WO2001019849A1 - A process for the preparation of h-tyr-d-ala-phe(f)-phe-nh¿2? - Google Patents

A process for the preparation of h-tyr-d-ala-phe(f)-phe-nh¿2? Download PDF

Info

Publication number
WO2001019849A1
WO2001019849A1 PCT/SE2000/001747 SE0001747W WO0119849A1 WO 2001019849 A1 WO2001019849 A1 WO 2001019849A1 SE 0001747 W SE0001747 W SE 0001747W WO 0119849 A1 WO0119849 A1 WO 0119849A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
derivative
ester
protecting group
benzyl
Prior art date
Application number
PCT/SE2000/001747
Other languages
French (fr)
Inventor
Mårten ELLBURG
Henry FRANZÈN
Maths Nilsson
Original Assignee
Astrazeneca Ab
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Astrazeneca Ab filed Critical Astrazeneca Ab
Priority to CA002383184A priority Critical patent/CA2383184A1/en
Priority to EP00963222A priority patent/EP1212350A1/en
Priority to JP2001523626A priority patent/JP2003509437A/en
Priority to AU74669/00A priority patent/AU7466900A/en
Publication of WO2001019849A1 publication Critical patent/WO2001019849A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1027Tetrapeptides containing heteroatoms different from O, S, or N
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • A61P29/02Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID] without antiinflammatory effect
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention is directed to a new process for the preparation of a tetrapeptide, more specifically the tetrapeptide H-Tyr-D-Ala-Phe(pF)-Phe- NH2, or a pharmaceutically acceptable salt thereof.
  • the present invention also relates to new intermediates used in the process.
  • WO 97/07129 discloses a process for producing inter alia the peptide H-Tyr-D-Ala- Phe(pF)-Phe-NH 2 .
  • the peptide H-Tyr-D-Ala-Phe(pF)-Phe-NH 2 is also disclosed in WO 97/07130.
  • Said peptide exhibits peripheral analgesic activity and selectivity for the ⁇ - subtype of opioid receptors, and is particularly suitable in pain therapy.
  • it is prepared using solid phase synthesis according to procedures well established in the art. The drawback with solid phase synthesis is that it is difficult to use in large-scale production, in addition to being expensive.
  • WO99/47548 discloses a process for the preparation of the tetrapeptide H-Tyr-D-Ala- Phe(pF)-Phe- NHb using stepwise synthesis.
  • the process of the present invention provides the tetrapeptide H-Tyr-D-Ala-Phe(pF)-Phe- NH 2 in a simpler manufacturing process with, e.g. easier purification of the final product.
  • the object of the present invention is to provide a novel process suitable for use in large-scale synthesis.
  • a further object of the present invention is to provide a process containing as few reaction steps as possible.
  • the present invention provides a new process for large-scale preparation of the peptide H- Tyr-D-Ala-Phe(pF)-Phe-NH2, which is the peptide of formula (I)
  • the process according to the present invention for preparing the compound of formula (I) is a fragment synthesis (2+2).
  • a fragment synthesis a plurality of intermediate compounds, are prepared in parallel and then coupled together to give the key intermediate(s) or the final compound.
  • This strategy should be compared to a traditional stepwise synthesis wherein a number of synthetic steps are performed sequential.
  • the different approaches in a stepwise vs. fragment synthesis are schematically shown in Figure 1 below.
  • A is an amino protecting group
  • R is an activating agent residue group
  • 2 R is H or a benzyl-like group; previously prepared by a pre-activation step or generated in situ, is reacted with the amino group of D-alanine, wherein the carboxyl group is protected as an ester, i.e. a compound of tthhee ffoorrmmuullaa DD--AAllaa--RR wwhheerreeiinn RR iiss tthhee eesstteerr ggrrooiup, e.g. OMe, in the presence of a solvent, providing a protected dipeptide derivative (IV)
  • A is an amino protecting group
  • R is an ester residue group
  • Step 2 A deprotection step wherein a protected dipeptide derivative (IV) prepared in the previous step, is deprotected by treatment with aqueous base or acid to give the dipeptide derivative (V),
  • A is an amino protecting group
  • activated tyrosine derivative is an activated ester or urethane protected N- carboxyanhydride (UNCA) of the structure (II')
  • A is an amino protecting group
  • the carboxyl group of D-alanine needs no protection, i.e. can be a compound of the formula D-Ala-OH, and the coupling reaction thereby provides a protected dipeptide derivative V, which can be used directly in the step 3 without any further deprotection.
  • UNCA-derivatives The preparation and use of UNCA-derivatives is discussed by Fehrentz et al. (1995). "The use of N-urethane protected N-carboxyanhydrides (UNCAs) in amino acid and peptide synthesis.” J. Pept. Sci., 1(2), 124-131 ; and by Fuller et al, (1996). "Urethane-protected a- aminoacid N-carboxanhydrides and peptide synthesis.” Biopolymers (Peptide Science), 40(2), 183-205 which are incorporated herein by reference.
  • the activated tyrosine derivative (II) can be reacted with the amino group of non-protected D-Ala, i.e. H-D-Ala-OH, providing dipeptide derivative (V) directly.
  • Step 1' A coupling step wherein an activated /7-fluorophenylalanine derivative (VI),
  • A is an amino protecting group
  • R is an activating agent residue group
  • previously prepared by a pre-activation step or generated in situ is reacted with the amino group of phenylalanine, wherein the carboxyl group is protected as an ester or amide, i.e. a compound of the formula Phe-R (VII), wherein R is -NH2 or an ester residue group, e.g. OMe, in the presence of a solvent, providing a protected dipeptide derivative (VIII)
  • A is an amino protecting group, and R is -NH2 or an ester residue group;
  • Step 2' A deprotection step wherein a protected dipeptide derivative (VIII) prepared in the previous step, is deprotected by either catalytic hydrogenation, base or acid treatment, depending on the amino protecting group used, to give the dipeptide derivative (IX),
  • R is -NH2 or an ester residue group
  • A is an amino protecting group
  • R is an activating agent residue group
  • A is an amino protecting group
  • R is -NH2 or an ester residue group
  • A is an amino protecting group
  • R 1 is -NH 2 .
  • the additional step described above may be prepared on the protected dipeptide derivative (VIII), if R is an ester, whereby the ester compound (VIII) is reacted with an amine in an organic alcohol, preferably ammonia in methanol, providing the protected dipeptide derivative (VHP),
  • A is an amino protecting group, and R 1 is -NH 7
  • Step 4 A deprotection step wherein the protected tetrapeptide derivative (X) is deprotected either by catalytic hydrogenation, or treatment with acid or base, depending on the amino protecting group used, providing the final tetrapeptide (I), which optionally may be converted to a pharmaceutically acceptable salt thereof
  • N -amino protecting group may be selected from any protecting group suitable in peptide synthesis, such as tert- butoxycarbonyl (Boc), 9-fluorenylmetoxycarbonyl (Fmoc) or benzyloxycarbonyl, often abbreviated Z-, just to mention three possible amino protecting groups
  • benzyloxycarbonyl is particularly preferred to be used in the present invention since it is easily removed by catalytic hydrogenation, and contrary to the protecting group Boc, it does not require neutralization of the liberated amine C i-C ⁇ alkyl esters and alkylaryl ester, such as benzyl, are preferred carboxyl protecting groups
  • Methyl esters are particularly preferred carboxyl protecting groups
  • Benzyl-like protecting groups are suitable tyrosine side-chain protecting groups to be used in the present invention Preferably no tyrosine side
  • the pre-activation step preceding steps 1 , 1 ' and 3, or the in situ generation of the activated amino acid derivatives (II), (VI) and (V), is achieved by reacting an amino acid, wherein the amino function has been protected by a suitable protecting group, such as tert- butoxycarbonyl (Boc), 9-fluorenylmetoxycarbonyl (Fmoc) or benzyloxycarbonyl (Z), which are either commercially available or available by techniques known in the art, with an activating agent in the presence of a suitable amine and an organic solvent, to give the activated amino acid derivative.
  • a suitable protecting group such as tert- butoxycarbonyl (Boc), 9-fluorenylmetoxycarbonyl (Fmoc) or benzyloxycarbonyl (Z), which are either commercially available or available by techniques known in the art
  • A is an amino protecting group
  • R is an activating agent residue group
  • Steps 1, Land 3 a variety of powerful solvents may be used, as long as the amino component is essentially soluble and available for immediate reaction with the activated peptide derivative.
  • suitable solvents for the coupling step are acetone, acetonitrile, DMF, N-methyl pyrrolidone (NMP), EtOAc, and mixtures thereof.
  • benzyl-like group denotes any substituted or un-substituted benzyl group that is hydrogenolyzed under similar reaction conditions as the benzyloxycarbonyl group.
  • C 1-C alkyl ' denotes a cyclic or linear, straight or branched, substituted or unsubstituted alkyl group having from 1 to 6 carbon atoms
