Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SUBSTITUDED BENZYLOXY-PHENYLMETHYLUREA DERIVATIVES
Document Type and Number:
WIPO Patent Application WO/2007/080109
Kind Code:
A1
Abstract:
The present invention relates to novel substituted benzyloxy-phenylmethylurea derivatives, processes for their preparation, and their use in medicaments, especially for the prophylaxis and treatment of diseases associated with Cold Menthol Receptor 1 (CMR-1) activity, in particular for the treatment of urological diseases or disorders, such as detrusor overactivity (overactive bladder), urinary incontinence, neurogenic detrusor oeractivity (detrusor hyperflexia), idiopathic detrusor overactivity (detrusor instability), benign prostatic hyperplasia, and lower urinary tract symptoms; chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, and inflammatory disorders such as asthma and chronic obstructive pulmonary (or airways) disease (COPD).

Inventors:
LAMPE THOMAS (DE)
ALONSO-ALIJA CRISTINA (DE)
BECK HARTMUT (DE)
ROSENTRETER ULRICH (DE)
SANDNER PETER (DE)
STAHL ELKE (DE)
STELTE-LUDWIG BEATRIX (DE)
Application Number:
PCT/EP2007/000192
Publication Date:
July 19, 2007
Filing Date:
January 11, 2007
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BAYER HEALTHCARE AG (DE)
LAMPE THOMAS (DE)
ALONSO-ALIJA CRISTINA (DE)
BECK HARTMUT (DE)
ROSENTRETER ULRICH (DE)
SANDNER PETER (DE)
STAHL ELKE (DE)
STELTE-LUDWIG BEATRIX (DE)
International Classes:
A61K31/17; A61K31/381; A61P13/00; C07C275/24; C07C275/26; C07C275/28; C07C275/30; C07D333/36
Domestic Patent References:
WO2006040136A12006-04-20
Other References:
D'AGOSTINO G ET AL: "An appraisal or recently patented compounds for bladder overactivity and urinary incontinence", EXPERT OPINION ON THERAPEUTIC PATENTS, ASHLEY PUBLICATIONS, GB, vol. 14, no. 7, 2004, pages 1041 - 1060, XP002408785, ISSN: 1354-3776
Attorney, Agent or Firm:
BAYER HEALTHCARE AG (Patents and Licensing, Leverkusen, DE)
Download PDF:
Claims:

We claim

1. A compound of the general formula (I)

wherein

R represents hydrogen or halogen,

R 2 represents hydrogen or halogen,

R 3 represents hydrogen or halogen,

R 4 represents chlorine, fluorine, nitro, trifluoromethyl, trifluoromethoxy, Ci-C 4 -alkyl or Ci-C 4 -alkoxy,

R represents hydrogen or halogen,

R 6 represents C 3 -C 8 -alkyl, C 3 -C 7 -cycloalkyl, C 6 -C| 0 -aryl, 5- to 10-membered heteroaryl or a group of the formula -Y-R 9 ,

wherein cycloalkyl can be further substituted with one to three identical or different radicals selected from the group consisting of trifluoromethyl and Ci-Q-alkyl,

and

wherein aryl and heteroaryl can be further substituted with one to three identical or different radicals selected from the group consisting of halogen, nitro, amino, hydroxy, trifluoromethyl, C,-C 6 -alkyl, Ci-C 6 -alkoxy and Ci-Cδ-alkylamino,

and

wherein

Y represents Ci-C 4 -alkandiyl,

R 9 represents C 3 -C 7 -cycloalkyl, phenyl or 5- to 10-membered heteroaryl,

wherein cycloalkyl can be further substituted with one to three identical or different radicals selected from the group consisting of trifluoromethyl and C r C 4 -alkyl,

and

wherein phenyl and heteroaryl can be further substituted with one to three identical or different radicals selected from the group consisting of halogen, nitro, amino, hydroxy, trifluoromethyl, Q-

C 6 -alkyl, Q-C ό -alkoxy and Ci-C 6 -alkylamino,

R 7 represents C|-C 6 -allcyl,

wherein alkyl is further substituted with one radical selected from the group consisting of amino, mono-alkylamino, Ci-Q-alkylcarbonylamino or Q-C 4 - alkoxycarbonylamino,

R 8 represents hydrogen or C|-C 4 -alkyl,

or one of its salts, hydrates and/or solvates.

2. A compound of general formula (I) according to Claim 1 , wherein

R 1 represents hydrogen or halogen,

R 2 represents hydrogen or halogen,

R 3 represents hydrogen,

R 4 represents chlorine, nitro, trifluoromethoxy, Ci-C 3 -alkyl or C|-C 3 -alkoxy,

R 5 represents hydrogen,

R 6 represents Cj-Cg-alkyl, C 3 -C 7 -cycloalkyl, phenyl, 5- or 6-membered heteroaryl or a group of the formula -Y-R 9 ,

wherein cycloalkyl can be further substituted with one to three identical or different radicals selected from the group consisting of trifluoromethyl and C,-C 4 -alkyl,

and

wherein phenyl and heteroaryl can be further substituted with one to three identical or different radicals selected from the group consisting of halogen, nitro, trifluoromethyl, Ci-C δ -alkyl and CpC δ -alkoxy,

and

wherein

Y represents Ci -C 4 -alkandiyl,

R 9 represents C 3 -C 7 -cycloalkyl, phenyl or 5- or 6-membered heteroaryl,

wherein cycloalkyl can be further substituted with one to three identical or different radicals selected from the group consisting of trifluoromethyl and Ci-C 4 -alkyl,

and

wherein phenyl and heteroaryl can be further substituted with one to three identical or different radicals selected from the group consisting of halogen, nitro, trifluoromethyl, C r C 6 -alkyl and C,-C 6 -alkoxy,

R 7 represents C|-C 3 -alkyl,

wherein alkyl is further substituted with one radical selected from the group consisting of amino, mono-alkylamino or Ci-C 4 -alkoxycarbonylamino,

R 8 represents hydrogen,

or one of its salts, hydrates and/or solvates.

3. A compound of general formula (I) according to Claim 1 or 2, wherein

R 1 represents hydrogen or fluorine,

R 2 represents hydrogen or fluorine,

R 3 represents hydrogen,

R 4 represents chlorine or methoxy,

R 5 represents hydrogen,

R 6 represents C 3 -C 6 -allcyl, C 3 -C 6 -cycloalkyl, phenyl, thienyl or a group of the formula

-Y-R 9 ,

wherein cycloalkyl can be further substituted with one trifluoromethyl group,

and

wherein phenyl and thienyl can be further substituted with one to three identical or different radicals selected from the group consisting of fluorine, chlorine, nitro, trifluoromethyl, methyl and methoxy,

and

wherein

Y represents methylen or ethan- 1 , 1 -diyl,

R 9 represents phenyl,

wherein phenyl can be further substituted with one to three identical or different radicals selected from the group consisting of fluorine, chlorine, nitro, trifluoromethyl, methyl and methoxy,

R 7 represents C,-C 2 -alkyl,

wherein alkyl is further substituted with one radical selected from the group consisting of amino or tert-butoxycarbonylamino,

R 8 represents hydrogen,

or one of its salts, hydrates and/or solvates.

Process for synthesizing a compound of general formula (I) according to Claim 1 , by

[A] condensing a compound of general formula (II)

wherein R , R , R , R , R , R and R have the meaning indicated in Claim 1 ,

with a compound of general formula (III)

R-N= -=n (HI).

wherein R 6 has the meaning indicated in Claim 1 ,

or

[B] condensing a compound of general formula (II) with a mixture of a carbonic acid derivative and a compound of general formula (IV)

R 6 \

NH. (IV) 1

wherein R 6 has the meaning indicated in Claim

or

[C] condensing a compound of general formula (V)

wherein R , R , R , R and R have the meaning indicated in Claim 1 ,

with a compound of general formula (VI)

wherein R 1 , R 2 and R 3 have the meaning indicated in Claim 1, and

X 1 represents a leaving group, such as halogen, preferably chlorine or bromine,

in the presence of a base.

5. A compound of general formula (I) according to Claim 1, 2 or 3 for the treatment of diseases or disorders.

6. Use of a compound of general formula (I) according to Claim 1, 2 or 3 for the preparation of medicaments.

7. Use according to Claim 6 for the preparation of medicaments for the treatment of urological diseases or disorders.

8. The composition containing at least one compound of general formula (I) according to Claim 1 , 2 or 3 and a pharmacologically acceptable diluent.

9. A composition according to Claim 8 for the treatment of urological diseases or disorders.

10. The process for the preparation of compositions according to Claim 8 and 9 characterized in that the compounds of general formula (I) according to Claim 1 , 2 or 3 together with customary auxiliaries are brought into a suitable application form.

A. CLASSIFlCATION Of SUB JECT MATTER ,

INV. A61K31/17 A61K31/381 A61P13/00 C07D333/36 C07C275/24 C07C275/26 C07C275/28 C07C275/30

According to International Patent Classification (IPC) or to both national classification and IPC

B. FIELDS SEARCHED

Minimum documentation searched (classification system followed by classification symbols)

C07C A61K A61P C07D

Documentation searched other than minimum documentation to the extent that such documents are included in the fields searched

Electronic data base consulted during the international search (name of data base and, where practical, search terms used)

EPO-Internal , BEILSTEIN Data, CHEM ABS Data

C. DOCUMENTS CONSIDERED TO BE RELEVANT

Category* Citation of document, with indication, where appropriate, of the relevant passages Relevant to claim No

D'AGOSTINO G ET AL: "An appraisal or 1 , 6 recently patented compounds for bladder overactivity and urinary incontinence"

EXPERT OPINION ON THERAPEUTIC PATENTS,

ASHLEY PUBLICATIONS, GB, vol. 14, no. 7, 2004, pages 1041-1060,

XP002408785

ISSN: 1354-3776 the whole document

P 5 X WO 2006/040136 A (BAYER HEALTHCARE AG 1-10 [DE]; LAMPE THOMAS [DE]; ALONSO-ALIJA CRISTINA [DE) 20 April 2006 (2006-04-20) page 1, lines 2-10; claim 1 and examples

D Further documents are listed in the continuation of Box C See patent family annex

* Special categories of cited documents

"T" later document published after the International filing date or priority date and not in conflict with the application but

"A" document defining the general state of the art which is not cited to understand the principle or theory underlying the considered to be of particular relevance invention

1 E" earlier document but published on or after the international 1 X" document of particular relevance, the claimed invention filing date cannot be considered novel or cannot be considered to

1 L" document which may throw doubts on pnoπty claιm(s) or involve an inventive step when the document is taken alone which is cited to establish the publication date of another 1 Y" document of particular relevance, the claimed invention citation or other special reason (as specified) cannot be considered to involve an inventive step when the

"O" document referring to an oral disclosure, use, exhibition or document is combined with one or more other such docuother means ments, such combination being obvious to a person skilled

"P" document published prior to the international filing date but In the art later than the priority date claimed '&' document member of the same patent family

Date of the actual completion of the international search Date of mailing of the international search report

21 May 2007 30/05/2007

Name and mailing address of the ISA/ Authorized officer

European Patent Office, P B 5818 Patentlaan 2 NL - 2280 HV Rijswijk TeI (+31-70) 340-2040, Tx 31 651 epo nl, Fax (+31-70) 340-3016 MATES VALDIVIELSO, J

Fonm PCT/ISA/210 (second sheet) (April 2005)

Patent document Publication Patent family Publication cited in search report date member(s) date

WO 2006040136 A 20-04-2006 NONE

Form PCT/ISA/210 (patent family annex) (April 2005)

Description:

Substituted benzyloxy-phenylmethylurea derivatives

The present invention relates to novel substituted benzyloxy-phenylmethylurea derivatives, processes for their preparation, and their use in medicaments, especially for the prophylaxis and treatment of diseases associated with Cold Menthol Receptor 1 (CMR-I) activity, in particular for the treatment of urological diseases or disorders, such as detrusor overactivity (overactive bladder), urinary incontinence, neurogenic detrusor overactivity (detrusor hyperflexia), idiopathic detrusor overactivity (detrusor instability), benign prostatic hyperplasia, and lower urinary tract symptoms; chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, and inflammatory disorders such as asthma and chronic obstructive pulmonary (or airways) disease (COPD).

