WO2007081694A2 - Derivatives of sulindac, use thereof and preparation thereof - Google Patents

Derivatives of sulindac, use thereof and preparation thereof Download PDF

Info

Publication number
WO2007081694A2
WO2007081694A2 PCT/US2007/000050 US2007000050W WO2007081694A2 WO 2007081694 A2 WO2007081694 A2 WO 2007081694A2 US 2007000050 W US2007000050 W US 2007000050W WO 2007081694 A2 WO2007081694 A2 WO 2007081694A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
methyl
acetamide
indene
Prior art date
Application number
PCT/US2007/000050
Other languages
French (fr)
Other versions
WO2007081694A3 (en
Inventor
Gary Piazza
Robert Reynolds
Original Assignee
Southern Research Institute
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 to EA200870148A priority Critical patent/EA016323B1/en
Priority to CN2007800019364A priority patent/CN101365461B/en
Priority to EP07716225A priority patent/EP1968616A4/en
Priority to NZ569430A priority patent/NZ569430A/en
Priority to AU2007205208A priority patent/AU2007205208B2/en
Priority to MX2008008717A priority patent/MX2008008717A/en
Application filed by Southern Research Institute filed Critical Southern Research Institute
Priority to CA2635093A priority patent/CA2635093C/en
Priority to JP2008549538A priority patent/JP5337491B2/en
Priority to BRPI0706283-4A priority patent/BRPI0706283A2/en
Publication of WO2007081694A2 publication Critical patent/WO2007081694A2/en
Publication of WO2007081694A3 publication Critical patent/WO2007081694A3/en
Priority to IL192427A priority patent/IL192427A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/26Sulfones; Sulfoxides having sulfone or sulfoxide groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton
    • C07C317/32Sulfones; Sulfoxides having sulfone or sulfoxide groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton with sulfone or sulfoxide groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/44Sulfones; Sulfoxides having sulfone or sulfoxide groups and carboxyl groups bound to the same carbon skeleton
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/095Sulfur, selenium, or tellurium compounds, e.g. thiols
    • A61K31/10Sulfides; Sulfoxides; Sulfones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C321/00Thiols, sulfides, hydropolysulfides or polysulfides
    • C07C321/24Thiols, sulfides, hydropolysulfides, or polysulfides having thio groups bound to carbon atoms of six-membered aromatic rings
    • C07C321/28Sulfides, hydropolysulfides, or polysulfides having thio groups bound to carbon atoms of six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/62Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atom of at least one of the thio groups bound to a carbon atom of a six-membered aromatic ring of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/06Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with radicals, containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/08Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon radicals, substituted by hetero atoms, attached to ring carbon atoms
    • C07D207/09Radicals substituted by nitrogen atoms, not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/08Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms
    • C07D211/10Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with radicals containing only carbon and hydrogen atoms attached to ring carbon atoms
    • C07D211/12Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with radicals containing only carbon and hydrogen atoms attached to ring carbon atoms with only hydrogen atoms attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/36Radicals substituted by singly-bound nitrogen atoms
    • C07D213/38Radicals substituted by singly-bound nitrogen atoms having only hydrogen or hydrocarbon radicals attached to the substituent nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/36Radicals substituted by singly-bound nitrogen atoms
    • C07D213/40Acylated substituent nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/56Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • C07D233/61Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms with hydrocarbon radicals, substituted by nitrogen atoms not forming part of a nitro radical, attached to ring nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/64Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms, e.g. histidine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/10Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms
    • C07D295/104Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms with the ring nitrogen atoms and the doubly bound oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/108Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms with the ring nitrogen atoms and the doubly bound oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/12Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms
    • C07D295/125Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/13Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/38Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/40Radicals substituted by oxygen atoms
    • C07D307/42Singly bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/38Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/52Radicals substituted by nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/58One oxygen atom, e.g. butenolide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/04One of the condensed rings being a six-membered aromatic ring
    • C07C2602/08One of the condensed rings being a six-membered aromatic ring the other ring being five-membered, e.g. indane

Abstract

Derivatives of sulindac are provided along with-pharmaceutical compositions containing them and use for precancerous conditions and treating cancer. Derivatives of sulindac are also suitable for treating chronic inflammatory conditions. A method for preparing the derivatives is also provided.

