CN104725380B - Lu Suo replaces the deuterated derivative of Buddhist nun - Google Patents

Lu Suo replaces the deuterated derivative of Buddhist nun Download PDF

Info

Publication number
CN104725380B
CN104725380B CN201310700192.2A CN201310700192A CN104725380B CN 104725380 B CN104725380 B CN 104725380B CN 201310700192 A CN201310700192 A CN 201310700192A CN 104725380 B CN104725380 B CN 104725380B
Authority
CN
China
Prior art keywords
compound
deuterium
pharmaceutically acceptable
hydrogen
acceptable salt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201310700192.2A
Other languages
Chinese (zh)
Other versions
CN104725380A (en
Inventor
I·R·西尔弗曼
J·F·刘
A·J·摩根
B·潘德雅
S·L·哈比森
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sun Pharmaceutical Industry Co ltd
Original Assignee
Concert Pharmaceuticals 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 Concert Pharmaceuticals Inc filed Critical Concert Pharmaceuticals Inc
Priority to CN201910481275.4A priority Critical patent/CN110229159B/en
Priority to CN201310700192.2A priority patent/CN104725380B/en
Publication of CN104725380A publication Critical patent/CN104725380A/en
Application granted granted Critical
Publication of CN104725380B publication Critical patent/CN104725380B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Abstract

The present invention relates to the deuterated derivatives that Lu's rope replaces Buddhist nun.A kind of embodiment of the invention provides the method for the compound or its pharmaceutically acceptable salt, the pharmaceutical composition containing the compound and treatment indication disclosed herein of formula A.