There is abundant direct or indirect evidence that shows the relation between Transient Receptor Potential (TRP) channel activity and diseases such as pain, ischaemia, and inflammatory disorders. Further, it has been demonstrated that TRP channels transduce reflex signals that are involved in the overactive bladder of patients who have damaged or abnormal spinal reflex pathways [De Groat WC: A neurologic basis for the overactive bladder. Urology 50 (6A Suppl): 36-52, 1997]. CMR-I , a nonselective cation channel is such a member of the TRP channel family (TRPM8). Recently, in 2002 the receptor was cloned and it was found to be sensitive to cold temperature and menthol and therefore named as cold menthol receptor - 1 (CMR-I) (McKemy et al, 2002; Peier et al., 2002). This receptor which is activated by 8 - 28°C temperature is expressed on the bladder urothelium and DRG (Dorsal Root Ganglia) and C-fibers. The intravesical ice water or menthol also induce C-fiber mediated spinal micturition reflex in patients with urgency and urinary incontinence (UI). Clinically CMR-I is supposed to mediate the bladder cooling reflex seen after ice water test in overactive patients.

Therefore antagonism of the CMR-I receptor leads to the blockage of neurotransmitter release, resulting in prophylaxis and treatment of the conditions and diseases associated with CMR-I activity.

Antagonists of the CMR-I receptor can be used for prophylaxis and treatment of the conditions and diseases including chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, inflammatory disorders, urinary incontinence (Ul) such as urge urinary incontinence (UUI), and/or overactive bladder, Lower urinary tract symptoms secondary to or independent of benign prostatic hyperplasia.

UI is the involuntary loss of urine. UUl is one of the most common types of UI together with stress urinary incontinence (SUI) which is usually caused by a defect in the urethral closure mechanism. UUl is often associated with neurological disorders or diseases causing neuronal damages such as dementia, Parkinson's disease, multiple sclerosis, stroke and diabetes, although it also occurs in individuals with no such disorders. One of the usual causes of UUI is overactive bladder (OAB) which is a medical condition referring to the symptoms of frequency and urgency derived from abnormal contractions and instability of the detrusor muscle.

There are several medications for urinary incontinence on the market today mainly to help treating UUI. Therapy for OAB is focused on drugs that affect peripheral neural control mechanisms or those that act directly on bladder detrusor smooth muscle contraction, with a major emphasis on development of anticholinergic agents. These agents can inhibit the parasympathetic nerves which control bladder voiding or can exert a direct spasmolytic effect on the detrusor muscle of the bladder. This results in a decrease in intravesicular pressure, an increase in capacity and a reduction in the frequency of bladder contraction. Orally active anticholinergic drugs which are commonly prescribed have serious drawbacks such as unacceptable side effects such as dry mouth, abnormal visions, constipation, and central nervous system disturbances. These side effects lead to poor compliance. Dry mouth symptoms alone are responsible for a 70% non-compliance rate with oxybutynin. The inadequacies of present therapies highlight the need for novel, efficacious, safe, orally available drugs that have fewer side effects.

In WO 03/037865 and Y. Lu, et al., Bioorg. Med. Chem. Lett. 2004, 14, 3957-3962 related benzyloxy-phenylmethylurea derivatives for the treatment of cancer and in WO 03/092670 for the treatment of female sexual dysfunction are described.

The present invention relates to compounds of the general formula (I)

wherein

R 1 represents hydrogen or halogen,

R 2 represents hydrogen or halogen,

R 3 represents hydrogen or halogen,

R 4 represents chlorine, fluorine, nitro, trifluoromethyl, trifluoromethoxy, C|-C 4 -alkyl or Ci-C 4 -alkoxy,

R 5 represents hydrogen or halogen,

R 6 represents C 3 -C 8 -alkyl, C 3 -C 7 -cycloalkyl, C 6 -Ci 0 -aryl, 5- to 10-membered heteroaryl or a group of the formula -Y-R 9 ,

wherein cycloalkyl can be further substituted with one to three identical or different radicals selected from the group consisting of trifluoromethyl and Ci-C 4 - alkyl,

and

wherein aryl and heteroaryl can be further substituted with one to three identical or different radicals selected from the group consisting of halogen, nitro, amino, hydroxy, trifluoromethyl, Ci-C 6 -alkyl, Ci-C 6 -alkoxy and C|-C 6 -alkylamino,

and

wherein

Y represents Ci -C 4 -alkandiyl,

R 9 represents C 3 -C 7 -cycloalkyl, phenyl or 5- to 10-membered heteroaryl,

wherein cycloalkyl can be further substituted with one to three identical or different radicals selected from the group consisting of trifluoromethyl and

Ci-C-alkyl,

and

wherein phenyl and heteroaryl can be further substituted with one to three identical or different radicals selected from the group consisting of halogen, nitro, amino, hydroxy, trifluoromethyl, C|-C 6 -alkyl, C|-C 6 -alkoxy and C]-C 6 -alkylamino,

R 7 represents C|-C 6 -alkyl,

wherein alkyl is further substituted with one radical selected from the group consisting of amino, mono-alkylamino, or Ci-C 4 -alkoxycarbonylamino,

R 8 represents hydrogen or C|-C 4 -alkyl,

and their salts, hydrates and/or solvates.

Physiologically acceptable salts are preferred in the context of the present invention.

Physiologically acceptable salts according to the invention are non-toxic salts which in general are accessible by reaction of the compounds (I) with an inorganic or organic base or acid conventionally used for this purpose. Non-limiting examples of pharmaceutically acceptable salts of compounds (I) include the alkali metal salts, e.g. lithium, potassium and sodium salts, the alkaline earth metal salts such as magnesium and calcium salts, the quaternary ammonium salts such as, for example, triethyl ammonium salts, acetates, benzene sulphonates, benzoates, dicarbonates, disulphates, ditartrates, borates, bromides, carbonates, chlorides, citrates, dihydrochlorides, fumarates, gluconates, glutamates, hexyl resorcinates, hydrobromides, hydrochlorides, hydroxy- naphthoates, iodides, isothionates, lactates, laurates, malates, maleates, mandelates, mesylates, methylbromides, methylnitrates, methylsulphates, nitrates, oleates, oxalates, palmitates, pantothenates, phosphates, diphosphates, polygalacturonates, salicylates, stearates, sulphates, succinates, tartrates, tosylates, valerates, and other salts used for medicinal purposes.

Hydrates of the compounds of the invention or their salts are stoichiometric compositions of the compounds with water, such as for example hemi-, mono-, or dihydrates.

Solvates of the compounds of the invention or their salts are stoichiometric compositions of the compounds with solvents.

The present invention includes both the individual enantiomers or diastereomers and the corresponding racemates or diastereomeric mixtures of the compounds according to the invention and their respective salts. In addition, all possible tautomeric forms of the compounds described above are included according to the present invention. The diastereomeric mixtures can be separated into the individual isomers by chromatographic processes. The racemates can be resolved into the respective enantiomers either by chromatographic processes on chiral phases or by resolution.

In the context of the present invention, the substituents, if not stated otherwise, in general have the following meaning:

Alkyl in general represents a straight-chain or branched saturated hydrocarbon radical having 1 to 6, preferably 1 to 4 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, iso- propyl, n-butyl, isobutyl, sec-butyl, tert-bulyl, pentyl, isopentyl, hexyl, isohexyl. The same applies to radicals such as alkoxy, alkylamino, alkylcarbonylamino, alkoxycarbonylamino and the like.

Alkandiyl in general represents a straight-chain or branched saturated alkandiyl radical having 1 to 4 carbon atoms. Non-limiting examples include methylen, ethan-l ,2-diyl, ethan-l .l-diyl, propan- 1 ,3-diyl, propan-l ,2-diyl, propan-2,2-diyl, butan-l ,4-diyl, butan-l ,3-diyl and butan-2,4-diyl.

Alkenyl in general represents a straight-chain or branched alkenyl radical having 2 to 6, preferably 2 to 4 carbon atoms. Non-limiting examples include vinyl, allyl, n-prop-1-en-l-yl, n-but-2-en-l-yl, 2-methylprop-l -en-1 -yl and 2-methylprop-2-en-l -yl.

Alkinyl in general represents a straight-chain or branched alkinyl radical having 2 to 6, preferably 2 to 4 carbon atoms. Non-limiting examples include ethinyl, propargyl (2-propinyl), 1-propinyl, but- 1-inyl, but-2-inyl.

Alkoxy illustratively and preferably represents methoxy, ethoxy, n-propoxy, isopropoxy, /er/-butoxy, n-pentoxy and n-hexoxy.

Alkylcarbonylamino in general represents a straight-chain or branched hydrocarbon radical having 1 to 6, preferably 1 to 4 carbon atoms which has a carbonylamino (-CO-NH-) function at the position of attachment and which is bonded to the carbonyl group. Non-limiting examples include formylamino, acetylamino, n-propionylamino, n-butyrylamino, isobutyrylamino, pivaloylamino, n- hexanoylamino.

Alkoxycarbonylamino illustratively and preferably represents methoxycarbonylamino, ethoxy- carbonylamino, n-propoxycarbonylamino, isopropoxycarbonylamino, tert-butoxycarbonylamino, n-pentoxycarbonylamino and n-hexoxycarbonylamino.

Alkylamino represents an alkylamino radical having one or two (independently selected) alkyl substituents, illustratively and preferably representing methylamino, ethylamino, n-propylamino, isopropylamino, tørt-butylamino, n-pentylamino, n-hexylamino, N,N-dimethylamino, N,N-diethyl- amino, N-ethyl-N-methylamino, N-methyl-N-n-propylamino, N-isopropyl-N-n-propylamino, N-tert- butyl-N-methylamino, N-ethyl-N-n-pentylamino and N-n-hexyl-N-methylamino.

Mono-alkylamino represents an alkylamino radical having one alkyl substituents, illustratively and preferably representing methylamino, ethylamino, n-propylamino, isopropylamino, fert-butylamino, n-pentylamino and n-hexylamino.

Cvcloalkyl in general represents a cyclic saturated hydrocarbon radical having 3 to 8, preferably 3 to 6 carbon atoms. Non-limiting examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclo- hexyl and cycloheptyl.

Aryl in general represents an aromatic mono- or bicyclic radical having 6 to 10 ring atoms, illustratively and preferably representing phenyl and naphthyl.

Heteroaryl per se and in heteroarylmethyl in general represents an aromatic mono- or bicyclic radical having 5 to 10 and preferably 5 or 6 ring atoms, and up to 5 and preferably up to 4 hetero- atoms selected from the group consisting of S, O and N, illustratively and preferably representing thienyl, furyl, pyrrolyl, pyrazolyl, thiazolyl, oxazolyl, isothiazolyl, isoxazolyl, imidazolyl, oxadiazolyl, thiadiazolyl, pyridyl, pyrimidyl, pyridazinyl, indolyl, indazolyl, benzofuranyl, benzo- thienyl, benzothiazolyl, quinolinyl, isoquinolinyl.

Halogen represents fluorine, chlorine, bromine and iodine.

In another preferred embodiment, the present invention relates to compounds of general formula (I), wherein

R 1 represents hydrogen or halogen,

R 2 represents hydrogen or halogen,

R 3 represents hydrogen,

R 4 represents chlorine, nitro, trifluoromethoxy, Ci-C 3 -alkyl or Ci-C 3 -alkoxy,

R 5 represents hydrogen,

R 6 represents C 3 -C 8 -alkyl, C 3 -C 7 -cycloalkyl, phenyl, 5- or 6-membered heteroaryl or a group of the formula -Y-R 9 ,

wherein cycloalkyl can be further substituted with one to three identical or different radicals selected from the group consisting of trifluoromethyl and C 1 -C 4 - alkyl.

and

wherein phenyl and heteroaryl can be further substituted with one to three identical or different radicals selected from the group consisting of halogen, nitro, trifluoromethyl, Ci-C 6 -alkyl and C|-C 6 -alkoxy,

and

wherein

Y represents CpGj-alkandiyl,

R 9 represents C 3 -C 7 -cycloalkyl, phenyl or 5- or 6-membered heteroaryl,

wherein cycloalkyl can be further substituted with one to three identical or different radicals selected from the group consisting of trifluoromethyl and C,-C 4 -alkyl,

and

wherein phenyl and heteroaryl can be further substituted with one to three identical or different radicals selected from the group consisting of halogen, nitro, trifluoromethyl, Ci-Cβ-alkyl and Ci-C 6 -alkoxy,

R 7 represents Ci-C 3 -alkyl,

wherein alkyl is further substituted with one radical selected from the group consisting of amino, mono-alkylamino or C,-C 4 -alkoxycarbonylamino,

R 8 represents hydrogen,

and their salts, hydrates and/or solvates.