Description

DERIVATIVES OF SULINDAC, USE THEREOF AND PREPARATION THEREOF
DESCRIPTION CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims priority from USSN 60/755,847, filed on January 4, 2006, entire disclosure of which is incorporated herein by reference
Technical Field
The present disclosure relates to certain derivatives of sulindac and especially to amide derivatives of sulindac. The present disclosure also relates to pharmaceutical compositions comprising the disclosed derivatives of sulindac, as well as a method of using the compounds in the treatment and prevention of precancerous conditions and cancer in a mammal. The present disclosure also relates to a method for producing the disclosed compounds.
BACKGROUND Even though significant advances have occurred in the treatment of cancer, it still remains a major health concern. Cancer has been reported as the leading cause of death in the United States with one of every four Americans likely to be diagnosed with the disease.
Included among the known chemotherapeutic drugs are carmustine, doxorubicin, methotrexate, paclitaxel, cyclophosphamide, procarbazine, and vinblastine, to name only a few. However, many chemotherapeutic drugs also produce undesirable side effects in the patient.
Certain nonsteroidal anti-inflammatory drugs (NSAIDs) have been recognized to have broad anticancer activity in animal models alone and in combination with chemotherapy or radiation. Representative examples include: Hial et al., "Alteration of tumor growth by aspirin and indomethacin: studies with two transplantable tumors in mouse" Eur. J. Pharm. 37: 367-376, 1976; Lynch et al., "Mechanism of inhibition of tumor growth by aspiring and indomethacin" Br. J. Cancer 38: 503-512, 1978; Bennett et al., "Increased survival of cancer- bearing mice treated with inhibitors of prostaglandin synthesis alone or with chemotherapy" Br. J. Cancer 45: 762- 768, 1982; Pollard and Luckert "Prolonged antitumor effect of indomethacin on autochthonous intestinal tumors in rats" J. Natl. Cancer Inst. 70: 1103-1105, 1983; Fulton, "Inhibition of experimental metastasis with indomethacin: role of macrophages and natural killer cells" Prostaglandins: 35: 413-425, 1988; Moorghen et al., "The effect of sulindac on colonic tumor formation in dimethylhydrazine-treated mice" Acta histochemica 29: 195-199, 1990; and Moorghen et al., "A protective effect of sulindac against chemically- induced primary colonic tumours in mice" J. of Path. 156: 341-347. Sulindac (Clinoril™) is a NSAID that has demonstrated anticancer activity. It has been recognized as having benefits for treating dysplasia as evidenced by a number of clinical trials in familial adenomatous polyposis patients which have shown the ability of sulindac to cause the regression of existing adenomas (size and number) and inhibit new adenoma (polyp) formation. For example, see Waddell et al, "Sulindac for polyposis of the colon". J. of Surg. 157: 175-179, 1989; Labayle et al., "Sulindac causes regression of rectal polyps in familial adenomatous polyposis" Gastroenterology 101: 635-639, 1991; Nugent et al., "Randomized controlled trial of the effect of sulindac on duodenal and rectal polyposis and cell proliferation in patients with familial adenomatous, polyposis" Br. J. Surg. 80: 1618- 1619, 1993; Giardiello, et al., "Treatment of colonic and rectal adenomas with sulindac in familial adenomatous polyposis" N. Eng. J. Med 328: 1313-6, 1993; and Winde et al., "Complete reversion and prevention of rectal adenomas in colectomized patients with familial adenomatous polyposis by rectal low-dose sulindac maintenance treatment." Dis. Colon Rectum 38: 813-83O5 1995.
The mechanism responsible for the anti-inflammatory efficacy and as well as the toxicity of NSAIDs and COX-2 selective inhibitors (gastrointestinal, renal, hematological, cardiovascular) has been shown to involve cyclooxygenase (COX)-I or COX-2 inhibition. Sulindac and certain other NSAIDs also have hepatic toxicity. For instance, see Vane, "Mode of action of aspirin and similar compounds" In Prostaglandin Synthetase Inhibitors, Eds Robinson, Raven Press, New York, NY, 1974; Eaker "Gastrointestinal injury related to the use of nonsteroidal anti-inflammatory drugs" Gastrointestinal Disease Today 6: 1-8, 1997; Wolfe et al., "Gastrointestinal toxicity of nonsteroidal anti-inflammatory drugs" N. Eng. J. Med 340: 1888-99, 1999; Palmer "Renal complications associated with use of nonsterdoidal anti-inflammatory agents" J. Invest. Medicine 43: 516-533, 1995; Tarazi et al., "Sulindac- associated hepatic injury: analysis of 91 cases reported to the Food and Drug Administration" Gastroenterology 104: 569-574, 1993; and Mukherjee et al. "Risk of cardiovascular events associated with selective COX-2 inhibitors" JAMA 286: 954-959, 2001.
Most investigators attribute the mechanism for the anticancer activity of NSAIDs to anti-inflammatory activity involving COX inhibition, although there is evidence for a COX- independent mechanism as mentioned below. For example, the activity of the sulfone metabolite of sulindac has been described which retains anticancer activity in preclinical and clinical trials but does not inhibit cyclooxygenase and displays less GI toxicity. See for example, Piazza et al., "Antineoplastic drugs sulindac sulfide and sulfone inhibit cell growth by inducing apoptosis" Cancer Res. 55: 3110-3116, 1995; Piazza etal., "Sulindac sulfone inhibits azoxymethane-induced colon carcinogenesis in rats without reducing prostaglandin levels" Cancer Res. 57: 2909-2915, 1997; Piazza etal., "Apoptosis primarily accounts for the growth inhibitory properties of sulindac metabolites and involves a mechanism that is independent of cyclooxygenase inhibition, cell cycle arrest, and p53 induction" Cancer Res. 57: 2452-2459, 1997; Piazza et al, "Exisulind a novel proapoptotic drug inhibits rat urinary bladder tumorigenesis" Cancer Res., 61: 3961-3968, 2001; and Chan "Nonsteroidal antiinflammatory drugs, apoptosis, and colon-cancer chemoprevention" The Lancet Oncology 3: 166-174, 2002.
There are publications suggesting that certain chemical modifications to the carboxylic acid moiety of NSAIDs will result in improved safety (i.e., as prodrugs or by localized release of nitric oxide). For example, see Mahmud et al., "A unifying hypothesis for the mechanism of NSAID related gastrointestinal toxicity". Ann. Rheumatic Diseases 55: 211-213, 1996; Venuti et al., "Synthesis and biological evaluation of (N,N,N,- trialkylammonium) alkyl esters and thioesters of carboxylic acid nonsteroidal anti- inflammatory drugs" Pharmaceutical Research 6: 867-873, 1989; Salimbeni et al., "New esters of N-arylanthranilic acids" Farmaco 30: 276-86, 1975; and Elliot et al. "A nitric oxide- releasing nonsteroidal anti-inflammatory drug accelerates gastric ulcer healing in rats" Gastroenterology 109: 524-530, 1995.
In addition, US Patents 5,401,774, 6,166,053 and 6,200,771 suggest certain modifications to sulindac sulfone which is not a NSAID.
A series of amide and ester derivatives of indomethacin and meclofenamic acid involving modifications to the carboxylic acid moiety were described by Marnett et al. These compounds were described as having safety advantages over the parent NSAIDs based on selectivity for the cyclooxygenase-2 isozyme. However, anticancer activity was not described and modifications to improve anticancer efficacy (potency) were not described. For example, see Kalgutkar et al., "Biochemical based design of cyclooxygenase-2 (COX-2) inhibitors: facile conversion of nonsteroidal anti-inflammatory drugs to potent and highly selective COX-2 inhibitors" Proc. Natl. Acad. Sci. 97: 925-930, 2000; Kalgutkar et al. "Amide derivatives of meclofenamic acid as selective cyclooxygenase-2 inhibitors" Bioorganic and Medicinal Chemistry Letters 12: 521-524, 2002; Katgutkar et al., "Ester and amide derivatives of the nonsteroidal anti-inflammatory drug, indomethacin, as selective cyclooxygenase-2 inhibitors" J. Med. Chem.43: 2860-2870, 2000; US Patent 5,973,191 to Marnett and Kalgutkar "Selective inhibitors of prostaglandin endoperoxide synthetase-2"; and US Patent 5,475,021 to Marnett and Kalgutkar "Compounds and compositions for inhibition of cyclooxygenase activity".
It cannot be predicted that chemical modifications to one family of NSAIDs are applicable to another family. For instance, meclofenamic acid belongs to the fenamate family, while sulindac belongs to the acetic acid family of NSAIDs and therefore are not structurally similar to each other. Additionally, not all amide modifications to the carboxylic acid result in increased COX-2 selectivity as demonstrated in this disclosure. Notwithstanding the advances in treatments for cancer and other diseases there still remains room for improved drugs that are effective for the desired treatment, while at the same time exhibiting reduced adverse side effects.
SUMMARY The present disclosure relates to derivatives of sulindac represented by the formula:
Figure imgf000006_0001
and pharmaceutically acceptable salts thereof, wherein X is CH3 S=O, CH3 S, HOS(=O)2, or CH3 S(O)2 ; Z is a halogen; Ri is (CHb)1nY; wherein .Y is selected from the group consisting of hydrogen , alkyl, amino, aminoalkyl, and a substituted or unsubstituted 5 "or 6 member ring;
R.2 is (CH2)mW; wherein W is selected from the group consisting of amino, aminoalkyl, and a substituted or unsubstituted 5 or 6 member ring; or wherein both Ri and R2 are interconnected and connected to the nitrogen to form a saturated or unsaturated 5 or 6 member ring, which can optionally contain a further hetero atom and can optionally be substituted.
The substituted or unsubstituted 5 or 6 member ring group for Y and W can be a saturated or unsaturated ring and includes carbon, and optionally a heteroatom such as N or O; and m is a whole number integer from 0 to 8, more typically 1 to 8 and even more typically 2-4.
Another aspect of the present disclosure relates to pharmaceutical compositions containing the above-disclosed compounds. Also disclosed are methods of using the compounds of the present disclosure in precancerous conditions and cancer in a mammal.
Another aspect of this disclosure is concerned with methods of using the compounds in treating chronic inflammatory diseases such as inflammatory bowel disease and certain neurodegenerative diseases including Alzheimer's disease.
A still further aspect of this disclosure is concerned with a method for preparing the above-disclosed compounds.
In particular, the present compounds can be produced by reacting sulindac with a compound represented by the formula: H2N(CH2)InRiR2.
Still other objects and advantages of the present disclosure will become readily apparent by those skilled in the art from the following detailed description, wherein it is shown and described only the preferred embodiments, simply by way of illustration of the best mode. As will be realized, the disclosure is capable of other and different embodiments, and its several details are capable of modifications in various obvious respects, without departing from the disclosure. Accordingly, the description is to be regarded as illustrative in nature and not as restrictive. BRIEF DESCRIPTION OF DRAWINGS
Figures IA and IB show growth inhibitory activity of derivatives according to the present disclosure compared with sulindac against Human HT29 colon tumor cells.
Figure 2 shows growth inhibitory activity of a derivative according to the present disclosure against a panel of histologically diverse tumor cell types.
Figures 3 A and 3B show cytostatic and cytotoxic activity, respectively, of a derivative according to the present disclosure compared with sulindac sulfide.
Figure 4 shows the oral bioavailability of a derivative according to the present disclosure. Figures 5 A and 5B show the reduced toxicity of a derivative according to the present disclosure in mice relative to sulindac.
Figure 6 shows antitumor activity of a derivative according to the present disclosure in athymic nude mice that were subcutaneously implanted with human colon HT29 colon tumor cells.
