NZ714267B2 - Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases - Google Patents

Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases Download PDF

Info

Publication number
NZ714267B2
NZ714267B2 NZ714267A NZ71426714A NZ714267B2 NZ 714267 B2 NZ714267 B2 NZ 714267B2 NZ 714267 A NZ714267 A NZ 714267A NZ 71426714 A NZ71426714 A NZ 71426714A NZ 714267 B2 NZ714267 B2 NZ 714267B2
Authority
NZ
New Zealand
Prior art keywords
alkyl
compound
aryl
carboxylic
halogen
Prior art date
Application number
NZ714267A
Other versions
NZ714267A (en
Inventor
Werner EMBRECHTS
Tim Hugo Maria Jonckers
Stefaan Julien Last
Gowan David Craig Mc
Serge Maria Aloysius Pieters
Pierre Jean Marie Bernard Raboisson
Original Assignee
Janssen Sciences Ireland Uc
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 Janssen Sciences Ireland Uc filed Critical Janssen Sciences Ireland Uc
Priority to NZ754320A priority Critical patent/NZ754320B2/en
Priority claimed from PCT/EP2014/063467 external-priority patent/WO2014207082A1/en
Publication of NZ714267A publication Critical patent/NZ714267A/en
Publication of NZ714267B2 publication Critical patent/NZ714267B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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

This invention concerns pyrrolo[3,2-d]pyrimidine derivatives of formula (I), processes for their preparation, pharmaceutical compositions, and their use in treatment and /or therapy of diseases.

Description

/063467 Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases This invention relates to pyrrolo[3,2-d]pyrimidine derivatives, processes for their preparation, pharmaceutical compositions, and their use in treatment and /or therapy of diseases.
The present invention relates to the use of pyrrolo[3,2-d]pyrimidine derivatives more specifically to the use of pyrrolo[3,2-d]pyrimidine derivatives in the treatment of viral infections, immune or inflammatory disorders, whereby the modulation, or agonism, of toll-like-receptors (TLRs) is involved. ike Receptors are primary transmembrane proteins terized by an extracellular leucine rich domain and a cytoplasmic extension that contains a conserved region. The innate immune system can recognize pathogen-associated molecular patterns via these TLRs expressed on the cell surface of certain types of immune cells. Recognition of n pathogens activates the production of cytokines and upregulation of co-stimulatory molecules on phagocytes.
This leads to the modulation of T cell our.
A majority of mammalian species have between ten and fifteen types of Toll-like receptors. Thirteen TLRs (named simply TLR1 to TLR13) have been identified in humans and mice together, and equivalent forms of many of these have been found in other ian species. However, equivalents of certain TLR found in humans are not present in all mammals. For example, a gene coding for a protein ous to TLR10 in humans is present in mice, but appears to have been damaged at some point in the past by a retrovirus. On the other hand, mice s TLRs 11, 12, and 13, none of which are represented in humans. Other mammals may express TLRs which are not found in humans. Other non-mammalian species may have TLRs distinct from mammals, as demonstrated by TLR14, which is found in the Takifugu pufferfish. This may complicate the process of using mental animals as models of human innate immunity.
For reviews on toll-like receptors see the following journal articles. Hoffmann, J.A., Nature, 426, p33-38, 2003; Akira, S., Takeda, K., and , T., Annual Rev.
Immunology, 21, p335-376, 2003; ch, R. J., Nature Reviews: Immunology, 4, p512-520, 2004.
Compounds ting activity on Toll-Like receptors have been previously described such as heterocyclic derivatives in WO2000/006577, adenine tives in W098/01448 and W099/28321, and dines in W02009/O67081.
In the treatment of certain viral infections, regular injections of interferon (lFN-alfa) can be administered, as is the case for tis C virus (HCV). Orally available small molecule IFN inducers offer the potential ages of reduced immunogenicity and convenience of administration. Thus, novel IFN inducers are potentially effective new class of drugs for the treatment of viral infections. For an e in the literature of a small molecule IFN inducer having antiviral effect see De Clercq, E.; ps, J.; De Somer, P. Science 1978, 200, 563-565.
Interferon or is also given to patients in combination with other drugs in the treatment of certain types of cancer. TLR 7/8 agonists are also of interest as vaccine adjuvants because of their ability to induce pronounced Th1 response.
However, there exists a strong need for novel Toll-Like receptor modulators having preferred selectivity, and an improved safety profile compared to the compounds of the prior art.
In accordance with the present invention a compound of formula (I) is provided and their pharmaceutically acceptable salt, solvate or polymorph thereof wherein R1 is H, fluorine or methyl; R2 is H, halogen or C13 alkyl; R3 is 01-6 alkyl optionally substituted by one or more substituents independently selected from aryloxy, heterocycle, n, aryl, mino, dialkylamino, C143 alkyl, carboxylic acid, carboxylic ester, carboxylic amide, nitrile, or 01-6 alkoxy; or wherein R3 is an alkylaryl optionally substituted by one or more substituents independently selected from n, aryloxy, aryl, alkylamino, dialkylamino, C143 alkyl, carboxylic acid, carboxylic ester, carboxylic amide, amide, nitrile, or 01-6 ; R4 is 01-6 alkyl ally substituted by one or more substituents independently selected from hydroxyl, 01-6 alkyl, C37 lkyl, 02-6 alkenyl or aryl optionally further substituted by 01-3 alkyl, and C37 cycloalkyl ally further substituted by 01-6 alkyl; or n R4 is an alkylaryl optionally tuted by one or more substituents independently selected from halogen, aryloxy, aryl, alkylamino, dialkylamino, C1-6 alkyl, carboxylic acid, carboxylic ester, carboxylic amide, sulfonamide, nitrile, or C 1-6 alkoxy, with the o that the compound does not satisfy any one of the following structural formulae: , ,and .