In another particularly preferred embodiment, the present invention relates to compounds of general formula (I), wherein

R 1 represents hydrogen or fluorine,

R 2 represents hydrogen or fluorine,

R 3 represents hydrogen,

R 4 represents chlorine or methoxy,

R 5 represents hydrogen,

R 6 represents C 3 -C 6 -alkyl, C 3 -C 6 -cycloalkyl, phenyl, thienyl or a group of the formula -Y-R 9 ,

wherein cycloalkyl can be further substituted with one trifluoromethyl group,

and

wherein phenyl and thienyl can be further substituted with one to three identical or different radicals selected from the group consisting of fluorine, chlorine, nitro, trifluoromethyl, methyl and methoxy,

and

wherein

Y represents methylen or ethan- 1 , 1 -diyl,

R 9 represents phenyl,

wherein phenyl can be further substituted with one to three identical or different radicals selected from the group consisting of fluorine, chlorine, nitro, trifluoromethyl, methyl and methoxy,

R 7 represents Ci-C 2 -alkyl,

wherein alkyl is further substituted with one radical selected from the group consisting of amino or tert-butoxycarbonylamino,

R 8 represents hydrogen,

and their salts, hydrates and/or solvates.

In another preferred embodiment, the present invention relates to compounds of general formula (I), wherein R 7 represents -CH 2 NH 2 or -CH 2 CH 2 NH 2 .

In another preferred embodiment, the present invention relates to compounds of general formula (I), wherein R 1 , R 2 and R 3 represent hydrogen.

In another preferred embodiment, the present invention relates to compounds of general formula (I), wherein R 1 represents halogen, R 2 represents hydrogen or halogen and R 3 represents hydrogen or halogen.

In another preferred embodiment, the present invention relates to compounds of general formula (I), wherein R 1 represents halogen, R 2 represents hydrogen or halogen and R 3 represents hydrogen.

In another preferred embodiment, the present invention relates to compounds of general formula (I), wherein R 1 represents fluorine, R 2 represents hydrogen or fluorine and R 3 represents hydrogen.

In another preferred embodiment, the present invention relates to compounds of general formula (0, wherein R 4 represents chlorine or methoxy.

In another preferred embodiment, the present invention relates to compounds of general formula (I), wherein R 6 represents phenyl, wherein phenyl can be further substituted with one to three identical or different radicals selected from the group consisting of fluorine, chlorine, nitro, trifluoromethyl, methyl and methoxy.

In another preferred embodiment, the present invention relates to compounds of general formula (I), wherein R 6 represents a group of the formula -Y-R 9 , wherein Y represents methylen or ethan- 1 ,1-diyl, and R 9 represents phenyl, wherein phenyl can be further substituted with one to three identical or different radicals selected from the group consisting of fluorine, chlorine, nitro, trifluoromethyl, methyl and methoxy.

Very particular preference is given to combinations of two or more of the abovementioned preference ranges.

The compounds of general formula (I) can be synthesized by

[A] condensing compounds of general formula (II)

wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 7 and R 8 have the meaning indicated above,

with compounds of general formula (III)

R 6 — N==O rø.

wherein R 6 has the meaning indicated above,

or

[B] condensing compounds of general formula (II) with a mixture of a carbonic acid derivative and compounds of general formula (IV)

^NH 2 (IV),

wherein R 6 has the meaning indicated above,

or

[C] condensing compounds of general formula (V)

wherein R 4 , R 5 , R 6 , R 7 and R 8 have the meaning indicated above,

with compounds of general formula (VI)

wherein R 1 , R 2 and R 3 have the meaning indicated above, and

X 1 represents a leaving group, such as halogen, preferably chlorine or bromine,

in the presence of a base.

In process [A] and [B] optionally a base can be added to the reaction mixture.

Amino groups in R 7 of compounds of general formula (II) are protected with acid labile groups, preferred is a boc-group. After the synthesis of compounds of general formula (I) this acid labile group can be cleaved via standard procedures known by a person skilled in the art. Compounds of general formula (I) are obtained. Preferred are acidic cleavage conditions.

If a salt of a compound of general formula (I), for example a hydrochloride or trifluoroacetate, is isolated the free base can be obtained by reversed phase chromatography of the salt using a mixture of acetonitile and water as eluent in the presence of a base. Preferably a RP 18 Phenomenex Luna C 18(2) column is used in the presence of diethylamine as base. Or the free base of a compound of general formula (I) can be obtained by neutralizing with a base and extraction.

The process [A] is in general carried out in a temperature range from -20 0 C to boiling point of the solvent, preferably from O 0 C to +40 0 C.

The process is generally carried out at normal pressure. However, it is also possible to carry it out at elevated pressure or at reduced pressure (for example in a range from 0.5 to 5 bar).

Suitable solvents for the process are ethers such as dioxan or tetrahydrofuran, or halogeno-hydro- carbons such as dichloromethane, dichloroethane or trichloromethane, or other solvents such as dimethylformamide, ethyl acetate or acetonitrile. It is also possible to use mixtures of the above- mentioned solvents. Preferred for the process is tetrahydrofuran or dichloromethane.

Suitable bases for the process are generally inorganic or organic bases. These preferably include alkali carbonates such as sodium or potassium carbonate or hydrogencarbonate, cyclic amines such as, for example, N-methylmorpholine, N-methylpiperidine, pyridine or 4-N,N-dimethylamino- pyridine, or (C|-C 4 )-trialkylamines such as, for example, triethylamine or diisopropylethylamine or polymer bound bases such as polymer bound tris-amine. Preference is given to polymer bound tris- amine.

The process [B] is in general carried out in a temperature range from room temperature to +40 0 C.

The process is generally carried out at normal pressure. However, it is also possible to carry it out at elevated pressure or at reduced pressure (for example in a range from 0.5 to 5 bar).

Suitable carbonic acid derivatives for the process are ν.ν-carbonyldiimidazole, phosgene, diphosgene, triphosgene, chloroformic acid phenyl ester or chloroformic acid 4-nitrophenyl ester. Preference is given to ν,ν-carbonyldiimidazole.

Suitable solvents for the process are ethers such as dioxan or tetrahydrofuran, or halogeno-hydro- carbons such as dichloromethane, dichloroethane or trichloromethane, or other solvents such as dimethylformamide, ethyl acetate or acetonitrile. It is also possible to use mixtures of the above- mentioned solvents. Preferred for the process is tetrahydrofuran or dichloromethane.

Suitable bases for the process are generally organic bases. These preferably include cyclic amines such as, for example, N-methylmorpholine, N-methylpiperidine, pyridine or 4-NN-dimethylamino- pyridine, or (Cι-Gi)-trialkylamines such as, for example, triethylamine or diisopropylethylamine. Preference is given to diisopropylethylamine.

The process [C] is in general carried out in a temperature range from 0 0 C to boiling point of the solvent, preferably from 20 0 C to boiling point of the solvent.

The process is generally carried out at normal pressure. However, it is also possible to carry it out at elevated pressure or at reduced pressure (for example in a range from 0.5 to 5 bar).

Optionally an alkali iodide such as sodium or potassium iodide can be added to the reaction mixture.

Suitable solvents for the process are ethers such as dioxan or tetrahydrofuran, or halogeno-hydro- carbons such as dichloromethane, dichloroethane or trichloromethane, or other solvents such as dimethylformamide, dimethylsulfoxide, ethyl acetate or acetonitrile. It is also possible to use mixtures of the above-mentioned solvents. Preferred for the process is acetonitrile.

Suitable bases for the process are generally inorganic or organic bases. These preferably include alkali carbonates such as sodium or potassium carbonate or hydrogen carbonate, cyclic amines such as, for example, N-methylmorpholine, N-methylpiperidine, pyridine or 4-N,N-dimethylamino- pyridine, or (Ci-GO-trialkylamines such as, for example, triethylamine or diisopropylethylamine. Preference is given to potassium carbonate.

The compounds of the general formula (III), (FV) and (VI) are known per se, or they can be prepared by customary methods.

The compounds of general formula (II) can be synthesized by condensing compounds of general formula (VII)

wherein R 1 , R 2 , R 3 , R 4 and R 5 have the meaning indicated above, with compounds of general formula (VIII)

wherein R 7 and R 8 have the meaning indicated above,

under conditions of a reductive amination.

The process is in general carried out in a temperature range from -20 0 C to boiling point of the solvent, preferably from 0 0 C to +40 0 C.

The process is generally carried out at normal pressure. However, it is also possible to carry it out at elevated pressure or at reduced pressure (for example in a range from 0.5 to 5 bar).

Suitable solvents for the process are halogeno-hydrocarbons such as dichloromethane, dichloro- ethane or trichloromethane, or alcohols such as methanol, ethanol, n-propanol, iso-propanol, n- butanol or tert-butano\, or a mixture of alcohol and water. Preferred for the process is methanol or a mixture of methanol and water.

Suitable reducing agents for the process are sodium borohydride or triacetoxyborohydride.

The compounds of the general formula (VIII) are known per se, or they can be prepared by customary methods.

The compounds of general formula (VII) can be synthesized by condensing compounds of general formula (IX)

wherein R 4 and R 5 have the meaning indicated above, with compounds of general formula (X)

wherein R 1 , R 2 and R 3 have the meaning indicated above, and

X 2 represents a leaving group, such as halogen, preferably chlorine or bromine,

in the presence of a base.

Optionally an alkali iodide such as sodium or potassium iodide can be added to the reaction mixture.

The process is in general carried out in a temperature range from 0 0 C to boiling point of the solvent, preferably from 20 0 C to boiling point of the solvent.

The process is generally carried out at normal pressure. However, it is also possible to carry it out at elevated pressure or at reduced pressure (for example in a range from 0.5 to 5 bar).

Suitable solvents for the process are ethers such as dioxan or tetrahydrofuran, or halogeno-hydro- carbons such as dichloromethane, dichloroethane or trichloromethane, or other solvents such as dimethylformamide, dimethylsulfoxide, ethyl acetate or acetonitrile. It is also possible to use mixtures of the above-mentioned solvents. Preferred for the process is acetonitrile.

Suitable bases for the process are generally inorganic or organic bases. These preferably include alkali carbonates such as caesium, sodium or potassium carbonate or hydrogencarbonate, cyclic amines such as, for example, N-methylmorpholine, N-methylpiperidine, pyridine or 4-N 1 N- dimethylaminopyridine, or (Ci-C 4 )-trialkylamines such as, for example, triethylamine or diiso- propylethylamine. Preference is given to potassium carbonate.

The compounds of the general formulas (IX) and (X) are known per se, or they can be prepared by customary methods.

The compounds of general formula (V) can be synthesized by treating compounds of general formula (Ia)

wherein R 4 , R 5 , R 6 , R 7 and R 8 have the meaning indicated above,

under reducing conditions.

The process is in general carried out in a temperature range from -20 0 C to boiling point of the solvent, preferably from 0 0 C to +4O 0 C.

The process is generally carried out at normal pressure. However, it is also possible to carry it out at elevated pressure or at reduced pressure (for example in a range from 0.5 to 5 bar).

Suitable solvents for the process are alcohols such as methanol, ethanol, n-propanol, iso-propanol, n-butanol or tert-butanol, or tetrahydrofuran, or a mixture of alcohol and water. Preferred for the process is methanol, ethanol, tetrahydrofuran or a mixture of ethanol and water.