Best and Various Modes The present disclosure is concerned with novel derivatives of sulindac represented by the formula:
Figure imgf000008_0001
and pharmaceutically acceptable salts thereof, wherein X is CH3 S=O, CH3 S, HOS(=O)2, or CH3 S(=O)2 ; and more typically X is CH3 S=O, CH3 S, or HOS(=O)2 :
Z is a halogen; and more typically Z is fluorine; Ri is (CH2)mY; wherein Y is selected from the group consisting of hydrogen , alkyl, amino, aminoalkyl, and a substituted or unsubstituted 5 or 6 member ring;
R.2 is (CH2)mW; wherein W is selected from the group consisting of amino, aminoalkyl, and a substituted or unsubstituted 5 or 6 member ring; or wherein both Ri and R2 are interconnected and connected to the nitrogen to form a saturated or unsaturated 5 or 6 member ring, which can optionally contain a further hetero atom such as N or O and can optionally be substituted.
The substituted or unsubstituted 5 or 6 member ring group for Y and W can be a saturated or unsaturated ring and includes carbon, and optionally a heteroatom such as N or O m is a whole number integer from 0 to 8, more typically 1 to 8 and even more typically 2-4.
Examples of 5 and 6 member ring groups are phenyl; N-heterocyclo groups such as pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrrolyl, pyrazolyl, pyrazinyl pyrimidinyl, pyridazinyl, imidazoyl and imidazolidinyl; O-heterocyelo groups such as furanyl and pyranyl; heterocyclo groups containing both N and O such as morpholinyl. When substituted these groups are typically substituted with an alkyl group, amino group or aminoalkyl group. It is understood that when the Y and/or W moieties area substituted 5 or 6 member ring group the substitution is in a position other than that connected to the (CH2)Hi or to the amido N atom. The alkyl group typically contains 1-12 carbon atoms. The alkyl group more typically contains 1-4 carbon atoms. Examples of suitable alkyl groups include methyl, ethyl and propyl. Examples branched alkyl groups include isopropyl and t-butyl. Examples of alkyl substituted aromatic groups (aralkyl) are phenyl C1.3 alkyl and benzyl. Examples of aminoalkyl groups are aminomethyl, aminodimethyl, aminoethyl, aminodiethyl, aminopropyl and aminodipropyl.
Examples of pharmaceutically acceptable acid addition salts include those derived from mineral acids, such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric and sulfuric acids, and organic acids, such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, and arylsulfonic, for example p-toluenesulfonic acid. Compounds according to the present disclosure can be prepared by known methods such as disclosed in March's Advanced Organic Chemistry, Reactions, Mechanisms and Structure, Fifth Ed. John Wiley & Sons, Inc, 2001, Chapter 10, pp 506-512, disclosure of which is incorporated herein by reference. It has been found according to the present disclosure that compounds disclosed are surprisingly and advantageously useful in treating mammalian cancer, especially human colorectal cancer. For example, a dimethylaminoethylamide derivative of sulindac, referred to here as SRI 21004, has shown potent in vitro growth inhibitory activity against human colon tumor cells. SRI 21004 and a sulfide derivative (N-[2-(Dimethylamino)ethyl]-5- fiuoro-2-methyl-l-[[4-(methylsulfide)phenyl]methylene]-lH-indene-3-acetamide5 SRI 21009) display cytotoxity ICso values (50% inhibitory concentration) of 29.18 and 1.47 uM, respectively, which compares to ICso values of 479.40 and 71.22 μM for sulindac and its sulfide metabolite, respectively. This observation was unexpected since the chemical modification resulted in reduced cyclooxygenase (COX), Types 1 and 2, inhibitory activity which is widely believed to be responsible for the antineoplastic activity of sulindac. However, COX inhibition and depletion of physiologically important prostaglandins is responsible for the toxicity of NSAIDs and COX-2 selective inhibitors (i.e., celecoxib and rofecoxib), and effectively limits their utility for cancer applications that require chronic exposure and high dosages. Compounds disclosed herein are deemed to have safety and efficacy advantages over NSAlDs and COX-2 selective inhibitors for cancer and possibly other indications involving chronic inflammation. These toxicities include gastrointestinal, renal and hematological disturbances which results from COX-I inhibition as well as cardiovascular toxicity which likely results from COX-2 inhibition. In addition, the modifications of this disclosure have the potential to display reduced hepatic toxicity which is a particular problem for sulindac and sulindac sulfone (exisulind).
The chemical modification according to this disclosure is to the carboxylic acid moiety of sulindac and is especially useful since the literature teaches that COX inhibition is necessary for the anticancer activity of NSAIDs and that COX-dependent toxicity is why NSAIDs have limitations for the treatment and prevention of cancer. The chemical modification according to this disclosure renders a net positive charge to an otherwise negatively charged species that is considered to be necessary for cyclooxygenase inhibitory activity, but was unexpectedly found to enhance anticancer activity through a cyclooxygenase-independent mechanism. Compounds according to the present disclosure can be prepared by the following schemes.
Figure imgf000011_0001
Method I. PyBoP, Pyridine, O0C- RT. (compounds 1-5 & 7-9) Method II. DCC, NHS, Pyridine, (compound β)
.CH3
1. R1=H R2 = -^ "CH3 SRI# 21004 (K252-39-1)
Figure imgf000011_0002
General Procedures for the Synthesis of Sulindac Derivatives (Compounds 1- 9): Compounds 1 - 5, and 7 - 9 are synthesized by method I and compound 6 is synthesized by method II. Method I:
Sulindac (60 mg, 0.17 mmol) is dissolved in anhydrous pyridine (5 mL) under an argon atmosphere and the solution is chilled in an ice/water bath (0 0C). The appropriate amine (0.033 mL, 0.25 mmol) followed by PyBOP (benzotriazole-1-yl-oxy- trispyrrolidinophosphonium hexafluorophosphate; 130 mg, 0.25 mmol) are added to the reaction. The reaction mixture is stirred at room temperature under argon overnight. Deionized water (5 mL) is added, and the reaction mixture is extracted with chloroform (2 x 30 mL), washed with H2O (10 mL), dried over Na2SO4, and concentrated on rotary evaporator. The resulting oil is re-dissolved in toluene and co-evaporated to remove residual pyridine. The crude product is further purified by column chromatography (60-200 mesh). The analytically pure compound is obtained by drying under vacuum overnight then at 78 0C for 4 hours.
Method II: Sulindac (100 mg, 0.28 mmol) is dissolved in anhydrous pyridine (5 mL) under an argon atmosphere. DCC (86.66 mg, 0.42 mmol), NHS (48.34 mg, 0.42 mmol) and the amine (0.044 mL, 0.34 mmol) are added sequentially to the solution, and the reaction mixture is stirred for 2 days. The reaction is quenched by the addition of deionized water (5 mL). The precipitated material (mostly N,N'-dicyclohexylurea) is removed by filtration, and the mixture is evaporated on a rotary evaporator. The residue is co-evaporated with toluene to remove residual pyridine. The crude product is purified by column chromatography (60-200 mesh) and pure compound is dried overnight under vacuum then at 78 0C for 4 hours.
EXAMPLE 1
N-[2-(Dimethylamino)ethyI]-5-fluoro-2-methyl-l-[[4-(methyIsulfinyl)pheαyl]methyIene]- lH-indene-3-acetamide (1):
Crude product is purified by column chromatography using CHCl3ZMeOH + 0.2% NH4OH (9:1). The product is obtained as a yellow solid in 98% yield. ESI-MS m/z: All [M+H]+. 1H NMR (CDCl3): δ 7.74-7.65 (2H, m, 2'-H, 3'-H), 7.17 (1Η, dd, J= 5.2 Hz, 8.4 Hz, 7-H), 6.88 (1Η, dd, J = 2.4 Hz, 8.9 Hz, 4-H), 6.59 (1Η, ddd, J= 2.4 Hz, 9.0 Hz, 11.0 Hz, 6-H), 6.19 (IH, br s, -Ni7CH2CH2N(CH3)2), 3.28 (2H, dd, J= 5.7 Hz5 11,2 Hz, -
NHCH2CH2N(CH3)2), 3.51 (2H, s, 3-CH2), 2.81 (3Η, s, 4'-CZf3), 2.33 (2H, t, J= 6.0 Hz, - NHCH2CH2N(CHs)2), 2.13 (6H, s, N(CH3)2, 2.12 (3Η, s, 2-CH3). CΗN Found: C, 67.99; Η, 6.45; N, 6.60. Calcd. for C24H27FN2O2S; C, 67.59; H, 6.38; N, 6.57.
EXAMPLE 2
5-Fluoro-2-methyI-iV-[(l-methyl-2-pyrroHdinyl)inethyI]-l-[[4- (methylsuIfinyl)phenyI]methyIene]-lH-indene-3-acetamide (2):
The crude product is purified by column chromatography using CHCl3ZMeOH + 0.2% NH4OH (8:1). Yellow solid, 91% yield. ESI-MS m/z: 467 [M+H]+. 1H NMR (DMSO-ck): δ 8.10 (IH3 1, J = 4.2 Hz, -NHCH2CH2), 7.79 (IH5 d, J= 8.4 Hz, 3'-H), 7.71 (IH, d,J= 8.2 Hz, 2'-H), 7.36 ( IH, s, 8-H), 6.71 (IH5 dd, J= 5.4 Hz, 8.5 Hz5 1-H), 3.41 (2H5 S5 1-CH2), 3.12- 3.06 (2Η, m5 -NHCH2CH2), 2.88-2.79 (IH, m, 2"-H), 2.82 (3Η, s, 4'-CH3), 2.18 (3Η, s, 2- CH3), 2.12 (3Η, s, N-CH3), 2.00-1.91 (3Η, m, 3"-Ho, 5"-CH2), 1.91-1.68 (2Η, m, 3"-H6), 1.68-1.50 (2Η, m, -NHCH2CH0, 4"H6), 1.40-1.07 (2Η, m, -NHCH2CH0, 4"-Hα). CΗN Found: C, 67.62; Η, 6.48; N56.22. Calcd. for C27Η3iFN2O2S • 0.8 H2O; C5 67.40; H, 6.83; N5 5.82.
EXAMPLE 3 5-Fluoro-2-methyl-l-[[4-(methylsulfmyl)phenyl]methylene]-N-[2-(l-piperazinyl)ethyl]- lH-indene-3-acetamide (3):
The crude product is purified by column chromatography using CHCI3/MeOH + 0.2% NH4OH (4:1) and further purified by preparative TLC using CHCl3/MeOH + 0.2% NH4OH (5:1). Yellow solid, 58% yield. ESI-MS m/z: 468 [M+H]+. 1H NMR (DMSO-d6): δ.7.97 (1 H5 1, J= 4.2 Hz5 -NHCH2CH2), 7.80 (2H5 d, J= 8.4 Hz5 3'-H, 5'-H), 7.72 (2Η, d, J= 8.2 Hz, 2'-H, 6'-H), 7.35 (IH5 s, 8-H), 7.15 (IH5 dd, J= 5.3 Hz, 8.4 Hz, T-H), 7.10 (IH5 dd, J= 2.4 Hz5 9.3 Hz, A-H), 6.71 (IH, ddd, J= 2.4 Hz, 9.6 Hz, 11.8 Hz5 6-H), 3.43 (2Η, s, 1-CH2), 3.16 (2Η, dd, J= 5.94 Hz, 11.9 Hz, -NHCH2CH2), 2.66 (4H, t, J= 4.7 Hz5 3"-H& 5"-H)5 2.51- 2.30 (6Η, m5 -NHCH2CH2, 2"-H& 6"-H), 2.18 (3Η, s, 2-CH3) CHN Found: C, 63.53; H, 6.46; N, 8.53. Calcd. for C26H30FN3O2S • 1.3 H2O; C, 63.60; H5 6.69; N, 8.55. EXAMPLE 4
5-FIuoro-2-methyI-l-[[4-(methylsuIfinyI)phenyI]methyIene]-N-[2-(l-piperidinyl)ethyI]- lH-indene-3-acetamide (4):
The crude product is purified by column chromatography using CHCl3/MeOH (9:1). Yellow solid, 89% yield. ESI-MS m/z: 467 [M+H]+. 1H NMR (DMSO-d6): δ. 7.95 (IH, t, J = 4.4 Hz5 -NHCH2CH2), 7.80 (2H5 d, J= 8.4 Hz, 3'-H, 5'-H), 7.71 (2Η, d, J= 8.4 Hz, 2'-H, 6'-H), 7.35 (1Η, s, 8-H), 7.15 (1Η, dd, J= 5.4 Hz, 8.5 Hz, T-H), 7.10 (IH5 dd, J= 2.5 Hz, 9.5 Hz, A-H), 6.71 (IH, ddd, J= 2.4 Hz, 9.6 Hz, 11.0 Hz5 6-H), 3.43 (2H, s, 1-CH2), 3.16 (2Η, dd, J= 5.0 Hz, 11.0 Hz, -NHCH2CH2), 2.82 (3H5 s, 4'-CH3), 2.40-2.36 (6Η, m, -NHCH2CH2, 2"-H2, 5"-H2), 2.18 (3Η, s, 2-CH3), 1.44-1.31(6H, m, 3"-H2, 4"-H2, 5"-H2). CΗN Found: C, 67.82; Η, 6.51; N, 5.78. Calcd. for C27H3IFN2O2S • 0.7 H2O; C, 67.67; H5 6.81; N5 5.84.
EXAMPLE 5
5-FIuoro-2-methyl-l-[[4-(methylsulfinyI)phenyI]methyIene]-iV-[2-(3-pyridinyl)ethyl]-lH- indene-3-acetamide (5): The crude product is purified by column chromatography using CHCl3ZMeOH (95:5) and further purified by preparative TLC using CHCl3/MeOH (98:2). Yellow solid, 62% yield. ESI-MS m/z: 461[M+H]+. 1H NMR (DMSO-d6): 58.38-8.42 (2H5 m, 4"-H& 6"-H)5 8.17 (IH5 1, J= 4.4 Hz, -NHCH2CH2), 7.78 (2H, d, 3'-H& 5'-H), 7.72 (2Η, d, 2'-H& 6'-H), 7.60-7.56 (1Η, m, 2"-H), 7.34 (IH5 s, 8-H), 7.27-7.23 (IH5 m, 3"-H)5 7.16 (IH5 dd5 J= 5.3 Hz5 8.4 Hz, 7-H)5 6.70 (1Η, ddd, J= 2.4 Hz, 9.5 Hz, 11.0 Hz, 6-H), 3.40 (2Η, s, 1-CH2), 2.31- 3.26 (2Η, m, -NHCH2CH2 ), 2.82 (3H, s, 2- CH3), 2.74 (2Η, t5 J= 6.8 Hz, -NHCH2CH2), 2.15 (3H3 s, 4'-CH3), CΗN Found: C, 69.48; Η, 5.43; N, 5.87. Calcd. for C27H25FN2O2S • 0.3 H2O; C, 69.60; H, 5.54; N5 6.01.
EXAMPLE 6 5-Fluoro-2-methyl-l-[[4-(methyIsulfinyl)phenyl]methylene]-N-[2-(4-morpholinyl)ethyl]- lH-indene-3-acetamide (6):
The crude product is purified by column chromatography using CHCb/MeOH (95:5). Yellow solid, 75% yield. ESI-MS m/z: 469 [M+H]+. 1H NMR (DMSO-d6): ζ 8.00 (IH5 1, J= 5.0 Hz5 NHCH2CH2), 7.79 (2H, d, J= 8.5 Hz, 3'-H, 5'-H), 7.71 (2Η, d, J= 8.2 Hz, 2'-H& 6'-H), 7.35 (IH5 s, 8-H), 7.15 (1Η, dd, J= 5.2 Hz, 8.4 Hz, 7-H)5 7.10 (1Η, dd, J= 2.4 Hz5 9.4 Hz, 4- H), 6.71 (1Η, ddd, J= 2.5 Hz5 9.6 Hz, 11.1 Hz5 6-H), 3.52 (4Η, t, J= 4.6 Hz3 3"-H, 4"-H)5 3.43(2Η, s, 1-CH2), 3.21-3.15 (2Η, m, -NHCH2CH2 ), 2.82 (3H5 s, 4'-CH3), 2.35-2.30(6Η, m, 2"-H, 5"-H, -NHCH2CH2), 2.18(3Η, S, 2-CH3). CΗN Found: C, 65.81 ; Η, 5.95; N, 5.83. Calcd. for C26H29FN2O3S • 0.4 H2O; C5 65.64; H5 6.31; N, 5.89.
