AU2002313540A1 - Antitumoral analogs - Google Patents

Antitumoral analogs

Info

Publication number
AU2002313540A1
AU2002313540A1 AU2002313540A AU2002313540A AU2002313540A1 AU 2002313540 A1 AU2002313540 A1 AU 2002313540A1 AU 2002313540 A AU2002313540 A AU 2002313540A AU 2002313540 A AU2002313540 A AU 2002313540A AU 2002313540 A1 AU2002313540 A1 AU 2002313540A1
Authority
AU
Australia
Prior art keywords
compound
nmr
mhz
hydrogen
alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2002313540A
Other versions
AU2002313540B2 (en
Inventor
Carmen Cuevas
Pilar Gallego
Ignacio Manzanares
Valentin Martinez
Simon Munt
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pharmamar SA
Original Assignee
Pharmamar SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB0119243.4A external-priority patent/GB0119243D0/en
Application filed by Pharmamar SA filed Critical Pharmamar SA
Publication of AU2002313540A1 publication Critical patent/AU2002313540A1/en
Application granted granted Critical
Publication of AU2002313540B2 publication Critical patent/AU2002313540B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Description

ANTITUMORAL ANALOGS
The present invention relates to derivatives of the ecteinascidins, particularly ecteinascidin 736 (ET-736), pharmaceutical compositions containing them and their use as antitumoral compounds.
BACKGROUND OF THE INVENTION
The ecteinascidins are exceedingly potent antitumour agents isolated from the marine tunicate Ecteinascidia turbinata. Several ecteinascidins have been reported previously in the patent and scientific literature. See, for example:
U.S. Patent No 5.256.663, which describes pharmaceutical compositions comprising matter extracted from the tropical marine invertebrate, Ecteinascidia turbinata, and designated therein as ecteinascidins, and the use of such compositions as antibacterial, antiviral, and/ or antitumour agents in mammals.
U.S. Patent No 5.089.273, which describes novel compositions of matter extracted from the tropical marine invertebrate, Ecteinascidia turbinata, and designated therein as ecteinascidins 729, 743, 745, 759A, 759B and 770. These compounds are useful as antibacterial and/or antitumour agents in mammals.
U.S. Patent No 5.149.804 which describes Ecteinascidins 722 and 736 (Et's 722 and 736) isolated from the Caribbean tunicate Ecteinascidia turbinata and their structures. Et's 722 and 736 protect mice in vivo at very low concentrations against P388 lymphoma, B 16 melanoma, and Lewis lung carcinoma.
U.S. Patent No 5.478.932, which describes ecteinascidins isolated from the Caribbean tunicate Ecteinascidia turbinata, which provide in vivo protection against P388 lymphoma, B 16 melanoma, M5076 ovarian sarcoma, Lewis lung carcinoma, and the LX- 1 human lung and MX- 1 human mammary carcinoma xenografts.
U.S. Patent No 5.654.426, which describes several ecteinascidins isolated from the Caribbean tunicate Ecteinascidia turbinata, which provide in vivo protection against P388 lymphoma, B 16 melanoma, M5076 ovarian sarcoma, Lewis lung carcinoma, and the LX-1 human lung and MX- 1 human mammary carcinoma xenografts.
U.S. Patent No 5.721.362 which describes a synthetic process for the formation of ecteinascidin compounds and related structures.
U.S. Patent No 6.124.292 which describes a series of new ecteinascidin- like compounds.
WO 0177115, WO 0187894 and WO 0187895, which describe new synthetic compounds of the ecteinascidin series, their synthesis and biological properties.
See also: Corey, E.J., J. Am. Chem. Soc, 1996, 118 pp. 9202-9203; Rinehart, et al., Journal of Natural Products, 1990, "Bioactive Compounds from Aquatic and Terrestrial Sources", vol. 53, pp. 771- 792; Rinehart et al., Pure and Appl. Chem., 1990, "Biologically active natural products", vol 62, pp. 1277- 1280; Rinehart, et al., J. Org. Chem., 1990, "Ecteinascidins 729, 743, 745, 759A, 759B, and 770: potent Antitumour Agents from the Caribbean Tunicate Ecteinascidia tuminata", vol. 55, pp. 4512-4515; Wright et al., J. Org. Chem., 1990, "Antitumour Tetrahydroisoquinoline Alkaloids from the Colonial ascidian Ecteinascidia turbinata", vol. 55, pp. 4508-4512; Sakai et al., Proc. Natl. Acad. Sci. USA 1992, "Additional anitumor ecteinascidins from a Caribbean tunicate: Crystal structures and activities in vivo", vol. 89, 1 1456- 1 1460; Science 1994, "Chemical Prospectors Scour the Seas for Promising Drugs", vol. 266, pp.1324; Koenig, K.E., "Asymmetric Synthesis", ed. Morrison, Academic Press, Inc., Orlando, FL, vol. 5, 1985, p. 71; Barton, et al., J. Chem Soc. Perkin Trans., 1 , 1982, "Synthesis and Properties of a Series of Sterically Hindered Guanidine bases", pp. 2085; Fukuyama et al., J. Am. Chem. Soc, 1982, "Stereocontrolled Total Synthesis of (+)-Saframycin B", vol. 104, pp. 4957; Fukuyama et al., J. Am. Chem. Soc, 1990, "Total Synthesis of (+) - Saframycin A", vol. 112, p. 3712; Saito, et al., J. Org. Chem., 1989, "Synthesis of Saframycins. Preparation of a Key tricyclic Lactam Intermediate to Saframycin A", vol. 54, 5391; Still, et al., J Org. Chem., 1978, "Rapid Chromatographic Technique for Preparative Separations with Moderate Resolution", vol. 43, p. 2923; Kofron, W.G.; Baclawski, L.M., J. Org. Chem., 1976, vol. 41, 1879; Guan et al., J. Biomolec Struc & Dynam., vol. 10, pp. 793-817 (1993); Shamma et al., "Carbon- 13 NMR Shift Assignments of Amines and Alkaloids", p. 206 (1979); Lown et al., Biochemistry, 21, 419-428 (1982); Zmijewski et al., Chem. Biol. Interactions, 52, 361-375 (1985); Ito, CRC Crit. Rev. Anal. Chem., 17, 65- 143 (1986); Rinehart et al., "Topics in Pharmaceutical Sciences 1989", pp. 613-626, D. D. Breimer, D. J. A. Cromwelin, K. K. Midha, Eds., Amsterdam Medical Press B. V., Noordwijk, The Netherlands (1989); Rinehart et al., "Biological Mass Spectrometry", 233-258 eds. Burlingame et al., Elsevier Amsterdam (1990); Guan et al., Jour. Biomolec. Struct. & Dynam., vol. 10 pp. 793-817 (1993); Nakagawa et al., J. Amer. Chem. Soc, 11 1 : 2721-2722 (1989);; Lichter et al., "Food and Drugs from the Sea Proceedings" (1972), Marine Technology Society, Washington, D.C. 1973, 117- 127; Sakai et al., J. Amer. Chem. Soc, 1996, 1 18, 9017; Garcϊa-Rocha et al., Brit. J. Cancer, 1996, 73: 875-883; and pommier et al., Biochemistry, 1996, 35: 13303- 13309;
In 2000, a hemisynthetic process for the formation of ecteinascidin compounds and related structures such as phthalascidin starting from natural bis(tetrahydroisoquinoline) alkaloids such as the saframycin and safracin antibiotics available from different culture broths was reported; See Manzanares et al., Org. Lett., 2000, "Synthesis of Ecteinascidin ET-743 and Phthalascidin Pt-650 from Cyanosafracin B", Vol. 2, No 16, pp. 2545-2548; and International Patent Application WO 00 69862.
Ecteinascidin 736 was first discovered by Rinehart and features a tetrahydro-β-carboline unit in place of the tetrahydroisoquinoline unit more usually found in the ecteinascidin compounds isolated from natural sources; See for example Sakai et al., Proc. Natl. Acad. Sci. USA 1992, "Additional antitumor ecteinascidins from a Caribbean tunicate: Crystal structures and activities in vivo", vol. 89, 11456-11460.
Et-736
WO 9209607 claims ecteinascidin 736, as well as ecteinascidin 722 with hydrogen in place of methyl on the nitrogen common to rings C and D of ecteinascidin 736 and O-methylecteinascidin 736 with methoxy in place of hydroxy on ring C of ecteinascidin 736.
Despite the positive results obtained in clinical applications in chemotherapy, the search in the field of ecteinascidin compounds is still open to the identification of new compounds with optimal features of cytotoxicity and selectivity toward the tumour and with a reduced systemic toxicity and improved pharmacokinetic properties.
SUMMARY OF THE INVENTION
The present invention is directed to compounds of the general formula
or la
la
wherein the substituent groups for R*., R2, R3, R4, and R5 are each independently selected from the group consisting of H, OH, OR', SH, SR', SOR', SO2R', C(=O)R', C(=O)OR', NO2, NH2, NHR', N(R')2, NHC(O)R', CN, halogen, =O, substituted or unsubstituted Ci-Cis alkyl, substituted or unsubstituted C2-C-.8 alkenyl, substituted or unsubstituted C2-Ci8 alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclic; wherein X is independently selected of OR', CN, (=O), or H. wherein each of the R' groups is independently selected from the group consisting of H, OH, NO2, NH2, SH, CN, halogen, =O, C(=O)H, C(=O)CH3, CO2H, substituted or unsubstituted Cj.-Ci8 alkyl, substituted or unsubstituted C2-Ci8 alkenyl, substituted or unsubstituted 02-C^ alkynyl, substituted or unsubstituted aryl; wherein m is 0, 1 or 2; and wherein n is 0, 1, 2, 3 or 4.
The present invention also relates to the synthesis of ecteinascidin 736 (where Ri, R3, R4, R5 = H , R2 = CH3CO- and X = OH) and related compounds of the general formula I or la.
In another aspect, the invention relates to pharmaceutical compositions comprising a compound of formula I.
In another aspect, the invention relates to the use of compounds of general formula I or la in the treatment of cancer.
As one group, the invention provides compounds of formula I or la, wherein:
Rx is hydrogen, hydroxy, halogen, alkoxy or aralkyl;
R2 and R3 are independently selected from hydrogen, R', C=OR', or
COOR', where R' is optionally substituted alkyl or alkenyl, the optional substituents being chosen from halo, amino including amino derived from amino acid, aryl or heterocyclic;
R4 is hydrogen, alkyl or C(=O)OR', where R' is alkylene.
R5 is hydrogen or alkyl;
X is hydrogen, hydroxy, cyano or keto; m is 0 or 1; and n is 0 or 1. Suitable halogen substituents in the compounds of the present invention include F, Cl, Br and I.
Alkyl groups preferably have from 1 to 24 carbon atoms. One more preferred class of alkyl groups has 1 to about 12 carbon atoms, yet more preferably 1 to about 8 carbon atoms, still more preferably 1 to about 6 carbon atoms, and most preferably 1, 2, 3 or 4 carbon atoms. Another more preferred class of alkyl groups has 12 to about 24 carbon atoms, yet more preferably 12 to about 18 carbon atoms, and most preferably 13, 15 or 17 carbon atoms. Methyl, ethyl and propyl including isopropyl are particularly preferred alkyl groups in the compounds of the present invention. As used herein, the term alkyl, unless otherwise modified, refers to both cyclic and noncyclic groups, although cyclic groups will comprise at least three carbon ring members.
Preferred alkenyl and alkynyl groups in the compounds of the present invention have one or more unsaturated linkages and from 2 to about 12 carbon atoms, more preferably 2 to about 8 carbon atoms, still more preferably 2 to about 6 carbon atoms, even more preferably 1, 2, 3 or 4 carbon atoms. The terms alkenyl and alkynyl as used herein refer to both cyclic and noncyclic groups, although straight or branched noncyclic groups are generally more preferred.
Preferred alkoxy groups in the compounds of the present invention include groups having one or more oxygen linkages and from 1 to about 12 carbon atoms, more preferably from 1 to about 8 carbon atoms, and still more preferably 1 to about 6 carbon atoms, and most preferably 1, 2, 3 or 4 carbon atoms.
Preferred alkylthio groups in the compounds of the present invention have one or more thioether linkages and from 1 to about 12 carbon atoms, more preferably from 1 to about 8 carbon atoms, and still more preferably 1 to about 6 carbon atoms. Alkylthio groups having 1, 2, 3 or 4 carbon atoms are particularly preferred.
Preferred alkylsulphinyl groups in the compounds of the present invention include those groups having one or more sulphoxide (SO) groups and from 1 to about 12 carbon atoms, more preferably from 1 to about 8 carbon atoms, and still more preferebly 1 to about 6 carbon atoms. Alkylsulphinyl groups having 1, 2, 3 or 4 carbon atoms are particularly preferred.
Preferred alkylsulphonyl groups in the compounds of the present invention include those groups having one or more sulphonyl (SO2) groups and from 1 to about 12 carbon atoms, more preferably from 1 to about 8 carbon atoms, and still more preferably 1 to about 6 carbon atoms. Alkylsulphonyl groups having 1, 2, 3 or 4 carbon atoms are particularly preferred.
Preferred aminoalkyl groups include those groups having one or more primary, secondary and/ or tertiary amine groups, and from 1 to about 12 carbon atoms, more preferably 1 to about 8 carbon atoms, still more preferably 1 to about 6 carbon atoms, even more preferably 1, 2, 3 or 4 carbon atoms. Secondary and tertiary amine groups are generally more preferred than primary amine moieties.
Suitable heterocyclic groups include heteroaromatic and heteroalicyclic groups. Suitable heteroaromatic groups in the compounds of the present invention contain one, two or three heteroatoms selected from N, O or S atoms and include, e.g., coumarinyl including 8-coumarinyl, quinolinyl including 8-quinolinyl, pyridyl, pyrazinyl, pyrimidyl, furyl, pyrrolyl, thienyl, thiazolyl, oxazolyl, imidazolyl, indolyl, benzofuranyl and benzothiazol. Suitable heteroalicyclic groups in the compounds of the present invention contain one, two or three heteroatoms selected from N, O or S atoms and include, e.g., tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholino and pyrrolindinyl groups.
Suitable carbocyclic aryl groups in the compounds of the present invention include single and multiple ring compounds, including multiple ring compounds that contain separate and/ or fused aryl groups. Typical carbocyclic aryl groups contain 1 to 3 separate or fused rings and from 6 to about 18 carbon ring atoms. Specifically preferred carbocyclic aryl groups include phenyl including substituted phenyl such as 2-substituted phenyl, 3-substituted phenyl, 2.3- substituted phenyl, 2.5-substituted phenyl, 2.3.5-substituted and 2.4.5-substituted phenyl, including where one or more of the phenyl substituents is an electron-withdrawing group such as halogen, cyano, nitro, alkanoyl, sulphinyl, sulphonyl and the like; naphthyl including 1- naphthyl and 2-naphthyl; biphenyl; phenanthryl; and anthracyl.
