by préparative reverse-phase HPLC on a Waters XBridge Ciecolumn 19 x 100 mm, 0.005 column eluting with a gradient of water in acetonitrile (0.03% ammonium hydroxide modifier). Analytical LCMS: rétention time 3.60 minutes (Atlantîs Cta 4.6 x 50 mm, 5 micrometer column; 95% water/acetonitrile linear gradient to 5% water/acetonitrile over 4 minutes; 0.05% trifluoroacetic modifier; flow rate 2.0 mL/minute; LCMS (ES+): 417.1 (M+H).
Example 14: 1-Methylcyclopropyl 4-{5-cvano-4-[(2.3-difluorophenoxv)methyi1-1Hpyrazol-1 -vl}piperidine-1 -carboxylate
The title compound was prepared using commercially available 2,3-diflurophenol, following procedures analogous to Example 13. The crude material (49 mg) was dissolved in dimethyl sulfoxide (0.9 mL) and purified by préparative reverse-phase
HPLC on a Waters XBridge C-i8 column 19 x 100 mm, 0.005 column eluting with a gradient of water in acetonitrile (0.03% ammonium hydroxide modifier). Analytical LCMS: rétention time 3.62 minutes (Atlantis Ci8 4.6 x 50 mm, 5 micrometer column; 95% water/acetonitrile linear gradient to 5% water/acetonitrile over 4 minutes; 0.05% trifluoroacetic modifier; flow rate 2.0 mL/minute; LCMS (ES+): 417.2 (M+H).
Example 15: 1-Methylcyclopropyl 4-{4“f(4-carbamovl-2-fluorophenoxv)methyll-5-cyano1 H-pyrazol-1 -yl)piperid ine-1 -carboxylate
A) ferf-Butyl 4-(4-((4-carbamoyl-2-fluorophenoxy)methvl)-5-cyano-1 H-pyrazol-1 vDpiperid ine-1-carboxylate
To a stirred solution of ferf-butyl 4-(5-cyano-4-(hydroxymethyl)-1 H-pyrazol-1yl)piperidine-1-carboxylate (Préparation 15) (200 mg, 0.65 mmol), 3-fluoro-4hydroxybenzamide (Préparation 23) (100 mg, 0.64 mmol) and triphenylphosphine (188 mg, 0.72 mmol) in 3 mL of 1,4-dioxane was added drop-wise diethyl azodicarboxylate (0.11 mL, 0.69 mmol). The resulting mixture was stirred overnight at room température before the mixture was concentrated in vacuo. The residue was purified by flash chromatography, eluting with a gradient of 30 to 70% ethyl acetate in heptane to give ferf-butyl 4-(4-((4-carbamoyl-2-fluorophenoxy)methyl)-5-cyano-1 H-pyrazol-1 yl)piperidine-1-carboxylate as a white solid (215 mg).
B) 4-((5-Cvano-1-(piperidin-4-vl)-1H-pvrazol-4-yl)methoxv)-3-fluorobenzamide
To a stirred solution of ferf-butyl 4-(4-((4-carbamoyl-2-fluorophenoxy)methyl)-5-cyano1 H-pyrazol-1-yl)piperidine-1-carboxylate (215 mg, 0.48 mmol) in 2 mL of dichloromethane was added 1 mL of trifluoroacetic acid at room température. After 1 hour the solution was concentrated in vacuo. The residue was purified by flash chromatography, eluting with a gradient mixture of 1 to 15% of methanol in
9l dichloromethane containing 2% of aqueous ammonia) to give 4-((5-cyano-1-(piperidin4-yl)-1H-pyrazol-4-yl)methoxy)-3-fluorobenzamide as a white solid (150 mg).
C) 1-Methylcyclopropyl 4-{4-[(4-carbamovl-2-fluorophenoxy)methyl1-5-cvano-1 Hpyrazol-1 -yl}piperidine-1 -carboxylate
To a stirred solution of 4-((5-cyano-1-(piperidin-4-yl)-1H-pyrazol-4-yl)methoxy)-3fluorobenzamide (40 mg, 0.12 mmol) in 1 mL of dichloromethane was added triethylamine (0.036 mL, 0.26 mmol), followed by 1-methylcyclopropyl 4-nitrophenyl carbonate (Préparation 26 and W009105717) (60 mg, 0.26 mmol) at room température. The resulting bright yellow mixture was stirred for 2 hours under a nitrogen atmosphère at 65 degrees Celsius. The reaction was cooled to room température, diluted with water and extracted twice with dichloromethane. The combined organic extracts were washed with saturated sodium bicarbonate, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo. The residue was purified by flash chromatography, eluting with a gradient of 40 to 90% ethyl acetate in heptane to give 1methylcyclopropyl 4-{4-[(4-carbamoyl-2-fluorophenoxy)methyl]-5-cyano-1 H-pyrazol-1 yl}piperidine-1-carboxylate as white solid (34 mg). 1H NMR (400 MHz, deuterochloroform) delta 0.59 - 0.67 (m, 2 H), 0.83 - 0.92 (m, 2 H), 1.54 (s, 3 H), 2.02 (d, J=4.10 Hz, 2 H), 2.04 - 2.22 (m, 2 H), 2.91 (br. s., 2 H), 4.11 - 4.43 (m, 2 H), 4.44 4.55 (m, 1 H), 5.15 (s, 2 H), 7.03 - 7.10 (m, 1 H), 7.52 - 7.62 (m, 2 H), 7.68 (s, 1 H). 1H NMR indicated the presence of less than 10% of what is believed to be the corresponding isopropyl carbamate dérivative (from the isopropyl 4-nitrophenyl carbonate contaminating the 1-methylcyclopropyl 4-nitrophenyl carbonate). LCMS (ES)
442.4 (M+1).
Example 16: 1-Methylcyclopropyl 4-(4-l(4-carbamovlphenoxv)methvl1-5-cvano-1 Hpvrazol-1-vl}piperidine-1-carboxylate
NHZ
The title compound was prepared using commercially available 4-hydroxybenzamide, following procedures analogous to Example 15.1H NMR (400 MHz, deuterochloroform) delta 0.57 - 0.67 (m, 2 H), 0.84 - 0.91 (m, 2 H), 1.56 (s, 3 H), 1.93 - 2.05 (m, 2 H), 2.05 2.19 (m, 2 H), 2.91 (t, 7=15.62 Hz, 2 H), 4.26 (br. s., 2 H), 4.44 - 4.55 (m, 1 H), 5.09 (s.
2 H), 6.96 - 7.04 (m, 2 H), 7.66 (s, 1 H), 7.75 - 7.82 (m, 2 H). 1H NMR indicated the presence of less than 10% of what is believed to be the corresponding isopropyl carbamate dérivative (from the isopropyl 4-nitrophenyl carbonate contaminating the 1methylcyclopropyl 4-nitrophenyl carbonate). LCMS (ES) 424.4 (M+1).
Example 17:1-Methylcyclopropyl 4-(5-cvano-4-((4-cvanophenoxv)methyl)-1H-pyrazol-1yl)piperidine-1-carboxylate
The title compound was prepared using commercially available 4-hydroxybenzonitrile, following procedures analogous to Example 15. The purification of the crude reaction mixture was performed by flash chromatography, eluting with a gradient mixture of ethyl acetate in heptane (0 to 100% ethyl acetate). 1H NMR (500 MHz, deuterochloroform) delta 0.60 - 0.70 (m, 2 H), 0.84 - 0.94 (m, 2 H), 1.23 -1.31 (m, 1 H), 1.56 (s, 3 H), 2.01 20 2.15 (m, 4 H), 2.93 (m, 2 H), 4.11 - 4.37 (m, 1 H), 4.49 - 4.55 (m, 1 H), 5.10 (s, 2 H),
7.03 (d, J=8.78 Hz, 2 H), 7.63 (d, J=8.78 Hz, 2 H), 7.67 (s, 1 H).
Exampie 18: Isopropyl 4-(4-((4-(1 H-pvrazol-1-vl)phenoxy)methyl)-5-cvano-1H-pyrazol-1vl )pipe rid i ne-1 -carboxvlate
The title compound was prepared using 4-(1H-pyrazol-1-yl)phenol (WO 2003072547 ), following a procedure analogous to Example 12. The purification of the crude reaction mixture was performed by flash chromatography, eluting with a gradient mixture of ethyl acetate in heptane (0 to 100% ethyl acetate). 1H NMR (500 MHz, deuterochloroform) delta 1.28 (d, 7=6.34 Hz, 6 H), 2.01 - 2.09 (m, 2 H), 2.17 (m, 2 H), 2.91 - 2.99 (m, 2 H),
4.37 (m, 2 H), 4.50 - 4.58 (m, 1 H), 4.93-4.98 (m, 1 H), 5.11 (s, 2 H), 6.47 (t, 7=2.07 Hz, 1 H), 7.07 (d, 7=9.03 Hz, 2 H), 7.64 (d, 7=9.03 Hz, 2 H), 7.70 (s, 1 H), 7.72 (d, 7=1.71
Hz, 1 H), 7.86 (d, 7=2.44 Hz, 1 H). LCMS (ES) 435.4(M+1 ).
Example 19: Isopropyl 4-(5-cvano-4-((2-fluoro-4-(1H-tetrazol-5-vl)phenoxy)methyl)-1Hpyrazol-1-yl)piperidine-1-carboxylate and Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2Htetrazol-5-vl)phenoxv)methvl)-1H-pyrazol-1-vl)piperidine-1-carboxylate
A) Isopropyl 4-(5-CYano-4-((2-fluoro-4-(1-((2-(trimethylsilyl)ethoxy)methyl)-1 H-tetrazol-5yl)phenoxv)methvl)-1H-pvrazol-1-yi)piperidine-1-carboxylate and Isopropyl 4-(5-cyano4-((2-fluoro-4-(2-((2-(trimethYlsilvl)ethoxv)methvl)-2H-tetrazol-5-vl)phenoxv)methyl)-1Hpyrazol-1 -Yl)piperidine-1 -carboxylate
To a stirred solution of isopropyl 4-(5-cyano-4-(hydroxymethyl)-1 H-pyrazol-1yl)pîperidine-1-carboxylate (94 mg, 0.322 mmol), 2-fluoro-4-(1-((2(trimethylsilyl)ethoxy)methyl)-1H-tetrazol-5-yl)phenol and 2-fluoro-4-(2-((2(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)phenol (Préparation 17) (100 mg, 0.322 mmol) and triphenylphosphine (110 mg, 0.42 mmol) in 5 mL of 1,4-dioxane was added
drop-wise diethyl azodicarboxylate (0.060 mL, 0.39 mmol). The resulting mixture was stirred overnight at room température before the mixture was concentrated in vacuo. The residue was purified by flash chromatography, eluting with a gradient of 10 to 40% ethyl acetate in heptane to give isopropyl 4-(5-cyano-4-((2-fluoro-4-(1-((2(trimethylsilyl)ethoxy)methyl)-1H-tetrazol-5-yl)phenoxy)methyl)-1 H-pyrazol-1yl)piperidine-1-carboxylate and isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-((2(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)phenoxy)methyl)-1 H-pyrazol-1yl)piperidine-1-carboxylate (140 mg, 74% yield).
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5yl)phenoxy)methyl)-1 H-pyrazol-1 -yl)piperidine-1 -carboxylate. 1H NMR (400 MHz, deuterochloroform) delta -0.05-0.01 (m, 9 H), 0.90- 1.00 (m, 2 H), 1.18-1.27 (m, 6 H), 2.02 (br. s., 2 H), 2.13 (m, 2 H) 2.93 (br. s., 2 H), 3.65 - 3.78 (m, 2 H), 4.30 (d, 7=7.22 Hz, 2 H), 4.46 - 4.58 (m, 1 H), 4.86 - 4.98 (m, 1 H), 5.16 (s, 2 H), 5.89 (s, 2 H), 7.09 7.18 (m, 1 H), 7.69 (s, 1 H), 7.88 - 7.96 (m, 2 H). LCMS (ES) 585.1 (M+1).
B) Isopropyl 4-(5-cyano-4-((2-fluoro-4-( 1 H-tetrazol-5-vl)phenoxy)methyl)-1 H-pyrazol-1 yl)piperidine-1-carboxylate and Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2H-tetrazol-5yl)phenoxv)methyl)-1 H-pyrazol-1-vl)piperidine-1-carboxylate
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(1 -((2-(trimethylsilyl)ethoxy)methyl)-1 H-tetrazol-5yl)phenoxy)methyl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate and isopropyl 4-(5-cyano4-((2-fluoro-4-(2-((2-(trimethylsilyl)ethoxy)methyl)-2H-tetrazol-5-yl)phenoxy)methyl)-1Hpyrazol-1 -yl)piperidine-1 -carboxylate (220 mg, 0.38 mmol) were dissolved in éthanol (3 mL) and a solution of aqueous 2 M hydrochloric acid (3 mL) was added drop-wise, The resulting mixture was stirred at 50 degrees Celsius for 4 hours before being cooled down to room température and filtered. The resulting white solid was washed with ethyl acetate and heptane (1/1 volume) and dried under reduced pressure to give the title compound (80 mg, 47% yield). 1H NMR (400 MHz, deutero dimethyl sulfoxide) delta
1.16 (d, 7=6.25 Hz. 6 H), 1.76 - 1.90 (m, 2 H), 1.98 (dd, 7=14.45, 3.12 Hz. 2 H). 2.99 (br. s., 2 H). 4.04 (d, 7=15.81 Hz. 2 H), 4.59 - 4.71 (m, 1 H). 4.70 - 4.82 (m. 1 H). 5.27 (s. 2 H), 7.47 - 7.57 (m, 1 H), 7.80 - 7.83 (m, 1 H), 7.83 - 7.87 (m. 1 H), 7.90 (s, 1 H). LCMS (ES) 455.0 (M+1).