  • alkyl include but are not limited to methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert - butyl, cyclohexyl. and cyclopentyl
  • substituted denotes a group that is substituted by one or more C
  • Suitable activating agents may be selected from those that generates any of the commonly used activated ammo acid derivatives including, but not limited to, carbod ⁇ mides, activated esters, azide, or anhydrides
  • Isobutylchloroformiate (iBuOCOCl) and 2-( lH- Benzot ⁇ azole-l-yl)- l , l,3,3-tetramethyluron ⁇ um tetrafluoroborate (TBTU) are the preferred activating agents together with UNCA-de ⁇ vatives
  • the amount of activating agent is between 0 9- 1 2 molar equivalents, preferably 0 95- 1 05 equivalents From a practical point of view the amount of activating agent shall be as close to 1 0 as possible
  • isobutylchloroformiate (iBuOCOCl) is the activating agent
  • the activated peptide derivative will have the following structure, exemplified on D-alanine,
  • the suitable amine may be selected from any tertiary amine However, NMM (N-methylmorphohne), di-isopropylethylamine and t ⁇ ethylamine are preferred.
  • the amount of amine is between 0 9-2 0 molar equivalents, compared to the acid, and preferable between 0.95 to 1.5 molar equivalents. From a practical point of view the amount of suitable amine shall be at least equal to the molar amount of activating agent used.
  • the organic solvent may be any organic solvent known to a person skilled in the art to be suitable in peptide chemistry. However, ethyl acetate, acetonitrile, acetone, tetrahydrofurane, DMF as well as mixtures thereof are preferred solvents in the pre- activation step.
  • the solvent used for the coupling step may be selected from a variety of solvents, as long as the amino component is essentially soluble and available for immediate reaction with the activated amino acid residue.
  • suitable solvents for the coupling steps are acetone, acetonitrile, DMF, N-methyl pyrrolidone (NMP), EtOAc, and mixtures thereof, of which acetone, EtOAc, NMP and DMF are preferred.
  • any temperature where the activated amino acid derivative is not degraded or the reaction rate is too slow may be used.
  • the preferred range when isobutyl chloroformate is used as the activating agent is from 0°C to -20°C, and particularly preferred is from -5°C to -15°C.
  • the preferred range when TBTU is used as the activating agent is around room temperature. The rate of addition is in both cases adjusted so that the preferred temperature is maintained in the reaction mixture.
  • the catalyst used for hydrogenation may be selected from a great variety of catalysts as will be appreciated by a person skilled in the art. However 5% Pd on carbon is preferred. Any solvent that can dissolve at least some of the peptide is possible to use except ketones, such as acetone, or those solvents that poison the catalyst or react with the components of the reaction. A person skilled in the art will appreciate the choice of solvent. DMF and NMP are the preferred solvents.
  • Hydrolysis of the ester residue group in compound (IV) can be achieved by any method known to the skilled person, e.g. aqueous acid, base treatment or hydro genolyzis, depending on the carboxyl protecting group used.
  • Cj-C ⁇ alkyl esters are preferred esters and treatment with aqueous base under standard conditions is the preferred method for ester hydrolysis.
  • the protected amino acid preferably oc using benzyloxycarbonyl- as N -amino protecting group
  • the method employed is based on the general method reviewed by J. Meienhofer in The Peptides,
  • the activation time can be extended to at least 30 min at a temperature about 0 - -15°C, contrary to the recommended 1-2 min at -15°C.
  • strictly anhydrous conditions are not necessary as otherwise is recommended. This allows the present method to be used for large-scale production where the longer reaction times allow a safe and reproducible process to be carried out.
  • the stereochemical integrity has been completely maintained and the chemical purity as well as yields have been typically over 90%.
  • the generated mixed anhydride is coupled with the slow addition of the amino component (amino acid/ peptide amide or ester) at about 0 - -15°C and the reaction mixture is then allowed to reach 20-30°C in about 30-60 min. or longer before crystallization of the product is initiated directly from the reaction mixture.
  • isobutylchloroformiate can be used in the key step in the fragment synthesis of the present invention, i.e. coupling step 3, without any substantial racemization of the D-alanine aminoacid fragment.
  • Another object of the present invention is to provide new intermediates that can be used in the preparation of compound of formula I.
  • a further aspect of the present invention is a compound of the general formula X'
  • A is tert-butoxycarbonyl (Boc), 9-fluorenylmethoxycarbonyl (Fmoc), benzyloxycarbonyl
  • R 1 is -NH 2 ,C r C 6 alkyl ester, benzyl ester, OH, 9-fluorenylmetyl ester or a substituted benzyl ester derivative:
  • R 2 is H, a benzyl-like group, tert. butyl group, or 9-flourenylmefhyl group, as intermediates for use according to the present invention.
  • Z-Tyr-D-Ala-OMe ( l.Og, 2.46mmol, of purity 95%) was dissolved in dioxane (8mL). 1M NaOH (aq) (5.2mL) was then charged and the reaction left over night. The solvents were removed by vacuum distillation. The residue was dissolved in EtOAc (250mL) and extracted, first with brine (4x75mL) followed by 1M KHSO 4 (3x75mL). The organic layer was then dried over MgSO (anh d ) for several hours before filtration. The filtrate was evaporated to dryness by vacuum distillation.
  • Example 3 Z-Tyr-OH (63,1 g, 200 mmol) and H-(D)-Ala-OMe x HCl (31,0 g, 222 mmol) were charged to a one liter reactor under nitrogen. Acetone (400 ml) was added and the slurry cooled to -20°C. Isobutylchloroformiat (30, 1 g, 220 mmol) was then added, quickly followed by N-Methylmorpholine (47,9 g, 472 mmol) while maintaining the temp at about -10°C. Upon completion of the NMM-charge the temp was allowed to reach 20°C, the precipitate filtered off and washed with Acetone (100ml).
  • a 250 mL flask was charged with H-D-Alanine-OH (4,52 g; 50,2 mmol; 2 eq), potassium carbonate anhydrate (7,05 g; 50,2 mmol; 2 eq) and polyethylene glycol 200 (50 mL; 11,1 mL/g Alanine), which was stirred at ambient temperature.
  • Another 250 mL flask was charged with Z-Tyrosine (8,00 g; 25,1 mmol) and EtOAc (100 mL; 12,6 mL/g Z- Tyrosine).
  • Z-Phe(pF)-OH ( 1 eq.) is first dissolved in acetonitrile (MeCN)( 1.7L/mole) and cooled before addition of z ' -Butylchloroformiate ( 1.05 eq). The reaction is then controlled by the rate of addition, (about 20 minutes) 15 min actual, of N-Methylmorpholine ( 1.4eq). A reaction temperature between 0 and -15°C is recommended where the reaction occurs immediately upon addition of N-Methylmorphohne, yet prevents the mixed anhydride from decomposing to rapidly
  • H-Phe- ⁇ H 2 x HCl ( 1 04 eq) is meanwhile dissolved in DMF (4 OL/mole), neutralized with s N-Methylmorpholine (1 04eq) and cooled to about -10°C This slurry is upon completion of the activation added at a rate that maintains the temperature around -10°C for about 15 minutes
  • Z-Phe(pF)-Phe-NH prepared in the previous step is mixed with DMF (3 5L/mole) and a 0 Pd/C catalyst (5% Pd) is added 5%, by weight and the resulting mixture hydrogenated for more than 0 5 hours at 25-30°C and about 3bar H 2
  • the reaction mixture is then filtered and cooled to about -15°C before the next step 99 6% purity in solution and >99% conversion of starting material
  • Example 8 Z-Tyr-(D)-Ala-OH (15.0 g, 39 mmol) and H-Phe(pF)-Phe-NH2xHCl (12.8 g, 35 mmol) were mixed with aceton (450 ml) in a one liter reactor. The slurry temperature was reduced to -10°C prior to addition of isobutylchloroformate (4.55 ml, 35 mmol). NMM (8.45 ml, 77 mmol) was slowly charged to maintain the temp at about -10°C. Upon completed addition the temp was increased to room temperature and 2M HCl (40 ml, 80 mmol) added followed by water (365 ml).
  • the free base compound I is dissolved in a mixture of water and acetone with one equivalent HCl added and clear filtered (146g/mole 25% HCl/H 2 O, 2L Acetone/mole in actual run).
  • the salt has a limited solubility in acetone and therefore the filter is washed once with an additional amount of the acetone/water (95:5) mixture (0.5L/mole).
  • the crystallization is initiated by a slow addition of acetone (3.4L/mole) at high agitation rate and then 1% w/w of seeding crystals is optionally added. After 30 minutes the first amount of MIBK (3L/mole) is slowly charged and left with slow stirring until the batch clearly thickens.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pain & Pain Management (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Rheumatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention discloses a new and improved process for the preparation of the tetrapeptide H-Tyr-D-Ala-Phe(F)-Phe-NH2 that is a peptide of formula (I), or a pharmaceutically acceptable salt thereof, as well as new intermediates in the preparation thereof. The novel process is a fragment synthesis and suitable for large-scale production.