References herein to substituted R' groups in the compounds of the present invention refer to the specified moiety, typically alkyl or alkenyl, that may be substituted at one or more available positions by one or more suitable groups, e.g., halogen such as fluoro, chloro, bromo and iodo; cyano; hydroxyl; nitro; azido; alkanoyl such as a Cl-6 alkanoyl group such as acyl and the like; carboxamido; alkyl groups including those groups having 1 to about 12 carbon atoms or from 1 to about 6 carbon atoms and more preferably 1-3 carbon atoms; alkenyl and alkynyl groups including groups having one or more unsaturated linkages and from 2 to about 12 carbon or from 2 to about 6 carbon atoms; alkoxy groups having those having one or more oxygen linkages and from 1 to about 12 carbon atoms or 1 to about 6 carbon atoms; aryloxy such as phenoxy; alkylthio groups including those moieties having one or more thioether linkages and from 1 to about 12 carbon atoms or from 1 to about 6 carbon atoms; alkylsulphinyl groups including those moieties having one or more sulphinyl linkages and from 1 to about 12 carbon atoms or from 1 to about 6 carbon atoms; alkylsulphonyl groups including those moieties having one or more sulphonyl linkages and from 1 to about 12 carbon atoms or from 1 to about 6 carbon atoms; aminoalkyl groups such as groups having one or more N atoms and from 1 to about 12 carbon atoms or from 1 to about 6 carbon atoms; carbocylic aryl having 6 or more carbons, particularly phenyl (e.g., R being a substituted or unsubstituted biphenyl moiety); and aralkyl such as benzyl; heterocyclic groups including heteroalicyclic and heteroaromatic groups, especially with 5 to 10 ring atoms of which 1 to 4 are heteroatoms, more preferably heterocyclic groups with 5 or 6 ring atoms and 1 or 2 heteratoms or with 10 ring atoms and 1 to 3 heteroatoms.
Preferred R' groups are present in groups of formula R', COR' or OCOR' and include alkyl or alkenyl, that may be substituted at one or more available positions by one or more suitable groups, e.g., halogen such as fluoro, chloro, bromo and iodo, especially ω-chloro or perfluoro; aminoalkyl groups such as groups having one or more N atoms and from 1 to about 12 carbon atoms or from 1 to about 6 carbon atoms, and especially including amino acid, notably glycine, alanine, arginine, asparagine, asparaginic acid, cy stein, glutamine, glutamic acid, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine or valine, especially protected forms of such amino acids; carbocylic aryl having 6 or more carbons, particularly phenyl; and aralkyl such as benzyl; heterocyclic groups including heteroalicyclic and heteroaromatic groups, especially with 5 to 10 ring atoms of which 1 to 4 are heteroatoms, more preferably heterocyclic groups with 5 or 6 ring atoms and 1 or 2 heteratoms or with 10 ring atoms and 1 to 3 heteroatoms, the heterocyclic groups optionally being substituted with one or more of the subsitituents permitted for R' and especially amino such as dimethylamino or with keto.
Without being exhaustive, preferred compounds of this invention have one or more of the following definitions:
Ri is H; hydroxy; halogen, especially F, alkyoxy (alkyl being 1 to 7 carbon atoms), especially C1 to C3 alkyl and benzyloxy, most especially methoxy and benzyloxy . Particularly preferred are H, OH or OMe.
R2 is H; or acetyl; alkyl-CO (alkyl being up to 25 carbon atoms, such as up to 17, 19 or 21 carbon atoms and preferably an odd number of carbon atoms corresponding to a fatty acid carboxylic acid of even number of carbon atoms or else a low number of carbon atoms such as 1 to 7) especially CH3-(CH2)n-CO- where n is for example 1 , 2, 4, 6, 12, 14 or 16; haloalkyl-CO-, especially trifluoromethylcarbonyl, heptafluorobutyrylcarbonyl or 3-chloropropionyl; arylalkyl-CO-; arylalkenyl-CO-, especially cinnamoyl-CO-; alkyl-O-CO-, especially t- butyl-O-CO- or alkenyl-O-CO-, especially allyl-O-CO- or vinyl-O-CO.
R3 is preferably H; alkenyl, especially allyl; alkyl-CO- (alkyl being up to 25 carbon atoms, such as up to 17, 19 or 21 carbon atoms and preferably an odd number of carbon atoms corresponding to a fatty acid carboxylic acid of even number of carbon atoms or else a low number of carbon atoms such as 1 to 6) especially CH3-(CH2)n-CO- where n is for example 1, 2, 4, 6, 12, 14 or 16; alkyl-O-CO-, especially t-butyl-O-CO-; alkenyl-O-CO, especially allyl-O-CO-and vinyl-O-CO.
R4 is preferably H, alkyl (alkyl being 1 to 6 carbon atoms) especially Ci to C3 alkyl; alkenyl especially allyl, alkenyl-O-CO- especially vinyl-O-CO and R4 is most especially H. Rs is H or alkyl (alkyl being 1 to 6 carbon atoms) and R5 is most especially H or Me.
X is a H, -CN or -OH, most especially -OH or -CN.
m is 0 or 1.
n is 0 or 1.
Compounds where RI is not hydrogen are one class of preferred compounds. See for example compounds 27 to 36. These compounds have higher activity, a wider therapeutic window and improved pharmacokinetic properties. Preferred substituents are methoxy, methyl, hydroxy, benzyloxy, fluoro.
Compounds wherein R3 is an ester or an ether are among the preferred compounds. In general they have improved toxicology properties and thus give a wider therapeutic window. Of those, compounds with an ester or carbonate at this position are most preferred, and in particular carbonates. Esters with bulky groups (long aliphatic or aromatic residues) give better results. Among the carbonates, terButyloxycarbonyl (tBOC) and vinyloxycarbonyl (VOC) are the most preferred substituents for these positions.
Compounds where R5 is not hydrogen are another class of preferred compounds. See for example compounds 37 to 44. These compounds tend to be less active (cytotoxic) but have lower toxicity and improved pharmacokinetic properties. When R5 is not hydrogen a chiral center is generated, and we have found that there is difference in activity between the diastereoisomers. Compounds wherein R2 is an ester or an ether are also preferred compounds. In general they have improved toxicology properties and thus give a wider therapeutic window. Of those, compounds with an ester or carbonate at this position are most preferred, and in particular carbonates. Esters with bulky groups (long aliphatic or aromatic residues) give better results. Among the carbonates, terButyloxycarbonyl (tBOC) and vinyloxycarbonyl (VOC) are the most preferred substituents for these positions.
There are compounds that have good ADME properties (absorption- distribution-metabolism-excretion) which are good indicative of pharmacokinetic s .
As mentioned above, compounds of the present invention, preferably those with bulky substituted groups, have a good therapeutic window and the estherification of the phenols with different acids and carbonates, results in a general enhancement of the pharmaceutical properties: there is a significant decrease in hepatocyte toxicity, and also a good profile on drug-drug interactions since these derivatives do not show cytochrome inhibition having moreover higher metabolic stability.
Several active antitumor compounds have been prepared and it is believed that many more compounds may be formed in accordance with the teachings of the present disclosure.
Antitumoural activities of these compounds include leukaemias, lung cancer, colon cancer, kidney cancer, prostate cancer, ovarian cancer, breast cancer, sarcomas and melanomas.
Another especially preferred embodiment of the present invention is pharmaceutical compositions useful as antitumour agents which contain as active ingredient a compound or compounds of the invention, as well as the processes for their preparation.
Examples of pharmaceutical compositions include any solid (tablets, pills, capsules, granules etc) or liquid (solutions, suspensions or emulsions) with suitable compositions or oral, topical or parenteral administration.
Administration of the compounds or compositions of the present invention may be any suitable method, such as intravenous infusion, oral preparation, intraperitoneal and intravenous preparation.
The compounds and compositions of this invention may be used with other drugs to provide a combination therapy. The other drugs may form part of the same composition, or be provided as a separate composition for administration at the same time or a different time. The identity of the other drug is not particularly limited, and suitable candidates include: a) drugs with antimitotic effects, especially those which target cytoskeletal elements, including microtubule modulators such as taxane drugs (such as taxol, paclitaxel, taxotere, docetaxel), podophylotoxins or vinca alkaloids (vincristine, vinblastine); b) antimetabolite drugs such as 5-fluorouracil, cytarabine, gemcitabine, purine analogues such as pentostatin, methotrexate); c) alkylating agents such as nitrogen mustards (such as cyclophosphamide or ifosphamide); d) drugs which target DNA such as the antracycline drugs adriamycin, doxorubicin, pharmorubicin or epirubicin; e) drugs which target topoisomerases such as etoposide; f) hormones and hormone agonists or antagonists such as estrogens, antiestrogens (tamoxifen and related compounds) and androgens, flutamide, leuprorelin, goserelin, cyprotrone or octreotide; g) drugs which target signal transduction in tumour cells including antibody derivatives such as herceptin; h) alkylating drugs such as platinum drugs (cis-platin, carbonplatin, oxaliplatin, paraplatin) or nitrosoureas; i) drugs potentially affecting metastasis of tumours such as matrix metalloproteinase inhibitors; j) gene therapy and antisense agents; k) antibody therapeutics;
1) other bioactive compounds of marine origin, notably the didemnins such as aplidine; m) steroid analogues, in particular dexamethasone; n) anti-inflammatory drugs, in particular dexamethasone; and o) anti-emetic drugs, in particular dexamethasone.
Yet another especially preferred embodiment of the present invention is the synthetic intermediates of the compounds of the present invention as described in detail below.
Finally, the present invention includes the synthetic processes described herein.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
One class of preferred compounds of this invention includes compounds of this invention which have one or more of the following substituents: Ri is hydrogen, hydroxy, halogen especially flouro, alkoxy especially methoxy, or aralkyl especially benzyl; R2 is hydrogen; alkyl, more preferably alkyl of 1 to 6 carbon atoms;
C(=O)R', where R' is alkyl, more preferably alkyl of 1 to 24 carbon atoms, especially 1 to 8 or 12 to 18 carbon atoms; haloalkyl, more preferably ω-chloro- or perfluoro- alkyl of 1 to 4 carbon atoms, especially ω-chloroethyl or perfluoromethyl, ethyl or propyl; heterocylicalkyl, more preferably an aylkyl of 1 to 6 carbon atoms with an ω-heterocyclic substituent suitably having 5 to 10 ring atoms and 1 to 4 heteroatoms, including fused heteroalicyclic with 3 hetero atoms, such as bio tin; aminoalkyl, more preferably alkyl of 1 to 6 carbon atoms, especially 2 carbon atoms, with an ω-amino group optionally protected for example with alkoxycarbonyl such as (CH3)3C-O-C=O- or other protecting group; arylalkylene, especially cinnamoyl; alkylene, especially vinyl or allyl; aralkyl, such as benzyl; or
C(=O)OR', where R' is alkyl, more preferably alkyl of 1 to 6 carbon atoms, especially branched alkyl; alkenyl, more preferably allyl;
R3 is hydrogen; alkyl, more preferably alkyl of 1 to 6 carbon atoms; (C=O)R', where R' is alkoxy, especially with an alkyl group of 1 to 6 carbon atoms; alkyl, more preferably alkyl of 1 to 24 carbon atoms, preferably 1 to 8 or 12 to 18 carbon atoms; haloalkyl, more preferably perfluoroalkyl of 1 to 4 carbon atoms, especially perfluoromethyl, ethyl or propyl; arylalkylene, especially cinnamoyl; heterocylicalkyl, more preferably an alkyl of 1 to 6 carbon atoms with an ω heterocyclic substituent suitably having 5 to 12 ring atoms and 1 to 4 heteroatoms, including fused heterocyclic with 3 ring atoms, such as biotin; heterocyclicalkyl, with preferably 1 carbon atom in the alkyl group, and more preferably heteroalicylicmethyl with 5 to 10 ring atoms and 1 to 4 ring atoms, especially fused heterocylic with 1 to 4 heteroatoms, such as dimethylaminocoumarin or coumarin; alkylene, especially allyl; aralkyl, such as benzyl;
(C=O)OR', where R' is alkyl, more preferably alkyl of 1 to 6 carbon atoms; alkylene, especially vinyl or allyl; aralkyl, such as benzyl.
R4 is hydrogen; alkyl, more preferably alkyl of 1 to 6 carbon atoms;
(C=O)OR', where R' is alkylene, especially vinyl.
R5 is hydrogen or alkyl.
X is hydrogen, hydroxy, cyano or keto.
m is 0 or 1.
n is 0 or 1.
In a related aspect of this invention, the compounds have one or more of the following features:
R2 is not acetyl. Preferably it has at least 4, 5 or 6 carbon atoms, for example up to 18 or 24 carbon atoms. Suitable substituents include esters COR', where R' is alkyl, alkenyl, often with one or more substituents. Alkyl, substituted alkyl, alkenyl and arylalkenyl are preferred, with suitable substituents including aryl, heterocyclic. Other defintions for R2 include esters of formula COR' derived from an amino acid, optionally a protected amino acid.
R3 is not hydrogen. Preferably it is R', COR' or COOR' where R' is a substituent with some bulk. Such bulky substituents include those with branched chain groups, unsaturated groups or cyclic groups including aromatic groups. Thus, branched alkyl, cycloalkyl, branched alkenyl, aryl, heteroaromatic and related groups are preferred for inclusion within the structure of the substituent R3. Preferably the total number of carbon atoms in R3 is 2 to 24, more preferably 6 to 18 carbon atoms. Typically R3 is an ester, ether or carbonate, being of formula COR', R' or COOR'.
R5 is not hydrogen. Preferably it is R', COR' or COOR' where R' is a substituent with some bulk. Such bulky substituents include those with branched chain groups, unsaturated groups or cyclic groups including aromatic groups. Thus, branched alkyl, cycloalkyl, branched alkenyl, aryl, heteroaromatic and related groups are preferred for inclusion within the structure of the substituent R5. Preferably the total number of carbon atoms in R5 is 2 to 24, more preferably 6 to 18 carbon atoms. Typically R4 is an ester, ether or carbonate, being of formula COR', R' or COOR'.
Examples of protecting groups for amino and other substituents are given in WO 0069862, and we expressly incorporate that disclosure.
This application claims priority of a British patent application. We expressly incorporate by reference any disclosure which is in the specification of that British priority application and which is not in the present application.
