Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
BIOCATALYSTS WITH AMINE ACYLASE ACTIVITY
Document Type and Number:
WIPO Patent Application WO/1997/041214
Kind Code:
A1
Abstract:
The invention belongs to the field of biotechnology. It concerns a biocatalyst, i.e. a dead or living microorganism or a polypeptide, preferably in isolated form, which exhibits acylase enzymatic activity without lipase- or esterase-activity. The biocatalyst is capable of stereoselectively hydrolysing a racemic acylamide which has an aliphatic acyl residue and which is not a derivative of a natural amino acid.

Inventors:
GHISALBA ORESTE (CH)
KITTELMANN MATTHIAS (DE)
LAUMEN KURT (DE)
WALSER-VOLKEN PAULA (CH)
Application Number:
PCT/EP1997/001866
Publication Date:
November 06, 1997
Filing Date:
April 14, 1997
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CIBA GEIGY AG (CH)
GHISALBA ORESTE (CH)
KITTELMANN MATTHIAS (DE)
LAUMEN KURT (DE)
WALSER VOLKEN PAULA (CH)
International Classes:
C12N9/80; C12P41/00; C12R1/01; C12R1/06; (IPC1-7): C12N9/80; C12P41/00
Domestic Patent References:
WO1982000253A11982-02-04
Foreign References:
EP0399589A11990-11-28
FR2438054A11980-04-30
EP0433117A11991-06-19
GB2246774A1992-02-12
Other References:
DATABASE WPI Section Ch Week 9435, Derwent World Patents Index; Class B02, AN 94-283347, XP002038271
Download PDF:
Claims:
Claims:
1. A biocataiyst exhibiting amine acylase enzymatic activity without lipase or esterase activity, which biocataiyst is capable of sterereoselectively hydrolysing a racemic acylamide of formula (1) wherein R2 is aryl or dC4aryl; or R1 and R2 together are forming a 5 to 7 membered ring substituted by or fused to aryl; R3 is an aliphatic acyl residue; and R1 and R3 together are chosen in order to form a compound of formula (1 ) that is not a derivative of a natural amino acid; wherein each of the residues can be substituted or unsubstituted.
2. The biocataiyst according to claim 1 selected from the group of (a) a polypeptide with said enzymatic activity and (b) a living microorganism or dead microorganism containing a polypeptide with said enzymatic activity or a cell extract of such a microorganism.
3. The biocataiyst according to claim 1 which is an enzyme with acylase activity and without lipase or esteraseactivity, and mutants, variants, and fragments of said acylase which exhibit acylase activity without lipase or esteraseactivity.
4. The biocataiyst according to claim 1 , capable of catalyzing reaction A R2 O R2 O R2 o * M amine acylase t II * II *%%> — ϊζδ — „.,<« . ff * Rc*NAR3 R (or S) amine acid S (or R) amide (1) (2) (3) (4) whereby in the formula (1) to (4) R1 is hydrogen, CιC8alkyl, C2Cβalkenyl, dCβalkoxy, C2Cθalkylcarboxy or carboxy; R2 is aryl or CιC4aryl; unsubstituted or substituted by dC4alkyl, dC4alkoxy, d C4hydroxyalkyl, CrC4aminoalkyl, CrC4haloalkyl, hydroxy, amino, halogeno, nitro, sulfo, or cayno; or wherein Ri and R2 together are forming a.
5. to 7 membered ring substituted by or fused to aryl, wherein the rings may contain one or two heteroatoms selected from nitrogen, sulfur and oxygen; and R3 is an aliphatic acyl residue.
6. 5 The biocataiyst according to claim 4 catalyzing reaction A, wherein R1 is hydrogen, d C4alkyl, C2C4alkenyl, dC4alkoxy or C2C4alkylcarboxy.
7. The biocataiyst according to claim 4 catalyzing reaction A, wherein R1 is hydrogen, d C3alkyl, C2C3alkenyl, CιC3alkoxy or C2C3alkylcarboxy.
8. The biocataiyst according to claim 4 catalyzing reaction A, wherein R1 is selected from the group consisting of hydrogen, methyl, ethyl, npropyl, isopropyl, propyl1en, propyl2en, prop2ylen, and methoxy.
9. The biocataiyst according to claim 4 catalyzing reaction A, wherein R 3 is dC4alkyl.
10. The biocataiyst according to claim 4 catalyzing reaction A, wherein R 3 is methyl.
11. The biocataiyst according to claim 4 catalyzing reaction A, wherein aryl is a single or double ring system having from 3 to 10 ring atoms, is bonded via a carbon atom or via a nitrogen atom and contains up to 4 hetero atoms selected from oxygen, nitrogen, sulfur, and sulfur linked to 1 or 2 oxygen atoms; which in addition may also be fused with 1 or 2 phenyl radicals or with 1 or 2 cycioalkyl radicals, cycioalkyl preferably having from 5 to 7 ring atoms; and which may be unsaturated or partially or fully saturated.
12. The biocataiyst according to claim 4 catalyzing reaction A, wherein aryl is phenyl, napthyl, biphenylyl, anthryl, fluorenyl, thienyl, furyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, thiazolyl, tetrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolyl, benzimidazolyl, quinolyl, isoquinolyl, 3,1benzofuranyl, chromanyl, cyclohexa[b]pyrrolyl, cyclohexa[b]pyridyl, [bjpyrimidinyl, pyrrolidinyl, pyrrolinyl, cyclohexa[b]pyrazinyl, cyclo hexa[b]pyrimidinyl, imidazolidyl, piperidyl, piperazinyl, morpholinyl, thiomorpholinyl, S,S dioxothiomorpholinyl, indolinyl, isoindolinyl, 4,5,6,7tetrahydro indolyl, 1 ,2,3,4 tetrahydroquinolyl or 1 ,2,3,4tetrahydroisoquinolyl.
13. The biocataiyst according to claim 4 catalyzing reaction A, wherein aryl is unsubstituted or substituted by one or more substituents selected from methyl, hydroxymethyl, 2 hydroxyethyl, hydroxy, methoxy; ethoxy, amino, methylamino, ethylamino, tertbutyl amino, dimethylamino, diethylamino, carboxy, methoxycarbonyl, isopropoxycarbonyl, secbutoxycarbonyl, tertbutoxycarbonyl, fluorine, chlorine, bromine, acetyl or pivaloyl, nitro, oxo and/or by cyano.
14. The biocataiyst according to claim 4 catalyzing reaction A, wherein R2 and R3 together form indane, tetraline or chromane, unsubstituted or substituted with methoxy, ethoxy, halogeno, methyl, ethyl, nitro, cyano, amino, hydroxy, trfluoromethyl.
15. The biocataiyst according to claim 4 catalyzing reaction A, wherein R2 is selected from the group consisting of (10) (11) (12) wherein R4 is hydrogen or dC4alkyl; n is an integral number selected from 0, 1 , 2, 3 and 4; R5 and R6 are each independent of each other hydrogen, dC4alkyl, d C4haloalkyl, halogeno, amino, cyano, nitro or C 1C4alkoxy.
16. The biocataiyst according to claim 4 catalyzing reaction A, wherein R2 is selected from the group consisting of (10) (11) (12) wherein R4 is hydrogen or methyl; n is an integral number selected from 0, 1 , 2 and 3; R5 and R6 are each independent of each other hydrogen, methyl, ethyl, trifluoromethyl, fluorine, chlorine, bromine, iodine, amino, nitro, cyano or methoxy.
17. The biocataiyst according to claim 4 catalyzing reaction A, wherein R2 is selected from the group consisting of (10) (11) (12) wherein R4 is hydrogen; n is an integral number selected from 0, 1 and 2; R5 and R6 are each independent of each other hydrogen, methyl, fluorine, chlorine, cyano, nitro or methoxy.
18. The biocataiyst according to claim 4 catalyzing reaction A, wherein R2 is selected from the group consisting of (13) (14) (15) wherein n is an integral number selected from 0, 1 and 2; R5 and R6 are each independent of each other hydrogen or cyano.
19. The biocataiyst according to claim 4 catalyzing reaction A, wherein R1 and R2 form together an unsubstituted or substituted cyclic heteroaliphatic radical selected from the group consisting of (16) (17) wherein X is oxygen, carbon or nitrogen; and R5 and R6 are each independent of each other hydrogen, dC4alkyl, CrC4haloalkyl, halogeno, amino, cyano, nitro or d dalkoxy.
20. The biocataiyst according to claim 4 catalyzing reaction A, wherein R1 and R2 form together an unsubstituted or substituted cyclic heteroaliphatic radical selected from the group consisting of (16) (17) wherein X is oxygen or carbon; and R5 and R6 are each independent of each other hydrogen, methyl, ethyl, iodo, bromo, trifluoromethyl, chloro, fluoro, amino, cyano, nitro, methoxy or ethoxy.
21. The biocataiyst according to claim 1 obtainable by a selection process comprising inoculating a selection medium with natural samples such as soil, water or plant silage, said selection medium comprising an acylamide as sole carbon source, which is used as racemate or, if a preselection on a R or Senantiomer specific acylase is intended, as R or Senantiomer.
22. The biocataiyst according to claim 1 obtainable by a selection process comprising inoculating a selection medium with natural samples such as soil, water or plant silage, said selection medium consisting of an acylamide as sole carbon source, which is selected from the group consisting of Nacetyl1 phenylethylamine as racemate or S or Renatiomer and Nacetyl2amino1phenyl4pentene as racemate or S or R enatiomer.
23. The biocataiyst according to claim 1 obtainable from a microorganism selected from the group consisting of Rhodococcus globerulus, Rhodococcus equi, and Arthrobacter aurescens.
24. The biocataiyst according to claim 1 obtainable from a microorganism selected from the group consisting of Rhodococcus globerulus K1/1 , DSM 10337, Rhodococcus equi Ac6, DSM 10278, and Arthrobacter aurescens AcR5b, DSM 10280.
25. The biocataiyst according to claim 1 which is a microorganism.
26. The biocataiyst according to claim 24 which is selected from the group consisting of genetically modified microorganisms and naturally occurring ones.
27. The biocataiyst according to claim 24, which is a recombinant microorganism transformed with a DNA sequence coding for an enzyme according to claim 1.
28. The biocataiyst according to claim 24, selected from the group consisting of Rhodococcus globerulus, Rhodococcus equi, and Arthrobacter aurescens.
29. A biocataiyst according to claim 27 selected from the group consisting of Rhodococcus globerulus K1/1 , DSM 10337, Rhodococcus equi Ac6, DSM 10278, and Arthrobacter aurescens AcR5b, DSM 10280.
30. A substantially purified biocataiyst according to claim 1 , isolated from a microorganism according to claim 27.
31. A substantially purified biocataiyst according to claim 1 , comprising an amino acid sequence selected from the group consisting of SEQ ID NOs 1 , 2, 3 and 4.
32. A process comprising the hydrolysis of a racemic Nacylamide acylamide which has an aliphatic acyl residue and which is not a derivative of a natural amino acid, characterized in that a biocataiyst according to claim 1 is used.
33. The process according to claim 31 , which has the following reaction scheme, R (or S) amine acid S (or R) amide d) (2) (3) (4) whereby in the formula (1) to (4) R1 is hydrogen, CιC8alkyl, C2CBalkenyl, CiCβalkoxy, C2Cβalkylcarboxy or carboxy; R2 is aryl or dC4aryl; unsubstituted or substituted by dC4alkyl. dC4alkoxy, Ci C4hydroxyalkyl, ddAminoalkyl, CιC4haloalkyl, hydroxy, amino, halogeno, nitro or sulfo, cayno; or wherein Ri and R2 together are forming a 5 to 7 membered ring substituted by aryl, wherein the ring may contain one or two heteroatoms selected from nitrogen, sulfur and oxygen; and substituted by or fused to aryl; R3 is an aliphatic acyl residue.
34. The process of claim 31 in which R3 is dC3alkyl if the biocataiyst is derived from Rhodococcus globerulus K1/1 , DSM 10337, or from Rhodococcus equi Ac6, DSM 10278.
35. The process of claim 31 in which the aliphatic residue is methyl if the biocataiyst is derived from Arthrobacter aurescens AcR5b, DSM 10280.
36. The process of claim 31 in which the biocataiyst is used in immobilized form.
37. The process of claim 31 wherein the pH is from 5.5 to 10.5 and the temperature is from 10 to 65 °C.
Description:
BIOCATALYSTS WITH AMINE ACYLASE ACTIVITY

There is a high and continuous need for enantiomerically pure pharmaceutical and agrochemical drugs avoiding the side effects triggered by the therapeutically ineffective enantiomer. The present invention helps to fulfill this need by providing novel enzymes capable of stereoselectively hydrolysing a racemic acylamide.