Description

A PROCESS FOR THE PREPARATION OF H-TYR-D-ALA-PHE(F)-PHE-NH2
FIELD OF THE INVENTION
The present invention is directed to a new process for the preparation of a tetrapeptide, more specifically the tetrapeptide H-Tyr-D-Ala-Phe(pF)-Phe- NH2, or a pharmaceutically acceptable salt thereof. In further aspects, the present invention also relates to new intermediates used in the process.
BACKGROUND AND PRIOR ART
WO 97/07129 discloses a process for producing inter alia the peptide H-Tyr-D-Ala- Phe(pF)-Phe-NH2. The peptide H-Tyr-D-Ala-Phe(pF)-Phe-NH2 is also disclosed in WO 97/07130. Said peptide exhibits peripheral analgesic activity and selectivity for the μ- subtype of opioid receptors, and is particularly suitable in pain therapy. Furthermore, it is prepared using solid phase synthesis according to procedures well established in the art. The drawback with solid phase synthesis is that it is difficult to use in large-scale production, in addition to being expensive.
WO99/47548 discloses a process for the preparation of the tetrapeptide H-Tyr-D-Ala- Phe(pF)-Phe- NHb using stepwise synthesis.
The process of the present invention provides the tetrapeptide H-Tyr-D-Ala-Phe(pF)-Phe- NH2 in a simpler manufacturing process with, e.g. easier purification of the final product.
Thus, the object of the present invention is to provide a novel process suitable for use in large-scale synthesis. A further object of the present invention is to provide a process containing as few reaction steps as possible. OUTLINE OF THE INVENTION
The present invention provides a new process for large-scale preparation of the peptide H- Tyr-D-Ala-Phe(pF)-Phe-NH2, which is the peptide of formula (I)
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof.
The process according to the present invention for preparing the compound of formula (I) is a fragment synthesis (2+2). In a fragment synthesis a plurality of intermediate compounds, are prepared in parallel and then coupled together to give the key intermediate(s) or the final compound. This strategy should be compared to a traditional stepwise synthesis wherein a number of synthetic steps are performed sequential. The different approaches in a stepwise vs. fragment synthesis are schematically shown in Figure 1 below.
B -→- C -→- D -→- E -→- F -→- G -→- product
Stepwise synthesis
G — *- product
Figure imgf000004_0001
Fragment synthesis
Figure 1. Stepwise vs. fragment synthesis
The process of the present invention can be described as comprising the steps shown in Figure 2 below;
2 step 1 step 2
+
Figure imgf000004_0002
Figure 2. Fragment synthesis according to the present invention
Step 1
A coupling step wherein an activated tyrosine derivative (II),
Figure imgf000005_0001
wherein
A is an amino protecting group, and
R is an activating agent residue group, and
2 R is H or a benzyl-like group; previously prepared by a pre-activation step or generated in situ, is reacted with the amino group of D-alanine, wherein the carboxyl group is protected as an ester, i.e. a compound of tthhee ffoorrmmuullaa DD--AAllaa--RR wwhheerreeiinn RR iiss tthhee eesstteerr ggrrooiup, e.g. OMe, in the presence of a solvent, providing a protected dipeptide derivative (IV)
Figure imgf000005_0002
wherein A is an amino protecting group,
R is an ester residue group, and
2 R is H or a benzyl-like group;
Step 2 A deprotection step wherein a protected dipeptide derivative (IV) prepared in the previous step, is deprotected by treatment with aqueous base or acid to give the dipeptide derivative (V),
Figure imgf000006_0001
wherein
A is an amino protecting group, and
2 R is H or a benzyl-like group.
However, if the activated tyrosine derivative is an activated ester or urethane protected N- carboxyanhydride (UNCA) of the structure (II')
Figure imgf000006_0002
wherein A is an amino protecting group, and
2 R is H or a benzyl-like group then the carboxyl group of D-alanine needs no protection, i.e. can be a compound of the formula D-Ala-OH, and the coupling reaction thereby provides a protected dipeptide derivative V, which can be used directly in the step 3 without any further deprotection.
The preparation and use of UNCA-derivatives is discussed by Fehrentz et al. (1995). "The use of N-urethane protected N-carboxyanhydrides (UNCAs) in amino acid and peptide synthesis." J. Pept. Sci., 1(2), 124-131 ; and by Fuller et al, (1996). "Urethane-protected a- aminoacid N-carboxanhydrides and peptide synthesis." Biopolymers (Peptide Science), 40(2), 183-205 which are incorporated herein by reference.
Alternatively, the activated tyrosine derivative (II) can be reacted with the amino group of non-protected D-Ala, i.e. H-D-Ala-OH, providing dipeptide derivative (V) directly.
Step 1' A coupling step wherein an activated /7-fluorophenylalanine derivative (VI),
Figure imgf000007_0001
wherein A is an amino protecting group, and R is an activating agent residue group; previously prepared by a pre-activation step or generated in situ, is reacted with the amino group of phenylalanine, wherein the carboxyl group is protected as an ester or amide, i.e. a compound of the formula Phe-R (VII), wherein R is -NH2 or an ester residue group, e.g. OMe, in the presence of a solvent, providing a protected dipeptide derivative (VIII)
Figure imgf000008_0001
wherein A is an amino protecting group, and R is -NH2 or an ester residue group;
Step 2' A deprotection step wherein a protected dipeptide derivative (VIII) prepared in the previous step, is deprotected by either catalytic hydrogenation, base or acid treatment, depending on the amino protecting group used, to give the dipeptide derivative (IX),
Figure imgf000008_0002
wherein
R is -NH2 or an ester residue group; Step 3
A coupling step wherein an activated dipeptide derivative (V),
Figure imgf000009_0001
wherein
A is an amino protecting group,
R is an activating agent residue group, and
2 R is H or a benzyl-like group previously prepared by a pre-activation step or generated in situ from compound (V), is reacted with the amino group of compound (IX) in the presence of a solvent, providing a protected tetrapeptide derivative (X)
Figure imgf000009_0002
wherein
A is an amino protecting group,
R is -NH2 or an ester residue group, and
2 R is H or a benzyl-like group An additional transformation step is performed if the protected tetrapeptide derivative (X). prepared in the previous step, is an ester. Thus the ester compound (X) wherein R is an ester residue group, e.g. OMe, is reacted with an amine in an organic alcohol, preferably ammonia in methanol, providing the protected dipeptide derivative (X'),
