Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
DERIVATIVES OF DOLASTATIN 10 AND AURISTATINS
Document Type and Number:
WIPO Patent Application WO/2014/174064
Kind Code:
A1
Abstract:
The present invention concerns a compound of following formula (I) where: - R1 is H or OH, - R2 is a (C1-C6)alkyl, COOH, COO-((C1-C6)alkyl) or thiazolyl group, - R3 is H or a (C1-C6)alkyl group, and - R4 is an aryl-(C1-C8)alkyl group substituted by one or more groups chosen from among OH and NR9R10 groups, or a pharmaceutically acceptable salt, hydrate or solvate thereof, and its uses in particular for the treatment of cancer, pharmaceutical compositions containing the same and the preparation methods thereof.

Inventors:
PEREZ MICHEL (FR)
RILATT IAN (FR)
LAMOTHE MARIE (FR)
Application Number:
PCT/EP2014/058427
Publication Date:
October 30, 2014
Filing Date:
April 25, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PF MEDICAMENT (FR)
International Classes:
C07D401/12; A61K31/401; A61K31/4025; A61K31/427; A61K31/4439; A61K33/243; A61K38/07; A61K38/08; A61K38/40; A61P35/00; C07D207/08; C07D417/12; C07D417/14; C07K5/02
Domestic Patent References:
WO2012059882A22012-05-10
WO2012041805A12012-04-05
WO2012123423A12012-09-20
WO2012166559A12012-12-06
WO2011154359A12011-12-15
Foreign References:
EP0731106A11996-09-11
Other References:
PETTIT G. R., J. AM. CHEM. SOC., vol. 109, 1987, pages 6883
PETTIT G. R., J. AM. CHEM. SOC., vol. 109, 1987, pages 7581
PETTIT, G. R., HETEROCYCLES, vol. 28, 1989, pages 553
PETTIT, G. R., J. AM. CHEM. SOC., vol. 111, 1989, pages 5015
PETTIT G. R., J. AM. CHEM. SOC., vol. 113, 1991, pages 6692
PETTIT G. R., J. MED. CHEM., vol. 33, 1990, pages 3133
MIYAZAKI K., PEPTIDE CHEMISTRY, vol. 31, 1993, pages 85
MIYAZAKI K, CHEM. PHAM. BULL., vol. 43, 1995, pages 1706
PETTIT G. R, ANTICANCER DRUG DESIGN, vol. 13, 1998, pages 243
PETTIT G. R., ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, 1998, pages 2961
MIYAZAKI K., CHEM. PHAM. BULL., vol. 43, 1995, pages 1706
T.W. GREENE: "Protective Groups in Organic Synthesis", 2006, JOHN WILEY & SONS
P.J. KOCIENSKI: "Protecting Groups", 1994, THIEME VERLAG
GREENE: "Protective Groups In Organic synthesis", 1981, JOHN WILEY & SONS
HARRISON ET AL.: "Compendium of Synthetic Organic Methods", vol. 1-8, 1971, J. WILEY & SONS
Attorney, Agent or Firm:
REGIMBEAU (Paris Cedex 17, Paris Cedex 17, FR)
Download PDF:
Claims:
CLAIMS

1. A compound of following formula (I):

(I) where:

- Ri is H or OH,

- R2 is a (Ci-C6)alkyl, COOH, COO-((Ci-C6)alkyl) or thiazolyl group,

- R3 is H or a (Ci-C6)alkyl group, and

- R4 is an aryl-(Ci-C8)alkyl group substituted by one or more groups chosen from among OH and NR9R10 groups with R9 and Rio each independently of one another representing H or a (Ci-C6)alkyl group,

or a pharmaceutically acceptable salt, hydrate or solvate thereof.

2. The compound according to claim 1, characterized in that:

- Ri=OH and R2 represents a (Ci-C6)alkyl group, or

- Ri=H and R2 represents a COOH, COO-(Ci-Ce)alkyl or thiazole group.

3. The compound according to claim 1 or 2, characterized in that Ri represents H and R2 represents COOH or COOMe.

4. The compound according to any one of claims 1 to 3, characterized in that R3 represents H or a methyl group.

5. The compound according to any one of claims 1 to 4, characterized in that R4 represents an aryl-(Ci-C4)alkyl group substituted by one group on the aryl moiety chosen from among OH and NR9R10, and notably being NR9R10.

6. The compound according to any one of claims 1 to 4, characterized in that R4 represents a phenyl-(Ci-C2)alkyl group substituted by one group on the phenyl moiety chosen from among OH and NR9R10, and notably being NR9R10.

7. The compound according to any one of claims 1 to 4, characterized in llowing formula:

wherein X0 represents OH or NR9R10, in particular NR9R10, and m represents an integer comprised between 1 and 8, advantageously m is 1 or 2.

8. The compound according to any one of claims 1 to 4, characterized in that R4 has the following formula:

wherein X0 represents NR9R10 and m represents 1 or 2.

and the pharmaceutically acceptable salts thereof such as the salts formed with trifluoroacetic acid.

10. A compound according to any one of claims 1 to 9 for use as a medicinal product. 11. A compound according to any one of claims 1 to 9 for use as medicinal product intended for the treatment of cancer or benign proliferative disorders.

12. A pharmaceutical composition comprising a formula (I) compound according to any one of claims 1 to 9 and at least one pharmaceutically acceptable excipient.

13. The pharmaceutical composition according to claim 12, further comprising another active ingredient, advantageously chosen from among anticancer agents, in particular comprising cytotoxic anticancer agents such as navelbine, vinflunine, taxol, taxoter, 5-fluorouracil, methotrexate, doxorabicin, camptothecin, gemcitabin, etoposide, cis-platin or carmustin; and hormonal anticancer agents such as tamoxifen or medroxyprogesterone.

14. A method for preparing a formula (I) compound according to any one of claims 1 to 9 comprising a condensation reaction between a compound of following formula (VI):

where Ri and R2 are as defined in claim 1 ,

a compound of following formul

where R3 is as defined in claim 1 , R4a represents an R4 group as defined in claim 1 , optionally in protected form, and X is OH or CI.

15. A method for preparing a formula (I) compound according to any one of claims 1 to 9 comprising a substitution reaction between a compound of following formula (VIII):

where Ri, R2 a,d R3 are as defined in claim 1, and

a compound of following formula (X):

where R4a represents an R4 group as defined in claim 1 optionally in protected form, Y is a leaving group such as CI, Br, I, OS02CH3, OS02CF3 or O-Tosyl.

16. A method for preparing a formula (I) compound according to any one of claims 1 to 9 where R4 represents a -CH2R4b group with R4b representing an aryl or aryl-(Ci-Cv)alkyl group substituted by one or more groups chosen from among OH and NR9R10 groups,

comprising a reductive amination reaction between a compound of following formula (VIII):

where Ri, R2 and R3 are as defined in claim 1, and

a compound of following formula (XI):

where R4b is as previously defined.

Description:
Derivatives of dolastatin 10 and auristatins

The subject of the present invention concerns novel derivatives of dolastatin 10 and auristatins, their methods of production, pharmaceutical compositions containing the same and the use thereof as medicinal product in particular in the treatment of cancer.

Dolastatin 10 (D10) is a cytotoxic peptide derivative isolated from a marine mollusc {Dolabella auricularia) whose absolute configuration was determined and later confirmed after total synthesis of the product (Pettit G. R. J. Am. Chem. Soc. 1987, 109, 6883; Pettit G. R. J. Am. Chem. Soc. 1987, 109, 7581; Pettit, G. R Heterocycles 1989, 28, 553; Pettit, G. R. J. Am. Chem. Soc. 1989,111, 5015; Pettit G. R. J. Am. Chem. Soc. 1991, 113, 6692). D10 is formed of 5 units called dolavaline (Dov), valine (Val), do laiso leucine (Dil), dolaproine (Dap) and dolaphenine (Doe). A certain number of analogues of this compound have been synthesised by modifying the nature of its component amino acids (Pettit G. R. J. Med. Chem. 1990, 33, 3133; Miyazaki K. Peptide Chemistry 1993, 31, 85; Miyazaki K. Chem. Pham. Bull. 1995, 43, 1706). Modifications of the C-terminal part (right end) have also been performed and have led to numerous derivatives which include auristatin E or F (Pettit G. R. Anticancer Drug

Design, 1998, 13, 243; Pettit G. R. Antimicrobial Agents And Chemotherapy, 1998, 2961).

Dolastatin 10

Auristatin F

The present invention has focused on modification of the N-terminal part (left end) of derivatives of dolastatin 10 and auristatins E and F. The few examples published in the literature on modifications made at this position have led to losses of activity (Miyazaki K. Chem. Pham. Bull. 1995, 43, 1706). The compounds described in the present invention differ from the prior art through their original chemical structures and also through their remarkable biological property that is fully unexpected having regard to the elements published in the literature. These remarkable activities result in making these compounds suitable for use in the treatment of cancer.

In addition, these compounds have the advantage of being both active as cytotoxic agents and more soluble than the parent compounds.

The subject of the present invention is thus a compound of following formula

(I):

where:

- Ri is H or OH,

- R 2 is a group: (Ci-C 6 )alkyl (e.g. methyl), COOH, COO-((Ci-C 6 )alkyl) (such as COOMe) or thiazolyl (such as thiazol-2-yl),

- ¾ is H or a (Ci-C 6 )alkyl group (such as methyl), in particular a (Ci-C6)alkyl group, and

- R 4 is an aryl-(Ci-Cs)alkyl group substituted by one or more groups (in particular one, preferably on the aryl part) chosen from among OH and NR 9 R 10 groups with R 9 and Rio each independently of one another representing H or a (Ci-Ce)alkyl group (such as methyl),

or a pharmaceutically acceptable salt, hydrate or solvate thereof.

The radicals R 2 to R 4 , and in particular R 4 , may be chiral groups and may be in the form of their different stereoisomers and optionally in the form of a mixture of stereoisomers.

By « stereoisomer », in the meaning of the present invention is meant a geometric isomer or an optical isomer.

Geometrical isomers result from the different position of the substituents on a double bond which may therefore have a Z or E configuration.

Optical isomers result in particular from the different position in space of the substituents on a carbon atom comprising 4 different substituents. This carbon atom then forms a chiral or asymmetric centre. Optical isomers comprise diastereoisomers and enantiomers. Optical isomers which are images of one another in a mirror but which cannot be superimposed are called « enantiomers ». Optical isomers which are not superimposable images of one another in a mirror are called « diastereoisomers ».

A mixture containing equal quantities of two individual enantiomer forms of opposite chirality is called a « racemic mixture ».

In the present invention by « pharmaceutically acceptable » is meant that which can be used in the preparation of a pharmaceutical composition which is generally, safe non-toxic and neither biologically nor otherwise undesirable, and which is acceptable for veterinary use as well as for human pharmaceutical use.

By « pharmaceutically acceptable salt, hydrate or solvate » of a compound is meant a salt, hydrate or solvate which is pharmaceutically acceptable as defined herein and which has the desired pharmacological activity of the parent compound.

Pharmaceutically acceptable salts notably comprise:

(1) the addition salts of a pharmaceutically acceptable acid formed with pharmaceutically acceptable inorganic acids such as hydrochloric, hydrobromic, phosphoric, sulfuric and similar acids; or formed with pharmaceutically acceptable organic acids such as acetic, trifluoroacetic, propionic, succinic, fumaric, malic, tartaric, citric, ascorbic, maleic, glutamic, benzoic, salicylic, toluenesulfonic, methanesulfonic, stearic, lactic and similar acids; and

(2) the addition salts of a pharmaceutically acceptable base formed when an acid proton present in the parent compound is either replaced by a metallic ion e.g. an alkaline metal ion, an alkaline-earth metal ion or an aluminium ion; or coordinated with a pharmaceutically acceptable organic base such as lysine, arginine and similar; or with a pharmaceutically acceptable inorganic base such as sodium hydroxide, potash, calcium hydroxide and similar.

These salts can be prepared from the compounds of the invention containing a base or acid function, and the corresponding acids or bases using conventional chemical methods.

The formula (I) compounds of the invention are preferably in salt form, and in particular a pharmaceutically acceptable acid addition salt.

Preferably, the compounds of formula (I) according to the present invention are in the form of a pharmaceutically acceptable acid addition salt, the acid possibly being trifluoroacetic acid, acetic acid or hydrochloric acid for example, and in particular trifluoroacetic acid.

The solvates comprise the conventional solvates obtained at the last preparation step of the compounds of the invention due to the presence of solvent, the solvent possibly being ethanol for example.

By « alkyl » in the present invention is meant a straight-chain or branched, saturated hydrocarbon chain. For example, mention can be made of methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl or hexyl groups.

By « (C x -C y )alkyl » in the meaning of the present invention is meant an alkyl chain such as defined above comprising x to y carbon atoms. Therefore, a (Ci-C 6 )alkyl group is an alkyl chain having 1 to 6 carbon atoms.

By « aryl » in the meaning of present invention is meant an aromatic hydrocarbon group preferably having 6 to 10 carbon atoms and able to comprise one or two fused rings. For example a phenyl or a naphthyl can be cited. Advantageously it is a phenyl.

By « aryl-(Ci-C8)alkyl » in the meaning of the present invention is meant an aryl group such as defined above linked to the remainder of the molecule via an alkyl group such as defined above and comprising 1 to 8, in particular 1 to 6, advantageously 1 to 4, preferably 1 or 2 carbon atoms. The aryl moiety is preferably a phenyl moiety. The (Ci- Cs)alkyl moiety is advantageously a (Ci-C4)alkyl, peferably a (Ci-C 2 )alkyl. In particular, the aryl-(Ci-C8)alkyl group is a benzyl or phenethyl group.

Among the compounds of the invention, one particularly appreciated class of compounds corresponds to the formula (I) compounds in which Ri is OH and R 2 represents a (Ci-C 6 )alkyl group, such as methyl.

Another particularly appreciated class of compounds corresponds to the formula (I) compounds in which Ri is a hydrogen and R 2 is a thiazole (in particular a thiazol-2- yl group).

Another class of particularly appreciated compounds corresponds to the formula (I) compounds in which Ri is a hydrogen and R 2 is a COO(Ci-Ce)alkyl group such as COOMe. Another class of particularly appreciated compounds corresponds to the formula (I) compounds in which Ri is a hydrogen and R 2 is a COOH group.

Therefore the compounds of the invention are advantageously formula (I) compounds in which:

- Ri=OH and R 2 =Me (methyl), or

- Ri=H and R 2 =COOH, COOMe or thiazol-2-yl.

According to one particular embodiment of the present invention, R 2 is more particularly a methyl, COOH, COOMe or thiazol-2-yl group.

Preferably, Ri is H and R 2 is COOH or COO(Ci-C 6 )alkyl, notably COOH or COOMe.

According to a first preferred embodiment, Ri is H and R 2 is COOH.

According to a second preferred embodiment, Ri is H and R 2 is COOMe.

R 3 particularly represents H or a methyl group, advantageously a methyl group.

R4 represents advantageously an aryl-(Ci-Cs)alkyl group, notably an aryl-(Ci- C4)alkyl group, such as an aryl-(Ci-C 2 )alkyl group, substituted by one group chosen from among OH and NR9R10, and notably being NR9R10.

R4 represents advantageously an aryl-(Ci-Cs)alkyl group, notably an aryl-(Ci- C4)alkyl group, such as an aryl-(Ci-C 2 )alkyl group, substituted by one group on the aryl moiety chosen from among OH and NR9R10, and notably being NR9R10.

The aryl group is advantageously a phenyl group.

Thus R4 can represent in particular a phenyl-(Ci-C 2 )alkyl substituted by one group (preferably on the phenyl moiety) chosen from among OH and NR9R10, and notably being NR9R10.

s have the following formula:

wherein X 0 represents OH or NR9R10, in particular NR9R10, and m represents an integer comprised between 1 and 8, notably between 1 and 4, and advantageously is 1 or 2.

According to a preferred embodiment, R4 has the following formula:

with Xo and m as defined previously, and in particular with and m=l or 2.

R4 may in particular be chosen from among:

Advantageously, the formula (I) compound is chosen from among the compounds 1 1-15, 19-20, 23-24, 27-29, 49-51 and 61-64 described in the examples below.

A further subject of the present invention is a formal (I) compound such as defined above for use as medicinal product, in particular for the treatment or prevention of cancer or benign proliferative disorders.

The present invention also concerns the use of a formula (I) compound such as defined above for producing a medicinal product, particularly intended for the treatment or prevention of cancer or benign proliferative disorders.

The present invention also concerns a method for treating or preventing cancer or benign proliferative disorders comprising the administration to a person in need thereof of an effective amount of a formula (I) compound such as defined above.

The cancer to be treated or prevented is more particularly cancer of the lung, pancreas, skin, head, neck, uterus, ovaries, anus, stomach, colon, breast, oesophagus, small intestine, thyroid gland, lymphatic system, prostate, kidney, or bladder, or an acute or chronic leukaemia, or a combination of two or more of these cancers.

By benign proliferative disorders is meant proliferating disorders which cannot give rise to metastases or which have not yet progressed towards a cancer (precancerous tumours). A further subject of the present invention is a pharmaceutical composition comprising a formula (I) compound such as defined above and at least one pharmaceutically acceptable excipient.

The active ingredient can be administered in unit forms of administration, in a mixture with conventional pharmaceutical carriers, to animals or to human beings. Suitable unit forms of administration comprise forms via oral route, forms for sublingual or buccal administration, forms for administration via parenteral route (subcutaneous, intradermal, intramuscular or intravenous), forms for topical administration (on the skin and mucosa, including intranasal and intraocular administration) and forms for rectal administration.