Suitable reducing agents for the process are metals such as palladium, platin, nickel or ruthenium or oxides thereof in the presence of hydrogen such as palladium on charcoal and hydrogen. Preferred for the process is palladium on charcoal and hydrogen.

The compounds of general formula (Ia) can be synthesized via process [A] or [B] or [C].

The above-mentioned process can be illustrated by the following scheme:

Scheme 1

potassium carbonate

H lN — YY CH H :

O CH, sodium borohydπde

isolation as free base or salt (TFA or HCI)

Alternatively above mentioned process can be conducted on solid support using polymer bound

diamines. Initially the diamines are attached to the resin via an acid labile linkage. In the final step of the synthesis the products are released from the solid support. The following scheme illustrates the process on solid phase:

Scheme 2

The compounds according to the invention exhibit an unforeseeable, useful pharmacological activity spectrum. They are therefore suitable for use as medicaments for the treatment and/or prophylaxis of disorders in humans and animals.

Surprisingly, the compounds of the present invention show excellent CMR-I antagonistic activity. They are, therefore suitable especially for the prophylaxis and treatment of diseases associated with CMR-I activity, in particular for the treatment of urological diseases or disorders, such as detrusor overactivity (overactive bladder), urinary incontinence, neurogenic detrusor oeractivity (detrusor hyperflexia), idiopathic detrusor overactivity (detrusor instability), benign prostatic hyperplasia, and lower urinary tract symptoms.

The compounds of the present invention are also effective for treating or preventing a disease selected from the group consisting of chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neuro- degeneration and/or stroke, as well as respiratory diseases and inflammatory diseases such as asthma, COPD and allergic rhinitis since the diseases also relate to CMR-I activity.

The compounds of the present invention are also useful for the treatment and prophylaxis of neuropathic pain, which is a form of pain often associated with herpes zoster and post-herpetic

neuralgia, painful diabetic neuropathy, neuropathic low back pain, posttraumatic and postoperative neuralgia, neuralgia due to nerve compression and other neuralgias, phantom pain, complex regional pain syndromes, infectious or parainfectious neuropathies like those associated with HIV infection, pain associated with central nervous system disorders like multiple sclerosis or Parkinson disease or spinal cord injury or traumatic brain injury, and post-stroke pain.

Furthermore, the compounds of the present invention are useful for the treatment of musculoskeletal pain, forms of pain often associated with osteoarthritis or rheumatoid arthritis or other forms of arthritis, and back pain.

In addition, the compounds of the present invention are useful for the treatment of pain associated with cancer, including visceral or neuropathic pain associated with cancer or cancer treatment.

The compounds of the present invention are furthermore useful for the treatment of visceral pain, e.g. pain associated with obstruction of hollow viscus like gallstone colik, pain associated with irritable bowel syndrome, pelvic pain, vulvodynia, orchialgia or prostatodynia, pain associated with inflammatory lesions of joints, skin, muscles or nerves, and orofascial pain and headache, e.g. migraine or tension-type headache.

The present invention further provides medicaments containing at least one compound according to the invention, preferably together with one or more pharmacologically safe excipient or carrier substances, and also their use for the above-mentioned purposes.

The active component can act systemically and/or locally. For this purpose, it can be applied in a suitable manner, for example orally, parenterally, pulmonally, nasally, sublingually, lingually, buccally, rectally, transdermally, conjunctivally, otically or as an implant.

For these application routes, the active component can be administered in suitable application forms.

Useful oral application forms include application forms which release the active component rapidly and/or in modified form, such as for example tablets (non-coated and coated tablets, for example with an enteric coating), capsules, sugar-coated tablets, granules, pellets, powders, emulsions, suspensions, solutions and aerosols.

Parenteral application can be carried out with avoidance of an absorption step (intravenously, intraarterially, intracardially, intraspinally or intralumbarly) or with inclusion of an absorption (intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneal^). Useful

parenteral application forms include injection and infusion preparations in the form of solutions, suspensions, emulsions, lyophilisates and sterile powders.

Forms suitable for other application routes include for example inhalatory pharmaceutical forms (including powder inhalers, nebulizers), nasal drops/solutions, sprays; tablets or capsules to be administered lingually, sublingually or buccally, suppositories, ear and eye preparations, vaginal capsules, aqueous suspensions (lotions, shake mixtures), lipophilic suspensions, ointments, creams, milk, pastes, dusting powders or implants.

The active components can be converted into the recited application forms in a manner known per se. This is carried out using inert non-toxic, pharmaceutically suitable excipients. These include inter alia carriers (for example microcrystalline cellulose), solvents (for example liquid polyethylene glycols), emulsifiers (for example sodium dodecyl sulphate), dispersing agents (for example polyvinylpyrrolidone), synthetic and natural biopolymers (for example albumin), stabilizers (for example antioxidants such as ascorbic acid), colorants (for example inorganic pigments such as iron oxides) or taste and/or odor corrigents.

For human use, in the case of oral administration, it is recommendable to administer doses of from 0.001 to 50 mg/kg, preferably of 0.01 mg/kg to 20 mg/kg. In the case of parenteral administration, such as, for example, intravenously or via mucous membranes nasally, buccally or inhalationally, it is recommendable to use doses of 0.001 mg/kg to 0.5 mg/kg.

In spite of this, it can be necessary in certain circumstances to depart from the amounts mentioned, namely as a function of body weight, application route, individual behaviour towards the active component, manner of preparation and time or interval at which application takes place. It can for instance be sufficient in some cases to use less than the aforementioned minimum amount, while in other cases the upper limit mentioned will have to be exceeded. In the case of the application of larger amounts, it can be advisable to divide them into a plurality of individual doses spread through the day.

The percentages in the tests and examples which follows are, unless otherwise stated, by weight; parts are by weight. Solvent ratios, dilution ratios and concentrations reported for liquid/liquid solutions are each based on the volume.

A. Examples

Abbreviations: aq. aqueous boc tert-butoxycarbonyl

CDCl 3 deutero chloroform cone. concentrated

DCI direct chemical ionisation (for MS)

DMAP 4-N,N-dimethylaminopyridine

DMF NN-dimethylformamide

DMSO dimethylsulfoxide

EI electron impact ionisation (for MS)

ESI electro-spray ionisation (for MS) h hour(s)

HOBT hydroxybenzotriazole

HPLC high pressure liquid chromatography

LC-MS liquid chromatography coupled with mass spectroscopy min minute(s)

Mp. melting point

MS mass spectroscopy

NMR nuclear magnetic resonance spectroscopy of th. of theoretical (yield)

RP reverse phase (for HPLC) rt room temperature

R, retention time (for HPLC) sat. saturated

TFA trifluoroacetic acid

THF tetrahydrofuran

LC-MS / HPLC methods: method 1 (LC-MS): Instrument MS: Micromass ZQ; Instrument HPLC: Waters Alliance 2795; column: Phenomenex Synergi 2μ Hydro-RP Mercury 20 mm x 4 mm; eluent A: 1 1 water + 0.5 ml 50% formic acid, eluent B: 1 1 acetonitrile + 0.5 ml 50% formic acid; gradient: 0.0 min 90% A — > 2.5 min 30% A → 3.0 min 5% A -» 4.5 min 5% A; flow: 0.0 min 1 ml/min → 2.5 min/3.0 min/4.5

min 2 ml/min; oven: 50 0 C; UV detection: 210 nm.

method 2 (HPLO: Instrument: HP 1100 with DAD detection; column: Kromasil 100 RP-18, 60 mm x 2.1 mm, 3.5 μm; eluent A: 5 ml HClO 4 (70%) /1 water, eluent B: acetonitrile; gradient: 0 min 2% B → 0.5 min 2% B → 4.5 min 90% B → 15.0 min 90%B → 15.2 min 2% B → 16 min 2% B; flow: 0.75 ml/min; oven: 30 0 C; UV detection: 210 nm.

method 3 (HPLC): Instrument: HP 1 100 with DAD-detection, column: Kromasil 100 RP-18, 60 mm x 2.1 mm, 3.5 μm; eluent A: 5 ml HClO 4 (70 %) /1 water, eluent B: acetonitrile; gradient: 0 min 2% B -» 0.5 min 2% B -> 4.5 min 90% B -» 6.5 min 90% B -» 6.7 min. 2% B -» 7.5 min 2% B; flow: 0.75 ml/min; column temperature: 30 0 C; UV detection: 210 nm.

method 4 (LC-MS): Instrument MS: Micromass ZQ; Instrument HPLC: Waters Alliance 2795; column: Merck Chromolith SpeedROD RP-18e 50 mm x 4.6 mm; eluent A: 1 1 water + 0.5 ml 50% formic acid; eluent B: 1 1 acetonitrile + 0.5 ml 50% formic acid; gradient: 0.0 min 10% B-> 3.0 min 95% B-> 4.0 min 95% B; flow: 0.0 min 1.0 ml/min-> 3.0 min 3.0 ml/min-> 4.0 min 3.0 ml/min; oven: 35°C; UV detection: 210 nm.

method 5 (LC-MS): instrument MS: Micromass ZQ; instrument HPLC: HP 1 100 Series; UV DAD; column: Phenomenex Synergi 2μ Hydro-RP Mercury 20 mm x 4 mm; eluent A: 1 1 water + 0.5 ml 50% formic acid, eluent B: 1 1 acetonitrile + 0.5 ml 50% formic acid; gradient: 0.0 min 90%A -> 2.5 min 30%A -> 3.0 min 5%A -» 4.5 min 5%A; flow: 0.0 min 1 ml/min, 2.5 min/3.0 min/4.5 min. 2 ml/min; oven: 5O 0 C; UV-detection: 210 nm.

method 6 (LC-MS): Instrument MS: Micromass Quattro LCZ; Instrument HPLC: HP 1 100 Series; UV DAD; column: Phenomenex Synergi 2μ Hydro-RP Mercury 20 mm x 4 mm; eluent A: 1 1 water + 0.5 ml 50% formic acid, eluent B: 1 1 acetonitrile + 0.5 ml 50% formic acid; gradient: 0.0 min 90% A → 2.5 min 30% A → 3.0 min 5% A → 4.5 min 5% A; flow: 0.0 min 1 ml/min → 2.5 min/3.0 min/4.5 min 2 ml/min; oven: 50 0 C; UV detection: 210 nm.

method 7 (HPLCV Instrument: HP 1 100 with DAD-detection; column: Kromasil 100 RP-18, 60 mm x 2.1 mm, 3.5 μm; eluent A: 5 ml HClO 4 (70 %) /1 water, eluent B: acetonitrile; gradient: 0 min 2% B -» 0.5 min 2% B -> 4.5 min 90% B ^ 9 min 90% B -» 9.2 min. 2% B ^ 10 min 2 % B; flow: 0.75 ml/min; column temperature: 30 0 C; UV detection: 210 nm.

Starting Materials and Intermediates:

Example IA

tert-buty\ (2-{[4-(benzyloxy)-3-methoxybenzyl]amino}ethyl)carbamate

33.8 g (139.6 mmol) 4-benzyloxy-3-methoxybenzaldehyde are dissolved in 160 ml methanol, 24.6 g (153.5 mmol) ter/-butyl(2-aminoethyl)carbamate are added and the resulting suspension is stirred for 2 h at rt. The mixture is cooled to 0 0 C and 26.4 g (697.9 mmol) sodium borohydride is added in portions. After stirring over night the solution is diluted with water and extracted three times with dichloromethane (aq. layer is saturated with sodium chloride). The combined organic layers are dried over magnesium sulfate and concentrated in vacuo. The crude product is purified by chromatography on silica gel (gradient dichloromethane / methanol 100: 1 , 50: 1 , 20: 1.) to yield 47.68 g (88% of th.) of the title compound.

LC-MS (method 1): R, = 1.50 min, m/z = 387 (M+H) +

1 H-NMR (300 MHz, DMSOd 6 ): δ = 7.50-7.25 (m, 5H), 6.90 (m, 2H), 6.80-6.70 (m, 2H), 5.05 (s, 2H), 3.75 (s, 3H), 3.60 (s, 2H), 3.00 (m, 2H), 2.50 (m, 2H), 1.40 (s, 9H).