EXAMPLE 7
5-FIuoro-2-methyl-l-[[4-(methyIsulfinyl)phenyI]methyIene]-iV-[2-(l-pyrroIidinyl)ethy]]- lH-indene-3-acetamide (7):
The crude product is purified by column chromatography using CHCl3ZMeOH (9:1) + 0.2% NH4OH. Yellow solid, 95% yield. ESI-MS m/z: 453 [M+H]+. 1HNMR (DMSO-d6): δ 8.12 (IH, t, J= 5.0 Hz5 NHCH2CH2), 7.78 (2H5 d, J= 8.4 Hz, 3'-H5 5'-H)5 7.71 (2H5 d, J= 8.2 Hz5 2'-H5 6'-H), 7.35 (IH5 S5 8-H)5 7.18-7.09 (2H5 m, 4-H5 7-H), 6.70 (1Η, ddd, J= 2.5 Hz5 9.5Hz5 10.9 Hz5 6-H), 3.44 (2H5 s, 1-CH2), 3.202Η, dd J= 6.6 Hz59.5 Hz5 -NHCH2CH2), 2.82 (3H5 s, 4'-CH3), 2.54-2.49 (4Η, m, 3-H, 4"-H5 -NHCH2CH2), 2.18 (3Η, s, 2-CH3), 1.69 (4Η,.s, T-H%5"-H). CHN Found: C, 66.59; H5 6.22; N5 6.04. Calcd. for C26H29FN2O2S • 0.8 H2O; C5 66.87; H5 6.60; N5 6.00.
EXAMPLE 8 iV£-Dimethyl-ΛT-[2-(dimethylamino)ethyl]-5-fluoro-l-[[4- ethylsu!finyl)phenyl]methyleαe]-lH-indene-3-acetamide (8):
The crude product is purified by column chromatography using CHCl3/MeOH (9:1). Yellow solid, 62% yield. ESI-MS m/z: 441 [M+H]+. 1H NMR (DMSO-d6) The partial double bond character of the amide bond gives rise two sets of resonances: δ 7.78 (2H5 d, J= 8.4 Hz, 3'-H5 5'-H), 7.72 (2Η, d, J= 8.5 Hz, 2'-H, 6'-H), 7.35 (1Η, s, Z-H), 7.15 (1Η, dd, J= 5.5 Hz5 8.46 Hz, 7-H), 7.01-6.95 (IH, m, 4-H), 6.71 (1Η, ddd, J= 2.5 Hz5 9.5Hz5 10.9 Hz, 6-H), 3.72 & 3.65 (2H, s, 1-CH2), 3.47 &3.39 (2Η, t5 J= 5.7 Hz5 -NHCH2CH2), 3.08 & 2.84 (3H5 s, - NCH3), 2.82 (3Η, s, 4'-CH3), 2.42 & 2.32 (2H51, J= 6.7 Hz5 -NHCH2CH2 S5 2-C), 2.20 (3H5 H3S5 -N(C), 2.13 (6Η, Ha)2). CΗN Found: C, 67.41; Η, 6.51; N5 6.29. Calcd. for C25H29FN2O2S • 0.3 H2O; C5 67.33; H, 6.69; N5 6.28.
EXAMPLE 9 iV-(2-Aminoethyl)-5-fluoro-2-methyl-l-[[4-(methylsulfinyl)phenyl]methylene]-lH- indene-3-acetamide (9): The crude product is purified by column chromatography using CHCl3/Me0H + 0.2% NH4OH (9:1). The product was obtained as a yellow solid in 95% yield. ESI-MS m/z: 399 [M+Hf. 1H NMR (DMSOd6): δ 8.09 (IH, t, J= 5.2 Hz, NHCH2CH2NH2), 7.78 (2H, d, J = 8.5 Hz, y-H, 5'-H), 7.72 (2Η, d, J= 8.1 Hz, 2'-H, 6'-H), 7.35 (1Η, s, 8-H), 7.18-7.10 (2Η, m, A-H, 7-H), 6.70 (1Η, ddd, J= 2.4 Hz, 9.7 Hz, 11.1 Hz, 6-H), 3.43 (2H, s, 3-CH2), 3.05 (2Η, dd, J= 6.1 Hz, 11.9 Hz, -NHCH2CH2NH2), 2.82 (3H, s, 4'-CH3), 2.57 (2Η, t, J= 6.3 Hz, - NHCH2CH2NH2), 2.18 (3H, ss 2-CH3). CΗN Found: C, 62.47; Η, 6.33; N5 6.33. Calcd. for C22H23FN2O2S • 1.4 H2O; C, 62.36; H, 6.14; N, 6.61.
EXAMPLE 10 iV-[2-(Dimethylamino)ethyl]-5-fluoro-2-methyH-[[4-(methylthio)phenyl]metl»ylene]- lH-indene-3-acetamide (General method: Olah etal. Synthesis 1978 137.):
Triphenylphosphine (0.502 g, 1.913 mmol) and I2 (0.486 g, 1.913 mmol) are stirred together in acetonitrile (5 mL) until a yellow slurry is obtained. A solution of the sulfoxide from example (0.068 g, 0.16 mol) in acetonitrile (5mL) is added to the yellow slurry followed by the addition of powdered NaI (0.358 g, 2.39 mmol). The reaction mixture is stirred under Argon overnight at room temperature. The reaction mixture is heated gently for 6h in an attempt to push to completion, and then left at room temperature overnight under Argon. The reaction is diluted with ether (250 mL), washed with Na2S2O3 saturated solution, 5% NaHCOa, H2O, concentrated, re-dissolved in CHCl3, dried over Na2SO4 and concentrated to a yellow residue. The crude material is purified by column chromatography using chloroform/methanol 20:1 to pack and elute the column of silica gel (230-400 mesh). A yellow solid is obtained in 44% yield (29 mg). FABMS m/z: 411 (M+H)+. 1H NMR: (CDCl3) H29389 δ 2.11 (6H, s, N(CH3)2, 2.20 (3Η, s, 2-CH3), 2.31 (2Η, t, -NHCH2CH2N(CHa)2, J=5.94 Hz), 2.55 (3H, s, 4'-CH3), 3.27 (2Η, dd, -NHCH2CH2N(CH3)2, J=5.82Hz, 11.42Hz), 3.51 (2H, s, 3-CH2), 6.17 (1Η, br. s, -NHCΗ22N(CΗ3)2 ), 6.60 (IH, ddd, 6-H), 6.87 (1Η, dd, 4-H, J= 2.41Ηz, 9.0 Hz)3.7.16 (lH,s, 10-H), 7.31-7.26 (1Η, m, 6'-H), 7.38 (1Η, dd, 7-H, J= 5.27 Hz, 8.46Hz), 7.44 (2H, d, V-HSc 5'-H); the spectrum supports submitted structure with some H2O present. CHN (C24H27N2OSF) Found: C, 69.84; H, 6.77; N, 6.89. Calcd. C, 70.21; H, 6.63; N, 6.82. In addition, the following compounds according to the present disclosure can be prepared by the following schemes:
Figure imgf000017_0001
Method I. PyBoP, Pyridine, 00C- RT. (compounds 1&2) Method 1I1II1IV. PyBoP, Pyridine, RT. (compound 3,4,5)
11. R1=H SRI# 21211 (K301-61)
Figure imgf000017_0002
O
12. R1=H R2 = U SRI# 21222 (K301-63-1)
HN^s. SRI# 21229 (K301-65-1)
13. R1=H R2 =
Figure imgf000017_0003
SRI# 21275 (K301-79-1)
Figure imgf000017_0004
General Procedures for the Synthesis of Sulindac Derivatives (Compounds 11-15):
Compounds 11 and 12 are synthesized by method I9 compound 13 is synthesized by method II, compound 14 is synthesized by method III, and compound 15 is synthesized by method IV.
Method I:
Sulindac (100 mg, 0.28 mmol) is dissolved in anhydrous pyridine (5 mL) under an argon atmosphere and solution is chilled in ice/water bath (~0 0C). Amine (0.045 mL, 0.42 mmol) followed by PyBOP (benzotriazole-1-yl-oxy-trispyrrolidinoρhosphonium hexafluorophosphate; 219 mg, 0.42 mmol) are added to the reaction. The reaction mixture is stirred at room temperature under argon overnight. Deionized water (5 mL) is added and reaction mixture is extracted with chloroform (2 x 30 mL), washed with H2O (10 mL), dried over Na2SO4, concentrated on rotary evaporator. It is re-dissolved in toluene and co- evaporated to remove residual pyridine. The crude product is purified by column chromatography (60-200 mesh). The pure compound is dried under vacuum for at least 48 hours.
Method II:
Sulindac (100 mg, 0.28 mmol) is dissolved in anhydrous pyridine (5 mL) under an argon atmosphere. Amine (77 mg, 0.42 mmol) followed by PyBOP (benzotriazole-1-yl-oxy- trispyrrolidinophosphonium hexafluorophosphate; 219 mg, 0.42 mmol) are added to the reaction. The reaction mixture is stirred at room temperature under argon overnight. Amine (26 mg, 0.14 mmol) is added to the reaction to increase the sulindac to amine ration 1:2. Reaction mixture is stirred at room temperature under argon for 72 hours. Deionized water (5 mL) is added to the reaction mixture to quench the reaction. The reaction mixture is co- evaporated with toluene to remove residual pyridine. The crude product is purified by column chromatography (60-200 mesh). The pure compound is dried under vacuum for at least 48 hours.
Method III:
Sulindac (100 mg, 0.28 mmol) is dissolved in anhydrous pyridine (5 mL) under an argon atmosphere. Amine (0.06 mL, 0.42 mmol) followed by PyBOP (benzotriazole-1-yl- oxy-trispyrrolidinophosphonium hexafluorophosphate; 219 mg, 0.42 mmol) are added to the reaction. Reaction mixture is stirred at room temperature under argon for 72 hours. Deionized water (5 mL) is added and reaction mixture is extracted with chloroform (2 x 30 mL), washed with H2O (10 mL), dried over Na2SO*, concentrated on rotary evaporator. It is re-dissolved in toluene and co-evaporated to remove residual pyridine. The crude compound is dried under vacuum for 72 hours. The crude product is purified by column chromatography (60-200 mesh). The pure compound is co-evaporated with ethanol (3 x 50 mL). Pure compound is dried on drying pistol overnight.
Method IV:
Sulindac (100 mg, 0.28 mmol) is dissolved in anhydrous pyridine (5 mL) under an argon atmosphere. Amine (95 mg, 0.42 mmol) followed by PyBOP (benzotriazole-1-yl-oxy- trispyrrolidinophosphonium hexafluorophosphate; 219 mg, 0.42 mmol) are added to the reaction. Reaction mixture is stirred at room temperature under argon for two weeks. Deionized water (5 mL) is added to the reaction mixture to quench the reaction. The reaction mixture is co-evaporated with toluene to remove residual pyridine. The crude product is purified by column chromatography (60-200 mesh). The pure compound is co-evaporated with ethanol (2 x 50 mL). Pure compound is dried on drying pistol overnight.
EXAMPLE 11
5-Fluoro-2-methyϊ-l-[[4-(methyIsulJRnyI)phenyl]methylene]-iV-(phenylmethyl)-lH- indene-3-acetamide (11): Crude product is purified by column chromatography using CHCb/MeOH (98:2).
The product is obtained as yellow solid in 91% yield. ESI-MS m/z: 446 [M+H]+. 1H NMR (DMSOd6): δ 8.64 (IH, t, J= 6.2 Hz, -NHCH2), 7.79 (2H, d, J= 8.3 Hz, 3'-H, 5'-H), 7.72 (2Η, d, J = 8.3 Hz, 2'-H, 6'-H), 7.35 (1Η, s, 8-H), 7.32-7.23 (5Η, m, C6H5), 7.17 (1Η, dd, J= 5.2 Hz, 8.4 Hz, 7-H), 7.13 (1Η, dd, J= 2.5 Hz, 9.5 Hz, 4-H), 6.71 (IH, ddd, J= 2.5 Hz, 8.5 Hz, 9.4 Hz, 6-H), 4.29 (2Η, d, J= 5.8 Hz, -NHCH2), 3.51 (2Η, s, 3-CH2), 2.82 (3Η, s, CH3SO-), 2.20. (3Η, s, 2-CH3). CΗN Found: C, 71.48; Η, 5.36; N, 3.22. Calcd. for C27H24FNO2S • 0.4 H2O; C, 71.63; H, 5.52; N, 3.09. EXAMPLE 12
S-Fluoro-N-Cl-furanylmethyO^-methyl-l-ft^CmethylsulfiπyOpheny^methylenel-lH- indene-3-acetamide (12):
Crude product is purified by column chromatography using CHCl3ZMeOH (95:5) and further purified by preparative TLC using CHCl3ZMeOH (95:5). Yellow solid, 34% yield. ESI-MS m/z: 436[M+H]+. 1H NMR (DMSO-d6): δ 8.61(1H, t, J= 5.3 Hz5 -NHCH2),7.79 (2H, d, J= 8.3 Hz, 3'-H5 5'-H), 7.72 (2H, d, J- 8.3 Hz, 2'-H, 6'-H), 7.57 (1Η, dd, J= 0.9 Hz5 1.9 Hz, 5"-H7.35 (1Η, s, S-H), 7.16 (1Η, dd, J= 5.3 Hz, 8.4 Hz, 7-H), 7.11 (1Η, dd, /= 2.4 Hz, 9.4 Hz, A-H), 6.71 (IH, ddd, J= 2.5 Hz, 8.4 Hz, 9.4 Hz, 6-H), 6.39 (IH, dd, J= 1.9 Hz, 3.2 Hz, 4"-H), 6.22 (1Η, dd, J= 0.8 Hz, 3.2 Hz, 3"-H), 4.28 (2Η, d, J= 5.6 Hz, -NHCH2), 3.47 (2Η, s5 3-CH2), 2.82 (3Η, s, CH3SO-), 2.18 3Η, s, Ϊ-CH^ CHN Found: C, 66.72; H, 4.88; N, 3.09. Calcd. for C25H22FNO3S • 0.75 H2O; C, 66.87; H, 5.27; N, 3.12.
EXAMPLE 13 5-Fluoro-Λ'-[2-(4-imidazolyI)ethyI]-2-πiethyl-l-[[4-(methylsulfiπyl)phenyI]methylene- lH-indene-3-acetamide (13):
Crude product is purified by column chromatography using CHCb/MeOH + 0.2% NH4OH (9:1) and further purified by preparative TLC using CHCl3ZMeOH + 0.2% NH4OH (8:1). Yellow solid, 54% yield of reacted material, 31% yield. ESI-MS m/z\ 450[M+H]+. 1H NMR (DMSO-dg The imidazole ring gives rise two sets of resonances δ 8.16(1H, br t, J= 5.3 Hz, -NHCH2CH2), 7.79 (2H, d, J = 8.4 Hz, 3'-H, 5'-H), 7.72 (2Η, d, J= 8.4 Hz, 2'-H, 6'-H), 7.50 (1Η, br s, 2"-H), 7.35 (1Η, s, 8-H), 7.16 (1Η, dd, J= 5.3 Hz, 8.4 Hz, 7-H), 7.09 (1Η, dd, J= 2.5 Hz, 9.4 Hz, A-H), 6.83 (IH, br s, 4"-H), 6.71 (1Η, ddd, J= 2.5 Hz7 8.4 Hz, 9.5 Hz, 6- H), 3.42 (2Η, s, 3-CH2), 3.26 (2Η, m, -NHCH2CH2), 2.82 (3H, s, CH3SO-), 2.65 (2Η, m, - NHCH2CH2), 2.17 DQ s, 2-CH3J CΗN Found: C, 64.22; Η, 5.37; N, 8.71. Calcd. for C25H24FN3O2S • 1.0 H2O; C, 64.22; H, 5.60; N, 8.99.
EXAMPLE 14
5-FIuoro-iV-[2-(iV,iV-diethyIainino)ethyl]-2-methyI-l-[[4- (methylsulfinyl)phenyl] rnethylene-lH-indene-3-acetamide (14) :
Crude product is purified by column chromatography using CHCl3/MeOH (95:5) and further purified by preparative TLC using CHCl3ZMeOH + 1.0% NH4OH (9: 1 ) and by preparative TLC using CHCl3ZMeOH + 1.0% NH4OH (7: 1 ). Yellow solid, 28% yield. ESI- MS m/z: 455[M+H]+. 1H NMR (DMSO-d6) δ 7.92(1H, t, J= 5.3 Hz, -NHCH2CH2),7.79 (2H, d, J= 8.4 Hz, 3'-H, 5'-H), 7.71 (2Η, d, J= 8.4 Hz, 2'-H, 6'-H), 7.35 (1Η, s, 8-H), 7.16 (1Η, dd, J= 5.2 Hz, 8.4 Hz, 7-H)5 7.10 (1Η, dd, J = 2.5 Hz, 9.4 Hz, 4-H), 6.72 (1Η, ddd, J= 2.5 Hz, 8.4 Hz, 9.4 Hz5 6-H), 3.43 (2H, s, 3-CH2), 3.12 (2Η, m, -NHCH2CH2), 2.82 (3H, s,
CH3SO-), 2.44 (4Η, m, -N(CH2CHs)2), 2.41 (2H, m, -NHCH2CH2), 2.18 3H3 s, 2-CH3), 0.91 (6Η, t, J= 7.1 Hz, -N(CH2CH3)^ CΗN Found: C, 66.67; Η, 6.98; N, 5.91. Calcd. for C26H31FN2O2S • 0.8 H2O; C5 66.58; H, 7.01; N, 5.97.
EXAMPLE 15 iV-[[4-(Dimethylainino)phenyI]inethyl]-5-fluoro-2-inethyl-l-[[4- (methyIsuIfinyl)phenyllmethylene]-lH-indene-3-acetamide (15):
Crude product is purified by column chromatography using CHCl3ZMeOH + 1.0% NH4OH (95:5) and further purified by preparative TLC using CHCl3ZMeOH (9:1). Yellow solid, 36% yield of reacted material, 26% yield. ESI-MS m/z: 489[M+H}+. 1HNMR (DMSO-ds) ζ 8.48(1H, t, J= 5.3 Hz, -NHCH2), 7.79 (2H, d, J= 8.4 Hz, 3'-H, 5'-H), 7.72 (2Η, d, J= 8.4 Hz, 2'-H, 6'-H), 7.35 (1Η, s, 8-H), 7.16 (1Η, dd, J= 5.3 Hz, 8.4 Hz5 7-H), 7.11 (IH, dd, J= 2.6 Hz, 9.6 Hz, 4-H), 7.08 (2Η, m, 2"-H), 6.71 (1Η, ddd, J= 2.4 Hz, 8.4 Hz, 9.5 Hz, 6-H), 6.66 (2Η, m, 3"-H), 4.15 (2Η, d, J= 5.6 Hz, -NHCH2), 3.47 (2Η, s, 1-CH2), 2.85 (6Η, s, -N(CHa)2) 2.82 (3Η, s, CH3SO-), 2.19.3Η, s, 2-CH3) CΗN Found: C, 71.07; Η, 6.06; N, 5.70. Calcd. for C29H29FN2O2S; C, 71.29; H, 5.98; N, 5.73.