Up to now enzymatic reactions on amines or amine-derivatives have been reported in the literature only for penicillins, cephaiosporins, glucosamines, amino acids, and their derivatives. Among the known examples in this field figure the resolution of racemic 2- aminobutanol via the hydrolysis of appropriate N-acyl-derivatives by acylases of microbial origin (Japanese Kokai JP 58-198,296 and JP 59-39,294) and the enantiomeric enrichment of different amines possessing the amino group on a secondary carbon atom through a process involving the action of omega amino acid transaminases (Stirling et al., US Patent No. 5,300,437). In JP 06-253,875 the stereospecific transfer of the amino group of L- alanine to acetophenone in the presence of e.g. Acinetobacter sp. MBA-15 (FERM P- 13432) for the production of S-1 -phenyl ethylamine is disclosed. US Patent 5,360,724 discloses the production of optically active 1 -aryl-2-amino propane by enantioselective transfer of the amino group in the racemate by means of amino acid transaminase from Bacillus mβgaterium. In DE 4332738 the production of optically active primary and secondary amines is disclosed; the process uses enantioselective acylation of a racemic amine with an activated ester as acyl donor in the presence of a hydrolase.

However, the present invention provides novel biocatalysts for enantioselective hydrolysis

Object of the invention

It is an object of the invention to provide novel biocatalysts for stereoselective hydrolysis of enantiomers in a racemic acylamide. Such biocatalysts are microorganisms capable of producing enzymes (acylases) capable of stereoselectively hydrolysing enantiomers in a racemic acylamide, or the enzymes themselves.

Moreover, it is an object to provide a process for the stereoselective hydrolysis of acylamides.

Summary of the invention

The invention concerns a biocataiyst, i.e. a dead or living microorganism or a polypeptide, preferably in isolated form, which exhibits acylase enzymatic activity without lipase- or esterase-activity. The biocataiyst is capable of stereoselectively hydrolysing a racemic acylamide which has an aliphatic acyl residue and which is not a derivative of a natural amino acid.

The invention accordingly also concerns microbial strains capable of producing an enzyme of the invention. Included are both genetically modified microorganisms and naturally occurring ones. The naturally occurring strains according to the invention are microorganisms obtainable by a selection process comprising inoculating a selection medium with a natural sample.

Another aspect of the invention is a process for the hydrolysis of a racemic N-acylamide which has an aliphatic acyl residue and which is not a derivative of a natural amino acid, characterized in that an enzyme according to the invention is used.

The process of the invention can be performed with free or immobilized enzyme, which can be used in enriched or purified form or in the form of a crude cell extract. In another embodiment of the invention a microorganism expressing an acylase of the invention is used for performing the reaction, i.e. the enzyme is in cell-bound form.

The process of the invention can be used for separating racemates, if one stereoisomer of the racemic acyl amide is specifically hydrolyzed. However, if the hydrolysis is not stereoselective (e.g. since the acylamide is not a racemate or since the enzyme hydrolyses both enantiomers of a certain racemic acylamide), the process of the invention can also be used for other purposes, e.g. in the chemistry of protecting groups.

Detailed description of the invention

The invention concerns a biocataiyst exhibiting amine acylase enzymatic activity without lipase- or esterase-activity, which biocataiyst is capable of stereoselectively hydrolysing a racemic acylamide of formula (1)

wherein R 2 is aryl or CrC 4 aryl; or R 1 and R 2 together are forming a 5 to 7 membered ring substituted by or fused to aryl;

R 3 is an aliphatic acyl residue; and

R 1 and R 3 together are chosen in order to form a compound of formula (1) that is not a derivative of a natural amino acid; wherein each of the residues can be substituted or unsubstituted.

A preferred embodiment is a biocataiyst, the substrate of which can be hydrolyzed to a primary amine. The biocataiyst according of the invention is preferably selected from the group of (a) a polypeptide with said enzymatic activity and (b) a living microorganism or dead microorganism containing a polypeptide with said enzymatic activity or a cell extract of such a microorganism. A dead microorganism in context with the present invention is e.g. a microorganism in a disintegrated form in which the cell wall and/or cell membrane is mechanically or chemically disrupted or removed.

The term without lipase- or esterase-activity in context with the present invention means that no enzymatic activity can be detected in the Api Zym Test (Bio Merieux SA, Marcy-L'Etoile, France). In this test the C4 esterase activity is tested with 2-naphtyl butyrate, C8 esterase activity with 2-naphtyl caproate, and C14 lipase activity with 2-naphtyl myristate.

In particular, the invention concerns a enzyme with amine acylase activity and without lipase- or esterase-activity and mutants, variants, and fragments of said acylase, which exhibit acylase activity and without lipase- or esterase-activity. A preferred embodiment is a polypeptide, the substrate of which can be hydrolyzed to a primary amine.

The polypeptide of the invention is herein also named "acylase of the invention" or "enzyme of the invention". If not stated otherwise, all these terms include not only the naturally occurring, authentic sequence of a polypeptide of the invention, which are the preferred embodiments of the invention, but also all mutants, variants and fragments thereof which exhibit acylase enzymatic activity, preferably the same stereoselective activity as the natural

enzyme. The polypeptide of the invention can be used in enriched or, preferably, purified form.

In a preferred embodiment of the invention a polypeptide of the invention is particularly capable of catalyzing the following stereoselective reaction (A)

.1 .

R- (or S) amine acid S- (or R) amide

(D (2) (3) (4) wherein the racemic acylamide of the formula (1) is stereoselectively hydrolyzed with an amine acylase of the invention, which is a polypeptide with amine acylase enzymatic activity but without lipase- or esterase-activity, and which is capable of stereoselectively hydrolysing the racemic acylamide of formula (1), and wherein said hydrolysis results in the R- (or S-) amine of the formula (2) and the acid of formula (3) and the S- (or R-) amide of formula (4), whereby whereby in the formula (1 ) to (4)

R 1 is hydrogen, CrC 8 alkyl, C 2 -C 8 alkenyl, CrC 8 alkoxy, C 2 -C 8 alkylcarboxy or carboxy; preferably CrC 4 alkyl, C 2 -C 4 alkenyl, Cι-C 4 alkoxy or C 2 -C 4 alkylcarboxy; more preferably Cι-C 3 alkyl, C 2 -C 3 alkenyl, Cι-C 3 alkoxy or C 2 -C 3 alkylcarboxy; and most preferably selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, iso-propyl, propyl-1 -en, propyl-2-en, prop-2-ylen, and methoxy; R 2 is aryl or Cι-C 4 aryl; unsubstituted or substituted by CrC 4 alkyl, Cι-C 4 alkoxy, Ci- C 4 hydroxyalkyl, CrC 4 aminoalkyl, CrC Λ haloalkyl, hydroxy, amino, halogeno, nitro, sulfo, or cayno; or wherein RT and R 2 together are forming a 5 to 7 membered ring substituted by or fused to aryl, wherein the rings may contain one or two heteroatoms selected from nitrogen, sulfur and oxygen; preferably indane, tetraline or chromane, unsubstituted or substituted with methoxy, ethoxy, halogeno, methyl, ethyl, nitro, cyano, amino, hydroxy, trfluoromethyl; and R 3 is an aliphatic acyl residue; preferably C 1 -C 4 alkyl; and more preferably methyl.

Aryl is, for example a homo- or heterocyclyl. A suitable ring systems is, for example, a single or double ring system having from 3 to 10 ring atoms, is bonded via a carbon atom or via a nitrogen atom and contains up to 4 hetero atoms selected from oxygen, nitrogen, sulfur, and sulfur linked to 1 or 2 oxygen atoms; which in addition may also be fused with 1 or 2 phenyl radicals or with 1 or 2 cycioalkyl radicals, cycioalkyl preferably having from 5 to 7 ring atoms; and which may be unsaturated or partially or fully saturated.

Examples for aryl are phenyl, napthyl, biphenylyl, anthryl, fluorenyl, thienyl, furyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, thiazolyl, tetrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolyl, benzimidazolyl, quinolyl, isoquinolyl, 3,1-benzofuranyl, chromanyl, cyclohexa- [b]pyrrolyl, cyclohexafb] pyridyl, [b] pyrimidinyl, pyrrolidinyl, pyrrolinyl, cyclohexa[b]pyrazinyl, cyclohexa[b]pyrimidinyl, imidazolidyl, piperidyl, piperazinyl, morpholinyl, thiomorpholinyl, S,S-dioxo-thiomorpholinyl, indolinyl, isoindolinyl, 4,5,6,7-tetrahydro indolyl, 1 ,2,3,4-tetra- hydroquinolyl or 1 ,2,3,4-tetrahydroisoquinolyl, for example one of the last-mentioned radicals, being unsubstituted or substituted by one or more substituents selected from lower alkyl, for example methyl, phenyl, 1 - or 2-naphthyl, phenyl-lower alkyl, for example benzyl, hydroxy-lower alkyl, for example hydroxymethyl or 2-hydroxyethyl, hydroxy, lower alkoxy, for example methoxy or ethoxy, amino, lower alkylamino, for example methyl-, ethyl- or tert- butyl-amino, di-lower alkylamino, for example dimethyl- or diethyl-amino, carboxy, lower alkoxycarbonyl, for example methoxy-, isopropoxy-, sec-butoxy- or tert-butoxy-carbonyl, phenyl- or naphthyl-lower alkoxycarbonyl, for example benzyloxycarbonyl, halogen, for example fluorine, chlorine, bromine or iodine, especially chlorine or bromine, lower alkanoyl, for example acetyl or pivaloyl, nitro, oxo and/or by cyano.

In a preferred embodiment aryl is unsubstituted or substituted by one or more substituents selected from methyl, hydroxymethyl, 2-hydroxyethyl, hydroxy, methoxy; ethoxy, amino, methyl-amino, ethyl-amino, tert-butyl-amino, dimethylamino, diethyl-amino, carboxy, methoxy-carbonyl, isopropoxy-carbonyl, sec-butoxy-carbonyl, tert-butoxy-carbonyl, fluorine, chlorine, bromine, acetyl or pivaloyl, nitro, oxo and/or by cyano.

In a preferred embodiment of the above reaction A, a racemic acylamide is hydrolyzed wherein R 3 is CrC 3 alkyl, more preferably dalkyl. In particular, if an acylase obtainable from Rhodococcus globerulus or Rhodococcus equi is used, R 3 is most preferably d-

C 3 alkyl; if an acylase obtainable from Arthrobacter aurescens is used, R 3 is most preferably dalkyl.

The term 'substituted' means that the moiety in question can be substituted by one to three identical or different substituents, preferably one or two identical or different, most preferably by one substituent selected from the group consisting of Cι-C 8 alkyl (preferably methyl), haloalkyl (preferably trifluoromethyl), halogen (preferably fluorine or chlorine), amino, nitro, and Cι-C β alkoxy (preferably methoxy).

In accordance with the present invention aryl standing alone or being member or an aralkyl moiety is a carbocyclic radical in which at least one ring is in the form of a 6-membered aromatic ring (i.e. a benzene ring). Preferred are phenyl, naphthyl, such as 1 - or 2- naphthyl, biphenylyl, such as, especially, 4-biphenylyl, anthryl, and fluorenyl, and also such ring systems having one or more fused saturated rings.

In a preferred embodiment aralkyl stands for an aliphatic radical substituted by an aryl moiety, wherein the aliphatic radical is an unbranched or branched C r C 8 alkylene (preferably CrC 3 alkylene, most preferably methylene or ethylene) and the aryl moiety is a carbocyclic radical as defined above. In a more preferred embodiment aralkyl stands for a radical of the formula 10, 11 or 12

(10) (11) (12) wherein

R 4 is hydrogen or C 1 -C 4 alkyl; preferably hydrogen or methyl; more preferably hydrogen; n is an integral number selected from 0, 1 , 2, 3 and 4; preferably 0, 1 , 2 and 3; more preferably 0, 1 and 2;

R 5 and R 6 are each independent of each other hydrogen, Cι-C 4 alkyl, C r C 4 haloalkyl, halogeno, amino, cyano, nitro or Cι-C 4 alkoxy; preferably hydrogen, methyl, ethyl, trifluoro¬ methyl, fluorine, chlorine, bromine, iodine, amino, nitro, cyano or methoxy; more preferably hydrogen, methyl, fluorine, chlorine, cyano, nitro or methoxy.

Further preferred compounds are of formula 13, 14 and 15

(13) (14) (15) wherein n is an integral number selected from 0, 1 and 2; R 5 and R 6 are each independent of each other hydrogen or cyano.

Further preferred compounds are of formulae 16 and 17

(16) (17) wherein X is oxygen, carbon or nitrogen; preferably oxygen and carbon; and R 5 and R 6 are each independent of each other hydrogen, C r C 4 alkyl, C 1 -C 4 haloalkyl, halogeno, amino, cyano, nitro or C r C 4 alkoxy; preferably hydrogen, methyl, ethyl, iodo, bromo, trifluoromethyl, chloro, fluoro, amino, cyano, nitro, methoxy or ethoxy.

If R 1 and R 2 form together an unsubstituted or substituted cyclic heteroaliphatic radical said radical preferably stands for a compound of formula (18)

Further examples for suitable compounds are:

An acylase of the invention is obtainable from a microorganism selectable by a process comprising inoculating a selection medium with natural samples such as soil, water, or plant silage, said selection medium comprising an acylamide as sole carbon source. The acylamide can be used as racemate or, if a preselection on a R- or S-enantiomer specific acylase is intended, as R- or S-enantiomer. In the preferred embodiments of the selection process, the acylamides described above in context with the hydrolysis reaction are used for the selection.