Description

Lu Suo replaces the deuterated derivative of Buddhist nun
Background of invention
There is undesirable absorption, distribution, metabolism and/or excretion (ADME) performance, this obstruction in current many drugs It is widely applied or limits its purposes in certain indications.Undesirable ADME performance is also drug candidate in clinical test The main reason for middle failure.Although preparation technique and prodrug strategies can be used for improving in some cases certain ADME performances, But these approach tend not to solve the problems, such as essence ADME existing for many drugs and drug candidate.One such problems It is tachymetabolism, some many drugs that should efficiently treat disease is caused to be removed too quickly from internal.Quick medicament is removed Possible solution be it is frequent or it is large dosage of medication to reach sufficiently high drug blood plasma level.However, which introduce permitted More potential treatment problems, as compliance of the patient to dosage regimen is poor, become side effect more sharply under higher dosage and The increase for the treatment of cost.The drug of rapid metabolization is also possible to that patient is made to be exposed to undesirable toxicity or reactive metabolin.
Another ADME limitation for influencing many drugs is the formation of toxicity or biological reactivity metabolin.Therefore, one Receive a bit drug patients may be poisoned or the safe dose of the drug may be restricted such that patient receives time most Good activating agent.In some cases, changing dosing interval or preparation way can help to reduce clinical adverse, but logical The formation of normal this undesirable metabolin is intrinsic for the metabolism of compound.
In the case where certain selections, metabolic poison will be co-administered with the drug of too fast removing.For treating HIV sense The protease inhibitors class drug of dye is exactly such situation.FDA (food and drug administration) recommend these drugs with Cytochrome P 450 enzymes 3A4(CYP3A4) (metabolism for being generally responsible for these drugs) inhibitor Ritonavir co-administered (ginseng See Kempf, D.J. etc., Antimicrobial agents and chemotherapy, 1997,41 (3): 654-60).However, Ritonavir causes adverse reaction, and the drug for having aggravated the HIV patient of the former different pharmaceutical that must have taken combination is negative Load.Similarly, in order to reduce treatment pseudobulbar mood (pseudobulbar affect) in dextromethorphan it is quick CYP2D6 metabolism, CYP2D6 inhibitor quinindium are added in dextromethorphan.However, quinindium has harmful side effect, This greatly limits it in latent purposes in combination therapy (referring to Wang, L etc., Clinical Pharmacology and Therapeutics,1994,56(6Pt1):659-67;And the FDA of the quinindium on www.accessdata.fda.gov Label).
Make us full in general, drug is not intended to reduce Drug total clearance in conjunction with cytochrome P 450 inhibitors The strategy of meaning.Inhibit the activity of CYP enzyme that may influence the metabolism and removing of the other drugs being metabolized by the same enzyme.CYP suppression System may cause other drugs and be accumulated in vivo to toxic level.
A kind of strategy for having potential attraction for improving drug metabolism performance is deuterium modification.In this approach, Ren Menshi Figure slows down the drug metabolism of CYP mediation by substituting one or more hydrogen atoms with D-atom or reduces undesirable metabolin Formation.Deuterium is safe and stable, on-radiation hydrogen isotope.Compared with hydrogen, deuterium and carbon form stronger key.In selection In the case of, the bond strength increase assigned by deuterium can improve efficacy of drugs, peace to generate with the ADME property of positive influence drug The potential of full property and/or tolerance.Meanwhile because the size and shape of deuterium and the size and shape of hydrogen are essentially identical, and only wrap Hydrogeneous original chemical entity compares, by the estimated biochemical activity and selectivity that will not affect that drug of deuterium substitution hydrogen.
Between past 35 years, the influence (ginseng to metabolic rate is replaced to the approval drug report of considerably less ratio deuterium See, for example, Blake, MI etc., J Pharm Sci, 1975,64:367-91;Foster,AB,Adv Drug Res,1985,14: 1-40("Foster");Kushner, DJ etc., Can J Physiol Pharmacol, 1999,79-88;Fisher, MB etc., Curr Opin Drug Discov Devel, 2006,9:101-09 (" Fisher ")).Many examples report in these documents The deuterium isotope effect (influence to the metabolic rate in deuterated site specific in substrate) of part rather than deuterate are for this The influence of drug overall metabolic stability, i.e., via the overall base consumption of metabolism.That is reported replaces to totality measurement deuterium Those of influence of metabolic stability research the result is that changeable and unpredictable.To some compounds, deuterate causes body Intracellular metabolite clearance rate reduces.To other compounds, without metabolic alterations.Compound also shows the increasing of metabolic clearance rate Add.The variability of deuterium effect also causes technical staff to suspect or abandon that deuterium modification is set as the feasible drug of the unfavorable metabolism of inhibition Stratagem omits (page 101 referring to page 35 of Foster and Fisher).
Influence of the deuterium modification to pharmacokinetic properties is not predictable, even if mixing known metabolism site in D-atom In the case where.People determine whether and how metabolic rate is different from only possible through the practical drug for preparing and testing deuterate The counterpart of its non-deuterate.See, e.g., Fukuto etc. (J.Med.Chem., 1991,34,2871-76).Many drugs have The multiple sites that may be metabolized.It needs to carry out the substituted site of deuterium and sees that the influence (if any) to metabolism must The degree of deuterium needed will be different various drugs.
Lu Suo for Buddhist nun (Ruxolitinib) phosphate be heteroaryl replace pyrrolo- [2,3-d] pyrimidine, also referred to as 3 (R)-cyclopenta -3- [4- (7H- pyrrolo- [2,3-d] pyrimidine-4-yl) -1H- pyrazol-1-yl] propionitrile phosphate, and (R) - 3- (4- (7H- pyrrolo- [2,3-d] pyrimidine-4-yl) -1H- pyrazol-1-yl) -3- cyclopenta propionitrile phosphate inhibits Janus Associated kinase (JAK) JAK1 and JAK2.These the kinase mediated cytokine profiles and life important for hematopoiesis and immune function The signal transduction of the long factor.JAK signal transduction is related to STAT(signal transducer and transcriptional activator) to cytokine receptor Recruitment, activation and then positioning of the STAT to nucleus, so as to cause the adjusting of gene expression.
Ratified Lu's rope for Buddhist nun's phosphate be used to treat in or high risk myelofibrosis patient, including primary bone Myelofibrosis after myelofibrosis and primary thrombocytosis after marrow fibrosis, polycythemia vera.Lu Suo is replaced Buddhist nun's phosphate it is existing also in clinical test for treating primary thrombocytosis, cancer of pancreas, prostate cancer, breast cancer, white Blood disease, non-Hodgkin lymphoma, Huppert's disease and psoriasis.
In human body by the hydroxylating on the position cycloalkyl moieties 2-, the hydroxylating on the position cycloalkyl moieties 3- and by ring The further oxidation ketone generated on the position cyclopentyl moiety 3- and the three kinds of metabolins generated be identified as it is active (referring to Shilling, A.D. etc., Drug Metabolism and Disposition, 2010,38 (11): 2023-2031;FDA regulation Information and US20080312258).
It for the relevant most common hematology adverse reaction of Buddhist nun's administration is decrease of platelet and anaemia to Lu's rope.It is most common Non-blood adverse reaction is bruise, dizziness and headache.
Although Lu's rope has beneficial activity for Buddhist nun, but exists to the new compound for treating above-mentioned disease and situation Be continuously needed.
Summary of the invention
The present invention relates to pyrrolo- [2,3-d] pyrimidines and its pharmaceutically acceptable salt that novel heteroaryl replaces. The present invention also provides the composition containing the compounds of this invention and the composition in treatment by applying to hypotype 1 and 2 (JAK1/JAK2) selectively Janus associated kinase inhibitor and by the purposes in the disease and situation of favourable therapeutic.
Detailed description of the invention
Attached drawing shows the result of the metabolic stability test of signified compound.
Detailed description of the invention
Definition
Term " treatment " refers to reduction, inhibition, mitigation, elimination, containment or stable disease (such as disease outlined herein Or disorder) development or progress, mitigate the severity of disease or improve related with disease symptom.
" disease " refers to damage or interferes any illness or disorder of the normal function of cell, tissue or organ.
It should be understood that depending on the source of chemical material used in synthesis, exist in the compound of synthesis natural same Some variations of the plain abundance in position.Therefore, Lu's rope will be inherently comprising the isotopic body of a small amount of deuterate for the product of Buddhist nun (isotopologue).Despite the presence of this variation, but compared with the degree that the stable isotope of the compounds of this invention replaces, The stabilization hydrogen of this natural abundance and the concentration of carbon isotope are still very low and inessential.See, for example, Wada, E etc., Seikagaku,1994,66:15;The such as Gannes, LZ, Comp Biochem Physiol Mol Integr Physiol, 1998,119:725。
In the compound of the present invention, any atom for not being specially designated as specific isotope means to indicate the atom Any stable isotope.Unless otherwise indicated, when a position is designated specifically as " H " or " hydrogen ", which should be managed Solution is with the hydrogen according to its natural abundance isotopics.Equally, unless otherwise indicated, when a position is designated specifically When for " D " or " deuterium ", which is interpreted as the abundance at least 3000 times of the natural abundance (it is 0.015%) greater than deuterium Deuterium (that is, at least 45% deuterium mixes).
Terms used herein " Isotope enrichment factor " refer to specific isotope isotope abundance and natural abundance it Between ratio.
In other embodiments, the compound of the present invention is for the Isotope enrichment factor of each specified D-atom At least 3500(at each specified D-atom 52.5% deuterium incorporation), the incorporation of the deuterium of at least 4000(60%), at least 4500 The deuterium incorporation of (67.5% deuterium incorporation), at least 5000(75%), the deuterium incorporation of at least 5500(82.5%), at least 6000(90% Deuterium incorporation), the incorporation of the deuterium of at least 6333.3(95%), the deuterium incorporation of at least 6466.7(97%), the deuterium incorporation of at least 6600(99%) Or the deuterium incorporation of at least 6633.3(99.5%).
Term " isotopic body " refers to wherein chemical structure and specific compound of the invention only in its isotopics side The different substance in face.
Term " compound ", when being related to the compound of the present invention, referring to may in addition in the composed atom of molecule There is the set of the molecule with identical chemical structure except isotope variation.Therefore those skilled in the art it is clear that By the compound represented by containing the particular chemical of D-atom indicated also include less amount of one in the structure or With the isotopic body of hydrogen atom at multiple specified deuterium positions.The relative quantity of such isotopic body will in the compounds of this invention Depending on many factors, including being used to manufacture the isotopic purity of the deuteration agents of the compound and being used to prepare describedization Close the doping efficiency of deuterium in each synthesis step of object.However, as described above, the overall relative quantity of such isotopic body will Lower than the 49.9% of compound.In other embodiments, the overall relative quantity of such isotopic body will be less than compound 47.5%, it is lower than 40%, is lower than 32.5%, be lower than 25%, be lower than 17.5%, be lower than 10%, be lower than 5%, is lower than 3%, lower than 1% or is lower than 0.5%。
The present invention also provides the salt of the compounds of this invention.Alkalinity of the salt of the compounds of this invention in acid and the compound It is formed between group (such as amido functional group) or between alkali and the acidic-group (such as carboxyl functional group) of the compound.Root According to another embodiment, the compound is pharmaceutically acceptable acid-addition salts.
Terms used herein " pharmaceutically acceptable " refer within the scope of reasonable medical judgment, are suitable for and people Contact without excessive toxicity, stimulation, allergy etc. with the tissue of other mammals, and with reasonable interests/wind The component that danger ratio matches." pharmaceutically acceptable salt ", which refers to, can directly or indirectly provide the present invention when being applied to recipient Any non-toxic salt of compound." pharmaceutically acceptable counter ion " is that Shi Weifei is discharged from salt being applied to recipient The ionic portions of the salt of toxicity.
The acid for being commonly used to form pharmaceutically acceptable salt includes inorganic acid such as hydrogen sulfide, hydrochloric acid, hydrobromic acid, hydrogen iodine Acid, sulfuric acid and phosphoric acid and organic acid for example p-methyl benzenesulfonic acid, salicylic acid, tartaric acid, liquor epinephrinae bitartratis ophthalmicus, ascorbic acid, maleic acid, Benzene sulfonic acid, fumaric acid, gluconic acid, glucuronic acid, formic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzene sulfonic acid, lactic acid, oxalic acid, To bromo-benzene sulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid and acetic acid and relevant inorganic acid and organic acid.Such medicine On therefore acceptable salt includes sulfate, pyrosulfate, disulfate, sulphite, bisulfites, phosphate, phosphorus Sour monohydric salt, dihydric phosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, capric acid Salt, caprylate, acrylates, formates, isobutyrate, caprate, enanthate, propiolate, oxalates, malonate, amber Amber hydrochlorate, suberate, sebacate, fumarate, maleate, butine -1,4- diacid salt, alkynes -1,6- diacid salt, benzene Formates, chloro-benzoate, methyl benzoic acid salt, dinitro-benzoate, hydroxy benzoate, methoxy benzoic acid salt, Phthalate, terephthalate, sulfonate, xylenesulfonate, phenyl acetate salt, phenylpropionic acid salt, phenylbutyric acid Salt, citrate, lactate, beta-hydroxy-butanoic acid salt, oxyacetate, maleate, tartrate, mesylate, propane sulfonic acid salt, Naphthalene -1- sulfonate, naphthalene-2-sulfonic acid salt, mandelate and other salt.In one embodiment, pharmaceutically acceptable acid adds Include the acid-addition salts formed with inorganic acid such as hydrochloric acid and hydrobromic acid at salt, is especially formed with organic acids such as maleic acid Acid-addition salts.
The compound of the present invention (such as compound of Formulas I or formula A) may be contained due to such as deuterium substitution or other reasons Asymmetric carbon atom.Therefore, the compound of the present invention can be used as the mixing of single enantiomter or two enantiomters Object exists.Therefore, the compound of the present invention can be used as racemic mixture or non-racemic mixture (scalemic Mixture) exist, or exist as the corresponding single stereoisomer for being substantially free of another possible stereoisomer. Terms used herein " being substantially free of other stereoisomers " refer to other stereoisomers, preferably for having less than 25% Other stereoisomers, more preferably less than 5% other stereoisomers and most preferably in less than 2% other solids less than 10% Isomers.The method of the single enantiomter of the given compound of acquisition or synthesis known in the art, and can be according to practical feelings Condition is applied to final compound or raw material or intermediate.
Unless otherwise indicated, when disclosed compound is named by the structure of not specified spatial chemistry or is described and has one When a or multiple chiral centers, it is understood to represent all possible stereoisomer of the compound.
