Method for treating diseases using HSP90-inhibiting agents in combination with enzyme inhibitors

Information

  • Patent Application
  • 20050020557
  • Publication Number
    20050020557
  • Date Filed
    May 27, 2004
    20 years ago
  • Date Published
    January 27, 2005
    19 years ago
Abstract
The present invention provides a method for treating cancer. The method involves the administration of an HSP90 inhibitor and an enzyme inhibitor, where the combined administration provides a synergistic effect. In one aspect of the invention, a method of treating cancer is provided where a subject is treated with a dose of an HSP90 inhibitor in one step and a dose of an enzyme inhibitor in another step. In another aspect of the invention, a method of treating cancer is provided where a subject is first treated with a dose of an HSP90 inhibitor and subsequently treated with a dose of an enzyme inhibitor. In another aspect of the invention, a method of treating cancer is provided where a subject is first treated with a dose of an enzyme inhibitor and subsequently treated with a dose of an HSP90 inhibitor.
Description
BACKGROUND OF THE INVENTION
Field of the Invention

This invention relates to methods for treating cancer in which an inhibitor of Heat Shock Protein 90 (“HSP90”) is combined with an enzyme inhibitor. More particularly, this invention relates to combinations of the HSP90 inhibitor geldanamycin and its derivatives, especially 17-alkylamino-17-desmethoxygeldanamycin (“17-AAG”) and 17-(2-dimethylaminoethyl)amino-17-desmethoxygeldanamycin (“17-DMAG”), with an enzyme inhibitor (e.g., SAHA and Iressa).


References

Agnew et al., “Clinical pharmacokinetics of 17-(allylamino)-17-demethoxy-geldanamycin and the active metabolite 17-(amino)-17-demethoxygeldanamycin given as a one-hour infusion daily for 5 days.” AACR, 2002.


An et al., “Depletion of p185erbB2, Raf-1 and mutant p53 proteins by geldanamycin derivatives correlates with antiproliferative activity.” Cancer Chemother. Pharmacol. 40:60-64, 1997.


Bagatell et al., “Induction of a heat shock factor 1-dependent stress response alters the cytotoxic activity of hsp90-binding agents.” Clin. Cancer Res. 6:3312-3318, 2000.


Bagatell et al., “Destabilization of steroid receptors by heat shock protein 90-binding drugs: a ligand-independent approach to hormonal therapy of breast cancer.” Clin. Cancer Res. 7:2076-2084, 2001.


Banerji et al., “A pharmacokinetically (PK)-pharmacodynamically (PD) driven Phase I trial of the HSP90 molecular chaperone inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG).” AACR, 2002.


Barent et al., “Analysis of FKBP51/FKBP52 chimeras and mutants for Hsp90 binding and association with progesterone receptor complexes.” Mol. Endocrinol. 12:342-354, 1998.


Bilodeau et al., “Tyrosine kinase inhibitors.” U.S. Pat. No. 6,245,759 issued Jun. 12, 2001.


Citri et al., “Drug-induced ubiquitylation and degradation of ErbB receptor tyrosine dinases: implications for cancer chemotherapy.” EMBO Journal 21:2407-2417, 2002.


Egorin et al., “Metabolism of 17-(allylamino)-17-demethoxygeldanamycin (NSC 330507) by murine and human hepatic preparations.” Cancer Res. 58:2385-2396, 1998.


Fraley et al., “Tyrosine kinase inhibitors.” U.S. Pat. No. 6,306,874 issued Oct. 23, 2001.


Fraley et al., “Tyrosine kinase inhibitors.” U.S. Pat. No. 6,313,138 issued Nov. 6, 2001.


Gaidigk et al., “AND(P)H:quinone oxidoreductase: polymorphisms and allele frequencies n Caucasian, Chinese and Canadian Native Indian and Inuit populations.” Pharmacogenetics 8:305-313, 1998.


Gelmon et al., “Anticancer agents targeting signaling molecules and cancer cell environment: challenges for drug development?” J. Natl. Cancer Inst. 91:1281-1287, 1999.


Goetz et al., “The Hsp90 chaperone complex as a novel target for cancer therapy.” Ann. Oncol. 14:1169-1176, 2003.


Goh et al., “Explaining interindividual variability of docetaxel pharmacokinetics and pharmacodynamics in Asians through phenotyping and genotyping strategies.” J. Clin. Oncol. 20:3683-3690, 2002.


Grenert et al., “The amino-terminal domain of heat shock protein 90 (hsp90) that binds geldanamycin is an ATP/ADP switch domain that regulates hsp90 conformation.” J. Biol. Chem. 272:23843-23850, 1997.


Johnson and Toft, “Binding of p23 and hsp90 during assembly with the progesterone receptor.” Mol. Endocrinol. 9:670-678, 1995.


Hart1 and Hayer-Hart1, “Molecular chaperones in the cytosol: from nascent chain to folded protein.” Science 195:1852-1858, 2002.


Hegde et al., “Short circuiting stress protein expression via a tyrosine kinase inhibitor, herbimycin A.” J. Cell Physiol. 165:186-200, 1995.


Hustert et al., “The genetic determinants of the CYP3A5 polymorphism.” Pharmacogenetics 11:773-779, 2001.


Kelland et al., “DT-Diaphorase expression and tumor cell sensitivity to 17-allylamino, 17-demethoxygeldanamycin, an inhibitor of heat shock protein 90.” J. Natl. Cancer Inst. 91:1940-1949, 1999.


Kuehl et al., “Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression.” Nat. Genet. 27:383-391, 2001.


Lawson et al., “Geldanamycin, an hsp90/GRP94-binding drug, induces increased transcription of endoplasmic reticulum (ER) chaperones via the ER stress pathway.” J. Cell Physiol. 174:170-178, 1998.