Preferred compounds are those of formula (I) n R3 is a CH2-aryl group (substituted or unsubstituted), and R1, R2, and R4 are described as above.
In a second embodiment are the compounds of formula (I) wherein R3 and R4 are both CH2-aryl groups ally further substituted as described above, and R1, and R2 are as described as above.
Other preferred embodiments are those of formula (I) wherein R1 is fluorine, R2 is hydrogen, and R3 and R4 are described as above.
The most preferred compound is compound of a (II) having the following chemical structure: (II) The compounds of formula (I) and (II) and their pharmaceutically acceptable salt, solvate or polymorph thereof have activity as pharmaceuticals, in particular as modulators of Toll-Like or (especially TLR7) activity. (followed by page 3a) -3a- In a particular aspect, the present invention provides the use of a compound of formula H2N N N N O R3 R4 (I) and their pharmaceutically acceptable salt, solvate or polymorph thereof wherein R1 is H, fluorine or methyl; R2 is H, n or C1-3 alkyl; R3 is C1-6 alkyl optionally substituted by one or more tuents independently selected from aryloxy, cycle, halogen, aryl, alkylamino, dialkylamino, C1-6 alkyl, carboxylic acid, carboxylic ester, carboxylic amide, nitrile, or C1-6 alkoxy; or wherein R3 is an alkylaryl optionally substituted by one or more substituents ndently selected from halogen, aryloxy, aryl, alkylamino, dialkylamino, C1-6 alkyl, carboxylic acid, ylic ester, carboxylic amide, sulfonamide, nitrile, or C 1-6 alkoxy; R4 is C1-6 alkyl optionally substituted by one or more substituents independently selected from hydroxyl, C1-6 alkyl, C3-7 cycloalkyl, C2-6 alkenyl or aryl optionally further substituted by C1-6 alkyl, and C3-7 cycloalkyl optionally further tuted by C1-6 alkyl; or wherein R4 is an alkylaryl optionally substituted by one or more substituents independently selected from halogen, aryloxy, aryl, alkylamino, dialkylamino, C1-6 alkyl, carboxylic acid, carboxylic ester, carboxylic amide, sulfonamide, nitrile, or C1-6 , in the manufacture of a medicament for the treatment of a disorder in which the modulation of TLR7 is involved.
In a further aspect the present invention provides a pharmaceutical composition comprising a compound of a (I) or (II) or a pharmaceutically acceptable salt, solvate or polymorph thereof together with one or more pharmaceutically acceptable excipients, diluents or carriers.
Furthermore a compound of formula (I) or (II) or a pharmaceutically acceptable salt, solvate or polymorph f ing to the current invention, or a pharmaceutical [FOLLOWED BY PAGE 4] composition comprising said compound of a (I) or (II) or a pharmaceutically acceptable salt, solvate or polymorph f can be used as a medicament.
Another aspect of the invention is that a compound of formula (I) or (II) or a ceutically acceptable salt, solvate or polymorph thereof, or said pharmaceutical composition comprising said compound of formula (I) or ‘II) or a pharmaceutically acceptable salt, solvate or polymorph thereof can be used accordingly in the ent of any disorder in which the modulation of TLR7 is involved.
The term "alkyl" refers to a straight-chain or branched-chain saturated aliphatic hydrocarbon containing the specified number of carbon atoms.
The term "halogen" refers to fluorine, ne, bromine or iodine.
The term "alkylaryl" refers to a straight-chain or branched-chain saturated aliphatic hydrocarbon containing the specified number of carbon atoms tuted by an aryl wherein "aryl" is defined as below.
The term "alkenyl" refers to an alkyl as defined above consisting of at least two carbon atoms and at least one carbon-carbon double bond.
The term "cycloalkyl" refers to a carbocyclic ring containing the ied number of carbon atoms.
The term "alkoxy" refers to an alkyl (carbon and hydrogen chain) group singular bonded to oxygen like for instance a methoxy group or ethoxy group.
The term "aryl" means an aromatic ring structure optionally comprising one or two atoms selected from N, O and S, in particular from N and O. Said aromatic ring structure may have 5, 6 or 7 ring atoms. In particular, said aromatic ring structure may have 5 or 6 ring atoms.
The term "aryloxy" refers to an aromatic ring ure. Said aromatic group is singularly bonded to oxygen.
The term "heterocycle" refers to molecules that are saturated or partially saturated and include tetrahydrofuran, e or other cyclic ethers. Heterocycles containing nitrogen include, for example azetidine, morpholine, piperidine, piperazine, pyrrolidine, and the like. Other heterocycles include, for example, thiomorpholine, dioxolinyl, and cyclic sulfones.
Pharmaceutically acceptable salts of the compounds of formula (I) and (II) include the acid on and base salts thereof. Suitable acid addition salts are formed from acids which form non-toxic salts. Suitable base salts are formed from bases which form non- toxic salts.