Apart from the carbon source, the selection medium also contains all essential ingredients necessary for allowing growth of microorganisms, such as mineral salts, N-sources, and trace elements.

In the most preferred embodiments, the selection medium comprises an acylamide selected from the group consisting of N-acetyl-1 -phenylethylamine as racemate or S- or R- enantiomer and N-acetyl-2-amino-1-phenyl-4-pentene as racemate or S- or R-enaπtiomer.

A suitable selection medium used for performing the invention comprises 3 g of the acylamide and further contains 3 ml trace element solution SL-6, and 1 I mineral salt solution ML1 (pH 7.0), whereby the composition of the mineral salt solution ML1 is 5 g K 2 HPO 4 , 0.2 g MgS0 4 χ 7H 2 O, 20 mg CaCI 2> 20 mg FeS0 4 χ7H 2 0, 1.5 g (NH 4 ) 2 S0 4 , and 1 I Aqua deion (pH 7.0); and the composition of the trace element solution SL-6 is 20 mg NiCI 2 χ 6H 2 O, 200 mg CoCI 2 χ 6H 2 O, 30 mg MnCI 2 χ4H 2 0, 10 mg CuCI 2 χ2H 2 0, 300 mg H 3 BO 3 , 30 mg Na 2 MoO 4 χ 2H 2 O, 100 mg 2nSO 4 χ 7H 2 O, and 1 I Aqua deion. This recipe, however, may of course be varied as long as sufficient trace elements, mineral salts and N-source are provided.

Preferably, an acylase of the invention is obtainable from a microorganism selected from the group consisting of Rhodococcus globerulus, Rhodococcus equi, and Arthrobacter aurescens, more preferably from a microorganism selected from the group consisting of Rhodococcus globerulus K1/1 , DSM 10337, Rhodococcus equi Ac6, DSM 10278, and Arthrobacter aurescens AcR5b, DSM 10280.

The term biocataiyst according to the present invention also includes living or dead microorganisms exhibiting the enzymatic activity of an acylase of the invention. Accordingly, the invention also concerns a microorganism expressing an acylase of the invention. Included are both genetically modified microorganisms and naturally occurring ones. While the former can be produced either by mutation or by genetic engineering, i.e. by transforming a microorganism such as bacteria, e.g. E. coli, or a yeast, e.g. Hansenula or Saccharomyces, with a gene coding for an acylase of the invention, the latter is obtainable from natural sources by a selection process comprising inoculating a selection medium with natural samples, e.g. soil, water, plant silage.

A gene coding for an acylase of the invention can, for example, be obtained by identifying at least a part of the sequence of an isolated acylase of the invention, deducing DNA sequences coding for the partial protein sequence, preparing an oligonucleotide or a mixture of oligonucleotides (taking into consideration the degeneracy of the genetic code), probing a DNA library derived from the microbial strain naturally expressing the desired acylase, isolating the gene, and cloning it into a suitable vector for transformation of the microorganism to be genetically modified. In another approach, the full sequence of the

isolated acylase can be determined and a DNA coding the protein can be produced synthetically. It is also very easily possible to screen a suitable DNA library in E. coli, for example, for growth on the corresponding amide as sole carbon source in order to obtain a transformed clone expressing the acylase.

A naturally occurring microorganism having acylase activity can be obtained by a process outlined hereinafter and can be converted by mutation in a manner known per se, for example using generally known mutagens, such as UV-rays or X-rays, or mutagenic chemicals, into mutants that are differentiated from their parents by improved properties, e.g. lower nutrient medium demands, higher growth rates and, especially, higher acylase activity. Such mutants can also occur spontaneously. The identification and isolation of such mutants is carried out also in a manner known per se: the acylase activity of colonies of such mutants is ascertained, for example, after disintegration of the cells, by adding specific amounts of a suitable acylamide substrate to aliquot portions of the cell residue and qualitatively or quantitatively determining the reaction products that are formed by means of chromatography, especially HPLC.

Isolation of a microorganism naturally expressing an acylase of the invention can be achieved by a selection process comprising inoculating a selection medium with natural samples such as soil, water, or plant silage, said selection medium comprising an acylamide as sole carbon source. The acylamide can be used as racemate or, if a preselection on a R- or S-enantiomer specific acylase is intended, as R- or S-enantiomer. In the preferred embodiments of the selection process, the acylamides described above in context with the hydrolysis reaction are used for the selection.

Apart from the carbon source, the selection medium also contains all essential ingredients necessary for allowing growth of microorganisms, such as mineral salts, N-sources and trace elements.

In the most preferred embodiments, the selection medium comprises an acylamide selected from the group consisting of N-acetyl-1 -phenylethylamine as racemate or S- or R- enantiomer and 2-amino-1 -phenyl-4-pentene as racemate or S- or R-enantiomer.

A suitable selection medium used for performing the invention comprises 3 g of the acylamide and further contains 3 ml trace element solution SL-6, and 1 I mineral salt solution ML1 (pH 7.0), whereby the composition of the mineral salt solution ML1 is 5 g K 2 HPO 4 0.2 g MgS0 4 χ 7H 2 0, 20 mg CaCI 2 . 20 mg FeSO 4 χ7H 2 O, 1.5 g (NH 4 ) 2 SO 4, and 1 I Aqua deion (pH 7.0); and the composition of the trace element solution SL-6 is 20 mg NiCI 2 χ 6H 2 O, 200 mg CoCI 2 χ 6H 2 O, 30 mg MnCI 2 χ 4H 2 O, 10 mg CuCI 2 χ2H 2 O, 300 mg H 3 BO 3 , 30 mg Na 2 MoO 4 χ 2H 2 O, 100 mg ZnSO 4 χ 7H2θ, and 1 I Aqua deion. This recipe, however, may of course be varied as long as sufficient trace elements, mineral salts, and N-source are provided.

A preferred microorganism is selected from the group consisting of Rhodococcus globerulus, Rhodococcus equi, and Arthrobacter aurescens, more preferably selected from the group consisting of Rhodococcus globerulus K1/1 , DSM 10337, Rhodococcus equi Ac6, DSM 10278, and Arthrobacter aurescens AcR5b, DSM 10280.

The acylase is isolated from the microorganism by methods well known in the art, in particular by the methods described in the examples.

Preferred is a substantially purified biocataiyst according to claim 1 , isolated from a microorganism as defined above; more preferably comprising an amino acid sequence selected from the group consisting of SEQ ID NOs 1 , 2, 3 and 4.

An important aspect of the invention relates to a process comprising the hydrolysis of a racemic N-acylamide which has an aliphatic acyl residue and which is not a derivative of a natural amino acid, characterized in that an acylase of the invention is used. The process comprises reaction A shown above. In the preferred embodiments of the process, residues R 1 , R 2 , and R 3 and the acylase have the meanings outlined above. In the case of the acylases of Rhodococcus globerulus K1/1 , DSM 10337 or Rhodococcus equi Ac6, DSM 10278, R 1 can also be urea.

The process of the invention (see process A above) can be performed with any "biocataiyst" having the activity of an enzyme of the invention. A "biocataiyst" according to the invention is, for example, a microorganism expressing an acylase of the invention, for example a

naturally occurring, a mutated or a recombinant microorganism defined above, a crude cell extract of such a microorganism, an enriched or a purified enzyme according to the invention. The term microorganism in this context includes the living microorganism or the dead microorganism, e.g. in a disintegrated form in which the cell wall and/or cell membrane is mechanically or chemically disrupted or removed.

A "biocataiyst" for use in a process of the invention may be immobilized. The immobilization of said "biocataiyst" can be carried out analogously to processes known per se, e.g. coupling to a solid support or enclosing in an enzyme membrane reactor.

A microorganism expressing acylase enzyme activity can be either a naturally occurring one, optionally converted by mutation as described above, or a microorganism transformed by genetic engineering techniques with a gene coding for an acylase of the invention to be able to produce the desired acylase. The hydrolysis of the acylamide substrate with microbial cell extract is preferably carried out in homogeneous aqueous solution at pH 5 to 10.5, more preferably at pH 6 to 9.5. For the stabilization of the pH value, the reaction is carried out in a manner known per se in buffered solution or using a pH-stat. The reaction temperature is approximately from 10 to 65 °C, more preferably from 20 to 50 °C, even more preferably from 20 to 30 °C. The acylamide substrate is used preferably in a concentration of 1 mM to 1 M, more preferably 10mM to 100 mM. However, if the substrate is less soluble, it is also possible to use a substrate suspension.

The enzymes isolated from Rhodococcus equi Ac6, DSM 10278, and Arthrobacter aurescens AcR5b, DSM 10280, differ in the pH and temperature optimum. For the enzyme of Rhodococcus equi Ac6, DSM 10278, the preferred pH range is 5 to 10.5, more preferred is 6 to 8, even more preferred is 6.5 to 7.5, and the preferred temperature range is 10 to 65 °C, more preferred is 20 to 37 °C, even more preferred is 25 to 30 C C. For the enzyme of Arthrobacter aurescens AcR5b, DSM 10280, the preferred pH range is 5.5 to 10.5, more preferred is 7 to 9.5, and the preferred temperature range is 10 to 65 °C, more preferred is 45 to 50 °C (if maximal activity is desired) or 20 to 30 °C (if maximal enzyme stability is desired).

The process according to the invention can be carried out either as a batch process or continuously in an enzyme membrane reactor (EMR). In the latter case, the enzyme

membrane reactor is preferably fitted with an ultrafiltration membrane having a separation limit of less than approximately 30 000, so that the enzymes contained in the reaction mixture are held back whilst the low-molecular-weight products and unreacted reactants pass through the membrane and the product can be isolated from the outflow. The reactor is preferably sterilized before use so that the addition of antibacterial substances can be dispensed with. The reactions are carried out in a manner analogous to that described above.

The process according to the invention can also be carried out by percolating the solution containing the acylamide substrate, which has been adjusted to a suitable pH value, through a solid carrier on which the acylase contained in the crude microbial extract has been immobilized (the matrix-bound enzyme preparation is obtainable, for example, by percolation of the crude microbial extract through CNBr-activated Sepharose, eupergite or the like).

Working up the reaction mixture and purification of the products and unhydrolyzed reactants or the unhydrolyzable enantiomer in accordance with the invention are carried out by customary processes known from the State of the Art. For example, the reaction mixture can be clarified by filtration or, preferably, centrifugation, and then the enzyme can be separated by ultrafiltration (membrane with separation limit of < 30 000 Daltons) and the remaining product can be washed out of the retentate by diafiltration. The actual purifi¬ cation is then carried out, for example, by chromatographic methods, e.g. gel chromato¬ graphy (inter alia Sephadex G-25), ion exchange chromatography, e.g. anion exchange chromatography, thin layer chromatography, HPLC or the like. Selective extraction of amide is performed, e.g., preferably at low pH values and after that the amine can be extracted at preferably alkaline pH values.

In order to obtain the cell extract used in accordance with the process, a microorganism having acylase activity, especially one of those mentioned above, is cultivated in an aqueous nutrient medium that contains assimilable carbon and nitrogen sources and also mineral salts, at a pH value of approximately from 6 to 9, preferably of approximately from 6.5 to 7, and at a temperature of approximately 28-40°C, especially approximately 28 to 30 °C, the biomass is removed and the cell extract is obtained. The fermentation time is so selected that optimum titers with respect to acylase activity are achieved.

When the cell density has reached an adequate value, the cultivation is discontinued. The culture broth is separated off in known manner, e.g. by centrifugation, and the sedimented cells are broken down in customary manner, e.g. by shaking with fine glass beads, by ultra¬ sound treatment, or using a French press. Insoluble cell components and, if used, glass beads, are removed, e.g., by centrifugation, and the residue is used as the enzyme source (crude extract). The residue, as an acylase-containing crude extract, can be used directly in the process according to the invention. Advantageously, however, in order to remove nucleic acids (viscous solutions!) the crude extract is treated with a polycationic agent, e.g. polyethyleneimine, a polyamine such as spermidine, streptomycin-sulfate, or ribonuclease or Mn 2+ salts, and the precipitated nucleic acids are removed by centrifugation.

Preferably, the crude cell extract is subjected to one or more conventional purification steps in order to remove interfering components from the extract.

The process of the invention can be used for separating racemates, if one stereoisomer of the racemic acyl amide is specifically hydrolyzed. However, if hydrolysis is not stereo¬ selective (e.g. since the acylamide is not a racemate or since the enzyme hydrolysis both enantiomers of a certain racemic acylamide), the process of the invention can be used in the chemistry of protecting groups. In this case, the C-atom to which R 2 and R 3 are bound in the substrates need not be chiral. In this case, 100 % of the substrate can be hydrolyzed. In one example, R 2 and R 3 are connected covalently to form a benzene ring; in another example, R 2 and R 3 are connected covalently to form a cyclopentane ring fused with benzene. In both cases, R 1 is preferentially methyl.