Such compositions may be in the form of a solid, liquid, emulsion, lotion or cream.

As solid compositions, for oral administration, use can be made of tablets, pills, powders (hard or soft gelatine capsules) or granules. In these compositions, the active ingredient of the invention is mixed with one or more inert diluents such as starch, cellulose, sucrose, lactose or silica, in a stream of argon. These compositions may also comprise substances other than diluents, for example one or more lubricants such as magnesium stearate or talc, a colouring agent, a coating (coated tablets) or a varnish.

As liquid compositions for oral administration, use can be made of solutions, suspensions, emulsions, syrups and elixirs that are pharmaceutically acceptable and contain inert diluents such as water, ethanol, glycerol, vegetable oils or paraffin oil. These compositions may comprise substances other than diluents; for example wetting, sweetening, thickening, flavouring or stabilising products.

The sterile compositions for parenteral administration may preferably be aqueous or non-aqueous solutions, suspensions or emulsions. As solvent or vehicle, use can be made of water, propyleneglycol, a polyethyleneglycol, vegetable oils, in particular olive oil, injectable organic esters e.g. ethyl oleate or other suitable organic solvents. These compositions may also contain adjuvants, in particular wetting, isotonic, emulsifying, dispersing and stabilising agents. Sterilisation can be performed in several manners, for example by sanitising filtration, by incorporating sterilising agents into the composition, by radiation or by heating. They can also be prepared in the form of solid sterile compositions which can be dissolved at the time of use in sterile water or any other injectable sterile medium.

The compositions for rectal administration are suppositories or rectal capsules which, in addition to the active ingredient, contain excipients such as cocoa butter, semi-synthetic glycerides or polyethyleneglycols.

The compositions for topical administration may for example be creams, lotions, eye drops, mouthwash, nasal drops or sprays.

The doses are dependent on the desired effect, on the length of treatment and the route of administration used. In general the physician will determine the suitable dosage in relation to the age, weight and all other factors particular to the subject to be treated.

Another active ingredient may be contained in the pharmaceutical compositions according to the present invention. In particular, it may be an anticancer agent, and in particular a cytotoxic anticancer agent such as navelbine, vinflunine, taxol, taxotere, 5- fluorouracil, methotrexate, doxorabicin, camptothecin, gemcitabin, etoposide, cis-platin or carmustine (also called BCNU); or a hormonal anticancer agent such as tamoxifen or medroxyprogesterone.

Radiation treatment (X-ray or gamma ray) may also be associated with the administering of a compound of the present invention. Such radiation can be given using an external source or by implanting minute internal radioactive sources.

The present invention also concerns the preparation of the formula (I) compounds according to the invention using the general methods described in the following synthesis schemes, optionally supplemented by any standard operation when needed that is described in the literature or well known to persons skilled in the art, or described in the examples in the experimental part hereof.

Scheme 1

Scheme 1 illustrates the first general method which can be used to prepare formula (I) compounds. In the above general formulas, R ls R 2> and R 3 are such as previously defined, R4 a represents a R 4 group such as previously defined optionally in protected form and G is a protective group.

The first step consists of the condensing of compound (II), protected on its amine function by a protective group G, with compound (III). X may represent a leaving group such as a chlorine. In this case the first step consists of the reaction between an acid chloride and an amine. This reaction can be conducted using methods and techniques well known to those skilled in the art. In one particularly appreciated method, the two entities are caused to react in the presence of an organic or inorganic base e.g. Et 3 N, iPr 2 NEt, pyridine, NaH, Cs 2 C0 3 , K 2 C0 3 in a solvent such as THF, dichloromethane, DMF, DMSO, at a temperature notably between -20°C and 100°C. X may also be a hydroxyl (OH). In this case, the first step is a condensation reaction between the carboxylic acid (II) and the amine (III). This reaction can be performed following methods and techniques well known to skilled persons. In one particularly appreciated method, these two entities are caused to react in the presence of a coupling agent such as l-(3-dimethylaminopropyl)-3-ethyl-carbodiimide (EDC), 3-hydroxy- l ,2,3-benzotriazin-4(3H)-one, a tertiary amine such as diisopropylethylamine, in a polar aprotic solvent such as dichloromethane or DMF, at a temperature notably between - 15°C and 40°C. In another particularly appreciated method, these two entities are caused to react in the presence of diethyl phosphorocyanidate (DEPC), a tertiary amine such as triethylamine, in a polar aprotic solvent such as dichloromethane or DMF, at a temperature of between -15°C and 40°C. Another particularly appreciated method consists of causing these two entities to react in the presence of 0-(7-azabenzotriazol- l-yl)-l,l,3,3-tetramethyl-uroniumhexafluorophosphate (HATU), a tertiary amine such as diisopropylethylamine, in a polar aprotic solvent such as dichloromethane or DMF, at a temperature of between -15°C and 100°C.

After deprotection of the intermediate using techniques well known to those skilled in the art (« Protective Groups in Organic Synthesis », T.W. Greene, John Wiley & Sons, 2006 and « Protecting Groups », P.J. Kocienski, Thieme Verlag, 1994), compound (IV) can be condensed with compound (V) following the methods and techniques described above to lead to compound (VI) after a deprotection step. This compound can then, after condensation with the intermediate (VII) and optional deprotection, lead to the formation of the formula (I) compounds. Compound (VI) can also be coupled with a compound (VIF) in which R' 3 is a precursor of R 3 , in particular an R 3 group protected by a protective group. Coupling followed by deprotection of group R' 3 to lead to R 3 can be carried out following the same procedures as described previously.

Scheme 2 Scheme 2 illustrates the second general method which can be used to prepare formula (I) compounds. In the above general formulas Ri, R 2 , and R 3 are such as previously defined, R 4a represents an R 4 group such as previously defined optionally in protected form, R 4b is a precursor of an R 4 group and G is a protective group.

At the first step, compound (IX) protected on its amine function by a protective group G is condensed with compound (VI). X may represent a leaving group e.g. a chlorine. In this case, the first step consists of the reaction between an acid chloride and an amine. This reaction can be performed using methods and techniques well known to persons skilled in the art. In one particularly appreciated method the two entities are caused to react in the presence of an organic or inorganic base such as Et 3 N, iPr 2 NEt, pyridine, NaH, Cs 2 C0 3 , K 2 C0 3 in a solvent such as THF, dichloromethane, DMF, DMSO at a temperature notably between -20° and 100°C. X may also represent a hydroxyl. In this case, the first step is a condensation reaction between the carboxylic acid (IX) and the amine (VI). This reaction can be conducted following methods and techniques well known to skilled persons. In one particularly appreciated method, the two entities are caused to react in the presence of l-(3-dimethylaminopropyl)-3-ethyl- carbodiimide (EDC), 3-hydroxy-l ,2,3-benzotriazin-4(3H)-one, a tertiary amine such as diisopropylethylamine, in a polar aprotic solvent such as dichloromethane or DMF, at a temperature notably between -15°C and 40°C. In another particularly appreciated method, these two entities are caused to react in the presence of diethyl phosphorocyanidate (DEPC), a tertiary amine such as triethylamine, in a polar aprotic solvent such as dichloromethane or DMF, at a temperature notably between -15°C and 40°C.

After deprotection of the intermediate, using techniques well known to skilled persons, the obtained compound (VIII) can lead to the formula (I) compounds after reaction with R 4 Y. In this case, Y is a leaving group such as CI, Br, I, OS0 2 CH 3 , OS0 2 CF 3 or O-Tosyl. The reaction is conducted in the presence of an organic or inorganic base such as Et 3 N, iPr 2 NEt, NaH, Cs 2 C0 3 , K 2 C0 3 , in a polar anhydrous solvent such as dichloromethane, THF, DMF, DMSO at a temperature notably between -20° and 100°C. In another particularly appreciated method, compound (VIII) is caused to react with an aldehyde of formula R 4b -CHO where R 4b corresponds to a precursor of R 4 . In this case, the reaction is a reductive amination in the presence of a reducing agent such as NaBH 4 , NaBH 3 CN, NaBH(OAc) 3 , in a polar solvent such as 1,2- dichloroethane, dichloromethane, THF, DMF, MeOH, in the optional presence of titanium isopropoxide (IV), at a pH which can be controlled by the addition of an acid such as acetic acid at a temperature notably between -20°C and 100°C.

In the foregoing synthesis schemes, a formula (I) compound may lead to another formula (I) compound after an additional reaction step such as saponification for example using methods well known to skilled persons whereby an R 2 group representing an ester, preferably a methyl ester, is changed to an R 2 group representing a carboxylic acid.

If it is desired to isolate a formula (I) compound containing at least one base function in the state of an acid addition salt, this is possible by treating the free base of the formula (I) compound (containing at least one base function) with a suitable acid, preferably in equivalent quantity. The suitable acid may in particular be trifluoroacetic acid.

A further subject of the present invention is therefore a first method for preparing a formula (I) compound, comprising a condensation reaction between a compound of following formula (VI):

where Ri and R 2 are such as defined previously, and

a compound of following formul

where R 3 is such as previously defined, R 4a corresponds to a R 4 group such as previously defined optionally in protected form, and X is OH or CI. When X = OH, the coupling reaction can be performed under peptide coupling conditions well known to persons skilled in the art.

Said peptide coupling can be performed in the presence of a coupling agent such as diisopropylcarbodiimide (DIC), dicyclohexylcarbodiimide (DCC), l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC), carbonyldiimidazole (CDI), 2-( 1 H-benzotriazole- 1 -yl)- 1 , 1 ,3 ,3-tetramethyluronium hexafluorophosphate (HBTU), 2-(lH-benzotriazole- 1 -yl)- 1 , 1 ,3,3-tetramethyluronium tetrafluoroborate (TBTU), 0-(7-azobenzotriazol- 1 -yl)- 1 , 1 ,3,3-tetramethyluronium hexafluorophosphate (HATU), diethyl phosphorocyanidate (DEPC) or (benzotriazol-l-yloxy) tripyrrolodinophosphonium hexafluorophosphate (PyBOP), optionally associated with a coupling auxiliary such as N-hydroxy succinimide (NHS), N-hydroxy benzotriazole (HOBt), 3,4-dihydro-3-hydroxy-4-oxo-l,2,3-benzotriazole (HOOBt), l-hydroxy-7- azabenzotriazole (HAt), N-hydroxysylfosuccinimide (sulfo NHS) or dimethyl aminopyridine (DMAP). Preferably the coupling agent is HATU or DEPC.

The reaction can also be performed in the presence of a base such as DIEA

(diisopropy lethy lamine) .

In particular, the peptide coupling is performed in the presence of HATU or DEPC and DIEA.

Said reaction can be carried out in a polar aprotic solvent such as dichloromethane (DCM) or dimethylformamide (DMF), in particular at a temperature of between -15°C and 40°C.

When X = CI, the condensation reaction will be conducted in the presence of a base which may be organic or inorganic, such as Et 3 N, iPr 2 NEt, pyridine, NaH, Cs 2 C0 3 , or K 2 C0 3 .

The reaction can be carried out in a solvent such as tetrahydrofuran (THF), dichloromethane (DCM), dimethylformamide (DMF), or dimethylsulfoxide (DMSO), in particular at a temperature of between -20° and 100°C. The compounds of formulas (VI) and (VII) can be prepared following synthesis protocols described in the experimental part below or following techniques known to those skilled in the art. A further subject of the present invention is a second method for preparing a formula (I) compound comprising a substitution reaction between a compound of following formula (VIII):

(VIII)

where Ri, R 2 an R 3 are such has previously defined, and

a compound of following formula (X):

where R4 a is an R4 group such as previously defined optionally in protected form, and Y is a leaving group such as CI, Br, I, OS0 2 CH 3 , OS0 2 CF 3 or O-Tosyl.

The substitution reaction will be notably conducted in the presence of a base which may be organic or inorganic such as Et 3 N, iPr 2 NEt, NaH, Cs 2 C0 3 , or K 2 C0 3 .

This reaction can be implemented in a polar solvent, preferably anhydrous, such as DCM, THF, DMF or DMSO, in particular at a temperature of between -20° and 100°C.

The compounds of formulas (VIII) and (X) can be prepared following the synthesis protocols described in the experimental part below or using techniques known to those skilled in the art. A further subject of the present invention is a third method for preparing a formula (I) compound in which R4 is a -CH 2 R4b group with R4b representing an aryl or aryl-(Ci-Cv)alkyl group substituted by one or more groups (in particular one, preferably on the aryl moiety) chosen from among OH and NR 9 R 10 groups,

comprising a reductive amination reaction between a compound of following formula (VIII):

where Ri, R 2 and R 3 are such as previously defined, and

a compound of following formula (XI):

where R4b is such as defined above.

The reductive amination reaction can be carried out in the presence of a reducing agent such as NaBH 4 , NaBH 3 CN or NaBH(OAc) 3 and optionally titanium isopropoxide (IV).

The pH can be controlled by adding an acid such as acetic acid, in particular to reach a pH of between 4 and 5.

This reaction can be implemented in a polar solvent such as DCE (1,2- dichloroethane), DCM, THF, DMF or methanol, in particular at a temperature of between -20° and 100°C.

The compounds of formulas (VIII) and (XI) can be prepared following synthesis protocols described in the experimental part below or using techniques known to those skilled in the art.

The compound obtained after the condensation/substitution/reductive amination step of one of the three above methods can be subjected to additional deprotection steps particularly concerning the substituents R 2 and R 4 and optionally additional functionalization steps using methods well known to skilled persons.

When R 2 represents a COOH group, the condensation/substitution/reductive amination step mentioned above can be performed from a compound of formula (VI) with an R 2 group representing a COO-((Ci-C 6 )alkyl) ester function, this ester function then possibly being saponified to yield a formula (I) compound with R 2 = COOH. When the R4 group comprises a NH function, this can be protected before performing the condensation/substitution/reductive amination reaction by substituting the nitrogen atom by an N-protective group such as a Boc or Fmoc group.

By « protective group » in the present invention is meant a group which selectively blocks a reactive site in a multifunctional compound such that a chemical reaction can selectively be carried out at another non-protected reactive site in the meaning conventionally associated therewith in chemical synthesis.

By « N-protective group » in the present invention is meant any substituent which protects the NH group against undesirable reactions such as the N-protective groups described in Greene, « Protective Groups In Organic synthesis », (John Wiley & Sons, New York (1981)) and Harrison et al. « Compendium of Synthetic Organic Methods », Vols. 1 to 8 (J. Wiley & sons, 1971 to 1996). The N-protective groups comprise carbamates, amides, N-alkylated derivatives, amino acetal derivatives, N- benzyl derivatives, imine derivatives, enamine derivatives and N-heteroatom derivatives. The N-protecting groups can be formyl; an aryl, such as a phenyl, optionally substituted with one or several methoxy groups such as /?-methoxyphenyl (PMP); an aryl-(Ci-C6)alkyl, such as a benzyl, the aryl moiety being optionally substituted with one or several methoxy groups, such as benzyl (Bn), /?-methoxybenzyl (PMB) or 3,4-dimethoxybenzyl (DMPM); -CO-R G pi such as acetyl (Ac), pivaloyl (Piv or Pv), benzoyl (Bz) or /?-methoxybenzylcarbonyl (Moz); -C0 2 -RGPI such as tbutyloxycarbonyl (Boc), trichloroethoxycarbonyl (TROC), allyloxycarbonyl (Alloc), benzyloxycarbonyl (Cbz or Z) or 9-fluorenylmethyloxycarbonyl (Fmoc); -S0 2 -RGPI such as phenylsulfonyl, tosyl (Ts or Tos) or 2-nitrobenzenesulfonyl (also called nosyl - Nos or Ns); and the like, with RGPI representing a (Ci-C 6 )alkyl optionally substituted with one or several halogen atoms such as F or CI; a (C 2 -Ce)alkenyl such as an allyl; an aryl, such as a phenyl, optionally substituted with one or several groups chosen among OMe (methoxy) and N0 2 (nitro); an aryl-(Ci-C 6 )alkyl, such as a benzyl, the aryl moiety being optionally substituted with one or several methoxy groups; or a 9-fluorenylmethyl group.

In particular, the N-protective group comprises formyl, acetyl, benzoyl, pivaloyl, phenylsulfonyl, benzyl (Bn), t-butyloxycarbonyl (Boc), benzyloxycarbonyl (Cbz), p-methoxybenzyloxycarbonyl, p-nitrobenzyl-oxycarbonyl, trichloroethoxy carbonyl (TROC), allyloxycarbonyle (Alloc), 9-fluorenylmethyloxycarbonyl (Fmoc), trifluoro-acetyl, benzyl carbamates (substituted or not) and similar. It may in particular be a Boc or Fmoc group.

The protection of the NH amine function by a Boc or Fmoc group and its subsequent deprotection, after the condensation/substitution/reductive amination reaction, are well known to persons skilled in the art and are described in particular in the experimental part below.

The formula (I) compound obtained with one of the three methods mentioned above can also be salified by adding a pharmaceutically acceptable base or acid, in particular a pharmaceutically acceptable acid such as trifluoroacetic acid. Said step can optionally be performed at the same time as another reaction step, in particular at the same time as a deprotection step when this must be performed in an acid medium for example.

The compound obtained with one of these three methods, optionally after additional step(s) for deprotection, functionalization and/or salification, can be separated from the reaction medium using methods well known to skilled persons, such as by extraction, solvent evaporation or by precipitation and filtration.

The compound may also be purified if necessary using techniques well known to skilled persons, e.g. by recrystallization if the compound is crystalline, by distillation, by silica gel column chromatography or high performance liquid chromatography (HPLC). The following examples illustrate the invention without however limiting the scope thereof.