Terms used herein " mammal " include people or non-human animal, as mouse, rat, cavy, dog, cat, horse, Ox, pig, monkey, chimpanzee, baboon or macaque.In one embodiment, the mammal is non-human animal.In another reality It applies in scheme, the mammal is people.
Terms used herein " stable compound " are referred to the stability and holding for being enough that them is allowed to manufacture The integrality of the compound sufficient time in order to be described in detail in this article purpose (for example, be configured to treatment product, supply For producing the disease of intermediate, separable or storable midbody compound, treatment in response to therapeutic agent of therapeutic compounds Disease or illness) compound.
" D " and " d " is all referring to deuterium." stereoisomer " refers to enantiomter and diastereoisomer." Tert " and " t- " all refers to " uncle "." US " refers to the U.S..
" being replaced with deuterium " refers to substituting one or more hydrogen atoms with the D-atom of respective numbers.
In entire this specification, variable, which can be, to be referred to (for example, " each R ") or can be and refer in particular to (such as R1、R2、R3 Deng).Unless otherwise specified, when variable refers to, it mean include the specific variable all specific concrete forms.
Therapeutic compound
A kind of embodiment of the invention provides the compound of formula A:
Or its pharmaceutically acceptable salt, in which:
Y1Selected from hydrogen and deuterium;
Each Y2Independently selected from hydrogen and deuterium, condition is attached to each Y on common carbon2It is identical;
Each Y3Independently selected from hydrogen and deuterium, condition is attached to each Y on common carbon3It is identical;
Y4Selected from hydrogen and deuterium;
Each Y5It is identical and selected from hydrogen and deuterium;And
Y6、Y7、Y8、Y9And Y10It is each independently selected from hydrogen and deuterium;
Condition is to work as Y1It is hydrogen, each Y2With each Y3It is hydrogen, Y4It is hydrogen and Y6、Y7、Y8、Y9And Y10Individually hydrogen when, respectively A Y5It is deuterium.
In an embodiment of formula A, each Y2It is identical, each Y3It is identical, and each Y5It is identical 's.In the one aspect of the embodiment, each Y2It is deuterium.Further, each Y3It is deuterium.Another further Aspect, each Y3It is hydrogen.In the another aspect of the embodiment, each Y2It is hydrogen.Further, each Y3It is deuterium.? Another further aspect, each Y3It is hydrogen.In any foregoing aspects of one embodiment, Y1It is deuterium.In any aforementioned aspects Another embodiment in, Y1It is hydrogen.In any foregoing aspects of embodiment particularly, Y1It is deuterium, Y4It is deuterium, and each Y5It is deuterium.In any foregoing aspects of another embodiment particularly, Y1It is deuterium, Y4It is deuterium, and each Y5It is hydrogen.It is in office In one foregoing aspects of another embodiment particularly, Y1It is deuterium, Y4It is hydrogen, and each Y5It is hydrogen.Any foregoing aspects of In another embodiment particularly, Y1It is hydrogen, Y4It is hydrogen, and each Y5It is hydrogen.It is any it is foregoing aspects of it is another particularly In embodiment, Y1It is hydrogen, Y4It is hydrogen, and each Y5It is deuterium.In any foregoing aspects of another embodiment particularly, Y1 It is hydrogen, Y4It is deuterium, and each Y5It is deuterium.In any foregoing aspects of another embodiment particularly, Y1It is hydrogen, Y4It is deuterium, And each Y5It is hydrogen.
In one embodiment, Y6It is deuterium.In the one aspect of the embodiment, Y7And Y8Individually deuterium.In the implementation The another aspect of scheme, Y7And Y8Individually hydrogen.
In one embodiment, Y6It is hydrogen.In the one aspect of the embodiment, Y7And Y8Individually deuterium.In the implementation The another aspect of scheme, Y7And Y8Individually hydrogen.
A kind of embodiment of the invention provides a kind of compound of Formulas I:
Or its pharmaceutically acceptable salt, in which:
Y1Selected from hydrogen and deuterium;
Each Y2Independently selected from hydrogen and deuterium, condition is attached to each Y on common carbon2It is identical;
Each Y3Independently selected from hydrogen and deuterium, condition is attached to each Y on common carbon3It is identical;
Y4Selected from hydrogen and deuterium;
Each Y5It is identical and selected from hydrogen and deuterium;And
Y6、Y7And Y8It is each independently selected from hydrogen and deuterium;Condition is to work as Y1It is hydrogen, each Y2With each Y3It is hydrogen, Y4It is hydrogen And Y6、Y7And Y8Individually hydrogen when, each Y5It is deuterium.
In one embodiment, each Y2It is identical, each Y3It is identical, and each Y5It is identical.At this The one aspect of embodiment, each Y2It is deuterium.Further, each Y3It is deuterium.In another further aspect, respectively A Y3It is hydrogen.In the another aspect of the embodiment, each Y2It is hydrogen.Further, each Y3It is deuterium.It is another into The aspect of one step, each Y3It is hydrogen.In any foregoing aspects of one embodiment, Y1It is deuterium.Any foregoing aspects of another In embodiment, Y1It is hydrogen.In any foregoing aspects of embodiment particularly, Y1It is deuterium, Y4It is deuterium, and each Y5It is deuterium. In any foregoing aspects of another embodiment particularly, Y1It is deuterium, Y4It is deuterium, and each Y5It is hydrogen.In any aforementioned side In another embodiment particularly in face, Y1It is deuterium, Y4It is hydrogen, and each Y5It is hydrogen.Any foregoing aspects of another more special In fixed embodiment, Y1It is hydrogen, Y4It is hydrogen, and each Y5It is hydrogen.In any foregoing aspects of another embodiment particularly In, Y1It is hydrogen, Y4It is hydrogen, and each Y5It is deuterium.In any foregoing aspects of another embodiment particularly, Y1It is hydrogen, Y4 It is deuterium, and each Y5It is deuterium.In any foregoing aspects of another embodiment particularly, Y1It is hydrogen, Y4It is deuterium, and each A Y5It is hydrogen.
In one embodiment, Y6It is deuterium.In the one aspect of the embodiment, Y7And Y8Individually deuterium.In the implementation The another aspect of scheme, Y7And Y8Individually hydrogen.
In one embodiment, Y6It is hydrogen.In the one side of the embodiment, Y7And Y8Individually deuterium.In the embodiment party The another aspect of case, Y7And Y8Individually hydrogen.
In one embodiment, which is the compound of Formulas I, wherein Y6、Y7And Y8Individually hydrogen, and the change Close object be selected from table 1(it is as follows) listed by any compound (Cmpd):
Table 1: the exemplary implementation scheme of Formulas I
Compound Y1 Each Y2 Each Y3 Y4 Each Y5
100 H H H D H
101 H H H H D
102 H H H D D
103 H H D H H
104 H H D D H
105 H H D H D
106 H H D D D
107 H D H H H
108 H D H D H
109 H D H H D
110 H D H D D
111 H D D H H
112 H D D D H
113 H D D H D
114 H D D D D
115 D H H H H
116 D H H D H
117 D H H H D
118 D H H D D
119 D H D H H
120 D H D D H
121 D H D H D
122 D H D D D
123 D D H H H
124 D D H D H
125 D D H H D
126 D D H D D
127 D D D H H
128 D D D D H
129 D D D H D
130 D D D D D
Or its pharmaceutically acceptable salt, any of them are not designated as the atom of deuterium and are deposited with its natural isotopic abundance ?.
In one embodiment, which is the compound of Formulas I, wherein Y6、Y7And Y8Individually deuterium, and the change Close object be selected from table 2(it is as follows) listed by any compound (Cmpd):
Table 2: the exemplary embodiment of Formulas I
Compound Y1 Each Y2 Each Y3 Y4 Each Y5
200 H H H D H
201 H H H H D
202 H H H D D
203 H H D H H
204 H H D D H
205 H H D H D
206 H H D D D
207 H D H H H
208 H D H D H
209 H D H H D
210 H D H D D
211 H D D H H
212 H D D D H
213 H D D H D
214 H D D D D
215 D H H H H
216 D H H D H
217 D H H H D
218 D H H D D
219 D H D H H
220 D H D D H
221 D H D H D
222 D H D D D
223 D D H H H
224 D D H D H
225 D D H H D
226 D D H D D
227 D D D H H
228 D D D D H
229 D D D H D
230 D D D D D
231 H H H H H
Or its pharmaceutically acceptable salt, any of them are not designated as the atom of deuterium and are deposited with its natural isotopic abundance ?.
In another set of embodiments, any atom for not being designated as deuterium is natural with it in any of the above-described embodiment Isotope abundance exists.
Following compounds can be used for preparing various compounds of the invention:
Or its salt, any of them are not designated as the atom of deuterium with the presence of its natural isotopic abundance.
The synthesis of the compound of Formulas I or formula A easily can synthesize chemist with reference to herein by common skill Disclosed exemplary synthesis is realized with embodiment.To similar related of the method for being used to preparation of compounds of formula I and its intermediate Method is disclosed in, such as United States Patent (USP) 7,598,257 and Organic Letters, 2009,11 (9): in 1999-2009.
Can use corresponding deuterate and optional other reagents and/or intermediate containing isotope retouched herein with synthesizing The compound stated or quote known in the art is held for isotope atom to be introduced to the standard synthetic protocols of chemical structure The such method of row.
Exemplary synthesis
The compound of Formulas I or formula A can by be similar to United States Patent (USP) 7,598,257 and Organic Letters, 2009,11 (9): those modes synthesized shown in 1999-2009 are prepared using suitable deuterate raw material.
The compound of Formulas I or formula A can also be prepared by scheme as follows.
Scheme 1: the preparation of the compound of Formulas I
Scheme 1 discloses the exemplary preparation of the compound of Formulas I, wherein Y1, each Y2With each Y3It is deuterium, and Y4, it is each A Y5、Y6、Y7And Y8It is hydrogen.In a manner of being similar to described in the WO2010/083283, the commercially available chloro- 7H- pyrrolo- of 4- [2, 3-d] pyrimidine 11(Aldrich) hydrogenated sodium and chlorination SEM handle to obtain 12, react to obtain 14 with commercially available 13.Instead of 11, Bromo- 7H- pyrrolo- [2,3-d] pyrimidine of 4- can also be used in the first step with provide SEM protection the bromo- 7H- pyrrolo- of 4- [2, 3-d] pyrimidine (being similar to 12), it can react with 13 to obtain 14.14 with such as the preparation disclosed in following scheme 2a 15 reaction is by with Lin, and similar mode described in the Org.Lett.2009,11,1999 such as Q. is carried out to obtain 16.The reaction carries out in the presence of such as Lin, the chiral ligand 27 of the preparation of the descriptions such as Q.16 by using NH4OH and I2Processing and It is converted into 17.17 SEM blocking group then uses LiBF4And NH4OH deprotects and provides the compound of Formulas I.
Scheme 2a: the preparation of compound 15
As shown in scheme 2a, commercially available 18 Yong Phosphonium ylide 20 and DCl/D2O processing is to generate 19, with 20 and DiBAl- H processing is to obtain 15.
Scheme 2b: the preparation of compound 23
Scheme 2c: the preparation of compound 26
Similar 15 compound can also be prepared.For example, as shown in scheme 2b, commercially available 21 can by in scheme 2a Disclosed similar fashion is converted into 23.For another example, as shown in scheme 2c, commercially available 24 can by with it is public in scheme 2a and scheme 2b The similar fashion opened is converted into 26.23 can by being converted into the compound of Formulas I with similar fashion disclosed in scheme 1, wherein Y1With each Y3It is deuterium, and Y4, each Y2, each Y5、Y6、Y7And Y8It is hydrogen.Similarly, 26 can by with disclosed in scheme 1 Similar fashion be converted into the compound of Formulas I, wherein Y1With each Y2It is deuterium, and Y4, each Y3, each Y5、Y6、Y7And Y8It is Hydrogen.
Particular approach and compound as shown above is not intended to restrictive.Regardless of pass through identical variable Name is (that is, R1、R2、R3Deng) be identified, the chemical structure in diagram is depicted with corresponding position in compounds herein formula herein Chemical group defines the variable that (part, atom etc.) is suitably limited herein.In the chemical combination for synthesizing another compound The adaptability of chemical group is within the knowledge of those of ordinary skill in the art in object structure.
The compound and its synthesis precursor of synthesis Formulas I or formula A are (including herein in diagram in the path that is not explicitly shown Those of) other methods within the scope of the means of the chemist of this field common skill.It can be used for synthesizing applicable The synthesis chemical conversion of compound and protection based method (protection and deprotection) are known in the art, and including for example, description Those of in the following documents method: Larock R, Comprehensive Organic Transformations, VCH Publishers(1989);Greene, TW etc., Protective Groups in Organic Synthesis, 3rd Ed., John Wiley and Sons(1999);Fieser, L etc., Fieser and Fieser ' s Reagents for Organic Synthesis,John Wiley and Sons(1994);And Paquette, L write, Encyclopedia of Reagents For Organic Synthesis, John Wiley and Sons (1995) and its later release.
It is contemplated by the invention that substituent group and variable combination only those result in the combination of stable compound.
Composition
The present invention also provides pyrogen-free pharmaceutical compositions, compound comprising a effective amount of Formulas I or formula A (for example including Formula described herein it is any) or the compound pharmaceutically acceptable salt and pharmaceutically acceptable carrier.It is described Carrier is compatible with the other compositions of preparation and pharmaceutically in the case where acceptable carrier, for the amount in drug Under will not it is harmful to its recipient in the sense that be " acceptable ".
Pharmaceutically acceptable carrier, adjuvant and the medium of pharmaceutical composition for use in the present invention include still unlimited In: ion-exchanger, aluminium oxide, aluminum stearate, lecithin, haemocyanin such as human serum albumins, buffer substance such as phosphorus Hydrochlorate, glycine, sorbic acid, potassium sorbate, the partial glyceride mixture of saturated vegetable fatty acid, water, salt or electrolyte are for example Protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salt, colloidal silica, magnesium trisilicate, polyvinylpyrrolidine Alkanone, cellulose base materials, polyethylene glycol, sodium carboxymethylcellulose, polyacrylate, wax, polyethylene-polyoxypropylene block are total Polymers, polyethylene glycol and lanolin.
If necessary, the compounds of this invention in pharmaceutical composition can be improved by method well-known in the art Solubility and bioavilability.A kind of method includes using lipid excipients in the formulation.Referring to " Oral Lipid-Based Formulations:Enhancing the Bioavailability of Poorly Water-Soluble Drugs (Drugs and the Pharmaceutical Sciences) " David J.Hauss chief editor, Informa Healthcare, 2007;" Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery:Basic Principles and Biological Examples " Kishor M.Wasan chief editor, Wiley- Interscience,2006。
Improve bioavilability another known method be using optionally with poloxamer (such as LUTROLTM and PLURONICTM (BASF Corporation)) or the amorphous shape prepared together of ethylene oxide and oxypropylene block copolymer The compounds of this invention of formula.Referring to United States Patent (USP) 7,014,866 and U.S. Patent Publication 20060094744 and 20060079502.
Pharmaceutical composition of the invention include be suitable for taking orally, rectum, nose, part (including cheek and sublingual), vagina or stomach Those of parenteral (including subcutaneous, intramuscular, intravenous and intradermal) application pharmaceutical composition.In certain embodiments, herein The compound transdermal administration (for example, using percutaneous plaster or iontophoretic technology) of structural formula.Other preparations mention in which can be convenient For for unit dosage form, such as tablet, sustained releasing type capsule and in liposome, and can be appointed by well known in pharmaceutical field Where prepared by method.See, e.g., Remington:The Science and Practice of Pharmacy, Lippincott Williams&Wilkins, Baltimore, MD (the 20th edition, 2000).
Such preparation method include will prepare the molecule of application and constitute one or more auxiliary elements ingredient (such as Carrier) the step of being combined.In general, by by active constituent and liquid-carrier, liposome or the solid carrier of subdivision or two Person is uniform closely to be combined, and then if necessary, prepares composition to product forming.