Figure imgf000010_0001
wherein
A is an amino protecting group,
R1 is -NH2, and
2 R is H or a benzyl-like group
Optionally, the additional step described above may be prepared on the protected dipeptide derivative (VIII), if R is an ester, whereby the ester compound (VIII) is reacted with an amine in an organic alcohol, preferably ammonia in methanol, providing the protected dipeptide derivative (VHP),
Figure imgf000010_0002
wherein
A is an amino protecting group, and R1 is -NH7
Step 4 A deprotection step wherein the protected tetrapeptide derivative (X) is deprotected either by catalytic hydrogenation, or treatment with acid or base, depending on the amino protecting group used, providing the final tetrapeptide (I), which optionally may be converted to a pharmaceutically acceptable salt thereof
A person skilled in the art will appreciate suitable amino, carboxyl and tyrosine side-chain ex protecting groups, which may be used in the present invention The N -amino protecting group may be selected from any protecting group suitable in peptide synthesis, such as tert- butoxycarbonyl (Boc), 9-fluorenylmetoxycarbonyl (Fmoc) or benzyloxycarbonyl, often abbreviated Z-, just to mention three possible amino protecting groups However, benzyloxycarbonyl is particularly preferred to be used in the present invention since it is easily removed by catalytic hydrogenation, and contrary to the protecting group Boc, it does not require neutralization of the liberated amine C i-C^ alkyl esters and alkylaryl ester, such as benzyl, are preferred carboxyl protecting groups Methyl esters are particularly preferred carboxyl protecting groups Benzyl-like protecting groups are suitable tyrosine side-chain protecting groups to be used in the present invention Preferably no tyrosine side-chain protecting group is used Reference is made to J Meienhofer in The Peptides Vol 1 Eds E Gross & J Meienhofer, Academic Press Inc London 1979, pp 264-309 The peptides, Vol 1-9, E Gross & J Meienhofer Eds Academic Press Inc , London 1979-198~, Houben-Weyl Methoden der organischen Chemie, E Muller, ed , Vol 15 Part I-II Thieme, Stuttgart 1974, and M Bodanszky Principles of peptide Synthesis Springer Verlag, Berlin 1984
The pre-activation step preceding steps 1 , 1 ' and 3, or the in situ generation of the activated amino acid derivatives (II), (VI) and (V), is achieved by reacting an amino acid, wherein the amino function has been protected by a suitable protecting group, such as tert- butoxycarbonyl (Boc), 9-fluorenylmetoxycarbonyl (Fmoc) or benzyloxycarbonyl (Z), which are either commercially available or available by techniques known in the art, with an activating agent in the presence of a suitable amine and an organic solvent, to give the activated amino acid derivative. A schematic representation of a pre-activation step is shown below;
Figure imgf000012_0001
(VI) aminoacid derivative activated aminoacid derivative wherein
A is an amino protecting group, and
R is an activating agent residue group;
For the coupling step, in Steps 1, Land 3 described above, a variety of powerful solvents may be used, as long as the amino component is essentially soluble and available for immediate reaction with the activated peptide derivative. Examples of suitable solvents for the coupling step are acetone, acetonitrile, DMF, N-methyl pyrrolidone (NMP), EtOAc, and mixtures thereof.
As used herein, the term " benzyl-like group " denotes any substituted or un-substituted benzyl group that is hydrogenolyzed under similar reaction conditions as the benzyloxycarbonyl group.
The term "pF' denotes a/?αra-fluoro substituent. The term "C 1-C alkyl ' denotes a cyclic or linear, straight or branched, substituted or unsubstituted alkyl group having from 1 to 6 carbon atoms Examples of said alkyl include but are not limited to methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert - butyl, cyclohexyl. and cyclopentyl
The term "substituted" denotes a group that is substituted by one or more C|-C6 alkyl, C C6 alkoxy, halogen, amino, thiol, nitro, hydroxy, Cι-C6 acyl or cyano groups
Possible as well as preferred reagents and reaction conditions in each step are the following
The pre-activation or in situ generation step
Suitable activating agents may be selected from those that generates any of the commonly used activated ammo acid derivatives including, but not limited to, carbodπmides, activated esters, azide, or anhydrides Isobutylchloroformiate (iBuOCOCl) and 2-( lH- Benzotπazole-l-yl)- l , l,3,3-tetramethyluronιum tetrafluoroborate (TBTU) are the preferred activating agents together with UNCA-deπvatives The amount of activating agent is between 0 9- 1 2 molar equivalents, preferably 0 95- 1 05 equivalents From a practical point of view the amount of activating agent shall be as close to 1 0 as possible When isobutylchloroformiate (iBuOCOCl) is the activating agent, the activated peptide derivative will have the following structure, exemplified on D-alanine,
Figure imgf000013_0001
The suitable amine may be selected from any tertiary amine However, NMM (N-methylmorphohne), di-isopropylethylamine and tπethylamine are preferred The amount of amine is between 0 9-2 0 molar equivalents, compared to the acid, and preferable between 0.95 to 1.5 molar equivalents. From a practical point of view the amount of suitable amine shall be at least equal to the molar amount of activating agent used.
The organic solvent may be any organic solvent known to a person skilled in the art to be suitable in peptide chemistry. However, ethyl acetate, acetonitrile, acetone, tetrahydrofurane, DMF as well as mixtures thereof are preferred solvents in the pre- activation step.
The coupling step; Steps 1, 1' and 3
The solvent used for the coupling step may be selected from a variety of solvents, as long as the amino component is essentially soluble and available for immediate reaction with the activated amino acid residue. Examples of suitable solvents for the coupling steps are acetone, acetonitrile, DMF, N-methyl pyrrolidone (NMP), EtOAc, and mixtures thereof, of which acetone, EtOAc, NMP and DMF are preferred.