EXAMPLES

I - Synthesis of the compounds of the invention

The following abbreviations are used in the following examples:

aq. Aqueous ee enantiomeric excess

equiv equivalent

ESI Electrospray ionisation

LC/MS Liquid Chromatography coupled with Mass Spectrometry

HPLC High Performance Liquid Chromatography

NMR Nuclear Magnetic Resonance

sat. saturated

UV ultraviolet

Reference Example 1

(S)-2-((S)-2-((3-aminopropyl)(methyl)amino)-3-methylbutanami do)- V- ((3R,4S,5S)-3-methoxy-l-((S)-2-((lR,2R)-l-methoxy-2-methyl-3 -oxo-3-(((S)- 2-phenyl-l-(thiazol-2-yl)ethyl)amino)propyl)pyrrolidin-l-yl) -5-methyl-l- oxoheptan-4-yl)- V,3-dimethylbutanamide, bis trifluoroacetic acid

Example 1A: (4R, 5S)-4-methyl-5-phenyl-3-propanoyl-l,3-oxazolidin-2- one

(4R, 5S)-4-methyl-5-phenyl-l,3-oxazolidin-2-one (5.8 g, 32.7 mmol, 1.00 equiv) was dissolved in tetrahydrofuran (THF, 120 mL) in an inert atmosphere. The mixture was cooled to -78°C and n-butyllithium (14.4 mL) was added drop-wise. After agitation for 30 minutes at -78°C, propanoyl chloride (5.7 mL) was added. Agitation was continued for 30 minutes at -78°C then overnight at ambient temperature. The reaction mixture was concentrated then re-dissolved in 200 mL of water. The pH of the solution was adjusted to 7 with sodium bicarbonate saturated aqueous solution. This aqueous phase was extracted 3 times with 100 mL of ethyl acetate (EtOAc). The organic phases were combined, dried over sodium sulfate, filtered and concentrated to yield 6.8 g (89 %) of compound 1A in the form of a yellow oil.

Example IB: tert-butyl (2S)-2-[(lR,2R)-l-hydroxy-2-methyl-3-[(4R,5S)-4- methy -2-oxo-5-phenyl- 1 ,3-oxazolidin-3yl]-3-oxopropyl]pyrrolidine- 1 -carboxylate

Compound 1A (17.6 g, 75.45 mmol, 1.00 equiv) was dissolved in dichloromethane (DCM, 286 mL) in an inert atmosphere. This solution was cooled with an ice bath. Triethylamine (TEA, 12.1 mL, 1.15 equiv) and Bu 2 BOTf (78.3 mL, 1.04 equiv) were added drop-wise whilst holding the temperature of the reaction mixture below 2°C. Agitation was continued at 0°C for 45 minutes, after which the reaction was cooled to -78°C. A solution of tert-butyl (2S)-2-formylpyrrolidine-l- carboxylate (8.5 g, 42.66 mmol, 0.57 equiv) in DCM (42 mL) was added drop-wise. Agitation was continued for 2 hours at -78°C, then for 1 hour at 0°C and finally 1 hour at ambient temperature. The reaction was neutralised with 72 mL of phosphate buffer (pH = 7.2 - 7.4) and 214 mL methanol, and cooled to 0°C. A solution of 30 % hydrogen peroxide in methanol (257 mL) was added drop-wise whilst maintaining the temperature below 10°C. Agitation was continued for 1 hour at 0°C. The reaction was neutralised with 142 mL of water, then concentrated under reduced pressure. The resulting aqueous solution was extracted 3 times with 200 mL EtOAc. The organic phases were combined, dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of EtOAc and petroleum ether (EtOAc:PE = 1 :8) to yield 13.16 g (40 %) of compound IB in the form of a colourless oil.

Example 1C: (2R,3R)-3-[(2S)-l-[(tert-butoxy)carbonyl]pyrrolidin-2-yl]-3- hydroxy-2-methylpropanoic acid

Compound IB (13.16 g, 30.43 mmol, 1.00 equiv) was dissolved in THF (460 mL) in the presence of hydrogen peroxide (30 % in water, 15.7 mL), then cooled with an ice bath. An aqueous solution of lithium hydroxide (0.4 mol/L, 152.1 mL) was added drop-wise whilst holding the reaction temperature below 4°C. The reaction mixture was agitated 2.5 hours at 0°C. An aqueous solution of Na 2 S0 3 (1 mol/L, 167.3 mL) was added drop-wise whist holding the temperature at 0°C. The reaction mixture was agitated 14 hours at ambient temperature, then neutralised with 150 mL of cold sodium bicarbonate saturated solution and washed 3 times with 50 mL of DCM. The pH of the aqueous solution was adjusted to 2-3 with a 1M aqueous solution of KHS0 4 . This aqueous solution was extracted 3 times with 100 mL of EtOAc. The organic phases were combined, washed once with saturated NaCl solution, dried over sodium sulfate, filtered and concentrated to yield 7.31 g (88 %) of compound 1C in the form of a colourless oil.

Example ID: (2R,3R)-3-[(2S)- l-[(tert-butoxy)carbonyl]pyrrolidin-2-yl]-3- methoxy-2-methylpropanoic acid

Compound 1C (7.31 g, 26.74 mmol, 1.00 equiv) was dissolved in an inert atmosphere in THF (135 mL) in the presence of iodomethane (25.3 mL). The reaction medium was cooled with an ice bath after which NaH (60 % in oil, 4.28 g) was added in portions. The reaction was left under agitation 3 days at 0°C and then neutralised with 100 mL of sodium bicarbonate saturated aqueous solution and washed 3 times with 50 mL ether. The pH of the aqueous solution was adjusted to 3 with 1M aqueous KHSO 4 solution. This aqueous solution was extracted 3 times with 100 mL of EtOAc. The organic phases were combined, washed once with 100 mL of Na 2 S 2 0 3 (5 % in water), once with NaCl-saturated solution, then dried over sodium sulfate, filtered and concentrated to yield 5.5 g (72 %) of compound ID in the form of a colourless oil. Example -methoxy-N-methyl-2-phenylacetamide

2-phenylacetic acid (16.2 g, 118.99 mmol, 1.00 equiv) was dissolved in dimethylformamide (DMF, 130 mL) then cooled to -10°C. Diethyl phosphorocyanidate (DEPC, 19.2 mL), methoxy(methyl)amine hydrochloride (12.92 g, 133.20 mmol, 1.12 equiv) and triethylamine (33.6 mL) were added. The reaction mixture was agitated 30 minutes at -10°C then 2.5 hours at ambient temperature. It was then extracted twice with 1 litre of EtOAc. The organic phases were combined, washed twice with 500 mL of NaHCC"3 (sat.), once with 400 mL of water, then dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with an EtOAc and PE mixture (1 : 100 to 1 :3) to yield 20.2 g (95 %) of compound IE in the form of a yellow oil.

Example IF: 2-phenyl-l-(l ,3-thiazol-2-yl)ethan-l-one

Tetramethylethylenediamine (TMEDA, 27.2 mL) was dissolved in THF 300 mL) in an inert atmosphere, then cooled to -78°C before the drop-wise addition of n- BuLi (67.6 mL, 2.5 M). 2-bromo-l,3-thiazole (15.2 mL) was added drop-wise and agitation was continued 30 minutes at -78°C. Compound IE (25 g, 139.50 mmol, 1.00 equiv) dissolved in THF (100 mL) was added drop-wise. Agitation was continued for 30 minutes at -78°C then 2 hours at -10°C. The reaction was neutralised with 500 mL of KHSO 4 (sat.), then extracted 3 times with 1 litre of EtOAc. The organic phases were combined, washed twice with 400 mL water and twice with 700 mL of NaCl (sat.), then dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of EtOAc and PE (1 : 100 to 1 : 10) to yield 25 g (88 %) of compound IF in the form of a yellow oil.

Example 1G: (lR)-2-phenyl-l-(l ,3-thiazol-2-yl)ethan-l-ol

In an inert atmosphere, a solution of compound IF (15 g, 73.8 mmol, 1.00 equiv.) in ether (300 mL) was added drop-wise to (+)-B-chloro diisopinocampheylborane ((+)-Ipc 2 BCl, 1 10.8 mL). The reaction mixture was agitated 24 hours at 0°C, then neutralised with 300 mL of a (1 : 1) mixture of NaOH (10 % in water) and H 2 0 2 (30 % in water), and finally extracted three times with 500 mL of EtOAc. The organic phases were combined, washed twice with 300 mL of K 2 C0 3 (sat.) and once with 500 mL of NaCl (sat.), then dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of EtOAc and PE (1 :20 to 1 :2) to yield 6.3 g (42 %) of compound 1G in the form of a white solid.

Example 1 -[(l S)-l-azido-2-phenylethyl]-l ,3-thiazole

Compound 1G (6 g, 29.23 mmol, 1.00 equiv.) was dissolved in an inert atmosphere in THF (150 mL) in the presence of triphenylphosphine (13 g, 49.56 mmol, 1.70 equiv.), then cooled to 0°C. Diethylazodicarboxylate (DEAD, 7.6 mL) was added drop-wise, followed by diphenylphosphorylazide (DPPA, 1 1 mL), the cold bath was then removed and the solution was left under agitation 48 hours at ambient temperature. The medium was concentrated under reduced pressure. The residue was purified on a silica column with a mixture of EtOAc and PE (1 : 100 to 1 :30) to yield 8 g of partly purified compound 1H in the form of a yellow oil. Compound 1H was used as such in the following step.

Example II: tert-butyl N-[(l S)-2-phenyl-l-(l ,3-thiazol-2-yl)ethyl] carbamate.

Compound 1H (6.5 g, 28.2 mmol, 1.00 equiv) was dissolved in an inert atmosphere in THF (100 mL) in the presence of triphenylphosphine (6.5 g, 33.9 mmol, 1.20 equiv.), and heated to 50°C for 2 hours. Ammonia (70 mL) was then added and heating was continued for 3 hours. The reaction was cooled, neutralised with 500 mL water, then extracted 3 times with 500 mL of EtOAc. The organic phases were combined and extracted twice with 500 mL of IN HC1. The aqueous phases were combined, brought to pH 8-9 by adding a sodium hydroxide solution (10 % in water), then extracted 3 times with 500 mL of DCM. The organic phases were combined, dried over sodium sulfate, filtered and concentrated to yield 4.8 g (83 %) of (lS)-2-phenyl- l-(l,3-thiazol-2-yl)ethan-l-amine in the form of a yellow oil. This compound was then protected with a Boc group ((tert-butoxy)carbonyl) so that it could be purified. It was dissolved in an inert atmosphere in 1,4-dioxane (40 mL), then cooled to 0°C. (Boc) 2 0 (10.26 g, 47.01 mmol, 2.00 equiv) diluted in 20 mL of 1,4-dioxane was added drop-wise. The cold bath was removed and the solution left under agitation overnight at ambient temperature before being neutralised with 300 mL of water and extracted twice with 500 mL of EtOAc. The organic phases were combined, dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of EtOAc and PE (1 : 100 to 1 :20, ee = 93 %). It was then recrystallized in a hexane/acetone mixture (~ 5-10 / 1, lg / 10 mL) to yield 6 g (84 %) of compound II in the form of a white solid (ee > 99 %).

Example 1J: tert-butyl (2S)-2-[(lR,2R)-l-methoxy-2-methyl-2-[[(lS)-2- phen - 1 -( 1 ,3-thiazol-2-yl)ethyl]carbamoyl]ethyl]pyrrolidine- 1 -carboxylate

Compound II (3 g, 9.86 mmol, 1.00 equiv) was dissolved in an inert atmosphere in 10 mL DCM. Trifluoroacetic acid (TFA, 10 mL) was added and the solution left under agitation overnight at ambient temperature, then concentrated under reduced pressure to yield 2.0 g (64 %) of (lS)-2-phenyl-l-(l,3-thiazol-2-yl)ethan-l-amine; trifluoroacetic acid in the form of a yellow oil. This intermediate was re-dissolved in 20 mL of DCM after which compound ID (1.8 g, 6.26 mmol, 1.05 equiv), DEPC (1.1 g, 6.75 mmol, 1.13 equiv) and diisopropylethylamine (DIEA, 1.64 g, 12.71 mmol, 2.13 equiv) were added. The reaction mixture was left under agitation overnight at ambient temperature, then concentrated under reduced pressure. The residue was purified on a silica column with a mixture of EtOAc and PE (1 : 100 to 1 :3) to yield 2.3 g (81 %) of compound 1 J in the form of a pale yellow solid.

Example IK: (2R,3R)-3-methoxy-2-methyl-N-[(l S)-2-phenyl-l-(l ,3-thiazol-2- yl)ethyl]-3- -pyrrolidin-2-yl]propanamide; trifluoroacetic acid

Compound 1J (2.25 g, 4.75 mmol, 1.00 equiv) was dissolved in an inert atmosphere in 10 mL of DCM. TFA (10 mL) was added and the solution left under agitation overnight at ambient temperature, then concentrated under reduced pressure to yield 2.18 g (94 %) of compound IK in the form of a yellow oil.

Example 1L: 2S,3S)-2-(benz lamino)-3-methylpentanoic acid

(2S,3S)-2-amino-3-methylpentanoic acid (98.4 g, 750 mmol, 1.00 equiv) was added at ambient temperature and in portions to a 2N sodium hydroxide solution (375 mL). Benzaldehyde (79.7 g, 751.02 mmol, 1.00 equiv) was quickly added and the resulting solution was agitated 30 minutes. Sodium borohydride (10.9 g, 288.17 mmol, 0.38 equiv) was added in small portions, whilst holding the temperature at between 5 and 15°C. Agitation was continued for 4 hours at ambient temperature. The reaction mixture was diluted with 200 mL of water, then washed twice with 200 mL of EtOAc. The pH of the aqueous solution was adjusted to 7 with a 2N hydrochloric acid solution. The formed precipitate was collected by filtering and gave 149.2 g (90 %) of compound 1L in the form of a white solid.

)-2-[benzyl(methyl)amino]-3-meth lpentanoic acid Compound 1L (25 g, 112.97 mmol, 1.00 equiv) was dissolved in an inert atmosphere in formic acid (31.2 g) in the presence of formaldehyde (36.5 % in water, 22.3 g). The solution was agitated 3 hours at 90°C then concentrated under reduced pressure. The residue was triturated in 250 mL of acetone, then concentrated. This trituration/evaporation operation was repeated twice with 500 mL of acetone to yield 21.6 g (81 %) of compound 1M in the form of a white solid.

Exam le IN: (2S ,3 S)-2-[benzyl(methyl)amino]-3-methylpentan- 1 -o 1

L1AIH 4 (0.36 g) was suspended in 10 mL of THF in an inert atmosphere at 0°C. Compound 1M (1.5 g, 6.37 mmol, 1.00 equiv) was added in small portions whilst holding the temperature at between 0 and 10°C. The reaction mixture was agitated 2 hours at 65°C, then again cooled to 0°C before being neutralised with successive additions of 360 of water, 1 mL of 15 % sodium hydroxide and 360 of water. The aluminium salts which precipitated were removed by filtering. The filtrate was dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of EtOAc and PE (1 :50) to yield 820 mg (58 %) of compound IN in the form of a pale yellow oil.

Example IO: (2S,3S)-2-[benzyl(methyl)amino]-3-methylpentanal

Oxalyl chloride (0.4 mL) was dissolved in DCM (15 mL) in an inert atmosphere.

The solution was cooled to -70°C and a solution of dimethylsulfoxide (DMSO (0.5 mL) in DCM (10 mL) was added drop-wise for 15 minutes. The reaction mixture was agitated 30 minutes after which a solution of compound IN (820 mg, 3.70 mmol, 1.00 equiv) in DCM (10 mL) was added drop-wise for 15 minutes. The reaction mixture was agitated a further 30 minutes at low temperature, then triethylamine (2.5 mL) was slowly added. The reaction mixture was agitated 1 hour at -50°C, the cold bath was then removed and the reaction neutralised with 25 mL of water whilst allowing the temperature to return to normal. The solution was washed once with 30 mL of NaCl- saturated aqueous solution, then dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of EtOAc and PE (1 :200) to yield 0.42 g (52 %) of compound lO in the form of a yellow oil.

Example IP: (2S,3S)-N-benz l-l , l-dimethox -N,3-dimethylpentan-2-amine

Compound lO (4.7 g, 21.43 mmol, 1.00 equiv) was dissolved in 20 mL of methanol at 0°C. Concentrated sulfuric acid (4.3 mL) was added drop-wise and agitation was continued for 30 minutes at 0°C. Trimethyl ortho formate (21.4 mL) was added, the cold bath removed and the reaction medium left under agitation for 3 hours at ambient temperature. The reaction medium was diluted with 200 mL of EtOAc, successively washed with 100 mL of 10 % Na 2 C0 3 and 200 mL of saturated NaCl, then dried over sodium sulfate, filtered and concentrated under reduced pressure to yield 3.4 g (60 %) of compound IP in the form of a pale yellow oil.