Example 2A

tert-buly\ (3 - { [4-(benzyloxy)-3 -methoxybenzyl]amino } propyl)carbamate

6.31 g (26.04 mmol) 4-benzyloxy-3-methoxybenzaldehyde are dissolved in 30 ml methanol, 4.99 g (28.64 mmol) tert-butyl (3-aminopropyl)carbamate are added and the resulting suspension is

stirred for 2 h at rt. The mixture is cooled to 0 0 C and 4.93 g (130.2 mmol) sodium borohydride is added in portions. After stirring over night the solution is diluted with water and extracted three times with dichloromethane (aq. layer is saturated with sodium chloride). The combined organic layers are dried over magnesium sulfate and concentrated in vacuo. The crude product is purified by chromatography on silica gel (gradient dichloromethane / methanol 100:1 , 50: 1 , 20: 1.) to yield 9.7 g (93% of th.) of the title compound.

HPLC (method 2): R, = 3.37 min,

MS: m/z = 401 (M+H) +

1 H-NMR (300 MHz, DMSOd 6 ): δ = 7.47-7.27 (m, 5H), 6.90-6.95 (m, 2H), 6.81-6.70 (m, 2H), 5.04 (s, 2H), 3.75 (s, 3H), 3.58 (s, 2H), 3.00-2.90 (m, 2H), 2.47-2.40 (m, 2H), 1.58-1.48 (m, I H), 1.45 (s, 9H).

Example 3A

4-[(2-fluorobenzyl)oxy] -3 -methoxybenzaldehyde

10.00 g (65.7 mmol) 4-hydroxy-3 -methoxybenzaldehyde and 13.67 g (72.3 mmol) 2-fluorobenzyl bromide are dissolved in 100 ml acetonitrile. 45.4 g (328.6 mmol) potassium carbonate and 10.91 g (65.7 mmol) potassium iodide are added and the mixture is heated to reflux during 3 h. After cooling to room temperature, water is added and the solution is extracted twice with ethyl acetate. The combined organic materials are washed with water, dried over magnesium sulfate and the solvent is evaporated under vacuum. Petroleum ether is added, the solid is triturated, filtered and dried to yield 17.0 g (99% of th.) of the title compound.

HPLC (method 3): R, = 4.56 min

MS (DCI): m/z = 261 (M+H) +

1 H-NMR (200 MHz, DMSOd 6 ): δ = 9.85 (s, IH), 7.60-7.20 (m, 7H), 5.20 (s, 2H), 3.80 (s, 3H).

Using an analogous procedure the following compounds are prepared:

Example 6A

tert-buly\ [2-({4-[(2-fluorobenzyl)oxy]-3-methoxybenzyl}amino)ethyl]car bamate

2.00 g (7.68 mmol) 4-[(2-fluorobenzyl)oxy]-3-methoxybenzaldehyde and 1.35 g (8.45 mmol) tert- butyl (2-aminoethyl)carbamate are dissolved in 40 ml methanol and stirred for 1 h at room temperature. The solution is cooled to 0 0 C and 1.45 g (38.4 mmol) sodium borohydride are carefully added. Water is added until a clear solution is formed and the mixture is stirred during 2h at room temperature. The mixture is concentrated under vacuum, the residue is diluted with dichloromethane and the organic phase is washed with brine, dried over magnesium sulfate and concentrated under vacuum to yield 2.40 g (68% of th.) of the title compound of sufficient purity to be used in the next step.

HPLC (method 3): R 1 = 4.47 min

MS (ESIpos): m/z = 405 (M+H) +

1 H-NMR (300 MHz, DMSOd 6 ): δ = 7.55-7.20 (m, 4H), 6.90 (m, 2H), 6.80-6.70 (m, 2H), 5.05 (s, 2H), 3.75 (s, 3H), 3.60 (s, 2H), 3.00 (m, 2H), 2.50 (m, 2H), 1.40 (s, 9H)

Using an analogous procedure the following compounds are prepared:

Example 9A

/er/-butyl {2-[{[(4-fluorophenyl)amino]carbonyl}(4-hydroxy-3-methoxyben zyl)amino]ethyl}- carbamate

1.33 g tert-buly\ [2-([4-(benzyloxy)-3-methoxybenzyl] {[(4-fluorophenyl)amino]carbonylamino)- ethyl]carbamate (2.54 mmol) are dissolved in 20 ml ethanol. 250 mg Pd/C (5%) are added and the mixture is stirred at rt under hydrogen atmosphere (normal pressure) for 1 h until the starting material is consumed. The mixture is filtrated through a plug of celite and the resulting filtrate is concentrated in vacuo. The crude product is purified by chromatography on silica gel (cyclohexane / ethyl acetate 2: 1) to yield 981 mg (89% of th.) of the title compound.

LC-MS (method 5): R, = 2.42 min, m/z = 434 (M+H) +

1 H-NMR (300 MHz, DMSO-d 6 ): δ = 8.89 (s, IH), 8.45 (s, IH), 7.54-7.45 (m, 2H), 7.12-7.06 (m, 2H), 6.95 (m, IH), 6.81 (s, IH), 6.74-6.65 (m, 2H), 4.46 (s, 2H), 3.75 (s, 3H), 3.35-3.26 (m, 2H), 3.10-3.01 (m, 2H), 1.38 (s, 9H).

Example IQA

N-[4-(benzyloxy)-3-methoxybenzyl]-N'-tritylethane-l ,2-diamine

14.56 g (60.1 mmol) 4-benzyloxy-3-methoxybenzaldehyde are dissolved in methanol and a solution of 20 g (approx. 66 mmol) N'-tritylethane-l,2-diamine (prepared from commercially available N'-tritylethane-l ,2-diamine hydrobromide) in methanol is added. To the mixture 20 ml dichloromethane are added and a white solid precipitated after 1 h at rt. The mixture is cooled to 0 0 C and 1 1.37 g (300.6 mmol) sodium borohydride is added in portions. After stirring over night the solution is diluted with water and the precipitated solid collected by filtration and washed three times with water. The residual solid is treated with ethanol and washed with ethanol. The

combined filtrates are evaporated in vacuo to yield 47.68 g (88% of th.) of the title compound. The material is not further purified.

LC-MS (method 5): R 1 = 2.35 min, m/z = 529 (M+H) +

1 H-NMR (400 MHz, DMSO-d 6 ): δ = 8.27 (s, IH), 7.48-7.10 (m, 26H), 5.12 (s, 2H), 3.80 (s, 3H), 3.68 (m, 2H), 2.72 (mt, IH), 2.25 (m, 2H).

Example HA

N-[4-(benzyloxy)-3-chlorobenzyl]-l ,2-diaminoethane trityl resin

4 g (approx. 4.8 mmol) 1 ,2-diaminoethane trityl resin are suspended in a mixture of methanol and dichloromethane. 3.55 g (14.4 mmol) 4-benzyloxy-3-chlorobenzaldehyde and 2.6 ml (24 mmol) trimethylorthoformiat are added and the mixture is agitated for 2 h at rt, before filtrated. The resin is washed two times with dichloromethane, suspended in 20 ml dichloromethane and 8 ml methanol. 907.9 mg (24 mmol) sodium borohydride are added in three portions and the mixture is vigorously agitated for 5 h at rt, before filtrated. The resin is washed successively with: DMF/methanol 1 : 1 (two times), methanol/water 1 : 1, methanol (two times) and dichloromethane

(four times). The resin can be used without further purification, drying to constant weight can be achieved in high vacuum.

Example 12A

N-[4-(benzyloxy)-3-methoxybenzyl]-l ,2-diaminorpopane trityl resin

2.00 g (approx. 2.0 mmol) trityl chloride resin (approx. 1 mmol/g loading, Novabiochem) are suspended in dichloromethane. 593.01 mg (8.00 mmol) propane-l ,3-diamine and 1.55 g N-ethyl-N- isopropylpropan-2-amine are added. The mixture is shaken at rt over night. The mixture is filtered and the remaining resin is washed three times with small portions of DMF and afterwards three times alternately with small portions of methanol and dichloromethane.

1.00 g (approx. 1.00 mmol) of the thus obtained resin are suspended in a mixture of dichloromethane and TMOF (1 : 1). 2.42 g (10 mmol) 4-(benzyloxy)-3-methoxybenzaldehyde are added and the mixture is shaken over night at rt. The resin is washed three times with DMF. After suspending the resin in 20 ml DMF, 1.287 g tetra-N-butyl ammonium borohydride are added and the mixture is stirred 30 min at rt. The mixture is cooled to -40 0 C and 5.7 ml (99 mmol) acetic acid are added and the mixture is slowly warmed up to rt. The mixture is filtered and the remaining resin is washed three times with DMF, and then successively twice with water, with a mixture of DMF and N-ethyl-N-isopropylpropan-2 -amine (1 : 1), with methanol and with dichloromethane. The resin is dried in vacuo.

Example 13A

N-[4-(benzyloxy)-3-methoxybenzyl]-N'-4-fluoro-phenyl-N-[2 -(tritylamino)ethyl]urea

2.5 g (4.79 mmol) N-[4-(benzyloxy)-3-methoxybenzyl]-N'-tritylethane-l ,2-diamine are dissolved in

4 ml dichlormethane. 0.54 ml (4.73 mmol) 4-fluorophenylisocyanate are added at rt. Stirring is continued for 45 min before solvents are removed by evaporation. The crude product is treated with ethanol and the solution is decanted from an oily residue. Upon standing crystallization starts and the precipitate is collected by filtration and dried in vacuo to yield 1.70 g (54% of th.) of the title compound. The material is not further purified.

LC-MS (method 1): R 1 = 2.91 min, m/z = 666 (M+H) +

'H-NMR (400 MHz, DMSOd 6 ): δ = 8.50 (s, I H), 7.50-7.05 (m, 21H), 6.94 (d, I H), 6.90 (d, IH), 6.75 (d, 1 H), 5.05 (s, 2H), 4.51 (s, 2H), 3.70 (s, 3H), 3.40 (m, 2H), 3.05 (t, I H), 2.14 (m, 2H).

Preparation Examples:

Example 1

/er/-butyl(2-{(anilinocarbonyl)[4-(benzyloxy)-3-methoxybe nzyl]amino}ethyl)carbamate

1.79 g (4.63 mmol) ter/-butyl(2-{[4-(benzyloxy)-3-methoxybenzyl]amino}ethyl)car bamate are dissolved in dichloromethane and at rt 0.7 ml (6.48 mmol) phenylisocyanate are added. After 1 h the reaction is complete, stirring is continued over night until the reaction mixture is concentrated in vacuo. The crude product is purified by chromatography on silica gel (gradient isohexane / ethyl acetate 10: 1 to 5: 1 to 2: 1 ) to yield 2.06 g (87% of th.) of the title compound.

LC-MS (method 5): R, = 2.93 min, m/z = 506 (M+H) +

1 H-NMR (400 MHz, DMSO-d 6 ): δ = 8.42 (s, IH), 7.52-7.21 (m, 9H), 7.05-6.89 (m, 4H), 6.77 (d, I H), 5.12 (s, 2H), 5.05 (s, 2H), 4.51 (s, 2H), 3.75 (3H), 3.35-3.30 (m, 2H), 3.07 (m, 2H), 1.38 (s, 9H).

Example 2

teA-/-butyl[2-((anilinocarbonyl) {4-[(2,4-difluorobenzyl)oxy]-3-methoxybenzyl}amino)ethyl]- carbamate

1.0 g (2.18 mmol) /erf-butyl[2-({4-[(2,4-difluorobenzyl)oxy]-3-methoxybenzyl}a mino)ethyl]- carbamate are dissolved in 30 ml dichloromethane and treated at rt with 260 mg (2.18 mmol)

phenylisocyanate. The mixture is stirred at rt over night, before concentrated in vacuo. The residual oil is purified by preparative reverse phase HPLC to afford 747 mg (63% of th.) of the title compound.

HPLC (method 7): R, = 5.19 min;

MS (ESI+): m/z = 542 (M+H) +

1 H-NMR (300 MHz, DMSOd 6 ): δ = 8.42 (s, IH), 7.59 (m, IH), 7.50 (d, 2H), 7.31 (dd, IH), 7.26 (t, 2H), 7.13 (m, IH), 7.03 (d, IH), 7.00-6.89 (m, 2H), 6.78 (dd, IH), 5.03 (s, 2H), 4.51 (s, 2H), 3.73 (s, 3H), 3.06 (m, 2H), 1.38 (s, 9H).

