WO2009091859A1 - Methods for treating cancer in patients having breast cancer resistance protein overexpression - Google Patents

Methods for treating cancer in patients having breast cancer resistance protein overexpression Download PDF

Info

Publication number
WO2009091859A1
WO2009091859A1 PCT/US2009/031052 US2009031052W WO2009091859A1 WO 2009091859 A1 WO2009091859 A1 WO 2009091859A1 US 2009031052 W US2009031052 W US 2009031052W WO 2009091859 A1 WO2009091859 A1 WO 2009091859A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
cancer
biomarker
patient
overexpression
Prior art date
Application number
PCT/US2009/031052
Other languages
French (fr)
Inventor
Francis Y. Lee
Jinping Gan
Hong Shen
Original Assignee
Bristol-Myers Squibb Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Priority to US12/812,777 priority Critical patent/US20110112155A1/en
Priority to EP09702397A priority patent/EP2238454A1/en
Publication of WO2009091859A1 publication Critical patent/WO2009091859A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention relates generally to the field of pharmacogenomics, and more specifically to methods and procedures to determine drug sensitivity in patients to allow the identification of individualized genetic profiles which will aid in treating diseases and disorders.
  • BCRP 72-kDa breast cancer resistance protein
  • ABSC human ATP binding cassette
  • BCRP is a half-transporter consisting of only 1 nucleotide binding domain followed by 1 membrane-spanning domain.
  • Current experimental evidence suggests that BCRP may function as a homodimer or homotetramer.
  • Overexpression of BCRP is associated with high levels of resistance to a variety of anticancer agents, including anthracyclines, mitoxantrone, and the camptothecins, by enhancing drug efflux.
  • BCRP expression has been detected in a large number of hematological malignancies and solid tumors, indicating that this transporter may play an important role in clinical drug resistance of cancers.
  • BCRP actively transports structurally diverse organic molecules, conjugated or unconjugated, such as estrone-3 -sulfate, 17 ⁇ -estradiol 17-( ⁇ -D-gIucuronide), and methotrexate.
  • BCRP is highly expressed in the placental syncytiotrophoblasts, in the apical membrane of the epithelium in the small intestine, in the liver canalicular membrane, and at the luminal surface of the endothelial cells of human brain microvessels.
  • BCRP BCRP substrates.
  • New prognostic and predictive markers which would facilitate an individualization of therapy for each patient, are needed to accurately predict patient response to treatments, such as small molecule or biological molecule drugs, in the clinic.
  • the problem may be solved by the identification of new parameters that could better predict the patient's sensitivity to treatment.
  • the classification of patient samples is a crucial aspect of cancer diagnosis and treatment.
  • the association of a patient's response to a treatment with molecular and genetic markers can open up new opportunities for treatment development in non-responding patients, or distinguish a treatment's indication among other treatment choices because of higher confidence in the efficacy. Further, the pre-selection of patients who are likely to respond well to a medicine, drug, or combination therapy may reduce the number of patients needed in a clinical study or accelerate the time needed to complete a clinical development program (Cockett, M. et al., Current Opinion in Biotechnology, 11 : 602-609 (2000)).
  • Microtubule-stabilizing agents such as ixabepilone (IXEMPRATM) and paclitaxel (TAXOL®), are commonly used for the treatment of many types of cancer, including breast and lung cancer.
  • the invention provides methods and procedures for determining patient sensitivity to one or more microtubule-stabilizing agents.
  • the invention relates to a method for treating cancer comprising identifying a mammal that overexpresses breast cancer resistance protein; and administering to said mammal a pharmaceutical composition comprising a therapeutically effective amount of ixabepilone, either alone or in combination with another agent.
  • the mammal is not administered an agent that is susceptible to breast cancer resistance protein overexpression resistance.
  • the cancer is breast and/or lung cancer.
  • the mammal further overexpresses at least one of BRCP (ABCG2), beta-tubulin UI (TUBB3), MDRl, MRPl, and a beta-tubulin mutant.
  • the mammal is a human.
  • the present invention provides a method of screening a biological sample, for example cells that do not respond, or that have stopped responding, or that have a diminished response, to one or more microtubule-stabilizing agents.
  • the present invention provides a method of screening cells from an individual suffering from cancer who is either being treated with one or more microtubule-stabilizing agents or is na ⁇ ve to said agents, and whose cells do not respond or have stopped responding or that have a diminished response to one or more microtubule-stabilizing agents, for overexpression of breast cancer resistance protein relative to a standard.
  • breast cancer resistance protein overexpression is present, administration of a therapeutically acceptable amount of ixabepilone, alone or in combination with one or more microtubule-stabilizing agents and/or other agent, such as a CTLA4 antagonist, is warranted to inhibit proliferation of said cells.
  • said cancer is breast and/or lung cancer.
  • the present invention provides a method of screening a biological sample, for example cells that do not respond, or that have stopped responding, or that have a diminished response, to one or more microtubule-stabilizing agents.
  • the present invention provides a method of screening cells from an individual suffering from cancer who is either being treated with one or more microtubule-stabilizing agents or is na ⁇ ve to said agents, and whose cells do not respond or have stopped responding or that have a diminished response to one or more microtubule-stabilizing agents, for overexpression of breast cancer resistance protein and beta-tubulin III relative to a standard.
  • the present invention provides a method of screening a biological sample, for example cells that do not respond, or that have stopped responding, or that have a diminished response, to one or more microtubule-stabilizing agents.
  • the present invention provides a method of screening cells from an individual suffering from cancer who is either being treated with one or more microtubule-stabilizing agents or is na ⁇ ve to said agents, and whose cells do not respond or have stopped responding or that have a diminished response to one or more microtubule-stabilizing agents, for overexpression of breast cancer resistance protein and MDRl relative to a standard. If breast cancer resistance protein and MDRl overexpression is present, administration of a therapeutically acceptable amount of ⁇ xabepilone, alone or in combination with one or more microt ⁇ bule-stabilizing agents and/or other agent, such as a CTLA4 antagonist, is warranted to inhibit proliferation of said cells. Wherein said cancer is breast and/or lung cancer.
  • the present invention provides a method of screening a biological sample, for example cells that do not respond, or that have stopped responding, or that have a diminished response, to one or more microtubule-stabilizing agents.
  • the present invention provides a method of screening cells from an individual suffering from cancer who is either being treated with one or more microtubule-stabilizing agents or is na ⁇ ve to said agents, and whose cells do not respond or have stopped responding or that have a diminished response to one or more microtubule-stabilizing agents, for overexpression of breast cancer resistance protein and MRPl relative to a standard.
  • breast cancer resistance protein and MRPl overexpression are present, administration of a therapeutically acceptable amount of ixabepilone, alone or in combination with one or more microtubule-stabilizing agents and/or other agent, such as a CTLA4 antagonist, is warranted to inhibit proliferation of said cells.
  • said cancer is breast and/or lung cancer.
  • the present invention provides a method of screening a biological sample, for example cells that do not respond, or that have stopped responding, or that have a diminished response, to one or more microtubule-stabilizing agents.
  • the present invention provides a method of screening cells from an individual suffering from cancer who is either being treated with one or more microtubule-stabilizing agents or is na ⁇ ve to said agents, and whose cells do not respond or have stopped responding or that have a diminished response to one or more microtubule-stabilizing agents, for overexpression of breast cancer resistance protein and beta-tubulin mutant relative to a standard.
  • breast cancer resistance protein and beta-tubulin mutant overexpression are present, administration of a therapeutically acceptable amount of ixabepilone, alone or in combination with one or more microtubule-stabilizing agents and/or other agent, such as a CTLA4 antagonist, is warranted to inhibit proliferation of said cells.
  • said cancer is breast and/or lung cancer.