Furthermore, we expressly incorporate by reference each of WO 0069862, WO 0177115, WO 0187894 and WO 0187895 for their discussion of substituents which correspond to the substituents of the present invention. Any definitions given in any of these earlier applications for a particular substituent can be adopted for a substituent of a compound of this invention.
Furthermore, we do not claim any of the compounds disclosed in the earlier applications, including WO 9209607, and we expressly disclaim any such compounds. We expressly incorporate by reference each of the earlier applications for the wording of any disclaimer which might be necessary.
Disclosed in international patent application WO 0069862 is compound 36 (an intermediate in the conversion of cyanosafracin B to Ecteinascidin 743).
This hemi-synthetic intermediate has served as the starting material for the synthesis of ecteinascidin 736, a further member of the naturally occurring ecteinascidin family with potential antitumor therapeutic activity.
The preferred method of producing ecteinascidin 736 and related compounds with different substituents in the tetrahydro-β-carboline unit and in the position 18 (-OR4) are described below in the following reaction scheme.
Scheme 1
36 Compound type I ET-736 (R„ R2, Rj =H)
As illustrated in Scheme 1, intermediate 36 can be converted to ET-736 (or a substituted derivative) in two steps.
The first step for producing the preferred compounds type I of the present invention from compound 36 is the introduction of the tetrahydro-β-carboline unit by reaction with the corresponding primary or secondary amine.
The second step is the transformation of the CN group into an OH group by reaction with silver nitrate in ACN/H2O.
Also is possible to obtain new derivatives with different substituent groups (-OR4, position 18 and =NRs) trough a acylation or alkylation reaction from the preferred compounds I. In all theses cases Ri and R2 in the starting material is an hydrogen atom. From the same intermediate and through an alkylation reaction with allyl bromide or an acylation reaction with vinylchloroformiate it can be obtained N and O allylated and N and O vinyl derivatives. All these compounds by reaction with silver nitrate lead to the final products wherein the CN group is transformed into an OH group. As the skilled artisan will readily appreciate, the reaction scheme described herein may be modified by use of a wide range of substituted primary amines to produce a series of substituted ecteinascidin 736 derivatives and the compounds generated therefore are to be considered as being part of this invention.
In particular the reaction conditions can be varied to suit other combinations of the substituent groups in the tetrahydro-β-carboline unit.
The preferred method of producing ecteinascidin 694 and related compounds with different substituents in the position 5 and 18 (-OR6 and -OR7) are described below in the following reaction scheme.
Scheme 2
In Scheme 2 the hydrolisis of the acetyl group in C-5 in basic conditions allows to prepare the intermediate with the hydroxyl group in this position. From this compound and by an acylation reaction with anhydrides, acid chlorides or carboxylic acids are prepared new derivatives mono-O sustitued and mono and di-O sustitued (in C-5 and C- 18). The reaction to tranform the CN group into the OH is performed in the classic conditions (silver nitrate in CH3CN/H2O). On the other hand Et-694 can be obtained from Et-736 through the hydrolisis of the acetyl group in C-5 with KOH/MeOH.
As the skilled artisan will readily appreciate, the reaction scheme described herein may be modified by use of a wide range of substituted primary amines to produce a series of substituted ecteinascidin 736-CN derivatives and the compounds generated therefore are to be considered as being part of this invention.
In particular the reaction conditions can be varied to suit other combinations of the substituent groups in the tetrahydro-β-carboline unit and in positions C-5 and C- 18.
The present invention will be further illustrated with reference to the following examples which aid in the understanding, but which are not to be construed as limitations thereof.
EXPERIMENTAL PART
Scheme 1
Method 1.- To a solution of 1 equiv. of starting material in acetic acid (5.33 E-5M) under argon at room temperature was added 3.5 equiv. of tryptamine. The reaction mixture was stirred during 24 h and then the acetic acid was evaporated. An aqueous saturated solution of NaHCO3 was added and the mixture was extracted with CH2CI2 and the organic layers were dried over Na2SO4. Flash chromatography gives pure compounds. Method 2.- To a solution of 1 equiv. of compound 1 in CH2CI2 (0.032M) under Argon at room temperature were added 2 equiv. of Et3N and 2 equiv. of the butyryl chloride or Boc anhydride (3 equiv.) or vinylchloroformiate. The reaction was followed by TLC and quenched with an aqueous saturated solution of NaHCO3, extracted with CH2CI2 and the organic layers dried over Na2SO4. Flash chromatography gives pure compound.
Method 3.- To a solution of 1 equiv. of compound 1 in DMF (0.032M) under Argon at room temperature were added 3 equiv. of CS2CO3 and 3 equiv. of the allyl bromide. The reaction was followed by TLC and quenched with an aqueous saturated solution of NaHCO3, extracted with CH2CI2 and the organic layers dried over Na2SO4. Flash chromatography gives a mixture of two pure compounds: compound 24 (ET-736-CN-A11) and compound 25 (ET-736-CN-diAll).
Method 4.- To a solution of 1 equiv. of starting material in CH3CN/H2O 3:2 (0.009M) were added 30 equiv. of AgNO3. After 24 h the reaction was quenched with a mixture 1 : 1 of saturated solutions of brine and NaHCO3, stirred for 10 min and diluted and extracted with CH2CI2. The organic layer was dried with Na2SO4. Chromatography gives pure compounds.
Method 1:
Compound 1: iH-NMR (300 MHz, CDC13): δ 7.74 (s, IH); 7.38 (d, IH); 7.25 (d, IH); 7.08 (t, IH); 7.00 (t, IH); 6.66 (s, IH); 6.22 (d, IH); 6.02 (d, IH); 5.79 (s, IH); 5.08(d, IH); 4.55(s, IH); 4.32(s, IH); 4.27(d, IH); 4.21(s, IH); 4.19(d, IH); 3.81 (s, 3H); 3.44-3.40(m, 2H); 3.18-2.78(m, 4H); 2.71-
2.51(m, 3H); 2.37(s, 3H); 2.26(s, 3H); 2.21(s, 3H); 2.06 (s, 3H).
!3C-NMR (75 MHz, CDCI3): δ 171.7, 168.9, 148.2, 145.9, 143.2, 141.3,
140.5, 135.7, 130.8, 130.6, 129.5, 127.0, 122.2, 120.9, 120.8, 1 19.5,
118.6, 118.4, 1 13.8, 111.1, 110.5, 102.2, 62.5, 61.5, 60.8, 60.5, 59.7, 55.9, 54.8, 42.1, 41.7, 40.0, 39.5, 29.9, 24.0, 21.7, 20.8, 16.1, 9.9. ESI-MS m/z: Calcd. for C41H41N5O8S: 763.3 Found (M+H+): 764.2.
Compound 2: Η-NMR (300 MHz, CDCI3): δ 7.64(s, IH); 7.12 (d, IH); 6.81 (d, IH); 6.73 (dd, IH); 6.65 (s, IH); 6.19 (s, IH); 6.00 (s, IH); 5.79 (s, IH); 5.0 (d, IH); 4.54 (s, IH); 4.30 (s, IH); 4.27 (d, IH); 4.20 (s, IH); 4.18 (d, IH); 3.80 (s, 3H); 3.78 (s, 3H); 3.43-3.40 (m, 2H); 3.18-2.77 (m, 4H); 2.66-2.49 (m, 3H); 2.37 (s, 3H); 2.34-2.20 (m, IH); 2.26 (s, 3H); 2.21 (s, 3H); 2.05 (s, 3H). 13C-NMR (75 MHz, CDCI3): δ 171.4, 168.6, 153.7, 148.0, 145.6, 142.9, 141.0, 140.2, 131.1, 130.6, 130.5, 129.2, 127.0, 120.6, 120.5, 118.2, 113.6, 111.9, 111.6, 1 10.0, 102.0, 100.3, 62.3, 61.2, 60.5, 60.2, 59.4, 55.7, 54.6, 54.5, 41.8, 41.4, 39.7, 39.2, 31.5, 29.6, 23.8, 22.6, 21.5, 20.5, 15.8, 14.4, 9.7. ESI-MS m/z: Calcd. for C42H43N5O9S: 793.3 Found (M+H+): 794.7. Compound 3: -Η-NMR (300 MHz, CDC13): δ
7.85(s, IH); 7.45-7.36(m, 5H); 7.01(t, IH);
6.91(t, IH); 6.65-6.63(m, 2H); 5.87(s, IH);
5.77(s, IH); 5.63(s, IH); 5.13(s, 2H); 5.05(d, 1H)' 4-53(s' 1H); 4.27-4.19(m, 4H); 3.80(s,
3H); 3.46-3.39(m 2H); 3.06-2.79(m, 4H); 2.68-2.50(m 2H); 2.42-2.20(m, IH); 2.36(s, 3H); 2.27(s, 3H); 2.20(s, 3H); 2.03(s, 3H). ESI-MS m/z: Calcd. for C48H47N5O9S: 869.3 Found (M+H+): 870.3.
Compound 4: iH-NMR (300 MHz, CDCI3): δ
7.36(s, IH); 7.44-7.25(m 5H); 7.13(d, IH);
6.91(d, IH); 6.82(dd, IH); 6.65(s, IH); 6.21(d,
IH); 6.01(d, IH); 5.80(s, IH); 5.08(d, IH);
5.03(s, 2H); 4.55(s, IH); 4.31(s, IH); 4.27(d, IH); 4.20-4.10(m, 3H); 3.81(s, 3H); 3.44-
3.40(m 2H); 3.18-2.77(m, 4H); 2.60-2.46(m, 2H); 2.37(s, 3H); 2.35-
2.19(m, IH); 2.26(s, 3H); 2.21(s, 3H); 2.06(s, 3H).
ESI-MS m/z: Calcd. for C48H47N5O9S: 869.3 Found (M+H+): 870.3.
Compound 5: *H-NMR (300 MHz, CDCI3): δ
7.63(s, IH); 7.15-7.11(m, 2H); 6.91(dd, IH); 6.65(s, IH); 6.21(d, IH); 6.01(s, IH); 5.78(s,
IH); 5.07(d, IH); 4.54(s, IH); 4.31(s, IH);
4.27(d, IH); 4.21-4.16(m, 2H); 3.81(s, 3H); 3.44-3.40(m, 2H); 3.17-2.77(m, 4H); 2.66- 2.46(m, 3H); 2.31(s, 6H); 2.26(s, 3H); 2.21(s, 3H); 2.06(s, 3H). ESI-MS m/z: Calcd. for C42H43N5O8S: 777.3 Found (M+Na+): 800.7. Compound 6: *H-NMR (300 MHz, CDC13): δ 7.66(s, IH); 6.95(d, IH); 6.64(s, 2H); 6.56(dd, IH); 6.15(s, IH); 5.97(s, IH); 5.81(s, IH); 5.06(d, IH); 4.53(s, IH); 4.29(s, IH); 4.26(d, IH); 4.19(s, IH); 4.17(d, IH); 3.80(s, 3H); 4.41- 3.39(m, 2H); 3.12-2.73(m, 4H); 2.55-2.27(m,
3H); 2.36(s, 3H); 2.25(s, 3H); 2.20(s, 3H); 2.04(s, 3H). ESI-MS m/z: Calcd. for C41H41N5O9S: 779.3 Found (M+H+): 780.3.
Compound 7: Η-NMR (300 MHz, CDCI3): δ 7.75(s, IH); 7.26(dd, IH); 6.93(dd, IH); 6.76(ddd, IH); 6.65(s, IH); 6.22(d, IH); 6.01(d, lH);5.79(s, IH); 5.08(d, IH); 4.55(s, IH); 4.31(s, IH); 4.25(d, IH); 4.20(s, IH); 4.18(dd, IH); 3.80(s, 3H); 3.43-3.40(m, 2H); 3.18-
2.77(m, 4H); 2.64-2.50(m, 3H); 2.36(s, 3H); 2.26(s, 3H); 2.21(s, 3H); 2.06(s, 3H). ESI-MS m/z: Calcd. for C41H40FN5O8S: 781.3 Found (M+H+): 782.3.
Compound 8: Η-NMR (300 MHz, CDCI3): δ 6.93(d, IH); 6.80(s, IH); 6.73(s, IH); 6.67(dd, IH); 6.46(s, IH); 6.20(s, IH); 6.06(s, IH); 5.72(s, IH); 4.96(d, IH); 4.45(s, IH); 4.37(d, IH); 4.25(d, IH); 4.05-4.01(m, 2H); 3.79(s, 3H); 3.63(d, IH); 3.39(d, IH); 3.03-3.91(m, 2H); 2.76-2.34(m, 5H); 3.30(s, 3H); 2.28(s, 3H); 2.21(s, 3H); 2.18(s, 3H); 2.04(s, 3H). ESI-MS m/z: Calcd. for C42H43N5O9S: 793.3 Found (M+H+): 794.3.
Compound 9: Η-NMR (300 MHz, CDCb): δ
7.63(s, IH); 7.24(d, IH); 6.75(d, IH); 6.66(dd, IH); 6.65(s, IH); 6.20(s, IH); 6.00(s, IH); 5.79(s, IH); 5.07(d, IH); 4.54(s, IH); 4.31(s, IH); 4.27(d, IH); 4.20(d, IH); 4.17(dd, IH); 3.80(s, 3H); 3.71(s, 3H); 3.43-
3.40(m, 2H); 3.16-2.78(m, 4H); 2.64-2.49(m, 3H); 2.36(s, 3H); 2.25(s, •3H); 2.21(s, 3H); 2.06(s, 3H). ESI-MS m/z: Calcd. for C42H43N5O9S: 793.3 Found (M+H+): 794.3.
Compound 10: H-NMR (300 MHz, CDCI3): δ 7.76 (s, IH); 7.14 (dd, IH); 7.00 (dd, IH); 6.81 (ddd, IH); 6.65 (s, IH); 6.21 (d, IH); 6.00 (d, IH); 5.79 (s, IH); 5.07 (d, IH); 4.55 (s, IH); 4.31 (s, IH); 4.27 (dd, IH); 4.20 (d, IH); 4.18 (dd, IH); 3.80(s, 3H); 3.44-3.40 (m, 2H); 3.16-2.77 (m, 4H); 2.64-2.44 (m, 3H); 2.37 (s, 3H); 2.26 (s,
3H); 2.21 (s, 3H); 2.05 (s, 3H).
ESI-MS m/z: Calcd. for C41H40FN5O8S: 781.3 Found (M+H+): 782.1.
Compound 11: Η-NMR (300 MHz, CDCI3): δ 7.48 (s, IH); 7.22 (d, IH); 6.96-6.88 (m, , 2H); 6.65 (s, IH); 6.15 (d, IH); 6.04 (d, IH); 5.78 (s, IH); 5.09 (d, IH); 4.55 (s, IH); 4.34 (s, IH); 4.28-4.20 (m, 3H); 3.81 (s, 3H); 3.48 (d, IH); 3.42 (d, IH); 3.12-2.78 (m, 4H); 2.69-2.43 (m, 3H); 2.37 (s, 3H); 2.36 (s, 3H); 2.28 (s, -3H); 2.21 (s, 3H); 2.06 (s, 3H). ESI-MS m/z: Calcd. for C42H43N5O8S: 777.3 Found (M+H+): 778.3
Compound 12 (first isomer): H-NMR (300 MHz, CDCI3): δ 7.68 (s, IH); 7.05 (d, IH); 6.63- 6.57 (m, , 3H); 6.22 (d, IH); 6.02 (d, IH); 5.73 (s, IH); 5.12 (d, IH); 4.58 (s, IH); 4.36 (s, IH); 4.34-4.22 (m, 3H); 3.80 (s, 3H); 3.47-3.42 (m, 2H); 3.05-2.86 (m, 2H); 2.67-2.35 (m, 2H); 2.32-2.05 (m, 3H); 2.31 (s, 3H); 2.28 (s, 3H); 2.15 (s, 3H); 2.03 (s, 3H); 1.07 (d, 3H). ESI-MS m/z: Calcd. for C42H43N5O9S: 793.2 Found (M+H+): 794.2.
Compound 13 (second isomer): H-NMR (300 MHz, CDCI3): δ 7.54 (s, IH); 7.08 (d, IH); 6.73 (d, IH); 6.63 (dd, IH); 6.57 (s, IH); 6.20 (d, IH); 6.00 (d, IH); 5.74 (s, IH); 5.02 (d, IH); 4.60 (s, IH); 4.33 (s, IH); 4.27 (d, IH); 4.22 (d, IH); 4.12 (dd, IH); 3.80 (s, 3H); 3.44-3.32 (m, 3H); 3.05-2.89 (m, 2H); 2.49-2.03 (m, 4H); 2.32 (s, 3H); 2.24 (s, 3H); 2.18 (s, 3H); 2.07 (s, 3H); 1.21 (d, 3H). ESI-MS m/z: Calcd. for C42H43N5O9S: 793.2 Found (M+H+): 794.2.
Compound 14 (first isomer): H-NMR (300
MHz, CDCI3): δ 7.98 (s, IH); 7.40 (dd, IH); 6.18 (t, IH); 7.04 (t, IH); 6.82 (s, IH); 6.16 (d, IH); 6.01 (d, IH); 5.74 (s, IH); 4.95 (d, IH); 4.64 (s,
IH); 4.40 (s, IH); 4.30-4.24 (m, 3H); 3.62 (s, 3H); 3.56 (d, IH); 3.50 (d, IH); 3.12-2.89 (m, 4H);
2.75-2.51 (m, 3H); 2.43 (s, 3H); 2.38-2.30 (m, 2H); 2.26 (s, 3H); 2.19 (s,
3H); 2.08 (s, 3H); 2.00 (s, 3H). ESI-MS m/z: Calcd. for C42H43N5O8S: 777.2 Found (M+H+): 778.2.
Compound 15 (second isomer): H-NMR (300 MHz, CDCI3): δ 7.36 (d, IH); 7.15-7.06 (m, 2H); 7.01 (ddd, IH); 6.93 (s, IH); 6.47 (s, IH); 6.22 (d, IH); 6.09 (d, IH); 5.72 (s, IH); 4.97 (d, IH); 4.46 (s, IH); 4.40 (d, IH); 4.26 (dd, IH); 4.03 (dd, IH); 4.02 (s, IH); 3.80 (s, 3H); 3.63 (d, IH); 3.39 (d, IH); 3.00-2.92 (m, 2H); 277-2.54 (m, 4H); 2.30 (s, 3H); 2.26- 2.25 (m, 2H); 2.23 (s, 6H); 2.19 (s, 3H); 2.05 (s, 3H). ESI-MS m/z: Calcd. for C42H43N5O8S: 777.2 Found (M+H+): 778.2.
Compound 16 (first isomer): *H-NMR (300 MHz, CDCI3): δ 7.82 (s, IH); 7.14 (dd, IH); 6.96 (dd, IH); 6.82 (ddd, IH); 6.61 (s, IH); 6.23 (d, IH); 6.02 (d, IH); 5.72 (s, IH); 5.12 (d, IH); 4.59 (s, IH); 4.37 (s, IH); 4.32-4.25 (m, 3H); 3.80 (s, 3H); 3.48-3.43 (m, 2H); 3.05-2.86 (m, 4H); 2.78-
2.70 (m, IH); 2.60-2.34 (m, 3H); 2.31 (s, 3H); 2.27 (s, 3H); 2.16 (s, 3H);
2.03 (s, 3H); 1.12 (d, 3H).
ESI-MS m/z: Calcd. for C42H42FN5O8S: 795.3 Found (M+H+): 796.2.
Compound 17 (second isomer): Η-NMR (300 MHz, CDCI3): δ 7.65 (s, IH); 7.18 (dd, IH); 6.99 (dd, IH); 6.83 (ddd, IH); 6.58 (s, IH); 6.22 (d, IH); 6.01 (d, IH); 5.74 (s, IH); 5.03 (d, IH); 4.61 (s, IH); 4.34 (s, IH); 4.27 (d, IH); 4.22 (d, IH); 4.14-4.10 (m, IH); 3.80(s, 3H); 3.44 (d, 2H); 3.38-3.30 (m, IH); 3.06-2.99 (m, 2H); 2.50 (dd, IH); 2.43 (d, IH); 2.32 (s, 3H); 2.24 (s, 3H); 2.18 (s, 3H); 2.16-2.1 1 (m, 2H); 2.08 (s, 3H); 1.20 (d, 3H). ESI-MS m/z: Calcd. for C42H42FN5O8S: 795.3 Found (M+H+): 796.2. Compound 18 (first isomer): H-NMR (300 MHz, CDCb): δ 7.83 (s, IH); 7.34 (d, IH); 7.24 (d, IH); 7.09 (ddd, IH); 7.00 (ddd, IH); 6.62 (s, IH); 6.24 (d, IH); 6.03 (d, IH); 5.73 (s, IH); 5.13 (d, IH); 4.59 (s, IH); 4.38 (s, IH); 4.33-4.27 (m, 3H); 3.80(s, 3H); 3.48-3.43 (m, 2H); 3.06-2.87 (m,
2H); 2.78-2.72 (m, IH); 2.61-2.24 (m, 4H); 2.32 (s, 3H); 2.27 (s, -3H);
2.16 (s, 3H); 2.03 (s, 3H); 1.13 (d, 3H).
ESI-MS m/z: Calcd. for C42H43N5O8S: 777.2 Found (M+H+): 778.2.