The compounds of the invention may also exist in unsolvated and solvated forms. The term "solvate" is used herein to describe a molecular complex comprising the compound of the invention and one or more pharmaceutically acceptable solvent molecules, for example, ethanol.
The term "polymorph" refers to the ability of the compound of the invention to exist in more than one form or crystal structure.
The compounds of the present invention may be administered as crystalline or amorphous products. They may be obtained for example as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, spray drying, or evaporative drying. They may be administered alone or in combination with one or more other nds of the invention or in combination with one or more other drugs.
Generally, they will be administered as a formulation in association with one or more pharmaceutically acceptable ents. The term "excipient" is used herein to describe any ingredient other than the compound(s) of the invention. The choice of excipient depends largely on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
The compounds of the present invention or any subgroup thereof may be formulated into various ceutical forms for administration es. As appropriate compositions there may be cited all compositions usually employed for systemically administering drugs. To prepare the pharmaceutical compositions of this invention, an effective amount of the particular compound, optionally in addition salt form, as the active ingredient is ed in intimate admixture with a pharmaceutically acceptable carrier, which carrier may take a wide variety of forms depending on the form of preparation desired for administration. These ceutical compositions are bly in unitary dosage form le, for e, for oral, rectal, or aneous administration. For example, in preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs, emulsions, and solutions; or solid rs such as starches, sugars, kaolin, ts, lubricants, binders, disintegrating agents and the like in the case of powders, pills, capsules, and tablets. e of their ease in administration, tablets and es represent the most advantageous oral dosage unit forms, in which case solid pharmaceutical carriers are obviously ed. Also included are solid form preparations that can be converted, shortly before use, to liquid forms. In the compositions suitable for percutaneous administration, the carrier optionally ses a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable ves of any nature in minor proportions, which additives do not introduce a significant deleterious effect on the skin. Said additives may facilitate the stration to the skin and/or may be helpful for preparing the desired compositions. These compositions may be administered in various ways, e.g., as a transdermal patch, as a spot-on, as an ointment. The compounds of the present invention may also be administered via inhalation or insufflation by means of methods and formulations employed in the art for administration via this way. Thus, in general the compounds of the present ion may be administered to the lungs in the form of a solution, a suspension or a dry powder.
It is especially advantageous to formulate the aforementioned pharmaceutical itions in unit dosage form for ease of administration and uniformity of dosage.
Unit dosage form as used herein refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Examples of such unit dosage forms are tablets (including scored or coated s), capsules, pills, powder s, wafers, suppositories, injectable solutions or suspensions and the like, and segregated multiples thereof.
Those of skill in the ent of infectious diseases will be able to determine the effective amount from the test results presented hereinafter. In general it is plated that an effective daily amount would be from 0.01 mg/kg to 50 mg/kg body weight, more preferably from 0.1 mg/kg to 10 mg/kg body weight. It may be appropriate to administer the required dose as two, three, four or more sub-doses at appropriate intervals throughout the day. Said sub-doses may be formulated as unit dosage forms, for e, containing 1 to 1000 mg, and in particular 5 to 200 mg of active ingredient per unit dosage form.
The exact dosage and frequency of administration depends on the particular compound of a (I) used, the particular condition being treated, the severity of the condition being treated, the age, weight and general physical condition of the particular patient as well as other tion the individual may be taking, as is well known to those skilled in the art. Furthermore, it is evident that the effective amount may be lowered or increased ing on the response of the treated subject and/or ing on the tion of the physician prescribing the compounds of the instant invention. The effective amount ranges mentioned above are therefore only guidelines and are not intended to limit the scope or use of the invention to any extent.
Experimental Section Scheme 1. Overall reaction scheme HOAR N H o\ 1N NaOH / |NYNTO / | \Y T —> N TRIPHENYLPHOSPHINE RESIN, N / N O H gfg'osxfne’ DIAD, THF,RT,1h CI J R CI A B HO/\ N NH2 R1 N NH2 / I Y / | Y RJ NaH 60%, NMP, CI R 05°C, 30' then RT 2h O\R1 C D Compounds of type A in scheme 1 can be functionalized with alcohols using Mitsunobu ions in a polar aprotic solvent, for example THF. The cleavage of the methyl carbamate was performed under basic conditions in 1,4-dioxane to form intermediate C.
The displacement of the chlorine in C was performed with an alcohol and a base (e.g.
NaH) in a polar aprotic solvent (e.g. NMP) to form compounds of the type D.
Preparation of intermediate A H O O O O- —> \/ o +OZ/‘< H+ Na" /\ 0— MeOH, RT,16h HN POCI3 "N m o a lift HO NANJLO/ DIPEA, CH3CN, 70°C C. N’ m o/ oethoxycarbonylpyrrole hydrochloride (25.8 g, 135.3 mmol) was ioned between dichloromethane and sat. NaHC03. The organic layer was dried over MgSO4, the solids were removed via filtration, and the solvent of the filtrate evaporated to dryness. The residue was dissolved in methanol (500 mL) together with 1,3- bis(methoxycarbonyl)methylthiopseudourea (32.1 g, 156 mmol) and acetic acid (39 mL, 677 mmol) and stirred 1 hour at room temperature. A precipitate appeared and stirring was continued overnight. Sodium methoxide (73.1 g, 1353 mmol) was added.