  • the diagnostic methods of the invention can be, for example, an in vitro method wherein the step of measuring in the mammal the level of at least one biomarker comprises taking a biological sample from the mammal and then measuring the level of the biomarker(s) in the biological sample.
  • the biological sample can comprise, for example, at least one of serum, whole fresh blood, peripheral blood mononuclear cells, frozen whole blood, fresh plasma, frozen plasma, urine, saliva, skin, hair follicle, bone marrow, or tumor tissue.
  • the level of the at least one biomarker can be, for example, the level of protein and/or mRNA transcript of the biomarker(s).
  • the invention also provides an isolated TUBB3 biomarker, an isolated BRCP (ABCG2) biomarker, an isolated MDRl (ABCBl) biomarker, an isolated MRPl (ABCCl) biomarker, and tubulin mutation biomarkers.
  • the biomarkers of the invention include nucleotide and amino acid sequences of full-length TUBB3, BRCP (ABCG2), MDRl (ABCBl), MRPl (ABCCl), and beta-tubulin mutations, as well as fragments and variants thereof.
  • the invention also provides a biomarker set comprising two or more biomarkers of the invention.
  • the invention also provides kits for measuring overexpression of breast cancer resistance protein, and uses thereof.
  • the invention also provides antibodies, including polyclonal or monoclonal, directed breast cancer resistance protein, and uses thereof. [0022] The invention will be better understood upon a reading of the detailed description of the invention when considered in connection with the accompanying Figures.
  • Figure 1 shows a schematic of the structure and mechanism of ixabepilone.
  • Figure 2 illustrates the issue of drug resistance for taxanes as well as providing examples of several mechanisms of how such resistance may arise.
  • Figure 3 shows the ability of ixabepilone to overcome multiple mechanisms of taxane-resi stance.
  • Figure 4 provides an illustration of ABC drug resistance transporters.
  • FIG. S shows the ability of ixabepilone to effectively overcome the BCRP overexpression-induced resistance to microtubulin stabilizers.
  • BCRP overexpressing HEK293 cells are denoted as “BCRP/HEK 293 Cells”, while control HEK293 cells are denoted as “HEK 293 Cells”.
  • Figure 6 shows the IC50 of paclitaxei, mitotropine, and ixabepilone in BCRP overexpressing HEK293 cells. As shown, ixabepilone had a very low IC50.
  • Figure 7 shows the detection of BCRP overexpressing cells using Hoescht 33342 dye and flow cytometry, and the abolition of BCRP overexpressing cells subsequent to the administration of the BCRP inhibitor, Fumitremorgin C.
  • Figure 8 shows the increased ability of ixabepilone to overcome BCRP- overexpression dependent resistance to taxanes in a human lung carcinoma cell line, H441.
  • ixabepilone refers to a compound having the following structure (I):
  • Compound (I) can also be referred to as (1S,3S,7S,1OR,11S,12S,16R)-7,11 - d ihydroxy-8 , 8, 10,12,16-pentamethyl-3-[( 1 E)- 1 -m ethyl-2-(2-methyI-4- thiazolyl)ethenyl]- 17-oxa-4-azabicyclo[ 14.1.0]heptadecane-5 ,9-dione in accordance with IUPAC nomenclature.
  • compositions Of (IS 5 SSJS 5 IOR 1 1 18,128,16R)-7 S 11- dihydroxy-8,8,10, 12, 16- ⁇ entamethyl-3-[( IE)-I -methyl-2-(2-methyl-4- thiazolylJethenylj- ⁇ -oxa ⁇ -azabicyclotM.l.OJheptadecane-S ⁇ -dione include all pharmaceutically acceptable compositions comprising (IS,3S,7S,1OR,11S,12S,16R)- 7,1 l-dihydroxy-8,8, 10, 12, 16-pentamethyl-3-[(l E)-I -methyl-2-(2-methyl-4- thiazolyl)ethenyl]-17-oxa-4-azabicyclo[14.1.0]heptadecane-5,9-dione and one or more diluents, vehicles and/or excipients
  • a pharmaceutical composition comprising (lS > 3S,7S,10R,l l
  • IXEMPRATM comprises (lS,3S 5 7S,10R,l lS,12S ! 16R)-7,n-dihydroxy- 8,8,10,12,16-pentamethyl-3-[(lE)-l-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-17-oxa- 4-azabicyclo[14.1.0]heptadecane-5,9-dione as the active ingredient, also referred to as ixabepilone, for IV infusion including inactive ingredients in the form of a diluent consisting of a sterile, non-pyrogenic of 52.8% (w/v) purified polyoxyethylated castor oil and 39.8% (w/v) dehydrated alcohol, USP. [0024]
  • Non-limiting examples of other epoth ⁇ ones for use in the methods and compositions of the present invention are encompassed by formula II:
  • Q is selected from the group consisting of:
  • G is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, heterocyclo,
  • W is O or N R !5 ;
  • X is O or H, H;
  • Y is selected from the group consisting of O; H, ORie; ORn, OR] 7 ; NOR] «; H, NHORi 9 ; H, NR 20 R 2 S; H, H; and CHR 22 ; wherein OR !7 , OR i7 can be a cyclic ketal;
  • Zi and Z 2 are independently selected from the group consisting of CH 2 , O, NR 23 , S, and SO 2 , wherein only one of Zi and Z 2 can be a heteroatom;
  • Bi and B 2 are independently selected from the group consisting of OR 24 , OCOR 25 , and and when Bi is H and Y is OH, H, they can form a six-membered ring ketal or acetal;
  • D is selected from the group consisting of NR 28 R 29 , NR 30 COR 31 and saturated heterocycle;
  • Ru R 2 , R 3 , R 4 , R 5 , R 6 , R?, Ri3, Rw, Ris, Rw, R2o» R21, R 22 , R 26 and R 27 are independently selected from the group consisting of H, alkyl, substituted alkyl, and aryl, and when Rj and R 2 are alkyl can be joined to form a cycloalkyl, and when R3 and R 4 are alkyl can be joined to form a cycloalkyl;
  • R 9 , Rio, R I O, Rj7, R 24> R 2 5 and R3 1 are independently selected from the group consisting of H, alkyl, and substituted alkyl;
  • R 11 , R i 2 , R 2 8, R3o, R32, and R 33 are independently selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl and heterocyclo;
  • Formula III provides another example of an epothilone suitable for use in the methods and compositions of the present invention:
  • R is selected from the group of H, alkyl, and substituted alkyl; R 1 is selected from the group consisting of:
  • G 1 is selected from the group of H, halogen, CN, alkyl and substituted alkyl
  • G 2 is selected from the group of H, alkyl, and substituted alkyl
  • G 3 is selected from the group of O, S, and NZ 1 ;
  • G 5 is selected from the group of halogen, N 3 , NCS, SH, CN, NC, N(Z 1 J 3 + and heteroaryl;
  • G 6 is selected from the group of H, alkyl, substituted alkyl, CF 3 , OZ 5 , SZ 5 , and NZ 5 Z 6 ;
  • G 7 is CZ 7 or N
  • G 8 is selected from the group of H, halogen, alkyl, substituted alkyl, OZ i0 , SZ 10 , NZ 10 Z 11 ;
  • G 10 is N or CZ 12 ;
  • G 11 is selected from the group of H 2 N, substituted H 2 N, alkyl, substituted alkyl, aryl, and substituted aryl;
  • Z 1 , Z 6 , Z 9 , and Z n are independently selected from the group H, alkyl, substituted alkyl, acyl, and substituted acyl;
  • Z 2 is selected from the group of H, alkyl, substituted alkyl, aryl, substituted aryl, and heterocycle;
  • Z 3 , Z 5 , Z 8 , and Z 10 are independently selected from the group H, alkyl, substituted alkyl, acyl, substituted acyl, aryl, and substituted aryl;
  • Z 4 is selected from the group of alkyl, substituted alkyl, aryl, substituted aryl, and heterocycle;
  • Z 7 is selected from the group of H, halogen, alkyl, substituted alkyl, aryl, substituted aryl, OZ 8 , SZ 8 , and NZ 8 Z 9 ;
  • Z 12 is selected from the group of H, halogen, alkyl, substituted alkyl, aryl, and substituted aryl; with the proviso that when R 1 is
  • G 1 S G 2 , G 3 and G 4 cannot simultaneously have the following meanings:
  • a preferred compound of Formula III of the invention is Formula Ilia:
  • P-Q is a C,C double bond or an epoxide
  • R is a H atom or a methyl group
  • G 1 is an H atom, an alkyl group, a substituted alkyl group or a halogen atom
  • G 2 is an H atom, an alkyl group or a substituted alkyl group
  • G 3 is an O atom, an S atom or an NZ 1 group with Z 1 being an H atom, an alkyl group, a substituted alkyl group, an acyl group, or a substituted acyl group;
  • a particularly preferred compound of Formula III is [IS- [lR*,3R*(E),7R*,10S*,l lR* > 12R*,16S*]]-3-[2-[2-(aminomethyl)-4-thiazolyl]-l- methylethenyl]-7 s 11 -dihydroxy-8,8, 10, 12, 16-pentamethyl-4, 17- dioxabicyclo[14.1.0]heptadecane-5,9-dione (Compound 4) and pharmaceutically acceptable salts thereof.