Compound 19 (second isomer): Η-NMR (300 MHz, CDCI3): δ 7.66 (s, IH); 7.37 (d, IH); 7.28 (d, IH); 7.10 (ddd, IH); 7.00 (ddd, IH); 6.58 (s, IH); 6.24 (d, IH); 6.02 (d, IH); 5.75 (s, IH); 5.03 (d, IH); 4.61 (s, IH); 4.35 (s, IH); 4.28 (dd, IH); 4.23 (d, IH); 4.15-4.08 (m, IH); 3.81 (s, 3H); 3.44 (d, 2H); 3.38-3.32 (m, IH); 3.07-2.90 (m, 2H); 2.58 (dd, IH); 2.45 (d, IH); 2.33 (s, 3H); 2.27-2.12 (m, 2H); 2.24 (s, -3H); 2.19 (s, 3H); 2.08 (s, 3H); 1.20 (d, 3H). ESI-MS m/z: Calcd. for C42H43N5O8S: 777.2 Found (M+H+): 778.2.
Method 2:
Compound 20: Η-NMR (300 MHz, CDCI3): δ 7.70 (s, IH); 7.38 (d, IH); 7.25 (d, IH); 7.10 (ddd, IH); 7.02 (ddd, IH); 7.01 (s, IH); 6.24 (d, IH); 6.03 (d, IH); 5.09 (d, IH); 4.44 (s, IH); 4.33 (s, IH); 4.22-4.18 (m, 2H) 3.81 (d, IH); 3.77 (s, 3H); 3.48-3.44 (m, 2H); 3.19-2.81 (m, 4H); 2.70-
2.48 (m, 3H); 2.62 (t, 2H); 2.37 (s, 3H); 2.34-2.15 (m, 2H); 2.29 (s, -3H);
2.18 (s, 3H); 2.04 (s, 3H); 1.95-1.82 (m, 2H); 1.10 (t, 3H).
ESI-MS m/z: Calcd. for C45H47N5O9S: 833.3 Found (M+H+): 834.2. Compound 21: *H-NMR (300 MHz, CDC13): δ 7.70 (s, IH); 7.38 (d, IH); 7.24 (d, IH); 7.09 (ddd> 1H); 7-00 (ddd' 1H)' 6-99 (s> 1H); 6-22 (d>
IH); 6.02 (d, IH); 5.08 (d, IH); 4.48 (s, IH); 4.32 (s, IH); 4.25-4.21 (m, 2H) 3.81 (s, 3H); 3.46-3.44 (m, 2H); 3.18-2.79 (m, 4H); 2.72-2.43 (m, 3H);
2.36 (s, 3H); 2.31 (s, -3H); 2.20 (s, 3H); 2.08 (s, 3H); 1.54 (s, 9H).
ESI-MS m/z: Calcd. for C46H49N5O10S: 863.3 Found (M+H+): 864.2.
Compound 22: Η-NMR (300 MHz, CDCI3): δ 7.70 (s, IH); 7.38 (d, IH); 7.26-6.99 (m, 5H); 6.24 (d, IH); 6.03 (d, IH); 5.09 (d, IH); 5.00 (dd, IH); 4.71 (dd, IH); 4.48 (s, IH); 4.33 (s, IH); 4.24-4.21 (m, 2H) 3.95 (d, IH); 3.81 (s, 3H); 3.48-3.45 (m, 2H); 3.18-2.81 (m, 4H); 2.72-2.45
(m, 3H); 2.38 (s, 3H); 2.34 (s, -3H); 2.30-2.11 (m, 2H); 2.20 (s, 3H); 2.08
(s, 3H).
ESI-MS m/z: Calcd. for C44H43N5O10S: 833.3 Found (M+H+): 834.2.
Compound 23: Η-NMR (300 MHz, CDCI3): δ 7.38 (d, IH); 7.20-7.13 (m, 3H); 7.07-6.98 (m, 2H); 6.88 (s, IH); 6.66 (s, IH); 6.18 (d, IH); 6.12 (d, IH); 4.95 (dd, IH); 4.79 (d, IH); 4.78 (dd, IH); 4.68 (dd, IH); 4.46 (dd, IH); 4.40 (d, IH); 4.34- 4.18 (m, 3H) 3.97 (d, IH); 3.89 (d, IH); 3.85 (s,
3H); 3.61 (d, IH); 3.41 (d, IH); 3.17-2.98 (m, 3H); 2.76-2.42 (m, 4H);
2.37 (s, 3H); 2.32 (s, 3H); 2.18 (s, 3H); 2.13 (s, 3H).
ESI-MS m/z: Calcd. for C47H45N5O12S: 903.2 Found (M+Na+): 926.1
Method 3: Compound 24: Η-NMR (300 MHz, CDC13): δ 7.71 (s, IH); 7.38 (d, IH); 7.24 (d, IH); 7.09 (ddd, IH); 7.00 (ddd, IH); 6.86 (s, IH); 6.22 (d, IH); 6.16-6.04 (m, IH); 6.02 (d, IH); 5.47 (dd, IH); 5.26 (dd, IH); 5.09 (d, IH); 4.83 (dd, IH); 4.52 (s, IH); 4.36 (dd, IH); 4.32 (s, IH); 4.24-
4.19 (m, 3H); 3.84 (s, 3H); 3.45-3.41 (m, 2H); 3.18-2.79 (m, 4H); 2.73-
2.47 (m, 3H); 2.33 (s, 3H); 2.31-2.26 (m, IH); 2.24 (s, 3H); 2.21 (s, 3H);
2.06 (s, 3H); 2.03 (d, IH).
ESI-MS m/z: Calcd. for C44H45N5O8S: 803.3 Found (M+H+): 804.3
Compound 25: *H-NMR (300 MHz, CDCI3): δ 7.39 (d, IH); 7.25-7.23 (m, IH); 7.096.98 (m, 3H); 6.80 (s, IH); 6.14-6.00 (m, IH); 6.10 (d, IH); 6.02 (d, IH); 5.60-5.40 (m, IH); 5.45 (dd, IH); 5.25 (dd, IH); 5.02-4.95 (m, 2H); 4.81 (dd, IH); 4.73-4.62 (m, IH); 4.55 (s, IH); 4.37-4.16 (m,
6H); 3.84 (s, 3H); 3.51 (d, IH); 3.45-3.38 (m, 2H); 3.05-2.89 (m, 3H);
2.70-2.50 (m, 3H); 2.33-2.16 (m, 2H); 2.31 (s, 3H); 2.23 (s, -3H); 2.21 (s,
3H); 2.03 (s, 3H).
ESI-MS m/z: Calcd. for C47H49N5O8S: 843.3 Found (M+H+): 844.2
Method 4:
Compound 26: H-NMR (300 MHz, CDC13): δ 7.70 (s, IH); 7.38 (d, IH); 7.24 (d, IH); 7.08 (t, IH); 7.00 (t, IH); 6.67 (s, IH); 6.20 (d, IH); 5.99 (d, IH); 5.74 (s, IH); 5.20 (d, IH); 4.82 (s, IH); 4.347-4.38 (m, 3H); 4.16-4.10 (m, 2H); 3.81 (s, 3H); 3.49 (d, IH); 3.22-3.13 (m, 2H); 3.00 (d,
IH); 2.88-2.79 (m, 2H); 2.71-2.52 (m, 3H); 2.37 (s, 3H); 2.28-2.24 (m, IH); 2.25 (s, 3H); 2.19 (s, 3H); 2.05 (s, 3H).
13C-NMR (75 MHz, CDCI3): δ 171.4, 168.7, 147.8, 145.4, 142.8, 141.0, 140.6, 135.4, 131.2, 130.9, 129.0, 126.8, 121.8, 121.3, 120.9, 119.1, 118.3, 118.1 , 115.5, 112.8, 110.8, 110.1, 101.7, 81.9, 62.3, 61.8, 60.2.57.6, 57.4, 55.8, 54.9, 42.1 , 41.2, 39.7, 39.2, 31.5, 23.5, 22.6, 21.5, 20.5, 15.8, 14.0, 9.6. ESI-MS m/z: Calcd. for C40H42N4O9S: 754.3 Found (M-H2O+H+): 737.2.
Compound 27: Η-NMR (300 MHz, CDCI3): δ
7.59 (s, IH); 7.13 (d, IH); 6.81 (s, IH); 6.73
(dd, IH); 6.67 (s, IH); 6.19 (d, IH); 5.99 (d,
IH); 5.74 (s, IH); 5.19 (d, IH); 4.82 (s, IH);
4.49-4.47 (m, 2H); 4.16-4.09 (m, 2H); 3.81 (s, 3H); 3.79 (s, 3H); 3.50-3.45 (m, 2H); 3.24-3.13
(m, 2H); 3.02 (d, IH); 2.88-2.79 (m, 2H); 2.67-2.48 (m, 3H); 2.37 (s, 3H);
2.30-2.24 (m, IH); 2.25 (s, 3H); 2.19 (s, 3H); 2.04 (s, 3H).
13C-NMR (75 MHz, CDCI3): δ 171.6, 154.0, 148.1 , 145.6, 143.1, 141.3,
140.9, 131.9, 131.4, 130.8, 129.3, 127.4, 121.5, 121.2, 1 15.7, 113.1,
1 12.1, 1 11.8, 110.1 , 102.0, 100.6, 82.1 , 62.6, 62.0, 60.5, 57.9, 57.6,
56.1, 56.0, 55.2, 42.4, 41.5, 40.0, 39.4, 31.8, 29.9, 23.8, 22.8, 21.8,
20.8, 16.0, 14.6, 9.9.
ESI-MS m/z: Calcd. for C40H42N4O9S: 784.4 Found (M-H2O+H+): 767.2. Compound 28: *H-NMR (300 MHz, CDC13): δ 7.81 (s, IH); 7.43-7.36 (m, 5H); 7.01 (d, IH); 6.91 (t, IH); 6.66 (s, IH); 6.63 (d, IH); 5.84 (s, IH); 5.75 (s, IH); 5.60 (s, IH); 5.20-5.09 (m, 3H); 4.78 (s, IH); 4.49 (d, IH); 4.44 (s, IH); 4.16 (s, IH); 4.14-4.12 (m, IH); 3.81 (s, 3H);
3.52 (d, IH); 3.47 (s, 2H); 3.22-2.80 (m, 5H); 2.68-2.51 (m, 2H); 2.36 (s, 3H); 2.39-2.21 (m, IH); 2.27 (s, 3H); 2.18 (s, 3H); 2.02 (s, 3H). 13C-NMR (75 MHz, CDCI3): δ 171.4, 148.0, 145.6, 145.2, 143.1 , 141.1, 140.8, 137.3, 131.6, 130.7, 129.3, 128.8, 128.5, 128.2, 128.0, 126.2, 121.4, 121.3, 119.8, 118.1, 115.5, 113.0, 111.8, 111.0, 103.8, 101.8, 82.1, 70.6, 62.9, 61.9, 60.5, 58.0, 57.7, 56.1, 55.1, 42.3, 41.5, 40.0, 39.5, 29.9, 23.9, 22.0, 20.7, 16.0, 9.8. ESI-MS m/z: Calcd. for C47H48N4O10S: 860.3 Found (M-H2O+H+): 843.3
Compound 29: Η-NMR (300 MHz, CDCI3): δ
7.59 (s, IH); 7.44-7.25 (m, 5H); 7.13 (d, IH); 6.91 (s, IH); 6.82 (d, IH); 6.66 (s, IH); 6.19 (s, IH); 5.98 (s, IH); 5.75 (s, IH); 5.19 (d, IH); 5.03 (s, 2H); 4.82 (s, IH); 4.49-4.47 (m, 2H); 4.17-4.09 (m, 2H); 3.81 (s, 3H); 3.49-3.47 (m, 2H); 3.24-2.80 (m, 5H); 2.64-2.50 (m, 3H); 2.37 (s, 3H); 2.25 (s, 3H); 2.19 (s, 3H); 2.05 (s, 3H).
13C-NMR (75 MHz, CDCI3): δ 171.4, 168.6, 153.2, 148.1, 145.7, 143.1, 141.3, 140.9, 137.9, 131.0, 129.7, 128.6, 127.9, 127.8, 127.4, 121.2, 115.7, 112.8, 111.8, 110.2, 102.3, 102.0, 82.1, 71.1, 62.5, 62.0, 60.5, 58.0, 57.6, 56.1, 55.2, 42.4, 41.5, 40.0, 39.4, 32.1 , 29.9, 29.5, 23.8, 22.9, 21.8, 20.8, 16.0, 14.6, 9.9. ESI-MS m/z: Calcd. for C47H48N4O10S: 860.3 Found (M-H2O+H+): 843.3 Compound 30: H-NMR (300 MHz, CDC13): δ 7.61 (s, IH); 7.16-7.11 (m, 2H); 6.91 (d, IH); 6.67 (s, IH); 6.20 (d, IH); 5.99 (d, IH); 5.75 (s, IH); 5.20 (d, IH); 4.82 (s, IH); 4.49 (d, IH); 4.35 (s, IH); 4.16 (d, 2H); 4.11 (dd, IH); 3.81 (s, 3H); 3.48 (s, IH); 3.23-2.79 (m, 5H); 2.67-2.47 (m, 3H); 2.37 (s, 6H); 2.25 (s, 3H); 2.19 (s, 3H); 2.05 (s, 3H).
13C-NMR (75 MHz, CDCI3): δ 174.5, 171.6, 168.9, 148.1, 145.6, 143.1, 141.3, 140.9, 134.0, 131.2, 128.6, 127.3, 123.6, 121.5, 121.2, 118.3, 115.7, 109.9, 102.0, 82.1, 62.6, 62.0, 60.5, 57.9, 57.7, 56.1, 56.2, 42.4, 41.5, 40.0, 39.4, 29.9, 23.8, 21.7, 21.6, 20.8, 16.0, 9.9. ESI-MS m/z: Calcd. for C41H44N4O9S: 768.3 Found (M-H2O+H+): 751.3.
Compound 31: Η-NMR (300 MHz, CDCI3): δ 7.60 (s, IH); 7.00 (d, IH); 6.69 (d, IH); 6.66 (s, IH); 6.69 (dd, IH); 6.16 (s, IH); 5.96 (s, IH); 5.78 (s, IH); 5.19 (d, IH); 4.82 (s, IH); 4.49 (d, IH); 4.46 (s, IH); 4.17 (d, IH); 4.10 (d, IH); 3.81 (s, 3H); 3.72-3.59 (m, 2H); 3.64 (d, 2H); 3.50 (d, IH); 3.23-2.76 (m, 4H); 2.55-2.29 (m, 3H); 2.37 (s, 3H); 2.25 (s, 3H); 2.19 (s, 3H); 2.03 (s. 3H). 13C-NMR (75 MHz, CDCI3): δ 171.3, 166.9, 149.2, 147.9, 145.4, 142.9, 141.0, 140.7, 131.2, 130.7, 127.5, 121.2, 120.9, 115.5, 111.5, 103.1, 101.8, 81.9, 62.3, 61.8, 60.3, 57.7, 57.4, 55.8, 54.9, 42.1.41.2, 39.6, 39.1, 29.6, 23.5, 22.6, 21.4, 20.5, 15.8, 14.1, 9.6. ESI-MS m/z: Calcd. for C40H42N4O10S: 770.3 Found (M-H2O+H+): 753.3. Compound 32: Η-NMR (300 MHz, CDC13): δ 7.72 (s, IH); 7.27 (dd, IH); 6.94 (dd, IH); 6.76 (ddd, IH); 6.66 (s, IH); 6.20 (s, IH); 5.99 (s, IH); 5.75 (s, IH); 5.19 (d, IH); 4.83 (s, IH); 4.49 (d, IH); 4.16-4.09 (m, 2H); 3.81 (s, 3H); 3.50-3.48 (m, IH); 3.48 (s, IH); 3.22-2.79 (m, 5H); 2.65-2.51 (m, 3H); 2.37 (s, 3H); 2.25 (s, 3H); 2.19 (s, 3H); 2.05 (s, 3H).
13C-NMR (75 MHz, CDCI3): δ 171.5, 169.0, 148.1, 145.7, 143.1, 141.3, 140.9, 131.5, 129.3, 123.7, 121.5, 121.1, 1 19.3, 119.1, 118.3, 115.7, 110.4, 108.2, 107.8, 102.0, 97.8, 97.5, 82.1, 62.4, 62.1 , 60.5, 57.9, 57.6, 56.1, 55.1, 42.4, 41.5, 39.9, 39.4, 29.9, 23.8, 21.7, 20.8, 16.0, 9.9. ESI-MS m/z: Calcd. for C40H41FN4O9S: 772.3 Found (M-H2O+H+): 755.3.
Compound 33: Η-NMR (300 MHz, CDCI3): δ 6.85 (d, IH); 6.80 (s, IH); 6.71-6.64 (m, 2H); 6.48 (s, IH); 6.18 (s, IH); 6.02 (s, IH); 5.74 (s, IH); 5.03 (d, IH); 4.88 (d, IH); 4.39 (d, IH); 4.36 (s, IH); 4.16 (d, IH); 3.98 (ddm IH); 3.80 (s, 3H); 3.71 (d, IH); 3.48 (s, IH); 3.22 (d, IH); 2.93-2.83 (m, 2H); 2.73-2.39 (m, 4H); 2.29 (s, 3H); 2.28-2.05 (m, IH); 2.26 (s, 3H); 2.22 (s, 3H); 2.18 (s, 3H); 2.03 (s, 3H).
13C-NMR (75 MHz, CDCI3): δ 168.9, 149.3, 147.6, 145.5, 142.7, 141.6, 140.7, 131.3, 131.1, 129.3, 126.9, 121.2, 1 15.7, 11 1.9, 111.0, 110.7, 103.1 , 102.0, 83.4, 69.8, 63.7, 60.2, 58.5, 57.7, 55.1 , 54.7, 59.5, 43.1, 41.4, 40.6, 35.0, 29.6, 24.6, 22.6, 21.1, 20.2, 15.5, 9.7. ESI-MS m/z: Calcd. for C41H44N4O10S: 784.3 Found (M-H2O+H+): 767.3. Compound 34: Η-NMR (300 MHz, CDC13) δ 7.58 (s, IH); 7.23 (d, IH); 6.76 (d, IH) 6.67 (dd, IH); 6.66 (s, IH); 6.19 (d, IH) 5.98 (d, IH); 5.74 (s, IH); 5.20 (d, IH); 4.82 (s, IH); 4.49 (d, IH); 4.47 (s, IH); 4.16 (d, IH); 4.10 (dd, IH); 3.81 (s, 3H); 3.78 (s, 3H);
3.51-3.47 (m, IH); 3.22-2.80 (m, 5H); 2.65.2.50 (m, 3H); 2.36 (s, 3H); 2.25 (s, 3H); 2.19 (s, 3H); 2.05 (s, 3H).
"C-NMR (75 MHz, CDCI3): δ 171.7, 168.9, 156.5, 148.1, 145.6, 143.1, 141.3, 140.9, 136.5, 131.5, 129.8, 129.3, 121.6, 121.5, 121.2, 119.2, 1 15.8, 113.1 , 110.3, 109.3, 102.2, 94.9, 82.2, 62.5, 62.0, 60.5, 57.9, 57.7, 56.1 , 55.8, 55.2, 42.3, 41.5, 40.0, 39.5, 29.9, 23.8, 21.8, 20.8, 16.0, 9.9. ESI-MS m/z: Calcd. for C41H44N4O10S: 784.3 Found (M-H2O+H+): 767.3.
Compound 35: iH-NMR (300 MHz, CDCI3): δ 7.73 (s, IH); 7.15 (dd, IH); 7.00 (dd, IH); 6.81 (ddd, IH); 6.67 (s, IH); 6.20 (d, IH); 5.99 (d, IH); 5.76(s, IH); 5.19 (d, IH); 4.83 (s, IH); 4.49 (d, 2H); 4.17 (d, IH); 4.12 (dd, IH); 3.81(s, 3H); 3.65-3.64 (m, IH); 3.50 (d, IH); 3.24-2.12 (m, 2H); 3.00 (d, IH); 2.89-2.80 (m, 2H); 2.66-2.45
(m, 3H); 2.37 (s, 3H); 2.25 (s, 3H); 2.20 (s, 3H); 2.05 (s, 3H).
13C-NMR (75 MHz, CDCI3): δ 171.4, 169.0, 159.4, 156.3, 148.1 , 145.7,
143.1 , 141.3, 140.9, 138.0, 132.2, 129.4, 127.4, 127.3, 121.4, 121.4,
121.2, 118.2, 1 15.7, 113.1, 1 1 1.8, 111.7, 110.4, 110.1 , 103.7, 103.4, 102.0, 82.1, 62.5, 62.1, 60.5, 57.9, 57.6, 56.1 , 55.1, 42.3, 41.4, 39.9, 39.4, 29.9, 23.8, 21.7, 20.8, 16.0, 9.9.
ESI-MS m/z: Calcd. for C40H41FN4O9S: 772.2 Found (M-H2O+H+): 755.2. Compound 36: Η-NMR (300 MHz, CDC13): δ 7.47 (s, IH); 7.22 (d, IH); 6.95-6.87 (m, 2H); 6.66 (s, IH); 6.13 (d, IH); 6.01 (d, IH); 5.76 (s, IH); 5.20 (d, IH); 4.84 (s, IH); 4.49 (d, IH); 4.46 (s, IH); 4.18-4.14 (m, 2H); 3.81 (s, 3H); 3.54 (d, IH); 3.48(s, IH); 3.22 (d, IH); 3.20- 2.80 (m, 4H); 2.70-2.42 (m, 3H); 2.36( s, 6H); 2.27 (s, 3H); 2.18 (s, 3H); 2.05 (s, 3H).
13C-NMR (75 MHz, CDCI3): δ 171.3, 169.0, 148.0, 145.6, 143.1, 141.4, 140.9, 135.3, 131.5, 131.1, 130.7, 129.4, 126.6, 122.8, 121.8, 121.3, 1 19.9, 119.6, 118.1 , 116.3, 115.8, 102.0, 82.1 , 62.1, 60.5, 58.0, 57.8, 56.1, 55.1 , 42.4, 41.5, 40.1 , 39.6, 29.9, 23.9, 21.9, 20.7, 16.6, 16.0, 9.9. ESI-MS m/z: Calcd. for C41H44N4O9S: 768.2 Found (M-H2O+H+): 751.2.
Compound 37 (first isomer): Η-NMR (300 MHz, CDCls): δ 7.70 (s, IH); 7.06 (d, IH); 6.67- 6.61 (m, 3H); 6.20 (d, IH); 5.98 (d, IH); 5.70 (s, IH); 5.20 (d, IH); 4.86 (s, IH); 4.53 (d, IH); 4.48 (s, IH); 4.18(s, IH); 3.80(s, 3H); 3.72-3.54 (m, 4H); 3.24-3.22 (m, IH); 3.01-2.56 (m, 5H); 2.31 (s, 3H); 2.27 (s, 3H); 2.15 (s, 3H); 2.02 (s.
3H); 1.10 (d, 3H).
ESI-MS m/z: Calcd. for C41H44N4O10S: 784.3 Found (M-H2O+H+): 767.3.
Compound 38 (second isomer): Η-NMR (300 MHz, CDCI3): δ 7.51 (s, IH); 7.10 (d, IH); 6.75 (d, IH); 6.64 (dd, IH); 6.59 (s, IH); 6.19 (d, IH); 5.97 (d, IH); 5.71 (s, IH); 5.15 (d, IH); 4.84 (s, IH); 4.53-4.50 (m, 2H); 4.16 (s, IH); 4.04 (dd, IH); 3.80(s, 3H); 3.65-3.63 (m, IH); 3.51-3.49 (m, IH); 3.40-2.36 (m, IH); 3.24-3.21 (m, IH); 3.03-2.84 (m, 2H); 2.50-2.41 (m, 2H); 2.32 (s, 3H); 2.23 (s, 3H); 2.16 (s, 3H); 2.06 (s. 3H); 1.20 (d,
3H).
ESI-MS m/z: Calcd. for C41H44N4O10S: 784.3 Found (M-H2O+H+): 767.3.
Compound 39 (first isomer): Η-NMR (300 MHz, CDCI3): δ 8.09 (s, IH); 7.41 (d, IH); 7.17 (t, IH); 7.03 (t, IH); 6.87 (d, IH); 6.83 (s, IH); 6.13 (d, IH); 5.98 (d, IH); 5.69 (s, IH); 5.02 (d, IH); 4.88 (s, IH); 4.55-4.16 (m, 4H); 3.64-3.56 (m, IH); 3.61 (s, 3H); 3.31-3.29 (m, IH); 3.22-2.80
(m, 3H); 2.68-2.46 (m, 3H); 2.41 (s, 3H); 2.27 (s, 3H); 2.20 (s, 3H); 2.07 (s, 3H); 1.99 (s, 3H). ESI-MS m/z: Calcd. for C41H44N4O9S: 768.2 Found (M-H O+H+): 751.2.
Compound 40 (second isomer): H-NMR (300 MHz, CDCI3): δ 7.36 (d, IH); 7.12-7.05 (m, 2H); 7.00 (ddd, IH); 6.92 (s, IH); 6.48 (s, IH); 6.20 (d, IH); 6.06 (d, IH); 5.70 (s, IH); 5.04 (d, IH); 4.88 (s, IH); 4.39-4.36 (m, IH); 4.15 (d, IH); 3.98 (dd, IH); 3.80 (s, 3H); 3.72-3.64 (m, 2H); 3.21 (d, IH); 2.95-2.84 (m, 2H); 2.73-2.55 (m, 4H); 2.29 ( s, 3H); 2.26 (s, 3H); 2.23 (s, 3H); 2.18 (s, 3H); 2.03 (s, 3H).
1 C-NMR (75 MHz, CDCI3): δ 169.1 , 167.5, 147.9, 145.7, 142.9, 141.9, 140.9, 136.0, 131.6, 129.4, 126.5, 122.5, 122.1, 121.5, 119.5, 118.7, 116.0, 11 1.5, 110.6, 102.2, 83.7, 63.9, 60.4, 58.8, 57.9, 55.4, 54.9, 49.7, 43.4, 41.7, 40.8, 32.1, 29.5, 24.9, 22.9, 21.5, 20.5, 15.8, 14.3, 9.9. ESI-MS m/z: Calcd. for C41H44N4O9S: 768.2 Found (M-H2O+H+): 751.2. Compound 41 (first isomer): *H-NMR (300 MHz, CDCI3): δ 7.84 (s, IH); 7.13 (dd, IH); 6.96 (dd, IH); 6.81 (ddd, IH); 6.62 (s, IH); 6.20 (d, IH); 5.99 (d, IH); 5.70 (s, IH); 5.19 (d, IH); 4.86 (s, IH); 4.52 (d, IH); 4.50 (s, IH); 4.16 (d, IH); 3.80 (s, 3H); 3.53 (d, IH); 3.49-3.48 (m, IH); 3.23 (d, IH); 3.00-2.71 (m, 3H); 2.62-2.41 (m, 2H); 2.31 (s, 3H); 2.27 (s, 3H); 2.14 (s, 3H); 2.02 (s, 3H); 1.13 (d, 3H). 13C-NMR (75 MHz, CDCI3): δ 170.5, 168.9, 159.4, 156.2, 147.6, 145.8, 143.0, 141.2, 133.2, 132.2, 131.7, 131.1, 129.2, 129.0, 127.3, 121.7, 115.6, 111.7, 1 11.6, 110.3, 109.9, 103.7, 103.4, 102.0, 81.8, 64.0, 62.0, 60.5, 58.0, 56.1 , 55.3, 44.0, 42.4, 41.5, 38.1, 32.1 , 29.5, 24.0, 22.9, 21.7, 20.7, 16.2, 14.3, 9.9. ESI-MS m/z: Calcd. for C41H43FN4O9S: 786.2 Found (M-H2O+H+): 769.3.