An exothermic reaction was observed and the reaction mixture was stirred overnight.
The mixture was t to pH 5 with acetic acid and the precipitate was isolated by filtration, triturated on the filter with water (2 x 350 mL), acetonitrile (350 mL) and diisopropylether (350 mL). The obtained methyl N-(4-hydroxy-5H-pyrrolo- [3,2-d]pyrimidinyl)carbamate was dried in the oven. methyl N-(4-hydroxy-5H-pyrrolo[3,2-d]pyrimidinyl)carbamate (25 g, 120 mmol) was dispensed in 350 mL acetonitrile in a 500 mL multi neck flask equipped with with an ad stirrer (300 rpm) at room temperature. POCI3 (22.1 mL, 238.2 mmol) was added and then the reaction e was heated to 70°C while ng.
Diisopropylethylamine (41.4 mL, 240.2 mmol) was added dropwise via a syringe pump at a flow of 0.2 mL/min.
The reaction mixture was cooled to room temperature and poured into a stirred solution of sodium acetate (78.8 g, 961 mmol) in water (500 mL) at 45°C. The organics were evaporated and the remaining liquid was stirred and cooled over an ice bath. The formed solid was isolated by filtration, washed with acetonitrile and triturated with diisopropylether to afford intermediate A, dried under . LC-MS m/z = 227 (M+H) Preparation of intermediate B Method 1.
HN DIAD ah \N 0 I + Cl / THF, rt 0/ 30min A B To a suspension of A (500 mg, 2.2 mmol), benzylalcohol (0.28 mL, 2.6 mmol) and triphenylphosphine (0.69 g, 2.6 mmol) in anhydrous THF (15 mL) was added DIAD (0.64 mL, 3.3 mmol) at room temperature. The on mixture was stirred at room temperature for 30 minutes. The mixture was concentrated under reduced pressure.
The product was purified via silica gel column tography using a heptanes to ethyl acetate gradient; 100-0 to 90-10. The product fractions were collected and concentrated under reduced pressure. The t was triturated in diisopropylether, isolated by filtration and dried under vacuum to afford B as a pale yellow solid. LC-MS m/z = 317 (M+H) Method 2 with resin bound triphenylphosphine.
To a suspension of A (700 mg, 3.1 mmol), benzylalcohol (0.39 mL, 3.7 mmol) and triphenylphosphine resin (2.6 g, 7.7 mmol) in ous THF (21 mL) was added DIAD (0.90 mL, 4.6 mmol) at room temperature. The reaction mixture was stirred at room temperature for 1h. The mixture was filtered over packed decalite and washed with methanol. The filtrate was trated in vacuo. The product was triturated in diisopropylether, isolated by filtration and dried under vacuum to afford a pale yellow solid, B. LC-MS m/z = 317 (M+H) Preparation of intermediate C Q/ ~ 1N NaOH \N O Q/NI I 1 ,4-dioxane, 60°C, 5h CI NAN H 2 CI NANJLO/ B (738 mg, 2.3 mmol) was dissolved in 1,4-dioxane (11 mL) in a 50 mL glass tube and NaOH (5.6 mL, 1N a.q.) was added. The mixture was heated to 60°C for 5h. The mixture was cooled and concentrated in vacuo. The residue was treated with water and the precipitate was ed by filtration and dried under vacuum to afford C as a solid.
The product was used as such in the next step. LC-MS m/z = 259 (M+H) Preparation of 1 and 2 Method 1.
Q/ ~ HCI 4N in Dioxane N HO/\/\ I MW120°C 10min—>N‘|\J\Q/ C N/)\NH2 c 1 Intermediate C (240 mg, 0.93 mmol), n-butylalcohol (3.2 mL, 35 mmol), and 4N HCI in dioxane (0.46 mL, 1.9 mmol) was placed into a 7 mL microwave vial. The vial was sealed and the mixture was heated in the microwave at 120°C for 10 minutes. The mixture was cooled and concentrated in vacuo. The residue was neutralized with sat.
NaHC03 solution and extracted with dichloromethane. The c layer was separated, dried (MgSO4), the solids were removed by filtration and the filtrate was concentrated under reduced pressure. The t was purified via silica gel column chromatography using a dichloromethane-methanol;100-0 to 95-5 gradient. The best fractions were collected and trated under d pressure. The product was triturated in diisopropylether and the solid was isolated by filtration and dried under vacuum to afford 1 as a white solid.
Method 2.
N, \O/\/N‘ \o/\/ \ N NaH 60% \N \ )\ I\ | N/ NH2 —’ + N\ A 0 O N c1 NMP, 06°C, 30' then RT2h ""2 OI \ Intermediate C2 (250 mg, 1.1 mmol), and 3-hydroxymethylmethylisoxazole (0.16 mL, 1.65 mmol) were dissolved in NMP (3 mL) in a 7 mL vial. The mixture was cooled on a ice bath and NaH (66 mg, 1.65 mmol, 60% dispersion in mineral oil) was added under N2 and the mixture was stirred at 0-5 °C for 30 s and then allowed to warm to room ature and continued stirring for 2h. Then crude reaction mixture was purified by preparatory HPLC (Stationary phase: RP Vydac Denali C18 10 pm, 200 g, 5 cm), mobile phase: 0.25% NH4OAc solution in water, CH3CN), the desired fractions were collected and concentrated in vacuo. The product was crystallized from CH3CN, isolated by filtration and dried under vacuum to afford a white solid, 2.