  • the phrase "microtubule modulating agent” is meant to refer to agents that either stabilize microtubule or destabilize microtubulin synthesis and/or polymerization.
  • Microtubulin modulatory agents either agonize or inhibit a cells ability to maintain proper microtubulin assemblies.
  • paclitaxel marketed as TAXOL®
  • TAXOL® causes mitotic abnormalities and arrest, and promotes microtubule assembly into calcium-stable aggregated structures resulting in inhibition of cell replication.
  • Epothilones mimic the biological effects of TAXOL®, (Bollag et al, Cancer Res., 55:2325-2333 (1995), and in competition studies act as competitive inhibitors of TAXOL® binding to microtubules.
  • epothilones enjoy a significant advantage over TAXOL® in that epothilones exhibit a much lower drop in potency compared to TAXOL® against a multiple drug-resistant cell line (Bollag et al. (1995)).
  • epothilones are considerably less efficiently exported from the cells by P-glycoprotein than is TAXOL® (Gerth et al. (1996)).
  • Ixabepilone is a semi-synthetic lactam analogue of patupilone that binds to tubulin and promotes tubulin polymerisation and microtubule stabilisation, thereby arresting cells in the G2/M phase of the cell cycle and inducing tumour cell apoptosis.
  • the therapeutic method of the invention comprises the administration of Formulas I, ⁇ , ED, and/or Ilia or analogs thereof.
  • a preferred epothilone analog for use in the methods of the invention is a compound of Formula II:
  • Q is selected from the group consisting of:
  • G is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, heterocyclo,
  • W is O or N Ri 5 ;
  • X is O or H, H; Y is selected from the group consisting of O; H, ORi 6 ; OR] 7 , ORi 7 ; NOR] g; H,
  • OR 19 can be a cyclic ketal
  • Z ⁇ and Z 2 are independently selected from the group consisting of CH 2 , O, NR 2 3, S, and SO 2 , wherein only one of Zi and Z 2 can be a heteroatom;
  • D is selected from the group consisting Of NR 2 SR 2 S, NR30COR3 1 and saturated heterocycle;
  • Ri, R 2 , R 3 , R 4 , Rs, Re, R 7 , Ri3 > Rw, Ri s, Ri9 > R 2 o, R21, R 22 , R26 and R 27 are independently selected from the group consisting of H, alkyl, substituted alkyl, and aryl, and when Ri and R 2 are alkyl can be joined to form a cycloalkyl, and when R3 and R 4 are alkyl can be joined to form a cycloalkyl;
  • R 9 , Rio, Ri ⁇ , Rn, R 24 , R 25 and R 3 ] are independently selected from the group consisting of H, alkyl, and substituted alkyl;
  • R 3 2j and R 33 are independently selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl and heterocyclo;
  • Another preferred epothiione for use in the present invention is a compound of Formula HI:
  • P-Q is a C 5 C double bond or an epoxide
  • R is selected from the group of H, alkyl, and substituted alkyl; R 1 is selected from the group consisting of:
  • G 1 is selected from the group of H 3 halogen, CN, alkyl and substituted alkyl;
  • G 2 is selected from the group of H f alkyl, and substituted alkyl;
  • G 3 is selected from the group of O, S, and NZ 1 ;
  • G 3 is selected from the group of halogen, N 3 , NCS, SH, CN, NC, N(Z 1 J 3 + and heteroaryl;
  • G 6 is selected from the group of H, alkyl, substituted alkyl, CF 3 , OZ 5 , SZ 5 , and NZ 5 Z 6 ;
  • G 7 is CZ 7 or N;
  • G 8 is selected from the group of H, halogen, alkyl, substituted alkyl, OZ 10 , SZ 10 , NZ 10 Z 11 ;
  • G 10 is N or CZ 12 ;
  • G 1 Ms selected from the group of H 2 N, substituted H 2 N, alkyl, substituted alkyl, aryl, and substituted aryl;
  • Z 1 , Z 6 , Z 9 , and Z 11 are independently selected from the group H, alkyl, substituted alkyl, acyl, and substituted acyl;
  • Z 2 is selected from the group of H, alkyl, substituted alkyl, aryl, substituted aryl, and heterocycle;
  • Z 3 , Z 5 , Z 8 , and Z 10 are independently selected from the group H, alkyl, substituted alkyl, acyl, substituted acyl, aryl, and substituted aryl;
  • Z 4 is selected from the group of alkyl, substituted alkyl, aryl, substituted aryl, and heterocycle;
  • Z 7 is selected from the group of H, halogen, alkyl, substituted alkyl, aryl, substituted aryl, OZ 8 , SZ S , and NZ 8 Z 9 ;
  • Z 12 is selected from the group of H, halogen, alkyl, substituted alkyl, aryl, and substituted aryl; with the proviso that when R 1 is
  • G ! , G 2 , G 3 and G 4 cannot simultaneously have the following meanings:
  • a preferred compound of Formula III of the invention is Formula Ilia:
  • P-Q is a C,C double bond or an epoxide
  • R is a H atom or a methyl group
  • G 1 is an H atom, an alkyl group, a substituted alkyl group or a halogen atom
  • G 2 is an H atom, an alkyl group or a substituted alkyl group
  • G 3 is an O atom, an S atom or an NZ 1 group with
  • Z 1 being an H atom, an alkyl group, a substituted alkyl group, an acyl group, or a substituted acyl group, and
  • Z 3 an H atom, an alkyl group, a substituted alkyl group, an acyl group or a substituted acyl group;
  • a particularly preferred compound of Formula III is [IS- [lR*,3R*(E),7R*,10S*,HR*,12R*,16S*]]-3-[2-[2-(ammomethyl)-4-thiazolyl]-l- methylethenyl] -7, 11 -dihydroxy-8,8, 10, 12, 16-pentamethyl-4, 17- dioxabicyclo[14.1.0]heptadecane-5,9-dkme (Compound 4) and pharmaceutically acceptable salts thereof.
  • lower alkyl or “lower alk” (as part of another group) refers to an unsubstituted alkyl group of 1 to 6, preferably 1 to 4, carbon atoms.
  • aralkyl refers to an aryl group bonded directly through a lower alkyl group.
  • a preferred araikyl group is benzyl.
  • aryl refers to a monocyclic or bicyclic aromatic hydrocarbon group having 6 to 12 carbon atoms in the ring portion.