Example 1Q: [[l-(tert-butoxy)ethenyl]oxy](tert-butyl)dimethylsilane

Diisopropylamine (20 g, 186.71 m mol, 1.08 equiv) was dissolved in 170 mL of THF in an inert atmosphere and cooled to -78°C. nBuLi (2.4 M, 78.8 mL) was added drop-wise and the solution agitated 30 minutes at low temperature (to give LDA- lithium diisopropylamide) before adding tert-butyl acetate (20 g, 172.18 mmol, 1.00 equiv). The reaction mixture was agitated 20 minutes at -78°C before adding hexamethylphosphoramide (HMPA, 25.8 mL) and a solution of tertbutyldimethylchlorosilane (TBDMSCl, 28 g, 185.80 mmol, 1.08 equiv) in 35 mL of THF. Agitation was continued for 20 additional minutes at low temperature, and the cold bath was then removed. The solution was concentrated under reduced pressure. The residue was re-dissolved in 100 mL of water and extracted 3 times with 100 mL of PE. The organic phases were combined, washed once with 500 mL of NaCl-saturated aqueous solution, dried over sodium sulfate, filtered and concentrated. The residue was purified by distillation to yield 16.6 g (83 %) of compound 1Q in the form of a colourless oil.

Example 1R: tert-butyl (3R,4S,5S)-4-[benzyl(methyl)amino]-3-methoxy-5- methyl he tanoate

Compound IP (2.0 g, 7.54 mmol, 1.00 equiv) and compound 1Q (2.6 g, 11.28 mmol, 1.50 equiv) were dissolved in 33 mL of DCM in an inert atmosphere. The solution was cooled to 0°C. DMF (1.2 g) was added drop-wise together with a solution of BF 3 Et 2 0 (2.1 g) in 7.5 mL of DCM. Agitation was continued for 24 hours at 0°C. The reaction medium was washed once with 30 mL of sodium carbonate (10 %) and twice with 50 mL of NaCl-saturated aqueous solution, then dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of EtOAc and PE (1 : 100) to yield 1.82 g (91 %) of compound 1R in the form of a yellow oil.

Example IS: (3R,4S,5S)-3-methoxy-5-methyl-4-(methylamino)heptanoate hydrochloride

Compound 1R (2.4 g, 6.87 mmol, 1.00 equiv) was dissolved in an inert atmosphere in 35 mL of ethanol in the presence of Pd/C (0.12 g) and concentrated hydrochloric acid (0.63 mL). The nitrogen atmosphere was replaced by a hydrogen atmosphere and the reaction medium was left under agitation 18 hours at ambient temperature. The reaction medium was filtered and concentrated under reduced pressure. The residue was triturated in 50 mL of hexane and the supernatant removed which, after drying under reduced pressure, gave 1.66 g (82 %) of compound IS in the form of a white solid.

Example IT: tert-butyl (3R,4S,5S)-4-[(2S)-2-[[(benzyloxy)carbonyl]amino]- N,3 -dimethylbutanamido] -3 -methoxy-5 -methylheptanoate

(2S)-2-[[(benzyloxy)carbonyl]amino]-3-methylbutanoic acid (15 g, 0.40 mmol, 1.00 equiv) was dissolved in 300 mL of DCM in the presence of DIEA (38.3 mL) and bromotripyrrolidinophosphonium hexafluorophosphate (PyBrOP, 32.3g). The solution was agitated 30 minutes at ambient temperature before adding compound IS (15.99g, 0.42 mmol, 1.07 equiv). The reaction medium was agitated 2 hours and then concentrated. The residue was purified in reverse phase (CI 8) with a mixture of acetonitrile (ACN) and water (30:70 to 100:0 in 40 minutes) to yield 17 g (58 %) of compound IT in the form of a colourless oil.

Example 1U: tert-butyl (3R,4S,5S)-4-[(2S)-2-amino-N,3-dimethylbutanamido]- 3 -methoxy-5 -methylheptanoate

Compound IT (76 mg, 0.15 mmol, 1.00 equiv) was dissolved in an inert atmosphere in 10 mL of ethanol in the presence of Pd/C (0.05 g). The nitrogen atmosphere was replaced by a hydrogen atmosphere and the reaction agitated 2 hours at ambient temperature. The reaction medium was filtered and concentrated under reduced pressure to yield 64 mg of compound 1U in the form of a colourless oil.

Example IV: (3R,4S,5S)-4-[(2S)-2-[[(9H-fluoren-9-ylmethoxy)carbonyl] amin -N,3-dimethylbutanamido]-3-methoxy-5-methylheptanoate

Compound 1U (18.19 g, 50.74 mmol, 1.00 equiv) was dissolved in 400 mL of a 1 ,4-dioxane/water mixture (1 : 1) in the presence of sodium bicarbonate (12.78 g, 152 mmol, 3.00 equiv) and 9H-fluoren-9-ylmethyl chloro formate (Fmoc-CI, 19.69 g, 76 mmol, 1.50 equiv), then agitated 2 hours at ambient temperature. The reaction medium was then diluted with 500 mL of water and extracted 3 times with 200 mL of EtOAc. The organic phases were combined, washed once with 200 mL of NaCl-saturated aqueous solution, dried over sodium sulfate, filtered and concentrated to yield 40 g of partly purified compound IV in the form of a pale yellow oil.

Example 1W: (3R,4S,5S)-4-[(2S)-2-[[(9H-fluoren-9-ylmethoxy)carbonyl] amino]- -dimethylbutanamido]-3-methoxy-5-methylheptanoic acid

Compound IV (40 g, 68.88 mmol, 1.00 equiv) was dissolved in a neutral atmosphere in 600 mL of DCM. TFA (300 mL) was added. The solution was agitated 2 hours at ambient temperature, then concentrated under reduced pressure. The residue was purified on a silica column with a mixture of methanol and DCM (1 : 10) to yield 23.6 g (65 %) of compound 1W in colourless oil form.

Example IX: 9H-fluoren-9-ylmethyl N-[(lS)-l-[[(3R,4S,5S)-3-methoxy-l- [(2S)-2-[(lR,2R)-l-methoxy-2-methyl-2-[[(lS)-2-phenyl-l-(l,3 -thiazol-2- yl)ethyl]carbamoyl] ethyljpyrrolidin- 1 -yl]-5-methyl- 1 -oxoheptan-4-yl] (methyl) carbamoyl]-2-methylpropyl]carbamate

Compound 1W (2.53 g, 4.82 mmol, 1.08 equiv) was dissolved in 20 mL of DCM in the presence of compound IK (2.18 g, 4.47 mmol, 1.00 equiv), DEPC (875 mg, 5.37 mmol, 1.20 equiv) and DIEA (1.25 g, 9.67 mmol, 2.16 equiv). The reaction mixture was left under agitation overnight at ambient temperature, then successively washed with 50 mL of saturated KHS0 4 and 100 mL of water, dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of methanol and DCM (1 :200 to 1 :40) to yield 2.8 g (71 %) of compound IX in the form of a pale yellow solid. Example 1Y: (2S)-2-amino-N-[(3R,5S)-3-methoxy-l-[(2S)-2-[(lR,2R)-l- methoxy-2-methyl-2- [ [( 1 S)-2-phenyl- 1 -( 1 ,3 -thiazol-2-yl)ethyl] carbamoyl] ethyl] pyrrolidin- 1 -yl] -5 -methyl- 1 -oxoheptan-4-yl]-N,3-dimethylbutanamide

Compound IX (2.8 g, 3.18 mmol, 1.00 equiv) was dissolved in acetonitrile

(ACN, 12 mL) in the presence of piperidine (3 mL) and left under agitation 18 hours at ambient temperature. The reaction was neutralised with 50 mL of water, then extracted twice with 100 mL of DCM. The organic phases were combined, dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of methanol and DCM (1 : 100 to 1 :40) to yield 1.2 g (57 %) of compound 1Y in the form of a yellow solid.

Example IZA: (2S)-2-[ [(tert-butoxy)carbonyl] (methyl)amino] -3 -methyl butanoic acid

(2S)-2-[[(tert-butoxy)carbonyl]amino]-3-methylbutanoic acid (63 g, 289.97 mmol, 1.00 equiv) was dissolved in an inert atmosphere in THF (1000 mL) in the presence of iodomethane (181 mL). The solution was cooled to 0°C before adding sodium hydride (1 16 g, 4.83 mol, 16.67 equiv) in small portions. The reaction mixture was agitated for 1.5 hours at 0°C, the cold bath was then removed and agitation continued for 18 hours. The reaction was neutralised with 200 mL of water and then concentrated under reduced pressure. The residual aqueous phase was diluted with 4 litres of water, washed once with 200 mL of EtOAc and its pH adjusted to between 3 and 4 with a IN solution of hydrochloric acid. The mixture obtained was extracted 3 times with 1.2 L of EtOAc. The organic phases were combined, dried over sodium sulfate, filtered and concentrated to yield 60 g (89 %) of compound IZA in the form of a yellow oil. Example 1ZB: benzyl (2S)-2- [ [(tert-butoxy)carbonyl] (methyl)amino] -3 -methyl butanoate

Compound 1ZA (47 g, 203.21 mmol, 1.00 equiv) was dissolved in DMF (600 mL) in the presence of L1 2 CO 3 (15.8 g, 213.83 mmol, 1.05 equiv). The solution was cooled to 0°C then benzyl bromide (BnBr 57.9 g, 338.53 mmol, 1.67 equiv) was added drop-wise. The reaction mixture was left under agitation overnight before being neutralised with 400 mL of water and filtered. The solution obtained was extracted twice with 500 mL of EtOAc. The organic phases were combined, dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of EtOAc and PE (1 : 100 to 1 :20) to yield 22.5 g (34 %) of compound 1ZB in the form of a yellow oil.

Example 1 -3-methyl-2- methylamino)butanoate hydrochloride

Compound 1ZB (22.5 g, 70.00 mmol, 1.00 equiv) was dissolved in 150 mL of DCM. Gaseous hydrochloric acid was bubbled. The reaction was agitated 1 hour at ambient temperature and then concentrated under reduced pressure to yield 17 g (94 %) of compound 1ZC in the form of a yellow solid.

Example 1ZD: tert-but l N-(3,3-diethoxypropyl)carbamate

3,3-diethoxypropan-l-amine (6 g, 40.76 mmol, 1.00 equiv) was dissolved in 1,4-dioxane (30 mL) in the presence of TEA (4.45 g, 43.98 mmol, 1.08 equiv), then cooled to 0°C. (Boc) 2 0 (9.6 g, 43.99 mmol, 1.08 equiv) diluted in 20 mL of 1,4-dioxane was added drop-wise. The solution was agitated 2 hours at 0°C then overnight at ambient temperature before being neutralised with 10 mL of water. The pH was adjusted to 5 with HCI (1 %). The solution was extracted 3 times with 50 mL of EtOAc. The organic phases were combined, dried over sodium sulfate, filtered and concentrated to yield 8.21 g (81 %) of compound 1ZD in the form of a pale yellow oil.

Example 1Z: tert-butyl N-(3-oxopropyl) carbamate

Compound 1ZD (8.20 g, 33.15 mmol, 1.00 equiv) was dissolved in 18.75 mL of acetic acid and left under agitation overnight at ambient temperature. The reaction medium was then extracted 3 times with 30 mL of EtOAc. The organic phases were combined, washed 3 times with 30 mL of saturated NaCl solution, dried over sodium sulfate, filtered and concentrated to yield 5 g (87 %) of compound 1ZE in the form of a dark red oil.

Example 1ZF: (2S)-2- [(3 - [[(tert-butoxy)carbonyl] amino]propyl)(methyl) amino]-3-methylbutanoic ac

Compound 1ZE (2.4 g, 13.86 mmol, 1.00 equiv) was dissolved in 50 mL of THF in the presence of compound 1ZC (3.56 g, 13.81 mmol, 1.00 equiv) and DIEA (9.16 mL, 4.00 equiv). The reaction mixture was agitated 30 minutes at ambient temperature before adding sodium triacetoxyborohydride (5.87 g, 27.70 mmol, 2.00 equiv). Agitation was continued overnight, then the reaction was neutralised with 100 mL of water and extracted 3 times with 50 mL of EtOAc. The organic phases were combined, dried over sodium sulfate, filtered and concentrated. The residue was partly purified on a silica column with a mixture of EtOAc and PE (1 :4). The crude product obtained was re-dissolved in 20 mL of methanol in the presence of Pd/C (1.2 g) and hydrogenated for 20 minutes at normal temperature and pressure. The reaction medium was filtered and concentrated under reduced pressure to yield 200 mg (5 %) of compound 1ZF in the form of a white solid.

Example 1ZG: tert-butyl N-(3-[[(lS)-l-[[(lS)-l-[[(3R,4S,5S)-3-methoxy-l- [(2S)-2-[(lR,2R)-l-methoxy-2-methyl-2-[[(lS)-2-phenyl-l-(l,3 -thiazol-2-yl)ethyl] carbamoyl]thyl]pyrrolidin-l-yl]-5-methyl-l -oxoheptan-4yl](rnethyl) carbamoyl]-2- methylpropyl]carbamoyl]-2-methylpropyl](methyl)amino]propyl) carbamate

Compound 1Y (50 mg, 0.08 mmol, 1.00 equiv) was dissolved in 2 mL of DMF in the presence of compound 1ZF (26.2 mg, 0.09 mmol, 1.20 equiv), DIEA (37.7 mL) and 0-(7-azabenzotriazol-l-yl)-7V r ,N,N',N , -tetramethyluronium hexafluorophosphate (HATU, 43.3 mg, 0.1 1 mmol, 1.50 equiv). The reaction was left under agitation overnight at ambient temperature, then diluted with 10 mL of water and extracted 3 times with 5 mL of EtOAc. The organic phases were combined, dried over sodium sulfate, filtered and concentrated to yield 100 mg of compound 1ZG in the form of a partly purified colourless oil.

Example 1: Compound 1ZG (90 mg, 0.10 mmol, 1.00 equiv) was dissolved in a neutral atmosphere in 2 mL of DCM and the solution was cooled with an ice bath. TFA (1 mL) was added and the reaction agitated for 2 hours at ambient temperature, then concentrated under reduced pressure. The residue was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep CI 8 OBD column, 5 μηι, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % of TFA; Gradient of 18 % to 31 % ACN in 7 minutes then 31 % to 100 % ACN in 2 minutes; Waters 2489 UV Detector at 254 nm and 220 nm). Compound 1 was obtained with a yield of 25 % (23 mg) in the form of a white solid.

LC/MS/UV (Atlantis T3 column, 3 μπι, 4.6 x 100 mm; 35°C; 1 mL / min, 30 % to 60 % ACN in water (20 mM ammonium acetate in 6 minutes); ESI (C 44 H 73 N 7 0 6 S, exact masse 827.53) m/z: 829 (MH + ), 5.84 min (93.7 %, 254 nm).

Ή NMR (300MHz, CD 3 OD, ppm): δ (Presence of rotamers) 7.85 - 7.80 (m, 1H); 7.69 - 7.66 (m, 1H), 7.40 - 7.10 (m, 5H), 5.80 - 5.63 (m, 1H), 4.80 - 4.65 (m, 2H), 4.22 -4.00 (m, 1H), 3.89 - 0.74 (m, 58H). Reference Example 2

(S)-2-((S)-2-(((2-aminopyridin-4-yl)methyl)(methyl)amino)-3- methylbutanamido)- V-((3R,4S,5S)-l-((S)-2-((lR,2R)-3-(((lS,2R)-l-hydroxy-l- phenylpropan-2-yl)amino)-l-methoxy-2-methyl-3-oxopropyl)pyrr olidin-l-yl)- 3-methoxy-5-methyl-l-oxoheptan-4-yl)- V,3-dimethylbutanamide,

trifluoroacetic acid

Example 2A: tert-butyl (S)-2-((lR,2R)-3-(((l S,2R)- 1 -hydroxy- 1 -phenylpropan- 2-yl)amino)- 1 -methoxy-2-methyl-3 -oxopropyl)pyrrolidine- 1 -carboxylate

Compound ID (2.5 g, 8.70 mmol, 1.00 equiv) and (l S,2R)-2-amino-l- phenylpropan-l-ol (1.315 g, 8.70 mmol, 1.00 equiv) were dissolved in an inert atmosphere in DMF (35 mL). The solution was cooled to 0 °C then DEPC (1.39 mL) and TEA (1.82 mL) were added drop-wise. The reaction mixture was agitated 2 hours at 0 °C then 4 hours at ambient temperature. The reaction mixture was diluted with 200 mL of water and extracted three times with 50 mL of EtOAc. The organic phases were combined, washed once with 50 mL of KHS0 4 (1 mol/L), once with 50 mL of NaHC0 3 (sat.), once with 50 mL of NaCl (sat.), then dried over sodium sulfate, filtered and concentrated under reduced pressure to yield 3.6 g (98 %) of compound 2A in the form of a yellow solid.

Example 2B: (2R,3R)-N-((1 S,2R)- 1-hydroxy- 1 -phenylpropan-2-yl)-3-methoxy- 2-methyl-3-((S)-pyrrolidin-2-yl)propanamide2,2,2-trifluoroac etate

Compound 2A (2.7 g, 6.42 mmol, 1.00 equiv) was dissolved in an inert atmosphere in DCM (40 mL) then cooled to 0 °C. TFA (25 mL) was added and the solution agitated for 2 hours at 0 °C. The reaction mixture was concentrated under reduced pressure to yield 4.4 g of compound 2B in the form of a yellow oil.