Example 20: Isopropyl 4-(5-cvano-4-((2-fluoro-4-(1-methvl-1H-tetrazol-5vl)phenoxv)methvl)-1H~pyrazol-1-vl)piperidine-1-carboxylate and
Example 21: Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-methvl-2H-tetrazol-5yl)phenoxv)methvl)-1H-pvrazol-1-vl)piperidine-1-carboxylate
To a solution of isopropyl 4-(5-cyano-4-((2-fluoro-4-(1 H-tetrazol-5-yl)phenoxy)methyl)1H-pyrazol-1-yl)piperidine-1-carboxylate and isopropyl 4-(5-cyano-4-((2-fluoro-4-(2Htetrazol-5-yl)phenoxy)methyl)-1H-pyrazol-1-yl)piperidine-1-carboxylate (70 mg, 0.15 mmol) at room température in tetrahydrofuran (2 mL) was added sodium hydride (14 mg, 0.31 mmol) in two portions, and the resulting mixture was stirred for 5 minutes, lodomethane (0.03 mL, 0.46 mmol) was then added and the reaction mixture was stirred at room température for an additional 16 hours. The reaction was quenched by addition of water and the mixture was diluted with ethyl acetate. The organic phase was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over magnésium sulfate, filtered and the filtrate was concentrated in vacuo. The residue was purified by flash silica gel chromatography, eluting with a gradient mixture of ethyl acetate in heptane (30 to 60% ethyl acetate) to give isopropyl 4-(5-cyano-4-((2-fluoro-4-(1-methyl-1 H-tetrazol20 5-yl)phenoxy)methyl)-1H-pyrazol-1-yl)piperidine-1-carboxylate (10 mg, 14% yield) and isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-methyl-2H-tetrazo1-5-yl)phenoxy)methyl)-1Hpyrazol-1-yl)piperidine-1-carboxylate (30 mg, 42% yield).
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(1-methyl-1 H-tetrazol-5-yl)phenoxy)methyl)-1Hpyrazol-1-yl)piperidine-1-carboxylate (Example 20). 1H NMR (400 MHz, deuterochloroform) delta 1.18 -1.28 (m, 6 H), 1.95 - 2.06 (m, 2 H), 2.13 (m, 2 H), 2.85 3.02 (m, 2 H), 4.17 (s, 3 H), 4.36 (d, J=10.15 Hz, 2 H), 4.46 - 4.57 (m, 1 H) 4.92 (spt, 1 H), 5.19 (s, 2 H), 7.17 - 7.24 (m, 1 H), 7.48 - 7.58 (m, 2 H), 7.70 (s, 1 H). LCMS (ES) 469.0 (M+1).
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-methyl-2H-tetrazol-5-yl)phenoxy)methy1)-1H30 pyrazol-1-yl)piperidine-1-carboxylate (Example 21). 1H NMR (400 MHz,
deuterochloroform) delta 1.24 (d, 7=6.25 Hz, 6 H) 1.95 - 2.05 (m, 2 H) 2.13 (m, 2 H)
2.93 (t, 7=12.59 Hz, 2 H) 4.31 (br. s., 2 H) 4.37 (s, 3 H) 4.51 (m, 1 H) 4.92 (m, 1 H) 5.16 (s, 2 H) 7.09 - 7.16 (m, 1 H) 7.69 (s, 1 H) 7.83 - 7.87 (m, 1 H) 7.87 - 7.90 (m, 1 H). LCMS (ES) 469.0 (M+1).
Example 22: Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-(2-hvdroxvethvl)-2H-tetrazol-5yl)phenoxy)methvl)-1 H-pyrazol-1 -yl)piperidine-1 -carboxylate
A) Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-(2-(trimethvlsilvloxv)ethvl)-2H-tetrazol-5vl)phenoxv)methvl)-1H-pvrazol-1-vl)piperidine-1-carboxylate
To a stirred solution of isopropyl 4-(5-cyano-4-(hydroxymethyl)-1 H-pyrazol-1 yl)piperidine-1-carboxylate (Préparation 5) (78 mg, 0.266 mmol), 2-fluoro-4-(2-(2(trimethylsilyloxy)ethyl)-2H-tetrazol-5-yl)phenol (Préparation 19) (90 mg, 0.27 mmol) and triphenylphosphine (77 mg, 0.29 mmol) in 5 mL of 1,4-dioxane was added dropwise diethyl azodicarboxylate (0.046 mL, 0.28 mmol). The resulting mixture was stirred for 15 hours at room température before the mixture was concentrated in vacuo. The residue was purified by flash chromatography, eluting with a gradient of 5 to 40% ethyl acetate in heptane to give isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-(2(trimethylsilyloxy)ethyl)-2H-tetrazol-5-yl)phenoxy)methyl)-1 H-pyrazol-1-yl)piperidine-1carboxylate (140 mg, 86% yield).
B) Isopropyl 4-(5-cvano-4-«2-fluoro-4-(2-(2-hydroxyethyl)-2H-tetrazol-5yl)phenoxy)methvl)-1 H-pyrazol-1 -yl)piperidine-1 -carboxylate
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-(2-(trimethylsilyloxy)ethyl)-2H-tetrazol-5yl)phenoxy)methyl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate (140 mg, 0.228 mmol) was dissolved in methanol (2 mL), and a solution of 4 M hydrochloric acid (1 mL) in 1,4dioxane was added drop-wise, The resulting mixture was stirred at room température for 2 hours before being concentrated under reduced pressure. The residue (160 mg) was divided and ca. 50 mg of the crude was purified by reverse-phase HPLC to give the
title compound {30 mg, 26%)(Column: Waters XBridge C18 19x100, 5 micrometer;
Mobile phase A: 0.03% ammonium hydroxide in water (v/v); Mobile phase B: 0.03% ammonium hydroxide in acetonitrile (v/v); Gradient: 85%water/15%acetonitrile linear to 0%water/100%acetonitrile in 8.5 minutes, hold at 0%water / 100%acetonitrile to 10.0 minutes. Flow: 25mL/min. Détection: 215 nm. LCMS (ES+): 499.5 (M+1).
Example 23: Isopropyl 4-(5-cvano-4-((2-fluoro-4-(1-(2-hvdroxvethyl)-1 H-tetrazol-5yl)phenoxv)methyl)-1 H-pyrazol-1 -yl)piperidine-1 -carboxylate
A) Isopropyl 4-(5-cyano-4-((2-fluoro-4-(1-(2-(trimethvlsilvloxv)ethyl)-1 H-tetrazol-5vl)phenoxy)methvl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate
To a stirred solution of isopropyl 4-(5-cyano-4-(hydroxymethyl)-1 H-pyrazol-1yl)piperidine-1 -carboxylate (43 mg, 0.15 mmol), 2-fluoro-4-(1-(2-(trimethylsilyloxy)ethyl)1 H-tetrazol-5-yl)phenol (préparation 20) (50 mg, 0.15 mmol) and triphenylphosphine (43 mg, 0.16 mmol) in 3 mL of 1,4-dioxane was added drop-wise dïethyl azod[carboxylate (0.025 mL, 0.16 mmol). The resulting mixture was stirred overnight at room température before the mixture was concentrated in vacuo. The residue was purified by flash chromatography, eluting with a gradient of 30 to 70% ethyl acetate in heptane to give isopropyl 4-(5-cyano-4-((2-fluoro-4-(1-(2-(trimethylsilyloxy)ethyl)-1H-tetrazol-5yl)phenoxy)methyl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate (50 mg, 55% yield).
B) Isopropyl 4-(5-cyano-4-((2-fluoro-4-( 1 -(2-hydroxvethyl)-1 H-tetrazol-5yl)phenoxv)methvl)-1 H-pyrazol-1-vl)Diperidine-1-carboxylate
Isopropyl 4-(5-cyano-4-((2-fiuoro-4-(1-(2-(trimethylsilyloxy)ethyl)-1H-tetrazol-5yl)phenoxy)methyl)-1H-pyrazol-1-yl)piperidine-1-carboxylate (50 mg, 0.082 mmol) was dissolved in methanol (2 mL) and a solution of 4 M hydrochloric acid (1 mL) in 1,4dioxane was added drop-wise, The resulting mixture was stirred at room température for 2 hours before being concentrated under reduced pressure. The residue (60 mg) was purified by reversed-phase HPLC to give the title compound (20 mg, 49% yield) (Column: Waters XBridge C1819x100, 5 micrometer; Mobile phase A: 0.03% ammonium hydroxide in water (v/v); Mobile phase B: 0.03% ammonium hydroxide in acetonitrile (v/v); Gradient: 80%water/20%acetonitrile linearto 0%water/100%acetonitrile in 8.5 minutes, hold at 0%water/ 100%acetonitrile to 10.0 minutes. Flow: 25mL/min. Détection: 215 nm
LCMS (ES+): 499.4 (M+1).
Example 24: 1 -Methylcyclopropyl 4-(5-cyano-4-(i2-fluoro-4-( 1 -methyl-1 H-tetrazol-5vl)phenoxvlmethvlÎ-1H-pvrazol-1-yl)piperidine-1-carboxylate
The title compound was prepared using 2-fluoro-4-(1-methyl-1 H-tetrazol-5-yl)phenol (Préparation 21), following procedures analogous to Example 15.1H NMR (400 MHz, deuterochloroform) delta 0.58 - 0.67 (m, 2 H), 0.83 - 0.92 (m, 2 H), 1.57 (s, 3 H), 1.94 2.05 (m, 2 H), 2.05 - 2.21 (m, 2 H), 2.92 (t, 7=12.98 Hz, 2 H), 4.17 (s, 3 H), 4.32 (br. s„ 2 H), 4.43 - 4.56 (m, 1 H), 5.19 (s, 2 H), 7.17 - 7.24 (m, 1 H), 7.48 - 7.58 (m, 2 H), 7.70 (s, 1 H). 1H NMR indicated the presence of less than 10% of what is believed to be the corresponding isopropyl carbamate dérivative (from the isopropyl 4-nitrophenyl carbonate contaminating the 1-methylcyclopropyl 4-nitrophenyl carbonate). LCMS (ES)
481.6 (M+1).
Example 25:1-Methylcyclopropyl 4-(5-cvano-4-([4-( 1 -methyl-1 H-tetrazol-5vl)phenoxylmethvlÎ-1H-pvrazol-1-vl)piperidine-1-carboxylate
The title compound was prepared using 4-(1-methyl-1H-tetrazol-5-yl)phenol (Préparation 22), following procedures analogous to Example 15, 1H NMR (400 MHz, deuterochloroform) delta 0.60 - 0.67 (m, 2 H), 0.83 - 0.91 (m, 2 H), 1.58 (s, 3 H), 1.96 - 2.06 (m, 2 H), 2.06 - 2.21 (m, 2 H), 2.84 - 3.00 (m, 2 H), 4.16 (s, 3
H), 4.33 (br. s., 2 H), 4.45 - 4.57 (m, 1 H), 5.12 (s, 2 H), 7.10 - 7.15 (m, 2 H), 7.68 (s, 1
H), 7.69 - 7.74 (m, 2 H). 1H NMR indicated the presence of less than 10% of what is believed to be the corresponding isopropyl carbamate dérivative (from the isopropyl 410 nitrophenyl carbonate contaminating the 1-methylcyclopropyl 4-nitrophenyl carbonate). LCMS (ES) 463.5 (M+1).
Example 26: 1-Methvlcyclopropvl 4-(4-((4-carbamovl-3-fluorophenoxy)methyl)-5-cvano1 H-pyrazol-1 -vl)piperidine-1 -carboxylate
NH,
The title compound was prepared using 2-fluoro-4-hydroxybenzamide (Préparation 24), following procedures analogous to Example 13.1H NMR (400 MHz, deuterochloroform) delta 0.57 - 0.65 (m, 2 H), 0.82 - 0.89 (m, 2 H), 1.53 (s, 3 H), 1.92 - 2.04 (m, 2 H), 2.10 (qd, .7=12.14, 4.20 Hz, 2 H), 2.90 (br. s., 2 H), 4.32 (br. s., 2 H), 4.49 (tt, J=11.25, 4.37
Hz, 1 H), 5.02 - 5.09 (m, 2 H), 6.00 (br. s., 1 H), 6.51 - 6.64 (m, 1 H), 6.69 (dd, J=13.66,
2.54 Hz, 1 H), 6.84 (dd, J=8.78, 2.54 Hz, 1 H), 7.64 (s, 1 H), 8.07 (t, J=9.08 Hz, 1 H). 1H
NMR indicated the presence of less than 10% of what is believed to be the corresponding isopropyl carbamate dérivative (from the isopropyl 4-nitrophenyl carbonate contaminating the 1-methylcyclopropyl 4-nitrophenyl carbonate). LCMS (ES) 25 442.4 (M+1).
ιοο
Example 27: Isopropyl 4-(5-cvano-4-{1-r2-fluoro-4-(methvlsulfonyl)phenoxv1ethvl}-1Hpyrazol-1 -yl)piperidine-1 -carboxylate
O
The title compound was prepared using 2-fluoro-4-(methylsulfonyl)phenol and isopropyl 4-(5-cyano-4-(1-hydroxyethyl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate (Préparation 25), following procedures analogous to Example 15. The sample was purified by reversed-phase HPLC (Column: Waters XBridge C18 19x100, 5 micrometer; Mobile phase A: 0.03% ammonium hydroxide in water (v/v); Mobile phase B: 0.03% ammonium hydroxide in acetonitrile (v/v); Gradient: 80%water/20%acetonitrile linear to 0%water/100%acetonitrile in 8.5 minutes, hold at 0%water / 100%acetonitrile to 10.0 minutes. Flow: 25mL/minute. LCMS ( ES+): 479.2 M+1).
Example 28: Isopropyl 4-(5-cvano-4-{1-[(2-methvlpvridin-3-vl)oxv1ethvl)-1H-pvrazol-1-
The title compound was prepared using 2-methylpyridin-3-ol and isopropyl 4-(5-cyano4-(1-hydroxyethyl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate (Préparation 25), following procedures analogous to Example 15. The sample was purified by reversed-phase HPLC (Column: Waters XBridge C18 19x100, 5 micrometer; Mobile phase A: 0.03% ammonium hydroxide in water (v/v); Mobile phase B: 0.03% ammonium hydroxide in acetonitrile (v/v); Gradient: 85%water/15%acetonitrile linear to 0%water/100%acetonitrile in 8.5 minutes, hold at 0%water / 100%acetonitrile to 10.0 minutes. Flow: 25mL/minute. LCMS (ES+): 398.2 M+1).