Lin et al., “Co-regulation of CYP3A4 and CYP3A5 and contribution to hepatic and intestinal midazolam metabolism.” Mol. Pharmacol. 62:162-172, 2002.


Morimoto et al., “The heat-shock response: regulation and function of heat-shock proteins and molecular chaperones.”Essays Biochem. 32:17-29, 1997.


Munster et al., “Phase I trial of 17-(allylamino)-17-demethoxygeldanamycin (17-AAG) in patients with advanced solid malignancies.” Proc. Am. Soc. Clin. Oncol, 83a, 2001.


Munster et al., “Modulation of Hsp90 function by ansamycins sensitizes breast cancer cells to chemotherapy-induced apoptosis in an RB- and schedule-dependent manner.” Clin. Cancer Res. 7:2228-2236, 2001.


Murakami et al., “Induction of hsp 72/73 by herbimycin A, an inhibitor of transformation by tyrosine kinase oncogenes.” Exp. Cell Res. 195:338-344, 1991.


Pratt and Toft, “Steroid receptor interactions with heat shock protein and immunophilin chaperones.” Endocr. Rev. 18:306-60, 1997.


Prodromou et al., “Identification and structural characterization of the ATP/ADP-binding site in the Hsp90 molecular chaperone.”Cell 90:65-75, 1997.


Richter and Buchner, “Hsp90: chaperoning signal transduction.” J. Cell. Physiol. 188:281-290, 2001.


Rosvold et al., “Identification of an NAD(P)H:quinone oxidoreductase polymorphism and its association with lung cancer and smoking.” Pharmacogenetics 5:199-206, 1995.


Schneider et al., “Pharmacologic shifting of a balance between protein refolding and degradation mediated by Hsp90.” Proc. Natl. Acad. Sci. USA 93:14536-14541, 1996.


Schnur et al., “erbB-2 oncogene inhibition by geldanamycin derivatives: synthesis, mechanism of action, and structure-activity relationships.” J. Med. Chem. 38:3813-20, 1995.


Schnur et al., “Inhibition of the oncogene product p185erbB-2 in vitro and in vivo by geldanamycin and dihydrogeldanamycin derivatives.” J. Med. Chem. 38:3806-3812, 1995.


Smith et al., “Progesterone receptor structure and function altered by geldanamycin, an hsp90-binding agent.” Mol. Cell Biol. 15:6804-6812, 1995.


Smith et al., “Identification of a 60-kilodalton stress-related protein, p60, which interacts with hsp90 and hsp70.” Mol. Cell Biol. 13:869-876, 1993.


Stebbins et al., “Crystal structure of an Hsp90-geldanamycin complex: targeting of a protein chaperone by an antitumor agent.” Cell 89:239-250, 1997.


Traver et al., “NAD(P)H:quinone oxidoreductase gene expression in human colon carcinoma cells: characterization of a mutation which modulates DT-diaphorase activity and mitomycin sensitivity.” Cancer Res. 52:797-802, 1992.


Whitesell et al., “Inhibition of heat shock protein HSP90-pp60v-src heteroprotein complex formation by benzoquinone ansamycins: essential role for stress proteins in oncogenic transformation.” Proc. Natl Acad. Sci. USA 91:8324-8328, 1994.


Young et al., “Hsp90: a specialized but essential protein-folding tool.” J. Cell Biol. 154:267-273, 2001.


Zou et al., “Repression of heat shock transcription factor HSF1 activation by HSP90 (HSP90 complex) that forms a stress-sensitive complex with HSF1.” Cell 94:471-480, 1998.


Discussion

Geldanamycin (figure below, R17═OCH3) is a benzoquinone ansamycin polyketide isolated from Streptomyces geldanus. Although originally discovered by screening microbial extracts for antibacterial and antiviral activity, geldanamycin was later found to be cytotoxic to certain tumor cells in vitro and to reverse the morphology of cells transformed by the Rous sarcoma virus to a normal state.
embedded image

Geldanamycin's nanomolar potency and apparent specificity for aberrant protein kinase dependent tumor cells, as well as the discovery that its primary target in mammalian cells is the ubiquitous Hsp90 protein chaperone, has stimulated interest in the development of this compound as an anti-cancer drug. However, the association of unacceptable hepatotoxicity with the administration of geldanamycin led to its withdrawal from Phase I clinical trials.


More recently, attention has focused on 17-amino derivatives of geldanamycin, in particular 17-(allylamino)-17-desmethoxygeldanamycin (“17-AAG”, R17═—NCH2CH═CH2). This compound has reduced hepatotoxicity while maintaining useful Hsp90 binding. Certain other 17-amino derivatives of geldanamycin, 11-oxogeldanamycin, and 5,6-dihydrogeldanamycin, are disclosed in U.S. Pat. Nos. 4,261,989, 5,387,584 and 5,932,566, each of which is incorporated herein by reference. Treatment of cancer cells with geldanamycin or 17-AAG causes a retinoblastoma protein-dependent G1 block, mediated by down-regulation of the induction pathways for cyclin D-cyclin dependent cdk4 and cdk6 protein kinase activity. Cell cycle arrest is followed by differentiation and apoptosis. G1 progression is unaffected by geldanamycin or 17-AAG in cells with mutated retinoblastoma protein; these cells undergo cell cycle arrest after mitosis, again followed by apoptosis.


The above-described mechanism of geldanamycin and 17-AAG appears to be a common mode of action among the benzoquinone ansamycins that further includes binding to Hsp90 and subsequent degradation of Hsp90-associated client proteins. Among the most sensitive client protein targets of the benzoquinone ansamycins are the Her kinases (also known as ErbB), Raf, Met tyrosine kinase, and the steroid receptors. Hsp90 is also involved in the cellular response to stress, including heat, radiation, and toxins. Certain benzoquinone ansamycins, such as 17-AAG, have thus been studied to determine their interaction with cytotoxins that do not target Hsp90 client proteins.