Using analogous procedure following examples are prepared:

example name structure analytical data

LC-MS (method 6): R 1 = 2.83 min, m/z = 556 tert-butyl [2-({4- (M+H) +

[(2,4-difluoro- benzyl)oxy]-3- 1 H-NMR (400 MHz, methoxybenzyl}- DMSO-d 6 ): δ = 8.32 (s,

{[(2-methyl- IH), 7.80 (s, IH), 7.59 phenyl)amino]- (m, IH), 7.35-7.20 (m, carbonyl}- 2H), 7.19-7.10 (m, 2H), amino)ethyl]- 7.08-6.98 (m, 2H), 6.92 carbamate (s, IH), 6.89 (m, IH), 6.80 (dd, IH), 5.05 (s, 2H), 4.50 (s, 2H), 3.75 (s, 3H), 3.31 (m, 2H), 3.09 (m, 2H), 2.13 (s, 3H), 1.37 (s, 9H).

General procedure A: preparation of ureas from secondary amines and isocyanates, use of polymer bound tris-amine as scavenger

The amine (1.0 eq.) is dissolved in dichloromethane (approx. 0.2 g/ml). Isocyanate (1.2 to 2 eq.) is added at rt. After completion of reaction (approx. 1 h) an excess of polymer bound tris-amine and dichloromethane are added to the mixture, the suspension is agitated for approx. 1 h, then filtrated. The resin is washed with dichloromethane and the combined filtrates are concentrated in vacuo. Further purification of the product can be achieved by preparative reverse phase HPLC (water/acetonitril).

General procedure B: preparation of ureas from secondary amines and isocyanates

The amine (1.0 eq.) is dissolved in dichloromethane (approx. 0.2 g/ml). Isocyanate (1.2 to 1.5 eq.) is added at rt. The reaction is generally complete after 1 h. After evaporation of the volatile components from reaction mixture the product can be purified by preparative reverse phase HPLC (water/acetonitril) or chromatography on silica gel.

General procedure C; preparation of ureas from secondary amines and isocyanates

The amine (1.0 eq.) is dissolved in dichloromethane (approx. 0.2 g/ml). Isocyanate (1.2 to 1.5 eq.) is added at rt. The mixture is stirred over night, while the product gradually precipitates. The crystalline solid is collected by filtration, washed with ethanol and dried in vacuo.

Following compounds are prepared according to general procedures A, B or C (method) from the corresponding starting materials:

=

=

=

Example 42

tert-butyl [2-({4-[(4-fluorobenzyl)oxy]-3-methoxybenzyl} {[(4-fluorophenyl)amino]carbonyl}- amino)-ethyl]carbamate

483 mg (1.11 mmol) tert-butyl {2-[{[(4-fluorophenyl)amino]carbonyl}(4-hydroxy-3-methoxy- benzyl)amino]ethyl} carbamate are dissolved in acetonitrile and 363 mg (1.1 1 mmol) caesium carbonate and 18.5 mg (0.1 1 mmol) potassium iodide are added. To the stirred mixture 0.175 ml (1.4 mmol) 4-fluorobenzylbromide are added and stirring is continued for 1 h at rt before, the reaction mixture is poured on water. The resulting precipitate is collected by filtration and washed 3 times with water and purified by column chromatography (silica gel, isohexane / ethyl acetate 4: 1 to 2: 1) to afford 353.5 mg (59% of th.) of the title compound.

LC-MS (method 1 ): R, = 2.70 min, m/z = 542 (M+H) +

1 H-NMR (300 MHz, DMSO-d 6 ): δ = 8.48 (s, IH), 7.55-7.45 (m, 4H), 7.26-6.90 (m, 7H), 6.77 (d, IH), 5.12 (s, 2H), 5.03 (s, 2H), 4.50 (s, 2H), 3.75 (3H), 3.32 (m, 2H), 3.06 (m, 2H), 1.38 (s, 9H).

Example 43

tert-butyl [2-({4-[(2,4-difluorobenzyl)oxy]-3-methoxybenzyl} {[(4-fluorophenyl)amino]carbonyl}- amino)ethyl]carbamate

473 mg (1.1 mmol) tert-butyl {2-[{[(4-fluorophenyl)amino]carbonyl}(4-hydroxy-3-methoxy- benzyl)amino]ethyl}carbamate are dissolved in acetonitrile and 356 mg (1.1 mmol) caesium carbonate and 18 mg (0.1 mmol) potassium iodide are added. To the stirred mixture 0.175 ml (1.36 mmol) 2,4-difluorobenzylbromide are added and stirring is continued for 1 h at rt before, the

reaction mixture is poured on water. The resulting precipitate is collected by filtration and washed three times with water and purified by column chromatography (silica gel, isohexane / ethyl acetate 4: 1 to 2: 1 ) to afford 536 mg (88% of th.) of the title compound.

LC-MS (method 1): R, = 2.75 min, m/z = 560 (M+H) +

1 H-NMR (300 MHz, DMSOd 6 ): δ = 8.49 (s, IH), 7.65-7.47 (m, 3H), 7.30 (m, IH), 7.16-6.93 (m, 5H), 6.90 (s, IH), 6.78 (d, IH), 5.04 (s, 2H), 4.51 (s, 2H), 3.74 (3H), 3.33 (m, 2H), 3.07 (m, 2H), 1.38 (s, 9H).

Example 44

tert -butyl {2-[[4-(benzyloxy)-3-methoxybenzyl]({[( IS)-I -phenylethyl]amino}carbonyl)amino]- ethyl} carbamate

200 mg (0.52 mmol) ter/-butyl (2-{[4-(benzyloxy)-3-methoxybenzyl]amino}ethyl)carbamate (1.0 eq.) are dissolved in 2 ml dichloromethane and treated with 0.09 ml (0.62 mmol) (S)-(-)-alpha- methylbenzylisocyanate at rt for 1 h. After evaporation of the volatile components from reaction mixture the product is purified by preparative reverse phase HPLC (water/acetonitrile) to yield the title compound.

LC-MS (method 6): R 1 = 2.86 min, m/z = 534 (M+H) +

1 H-NMR (400 MHz, DMSO-d 6 ): δ = 7.45-7.16 (m, 9H), 6.95 (d, IH), 6.89 (t, IH), 6.80 (s, IH), 6.74 (d, 1 H), 6.69 (d, IH), 5.05 (s, 2H), 4.89 (m, IH), 4.49 (s, 2H), 3.68 (s, 3H), 3.23-3.12 (m, 2H), 3.02-2.95 (m, 2H), 1.38 (s, 12H).

Example 45

tert-butyl {2-[[4-(benzyloxy)-3-methoxybenzyl]({[(lS)-l-(4-fluorophenyl )ethyl]amino}carbonyl)- amino]ethyl } carbamate

3.0 g (21.56 mmol) (lS)-l-(4-fluorophenyl)ethylamine and 4.5 ml (25.87 mmol) diisopropylethylamine are dissolved in 20 ml dichloromethane and cooled to 0 0 C. 4.56 g (22.63 mmol) 4-nitrophenylchloroformate is added and after 5 min the cooling bath is removed. Stirring is continued for 1.5 h until 4 g (10.35 mmol) ter/-butyl (2-{[4-(benzyloxy)-3-methoxybenzyl]- amino}ethyl)carbamate is added to the suspension. Additional 2.7 ml (15.5 mmol) diisopropylethylamine is added and stirring is continued for 3 h at rt before the reaction mixture is poured on water. The mixture is extracted three times with ethyl acetate and the combined organic layers are washed with buffer solution (pH 7) and brine, dried over sodium sulfate and concentrated in vacuo. The product is isolated from the crude mixture by chromatography on silica gel (gradient dichloromethane to dichloromethane/methanol 100:1) followed by chromatography on silica gel (gradient cyclohexane / ethyl acetate 5:1 - 3: 1 - 2: 1) to yield 2.5 g (44% of th.) of the title compound.

LC-MS (method 1): R 1 = 2.73 min, m/z = 552 (M+H) +

1 H-NMR (400 MHz, DMSO-d 6 ): δ = 7.47-7.31 (m, 6H), 7.10 (t, 2H), 6.97 (d, IH), 6.89 (t, IH), 6.79 (s, IH), 6.75 (d, IH), 6.68 (d, IH), 5.05 (s, 2H), 4.89 (m, IH), 4.48 (s, 2H), 3.68 (s, 3H), 3.23- 3.15 (m, 2H), 3.02-2.94 (m, 2H), 1.38 (s, 12H).

Using an analogous procedure the following compounds are prepared:

Example 48

tert-butyl {2-[{4-[(2,4-difluorobenzyl)oxy]-3-methoxybenzyl}({[(l S)-l -(4-fluorophenyl)ethyl]- arnino}carbonyl)amino]ethyl}carbamate

620 mg (4.46 mmol) (15)-l-(4-fluorophenyl)ethylamine and 0.93 ml (5.37 mmol) diisopropyl- ethylamine are dissolved in 10 ml dichloromethane and cooled to 0 0 C. 943 mg (4.68 mmol) 4- nitrophenylchloroformate is added and after 5 min the cooling bath is removed. Stirring is continued for 1.5 h until 750 mg (1.78 mmol) ter/-butyl [2-({4-[(2,4-difluorobenzyl)oxy]-3- methoxybenzyl}amino)ethyl]carbamate and 0.79 ml (4.44. mmol) diisopropylethylamine dissolved in 2.5 ml dichloromethane are added to the suspension. The mixture is stirred at rt before poured on IN hydrochloric acid. The aqueous layer is extracted with dichloromethane and the combined organic layers are washed with brine, dried over magnesium sulfate and concentrated in vacuo. The product is isolated from the crude mixture by chromatography on silica gel (gradient dichloromethane to dichloromethane/methanol 100: 1) to yield 904 mg (87% of th.) of the title compound.

LC-MS (method 1 ): R 1 = 2.78 min, m/z = 588 (M+H) +

1 H-NMR (400 MHz, DMSO-d 6 ): δ = 7.60 (q, IH), 7.38-7.28 (m, 3H), 7.17-7.07 (m, 3H), 7.00 (d, IH), 6.89 (t, IH), 6.80 (s, IH), 6.76 (d, IH), 6.70 (d, IH), 5.05 (s, 2H), 4.90 (m, IH), 4.48 (s, 2H), 3.65 (s, 3H), 3.22-3.15 (m, 2H), 3.02-2.95 (m, 2H), 1.37 (s, 12H).

Example 49

N-(2-aminoethyl)-N-{4-[(2,4-difluorobenzyl)oxy]-3-methoxy benzyl}-N'-phenylurea trifluoroacetate

To a solution of 448 (0.83 mmol) tert-buty\ [2-((anilinocarbonyl){4-[(2,4-difluorobenzyl)oxy]-3- methoxybenzyl}amino)ethyl]carbamate in 5 ml dichloromethane is added at 0 0 C 0.9 ml TFA. The mixture is allowed to warm to rt, before evaporated to dryness. The residual oil is treated with petroleum ether. Supernatant layer is discarded and the residue is dissolved in diethyl ether, again the supernatant layer is discarded and the oily residue dried at high vacuum. After chromatography (silica gel 60; dichloromethane / methanol 20:1) 126 mg (25% of th.) of the title compound are obtained.

HPLC (method 7): R 1 = 4.31 min;

MS (ESI+): m/z = 442 (M+H) +

1 H-NMR (300 MHz, DMSO-d 6 ): δ = 8.54 (s, IH), 7.72 (bs, 2H), 7.59 (m, IH), 7.49 (d, 2H), 7.36- 7.19 (m, 3H), 7.13 (m, IH), 7.08 (d, I H), 6.99 (t, IH), 6.91 (s, IH), 6.79 (dd, IH), 5.05 (s, 2H), 4.59 (s, 2H), 3.73 (s, 3H), 3.48 (m, 2H), 2.95 (m, 2H).