Example 29: Isopropyl 4-(5-cyano-4-{2-[2-fluoro-4-(methvlsulfonvl)phenvllpropyl}-1 Hpyrazol-1-yl)piperidine-1-carboxylate
101
A) Isopropyl 4-(5-cvano-4-vinyl-1H-pvrazol-1-vl)piperidine-1-carboxylate
To a stirred mixture of (methyl)-triphenylphosphonium bromide (323 mg, 0.88 mmol) in tetrahydrofuran (5 mL) at -78 degrees Celsius was added drop-wise n-butyllithium (0.360 mL, 0.89 mmol, 2.5 M in hexanes). The resulting yellow mixture was stirred at 78 degrees Celsius for 30 minutes, and then a solution of isopropyl 4-(5-cyano-4-formyl1H-pyrazol-1-yl)piperidine-1-carboxylate (Example 9, Step A) (171 mg, 0.59 mmol) in tetrahydrofuran (2.5 mL) was added, The cold bath was removed, and the reaction mixture was stirred for 3.75 hours at room température. The reaction was quenched with saturated aqueous ammonium chloride, and the mixture was extracted twice with ethyl acetate. The combined extracts were washed sequentially with water and brine and then dried over sodium sulfate. The mixture was filtered, and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography eluting with a gradient mixture of ethyl acetate in heptane (10 to 100%) to give the title compound as a clear oil (116 mg, 68%). 1H NMR (500 MHz, deuterochloroform) delta 0.88 (d, /=6.10 Hz, 6 H), 1.55-1.67 (m, 2 H), 1.68 - 1.84 (m, 2 H), 2.43 - 2.73 (m, 2 H),
3.95 (br. s., 2 H), 4.04 - 4.21 (m, 1 H) 4.44 - 4.67 (m, 1 H), 5.02 (d, J=11.22 Hz, 1 H),
5.43 (d, /=17.81 Hz, 1 H), 6.20 (dd, /=17.81, 11.22 Hz, 1 H), 7.27 (s, 1 H).
B) (E,Z)-lsopropyl 4-(5-cyano-4-(2-(2-fluoro-4-(methvlsulfonyl)phenvl)prop-1 -envl)-1 Hpyrazol-1-yl)piperidine-1-carboxylate
To a solution of isopropyl 4-(5-cyano-4-vinyl-1H-pyrazol-1-yl)piperidine-1 -carboxylate (116 mg, 0.4 mmol) and 2-fluoro-4-(methylsulfonyl)-1-(prop-1-en-2-yl)benzene (Préparation 29) (43 mg, 0.20 mmol) in anhydrous dichloromethane (2 mL) was added the second génération Hoveyda-Grubbs catalyst (commercially available from Aldrich) (12.5 mg, 0.020 mmol). The green solution was heated at 40 degrees Celsius for 72 hours periodically adding dichloromethane. The material was concentrated under reduced pressure, and the residue purified by silica gel chromatography (10 to 100% ethyl acetate in heptane) to give the product as an impure oil (8 mg, 8%). This material was used as is. LCMS (APCI): 473.2 (M - 1 ).
102
C) Isopropyl 4-(5-cvano-4-(2-[2-fluoro-4-(methvlsulfonyl)phenyl1propvlÎ-1 H-pyrazol-1vl)piperidine-1-carboxylate
A solution of (E,Z)-isopropyl 4-(5-cyano-4-(2-(2-fluoro-4-(methylsulfonyl)-phenyl)prop-1enyl)-1H-pyrazol-1-yl)piperidine-1-carboxylate (8 mg, 0.02 mmol) in ethyl acetate (3 mL) was hydrogenated on the H-Cube™ at the full hydrogen setting using a 10% palladium on carbon cartridge at a flow rate of 1 mL/minute. The material was concentrated in vacuo, and the residue (4 mg) was purified by reversed-phase HPLC (Column: Waters XBridge C18 19x100, 5 micrometer; Mobile phase A: 0.03% ammonium hydroxide in water (v/v); Mobile phase B: 0.03% ammonium hydroxide in acetonitrile (v/v);
Gradient: 80%water/ 20%acetonitrile linear to 0%water/100%acetonitrile in 8.5 minutes, hold at 0% water /100% acetonitrile to 10.0 minutes. Flow: 25mL/minute) to give the title compound (1.9 mg, 23%); LCMS (ES+): 477.2 (M+1).
Example 30: 1-Methylcyclopropyl 4-(5-cvano-4-([(2-methvlpyridin-3-vl)oxvlmethvl]-1Hpyrazol-1 -yl)piperidine-1 -carboxylate
The title compound was prepared using 2-methylpyridin-3-ol, following procedures analogous to Example 13. The crude material was purified by flash chromatography, eluting with a gradient mixture of ethyl acetate in heptane (60 to 100% ethyl acetate) to give 77 mg of the title compound as a white solid. 1H NMR (400 MHz, deuterochloroform) delta 0.60 - 0.66 (m, 2 H), 0.83 - 0.90 (m, 2 H), 1.55 (s, 3 H), 1.96 2.05 (m, 2 H), 2.05 - 2.20 (m, 2 H), 2.49 (s, 3 H), 2.84 - 2.98 (m, 2 H), 4.11 - 4.42 (m, 2
H), 4.46 - 4.55 (m, 1 H), 5.04 (s, 2 H), 7.06 - 7.16 (m, 2 H), 7.65 (s, 1 H), 8.12 (dd
J=4.49, 1.56 Hz, 1 H).
Example 31: 1-Methylcyclopropvl 4-{5-cvano-4-[(2,3,6-trifluorophenoxv)methyll-1Hpyrazol-1 -yl)piperidine-1 -carboxylate
103
A) tërt-Butvl 4-(5-cyano-4-((2,3,6-trifluorophenoxy)methyl)-1H-pyrazol-1-vl)piperidine-1carboxylate terf-Butyl 4-(5-cyano-4-((methylsulfonyloxy)m ethyl )-1 H-pyrazol-1-yl)piperidine-15 carboxylate (Préparation 16) (87.8 mg, 0.228 mmol), 2,3,6-trifluorophenol (51.7 mg, 0.342 mmol ), and césium carbonate (149 mg, 0.456 mmol) were placed in microwave vial and dissolved in acetonitrile (3 mL). The vial was heated in a microwave reactor at 110 degrees Celsius for 20 minutes. The mixture was concentrated under reduced pressure, and the residue was taken up in 1 N sodium hydroxide solution (5 mL) and extracted three times with dichloromethane. The combined organic extracts were washed with brine, dried over sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude material was purified by chromatography eluting with a 0 to 30 % ethyl acetate in heptane gradient to give 36.2 mg of tert-butyl 4-(5cyano-4-((2,3,6-trifluorophenoxy)methyl)-1 H-pyrazol-1 -yl)piperidine-1 -carboxylate as a 15 clearoil.
B) 1-Methvlcyclopropyl 4-f5-cyano-4-[(2,3,6-trifluorophenoxv)methvl1-1 H-pyrazol-1yllpiperidine-l -carboxylate
1-Methylcyctopropyl 4-{5-cyano-4-[(2,3,6-trifluorophenoxy)methyl]-1 H-pyrazol-1 20 yl)piperidine-1 -carboxylate was prepared using commercially available 2,3,6-trifluoro phénol, following procedures analogous to Example 13 (B and C). The crude material (17.1 mg) was purified by préparative reverse-phase HPLC on a Sepax 2-Ethyl Pyridine column 250 x 21.2 mm, 0.005 eluting with a gradient of éthanol in heptane. Analytical LCMS: rétention time 11.769 minutes (Phenomenex Luna (2) Cis 150 x
3.0mm, 5 micrometer column; 95% water/methanol linear gradient to 100% methanol over 12.5 minutes; 0.1% formic acid modifier; flow rate 0.75 mL/minute; LCMS (ES+): 456.9 ( M + Na). 1H NMR (500 MHz, deuterochloroform) delta 0.64 - 0.66 (m, 2 H), 0.88 - 0.91 (m, 2 H), 1.57 (s, 3 H), 2.00 (d, 7=10.49 Hz, 2 H), 2.07 - 2.18 (m, 2 H), 2.91 -
2.95 (m, 2 H), 4.18 (br. s., 1 H),4.36(br. s„ 1 H), 4.50 (tt, 7=11.34, 4.15 Hz, 1 H), 5.19 (s, 2 H), 6.83 - 6.90 (m, 2 H), 7.67 (s, 1 H).
y
104
Example 32: Isopropyl 4-{5-cvano-4-i(2,3l6-trifluorophenoxv)methyl1-1 H-pyrazol-1 yl}piperidine-1 -carboxylate
The title compound was prepared using commercially available 2,3,6-trifulorophenol following procedures analogous to Example 11. The crude material was purified by column chromatography eluting with a 0 to 25% ethyl acetate in heptane gradient to give isopropyl 4-{5-cyano-4-[(2,3,6-trifluorophenoxy)methyl]-1 H-pyrazol-1-yljpiperidine1-carboxylate as a clear oil. 1H NMR (500 MHz, deuterochloroform) delta 1.26 (d, 7=6.10 Hz, 6 H), 2.01 (d, 7=11.22 Hz, 2 H) 2.13 (qd, 7=12.28, 4.64 Hz, 2 H), 2.88 - 3.01 (m, 2 H), 4.32 (br. s., 2 H) 4.51 (tt, 7=11.34, 4.15 Hz, 1 H), 4.90-4.98 (m,1 H), 5.18 (s,
H), 6.82 - 6.92 (m, 2 H), 7.67 (s, 1 H); LCMS (ES+): 423.4 ( M + H).
Example 33: Isopropyl 4-(5-cvano-4-([2-fluoro-4-(1-methvl-1H-imidazol-2yl)phenoxv]methvl}-1 H-pyrazol-1 -ylÎpiperidine-1 -carboxylate
The title compound was prepared from 2-fluoro-4-(1-methyl-1H-imidazol-2-yl)phenol (Préparation 28) and isopropyl 4-(5-cyano-4-((methylsulfonyloxy)methyl)-1 H-pyrazol-1yl)piperidine-1-carboxylate (Préparation 10) following procedures analogous to Example
11. The crude material was purified by préparative reverse-phase HPLC on a Sepax
Silica 250 x 21.2mm, 0.005 mm, eluting with a gradient of éthanol in heptane. Analytical LCMS: rétention time 8.598 minutesfPhenomenex Luna (2) Cie 150 x 3.0mm, 5 micrometer column; 95% water/methanol linear gradient to 100% methanol over 12.5 minutes; 0.1%formic acid modifier; flow rate 0.75 mL/minute; LCMS (ES +): 467.0 (M +
H). 1H NMR (500 MHz, deuterochloroform) delta 1.27 (d, 7=6.10 Hz, 6 H), 1.97 - 2.09 (m, 2 H), 2.16 (m, 2 H), 2.93 - 2.98 (m, 2 H), 3.76 (s, 3 H) 4.25 - 4.43 (m, 2H), 4.50 16400
105
4.57(m, 1 H), 4.91-4.99 (m, 1 H), 5.17 (s, 2 H), 6.97 (s, 1 H), 7.11 (s, 1 H), 7.12-7.15 (m, 1 H), 7.42 (dd, 7=11.71, 1.95 Hz, 1 H), 7.38 - 7.44 (m, 1 H), 7.72 (s, 1 H).
Example 34: Isopropyl 4-(5-cyano-4-{[2-fluoro-4-(1-methyl-1 H-imidazol-5yl)phenoxv1methyl}-1 H-pyrazol-1 -yl)piperidine-1 -carboxylate
The title compound was prepared from 2-fluoro-4-(1 -methyl-1 H-imidazol-5-yl)phenol (Préparation 27) and Isopropyl 4-(5-cyano-4-((methylsulfonyloxy)methyl)-1 H-pyrazol-1 yl)piperidine-1-carboxylate (Préparation 10) following procedures analogous to Example
11. The crude material was purified by préparative reverse-phase HPLC on a Sepax Silica 250 x 21.2mm, 0.005 eluting with a gradient of éthanol in heptane. Analytical LCMS: rétention time 8.797 minutes(Phenomenex Luna (2) Cia 150 x 3.0mm, 5 micrometer column; 95% water/methanol linear gradient to 100% methanol over 12.5 minutes; 0.1% formic acid modifier; flow rate 0.75 mL/minute; LCMS (ES +): 467.0 (M + H). 1H NMR (500 MHz, deuterochloroform) delta 1.27 (d, 7=6.34 Hz, 6 H), 2.03 (d, 7=11.22 Hz, 2 H), 2.11 - 2.20 (m, 2 H), 2.95 (br. s., 2 H), 3.66 (s, 3 H), 4.34 (br. s., 2 H), 4.50 - 4.57 (m,1 H), 4.94 (dt, 7=12.44, 6.22 Hz, 1 H), 5.15 (s, 2 H), 7.07 (s, 1 H), 7.10 -
7.17 (m, 3 H), 7.51 (s, 1 H), 7.71 (s, 1 H).
Example 35: Isopropyl 4“[5“Cyano-4-((r2-methvl-6-(1H-1,2,4-triazol-1-yl)pvridin-3ylloxylmethyD-l H-pyrazol-1-vllpiperidine-1-carboxylate
106
The title compound was prepared using 2-methyl-6-(1H-1,2,4-triazol-1-yl)pyridin-3-ol following procedures analogous to Example 12. The sample was purified by reversedphase HPLC (Column: Waters XBridge C18 19x100, 5 micrometer; Mobile phase A: 0.03% ammonium hydroxide in water (v/v); Mobile phase B: 0.03% ammonium hyrdroxide in acetonitrile (v/v); Gradient: 80%water/20%acetonitrile linear to 0%water/100%acetonitrile in 8.0 minutes, hold at 0% water /100% acetonitrile to 9,5 minutes. Flow: 25mL/minute, LCMS (MS ES+:451.1).