Method IV
Figure imgf000022_0002
Figure imgf000022_0001
(SRI # 21387)
Example 16
Ethanone, 2-[5-fluoro-2-methyI-l-[[4-(niethylsuIfonyl)phenyl]methyIenel-lH-inden-3- yl]-l-(4-methyIpiperazinyl)
Crude product was purified by column chromatography using CHCl3ZMeOH + 0.2% NH4OH (95:5). The product is obtained as a yellow solid in modest yield. ESI-MS m/z: 439.23 [M+H]+. 1H NMR (CDCl3, 300 MHz): δ 7.73-7.64 (4H, m, 2'-H5 3'-H, 5'-H, 6'-H)3 7.16 (1Η, dd, 3J = 8.7 Hz, 4J = 5.4 Hz, 7-H)5 7.12 (1Η, s, S-H), 6.90 (1Η, dd, 3J = 9.0 Hz, 4J = 2.4 Hz, A-H), 6.58-6.51 (IH, m, 6-H), 3.69 (2H, m, CH2NCH3), 3.60 ( 2H, s> CH2CO), 3.55 (2H, m CH2NCH3),.2.81 (3H> s, SOCH3), 2.42-2.37 (4H, m, CONCH2), 2.30 (3Η, s, NCHj), 2.18 (3Η, s, 2-CH3). Anal. Calcd for C25H27FN2O2S • 0.1 CH3COOH: C, 65.17; H, 5.87; N, 5.63. Found: C, 64.80; H, 6.15; N, 5.74.
Compounds 17, 18, 19 are synthesized by method V and compounds 20 is synthesized by method IV. Method V
O-(BenzotriazoI-l-yl)-N,N,N\N'-tetramethyluronium hexafluorophosphate, (HBTU) (114 mg, 0.2996 mmol) is added to a solution of sulindac sulfide or sulindac (0.2497 mmol), the appropriate amine (0.2996 mmol) and Et3N (0.07 mL, 0.4994 mmol) in dry acetonϊtrile (10 mL) at room temperature under argon atmosphere. Reaction mixture is stirred at room temperature for 30 minutes. Saturated brine (30 mL) is added to the reaction mixture and extracted with CHCI3 (2 x 30 mL). The combined organic extracts are dried over anhydrous Na2SO4 and the solvent is evaporated off under reduced pressure. The crude product is purified by flash column chromatography (60-200 mesh). Method VI
Diphenylphosphoryl azide, DPPA (0.052 mL, 0.2417 mmol) is added to a solution of sulindac sulfone (75 mg, 0.2014 mmol) and N,N-dimethylethylenediamine (0.026 mL, 0.2417 mmol) in dry DMF (2 mL) at 00C under argon atmosphere. Reaction mixture is allowed to room temperature and stirred overnight. Aqueous Na2COs (30 mL) is added to the reaction mixture and extracted with CHCI3 (2 x 30 mL). The combined organic extracts are dried over anhydrous Na2SO4 and the solvent is evaporated off under reduced pressure. The crude product is purified by flash column chromatography (60-200 mesh).
EXAMPLE 17
N-[[4-(Dimethylamino)phenyl]methyI]-(Z)-5-fluoro-2-methyl-3-[[4- (methylthio)phenyl]rπethylene]-lH-indene-3-acetamide
Crude product is purified by column chromatography using CHCU/Petroleum ether + 0.1% NH4OH (4:1)). The product is obtained as a yellow solid in 98% yield. ESI-MS m/z: 473.26 [M+H]\ 1H NMR (CDCl3, 300 MHz): δ 7.45 (2H, d, 3J = 8.1 Hz, 3'-H, 5'-H), 7.38 (IH5 dd, 3J = 8.4 Hz, 4J = 5.1 Hz, 1-H), 7.30 (2H, dd, 3J = 8.4 Hz, 2'-H, 6'-H), 7.14 (1Η, s, 8-H), 7.03 (2Η, d, J = 8.7 Hz, 2"-H, 6"-H), 6.87 (1Η, dd, 3J = 9.0 Hz, 4J = 2.4 Hz, 4-H)5 6.64-6.56 (3Η, m, 6-H, 3"-H, 5"-H), 5.75 (m, bs, NH), 4.32 ( 2Η, d5 J = 5.6 Hz, NH-CH2-), 3.56 ( 2Η, s, - CH2-CO), 2.90 (6H, s, N(CH3)2), 2.54 (3Η, s, -SCHj)52.17 (3Η, s, 2-CH5). Anal. Calcd for C29H29FN2OS: C, 73.70; H, 6.18; N, 5.93. Found: C, 73.74; H5 6.17; N5 5.72. EXAMPLE 18
N^-Dimethyl-(Z)-5-fluoro-2-methyl-N-[2-(methylamino)ethyl]-3-[|;4- (methylthio)phenyl]methylene] ]-lH-indene-3-acetamide Crude product is purified by column chromatography using CHC^/MeOH + 0.1% NH4OH (97:1). The product is obtained as a yellow solid in 82% yield. ESI-MS m/z: 411.26 [M+H]+, 821.49 [2M+H]+. 1HNMR (CDCl3, 300 MHz): δ 7.45 (2H, d, 3J = 8.1 Hz, 3'-H, 5'-H), 7.65 (2Η, d, J =8.1 Hz, 2'-H5 6'-H), 7.15-7.10 ( 2Η, m's, 4-H5 8-H)29 6.96-6.90 (IH5 m, 1-H), 6.59-6.52 (IH, m, 6-H)5 3.69, 3.62 ( 2Η, 2s, -CH2-CO), 3.55-3.46 (2Η, m, N(CH3)-CH2), 3.09, 2.97 (3Η, 2s, CO-N(CH3)), 2.79-2.74 (2Η, m, CH2-NH(CH3)), 2.54 (3H, s, -SCHj)5 2.45, 2.42 (3Η, 2s, NHCHj), 2.18 (3Η, s, 2-CH3). Anal. Calcd for C24H27FN2OS - 0.35 H2O: C, 69.15; H, 6.69; N, 6.71. Found: C, 69.00; H, 6.50; N, 6.29. EXAMPLE 19
N-[[4-(Dimethylamino)phenyl]-(Z>-5-fluoro-2-methyI-3-[[4- - (methylsulfinyl)phenyl]methylene] ]-lH-indene-3-acetamide
Crude product is purified by column chromatography using CHCla/MeOH + 0.1% NH4OH (99:1). The product is obtained as a yellow solid in 98% yield. ESI-MS m/z: 475.21 [M+H]+ 949.42 [2M+H]\ 1H NMR (CDCl3, 300 MHz): δ 7.75-7.67 (4H, m, 2'-H, V-H 5'-H, 6'-H)3 7.25 (2Η, d, 3Jf = 9.0 Hz, 2"-H5 6"-H), 7.22 (1Η, s, 8-H), 7.20 (1Η, dd, 3J = 8.4 Hz, 4J = 5.1 Hz, 7-H), 7.13 (1Η, bs, NH), 6.95 (1Η, dd, 3J = 8.7 Hz, 4J = 2.4 Hz, A-H), 6.66 (2H, d, 3J = 9.0 Hz, 3"-H5 5"-H), 6.67-6.57 (1Η, m, 6-H), 3.66 (2Η, s, CH2-CO), 2.89 (6Η, s, N(CHj)2), 2.82 (3H, s, -SOCH3), 2.27 (3H, s, 2-CH5). Anal. Calcd for C28H27FN2O2S • 1.0 H2O: C, 68.27; H, 5.93; N, 5.69. Found: C5 68.34; H, 5.66; N, 5.80. EXAMPLE 20
N-[2-(Dimethylamino)ethyl]-(Z)-5-fluoro-2-methyl-3-ΪI4- (methylsulfonyl)phenyl]methylene] ]-lH-indene-3-acetamide Crude product is purified by column chromatography using CHCb/MeOH + 0.1% NH4OH (49:1). The product is obtained as a yellow solid in 94% yield. ESl-MS m/z: 443.22 [M+H]+. 1H NMR (CDCl3, 300 MHz): δ 8.02 (2H, d, 3J = 8.4 Hz, 3'-H, 5'-H)5 7.72 (2Η, d, 3J = 8.7 Hz5 2'-H, 6'-H), 7.13 (1Η, s, 8-H), 7.11 (1Η, dd, 3J = 8.4 Hz5 4J = 5.1 Hz5 7-H), 6.90 (IH5 dd, 3J - 9.0 Hz, 4J = 2.7 Hz5 4-H)5 6.61-6.54 (IH5 m5 6-H)5 6.31 (IH5 bs, NH)5 3.50 ( 2Η, S5 CH2-CO), 3.30 ( 2H, q, J = 5.7 Hz5 NH-CH2), 3.14 (3H5 s, SO2CH3), 2.36 (2H, t5 CH2-N(CHs)2), 2.21 (3H5 s, 2-CHj)5 2.16 (6Η, s, N(CHj)2). Anal. Calcd for C24H27FN2O3S • 0.25 H2O: C, 64.48; H, 6.20; N, 6.27. Found: C5 64.54; H, 6.05; N, 6.29.
EXAMPLE 21 N-[[4-(DimethyIamino)phenyl]-CZ)-5-fluoro-2-raethyl-3-[[4- (methylthio)phenyl) methylene] ]-lH-indene-3-acetamide
Crude product is purified by column chromatography using 2% MeOH-CHC^ + 0.1% NH4OH. The product is obtained as a yellow solid in 100% yield. ESI-MS m/z: 459.25 [M+H]+, 917.49 [2M+H]+. 1H NMR (CDCl3, 300 MHz): δ 7.47 (2H, d, 3J = 8.2 Hz, 2'-H, 6'- H), IAA (1Η, dd, 3J = 8.4 Hz, 4J = 5.2 Hz, 7-H)5 7.31 (2H5 d, 3J - 8.2 Hz, 3'-H5 5'-H)5 7.22 (2H5 dd J = 9.0Hz5 2"-H, 6"-H) 7.21 (IH5 S5 8-H)5 7.08 (1Η, bs, NH), 6.93 (1Η, dd, 3J = 8.8 Hz5 4J = 2.4 Hz, A-H), 6.67-6.60 (3H5 overlapping m's, 3"-H, 5"-H, 6-H), 3.66 ( 2Η, s, CH2- CO)5 2.89 (6H, s, N(CHj)2), 2.56 (3Η, s, -SCH3), 2.26 (3H, S5 2-CH3). Anal. Calcd for C2βH27FN2OS«0.25 H2O: C5 72.62; H5 5.99; N, 6.05. Found: C5 72.41; H5 5.61; N, 5.74.
Experimental Results
In vitro cytotoxicity testing using the human HT29 colon tumor cell line reveals significant potency improvements of the sulindac derivatives SRl 21004(sulfoxide) and SRI 21009(sulfide) relative to the corresponding forms of sulindac. Growth inhibitory activity of SRI derivatives and sulindac using the tritiated thymidine incorporation assay (left panel) or the MTS cell viability assay (right panel) are shown in Figures IA and IB. Note the increased potency of SRl derivatives compared to sulindac and its sulfone and sulfide metabolites, Also note that higher concentrations of sulindac and its metabolites were required to reduce viable cell number compared to concentrations required to inhibit proliferation, which was not the case for the SRI derivatives. This indicates greater potential of the SRI derivatives to kill tumor cells compared to sulindac and its metabolites.
Figure 2 shows growth inhibitory activity of SRI 21009 against a panel of histologically disease tumor cell types. Note the enhanced sensitivity of tumor cells derived from colorectal tumors compared with other tumor types.
Figures 3A and 3B show cytostatic and cytotoxic activity, respectively, of SRI 21004 compared with sulindac sulfide. Cytotoxic activity is defined as the ability the treatment to reduce cell number below the number when treatment was initiated and is an indicator of chemotherapeutic efficacy. Cytostasis is defined as the ability of the treatment to reduce cell number relative to untreated cells and is an indicator of chemopreventive efficacy. Note that SRI 21004 displays greater potential for chemotherapeutic efficacy compared with sulindac sulfide and is consistent with greater ability to kill tumor cells as indicated in Figure IB.
Figure 4 shows the oral bioavailability of SRI 21009. The achievable plasma levels following the oral administration of SRI 21009 at a single dose of 200 mg/kg exceeded the IC50 value for in vitro growth inhibition. Negligible amounts of sulindac sulfoxide, sulfone, or sulfide are detected in the plasma following SRI 21009 administration. Figures 5 A and 5B show the reduced toxicity of SRI 21009 in mice relative to sulindac. Following a once a day dosing schedule, the maximum tolerated dose of sulindac is estimated to be 50 mg/kg in comparison to 300 mg/kg for SRI 21009. Figure 6 shows antitumor activity of SRI 21009 in athymic nude mice that are subcutaneously implanted with human colon HT29 colon tumor cells. For this study, SRI 21009 is administered orally once every two days at a dose of 700 mg/kg. SRI 21009 administered at 300 mg/kg is as effective as sulindac at 50 mg/kg using a once a day dosing schedule.
Table A below shows the reduced COX-I and COX-2 inhibitory activity of SRI 21009 in comparison to a non-selective COX inhibitor (sulindac sulfide), a COX-I selective inhibitor (indomethacin), and a COX-2 selective inhibitor (rofecoxib). The inhibitory activity of the compounds is expressed as an IC50 value (50% inhibitory concentration).
Table A COX-I and Cox-2 inhibitory activity of known NSAIDs and SRI 210009
Figure imgf000027_0001
Additional derivatives of sulindac sulfoxide are synthesized and tested for growth inhibitory activity against human HT29 colon tumor cells are shown in Table 1 below. AU derivatives are more active than sulindac sulfoxide, with several displayed comparable or greater activity compared SRI 21004. Table B
Figure imgf000028_0001
Figure imgf000029_0001
In above tables-Growth inhibitory activity of sulindac and a group of SRI derivatives against the human HT29 colon tumor cell line. Activity is expressed as potency by the IC50 value (50% inhibitory concentration). It is noted that 4-N, N dimethyl phenylamide compounds of this disclosure are not especially preferred since they are less active than the 4- N5N dimethyl benzylamide compounds. For instance, compare compound 21590 and 21623 to 21275 and 21540, respectively. Also, it is believed that the SCH3 compound (21623) is less active than the corresponding SOCH3 (21590) due at least in part to being less hydrophilic resulting in the noted solubility issues during the bioassay. In keeping with the present disclosure, the derivatives of sulindac can be used alone or in appropriate association, and also may be used in combination with pharmaceutically acceptable carriers and other pharmaceutically active compounds such as various cancer treatment drugs including NSAIDs and/or along with radiation. The active agent may be present in the pharmaceutical composition in any suitable quantity. The pharmaceutical compositions can include components typically used for such purposes.
The pharmaceutically acceptable carriers described herein, for example, vehicles, adjuvants, excipients, or diluents, are well-known to those who are skilled in the art. Typically, the pharmaceutically acceptable carrier is chemically inert to the active compounds and has no detrimental side effects or toxicity under the conditions of use. The pharmaceutically acceptable carriers can include polymers and polymer matrices. Another additive for the pharmaceutical compositions can be cyclodextrin for enhancing absorption of the active agent.
The choice of carrier will be determined in part by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical composition of the present invention. The following formulations for oral, aerosol, parenteral, subcutaneous, intravenous, intraarterial, intramuscular, intraperitoneal, intrathecal, rectal, and vaginal administration are merely exemplary and are in no way limiting.