In some embodiments, the compound is administered orally.The orally administered present composition can be with Discrete the unit such as respectively capsule containing predetermined amounts of active ingredients, sachet or tablet;Powder or particle;Waterborne liquid is non- Solution or suspension in waterborne liquid;Oil-in-water liq emulsion;Water-in-oil liquid emulsion;Filled liposome;Or as big Pill etc. exists.Perle can be used for advantageously improving compound absorptivity comprising such suspension.
In the tablet being administered orally, commonly utilized carrier includes lactose and cornstarch.Lubricant is for example hard Fatty acid magnesium is also usually to add.For being administered with capsules per os, useful diluent includes lactose and dry corn Starch.When aqueous suspension is administered orally, active constituent is combined with emulsifier and suspending agent.If desired, can add certain Sweetener and/or flavoring agent and/or colorant.
The composition for being suitable for being administered orally is included in flavoured base (usually sucrose and Arabic gum or bassora gum) Dragee comprising the ingredient and in inert base (such as gelatin and glycerol or sucrose and Arabic gum) comprising activity The pastille of ingredient.
Parenterally administrated composition includes aqueous and non-aqueous aseptic parenteral solution, be may include anti-oxidant Agent, buffer, bacteriostatic agent and the solute for making the blood of preparation and intended recipient isotonic;And aqueous and non-aqueous nothing Bacteria suspension may include suspending agent and thickener.The preparation can be provided in unit dose or multi-dose container, such as The ampoule and bottle of sealing, and can be stored under the conditions of freeze-drying (freeze-drying), it is only necessary to nothing will be being added before use The liquid-carrier of bacterium, such as water for injection.Injection solution and suspension immediately can be prepared from sterile powder, particle and tablet.
Such injection solution can be sterile aqueous or oily injection suspension form.This suspension can be according to this Technology known to field is prepared using dispersion appropriate or wetting agent (such as Tween 80) and suspending agent.Aseptic injection preparation The aseptic injectable solution or suspension being also possible in parenteral acceptable non-toxic diluent or solvent, such as 1, Solution in 3- butanediol.The acceptable medium and solvent that can be used include mannitol, water, Ringer's solution and isotonic chlorine Change sodium solution.In addition, sterile fixed oil is conventionally used as solvent or suspension media.In this regard, can be used any mild Fixed oil, glyceryl monoacetate or diglyceride including synthesis.Fatty acid, such as oleic acid and its glyceride ester derivatives, It can be used for preparing injection, and natural pharmaceutically acceptable oil, such as olive oil or castor oil, especially theirs is poly- Oxygen ethylization form.These oil solutions or suspension also may include long-chain alcohol diluents or dispersing agent.
Pharmaceutical composition of the invention can be applied using the suppository form for rectally.These compositions can pass through The compound of the present invention is mixed with nonirritant excipient appropriate to prepare, the excipient be at room temperature solid still It is liquid under rectal temperature, therefore is melted in the rectum to release active component.Such material includes, but are not limited to Cocoa butter, beeswax and polyethylene glycol.
Pharmaceutical composition of the invention can be administered by nose aerosol or sucking.Such composition is according to medicine Prepared by technology well known to object preparation technique field, and benzyl alcohol or other suitable preservatives can be used, improve biology benefit Other known solubilisings of sorbefacient, fluorohydrocarbon and/or technical field of expenditure or dispersing agent are prepared into the solution of salt water.Ginseng See, for example, Rabinowitz JD and Zaffaroni AC, United States Patent (USP) 6,803,031, are assigned to Alexza Molecular Delivery Corporation。
When required treatment is related to the readily accessible region of topical application or organ, the part of pharmaceutical composition of the invention Application is particularly useful.For the topical application of local skin, pharmaceutical composition should be suspended or dissolved in carrier with containing In the suitable ointment of active component prepare.Carrier for local application the compounds of this invention includes but is not limited to: Mineral oil, Albolene, albolene, propylene glycol, PULLRONIC F68 compound, emulsifying wax and water.Alternatively, institute Stating pharmaceutical composition can be prepared with the washing lotion containing the reactive compound being suspended or dissolved in carrier or creams.Suitable load Body includes but is not limited to: mineral oil, monostearate anhydrosorbitol, polysorbate 60, cetyl ester wax, cetearyl alcohol Alcohol, 2- octyldodecanol, benzyl alcohol and water.Pharmaceutical composition of the invention can also be by rectal suppository formulation or to be suitble to Enema agent be applied topically to lower intestine.The invention also includes topical transdermal patches and iontophoresis to apply.
This therapeutic agent application can be it is local, so as to purpose site apply.Various technologies can be used in purpose Site provides this composition, such as injects, uses conduit, trochar, projectile, pluronic gel, bracket, Sustained drug Release polymers are provided for reaching other internal devices.
Therefore, according to yet another embodiment, the compounds of this invention can be added into composition for coating and can plant The medical device entered, such as prosthese, artificial valve, blood vessel graft, bracket or conduit.Implantable device through coating it is appropriate Coating and to be typically prepared be known in the art and in United States Patent (USP) 6,099,562;It is illustrated in 5,886,026 and 5,304,121 Explanation.Coating is usually biocompatible polymeric material, for example, aquogel polymer, poly- tetramethyldisiloxane, polycaprolactone, Polyethylene glycol, polylactic acid, ethane-acetic acid ethyenyl ester and its mixture.The coating can be optionally further by fluorine silicon oxygen appropriate The external coating of alkane, polysaccharide, polyethylene glycol, phosphatide or combinations thereof covers, to assign composition control release characteristics.For invading Property device coating be included in the definition of pharmaceutically acceptable carrier, adjuvant or medium, it is as used herein that A little terms.
According to another embodiment, the present invention provides the method for coating implantable medical device, including makes described device The step of being contacted with coating composition described above.Those skilled in the art are it is evident that the implantation that is coated in of device is fed Occur before in newborn animal.
According to another embodiment, the present invention provides the method for impregnating implantable drug release device, including makes described The step of drug release device and the compound of the present invention or composition contact.Implantable drug release device includes but is not limited to Biodegradable polymer capsule or bullet, nondegradable diffusible polymer capsule and biodegradable polymer Thin slice.
According to another embodiment, present invention offer is coated with the compounds of this invention or the combination containing the compounds of this invention The implantable medical device of object, so that the compound has therapeutic activity.
According to another embodiment, the present invention provides dipping or contains the compound of the present invention or include chemical combination of the present invention The implantable drug release device of the composition of object, so that the compound discharges from described device and is therapeutic activity 's.
When being close to when organ or tissue because of removing from patient, this organ or tissue, which can be bathed in, contains this hair In the medium of bright composition, composition of the invention can be coated on organ or the present composition can be with any other side Just mode applies.
In another embodiment, composition of the invention further includes second therapeutic agent.The second therapeutic agent Can have or show when being applied together with having and Lu's rope is for the compound of the identical mechanism of action of Buddhist nun selected from any known The compound or therapeutic agent of favorable property.These reagents include being shown as those of to be used in combination with Lu's rope for Buddhist nun.
Preferably, the second therapeutic agent is the reagent that can be used to treat or prevent disease or situation selected from the following: bone Marrow fibrosis (including myelofibrosis after primary myelofibrosis, polycythemia vera, polycythemia vera, Myelofibrosis and primary thrombocytosis after chronic idiopathic myelofibrosis, primary thrombocytosis), pancreas Gland cancer, prostate cancer, breast cancer, leukaemia, non-Hodgkin lymphoma, Huppert's disease, psoriasis and alopecia areata.
In one embodiment, the second therapeutic agent is selected from lenalidomide, pabishta, capecitabine, Yi Ximei It is smooth and combinations thereof.
In another embodiment, the present invention provides the compounds of this invention and one or more above-mentioned any second treatments The independent dosage form of agent, wherein the compound is associated with each other with second therapeutic agent.Term as used herein " is associated with each other " Refer to that independent dosage form is packed together or is connected with each other to be readily appreciated that the independent dosage form is intended to together in another way Sale and application (be separated by within less than 24 hours and sequentially apply or simultaneously apply).
In pharmaceutical composition of the present invention, the compounds of this invention exists with effective quantity.As it is used herein, term " has Effect amount " refers to and is enough to treat when with the application of correct dosage regimen the amount of (therapeutic or prophylactically) target conditions.
Correlation for animal and the dosage (being based on every square metre of body surface area of milligram) of people is described in In Freireich etc., (1966) Cancer Chemother.Rep50:219.Body surface area can be close from the height and weight of patient As determine.See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardsley, N.Y., 1970, 537。
In one embodiment, the effective dose scope of the compounds of this invention can be from 1mg to 500mg, such as 5mg is arrived 100mg, such as 5mg to 50mg.The example of range is from 40mg to 50mg, from 25mg to 40mg, from 25mg to 50mg, from 20mg To 40mg, from 20mg to 50mg, from 10mg to 25mg, from 10mg to 20mg, from 5mg to 25mg, from 5mg to 20mg, Yi Jicong 5mg to 10mg.In one embodiment, the dosage of 10mg, 20mg, 40mg and 50mg are administered once a day.Implement at one In scheme, the dosage of 5mg, 10mg, 20mg, 40mg and 50mg are applied twice daily.
As those skilled in the art recognize, effective dose can also change, this depends on treated disease, disease At severity, the approach of administration, the gender of subject, age and general health, the use of excipient and other treatment Reason method shares a possibility that as used other reagents and the judgement of attending physician.For example, the guidance of selection effective dose can The provisioning information of Buddhist nun is replaced to determine by reference to Lu's rope.
For the pharmaceutical composition comprising second therapeutic agent, the effective quantity of second therapeutic agent is commonly used in the medicine is used only About 20% to the 100% of the dosage of the monotherapy of agent.Preferably, effective quantity is about 70% to the 100% of normal monotherapy dosage. The normal monotherapy dosage of these second therapeutic agents is well known to those skilled in the art, and is write for example, see Wells etc., Pharmacotherapy Handbook, second edition, Appleton and Lange, Stamford, Conn. (2000);PDR Pharmacopoeia,Tarascon Pocket Pharmacopoeia2000,Deluxe Edition,Tarascon Publishing, Loma Linda, Calif. (2000), the bibliography are respectively incorporated herein in full by reference.
It is expected that above-mentioned some second therapeutic agents will be cooperateed with the compounds of this invention and be played a role.When such case is sent out When raw, the effective dose of second therapeutic agent and/or the compounds of this invention will be allowed to reduce relative to dosage needed for monotherapy. This has the application for making that second therapeutic agent or the toxic side effects of the compounds of this invention minimize, the collaboration of effect improves, improves Or ease of use and/or compound preparation or preparation whole cost reduce the advantages of.
Treatment method
In another embodiment, the present invention provides Janus associated kinase (JAK) JAK1 in a kind of inhibition cell and The method of one or more of JAK2, compound or its pharmacy including making cell and one or more Formulas I herein or formula A Upper acceptable salt is in contact.
According to another embodiment, the present invention provides treat valuably to be controlled with Lu's rope for Buddhist nun in the subject of needs The method of the disease for the treatment of includes the steps that applying a effective amount of the compound of the present invention or composition to the subject.At one In embodiment, which is the patient for needing this treatment.This kind of disease is well known in the art, and it is following (but not It is limited to) it discloses in patent: United States Patent (USP) 7,598,257.This kind of disease include but is not limited to be related to the disease of immune system, including Such as organ transplant rejection (such as allograft rejection and graft versus host disease(GVH disease));Autoimmune disease is such as more Hair property sclerosis, rheumatoid arthritis, juvenile arthritis, type-1 diabetes mellitus, lupus, psoriasis, inflammatory bowel disease, ulcer Property colitis, Crohn disease, myasthenia gravis, immunoglobulin nephrosis, autoimmune thyroid disease;Allergic conditions are such as Asthma, food hypersenstivity, allergic dermatitis and rhinitis;Viral disease such as epstein-Barr virus (EBV), hepatitis B, third Type hepatitis, HIV, HTLV1, varicella virus (VZV) and human papilloma virus (HPV);Skin disease such as psoriasis (such as psoriasis vulgaris), allergic dermatitis, fash, skin irritatin, sensitization of skin reaction (such as contact dermatitis or allergy Property contact dermatitis);Cancer, cancer (such as prostate cancer, kidney, liver cancer, pancreas including those characterized by entity tumor Cancer, gastric cancer, breast cancer, lung cancer, the cancer of incidence, thyroid cancer, glioblastoma, Kaposi's sarcoma, Frank Castleman Disease, melanoma), hematologic cancers (such as lymthoma, leukaemia such as acute lymphatic leukemia or Huppert's disease) and (example includes that Sai Zharui syndrome and gill fungus sample are true for cutaneum carcinoma such as skin T cell lymphoma (CTCL) and cutaneous B-cell lymphoma Bacterium disease;Myeloproliferative disease (MPD) such as polycythemia vera (PV), primary thrombocytosis (ET), marrow sample Raw myelofibrosis (MMM), chronic myelomonocytic leukaemia (CMML), hypereosinophilia syndrome (HES), it is System property mast cell disease (SMCD);Inflammation and inflammatory disease such as ocular inflammatory disease (such as iritis, uveitis, sclera Scorching, conjunctivitis or relevant disease), (such as the upper respiratory tract includes the inflammatory disease of nose, nasal sinus for the diseases associated with inflammation of respiratory tract Disease, if the diseases associated with inflammation of rhinitis or nasosinusitis or lower respiratory tract includes bronchitis, chronic obstructive pulmonary disease etc.), inflammatory flesh Disease such as myocarditis;Systemic inflammatory response syndrome (SIRS) and septic shock;Ischemical reperfusion injury or with inflammatory ischemic The relevant disease of event or situation such as apoplexy or sudden cardiac arrest;Apositia;Cachexia;Fatigue is as caused by cancer or and cancer Relevant fatigue;Restenosis;Sclerodermatitis (sclerodermitis);Fibrosis;With anoxic or proliferation of astrocytes Relevant situation, such as diabetic retinopathy, cancer or neurodegenerative disease;Gout;Due to for example benign forefront The forefront hylperadenosis of gland hypertrophy or benign prostatic hyperplasis.