Example 2C: (9H-fluoren-9-yl)methyl ((S)- 1 -(((3R,4S,5 S)- 1 -((S)-2-((l R,2R)-3- ((( 1 S ,2R)- 1 -hydroxy- 1 -phenylpropan-2-yl)amino)- 1 -methoxy-2-methyl-3 -oxopropyl) pyrrolidin- 1 -yl)-3 -methoxy-5 -methyl- 1 -oxoheptan-4-yl) (methyl)amino)-3 -methyl- 1 - oxobutan-2-yl)carbamate

Compounds 2B (4.4 g, 10.13 mmol, 1.00 equiv) and 1W (5.31 g, 10.12 mmol, 1.00 equiv) were dissolved in an inert atmosphere in DCM (45 mL). The solution was cooled to 0°C then DEPC (1.62 mL) and DIEA (8.4 mL) were added drop-wise. The reation mixture was agitated for 2 hours at 0 °C then at ambient temperature overnight. The reaction mixture was diluted with 100 mL of water and extracted three times with 50 mL of DCM. The organic phases were combined, washed once with 50 mL of KHSO 4 (1 mol/L), once with 50 mL of NaHC0 3 (sat.), once with 50 mL of NaCl (sat.), then dried over sodium sulfate, filtered and concentrated under pressure to yield 3.3 g (39 %) of compound 2C in the form of a yellow solid.

Example 2D: (S)-2-amino-N-((3R,4S,5S)-l-((S)-2-((lR,2R)-3-(((l S,2R)-l- hydroxy- 1 -phenylpropan-2-yl)amino)- 1 -methoxy-2-methyl-3 -oxopropyl)pyrrolidin- 1 - yl)-3 -methoxy-5 -methyl- 1 -oxoheptan-4-yl)-N,3 -dimethylbutanamide

Compound 2C (300 mg, 0.36 mmol, 1.00 eq.) was dissolved in an inert atmosphere in ACN (2 mL) and piperidine (0.5 mL). The solution was left under agitation at ambient temperature overnight then evaporated to dryness under reduced pressure. The residue was purified on a silica column with a mixture of DCM and MeOH (1 : 100) to yield 150 mg (68 %) of compound 2D in the form of a white solid.

Example 2E: methyl 2-((tert-butoxycarbon l)amino)isonicotinate

Methyl 2-aminopyridine-4-carboxylate (2 g, 13.14 mmol, 1.00 equiv) was dissolved in tert-butanol (20 mL) after which di-tert-butyl dicarbonate (4.02 g, 18.42 mmol, 1.40 equiv) was added. The reaction mixture was agitated at 60°C overnight then the reaction was halted through the addition of an aqueous 1M NaHC0 3 solution (50 mL). The solid was recovered by filtration, washed with 50 mL of EtOH then dried in vacuo to yield 2.5 g (75 %) of compound 2E in the form of a white solid.

Example 2F: tert-butyl (4-(hydroxymethyl)pyridin-2-yl)carbamate

Compound 2E (2.5 g, 9.91 mmol, 1.00 equiv) and CaCl 2 (1.65 g) were dissolved in EtOH (30 mL). The solution was cooled to 0°C then NaBH 4 (1.13 g, 29.87 mmol, 3.01 equiv) was gradually added. The solution was left under agitation overnight at ambient temperature then the reaction was halted with the addition of water (50 mL). The mixture was extracted three times with 20 mL of EtOAc. The organic phases were combined, washed twice with 20 mL of NaCl (sat.) then dried over sodium sulfate, filtered and concentrated under reduced pressure to yield 2.0 g (90 %) of compound 2F in the form of a colourless solid. Example 2G: tert-butyl (4-formylpyridin-2-yl)carbamate

Compound 2F (2.5 g, 11.15 mmol, 1.00 equiv) was dissolved in DCE (25 mL) then 19.4 g (223.14 mmol, 20.02 equiv) of Mn0 2 were added. The mixture was left under agitation overnight at 70 °C then the solids were removed by filtering. The filtrate was evaporated to dryness to yield 1.4 g (57 %) of compound 2G in the form of a white solid.

Example 2H: benzyl (S)-2-(((2-((tert-butoxycarbonyl)amino)pyridin-4-yl) methyl) (methyl)amino)-3-methylbutanoate

Compound 2G (2.3 g, 10.35 mmol, 1.00 equiv) was dissolved in 25 mL of THF in the presence of compound 1ZC (2.93 g, 11.37 mmol, 1.10 equiv), DIEA (5.39 g, 41.71 mmol, 4.03 equiv) and NaBH(OAc) 3 (4.39 g, 20.71 mmol, 2.00 equiv). The reaction mixture was agitated for 6 hours at ambient temperature then neutralised with 60 mL of NaHC0 3 (sat.) and extracted 3 times with 20 mL of AcOEt. The organic phases were combined, washed twice with 20 mL of NaCl (sat.), dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of EtOAc and PE (1 : 15) to yield 2.7 g (61 %) of compound 2H in the form of a white solid.

Example 21: (S)-2-(((2-((tert-butoxycarbonyl)amino)pyridin-4-yl)methyl)

(methyl)amino)-3-methylbutanoic acid

Compound 2H (500 mg, 1.17 mmol, 1.00 equiv) was dissolved in 10 mL of AcOEt and 2 mL of methanol in the presence of Pd/C (250 mg), and hydrogenated for 3 hours at ambient temperature and atmospheric pressure. The reaction medium was filtered and concentrated under reduced pressure to yield 254 mg (64 %) of compound 21 in the form of a colourless solid.

Example 2J: tert-butyl (4-((3S,6S,9S,10R)-9-((S)-5ec-butyl)-10-(2-((S)-2- ((lR,2R)-3-(((l S,2R)-l -hydroxy-l-phenylpropan-2-yl)amino)-l-methoxy-2-methyl-3- oxopropyl)pyrrolidin-l-yl)-2-oxoethyl)-3,6-diisopropyl-2,8-d imethyl-4,7-dioxo-l l-oxa-

2,5,8-triazadodecyl)pyridin-2-yl) carbamate

Compound 2 J was prepared in similar manner to compound 1ZG from the amine 2D (85.2 mg, 0.14 mmol, 1.50 equiv), the acid 21 (31.7 mg, 0.09 mmol, 1.00 equiv), HATU (42.9 mg, 0.1 1 mmol, 1.20 equiv) and DIEA (36.7 mg, 0.28 mmol, 3.02 equiv) in DMF (3 mL). After evaporation to dryness, 100 mg of crude product were obtained in the form of a white solid.

Example 2: Compound 2 J (100 mg, 0.1 1 mmol, 1.00 equiv) was dissolved in 2 mL of DCM and 1 mL of TFA. The reaction was agitated for 1 hour at ambient temperature, then concentrated under reduced pressure. The residue (80 mg) was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep CI 8 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2489 UV Detector at 254 nm and 220 nm). Compound 2 was obtained with a yield of 6 % (6.3 mg) in the form of a white solid.

LC/MS/UV (Ascentis Express CI 8 column, 2.7 μηι, 4.6 x 100 mm; 40°C; 1.8mL/min, from 10 % to 95 % ACN in water (0.05 % TFA) in 6 minutes); ESI (C 45 H 73 N 7 0 7 , exact mass 823.56) m/z: 824.5 (MH + ) and 412.9 (M.2H + /2, 100 %), 3.21 min (99.2 %, 210 nm).

'H NMR (400MHZ, CD 3 OD, ppm): 5 (Presence of rotamers) 7.81 - 7.79 (m, 1 H); 7.39 - 7.29 (m, 5H); 6.61 - 6.59 (m, 2H); 4.84 - 4.52 (m, 1 H); 4.32 - 4.02 (m, 1 H); 3.90 - 2.98 (m, 10H); 2.90 - 2.78 (m, 1 H); 2.55 - 0.81 (m, 39H). Reference Example 3

methyl ((S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)- V,3-dimethyl-2-((S)-3- methyl-2-(methyl(pyridin-4-ylmethyl)amino)butanamido)butanam ido)-3- methoxy-5-methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2- methylpropanamido)-3-phenylpropanoate, trifluoroacetic acid

Compound ID (3 g, 10.44 mmol, 1.00 equiv) and methyl (S)-2-amino-3- phenylpropanoate (2.25 g, 12.55 mmol, 1.20 equiv) were dissolved in an inert atmosphere in DMF (40 mL). The solution was cooled to 0 °C then DEPC (1.67 mL, 1.05 equiv) and TEA (3.64 mL, 2.50 equiv) were added drop-wise. The reaction mixture was agitated 2 hours at 0 °C then at ambient temperature overnight. The reaction mixture was diluted with 100 mL of water and extracted three times with 50 mL EtOAc. The organic phases were combined, washed once with 100 mL of KHS0 4 (1 mol/L), once with 100 mL of NaHC0 3 (sat.), once with 100 mL of NaCl (sat.), then dried over sodium sulfate, filtered and concentrated under pressure to yield 4 g (85 %) of compound 3A in the form of a colourless oil.

Example 3B: 2,2,2-trifluoroacetate of methyl (S)-2-((2R,3R)-3-methoxy-2- methyl-3 -((S)-pyrrolidin-2-yl)propanamido)-3 -phenylpropanoate

Compound 3 A (5 g, 1 1.15 mmol, 1.00 equiv) was dissolved in an inert atmosphere in DCM (40 mL). TFA (25 mL) was added and the solution agitated for 2 hours. The reaction mixture was concentrated under reduced pressure to yield 8 g of compound 3B in the form of a yellow oil.

Example 3C: methyl (S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-((((9H- fluoren-9-yl)methoxy)carbonyl)amino)-N,3-dimethylbutanamido) -3-methoxy-5-methyl heptanoyl)pyrro lidin-2-yl)-3 -methoxy-2-methylpropanamido)-3 -phenylpropanoate

Compounds 3B (8.03 g, 17.36 mmol, 1.00 equiv) and 1W (9.1 g, 17.34 mmol, 1.00 equiv) were dissolved in an inert atmosphere in DCM (80 mL). The solution was cooled to 0 °C then DEPC (2.8 mL) and DIEA (12 mL) were added drop-wise. The reaction mixture was agitated for 2 hours at 0 °C then at ambient temperature overnight. The reaction mixture was diluted with 200 mL of water and extracted three times with 50 mL of DCM. The organic phases were combined, washed once with 50 mL of KHSO 4 (1 mol/L), once with 50 mL of NaHC0 3 (sat.), once with 50 mL of NaCl (sat.), then dried over sodium sulfate, filtered and concentrated under reduced pressure to yield 5 g (34 %) of compound 3C in the form of a yellow solid.

Example 3D: methyl (S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-amino-N,3- dimethylbutanamido)-3 -methoxy-5 -methylheptanoyl)pyrrolidin-2-yl)-3 -methoxy-2- methylpropanamido)-3-phenylpropanoate

Compound 3C (5.5 g, 6.43 mmol, 1.00 equiv) was dissolved in an inert atmosphere in a solution of tetrabutylammonium fluoride (TBAF, 2.61 g, 9.98 mmol, 1.55 quiv) in DMF (100 mL). The solution was agitated at ambient temperature for 2 hours then diluted with 100 mL of water and extracted three times with 50 mL of EtOAc. The organic phases were combined then dried over sodium sulfate, filtered and concentrated under reduced pressure to yield 3.3 g (81 %) of compound 3D in the form of a yellow solid.

Example 3E: benzyl (S)-3-methyl-2-(methyl(pyridin-4-ylmethyl)amino) butanoate

Pyridine-4-carbaldehyde (1 g, 9.34 mmol, 1.00 equiv) was dissolved in 10 mL of 1 ,2-dichloroethane (DCE) in the presence of compound 1ZC (2.9 g, 11.25 mmol, 1.21 equiv) and titanium isopropoxide (IV) (4.19 mL, 1.40 equiv). The mixture was agitated at ambient temperature for 30 minutes then 2.77 g of NaBH(OAc)3 (13.07 mmol, 1.40 equiv) were added. The reaction medium was left under agitation overnight then neutralised with 100 mL of water and the mixture extracted 3 times with 50 mL of AcOEt. The organic phases were combined and evaporated to dryness. The residue was purified on a silica column with a mixture of EtOAc and PE (1 :20) to yield 1.3 g (45 %) of compound 3E in the form of a colourless oil.

Example 3F: (S)-3-methyl-2-(methyl(pyridin-4-ylmethyl)amino)butanoic acid Compound 3E (800 mg, 2.56 mmol, 1.00 equiv) was dissolved in 30 mL of AcOEt in the presence of Pd/C (300 mg) and hydrogenated for 3 hours at ambient temperature and atmospheric pressure. The reaction medium was filtered and concentrated under reduced pressure. The residue was purified on a silica column with a mixture of DCM and MeOH ( 100 : 1 to 5 : 1 ) to yield 100 mg ( 18 %) of compound 3F in the form of a white solid.

Example 3: Compounds 3D (50 mg, 0.08 mmol, 1.00 equiv) and 3F (26.34 mg, 0.12 mmol, 1.50 equiv) were dissolved in 3 mL of DCM. The solution was cooled to 0 °C then 0.018 mL of DEPC and 0.0392 mL of DIE A were added. The reaction was agitated at 0°C for 2 hours then at ambient temperature overnight. The reaction medium was concentrated under reduced pressure and the residue (70 mg) was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep CI 8 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % of TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2545 UV Detector at 254 nm and 220 nm). Compound 3 was obtained with a yield of 27 % (20 mg) in the form of a white solid.

LC/MS/UV (Ascentis Express C18 column, 2.7 μιη, 4.6 x 100 mm; 40°C; 1.5 mL/min, 10 % to 95 % ACN in water (0.05 % TFA) in 8 minutes); ESI (C 46 H 72 N 6 O 8 , exact mass 836.5) m/z: 837.5 (MH + ) and 419.4 (M.2H + /2 (100 %)), 7.04 min (90.0 %, 210 nm).

1H NMR (400MHz, CD 3 OD, ppm): δ (Presence of rotamers) 8.76 - 8.74 (m, 2H); 8.53 - 8.48 (m, 0.4H, NHCO incomplete exchange); 8.29 - 8.15 (m, 0.8H, NHCO incomplete exchange); 8.01 (s, 2H), 7.31 - 7.22 (m, 5H), 4.88 - 4.68 (m, 3H); 4.31 - 4.07 (m, 2H); 3.94 - 2.90 (m, 18H); 2.55 - 0.86 (m, 38H).

Reference Example 4

(S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)- V,3-dimethyl-2-((S)-3-methyl-2- (methyl(pyridin-4-ylmethyl)amino)butanamido)butanamido)-3-me thoxy-5- methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropanami do)-3- phenylpropanoic acid, trifluoroacetic acid

Example 4: Compound 3 (100 mg, 0.11 mmol, 1.00 equiv) was dissolved in a mixture of water (5 mL), ACN (5 mL) and piperidine (2.5 mL). The reaction mixture was left under agitation overnight then concentrated under reduced pressure. The residue was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep CI 8 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2545 UV Detector at 254 nm and 220 nm), to yield 20 mg (20 %) of compound 4 in the form of a white solid.

LC/MS/UV (Ascentis Express C18 column, 2.7 μιη, 4.6 x 100 mm; 40°C;

1.5 mL/min, 10 % to 95 % ACN in water (0.05 % TFA) in 8 minutes); ESI (C 45 H 70 N 6 O 8 , exact mass 822.5) m/z: 823.5 (MH + ) and 412.4 (M.2H + /2, 100 %), 6.84 min (89.1 %, 210 nm).

1H NMR (400MHz, CD 3 OD, ppm): δ (Presence of rotamers) 8.79 - 8.78 (m, 2H); 8.09 (m, 2H); 7.30 - 7.21 (m, 5H); 4.80 - 4.80 (m, 1H), 4.36 - 0.87 (m, 58H).

Reference Example 6

methyl (S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-((S)-2-((3-amin opropyl) (methyl)amino)-3-methylbutanamido)- V,3-dimethylbutanamido)-3-methoxy-5- methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropanami do)-3- phenylpropanoate, bis trifluoroacetic acid

Example 6A: methyl (2S)-2-[(2R)-2-[(R)-[(2S)-l-[(3R,4S,5S)-4-[(2S)-2-[(2S)- 2- [(3 -[ [(tert-butoxy)carbonyl] amino]propyl)(methyl)amino] -3-methyl butanamido] -N,3 - dimethylbutanamido]-3-methoxy-5-methylheptanoyl]pyrrolidin-2 -yl](methoxy)methyl] propanamido] -3 -phenylpropanoate

Compound 3D (157.5 mg, 0.25 mmol, 1.00 equiv) was dissolved at 0°C in an inert atmosphere in 3 mL of DCM in the presence of carboxylic acid 1ZF (78.7 mg, 0.27 mmol, 1.10 equiv), DEPC (46 μΐ) and DIEA (124 μΐ). The reaction mixture was agitated 2 hours at low temperature and the cold bath was then removed and agitation continued for 4 hours. It was then concentrated under reduced pressure to yield 200 mg of compound 6A in the form of a crude yellow oil. It was used as such in the following step.

Example 6: Compound 6A (200 mg, 0.22 mmol, 1.00 equiv) was dissolved in an inert atmosphere at 0°C in 2 mL of DCM. TFA (1 mL) was added drop-wise and the cold bath removed. The reaction mixture was agitated 1 hour at ambient temperature then concentrated under reduced pressure. The residue was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep CI 8 OBD column, 5 μηι, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2489 UV Detector at 254 nm and 220 nm), to yield 60 mg (26 %, yield in 2 steps) of compound 6 in the form of a white solid.

LC/MS/UV (Zorbax Eclipse Plus C8, 3.5 μη , 4.6 x 150 mm; 1 mL/min, 40°C, 30 to 80 % methanol in water (0.1 % H 3 P0 4 ) in 18 minutes); ESI (C 43 H 74 N 6 0 8 , exact mass 802.56) m/z: 804 (MH + ); 1 1.50 min (91.5 %, 210 nm).