Formulations suitable for oral administration can include (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granule; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions. Liquid formulations may include diluents, such as water, cyclodextrin, dimethyl sulfoxide and alcohols, for example, ethanol, benzyl alcohol, propylene glycol, glycerin, and the polyethylene alcohols including polyethylene glycol, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent. Capsule forms can be of the ordinary hard-or soft- shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and corn starch. Tablet forms can include one or more of the following: lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guargum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers. Lozenge forms can comprise the active ingredient in a flavor, such as sucrose and acacia or tragacaπth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acadia, emulsions, and gels containing, the addition to the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acadia, emulsions, and gels containing, in addition to the active ingredient, such carriers as are known in the art.
The derivatives of sulindac alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation. These aerosol formulations can be placed into pressurized acceptable propel lants, such as dichlorodifluoromethane, propane, and nitrogen. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer.
Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blopd of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The compound can be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol such as poly(ethyleneglycol) 400, glycerol ketals, such as 2,2- dimethyl-1, 3-dioxolane-4-methanol, ethers, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcelluslose, or emulsifying agents and other pharmaceutical adjuvants.
Oils, which can be used in parenteral formulations include, petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters. Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts, and suitable detergents include (a) cationic detergents such as, for example, dimethyldialkylammoniurn halides, and alkylpyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolarnides, and polyoxyethylene polypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl β-aminopropionates, and 2-alkylimidazoline quaternary ammonium salts, and (e) mixtures thereof.
The parenteral formulations typically contain from about 0.5% to about 25% by , weight of the active ingredient in solution. Suitable preservatives and buffers can be used in such formulations. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-Hpophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations ranges from about 5% to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol. Pharmaceutically acceptable excipients are also well-known to those who are skilled in the art. The choice of excipient will be determined in part by the particular compound, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical composition of the present disclosure. The following methods and excipients are merely exemplary and are in no way limiting. The pharmaceutically acceptable excipients preferably do not interfere with the action of the active ingredients and do not cause adverse side-effects. Suitable carriers and excipients include solvents such as water, alcohol, and propylene glycol, solid absorbants and diluents, surface active agents, suspending agent, tableting binders, lubricants, flavors, and coloring agents. The formulations can be presented in unit-does or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets. The requirements for effective pharmaceutical carriers for injectable compositions are well known to those of ordinary skill in the art. See Pharmaceutics and Pharmacy Practice, J.B. Lippincott Co., Philadelphia, PA, Banker and Chalmers, Eds., 238-250 (1982) and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., 622-630 (1986).
Formulations suitable for topical administration include lozenges comprising the active ingredient in a flavor, such as sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier; as well as creams, emulsions, and gels containing, in addition to the active ingredient, such carriers as are known in the art.
Additionally, formulations suitable for rectal administration may be presented as suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases. Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
One skilled in the art will appreciate that suitable methods of exogenously administering a compound of the present disclosure to an animal are available, and , although more than one route can be used to administer a particular compound, a particular route can provide a more immediate and more effective reaction than another route.
The present disclosure further provides a method of treating precancerous conditions or dysplosia (i.e. - intraepithelial neoplasia) as well as cancer in a mammal, especially humans. The method comprises administering an effective treatment amount of a derivative of sulindac disclosed above to the mammal. As regards these applications, the present method includes the administration to an animal, particularly a mammal, and more particularly 'a human, of a therapeutically effective amount of the compound effective in the inhibition of neoplasia and tumor growth and treating malignant disease including metastases, especially colorectal cancer. The method also includes the administration of a therapeutically effect amount of the compound for the treatment of and precancerous lesions such as adenomatous polyps of the colon and other dysplastic lesions of the skin (actinic keratosis), bladder, cervix, esophagus, oral cavity, lung, prostate and breast sometimes referred to as intraepithelial neoplasia. The disclosed compounds and compositions can be administered to treat a number of cancers, including leukemias and lymphomas such as acute lymphocytic leukemia, acute nonlymphocytic leukemias, chronic lymphocytic leukemia, chronic myelogenous leukemia, Hodgkin's Disease, non-Hodgkin's lymphomas, and multiple myeloma, childhood solid tumors such as brain tumors, neuroblastoma, retinoblastoma, Wilms Tumor, bone tumors, and soft-tissue sarcomas, common solid tumors of adults such as lung cancer, breast cancer, prostate cancer, urinary cancers, uterine cancers, oral cancers, pancreatic cancer, melanoma and other skin cancers, stomach cancer, ovarian cancer, brain tumors, liver cancer, laryngeal cancer, thyroid cancer, esophageal cancer, and testicular cancer. The present disclosure also relates to treating certain chronic inflammatory conditions which NSAIDs have shown benefit, but may be contraindicated due to cyclooxygenase inhibition (i.e. - inflammatory bowel disease) or do not appear to require cyclooxygenase inhibition for efficacy such as certain neurodegenerative diseases, including Alzheimer's Disease. The dose administered to an animal, particularly a human, in the context of the present invention should be sufficient to affect a therapeutic response in the animal over a reasonable time frame. One skilled in the art will recognize that dosage will depend upon a variety of factors including the condition of the an imal, the body weight of the animal, as well as the severity and stage of the cancer. A suitable dose is that which will result in a concentration of the active agent in tumor tissue which is known to affect the desired response. The preferred dosage is the amount which results in maximum inhibition of cancer, without unmanageable side effects.
The total amount of the compound of the present disclosure administered in atypical treatment is typically between about 10 mg/kg and about 1000 mg/kg of body weight for mice, and between about 100 mg/kg and about 500 mg/kg of body weight, and more typically between 200 mg/kg and about 400 mg/kg of body weight for humans per daily dose. This total amount is typically, but not necessarily, administered as a series of smaller doses over a period of about one time per day to about three times per day for about 24 months, and more typically over a period of twice per day for about 12 months. The size of the dose also will be determined by the route, timing and frequency of administration as well as the existence, nature and extent of any adverse side effects that might accompany the administration of the compound and the desired physiological effect. It will be appreciated by one of skill in the art that various conditions or disease states, in particular chronic conditions or disease states, may require prolonged treatment involving multiple administrations.
The method disclosed also comprises further administering a chemotherapeutic agent other than the derivatives of the present invention. Any suitable chemotherapeutic agent can be employed for this purpose. The chemotherapeutic agent is typically selected from the group consisting of alkylating agents, antimetabolites, natural products, anti-inflammatory agents, hormonal agents, molecular targeted drugs, anti-angiogenic drugs, and miscellaneous agents.
Examples of alkylating chemotherapeutic agents include carmustine, chlorambucil, . cisplatin, lomustine, cyclophosphamide, melphalan, mechlorethamine, procarbazine, thiotepa, uracil mustard, triethylenemelamine, busulfan, pipobroman, streptozocin, ifosfamide, dacarbazine, carboplatin, and hexamethylmelamine.
Examples of chemotherapeutic agents that are antimetabolites include cytosine arabinoside fluorouracil, gemcitabine, mercaptopurine, methotrexate, thioguanine, floxuridine, fludarabine, and cladribine.
Examples of chemotherapeutic agents that are natural products include actϊnomycin
D, bleomycin, camptothecins, daunomycin, doxorubicin, etoposide, mitomycin C, paclitaxel, taxoteredocetaxel, tβnisposide, vincristine, vinblastine, vinorelbine, idarubicin, mitoxantrone, mithramycin and deoxycoformycin. Examples of hormonal agents include antiestrogen receptor antagonists such as tamoxifen and fluvestrant, aromatase inhibitors such as anastrozole, androgen receptor antagonists such as cyproterone and flutamine, as well as gonadotropin release hormone agonists such as leuprolide. Examples of anti-inflammatory drugs include adrenocorticoids such as prednisone, and nonsteroidal anti-inflammatory drugs such as sulindac or celecoxib. Examples of molecular targeted drugs include monoclonal antibodies such as rituximab, cetuximab, trastuzumab and small molecules such as imatinib, erlotinib, ortizumib.
Examples of anti-angiogenic drugs include thalidomide and bevacizimab. Examples of the aforesaid miscellaneous chemotherapeutic agents include mitotane, arsenic trioxide, tretinoin, thalidomide, levamisole, L-asparaginase and hydroxyurea. The term "comprising" (and its grammatical variations) as used herein is used in the inclusive sense of "having" or "including" and not in the exclusive sense of "consisting only of." The terms "a" and "the" as used herein are understood to encompass the plural as well as the singular. The foregoing description illustrates and describes the disclosure. Additionally, the disclosure shows and describes only the preferred embodiments but, as mentioned above, it is to be understood that it is capable to use in various other combinations, modifications, and environments and is capable of changes or modifications within the scope of the invention concepts as expressed herein, commensurate with the above teachings and/or the skill or knowledge of the relevant art. The embodiments described herein above are further intended to explain best modes known by applicant and to enable others skilled in the art to utilize the disclosure in such, or other, embodiments and with the various modifications required by the particular applications or uses thereof. Accordingly, the description is not intended to limit the invention to the form disclosed herein. Also, it is intended to the appended claims be construed to include alternative embodiments.
All publications and patent applications cited in this specification are herein incorporated by reference, and for any and all purposes, as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference.