Example 50

N-(2-aminoethyl)-N-{4-[(2-fluorobenzyl)oxy]-3-methoxybenz yl}-N'-phenylurea trifluoroacetate

To a solution of 300 mg (0.57 mmol) tert-bulyl [2-((anilinocarbonyl){4-[(2-fluorobenzyl)oxy]-3- methoxybenzyl}amino)ethyl]carbamate in 10 ml dichloromethane is added at 0 0 C 10 ml TFA. After 15 min at 0 0 C the mixture is allowed to warm to rt, before evaporated to dryness. Chromatography (dichloromethane / methanol 20: 1) affords 95 mg (31% of th.) of the title compound.

HPLC (method 7): R, = 4.28 min;

MS (ESI+): m/z = 424 (M+H) +

1 H-NMR (300 MHz, DMSO-d 6 ): δ = 7.54 (m, IH), 7.48-7.37 (m, 3H), 7.31-7.18 (m, 4H), 7.06 (d, IH), 6.99-6.91 (m, 2H), 6.81 (dd, IH), 5.08 (s, 2H), 4.52 (s, 2H), 3.72 (s, 3H), 3.38 (m, 2H), 2.83 (m, 2H).

Example 51

N-(2-aminoethyl)-N-{4-[(2-fluorobenzyl)oxy]-3-methoxybenz yl}-N'-phenylurea

150 (0.29 mmol) tert-butyl [2-((amlinocarbonyl){4-[(2-fluorobenzyl)oxy]-3-methoxybenzyl }- amino)ethyl]carbamate are treated with 30 ml 4N hydrogen chloride in dioxane, the mixture is stirred for 2 h, before evaporated to dryness. Diethyl ether is added to the residue and decanted. The residual oil is purified by chromatography (dichloromethane / methanol 9: 1) to afford 4.3 mg (3.3% of th.) of the title compound.

HPLC (method 7): R, = 4.26 min;

MS (ESI+): m/z = 424 (M+H) +

1 H-NMR (400 MHz, DMSOd 6 ): δ = 10.00 (bs, I H), 7.54 (t, IH), 7.47-7.37 (m, 3H), 7.29-7.18 (m, 4H), 7.05 (d, I H), 6.95 (s, IH), 6.91 (d, IH), 6.81 (d, IH), 5.08 (s, 2H), 4.45 (s, 2H), 3.74 (s, 3H), 3.28 (m, 2H), 2.72 (t, 2H).

Example 52

N-(2-aminoethyl)-N-{4-[(2,4-difluorobenzyl)oxy]-3-methoxy benzyl}-N'-phenylurea

To a solution of 326 (0.602 mmol) tert-butyl [2-((anilinocarbonyl){4-[(2,4-difluorobenzyl)oxy]-3- methoxybenzyl}amino)ethyl]carbamate in 7 ml dichloromethane is added at 0 0 C 7 ml TFA. The mixture is allowed to warm to rt, before evaporated to dryness. The residual oil is treated with petroleum ether. Supernatant layer is discarded and the residue is dissolved in diethyl ether, again the supernatant layer is discarded and the oily residue dried at high vacuum. After chromatography (silica gel 60; dichloromethane / methanol 20: 1) 53 mg (19% of th.) of the title compound are obtained.

HPLC (method 7): R 1 = 4.33 min;

MS (ESI+): m/z = 442 (M+H) +

1 H-NMR (300 MHz, DMSO-d 6 ): δ = 10.1 1 (bs, IH), 7.59 (m, IH), 7.42 (d, 2H), 7.36-7.19 (m, 3H), 7.13 (m, I H), 7.05 (d, IH), 6.95 (s, I H), 6.91 (d, IH), 6.82 (dd, I H), 5.05 (s, 2H), 4.45 (s, 2H), 3.73 (s, 3H), 3.24 (m, 2H), 2.71 (t, 2H).

Example 53

N-(2-aminoethyl)-N'-(2-chlorophenyl)-N-{4-[(2,4-difluorob enzyl)oxy]-3-methoxybenzyl}urea

To a solution of 265 (0.46 mmol) tert-butyl [2-({[(2-chlorophenyl)amino]carbonyl} {4-[(2,4- difluorobenzyl)oxy]-3-methoxybenzyl}amino)ethyl]carbamate in 6 ml dichloromethane is added at 0°C 6 ml TFA. The mixture is allowed to warm to rt and stirred for 1 h, before evaporated to dryness. The residual oil is treated with petroleum ether. The supernatant layer is discarded and the oily residue dried at high vacuum. The residue is dissolved in dichloromethane and washed with IN sodium hydroxide solution, dried over magnesium sulfate and concentrated in vacuo to afford 96.5 mg (40% of th.) of the title compound.

HPLC (method 7): R, = 4.45 min;

MS (ESI+): m/z = 475 (M+H) +

1 H-NMR (300 MHz, DMSO-d 6 ): δ = 7.75 (d, IH), 7.59 (m, IH), 7.42 (d, IH), 7.36-7.22 (m, 2H), 7.13 (m, I H), 7.05 (d, 2H), 6.99 (s, I H), 6.85 (d, IH), 5.05 (s, 2H), 4.45 (s, 2H), 3.73 (s, 3H), 3.15 (m, 2H), 2.73 (m, 2H).

The following examples are prepared from the corresponding Boc protected starting materials (examples cf. above) using analogous procedures:

General procedure D: cleavage of the Boc group

A stirred solution of a BOC derivative in 1 ,4-dioxane (approx. lg/ml) is treated with approx. 10 equivalents of hydrochloric acid in 1 ,4-dioxane (4 molar solution) at rt for approx. 1 h. Methanol is added to the mixture until a homogeneous solution is obtained. The product is isolated from the crude solution after purification by reverse phase HPLC (water-acetonitrile gradient) and concentration in vacuo as a hydrochloric acid salt.

The following examples are prepared according to general procedure D from the corresponding BOC protected starting materials (examples cf. above):

=

=

N-(3-aminopropyl)-N-[4-(benzyloxy)-3-methoxybenzyl]-N'-(2 -methylphenyl)urea

300 mg (0.30 mmol) loaded resin (approx. 1 mmol/g loading) are suspended in 5 ml ethyl acetate. 80 mg (0.60 mmol) 1 -isocyanato-2-methylbenzene are added. The mixture is shaken at rt for one hour. The mixture is filtered and the remaining resin is washed three times with small portions of DMF and afterwards three times alternately with small portions of methanol and dichloromethane. The resin is treated with a solution of 1 % (v/v) TFA in dichloromethane for 15 min. The cleavage mixture is filtered and the resin washed twice with dichloromethane. The filtrate is concentrated in vacuo and the residue is purified via prep. HPLC to yield 5 mg (4% of th. based on calculated initial loading) of the title compound.

LC-MS (method 5): R, = 1.80 min, m/z = 434 (M+H) +

1 H-NMR (400 MHz, DMSO-d 6 ): δ = 8.00 (bs, IH), 7.65 (bs, 2H), 7.50-5.80 (m, 12H), 5.10 (s, 2H), 4.50 (s, 2H), 3.80 (s, 3H), 3,40 (m, 2H, assumed signal, disguised by water signal), 2.80 (m, 2H), 2.10 (m, 3H), 1.80 (m, 2H).

Example 96

N-(2-aminoethyl)-N-[4-(benzyloxy)-3-methoxybenzyl]-N'-(4- fluorophenyl)urea

2.48 g (3.725 mmol) N-[4-(benzyloxy)-3-methoxybenzyl]-N'-4-fluoro-phenyl-N-[2-(t ritylamino)- ethyl]urea are dissolved in 20 ml dichloromethane and treated at rt in portions with a total of 5 ml TFA. After reaction is complete the yellow reaction mixture is treated with 10 ml methanol (mixture becomes colorless), diluted with dichloromethane and washed with saturated sodium carbonate solution and brine, dried over magnesium sulfate and concentrated in vacuo. The residue is purified by column chromatography (silica gel, gradient: dichloromethane - dichloromethane / methanol 20: 1 to 5: 1) to yield 1.0 g (63% of th.) of the title compound.

LC-MS (method 6): R, = 1.59 min, m/z = 424 (M+H) +

Example 97

N-(2-aminoethyl)-N-[4-(benzyloxy)-3-methoxybenzyl]-N'-(4- fluorophenyl)urea hydrochloride

1.0 g (2.36 mmol) N-(2-aminoethyl)-N-[4-(benzyloxy)-3-methoxybenzyl]-N'-(4-flu orophenyl)urea are dissolved in 10 ml dioxane and treated at rt with 0.59 ml 4M hydrochloric acid in dioxane. After 10 min the mixture is evaporated to dryness. The residue is re-dissolved in a small amount of methanol and purified directly by reverse phase HPLC (water/acetonitril) to yield 752 mg (69% of th.) of the title compound.

LC-MS (method 6): R 1 = 1.69 min, m/z = 424 (M-HC1+H) +

1 H-NMR (400 MHz, DMSOd 6 ): δ = 8.71 (s, IH), 8.02 (bs, 3H), 7.60-7.52 (m, 2H), 7.45-7.30 (m, 5H), 7.09 (t, IH), 7.03 (d, IH), 6.95 (m, IH), 6.80 (d, IH), 5.05 (s, 2H), 4.55 (s, 2H), 3.76 (s, 3H), 3.49 (t, 2H), 2.94 (m, 2H).

Example 98

N-(2-aminoethyl)-N-[4-(benzyloxy)-3-methoxybenzyl]-N'-[(1 5)-l -(phenyl)ethyl]urea hydrochloride

760 mg (1.42 mmol) tert-butyl {2-[[4-(benzyloxy)-3-methoxybenzyl]({[(lS)-l -phenylethyl]amino}- carbonyl)amino]ethyl}carbamate are dissolved in 3.8 ml 1 ,4-dioxane. The solution is treated with 3.5 ml of hydrochloric acid in 1 ,4-dioxane (4 molar solution) at rt for 1 h. The product is isolated from the crude mixture after direct purification by reverse phase HPLC (water - acetonitrile gradient) to yield after evaporation of solvents 467 mg (68% of th.) of the title compound.

LC-MS (method 5): R, = 1.76 min, m/z = 434 (M-HC1+H) +

1 H-NMR (400 MHz, DMSO-d 6 ): δ = 7.91 (bs, 3H), 7.48-7.18 (m, 10H), 6.99 (m, 2H), 6.86 (s, IH), 6.72 (d, I H), 5.08 (s, 2H), 4.94 (m, IH), 4.45 (q, I H), 3.70 (s, 3H), 3.50-3.3 (m, 2H), 2.87-2.81 (m, 2H), 1.40 (d, 3H).

Example 99

N-(2-aminoethyl)-N-[4-(benzyloxy)-3-methoxybenzyl]-N'-[(1 5)-l-(4-fluorophenyl)ethyl]urea hydrochloride

1600 mg (2.9 mmol) tert-butyl {2-[[4-(benzyloxy)-3-methoxybenzyl]({[(lS)-l-(4-fluorophenyl )- ethyl]amino}carbonyl)amino]ethyl}carbamate are dissolved in 5 ml 1 ,4-dioxane. The solution is treated with 7.25 ml of hydrochloric acid in 1 ,4-dioxane (4 molar solution) at rt for 1 h. The product is isolated from the crude mixture after direct purification by reverse phase HPLC (water- acetonitrile gradient) to yield after evaporation of solvents 891 mg (63% of th.) of the title compound.

LC-MS (method 5): R, = 1.79 min, m/z = 452 (M-HC1+H) +

1 H-NMR (400 MHz, DMSOd 6 ): δ = 7.90 (bs, 3H), 7.46-7.31 (m, 7H), 7.12-6.97 (m, 4H), 6.82 (s, I H), 6.71 (d, I H), 5.07 (s, 2H), 4.93 (m, IH), 4.45 (q, IH), 3.70 (s, 3H), 3.47-3.3 (m, 2H), 2.85- 2.80 (m, 2H), 1.38 (d, 3H).