U.S. Pat. Nos. 6,245,759, 6,306,874 and 6,313,138, each of which is incorporated herein by reference, disclose compositions comprising certain tyrosine kinase inhibitors together with 17-AAG and methods for treating cancer with such compositions. Münster, et al., “Modulation of Hsp90 function by ansamycins sensitizes breast cancer cells to chemotherapy-induced apoptosis in an RB- and schedule-dependent manner,” Clinical Cancer Research (2001) 7:2228-2236, discloses that 17-AAG sensitizes cells in culture to the cytotoxic effects of Paclitaxel and doxorubicin. The Münster reference further discloses that the sensitization towards paclitaxel by 17-AAG is schedule-dependent in retinoblastoma protein-producing cells due to the action of these two drugs at different stages of the cell cycle: treatment of cells with a combination of paclitaxel and 17-AAG is reported to give synergistic apoptosis, while pretreatment of cells with 17-AAG followed by treatment with paclitaxel is reported to result in abrogation of apoptosis. Treatment of cells with paclitaxel followed by treatment with 17-AAG 4 hours later is reported to show a synergistic effect similar to coincident treatment.


Citri, et al., “Drug-induced ubiquitylation and degradation of ErbB receptor tyrosine kinases: implications for cancer chemotherapy,” EMBO Journal (2002) 21:2407-2417, discloses an additive effect upon co-administration of geldanamycin and an irreversible protein kinase inhibitor, CI-1033, on growth of ErbB2-expressing cancer cells in vitro. In contrast, an antagonistic effect of CI-1033 and anti-ErB2 antibody, Herceptin is disclosed.


Thus, while there has been a great deal of research interest in the benzoquinone ansamycins, particularly geldanamycin and 17-AAG, there remains a need for effective therapeutic regimens to treat cancer or other disease conditions characterized by undesired cellular hyperproliferation using such compounds, whether alone or in combination with other agents.


SUMMARY OF THE INVENTION

The present invention provides a method for treating cancer. The method involves the administration of an HSP90 inhibitor and an enzyme inhibitor, where the combined administration provides a synergistic effect.


In one aspect of the invention, a method of treating cancer is provided where a subject is treated with a dose of an HSP90 inhibitor in one step and a dose of an enzyme inhibitor in another step.


In another aspect of the invention, a method of treating cancer is provided where a subject is first treated with a dose of an HSP90 inhibitor and subsequently treated with a dose of an enzyme inhibitor.


In another aspect of the invention, a method of treating cancer is provided where a subject is first treated with a dose of an enzyme inhibitor and subsequently treated with a dose of an HSP90 inhibitor.


In another aspect of the invention, a method of treating cancer is provided where a subject is first treated with a dose of an enzyme inhibitor (e.g., SAHA or Iressa). After waiting for a period of time sufficient to allow development of a substantially efficacious response of the enzyme inhibitor, a formulation comprising a synergistic dose of a benzoquinone ansamycin together with a second sub-toxic dose of the enzyme inhibitor is administered.


In another aspect of the invention, a method of treating cancer is provided where a subject is treated first with a dose of a benzoquinone ansamycin, and second, a dose of an enzyme inhibitor. After waiting for a period of time sufficient to allow development of a substantially efficacious response of the enzyme inhibitor, a formulation comprising a synergistic dose of a benzoquinone ansamycin together with a second sub-toxic dose of the enzyme inhibitor drug is administered.


In another aspect of the invention, a method for treating cancer is provided where a subject is treated with a dose of an HSP90 inhibitor in one step and a dose of an enzyme inhibitor in another step, and where a side effect profile for the combined, administered drugs is substantially better than for the enzyme inhibitor alone.


In another aspect of the invention, a method for treating breast or colorectal cancer is provided where a subject is treated with a dose of an HSP90 inhibitor in one step and a dose of an enzyme inhibitor in another step. The HSP90 inhibitor for this aspect is typically 17-AAG, while the enzyme inhibitor is usually SAHA or Iressa. For the treatment of colorectal cancer or breast cancer, the enzyme inhibitor is typically administered before the 17-AAG.


Definitions

“Enzyme inhibitor” refers to a drug that stops, prevents or reduces the activity of an enzyme or a prodrug thereof. Enzyme inhibitors include, without limitation, histone deacetylation inhibitors (e.g., SAHA) and tyrosine kinase inhibitors (e.g., Iressa).


“HSP90 inhibitor” refers to a compound that inhibits the activity of heat shock protein 90, which is a cellular protein responsible for chaperoning multiple client proteins necessary for cell signaling, proliferation and survival. One class of HSP90 inhibitors is the benzoquinone ansamycins. Examples of such compounds include, without limitation, geldanamycin and geldanamycin derivatives (e.g., 17-alkylamino-17-desmethoxy-geldanamycin (“17-AAG”) and 17-(2-dimethylaminoethyl)amino-17-desmethoxy-geldanamycin (“17-DMAG”). See Sasaki et al., U.S. Pat. No. 4,261,989 (1981) for synthesis of 17-AAG and Snader et al., US 2004/0053909 A1 (2004) for synthesis of 17-DMAG. In addition to 17-AAG and 17-DMAG, other preferred geldanamycin derivatives are 11-O-methyl-17-(2-(1-azetidinyl)ethyl)amino-17-demethoxygeldanamycin (A), 11-O-methyl-17-(2-dimethylaminoethyl)amino-17-demethoxygeldanamycin (B), and 11-O-methyl-17-(2-(1-pyrrolidinyl)ethyl)amino-17-demethoxygeldanamycin (C), whose synthesis is described in the co-pending commonly U.S. patent application of Tian et al., Ser. No. 10/825,788, filed Apr. 16, 2004, and in Tian et al., PCT application no. PCT/US04/11638, filed Apr. 16, 2004; the disclosures of which are incorporated herein by reference. Additional preferred geldanamycin derivatives are described in Santi et al., US 2003/0114450 A1 (2003), also incorporated by reference.
embedded image