In one particular embodiment, it is fine selected from marrow to be used to the treatment in the subject of needs to method of the invention Bone after myelofibrosis, primary thrombocytosis after dimensionization, including primary myelofibrosis, polycythemia vera Marrow fibrosis, primary thrombocytosis or combinations thereof;Cancer of pancreas;Prostate cancer;Breast cancer;Leukaemia;Non-Hodgkin's leaching Bar tumor;Huppert's disease;The disease or situation of psoriasis and combinations thereof.
In another specific embodiment, method of the invention is used to the treatment in the subject of needs and is selected from marrow Myelofibrosis and primary thrombocytosis after fibrosis, including primary myelofibrosis, polycythemia vera The disease or situation of myelofibrosis afterwards.
Needing the identification of the subject of this treatment can be carried out by the judgement of subject or health care professional, and It can be subjective (such as opinion) or (can objectively be measured by inspection or diagnostic method).
In another embodiment, any above treatment method includes further applying jointly to the subject of needs The step of with one or more second therapeutic agents.Second therapeutic agent can be used for applying jointly with Lu's rope for Buddhist nun selected from any known Second therapeutic agent.The selection of second therapeutic agent additionally depends on specified disease to be treated or situation.It can in the method for the present invention The example of the second therapeutic agent of use is the above-mentioned composition combined to contain the compound of the present invention and second therapeutic agent Those of in.
Particularly, conjoint therapy of the invention includes the compound and for the subjects of needs being co-administered Formulas I or formula A Two therapeutic agents treat conditions (specific second therapeutic agent is shown in the subsequent bracket of the indication): myelofibrosis (lenalidomide or pabishta);Cancer of pancreas (capecitabine) and breast cancer (Exemestane).
Term " co-administration " used in the present invention refers to that second therapeutic agent can be used as single formulation and (such as contain this hair The present composition of bright compound and second therapeutic agent described above) part applied together with the compounds of this invention, or It is applied together with the compounds of this invention as individual multi-form.Alternatively, other medicaments can before applying the compounds of this invention, Simultaneously or after apply.In the treatment of this conjoint therapy, the compounds of this invention and second therapeutic agent are all by conventional method Application.The present composition containing the compounds of this invention and second therapeutic agent is applied to patient to be not excluded for over the course for the treatment of Another time the identical therapeutic agent, any other second therapeutic agent or any compound of the invention is administered alone.
The effective quantity of these second therapeutic agents is known in those skilled in the art, and is administered and instructs to mention herein And patent and disclosed patent application and Wells etc. write, Pharmacotherapy Handbook, second edition, Appleton and Lange,Stamford,Conn.(2000);PDR Pharmacopoeia,Tarascon Pocket Pharmacopoeia2000,Deluxe Edition,Tarascon Publishing,Loma Linda,Calif.(2000) It is found in other medical articles.However, those skilled in the art are easily determined the best available amount range of second therapeutic agent.
In the embodiment of the present invention that wherein second therapeutic agent is applied to subject, the effective quantity of the compounds of this invention is small Its effective quantity in the case where not applying second therapeutic agent.In another embodiment, the effective quantity of second therapeutic agent is small Its effective quantity in the case where not applying the compounds of this invention.By this method, the high agent with any medicament can be minimized Measure relevant undesirable side effect.Other possible advantage (including but not limited to improved dosage regimen and/or reduced medicines Object cost) it will be apparent to those skilled in the art.
At yet another aspect, the present invention provides the compounds of Formulas I or formula A to be controlled individually or with one or more above-mentioned second It treats agent and is used to treat or prevent disease as described above, mistake in subject as single composition or independent dosage form in preparation together Purposes in tune or the drug of symptom.Another aspect of the present invention be the compound of Formulas I or formula A be used in subject treatment or Prevent disease, imbalance or symptom described herein.
Embodiment
Embodiment 1. (R) -3- (4- (7H- pyrrolo- [2,3-d] pyrimidine-4-yl) -1H- pyrazol-1-yl) -3- (2,2,5, 5-d4Cyclopenta) propionitrile (compound 107) synthesis.
The preparation of 3. compound 107 of scheme
Step 1.2,2,5,5-d4Pentamethylene -1,1- diethyl dicarboxylate (32).Diethyl malonate (6.57mL, The ethanol solution (32.3mL, 86.6mmol) of the sodium ethoxide of 21 weight % is added in ethyl alcohol (40mL) solution 43.3mmol), with Tetra- deuterium -1,4- dibromobutane of 1,1,4,4- (31,5.53mL, 45.5mmol, CDN Isotopes, 98 atom % deuteriums) are added afterwards.Institute It obtains solution to stir under reflux 2 hours, then cools to room temperature and diluted with excessive water.Most of second is then removed by distillation Alcohol, and obtained aqueous solution is extracted with ethyl acetate (3x75mL).Organic layer merges, and is washed with brine, dry (Na2SO4), filtering And be concentrated under reduced pressure to obtain the 32 of yellow oil, enter subsequent step (9.45g, 100%) without further purification.
Step 2.2,2,5,5-d4Pentamethylene -1- carboxylic acid (33).Ethyl alcohol (20mL) to 32 (9.45g, 43.3mmol) is molten The sodium hydroxide solution (20mL) of 5M is added in liquid.It then adds other water (15mL), stirring 3 is small under reflux for reaction When.After being cooled to room temperature, which dilutes and most of ethyl alcohol is removed by distillation.Make the aqueous solution with 1N hydrochloric acid Aobvious acidity (pH < 2), is then extracted with diethyl ether (3x50mL).Organic layer merges, dry (Na2SO4), it filters and is concentrated under reduced pressure. Gained light orange solid is transferred in pressure flask, and water (140mL) is added.The pressure flask is sealed, and is reacted at 160 DEG C Lower stirring 15 hours, then cools to room temperature.Reaction 1N hydrochloric acid is simultaneously extracted with diethyl ether (3x50mL).It is organic laminated And dry (Na2SO4), it filters and is concentrated under reduced pressure to obtain the 33 of amber oily (4.37g, 86%), make without further purification With.
Step 3.2,2,5,5-d4- N- methoxy-. N-methyl cyclopentane formamide (34).At 0 DEG C, to 33 (4.37g, N, hydrochloride (4.33g, 44.4mmol), the TBTU of O- dimethyl hydroxylamine are added in acetonitrile (60mL) solution 37.0mmol) (12.5g, 38.9mmol) and N, N- diisopropylethylamine (19.0mL, 111mmol).Reaction is stirred at room temperature 15 hours, so It is extracted afterwards with 1N hydrochloric acid and with ethyl acetate (3x50mL).Organic phase merges, with saturation NaHCO3Washing, it is dry (Na2SO4), it filters and is concentrated under reduced pressure.Products therefrom column chromatography (SiO2, 0-50% ethyl acetate/hexane) and it purifies to obtain 34 (2.22g, 37%) of bright oily.MS(ESI)162.3[(M+H)+]。
Step 4.2,2,5,5-d4Pentamethylene -1- formaldehyde (35).At 0 DEG C, to the THF of 34 (2.22g, 13.8mmol) The LiAlH of 1M is added dropwise in (50mL) solution4THF (24.8mL, 24.8mmol) solution.Reaction stirring 1 at 0 DEG C is small When, then it is quenched by the way that NaOH (940 μ L) and the water (2.82mL) of water (940 μ L), 15% is sequentially added dropwise.The reactant being quenched It is stirred at room temperature 30 minutes, then passes throughIt filters and is concentrated under reduced pressure.Gained grease is used in combination with 1N hydrochloric acid Diethyl ether (3x50mL) extraction.Organic layer merges, dry (Na2SO4), it filters and is concentrated under reduced pressure to obtain the 35 of transparent oil (850mg, 60%), uses without further purification.
Step 5.3- (2,2,5,5-d4Cyclopenta) acrylonitrile (36).At 0 DEG C, to the THF of the potassium tert-butoxide of 1M The THF (12mL) that cyanogen methylphosphonic acid diethylester (1.48mL, 9.15mmol) is added dropwise in (8.74mL, 8.74mmol) solution is molten Liquid.Reaction is heated to room temperature, is stirred 15 minutes, is cooled to 0 DEG C.Aldehyde 35 (850mg, 8.32mmol) is as THF's (3mL) Solution is added dropwise.Reaction is stirred at room temperature 48 hours, is then diluted with excessive water and uses diethyl ether (1x50mL) and second Acetoacetic ester (3x50mL) extraction.Organic layer merges, dry (Na2SO4), it filters and is concentrated under reduced pressure to obtain the 36 of light orange oily (1.17g, > 100%), uses without further purification.
Step 6. (+/-)-(4- (1- (2- cyano -1- (2,2,5,5-d4Cyclopenta) ethyl) -1H- pyrazoles -4- base) - 7H- pyrrolo- [2,3-d] pyrimidin-7-yl) methyl pivalate ((+/-) 38).To 37 (400mg, 1.34mmol, in Lin, Q. Prepared described in equal Org.Lett., 2009,11,1999-2002) acetonitrile (10mL) solution in be added 36 (418mg, 3.34mmol), DBU (421 μ L, 2.81mmol) then is added.Reaction is stirred at room temperature 15 hours, is then concentrated under reduced pressure.Institute Crude mixture is obtained to be diluted with water and ethyl acetate (3x50mL) is used to extract.Organic layer merges, dry with 1N salt acid elution (Na2SO4), it filters and is concentrated under reduced pressure.First use positive column chromatography (SiO2, 0-60% ethyl acetate/hexane) and purifying, then with reversed column Chromatography (C18, the 5-70% acetonitrile/water containing 0.1% formic acid) purifying obtains (+/-) 38 (68mg, 12%) of white foam.1H NMR(DMSO-d6,400MHz)8.84(s,1H),8.79(s,1H),8.40(s,1H),7.74(d,J=3.8Hz,1H),7.12 (d,J=3.8Hz,1H),6.24(s,2H),4.54(td,J=9.7,4.3Hz,1H),3.30-3.15(m,2H),2.39(d,J= 9.8Hz,1H),1.68–1.36(m,4H),1.08(s,9H);MS(ESI)425.3[(M+H)+].
Step 7. (R)-(4- (1- (2- cyano -1- (the deuterated cyclopenta of 2,2,5,5- tetra-) ethyl) -1H- pyrazoles -4- base) - 7H- pyrrolo- [2,3-d] pyrimidin-7-yl) methyl pivalate ((R) -38).Racemic compound (+/-) 38 (62mg) with The concentration of 30mg/mL is dissolved in acetonitrile, and with (+/-) 38 solution of 500 μ L of per injection in Daicel ChiralPak AD Chiral resolution: 30% isopropanol (+0.1% diethyl is carried out by preparative HPLC using isocratic method on column (20x250mm, 10 μm) Amine)/70% hexane (+0.1% diethylamine), flow velocity 17mL/min.In these cases, to elute (S) -38 at 15.0 minutes Baseline separation is realized with (R) -38 is eluted at 20.2 minutes.
Fraction containing each enantiomter merges and concentration, generates the colorless film and 29mg of (S) -38 of 28mg (R) -38 colorless film.
Step 8. (R) -3- (4- (7H- pyrrolo- [2,3-d] pyrimidine-4-yl) -1H- pyrazol-1-yl) -3- (2,2,5,5- Four deuterated cyclopenta) propionitrile (compound 107).Compound (R) -38 (28mg, 0.066mmol, 1 equivalent) is in the scintillation vial of 20mL In be dissolved in methanol (1mL).It is added sodium hydroxide (the 1M solution of 0.13mL, 0.13mmol, 2 equivalents), and reacts in room temperature Lower stirring 18 hours.Reaction is diluted with water (10mL) and salt water (20mL).The aqueous mixture is extracted with ethyl acetate (2x20mL) It takes.Combined organic layer is washed with salt water (20mL), is dried, filtered and is evaporated with sodium sulphate.Crude material with Analogix from The purifying of dynamicization chromatographic system, is eluted with the 0-6% methanol in methylene chloride.Product fraction merges and evaporates, and generates white foam Compound 107.Chiral purity is > 99%ee (Chiralpak OD4.6x250mm, 10um, 70% (+0.1% diethylamine of hexane)+ 30% (+0.1% diethylamine of isopropanol), 1mL/min, 254nm retention time=8.85min).
Embodiment 2. (R) -3- (4- (7H- pyrrolo- [2,3-d] pyrimidine-4-yl) -1H- pyrazol-1-yl) -3- (3,3,4, 4-d4Cyclopenta) propionitrile (compound 103) synthesis.
The preparation of 4. compound 103 of scheme
Step 1.3,3,4,4-d4Pentamethylene -1,1- diethyl dicarboxylate (40).To diethyl malonate (3.25mL, The ethanol solution (16.0mL, 42.8mmol) of the sodium ethoxide of 21 weight % is added in ethyl alcohol (20mL) solution 21.4mmol), so Deuterated -1,4- the dibromobutane of 2,2,3,3- tetra- (39,4.95g, 22.5mmol, CDN Isotopes, 98 atom % deuteriums) are added afterwards. Acquired solution stirs 2 hours under reflux, then cools to room temperature and is diluted with excessive water.Major part is then removed by distillation Ethyl alcohol, and obtained aqueous solution is extracted with ethyl acetate (3x75mL).Organic layer merges, and is washed with brine, dry (Na2SO4), mistake It filters and is concentrated under reduced pressure to obtain the 40 of yellow oil, enter subsequent step (4.67g, 100%) without further purification.
Step 2.3,3,4,4-d4Pentamethylene -1- carboxylic acid (41).Ethyl alcohol (10mL) to 40 (4.67g, 21.4mmol) is molten The sodium hydroxide solution (10mL) of 5M is added in liquid.It then adds other water (10mL), and it is small to react stirring 3 under reflux When.After being cooled to room temperature, which dilutes and most of ethyl alcohol is removed by distillation.Make the aqueous solution with 1N hydrochloric acid In acid (pH < 2), then extracted with diethyl ether (3x50mL).Organic layer merges, dry (Na2SO4), it filters and is concentrated under reduced pressure. Gained light orange solid is transferred in pressure flask, and water (70mL) is added.The pressure flask is sealed, and is reacted 160 It stirs 15 hours, then cools to room temperature at DEG C.Reaction 1N hydrochloric acid is simultaneously extracted with diethyl ether (3x50mL).Organic layer Merge, dry (Na2SO4), it filters and is concentrated under reduced pressure to obtain the 41 of amber oily (1.93g, 76%), make without further purification With.
Step 3.3,3,4,4-d4- N- methoxy-. N-methyl cyclopentane formamide (42).At 0 DEG C, to 41 (1.93g, N, O- dimethyl hydroxylamine hydrochloride (1.91g, 19.6mmol), TBTU is added in acetonitrile (30mL) solution 16.3mmol) (5.50g, 17.1mmol) and N, N- diisopropylethylamine (8.52mL, 48.9mmol).Reaction is stirred at room temperature 15 hours, so It is extracted afterwards with 1N hydrochloric acid and with ethyl acetate (3x50mL).Organic phase merges, with saturation NaHCO3Washing, it is dry (Na2SO4), it filters and is concentrated under reduced pressure.Products therefrom column chromatography (SiO2, 0-40% acetone/hexane) and it purifies to obtain clean oil 42 (1.47g, 56%) of shape.MS(ESI)162.3[(M+H)+]。
Step 4.3,3,4,4-d4Pentamethylene -1- formaldehyde (43).At 0 DEG C, to the THF of 42 (1.47g, 9.12mmol) The LiAlH of 1M is added dropwise in (35mL) solution4THF (16.4mL, 16.4mmol) solution.It is small that reaction is stirred at room temperature 1 When, then it is quenched at 0 DEG C by the way that NaOH (623 μ L) and the water (1.87mL) of water (623 μ L), 15% is sequentially added dropwise.It is quenched Reaction is stirred at room temperature 30 minutes, then passes throughIt filters and is concentrated under reduced pressure.Gained grease 1N hydrochloric acid And it is extracted with diethyl ether (3x50mL).Organic layer merges, dry (Na2SO4), it filters and is concentrated under reduced pressure to obtain clear oil 43 (767mg, 82%), use without further purification.
Step 5.3- (3,3,4,4-d4Cyclopenta) acrylonitrile (44).At 0 DEG C, to cyano methyl diethyl phosphonate The THF (3.75mL, 3.75mmol) of the potassium tert-butoxide of 1M is added dropwise in THF (10mL) solution of (0.607mL, 3.75mmol) Solution.Reaction is stirred 1 hour at 0 DEG C.THF (3mL) solution of aldehyde 43 (767mg, 7.51mmol) is added dropwise.Reaction exists It stirs 15 hours at room temperature, then diluted with excessive 1:1 water/salt water and MTBE (3x50mL) is used to extract.Organic layer merges, and does Dry (Na2SO4), it filters and is concentrated under reduced pressure.Gained grease is dissolved in CH2Cl2In (100ml) and use NaHSO3(3x25mL) is washed It washs.Organic phase is dried (Na2SO4), 44 (537mg, 57%) of light orange oily are filtered and obtain after being concentrated under reduced pressure, without pure Change and uses.