Example 36: Isopropyl 445-cvano-4-({[2-methyl-6-(1 H-1,2,4-triazoÎ-1-vl)pyridin-3yl1amino)methvl)-1 H-pyrazol-1 -yllpiperidine-1 -carboxylate
To a stirred solution of isopropyl 4-(5-cyano-4-((methylsulfonyloxy)methyl)-1 H-pyrazol1-yl)piperidine-1-carboxylate (Préparation 10) (44 mg, 0.12 mmol) in 0.75 mL of tetrahydrofuran was added /V,A/-diisopropylethylamine (0.042 mL, 0.24 mmol) followed by 2-methyl-6-(1H-1,2,4-triazol-1-yl)pyridin-3-amine (21 mg, 0.12 mmol). The reaction mixture was heated at 60 degrees Celsius for 16 hours before it was cooled to room température and diluted with water and brine. The mixture was then extracted three times with 15 mL ethyl acetate. The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated in vacuo to give 52 mg of a yellow foam. The sample was purified by reversed-phase HPLC (Column: Waters Sunfire C18 19x100, 5 micrometer; Mobile phase A: 0.05% trifluoroacetic acid in water (v/v); Mobile phase B: 0.05% trifluoroacetic acid in acetonitrile (v/v));
Gradient: 90%water/10%acetonitrile linear to 0%water/100%acetonitrile in 8.5 minutes, hold at 0% water /100% acetonitrile to 10.0 minutes. Flow: 25mL/minute. LCMS (MS ES+: 450.1).
Example 37: Isopropyl 4-[5-cvano-4-({r2-methvl-6-(methylsulfonyl)pvridin-3vÎlaminoÎmethvl)-1H-pvrazol-1-yl1piperidine-1-carboxylate
107
The title compound was prepared using 2-methyl-6-(methylsuifonyl)pyridin-3-amine following procedures analogous to Example 36. The sample was purified by reversedphase HPLC (Column: Waters XBridge C18 19x100, 5 micrometer; Mobile phase
A: 0.03% ammonium hydroxide in water (v/v); Mobile phase B: 0.03% ammonium hyrdroxide in acetonitrile (v/v); Gradient: 85%water/15%acetonitrile linear to 0%water/100%acetonitrile in 8.5 minutes, hold at 0% water /100% acetonitrile to 10.0 minutes. Flow: 25mL/minute. LCMS (ES+): 461.0 (M+1).
Example 38: 1-Methvlcyclopropyl 4-(5-cvano-4-(F4-(1 H-tetrazol-1-vl)phenoxv1methylT
H-pyrazol-1 -yl)piperidine-1 -carboxylate
The title compound was prepared using commercially available 4-tetrazol-1-yl-phenol following procedures analogous to Example 15. The crude material was purified by flash chromatography eluting with a gradient from 0% to 75% ethyl acetate in heptanes, 1H NMR (400 MHz, deuterochloroform) delta ppm 0.60 - 0,66 (m, 2 H) 0.84 - 0.90 (m, 2
H) 1.19 (t, J=7.03 Hz, 1 H) 1.55 (s, 3 H) 2.03 (br. s., 2 H) 2.06 - 2.19 (m, 2 H) 2.92 (br.
s., 2 H) 3,46 (q, J=7.09 Hz, 1 H) 4.46 - 4.56 (m, 1 H) 5.11 (s, 2 H) 7.11 - 7.16 (m, 2 H) 7.60 - 7.65 (m, 2 H) 7.68 (s, 1 H) 8.90 (s, 1 H)
Example 39: 1-[1-(5-Ethvlpvrimidin-2-vl)piperidin-4-yll-4-(i4-(1H-tetrazol-1vl)phenoxvlmethvl}-1H-pyrazole-5-carbonitrile
108 f A) tert-Butyl 4-(5-cyano-4-f[4-(1 H-tetrazol-1-yl)phenoxy]methvlÎ-1 H-pyrazol-1vl)pÎperidine-1-carboxylate
To a stirred, cold (0 degrees Celsius) solution of triphenylphosphine (283 mg, 1.08 mmol) in tetrahydrofuran (2 mL) was added diethylazodicarboxylate (0.17 mL, 1.1 mmol) drop wise. The cold reaction mixture was stirred for 20 minutes before a solution of 4-tetrazol-1-yl-phenol (165.5 mg, 1.021 mmol) in tetrahydrofuran was added. After 35 minutes a solution of tert-butyl 4-(5-cyano-4-(hydroxymethyl)-1H-pyrazol-1-yl)piperidine1-carboxylate (Préparation 15)(300 mg, 0.979 mmol) in tetrahydrofuran was added and the reaction was slowly allowed to warm up to room température overnight. The reaction was concentrated under reduced pressure and the residue was purified by flash chromatography eluting with a gradient from 0% to 80% ethyl acetate in heptanes to give the title compound as a white fluffy solid (304 mg, 68%). 1H NMR (400 MHz, deuterochloroform) delta ppm 1.46 (s, 9 H) 2.03 (s, 2 H) 2.06 - 2.20 (m, 2 H) 2.90 (br. s., 2 H) 4.28 (br. s„ 2 H) 4.46 - 4.56 (m, 1 H) 5.12 (s, 2 H) 7.10 - 7.18 (m, 2 H) 7.59 - 7.66 (m, 2 H) 8.90 (s, 1 H); LCMS (ES) 451.1 (M+1 )
B) 1-Piperidin-4-yl-4-if4-(1 H-tetrazol-1-vl)phenoxvlmethyl}-1 H-pvrazole-5-carbonitrile tert-Butyl 4-(5-cyano-4-{[4-(1 H-tetrazol-1-yl)phenoxy]methyl}-1 H-pyrazol-1-yl)piperidine1-carboxylate (298 mg, 0.663 mmol) was dissolved in dichloromethane (1.6 mL). Trifluoroacetic acid (0.15 mL) was added and the reaction was stirred at room température under nitrogen for 1.5 hours. The reaction was concentrated and used as is in the following step without further purification. LCMS (ES+) 351.1 (M+1 )
C) 1-Methylcyclopropyl 4-(5-cyano-4-{|4-(1 H-tetrazol-1 -vl)phenoxvlmethvl)-1 H-pyrazol-
-vl)piperidine-1 -carboxylate
1-Piperidin-4-yl-4-{[4-(1H-tetrazol-1-yl)phenoxy]methyl)-1H-pyrazole-5-carbonitrile (30 mg, 0.086 mmol) and diisopropylethylamine (0.12 ml, 0.688 mmol) were dissolved in acetonitrile (2 mL) in a sealed tube. 2-Chloro-5-ethylpyrimidine (0.020mL,
0.2 mmol) was added and the reaction was heated at 120 degrees Celsius for 18 hours and at room température for 36 hours. The reaction mixture was concentrated under 30 reduced pressure and the crude material was purified by flash chromatography eluting with a gradient from 0% to 70% ethyl acetate in heptanes to give a brown solid. The solid was triturated with minimal amounts of ether to give the title compound as a light brown solid (3 mg, 8%). LCMS (ES+) 457.1 (M+1) 1H NMR (400 MHz, deuterochloroform) delta ppm 1,16 -1.22 (m, 3 H) 2.10 (br. s., 2 H) 2.13 - 2.25 (m, 2 H)
2.43 - 2.50 (m, 2 H) 3.00 - 3.10 (m, 2 H) 3.43 - 3.50 (m, 1 H) 4.88 - 4.96 (m, 2 H) 5.12 (s,
H) 7.10 - 7.16 (m, 2 H) 7.60 - 7.64 (m, 2 H) 7.66 (s, 1 H) 8.12 - 8.24 (m, 2 H) 8.90 (s, 1 H)
Example 40: Isopropyl 4-(5-cyano-4-F(3-cyanophenoxv)methvl1-1H-pvrazol-1vl)piperidine-1 -carboxylate
0'
The title compound was prepared using commercially available 3-cyanophenol, following procedures analogous to Example 12. The crude material was purified by flash chromatography eluting with a gradient of 0% to 40% ethyl acetate in heptane to give 16.4 mg (62%) of the title compound as a clear colorless residue. 1H NMR (400 MHz, deuterochloroform) delta ppm 1.28 (d, J=6.25 Hz, 6 H) 1.99 - 2.09 (m, 2 H) 2.10 -
2.24 (m, 2 H) 2.88 - 3.06 (m, 2 H) 4.35 (br. s., 2 H) 4.48 - 4.60 (m, 1 H) 4.90 - 5.01 (m, 1 H) 5.08 (s, 2 H) 7.19 - 7.25 (m, 2 H) 7.32 (d, J=7.82 Hz, 1 H) 7.39 - 7.47 (m, 1 H) 7.68 (s, 1 H)
Example 41: Isopropyl 4-(5-cvano-4-[(4-cvano-3-methylphenoxv)methvll-1 H-pyrazol-1ylÎpiperidine-1 -carboxylate
The title compound was prepared using 4-hydroxy-2-methylbenzonitrile, following procedures analogous to Example 12. The crude material was purified by flash chromatography eluting with a gradient from 0% to 40% ethyl acetate in heptanes to give 18.8 mg (69%) of the title compound as a clear residue. 1H NMR (400 MHz, deuterochloroform) delta ppm 1.27 (d, J=6.25 Hz, 6 H) 1.96 - 2.08 (m, 2 H) 2.09 - 2.23 (m, 2 H) 2.54 (s, 3 H) 2.96 (t, J=12.51 Hz, 2 H) 4.35 (br. s., 2 H) 4.54 (tt, J=11.29, 4.15
110
Hz, 1 H) 4.95 (spt, J=6.25 Hz, 1 H) 5.09 (s, 2 H) 6.85 (dd, J=8.60, 2.35 Hz, 1 H) 6.90 (s, 1 H) 7.57 (d, J=8.60 Hz, 1 H) 7,67 (s, 1 H)
Example 42 : Isopropyl 4-(5-cyano-4-[(4-cyanophenoxv)methvll-1 H-pyrazol-1 5 vllpiperidine-l-carboxylate
The title compound was prepared using commercially available 4-cyanophenol, foilowing procedures analogous to Example 12. The crude material was purified by flash chromatography eluting with a gradient from 0% to 40% ethyl acetate in heptane 10 to give 14.7mg (56%) of the title compound as a sticky white solid. 1H NMR (400 MHz, deuterochloroform) delta ppm 1.27 (d, J=6.25 Hz, 6 H) 1.95 - 2.08 (m, 2 H) 2.16 (m, 2 H) 2.85 - 3.08 (m, 2 H) 4.35 (br. s., 2 H) 4.54 (tt, J=11.29, 4.15 Hz, 1 H) 4.95 (dt,
J=12.51, 6.25 Hz, 1 H) 5.11 (s, 2 H) 7.04 (d, J=8.99 Hz, 2 H) 7.64 (d, J=8.99 Hz, 2 H) 7.68 (s, 1 H)
Example 43: 4-[(4-Cyano-2-fluorophenoxy)methvl1-1-ri-(5-ethvlpvrimidin-2-vl)piperidin4-yll-1H-pyrazole-5-carbonitrile
The title compound was prepared using commercially available 4-cyano-2-fluorophenol, 20 foilowing procedures analogous to Example 39. The crude material was purified by flash chromatography eluting with a gradient from 0% to 1.5% methanoi in dichloromethane. The resulting solids were further purified via recrystallization from 10% methanol/ethyl acetate to give 3.67 g (60%) of clean product as a nearly white solid. 1H NMR (500 MHz, deuterochloroform) delta ppm 1.22 (t, J=7.56 Hz, 3 H) 2.07 25 2.15 (m, 2 H) 2.15 - 2.28 (m, 2 H) 2.50 (q. J=7.56 Hz, 2 H) 3.03 - 3.13 (m, 2 H) 4.66 (tt,
7=11.44, 4.18 Hz, 1 H) 4.95 (d, 7=13.66 Hz, 2 H) 5.19 (s, 2 H) 7.13 (t, 7=8.17 Hz, 1 H)
7.42 (dd, 7=10.37,1.83 Hz, 1 H) 7.47 (d, 7=8.29 Hz, 1 H) 7.70 (s, 1 H) 8.21 (s, 2 H)
Example 44: terf-Butyl 4-(5-cvano-4-f(4-cyano-2-fluorophenoxv)methvll-1H-pyrazol-1yiypiperidine-l-carboxylate
The title compound was prepared using commercially available 4-cyano-2-fluorophenol, foliowing procedures analogous to Example 15. The crude material was purified by flash chromatography eluting with a gradient from 10% to 40% ethyl acetate in heptanes to give the title compound (21 g, 100%). 1H NMR (deuterochloroform) delta ppm 7.71 (s, 1H), 7.44 - 7.48 (m, 1H), 7.40 - 7.43 (m, 1H), 7.09 - 7.15 (m, 1 H), 5.18 (s, 2H), 4.48 4.56 (m, 1H), 4.22 - 4.38 (m, 2H), 2.84 - 3.01 (m, 2H), 2.09 - 2.19 (m, 2H), 1.99 - 2.06 (m, 2H), 1.49 (s, 9H)
Example 45: Isopropyl 4-f5-cyano-4-K2-cvano-4-fluorophenoxv)methyl1-1 H-pyrazol-1yQpiperidine-1-carboxylate
The title compound was prepared using commercially available 2-cyano-4-fluorophenol, foliowing procedures analogous to Example 15. The crude material was purified by HPLC (Column Waters Atlantis dC18 4.6x50mm, 5 micrometer; Modifier: 0.05% trifluoroacetic acid; Gradient: 95% water / 5% acetonitrile linear to 5% water / 95% acetonitrile over 4.0 min, HOLD at 5% water / 95% acetonitrile to 5.0 min; Flow: 2.0 mL/min) to give 35.8 mg (73%) of the title compound. LCMS (ES+): 412. 0 (M+1 )
Example 46: Isopropyl 4-(5-cyano-4-{|4-(dimethvlcarbamovl)-2-fiuorophenoxylmethvl)1 H-pyrazol-1-yl)piperidine-1-carboxylate
112
The title compound was prepared using 3-fluoro-4-hydroxy-N,N-dimethylbenzamide (Préparation 31 B), following procedures analogous to Example 15. The crude material was purified by HPLC (Column Waters Atlantis dC18 4.6x50mm, 5 micrometer;
Modifier: 0.05% trifluoroacetic acid; Gradient: 95% water / 5% acetonitrile linear to 5% water / 95% acetonitrile over 4.0 min, HOLD at 5% water / 95% acetonitrile to 5.0 min; Flow: 2.0mL/min) to give 6.8 mg (12%) of the title compound. LC/MS (ES+): 458.0 (M+1)
Example 47: 1-Methylcvclopropyl 4-(5-cyano-4-ff4-(dimethylcarbamovl)-2fluorophenoxylmethyl)-1H-pyrazol-1-vl)piperidine-1-carboxylate
The title compound was prepared using 3-fluoro-4-hydroxy-N,N-dimethylbenzamide (Préparation 31 B), following procedures analogous to Example 15. The crude material was purified by HPLC (Column Waters Atlantis dC18 4.6x50 mm, 5 micrometer;
Modifier: 0.05% trifluoroacetic acid; Gradient: 95% water i 5% acetonitrile linear to 5% water / 95% acetonitrile over 4.0 min, HOLD at 5% water / 95% acetonitrile to 5.0 min; Flow: 2.0mL/min) to give 28.7 mg (51%) of the title compound. LC/MS ( ES+): 470,1 (M+1)
Example 48: 1-Methvlcvclopropyl 4-(5-cvano-4-{[2-fluoro-4(mefhvlcarbamovl)phenoxv1methvlMH-pvrazol-1-yl)piperidine-1 -carboxylate
113
The title compound was prepared using 3-fluoro-4-hydroxy-N-methylbenzamide (Préparation 31 A), following procedures analogous to Example 15. The crude material was purified by HPLC (Column Waters Atlantis dC18 4.6x50 mm, 5 micrometer;
Modifier: 0.05% trifluoroacetic acid; Gradient: 95% water / 5% acetonitrile linear to 5% water / 95% acetonitrile over 4.0 min, Hold at 5% water / 95% acetonitrile to 5.0 min; Flow: 2.0mL/min) to give 35.6 mg (65%) of the title product. LC/MS (ES+): 456.0 (M+1 )
Example 49: 4-((5-Cyano-1 -[1 -(5-ethvÎpvrimidin-2-vl)piperidin-4-vn-1 H-pyrazol-410 vlÎmethoxvl-S-fluoro-N.N-dimethylbenzamide
The title compound was prepared using 3-fluoro-4-hydroxy-N,N-dimethylbenzamide (Préparation 31 B), following procedures analogous to Example 39. The crude material was purified by HPLC (Column Waters Atlantis dC18 4.6x50 mm, 5 micrometer;
Modifier: 0.05% trifluoroacetic acid; Gradient: 95% water / 5% acetonitrile linear to 5% water / 95% acetonitrile over 4.0 min, HOLD at 5% water / 95% acetonitrile to 5.0 min; Flow: 2.0mL/min) to give 26.7 mg of the title product. LC/MS (ES+): 478.0 (M+1)
Example 50: 1-Methvlcyclopropyl 4-(5-cyano-4-[(4-cyano-2-fluorophenoxy)methyÎ1-1 H20 pvrazol-1-yl)piperidine-1-carboxylate
114
The synthesis is outlined in Scheme 5 below.