Compound 42 (second isomer): H-NMR (300 MHz, CDCI3): δ 7.62 (s, IH); 7.18 (dd, IH); 6.99 (dd, IH); 6.82 (ddd, IH); 6.59 (s, IH); 6.20 (d, IH); 5.98 (d, IH); 5.71 (s, IH); 5.15 (d, IH); 4.85 (s, IH); 4.52(s, IH); 4.50 (d, IH); 4.16 (d, IH); 4.05 (dd, IH); 3.80 (s, 3H); 3.50-3.48 (m, IH); 3.42-3.36 (m, IH); 3.23 (d, IH); 3.00-2.81 (m, 2H); 2.50 (dd, IH); 2.44 (d, IH); 2.32 (s, 3H); 2.24 (s, 3H); 2.16 (s, 3H); 2.07 (s, 3H); 1.23 (d, 3H). 13C-NMR (75 MHz, CDCI3): δ 171.9, 168.7, 156.2, 147.8, 145.6, 143.3, 141.8, 140.9, 132.7, 131.4, 131.1 , 129.4, 129.0, 121.8, 121.4, 115.8, 113.1 , 111.9, 1 11.8, 110.4, 1 10.0, 103.7, 103.4, 102.0, 81.9, 63.4,
61.8, 60.6, 58.0, 56.2, 55.2, 46.6, 42.3, 41.5, 41.0, 32.1, 29.5, 23.9,
22.9, 21.9, 20.7, 16.1, 14.3, 9.9.
ESI-MS m/z: Calcd. for C41H43FN4O9S: 786.2 Found (M-H O+H+): 769.3. Compound 43 (first isomer): Η-NMR (300 MHz, CDCls): δ 7.85 (s, IH); 7.33 (d, IH); 7.23 (d, IH); 7.07 (t, IH); 6.99 (t, IH); 6.63 (s, IH); 6.22 (d, IH); 6.00 (d, IH); 5.70 (s, IH); 5.20 (d, IH); 4.86 (s, IH); 4.52 (d, IH); 4.48 (s, IH); 4.16 (d, IH); 3.80 (s, 3H); 3.53 (d, IH); 3.22 (d, IH);
3.01-2.73 (m, 3H); 2.62-2.48 (m, 2H); 2.39-2.17 (m, IH); 2.31 ( s, 3H); 2.27 (s, 3H); 2.14 (s, 3H); 2.02 (s, 3H); 1.14 (d, 3H). 13C-NMR (75 MHz, CDCI3): δ 170.6, 168.8, 147.6, 145.7, 143.0, 141.2, 135.7, 131.8, 131.3, 129.2, 127.0, 122.0, 121.8, 121.7, 1 19.3, 118.5, 115.6, 11 1.0, 1 10.2, 102.0, 81.8, 64.0, 61.9, 60.5, 58.1 , 58.0, 56.1 , 55.3, 44.0, 42.4, 41.5, 38.1, 32.1 , 29.5, 24.0, 22.9, 21.8, 20.7, 16.2, 14.3, 9.9. ESI-MS m/z: Calcd. for C41H44N4O9S: 768.2 Found (M-H2O+H+): 751.3.
Compound 44 (second isomer): Η-NMR (300 MHz, CDCI3): δ 7.62 (s, IH); 7.36 (d, IH); 7.27 (d, IH); 7.09 (t, IH); 7.00 (t, IH); 6.59 (s, IH); 6.21 (d, IH); 5.98 (d, IH); 5.71 (s, IH); 5.15 (d, IH); 4.84 (s, IH); 4.51 (d, 2H); 4.17-4.16 (m, IH); 4.05 (dd, IH); 3.80 (s, 3H); 3.49-3.48 (m, IH); 3.42-3.38 (m, IH); 3.24-3.22 (m, IH); 3.03-2.81 (m, 2H); 2.57 (dd, IH); 2.46 (d, IH); 2.32 ( s, 3H); 2.24 (s, 3H); 2.17-2.12 (m, IH); 2.16 (s, 3H); 2.07 (s, 3H); 1.23 (d, 3H). ESI-MS m/z: Calcd. for C41H44N4O9S: 768.2 Found (M-H2O+H+): 751.3.
Compound 45: Η-NMR (300 MHz, CDCI3): δ 7.69 (s, IH); 7.38 (d, IH); 7.24 (d, IH); 7.09 (ddd, IH); 7.03 (s, IH); 7.00 (ddd, IH); 6.22 (d, IH); 6.00 (d, IH); 5.20 (d, IH); 4.83 (s, IH); 4.50 (s, IH); 4.38 (s, IH); 4.33 (s, IH); 4.12 (dd, IH); 3.77 (s, 3H); 3.68-3.66 (m, IH); 3.51-3.49 (m, IH); 3.24-2.85 (m, 4H); 2.70-2.49 (m, 2H); 2.62 (t, 2H); 2.37 ( s, 3H); 2.28 (s, 3H); 2.15 (s, 3H); 2.06 (s, 3H); 1.94-1.83 (m, 2H); 1.10 (t, 3H). ESI-MS m/z: Calcd. for C44H48N4O10S: 824.3 Found (M-H2O+H+): 807.2.
Compound 46: Η-NMR (300 MHz, CDCI3): δ .67 (s, IH); 7.38 (d, IH); 7.24 (d, IH); 7.09 (ddd, IH); 7-00 (ddd> 1H); 7-00 (s> 1H) 6-21 (d> 1H)» 6-00
(d, IH); 5.20 (d, IH); 4.83 (s, IH); 4.51 (d, IH);
4.39 (s, IH); 4.15 (d, IH); 3.81 (s, 3H); 3.52 (d, IH); 3.26 (d, IH); 3.19-3.11 (m, IH); 3.06-2.81
(m, 3H); 2.72-2.44 (m, 3H); 2.36 ( s, 3H); 2.31 (s, 3H); 2.17 (s, 3H); 2.03 (s, 3H); 1.54 (s, 9H).
13C-NMR (75 MHz, CDCI3): δ 171.5, 169.2, 151.4, 148.2, 145.7, 144.4, 141.3, 140.9, 135.7, 131.4, 131.1, 130.9, 127.4, 127.1, 124.4, 122.1, 121.5, 1 19.4, 118.7, 115.6, 1 11.1 , 102.0, 83.3, 81.8, 62.8, 61.9, 60.2, 57.8, 56.3, 56.1, 42.3, 41.5, 39.8, 39.4, 29.9, 27.8, 23.6, 21.7, 20.5, 16.0, 14.3, 9.9. ESI-MS m/z: Calcd. for C45H50N4O11S: 854.3 Found (M-H2O+H+): 837.2.
Compound 47: --H-NMR (300 MHz, CDCI3): δ 7.68 (s, IH); 7.38 (d, IH); 7.24 (d, IH); 7.19 (dd, IH); 7.10 (ddd, IH); 7.06 (s, IH); 7.00 (ddd, IH); 6.21 (d, IH); 6.01 (d, IH); 5.20 (d, IH); 5.00 (dd, IH); 4.83 (s, IH); 4.70 (dd, IH); 4.51 (s, IH); 4.40 (d, IH); 4.15 (dd, IH); 3.83 (dd, IH); 3.82 (s, 3H); 3.53 (d, IH); 3.28-3.03 (m, 3H); 2.94-2.83 (m, 2H); 2.72-2.46 (m, 3H); 2.38 ( s, 3H); 2.33 (s, 3H); 2.17 (s, 3H); 2.07 (s, 3H). 13C-NMR (75 MHz, CDCI3): δ 171.5, 169.0, 150.8, 148.0, 145.8, 144.0, 143.1, 141.3, 140.9, 135.7, 130.8, 128.2, 127.1 , 122.2, 120.0, 119.5, 118.7, 115.5, 113.3, 111.1, 110.5, 102.1, 98.7, 81.9, 62.8, 62.0, 60.5, 57.7, 56.2, 56.0, 42.3, 41.8, 39.9, 39.5, 32.1, 29.5, 23.7, 21.8, 20.5,
16.0, 14.3, 9.9.
ESI-MS m/z: Calcd. for C43H44N4O11S: 824.2 Found (M-H2O+H+): 807.2.
Compound 48: iH-NMR (300 MHz, CDCI3): δ 7.70 (s, IH); 7.37 (d, IH); 7.24 (d, IH); 7.08 (ddd, IH); 7.00 (ddd, IH); 6.87 (s, IH); 6.20 (d, IH); 6.17-6.05 (m, IH); 5.99 (d, IH); 5.47 (dd, IH); 5.25 (dd, IH); 5.21 (d, IH); 4.82 (s, IH); 4.81 (dd, IH); 4.50 (d, IH); 4.44 (s, IH); 4.36 (dd, IH); 4.13 (dd, IH); 4.11 (s, IH); 3.84 (s, 3H); 3.51 (d, IH); 3.24- 3.00 (m, 3H); 2.89-2.80 (m, 2H); 2.73-2.48 (m, 3H); 2.33 ( s, 3H); 2.31- 2.26 (m, IH); 2.23 (s, 3H); 2.19 (s, 3H); 2.05 (s, 3H). 13C-NMR (75 MHz, CDCb): δ 171.3, 168.6, 150.5, 148.6, 145.3, 140.9, 140.6, 135.5, 134.6, 131.0, 130.7, 126.7, 124.7, 121.7, 121.3, 119.0, 118.3, 116.2, 118.4, 110.8, 109.9, 101.7, 81.6, 72.6, 62.4, 61.6, 59.3, 57.6, 57.5, 55.9, 55.1, 42.0, 41.3, 39.4, 39.0, 29.2, 23.5, 22.5, 21.4, 20.3, 15.7, 14.0, 9. ESI-MS m/z: Calcd. for C43H46N4O9S: 794.3 Found (M-H O+H+): 777.2.
Compound 49: H-NMR (300 MHz, CDCI3): δ 7.60 (s, IH); 7.42-7.39 (m, 2H); 7.09-7.00 (m, 2H); 6.81 (s, IH); 6.16-6.04 (m, 2H); 6.07 (s, IH) 5.99 (s, IH); 5.52-5.43 (m, 2H); 5.24 (d, IH) 5.11 (d, IH); 4.96 (d, IH); 4.80-4.32 (m, 6H) 4.13-4.10 (m, 2H); 3.81 (s, 3H); 3.58-3.56 (m, IH); 3.46-3.40 (m, IH); 3.24-3.20 (m, IH); 2.99-2.87 (m, 2H); 2.67-2.53 (m, 2H); 2.31 (s, 3H); 2.23 (s, 3H); 2.19 (s, 3H); 2.02 (s, 3H). ESI-MS m/z: Calcd. for C46H50N4O9S: 834.3 Found (M-H2O+H+): 817.2. Scheme 2
To a solution of ET-736-CN in THF/H2O 3: 1 (0.027M) were added 15 equiv. of KOH. The reaction mixture was stirred at room temperature for 5 h. After this time the reaction was quenched with an aqueous saturated solution of NaCl, extracted with CH2CI2. The organic layer was dried over Na2SO4. Chromatography gives pure compound 50.
"-H-NMR (300 MHz, CDCI3): δ 7.59 (s, IH); 7.40 (d, IH); 7.25 (d, IH); 7.11 (ddd, IH); 7.02 (ddd, IH); 6.67 (s, IH); 6.16 (d, IH); 5.93 (d, IH); 5.90 (s, IH); 5.62 (s, IH); 5.06 (d, IH); 4.46 (d, IH); 4.36 (s, IH); 4.31 (dd, IH); 4.19 (d, IH); 4.12 (dd, IH); 3.82 (s, 3H); 3.55 (d, IH); 3.42 (d, IH); 3.20-2.80 (m, 4H); 2.69-2.53 (m, 3H); 2.38 (s, 3H); 2.21 (s, 3H); 2.18 (s, 3H). ESI-MS m/z: Calcd. for C39H39N5O7S: 721.3 Found (M+H+): 722.2.
Derivatives of Et-694:
Method 5: To a solution of 1 equiv. of ET-694-CN, compound 50 in CH2CI2 (0.032M) under argon at room temperature were added 2 equiv. of pyridine and 2 equiv. of acid chloride, chlorofomiate or anhydride. The reaction was followed by TLC and quenched with an aqueous saturated solution of NaHCO3, extracted with CH2CI2 and the organic layers dried over Na2SO4. Flash chromatography gives pure compounds. Compound 51: "-H-NMR (300 MHz, CDCI3): δ 7.65 (s, IH); 7.40 (d, IH); 7.26 (d, IH); 7.12 (s, IH); 7.11 (ddd, IH); 7.04 (ddd, IH); 6.30 (d, IH); 6.19 (d, IH); 5.10 (d, IH); 4.40-4-30 (m, 2H); 4.23-4.16 (m, IH); 3.76 (s, 3H); 3.50-3.44 (m, 2H); 3.19-
3.13 (m, 2H); 3.03-2.83 (m, 2H); 2.66-2.47 (m, 3H); 2.40 (s, 3H); 3.36-
2.22 (m, 2H); 2.16 (s, 3H); 2.07 (s, 3H).
ESI-MS m/z: Calcd. for C43H38F7N5O8S: 917.2 Found (M+H+): 918.1.
Compound 52: Η-NMR (300 MHz, CDCI3): δ 7.72 (d, IH), 7.38 (d, IH), 7.26 (d, IH), 7.10 (t, 1H).7.00 (t, IH), 6.65 (s, IH), 6.24 (d, IH), 6.02 (d, IH), 5.74 (s, IH), 5.08 (d, IH), 4.54 (broad s, IH), 4.33 (s, IH), 4.27 (d, IH), 4.21 (s, IH), 4.20 (d, IH), 3.80 (s, 3H), 3.43 (m, 2H), 3.20-2.81 (m, 4H), 2.64-2.58 (m, 3H), 2.53 (t, 2H), 2.37 (s, 3H), 2.26 (m, IH), 2.21 (s, 3H), 2.05 (s, 3H), 1.74 (sext, 2H), 1.01 (t, 3H). ESI-MS m/z: Calcd. for C43H45N5O8S: 791.3 Found (M+H+): 792.2.
Compound 53: Η-NMR (300 MHz, CDCI3): δ 7.71 (s, IH), 7.39 (d, IH), 7.26 (d, IH), 7.11 (t, IH), 7.02 (t, IH), 6.66 (s, IH), 6.26 (s, IH), 6.04 (s, IH), 5.80 (s, IH), 5.09 (d, IH), 4.52 (broad s, IH), 4.34 (s, IH), 4.27 (d, IH), 4.22 (d, IH), 4.20 (m, IH), 3.82 (s, 3H), 3.79 (m, 2H), 3.42 (m, 2H), 3.16 (m, IH), 3.07-2.81 (m, 5H), 2.64- 2.50 (m, 3H), 2.37 (s, 3H), 2.25 (m, IH), 2.21 (s, 3H), 2.08 (s, 3H). ESI-MS m/z: Calcd. for C42H42CIN5O8S: 811.2 Found (M+H+): 812.2. Compound 54: This product was obtained with 4 equiv. of cinnamoyl chloride and 4 equiv. of pyridine. "-H-NMR (300 MHz, CDC13): δ 7.83 (d, IH), 7.75 (s, IH), 7.58 (m, 2H), 7.46 (m, 3H), 7.39 (d, IH), 7.27 (d, IH), 7.1 1 (t, IH), 7.02 (t, IH), 6.61 (s, IH) 6.58 (d, IH), 6.26 (s, IH), 6.05 (s, IH), 5.52 (s, IH), 5.09 (d, IH), 4.60 (broad s, IH), 4.37 (s, IH), 4.27 (d, IH), 4.25 (s, IH), 4.23 (m, IH), 3.47 (s, 3H), 3.45 (m, 2H), 3.15 (m, IH), 3.04 (d, IH), 2.96 (m, IH), 2.84 (m, IH), 2.70- 2.53 (m, 3H), 2.33 (m, IH), 2.29 (s, 3H), 2.21 (s, 3H), 2.12 (s, 3H).
ESI-MS m/z: Calcd. for C48H45N5O8S: 851.3 Found (M+H+): 852.2.
Compound 55: This product was obtained with 6 equiv. of allylchloroformiate and 6 equiv. of pyridine.
Η-NMR (300 MHz, CDCI3): δ 7.74 (s, IH), 7.39 (d, IH), 7.25 (d, IH), 7.10 (t, IH), 7.01 (t, IH), 6.65 (s, IH), 6.25 (s, IH), 6.03 (s, IH), 5.90 (ddd, IH), 5.78 (s, IH), 5.37 (d, IH), 5.24 (d, IH), 5.08 (d, IH), 4.61 (m, 3H), 4.32 (s, IH), 4.28 (d, IH), 4.22 (s, IH), 4.20 (d, IH), 3.80 (s, 3H), 3.42 (m, 2H), 3.18 (m, IH), 3.09-2.81 (m, 3H), 2.59 (m, 3H), 2.37 (s, 3H), 2.26 (m, IH), 2.21 (s, 3H), 2.12 (s, 3H). ESI-MS m/z: Calcd. for C43H43N5O9S: 805.3 Found (M+H+): 806.3 Compound 56: This product was obtained with 3 equiv. of trifluoroacetic anhydride and 3 equiv. of pyridine iH-NMR (300 MHz, CDC13): δ 7.66 (s, IH), 7.40 (d, IH), 7.26 (d, IH), 7.1 1 (t, IH), 7.02 (t, IH), 6.65 (s, IH), 6.31 (d, IH), 6.08 (d, IH), 5.74 (s,
IH), 5.1 1 (d, IH), 4.55 (s, IH), 4.36 (s, IH), 4.28 (d, IH), 4.25 (s, IH),
4.23 (d, IH), 3.79 (s, 3H), 3.46 (m, 2H), 3.15 (m, IH), 3.09-2.46 (m, 6H),
2.36 (s, 3H), 2.23 (s, 3H), 2.20 (m, IH), 2.01 (s, 3H).
ESI-MS m/z: Calcd. for C41H38F3N5O8S: 817.2 Found (M+H+): 818.2
Compound 57: Η-NMR (300 MHz, CDCI3): δ 7.74 (s, IH), 7.40 (d, IH), 7.27 (d, IH), 7.1 1 (t, IH), 7.02 (t, IH), 6.65 (s, IH), 6.23 (s, IH), 6.02 (s, IH), 5.74 (s, IH), 5.09 (d, IH), 4.66 (s, IH), 4.32-4.21 (m, 4H), 3.81 (s, 3H), 3.40 (m, 2H), 3.21-2.86 (m, 3H), 2.80 (m, IH),
2.64 (m, 3H), 2.37 (s, 3H), 2.29 (m, IH), 2.21 (s, 3H), 2.11 (s, 3H), 1.45
(s, 9H).
ESI-MS m/z: Calcd. for C44H47N5O9S: 821.3 Found (M+H+): 822.0.
Compound 58: H-NMR (300 MHz, CDCI3): δ 7.72 (s, IH), 7.39 (d, IH), 7.26 (d, IH); 7.11 (t, IH), 7.02 (t, IH), 6.99 (s, IH), 6.24 (s, IH), 6.03 (s, IH), 5.09 (d, IH), 4.61 (s, IH), 4.30 (s, IH), 4.20 (m, 2H), 3.98 (s, IH), 3.83 (s, 3H), 3.45 (m, 2H), 3.21-2.90 (m, 3H), 2.80
(m, IH), 2.59 (s, 3H), 2.36 (s, 3H), 2.31 (m, IH), 2.20 (s, 3H), 2.12 (s,
3H), 1.54 (s, 9H), 1.45 (s, 9H).
ESI-MS m/z: Calcd. for C49H55N5O11S: 921.4 Found (M+H+): 922.3. The monoBoc derivative in C-5 was obtained with 6 equiv. of Boc anhydride and 6 equiv of pyridine. With these conditions traces of diBoc derivative in C-5 and C- 18 was isolated as a secondary product. This last compound can be obtained as the major product when the reaction was performed with TEA as base.
Compound 59: Η-NMR (300 MHz, CDC13): δ 7.71 (s, IH), 7.39 (d, IH), 7.26 (d, IH), 7.10 (t, IH), 7.04 (t, IH), 6.95 (dd, IH), 6.65 (s, IH), 6.26 (s, IH), 6.04 (s, IH), 5.78 (s, IH), 5.09 (d, IH), 4.99 (dd, IH), 4.63 (s, IH), 4.60 (dd, IH), 4.33 (s, IH), 4.29 (d, IH), 4.22 (s, IH), 4.21 (d,
IH), 3.78 (s, 3H), 3.42 (m, 2H), 3.21-2.79 (m, 4H), 2.63 (m, 3H), 2.37 (s,
3H), 2.27 (m, IH), 2.22 (s, 3H), 2.13 (s, 3H).
ESI-MS m/z: Calcd. for C42H41N5O9S: 791.3 Found (M+H+): 792.1
Compound 60: Η-NMR (300 MHz, CDCI3): δ 7.70 (s, IH), 7.40 (d, IH), 7.26 (d, IH), 7.18 (dd, IH), 7.11 (t, IH), 7.05 (s, IH), 7.02 (t, IH), 6.97 (dd, IH), 6.27 (s, IH), 6.05 (s, IH), 5.10 (d, IH), 5.09-5.00 (m, IH), 5.05 (s, IH), 4.72 (dd, IH), 4.60 (dd, IH), 4.56 (s, IH), 4.33 (s,
IH), 4.22 (m, 2H), 3.97 (d, IH), 3.78 (s, 3H), 3.46 (m, 2H), 3.18 (m, IH),
3.11 (d, IH), 2.97 (dd, IH), 2.85 (m, IH), 2.71-2.51 (m, 3H), 2.37 (s,
3H), 2.32 (m, IH), 2.21 (s, 3H), 2.16 (s, 3H).
ESI-MS m/z: Calcd. for C45H43N5O11S: 861.3 Found (M+H+): 862.7.
Method 6: To a solution of 1 equiv. of ET-694-CN, compound 50 in CH2CI2 (0.032M) under argon at room temperature were added 2 equiv. of acid, 2 equiv. of EDC.HC1 and 2 equiv. of DMAP. The reaction was followed by TLC and quenched with an aqueous saturated solution of NaHCO3, extracted with CH2CI2 and the organic layers dried over Na2SO4. Flash chromatography gives pure compounds.
Compound 61: *H-NMR (300 MHz, CDCI3): δ 7.70 (s, IH), 7.39 (d, IH), 7.26 (d, IH), 7.11 (t, IH), 7.00 (t, IH), 6.65 (s, IH), 6.23 (s, IH), 6.03 (s, IH), 5.72 (s, IH), 5.09 (d, IH), 4.57 (broad s, IH), 4.33 (s, IH), 4.26 (d, IH), 4.21 (s, IH), 4.19 (d, IH), 3.80 (s, 3H), 3.44 (m, 2H), 3.16 (m, IH), 3.03 (d, IH), 3.00-2.89 (m, 2H), 2.68-2.52 (m, 3H), 2.54 (t, 2H), 2.37 (s, 3H), 2.31 (m, IH), 2.21 (s, 3H), 2.04 (s, 3H), 1.65 (m, 2H), 1.29 (m, 8H), 0.87 (m, 3H). ESI-MS m/z: Calcd. for C47H53N5O8S: 847.4 Found (M+H+): 848.3.
Method 4.- To a solution of 1 equiv. of starting material in CH3CN/H2O 3:2 (0.009M) were added 30 equiv. of AgNO3. After 24 h the reaction was quenched with a mixture 1 : 1 of saturated solutions of brine and NaHCO3, stirred for 10 min and diluted and extracted with CH2CI2. The organic layer was dried with Na2SO4. Chromatography gives pure compounds.
Compound 62: Η-NMR (300 MHz, CDCI3): δ 7.65 (s, IH); 7.40 (d, IH); 7.25 (d, IH); 7.10 (ddd, IH); 7.01 (ddd, IH) 6.66 (s, IH); 6.27 (d, IH); 6.04 (d, IH) 5.69 (s, IH); 5.23 (d, IH); 4.85 (s, IH) 4.51 (d, IH); 4.47 (s, IH); 4.19 (s, IH) 4.15 (dd, IH); 3.78 (s, 3H); 3.52-3.50 (m, IH); 3.26-3.14 (m, 2H); 3.04- 2.80 (m, 3H); 2.72-2.47 (m, 3H); 2.36 (s, 3H); 2.20 (s, 3H); 2.05 (s, 3H). ESI-MS m/z: Calcd. for C42H39F7N4O9S: 908.3 Found (M+H+): 909.2.
Compound 63: Η-NMR (300 MHz, CDC13): δ 7.71 (d, IH), 7.38 (d, IH), 7.26 (d, IH), 7.09 (t, IH), 7.03 (t, IH), 6.67 (s, IH), 6.21 (d, IH), 5.99 (d, IH), 5.71 (broad s, IH), 5.18 (d, IH), 4.83 (s, IH), 4.50 (d, IH), 4.46 (broad s, IH), 4.17 (d, IH), 4.12 (d, IH), 3.81 (s, 3H), 3.51
(d, IH), 3.24-3.18 (m, 2H), 3.00 (d, IH), 2.85 (m, 2H), 2.70-2.50 (m, 5H),
2.37 (s, 3H), 2.27 (m, IH), 2.19 (s, 3H), 2.04 (s, 3H), 1.74 (sext, 2H),
1.01 (t, 3H).
ESI-MS m/z: Calcd. for C42H46N4O9S: 782.3 Found (M+H+): 783.2.
Compound 64: Η-NMR (300 MHz, CDCI3): δ 7.71 (s, IH), 7.39 (d, IH), 7.25 (d, IH), 7.10 (t, IH), 7.01 (t, IH), 6.66 (s, IH), 6.22 (s, IH), 6.00 (s, IH), 5.90 (ddd, IH), 5.74 (broad s, IH), 5.37 (d, IH), 5.22 (t, IH), 4.83 (s, IH), 4.59 (m, 2H), 4.49 (s, IH),
4.29 (dd, IH), 4.15 (m, 2H), 3.80 (s, 3H), 3.65 (m, IH), 3.51 (m, 2H),