“MTD” refers to maximum tolerated dose. The MTD for a compound is determined using methods and materials known in the medical and pharmacological arts, for example through dose-escalation experiments. One or more patients is first treated with a low dose of the compound, typically about 10% of the dose anticipated to be therapeutic based on results of in vitro cell culture experiments. The patients are observed for a period of time to determine the occurrence of toxicity. Toxicity is typically evidenced as the observation of one or more of the following symptoms: vomiting, diarrhea, peripheral neuropathy, ataxia, neutropenia, or elevation of liver enzymes. If no toxicity is observed, the dose is increased about 2-fold, and the patients are again observed for evidence of toxicity. This cycle is repeated until a dose producing evidence of toxicity is eached. The dose immediately preceding the onset of unacceptable toxicity is taken as the MTD.


“Side effects” refer to a number of toxicities typically seen upon treatment of a subject with an antineoplastic drug. Such toxicities include, without limitation, anemia, anorexia, bilirubin effects, dehydration, dermatology effects, diarrhea, dizziness, dyspnea, edema, fatigue, headache, hematemesis, hypokalemia, hypoxia, musculoskeletal effects, myalgia, nausea, neuro-sensory effects, pain, rash, serum glutamic oxaloacetic transaminase effects, serum glutamic pyruvic transaminase effects, stomatitis, sweating, taste effects, thrombocytopenia, voice change, and vomiting.


“Side effect grading” refers to National Cancer Institute common toxicity criteria (NCI CTC, Version 2). Grading runs from 1 to 4, with a grade of 4 representing the most serious toxicities.


Combination Therapy

The present invention provides a method for treating cancer. The method involves the administration of an HSP90 inhibitor and an enzyme inhibitor, where the combined administration provides a synergistic effect.


Suitable HSP90 inhibitors used in the present invention include benzoquinone ansamycins. Typically, the benzoquinone ansamycin is geldanamycin or a geldanamycin derivative. Preferably, the benzoquinone ansamycin is a geldanamycin derivative selected from a group consisting of 17-alkylamino-17-desmethoxy-geldanamycin (“17-AAG”) and 17-(2-dimethylaminoethyl)amino-17-desmethoxy-geldanamycin (“17-DMAG”).


Enzyme inhibitors employed in the present method include, without limitation, histone deacetylation inhibitors (e.g., SAHA) and tyrosine kinase inhibitors (e.g., Iressa).


The dose of enzyme inhibitor used as a partner in combination therapy with an HSP90 inhibitor (e.g., benzoquinone ansamycin) is determined based on the maximum tolerated dose observed when the enzyme inhibitor is used as the sole therapeutic agent. In one embodiment of the invention, the dose of enzyme inhibitor when used in combination therapy with a benzoquinone ansamycin is the MTD. In other embodiments of the invention, the dose of enzyme inhibitor when used in combination therapy with a benzoquinone ansamycin is between about 1% of the MTD and the MTD, between about 5% of the MTD and the MTD, between about 5% of the MTD and 75% of the MTD, or between about 25% of the MTD and 75% of the MTD.


Use of the benzoquinone ansamycin allows for use of a lower therapeutic dose of an enzyme inhibitor, thus significantly widening the therapeutic window for treatment. In one embodiment, the therapeutic dose of enzyme inhibitor is lowered by at least about 10%. In other embodiments the therapeutic dose is lowered from about 10 % to 20%, from about 20% to 50%, from about 50% to 200%, or from about 100% to 1,000%.


For the treatment of a variety of carcinomas, the typical oral dose of various enzyme inhibitors is as follows: SAHA—up to 500 mg/day; Iressa—250 mg/day.


The synergistic dose of the benzoquinone ansamycin used in combination therapy is determined based on the maximum tolerated dose observed when the benzoquinone ansamycin is used as the sole therapeutic agent. Clinical trials have determined an MTD for 17-AAG of about 40 mg/m2 utilizing a daily ×5 schedule, and MTD of about 220 mg/m2 utilizing a twice-weekly regimen, and an MTD of about 308 mg/m2 utilizing a once-weekly regimen. In one embodiment of the invention, the dose of the benzoquinone ansamycin when used in combination therapy is the MTD. In other embodiments of the invention, the does of the benzoquinone ansamycin when used in combination therapy is between about 1% of the MTD and the MTD, between about 5% of the MTD and the MTD, between about 5% of the MTD and 75% of the MTD, or between about 25% of the MTD and 75% of the MTD.


Where the benzoquinone ansamycin is 17-AAG, and the administration of compound is weekly, its therapeutic dose is typically between 50 mg/m2 and 450 mg/m2. Preferably, the dose is between 150 mg/m2 and 350 mg/m2, and about 308 mg/m2 is especially preferred. Where the administration of compound is biweekly (i.e., twice per week), the therapeutic dose of 17-AAG is typically between 50 mg/m2 and 250 mg/m2. Preferably, the dose is between 150 mg/m2 and 250 mg/m2, and about 220 mg/m2 is especially preferred.