Any temperature where the activated amino acid derivative is not degraded or the reaction rate is too slow may be used. The preferred range when isobutyl chloroformate is used as the activating agent is from 0°C to -20°C, and particularly preferred is from -5°C to -15°C. The preferred range when TBTU is used as the activating agent is around room temperature. The rate of addition is in both cases adjusted so that the preferred temperature is maintained in the reaction mixture.
The deprotection step; Step V and 4
The catalyst used for hydrogenation may be selected from a great variety of catalysts as will be appreciated by a person skilled in the art. However 5% Pd on carbon is preferred. Any solvent that can dissolve at least some of the peptide is possible to use except ketones, such as acetone, or those solvents that poison the catalyst or react with the components of the reaction. A person skilled in the art will appreciate the choice of solvent. DMF and NMP are the preferred solvents.
The deprotection step; Step 2
Hydrolysis of the ester residue group in compound (IV) can be achieved by any method known to the skilled person, e.g. aqueous acid, base treatment or hydro genolyzis, depending on the carboxyl protecting group used. Cj-Cό alkyl esters are preferred esters and treatment with aqueous base under standard conditions is the preferred method for ester hydrolysis.
In a preferred embodiment of the present invention the protected amino acid, preferably oc using benzyloxycarbonyl- as N -amino protecting group, is activated as a mixed anhydride with isobutyloxycarbonylchloride, or a similar type of chloroformate. The method employed is based on the general method reviewed by J. Meienhofer in The Peptides,
Vol. l, Eds: E. Gross & J. Meienhofer, Academic Press, Inc, London 1979, pp. 264-309.
We have surprisingly found that the activation time can be extended to at least 30 min at a temperature about 0 - -15°C, contrary to the recommended 1-2 min at -15°C. We also found that strictly anhydrous conditions are not necessary as otherwise is recommended. This allows the present method to be used for large-scale production where the longer reaction times allow a safe and reproducible process to be carried out. The stereochemical integrity has been completely maintained and the chemical purity as well as yields have been typically over 90%. The generated mixed anhydride is coupled with the slow addition of the amino component (amino acid/ peptide amide or ester) at about 0 - -15°C and the reaction mixture is then allowed to reach 20-30°C in about 30-60 min. or longer before crystallization of the product is initiated directly from the reaction mixture.
We have also surprisingly found that when using the present method, if appropriately selected solvent combinations are used, there is no need for a separate washing step prior to crystallization. DMF, acetonitrile, EtOAc and water are preferably used as solvents. A controlled crystallization achieves an excellent purification, shortens the filtering or centrifugation time during work up, as well as shortens the drying time, if dry intermediates are required. One important factor is to generate sufficiently large crystals with a relatively narrow size distribution not to block the filter medium or centrifugation cloth. It is very common for peptides in particular to generate gels or amorphous crystals that are almost impossible to filter.
We have further surprisingly found that isobutylchloroformiate can be used in the key step in the fragment synthesis of the present invention, i.e. coupling step 3, without any substantial racemization of the D-alanine aminoacid fragment.
INTERMEDIATES
Another object of the present invention is to provide new intermediates that can be used in the preparation of compound of formula I.
Accordingly, a further aspect of the present invention is a compound of the general formula X'
Figure imgf000016_0001
wherein A is tert-butoxycarbonyl (Boc), 9-fluorenylmethoxycarbonyl (Fmoc), benzyloxycarbonyl
(Z) or a substituted benzyloxycarbonyl derivative;
R1 is -NH2,CrC6 alkyl ester, benzyl ester, OH, 9-fluorenylmetyl ester or a substituted benzyl ester derivative:
R2 is H, a benzyl-like group, tert. butyl group, or 9-flourenylmefhyl group, as intermediates for use according to the present invention.
DETAILED DESCRIPTION OF THE INVENTION
The preparation of the peptide H-Tyr-D-Ala-Phe(pF)-Phe-NH, or a pharmaceutically acceptable salt thereof, will now be described in more detail by the following Examples, which should not be construed as limiting the invention. Furthermore, Scheme 1 below provides a detailed overview of the synthetic route followed for the preparation of the peptide of the formula (I) according to the present invention using an alanine derivative, wherein the carboxyl group is protected as an amid. The compound numbers referred to in the Examples below corresponds to the compound numbering in Scheme 1.
Figure imgf000017_0001
Z-Phe(pF)-OH iBuOCOCl zv rmn. z-Phe(pF)-OCOOiBu (6)
-10°C - +25°C EtOAc + DMF
Figure imgf000017_0002
H-Phe(pF)-Phe-NH2 40°C
Z-Phe(pF)-Phe-NH2 (9)
Scheme 1 (8)
Figure imgf000018_0001
Scheme 1, cont'd EXAMPLES
Step l Preparation of Z-Tyr-D-Ala-OMe (Compound 4 in Scheme 1)
Example 1
2,85g (22mmol) DEEA was added dropwise to an ice-cool solution of Z-Tyr-OH (2), H-D- Ala-OMe (3) and TBTU in 10ml of EtOAc (lOmmol each). The reaction mixture was left to assume room temperature after 25min at 4°C. The solvents were then evaporated and the residue partitioned between EtOAc and water. The organic phase was washed with 3 x 1M Na2CO3, 3x 1M KHSO4 and brine before drying over Na2SO4. The product was crystallized by the addition of light petroleum ether to yield 3.2g (80%) and 99% pure.
Step 2
Preparation of Z-Tyr-D-AIa-OH (Compound 5 in Scheme 1)
Example 2
Z-Tyr-D-Ala-OMe ( l.Og, 2.46mmol, of purity 95%) was dissolved in dioxane (8mL). 1M NaOH(aq) (5.2mL) was then charged and the reaction left over night. The solvents were removed by vacuum distillation. The residue was dissolved in EtOAc (250mL) and extracted, first with brine (4x75mL) followed by 1M KHSO4 (3x75mL). The organic layer was then dried over MgSO (anh d) for several hours before filtration. The filtrate was evaporated to dryness by vacuum distillation. The residual oil was dissolved in Acetone EtOAc (2: 1, 5mL) and then isopropyl ether (30mL) was charged slowly in order to precipitate the product. The solid was filtered off and dried under reduced pressure at 30°C, yielding 0.8g (86%) of the product with a purity of 99% (HPLC).