3.18 (m, IH), 3.09-2.81 (m, 3H), 2.59 (m, 3H), 2.37 (s, 3H), 2.26 (m,
IH), 2.21 (s, 3H), 2.12 (s, 3H).
ESI-MS m/z: Calcd. for C42H44N4O10S: 796.3 Found (M+H+): 797.0
Compound 65: iH-NMR (300 MHz, CDCI3): δ 7.74 (s, IH), 7.40 (d, IH), 7.27 (d, IH), 7.1 1 (t, IH), 7.02 (t, IH), 6.65 (s, IH), 6.23 (s, IH),
6.02 (s, IH), 5.74 (broad s, IH), 5.20 (d, IH), 4.82 (s, IH), 4.58 (s, IH), 4.49 (m, IH), 4.13 (m, 2H), 3.81 (s, 3H), 3.49 (m, IH), 3.21 (m, 2H), 3.02 (d, IH), 2.80 (m, 3H), 2.64 (m, 2H), 2.37 (s, 3H), 2.29 (m, IH), 2.21 (s, 3H), 2.1 1 (s, 3H), 1.45 (s, 9H). ESI-MS m/z: Calcd. for C43H48N4O10S: 812.3 Found (M+H+): 813.0.
Compound 66: "-H-NMR (300 MHz, CDCb): δ 7.71 (s, IH), 7.39 (d, IH), 7.26 (d, IH); 7.1 1 (t, IH), 7.02 (t, IH), 6.99 (s, IH), 6.21 (s, IH), 6.00 (s, IH), 5.19 (d, IH), 4.80 (s, IH), 4.51 (m, 2H), 4.16 (m, 2H), 3.83 (s, 3H), 3.54 (m, IH), 3.28-3.04 (m, 3H), 2.92-2.78 (m, 2H),
2.59 (m, 3H), 2.36 (s, 3H), 2.31 (m, IH), 2.18 (s, 3H), 2.11 (s, 3H), 1.54
(s, 9H), 1.45 (s, 9H).
ESI-MS m/z: Calcd. for C48H56N4O12S: 912.4 Found (M+H+): 913.1.
Compound 67: Η-NMR (300 MHz, CDCI3): δ 7.70 (s, IH), 7.39 (d, IH), 7.26 (d, IH), 7.10 (t, IH), 7.01 (t, IH), 6.95 (dd, IH), 6.66 (s, IH), 6.24 (s, IH), 6.01 (s, IH), 5.75 (s, IH), 5.21 (d, IH), 4.99 (dd, IH), 4.84 (s, IH), 4.58 (dd, IH), 4.55 (s, IH), 4.51 (s, IH), 4.20 (s, IH), 4.15 (d, IH), 3.78 (s, 3H), 3.49 (m, IH), 3.21 (m, 2H), 3.00 (d, IH), 2.86 (m, 2H), 2.59 (m, 3H), 2.37 (s, 3H), 2.27 (m, IH), 2.20 (s, 3H), 2.12 (s, 3H). ESI-MS m/z: Calcd. for C41H42N4O10S: 782.3 Found (M+H+): 783.1
Compound 68: Η-NMR (300 MHz, CDCI3): 7.67 (s, IH), 7.39 (d, IH), 7.24 (d, IH), 7.17 (dd, IH), 7.10 (t, IH), 7.05 (s, IH), 7.02 (t, IH), 6.97 (dd, IH), 6.24 (s, IH), 6.02 (s, IH), 5.21
(d, IH), 5.06 (dd, IH), 5.01 (dd, IH), 4.83(s, IH); 4.72 (dd, IH), 4.60 (dd, IH), 4.51 (s, IH), 4.48 (s, IH), 4.15 (dd, IH), 3.86(d, IH), 3.78 (s, 3H), 3.54 (d, IH), 3.28-3.18 (m, 2H), 3.07 (d, IH), 2.94-2.84(m, 2H), 2.67- 2.52 (m, 3H), 2.37 (s, 3H), 2.32 (m, IH), 2.18 (s, 3H), 2.14 (s, 3H). ESI-MS m/z: Calcd. for C44H44N4O12S: 852.3 Found (M+H+): 853.5
BIOASSAYS FOR ANTITUMOR SCREENING
The finality of these assays is to interrupt the growth of a "in vitro" tumor cell culture by means of a continued exhibition of the cells to the sample to be testing.
CELL LINES
Name N° ATCC Species Tissue Characteristics
P-388 CCL-46 mouse ascites fluidlymphoid neoplasm
K-562 CCL-243 human leukemia erythroleukemia (pleural effusion)
A-549 CCL- 185 human lung lung carcinoma "NSCL"
SK-MEL-28 HTB-72 human melanoma malignant melanoma
HT-29 HTB-38 human colon colon adenocarcinoma
LoVo CCL-229 human colon colon adenocarcinoma
LoVo-Dox human colon colon adenocarcinoma (MDR)
SW620 CCL-228 human colon colon adenocarcinoma (lymph node metastasis)
DU- 145 HTB-81 human prostate prostate carcinoma, not androgen receptors
LNCaP CRL- 1740 human prostate prostate adenocarcinoma, with androgen receptors
SK-BR-3 HTB-30 human breast breast adenocarcinoma, Her2/neu+, (pleural effusion) MCF-7 HTB-22 human breast breast adenocarcinoma, (pleural effusion)
MDA-MB- HTB-26 human breast breast adenocarcinoma,
231 Her2/neu+, (pleural effusion)
IGROV- 1 human ovary ovary adenocarcinoma
IGROV-ET human ovary ovary adenocarcinoma, characterized as ET-743 resistant cells
SK-OV-3 HTB-77 human ovary ovary adenocarcinoma (malignant ascites)
OVCAR-3 HTB-161 human ovary ovary adenocarcinoma
HeLa CCL-2 human cervix cervix epitheloid carcinoma
HeLa-APL CCL-3 human cervix cervix epitheloid carcinoma, characterized as aplidine resistant cells
A-498 HTB-44 human kidney kidney carcinoma
PANC-1 CRL-1469 human pancreas pancreatic epitheloid carcinoma
HMEC1 human endotheliu m
Inhibition of cells growth by colorimetric assay
A colorimetric type of assay, using sulphorhoda ine B (SRB) reaction has been adapted for a quantitative measurement of cell growth and viability [following the technique described by Philip Skehan, et al. (1990), New colorimetric cytotoxicity assay for anticancer drug screening, J. Natl. Cancer Inst, 82: 1107-1112]
This form of the assay employs 96 well cell culture microplates of 9 mm diameter (Faircloth, 1988; Mosmann, 1983). Most of the cell lines are obtained from American Type Culture Collection (ATCC) derived from different human cancer types.
Cells are maintained in RPMI 1640 10% FBS, supplemented with 0.1 g/1 penicillin and 0.1 g/1 streptomycin sulphate and then incubated at 37°C, 5% CO2 and 98% humidity. For the experiments, cells were harvested from subconfluent cultures using trypsin and resuspended in fresh medium before plating.
Cells are seeded in 96 well microtiter plates, at 5 x 103 cells per well in aliquots of 195 μl medium, and they are allowed to attach to the plate surface by growing in drug free medium for 18 hours. Afterward, samples are added in aliquots of 5 μl in a ranging from 10 to 10'8 μg/ml, dissolved in DMSO/EtOH/PBS (0.5:0.5:99). After 48 hours exposure, the an ti tumor effect are measured by the SRB methodology: cells are fixed by adding 50 μl of cold 50% (wt/vol) trichloroacetic acid (TCA) and incubating for 60 minutes at 4°C. Plates are washed with deionized water and dried. One hundred μl of SRB solution (0.4% wt/vol in 1% acetic acid) is added to each microtiter well and incubated for 10 minutes at room temperature. Unbound SRB is removed by washing with 1% acetic acid. Plates are air dried and bound stain is solubilized with Tris buffer. Optical densities are read on a automated spectrophotometric plate reader at a single wavelength of 490 nm.
The values for mean +/- SD of data from triplicate wells are calculated. Some parameters for cellular responses can be calculated: GI = growth inhibition, TGI = total growth inhibition (cytostatic effect) and LC = cell killing (cytotoxic effect).
Obtained results may predict the usefulness of a certain drug as a potential cancer treatment. For this technique, compounds which show GI50 values smaller than 10 μg/ml are selected to continue with further studies. GI50 d ata allow to predict that not only could a drug be cystostatic, but also it could have a potential in terms of tumor reduction.
Activity Data (Molar).
TOXICITY DATA
Toxicity was asssessed by the methods reported in Toxicology in Nitro, 15 (2001) 571-577, J. Luber Νarod et al: "Evaluation of the use of in vitro methodologies as tools for screening new compounds for potential in vivo toxicity".
Methods
In order to assess the cytotoxicity of the drugs to normal cells, we used 96 well plates plated at a density of 5000 cells per well (except for the FDC-Pl which were plated at 12,000 cells per well) with normal cell lines (ATCC, Table 1) maintained as per the directions of the ATCC: AML-12, normal mouse liver cells; ΝRK-52E, normal rat kidney cells; L8, normal rat skeletal muscle cells; FDC-Pl, normal mouse myelogeous stem cells; and H9c2 (2-1), normal rat cardiac muscle cells. The cells in each plate were permitted to settle overnight before adding the test drug. In addition, primary neuronal cultures were prepared from embryonic (day e- 17) whole brain (forebrain and brainstem) and spinal cord using established methods (Federoff and Richardson, 1997). To each well (lOOμl medium) lOμl of drug in media was added at varying concentrations (1x10- 10-0.0 lmg/ ml final concentration) and further incubated overnight at 37°C with 5% CO2. After 24h the following assays were performed. All experiments were repeated at least 3 times and were assayed in duplicate.
l .MTS assay (CellTiter 96 aqueous) was performed according to the manufacturer's (Promega) directions (for all cell types). Cell viability (mitochondrial activity) is determined via enzymatic conversion of the formazan substrate.
In vitro evaluation of the compounds for ADME-TOX profile
Partition Coefficient (log D)
The partition coefficient of a chemical compound provides a thermodynamic measure of its hydro philicity-lipophylicity balance. Lipophilicity is a major structural factor that influences the pharmacokinetic and pharmacodynamic behavior of compounds. The partition coefficient between water or buffer and 1 -octanol is the most widely used measure of chemical compound lipophilicity.
The measurement of partition coefficient was evaluated based on a miniaturized shake-flask procedure. Buffer (Dulbecco's PBS, pH 7.40) was used as the aqueous phase. The tested compound was dissolved in DMSO, at the concentration of 100 μM. The final DMSO concentration (1%.) during the octanol-buffer partitioning are very low to avoid bias on the partitioning. The amount of compound in the buffer phase was determined by HPLC with photodiode array detection after an equilibration phase of 60 min. The amount of compound in the octanol phase is calculated by subtraction of the amount of compound in buffer from the total amount of compound, which is determined from a calibration sample.
Log D is calculated as the Logio of the amount of compound in the octanol phase divided by the amount of compound in the buffer phase. The effective range of the log D microassay is approximately -0.5 to +4.5.
In vitro Intestinal Absorption Assays
The intestinal epithelium permeability is a critical characteristic that determines the rate and extent of human absorption and ultimately the bioavailability of a drug candidate. Caco-2 permeability assay allows a rapid assessment of membrane permeability and thus helps to rank- order compounds in terms of their absorption potential.
The Caco-2 cell line is a human colon adenocarcinoma cell line that differentiates in culture and resembles the epithelial lining of the human small intestine. It has been widely used as an in vitro intestinal epithelial model for drug transport and permeability screening of discovery compounds.
The apparent permeability coefficients (Papp) was determined in the apical-to-basolateral (A-to-B) direction across the cell monolayers (TC-7 sub-clone of the Caco-2) cultured on polycarbonate membrane filters. Compounds were tested at 50 μM with at a final DMSO concentration of 1%. Samples were analyzed by HPLC-MS or HPLC-MS/MS.
The test compound was added to the apical side and the Papp was determined based on the rate of appearance of the test compound in the basolateral side after 2h-incubation. Two reference compounds, propranolol (highly permeable) and ranitidine (poorly permeable), are tested in each assay as controls. Results from this assay can be used to rank-order compounds in terms of their absorption potential. Compounds with PapP equals to or greater than 20 x 10-6 cm/s could be considered highly permeable and are likely to be "not permeability- limited". Compounds with Papp less than 5 x 10-6 cm/s are considered poorly permeable and are likely to be "permeability-limited". Compounds with PapP greater than 5 x 10-6 but less than 20 x 10-6 cm/s" are considered to have medium permeability.
Metabolism Hepatic metabolism is a primary determinant of pharmacokinetic behavior, and rapid first-pass metabolism is a major cause of low bioavailability. Pooled liver microsomes and recombinant cytochrome P450's are used for the metabolic assessment of hits, leads, and new pharmaceutical compounds. The results of the metabolic screening studies are useful in:
• Determining the initial rate at which compounds are metabolized
• Investigating the major pathways of drug metabolism
• Predicting in vivo pharmacokinetic behavior
• Investigating the potential for drug-drug interactions
The metabolic stability was determined used human liver S9 homogenate that including both microsomal and cytosolic enzyme activities. The test compound was diluted in methanol (0.625%) and acetonitrile (0.625%) at the concentration 1 μM and incubated in the human liver pool (protein = Img/mL) during 60 min at 37°C. Peak areas corresponding to all analytes (metabolic products) were determined by HPLC-MS/MS. Areas were recorded and ratios of peak areas of analytes to that of internal standard for each analyte were determined. The ratio of precursor compound remaining after 60 minutes and the amount remaining at time zero, expressed as per cent, are reported as metabolic stability. Higher values mean higher metabolic stability.
Inhibition of cvtochromes P450 (CYP450)
The cytochromes P450 are a group of related enzymes primarily located in the liver and responsible of the metabolism of drugs. The inhibition of these CYP by drugs are related with drug-drug interactions and toxicities. CYP3A4 is the most common form of the CYP3A enzymes found in adults and is the form implicated in most drug interactions. CYP2D6 metabolize more than 25% of the clinically useful medications.
For CYP2D6 inhibition assay, the compound are tested at 10 μM in duplicate with a 0.25% final concentration of both methanol and acetonitrile in presence of the fluorescent substrate AMMC (3-[2-7Y,7 - diethyl-7Y-methylammonium)ethyl]-7-methoxy-4-methylcoumarin) at the concentration of 1.5 μM). The conversion of the AMMC in AHMC (3-[2- 7Y,iV-diethyl-7Y-methylammonium)ethyl]-7-hydroxy-4-methylcoumarin) is determined spectrofluorimetry after incubation with the enzyme during 450 min at 37aC.
For CYP3A4 inhibition assay, the compound were tested at 10 μM in duplicate with a 0.25% final concentration of both methanol and acetonitrile in presence of the fluorescent substrate BFC (50 μM). The conversion of the BFC (7-benzyloxy-4-trifluoromethylcoumarin) in HFC (7-hydroxy-4-trifluoromethylcoumarin) was determined spectro- fluormetry after incubation with the enzyme during 30 min at 37aC .
For both assays, the fluorescent intensity measured at t = 0 is subtracted from that measured after the appropriate incubation time. The ratio of signal-to-noise is calculated by comparing the fluorescence in incubations containing the test compound to the control samples containing the same solvent vehicle. The percent of control activity is calculated and reported as percent inhibition.
In vitro Safety Assessment. Cell Viability
The toxic potential of compounds was investigated in vitro using primary human hepatocytes (HEPG2). The compounds were tested at 30 μM in duplicate with a final DMSO concentration of 1%. After incubation during 24 h at 37°C the cell viability was determined by the conversion of oxided alarmarBlue (resazurin) to reduced alarmarBlue (resorufin). Chlorpromazine was used as reference compound. Results are expressed as a percent of inhibition of control values.
Results of the studies ADME -TOX