The following examples are illustrative, however, should not be construed to limit the present invention.

Examples:

Example 1 : Isolation of microorganisms with (S)-1-phenylethylamine acylase activity

16 soil samples are suspended in the mineral salt solution ML1 described below, which has been sterilized before by autoclavation at 120°C for 20 min. 0.1 ml of these solutions and 12 water samples from nature and from a sewage purification plant are used for the

inoculation of 4 ml of liquid medium (= enrichment medium). In parallel control cultures with enrichment medium not containing racemic N-acetyl-1 -phenylethylamine are inoculated. The composition of the mineral salt solution ML1 is:

Prior to inoculation the medium is filled into 16 ml test tubes and sterilized in an autoclave for 20 min at 121 °C.

The tubes are incubated in an inclinated position at 28°C on a rotary shaking machine with 220 rpm. Cultures with growth, observed as turbidity, in the presence of N-acetyl-1 - phenylethylamine, but not in its absence, are further inoculated into 4 ml of sterile enrichment medium and also into control tubes without substrate (= second propagation).

Cultures of the fifth propagation were diluted 10 5 and 10 6 fold in sterile 69 mM potassium phosphate buffer, pH 7, and plated on solid medium consisting of the enrichment medium plus 20 g/l agar and on control plates without N-acetyl-1 -phenylethylamine. Colonies morphologically different from those on the control plates are streaked - for cell isolation

and determination of the enantioselectivity of the N-acetyl-1 -phenylethylamine utilization - on selective agar plates containing enrichment medium plus 20 g/l agar, where the racemic N-acetyl-1 -phenylethylamine is substituted by a) 3 g/l (S)-N-acetyl-1 -phenylethylamine or b) (/?)-N-acetyl-1 -phenylethylamine. The plates are incubated for 3 to 10 days at 28°C. Strains growing only on one of the enantiomers of N-acetyl-1 -phenylethylamine and appearing pure by colony morphology are cultured for 24 - 48 h at 28°C on a shaker at 220 rpm in 25 ml medium (100 ml flasks) of the following composition (= basic medium with (R) or (S)-N-acetyl-1 -phenylethylamine):

0.5 ml of these precultures are used to inoculate the main cultures possessing the same medium and scale. After 24 - 48 h of incubation at 220 rpm and 28°C the contents of the flasks is centrifuged (15 min. 9000 x g) in a refrigerated Superspeed Centifuge (Dupont Co., Welmington, Delaware, USA), and the sedimented cells are suspended in 1 ml of 69 mM potassium phosphate buffer with pH 7. In Eppendorf vials 0.6 ml of these suspensions are mixed with 1.2 g of glass beads (diameter of 0.1 - 0.25 mm, Carl Roth GmbH, Karlsruhe, Germany) and are shaken for 10 min. in a glass bead mill (Retsch Co., Haan, Germany) at maximum velocity. After cooling the vials in an ice bath and centrifugation at 11500 rpm in a Biofuge 15 (Heraeus Sepatech, Osterode Germany) in order to remove the cell debris and the glass beads the supernatants (= crude cell extracts) are used as the enzyme source.

The assays for acylase activity are prepared in Eppendorf vials as follows:

After an incubation at 30°C for a time period allowing the liberation of 0 to 3 mM of 1 - phenylethylamine 400 μl of ice cooled acetone are added to stop the reaction. The tube is mixed vigorously, put on ice for about 15 min., and then is centrifuged for 4 min at 1 1500 rpm in a Biofuge 15 (Heraeus Sepatech, Osterode Germany). The supernatant is subjected to quantitative HPLC analysis. If activity assays contain crude extracts of cells grown in the

presence of N-acetyl-1 -phenylethylamine a control assay without substrate has to be prepared in parallel in order to determine the 1 -phenylethylamine content of the crude extract. For HPLC analysis sample volumes of 20 μl are injected onto a RP-8 column (LiChroCART 125-4, LiChrospher 100 RP-8 (5mm), guard column: LiChroCART 4-4, LiChrospher 100 RP-8 (5mm), Merck Co., Darmstadt Germany). The following gradient with a flow rate of 1.25 ml/min. is applied for the separation of the substrate N-acetyl-1 - phenylethylamine and the product 1 -phenylethylamine:

Solvent A: potassium phosphate buffer, 3 mM, pH 3 Solvent B: 10 % v/v of solvent A and 90 % v/v of acetonitrile

The substances in the eluate are detected by measuring the UV absorbance at 215 nm. The concentrations of the reaction product 1 -phenylethylamine are calculated via the peak area using a calibration curve from 0 to 5 mM.

One unit (U) of acylase activity is defined as the amount of enzyme catalyzing the liberation of 1 μmol of 1 -phenylethylamine per minute. The acylase concentration in the samples is calculated according to the following formula:

U/ml [μmol/(mlχmin)] = 1 -phenylethylamine concentration in the HPLC vial [mM] / (incubation time[min] x volume of the acylase containing sample [20 μl]) x final assay volume [800 μl] x dilution factor (dilution of the acylase preparation before the test).

The specific activity is expressed as Units of acylase activity per mg of protein in the enzyme preparation determined by the BioRad Protein Assay (Biorad Co., Glattbrugg, Switzerland). The protein concentration of the sample is calculated by using a calibration curve with 0 - 1 mg/ml of bovine serum albumine.

26 bacterial strains were isolated possessing (S) -N-acetyl-1 -phenylethylamine acylase activity. No strain was obtained containing an (ft)-specific enzyme. Table 1 shows the specific activities of the 4 best organisms. Rhodococcus equi Ac6 (= DSM 10728) shows the highest specific activity in the crude extract and also the highest enzyme yield (45.2 U/l culture broth) and therefore, is selected as the producer of a (S)-1 -phenylethylamine acylase. In the following the acylase of strain Ac6 is referred to as Ac6-acylase. In the examples given below the standard incubation time for shake flask cultures with strain Ac6 is 48 h.

Table 1 : Production of (S)-N-acetyl-1 -phenylethylamine acylase by the 4 favorite strains

*) U/mg pro tβjn in the crude cell extract

Example 2: Growth and induction of the acylase of Rhodococcus eaui Ac6 a) Variation of the inducer and investigation of the enantioselectivitv of the acylase reaction R. equi Ac6 is grown in basic medium containing 3g/l (S)-N-acetyl-1 -phenylethylamine or 3g/l (fl)-N-acetyl-1 -phenylethylamine or no N-acetyl-1 -phenylethylamine for 91 h by the shake flask culture technique described in example 1. By applying the methods given in example 1 the crude extracts are prepared and subjected to measurement of the protein concentration and the acylase activity using (S)-N-acetyl-1 -phenylethylamine and (R)-t-i- acetyl-1 -phenylethylamine as the substrates.

Table 2: Induction of R. equi Ac6 acylase

The data given in table 2 show that (S)-N-acetyl-1 -phenylethylamine has to be present in the growth medium to induce the acylase expression. (f?)-N-acetyl-1 -phenylethylamine is not effective as an inducer of the acylase of strain Ac6 and inhibits the growth of the bacterium. Furthermore, the acylase acts highly ( S)-enantiospecific.

b) Variation of the concentration of ( S)-acetyl 1 -phenylethylamine

R. equi Ac6 cells are inoculated into three 1 I Erlenmeyer flask containing each 100 ml of basic medium with 10 g/l yeast extract and 10 g/l meat extract as additional nutrients. The cells are grown (shake flask technique see example 1) until the OD 660 reaches about 2. Then the culture is split into eight portions of 25 ml and (S)-N-acetyl-1 -phenylethylamine is added to the shake flasks in final concentrations of 0 - 7 g/l in steps of 1 g/l. The cells are further incubated for 25 h. Then the protein concentrations and acylase activities of the crude extracts are measured as described in example 1.

More than 2 g/l (S)-N-acetyl-1 -phenylethylamine in the medium affect the acylase production negatively. (S)-N-acetyl-1 -phenylethylamine inhibits the growth of the cells: the ODsso decreases with increasing concentrations of (S)-N-acetyl-l -phenylethylamine in the medium.

In another experiment (S)-N-acetyl-1 -phenylethylamine is added to the culture medium in final concentrations of 1 to 5 g/l in steps of 1 g/l directly after inoculation to one series of shake flasks (see example 1 ) and after 72 h of growth to another. Basic medium without peptone supplemented with 20 g/l meat extract and 20 g/l yeast extract is used. 96 h after inoculation the cells are harvested by centrifugation and the protein concentration and acylase activity in the crude extract are measured (see example 1).

3 g/l (S) -N-acetyl-1 -phenylethylamine result in the maximal acylase yield (60 U/I cu iture medium) if the inducer is added to the medium directly after inoculation, whereas 2 g/l (S)-N-acetyl-1 - phenylethylamine lead to the maximal acylase production (20 U/lcuiture medium) if the inducer is added 72 h after inoculation. By starting the cultivation in the presence of (S)-N-acetyl-l- phenylethylamine the acylase yield is three fold higher than in the culture where the inducer is added after 72 h of growth.

c) Variation of the start pH-value

Shake flask cultures containing basic medium with 3g/l (S)-N-acetyl-1 -phenylethylamine varying in pH from 2.5 to 9.5 in steps of 0.5 pH units are grown for 65 h. Then the O.D.660 of the cultures as well as the protein concentrations and acylase activities of the crude extracts are measured (see example 1).

Optimum growth of Rhodococcus equi Ac6 and maximum acylase yield both are obtained between pH 5.5 and 7. The specific acylase activity, however, is rather constant over a broad range from pH 5.5 to pH 9.0.

d) Variation of the nutrients

Shake flask cultures containing basic medium without peptone, with a lower yeast extract content of 0.5 g/l, 3 g/l racemic N-acetyl-1 -phenylethylamine, and 2 g/l of an additional nutrient are incubated for 45 and 111 h, respectively. After determination of the O.D.660 the cells are harvested, disrupted, and the protein concentrations and acylase activities of the crude extracts are determined (see example 1).

Table 3: Growth and acylase production of Rhodococcus equi Ac6 with different nutrients

The highest acylase yield after 45 h as well as after 1 1 1 h of incubation are achieved with yeast extract (see table 3). The expression of the acylase is positively influenced by the presence of L-glutamate and complex nutrients in the medium, which leads to higher acylase yields (U/lmedwm), but not to increased specific activities. Cultures grown without additional nutrient show the highest specific acylase activity, but a low overall enzyme yield [U/l cu K ure broth] and a low cell density (O.D.660).

Example 3: Production of the acylase of Rhodococcus eaui Ac6 bv fermentation on 20 I scale

For the fermentation of Rhodococcus equi on 20 I scale a 30 I bioreactor is used equipped with an automatic regulation of the pH, the temperature, and the concentration of the dissolved oxygen (pO 2 ) and with an anti foam system. The fermentor is filled with 20 I of basic medium containing 3 g/l of racemic N-acetyl-1 -phenylethylamine and 5 g/l of yeast extract. After autoclavation of the medium for 30 min. at 121°C and cooling down to 28°C the bioreactor is inoculated to an initial O.D.660 of 0.05 with 1 I of shake flask culture, which has been grown before in 5 portions of 200 ml in 1 I Erlenmeyer flasks during 84 h at 28°C and 220 rpm. The conditions during the fermentation are:

Samples of 25 ml culture medium are taken at different time points. The O.D.660 values of the culture broth as well as the protein contents and the acylase activities of the crude extracts are measured (see example 1).

The maximum enzyme yield (U/I cu ιture medwm) is reached after 30 h of incubation and remains constant at least during the next 18 h. After 48 h of cultivation the fermentation broth is cooled down to 15°C. By continuous flow centrifugation using a Sorvall TZ-28 rotor in a refrigerated Sorvall RC5B Superspeed Centrifuge (Dupont Co., Wilmington, Delaware, USA) 41 g of cells (wet weight) are harvested containing 680 U of Ac6-acylase with a specific activity of 0.19 U/mg protein in the crude extract. The cells are resuspended in 103 ml of 69 mM potassium phosphate buffer, pH 7, and stored at -20°C.