Table 1. Compounds of formula (I) and corresponding analytical data. Compounds were prepared according to the methods described in the experimental section.
LC Method, LC-MS Mass # URE 1 H NMR Rt (min). Found (M+H) 1H NMR (400 MHz, DMSO-de) 5 ppm 0.85 (t, J=7.37 Hz, 3 H) < 2 - 1.26 (dq, J=15.02, 7.39 Hz, 2 H) 1.56 - 1.63 (m, 2 H) 4.30 (t, AHZ J=6.38 Hz, 2 H) 5.39 (s, 2 H) B, 1.98 297 .72 (s, 2 H) 6.08 (d, J=3.08 Hz, 1 H) 7.03 - 7.08 (m, 2 H) 7.19 - 7.25 (m, 1 H) 7.26 - 7.32 (m, 2 H) 7.48 (d, J=3.08 Hz, 1 H) 1H NMR (400 MHz, DMSO- d6) 5 ppm 2.41 (d, J=0.66 Hz, 3 H) \N 3.17 (s, 3 H) 3.57 (t, J=5.50 Hz, AHZ 2 H) 4.29 (t, J=5.50 Hz, 2 H) 5.50 A, 0.69 304 (s, 2 H) 5.82 (s, 2 H) 6.03 (d, J=2.86 Hz, 1 H) 6.37 (d, J=0.88 Hz, 1 H)7.35 (d, J=2.86 Hz, 1 H) 1H NMR (400 MHz, DMSO- d6) 5 q: ppm 2.33 - 2.38 (m, 3 H) 3.79 (s, 3H)534(s 2H)538(s 2H) INJ‘NHz 5.7,5(s1H)586(s 2H)6.12 (d, J:3.0,8Hz 1 - 6.47 B, 1.62 366 (m, 1 H) 6.78 (td, J=7.48, 0.66 Hz, 1 H)7.00 (d, J=7.92 Hz, 1 H) 7.24 (td, J=7.80, 1.80 Hz, 1 H) 7.43 (d, J=2.86 Hz, 1 H) -1 2- LC Method, LC-MS Mass # STRUCTURE 1H NMR Rt (min) Found (M+H) 1H NMR (400 MHz, DMSO-de) 5 ppm 0.77 (t, J=7.4 Hz, 3 H), 1.12 / (dq, J=15.0, 7.4 Hz, 2 H), 1.40 - \N / 1.50 (m, 2 H), 4.21 (t, J=6.4 Hz, | 2 H), 5.49 (s, 2 4 9 AH, (s, 2 H), 5.73 6 A, 0.81 298 f H), 6.11 (d, J—2.9 Hz, 1 H), 6.65 (d, J=7.9 Hz, 1 H), 7.21 - 7.28 (m, 1 H), 7.47 (d, J=3.1 Hz, 1 H), 7.69 (td, J=7.7, 1.8 Hz, 1 H), 8.47 - 8.53 (m, 1 H) 1H NMR (400 MHz, DMSO-de) 5 ppm 2.35 (s, 3 H) 5.37 (s, 2 H) / 5.47 (s, 2 H) 5.84 - 5.90 (m, 3 H) N 6.14 (d, J=2.86 Hz, 1 H) 6.72 (d, J=7.92 Hz, 1 H) 7.24 (dd, B, 1.29 337 _, J=6.93, 4.95 Hz, 1 H) 7.52 (d, J=3.08 Hz, 1 H) 7.65 (td, J=7.70, 1.76 Hz, 1 H) 8.47 (d, J=4.18 Hz, 1 H) 1H NMR (400 MHz, DMSO-de) 5 ppm 2.57 (s, 3 H) 5.45 (s, 2 H) / 5.51 (s, 2 H) 5.85 (s, 2 H) 6.13 N (d, J=2.86 Hz, 1 H) 6.85 (d, J=7.70 Hz, 1 H) 7.22 (dd, B, 1.14 338 >=N , 5.06 Hz, 1 H) 7.52 (d, J=3.08 Hz, 1 H) 7.64 (td, J=7.65, 1.65 Hz, 1 H) 8.43 (d, J=4.18 Hz, 1 H) LC Method, LC-MS Mass # URE 1 H NMR Rt (min) Found (M+H) 1H NMR (400 MHz, DMSO-ds) 5 /0 ppm 2.36 (s, 3 H) 3.74 (s, 3 H) C," 3.71 ‘ (s, 3 H)5.29 (s, 2 H)5.40 7 "39% (s, 2 H) 5.85 (s, 2 H) 5.93 (s, 1 B 1.45 397 0 HM H) 6.12 (d, J=3.08 Hz, 1 H) 6.29 02k (d, J=7.92 Hz, 1 H)7.11 (d, J=7.92 Hz, 1 H) 7.50 (d, J=3.08 Hz, 1 H) 1H NMR (400 MHz, DMSO- d6) 5 q _ ppm 2.36 (s, 3 H) 3.80 (s, 3 H) .31 o\ \N (s, 2 H) 5.48 (s, 2 H) 5.75 - 8 o N/ H: 5.81 (m, 3 H) 6.07 (d, J=2.86 Hz, A, 0.72 367 07k 1 H) 7.25 (dd, J=8.25, 4.73 Hz, 1 H) 7.36 - 7.41 (m, 2 H) 7.90 (dd, J=4.73, 0.99 Hz, 1 H) 1H NMR (400 MHz, DMSO-ds) 5 ppm 2.23 (d, J=1.10 Hz, 3 H) N 3.77 (s, 3 H) 5.32 (s, 2 H) 5.45 N‘ (s,2 H)5.77 (s,2 H)6.07 (d, 9 A; | :" J=2.86 Hz, 1 H)6.79 (d,J=1.10 B, 1.26 367 yr» N "2 Hz, 1 H)7.21 (dd, J=8.25, 4.73 Hz, 1 H) 7.33 (dd, J=8.36, 1.32 Hz, 1 H)7.37 (d, J=2.86 Hz, 1 H) 7.88 (dd, J=4.73, 1.21 Hz, 1 H) 1H NMR (400 MHz, DMSO-ds) 5 N ppm 2.34 - 2.41 (m, 3 H) 5.49 (s, \ I ‘ 2 H) 5.58 (s, 2 H) 5.88 (s, 2 H) N | l 6.15(d,J=2.86 Hz, 1 H)6.72 (d, B, 1.28 353 V0 / N Hz J=7.92 Hz, 1 H)7.20- 7.25 (m, 1 H) 7.43 (d, J=1.10 Hz, 1 H) 7.52 (d, J=2.86 Hz, 1 H) 7.63 (td, J=7.70, 1.76 Hz, 1 H) 8.46 (dd, WO 07082 LC Method, LC-MS Mass # STRUCTURE 1 H NMR Rt (min). Found (M+H) J=4.73, 0.77 Hz, 1 H) 1H NMR (400 MHz, DMSO-de) 5 ppm 2.24 (s, 3 H) 5.39 (s, 2 H) //N 5.43 (s, 2 H) 5.85 (s, 2 H) 6.13 \ ‘ (d, J=2.86 Hz, 1 H) 6.76 (d, N | J=7.70 Hz, 1 H)6.81 (s, 1 H) B, 1.18 337 Sj/N) / N Hz 7.21 (dd, J=6.93, 5.17 Hz,1 H) 7.52 (d, J=2.86 Hz, 1 H) 7.62 (td, J=7.65, 1.43 Hz, 1 H) 8.40 - 8.45 (m, 1 H) Analytical Methods.
LCMS General Procedure The High Performance Liquid Chromatography (HPLC) measurement was performed using a LC pump, a diode-array (DAD) or a UV detector and a column as specified in the respective methods. If necessary, onal detectors were included (see table of methods below).
Flow from the column was brought to the Mass Spectrometer (MS) which was configured with an atmospheric pressure ion source. It is within the knowledge of the d person to set the tune parameters (e.g. scanning range, dwell time...) in order to obtain ions allowing the identification of the compounds nominal monoisotopic molecular weight (MW). Data acquisition was performed with appropriate re.
Compounds are described by their experimental retention times (Rt) and ions. If not ied differently in the table of data, the reported molecular ion corresponds to the [M+H]+ (protonated molecule) and/or [M-H]— (deprotonated molecule). In case the compound was not directly ionizable the type of adduct is ied (i.e. [M+NH4]+, [M+HCOO]—, etc...). For molecules with multiple isotopic patterns (Br, Cl..), the reported value is the one ed for the lowest isotope mass. All results were obtained with mental uncertainties that are commonly associated with the method used.
Hereinafter, "SQD" means Single Quadrupole Detector, "MSD" Mass Selective Detector, "RT" room temperature, "BEH" bridged ethylsiloxane/silica hybrid, "DAD" Diode Array or, "HSS" High Strength silica., "Q-Tof’ Quadrupole f-flight mass spectrometers, "CLND", uminescent Nitrogen Detector, "ELSD" ative Light Scanning Detector, LC-MS Method codes (Flow sed in mL/min; column temperature (Col T) in °C; Run time in s).
Instrument Column Mobile phase Gradient A: 10mM Waters: Waters: CH3COONH4 From 95% A Acquity® BEH C18 in 95% H20 + to 5% A in UPLC®-DAD (1.7um, 5% CH3CN 1.3 min, held and SQD 2.1*50mm) for 0.7 min.
B: CH3CN A: 10mM From 100% Waters: Waters: CH3COONH4 Ato 5% A in Acquity® HSS T3 in 95% H20 + 2.10min, to UPLC®-DAD (1.8um, 5% CH3CN 0% A in 0.90 and SQD 2.1*100mm) min, to 5% A B: CH3CN in 0.5min Biolo ical t of com ounds of formula I and II Description of Biological Assays Assessment of TLR7 and TLR8 activity The ability of compounds to activate human TLR7 and/or TLR8 was assessed in a cellular reporter assay using HEK293 cells transiently transfected with a TLR7 or TLR8 expression vector and NFKB-IUC reporter construct.