o
Scheme 5
A) fôrt-Butvl 4-[5-cyano-4-(ethoxvcarbonyl)-1 H-pyrazol-1-vllpiperidine-1-carboxylate
Ethyl 5-cyano-1H-pyrazole-4-carboxylate (Jubilant Chemsys Ltd. D-12, Sector-59, 201 301, Noida, U.P. India) (50 g, 300 mmol), fert-butyl 4-hydroxypiperidine-1-carboxylate (67 g, 333 mmol), and triphenylphosphine (111 g, 420 mmol) were dissolved in 2-methyl tetrahydrofuran (200 mL) and cooled to 0 degrees Celsius. A 40% solution of diethyl azodicarboxylate in toluene (76.5 mL, 420 mmol) was added drop wise. Once the addition was complété, the reaction was warmed up to room température over 1 hour and then allowed to stir at room température for 18 hours. Under vigorous stirring, heptane (1400 mL) was carefully added and a suspension formed after 1 hour. The solids were filtered off, and the filtrate was washed with a mixture of heptane (400 mL) and ethyl acetate (200 mL). The filtrate was then concentrated and the residue was purified by flash chromatography eluting with 25% ethyl acetate in heptanes and then re-crystallized from ethyl acetate-heptane to give the desired product (35.2 g, 33%). 1H NMR (deuterochloroform) delta ppm 7.97 (s, 1 H), 4.49 - 4.59 (m, 1 H), 4.36 (q, J = 7.1 Hz, 2H), 4.22 - 4.30 (m, 2H), 2.80 - 2.99 (m, 2H), 2.06 - 2.19 (m, 2H), 1.93 - 2.02 (m, 2H), 1.46 (s, 9H), 1.37 (t, J = 7.1 Hz, 3H)
B): tert-Butvl 4-[5-cyano-4-(hvdroxymethvl)-1 H-pyrazol-l-yllpiperidine-l -carboxylate
terf-Butyl 4-[5-cyano-4-(ethoxycarbonyl)-1H-pyrazol-1-yl]piperidine-1 -carboxylate (45.5 g, 131 mmol) was dissolved in tetrahydrofuran (350 mL) and cooled to -78 degrees Celsius. A 1.5M solution of diisobutylaluminum hydride in toluene (50 g, 350 mmol) was added drop wise over 75 minutes maintaining the internai température between -65 degrees Celsius and -60 degrees Celsius. Once the addition was complété, the reaction mixture was warmed to -10 degrees Celsius for 90 minutes. While maintaining a température of -10 degrees Celsius, an aqueous 4 M solution of potassium hydroxide (350 mL, 10.7 eq) was carefully added drop wise. Once addition was complété, the
reaction mixture was slowly allowed to corne up to room température with vigorous stirring and then allowed to stir at room température for 20 hours. Methyl ferf-butyl ether (200 mL) and heptanes (400 mL) were added and the organic phase was separated. The organic phase was washed with 1 M aqueous potassium hydrogen sulfate, brine, and dried over a mixture of magnésium sulfate and 30 g of silica gel. The solids were filtered off and the filtrate was concentrated under reduced pressure. Upon concentration a precipitate began to form. The resulting wet residue was triturated with 500 mL of 10% methyl teri-butyl ether in heptane at 60 degrees Celsius for 1 hour and the suspension was slowly cooled to room température under stirring. The resulting solids were filtered off and dried in a vacuum oven set at 40 degrees Celsius to give tert-butyl 4-[5-cyano-4-(hydroxymethyl)-1 H-pyrazol-1-yl]piperidine-1-carboxylate (31.2g, 78%). 1H NMR (deuterochloroform) delta ppm 7.59 (s, 1H), 4.70 (d, J - 5.5 Hz, 2H), 4,41 - 4.51 (m, 1 H), 4.17 - 4.32 (m, 2H), 2.81 - 2.96 (m, 2H), 2.01 - 2.16 (m, 3H), 1.94 2.00 (m, 2H), 1.45 (s, 9H)
C) tert-Butyl 4-{5-cvano-4-[(4-cvano-2-fluorophenoxy)methvl1-1 H-pyrazol-1yl)piperidine-1 -carboxylate
To a 4 L bottle was charged tert-butyl 4-[5-cyano-4-(hydroxymethyl)-1 H-pyrazol-1 yl]piperidîne-1-carboxylate (336 g, 1.10 moles), triphenylphosphine (359.58 g, 1.37 moles), 4-cyano-2-fluorophenol (157.89 g, 1.15 moles), and 2-methyltetrahydrofuran (2.02 L, 20.10 moles). The mixture was stirred into solution and kept under nitrogen at room température. To another 4 L bottle was charged, a solution of diethyl diazenedicarboxylate in toluene (564.33 mL, 620.76 g, 1.43 moles) and 2methyltetrahydrofuran (2.12 L, 21.11 moles). The mixture was agitated to ensure complété solution, and kept under nitrogen at room température. A single peristaltic pump was used (two feed lines) to pump the two streams to a T-piece (stainless) followed by 100 mL of coil volume (1/8 followed by 1/4 ID PTE tubing) with a combined flow rate of 20 mL/min. After 8 hours of flowing, the feed bottles were
117
emptied, and 2 x 25 mL of methyltetrahydrofuran was used to rinse the bottles, and pumped through the lines. The product stream was used as is in the following reaction.
D) 4-r(4-Cvano-2-fluorophenoxv)methvll-1-piperidin-4-vl-1H-pvrazole-5-carbonitrile tosylate sait
The stream of tert-butyl 4-{5-cyano-4-[(4-cyano-2-fluorophenoxy)methyl]-1H-pyrazol-1yl}piperidine-1-carboxylate collected in Step C was split into two 5 L single neck flasks. p-Toluenesulfonic acid monohydrate (344.22 g, 1.81 moles) was split charged into the mixture and the flask was heated using a rotary evaporator bath kept at 75 degrees Celsius for 8 hours. The reaction was allowed to cool to room température and granulated overnight. The mixture was filtered and pulled dry under vacuum for 3 hours to give the target product as the tosylate sait (480 g, 88% over two steps).
118
E) 1-Methylcyclopropyl 4-{5-cvano-4-[(4-cvano-2-fluorophenoxy)methyl1-1 H-pyrazol-1yll· pipe rid i ne-1 -carboxylate
The tosylate sait of 4-[(4-cyano-2-fluorophenoxy)methyl]-1-piperidin-4-yl-1H-pyrazole-55 carbonitrile (478 g, 960.71 mmoles) was dissolved in 2-methyltetrahydrofuran (2,39 L,
23.83 moles) and water (478.00 mL) in a 4L bottle. Triethylamine (200,86 mL, 1.44 moles) and 1-methylcyclopropyl 4-nitrophenyl carbonate (Préparation 26) (229.83 g, 960.71 mmoles) were added and stirred for 48 hours. The reaction mixture was washed with aqueous 1N sodium hydroxîde (1 L). The mixture was stirred and the layers separated. The organic layer was washed several times with aqueous 1N sodium hydroxîde (1 L), dried over magnésium sulfate, filtered and the filtrate was concentrated under reduced pressure to give bright yellow solids. These solids were slurried in ethyl acetate at room température overnight. The solids were filtered and the resulting pale yellow solids were re-slurried in ethyl acetate (3 volumes), filtered and pulled dry under vacuum to give the target compound as an off-white solids (313 g, in two batches, 77%). 1H NMR (400 MHz, deuterochloroform) delta ppm 0,60 - 0.66 (m, 2 H) 0.84 - 0.90 (m, 2 H) 1.55 (s, 3 H) 1.96 - 2.04 (m, 2 H) 2.11 (qd, 7=12.10, 4.68 Hz, 2 H) 2.92 (br. s„ 2 H) 4.07-4.41 (m, 2 H) 4.50 (tt, 7=11.27, 4.15 Hz, 1 H) 5.16 (s, 2 H) 7.09 (t, 7=8.20 Hz, 1 H) 7.36 - 7.46 (m, 2 H) 7.68 (s, 1 H).
Melting point = 144.6 degrees Celsius
Combustion Analysis for (Quantitative Technologies Inc. (QTI)
291 Route 22 East
Salem Ind. Park- Bldg 5
Whitehouse NJ 08888-0470
C22H22FN5O3
C (Theoretical=62.40%)
62.28 %
62.29 %
H (Theoretical=5.24%)
119
5.17%
5.13%
N (Theoretical=16.54%)
16.42 %
16.50%
Example 51: ferf-Butyl (3S.4S)-4-(5-cvano-4-([2-fluoro-4(methylcarbamovl)phenoxylmethvl)-1H-pvrazol-1-yl)-3-fluoropiperidine-1-carboxylate
o'
A) ferf-Butyl (3S,4S)-4-[5-cyano-4-(ethoxvcarbonyl)-1 H-pvrazol-1-vl1-3-fluoropiperidine1-carboxylate ferf-Butyl (3S,4S)-4-[5-cyano-4-(ethoxycarbonyl)-1 H-pyrazol-1 -yl]-3-fluoropiperidine-1 carboxylate was prepared from ethyl 5-cyano-1 H-pyrazole-4-carboxylate and (3S.4R)tert-butyl 3-fluoro-4-hydroxypiperidine-1-carboxylate (Préparation 43 B) in a manner similar to that described for the préparation of ferf-butyl 4-[5-cyano-4-(ethoxycarbonyl)1 H-pyrazol-1 -yl]piperidine-1 -carboxylate (Example 50, Step A). The crude material was purified by flash chromatography eluting with a gradient from 0% to 30% ethyl acetate in heptanes to give the desired product as a thick clear oil, (149.4 mg, 32%).
B) fert-Butvl (3S,4S)-4-f5-cyano-4-(hydroxymethvl)-1 H-pvrazol-1-vl1-3-fluoropiperidine-
1-carboxylate ferf-Butyl (3S,4S)-4-[5-cyano-4-(hydroxymethyl)-1 H-pyrazol-1 -yl]-3-fluoropiperidine-1carboxylate was prepared from ferf-butyl (3S,4S)-4-[5-cyano-4-(ethoxycarbonyl)-1 Hpyrazol-1-yl]-3-fluoropiperidine-1 -carboxylate in a manner similar to that described for the préparation of ferf-butyl 4-[5-cyano-4-(hydroxymethyl)-1 H-pyrazol-1-yl]piperidine-1carboxylate (Example 50, Step B). The crude product was purified by flash
I20 chromatography eluting with a gradient from 5% to 50% ethyl acetate in heptanes to give the desired product as a thick clear oil that solidified upon standing (74 mg, 56%).