Example 100

N-(2-aminoethyl)-N-[4-(benzyloxy)-3-chlorobenzyl]-N'-(4-f luorophenyl)urea

N-[4-(benzyloxy)-3-chlorobenzyl]-l ,2-diaminoethane trityl resin (approx. 0.8 mmol loading on resin) is suspended in dichloromethane and treated at rt with 0.175 ml (1.52 mmol) 4- fluorophenylisocyante. The mixture is gentile agitated for 1 h at rt until more dichloromethane is added. The mixture is filtrated and the resin washed three times with dichloromethane. After addition of 4 ml dichloromethane the resin is gentile agitated and treated with 0.32 ml TFA at rt, shaking is continued before the mixture is filtrated and the resin washed with dichloromethane (filtrates are collected and combined). The cleavage procedure is repeated once and all filtrates are

combined and washed with saturated sodium carbonate solution and with brine, dried over magnesium sulfate and concentrated in vacuo. The product is isolated from the crude mixture by chromatography on silica gel (gradient dichloromethane to dichloromethane/methanol 500:1 ) to yield 220 mg (64% of th. from calculated resin loading) of the title compound.

LC-MS (method 6): R 1 = 1.86 min, m/z = 428 (M+H) +

1 H-NMR (300 MHz, DMSOd 6 ): δ = 7.49-7.32 (m, 8H), 7.20 (m, 2H), 7.08 (m, 2H), 5.19 (s, 2H), 4.92 (s, 2H), 3.22 (m, 2H), 2.70 (t, 2H).

Example 101

N-(2-aminoethyl)-N-[4-(benzyloxy)-3-chlorobenzyl]-N'-(4-f luorophenyl)urea hydrochloride

195 mg (0.456 mmol) N-(2-aminoethyl)-N-[4-(benzyloxy)-3-chlorobenzyl]-N'-(4-fluo rophenyl)- urea are dissolved in 2 ml diethyl ether and treated at rt with 0.455 ml (0.455 mmol) IN hydrochloric acid in diethyl ether. The mixture is concentrated in vacuo to give an oily residue. The product is isolated as the hydrochloride from the crude mixture after direct purification by reverse phase HPLC (water - acetonitrile gradient).

LC-MS (method 1): R, = 1.68 min, m/z = 428 (M-HC1+H) +

1 H-NMR (300 MHz, DMSO-d 6 ): δ = 8.68 (s, IH), 7.92 (bs, 3H), 7.57-7.30 (m, 7H), 7.22 (m, 2H), 7.08 (t, 2H), 5.20 (s, 2H), 4.55 (s, 2H), 3.50 (m, 2H), 2.95 (m, 2H).

Example 102

N-(2-aminoethyl)-N- {4-[(2,4-difiuorobenzyl)oxy]-3-methoxybenzyl } -N'-[( 1 S)- 1 -(4-fluorophenyl)- ethyl]urea hydrochloride

904 mg (1.54 mmol) tert-butyl {2-[{4-[(2,4-difluorobenzyl)oxy]-3-methoxybenzyl}({[(l S)-l-(4- fluorophenyl)ethyl]amino}carbonyl)amino]ethyl}carbamate are dissolved in 4.5 ml 1 ,4-dioxane. The solution is treated with 3.8 ml hydrochloric acid in 1 ,4-dioxane (4 molar solution) at rt for 1 h. The product is isolated from the crude mixture after direct purification by reverse phase HPLC (water - acetonitrile gradient) to yield after evaporation of solvents 425 mg (53% of th.) of the title compound.

LC-MS (method 1): R, = 1.55 min, m/z = 488 (M-HC1+H) +

1 H-NMR (400 MHz, DMSO-d 6 ): δ = 7.90 (bs, 3H), 7.59 (q, IH), 7.49-7.28 (m, 3H), 7.18-6.97 (m, 5H), 6.82 (s, IH), 6.73 (d, I H), 5.07 (s, 2H), 4.93 (m, IH), 4.45 (dd, IH), 3.69 (s, 3H), 3.48-3.30 (m, 2H), 2.85 (m, 2H), 1.39 (d, 3H).

Example 103

N-(2-aminoethyl)-N-[4-(benzyloxy)-3-chlorobenzyl]-N'-[(lS )-l-(4-fluorophenyl)ethyl]urea

350 mg (2.52 mmol) (S)-I -(4-fluorphenyl)ethylamine are dissolved in 10 ml dichloromethane and cooled to 0 0 C. 532 mg (2.64 mmol) 4-nitrophenylchloroformate were added at once, the cooling bath is removed after 5 min and the mixture is stirred for 1.5 h (warmed up to rt and a slid gradually precipitates). 1.8 g N-[4-(benzyloxy)-3-chlorobenzyl]-l ,2-diaminoethane trityl resin (approx. 1 mmol loading on resin) is added at rt and stirring is continued for 3 h. The mixture is filtered and the remaining resin is washed five times with dichloromethane. The resin is suspended in 10 ml dichloromethane, treated with 2 ml TFA (resin turns dark-red) at rt with gentile shaking and further agitated for 10 min. The cleavage mixture is filtrated and the resin washed four times

with dichloromethane. The filtrates are collected and combined. The cleavage procedure is repeated once and all filtrates are combined and washed three times with saturated sodium carbonate solution, water and brine, dried over magnesium sulfate and concentrated in vacuo to yield after evaporation of solvents 687 mg (84% of th. based on calculated initial loading) of the title compound. The product is used without further purification.

LC-MS (method 5): R 1 = 1.97 min, m/z = 456 (M-HCHH) +

1 H-NMR (400 MHz, DMSOd 6 ): δ = 7.48-7.08 (m, 13H), 5.19 (s, 2H), 4.85 (m, IH), 4.35 (dd, 2H), 3.13 (m, 2H), 2.51 (m, 2H), 1.62 (bs, 2H), 1.35 (d, 3H).

Example 104

N-(2-aminoethyl)-N-[4-(benzyloxy)-3-chlorobenzyl]-N'-[(15 )-l-(4-fluorophenyl)ethyl]urea hydrochloride

680 mg (1.49 mmol) N-(2-aminoethyl)-N-[4-(benzyloxy)-3-chlorobenzyl]-N'-[(lS)-l -(4-fluoro- phenyl)ethyl]urea are dissolved in 2 ml 1,4-dioxane. The solution is treated with 0.38 ml of hydrochloric acid in 1 ,4-dioxane (4 molar solution) at rt for 10 min, before volatile components are removed in vacuo. The product is isolated by direct purification via reverse phase HPLC (water - acetonitrile gradient) to yield after evaporation of solvents 523 mg (71% of th.) of the title compound.

LC-MS (method 5): R t = 1.99 min, m/z = 456 (M-HC1+H) +

1 H-NMR (400 MHz, DMSOd 6 ): δ = 7.98 (bs, 3H), 7.48-7.20 (m, 9H), 7.15-7.01 (m, 4H), 5.21 (s, 2H), 4.90 (m, I H), 4.44 (dd, 2H), 3.50-3.30 (m, 2H), 2.86 (m, 2H), 1.39 (d, 3H).

B. Evaluation of physiological activity

The potential Cold Menthol Receptor - 1 (CMR-I) antagonistic activity of the compounds of the invention may be demonstrated, for example, using the following assays:

Measurement of the menthol-induced Ca 2+ influx in HEK293 Cell expressing CMR-I receptor (Assay 1)

A cell-based calcium influx assay using HEK293 cells stably expressing human CMR-I is used to identify CMR-I receptor-antagonists. Menthol, a CMR-I specific agonist, is used for stimulation of these cells, inducing an increase in intracellular calcium. This menthol-induced Ca 2+ increase is traced by fluorescence measurement. Therefore the cells are loaded with fluo4-AM prior to stimulation. For testing inhibitors the cells are preincubated with various concentrations of the compound before menthol stimulation. The potency of potential CMR-I inhibitors is quantified by measuring decrease of fluorescence .

Table A

Measurement of the menthol-induced Ca 2+ influx in primary cultured rat dorsal root ganglia neurons (Assay 2)

Since CMR-I is expressed on DRG (C-fibers), in which this receptor mediates the altered afferent information in overactive bladder; primary cultures of rat DRG are used as functional in vitro test. Stimulation of the cells is done with menthol and cold and the induced calcium influx is quantified by fluorescence in the presence or absence of CMR-I inhibitors.

Preparation of primary cultured rat DRG neurons: DRG are prepared from Zucker rats (30 days in age) and neuronal cells are dispersed in 0.1% collagenase. After removal of Schwann cells by

adhering to a culture plate, non-adherent neuronal cells are recovered and cultured on laminin- and poly-D-lysine coated 384 well plates for 2 days in the presence of 50 ng/ml rat NGF and 50 μM 5- fiuorodeoxyuridine.

Measurement of Ca 2+ : Rat DRG neurons are suspended in a culture medium and seeded into 384- well plates (black walled clear-base / Nalge Nunc International). Following the culture for 48 hrs the medium is changed to 2 μM Fluo-4 AM (Molecular Probes) and 0.02% Puronic F-127 in assay buffer (Hank's balanced salt solution (HBSS), 17 mM HEPES (pH7.4), 1 mM Probenecid, 0.1% bovine serum albumin (BSA)) and the cells are incubated for 60 min at 25°C. After washing twice with assay buffer the cells are incubated with a test compound or vehicle (dimethylsulfoxide) for 20 min at 25°C. The fluorescence change indicating mobilization of cytoplasmic Ca 2+ is measured for 60 sec after the stimulation with 50 μM menthol. The fluorescence change is calculated in the samples treated with a test compound and vehicle respectively. Inhibitory effect of the compound is calculated by a comparison of the values.

Measurement of the micturition frequency in guinea pigs in vivo (Assay 3)

Experiments are performed according to the principles of the national law for the protection of laboratory. Female Guinea Pigs (300-350g) are anaesthetized with urethane (1 mg/kg i.p.). A midline abdominal incision is performed, both ureters are exposed and ligated, a catheter is implanted in the bladder pole and the abdomen is closed. For administration of the compounds the vena jugularis is exposed and canulated with a catheter. After this surgery the bladder catheter is connected via a t-shaped tube to an infusion pump (Braun Perfusor® compact) and to a pressure transducer (BioResearch Center, MLT0698, Nagoya). Saline is infused and intrabladder pressure is registered. After 1 h of equilibration period and the establishment of constant voiding cycles, menthol (0.6 mM) is added to the infused saline. At this point also vehicle (control group) or CMR-I inhibitors are administered i.v. as bolus injection. The effect of treatment on the micturition interval (corresponding to bladder capacity) and micturition pressure is calculated and compared between vehicle-treated and compound-treated groups.

C ; Operative examples relating to pharmaceutical compositions

The compounds according to the invention can be converted into pharmaceutical preparations as follows:

Tablet

Composition:

100 mg of the compound of Example 1 , 50 mg of lactose (monohydrate), 50 mg of maize starch (native), 10 mg of polyvinylpyrrolidone (PVP 25) (from BASF, Ludwigshafen, Germany) and 2 mg of magnesium stearate.

Tablet weight 212 mg, diameter 8 mm, curvature radius 12 mm.

Preparation:

The mixture of active component, lactose and starch is granulated with a 5% solution (m/m) of the PVP in water. After drying, the granules are mixed with magnesium stearate for 5 min. This mixture is moulded using a customary tablet press (tablet format, see above). The moulding force applied is typically 15 kN.

Orally administrable suspension

Composition:

1000 mg of the compound of Example 1 , 1000 mg of ethanol (96%), 400 mg of Rhodigel (xanthan gum from FMC, Pennsylvania, USA) and 99 g of water.

A single dose of 100 mg of the compound according to the invention is provided by 10 ml of oral suspension.

Preparation:

The Rhodigel is suspended in ethanol and the active component is added to the suspension. The water is added with stirring. Stirring is continued for about 6h until the swelling of the Rhodigel is complete.

Solution for intravenous administration:

Composition:

1 mg of the compound of Example 1 , 15 g of polyethylene glycol 400 and 250 g of water for injection.

Production:

The compound of Example 1 is dissolved with polyethylene glycol 400 in the water with stirring., The solution is sterilized by filtration (pore diameter 0.22 μm) and dispensed under aseptic conditions into heat-sterilized infusion bottles. These are closed with infusion stoppers and crimped caps.