  • exemplary of aryl herein are phenyl, naphthyl and biphenyl groups.
  • heterocyclo refers to a fully saturated or unsaturated, aromatic or nonaromatic cyclic group which is a 4 to 7 membered monocyclic, 7 to 11 membered bicyclic, or 10 to 15 membered tricyclic ring system which has at least one heteroatom in at least one carbon atom-containing ring.
  • Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3 or 4 heteroatoms selected from nitrogen, oxygen and sulfur where the nitrogen and sulfur heteroatoms may also optionally be oxidized and the nitrogen heteroatoms may also optionally be quaternized.
  • the heterocyclo group may be attached at any heteroatom or carbon atom.
  • Exemplary monocyclic heterocyclo groups include pyrrolidinyl, pyrrolyl, indolyl, pyrazolyl, oxetanyl, pyrazolinyl, imidazolyl, imidazolinyl, imidazolidinyl, oxazolyl, oxazolidinyl, isoxazolinyl, isoxazolyl, thiazolyl, thiadiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, furyl, tetrahydrofiiry ⁇ , thienyl, oxadiazolyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, 2-oxazepinyI, azepinyl, 4-piperidonyl, pyrldyl.
  • N-oxo-pyridyl N-oxo-pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, tetrahydrothiopyranyl, tetrahydropyranyl, morphol ⁇ ny!, thiamorpholinyl, thiamorpholinyl sulfoxide, tetrahydroth ⁇ opyranylsulfone, thiamorpholinyl sulfone, 1,3-dioxolane, tetrahydro-l,l-dioxothienyl ; dioxanyl, isothiazolidinyl, thietanyl, thiiranyl, triazinyl, triazolyl, and the like.
  • Exemplary bicyclic heterocyclo groups include benzothiazolyl, benzoxazolyl, benzothienyl, quinolinyl, quinolinyl-N-oxide, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyi, benzofuryl, chromonyl, coumarinyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl (such as fijro[2,3-c]pyridinyl, furo[3,l-b]pyridinyl or furo[2,3 ⁇ b]pyridinyl), dihydroisoindolyl, dihydroquinazolinyl (such as 3,4 ⁇ dihydro-4-oxo-quinazolinyl), benzisothiazolyl, benziso
  • a group When a group is referred to as being optionally substituted, it may be substituted with one to five, preferably one to three, substituents such as F, Cl, Br, I, trifluoromethyl, trifluoromethoxy, hydroxy, lower alkoxy, cycloa ⁇ koxy, heterocyclooxy, oxo, lower alkanoyl, aryloxy, lower alkanoyloxy, amino, lower alkylamino, arylamino, aralkylamino, cycloalkylamino, heterocycloamino, disubstituted amines in which the two amino substituents independently are selected from lower alkyl, aryl or aralkyl, lower alkanoylamino, aroylamino, aralkanoylamino, substituted lower alkanoylamino, substituted arylamino, substituted aralkylanoylamino, thiol, lower alkylthio, aryl
  • a particularly preferred epothilone analog for use in the methods of the invention is Compound 1: [lS-[lR*,3R*(E),7R*,10SMlRM2RM6S*]]-7, ⁇ - dihydroxy-8, 8, 10, 12, 16-pentamethyl-3-[ 1 -methyl-2-(2-methyl-4-thiazolyl)ethenyl] -4- aza-17-oxabicyclo[14.1.0]heptadecane-5,9-dione.
  • Another exemplary epothilone is [lS-[lR*,3R*(E),7R*.10S*,HR*,12R*,I6S*]3-3-[2-[2-(aminomethyl)-4-11iiazolyl]-l- methylethenyl] -7, 11 -dihydroxy-8 ,8, 10,12,16-pentamethyl-4 5 17- dioxabicycIo[14.1.0]heptadecane-5,9-dione., Compound 4.
  • Compound I 5 an exemplary epothilone analog of the invention, is a semisynthetic epothilone analog and has a mode of action analogous to paclitaxel (i.e., microtubule stabilization).
  • paclitaxel i.e., microtubule stabilization
  • Compound 1 has demonstrated significant improvement over paclitaxel in several critical aspects.
  • Compound 1 exhibits a very impressive and broad spectrum of antitumor activity against paclitaxel-sensitive (A2780, HCTl 16 and LS174T) and, more importantly, as well as paclitaxel-resistant human colon tumors (HCTl 16/VM46), ovarian carcinoma (Pat-7 and A2780Tax) and breast carcinoma (Pat-21) models.
  • Compound 1 is orally efficacious; the antitumor activity produced after oral administration is comparable to that produced by parenteral administration of the drug. These preclinical efficacy data indicate that Compound 1 demonstrates improved clinical efficacy in TAXOL®- insensitive and sensitive disease types.
  • Compound 2 (R)-2 ? 3,4,5-tetrahydro-l-(lH-imidazol-4-ylmethyl)-3- (phenyImethyl)-4-(2-thienylsulfonyl)-lH-l,4-benzodiazepine-7-carbonitrile s hydrochloride salt.
  • Compound 3 A CDK inhibitor is shown below
  • Combinations of a microtubulin-stabilizing agent with another agent is contemplated by the present invention, and may include the addition of an antiproliferative cytotoxic agent.
  • Classes of compounds that may be used as anti- proliferative cytotoxic agents include the following: co-stimulatory modulating agents including, without limitation, CTLA4 antagonists, ip ⁇ limumab, agatolimod, belatacept, bl ⁇ natumomab, CD40 ligand, anti- B7-1 antibody, anti-B7-2 antibody, anti-B7-H4 antibody, AG4263, eritoran, anti- OX40 antibody, ISF-154, and SGN-70; alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes): Uracil mustard, Chlormethine, Cyclophosphamide (CYTOXAN®), Ifosf
  • anti-proliferative cytotoxic agents contemplated by the present invention are navelbene, CPT-I l, anastrazole, letrazole, capecitabine, reloxafine, cyclophosphamide, ifosamide, and droloxafme.
  • Suitable anti-CTLA4 antagonist agents for use in the methods of the invention include, without limitation, anti ⁇ CTLA4 antibodies, human anti-CTLA4 antibodies, mouse anti-CTLA4 antibodies, mammalian anti-CTLA4 antibodies, humanized anti-CTLA4 antibodies, monoclonal anti-CTLA4 antibodies, polyclonal anti-CTLA4 antibodies, chimeric anti-CTLA4 antibodies, MDX-010 (ipilimumab), tremelimumab, ant ⁇ -CD28 antibodies, anti-CTLA4 adnectins, anti-CTLA4 domain antibodies, single chain anti-CTLA4 fragments, heavy chain anti-CTLA4 fragments, light chain anti-CTLA4 fragments, inhibitors of CTLA4 that agonize the co- stimuiatory pathway, the antibodies disclosed in PCT Publication No.
  • CTLA-4 antibodies are described in U.S. Patent Nos. 5,811,097, 5,855,887, 6,051,227, and 6,984,720; in PCT Publication Nos. WO 01/14424 and WO 00/37504; and in U.S. Publication Nos. 2002/0039581 and 2002/086014.
  • Other anti-CTLA-4 antibodies that can be used in a method of the present invention include, for example, those disclosed in: WO 98/42752; U.S. Patent Nos.
  • Additional anti-CTLA4 antagonists include, but are not limited to, the following: any inhibitor that is capable of disrupting the ability of CD28 antigen to bind to its cognate ligand, to inhibit the ability of CTLA4 to bind to its cognate ligand, to augment T cell responses via the co-stimulatory pathway, to disrupt the ability of B7 to bind to CD28 and/or CTLA4, to disrupt the ability of B7 to activate the co- stimulatory pathway, to disrupt the ability of CD80 to bind to CD28 and/or CTLA4, to disrupt the ability of CD 80 to activate the co-stimulatory pathway, to disrupt the ability of CD86 to bind to CD28 and/or CTLA4, to disrupt the ability of CD86 to activate the co-stimu!atory pathway, and to disrupt the co-stimulatory pathway, in general from being activated.