]H NMR (300MHz, CD 3 OD, ppm): δ (Presence of rotamers) 8.52 (d, 0.3H,

NHCO incomplete exchange); 8.25 (d, 0.5H, NHCO incomplete exchange); 7.30-7.22 (m, 5H); 4.9^.6 (m, 3H); 4.2-4.0 (m, 1 H); 4.0-0.86 (m, 61 H). Reference Example 7

(S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-((S)-2-((3-amin opropyl) (methyl)amino)-3-methylbutanamido)- V,3-dimethylbutanamido)-3-methoxy-5- methylheptanoyl) pyrrolidin-2-yl)-3-methoxy-2-methylpropanamido)-3- lylpropanoic acid, bis trifluoroacetic acid

Example 7: Compound 6 (70 mg, 0.08 mmol, 1.00 equiv) was dissolved in a mixture of water (5 mL), ACN (2.5 mL) and piperidine (5 mL). The reaction mixture was left under agitation overnight at ambient temperature, then concentrated under reduced pressure. The residue was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep C18 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; UV Waters 2489 UV Detector at 254 nm and 220 nm), to yield 14.6 mg (21 %) of compound 7 in the form of a white solid.

LC/MS/UV (Ascentis Express C18, 2.7 μιη, 4.6 x 100 mm; 1.5 mL/min, 40°C, 0 to 80 % methanol in water (0.05 % TFA) in 8 minutes); ESI (C 42 H 72 N 6 O 8 , exact mass 788.54) m/z: 790 (MH + ), 5.71 min (96.83 %, 210 nm).

1H NMR (300MHz, CD 3 OD, ppm): δ (Presence of rotamers) 8.42 (d, 0.3H, NHCO incomplete exchange); 8.15 (d, 0.2H, NHCO incomplete exchange); 7.31-7.21 (m, 5H); 4.9-4.6 (m, 3H); 4.25-4.0 (m, 1H); 4.0-0.86 (m, 59H).

Example 11

(S)- V-((3R,4S,5S)-3-methoxy-l-((S)-2-((lR,2R)-l-methoxy-2-methyl -3-oxo-3- (((S)-2-phenyl-l-(thiazol-2-yl)ethyl)amino)propyl)pyrrolidin -l-yl)-5-methyl-l- oxoheptan-4-yl)- V,3-dimethyl-2-((S)-3-methyl-2-(methyl(4-

(methylamino)phenethyl)amino) butanamido)butanamide, trifluoroacetic acid

Example 11 A: tert-butyl N-[4-(2-hydroxyethyl)phenyl] carbamate

Compound 11A was obtained with a yield of 75 % after reaction at ambient temperature of 2-(4-aminophenyl)ethanol with BOC 2 0 in THF.

Example 11B: tert-butyl N-[4-(2-oxoethyl)phenyl]carbamate

Compound 11A (2.5 g, 10.5 mmol, 1.00 equiv) was dissolved in 25 mL of DCM then cooled to -78°C. A Dess-Martin Periodinane solution (DMP, 6.71 g, 15.8 mmol, 1.5 equiv) in DCM (10 mL) was added drop-wise. The cold bath was removed and agitation continued for 1 hour at ambient temperature. The reaction was neutralised with 60 mL of a 50/50 mixture of sodium bicarbonate-saturated aqueous solution and Na 2 S 2 03-saturated aqueous solution. The resulting solution was extracted 3 times with 30 mL of EtOAc. The organic phases were combined, washed twice with NaCl- saturated aqueous solution, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified on silica gel (EtOAc/PE 1/15) to yield 1.0 g (40 %) of compound 11B in the form of a pale yellow solid.

Example 11C: benzyl (2S)-2-[[2-(4-[[(tert-butoxy)carbonyl]amino]phenyl) ethyl](methyl)amino]-3-methylbutanoate

Compound 1ZC (3.5 g, 13.6 mmol, 1.1 equiv) was dissolved in THF (30 mL) in the presence of DIEA (6.4 g, 49.7 mmol, 4.0 equiv), aldehyde 11B (2.9 g, 12.3 mmol, 1.0 equiv) and sodium triacetoxyborohydride (5.23 g, 49.7 mmol, 2.0 equiv). The reaction mixture was left under agitation overnight at ambient temperature, then neutralised with 60 mL of sodium bicarbonate-saturated solution. The resulting solution was extracted 3 times with 30 mL EtOAc. The organic phases were combined, washed twice with NaCl-saturated aqueous solution, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified on silica gel (EtOAc/PE 1 :20) to yield 3.7 g (68 %) of compound 11C in the form of a yellow oil.

Example IIP: (2S)-2-[[2-(4-[[(tert-butoxy)carbonyl]amino]phenyl)ethyl] (methyl)amino] -3 -methylbutanoic acid

Compound 11C (2 g, 4.5 mmol, 1 equiv) was dissolved in 10 mL of methanol in the presence of Pd/C (2 g) and hydrogenated for 2 hours at normal temperature and pressure. The reaction medium was filtered and concentrated under reduced pressure to yield 1.2 g (75 %) of compound 11D in the form of a yellow oil.

Example HE: (2S)-2-[[2-(4-[[(tert-butoxy)carbonyl](methyl)amino]phenyl) ethyl] (methyl) amino]-3-methylbutanoic acid

Compound 11D (1.2 g, 3.4 mmol, 1.00 equiv) was dissolved in an inert atmosphere in THF (20 mL). The reaction medium was cooled with an ice bath after which NaH (60 % in oil, 549 mg, 13.7 mmol, 4.0 equiv) was added in portions, followed by iodomethane (4.9 g, 34 mmol, 10 equiv). The reaction was left under agitation overnight at ambient teperature, then neutralised with water and washed with 100 mL of EtOAc. The pH of the aqueous solution was adjusted to 6-7 with IN HC1. This aqueous solution was extracted 3 times with 100 mL of EtOAc. The organic phases were combined, dried over sodium sulfate, filtered and concentrated to yield 800 mg (64 %) of compound HE in the form of a yellow solid. Example 11F: tert-butyl N-[4-(2-[[(l S)-l-[[(l S)-l-[[(3R,4S,5S)-3-methoxy-l- [(2S)-2- [( 1 R,2R)- 1 -methoxy-2-methyl-2-[ [( 1 S)-2-phenyl- 1 -( 1 ,3-thiazol-2-yl) ethyl] carbamoyl] ethyl]pyrrolidin- 1 -yl] -5 -methyl- 1 -oxoheptan-4yl] (methyl)carbamoyl] - 2-methylpropyl] carbamoyl] -2 -methylpropyl] (methyl)amino] ethyl)phenyl] -N-methyl carbamate

Compound 11F was prepared in similar manner to compound 6A from the amine 1Y (150 mg, 0.22 mmol, 1.2 equiv) and the acid HE (70 mg, 0.19 mmol, 1.0 equiv). After purification on silica gel (EtOAc/PE 1 : 1) 100 mg (52 %) of desired product were obtained in the form of a pale yellow solid.

Example 11: Compound 11 was prepared in the same manner as for compound 1 from the intermediate 11F (100 mg, 0.1 mmol). The residue was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep CI 8 OBD column, 5 μη , 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2489 UV Detector at 254 nm and 220 nm). Compound 11 was obtained with a yield of 39 % (39.7 mg) in the form of a white solid.

LC/MS/UV (Eclipse Plus C8, 3.5 pm, 4.6 x 150 mm; 1 mL/min, 40°C, 50 to 95 % methanol in water (0.05 % TFA) in 18 minutes); ESI (C 5 oH7 7 N 7 0 6 S, exact mass 903.57) m/z: 904.5 (MH + ), 7.53 min (93.68 %, 254 nm).

] H NMR (300MHz, CD 3 OD, ppm): δ (Presence of rotamers) 8.84 (d, 0.5H, NHCO incomplete exchange); 8.7-8.5 (m, 0.9H, NHCO incomplete exchange); 7.76- 7.73 (m, 1H); 7.55 - 7.4 (m, 1H); 7.28-7.22 (m, 7H); 7.08-7.05 (m, 2H); 5.51-5.72 (m, 1H); 4.9^.80 (m, 2H); 4.3-0.7 (m, 60H). Example 12

methyl (S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)- V,3-dimethyl-2-((S)-3- methyl-2-(methyl(4-(methylamino)phenethyl)amino)butanamido)b utanamido)-3- methoxy-5-methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2- methylpropanamido)-3- phenylpropanoate, trifluoroacetic acid

Example 12: In the same manner as for the final phases in the synthesis of compound 1, compound 12 was prepared in two steps from the amine 3D (118 mg, 0.19 mmol) and the acid HE (82 mg, 0.22 mmol). The final residue was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep CI 8 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2489 UV Detector at 254 nm and 220 nm). Compound 12 was obtained with a yield of 7 % (13.7 mg) in the form of a white solid.

LC/MS/UV (Eclipse Plus C8, 3.5 μιη, 4.6 x 150 mm; 1 mL/min, 40°C, 40 to 95

% methanol in water (0.05 % TFA) in 18 minutes); ESI (C 4 H 78 N 6 O 8 , exact mass 878.59) m/z: 879.7 (MH + ), 10.07 min (90.6 %, 254 nm).

1HNMR (300MHz, CD 3 OD, ppm): δ (Presence of rotamers) 7.40 (se, 2H); 7.38-7.22 (m, 7H); 4.95-4.7 (m, 3H); 4.2-4.0 (m, 1H); 3.9-0.86 (m, 62H).

Example 13

(S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)- V,3-dimethyl-2-((S)-3-methyl-2- (methyl(4-(methylamino)phenethyl)amino)butanamido)butanamido )-3-methoxy- 5-methyl heptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropanamido)-3- phenylpropanoic acid, trifluoroacetic acid

Example 13: Compound 13 was prepared in the same manner as for compound 7 from compound 12 (100 mg, 0.10 mmol). The residue was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep CI 8 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2489 UV Detector at 254 nm and 220 nm). Compound 13 was obtained with a yield of 20 % (20 mg) in the form of a white solid.

LC/MS/UV (Ascentis Express CI 8, 2.7 μιη, 4.6 x 100 mm; 1.5 mL/min, 40°C, 10 to 95 % methanol in water (0.05 % TFA) in 8 minutes); ESI exact mass 864.57) m/z: 865.6 (MH + ), 6.05 min (90.9 %, 210 nm).

1H NMR: (300MHz, CD 3 OD, ppm): δ (Presence ofrotamers) 7.32-7.19 (m, 9H); 4.9-4.65 (m, 3H); 4.2-4.0 (m, 1H); 3.9-0.86 (m, 59H). Example 14

(S)-2-((S)-2-((3-aminobenzyl)(methyl)amino)-3-methylbutanami do)- V- ((3R,4S,5S)-3-methoxy-l-((S)-2-((lR,2R)-l-methoxy-2-methyl-3 -oxo-3-(((S)- 2-phenyl-l-(thiazol-2-yl)ethyl)amino)propyl)pyrrolidin-l-yl) -5-methyl-l- oxoheptan-4-yl)- V,3-dimethylbutanamide, trifluoroacetic acid

Example 14A: tert-butyl (3-(hydroxymethyl)phenyl) carbamate

(3-aminophenyl)methanol (3 g, 24.36 mmol, 1.00 equiv) was dissolved in THF (60 mL) after which di-tert-butyl dicarbonate (6.38 g, 29.23 mmol, 1.20 equiv) was then added. The reaction mixture was left under agitation overnight at ambient temperature and the reaction was then diluted by adding 200 mL of water. The product was extracted 3 times with 100 mL of AcOEt and the organic phases were then recombined, dried over sodium sulfate, filtered and concentrated under reduced pressure to yield the crude product (13.85 g of compound 14A) in the form of a yellow oil.

Exam le 14B rt-but l 3-form l hen l carbamate

Compound 14A (13.8 g, 61.81 mmol, 1.00 equiv) was dissolved in DCE (400 mL) and Mn0 2 (54 g, 621.14 mmol, 10.05 equiv) was then added. The mixture was left under agitation at ambient temperature for 3 days after which the solids were removed by filtering. The filtrate was evaporated to dryness and the residue was purified on a silica column with a mixture of EtOAc and PE (1 :30) to yield 3 g (22 %) of compound 14B in the form of a white solid.

Example 14C: benzyl (S)-2-((3-((tert-butoxycarbonyl)amino)benzyl) (methyl) amino)-3 -methylbutano

Compound 14B (1 g, 4.52 mmol, 1.00 equiv) was dissolved in 20 mL of THF in the presence of compound 1ZC (1.16 g, 4.50 mmol, 1.00 equiv), DIEA (3 mL) and NaBH(OAc) 3 (1.92 g, 9.06 mmol, 2.01 equiv). The reaction mixture was left under agitation overnight at ambient temperature and then neutralised with 100 mL of water and extracted 3 times with 50 mL of AcOEt. The organic phases were combined, dried over sodium sulfate, filtered and concentrated. The residue was purified on a silica column with a mixture of EtOAc and PE (1 :50) to yield 1.9 g (99 %) of compound 14C in the form of a white solid.

Exemple 14D: (S)-2-((3-((iert-butoxycarbonyl)amino)benzyl)(methyl)amino)- 3-methylbutanoic acid

Compound 14C (1 g, 2.34 mmol, 1.00 equiv) was dissolved in 30 mL of AcOEt and 4 mL of methanol in the presence of Pd/C (400 mg) and hydrogenated for 1 hour at ambient temperature and atmospheric pressure. The reaction medium was filtered and concentrated under reduced pressure to yield 680 mg (86 %) of compound 14D in the form of a white solid.

Example 14E: tert-butyl (3-((3S,6S,9S,10R)-9-((S)-sec-butyl)-3,6-diisopropyl- 10-(2-((S)-2-(( 1 R,2R)- 1 -methoxy-2-methyl-3 -oxo-3 -(((S)-2-phenyl- 1 -(thiazol-2-yl) ethyl)amino)propyl)pyrrolidin-l -yl)-2-oxoethyl)-2,8-dimethyl-4,7-dioxo-l l-

amine 1Y (100 mg, 0.15 mmol, 1.00 equiv), the acid 14D (102.27 mg, 0.30 mmol, 2.00 equiv), DEPC (0.053 mL) and DIEA (0.046 mL) in DCM (3 mL). The crude product (80 mg) was purified on a silica column with a mixture of EtOAc and PE (1 : 1) to yield 100 mg (67 %) of compound 14E in the form of a pale yellow solid.

Example 14: Compound 14 was synthesised in the same manner as for compound 2 from the intermediate 14E (100 mg, 0.10 mmol, 1.00 equiv). The crude product (80 mg) was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep CI 8 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2545 UV Detecctor at 254 nm and 220 nm). Compound 14 was obtained with a yield of 10 % (10 mg) in the form of a white solid.

LC/MS/UV (Eclipse plus C8 column, 3.5 μηι, 4.6 x 150 mm; 40°C; 1.0 mL / min, 40 % to 95 % MeOH in water (0.05 % TFA) in 18 minutes); ESI (C 48 H 73 N 7 0 6 S, exact mass 875.5) m/z: 876.5 (MH + ) and 438.9 (M.2H + /2, 100 %), 1 1.35 min (95.6 %, 210 nm).

1H NMR (400MHz, CD 3 OD, ppm): δ (Presence of rotamers) 8.92 - 8.86 (m, 0.4H, NH incomplete exchange); 8.70 - 8.54 (m, 0.6H, NH incomplete exchange); 7.88 - 7.78 (m, 1H); 7.60 - 7.50 (m, 1H); 7.45 - 6.97 (m, 9H); 5.80 - 5.65 (m, 1H); 4.85 - 4.70 (m, 1H); 4.40 - 0.80 (m, 56H).

Example 15

methyl (S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-((S)-2-((3-amin obenzyl) (methyl)amino)-3-methylbutanamido)-iV,3-dimethylbutanamido)- 3-methoxy-5- methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropanami do)-3- phenylpropanoate, trifluoroacetic acid

Example 15A: methyl (S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)^-((S)-2-((S)- 2-((3-((/eri-butoxycarbonyl)amino)benzyl)(methyl)amino)-3-me thylbutanamido)- N,3-dimethylbutanamido)-3-methoxy-5-methylheptanoyl)pyrrolid in-2-yl)-3- methoxy-2-methylpropanamido)-3-phenylpropanoate

Compound 15A was synthesised in the same manner as for compound 3 from the amine 3D (200 mg, 0.32 mmol, 1.00 equiv), the acid 14D (212.6 mg, 0.63 mmol, 2.00 equiv), DEPC (0.1103 mL) and DIEA (0.157 mL, 3.00 equiv) in DCM (5 mL). The crude product was purified on a silica column with a mixture of EtOAc and PE (1 : 1) to yield 200 mg (67 %) of compound 15A in the form of a yellow solid.

Example 15: Compound 15 was synthesised in the same manner as for compound 2 from the intermediate 15A (200 mg, 0.21 mmol, 1.00 equiv). The crude product was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep CI 8 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters UV Detector 2545 at 254 nm and 220 nm). Compound 15 was obtained with a yield of 19 % (38.6 mg) in the form of a white solid.

LC/MS/UV (Ascentis Express C18 column, 2.7 μιη, 4.6 x 100 mm; 40°C; 1.5 mL/min, 10 % to 95 % MeOH in water (0.05 % TFA) in 8 minutes); ESI (C 47 H 74 N 6 O 8 , exact mass 850.5) m/z: 851.5 (MH + ) and 426.4 (M.2H + /2, 100 %), 6.61 min (91.1 %, 210 nm).

1H NMR (400MHz, CD 3 OD, ppm): δ (Presence of rotamers) 7.53 - 7.42 (m, 1H); 7.35 - 7.18 (m, 8H); 4.88 - 4.79 (m, 2H); 4.42 - 4.00 (m, 3H); 3.93 - 2.71 (m, 22H); 2.61 - 0.81 (m, 33H).