Briefly, HEK293 cells were grown in culture medium (DMEM supplemented with 10% FCS and 2 mM Glutamine). For transfection of cells in 15 cm dishes, cells were detached with Trypsin-EDTA, transfected with a mix of CMV-TLR7 or TLR8 plasmid WO 07082 (1700 ng), NFKB-IUC plasmid (850 ng) and a transfection reagent and incubated for 48 h at 37°C in a humidified 5% C02 atmosphere. Transfected cells were then washed in PBS, detached with Trypsin-EDTA and resuspended in medium to a density of 1.25 X 105 cells/mL. Forty microliters of cells were then dispensed into each well in ll plates, where 200 nL of compound in 100% DMSO was already present. Following 6 hours incubation at 37°C, 5% C02, the luciferase activity was determined by adding 15 uL of Steady Lite Plus substrate (Perkin Elmer) to each well and readout performed on a ViewLux ultraHTS microplate imager (Perkin Elmer). Dose response curves were generated from measurements performed in quadruplicates. Lowest effective concentrations (LEC) values, defined as the concentration that induces an effect which is at least two fold above the standard ion of the assay, were determined for each compound.
Compound toxicity was determined in el using a similar dilution series of compound with 40 uL per well of cells transfected with the R7 construct alone (1.25 x 105 cells/mL), in 384-well plates. Cell viability was measured after 6 hours incubation at 37°C, 5% C02 by adding 15 uL of ATP lite (Perkin Elmer) per well and reading on a ViewLux ultraHTS microplate imager (Perkin Elmer). Data was reported as 0050.
In parallel, a similar dilution series of compound was used (200 nL of compound in 100% DMSO) with 40 uL per well of cells transfected with NFkB-luc reporter construct alone (1.25 x 105 cells/mL). Six hours after incubation at 37°C, 5% C02, the luciferase activity was determined by adding 15 uL of Steady Lite Plus substrate (Perkin Elmer) to each well and readout performed on a ViewLux ultraHTS microplate imager (Perkin Elmer). Counterscreen data is ed as LEC.
Activation of ISRE er elements The potential of compounds to induce lFN-l was also evaluated by measuring the activation of interferon-stimulated responsive elements (ISRE) by conditioned media from PBMC. The ISRE element of sequence GAAACTGAAACT is highly responsive to the STAT1-STAT2-IRF9 transcription factor, activated upon binding of lFN-l to their receptor IFNAR ech, PT3372-5W). The d plSRE-Luc from Clontech (ref. 631913) contains 5 copies of this ISRE element, ed by the firefly luciferase ORF.
A HEK293 cell line stably transfected with plSRE-Luc (HEK-lSREluc) was established to e the conditioned PBMC cell culture media.
Briefly, PBMCs were ed from buffy coats of at least two donors using a standard Ficoll centrifugation protocol. lsolated PBMCs were resuspended in RPMI medium supplemented with 10% human AB serum and 2 x 105 cells/well were dispensed into 384-well plates containing compounds (70 uL total volume). After overnight incubation, uL of supernatant was transferred to 384-well plates containing 5 X 103 HEK-lSREluc cells/well in 30 uL (plated the day before). Following 24 hours of incubation, activation of the ISRE elements was measured by assaying luciferase activity using 40 uL/well Steady Lite Plus substrate (Perkin Elmer) and measured with ViewLuX TS microplate imager (Perkin Elmer). The ating activity of each compound on the HEK-lSREluc cells was reported as LEC value, defined as the compound concentration applied to the PBMCs resulting in a luciferase activity at least two fold above the rd deviation of the assay. The LEC in turn indicates the degree of ISRE activation on transfer of a d amount of PBMC culture medium. inant interferon d-2a (Roferon-A) was used as a standard control compound.
Table 2. Activity of compounds of formula (I).
All compounds demonstrated a CC50 >24uM.
Human TLR 7 (LEC) Human TLR 8 HEK-ISRE luc uM (LEC) uM (LEC) uM 1 0.6 >25 0.4 2 2.7 >25 0.5 3 0.1 >25 0.03 4 1.4 >25 0.6 0.4 >25 0.1 6 3.9 >25 2 7 0.08 >25 0.03 8 0.03 >25 0.01 9 0.07 >25 NA WO 07082 Human TLR 7 (LEC) Human TLR 8 HEK-ISRE luc "M (LEC) "M (LEC) "M 0.5 >25 NA 11 0.6 >25 NA NA = not available

Claims (12)

Claims
1. A compound of formula (I) H2N N N N O R3 R4 (I) and their pharmaceutically able salt, e or polymorph thereof wherein 5 R1 is H, fluorine or methyl; R2 is H, halogen or C1-3 alkyl; R3 is C1-6 alkyl optionally substituted by one or more substituents ndently selected from aryloxy, heterocycle, halogen, aryl, alkylamino, dialkylamino, C1-6 alkyl, carboxylic acid, carboxylic ester, carboxylic amide, nitrile, or C1-6 ; 10 or wherein R3 is an alkylaryl optionally substituted by one or more tuents independently selected from halogen, aryloxy, aryl, alkylamino, dialkylamino, C1-6 alkyl, carboxylic acid, carboxylic ester, carboxylic amide, sulfonamide, nitrile, or C1-6 alkoxy; R4 is C1-6 alkyl optionally substituted by one or more substituents independently 15 selected from hydroxyl, C1-6 alkyl, C3-7 cycloalkyl, C2-6 alkenyl or aryl optionally further substituted by C1-6 alkyl, and C3-7 cycloalkyl ally further substituted by C1-6 alkyl; or wherein R4 is an alkylaryl optionally substituted by one or more substituents independently 20 selected from halogen, aryloxy, aryl, alkylamino, dialkylamino, C1-6 alkyl, carboxylic acid, carboxylic ester, ylic amide, sulfonamide, e, or C1-6 alkoxy, with the proviso that the compound does not satisfy any one of the following structural formulae: , ,and .