C) tert-Butyl (3S,4S)-4-(5-cvano-4-ff2-fluoro-4-(methvlcarbamoyl)phenoxy1methyl}-1 Hpyrazol-1-vl)-3-fluoropiperidine-1-carboxylate
The title compound was prepared using 3-fluoro-4-hydroxy-N-methylbenzamide (Préparation 31A), following procedures analogous to Example 50. The crude material was purified by HPLC (Column: Waters Xbridge C12 4.6x50mm, 5 micrometer; Modifier: 0.05% Ammonium hydroxide; Gradient: 95% water / 5% acetonitrile linear to 5% water / 95% acetonitrile over 4.0 min, HOLD at 5% water / 95% acetonitrile to 5.0 min; Flow: 2.OmL/min) to give the desired product. LC/MS (ES+): 476.4 (M+1)
Example: 52: tert-Butyl (3R,4S)-4-(5-cvano-4-i[2-fluoro-4(methvlcarbamoyl)phenoxvlmethvlÎ-1H-pvrazol-1-vl)-3-fluoropiperidine-1-carboxylate
0’
The title compound was prepared using 3-fluoro-4-hydroxy-N-methylbenzamide (Préparation 31 A), following procedures analogous to Example 51. The crude material was purified by HPLC (Column: Waters Xbridge C12 4.6x50 mm, 5 micrometer; Modifier: 0.05% Ammonium hydroxide; Gradient: 95% water / 5% acetonitrile linear to 5% water / 95% acetonitrile over 4.0 min, HOLD at 5% water / 95% acetonitrile to 5.0 min; Flow: 2.OmL/min) to give the desired product. LC/MS (ES+): 476,4 (M+1)
Example 53: 1-Methylcvclopropyl (3S,4S)-4-(5-cyano-4-ff2-fluoro-4(methvlcarbamovl)phenoxylmethvlΐ-1H-pvrazol·1-vl)-3-fluoropiperidine-1-carboxvlate
121
The title compound was prepared using 3-fluoro-4-hydroxy-N-methylbenzamide (Préparation 31 A), foilowing procedures analogous to Examples 50 and 51. The crude material was purified via HPLC (Column: Princeton 2-ethyl pyridine 250 x 21.2 mm 5 micrometer; Gradient: 95% heptane / 5% éthanol for 1.5 minutes, linear to 0% heptane /100% éthanol over 10 min, HOLD at 0% heptane /100% éthanol to 5.0 minfor 1 minute and linear to 95% heptane / 5% éthanol ; Flow: 28 mL/min) to give the desired product. LC/MS (ES+): 473.9 (M+1)
Example 54: 1-Methylcyclopropyl (3R,4R)-4-(5-cyano-4-{[2-fluoro-4(methvlcarbamovDphenoxvlmethylM H-pyrazol-1 -yl)-3-fluoropiperidine-1 -carboxylate
The title compound was prepared using 3-fluoro-4-hydroxy-N-methylbenzamide (Préparation 31 A), foilowing procedures analogous to Examples 50 and 51. The crude material was purified via HPLC (Column: Princeton 2-ethyl pyridine 250 x 21.2mm, 5 micrometer; Gradient: 95% heptane / 5% éthanol for 1.5 minutes, linear to 0% heptane 20 / 100% éthanol over 10min, Hold at 0% heptane /100% éthanol to 5.0 minfor 1 minute and linear to 95% heptane / 5% éthanol ; Flow: 28 mL/min) to give the desired product. LC/MS (ES+): 473.9 (M+1)
Example 55: tert-Butyl (3S,4S)-4-(5-cyano-4-ffî2-methvlpvridin-3-vl)oxv]methvlÎ-1H25 pvrazol-1-vl)-3-fluoropiperidine-1-carboxylate <
122
tert-Butyl 4-(5-cyano-4-{[(methylsulfonyl)oxy]methyl}-1H-pyrazol-1-yl)-3-fluoropiperidine1-carboxylate (Préparation 42) (33 mg, 0.082 mmol) was dissolved in acetonitrile (3 mL) and césium carbonate (53 mg, 0.164mmol) and 3-hydroxy-2-methylpyridine (9 mg, 0.082 mmol) were added. The reaction mixture was heated to 80 degrees Celsius for
1.5 hour. The reaction was cooled to room température and concentrated under reduced pressure. The crude residue was diluted with water and extracted with ethyl acetate (3x). The combined organic extracts were washed with aqueous 0.5N sodium hydroxide, water and brine and dried over sodium sulfate, filtered and the filtrate was concentrated under reduced pressure. The crude residue was purified by flash chromatography eluting with a gradient from 30% to 100% ethyl acetate in heptanes to give the racemic product as an amber oil (30 mg, 70%). 1H NMR (500 MHz, deuterochloroform) delta ppm 1.50 (s, 9 H) 2.11 (m, 1 H) 2.25 - 2.39 (m, 1 H) 2.53 (s, 3 H) 2.93 (br. s., 2 H) 4.30 (br. s., 1 H) 4.43 - 4.71 (m, 2 H) 4.72 - 4.91 (m, 1 H) 5.09 (s, 2 H) 7.09 - 7.20 (m, 2 H) 7.75 (s, 1 H) 8.16 (d, J=3.90 Hz, 1 H)
Example 56: tert-Butyl (3S,4R)-4-(5-cvano-4-f[(2-methvlpvridin-3-vl)oxylmethvl|-1Hpyrazol-1 -yi)-3-fluoropiperidine-1 -carboxylate
The title compound was prepared using commercially available 3-hydroxy-2methylpyridine, following procedures analogous to Example 55. The crude material was purified by flash chromatography eluting with a gradient from 30% to 100% ethyl acetate in heptane to give the desired product as an amber oil. 1H NMR (500 MHz, deuterochloroform) delta ppm 1.50 (s, 9H), 2.01 - 2.08 (m, 1 H) 2.52 (s, 3 H) 2.74 - 2.88 (m, 1 H) 2.94 - 3.14 (m, 1 H) 3.14 - 3.34 (m, 1 H) 4.27 - 4.57 (m, 2 H) 4.61 - 4.75 (m, 1
123
H) 4.80 - 5.01 (m, 1 H) 5.09 (s, 2 H) 7.10 - 7.18 (m, 2 H) 7.73 (s, 1 H) 8.15 (d, J=3.66
Hz, 1 H)
Example 57: 1-Methvlcyclopropyl (3S,4R)-4-(5-cvano-4-ffî2-methvlpyridin-3¥l)oxylmethvlMH-Dvrazol-1-yl)-3-fluoropiperidine-1-carboxylate
The title compound was prepared using commercially available 3-hydroxy-2methylpyridine, following procedures analogous to Example 55. The crude material was purified by flash chromatography, eluting with a gradient of 40% to 100% ethyl acetate in heptanes to give the desired racemtc product as a white solid. 1H NMR (400 MHz, deuterochloroform) delta ppm 0.66 (br. s., 2 H) 0.91 (br. s., 2 H) 1.57 (s, 3 H) 2.05 (d, J=12.10 Hz, 1 H) 2.52 (s, 3 H) 2.82 (br. d, J=9.00 Hz, 1 H) 3.05 (br. d, J=9.00 Hz, 1 H) 3.15 - 3.40 (m, 1 H) 4.20 - 4.60 (m, 2 H) 4.60 - 4.77 (m, 1 H) 4.77 - 5.03 (m, 1 H) 5.09 (s, 2 H) 7.06 - 7.21 (m, 2 H) 7.73 (s, 1 H) 8.16 (d, J=4.29 Hz, 1 H)
Example 58: 1-Methvlcvclopropyl (3S,4R)-4-(5-cvano-4-{[(2-methvlpyridin-3vl)oxv1methvÎMH-pvrazol-1-vl)-3-fluoropiperidine-1-carboxylate
The title compound was prepared using commercially available 3-hydroxy-2methylpyridine, following procedures analogous to Example 55. The crude material was purified by flash chromatography eluting with a gradient of 40% to 100% ethyl acetate in heptanes to give the racemic product which was further purified by chiral HPLC with the following conditions: Column: chiralcel OJ-H 4.6mm x 25cm; Mobile Phase: 85/15
124 carbon dioxide/methanol, Modifier: 0.2% isopropylamine; Flow Rate: 2.5mL/minute to give the title compound. LC/MS (ES+): 414.1 (M+1)
Example 59: 1-Methylcyclopropyl (3R,4S)-4-(5-cvano-4-(i(2-methylpyridin-3vl)oxy]methvlÎ-1 H-pyrazol-1-vlF3-fluoropiperidine-1-carboxylate
The title compound was prepared using commercially available 3-hydroxy-2methylpyridine, following procedures analogous to Example 55. The crude material was purified by flash chromatography eluting with a gradient from 40% to 100% ethyl acetate in heptanes to give the racemic product which was further purified by chiral HPLC with the following conditions: Column: chiralcel OJ-H 4.6mm x 25cm; Mobile Phase: 85/15 carbon dioxide/methanol, Modifier: 0.2% isopropylamine; Flow Rate: 2.5ml_/minute to give the title compound. LC/MS (ES+): 414.1 (M+1)
Example 60: tert-Butyl 4-(5-cyano-4-f[4-(1 H-1,2,3-triazol-1-yl)phenoxv]methvlÎ-1 Hpyrazol-1 -vDpiperidine-1 -carboxylate
The title compound was prepared using 4-(1 H-1,2,3-triazol-1-yl)phenol (US Patent Application No. PCT/US2009/038315, Publication No. WO 2009/129036 A1) following procedures analogous to Example 15. The crude material was purified by HPLC (Column: Phenomenex Gemini C18 250x21.2 mm, 8 micrometer; Mobile Phase: from 50% acetonitrile (ammonia pH 10) in water (ammonia pH 10) to 55% acetonitrile (ammonia pH 10) in water (ammonia pH 10); Flow Rate: 25mL/minute; wavelength: 220 nm) to give the title compound. LC/MS (ES+): 450.1 (M+1)
Example 61: tert-Butyl 4-(5-cyano-4-(r4-(2H-1 ,2,3-triazol-2-vl)phenoxv]methyl)-1 Hpyrazol-1 -vl)piperidine-1 -carboxylate
The title compound was prepared using 4-(2H-1,2,3-triazol-2-yl)phenol (US Patent Application No. PCT/US2009/038315, Publication No. WO 2009/129036 A1) following procedures analogous to Example 15. The crude material was purified by HPLC (Column: Phenomenex Gemini C18 250x21.2 mm, 8 micrometer; Mobile Phase: 63% acetonitrile (ammonia pH 10) in water (ammonia pH 10); Flow Rate: 25mL/minute; wavelength: 220 nm) to give the title compound. LC/MS (ES+): 450.1 (M+1) Example 62: 1-Methvlcyclopropyl 4-(4-((4-(1 H-1,2,3-triazol-1-vl)phenoxv)methyl)-5cyano-1 H-pyrazol-1 -yl)piperidine-1 -carboxylate
O^O
Φ
The title compound was prepared in a manner analogous to Example 60 starting with
Example 60. The crude material was purified by reverse phase HPLC:
Column: Kromasil Eternity-5-C18 150x30 mmx 5 micrometer
Mobile phase: from 38% acetonitrile (0.225% formic acid) in water (0.225% formic acid) to 58% acetonitrile (0.225% formic acid) in water (0.225% formic acid)
126
Flow rate: 30 mL/min
Wavelength: 220 nm
1H NMR (400 MHz, deuterochlorform): delta ppm 7.92 (s, 1H), 7.84 (s, 1H), 7.68 (d, 3H), 7.11 (t, 2H), 5.11 (s, 2H), 4.53 (m, 1H), 4.26 (m, 2H), 2.93 (s, 2H), 2.12(t, 2H), 2.03 5 (d, 2H) 1.56 (s, 3H), 0.88 (t, 2H), 0.64 (t, 2H)
Example 63:1-Methylcyclopropyl 4-(4-((4-(2H-1,2,3-triazol-2-vl)phenoxy)methyl)-5cyano-1 H-pyrazol-1 -vl)piperidine-1 -carboxylate
The title compound was prepared in a manner analogous to Example 61 starting with Example 61. The crude residue was purified by préparative HPLC to yield 50 mg (39%) of the title compound as a white solid:
Column: Boston Symmetrix ODS-H 150x30 mm x 5 micrometer
Mobile phase: from 50% acetonitrile (0.225% formic acid) in water (0.225% formic acid) to 70% acetonitrile (0.225% formic acid) in water (0.225% formic acid)
Flow rate: 30 mL/min
Wavelength: 220 nm 1H NMR (400 MHz, deuterochloroform): delta ppm 8.01 (d, 2H), 7.78 (s, 2H), 7.69 (s,
1H), 7.07 (d, 2H), 5.10 (s, 2H), 4.51 (m, 1H), 4.33 (m, 2H), 2.93 (s, 2H), 2.11 (t, 2H),
2.03 (d, 2H) 1.56 (s, 3H), 0.80 (s, 2H), 0.65 (d, 2H).
Example 64: tert-Butyl 4-r5-cvano-4-(ÎH-(methvlsulfonvl)piperidin-4-vl1oxv}methyl)-1Hpyrazol-1-yl]piperidine-1-carboxylate
127
0^0
The title compound was prepared in a manner analogous to Example 13. The crude compound was purified by siiica gel chromatography using an 1:4 mixture of petroleum ether and ethyl acetate.
1H NMR (400 MHz, deuterochloroform): delta ppm 7.54 (s, 1 H), 4.53 (s, 2H), 4.48 (m,
H), 4.28 (br, 2H), 3.69 (m, 1 H), 3.31 (m, 4H), 2.90 (m, 2H), 2.79 (s, 3H), 2.11 (m, 2H), 1.88-2.00 (m,6H), 1.47 (s, 9H).