Examples 19 and 20

Compounds 19 and 20 were prepared in the same manner as for compound 1, from the amines 1Y and 1ZC and corresponding aldehydes.

The tert-butyl 4-formylphenyl carbonate involved in the preparation of compound 19 was prepared in a single step as follows: 4-hydroxybenzaldehyde (2.5 g, 20.5 mmol, 1.0 equiv) was dissolved in an inert atmosphere in THF (20 mL) in the presence of 18-crown-6 (0.25 g) and potassium carbonate (5 g). The reaction mixture was cooled to 0°C and the di-tert-butyl dicarbonate (5.8 g, 26.58 mmol, 1.30 equiv) was then added. Agitation was continued for 1 hour at low temperature after which the reaction was neutralised with 30 mL of water. The resulting solution was extracted three times with 200 mL of EtOAc. The organic phases were combined, dried over anhydrous sodium sulfate filtered and concentrated under reduced pressure. The residue was purified on silica gel (EtOAc/PE 1 : 10) and yielded 4.2 g (92 %) of tert-butyl 4- formylphenyl carbonate in the form of a pale yellow solid.

The 4-nitrobenzaldehyde involved in the preparation of compound 20 was commercial.

The synthesis of compound 20 was completed by reducing the nitro group. This was performed as follows: (2S)-N-[(3R,4S,5S)-l-[(2S)-2-[(lR,2R)-2-[[(l S,2R)-l- hydroxy- 1 -phenylpropan-2-yl] carbamoyl] - 1 -methoxy-2-methylethylpyrrolidin- 1 -yl] -3 - methoxy-5 -methyl- 1 -oxoheptan-4-yl] -N,3 -dimethyl-2- [(2S)-3-methyl-2- [methyl[(4- nitrophenyl)methyl]amino]butanamido]butanamide (40 mg, 0.05 mmol, 1.0 equiv) was dissolved in 15 mL of ethanol. Dihydrated tin chloride (II) (317 mg, 1.4 mmol, 30 equiv) was added and the solution left under agitation for 3 days at ambient temperature. The reaction was neutralised with 50 mL of water, then extracted three times with 50 mL of EtOAc. The organic phases were combined, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to yield compound 20 in the crude state.

Name X R Purity* Quantity

(S)-2-((S)-2-((4-hydroxybenzyl)

(methyl)amino)-3 -methyl

butanamido)-N-((3R,4S,5S)-3- methoxy- 1 -((S)-2-((l R,2R)- 1 - methoxy-2-methyl-3-oxo-3-(((S)-2- 1 93.2 % 21.6 mg phenyl- 1 -(thiazol-2-yl)ethyl)amino)

propyl)pyrrolidin- 1 -yl)-5 -methyl- 1 - oxoheptan-4-yl)-N,3-dimethyl

butanamide, trifluoroacetic acid Name X R Purity* Quantity

(S)-2-((S)-2-((4-aminobenzyl)

(methyl)amino)-3 -methyl

butanamido)-N-((3R,4S,5S)-3- methoxy- 1 -((S)-2-((l R,2R)- 1 - methoxy-2-methyl-3-oxo-3-(((S)-2- 1 96.7 % 21.1 mg phenyl- 1 -(thiazol-2-yl)ethyl)amino)

propyl)pyrrolidin- 1 -yl)-5 -methyl- 1 - oxoheptan-4-yl)-N,3-dimethyl

butanamide, trifluoroacetic acid

* The compounds were purified by preparative H PLC (Pre-HPLC-001

SHIMADZU, SunFire Prep C18 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2489 UV Detector at 254 nm and 220 nm), to give the corresponding TFA salts in the form of white solids.

Characterization of the end products: Compound 19 LC/MS/UV ESI: (C 48 H 72 N 6 O 7 S, exact mass 876.52) m/z 877 (MH + ), 439 [100 %, (M.2H + )/2]; UV: RT = 1.76 min (93.2 %, 220 nm). Compound 20 1H NMR: (400MHz, CD 3 OD, ppm): δ (Presence of rotamers) 7.85-7.80 (m, 1H); 7.6-7.5 (m, 1H); 7.4-7.15 (m, 5H); 7.1-7.05 (m, 2H); 6.73-6.70 (m, 2H); 5.8-5.55 (m, 1H); 5.0-4.7 (m, 2H); 4.25-4.05 (m, 1H); 4.0-0.8 (m, 54H). LC/MS/UV ESI: (C 48 H 73 N 7 O 7 S, exact mass 875.53) m/z 876 (MH + ), 439 [75 %, (M.2H + )/2]; UV: RT = 4.83 min (96.8 %, 254 nm). 1H NMR (400MHz, CD3OD, ppm): δ (Presence of rotamers) 7.85-7.80 (m, 1H); 7.6-7.5 (m, 1H); 7.4-7.1 (m, 7H); 6.76-6.72 (m, 2H); 5.8-5.55 (m, 1H); 4.9-4.65 (m, 2H); 4.25-4.05 (m, 1H); 4.0-0.8 (m, 54H). Examples 23 and 24

Compounds 23 and 24 were prepared in the same manner as for compounds 19 20, replacing the amine 1Y by the amine 2D.

* The compounds were purified by preparative HPLC (Pre-HPLC-001

SHIMADZU, SunFire Prep C18 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2489 UV Detector at 254 nm and 220 nm), to give the corresponding TFA salts in the form of white solids.

Characterization of the end products: Compound 23 LC/MS/UV (ESI) (C 4 6H73N 5 0 8 , exact mass 823.55) m/z 824 (MH + ), 846 (MNa + ), 413 (100 %, (M.2H + )/2); UV: 4.76 min (98.5 %, 215 nm). 1H NMR (400MHz, CDC1 3 , ppm): δ (Presence of rotamers) 7.5-7.2 (m, 5H); 7.9-7.75 (m, 2H); 5.5-5.3 (m, 1H); 4.9-4.6 (m, 2H); 4.55-4.15 (m, 4H); 4.0-0.8 (m, 55H). Compound 24 LC/MS/UV (ESI) (C 4 6H 74 N 6 0 7 , exact mass 822.56) m/z 823 (MH + ), 845 (MNa + ), 861 (MK + ); UV: 3.68 min (99.15 %, 254 nm). 1H NMR (400MHz, CD 3 OD, ppm): δ (Presence of rotamers) 8.0-7.7 (m, 0.5H, NHCO incomplete exchange); 7.5-7.0 (m, 7H); 6.75-6.65 (m, 2H); 4.85-4.5 (m, 2H); 4.4-4.05 (m, 2H); 4.0-0.8 (m, 56H).

Example 27

methyl (S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-((S)-2-((4- hydroxyphenethyl)(methyl)amino)-3-methylbutanamido)-/V,3- dimethylbutanamido)-3-methoxy-5-methylheptanoyl)pyrrolidin-2 -yl)-3- methoxy-2-methylpropanamido)-3- phenylpropanoate, trifluoroacetic acid

Example 27: Compound 27 was prepared in the same manner as for compound

3 from the amine 3D (70 mg, 0.11 mmol, 1.00 equiv), the acid 49C (55.5 mg, 0.22 mmol, 2.00 equiv), DEPC (0.034 mL, 2.00 equiv) and DIEA (0.055 mL, 3.00 equiv) in DCM (3 mL). The crude product was purified by preparative HPLC (Pre- HPLC-001 SHIMADZU, SunFire Prep C18 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 45 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2545 UV Detector at 254 nm and 220 nm). Compound 27 was obtained with a yield of 3 % (2.9 mg) in the form of a white solid. LC/MS/UV (Eclipse Plus C8 column, 3.5 pm, 4.6 x 150 mm; 40°C; 1.5 mL/min, 10 % to 95 % MeOH in water (0.05 % TFA) in 8 minutes); ESI (C 48 H 75 N 5 0 9 , exact mass 866.56) m/z: 866.5 (MH + ) and 433.9 (M.2H + /2, 100 %), 6.61 min (89.1 %, 210 ran).

1H NMR (400MHz, CD 3 OD, ;?pm): δ (Presence of rotamers) 8.70 - 8.49 (m, 0.9 H, NH/OH incomplete exchange); 8.30 - 8.22 (m, 0.3H, NH incomplete exchange); 7.36 - 7.02 (m, 7H); 6.86 - 6.62 (m, 2H); 4.82 - 4.69 (m, 2H); 4.20 - 4.03 (m, 1H); 3.91 - 3.33 (m, 12H); 3.30 - 2.90 (m, 17H); 2.55 - 0.80 (m, 35H).

Example 28

(S)-2-((S)-2--((3-aminobenzyl)(methyl)amino)-3-methylbutanam ido)- V- ((3R,4S,5S)-l-((S)-2-((lR,2R)-3-(((lS,2R)-l-hydroxy-l-phenyl propan-2- yl)amino)-l-methoxy-2-methyl-3-oxopropyl)pyrrolidin-l-yl)-3- methoxy--5- methyl— l-oxoheptan- — yl)- N,3— dimethylbutanamide, trifluoroacetic acid

Example 28A: teri-butyl (3-((3S,6S,9S,10R)-9-((S)-5ec-butyl)-10-(2-((S)- 2-(( 1 R,2R)-3-((( 1 S,2R)-1 -hydroxy-1 -phenylpropan-2-yl)amino)-l -methoxy-2- methyl-3-oxopropyl)pyrrolidin-l-yl)-2-oxoethyl)-3,6-diisopro pyl-2,8-dimethyl- 4,7-dioxo-l l-oxa-2,5,8-triazadodecyl)phenyl)carbamate

amine 2D (100 mg, 0.17 mmol, 1.00 equiv), the acid 14D (1 11.25 mg, 0.33 mmol, 2.00 equiv), DEPC (0.058 mL) and DIEA (0.05 mL) in DCM (3 mL). The residue was purified on a silica column with a mixture of EtOAc and hexane (1 : 1) to yield 100 mg

(66 %) of compound 28A in the form of a white solid.

Example 28: Compound 28 was synthesised in the same manner as for compound 2 from the intermediate 28A (100 mg, 0.11 mmol, 1.00 equiv). The crude product (80 mg) was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU,

SunFire Prep C18 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to

100 % ACN in 2 minutes; Waters 2545 UV Detector at 254 nm and 220 nm).

Compound 28 was obtained with a yield of 20 % (20 mg) in the form of a white solid.

LC/MS/UV (Ascentis Express C18 column, 2.7 μιη, 4.6 x 100 mm; 40°C;

1.5 mL/min, 10 % to 95 % MeOH in water (0.05 % TFA) in 8 minutes);

ESI (C 46 H 74 N 6 O 7 , exact mass 822.56) m/z: 823.5 (MH + ) and 412.4 (M.2H + /2, 100 %),

12.45 min (87.2 %, 210 nm).

1H NMR: (400MHz, CD 3 OD, ppm): δ (Presence of rotamers) 7.47 - 7.20 (m, 5H); 7.10 - 7.01 (m, 1H); 6.80 - 6.56 (m, 3H); 4.82 - 4.52 (m, 3H); 4.33 - 4.03 (m, 2H);

3.91 - 3.82 (m, 0.5H); 3.75 - 3.35 (m, 9.5H); 3.28 - 3.10 (m, 2H); 2.79 - 2.90 (m, 1H);

2.60 - 2.40 (m, 2H); 2.30 - 0.80 (m, 40H).

Example 29

(S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-((S)-2-((3-amin obenzyl)

(methyl)amino)-3-methylbutanamido)- V,3-dimethylbutanamido)-3-methoxy-5- methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropanami do)-3- phenylpropanoic acid, trifluoroacetic acid

Example 29: Compound 15 (100 mg, 0.10 mmol, 1.00 equiv) was dissolved in a mixture of water (5 mL), ACN (5 mL) and piperidine (2.5 mL). The reaction mixture was left under agitation overnight at ambient temperature and then concentrated under reduced pressure. The residue was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, SunFire Prep C18 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2545 UV Detector at 254 nm and 220 nm), to yield 20 mg (20 %) of compound 29 in the form of a white solid.

LC/MS/UV (Eclipse Plus C8 column, 3.5 μιη, 4.6 x 150 mm; 40°C; 1.0 mL/min, 40 % to 95 % MeOH in water (0.05 % TFA) in 18 minutes); ESI (C 46 H 72 N 6 O 8 , exact mass 836.54) m/z: 837.5 (MH + ) and 419.4 (M.2H + /2, 100 %), 10.61 min (92.5 %, 210 nm).

1H NMR: (400MHz, CD 3 OD, ppm): δ (Presence of rotamers) 7.38 - 7.15 (m, 6H);

7.00 - 6.99 (m, 3H); 4.85 - 4.68 (m, 2H); 4.37 - 3.38 (m, 11H); 3.31 - 2.70 (m, 8H); 2.60 - 0.82 (m, 35H).

Example 49

(S)-2-((S)-2-((4-hydroxyphenethyl)(mthyl)amino)-3-methylbuta namido)- V- ((3R,4S,5S)-3-mthoxy-l-((S)-2-((lR,2R)-l-methoxy-2-methyl-3- oxo-3-(((S)-2- phenyl-l-(thiazol-2-yl)ethyl)amino)propyl)pyrrolidin-l-yl)-5 -methyl-l- oxohe tan-4-yl)- V,3-dimethylbutanamide, trifluoroacetic acid

4-(2-hydroxyethyl)phenol (4 g, 28.95 mmol, 1.00 quiv) was dissolved in DMSO (32 mL) and TEA (8.8 mL, 2.20 equiv) was then added dropwise. A solution of S0 3 .Py (10 g, 2.20 equiv) in DMSO (36 mL) was added and the mixture was left under agitation overnight at ambient temperature. The reaction mixture was neutralised with 250 mL of water and extracted 3 times with 100 mL of AcOEt. The organic phases were combined, washed 5 times with water (100 mL) then twice with 150 mL of NaCl (sat.), dried over sodium sulfate, filtered and concentrated. The residue was purified on silica gel (EtOAc/PE (1 : 10) to yield 1 g (25 %) of compound 49A in the form of a colourless oil.

Example 49B: benzyl (S)-2-((4-hydroxyphenethyl)(methyl)amino)-3 -methyl butanoa

Compound 49B was synthesised in the same manner as for compound 14C from the amine 1ZC (1.5 g, 5.82 mmol, 0.99 equiv), the aldehyde 49A (800 mg, 5.88 mmol, 1.00 equiv), NaBH(OAc) 3 (2.7 g, 12.74 mmol, 2.17 equiv) and DIEA (4.23 mL) in THF (25 mL). The reaction mixture was neutralised with 50 mL of water and extracted 3 times with 50 mL of AcOEt. The organic phases were combined, dried over sodium sulfate, filtered and concentrated. The residue was purified on silica gel (EtOAc/PE (1 :10) to yield 600 mg (37 %) of compound 49B in the form of a white solid.

Example 49C: (S)-2-((4-hydroxyphenethyl)(methyl)amino)-3-methylbutanoic acid

Compound 49B (0.5 g, 1.46 mmol, 1.00 equiv) was dissolved in 40 mL of MeOH in the presence of Pd/C (250 mg) and hydrogenated for 3 hours at ambient temperature and atmospheric pressure. The reaction medium was filtered and concentrated under reduced pressure to yield 0.4 g of compound 49C in the form of a white solid.

Example 49: Compound 49 was synthesised in the same manner as for compound 3 from the amine 1Y (53.4 mg, 0.08 mmol, 2.00 equiv), the acid 49C (70 mg, 0.28 mmol, 1.00 equiv), DEPC (0.032 mL, 2.00 equiv) and DIEA (0.053 mL, 3.00 equiv) in DCM (3 mL). The residue was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, Atlantis Prep OBD T3 column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 45 % ACN in 10 minutes then 45 % to 100 % ACN in 2 minutes; Waters 2545 UV Detector at 254 nm and 220 nm), to yield 3 mg (1 %) of compound 49 in the form of a white solid.

LC/MS/UV (Ascentis Express C18 column, 2.7 μιη, 4.6 x 100 mm; 40°C;

1.5 mL/min, 10 % to 95 % MeOH in water (0.05 % TFA) in 8 minutes); ESI (C 49 H 74 N 6 O 7 S, exact mass 890.5) m/z: 891.5 (MH + ) and 446.4 (M.2H + /2, 100 %), 6.69 min (100 %, 210 nm).

1H NMR: (400MHz, CD 3 OD, ppm): δ (Presence of rotamers) 8.92 - 8.87 (m, 0.5 H, NHCO, incomplete exchange), 8.70 - 8.63 (m, 0.4 H, NHCO, incomplete exchange), 8.85 - 8.77 (m, 1H), 7.59 - 7.51 (m, 1H), 7.35 - 7.03 (m, 7H), 6.82 - 6.71 (m, 2H), 5.77 - 5.58 (m, 1H), 5,81 - 5.70 (m, 1H), 4.21 - 0.80 (m, 58H).

Example 50

(S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-((S)-2-((4- hydroxyphenethyl)(methyl)amino)-3-methylbutanamido)- V,3- dimethylbutanamido)-3-methoxy-5-methylheptanoyl)pyrrolidin-2 -yl)-3- methoxy-2-m thylpropanamido)-3-phenylpropanoic acid, trifluoroacetic acid

Example 50: Compound 50 was prepared in the same manner as for compound

4, from compound 27 (100 mg, 0.10 mmol, 1.00 equiv). The residue was purified by preparative HPLC (Pre-HPLC-001 SHIMADZU, Atlantis Prep OBD T3 column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 20 % to 40 % ACN in 10 minutes then 40 % to 100 % ACN in 2 minutes; Waters 2545 UV Detector at 254 nm and 220 nm), to yield 10.7 mg (11 %) of compound 50 in the form of a white solid.