Where the present method involves the administration of 17-AAG and SAHA, a dosage regimen involving one or more administration of the combination per week is typical. Oftentimes, the combination is administered 2, 3, 4, 5, 6, or 7 times per week. Tables 1 and 2 below show a number of SAHA/17-AAG dosage combinations (i.e., dosage combinations 0001 to 0080).

TABLE 1SAHA/17-AAG dosage combinations.30-100100-150150-200mg/m2mg/m2mg/m2200-250 mg/m217-AAG17-AAG17-AAG17-AAG  0-50 mg/day0001000200030004SAHA 50-100 mg/day0005000600070008SAHA100-150 mg/day0009001000110012SAHA150-200 mg/day0013001400150016SAHA200-250 mg/day0017001800190020SAHA250-300 mg/day0021002200230024SAHA300-350 mg/day0025002600270028SAHA350-400 mg/day0029003000310032SAHA400-450 mg/day0033003400350036SAHA450-500 mg/day0037003800390040SAHA









TABLE 2










SAHA/17-AAG dosage combinations continued.












250-300
300-350
350-400




mg/m2
mg/m2
mg/m2
400-450 mg/m2



17-AAG
17-AAG
17-AAG
17-AAG














  0-50 mg/day
0041
0042
0043
0044


SAHA


‘150-100 mg/day
0045
0046
0047
0048


SAHA


100-150 mg/day
0049
0050
0051
0052


SAHA


150-200 mg/day
0053
0054
0055
0056


SAHA


200-250 mg/day
0057
0058
0059
0060


SAHA


250-300 mg/day
0061
0062
0063
0064


SAHA


300-350 mg/day
0065
0066
0067
0068


SAHA


350-400 mg/day
0069
0070
0071
0072


SAHA


400-450 mg/day
0073
0074
0075
0076


SAHA


450-500 mg/day
0077
0078
0079
0080


SAHA









Where the present method involves the administration of 17-AAG and Iressa, a dosage regimen involving one or more administration of the combination per week is typical. Oftentimes, the combination is administered 2, 3, 4, 5, 6, or 7 times per week. Tables 3 and 4 below show a number of Iressa/17-AAG dosage combinations (i.e., dosage combinations 0081 to 0160).

TABLE 3Iressa/17-AAG dosage combinations.30-100100-150150-200mg/m2mg/m2mg/m2200-250 mg/m217-AAG17-AAG17-AAG17-AAG  0-25 mg/day0081008200830084Iressa 25-50 mg/day0085008600870088Iressa 50-75 mg/day0089009000910092Iressa 75-100 mg/day0093009400950096Iressa100-125 mg/day0097009800990100Iressa125-150 mg/day0101010201030104Iressa150-175 mg/day0105010601070108Iressa175-200 mg/day0109011001110112Iressa200-225 mg/day0113011401150116Iressa225-250 mg/day0117011801190120Iressa









TABLE 4










Iressa/17-AAG dosage combinations continued.












250-300
300-350
350-400




mg/m2
mg/m2
mg/m2
400-450 mg/m2



17-AAG
17-AAG
17-AAG
17-AAG














  0-25 mg/day
0121
0122
0123
0124


Iressa


 25-50 mg/day
0125
0126
0127
0128


Iressa


 50-75 mg/day
0129
0130
0131
0132


Iressa


 75-100 mg/day
0133
0134
0135
0136


Iressa


100-125 mg/day
0137
0138
0139
0140


Iressa


125-150 mg/day
0141
0142
0143
0144


Iressa


150-175 mg/day
0145
0146
0147
0148


Iressa


175-200 mg/day
0149
0150
0151
0152


Iressa


200-225 mg/day
0153
0154
0155
0156


Iressa


225-250 mg/day
0157
0158
0159
0160


Iressa









The method of the present invention may be carried out in at least two basic ways. A subject may first be treated with a dose on an HSP90 inhibitor and subsequently be treated with a dose of an enzyme inhibitor. Alternatively, the subject may first be treated with a dose of an enzyme inhibitor and subsequently be treated with a dose of an HSP90 inhibitor. The appropriate dosing regimen depends on the particular enzyme inhibitor employed.


In another aspect of the invention, a subject is first treated with a dose of an enzyme inhibitor (e.g., SAHA or Iressa). After waiting for a period of time sufficient to allow development of a substantially efficacious response of the enzyme inhibitor, a formulation comprising a synergistic dose of a benzoquinone ansamycin together with a second sub-toxic dose of the enzyme inhibitor is administered. In general, the appropriate period of time sufficient to allow development of a substantially efficacious response to the enzyme inhibitor will depend upon the pharmacokinetics of the enzyme inhibitor, and will have been determined during clinical trials of therapy using the enzyme inhibitor alone. In one embodiment of the invention, the period of time sufficient to allow development of a substantially efficacious response to the enzyme inhibitor is between about 1 hour and 96 hours. In another aspect of the invention, the period of time sufficient to allow development of a substantially efficacious response to the enzyme inhibitor is between about 2 hours and 48 hours. In another embodiment of the invention, the period of time sufficient to allow development of a substantially efficacious response to the enzyme inhibitor is between about 4 hours and 24 hours.


In another aspect of the invention, a subject is treated first with one of the above-described benzoquinone ansamycins, and second, a dose of an enzyme inhibitor, such as, but not limited to, SAHA and Iressa. After waiting for a period of time sufficient to allow development of a substantially efficacious response of the enzyme inhibitor, a formulation comprising a synergistic dose of a benzoquinone ansamycin together with a second sub-toxic dose of the enzyme inhibitor is administered. In general, the appropriate period of time sufficient to allow development of a substantially efficacious response to the enzyme inhibitor will depend upon the pharmacokinetics of the enzyme inhibitor, and will have been determined during clinical trials of therapy using the enzyme inhibitor alone. In one embodiment of the invention, the period of time sufficient to allow development of a substantially efficacious response to the enzyme inhibitor is between about 1 hour and 96 hours. In another aspect of the invention, the period of time sufficient to allow development of a substantially efficacious response to the enzyme inhibitor is between about 2 hours and 48 hours. In another embodiment of the invention, the period of time sufficient to allow development of a substantially efficacious response to the enzyme inhibitor is between about 4 hours and 24 hours.