Example 3 Z-Tyr-OH (63,1 g, 200 mmol) and H-(D)-Ala-OMe x HCl (31,0 g, 222 mmol) were charged to a one liter reactor under nitrogen. Acetone (400 ml) was added and the slurry cooled to -20°C. Isobutylchloroformiat (30, 1 g, 220 mmol) was then added, quickly followed by N-Methylmorpholine (47,9 g, 472 mmol) while maintaining the temp at about -10°C. Upon completion of the NMM-charge the temp was allowed to reach 20°C, the precipitate filtered off and washed with Acetone (100ml). The product was slowly charged to NaOH (20 g, 500 mmol) in water (600 ml) at RT. HCl, 32% (60 ml) was added followed by evaporation of half the reaction volume at 70°C. Upon cooling and seeding the product crystallised. The product was isolated by filtration and dried in vacuum to yield 19.3 g Z- Tyr-(D)-Ala-OH (25%).
Example 4
A 250 mL flask was charged with H-D-Alanine-OH (4,52 g; 50,2 mmol; 2 eq), potassium carbonate anhydrate (7,05 g; 50,2 mmol; 2 eq) and polyethylene glycol 200 (50 mL; 11,1 mL/g Alanine), which was stirred at ambient temperature. Another 250 mL flask was charged with Z-Tyrosine (8,00 g; 25,1 mmol) and EtOAc (100 mL; 12,6 mL/g Z- Tyrosine). After cooling (jacket temperature -17°C) to -9°C Isobutylchloroformate (3,55 mL: 30,1 mmol; 1,2 eq) was charged. N-Methylmorpholine (3,35 mL; 30,1 mmol; 1 ,2 eq) was charged keeping the temperature below -7°C. The cool mixture was charged to the first 250 mL flask containing the alanine via a peristaltic pump. The conversion was 90% by HPLC. The product was not isolated, but ready to use in the next step.
Step 1'
Preparation of Z-Phe(pF)-Phe-ΝH2 (Compound 8 in Scheme 1)
Example 5
Z-Phe(pF)-OH ( 1 eq.) is first dissolved in acetonitrile (MeCN)( 1.7L/mole) and cooled before addition of z'-Butylchloroformiate ( 1.05 eq). The reaction is then controlled by the rate of addition, (about 20 minutes) 15 min actual, of N-Methylmorpholine ( 1.4eq). A reaction temperature between 0 and -15°C is recommended where the reaction occurs immediately upon addition of N-Methylmorphohne, yet prevents the mixed anhydride from decomposing to rapidly
H-Phe-ΝH2 x HCl ( 1 04 eq) is meanwhile dissolved in DMF (4 OL/mole), neutralized with s N-Methylmorpholine (1 04eq) and cooled to about -10°C This slurry is upon completion of the activation added at a rate that maintains the temperature around -10°C for about 15 minutes
After completion of the coupling the product was crystallized from the reaction mixture by slow addition of 50% Ethanol/water (3 6L/mole) After 30 min a total of 2 85L/mole water lo in three portions were charged with about 25 min wait between each addition and at temperature of about 20°C The crystals can after about 17 hours be filtered or centπfuged and washed with 50% Ethanol/water followed by several portions of acetonitrile before drying under vacuum at 20-60°C Yield 90% and 99 9% purity
15 Step 2'
Preparation of H-Phe(pF)-Phe-ΝH2 (compound 9 in Scheme 1)
Example 6
Z-Phe(pF)-Phe-NH prepared in the previous step is mixed with DMF (3 5L/mole) and a 0 Pd/C catalyst (5% Pd) is added 5%, by weight and the resulting mixture hydrogenated for more than 0 5 hours at 25-30°C and about 3bar H2 The reaction mixture is then filtered and cooled to about -15°C before the next step 99 6% purity in solution and >99% conversion of starting material
5 Step 3
Preparation of Z-Tyr-D-Ala- PhefoFVPhe-NH? (compound 10 in Scheme 1)
Example 7
The two building blocks Z-Tyr-D-Ala-OH ( 1 29mmol, 1 00 eq) and H-Phe( F)-Phe-NH:x o HCl (1 29mmol, 1 OOeq) were added to a round bottom flask containing solvent (DMF, 5mL) and the coupling reagent TBTU (1.28mmol, 0.99eq). The base (DIEA, 2.58mmol, 2. OOeq) was charged to this slurry. The reaction was monitored by HPLC and was terminated after 4h. The solvent was then evaporated. 22mL of a solvent mixture (EtOAc/Acetone MeOH 5: 10: 1) was charged to 2.4g of the remaining oil to yield the product as a solid, which was filtered and washed with acetone (3x5mL). The moist crystals were dried under reduced pressure at 30°C over night. The overall yield was 0.74g (80%) corrected for the purity 97% (HPLC).
Example 8 Z-Tyr-(D)-Ala-OH (15.0 g, 39 mmol) and H-Phe(pF)-Phe-NH2xHCl (12.8 g, 35 mmol) were mixed with aceton (450 ml) in a one liter reactor. The slurry temperature was reduced to -10°C prior to addition of isobutylchloroformate (4.55 ml, 35 mmol). NMM (8.45 ml, 77 mmol) was slowly charged to maintain the temp at about -10°C. Upon completed addition the temp was increased to room temperature and 2M HCl (40 ml, 80 mmol) added followed by water (365 ml). The mixture was heated to reflux, ca 70°C, and acetone removed. The precipitate was filtered off and washed with acetonitril/water 3:2 (250 ml x 2) and dried to yield 18.9 g (77 %) of the title compound.
Step 4 Preparation of H-Tyr-D-Ala-Phe(pF)-Phe-NH2 (compound I in Scheme 1)
Example 9
Compound 4 is mixed with DMF (2-2.6L/mole) and a 5 % Pd/C catalyst is added (6-7%, by weight) and the resulting mixture hydrogenated for more than 0.5 hours at 20-50°C and 3bar H2. The reaction mixture is then filtered to remove the Pd/C before crystallizing the product by addition of EtOAc until all substance has crystallized (typically lOL/mole). The solid is separated by filtration or centrifugation and washed with EtOAc prior to drying under vacuum at 20-60°C. Preparation of H-Tyr-D-Ala-Phe(pF)-Phe-NH? hvdrochloride
Example 10
The free base compound I is dissolved in a mixture of water and acetone with one equivalent HCl added and clear filtered (146g/mole 25% HCl/H2O, 2L Acetone/mole in actual run). The salt has a limited solubility in acetone and therefore the filter is washed once with an additional amount of the acetone/water (95:5) mixture (0.5L/mole). The crystallization is initiated by a slow addition of acetone (3.4L/mole) at high agitation rate and then 1% w/w of seeding crystals is optionally added. After 30 minutes the first amount of MIBK (3L/mole) is slowly charged and left with slow stirring until the batch clearly thickens. MLBK (3L/mole) is charged three additional times separated by 30-60 minutes while maintaining the reactor inner temperature at about 20°C. The solid is then separated by centrifugation or filtration and washed with MLBK before drying under vacuum at 20- 50°C for more than 16 hours or until the solvent levels are lower than specified in the release specifications.