Example 4: Purification of the acylase of Rhodococcus equi Ac6 a) Preparation of the crude extract in preparative scale

100 ml of cell suspension prepared as described in example 3 are thawn and poured into a 600 ml glass beaker containing 150 ml of glass beads (diameter of 0.1 - 0.25 mm). The beaker is cooled in an ice bath during the cell disruption with a Philips HR1385 professional handblender, which is operated 6 - 8 times for 2 min. interrupted by breaks for 5 min. After sedimentation of the glass beads the supernatant is removed and 150 ml of 69 mM potassium phosphate buffer pH 7.0 are added to the glass beads. By short mixing and settling of the glass beads a second supernatant is obtained. The two supernatants are combined and centrifuged (20 min., 9000 rpm) in a Sorvall RC5B refrigerated Superspeed Centrifuge using a Sorvall GS-3 aluminum rotor. The supernatant of the centrifugation is used as the crude cell extract for the purification of the acylase. The crude extract can be stored at -20°C for at least two month without loss of activity, and intermediate thawing and freezing only results in a loss of activity of 5 %.

b) Ammonium sulfate precipitation

To 100 ml of ice cooled crude extract (preparation see example 4 a) crystalline ammonium sulfate is added slowly to a final concentration of 25 % saturation. After the ammonium sulfate is dissolved under gentle stirring, the solution is kept on ice for about 30 min. and then the precipitated protein is sedimented by centrifugation for 60 min. at 9000 rpm in a Du Pont RC5B refrigerated Superspeed Centrifuge using a Sorvall GS-3 aluminum rotor. Further ammonium sulfate was added to the supernatant to a final concentration of 85 % and a second protein precipitation is performed as described above. The precipitated protein comprising the Ac6-acylase is dissolved in 150 ml of an ammonium sulfate solution (concentration 30 % saturation) in 69 mM potassium phosphate buffer, pH 7.

c) Hydrophobic interaction chromatography (HIC) on Butyl- Fractogel

For HIC a Merck Fraktogel TSK Butyl-650(S) column (26 x 310 mm) is equilibrated with a 30% saturated (NH 4 ) 2 SO 4 solution (in phosphate buffer, 69 mM, pH 7.0). After loading the column with 75 ml of the final protein solution obtained after the ammonium sulfate precipitation in example 4 b) the column is washed with a 30% saturated ammonium sulfate solution (in phosphate buffer, 69 mM, pH 7.0) until the UV detector signal reaches the baseline level again. Then a gradient decreasing from 30% to 0% ammonium sulfate with a

total volume of 825 ml and a flow rate of 1.8 ml/min. is applied to the column. The fraction size is 12 ml. The Ac6-acylase activity of the fractions is measured (see example 1).

The Ac6-acylase elutes from the column between 8 to 4 % saturation of ammonium sulfate. The 5 most active fractions are pooled and desalted by diafiltration and concentrated to a final volume of 23 ml in a CEC column concentrator equipped with an YM 30000 membrane (Amicon Inc., Beverly, MA, USA). The diafiltration is achieved by repeatedly adding potassium phosphate buffer, 69 mM, pH 7, to the concentrated protein solution and ensuing a reconcentration step.

Table 4 summarizes the results of the purification. The Ac6-acylase preparation obtained by only one chromatographic step appears homogenous by SDS polyacrylamide gel electrophoresis performed with a Phast System using gradient Phast Gels of the type 10-15 (containing 10 - 15 % polyacrylamide) (Pharmacia Co., Dϋbendorf, Switzerland) and coomassie blue staining. For the electrophoresis and the staining procedure the standard methods given by Pharmacia for Phast Gel gradient media are used (Pharmacia Phast System separation technique file no. 110: "SDS-PAGE", and file no. 200: "Fast Coomassie Blue Staining", Pharmacia, Uppsala, Sweden).

Table 4: Purification of the Ac6-acylase

HIC: Hydrophobic Interaction Chromatography, UF: Ultrafiltration

If a crude extract of a lesser specific activity is used, it is necessary to subject the acylase further chromatographic purification steps to obtain a homogenous enzyme preparation, e.g. by amino exchange chromatography on Mono-Q or gel filtration on Superose 12 HR (Pharmacia, Uppsala, Sweden) described in example 5.

Example 5: Determination of the molecular weight and the subunit structure of the Ac6- acylase

A Pharmacia Superose 12HR 10/30 gel filtration column (10 x 300 mm) is used to determine the molecular weight of the native Ac6-acylase. The elution is performed using 69 mM potassium phosphate buffer, pH 7.0, supplemented with NaCl 100 mM, with a flow rate of 0.3 ml/mm. 50 μl of the purified and concentrated Ac6-acylase obtained in example 4 c are applied to the column. The molecular weight of the native enzyme is determined using a calibration curve representing the rWvalues of the calibration proteins (see table 5) against the logarithm of the molecular weight. The K A v value is defined as follows:

KAV = (V β iubon-VrJ/fVcoiumn-Vo)

V 0 = void volume = V β ιm l0n of dextran blue = 7.65 ml, Vcoiumn = column volume = 23.6 ml.

A molecular weight of 94000 ± 3000 (K A V = 0.33 ± 0.04) is measured for the native Ac6- acylase.

Table 5: Calibration proteins for the determination of the molecular weight of the native Ac6- acylase by gel filtration

*) used for the determination of V<

In order to determine the subunit structure SDS-polyacrylamide gelelectrophoresis is applied as described in example 4c. The molecular weight of the denatured acylase is determined using a calibration curve showing the migration distances against the logarithm

of the molecular weights of the marker proteins of a Pharmacia Low Molecular Weight Calibration Kit.

The denatured Ac6-acylase has a molecular weight of 50000 ± 2000. This result in comparison with the molecular weight determined for the native enzyme indicates a homodimeric substructure for the Ac6-acylase.

Example 6: Determination of the isoelectric point (IEP.

Isoelectric focusing (IEF) is done on gels of the type Pharmacia Phast Gel IEF 3-9 (forming a gradient from pH 3 to pH 9) using the Pharmacia Phast System. The IEP of the enzyme is determined in comparison with the proteins of the Pharmacia Low pl Calibration Kit (pH 2.5 - pH 6.5). For the isoelectric focusing and the staining procedure the standard methods given by Pharmacia for Phast Gel IEF media are used (Pharmacia Phast System separation technique file no. 100: "IEF and titration curve analysis", and file no. 200: "Fast Coomassie Blue staining", Pharmacia, Uppsala, Sweden). The IEP of the Ac6-acylase is around pH 3.5.

Example 7: Dependence of the reaction velocity of the Ac6-acylase on the pH value

Enzyme tests as described in example 1 are performed in buffers ranging in pH from 5.71 to 9.88 (pH 5.71 , 6.09, 6.58, 7.0, 7.47: potassium phosphate buffer, 69 mM; pH 7.48, 8.0, 8.4, 8.82, 9.10: Tris-HCl buffer, 100 mM; pH 9.38, 9.88: glycine-NaOH buffer, 100 mM). The incubation time is 30 min. using the final acylase preparation obtained in example 4c diluted 20 times as the enzyme source.

The Ac6-acylase shows a broad activity optimum between pH 6.0 and pH 8.5 with a maximum between pH 6.5 and pH 7.0.

Example 8: Influences of activators and inhibitors on the Ac6-acylase activity

In the presence of various potential enzyme activators and inhibitors activity assays are performed according to the scheme given in example 1 by applying the following conditions and final concentrations:

Table 6: Activity of the Ac6-acylase in the presence of potential enzyme activators and inhibitors

Relative activities given in brackets indicate formation of precipitates during the enzyme assays.

None of the tested metal cations except Ni 2+ and to some extent Zn 2+ in 1 mM concentration affect the acylase activity much (see table 6). In 10 mM concentration Zn 2 \ Ni 2 \ Cu 2+ and Cd 2+ show considerable inhibition of the acylase.

Chelating agents and thiol reducing agents do not affect the acylase activity, whereas 10 mM iodoacetamide and phenylmethylsulfonyl fluoride already in a concentration of 0.01 mM are strongly inhibitory. The strong inhibition by phenylmethylsulfonyl fluoride suggests a serine protease reaction mechanism for the Ac6-acylase.

Example 9: Temperature stability of the Ac6-acylase

Samples of the final acylase preparation obtained in example 4 c are diluted 45 fold with 69 mM potassium phosphate buffer, pH 7, and incubated for various time periods at different temperatures. After the temperature treatment standard activity assays (see example 1 ) are carried out at 30°C.

Table 6: Temperature stability of the Ac6-acylase

Time (min.) Residual activity (%)

30°C 37°C 39.5°C 44°C

15 98 92 85 23

30 96 90 77 11

60 96 88 62 7.0

120 94 83 45 6.3

240 94 84 28 4.7

As it can be seen in table 6 the acylase retains almost all of its initial activity after 4 h of incubation at 30 and 37°C, whereas at 39.5 and 44°C the enzyme is deactivated considerably.

At 30°C even after 34 days still 94 % of the initial acylase activity are present.

Example 10: Dependence of the reaction velocity of the Ac6-acylase on the concentration of (S)-N-acetyl-1 -phenylethylamine

Activity assay under the standard conditions given in example 1 with an incubation time of 10 min. are performed using different initial concentrations of (S)-N-acetyl-1 -phenylethyl¬ amine and the final Ac6-acylase preparation obtained in example 4c, which has been diluted 5 fold with 69 mM potassium phosphate buffer, pH 7, as the enzyme source. The Michaelis constant K m is calculated from the substrate concentrations and the corresponding reaction velocities by numerical nonlinear regression with the program "SigmaPlot for Windows" (Jandel Scientific GmbH, Schimmelbusch, Germany) to the formula: v = V m x S / (S + K m ), v: reaction velocity (U/ml = mM/min.), V m : maximum reaction velocity (U/ml), S = (S)-N-acetyl-1 -phenylethylamine concentration (mM).

The K m value for (S) -N-acetyl-1 -phenylethylamine is 0.6 mM ± 0.1 mM.

Example 11 : Isolation of microorganisms with (ffl-1 -phenylethylamine acylase activity In general the procedure given in example 1 is applied with the following modifications:

• 43 soil and water samples are investigated.

• The liquid medium for the enrichment cultures and the selective agar plates, on which the diluted enrichment cultures are plated to obtain single colonies, contains 3 g/l (R)-H- acetyl-1 -phenylethylamine instead of racemic N-acetyl-1 -phenylethylamine and in addition 2 ml/l of the following vitamin solution:

• The liquid medium for the first propagation is supplemented with 0.5 g/l, for the second propagation with 0.2 g/l of sodium acetate in order to facilitate the growth of acetic acid consuming microorganisms in the beginning of the enrichment.

• Only 3 propagations of enrichment culture are performed.

• The basic medium to grow the cells for the activity tests only contains 1 g/l of yeast extract and peptone.

• (R)-N-acetyl-1 -phenylethylamine is used as the substrate for the activity assays with the AcR5b-acylase.

Only 3 strains with a (R)-specific acylase activity could be isolated (see table 7). Arthrobacter aurescens AcR5b (= DSM 10280) shows the highest specific activity and is selected as the producer of the (R)-N-1 -phenylethylamine acylase, which is in the following referred to as AcR5b-acylase. In the examples given below the standard incubation time for shake flask cultures with strain AcR5b is 23 h.

Table 7: Production of (R)-N-acetyl-1 -phenylethylamine acylase activity by the three (R)- specific strains

Strain Incubation O.D.660 Acylase yield with Acylase yield with Specific activity time (days) (S)-amide as the (R)-amide as the with (R) -amide enzyme substrate enzyme substrate (U/mg pr0 iβin in the crude 1

(U/lculture broth) (U/lculture broth) extract)

AcR5b 1 3.08 0.0 137 0.98

AcR11a 2 4.16 0.0 152 0.84

AcR30a 4 2.39 4.8 24 0.17

(S) and (R)-amide: (S) and (R)-N-acetyl-1 -phenylethylamine

Example 12: Growth and induction of the acylase of Arthrobacter aurescens AcR5b a) Variation of the concentration of ( R)-acetyl 1 -phenylethylamine

Arthrobacter aurescens AcR5b was cultured in shake flasks (see example 11 ) using the basic medium, in which the concentration of (f?)-N-acetyl-1 -phenylethylamine was varied between 0.5 to 4 g/l in steps of 0.5 g/l. The O.D.660, the acylase-activity, and the protein content of the crude extracts are measured as described in example 11.

The maximum amount of AcR5b-acylase is produced in the medium without (R)-N-acetyM - phenylethylamine. Therefore, the enzyme is expressed constitutively. Furthermore, the specific activity of the acylase decreases with increasing concentrations of (R)-N-acetyl-1 - phenylethylamine in the culture broth.

b) Variation of the start pH-value

Shake flask cultures containing basic medium without (R)-N-acetyl-1 -phenylethylamine varying in pH from 3.5 to 11 in steps of 0.5 pH units are grown for 23 h. Then the ODβeo of the cultures as well as the protein concentrations and acylase activities of the crude extracts are measured (see example 11).

Optimum growth of Arthrobacter aurescens AcR5b and maximum acylase yield (U/l culture broth) are both obtained between pH 5.5 and 9. No growth occurs below pH 4.5 and pH 10. For further cultivation of strain AcR5b pH 7 is chosen.

c) Variation of the temperature

Shake flask cultures including the measurement of the O.D.660, the acylase activity, and the protein content of the crude extracts are performed with strain AcR5b according to example 1 1 by varying the temperature between 22 - 36°C in steps of 2°C. The basic medium without (R)-N-acetyl-l -phenylethylamine is used.

Strain AcR5b grows most rapidly between 28 and 32°C . No growth occurs at 36°C. At 28°C the maximum enzyme yield (U/lcuiture broth) is obtained.

d) Variation of the nutrients

In shake flask cultures with strain AcR5b the basic medium without (R)-N-acetyl-1 - phenylethylamine is supplemented with different nutrients at a concentration of 5 g/l, respectively. The O.D.660, the acylase-activity, and the protein content of the crude extracts are determined as described in example 11.