Example 65: terf-Butyl 4-[5-cvano-4-((2-fluoro-4-[(2hvdroxvethvlXmethvl)carbamoyl1phenoxvÎmethyl)-1H-pyrazol-1-vl1piperidine-1carboxylate
The title compound was prepared in a manner analogous to Example 46. The crude materiai was purified by reverse phase HPLC:
Column: Phenomenex Gemini C18 250x21.2 mm x 8 micrometer
128
Mobile phase: from 40% acetonitrile (ammonia pH 10) in water (ammonia pH 10) to
60% acetonitrile (ammonia pH 10) in water (ammonia pH 10)
Flow rate: 25 mL/min
Wavelength: 220 nm 1H NMR (400 MHz, deuterochloroform): delta ppm 7.69 (s, 1H), 7.28 (d, 1H), 7.24 (s,
1H), 7.05 (d, 1H), 5.18 (s, 2H). 4.50 (q, 1H), 4.29 (d, 2H), 3.90 (s, 2H), 3.71 (s, 2H),
3.10 (s, 3H), 2.91 (s, 2H), 2.14 (q, 2H), 1.99 (s, 2H), 1,48 (s, 9H).
Example 66: tert-Butyl 4-[5-cvano-4-(f2-fluoro-4-Î(3-hvdroxvpyrrolidin-1vl)carbonvllphenoxv)methvl)-1 H-pyrazol-1-yllpiperidine-1-carboxylate
The title compound was prepared in a manner analogous to Example 46. The crude material was purified by reverse phase HPLC:
Column: Phenomenex Gemini C18 250x21.2 mm x 8 micrometer
Mobile phase: from 40% acetonitrile (ammonia pH 10) in water (ammonia pH 10) to
60% acetonitrile (ammonia pH 10) in water (ammonia pH 10)
Flow rate: 25 mL/min
Wavelength: 220 nm 1H NMR (400 MHz, deuterochloroform): delta ppm 7.69 (s, 1H), 7.32 (d, 2H), 7.05 (t, 1H), 5.14 (s, 2H), 4.52 (q, 2H), 4.29 (s, 2H), 3.78 (d, 2H), 3.64 (d, 1H), 3.45 (d, 1H),
2.90 (s, 2H), 2.14 (q, 2H), 2.00 (d, 4H), 1.47 (s, 9H).
Example 67: tert-Butyl 4-(4-{i4-(azetidin-1-ylcarbonvl)-2-fluorophenoxy1methvl)-5-cyano1 H-pyrazol-1-vl)piperidine-1 -carboxylate
129
The title compound was prepared in a manner analogous to Example 46. The crude material was purified by reverse phase HPLC:
Column: Phenomenex Gemini C18 250x21.2 mm x 8 micrometer
Mobile phase: from 40% acetonitrile (ammonia pH 10) in water (ammonia pH 10) to
60% acetonitrile (ammonia pH 10) in water (ammonia pH 10)
Flow rate: 25 mL/min
Wavelength: 220 nm 1H NMR (400 MHz, deuterochloroform): delta ppm 7.69 (s, 1H), 7.43 (d, 1H), 7.41 (s, 10 1 H), 7.04 (t, 1 H), 5.14 (s, 2H), 4.50 (q, 1 H), 4.31 (d, 6H), 2.90 (d, 2H), 2.38 (q, 2H),
2.15 (q, 2H), 2.08 (d, 2H), 1.47 (s, 9H).
Example 68: 1-Methylcyclopropyl 4-[5-cvano-4-({[1-(methylsulfonvl)piperidin-4yl1oxv)methyl)-1 H-pyrazol-1 -yllpiperidine-1 -carboxylate
The title compound was prepared in a manner analogous to Example 64. The crude material was purified by reverse phase HPLC:
Column: Phenomenex SynergiC18 150x30 mm x4 micrometer
130
Mobile phase: 43% acetonitrile (0.225% formic acid) in water (0.225% formic acid) to
53% acetonitrile (0.225% formic acid) in water (0.225% formic acid)
Flow Rate: 30mL/min
1H NMR (400 MHz, deuterochloroform): delta ppm 7.54 (s, 1H), 4.53 (s, 2H), 4.48 (m,
1 H), 4.35 (d, 2H), 3.69 (m, 1 H), 3.31 (m, 4H), 2.91 (m, 2H), 2.78 (s, 3H), 2.12(m, 2H),
1.87-1.98 (m, 6H), 1.56 (s, 3H), 0.88 (t, 2H), 0.66 (t, 2H).
Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application for ail purposes.
It will be apparent to those skilled in the art that various modifications and variations can be made in the présent invention without departing from the scope or spirit of the invention. Other embodiments of the invention will be apparent to those skilled in the art from considération of the spécification and practice of the invention disclosed herein. It is intended that the spécification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.

Claims (5)

What is claimed is:
1 -Methylcyclopropyl 4-{5-cyano-4-[(4-cyano-2-fluorophenoxy)methyl]-1 H-pyrazol-1 y l}pi perid i n e-1 -carboxylate;
or a pharmaceutically acceptable sait thereof.
1-[1 -(5-Ethylpyrimidin-2-yl)piperidin-4-yll-4-{[4-(1 H-tetrazol-1-yl)phenoxy]methyl}-1 H pyrazole-5-carbonitrile; 4-[(4-Cyano-2-fluorophenoxy)methyl]-1-[1-(5-ethylpyrimidin-2-yl)piperidin-4-yl]-1Hpyrazole-5-carbonitrile; and
1-Methylcyclopropyl 4-(5-cyano-4-{[4-(1 H-tetrazol-1 -yl)phenoxy]methyl}-1 H-pyrazol-1yl)piperidine-1-carboxylate;
1-Methylcyclopropyl 4-(5-cyano-4-{[2-fluoro-4-(1-methyl-1H-tetrazol-5yl)phenoxy]methyl}-1 H-pyrazol-1 -yl Jpiperid ine-1 -carboxylate;
1-methylcyclopropyl 4-(4-((4-(azetidine-1-carbonyl)-2-fluorophenoxy)methyl)-5-cyano1 H-pyrazol-1-yl)piperid ine-1-carboxylate;
isopropyl 4-(4-((4-(azetidine-1-carbonyl)-2-fluorophenoxy)methyl)-5-cyano-1H-pyrazol1 -yl)piperidine-1 -carboxylate; and
1-(1-(5-ethylpyrimidin-2-yl)piperidin-4-yl)-4-((2-fluoro-4-(1-methyl-1H-tetrazol-5yl)phenoxy)methyl)-1H-pyrazole-5-carbonitrile;
1-methylcyclopropyl 4-(5-cyano-4-((2-methyl-6-(1 H-1,2,3-triazol-1-yl)pyridin-3yloxy)methyl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate;
1-(1-(5-ethylpyrimidin-2-yl)piperidin-4-yl)-4-((2-methyl-6-(1H-112,3-triazol-1-yl)pyridin-3yloxy)methyl)-1H-pyrazole-5-carbonitrile;
isopropyl 4-(5-cyano-4-((2-methyl-6-(1 H-1,2,3-triazol-1 -y I )pyr idin-3-y loxy)methyl)-1 Hpyrazol-1-yl)piperidine-1-carboxylate;
1-(1-(5-ethylpyrimidin-2-yl)piperidin-4-yl)-4-((3-fluoro-4-(1 H-tetrazol-1 yl)phenoxy)methyl)-1H-pyrazole-5-carbonitrile;
1-methylcyclopropyl 4-(5-cyano-4-((3-fluoro-4-(1 H-tetrazol-1-yl)phenoxy)methyl)-1 Hpyrazol-1 -yl )p i perid i ne-1 -carboxylate;
isopropyl 4-(5-cyano-4-((3-fluoro-4-(1 H-tetrazol-1-yl)phenoxy)methyl)-1 H-pyrazol-1yl ) pîperid in e-1 -carboxylate;
1-methylcyclopropyl 4-(4-((5-(1 H-1 l2,3-triazol-1-yl)pyridin-2-yloxy)methyl)-5-cyano-1 Hpyrazol-1 -yl )pi perid ine-1 -carboxylate;
isopropyl 4-(4-((5-(1 H-1,2,3-triazol-1-yl)pyridin-2-yloxy)methyl)-5-cyano-1 H-pyrazol-1 yl)piperidine-1-carboxylate;
1 -(1 -(5-ethylpyrimidin-2-yl)piperidin-4-yl)-4-((2-fluoro-4-(2H-1,2,3-triazol-2yl)phenoxy)methyl)-1H-pyrazole-5-carbonitrile;
1-methylcyclopropyl 4-(5-cyano-4-((2-fluoro-4-(2H-1,2,3-triazol-2-yl)phenoxy)methyl)1 H-pyrazol-1-yl)piperidine-1-carboxylate;
isopropyl 4-(5-cyano-4-((2-fluoro-4-(2H-1,2,3-triazol-2-yl)phenoxy)methyl)-1 H-pyrazol-1 yl )pi perid i ne-1 -carboxylate;
135
1-(1-(5-ethylpyrimidin-2-yl)piperidin-4-yl)-4-((2-fluoro-4-(1 H-1,2,3-triazol-1yl)phenoxy)methyl)-1H-pyrazole-5-carbonitrile;
isopropyl 4-(4-((4-(1 H-1,2,3-triazol-1-yl)phenoxy)methyl)-5-cyano-1H-pyrazol-1yl)piperidine-1-carboxylate;
1-methylcyclopropyl 4-(5-cyano-4-((2-fluoro-4-(1 H-1,2,3-triazol-1-yl)phenoxy)methyl)1 H-pyrazol-1-yl)piperidine-1-carboxylate;
isopropyl 4-(5-cyano-4-((2-fluoro-4-(1 H-1,2,3-triazol-1-yl)phenoxy)methyl)-1 H-pyrazol-1yl)piperidine-1-carboxylate;
1-Methylcyclopropyl 4-[5-cyano-4-({[1-(methylsulfonyl)piperidin-4-yl]oxy}methyl)-1Hpyrazol-1-yl]piperidine-1-carboxylate;
1-Methylcyclopropyl 4-(4-((4-(2H-1,2,3-triazol-2-yl)phenoxy)methyl)-5-cyano-1 Hpyrazol-1 -yl )piperidine-1 -carboxylate;
tert-Butyl 4-[5-cyano-4-({[1-(methylsulfonyl)piperidin-4-yl]oxy}methyl)-1 H-pyrazol-1 yljpiperîd in e-1 -carboxylate;
tert-Butyl 4-[5-cyano-4-((2-fluoro-4-[(2hydroxyethyl)(methyl)carbamoyl]phenoxy}methyl)-1 H-pyrazol-1-yl]piperidine-1carboxylate;
tert-Butyl 4-[5-cyano-4-({2-fluoro-4-[(3-hydroxypyrrolidin-1-yl)carbonyl]phenoxy)methyl)1 H-pyrazol-1 -y IJpi perid ine-1 -carboxylate;
tert-Butyl 4-(4-{[4-(azetidin-1-ylcarbonyl)-2-fluorophenoxy]methyl)-5-cyano-1H-pyrazol1 -y J )p i perid i ne-1 -carboxylate;
1-Methylcyclopropyl 4-(4-((4-(1 H-1,2,3-triazol-1-yl)phenoxy)methyl)-5-cyano-1 Hpyrazol-1 -yl )piperidine-1 -carboxylate;
1-Methylcyclopropyl (3R,4S)-4-(5-cyano-4-{[(2-methylpyridin-3-yl)oxy]methyl}-1 Hpyrazol-1-yl)-3-fluoropiperidine-1-carboxylate;
tert-Butyl 4-(5-cyano-4-{[4-(1H-1,2,3-triazol-1-yl)phenoxy]methyl}-1 H-pyrazol-1yl)piperidine-1-carboxylate;
134 tert-Butyl 4-(5-cyano-4-([4-(2H-1,2,3-triazol-2-yl)phenoxy]methyl}-1 H-pyrazol-1 yl ) piperid i n e-1 -carboxylate;
1 -Methylcyclopropyl (3S,4R)-4-(5-cyano-4-{[(2-methylpyridin-3-yl)oxy]methyl}-1 Hpyrazol-1 -yl )-3-f lu oro pi pe rid ine-1 -carboxylate;
1 -Methylcyclopropyl (3S,4R)-4-(5-cyano-4-{[(2-methylpyridin-3-yl)oxy]methyl}-1 Hpyrazol-1 -yl)-3-fluoropiperîdine-1 -carboxylate;
1 -Methylcyclopropyl (3R,4R)-4-(5-cyano-4-{[2-fluoro-4(methylcarbamoyl)phenoxy]methyl)-1H-pyrazol-1-yl)-3-fluoropiperidine-1 -carboxylate; tert-Butyl (3S,4S)-4-(5-cyano-4-{[(2-methylpyridin-3-yl)oxy]methyl}-1 H-pyrazol-1-yl)-3fluoropiperidine-1 -carboxylate;
tert-Butyl (3S,4R)-4-(5-cyano-4-{[(2-methylpyridin-3-yl)oxy]methyl}-1 H-pyrazol-1-yl)-3fluoropiperidine-1 -carboxylate;
1 -Methylcyclopropyl (3S,4S)-4-(5-cyano-4-{[2-fluoro-4(methylcarbamoyl)phenoxy]methyl}-1H-pyrazol-1-yl)-3-fluoropiperidine-1-carboxylate;
1-Methylcyclopropyl 4-{5-cyano-4-[(4-cyano-2-fluorophenoxy)methyl]-1 H-pyrazol-1yl)piperidine-1-carboxylate;
tert-Butyl (3S,4S)-4-(5-cyano-4-{[2-fluoro-4-(methylcarbamoyl)phenoxy]methyl}-1Hpyrazol-1 -yl)-3-fluoropiperidine-1 -carboxylate;
tert-Butyl (3R,4S)-4-(5-cyano-4-{[2-fluoro-4-(methylcarbamoyl)phenoxy]methyl}-1Hpyrazol-1-yl)-3-fluoropiperidine-1 -carboxylate;
1-Methylcyclopropyl 4-(5-cyano-4-{[2-fluoro-4-(methylcarbamoyl)phenoxy]methyl)-1Hpyrazol-1 -yl)piperidine-1 -carboxylate;
1 -Methylcyclopropyl 4-(5-cyano-4-([4-(dimethylcarbamoyl)-2-fluorophenoxy]methyl}-1 Hpyrazol-1-yl)piperidine-1-carboxylate;
1-[1-(5-Ethylpyrimidin-2-yl)piperidin-4-yl]-4-{[4-(1H-tetrazol-1-yl)phenoxy]methyl}-1Hpyrazole-5-carbonitrile;
Isopropyl 4-{5-cyano-4-[(3-cyanophenoxy)methyl]-1 H-pyrazol-1-yl}piperidine-1carboxylate;
Isopropyl 4-{5-cyano-4-[(4-cyano-3-methylphenoxy)methyl]-1 H-pyrazol-1 -yl}piperidine1 -carboxylate;
Isopropyl 4-{5-cyano-4-[(4-cyanophenoxy)methyl]-1 H-pyrazol-1-yl}piperidine-1carboxylate;
1-Methylcyclopropyl 4-(5-cyano-4-{[4-(1 H-tetrazol-1-yl)phenoxy]methyl}-1 H-pyrazol-1yl )piperid ine-1 -carboxylate;
1-Methylcyclopropyl 4-{5-cyano-4-[(2,3,6-trifluorophenoxy)methyl]-1 H-pyrazol-1yl)pi perid in e-1 -carboxylate;
Isopropyl 4-{5-cyano-4-[(2,3,6-trifluorophenoxy)methyl]-1 H-pyrazol-1 -yl}piperidine-1carboxylate;
Isopropyl 4-(5-cyano-4-{[2-fluoro-4-(1 -methyl-1 H-imidazol-2-yl)phenoxy]methyl}-1 Hpyrazol-1 -y I )pi perid i ne-1 -carboxylate;