Claims

1. A process for the preparation of the tetrapeptide H-Tyr-D-Ala-Phe(pF)-Phe-NH2 of the formula (I)
Figure imgf000024_0001
or a pharmaceutically acceptable salt thereof, comprising a coupling step wherein an activated dipeptide derivative (V),
Figure imgf000024_0002
wherein
A is an amino protecting group,
R is an activating agent residue group, and
2 R is H or a benzyl-like group is reacted with the amino group of a compound (IX)
Figure imgf000025_0001
in the presence of a solvent under standard conditions, to give a protected tetrapeptide derivative (X)
Figure imgf000025_0002
wherein
A is an amino protecting group,
R is -NH2 or an ester residue group, and
2 R is H or a benzyl-like group which is thereafter deprotected either by catalytic hydrogenolyzis, aqueous base or aqueous acid treatment, under standard conditions, to give the tetrapeptide (I)..
2. A process according to claim 1 , characterized in comprising an additional coupling step wherein an activated tyrosine derivative (II),
Figure imgf000026_0001
wherein
A is an amino protecting group, and R is an activating agent residue group, and
R is H or a benzyl-like group; is reacted with the amino group of D-alanine,
D-Ala-R1 (III), wherein R is an ester group, in the presence of a solvent under standard conditions, to give a protected dipeptide derivative (IV)
Figure imgf000026_0002
wherein
A is an amino protecting group,
R is an ester residue group, and
2 R is H or a benzyl-like group; which is thereafter a) deprotected by aqueous base treatment, under standard conditions, and b) treated with an activating agent to give an activated dipeptide derivative (V).
3. A process according to claim 1, characterized in comprising an additional coupling step wherein an activated tyrosine derivative (II),
Figure imgf000027_0001
wherein
A is an amino protecting group, and o R is an activating agent residue group, and
2 R is H or a benzyl-like group; is reacted with the amino group of D-alanine,
D-Ala-R1 (III), wherein R .1 i .s OH, 5 in the presence of a solvent under standard conditions, to give dipeptide derivative (V),
Figure imgf000027_0002
o wherein A is an amino protecting group, and
2 R is H or a benzyl-like group; which is thereafter treated with an activating agent to give an activated dipeptide derivative
(V).
4. A process according to claim 1 , characterized in comprising an additional coupling step wherein an activated -fluorophenylalanine derivative (VI),
Figure imgf000028_0001
wherein
A is an amino protecting group, and
R is an activating agent residue group; is reacted with the amino group of phenylalanine
Phe-R1 (VII) wherein R is -NH2 or an ester residue group, in the presence of a solvent under standard conditions, providing a protected dipeptide derivative (VIII)
Figure imgf000029_0001
wherein
A is an amino protecting group, and
R is -NH2 or an ester residue group; which is thereafter deprotected, under standard conditions, to give (IX).
5. A process according to claim 1, characterized in that the activated amino acid derivative used in at least one of the coupling steps is selected from a group consisting of a carbodiimide, an activated ester, an azide, or an anhydride.
6. A process according to claim 1, characterized in that benzyloxycarbonyl is the amino protecting group, methyl ester is the carboxyl protecting group, -NH2 is the phenylalanine carboxyl protecting group, the activated amino acid derivative is prepared from isobutylchloroformiate or 2-(lH-Benzotriazole-l-yl)-l,l,3,3-tetramethyluronium tetrafluoroborate and no tyrosine side-chain protecting group is used.
7. A process according to claim 1, characterized in that the solvent used in at least one of the coupling steps is acetone, acetonitrile, NMP, DMF, EtOAc or a mixture thereof.
8. A process according to claims 1, wherein the solvent used in at least one of the coupling steps is DMF.
9. A process according to claim 1 characterized in that the deprotection step of the amino protecting group is performed using palladium on charcoal.
10. A process according to claim 1, characterized in that at least one of the coupling reactions are performed at a temperature from 0°C to -20 °C.
1 1. A process according to claim 10, wherein the temperature is from -5 °C to -15 °C.
12. A peptide of the formula (I)
Figure imgf000030_0001
or a pharmaceutically acceptable salt thereof, prepared according to the process of claim 1.
13. A peptide according to claim 12, in the form of a hydrochloride salt.
14. A protected peptide derivative of the formula (X')
Figure imgf000030_0002
wherein
A is tert-butoxycarbonyl (Boc), 9-fluorenylmethoxycarbonyl (Fmoc), benzyloxycarbonyl
(Z) or a substituted benzyloxycarbonyl derivative;
R is -NH2, Ci-Cό alkyl ester, benzyl ester, OH, 9-fluorenylmetyl ester or a substituted benzyl ester derivative:
2 R is H, a benzyl-like group, tert. butyl group, or 9-flourenylmethyl group,
PCT/SE2000/001747 1999-09-15 2000-09-07 A process for the preparation of h-tyr-d-ala-phe(f)-phe-nh¿2? WO2001019849A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA002383184A CA2383184A1 (en) 1999-09-15 2000-09-07 A process for the preparation of h-tyr-d-ala-phe(f)-phe-nh2
EP00963222A EP1212350A1 (en) 1999-09-15 2000-09-07 A process for the preparation of h-tyr-d-ala-phe(f)-phe-nh 2?
JP2001523626A JP2003509437A (en) 1999-09-15 2000-09-07 Method for producing H-TYR-D-ALA-PHE (F) -PHE-NH2
AU74669/00A AU7466900A (en) 1999-09-15 2000-09-07 A process for the preparation of h-tyr-d-ala-phe(f)-phe-nh2

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
SE9903291-4 1999-09-15
SE9903291A SE9903291D0 (en) 1999-09-15 1999-09-15 New process

Publications (1)

Publication Number Publication Date
WO2001019849A1 true WO2001019849A1 (en) 2001-03-22

Family

ID=20416994

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SE2000/001747 WO2001019849A1 (en) 1999-09-15 2000-09-07 A process for the preparation of h-tyr-d-ala-phe(f)-phe-nh¿2?

Country Status (6)

Country Link
EP (1) EP1212350A1 (en)
JP (1) JP2003509437A (en)
AU (1) AU7466900A (en)
CA (1) CA2383184A1 (en)
SE (1) SE9903291D0 (en)
WO (1) WO2001019849A1 (en)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2088154A1 (en) 2004-03-09 2009-08-12 Ironwood Pharmaceuticals, Inc. Methods and compositions for the treatment of gastrointestinal disorders
WO2010065751A2 (en) 2008-12-03 2010-06-10 Synergy Pharmaceuticals, Inc. Formulations of guanylate cyclase c agonists and methods of use
EP2246360A1 (en) 2003-01-28 2010-11-03 Ironwood Pharmaceuticals, Inc. Compositions for the treatment of gastrointestinal disorders
CN101970453A (en) * 2008-03-10 2011-02-09 索尔维公司 Peptide synthesis method using n-carboxyanhydride (unca)
WO2011069038A2 (en) 2009-12-03 2011-06-09 Synergy Pharmaceuticals, Inc. Agonists of guanylate cyclase useful for the treatment of hypercholesterolemia, atherosclerosis, coronary heart disease, gallstone, obesity and other cardiovascular diseases
WO2012118972A2 (en) 2011-03-01 2012-09-07 Synegy Pharmaceuticals Inc. Process of preparing guanylate cyclase c agonists
WO2013138352A1 (en) 2012-03-15 2013-09-19 Synergy Pharmaceuticals Inc. Formulations of guanylate cyclase c agonists and methods of use
WO2014029983A1 (en) 2012-08-21 2014-02-27 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2014151206A1 (en) 2013-03-15 2014-09-25 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase and their uses
WO2014151200A2 (en) 2013-03-15 2014-09-25 Synergy Pharmaceuticals Inc. Compositions useful for the treatment of gastrointestinal disorders
EP2810951A2 (en) 2008-06-04 2014-12-10 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal disorders, inflammation, cancer and other disorders
WO2014197720A2 (en) 2013-06-05 2014-12-11 Synergy Pharmaceuticals, Inc. Ultra-pure agonists of guanylate cyclase c, method of making and using same
EP2998314A1 (en) 2007-06-04 2016-03-23 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal disorders, inflammation, cancer and other disorders
EP3241839A1 (en) 2008-07-16 2017-11-08 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal, inflammation, cancer and other disorders
WO2018129552A1 (en) 2017-01-09 2018-07-12 Ardelyx, Inc. Compounds useful for treating gastrointestinal tract disorders
WO2018129556A1 (en) 2017-01-09 2018-07-12 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2018129557A1 (en) 2017-01-09 2018-07-12 Ardelyx, Inc. Inhibitors of nhe-mediated antiport
EP3351248A1 (en) 2008-12-31 2018-07-25 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US10272079B2 (en) 2013-04-12 2019-04-30 Ardelyx, Inc. NHE3-binding compounds and methods for inhibiting phosphate transport
US10376481B2 (en) 2012-08-21 2019-08-13 Ardelyx, Inc. Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4540568B2 (en) * 2005-07-26 2010-09-08 株式会社トクヤマ Method for producing L-carnosine