Among the best results regarding the total enzyme yield (U/l culture broth) are those obtained with sorbitol and meat extract, which are chosen for the further cultivation of strain AcR5b.

Table 8: Growth and acylase production of Arthrobacter aurescens AcR5b with different nutrients

Additional nutrient O.D.660 Acylase yield Specific activity

[U/lculture broth] [U/mg Dra teinl

None 1.50 134 1.12

Carbon sources:

D-Fructose 3.97 169 1.11

D-Galactose 4.64 173 1.19

D-Glucose 4.82 176 0.96

D-Maltose Monohydrate 5.18 157 1.06

Malt extract 4.63 189 0.87

Saccharose 6.08 156 0.75

Starch (soluble) 1.64 161 1.11

Glycerol 7.08 182 0.79

D-Mannitol 6.05 174 0.89

D-Sorbitol 5.9 217 1.00

Sodium acetate 2.68 180 0.70

Tri-sodium citrate dihydrate 2.77 153 0.66

Nitrogen sources:

AC-Broth (Difco) 4.23 190 1.17

Bacto-Soytone 4.31 190 0.86

Casamino acids 3.75 160 1.32

Casein hydrolysate 3.53 199 1.21

Casitone 4.75 210 1.05

**Corn Steep Liquor* 4.48 197 1.45

Fish protein hydrolysate* 4.20 93 0.52

L-Glutamic acid 4.88 203 1.04

L-Glutamine 4.49 159 1.1 1

Meat extract 3.38 212 0.87

Peptone from casein 4.63 21 1 1.05

Peptone from soybeans 3.95 175 1.07

Peptone C (technical grade) 2.75 136 1.32

Soybean powder full fat* 1.02 6.5 0.09

Yeast extract (Difco) 3.94 157 0.63

Yeast extract, technical grade 3.86 168 1.01

*not completely dissolved, **formation of cell aggregates

e) Variation of the concentration ratio between the carbon and nitrogen source

In shake flask cultures with strain AcR5b the mineral solution ML1 without ammonium sulfate is supplemented with sorbitol and meat extract at different ratios, but the total concentration of both nutrients was held constant at 7 g/l. The O.D.660, the acylase-activity, and the protein content of the crude extracts are determined as described in example 11.

The optimum yield of the AcR5b-acylase (375 U/l culture broth) was obtained at a ratio of sorbitol to meat extract of 4 : 3.

f) Variation of the total nutrient concentration

The mineral solution ML1 without ammonium sulfate is supplemented with sorbitol and a complex nitrogen source in the concentration ratio of 4 : 3 by varying the total concentration of both nutrients (7, 14, 21, 28, and 42 g/l). Meat extract, peptone from casein, and yeast extract of technical grade are used as the complex nitrogen sources. The shake flask culture (see example 11 ) is performed in 1 I Erlenmeyer flasks filled with 200 ml of medium, which have been inoculated with 3 ml of a preculture. After 11 , 20, 27, 35, 46.5, and 70 h of incubation samples of 25 ml are removed from the flasks and the O.D.660, the acylase activity, and the protein content of the crude extracts are measured.

The maximum AcR5b-acylase yield was obtained with peptone (table 8) after 70 h of incubation.

Table 9: Growth and acylase production by Arthrobacter aurescens AcR5b with different complex nitrogen sources after 70 h of incubation at the total nutrient concentration resulting in the maximum enzyme yield

Nitrogen source Total nutrient O.D.660 Acylase yield Specific activity concentration (g/l) (U/Uure b r ot h ) (U/mg Dr o tein )

Peptone from casein 28 16.0 1470 4.5

Meat extract 21 9.7 1070 4.2

Yeast extract 21 8.2 950 3.5

Example 13: Production of the acylase of Arthrobacter aurescens AcR5b bv fermentation on 20 I scale

The fermentative production of the AcRSb-acylase on 20 I scale is performed as described in example 3 with the following modifications:

• The composition of the culture medium is peptone from casein 12 g/l, sorbitol 16 g/l, antifoam agent SAG 471 1 ml/I, dissolved in mineral salt solution ML1 (example 1 ) without ammonium sulfate, pH 7.

• The agitator speed is adjusted to 500 - 800 rpm, the set point for the dissolved oxygen concentration to 40 % saturation, and the airflow rate to 15 l/min (= 0.75 WM).

• The bioreactor is inoculated with 0.5 1 of preculture grown in portions of 2 times 250 ml in 1 I Erlenmeyer flasks at 28°C and 220 rpm for 19 h.

• The cells are harvested by continuous centrifugation after 33 h of cultivation.

The specific activity and also the total acylase yield still increase significantly in the stationary growth phase up to 5.26 U/mg pro tein and 6420 U/I cu itu r e br ot h , respectively, at an O.D.660 value of 25. The wet cell mass is resuspended in 69 mM potassium phosphate buffer, pH 7, to a final concentration of 40 % weight per volume.

Example 14: Purification of the acylase of Arthrobacter aurescens AcR5b a) Preparation of the crude cell extract on preparative scale

100 ml of the thawn cell suspension obtained in example 13 are cooled in an ice bath and sonicated with a Sonicator W-385 (Heat System Ultrasonics, Farmingdale, USA) using the 1/2 " standard horn under the following conditions: Cycle time 1 sec, duty cycle 50 %, output control 8, and sonicating time 25 min. After removing the cell debris by centrifu¬ gation in a refrigerated centrifuge at 13400 g for 20 min. the supernatant is used as the crude cell extract for the further purification.

b) Precipitation of nucleic acids with polyethylene imine

The crude extract obtained in example 14 a) is mixed with a 10 % (weight / volume) solution of polyethylene imine with a molecular weight of 30000 - 40000, pH 7, to a final

conceπtration of the polymer of 0.3 %. The solution is stirred for 30 min. at 0°C and the precipitate is removed by centrifugation at 27000 g and 4°C for 30 min.

c) Ammonium sulfate precipitation

To the supernatant from example 14 b) solid ammonium sulfate is added a final concentration of 30 % saturation. The mixture is gently stirred at 0°C and then centrifuged at 4°C and 27000 g for 30 min. To the supernatant further ammonium sulfate is added to a final concentration of 70 % saturation. The precipitated protein containing the acylase is sedimented by centrifugation (see above) and dissolved in 40 ml of 69 mM potassium phosphate buffer, pH 7.

d) HIC on Butyl-Fractooel

5.76 g of ammonium sulfate are dissolved in the final protein solution from example 14 c) to reach a final concentration of 25 % saturation. The HIC is performed according to example 4 c) with the only exceptions, that the column is preequilibrated with a 25 % ammonium sulfate solution and the gradient is run from 25 to 0 % saturation of ammonium sulfate.

After washing the non bound protein from the column with a 25 % ammonium sulfate solution the AcRSb-acylase elutes between 13 and 10 % of ammonium sulfate. The enzyme represents the only protein peak of the whole chromatogram. The active fractions are pooled, desalted, and concentrated as described is example 4 c).

e) Anion exchange chromatography on Macro Prep High-Q

A 16 x 125 mm column filled with Macro Prep High-Q support (Biorad, Glattbrugg, Switzerland) is equilibrated with 250 ml of 69 mM potassium phosphate buffer pH 7. After half of the concentrated protein solution obtained in example 14 d) is loaded onto the gel a gradient from 0 to 1 M of NaCl in 70 mM potassium phosphate buffer, pH 7, with a volume of 125 ml is applied to the column. The flow rate is 1 ml/min., the fraction size 1.5 ml. The protein concentration in the eluate is monitored via a UV detector signal and the acylase activity and the protein content of the active fractions are measured as described in example 11.

The acylase activity elutes between 0.4 and 0.6 M NaCl. The activity peak is identical with the only UV detector peak of the chromatogram. The active fractions are pooled and concentrated by ultrafiltration (see example 4 c) to a final protein concentration of 2 mg/ml.

Table 10 summarizes the purification of the AcR5b-acylase. The AcRSb-acylase fractions obtained by anion exchange chromatography all show two bands by SDS polyacrylamide gel electrophoresis (SDS-PAGE, see example 4 c) and appear homogenous by native PAGE, which is also performed with a Phast System using gradient Phast Gels of the type 8 - 25 (containing 8 - 25 % polyacrylamide) (Pharmacia Co., Dϋbendorf, Switzerland). The gels are stained with coomassie blue (see example 4 c). For the native PAGE the standard method given by Pharmacia for Phast Gel gradient media is used (Pharmacia Phast System separation technique file no. 120: "Native PAGE".

Table 10: Purification of the AcR5b-acylase

Purification step Total acylase Recovery (%) Specific activity Enrichment

(U) (U/mg) factor

Cell breakage 5593 100 1.9 1.0

Polyethylene imine 5262 94 3.0 1.6 precipitation ammonium sulfate 4408 79 2.8 1.5 precipitation

HIC + UF-diafiltration / 4186 75 8.2 4.3 concentration

Anion exchange 3725 67 10.0 5.2 chromatography

Example 15: Determination of the molecular weight and the subunit structure The molecular weight of the native AcR5b-acylase is 220000 ± 10000 measured by gel chromatography according to example 5. In the SDS-PAGE (see example 5) the denatured acylase of Arthrobacter aurescens AcR5b shows two bands corresponding to a molecular weight of 89000 ± 3000 and 16000 ± 1000. These results indicate that the AcR5b-acylase is a tetramer consisting of 2 identical large and two identical small subunits (subunit structure of the type α 2 β 2 ).

Example 16: Dependence of the reaction velocity of the AcR5b-acylase on the pH value Enzyme tests as described in example 1 with (R)-N-acetyl-l -phenylethylamine are performed in buffers ranging in pH from 5.5 to 11.0 (pH 5.5, 6.0, 6.5, 7.0, 7.5, 8.0:

potassium phosphate buffer, 69 mM; pH 7.5, 8.0, 8.5, 9.0: Tris-HCl buffer, 100 mM; pH 8.5, 9.0, 9.5, 10.0, 10.5, 11.0: glycine-NaOH buffer, 100 mM). The incubation time is 3 min. using the final acylase preparation obtained in example 4 d as the enzyme source.

The acylase activity shows a broad pH optimum between pH 7.5 and 9 with a maximum at pH 8.

Example 17: Influence of activators and inhibitors on the AcRSb-acylase activity 40 μl of the concentrated enzyme solution from example 4 d) are mixed with 360 μl of 1 and 10 mM solutions of the potential activators / inhibitors in 100 mM Tris-HCl buffer, pH 8, and incubated at room temperature for 30 min. These preincubated acylase solutions are used as the enzyme source for activity assays in the presence of the activator / inhibitor (see example 8) in the concentration used for the preincubation. The incubation time is 12 min. For the EDTA reactivation the acylase is first preincubated with the metal cation at 1 mM concentration (see above). After adding EDTA to a final concentration of 10 mM in form of a 110 mM solution in 100 mM Tris-HCl, pH 8, the acylase is preincubated a second time for 30 min., before the standard activity assay is performed.

6 of the bivalent metal cations inhibit the enzyme remarkably, but in all cases the activity can at least partially be restored with EDTA. EDTA alone and FeCI 3 enhance the acylase activity. Dithiothreitol, iodoacetamide, and phenymethylsulfonyl fluoride inhibit the enzyme at a low to moderate degree.

Table 11 : Activity of the AcR5b-acylase in the presence of potential enzyme activators and inhibitors

Compound Relative activity (%)

None 100

1 mM 10 mM EDTA reactivation

Bivalent metal cations:

CoCI 2 26 (15) 91

MnCI 2 99 (60) n.d.

ZnCI 2 <1.5 (0) 75

MgCI 2 103 93 n.d.

NiCI 2 14 0 85

CaCI 2 92 88 n.d.

CuCI 2 50 0 86

PbCI 2 79 9 102

BaCI 2 92 89 n.d.

CdCI 2 5 0 79

FeCI 3 121 132 n.d. vcι 3 96 (106) n.d.

Chelating aqents:

EDTA 119 121 n.d.

Citrate 122 100 n.d.

Thiol reducing agents:

Dithiothreitol 35 5.4 n.d.

Mercaptoethanol 115 69 n.d.

Glutathion, reduced 113 1 12 n.d.

Thiol orouD blocking agents:

Iodoacetamide 96 29 n.d.

Protease inhibitor:

Phenylmethyl¬ 44 n.d. n.d. sulfonyl fluoride

Relative activities in brackets indicate formation of precipitates in course of the enzyme assays, n.d.: not determined.

Example 18: Stability of the AcR5b-acylase a) Temperature stability

Samples of the final acylase preparation obtained in example 14 d are diluted 10 fold with 69 mM potassium phosphate buffer, pH 7, and incubated for 30 min. at different temperatures. A sample incubated on ice serves as the 100% standard. Afterwards the remaining acylase activities of the samples are measured via the standard activity assays (see example 11).