  • Ipilimumab refers to an anti-CTLA-4 antibody, and is a fully human IgGi M antibody derived from transgenic mice having human genes encoding heavy and light chains to generate a functional human repertoire. Ipilimumab can also be referred to by its CAS Registry No. 477202-00-9, and is disclosed as antibody IODI in PCT Publication No. WO01/14424, incorporated herein by reference in its entirety and for all purposes.
  • Ipilimumab describes a human monoclonal antibody or antigen-binding portion thereof that specifically binds to CTLA4, comprising a light chain variable region and a heavy chain variable region having a light chain variable region comprised of SEQ ID NO:5, and comprising a heavy chain region comprised of SEQ ID NO:6.
  • Pharmaceutical compositions of Ipilimumab include all pharmaceutically acceptable compositions comprising Ipilimumab and one or more diluents, vehicles and/or excipients. Examples of a pharmaceutical composition comprising Ipilimumab are provided in PCT Publication No. WO2007/67959. Impi ⁇ imumab may be administered by LV.
  • Light Chain Variable Region for lmpilimumab :
  • the administration of one or more anti-CTLA4 antagonists may be administered either alone or in combination with a peptide antigen (e.g., gplOO), in addition to an antiproliferative agent disclosed herein.
  • a peptide antigen e.g., gplOO
  • a non- limiting example of a peptide antigen would be a gplOO peptide comprising, or alternatively consisting of, the sequence selected from the group consisting of: IMDQVPFSV (SEQ ID NO:3), and YLEPGPVTV (SEQ ID NO:4).
  • Such a peptide may be administered orally, or preferably by injection sx. at I mg emulsified in incomplete Freund's adjuvant (IFA) injected s.c. in one extremity, and 1 mg of either the same or a different peptide emulsified in IFA may be injected in another extremity.
  • IFA incomplete Freund's adjuvant
  • the present invention also encompasses a pharmaceutical composition useful in the treatment of cancer, comprising the administration of a therapeutically effective amount of a microtubulin-stabilizing agent, either alone or in combination with another agent, with or without pharmaceutically acceptable carriers or diluents.
  • the compositions of the present invention may further comprise one or more pharmaceutically acceptable additional ingredient(s) such as alum, stabilizers, antimicrobial agents, buffers, coloring agents, flavoring agents, adjuvants, and the like.
  • the Formula I, H 5 HI, and/or IHa, or analogs thereof compounds, CTLA4 antagonist agents, antineoplastic agents, and compositions of the present invention may be administered orally or parenterally including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
  • the antineoplastic agents, Formulas I, ⁇ , UI, and/or HIa or analogs thereof compounds and compositions of this invention may be administered, for example, in the form of tablets or capsules, powders, dispersible granules, or cachets, or as aqueous solutions or suspensions.
  • carriers which are commonly used include lactose, corn starch, magnesium carbonate, talc, and sugar, and lubricating agents such as magnesium stearate are commonly added.
  • useful carriers include lactose, corn starch, magnesium carbonate, talc, and sugar.
  • emulsifying and/or suspending agents are commonly added.
  • sweetening and/or flavoring agents may be added to the oral compositions.
  • sterile solutions of the active ingredient(s) are usually employed, and the pH of the solutions should be suitably adjusted and buffered.
  • the total concentration of the solute(s) should be control !ed in order to render the preparation isotonic.
  • a low melting wax such as a mixture of fatty acid glycerides or cocoa butter is first melted, and the active ingredient is dispersed homogeneously in the wax, for example by stirring. The molten homogeneous mixture is then poured into conveniently sized molds and allowed to cool and thereby solidify.
  • Liquid preparations include solutions, suspensions and emulsions. Such preparations are exemplified by water or water/propylene glycol solutions for parenteral injection. Liquid preparations may also include solutions for intranasal administration.
  • Aerosol preparations suitable for inhalation may include solutions and solids in powder form, which may be in combination with a pharmaceutically acceptable carrier, such as an inert compressed gas.
  • solid preparations which are intended for conversion, shortly before use, to liquid preparations for either oral or parenteral administration.
  • liquid forms include solutions, suspensions and emulsions.
  • the compounds of Formulas I, II, IB, and/or Ilia or analogs thereof, as well as the anti-CTLA4 agents and antineoplastic agents, described herein may also be delivered transdermally.
  • the transdermal compositions can take the form of creams, lotions, aerosols and/or emulsions and can be included in a transdermal patch of the matrix or reservoir type as are conventional in the art for this purpose.
  • the combinations of the present invention may also be used in conjunction with other well known therapies that are selected for their particular usefulness against the condition that is being treated.
  • the active ingredient(s) of the microtubulin- stabilizing agents, or combination compositions, of this invention are employed within the dosage ranges described below.
  • the anti-CTLA4 agent, and Formulas I, II, 133, and/or Ilia or analogs thereof compounds may be administered separately in the dosage ranges described below.
  • the anti-CTLA4 agent is administered in the dosage range described below following or simultaneously with administration of the Formulas I, II, III, and/or Ilia or analogs thereof compound in the dosage range described below.
  • the clinician may utilize preferred dosages as warranted by the condition of the patient being treated.
  • the compound of Formula I may preferably be administered at about 40 mg/m 2 every 3 weeks.
  • Compound I may preferably be administered at about 25-60 mg/m 2 every 3 weeks.
  • Compound 2 may preferably be administered at a dosage ranging from about 25-500 mg/m 2 every three weeks for as long as treatment is required.
  • the anti-CTLA4 antibody may preferably be administered at about 0.3 - 10 mg/kg, or the maximum tolerated dose.
  • a dosage of CTLA-4 antibody is administered about every three weeks.
  • the CTLA-4 antibody may be administered by an escalating dosage regimen including administering a first dosage of CTLA-4 antibody at about 3 mg/kg, a second dosage of CTLA-4 antibody at about 5 mg/kg, and a third dosage of CTLA-4 antibody at about 9 mg/kg.
  • the escalating dosage regimen includes administering a first dosage of CTLA-4 antibody at about 5 mg/kg and a second dosage of CTLA-4 antibody at about 9 mg/kg.
  • the present invention provides an escalating dosage regimen, which includes administering an increasing dosage of CTLA-4 antibody about every six weeks.
  • a stepwise escalating dosage regimen which includes administering a first CTLA-4 antibody dosage of about 3 mg/kg, a second CTLA-4 antibody dosage of about 3 mg/kg, a third CTLA-4 antibody dosage of about 5 mg/kg, a fourth CTLA-4 antibody dosage of about 5 mg/kg, and a fifth CTLA-4 antibody dosage of about 9 mg/kg.
  • a stepwise escalating dosage regimen is provided, which includes administering a first dosage of 5 mg/kg, a second dosage of 5 mg/kg, and a third dosage of 9 mg/kg.
  • the actual dosage employed may be varied depending upon the requirements of the patient and the severity of the condition being treated. Determination of the proper dosage for a particular situation is within the skill of the art. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small amounts until the optimum effect under the circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day if desired. Intermittent therapy (e.g., one week out of three weeks or three out of four weeks) may also be used.
  • Certain cancers can be treated effectively with compounds of Formulas I, II, III, and/or IEEa and a one or more anti-CTLA4 agents.
  • Such triple and quadruple combinations can provide greater efficacy.
  • the dosages set forth above can be utilized.
  • the present invention encompasses a method for the synergistic treatment of cancer wherein anti-CTLA4 agent and a Formulas I, II, ITI, and/or Ilia compound are administered simultaneously or sequentially.