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4797469A (en) * 1984-07-10 1989-01-10 Sanofi Synthesis of hGRF (Somatocrinin) in liquid phase and intermediate peptides
WO1997007129A1 (en) * 1995-08-18 1997-02-27 Biochem Pharma Inc. Solution synthesis of peripheral acting analgesic opioid tetrapeptides
WO1999047548A1 (en) * 1998-03-16 1999-09-23 Astrazeneca Ab Process for the preparation of a tetrapeptide

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4797469A (en) * 1984-07-10 1989-01-10 Sanofi Synthesis of hGRF (Somatocrinin) in liquid phase and intermediate peptides
WO1997007129A1 (en) * 1995-08-18 1997-02-27 Biochem Pharma Inc. Solution synthesis of peripheral acting analgesic opioid tetrapeptides
WO1999047548A1 (en) * 1998-03-16 1999-09-23 Astrazeneca Ab Process for the preparation of a tetrapeptide

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2246360A1 (en) 2003-01-28 2010-11-03 Ironwood Pharmaceuticals, Inc. Compositions for the treatment of gastrointestinal disorders
EP2088154A1 (en) 2004-03-09 2009-08-12 Ironwood Pharmaceuticals, Inc. Methods and compositions for the treatment of gastrointestinal disorders
EP2998314A1 (en) 2007-06-04 2016-03-23 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal disorders, inflammation, cancer and other disorders
CN101970453A (en) * 2008-03-10 2011-02-09 索尔维公司 Peptide synthesis method using n-carboxyanhydride (unca)
EP2810951A2 (en) 2008-06-04 2014-12-10 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal disorders, inflammation, cancer and other disorders
EP3241839A1 (en) 2008-07-16 2017-11-08 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal, inflammation, cancer and other disorders
WO2010065751A2 (en) 2008-12-03 2010-06-10 Synergy Pharmaceuticals, Inc. Formulations of guanylate cyclase c agonists and methods of use
EP3939964A1 (en) 2008-12-31 2022-01-19 Ardelyx, Inc. Combinations for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
EP3351248A1 (en) 2008-12-31 2018-07-25 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
EP2923706A1 (en) 2009-12-03 2015-09-30 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of hypercholesterolemia
WO2011069038A2 (en) 2009-12-03 2011-06-09 Synergy Pharmaceuticals, Inc. Agonists of guanylate cyclase useful for the treatment of hypercholesterolemia, atherosclerosis, coronary heart disease, gallstone, obesity and other cardiovascular diseases
WO2012118972A2 (en) 2011-03-01 2012-09-07 Synegy Pharmaceuticals Inc. Process of preparing guanylate cyclase c agonists
WO2013138352A1 (en) 2012-03-15 2013-09-19 Synergy Pharmaceuticals Inc. Formulations of guanylate cyclase c agonists and methods of use
EP4309673A2 (en) 2012-03-15 2024-01-24 Bausch Health Ireland Limited Formulations of guanylate cyclase c agonists and methods of use
EP3708179A1 (en) 2012-03-15 2020-09-16 Bausch Health Ireland Limited Formulations of guanylate cyclase c agonists and methods of use
US10376481B2 (en) 2012-08-21 2019-08-13 Ardelyx, Inc. Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2014029983A1 (en) 2012-08-21 2014-02-27 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2014151200A2 (en) 2013-03-15 2014-09-25 Synergy Pharmaceuticals Inc. Compositions useful for the treatment of gastrointestinal disorders
WO2014151206A1 (en) 2013-03-15 2014-09-25 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase and their uses
US10940146B2 (en) 2013-04-12 2021-03-09 Ardelyx, Inc. NHE3-binding compounds and methods for inhibiting phosphate transport
US10272079B2 (en) 2013-04-12 2019-04-30 Ardelyx, Inc. NHE3-binding compounds and methods for inhibiting phosphate transport
WO2014197720A2 (en) 2013-06-05 2014-12-11 Synergy Pharmaceuticals, Inc. Ultra-pure agonists of guanylate cyclase c, method of making and using same
WO2018129557A1 (en) 2017-01-09 2018-07-12 Ardelyx, Inc. Inhibitors of nhe-mediated antiport
WO2018129556A1 (en) 2017-01-09 2018-07-12 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US11147884B2 (en) 2017-01-09 2021-10-19 Ardelyx, Inc. Inhibitors of NHE-mediated antiport
US11242337B2 (en) 2017-01-09 2022-02-08 Ardelyx, Inc. Compounds useful for treating gastrointestinal tract disorders
WO2018129552A1 (en) 2017-01-09 2018-07-12 Ardelyx, Inc. Compounds useful for treating gastrointestinal tract disorders

Also Published As

Publication number Publication date
AU7466900A (en) 2001-04-17
JP2003509437A (en) 2003-03-11
EP1212350A1 (en) 2002-06-12
SE9903291D0 (en) 1999-09-15
CA2383184A1 (en) 2001-03-22

Similar Documents

Publication Publication Date Title
WO2001019849A1 (en) A process for the preparation of h-tyr-d-ala-phe(f)-phe-nh¿2?
Brady et al. Large-scale synthesis of a cyclic hexapeptide analog of somatostatin
RU2602042C2 (en) Method for production of degarelix and intermediate compounds thereof
US6235876B1 (en) Liquid phase process for the preparation of GNRH peptides
GB2058079A (en) TRH analogues
US6211333B1 (en) Method for producing peptides and their salts which have an agonist activity of luteinizing hormone releasing hormones secreted from the hypothalamus
HU182866B (en) Process for preparing new tetrapeptide derivatives
Katakai Peptide synthesis using o-nitrophenylsulfenyl N-carboxy. alpha.-amino acid anhydrides
WO2010040660A1 (en) Pseudoproline dipeptides
CA2315222A1 (en) Branched building units for synthesizing cyclic peptides
US3592836A (en) Aryloxycarbonyl fluorides and amino acids and their production
BE1005720A3 (en) Process for peptide synthesis and new intermediate synthesis.
HU208838B (en) Method for producing peptones containing aza aminoacides by means of solid-phase synthesis
AU2968899A (en) Process for the preparation of a tetrapeptide
EP0410182A2 (en) A new technique for rapid peptide coupling
Gros et al. Stereochemical control in the preparation of α-amino N-methylthiazolidine masked aldehydes used for peptide aldehydes synthesis
US3780015A (en) Process for preparing lysine containing peptides
JP3759821B2 (en) Peptide production method
JP2748897B2 (en) Novel arginine derivative and method for producing peptide using the same
RU2111972C1 (en) METHOD OF SYNTHESIS OF δ-SLEEP PEPTIDE
JP4387352B2 (en) Peptide production method
MXPA00008828A (en) Process for the preparation of a tetrapeptide
EP0564587A1 (en) Solution phase process for synthesis of peptide
Sureshbabu et al. HOAt. DCHA as co-coupling agent in the synthesis of peptides employing Fmoc-amino acid chlorides as coupling agents: Application to the synthesis of β-casomorphin
PL194997B1 (en) Method of obtaining l-pyroglutamyl-l-istidyl-l-triptophyl-l-seril-l-thyrosil-d-(o-t-butyl)-seril-l-leucil-larginyl-l-prolyl-azaglicin acetate

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 09701488

Country of ref document: US

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2000963222

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2383184

Country of ref document: CA

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2001 523626

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 2000963222

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 2000963222

Country of ref document: EP