Between 21 and 30 C C no loss of acylase activity is observed (see table 12). Whereas at 50°C still 60 % of the initial activity are retained after 30 min. of incubation, at 56°C the enzyme is completely deactivated.

Table 12: Residual activity of the AcR5b-acylase after incubation at different temperatures for 30 min.

Temperature (°C): 0 21 25 30 35 40 45 50 56 60 Residual activity (%): 100 95 102 100 89 82 68 60 0 0

When the diluted acylase (see above) is incubated at 0 - 4°C for 16 and for 135 days, the relative residual activities are still 100 and 95 % of the initial value. After an incubation at 23° and 30°C for 16 days, 86 and 65 % of the initial activity are retained.

b) pH-Stabilitv

The final enzyme preparation from example 4 d) is diluted 20 fold with the buffers given in example 16 and incubated at room temperature (about 23°C) for one week. Then the remaining activity of the samples is measured as described in example 11. 100 % activity correspond to the value obtained immediately after the dilution with the pH 7 buffer.

The enzyme is rather stable between pH 7 and 9 (100 % residual activity), but beyond these limits the stability declines sharply.

c) Stability during freezing

The final acylase preparation obtained in example 14 d) is diluted 20 fold with 69 mM potassium phosphate buffer (pH 7), with and without cryoprotective agents (see table 13). The enzyme solutions are frozen at -20 and -80 C C. After 2.5 h and 9 days the samples are thawn and the residual acylase activity is measured as described in example 11.

The data given in table 13 demonstrate that the acylase is deactivated by storage in the frozen state. The stability can remarkably be enhanced by addition of cryoprotective agents.

Table 13: Influence of freezing on the AcRSb-acylase activity

Cryoprotective agent Relative activity (%)

2.5 h, 9 days, 2.5 h, 9 days, -20°C -20°C -80°C -80°C

None 67 0 100 34

5% Glycerol 100 89 100 97 50% Glycerol 77 98 100 90 10% (NH 4 ) 2 SO 4 100 81 92 83 10% Saccharose 100 100 100 92

Example 19: Dependence of the reaction velocity of the AcRδb-acylase on the concentration of the substrate

Activity assays are performed under the standard conditions given in example 11 with an incubation time of 30 min. using different initial concentrations of (R)-N-acetyl-1 - phenylethylamine and racemic N-acetyl-(m-cyano-phenyl)ethylamine. The final AcR5b- acylase preparation obtained in example 4 d, which has been diluted 20 fold with 69 mM potassium phosphate buffer, pH 7, serve as the enzyme source. The Michaelis constants (K m ) and the maximum reaction velocities are calculated as described in example 9.

The K m values for (/?) -N-acetyl-1 -phenylethylamine and the racemic (m-cyano) derivative are 5.7 and 10.4 mM, the relative maximum reaction velocities are 100 and 23 %, respectively.

Example 20: Isolation of microorganisms with (S)-2-amino-1 -phenyl-4-pentene acylase activity

In general the procedure given in example 1 is applied with the following modifications:

• 74 Soil and water samples are used to inoculate enrichment cultures with pH 6, pH 7, and pH 9, incubated at 28°C, and with pH 7, incubated at 37°C.

• The medium for the first propagation contains 0.5 g/l of sodium acetate as the sole carbon source, the medium for the second and third propagation 1 g/l of racemic 2- acetylamino-1 -phenyl-4-pentene.

• Strain cultivation is performed at the same temperature and pH used in the corresponding enrichment cultures.

• The organisms from the enrichment cultures are isolated on Plate Count Agar (Fluka Co., Buchs, Switzerland).

The medium for growing the strains for the activity assays consists of 1 g/l of racemic 2- acetylamino-1-phenyl-4-pentene, 1 g/l yeast extract, and 1 g/l peptone in the mineral salt solution ML1. The incubation time for shake flask cultures is 48 - 72 h.

In order to test the activity and enantioselectivity of the acylase activity assays are performed with 5.47 mM of (S) and (R)-2-acetylamino-1 -phenyl-4-pentene, respectively, as the substrate and 40 μl of crude cell extract as the enzyme source in a total volume of 400 μl. After an appropriate incubation time 200 μl of ice cooled acetone are mixed with 200 μl of the assay. 600 μl of 69 mM potassium phosphate buffer, pH 7, are added, and after centrifugation at 1 1500 rpm the concentration of 2-amino-1-phenyl-4-pentene in the supernatant is analyzed by HPLC (detection wavelength 210 nm).

18 organisms are isolated which hydrolyze preferentially one enantiomer of 2-acetylamino- 1 -phenyl-4-pentene, but only for one strain, Rhodococcus globerulus K1/1 (= DSM 10337), exclusive cleavage of the (S)-enantiomer was observed in the enzyme assays.

Example 21 : Growth and induction of the acylase of Rhodococcus globerulus K1 /1 a) Variation of the inducer

Strain K1/1 is grown in shake flask culture using the medium from example 21 , but with 2 g/l of each yeast extract and peptone, with racemic 2-acetylamino-1 -phenyl-4-pentene or alternatively racemic N-acetyl-1 -phenylethylamine as the inducer in concentrations of 0, 4.92, 7.38, and 9.84 mM.

The measurement of the acylase activity and the O.D.660 reveals, that the acylase is only formed by strain K1/1 in the presence of an inducing substance, which can either be racemic 2-acetylamino-1 -phenyl-4-pentene or racemic N-acetyl-1 -phenylethylamine. Concentrations of 2-acetylamino-1 -phenyl-4-pentene higher than 4.92 mM are inhibitory for the growth and the enzyme formation of strain K1/1. N-acetyl-1 -phenylethylamine at concentrations of 7.38 and 9.84 mM, however, do not inhibit the growth, but neither leads to an increased cell density nor to an higher acylase yield (U/l cu |ture broth)-

b) Variation of the start pH-value and the temperature

Strain K1/1 is grown in shake flasks (see example 21 a) in the presence of 4.92 mM racemic 2-acetylamino-1 -phenyl-4-pentene by varying the start pH-value between 4 and 9 in steps of 0.5 pH values at 28°C and by varying the incubation temperature between 20 and

40°C in steps of 5 C C at pH 7. After 72 h of incubation the O.D.660 and the acylase activity of the crude extracts are measured.

The best production of the acylase of Rhodococcus globerulus K1/1 is achieved at pH 6.5 and 30°C (28 U/lcuKure broth, 0.4 U/mg p rotein in the crude extract) •

Example 22: Enzymatic hydrolysis with highly purified acylase from Arthrobacter aurescens Ac5R

To 10 ml of a 20 mM solution of each of the racemic amides shown below 1 in 0.1 M phosphate buffer pH 7.0, 5 ml (1.3 units) of the enzyme is added and the solutions are shaken at 100 rpm and 30°C.

R 2 0 J 2 i t , II amine acylase T r o

R' /C* N^R 3

H I H (+/-)-! a-12a (+)- or (-)-1b-12b (-)- or (+)-la-12a

2a: R 7 = H, R β = CN

7a 8a 9a 10a: R 9 = -CH 2 CH=CH 2 11a: R 9 = -CH 2 CH 2 CH 3 12a: R 9 = -CH 2 CH 3

When the conversion reaches about 50 % (checked by HPLC) the solutions are acidified to pH 2 by adding 1 N HCI and extracted three times with dichloromethane to obtain the unconverted amides. The aqueous layers are neutralized with 1 N NaOH solution and extracted again three times with dichloromethane to recover the formed amines. Both organic phases are dried with MgSO 4 , filtered and the solvent is distilled off. The amides are used directly for optical purity determinations (HPLC), whereas the obtained amines are converted to the acetamides by adding 100 μl triethylamine and 100 μl acetic anhydride to each sample and then analyzed for the optical purity by "chiral" HPLC. The results are summarized in table 14.

Table 14: Conversion and enantioselectivity of the enzymatic hydrolysis with highly purified acylase from Arthrobacter aurescens Ac5R

Substrate Incubation Conversion e.e. e.e. Selectivity E time (h) (%) (S)-Amide (R)-Amine

1a 22 50.3 >99.9 98.2 >500

2a 75 47.4 82.4 91.5 60

3a 75 51.3 >99.9 95.0 320

4a 139 46.0 87.4 >99.9 >500

5a 46 50.1 96.1 95.7 180

6a 194 35.2 46.6 90.5 32

8a 22 47.4 82.4 91.5 60

Example 23: Enzymatic hydrolysis with partially purified acylase from Rhodococcus equi Ac6

The experiments are carried out in the same manner as described in example 22. 200 ml (1.1 units) enzyme are used for each substrate listed in table 15.

Table 15: Conversion and enantioselectivity of the enzymatic hydrolysis with partially purified acylase from Rhodococcus equi Ac6

Substrate Incubation Conversion e.e. e.e. Selectivity E time (h) (%) (R)-Amide (S)-Amine

1a 18 50.6 99.3 96.8 350

2a 19 46.1 85.5 >99.9 >500

3a 19 49.4 97.4 >99.9 >500

4a 19 50.4 >99.9 98.5 >500

5a 20 52.3 96.7 88.2 65

7a 19 14.4 16.4 97.4 88

8a 20 47.9 90.1 97.8 280

9a 20 51.7 >99.9 92.1 230

Example 24: Amide hydrolysis with whole cells of Rhodococcus globerulus K1/1

To 50 ml of a 10 mM solution in phosphate buffer pH 7.0 of each racemic amide listed in table 16, whole cells of Rhodococcus globerulus K1/1 (each obtained from 50 ml shake flask medium (see example 21), ODβeo = 2.0) is added. The deacetylation is performed at 30°C under continuous shaking (200 rpm). The reaction is monitored by HPLC and after the conversion is about 50 % the reaction mixtures are worked up in the same manner as described in example 22. The results are summarized in table 16.

Table 16: Conversion and enantioselectivity of the enzymatic hydrolysis with whole cells of Rhodococcus globerulus K1/1

Substrate Incubation Conversion e.e. e.e. Selectivity E time (h) (%) (R)-Amide (S)-Amine

1a 3.0 50.6 >99.9 97.5 >500

4a 3.5 52.6 >99.9 90.0 140

6a 3.5 51.7 >99.9 93.5 290

10a 3.5 51.7 95.7 90.0 70

11a 4.0 55.7 97.8 77.8 35

12a 2.0 49.4 90.7 93.0 87

12a 3.5 53.1 >99.9 87.9

Example 25: Preparative hydrolysis of racemic 1a with acylase from Arthrobacter aurescens Ac5R.

To 1 g (6.13 mmol) racemic N-acetyl 1 -phenylethylamine (+/-)-1a, partially dissolved in 100 ml 0.1 M phosphate buffer pH 7.0, 100 ml (25.3 units) acylase from Arthrobacter aurescens Ac5R is added and the mixture is shaken (100 rpm) at 20°C for 27 hours. After acidification (pH 2) by addition of HCI the reaction mixture is extracted three times with 100 ml dichloromethane. The combined organic layers are dried (MgSO 4 ) and the solvent is distilled off after filtration, yielding 0.496 g (49.6 %) of (S)-N-acetyl 1 -phenylethylamine (-)- 1a ([α]° 2 o = -138.0° (c = 1.0 EtOH), enantiomeric ratio S/R = 96.8:3.2) as a white solid. The aqueous phase is neutralized by addition of NaHCO 3 and extracted again with dichloro¬ methane. Usual workup leads to 0.323 g (43.5 %) of (/?)-1 -phenylethylamine (+)-1b ([αf∞ = +29.4° (c = 2.2 EtOH), enantiomeric ratio R/S = 99.7:0.3) as colorless oil.

Example 26 Purification of the acylase of Rhodococcus globerulus K1/1 26.1. Preparation of the crude extract in preparative scale.

100ml of cell suspension (40% w/w) are poured into a 600ml glass beaker and placed in a bucket of ice. The beaker has to remain in an ice bath during sonification. A one inch standard horn is used under the following conditions: cycle time: 1 second duty cycle: 50 % output control: 8 total sonification time: 20 minutes (without interruption) After 20 minutes sonicating (40 minutes including interruption) the sample is centrifuged for 20 minutes at 9000rpm in a Sorvall RC5B refrigerated (4°C) superspeed centrifuge using a Sorvall GS-3 aluminum rotor. The supernatant is the crude extract.

The standard assay for K1/1 -acylase activity used for monitoring the purification and for the enzymological characterization is performed by mixing 20 μl of an enzyme sample with 380 μl of a 5.55 mM solution of (S)-2-acetylamino-1-phenyl-4-pentene in 69 mM potssium phosphate buffer, pH 7, and incubating at 23°C. The dilution of the acylase and the incubation time are chosen so that not more than 1.3 mM of product are formed. The reaction is stopped by addition of 800 μl ice cold methanol. After centrifugation for 4 min at 11500 rpm in a Biofuge 15 (Heraeus Sepatech, Osterode, Germany) the supernatant is subjected to quantitative HPLC analysis (see example 1).