2. A compound according to claim 1 wherein R3 is a CH2-aryl group (substituted or unsubstituted), and R1, R2, and R4 are described as in claim 1. 5
3. A compound of a (I) wherein R3 and R4 are both CH2-aryl groups optionally further substituted as described in claim 1, and n R1, and R2 are as described in claim1.
4. A compound according to claim 1 wherein R1 is fluorine, R2 is en, and R3 and 10 R4 are described as in claim 1.
5. A compound according to claim 1 having the following chemical structure:
6.(II). 15 6. A compound according to claim 1, wherein the compound satisfies any one of the following formulae: , , , , ,, , , , and . 5
7. A pharmaceutical composition comprising a compound of formula (I) or (II) or a ceutically acceptable salt, solvate or polymorph thereof according to claim 1, 5 or 6 er with one or more pharmaceutically acceptable excipients, diluents or carriers. 10
8. A compound of a (I) or (II) or a pharmaceutically acceptable salt, solvate or polymorph thereof according to claim 1, 5 or 6, or a pharmaceutical composition comprising said compound of formula (I) or (II) or a pharmaceutically acceptable salt, solvate or rph thereof according to claim 7 for use as a medicament. 15
9. Use of a compound of formula (I) or (II) or a ceutically acceptable salt, solvate or polymorph thereof according to claim 1, 5 or 6 or said pharmaceutical composition comprising said compound of formula (I) or (II) or a pharmaceutically acceptable salt, solvate or polymorph thereof according to claim 7 in the manufacture of a ment for treatment of any disorder in which the modulation of TLR7 is involved.
10. The use of a compound of formula (I) H2N N N N O R3 5 R4 (I) and their pharmaceutically able salt, solvate or polymorph thereof wherein R1 is H, ne or methyl; R2 is H, halogen or C1-3 alkyl; R3 is C1-6 alkyl optionally substituted by one or more substituents independently selected 10 from y, heterocycle, halogen, aryl, alkylamino, dialkylamino, C1-6 alkyl, carboxylic acid, carboxylic ester, carboxylic amide, nitrile, or C1-6 ; or wherein R3 is an ryl ally substituted by one or more substituents independently selected from halogen, aryloxy, aryl, alkylamino, dialkylamino, C1-6 alkyl, ylic acid, 15 carboxylic ester, carboxylic amide, sulfonamide, nitrile, or C 1-6 alkoxy; R4 is C1-6 alkyl optionally substituted by one or more substituents independently selected from hydroxyl, C1-6 alkyl, C3-7 cycloalkyl, C2-6 alkenyl or aryl optionally further substituted by C1-6 alkyl, and C3-7 cycloalkyl optionally further substituted by C1-6 alkyl; or wherein R4 is an alkylaryl optionally substituted by one or more substituents 20 independently selected from halogen, y, aryl, alkylamino, dialkylamino, C1-6 alkyl, carboxylic acid, carboxylic ester, carboxylic amide, sulfonamide, nitrile, or C1-6 alkoxy, in the manufacture of a medicament for the treatment of a disorder in which the modulation of TLR7 is involved. 25
11. The compound of claim 1, substantially as herein described with reference to any one of the Examples thereof.
12. The use of claim 10, ntially as herein described with reference to any one of the Examples thereof.
NZ714267A 2013-06-27 2014-06-26 Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases NZ714267B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
NZ754320A NZ754320B2 (en) 2014-06-26 Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP13174108.4 2013-06-27
EP13174108 2013-06-27
PCT/EP2014/063467 WO2014207082A1 (en) 2013-06-27 2014-06-26 Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases

Publications (2)

Publication Number Publication Date
NZ714267A NZ714267A (en) 2021-06-25
NZ714267B2 true NZ714267B2 (en) 2021-09-28

Family

ID=

Similar Documents

Publication Publication Date Title
AU2018274911B2 (en) Pyrrolo[3,2-c]Pyrimidine derivatives for the treatment of viral infections and other diseases
AU2013328732B2 (en) Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
DK2812331T3 (en) PIPERIDINOPYRIMIDINE DERIVATIVES FOR TREATING VIRUS INFECTIONS
US9284304B2 (en) Substituted pyrimidines as toll-like receptor modulators
AU2013326579B2 (en) 1,2,4-triazine derivatives for the treatment of viral infections.
CA3027471A1 (en) Dihydropyranopyrimidines for the treatment of viral infections
JP2015537020A (en) Heterocyclic substituted 2-amino-quinazoline derivatives for the treatment of viral infections
AU2014270418B2 (en) Pyridone derivatives for the treatment of viral infections and further diseases
NZ714267B2 (en) Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
NZ754320B2 (en) Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
EA040150B1 (en) METHOD FOR OBTAINING PYRROLO[3,2-D]PYRIMIDINE DERIVATIVES
NZ713679B2 (en) Pyridone derivatives for the treatment of viral infections and further diseases