  • a pharmaceutical formulation comprising anti-CTLA4 agent(s) and a Formulas I, H, III, and/or EUa compound may be advantageous for administering the combination for one particular treatment, prior administration of the anti-CTLA4 agent(s) may be advantageous in another treatment.
  • anti-CTLA4 agent(s) and Formulas I, II, III, and/or Ilia compound may be used in conjunction with other methods of treating cancer (preferably cancerous tumors) including, but not limited to, radiation therapy and surgery.
  • cancer preferably cancerous tumors
  • a cytostatic or quiescent agent if any, may be administered sequentially or simultaneously with any or all of the other synergistic therapies.
  • the combinations of the instant invention may also be co-administered with other well known therapeutic agents that are selected for their particular usefulness against the condition that is being treated. Combinations of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when a multiple combination formulation is inappropriate.
  • the chemotherapeutic agent(s) and/or radiation therapy can be administered according to therapeutic protocols well known in the art. It will be apparent to those skilled in the art that the administration of the chemotherapeutic agent(s) and/or radiation therapy can be varied depending on the disease being treated and the known effects of the chemotherapeutic agent(s) and/or radiation therapy on that disease.
  • the therapeutic protocols e.g., dosage amounts and times of administration
  • the administered therapeutic agents i.e., anti- CTLA4 agent(s)
  • the therapeutic protocols can be varied in view of the observed effects of the administered therapeutic agents (i.e., anti- CTLA4 agent(s)) on the patient, and in view of the observed responses of the disease to the administered therapeutic agents.
  • a compound of Formula I, ⁇ , ID or Formula ma is administered simultaneously or sequentially with an anti-CTLA4 agent.
  • an anti-CTLA4 agent it is not necessary that the anti-CTLA4 therapeutic agent(s) and compound of Formulas I, II, HE, and/or IHa, be administered simultaneously or essentially simultaneously.
  • the advantage of a simultaneous or essentially simultaneous administration is well within the determination of the skilled clinician.
  • the compound of Formulas I, II, IH, and/or HIa, and anti- CTLA4 agent(s) do not have to be administered in the same pharmaceutical composition, and may, because of different physical and chemical characteristics, have to be administered by different routes.
  • the compound of Formula I, ⁇ , HI, or IV may be administered intravenously to generate and maintain good blood levels thereof, while the anti-CTLA4 agent(s) may also be administered intravenously.
  • the compound of Formula I, II, HI, or IV may be administered orally to generate and maintain good blood levels thereof, while the anti-CTLA4 agent(s) may also be administered intravenously.
  • the compound of Formula I, H, IH, or rV may be administered intravenously to generate and maintain good blood levels thereof, while the anti-CTLA4 agent(s) may also be administered orally.
  • the determination of the mode of administration and the advisability of administration, where possible, in the same pharmaceutical composition, is well within the knowledge of the skilled clinician.
  • the initial administration can be made according to established protocols known in the art, and then, based upon the observed effects, the dosage, modes of administration and times of administration can be modified by the skilled clinician.
  • the initial order of administration of the compound of Formulas I, ⁇ , III, and/or Ilia, and the anti-CTLA4 agent(s) may be varied.
  • the compound of Formulas I, II, in, and/or IUa or analogs thereof may be administered first followed by the administration of the anti-CTLA4 agent(s); or the anti-CTLA4 agent(s) may be administered first followed by the administration of the compound of Formulas I, II, III, and/or Ilia. This alternate administration may be repeated during a single treatment protocol.
  • the a ⁇ t ⁇ -CTLA4 agent(s) may be administered initially.
  • the treatment is then continued with the administration of the compound of Formulas I, II, III, and/or IHa or analogs thereof and optionally followed by administration of a cytostatic agent, if desired, until the treatment protocol is complete.
  • the administration of the compound of Formulas I, II, III, and/or Ilia or analogs thereof and optionally followed by administration of a cytostatic agent may be administered initially.
  • the treatment is then continued with the administration of the anti-CTLA4 agent(s), until the treatment protocol is complete.
  • the practicing physician can modify each protocol for the administration of a component (therapeutic agent - i.e., compound of Formulas I, ⁇ , III, and/or Ilia or analogs thereof, anti-CTLA4 agent(s)) of the treatment according to the individual patient's needs, as the treatment proceeds.
  • a component i.e., compound of Formulas I, ⁇ , III, and/or Ilia or analogs thereof, anti-CTLA4 agent(s)
  • the attending clinician in judging whether treatment is effective at the dosage administered, will consider the general well-being of the patient as well as more definite signs such as relief of disease-related symptoms, inhibition of tumor growth, actual shrinkage of the tumor, or inhibition of metastasis.
  • Size of the tumor can be measured by standard methods such as radiological studies, e.g., CAT or MRI scan, and successive measurements can be used to judge whether or not growth of the tumor has been retarded or even reversed. Relief of disease-related symptoms such as pain, and improvement in overall condition can also be used to help judge effectiveness of treatment.
  • the present invention provides methods for the treatment of a variety of cancers, including, but not limited to, the following: carcinoma including that of the bladder (including accelerated and metastatic bladder cancer), breast, colon (including colorectal cancer), kidney, liver, lung (including small and non-small cell lung cancer and lung adenocarcinoma), ovary, prostate, testes, genitourinary tract, lymphatic system, rectum, larynx, pancreas (including exocrine pancreatic carcinoma), esophagus, stomach, gall bladder, cervix, thyroid, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-ce ⁇ l lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, histiocytic
  • disorders include urticaria pigmentosa, mastocytosises such as diffuse cutaneous mastocytosis, solitary mastocytoma in human, as well as dog mastocytoma and some rare subtypes like bullous, erythrodermic and teleangiectat ⁇ c mastocytosis, mastocytosis with an associated hematological disorder, such as a myeloproliferative or myelodysplastic syndrome, or acute leukemia, myeloproliferative disorder associated with mastocytosis, mast cell leukemia, in addition to other cancers.
  • mastocytosises such as diffuse cutaneous mastocytosis, solitary mastocytoma in human, as well as dog mastocytoma and some rare subtypes like bullous, erythrodermic and teleangiectat ⁇ c mastocytosis
  • mastocytosis with an associated hematological disorder such as a myeloproliferative or myelodysplastic syndrome, or acute
  • carcinoma including that of the bladder, urothelial carcinoma, breast, colon, kidney, liver, lung, ovary, pancreas, stomach, cervix, thyroid, testis, particularly testicular seminomas, and skin; including squamous cell carcinoma; gastrointestinal stromal tumors ("GIST"); hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B- cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma and Burketts lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias and promyelocytic leukemia; tumors of mesenchymal origin, including fibrosarcoma and rhab
  • the invention is used to treat accelerated or metastatic cancers of the breast and/or lung.
  • the invention includes individual biomarkers and biomarker sets having both diagnostic and prognostic value in proliferative disease areas in which microtubulin status is of importance, e.g. ' , in cancers or tumors, or in disease states in which cell signaling and/or cellular proliferation controls are abnormal or aberrant.
  • the biomarker sets comprise a plurality of biomarkers that highly correlate with resistance or sensitivity to one or more microtubulin-stabilizing agents.
  • the biomarkers and biomarker sets of the invention enable one to predict or reasonably foretell the likely effect of one or more microtubulin-stabilizing agents in different biological systems or for cellular responses merely based upon whether one or more of the biomarkers of the present invention are overexpressed relative to normal.
  • biomarkers and biomarker sets can be used in in vitro assays of cellular proliferation by sample cells to predict in vivo outcome.
  • the various biomarkers and biomarker sets described herein, or the combination of these biomarker sets with other biomarkers or markers can be used, for example, to predict and monitor how patients with cancer might respond to therapeutic intervention with one or more microtubulin-stabilizing agents.