Isopropyl 4-(5-cyano-4-{[2-fluoro-4-(1 -methyl-1 H-imidazol-5-yl)phenoxy]methyl}-1 Hpyrazol-1 -yl )p i perid ine-1 -carboxylate;
Isopropyl 4-[5-cyano-4-({[2-methyl-6-(1 H-1 ^Atriazol-l-yOpyridin-S-ylJoxyJmethylJ-l Hpyrazol-1 -y l]p i perid ine-1 -carboxylate;
Isopropyl 4-[5-cyano-4-({[2-methyl-6-(1 H-1,2,4-triazol-l-yl)pyridin-3-yl]amino}methyl)1 H-pyrazol-1 -yl]piperidine-1 -carboxylate;
Isopropyl 4-[5-cyano-4-({[2-methyl-6-(methylsulfonyl)pyridin-3-yl]amino}methyl)-1 Hpyrazol-1 -y l] p ip erid ine-1 -carboxylate;
1-Methylcyclopropyl 4-(5-cyano-4-{[(2-methylpyridin-3-yl)oxy]methyl}-1 H-pyrazol-1yl)piperidine-1 -carboxylate;
1-Methylcyclopropyl 4-(4-((4-carbamoyl-3-fluorophenoxy)methyl)-5-cyano-1 H-pyrazol-1 35 yl)piperidine-1 -carboxylate;
132
Isopropyl 4-(5-cyano-4-{1-[2-fluoro-4-(methylsulfonyl)phenoxy]ethyl}-1 H-pyrazol-1 yl )piperid i ne-1 -carboxylate;
Isopropyl 4-(5-cyano-4-{1-[(2-methylpyridin-3-yl)oxy]ethyl}-1 H-pyrazol-1 -yl)piperidine-1carboxylate;
Isopropyl 4-(5-cyano-4-{2-[2-fluoro-4-(methylsulfonyl)phenyl]propyl}-1 H-pyrazol-1 yl)piperidine-1-carboxylate;
1-Methylcyclopropyl 4-(5-cyano-4-{[4-(1 -methyl-1 H-tetrazol-5-yl)phenoxy]methyl}-1Hpyrazol-1 -yl )piperid ine-1 -carboxylate;
1 -Methylcyclopropyl 4-{4-[(4-carbamoylphenoxy)methyl]-5-cyano-1 H-pyrazol-1 y l}p i pe rid i ne-1 -carboxylate;
15 1 -Methylcyclopropyl 4-(5-cyano-4-((4-cyanophenoxy)methyl)-1 H-pyrazol-1-yl)piperidine1-carboxylate;
Isopropyl 4-(4-((4-(1 H-pyrazol-1-yl)phenoxy)methyl)-5-cyano-1 H-pyrazol-1-yl)piperidine1-carboxylate;
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(1 H-tetrazol-5-yl)phenoxy)methyl)-1 H-pyrazol-120 yl)piperidine-1-carboxylate and Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2H-tetrazol-5yl)phenoxy)methyl)-1 H-pyrazol-1-yl)piperidine-1-carboxylate;
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(1-methyl-1 H-tetrazol-5-yl)phenoxy)methyl)-1Hpyrazol-1 -yl )piperid ine-1 -carboxylate;
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-methyl-2H-tetrazol-5-yl)phenoxy)methyl)-1H25 pyrazol-1 -yl )pi pe rid rn e-1 -carboxylate;
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(2-(2-hydroxyethyl)-2H-tetrazol-5yl)phenoxy)methyl)-1 H-pyrazol-1 -yl)piperidine-1 -carboxylate;
Isopropyl 4-(5-cyano-4-((2-fluoro-4-(1-(2-hydroxyethyl)-1H-tetrazol-5yl)phenoxy)methyl)-1H-pyrazol-1-yl)piperidine-1-carboxylate;
30 1 -Methylcyclopropyl 4-(5-cyano-4-{[2-fluoro-4-( 1 -methyl-1 H-tetrazol-5yl)phenoxy]methyl}-1H-pyrazol-1-yl)piperidine-1-carboxylate;
1 -Methylcyclopropyl 4-{4-[(4-carbamoyl-2-fluorophenoxy)methyl]-5-cyano-1 H-pyrazol-1 y IJpiperid ine-1 -carboxylate;
1 -Methylcyclopropyl 4-{5-cyano-4-[(2,3-difluorophenoxy)methyl]-1 H-pyrazol-1 10 yl}piperidine-1-carboxylate;
1. A compound selected from the group consisting of:
Isopropyl 4-{5-cyano-4-[(2,4-difluorophenoxy)methyl]-1 H-pyrazol-1-yl}piperidine-15 carboxylate;
Isopropyl 4-{5-cyano-4-[(2-methylphenoxy)methyl]-1 H-pyrazol-1-yl}piperidine-1carboxylate; 1-Methylcyclopropyl 4-{5-cyano-4-[(2,5-difluorophenoxy)methyl]-1H-pyrazol1 -y I} piperid in e-1 -carboxylate;
2. A compound selected from the group consisting of:
3. A pharmaceutical composition comprisîng a compound according to any of daims 1-2, présent in a therapeutically effective amount, in admixture with at least one pharmaceutically acceptable excipient.
4. The composition of claim 3 further comprisîng at least one additional pharmaceutical agent selected from the group consisting of an anti-obesity agent and an anti-diabetic agent.
5. The composition of Claim 4 wherein said anti-obesity agent is selected from the group consisting of dirlotapide, mitratapide, implitapide, R56918 (CAS No. 403987), CAS No. 913541-47-6, lorcaserin, cetilistat, PYY3.36. naltrexone, oleoyl-estrone, obinepitide, pramlintide, tesofensine, leptin, liraglutide, bromocriptine, orlistat, exenatide, AOD-9604 (CAS No. 221231-10-3) and sibutramine.
6. The composition of Claim 4 wherein said anti-diabetic agent is selected from the group consisting of metformin, acetohexamide, chlorpropamide, diabinese, glibenclamide, glipizide, glyburide, glimepiride, gliclazide, glipentide, gliquidone, glisolamide, tolazamide, tolbutamide, tendamistat, trestatin, acarbose, adiposine, camiglibose, emiglitate, miglitol, voglibose, pradimicin-Q, salbostatin, balaglitazone, ciglitazone, darglitazone, englitazone, isaglitazone, pioglitazone, rosiglitazone, troglitazone, exendin-3, exendin-4, trodusquemine, reservatrol, hyrtiosal extract, sitagliptin, vlldagliptin, alogliptin and saxagliptin.
7. Use of a compound according to claim 1 or 2 in the manufacture of a pharmaceutical composition for the treatment of diabètes.
137
8. Use of a compound of claim 1 or 2 in the manufacture of a pharmaceutical composition for treating a metabolic or metabolic-related disease, condition or disorder.
9. Use of a compound of claim 1 or 2 in the manufacture of a pharmaceutical composition for treating a condition selected from the group consisting of hyperlipidemia, Type I diabètes, Type II diabètes mellitus, idiopathic Type I diabètes (Type Ib), latent autoimmune diabètes in adults, early-onset Type 2 diabètes, youth-onset atypical diabètes, maturity onset diabètes of the young, malnutrition-related diabètes, gestational diabètes, coronary heart disease, ischémie stroke, restenosis after angioplasty, peripheral vascular disease, intermittent claudication, myocardial infarction (e.g. necrosis and apoptosis), dyslipidemia, post-prandial lipemia, conditions of impaired glucose tolérance, conditions of impaired fasting plasma glucose, metabolic acidosis, ketosis, arthritis, obesity, osteoporosis, hypertension, congestive heart failure, left ventricular hypertrophy, peripheral arterial disease, diabetic retinopathy, macular degeneration, cataract, diabetic nephropathy, glomerulosclerosis, chronic rénal failure, diabetic neuropathy, metabolic syndrome, syndrome X, premenstrual syndrome, coronary heart disease, angina pectoris, thrombosis, atherosclerosis, myocardial infarction, transient ischémie attacks, stroke, vascular restenosis, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertrygliceridemia, insulin résistance, impaired glucose metabolism, conditions of impaired glucose tolérance, conditions of impaired fasting plasma glucose, obesity, erectile dysfunction, skin and connective tissue disorders, foot ulcérations and ulcerative colitis, endothélial dysfunction and impaired vascular compliance, hyper apo B lipoproteinemia, Alzheimer’s disease, schizophrenia, impaired cognition, inflammatory bowel disease, ulcerative colitis, Crohn’s disease, and irritable bowel syndrome.
10. Use of a compound according to any of claims 1 to 2 in admixture with at least one pharmaceutically acceptable excipient in the manufacture of a first composition, and at least one additional pharmaceutical agent selected from the group consisting of an anti-obesity agent and an anti-diabetic agent and at least one pharmaceutically acceptable sait in the manufacture of a second composition, for treating a metabolic or metabolic-related disease, condition or disorder, said compositions being for separate administration to a patient in need of such treatment.
I38
11. The use of claim 10 wherein said first composition and said second composition are for simultaneous administration.
4-((4-(azetidine-1-carbonyl)-2-fluorophenoxy)methyl)-1-(1-(5-ethylpyrimidin-2yl)piperidin-4-yl)-1 H-pyrazole-5-carbonitrile;
or a pharmaceutically acceptable sait thereof.
4-((5-(1 H-1,2,3-triazol-1-yl)pyridin-2-yloxy)methyl )-1-(1 -(5-ethylpyrimidin-2-yl)piperidin4-yl)-1H-pyrazole-5-carbonitrile;
4-((4-(2H-1,2,3-triazol-2-yl)phenoxy)methyl)-1-(1-(5-ethylpyrimidin-2-yl)piperidrn-4-yl)1 H-pyrazole-5-carbonitrile;
4-((4-(1 H-1,2,3-triazol-1 -yl)phenoxy)methyl)-1 -(1-(5-ethylpyrimidin-2-yl)piperidin-4-yl)1 H-pyrazole-5-carbonitrile;
isopropyl 4-(4-((4-(2H-1,2,3-triazol-2-yl)phenoxy)methyl)-5-cyano-1 H-pyrazol-1yl)piperidine-1-carboxylate;
4-({5-Cyano-1-[1-(5-ethylpyrimidin-2-yl)piperidin-4-yl]-1H-pyrazol-4-yl}methoxy)-3-fluoroN,N-dimethylbenzamide;
4-[(4-Cyano-2-fluorophenoxy)methyl]-1 -[1 -(5-ethyl pyrimidin-2-y I )pi peridin-4-yl]-1 Hpyrazole-5-carbonitrile;
133 tert-Butyl 4-{5-cyano-4-[(4-cyano-2-fluorophenoxy)methyl]-1 H-pyrazol-1 -yl}piperidine-1 carboxylate;
Isopropyl 4-{5-cyano-4-[(2-cyano-4-fluorophenoxy)methyl]-1 H-pyrazol-1 -ylJpiperidine-1 carboxylate;
Isopropyl 4-(5-cyano-4-{[4-(dimethylcarbamoyl)-2-fluorophenoxy]methyl}-1 H-pyrazol-1 y I )pi perid i ne-1 -carboxylate;
5 12. The use of claim 10 wherein said first composition and said second composition are for sequential administration and in any order.
OA1201300182 2010-11-23 2011-11-09 4-(5-cyano-pyrazol-1-yl)-piperidine derivatives as GPR 119 modulators. OA16400A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US61/416,441 2010-11-23

Publications (1)

Publication Number Publication Date
OA16400A true OA16400A (en) 2015-10-07

Family

ID=

Similar Documents

Publication Publication Date Title
AU2010255422B2 (en) 1- ( piperidin-4-yl) -pyrazole derivatives as GPR 119 modulators
US20120052130A1 (en) Gpr 119 modulators
EP2427450A1 (en) Gpr 119 modulators
EP2401265B1 (en) Derivatives of 1-pyri(mid)in-2-yl-pyrazole-4-carboxylic acid which are useful for therapy or prophylaxis of cardiovascular diseases
AU2011333427A1 (en) 4- (5-cyano-pyrazol-1-yl) -piperidine derivatives as GPR 119 modulators
US20100285145A1 (en) Gpr 119 modulators
EP2408780A2 (en) 3-oxa-7-azabicycloý3.3.1¨nonanes
JP5451633B2 (en) Triazole oxadiazole derivatives
US20120295845A1 (en) Imidazo-pyrazoles as gpr119 inhibitors
WO2012117996A1 (en) Gpr119 agonist
US20130072427A1 (en) Gpr 119 modulators
OA16400A (en) 4-(5-cyano-pyrazol-1-yl)-piperidine derivatives as GPR 119 modulators.