Claims

CLAIMSWhat is claimed is:
1. Compound represented by the formula:
Figure imgf000037_0001
and pharmaceutically acceptable salts thereof, , wherein X is CH3 S=O5 CH3 S, HOS(=0)2, or CH3 S(=O)2 ;
Z is a halogen;
Ri is (CH2)mY; wherein Y is selected from the group consisting of hydrogen , alkyl, amino, aminoalkyl, and a substituted or unsubstituted 5 or 6 member ring;
R2 is (QHb)1nW; wherein W is selected from the group consisting of amino, aminoalkyl, and a substituted or unsubstituted 5 or 6 member ring; or wherein both Ri and R2 are interconnected and connected to the nitrogen to form a saturated or unsaturated 5 or 6 member ring, which can optionally contain a further hetero atom and can optionally be substituted, and m is a whole number integer from 0 to 8.
2. The compound of claim 1 wherein X is CH3S(=O)2.
3. The compound of claim 1 wherein X is CH3S.
4. The compound of claim 1 wherein X is CH3S=O.
5. The compound of claim 1 bemgN-[2-(dimethylamino)ethyl]-5-fluoro-2-methyl-l-[[4- (methylsulfmyl)phenyl]methylene]-lH-indene~3-acetamide or pharmaceutically acceptable salts thereof.
6. The compound of claim 1 being 5-fluoro-2-methyl-JV-[(l-methyI-2- pyrrolidinyl)methy I]- 1 -[[4-(methy lsulfinyl)phenyl]methylene]- 1 H-indene-3-acetamide or pharmaceutically acceptable salt thereof.
7. The compound of claim 1 being 5-fluoro-2-methyl-l-[[4-
(methylsulfiny^phenyllmethylenej-N-p-Cl-piperaziny^ethyll-lH-indene-S-acetamide or pharmaceutically acceptable salt thereof.
8. The compound of claim 1 being 5-fluoro-2-methyl-l-[[4-
(methylsulfinyl)phenyl]methylene]-N-[2-(l-piperidinyl)ethyl]-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
9. The compound of claim 1 being 5-fluoro-2-methyl-l-[[4- (methylsulfinyQphenylJmethyleneJ-iV-P-CS-pyridinyOethyO-lH-indene-S-acetamide or pharmaceutically acceptable salt thereof.
10. The compound of claim 1 being 5-fluoro-2-methyl-l-[[4-
(methylsulfinyl)phenyljmethylene]-N-[2-(4-morpholinyl)ethyl]-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
11. The compound of claim 1 being 5-fluoro-2-methyl-l-[[4-
(methylsulfinyOphenylJmethylenej-iV-P-Cl-pyrrolidinyOethylJ-lH-indene-S-acetamide or pharmaceutically acceptable salt thereof.
12. The compound of claim 1 being N,2-dimethyl-N-[2-(dimethylamino)ethyl]-5-fiuoro- 1 -[[4-ethylsulfinyI)phenyl]methyIene]-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
13. The compound of claim 1 beingN-(2-aminoethyI)-5-fluoro-2-methyl-l-[[4- (methylsuifinyl)phenyl]methylene]-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
14. The compound of claim 1 beingiV-[2-(diπiethylammo)ethyl]-5-fluoro-2-methyl-l-[[4- (methylthio)phenyl]methylene]-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
15. The compound of claim 1 being 5-fluoro-2-methyl-l-[[4- (methylsulfiny^phenyljmethylenej-iV-CphenylmethyO-lH-indene-S-acetamide or pharmaceutically acceptable salt thereof.
16. The compound of claim 1 being 5-fluoro-N-(2-furanylmethyl)-2-methyl-l-[[4- (methylsulfinyl)phenyl]methylene]-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
17. The compound of claim 1 being 5-fiuoro-N-[2-(4-imidazolyl)ethyl]-2-methyl-l-[[4- (methylsulfinyl)phenyl]methylene-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
18. The compound of claim 1 being 5-fluoro-N-[2-(ΛζN-diethylamino)ethyl]-2-methyl-l- [[4-(methylsuIfinyl)phenyl]methylene-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
19. The compound of claim 1 being Ν-[[4-(dimethylamino)phenyl]methyl]-5-fluoro-2- methyl-l-[[4-(methylsulfinyl)phenyl]methylene]-lH-iήdene-3-acetamide or pharmaceutically acceptable salt thereof.
20. The compound of claim 1 being ethanone, 2-[5-fiuoro-2-methyl-l -[[4- (methylsulfonyl)phenyl]methylene]-lH-inden-3-yl]-l-(4-methylpiperazinyl) or pharmaceutically acceptable salt thereof.
21. The compound of claim 1 being Ν-[[4-(dimethylamino)phenyl]methyl]-(Z)-5-fluoro- 2-methyl-3-[[4-(methylthio)phenyl]methylene]-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
22. The compound of claim 1 being Ν,2-dimethyl-;(Z)-5-fluoro-2-methyl-Ν-[2- (methylamino)ethyl]-3-[[4-(methylthio)phenyl]methylene] ]-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
23. The compound of claim 1 being N-[[4-(dimethylamino)phenyl]-(Z)-5-fluoro-2- methyl-3-[[4-(methylsulfinyl)phenyl]methylene] ]-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
24. The compound of claim 1 being N-[2-(dimethylamino)ethyl]-(Z)-5-fluoro-2-methyl- 3-[[4-(methylsulfonyl)ρhenyl]methylene] ]-lH-indene-3-acetamide or pharmaceutically acceptable salt thereof.
25. The compound of claim 1 being N-[2-(dimethylamino)ethyl]-5-fluoro-2-methyl-l- [[4-(methylsulfide)phenyl]methylene]-lH-indene-3-acetamide or the pharmaceutically acceptable salt thereof.
26. A pharmaceutical composition comprising at least one compound according to anyone of claims 1-25 and a pharmaceutically acceptable carrier.
27. A method of treating a precancerous condition or cancer in a mammal comprising administering to the mammal an effective treatment amount of at least one compound according to anyone of claims 1-25.
28. The method of claim 27, wherein the cancer is colon cancer.
29. The method of claim 27 wherein the treatment amount is from about 10 mg/kg to about 1000 mg/kg of the body weight of the mammal.
30. The method of claim 27 wherein the treatment amount is from about 100 mg/kg to about 500 mg/kg of the body weight of the mammal.
31. The method of claim 27 wherein the treatment is carried out over a period of from one time per day to about 3 times per day for 24 months.
32. The method of claim 27 wherein the derivative is administered orally, intravenously or intraperitoneally.
33. The method of claim 28 wherein the mammal is human.
34. A method for preparing a compound according to anyone of claims 1-25 which comprises reacting sulindac with a compound represented by the formula: NH2(CH2)mNRt R2-
PCT/US2007/000050 2006-01-04 2007-01-04 Derivatives of sulindac, use thereof and preparation thereof WO2007081694A2 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
CN2007800019364A CN101365461B (en) 2006-01-04 2007-01-04 Derivatives of sulindac, use thereof and preparation thereof
EP07716225A EP1968616A4 (en) 2006-01-04 2007-01-04 Derivatives of sulindac, use thereof and preparation thereof
NZ569430A NZ569430A (en) 2006-01-04 2007-01-04 Derivatives of sulindac, use thereof and preparation thereof
AU2007205208A AU2007205208B2 (en) 2006-01-04 2007-01-04 Derivatives of sulindac, use thereof and preparation thereof
MX2008008717A MX2008008717A (en) 2006-01-04 2007-01-04 Derivatives of sulindac, use thereof and preparation thereof.
EA200870148A EA016323B1 (en) 2006-01-04 2007-01-04 Derivatives of sulindac, use thereof and preparation thereof
CA2635093A CA2635093C (en) 2006-01-04 2007-01-04 Derivatives of sulindac, use thereof and preparation thereof
JP2008549538A JP5337491B2 (en) 2006-01-04 2007-01-04 Sulindac derivatives, their use and their preparation
BRPI0706283-4A BRPI0706283A2 (en) 2006-01-04 2007-01-04 compound, pharmaceutical composition and methods of compound preparation and treatment of precancerous or cancerous conditions in mammals
IL192427A IL192427A0 (en) 2006-01-04 2008-06-24 Derivatives of sulindac, use thereof and preparation thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US75584706P 2006-01-04 2006-01-04
US60/755,847 2006-01-04

Publications (2)

Publication Number Publication Date
WO2007081694A2 true WO2007081694A2 (en) 2007-07-19
WO2007081694A3 WO2007081694A3 (en) 2007-11-01

Family

ID=38256870

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/000050 WO2007081694A2 (en) 2006-01-04 2007-01-04 Derivatives of sulindac, use thereof and preparation thereof

Country Status (13)