Step 6. (+/-)-(4- (1- (2- cyano -1- (3,3,4,4-d4Cyclopenta) ethyl) -1H- pyrazoles -4- base) - 7H- pyrrolo- [2,3-d] pyrimidin-7-yl) methyl pivalate ((+/-) 45).To 37 (514mg, 1.72mmol, such as Lin, Q. Prepared described in equal Org.Lett., 2009,11,1999-2002) acetonitrile (15mL) solution in be added 44 (537mg, 4.29mmol), DBU (540 μ L, 3.61mmol) then is added.Reaction is stirred at room temperature 15 hours, then dense under reduced vacuum Contracting.Gained crude mixture is diluted with water and ethyl acetate (3x50mL) is used to extract.Organic layer merges, dry with 1N salt acid elution (Na2SO4), it filters and is concentrated under reduced pressure.With positive column chromatography (SiO2, 0-60% ethyl acetate/hexane) and white bubble is obtained after purification (+/-) 45 (368mg, 50%) of foam.1H NMR(DMSO-d6,400MHz)δ8.84(s,1H),8.79(s,1H),8.40(s, 1H),7.75(d,J=3.7Hz,1H),7.12(d,J=3.7Hz,1H),6.24(s,2H),4.53(td,J=9.7,4.2Hz,1H), 3.32–3.14(m,2H),2.41(q,J=8.7Hz,1H),1.79(dd,J=12.6,7.6Hz,1H),1.36–1.11(m,3H), 1.08(s,9H).;MS(ESI)425.2[(M+H)+]。
Step 7. (R)-(4- (1- (2- cyano -1- (3,3,4,4-d4Cyclopenta) ethyl) -1H- pyrazoles -4- base) -7H- Pyrrolo- [2,3-d] pyrimidin-7-yl) methyl pivalate ((R) -45).Racemic compound (+/-) 45 (151mg) is with 30mg/ The concentration of mL is dissolved in acetonitrile, and with (+/-) 45 solution of 1000 μ L of per injection in Daicel ChiralPak AD column Chiral resolution is carried out by preparative HPLC with isocratic method on (20x250mm, 10 μm): 30% isopropanol (+0.1% diethylamine)/ 70% hexane (+0.1% diethylamine), flow velocity 17mL/min.In these cases, eluted at 15.5 minutes with (S) -45 and (R) -45 realization baseline separation is eluted at 20.7 minutes.
Fraction containing each enantiomter individually merges and is concentrated to generate the colorless film of (S) -45 of 51mg And the colorless film of (R) -45 of 53mg.
Step 8. (R) -3- (4- (7H- pyrrolo- [2,3-d] pyrimidine-4-yl) -1H- pyrazol-1-yl) -3- (3,3,4,4- d4Cyclopenta) propionitrile (compound 103).(R) -45 (53mg, 0.13mmol, 1 equivalents) are dissolved in first in the scintillation vial of 20mL In alcohol (2mL).It is added sodium hydroxide (the 1M solution of 0.25mL, 0.25mmol, 2 equivalents), reaction mixture is stirred at room temperature 18 hours.Reaction mixture water (10mL) and salt water (20mL) dilution.The aqueous mixture is with ethyl acetate (2x20mL) Extraction.Combined organic layer is washed with salt water (20mL), is dried, filtered and concentrated with sodium sulphate.Thick material with Analogix from The purifying of dynamicization chromatographic system, is eluted with the 0-6% methanol in methylene chloride.Product fraction merges and evaporates to generate white foam Compound 103, purity be~90%, not completely de-protected methylol intermediate be major impurity.Further chromatography Its purity can not be further increased.The 90% pure substance is dissolved in THF (2mL), and with it is a few drop 10% aqueous hydroxide Sodium is handled 8 hours at 40 DEG C, causes to be fully converted to compound 103.The reaction mixture water (10mL) dilutes and uses second Acetoacetic ester (2x10mL) extraction.Combined organic layer is dried, filtered and concentrated into white foam with sodium sulphate.The foam is dissolved It in least acetonitrile, is diluted with water, and is lyophilized to provide the compound 103 (14mg, 35% yield) of white solid.Chiral purity Degree is > 99%ee (Chiralpak OD4.6x250mm, 10um, 70% (+0.1% diethylamine of hexane)+30% (isopropanol+0.1% two Ethamine), 1mL/min, 254nm retention time=7.56min).
Embodiment 3. (R) -3- (4- (7H- pyrrolo- [2,3-d] pyrimidine-4-yl) -1H- pyrazol-1-yl) -3- (cyclopenta - d9) propionitrile (compound 127) synthesis.
The preparation of 5. compound 127 of scheme
Step 1.2,2,3,3,4,4,5,5-d8Pentamethylene -1,1- diethyl dicarboxylate (47).To diethyl malonate Be added in ethyl alcohol (40mL) solution of (6.24mL, 41.1mmol) sodium ethoxide of 21 weight % ethanol solution (30.7mL, 82.2mmol), 1,1,2,2,3,3,4,4- eight deuterated-Isosorbide-5-Nitrae-dibromobutane (46,9.67g, 43.2mmol, CDN is then added Isotopes, 98 atom % deuteriums).Acquired solution stirs 2 hours under reflux, then cools to room temperature and is diluted with excessive water.With After be removed by distillation most of ethyl alcohol, and obtained aqueous solution is extracted with ethyl acetate (3x75mL).Organic layer merges, and uses salt water Washing, dry (Na2SO4), filter and be concentrated under reduced pressure to obtain the 47 of yellow oily (9.12g, 100%), without further purification and into Enter subsequent step.
The complete deuterated pentamethylene -1- carboxylic acid (48) of step 2..Into ethyl alcohol (20mL) solution of 47 (9.12g, 41.1mmol) The sodium hydroxide solution (20mL) of 5M is added.It then adds other water (15mL), reaction is stirred 3 hours under reflux.It is cold But to after room temperature, reaction excessive water dilutes and most of ethyl alcohol is removed by distillation.Make the aqueous solution in acidity with 1N hydrochloric acid (pH < 2) are then extracted with diethyl ether (3x50mL).Organic layer merges, dry (Na2SO4), it filters and is concentrated under reduced pressure.Gained is shallow Orange solids are transferred in pressure flask, and D is added2O(120mL).The pressure flask is sealed, and is reacted at 160 DEG C Stirring 15 hours, then cools to room temperature.Reaction 1N hydrochloric acid is simultaneously extracted with diethyl ether (3x50mL).Organic layer merges, Dry (Na2SO4), it filters and is concentrated under reduced pressure to obtain the 48 of yellow oily (4.58g, 90%), use without further purification.
Step 3.N- methoxy-. N-methyl (pentamethylene-d9) formamide (49).At 0 DEG C, to 48 (4.58g, 37.2mmol) Acetonitrile (60mL) solution in be added N, O- dimethyl hydroxylamine hydrochloride (4.35g, 44.6mmol), TBTU (12.5g, 39.1mmol) and N, N- diisopropylethylamine (19.4mL, 112mmol).Reaction is stirred at room temperature 15 hours, then uses 1N salt Acid dilution is simultaneously extracted with ethyl acetate (3x50mL).Organic phase merges, with saturation NaHCO3Washing, dry (Na2SO4), filtering is simultaneously It is concentrated under reduced pressure.Products therefrom column chromatography (SiO2, 0-50% ethyl acetate/hexane) and it purifies to obtain the 49 of clear oil (3.41g,55%)。MS(ESI)167.2[(M+H)+]。
The complete deuterated pentamethylene -1- formaldehyde (50) of step 4..At 0 DEG C, the THF (80mL) to 49 (3.41g, 20.5mmol) is molten The LiAlH of 1M is added dropwise in liquid4THF (37.0mL, 37.0mmol) solution.Reaction is stirred at room temperature 1 hour, then exists D is added dropwise by sequence at 0 DEG C2O (1.41mL), 15% NaOD/D2O (1.41mL) and D2O (4.23mL) is quenched.What this was quenched Reaction is stirred at room temperature 30 minutes, then passes throughIt filters and is concentrated under reduced pressure.Gained grease 1N DCl/D2O It dilutes and diethyl ether (3x50mL) is used to extract.Organic layer merges, dry (MgSO4), it filters and is concentrated under reduced pressure to obtain clean oil 50 (1.79g, 82%) of shape, use without further purification.
Step 5.3- (complete deuterated cyclopenta) acrylonitrile (51).At 0 DEG C, to cyano methyl diethyl phosphonate (1.35mL, THF (8.34mL, 8.34mmol) solution of the potassium tert-butoxide of 1M is added dropwise in THF (25mL) solution 8.34mmol).Reaction It is stirred 1 hour at 0 DEG C.Then THF (5mL) solution of aldehyde 50 (1.79g, 16.7mmol) is added dropwise.Reaction is in room temperature Then lower stirring 15 hours is diluted with excessive 1:1 water/salt water and MTBE (3x50mL) is used to extract.Organic layer merges, dry (Na2SO4), it filters and is concentrated under reduced pressure.Organic phase merges, dry (Na2SO4), it filters and is concentrated under reduced pressure to obtain light orange oily 51 (1.61g, 74%), use without further purification.
Step 6. (+/-)-(4- (1- (2- cyano -1- (cyclopenta-d9) ethyl) -1H- pyrazoles -4- base) -7H- pyrrolo- [2,3-d] pyrimidin-7-yl) methyl pivalate ((+/-) 52).To 37 (619mg, 2.07mmol, in Lin, Q. etc. Org.Lett., described in 2009,11,1999-2002 prepare) acetonitrile (15mL) solution in be added 51 (673mg, 5.17mmol), DBU (650 μ L, 4.35mmol) then is added.Reaction is stirred at room temperature 15 hours, is then concentrated under reduced pressure.Institute Crude mixture is obtained to be diluted with water and ethyl acetate (3x50mL) is used to extract.Organic layer merges, dry with 1N salt acid elution (Na2SO4), it filters and is concentrated under reduced pressure.With positive column chromatography (SiO2, 0-60% ethyl acetate/hexane) purifying obtain white foam (+/-) 52 (447mg, 50%).1H NMR(DMSO-d6,400MHz)δ8.84(s,1H),8.79(s,1H),8.39(s,1H), 7.75(d,J=3.7Hz,1H),7.12(d,J=3.7Hz,1H),6.24(s,2H),4.53(dd,J=9.6,4.2Hz,1H), 3.32–3.13(m,2H),1.08(s,9H).;MS(ESI)430.3[(M+H)+]。
Step 7. (R)-(4- (1- (2- cyano -1- (cyclopenta-d9) ethyl) -1H- pyrazoles -4- base) -7H- pyrrolo- [2, 3-d] pyrimidin-7-yl) methyl pivalate ((R) -52).Racemic compound (+/-) 52 (162mg) is with the concentration of 30mg/mL Be dissolved in acetonitrile, and with (+/-) 52 solution of 1000 μ L of per injection Daicel ChiralPak AD column (20x250mm, 10 μm) on isocratic method chiral resolution is carried out by preparative HPLC: 30% isopropanol (+0.1% diethylamine)/70% hexane (+ 0.1% diethylamine), flow velocity 17mL/min.In these cases, it is eluted at 15.4 minutes with (S) -52 and (R) -52 exists Baseline separation is realized in elution at 20.5 minutes.
Fraction containing each enantiomter individually merge and be concentrated with generate the colorless film of (S) -52 of 61mg with And the colorless film of (R) -52 of 63mg.
Step 8. (R) -3- (4- (7H- pyrrolo- [2,3-d] pyrimidine-4-yl) -1H- pyrazol-1-yl) -3- (cyclopenta - d9) propionitrile (compound 127).(R) -52 (60mg, 0.14mmol, 1 equivalents) are dissolved in methanol (2mL) in the scintillation vial of 20mL In.It is added sodium hydroxide (the 1M solution of 0.28mL, 0.28mmol, 2 equivalents), and to be stirred at room temperature 18 small for reaction mixture When.Reaction mixture water (10mL) and salt water (20mL) dilution.The aqueous mixture is extracted with ethyl acetate (2x20mL). Combined organic layer is washed with salt water (20mL), is dried, filtered and concentrated with sodium sulphate.Thick material automates color with Analogix Spectra system purifying, with 0-6% methanol/elution in methylene chloride.Product fraction merges and evaporates to generate the chemical combination of white foam Object 127 (34mg), purity are~90%, and de-protected methylol intermediate is not major impurity completely.Further chromatography Its purity can not be further increased.90% pure substance is dissolved in THF (2mL), and is existed with the aqueous NaOH of several drops 10% It is handled 8 hours at 40 DEG C, causes to be fully converted to compound 127.The reaction mixture is diluted with water (10mL) and with acetic acid second Ester (2x10mL) extraction.Organic layer after merging is dried, filtered and concentrated into white foam with sodium sulphate.The foam is dissolved in It in least acetonitrile, is diluted with water, and is lyophilized to provide the compound 127 (19mg, 42% yield) of white solid.Chiral purity For > 99%ee (+30% (+0.1% diethyl of isopropanol of Chiralpak OD4.6x250mm, 10um, 70% (+0.1% diethylamine of hexane) Amine), 1mL/min, 254nm retention time=7.55min).
Embodiment 4.CYP3A4SupersomesTMThe assessment of middle metabolic stability
People CYP3A4SupersomesTMThe assessment of the metabolic stability of middle compound 103,107 and 127
SUPERSOMESTMAnalysis.Test-compound is prepared in DMSO, 103,107,127 and Lu Suo of compound is for Buddhist nun's 10mM stock solution.The 10mM stock solution is diluted to 15.6 μM in acetonitrile (ACN).People CYP3A4supersomesTM (1000pmol/mL is purchased from BD GentestTMProducts and Services) in the kaliumphosphate buffer of 0.1M (pH7.4, the MgCl containing 3mM2) in be diluted to 62.5pmol/mL.Diluted supersomes is added to 96- in triplicate In the hole of hole polypropylene board.10 μ L aliquots of 15.6 μM of test-compounds are added in the supersomes, and are mixed Object is preheated 10 minutes.By the way that the NADPH solution of preheating is added with initiation reaction.Final reaction volume is 0.5mL, and is wrapped It is contained in the kaliumphosphate buffer (MgCl of pH7.4,3mM of 0.1M2) in 50pmol/mL CYP3A4supersomesTM、0.25 μM test-compound and 2mM NADPH.The reaction mixture incubated at 37 DEG C, and at 0,5,10,20 and 30 minute 50 μ L aliquots are pipetted, and are added in the 96- orifice plate that there is the ice-cold CAN of interior target containing 50 μ L to stop reaction.The plate is at 4 DEG C Lower storage 20 minutes, hereafter 100mL water is added in the hole of plate, is centrifuged the protein balling-up so that precipitating later.Supernatant It is transferred in another 96- orifice plate, and by using the mass spectrometric LC-MS/MS of Applied Bio-systems API4000 To analyze remaining size of population.
Data analysis: test-compound is calculated from linear regression slope of the LN (the remaining parent of %) to incubative time relationship Vitro half-lives (t1/2Value):
External t1/2=0.693/k, wherein k=- [% remains parent (ln) to the slope of the linear regression of incubative time].
The experiment as the result is shown in table 3 and Fig. 1.As shown in table 3, it is 14.5 that Lu's rope, which is calculated, for the half-life period of Buddhist nun Minute.On the contrary, each compound 103,107 and 127 is more stable in the supersomes, half-life period calculated is respectively 16.9,17.9 and 32.0 minutes.The half-life period of this representation compound 103 improves 17%, and the half-life period of compound 107 improves 23%, and the half-life period of compound 127 improves 121%.
Table 3: in people CYP3A4SupersomesTMThe opposite Lu's rope of middle compound 103,107 and 127 replaces the metabolic stability of Buddhist nun
* % Δ=[(deuterated substance)-(non-deuterated substance)] (100)/(non-deuterated substance)
Metabolic stability evaluation of the embodiment 5. in human liver's microsome
Microsomal assay: people's liver microsomes (20mg/mL) are obtained from Xenotech, LLC (Lenexa, KS).Reduction form β-nicotinamide-adenine dinucleotide phosphate (NADPH), magnesium chloride (MgCl2) and dimethyl sulfoxide (DMSO) be purchased from Sigma- Aldrich。
The measurement of metabolic stability: the test-compound stock solution of 7.5mM is prepared in DMSO.The deposit of the 7.5mM Solution is diluted to 12.5-50 μM with acetonitrile (ACN).Kaliumphosphate buffer of the people's liver microsomes of 20mg/mL in 0.1M (pH7.4 contains 3mM MgCl2) in be diluted to 0.625mg/mL.The diluted microsome is added to 96- hole depth in triplicate In the hole of hole polypropylene board.10 μ L aliquots of 12.5-50 μM of test-compound are added in microsome, and mixture is pre- Heat 10 minutes.The NADPH solution of preheating is added with initiation reaction.Final reaction volume is 0.5mL, and included in 0.1M's Kaliumphosphate buffer (the MgCl of pH7.4,3mM2) in 0.5mg/mL people's liver microsomes, 0.25-1.0 μM of test-compound And the NADPH of 2mM.The reaction mixture incubates at 37 DEG C, and pipettes 50 μ L aliquots at 0,5,10,20 and 30 minute, It is added in the shallow bore hole 96- orifice plate that there is the ice-cold CAN of interior target containing 50 μ L to stop reaction.The plate stores 20 points at 4 DEG C Clock, hereafter 100 μ L water are added in the hole of plate, are centrifuged the protein balling-up so that precipitating later.Supernatant is transferred to separately In one 96- orifice plate, and by remaining to analyze with the mass spectrometric LC-MS/MS of Applied Bio-systems API4000 Size of population.Compound and positive control 7-ethoxy coumarin (1 μM) to non-deuterated Formulas I or formula A follow same journey Sequence.It is tested in triplicate.
Data analysis: test-compound is calculated from linear regression slope of the remaining parent of % (ln) to incubative time relationship External t1/2:
External t1/2=0.693/k
K=- [% remains parent (ln) to the slope of the linear regression of incubative time]
Data analysis is carried out with Microsoft Excel Software.
Without further illustrating, it is believed that those skilled in the art can utilize the reality of the description and explanation of front Example is applied to make and use the compound of the present invention and implement claimed method.It should be understood that the discussion above and reality It applies example and only illustrates the detailed description of certain preferred embodiments.It is various improvement and equivalent can not depart from it is of the invention It is made under spirit and scope, this will be apparent for those of ordinary skill in the art.