Claims (14)

1. A compound of the general formula I:
wherein the groups Ri, R2, R3, R4, and R5 are each independently selected from the group consisting of H, OH, OR', SH, SR', SOR', SO R', C(=O)R', C(=O)OR', NO2, NH2, NHR', N(R')2, NHC(O)R', CN, halogen, =O, substituted or unsubstituted C1-C25 alkyl, substituted or unsubstituted C2-C18 alkenyl, substituted or unsubstituted C2-C18 alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclic; wherein X is independently selected of OR', CN, (=O), or H. wherein each of the R' groups is independently selected from the group consisting of H, OH, NO2, NH2, SH, CN, halogen, =O, C(=O)H, C(=O)CH3, CO2H, substituted or unsubstituted Ci-C25 alkyl, substituted or unsubstituted 02-0^ alkenyl, substituted or unsubstituted C2-C1.8 alkynyl, substituted or unsubstituted aryl; wherein m is 0, 1 or 2; and wherein n is 0, 1, 2, 3 or 4.
2. A compound of formula I, wherein:
Ri is hydrogen, hydroxy, halogen, alkyl, alkoxy or aralkyl;
R2 and R3 are independently selected from hydrogen, R', C=OR', or
COOR', where R' is optionally substituted alkyl or alkenyl, the optional substituents being chosen from halo, amino including amino derived from amino acid, aryl or heterocyclic; R4 is hydrogen, alkyl, alkylene or C(=O)OR', where R' is alkylene;
R5 is hydrogen or alkyl;
X is hydrogen, hydroxy, cyano or keto; m is 0 or 1 ; and n is 0 or 1.
3. A compound according to claim 1 or 2, wherein R-. is hydrogen; hydroxy; fluorine, methyl, methoxy or benzyloxy, and n is 0 or 1.
4. A compound according to claim 1 , 2 or 3 wherein R2 is hydrogen; acetyl; trifluoromethylcarbonyl, heptafluorobutyrylcarbonyl, 3- chloropropionyl; cinnamoyl-CO-; t-butyl-O-CO-, allyl-O-CO- or vinyl-O- CO.
5. A compound according to any preceding claim wherein R3 is hydrogen; allyl; CH3-(CH )n-CO- where n is 1, 2, 4, 6, 12, 14 or 16; t- butyl-O-CO-; allyl-O-CO- or vinyl-O-CO.
6. A compound according to any preceding claim wherein R4 is hydrogen; Ci to C3 alkyl; allyl; or vinyl-O-CO.
7. A compound according to any preceding claim wherein R5 is hydrogen or methyl and m is 0 or 1.
8. A compound according to any preceding claim, wherein X is hydroxy or cyano.
9. A compound according to any preceding claim, wherein Ri is not hydrogen.
10. A compound according to any preceding claim wherein R2 is not acetyl.
11. A compound according to any preceding claim wherein R3 is not hydrogen.
12. A compound according to any preceding claim wherein R5 is not hydrogen.
13. A pharmaceutical composition which contains a compound according to any preceding claim together with a pharmaceutically acceptable diluent.
14. The use of a compound of general formula I according to claim 1 in the preparation of a medicament for treatment of cancer.
AU2002313540A 2001-08-07 2002-08-06 Antitumoral analogs Active 2027-08-06 AU2002313540B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GBGB0119243.4A GB0119243D0 (en) 2001-08-07 2001-08-07 Antitumoral analogs of ET-743
GB0119243.4 2001-08-07
PCT/GB2002/003592 WO2003014127A1 (en) 2001-08-07 2002-08-06 Antitumoral analogs