  • TUBB3 overexpression of TUBB3 resulted in xenografts that were resistant to docetaxel and vinorelbine, yielding antitumor efficacy ranging 0.2-0.9 and 0.1-0.3 LCK, respectively.
  • ixabepilone was active in all 4 tumors in which TUBB3 was overexpressed, yielding 1.6-4.2 LCK (see Table 1 ) when tested at their maximum tolerated doses (MTD).
  • MTD maximum tolerated doses
  • Measuring the level of expression of a biomarker and biomarker set provides a useful tool for screening one or more tumor samples before treatment of a patient with the microtubulin-stabilizing agents.
  • the screening allows a prediction of whether the cells of a tumor sample will respond favorably to the microtubulin- stabilizing agents, based on the presence or absence of over-expression - such a prediction provides a reasoned assessment as to whether or not the tumor, and hence a patient harboring the tumor, will or will not respond to treatment with the microtubulin-stabilizing agents.
  • a difference in the level of the biomarker that is sufficient to indicate whether the mammal will or will not respond therapeutically to the method of treating cancer can be readily determined by one of skill in the art using known techniques.
  • the increase or decrease in the level of the biomarker can be correlated to determine whether the difference is sufficient to identify a mammal that will respond therapeutically.
  • the difference in the level of the biomarker that is sufficient can, in one aspect, be predetermined prior to determining whether the mammal will respond therapeutically to the treatment.
  • the difference in the level of the biomarker is a difference in the mRNA level (measured, for example, by RT-PCR or a microarray), such as at least about a two-fold difference, at least about a three-fold difference, or at least about a four-fold difference in the level of expression, or more.
  • the difference in the level of the biomarker is determined at the protein level by mass spectral methods or by FISH or by IHC.
  • the difference in the level of the biomarker refers to a p-value of ⁇ 0.05 in Anova analysis.
  • the difference is determined in an ELISA assay.
  • the biomarker or biomarker set can also be used as described herein for monitoring the progress of disease treatment or therapy in those patients undergoing treatment for a disease involving a microtubulin-stabilizing agent.
  • the biomarkers also serve as targets for the development of therapies for disease treatment. Such targets may be particularly applicable to treatment of cancer, such as, for example, breast and/or lung cancer.
  • biomarkers are differentially expressed in sensitive and resistant cells, their expression patterns are correlated with relative intrinsic sensitivity of cells to treatment with microtubulin-stabilizing agents. Accordingly, the biomarkers over expressed in resistant cells may serve as targets for the development of new therapies for the tumors which are resistant to microtubulin-stabilizing agents.
  • the level of biomarker protein and/or mRNA can be determined using methods well known to those skilled in the art. For example, quantification of protein can be carried out using methods such as ELISA, 2-dimensional SDS PAGE 5 Western blot, immunoprecipitation, immunohistochemistry, fluorescence activated cell sorting (FACS), or flow cytometry. Quantification of mRNA can be carried out using methods such as PCR 5 array hybridization, Northern blot, in ⁇ situ hybridization, dot- blot, TAQMAN®, or RNAse protection assay.
  • the present invention encompasses the use of any one or more of the following as a biomarker for use in predicting microtubulin-stabilizing agent response: TUBB3, BRCP, MDRl, MRPl, and beta-tubulin mutations.
  • the present invention also encompasses any combination of the aforementioned biomarkers, including, but not limited to: TUBB3, BRCP, MDRl, MRPl, and beta-tubulin mutations; TUBB3, BRCP, MDRl, MEPl; TUBB3, BRCP, MDRl; BRCP, MDRl, MEPl, and beta-tubulin mutations; BRCP, MDRl, MRPl; MDRl, MRPl 5 and beta-tubulin mutations; TUBB3 and BRCP; TUBB3 and MDRl; TUBB3 and MRPl; TUBB3 and beta-tubulin mutations; BRCP and MDRl; BRCP and MRPl; BRCP and beta-tubulin mutations; MDRl and MRPl; MDRl and beta- tubulin mutations; and/or MRPl and beta-tubulin mutations.
  • Embodiments of the invention include measuring changes in the levels of mRNA and/or protein in a sample to determine whether said sample contains increased expression of TUBB 3, BRCP, MDRl, MPRl, and/or beta- tubulin mutations. In one aspect, said samples serve as surrogate tissue for biomarker analysis. These biomarkers can be employed for predicting and monitoring response to one or more microtubulin-stabilizing agents.
  • the biomarkers of the invention are one or more of the following: TUBB3, BRCP 9 MDRl, MPRl, and/or beta-tubulin mutations, including both polynucleotide and polypeptide sequences.
  • the biomarkers of the invention are nucleotide sequences that, due to the degeneracy of the genetic code, encodes for a polypeptide sequence provided in the sequence listing.
  • the biomarkers serve as useful molecular tools for predicting and monitoring response to microtubulin-stabilizing agents.
  • Methods of measuring the level of any given marker described herein may be performed using methods well known in the art, which include, but are not limited to PCR; RT-PCR; FISH; IHC; immuno-detection methods; immunoprecipitation; Western Blots; ELISA; radioimmunoassays; PET imaging; HPLC; surface plasmon resonance, and optical spectroscopy; and mass spectrometry, among others.
  • the biomarkers of the invention may be quantified using any immunospecific binding method known in the art.
  • the immunoassays which can be used include but are not limited to competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, to name but a few.
  • competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric
  • Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X- 100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2, 1% TRASYLOL®) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding the antibody of interest (i.e., one directed to a biomarker of the present invention) to the cell lysate, incubating for a period of time (e.g., 1-4 hours) at 4° C, adding protein A and/or protein G SEPHAROSE® beads to the cell lysate, incubating for about an hour or more at 4° C, washing the beads in lysis buffer and resuspending the beads in SDS/sample buffer.
  • a lysis buffer such as
  • the ability of the antibody of interest to immunoprecipitate a particular antigen can be assessed by, e.g., western blot analysis.
  • One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the antibody to an antigen and decrease the background (e.g., pre-clearing the cell lysate with SEPHAROSE® beads).
  • immunoprecipitation protocols see, e.g., Ausubel et al., eds., Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.16.1 (1994).
  • J Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%- 20% SDS- PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or nonfat milk), washing the membrane in washing buffer (e.g., PBS-Tween 20), blocking the membrane with primary antibody (the antibody of interest) diluted in blocking buffer, washing the membrane in washing buffer, blocking the membrane with a secondary antibody (which recognizes the primary antibody, e.g., an anti-human antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., 32P or 1251) diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen.
  • ELISAs comprise preparing antigen, coating the well of a 96 well microtiter plate with the antigen, adding the antibody of interest conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the well and incubating for a period of time, and detecting the presence of the antigen.
  • a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase)
  • a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase)
  • a second antibody conjugated to a detectable compound may be added following the addition of the antigen of interest to the coated well.
  • ELISAs see, e.g., Ausubel et al., eds., Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 11.2.1 (1994).
  • identifying the relative quantitation of the biomarker polypeptide(s) may be performed using tandem mass spectrometry; or single or multi dimensional high performance liquid chromatography coupled to tandem mass spectrometry.
  • the method takes into account the fact that an increased number of fragments of an identified protein isolated using single or roulti dimensional high performance liquid chromatography coupled to tandem mass spectrometry directly correlates with the level of the protein present in the sample.
  • Such methods are well known to those skilled in the art and described in numerous publications, for example, 2-D Proteome Analysis Protocols, AJ. Link, ed., Humana Press (1999), ISBN: 0896035247; Mass Spectrometry of Proteins and Peptides, J.R. Chapman, ed., Humana Press (2000), ISBN: 089603609X.
  • the terms “modulate” or “modulates” or “modulators” refer to an increase or decrease in the amount, quality or effect of a particular activity, or the level of DNA, RNA, or protein detected in a sample.