Claims (13)

1. a kind of compound selected from the following:
Or the pharmaceutically acceptable salt of any aforesaid compound, any of them are not designated as the atom of deuterium with its natural same position Plain abundance, which exists and is wherein specifically designated, to be mixed for the position of " deuterium " at least 90% deuterium.
2. compound as claimed in claim 1 or its pharmaceutically acceptable salt, wherein being specifically designated has extremely for the position of " deuterium " Few 95% deuterium incorporation.
3. compound as claimed in claim 1 or its pharmaceutically acceptable salt, wherein being specifically designated has extremely for the position of " deuterium " Few 97% deuterium incorporation.
4. compound as claimed in claim 1, wherein the compound is
Or its pharmaceutically acceptable salt, any of them be not designated as the atom of deuterium with its natural isotopic abundance exist and its In the position that is specifically designated as " deuterium " mixed at least 90% deuterium.
5. compound as claimed in claim 4 or its pharmaceutically acceptable salt, wherein being specifically designated has extremely for the position of " deuterium " Few 95% deuterium incorporation.
6. compound as claimed in claim 4 or its pharmaceutically acceptable salt, wherein being specifically designated has extremely for the position of " deuterium " Few 97% deuterium incorporation.
7. the medicine of compound or its pharmaceutically acceptable salt and pharmaceutically acceptable carrier comprising any one of claim 1-3 Compositions.
8. pharmaceutical composition as claimed in claim 7 is further included selected from lenalidomide, pabishta, capecitabine, Yi Ximei Smooth and combinations thereof therapeutic agent.
9. the medicine of compound or its pharmaceutically acceptable salt and pharmaceutically acceptable carrier comprising any one of claim 4-6 Compositions.
10. the compound of any one of claim 1-6 or the pharmaceutical composition of any one of claim 7-9 are in preparation for treating It is the myelofibrosis of the subject needed, cancer of pancreas, prostate cancer, breast cancer, leukaemia, non-Hodgkin lymphoma, multiple Purposes in the drug of myeloma, psoriasis or combinations thereof.
11. such as the purposes of claim 10, wherein the myelofibrosis is primary myelofibrosis, polycythemia vera Myelofibrosis, primary thrombocytosis or combinations thereof after myelofibrosis, primary thrombocytosis after disease.
12. the purposes of claim 10, the drug with selected from lenalidomide, pabishta, capecitabine, Exemestane and its Combined therapeutic agent uses.
13. the compound of any one of claim 1-6 or the pharmaceutical composition of any one of claim 7-9 are in preparation for inhibiting Purposes in one of JAk1 and JAk2 or a variety of drugs.
CN201310700192.2A 2013-12-18 2013-12-18 Lu Suo replaces the deuterated derivative of Buddhist nun Active CN104725380B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201910481275.4A CN110229159B (en) 2013-12-18 2013-12-18 Deuterated derivative of ruxotinib
CN201310700192.2A CN104725380B (en) 2013-12-18 2013-12-18 Lu Suo replaces the deuterated derivative of Buddhist nun