Publications (2)

Publication Number Publication Date
AU2002313540A1 true AU2002313540A1 (en) 2003-06-19
AU2002313540B2 AU2002313540B2 (en) 2008-05-22

Family

ID=9919954

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2002313540A Active 2027-08-06 AU2002313540B2 (en) 2001-08-07 2002-08-06 Antitumoral analogs

Country Status (23)

Country Link
US (1) US7763615B2 (en)
EP (3) EP1806349A1 (en)
JP (2) JP4684552B2 (en)
KR (1) KR20040032150A (en)
CN (1) CN100334094C (en)
AT (1) ATE374777T1 (en)
AU (1) AU2002313540B2 (en)
CA (1) CA2455768C (en)
CY (1) CY1107830T1 (en)
DE (1) DE60222775T2 (en)
DK (1) DK1414828T3 (en)
ES (1) ES2294151T3 (en)
GB (1) GB0119243D0 (en)
HK (1) HK1065786A1 (en)
HU (1) HU229779B1 (en)
IL (2) IL160051A0 (en)
MX (1) MXPA04001240A (en)
NO (1) NO337476B1 (en)
NZ (1) NZ530837A (en)
PL (1) PL218295B1 (en)
PT (1) PT1414828E (en)
RU (1) RU2283842C2 (en)
WO (1) WO2003014127A1 (en)

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0202544D0 (en) * 2002-02-04 2002-03-20 Pharma Mar Sa The synthesis of naturally occuring ecteinascidins and related compounds
AU2009204048B2 (en) * 2008-01-11 2013-08-01 Albany Molecular Research, Inc. (1-azinone) -substituted pyridoindoles as MCH antagonists
WO2011003012A1 (en) * 2009-07-01 2011-01-06 Albany Molecular Research, Inc. Azinone-substituted azapolycycle mch-1 antagonists, methods of making, and use thereof
US8629158B2 (en) * 2009-07-01 2014-01-14 Albany Molecular Research, Inc. Azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine MCH-1 antagonists, methods of making, and use thereof
US8637501B2 (en) * 2009-07-01 2014-01-28 Albany Molecular Research, Inc. Azinone-substituted azepino[b]indole and pyrido-pyrrolo-azepine MCH-1 antagonists, methods of making, and use thereof
US9073925B2 (en) * 2009-07-01 2015-07-07 Albany Molecular Research, Inc. Azinone-substituted azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine MCH-1 antagonists, methods of making, and use thereof
JP6037337B2 (en) * 2010-05-25 2016-12-07 ファルマ、マール、ソシエダード、アノニマPharma Mrs,S.A. Synthetic methods for the production of echinasaidin compounds
BR122017028570B1 (en) * 2010-11-12 2022-03-03 Pharma Mar, S.A USE OF PM01183, OR A PHARMACEUTICALLY ACCEPTABLE SALT THEREOF, IN SYNERGIC COMBINATION WITH A TOPOISOMERASE I AND/OR II INHIBITOR AND KIT
ES2688086T3 (en) 2010-11-26 2018-10-30 Nec Corporation Video decoding device, video decoding method and program
WO2012088124A2 (en) 2010-12-21 2012-06-28 Albany Molecular Research, Inc. Tetrahydro-azacarboline mch-1 antagonists, methods of making, and uses thereof
WO2012088038A2 (en) 2010-12-21 2012-06-28 Albany Molecular Research, Inc. Piperazinone-substituted tetrahydro-carboline mch-1 antagonists, methods of making, and uses thereof
CN108271369A (en) 2015-10-23 2018-07-10 法尔玛赞公司 It is used to prepare the new process of tryptamines and its derivative
JOP20190254A1 (en) 2017-04-27 2019-10-27 Pharma Mar Sa Antitumoral compounds
TWI824043B (en) * 2018-10-25 2023-12-01 西班牙商瑪製藥股份有限公司 Drug antibody conjugates
BR112022003149A2 (en) 2019-09-03 2022-05-17 Pharma Mar Sa Lurbinectedin in the treatment of malignant mesothelioma
CN112574234B (en) * 2019-09-27 2022-05-24 江苏恒瑞医药股份有限公司 Preparation method of ecteinascidin derivative
CN115151259A (en) * 2019-11-21 2022-10-04 法马马有限公司 Method for treating small cell lung cancer with lubicatin preparation
WO2021228414A1 (en) 2020-05-14 2021-11-18 Pharma Mar, S.A. Methods of treating small cell lung cancer with lurbinectedin formulations
MA56827B2 (en) 2019-11-21 2023-09-27 Pharma Mar Sa METHODS OF TREATING SMALL CELL LUNG CANCER WITH LURBINECTEDIN FORMULATIONS
EP4138923A1 (en) 2020-04-21 2023-03-01 Pharma Mar, S.A. Drug antibody conjugates
TW202144369A (en) * 2020-04-26 2021-12-01 大陸商江蘇恆瑞醫藥股份有限公司 Ascidianin derivatives, preparation method and medical use thereof
JP2023541010A (en) 2020-09-04 2023-09-27 ファルマ、マール、ソシエダード、アノニマ Combination of lurbinectedin and immune checkpoint inhibitors
WO2022101255A1 (en) 2020-11-10 2022-05-19 Pharma Mar, S.A. Lurbinectedin and irinotecan combinations
CA3229559A1 (en) * 2021-08-31 2023-03-09 Srinivasa Chary CHINTALAPATI Novel crystalline polymorph of lurbinectedin and improved process for the preparation of lurbinectedin
WO2023079177A1 (en) 2021-11-08 2023-05-11 Pharma Mar, S.A. Lurbinectedin and atezolizumab combinations
CN115246846A (en) * 2021-11-19 2022-10-28 江苏慧聚药业股份有限公司 New crystal form of rubitinid and preparation thereof
CN115304619A (en) * 2022-04-08 2022-11-08 上海皓元医药股份有限公司 Crystal form of rubitinid and preparation method thereof
CN114940682A (en) * 2022-05-18 2022-08-26 博瑞制药(苏州)有限公司 Crystal form of ribitdine, preparation method and application thereof

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS59225189A (en) 1983-06-03 1984-12-18 Shionogi & Co Ltd Quinonamin derivative and its preparation
JPS6084288A (en) 1983-10-13 1985-05-13 Shionogi & Co Ltd Cyanoquinonamine acetate and its production
US5089273A (en) 1986-06-09 1992-02-18 Board Of Trustees Of The University Of Illinois Ecteinascidins 729, 743, 745, 759A, 759B and 770
AU589282B2 (en) 1986-06-09 1989-10-05 University Of Illinois Ecteinascidins 729, 743, 745, 759a, 759b and 770
US5256663A (en) 1986-06-09 1993-10-26 The Board Of Trustees Of The University Of Illinois Compositions comprising ecteinascidins and a method of treating herpes simplex virus infections therewith
US5149804A (en) * 1990-11-30 1992-09-22 The Board Of Trustees Of The University Of Illinois Ecteinascidins 736 and 722
US5478932A (en) 1993-12-02 1995-12-26 The Board Of Trustees Of The University Of Illinois Ecteinascidins
US5721362A (en) 1996-09-18 1998-02-24 President And Fellows Of Harvard College Process for producing ecteinascidin compounds
US5985876A (en) 1997-04-15 1999-11-16 Univ Illinois Nucleophile substituted ecteinascidins and N-oxide ecteinascidins
IL138856A0 (en) 1998-04-06 2001-10-31 Univ Illinois Semi-synthetic ecteinascidins
CN100548988C (en) 1998-05-11 2009-10-14 法马马有限公司 The metabolite of ET 743
US6124292A (en) 1998-09-30 2000-09-26 President And Fellows Of Harvard College Synthetic analogs of ecteinascidin-743
MY130271A (en) 1999-05-14 2007-06-29 Pharma Mar Sa Hemisynthetic method and new compounds
EP1254140A4 (en) 2000-01-19 2003-03-12 Univ Columbia Compounds of the saframycin-ecteinascidin series, uses, and synthesis thereof
WO2001077115A1 (en) 2000-04-12 2001-10-18 Pharma Mar, S.A. Antitumoral ecteinascidin derivatives
MXPA02011319A (en) 2000-05-15 2003-06-06 Pharma Mar Sa Antitumoral analogs of et 743.

Similar Documents

Publication Publication Date Title
EP1414828B1 (en) Antitumoral analogs
AU2002313540A1 (en) Antitumoral analogs
US8076337B2 (en) Antitumoral derivatives of ET-743
EP2305689B1 (en) The synthesis of naturally occuring ecteinascidins and related compounds
AU2002317967A1 (en) New antitumoral derivatives of ET-743