As noted above, the combination of an HSP90 inhibitor and an enzyme inhibitor allows for the use of a lower therapeutic dose of the enzyme inhibitor for the treatment of cancer. That a lower dose of enzyme inhibitor is used oftentimes lessens the side effects observed in a subject. The lessened side effects can be measured both in terms of incidence and severity. Severity measures are provided through a grading process delineated by the National Cancer Institute (common toxicity criteria NCI CTC, Version 2). For instance, the incidence of side effects are typically reduced 10%. Oftentimes, the incidence is reduced 20%, 30%, 40% or 50%. Furthermore, the incidence of grade 3 or 4 toxicities for more common side effects associated with enzyme inhibitor administration (e.g., anemia, anorexia, diarrhea, fatigue, nausea and vomiting) is oftentimes reduced 10%, 20%, 30%, 40% or 50%.


Formulations used in the present invention may be in any suitable form, such as a solid, semisolid, or liquid form. See Pharmaceutical Dosage Forms and Drug Delivery Systems, 5th edition, Lippicott Williams & Wilkins (1991), incorporated herein by reference. In general the pharmaceutical preparation will contain one or more of the compounds of the present invention as an active ingredient in admixture with an organic or inorganic carrier or excipient suitable for external, enteral, or parenteral application. The active ingredient may be compounded, for example, with the usual non-toxic, pharmaceutically acceptable carriers for tablets, pellets, capsules, suppositories, pessaries, solutions, emulsions, suspensions, and any other form suitable for use. The carriers that can be used include water, glucose, lactose, gum acacia, gelatin, mannitol, starch paste, magnesium trisilicate, talc, corn starch, keratin, colloidal silica, potato starch, urea, and other carriers suitable for use in manufacturing preparations in solid, semi-solid, or liquefied form. In addition, auxiliary stabilizing, thickening, and coloring agents and perfumes may be used. Where applicable, the compounds useful in the methods of the invention may be formulated as microcapsules and nanoparticles. General protocols are described, for example, by Microcapsules and Nanoparticles in Medicine and Pharmacy by Max Donbrow, ed., CRC Press (1992) and by U.S. Pat. Nos. 5,510,118, 5,534,270 and 5,662,883 which are all incorporated herein by reference. By increasing the ratio of surface area to volume, these formulations allow for the oral delivery of compounds that would not otherwise be amenable to oral delivery. The compounds useful in the methods of the invention may also be formulated using other methods that have been previously used for low solubility drugs. For example, the compounds may form emulsions with vitamin E or a PEGylated derivative thereof as described by PCT publications WO 98/30205 and WO 00/71163, each of which is incorporated herein by reference. Typically, the compound useful in the methods of the invention is dissolved in an aqueous solution containing ethanol (preferably less than 1% w/v). Vitamin E or a PEGylated-vitamin E is added. The ethanol is then removed to form a pre-emulsion that can be formulated for intravenous or oral routes of administration. Another method involves encapsulating the compounds useful in the methods of the invention in liposomes. Methods for forming liposomes as drug delivery vehicles are well known in the art. Suitable protocols include those described by U.S. Pat. Nos. 5,683,715, 5,415,869, and 5,424,073 which are incorporated herein by reference relating to another relatively low solubility cancer drug paclitaxel and by PCT Publicaton WO 01/10412 which is incorporated herein by reference relating to epothilone B. Of the various lipids that may be used, particularly preferred lipids for making encapsulated liposomes include phosphatidylcholine and polyethyleneglycol-derivatized distearyl phosphatidyl-ethanoloamine.


Yet another method involves formulating the compounds useful in the methods of the invention using polymers such as biopolymers or biocompatible (synthetic or naturally occurring) polymers. Biocompatible polymers can be categorized as biodegradable and non-biodegradable. Biodegradable polymers degrade in vivo as a function of chemical composition, method of manufacture, and implant structure. Illustrative examples of synthetic polymers include polyanhydrides, polyhydroxyacids such as polylactic acid, polyglycolic acids and copolymers thereof, polysters, polyamides, polyorthoesters and some polyphosphazenes. Illustrative examples of naturally occurring polymers include proteins and polysaccharides such as collagen, hyaluronic acid, albumin, and gelatin.


Another method involves conjugating the compounds useful in the methods of the invention to a polymer that enhances aqueous solubility. Examples of suitable polymers include polyethylene glycol, poly-(d-glutamic acid), poly-(1-glutamic acid), poly-(1-glutamic acid), poly-(d-aspartic acid), poly-(1-aspartic acid) and copolymers thereof. Polyglutamic acids having molecular weights between about 5,000 to about 100,000 are preferred, with molecular weights between about 20,000 and 80,000 being more preferred wand with molecular weights between about 30,000 and 60,000 being most preferred. The polymer is conjugated via an ester linkage to one or more hydroxyls of an inventive geldanamycin using a protocol as essentially described by U.S. Pat. No. 5,977,163 which is incorporated herein by reference.


In another method, the compounds useful in the methods of the invention are conjugated to a monoclonal antibody. This method allows the targeting of the inventive compounds to specific targets. General protocols for the design and use of conjugated antibodies are described in Monoclonal Antibody-Based Therapy of Cancer by Michael L. Grossbard, ED. (1998), which is incorporated herein by reference.