26.2. Nucleic acid precipitation.

3.25ml of a 10% PEI solution (adjusted to pH 7.5 with 6N HCI) are added to 105ml of cell homogenate (from sonification step) to reach a 0.3% concentration. The mixture is kept on ice for 30 minutes and gently stirred. Then the mixture is centrifuged for 20 minutes at 9000 rpm at 4°C. The supernatant is then used for ammonium sulfate precipitation.

26.3. Ammonium sulfate precipitation.

18.81 g of crystalline (NH 4 ) 2 SO 4 are slowly added to 90ml of crude extract after nucleic acid precipitation to reach a concentration of 35 % saturation. The mixture is kept on ice for 30 minutes and gently stirred. Then the mixture is centrifuged for 20 minutes at 9000rpm at 4°C. The supernatant is subjected to chromatography on an FPLC-system (Pharmacia, Uppsala, Sweden) for further purification.

26.4. FPLC.

Two different column types (hydrophobic interaction and gel filtration) coupled to the FPLC equipment are used to purify the acylase.

26.4.1. Hydrophobic interaction chromatography (HIC).

For HIC a Merck Fractogel TSK Butyl-650(S) column (26 x 310mm, volume: 165ml) is prepared and equilibrated with a 35% saturated (NH 4 ) 2 SO 4 solution (in phosphate buffer pH7.0). After loading the column with 90ml of supernatant from the (NH 4 ) 2 SO 4 precipatation the column is washed with 35% saturated (NH 4 ) 2 SO 4 solution until no more protein is washed off the column. Then a (NH 4 );>SO 4 gradient spanning from 35% to 0% saturation with a total volume of 1150ml is applied to the column. The flow is adjusted to 1.8ml/min, fractions of 12ml are collected and tested for acylase activity.

The acylase elutes between 1 1.5% and 7.5% (NH 4 ) 2 S0 4 . Active fractions are pooled (140ml).

26.4.2. Ultrafiltration.

Concentration and desalination of the HIC fraction pool (140 ml) containing the Rhodococcus globerulus K1/1 acylase are performed in a CEC1 Amicon ultrafiltration chamber. A YM30 (MW cut off 30000Da) membrane is utilized. Desalination is achieved by diafiltration and repeated additions of (NH 4 ) 2 SO 4 -free potassium phosphate buffer (69mM, pH7.0) to the concentrated sample followed by a reconcentration step. The HIC fraction pool is concentrated to 4.5ml.

26.4.3. Gel filtration.

A Pharmacia Superose 12HR 10/30 gel filtration column (10 x 300mm) is utilized as the final purification step. The elution is performed using potassium phosphate buffer (69mM, pH7.0) supplemented with 100mM NaCl with a flow rate of 0.3ml/min. 200μl of the concentrated and desalted acylase fraction obtained from ultrafiltration are applied to the column. Fractions of 0.5ml are collected and analyzed for the presence of acylase.

26.4.4. Purification scheme for the Rhodococcus globerulus K1/1 acylase.

*Only 200ml of 4.5ml concentrated and desalted HIC fraction (see Example 26.4.2) are applied to the Pharmacia Superose 12HR column. The data given in the table are calculated for the whole volume of the concentrated HIC fraction (4.5ml).

Example 27: Characterization data of the Rhodococcus globerulus K1/1 acylase

27.1. Activity of the Rhodococcus globerulus K1/1 acylase in the presence of potential enzyme activators and inhibitors:

The K1/1 -acylase is inhibited strongly only by 10 mM Zn 2+ and phenymethylsulfonylfluoride. The severe inhibition by the latter compound even at very low concentrations indicate a serine protease like reaction mechanism of the enzyme.

27.2. General characterization data of the Rhodococcus globerulus K1/1 acylase:

27.2.1. The molecular weight of the acylase is 54.6kDa on SDS-PAGE (see example 4.c. and 5) and 92.3kDa when subjected to gel filtration (see example 5) suggesting a homodimeric structure of the native acylase.

27.2.2. The isoelectric point of the acylase is measured as described in example 6 to be around pH 4.0.

27.2.3. For the determination of activity and stability of the acylase under varying pH conditions the following buffer solutions are utilized: pH5.5 - pH7.5 phosphate buffer pH7.5 - pH9.0 Tris buffer pH9.0 - pH10.5 glycine buffer

For the determination of pH-stability acylase samples are incubated for 2h at 23°C in buffers of various pH. Then the enzyme samples are subjected to standard activity assays (see example 26).

The acylase shows a broad stability optimum between pH5.5 and pH7.5.

The activity of the K1/1 -acylase is measured in standard activity assays (see example 26) under various pH conditions.

The acylase has an optimal activity between pH7.0 and pH7.5 with a sharper decrease of activity on the acidic side of the optimum.

27.2.4. Temperature stability of the Rhodococcus globerulus K1/1 acylase.

Samples of acylase are incubated at various temperatures ranging from 0°C to 65°C for 30 min. After cooling in an ice bath the acylase samples are subjected to standard activity assays (see example 26).

The acylase shows no loss of activity when incubated for 30 min at up to 50°C. Acylase incubated at 55°C looses 40% of its activity whereas acylase incubated at higher temperatures (60°C, 65°C) is fully inactivated within 30min of incubation.

27.2.5. N-terminal amino acid sequence of the Rhodococcus globerulus K1/1 acylase For the determination of the N-terminal amino acid sequence of the K1/1 -acylase a HP G1005A N-terminal Protein Sequencing Systems is utilized performing automated Edman degradation chemistry on protein samples retained on miniature adsorptive biphasic columns consisting of of a reverse-phase sample column mated to a strong anion exchange column. Analysis of PTH-amino acids is performed by an on-line HP1090 HPLC system (Hewlett Packard, Palo Alto, CA, USA).

The N-terminal amino acid sequence (50 residues) is shown in SEQ ID NO 1 :

SQSEIVWASASELAARVRERSLTPVEIGDAMIEHIDAVNPS INAWQFDR .

Example 28: N-terminal amino acid seguence of the (S)-N-acetvH -phenylethylamine acylase of Rhodococcus egui Ac6

The determination of the N-terminal sequence is performed as described in example 27.2.5.

The N-terminal amino acid sequence (30 residues) of the Ac6-acylase is shown in SEQ ID

NO 2: MNTSDPGWMSATEMAAQVASKSLSPNEIAE .

Example 29: N-terminal amino acid sequence of the (R.-specific acylase of Arthrobacter aurescens AcR5b

The determination of the N-terminal sequence is performed as described in example 27.2.5.

The N-terminal amino acid sequence of the small subunit (16kDa) is shown in SEQ ID NO

3: PITNPA??RDHAE, the N-terminal amino acid sequence of the large subunit (89kDa) is sown in SEQ ID NO 4:

TAIΓIΓGY?DTPSVAPGE (?: amino acid unkown, small caps: amino acid not certain).

Example 30: Preparation of (S)-2-amino-1-phenyl-4-pentene using the purified K1/1 acylase

To 50 ml of a 10 mM solution of racemic 2-acetylamino-1 -phenyl-4-pentene in phosphate buffer (pH 7.0) 7 ml of the K1/1 acylase are added (0.456 U), which has been purified by HIC and concentrated by ultrafiltration (example 26.5 and 26.6). The deacylation is performed at 30°C under continuous shaking at 200 rpm. The reaction is monitored by HPLC (see example 1), and after the conversion is about 50 % the solution is acidified to pH 2 with 1 N HCI and extracted three times with one volume of dichloromethane to obtain

the unconverted amides. The aqueous phase is neutralized with 1 N NaOH and extracted again three times with dichloromethane to recover the formed amines. Both organic phases are dried with MgS0 4 , filtered and the solvent is distilled off. The amides are used directly for optical purity determinations via HPLC under the following conditions: Chiralcel OJ column, hexane / isopropanol 9 : 1 as the eluent, flow rate 1 ml/min., detection at 208 nm. The obtained amines are converted to the acetamides by adding 100 ml triethylamine and 100 ml acetic anhydride to each sample before being also analyzed for the optical purity by HPLC.

At 50.3 % conversion (S)-2-amino-1-phenyl-4-pentene is obtained with 87.4 % ee, (R)-N-2- acetylamino-1 -phenyl-4-pentene with 98.8 % ee, corresponding to an E-value of 74.9 %.

Example 31 : Preparation of (S)-2-amino-1 (4-chlorophenyl)-4-oentene using the K1/1- acylase in form of whole cells of Rhodococcus globerulus K1/1

Strain K1/1 is grown according to example 3 in a 30 I fermenter filled with 20 I of basic medium containing 1 g/l of racemic N-acetyl-1 -phenylethylamine. The cells are harvested by continuous centrifugation and resuspended in the reaction mixture, which is prepared by adding a solution of 80 g racemic 2-acetylamino-1 (4-chlorophenyl)-4-pentene in 500 ml methanol to 20 I of potassium phosphate buffer, 69 mM, pH 7. After stirring for 21 h at 28°C and 500 rpm the amine formed and the unreacted amide are separated and analyzed for their optical purity analogously to the proceedure given in example 30.

31.1 g (94.5 % yield) of (S)-2-amino-1 (4-chlorophenyl)-4-pentene with > 99.9 % ee and 38.6 g ( 97 % yield) of (R)-2-acetylamino-1 (4-chlorophenyl)-4-pentene with 98.5 % ee are isolated.

Deposition of Microorganisms

The following microorganisms are deposited according to the Budapest Treaty with the DSM - Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH, Mascheroder Weg 1 b, D-38124 Braunschweig:

Rhodococcus globerulus K1/1 , DSM 10337 (deposited September 23, 1995) Rhodococcus equi Ac6, DSM 10278 (deposited September 23, 1995) Arthrobacter aurescens AcR5b, DSM 10280 (deposited September 23, 1995)

SEQUENCE LISTING

(1) GENERAL INFORMATION:

(i) APPLICANT:

(A) NAME: Novartis AG

(B) STREET: Schartzwaldallee

(C) CITY: Basel

(E) COUNTRY: Switzerland

(F) POSTAL CODE (ZIP): 4002

(G) TELEPHONE: +41 61 696 11 11 (H) TELEFAX: + 41 61 696 79 76 (I) TELEX: 962 991

(ii) TITLE OF INVENTION: Novel Biocataiyst

(iii) NUMBER OF SEQUENCES: 4

(iv) COMPUTER READABLE FORM:

(A) MEDIUM TYPE: Floppy disk

(B) COMPUTER: IBM PC compatible

(C) OPERATING SYSTEM: PC-DOS/MS-DOS

(D) SOFTWARE: Patentln Release #1.0, Version #1.30 (EPO)

(2) INFORMATION FOR SEQ ID NO: 1:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 50 amino acids

(B) TYPE: amino acid

(C) STRANDEDNESS: unknown

(D) TOPOLOGY: unknown

(ii) MOLECULE TYPE: DNA (genomic)

(ix) FEATURE:

(A) NAME/KEY: Peptide

(B) LOCATION:41

(D) OTHER INFORMATION:/product= "amino acid is uncertain"

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1:

Ser Gin Ser Glu lie Val Trp Ala Ser Ala Ser Glu Leu Ala Ala Arg 1 5 10 15

Val Arg Glu Arg Ser Leu Thr Pro Val Glu lie Gly Asp Ala Met lie 20 25 30

Glu His lie Asp Ala Val Asn Pro Ser lie Asn Ala Val Val Gin Phe 35 40 45

Asp Arg 50

(2) INFORMATION FOR SEQ ID NO: 2:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 30 arriino acids

(B) TYPE: amino acid

(C) STRANDEDNESS: unknown

(D) TOPOLOGY: unknown

(ii) MOLECULE TYPE: DNA (genomic)

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2:

Met Asn Thr Ser Asp Pro Gly Trp Met Ser Ala Thr Glu Met Ala Ala 1 5 10 15

Gin Val Ala Ser Lys Ser Leu Ser Pro Asn Glu lie Ala Glu 20 25 30

(2) INFORMATION FOR SEQ ID NO: 3:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 13 amino acids

(B) TYPE: amino acid

(C) STRANDEDNESS: unknown

(D) TOPOLOGY: unknown

(ii) MOLECULE TYPE: DNA (genomic)

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3:

Pro lie Thr Asn Pro Ala Xaa Xaa Arg Asp His Ala Glu

1 5 10

(2) INFORMATION FOR SEQ ID NO: 4:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 18 amino acids

(B) TYPE: amino acid

(C) STRANDEDNESS: unknown

(D) TOPOLOGY: unknown

(ii) MOLECULE TYPE: DNA (genomic)

(ix) FEATURE:

(A) NAME/KEY: Modified-site

(B) LOCATIONS

(D) OTHER INFORMATION:/product= "amino acid is uncertain"

(ix) FEATURE:

(A) NAME/KEY: Modified-site

(B) LOCATIONS

(D) OTHER INFORMATION:/product= "amino acid is uncertain"

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4:

Thr Ala lie Arg lie Arg Gly Tyr Xaa Asp Thr Pro Ser Val Ala Pro 1 5 10 15

Gly Glu