Country Link
US (1) US8044048B2 (en)
EP (1) EP1968616A4 (en)
JP (1) JP5337491B2 (en)
KR (1) KR20080091134A (en)
CN (1) CN101365461B (en)
AU (1) AU2007205208B2 (en)
BR (1) BRPI0706283A2 (en)
CA (1) CA2635093C (en)
EA (1) EA016323B1 (en)
IL (1) IL192427A0 (en)
MX (1) MX2008008717A (en)
NZ (1) NZ569430A (en)
WO (1) WO2007081694A2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010008427A1 (en) * 2008-04-11 2010-01-21 Ludwig Institute For Cancer Research Ltd. Tryptophan catabolism in cancer treatment and diagnosis
WO2014068461A2 (en) * 2012-11-02 2014-05-08 Mahesh Kandula Compositions and methods for the treatment of acute inflammation
US9365528B2 (en) 2011-04-01 2016-06-14 Southern Research Institute Derivatives of sulindac, use thereof and preparation thereof
WO2016099452A1 (en) * 2014-12-16 2016-06-23 Adt Pharmaceuticals, Inc. Ras-inhibiting indenyl acetamide compounds, compositions, and uses
EP3233794A1 (en) * 2014-12-16 2017-10-25 ADT Pharmaceuticals, Inc. Indenyl compounds, pharmaceutical compositions, and medical uses thereof
US11104658B2 (en) 2014-12-16 2021-08-31 Adt Pharmaceuticals, Llc Method of treating or preventing Ras-mediated diseases
EP3687972A4 (en) * 2017-09-28 2021-09-29 Medicon Pharmaceuticals, Inc. Anti-inflammatory, anti-cancer, and anti-angiogenic compounds, pharmaceutical compositions, and methods of making and using thereof
US11186596B2 (en) 2018-04-26 2021-11-30 Adt Pharmaceuticals, Llc Anticancer indenes, indanes, azaindenes, azaindanes, pharmaceutical compositions and uses

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103002887B (en) * 2010-05-06 2016-12-21 桑福德-伯纳姆医学研究所 Relate to the method and composition of Retinoid receptor selective channel
WO2018170410A1 (en) * 2017-03-17 2018-09-20 University Of South Alabama Derivatives of sulindac can protect normal cells against oxidative damage
US10329249B1 (en) 2018-10-03 2019-06-25 King Saud University Sulindac derivatives

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AT290523B (en) 1962-01-05 1971-06-11 Merck & Co Inc Process for the production of new α- (3-indolyl) -carboxylic acids
US3647858A (en) 1970-05-01 1972-03-07 Merck & Co Inc Process for preparing 1-benzylidene-3-indenyl acetic acids
US3654349A (en) 1970-05-01 1972-04-04 Merck & Co Inc Substituted indenyl acetic acids
US3851063A (en) 1972-07-21 1974-11-26 Merck & Co Inc Treatment of pain,fever or inflammation
US4423075A (en) 1980-06-19 1983-12-27 Ayerst, Mckenna & Harrison Inc. Aldose reductase inhibition by 5-fluoro-2-methyl-1-[[4-(methylsulfonyl)phenyl]methylene]-1H-indene-3-acetic acid
US5401774A (en) 1991-03-08 1995-03-28 University Of Arizona Method for treating patients with precancerous lesions by administering substituted sulfonyl idenyl acetic and propionic acids and esters to patients with lesions sensitive to such compounds
EP0508586B1 (en) * 1991-03-08 1995-05-31 Fgn, Inc. Substituted indenyl compounds
US20050090553A1 (en) 1992-06-30 2005-04-28 Shapiro Howard K. Compositions and method for treatment of chronic inflammatory diseases
US5475021A (en) 1993-12-03 1995-12-12 Vanderbilt University Compounds and compositions for inhibition of cyclooxygenase activity
US6063818A (en) 1996-06-13 2000-05-16 Cell Pathways Inc. Substituted benzylidene indenyl formamides, acetamides and propionamides
HUP9904696A2 (en) 1996-11-12 2000-05-28 American Home Products Corporation Indene inhibitors of cox-2
US5973191A (en) 1996-12-30 1999-10-26 Vanderbilt University Selective inhibitors of prostaglandin endoperoxide synthase-2
US6071934A (en) * 1997-03-25 2000-06-06 Cell Pathways, Inc. Indenyl hydroxamic acids, (hydroxy) ureas and urethanes for treating patients with precancerous lesions
CA2238283C (en) 1997-05-30 2002-08-20 Cell Pathways, Inc. Method for identifying compounds for inhibition of neoplastic lesions, pharmaceutical compositions from such compounds and uses of such compounds and compositions for treating neoplastic lesions
US5858694A (en) 1997-05-30 1999-01-12 Cell Pathways, Inc. Method for identifying compounds for inhibition of cancerous lesions
CN100436418C (en) 1997-12-12 2008-11-26 Osi制药公司 N-Benzyl-3-indenylacetamides derivatives for treating neoplasia
US5942520A (en) 1998-01-27 1999-08-24 Cell Pathways, Inc. Method for inhibiting neoplastic cells by exposure to substituted N-cycloalkylmethyl-1-H-pyrazolo (3,4-B) quinolone-4 amines
US6388054B1 (en) * 1998-08-20 2002-05-14 John M. Stewart Anti-cancer compounds
US6200771B1 (en) 1998-10-15 2001-03-13 Cell Pathways, Inc. Method of using a novel phosphodiesterase in pharmaceutical screeing to identify compounds for treatment of neoplasia
US6130053A (en) * 1999-08-03 2000-10-10 Cell Pathways, Inc. Method for selecting compounds for inhibition of neoplastic lesions
US6235776B1 (en) * 1998-11-12 2001-05-22 Cell Pathways, Inc. Method for treating a patient with neoplasia by treatment with a paclitaxel derivative
US6207700B1 (en) 1999-01-07 2001-03-27 Vanderbilt University Amide derivatives for antiangiogenic and/or antitumorigenic use
ITMI20030617A1 (en) * 2003-03-28 2004-09-29 Dinamite Dipharma S P A In Forma A Bbreviata Diph PROCEDURE FOR THE PREPARATION OF AN ORGANIC COMPOUND CONTAINING A SULFINYL OR SULFONYL GROUP
JP2004315511A (en) * 2003-03-31 2004-11-11 Taisho Pharmaceut Co Ltd Mch receptor antagonist
CA2562783A1 (en) * 2004-04-26 2005-12-01 Vanderbilt University Indoleacetic acid and indenacetic acid derivatives as therapeutic agents with reduced gastrointestinal toxicity
JP2006124387A (en) * 2004-09-30 2006-05-18 Taisho Pharmaceut Co Ltd New quinoline, tetrahydroquinazoline, and pyrimidine derivative, and treating method associated with their use

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
None
See also references of EP1968616A4

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010008427A1 (en) * 2008-04-11 2010-01-21 Ludwig Institute For Cancer Research Ltd. Tryptophan catabolism in cancer treatment and diagnosis
US9365528B2 (en) 2011-04-01 2016-06-14 Southern Research Institute Derivatives of sulindac, use thereof and preparation thereof
WO2014068461A2 (en) * 2012-11-02 2014-05-08 Mahesh Kandula Compositions and methods for the treatment of acute inflammation
WO2014068461A3 (en) * 2012-11-02 2014-12-24 Mahesh Kandula Compositions and methods for the treatment of acute inflammation
US10981886B2 (en) 2014-12-16 2021-04-20 Adt Pharmaceuticals, Llc Indenyl compounds, pharmaceutical compositions, and medical uses thereof
US11407727B2 (en) 2014-12-16 2022-08-09 Adt Pharmaceuticals, Llc Indenyl compounds, pharmaceutical compositions, and medical uses thereof
US10526307B2 (en) 2014-12-16 2020-01-07 Adt Pharmaceuticals, Llc Indenyl compounds, pharmaceutical compositions, and medical uses thereof
US10975054B2 (en) 2014-12-16 2021-04-13 Adt Pharmaceuticals, Llc Indenyl compounds, pharmaceutical compositions, and medical uses thereof
WO2016099452A1 (en) * 2014-12-16 2016-06-23 Adt Pharmaceuticals, Inc. Ras-inhibiting indenyl acetamide compounds, compositions, and uses
US11104658B2 (en) 2014-12-16 2021-08-31 Adt Pharmaceuticals, Llc Method of treating or preventing Ras-mediated diseases
US11130744B2 (en) 2014-12-16 2021-09-28 Adt Pharmaceuticals, Llc Indenyl compounds, pharmaceutical compositions, and medical uses thereof
EP3233794B1 (en) * 2014-12-16 2023-02-01 ADT Pharmaceuticals, LLC Indenyl compounds, pharmaceutical compositions and medical uses thereof
EP4053105A1 (en) * 2014-12-16 2022-09-07 ADT Pharmaceuticals, LLC Indenyl compounds, pharmaceutical compositions, and medical uses thereof
US11198679B2 (en) 2014-12-16 2021-12-14 Adt Pharmaceuticals, Llc Method of treating or preventing Ras-mediated diseases
EP3233794A1 (en) * 2014-12-16 2017-10-25 ADT Pharmaceuticals, Inc. Indenyl compounds, pharmaceutical compositions, and medical uses thereof
EP3687972A4 (en) * 2017-09-28 2021-09-29 Medicon Pharmaceuticals, Inc. Anti-inflammatory, anti-cancer, and anti-angiogenic compounds, pharmaceutical compositions, and methods of making and using thereof
US11752146B2 (en) 2017-09-28 2023-09-12 Medicon Pharmaceuticals, Inc. Anti-inflammatory, anti-cancer, and anti-angiogenic compounds, pharmaceutical compositions, and methods of making and using thereof
US11186596B2 (en) 2018-04-26 2021-11-30 Adt Pharmaceuticals, Llc Anticancer indenes, indanes, azaindenes, azaindanes, pharmaceutical compositions and uses
US11680073B2 (en) 2018-04-26 2023-06-20 Adt Pharmaceuticals, Llc Anticancer indenes, indanes, azaindenes, azaindanes, pharmaceutical compositions and uses

Also Published As

Publication number Publication date
WO2007081694A3 (en) 2007-11-01
JP2009522364A (en) 2009-06-11
CA2635093A1 (en) 2007-07-19
EP1968616A2 (en) 2008-09-17
EA200870148A1 (en) 2008-12-30
BRPI0706283A2 (en) 2011-03-22
MX2008008717A (en) 2008-09-10
JP5337491B2 (en) 2013-11-06
US8044048B2 (en) 2011-10-25
IL192427A0 (en) 2009-02-11
EP1968616A4 (en) 2009-10-21
CN101365461B (en) 2013-07-24
NZ569430A (en) 2012-05-25
US20070244122A1 (en) 2007-10-18
CA2635093C (en) 2012-06-26
AU2007205208A1 (en) 2007-07-19
KR20080091134A (en) 2008-10-09
CN101365461A (en) 2009-02-11
EA016323B1 (en) 2012-04-30
AU2007205208B2 (en) 2012-11-01

Similar Documents

Publication Publication Date Title
US8044048B2 (en) Derivatives of sulindac, use thereof and preparation thereof
AU2019261938A1 (en) Small molecule degraders of polybromo-1 (PBRM1)
JP2004520282A (en) Cyanoguanidine prodrug
WO2020257278A2 (en) Small molecule target bromo/acetyl proteins and uses thereof
US9365528B2 (en) Derivatives of sulindac, use thereof and preparation thereof
CS274446B2 (en) Method of 1-acyl-3-/4-/2-pyrimidinyl/-1-piperazinyl/propanes production
US8236850B2 (en) Dimeric derivatives of artemisinin and application in anticancer therapy
ES2636469T3 (en) New derivatives of pyrido [3,4-c] [1,9] phenanthroline and 11,12-dihydropyrid [3,4-c] [1,9] phenanthroline and their use, particularly for the treatment of cancer
US6376518B1 (en) Acyl derivatives of 4-demethylpenclomedine, use thereof and preparation thereof
AU2006210831B2 (en) Derivatives of 4-demethylpenclomedine, use thereof and preparation thereof
KR20180011949A (en) Novel 2-amine substituted 1,4-naphthoquinone compounds and pharmaceutical composition for preventing or treating cancer comprising the same as an active ingredient
DK3186268T3 (en) ESTERS OF STEROID LACTAM AND BIS (2-CHLORETHYL) -AMINOPHENOXY PROPIONIC ACID DERIVATIVES
KR20230118883A (en) Imidazole compounds as ENPP1 inhibitors
NZ616076B2 (en) Derivatives of sulindac, use thereof and preparation thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2007205208

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 569370

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2635093

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 569430

Country of ref document: NZ

REEP Request for entry into the european phase

Ref document number: 2007716225

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/008717

Country of ref document: MX

Ref document number: 2008549538

Country of ref document: JP

Ref document number: 2007716225

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200780001936.4

Country of ref document: CN

Ref document number: 1020087016410

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2007205208

Country of ref document: AU

Date of ref document: 20070104

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 200870148

Country of ref document: EA

ENP Entry into the national phase

Ref document number: PI0706283

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080630