The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the subject treated and the particular mode of administration. For example, a formulation for intravenous use comprises an amount of the inventive compound ranging from about 1 mg/mL to about 25 mg/mL, preferably from about 5 mg/mL, and more preferably about 10 mg/mL. Intravenous formulations are typically diluted between about 2 fold and about 30 fold with normal saline or 5% dextrose solution prior to use.


Preferably, 17-AAG is formulated as a pharmaceutical solution formulation comprising 17-AAG in an concentration of up to 15 mg/mL dissolved in a vehicle comprising (i) a first component that is ethanol, in an amount of between about 40 and about 60 volume %; (ii) a second component that is a polyethoxylated castor oil, in an amount of between about 15 to about 50 volume %; and (iii) a third component that is selected from the group consisting of propylene glycol, PEG 300, PEG 400, glycerol, and combinations thereof, in an amount of between about 0 and about 35 volume %. The aforesaid percentages are volume/volume percentages based on the combined volumes of the first, second, and third components. The lower limit of about 0 volume % for the third component means that it is an optional component; that is, it may be absent. The pharmaceutical solution formulation is then diluted into water to prepare a diluted formulation containing up to 3 mg/mL 17-AAG, for intravenous formulation.


Preferably, the second component is Cremophor EL and the third component is propylene glycol. In an especially preferred formulation, the percentages of the first, second, and third components are 50%, 20-30%, and 20-30%, respectively.


Other formulations designed for 17-AAG are described in Tabibi et al., U.S. Pat. No. 6,682,758 B1 (2004) and Ulm et al., WO 03/086381 A1 (2003); the disclosures of which are incorporated herein by reference.


The method of the present invention is used for the treatment of cancer. In one embodiment, the methods of the present invention are used to treat cancers of the head and neck, which include, but are not limited to, tumors of the nasal cavity, paranasal sinuses, nasopharynx, oral cavity, oropharynx, larynx, hypopharynx, salivary glands, and paragangliomas. In another embodiment, the compounds of the present invention are used to treat cancers of the liver and biliary tree, particularly hepatocellular carcinoma. In another embodiment, the compounds of the present invention are used to treat intestinal cancers, particularly colorectal cancer. In another embodiment, the compounds of the present invention are used to treat ovarian cancer. In another embodiment, the compounds of the present invention are used to treat small cell and non-small cell lung cancer. In another embodiment, the compounds of the present invention are used to treat breast cancer. In another embodiment, the compounds of the present invention are used to treat sarcomas, including fibrosarcoma, malignant fibrous histiocytoma, embryonal rhabdomysocarcoman, leiomysosarcoma, neuro-fibrosarcoma, osteosarcoma, synovial sarcoma, liposarcoma, and alveolar soft part sarcoma. In another embodiment, the compounds of the present invention are used to treat neoplasms of the central nervous systems, particularly brain cancer. In another embodiment, the compounds of the present invention are used to treat lymphomas which include Hodgkin's lymphoma, lymphoplasmacytoid lymphoma, follicular lymphoma, mucosa-associated lymphoid tissue lymphoma, mantle cell lymphoma, B-lineage large cell lymphoma, Burkitt's lymphoma, and T-cell anaplastic large cell lymphoma.







EXAMPLES

The following Examples are provided to illustrate certain aspects of the present invention and to aid those of skill in the art in practicing the invention.


Materials and Methods

Cell Line and Reagents


Human colon adenocarcinoma cell line, DLD-1, and human breast adenocarcinoma cell line, SKBr-3, were obtained from American Type Culture Collection (manassas, Va.). DLD- 1 cells were maintained in RPMI 1640 medium supplemented with 10% fetal bovine serum, and SKBr-3 cells were cultured in McCoy's Sa medium supplemented with 10% fetal bovine serum. 17-DMAG and 17-AAG were obtained using published procedures. Other cytotoxic agents were purchased commercially from suppliers such as Sigma Chemical Co. (St. Louis, Mo.) and Sequoia Research Products (Oxford, UK).


Cell Viability Assay and Combination Effect Analysis


Cells were seeded in duplicate in 96-well microtiter plates at a density of 5,000 cells per well and allowed to attach overnight. Cells were treated with 17-AAG or 17-DMAG and the corresponding enzyme inhibitor at varying concentrations, ranging from 0.5 picomolar (“pM”) to 50 micromolar (“μM”), for 3 days. Cell viability was determined using the MTS assay (Promega). For the drug combination assay, cells were seeded in duplicate in 96-well plates (5,000 cells/well). After an overnight incubation, cells were treated with drug alone or a combination and the IC50 value (the concentration of drug required to inhibit cell growth by 50%) was determined. Based on the IC50 values of each individual drug, combined drug treatment was designed at constant ratios of two drugs, i.e., equivalent to the ratio of their IC50. Two treatment schedules were used: In one schedule, the cells were exposed to 24 hours of 17-AAG or 17-DMAG. The drug was then added to the cells and incubated for 48 hours. In another schedule, cells were exposed to the drug alone for 24 hours followed by addition of 17-AAG or 17-DMAG for 48 hours. Cell viability was determined by the MTS assay.


Synergism, additivity or antagonism was determined by median effect analysis using the combination index (CI) calculated using Calcusyn (Biosoft, Cambridge, UK). The combination index is defined as follows:

CI=[D]1/[Dx]1+[D]2/[Dx]2

The quantities [D]1 and [D]2 represent the concentrations of the first and second drug, respectively, that in combination provide a response of x % in the assay. The quantities [Dx] and [Dx]2 represent the concentrations of the first and second drug, respectively, that when used alone provide a response of x % in the assay. Values of CI<1, CI=1, and CI>1 indicated drug-drug synergism, additivity, and antagonism respectively (Chou and Talalay 1984). The “enhancing” effect of two drugs can also be determined.