LC/MS/UV (Ascentis Express C18 column, 2.7 μιη, 4.6 x 100 mm; 40°C; 1.5 mL/min, 10 % to 95 % MeOH in water (0.05 % TFA) in 8 minutes); ESI (C 47 H 73 N 5 O 9 , exact mass 851.5) m/z: 852.5 (MH ) and 426.8 (M.2H72, 100 %), 6.46 min (91.7 %, 210 nm).

1H NMR: (400MHz, CD 3 OD, ppm): δ (Presence of rotamers) 7.34 - 7.15 (m, 5H); 7.15 - 7.04 (se, 2H), 6.82 - 6.83 (m, 2H), 4.83 - 4.70 (m, 1H), 4.21 - 4.00 (m, 1H), 3.90 - 3.80 (m, 1H), 3.74 - 3.62 (m, 1H), 3.57 - 2.86 (m, 20H), 2.56 - 0.80 (m, 36H).

Example 51

methyl (S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-((S)-2-((4- hydroxybenzyl)(methyl)amino)-3-methylbutanamido)- V,3- dimethylbutanamido)-3-methoxy-5-methylheptanoyl)pyrrolidin-2 -yl)-3- methoxy-2-methylpropanamido)-3- phenylpropanoate, trifluoroacetic acid

Example 51A: tert-butyl (4-formylphenyl)carbonate

4-hydroxybenzaldehyde (3.0 g, 24 mmol) was dissolved in 30 mL of DCM in the presence of 4-DMAP (300 mg, 2.46 mmol, 0.1 equiv.) and di-tert-butyl dicarbonate (5.35 g, 24 mmol, 1.0 equiv.) and agitated 1 hour at ambient temperature. The solution was then diluted with 200 mL of water and extracted 3 times with 100 mL of DCM. The organic phases were combined, dried over sodium sulfate, filtered and concentrated under reduced pressure to yield 5 g (92 %) of compound 51A in the form of a white solid.

Example 51B: benzyl (S)-2((4-((tert-butoxycarbonyl)oxy)benzyl)(methyl) amino)-3 -methylbutanoate

NaBH(OAc) 3 DIEA.THF

Compound 51A (220 mg, 0.99 mmol) was dissolved in 5 mL of THF in the presence of compound IZC (255 mg, 0.99 mmol, 1.0 equiv.), NaBH(OAc) 3 (420 mg, 2 mmol, 2.0 equiv.) and DIEA (654 μΐ) and agitated overnight at ambient temperature. 5 The solution was then diluted with 100 mL of water and extracted 3 times with 50 mL of EtOAc. The organic phases were combined, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified on a silica column with a mixture of EtOAc and PE (1 : 100) to yield 200 mg (47 %) of compound 51B in the form of a white solid.

0 Example 51C: (S)-2-((4-((iert-butoxycarbonyl)oxy)benzyl)(methyl)amino)-3- methyl butanoic acid

Compound 51C was prepared by hydro genation of compound 5 IB (200 mg), following the protocol used for the preparation of compound 3F.

5 Example 51D: methyl (S)-2-((2R,3R)-3-((S)-l-((3R,4S,5S)^l-((S)-2-((S)-

2-((4-((teri-butoxycarbonyl)oxy)benzyl)(methyl)amino)-3-m ethylbutanamido)-N,3- dimethylbutanamido)-3-methoxy-5-methylheptanoyl)pyrrolidin-2 -yl)-3-methoxy-2- methylpropanamido)-3-phenylpropanoate

0 Compound 51D was prepared by coupling compound 51C with amine 3D, following the protocol used for the preparation of compound 3 to obtain the desired product in the form of yellow oil with a yield of 60 %. Example 51: Compound 51D (80 mg, 0.08 mmol) was dissolved in 1 mL of DCM in the presence of 0.5 mL TFA, agitated 2 hours at ambient temperature and then concentrated under reduced pressure. The residue was purified by preparative HPLC (Pre-HPLC-010, SunFire Prep C18 OBD column, 5 μιη, 19 x 150 mm; Eluting phase: water / ACN buffered with 0.05 % TFA; Gradient of 23 % to 40 % ACN in 10 minutes then 40 % to 95 % ACN in 2 minutes; Waters 2489 UV Detector at 254 nm and 220 nm). Compound 51 was obtained with a yield of 24 % (20 mg) in the form of a white solid.

LC/MS/UV (Zorbax SB-Aq, 1.8 μιη, 4.6 x 100 mm; 2 % MeOH in water (0.05 % TFA) for 1 minute then 2 % to 95 % MeOH in 13 minutes); ESI

exact mass 851.54) m/z: 874.5 (MNa + ), 426.9 (M.2H + /2); 12.48 min (96 %, 210 nm).

1H NMR: (300MHz, CD 3 OD, ppm): δ (Presence of rotamers) 8.1 - 8.6 (m, 0.9H, NHCO incomplete exchange); 7.29 - 7.27 (m, 2H), 7.25 - 6.86 (m, 5H), 6.84 - 6.83 (m, 2H), 4.83 - 4.72 (m, 3H), 4.26 - 0.82 (m, 58H).

Example 61

(S)-2-((S)-2-((4-aminophenethyl)(methyl)amino)-3-methylbutan amido)- V- ((3R,4S,5S)-3-methoxy-l-((S)-2-((lR,2R)-l-methoxy-2-methyl-3 -oxo-3-(((S)-2- phenyl-l-(thiazol-2-yl)ethyl)amino)propyl)pyrrolidin-l-yl)-5 -methyl-l-oxoheptan- 4-yl)- V,3-dimethylbutanamide

Example 61A: N-(4-aminophenethyl)-N-methyl-Z-valine dihydrochloride

Compound 11D (962 mg, 2.75 mmol) was dissolved in 10 ml of a commercially available solution of HC1 in propan-2-ol (5 - 6 M), and stirred at room temperature for 2 hours. TLC analysis indicated complete consumption of starting material. The solvent was evaporated under reduced pressure, and the resulting yellow solid triturated with Et 2 0 (2 x 10 ml). The product was dried under vacuum to furnish compound 61 A as a yellow solid (322 mg, 47 %).

Example 61: Carboxylic acid 61 A (73 mg, 0.23 mmol, 1 eq.) and amine 1Y (150 mg, 0.23 mmol, 1 eq.) were dissolved in dry DMF (2 ml). DIEA (158 μΐ, 0.90 mmol, 4 eq.) and DECP (51 μΐ, 0.34 mmol, 1.5 eq.) were added and the reaction stirred for 4 hours at room temperature. Analysis by LC-MS showed complete consumption of the starting material. The solvent was evaporated under reduced pressure, and the residue purified by flash chromatography on silica gel (DCM/MeOH) to furnish compound 61 as a light yellow solid (83 mg, 40 %).

1H NMR: (500MHz, DMSO-d 6 , ppm): δ (Presence of rotamers), 8.86 (d, 0.5H,

NHCO); 8.65 (d, 0.5H, NHCO), 8.11-8.05 (m, 1H, NHCO), 7.80 (d, 0.5H, thiazole), 7.78 (d, 0.5H, thiazole), 7.65 (d, 0.5H, thiazole), 7.63 (d, 0.5H, thiazole), 7.32 - 7.12 (m, 5H), 6.83 (d, J=8.3 Hz, 2H), 6.45 (d, J=8.3 Hz, 2H), 5.56 - 5.49 (m, 0.5 H), 5.42 - 5.35 (m, 0.5H), 4.78 (s, 2H, NH 2 ), 4.74 - 4.46 (m, 2H), 4.01 - 0.66 (m, 57H).

HPLC (Xbridge Shield C18, 3.5 μιη, 4.6 x 50 mm ; 3.5 ml/min, 40°C, 0 to 95 %

MeCN in water (0.1 % TFA) in 2.25 minutes then 95 % MeCN for 0.5 minutes, Tr = 1.31 min (96.5 %, 220 nm).

m/z (Q-TOF ESI + ) 890.5558 (2%, MH + , C 49 H 76 N 7 O 6 S requires 890.5572), 445.7834 (100 %, (MH 2 ) 2+ , C 49 H 77 N 7 O 6 S requires 445.7823).

Example 62

Methyl ((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-((S)-2-((4- aminophenethyl)(methyl)amino)-3-methylbutanamido)- V,3-dimethylbutanamido)- 3-methoxy-5-methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-met hylpropanoyl)-L- phenylalaninate

Example 62: Compound 62 was prepared in the same manner as for compound 61, using carboxylic acid 61A (69 mg, 0.21 mmol, 1 eq.), amine 3D (135 mg, 0.21 mmol, 1 eq.), DIEA (75 μΐ, 0.43 mmol, 2 eq.) and DECP (49 μΐ, 0.32 mmol, 1.5 eq.). The crude product was purified by flash chromatography on silica gel (DCM/MeOH) to furnish compound 62 as a yellowish solid (82 mg, 45 %).

1H NMR: (500MHz, OMSO-d 6 , ppm): δ (Presence of rotamers), 8.50 (d, J=8.3, 0.5H, NHCO); 8.27 (d, J=8.0, 0.5H, NHCO), 8.15-8.04 (m, 1H, NHCO), 7.27 - 7.13 (m, 5H), 6.86 - 6.79 (m, 2H), 6.48 - 6.42 (m, 2H), 4.78 (s, 2H, NH 2 ), 4.74 - 4.44 (m, 3H), 4.01 - 3.72 (m, 1.5H), 3.66 (s, 1.5H, C0 2 Me), 3.63 (s, 1.5H, C0 2 Me), 3.57 - 0.65 (m, 55.5H).

HPLC (Xbridge Shield C18, 3.5 μιη, 4.6 x 50 mm ; 3.5 ml/min, 40°C, 0 to 95 % MeCN in water (0.1 % TFA) in 2.25 minutes then 95 % MeCN for 0.5 minutes, Tr = 1.29 min (95.3 %, 220 nm).

m/z (Q-TOF ESI + ) 865.5800 (2%, MH + , C48H77N6O8 requires 865.5797), 433.2937 (100 %, (MH 2 ) 2+ , C 48 H 7 8N 6 0 8 requires 433.2935).

Example 63

((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-((S)-2-((4-aminophene thyl)(methyl)amino)- 3-methylbutanamido)- V,3-dimethylbutanamido)-3-methoxy-5- methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropanoyl )-L-phenylalanine

2,2,2-trifluoroacetate

Example 63: Compound 62 (23 mg, 0.03 mmol) was dissolved in a mixture of water (1 ml) and acetonitrile (1 ml). Piperidine (0.75 ml) was added and the mixture stirred at room temperature for 5 hours. TLC analysis indicated complete consumption of the starting material. The solvent was evaporated under reduced pressure, and the residue purified by preparative HPLC (SunFire Prep column C 18 OBD, 5 μιη, 19 x 150 mm; Mobile phase: water/MeCN buffered with 0.1 % TFA; Gradient of 20 % to 40 % MeCN in 10 minutes, then from 40 % to 100 % MeCN in 2 minutes; Detector UV Waters 2545 at 254 nm et 220 nm). Compound 63 was obtained as a white solid (14 mg, 66 %).

1H NMR: (500MHz, DMSO-d 6 , ppm): δ (Presence ofrotamers), 12.7 (s(br), 1H, C0 2 H), 9.58 (m(br), 1H); 9.04 - 8.89 (m, 1H), 8.41 (d, 0.6H, NHCO), 8.15 (d, 0.4H, NHCO), 7.27 - 7.13 (m, 5H), 7.13 - 6.99 (m(br), 2H), 6.90 - 6.64 (s(br), 2H), 4.77 - 3.40 (m, 10H), 3.34 - 2.75 (m, 20H), 2.34 - 1.94 (m, 4H), 1.90 - 0.7 (m, 25H).

HPLC (Xbridge Shield C I 8, 3.5 μιη, 4.6 x 50 mm ; 3.5 ml/min, 40°C, 0 to 95 % MeCN in water (0.1 % TFA) in 2.25 minutes then 95 % MeCN for 0.5 minutes, Tr = 1.24 min (100 %, 220 nm).

m/z (Q-TOF EST ) 851.5641 (6%, MH + , C 4 7H7 5 N 6 0 8 requires 851.5641), 426.2854 (100 %, (MH 2 ) 2+ , requires 426.2857).

Example 64

(S)-2-((S)-2-((4-aminophenethyl)(methyl)amino)-3-methylbutan amido)- V- ((3R,4S,5S)-l-((S)-2-((lR,2R)-3-(((lS,2R)-l-hydroxy-l-phenyl propan-2-yl)amino)- l-methoxy-2-methyl-3-oxopropyl)pyrrolidin-l-yl)-3-methoxy-5- methyl-l- oxoheptan-4-yl)- V,3-dimethylbutanamide

Compound 64 was prepared in the same manner as for compound 61, using carboxylic acid 61A (93 mg, 0.29 mmol, 1 eq.), amine 2D (174 mg, 0.29 mmol, 1 eq.), DIEA (100 μΐ, 0.58 mmol, 2 eq.) and DECP (66 μΐ, 0.43 mmol, 1.5 eq.). The crude product was purified by flash chromatography on silica gel (DCM/MeOH) to furnish compound 64 as an off-white solid (51 mg, 21 %).

1H NMR: (500MHz, DMSO-d 6 , ppm): δ (Presence of rotamers), 9.61 (m(br), 1H); 9.05 - 8.89 (m, 1H), 7.93 (d, 0.6H, NHCO), 7.64 (d, 0.4H, NHCO), 7.36 - 6.98 (m, 7H), 6.92 - 6.70 (m(br), 2H), 5.45 (s(br), 1H), 4.80 - 4.41 (m, 3H), 4.06 - 3.44 (m, 4H), 3.37 - 2.79 (m, 18H), 2.45 - 2.21 (m, 3H), 2.17 - 0.70 (m, 35H).

HPLC (Xbridge Shield C18, 3.5 μιη, 4.6 x 50 mm ; 3.5 ml/min, 40°C, 0 to 95 % MeCN in water (0.1 % TFA) in 2.25 minutes then 95 % MeCN for 0.5 minutes, Tr = 1.20 min (100 %, 220 nm).

m/z (Q-TOF ESI + ) 837.5826 (33%, MH + , C47H77N6O7 requires 837.5848),

419.2956 (100 %, (MH 2 ) 2+ , C47H76N6O8 requires 419.2961).

II - Biological activity of the compounds of the invention

Method:

Cell culture. A549 (Non Small Cell Lung Cancer - ATCC CCL- 185) and MDA-

MB-231 (breast adenocarcinoma - ATCC HTB-26) cells were cultured in Minimum Essential Medium Eagle (MEM) with 5% fetal calf serum (FCS) and Dulbecco's modified Eagle Medium (DMEM) with 10% FCS respectively. MCF7 (breast ductal carcinoma - ATCC HTB-22) and SN-12C (kidney carcinoma - ATCC) cells were maintained in RPMI1640 medium (without phenol red for MCF7 cells) containing 10% FCS. All the media were supplemented with fungizone (1.25 μg/mL) and penicillin- streptomycin (100 U / 100 μg/mL). Cells were cultured under standard conditions in an incubator at 37°C, 5%> C0 2 and 95%> atmospheric humidity. Antiproliferative activity on 4 tumor cell lines. Compounds according to the invention were investigated for their antiproliferative activity using an ATPlite proliferation assay (Perkin Elmer, Villebon sur Yvette, France) on a comprehensive panel of 4 cell lines. Cells were seeded in 96 well plates (10 3 cells/well for A549, 2.10 3 for MCF7, MDA-MB-231 and SN12C) at day 0 at a concentration to ensure cells remained in logarithmic cell growth phase throughout the 72 h drug treatment period. After a 24h incubation period, all the cells were treated with serial dilutions of the tested compounds (11 of a 10X solution in 1% DM SO - 6 wells/ condition). To avoid adherence of the compounds onto the tips, tips were changed between two consecutive dilutions. Cells were then placed in 37°C, 5% C0 2 incubator. On day 4, cell viability was evaluated by dosing the ATP released by viable cells. The number of viable cells was analyzed in comparison with the number of solvent treated cells. The EC50 values were determined with curve fitting analysis (non linear regression model with a sigmoidal dose response, variable hill slope coefficient), performed with the algorithm provided by the GraphPad Software (GraphPad Software Inc., CA, USA).

Results:

Various compounds:

Various compounds according to the invention were tested to determine their antiproliferative activity on the MDA-MB-231 cell line following the above-described method. The measured activities gave values of EC50 < 0.1 μΜ.

The few following examples chosen from among the compounds according to the invention illustrate their fully remarkable antiproliferative properties:

Example 12: EC 50 = 5.80xl0 ~10 M; Example 13: EC 50 = 7.95xl0 ~8 M; Example 15: EC 50 = 1.70xl0 "10 M; Example 27: EC 50 = 1.20xl0 "10 M.

Various cell lines:

Compound 15 was tested on different cell lines (A549, MDA-MB-231, MCF-7, SN12C) following the above-described method. The measured activities gave values of ECso < 0.1 μΜ.

ECso (M) A549 MDA-MB-231 MCF-7 SN12C

Compound 15 1.45xlO "llJ 1.70xlO "llJ 7.15xlO "llJ 2.18χ10 "ιυ Comparative examples:

The substitution on the phenyl ring (amino/hydroxyl v. carboxyl) was studied in the comparative examples below showing the improved antiproliferative activity of the drugs according to the invention comprising an amino or hydroxyl substituent.