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201310700192.2A CN104725380B (en) 2013-12-18 2013-12-18 Lu Suo replaces the deuterated derivative of Buddhist nun

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN201910481275.4A Division CN110229159B (en) 2013-12-18 2013-12-18 Deuterated derivative of ruxotinib

Publications (2)

Publication Number Publication Date
CN104725380A CN104725380A (en) 2015-06-24
CN104725380B true CN104725380B (en) 2019-06-28

Family

ID=53449858

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201310700192.2A Active CN104725380B (en) 2013-12-18 2013-12-18 Lu Suo replaces the deuterated derivative of Buddhist nun
CN201910481275.4A Active CN110229159B (en) 2013-12-18 2013-12-18 Deuterated derivative of ruxotinib

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN201910481275.4A Active CN110229159B (en) 2013-12-18 2013-12-18 Deuterated derivative of ruxotinib

Country Status (1)

Country Link
CN (2) CN104725380B (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105777754B (en) * 2014-12-16 2019-07-26 北京赛林泰医药技术有限公司 Pyrrolopyrimidine compounds
AU2017261286B2 (en) * 2016-05-04 2023-03-23 Sun Pharmaceutical Industries, Inc. Treatment of hair loss disorders with deuterated JAK inhibitors
JP6737952B2 (en) * 2016-08-17 2020-08-12 深▲セン▼市塔吉瑞生物医▲薬▼有限公司Shenzhen TargetRx,Inc. Fused bicyclic compounds for inhibiting tyrosine kinase activity
DK3866931T3 (en) * 2018-10-18 2023-04-24 Oncopeptides Ab Compounds containing deuterium

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101448826A (en) * 2005-12-13 2009-06-03 因塞特公司 Heteroaryl-substituted pyrrolo [2,3-b] pyrroles and pyrrolo [2,3-b] pyrimidines as inhibitors of Bissaelectrical kinases

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101448826A (en) * 2005-12-13 2009-06-03 因塞特公司 Heteroaryl-substituted pyrrolo [2,3-b] pyrroles and pyrrolo [2,3-b] pyrimidines as inhibitors of Bissaelectrical kinases

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Metabolism,Excretion,and Pharmacokinetics of [14C]INCB018424,a Selective Janus Tyrosine Kinase 1/2 Inhibitor,in Humans;Adam D. Shilling,等;《DRUGMETABOLISM AND DISPOSITION》;20101231;第38卷(第11期);第2026页第2栏第1段,第2028页图2

Also Published As

Publication number Publication date
CN110229159A (en) 2019-09-13
CN104725380A (en) 2015-06-24
CN110229159B (en) 2021-08-24

Similar Documents

Publication Publication Date Title
US20230355629A1 (en) Deuterated derivatives of ruxolitinib
AU2016238877B2 (en) Deuterated derivatives of ruxolitinib
JP2023154048A (en) Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making modulator
US8921366B2 (en) Substituted triazolo-pyridazine derivatives
CN101815717A (en) The meta-bolites of JANUS kinase inhibitor (R)-3-(4-(7H-pyrrolo-[2,3-d] pyrimidine-4-yl)-1H-pyrazol-1-yl)-3-cyclopentyl propionitrile
CN106170290A (en) Substituted di-amino-pyrimidine based compound, combinations thereof and treatment method thereof
CN104725380B (en) Lu Suo replaces the deuterated derivative of Buddhist nun
TW201602104A (en) New 1-(4-pyrimidinyl)-1H-pyrrolo[3,2-c]pyridine derivatives as NIK inhibitors
WO2019001425A1 (en) Deuterated osimertinib derivative and application thereof
CN107759600A (en) Crystallization as the Pyrrolopyrimidine compounds of JAK inhibitor
JP2022526926A (en) Imidazolonyl quinoline compounds and their therapeutic use
CN104098570A (en) Crystal form of Ticagrelor Brilinta and preparation method and purpose thereof
JP6367545B2 (en) Deuterated derivatives of ruxolitinib
CN101160312A (en) Tricyclic azole derivatives, their manufacture and use as pharmaceutical agents
JP2021523120A (en) Solid form of CERDULATINIB
KR102433283B1 (en) Deuterated derivatives of ruxolitinib
CN106459056A (en) Substituted triazolobenzodiazepines
JP6830460B2 (en) Deuterated derivative of ruxolitinib
WO2012129381A1 (en) Deuterated preladenant
WO2022199503A1 (en) Use of heterocyclic compound in treating diseases related to kinase drug-resistant mutation and method therefor
WO2014081816A1 (en) Fluoro-derivatives of pyrazole-substituted amino-heteroaryl compounds
JPH02289551A (en) Ameliorant containing pyrimidine derivative as active ingredient for cerebral function

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
TR01 Transfer of patent right
TR01 Transfer of patent right

Effective date of registration: 20231012

Address after: new jersey

Patentee after: Sun Pharmaceutical Industry Co.,Ltd.

Address before: Massachusetts

Patentee before: Concert Pharmaceuticals, Inc.