Results

17-AAG Combination in DLD-1 Cells


The following table provides CI values for combinations of 17-AAG and the enzyme inhibitor Iressa in a DLD-1 cell assay. “Pre-administration” refers to the administration of 17-AAG to the cells before the administration of enzyme inhibitor; “post-administration” refers to the administration of 17-AAG to the cells after the administration of enzyme inhibitor.

TABLE 5CI values for combinations in DLD-1 cells(human colorectal cancer cells).EnzymeInhibitor17-AAG Pre-Administration17-AAG Post-AdministrationIressa0.76 ± 0.370.41 ± 0.01

17-AAG Combination in SKSBr-3 Cells


The following table provides CI values for combinations of 17-AAG and the enzyme inhibitors SAHA, Trichostatin A, and Iressa in an SKBr-3 cell assay.

TABLE 6CI values for combinations in SKBr cells(human breast cancer cells).17-AAGEnzyme Inhibitor17-AAG Pre-AdministrationPost-AdministrationSAHA1.05 ± 0.030.15 ± 0.12Trichostatin A1.16 ± 0.02 0.91 ± 0.007Iressa0.77 ± 0.290.67 ± 0.02


Additional Observations


Additional analysis indicated that both 17-AAG and 17-DMAG reduced the expression of ErbB2 protein in SKBr3 and glioma cells. This observation, taken in combination with the results reported above, indicates that combinations of 17-AAG or 17-DMAG with any of the enzyme inhibitors above that are known to be useful to treat diseases characterized by elevated ErbB2 protein expression (i.e., levels of expressions of ErbB2 protein greater than those found in healthy cells).

Claims
  • 1. A method for treating breast cancer in a patient, wherein the method comprises administering an HSP90 inhibitor and an enzyme inhibitor to the patient.
  • 2. The method of claim 1, wherein the HSP90 inhibitor is administered to the patient before the enzyme inhibitor.
  • 3. The method of claim 1, wherein the HSP90 inhibitor is administered to the patient after the enzyme inhibitor.
  • 4. The method of claim 2, wherein the HSP90 inhibitor is geldanamycin or a geldanamycin derivative.
  • 5. The method of claim 3, wherein the HSP90 inhibitor is geldanamycin or a geldanamycin derivative.
  • 6. The method of claim 4, wherein the HSP90 inhibitor is a geldanamycin derivative, and wherein the derivative is 17-AAG.
  • 7. The method of claim 5, wherein the HSP 90 inhibitor is a geldanamycin derivative, and wherein the derivative is 17-AAG.
  • 8. The method of claim 6, wherein the enzyme inhibitor is SAHA or Iressa.
  • 9. A method for treating colorectal cancer in a patient, wherein the method comprises administering an HSP90 inhibitor and an enzyme inhibitor to the patient.
  • 10. The method of claim 9, wherein the HSP90 inhibitor is administered to the patient after the enzyme inhibitor.
  • 11. The method of claim 10, wherein the HSP90 inhibitor is administered to the patient before the enzyme inhibitor.
  • 12. The method of claim 10, wherein the HSP90 inhibitor is geldanamycin or a geldanamycin derivative.
  • 13. The method of claim 11, wherein the HSP90 inhibitor is geldanamycin or a geldanamycin derivative.
  • 14. The method of claim 12, wherein the HSP90 inhibitor is a geldanamycin derivative, and wherein the derivative is 17-AAG.
  • 15. The method of claim 13, wherein the HSP90 inhibitor is a geldanamycin derivative, and wherein the derivative is 17-AAG.
  • 16. The method of claim 14, wherein the enzyme inhibitor is Iressa.
  • 17. The method of claim 1, wherein the enzyme inhibitor is a histone deacetylase inhibitor.
  • 18. The method of claim 1, wherein the enzyme inhibitor is a tyrosine kinase inhibitor.
  • 19. The method of claim 9, wherein the enzyme inhibitor is a histone deacetylase inhibitor.
  • 20. The method of claim 9, wherein the enzyme inhibitor is a tyrosine kinase inhibitor.
  • 21. The method of claim 1, wherein the HSP90 inhibitor is 17-AAG, and wherein the administration of 17-AAG and the enzyme inhibitor is performed once per week.
  • 22. The method of claim 1, wherein the HSP90 inhibitor is 17-AAG, and wherein the administration of 17-AAG and the enzyme inhibitor is performed twice per week.
  • 23. The method of claim 9, wherein the HSP90 inhibitor is 17-AAG, and wherein the administration of 17-AAG and the enzyme inhibitor is performed once per week.
  • 24. The method of claim 9, wherein the HSP90 inhibitor is 17-AAG, and wherein the administration of 17-AAG and the enzyme inhibitor is performed twice per week.
  • 25. The method of claim 21, wherein the therapeutic dose of 17-AAG is between 50 mg/m2 and 450 mg/m2.
  • 26. The method of claim 22, wherein the therapeutic dose of 17-AAG is between 50 mg/m2 and 250 mg/m2.
  • 27. The method of claim 23, wherein the therapeutic dose of 17-AAG is between 50 mg/m2 and 450 mg/m2.
  • 28. The method of claim 24, wherein the therapeutic dose of 17-AAG is between 50 mg/m2 and 250 mg/m2.
CROSS REFERENCE TO RELATED U.S. PATENT APPLICATIONS

The present application claims the benefit of Provisional Patent Application No. 60/474,906, which was filed May 30, 2003, under 35 U.S.C. § 119(e). The provisional application is hereby incorporated-by-reference into this application for all purposes.

Provisional Applications (1)
Number Date Country
60474906 May 2003 US