  • the following examples are presented primarily for the purpose of illustrating more specific details thereof. The scope of the invention should not be deemed limited by the examples, but to encompass the entire subject matter defined by the claims. EXAMPLE 1 - METHOD OF ASSESSING THE RESPONSE OF TUBB3 OVEREXPRESSING TUMOR XENOGRAFTS TO THE ADMINISTRATION
  • Cancer cell lines overexpressing TUBB3 were evaluated in vivo in mice for sensitivity to ixabepilone, docetaxel and vinorelbine. These include DU4475 and PAT21 breast, as well as Hl 155 and LX-I lung cancer lines. BCRP overexpressing HEK-293 cell line was studied in vitro for sensitivity to ixabepilone, paclitaxel and mitoxantrone.
  • HEK and HEK/BCRP cells were maintained in RPMI-1640 (Gibco) supplemented with 10% heat-inactivated fetal bovine serum and 25 mM HEPES at 37°C/5% CO2.
  • Cell growth assays were conducted in 6-well culture plates. Cells were plated at a density of 4 x 104 cells/well overnight. Compounds were then added (total DMSO content not exceeding 0.1%). Cell growth was determined by the direct counting of cell number, following trypsinization, using a Coulter Channelyzer.
  • Tumor response was determined by measurement of tumors by caliper twice a week
  • Efficacy evaluation in nude mice demonstrated that the 4 xenografts overexpressing TUBB3 were resistant to docetaxel and vinorelbine, yielding antitumor efficacy ranging 0.2-0.9 and 0.1-0.3 LCK 3 respectively (see Fig. 5).
  • ixabepilone was active in all 4 tumors, yielding 1.6-4.2 LCK (Table 1) when tested at their maximum tolerated doses (MTD).
  • This resistance can be reversed by fumitremorg ⁇ n C, a selective inhibitor of BCRP (see Fig. 7).
  • Ixabepilone's clinical activity has been demonstrated in metastatic breast cancer patients who developed resistance to other chemotherapy regimens, including anthracyclines and taxanes.
  • ixabepilone was recently FDA-approved in the United States combination with capecitabine for the treatment of patients with metastatic or locally advanced breast cancer after failure of an anthracycline and a taxane, and as monotherapy for the treatment of metastatic or locally advanced breast cancer in patients after failure of an anthracycline, a taxane, and capecitabine.
  • the results from the present study add to accumulating data supporting the inclusion of ixabepilone as a key component of breast cancer treatment.
  • Total RNA may be purified using RNEASY® system (Qiagen, CA 5 USA).
  • Mixed Oligo-d(T)is primers may be used to generate single-stranded cDNAs using the SUPERSCRIPT® First-strand Synthesis kit (Invitrogen, CA, USA).
  • Levels for each gene of interest and GAPDH transcripts may be analyzed using an Applied Biosystems 7900HT Sequence Detection System.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • Hospice & Palliative Care (AREA)
  • Microbiology (AREA)
  • Pathology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A method for treating cancer comprising identifying a mammal that overexpresses breast cancer resistance protein; and administering to said mammal a pharmaceutical composition comprising a therapeutically effective amount of ixabepilone. In one aspect, the mammal is not administered an agent that is susceptible to breast cancer resistance protein overexpression resistance. In another aspect, the cancer is breast and/or lung cancer.

Description

METHODS FOR TREATING CANCER IN PATIENTS HAVING BREAST
CANCER RESISTANCE PROTEIN OVEREXPRESSION
[0001] This application claims benefit to provisional application U.S. Serial No. 61/021,149, filed January 15, 2008; under 35 U.S.C. 119(e). The entire teachings of the referenced applications are incorporated herein by reference.
FIELD OF THE INVENTION
[0002] The present invention relates generally to the field of pharmacogenomics, and more specifically to methods and procedures to determine drug sensitivity in patients to allow the identification of individualized genetic profiles which will aid in treating diseases and disorders.
BACKGROUND OF THE INVENTION [0003] The 72-kDa breast cancer resistance protein (BCRP) is the second member of the subfamily G of the human ATP binding cassette (ABC) transporter superfamily and thus also designated as ABCG2. Unlike P-glycoproteϊn and MRPl, which are arranged in 2 repeated halves^ BCRP is a half-transporter consisting of only 1 nucleotide binding domain followed by 1 membrane-spanning domain. Current experimental evidence suggests that BCRP may function as a homodimer or homotetramer. Overexpression of BCRP is associated with high levels of resistance to a variety of anticancer agents, including anthracyclines, mitoxantrone, and the camptothecins, by enhancing drug efflux. BCRP expression has been detected in a large number of hematological malignancies and solid tumors, indicating that this transporter may play an important role in clinical drug resistance of cancers. In addition to its role to confer resistance against chemotherapeutic agents, BCRP actively transports structurally diverse organic molecules, conjugated or unconjugated, such as estrone-3 -sulfate, 17β-estradiol 17-(β-D-gIucuronide), and methotrexate. BCRP is highly expressed in the placental syncytiotrophoblasts, in the apical membrane of the epithelium in the small intestine, in the liver canalicular membrane, and at the luminal surface of the endothelial cells of human brain microvessels. This strategic and substantial tissue localization indicates that BCRP also plays an important role in absorption, distribution, and elimination of drugs that are BCRP substrates. See, Mao et al., AAPS Journal, 07(01):E118-E133 (2005). [0004] New prognostic and predictive markers, which would facilitate an individualization of therapy for each patient, are needed to accurately predict patient response to treatments, such as small molecule or biological molecule drugs, in the clinic. The problem may be solved by the identification of new parameters that could better predict the patient's sensitivity to treatment. The classification of patient samples is a crucial aspect of cancer diagnosis and treatment. The association of a patient's response to a treatment with molecular and genetic markers can open up new opportunities for treatment development in non-responding patients, or distinguish a treatment's indication among other treatment choices because of higher confidence in the efficacy. Further, the pre-selection of patients who are likely to respond well to a medicine, drug, or combination therapy may reduce the number of patients needed in a clinical study or accelerate the time needed to complete a clinical development program (Cockett, M. et al., Current Opinion in Biotechnology, 11 : 602-609 (2000)). [0005] The ability to determine which patients are responding to anti-angiogenesis therapies (such as microtubule-stabilizing agents) or predict drug sensitivity in patients is particularly challenging because drug responses reflect not only properties intrinsic to the target cells, but also a host's metabolic properties. Efforts to use genetic information to predict or monitor drug response have primarily focused on individual genes that have broad effects, such as the multidrug resistance genes mdrl and mrpl (Sonneveld, P., J. Intern. Med, 247:521-534 (2000)). [0006] The development of microarray technologies for large scale characterization of gene mRNA expression pattern has made it possible to systematically search for molecular markers and to categorize cancers into distinct subgroups not evident by traditional histopathological methods (Khan, J. et al., Cancer Res., 58:5009-5013 (1998); Alizadeh, AA. et al., Nature, 403:503-511 (2000); Bittner, M. et al., Nature, 406:536-540 (2000); Khan, J. et al., Nature Medicine, 7(6):673-679 (2001); and Golub, T.R. et al., Science, 286:531-537 (1999); Alon, U. et al., Proc. Natl Acad. ScL USA, 96:6745-6750 (1999)). Such technologies and molecular tools have made it possible to monitor the expression level of a large number of transcripts within a cell population at any given time (see, e.g., Schena et al., Science, 270:467-470 (1995); Lockhart et al., Nature Biotechnology, 14:1675-
1680 (1996); Blanchard et al., Nature Biotechnology, 14:1649 (1996); U.S. Patent No.
5,569,588 to Ashby et al.).
[0007] Recent studies demonstrate that gene expression information generated by microarray analysis of human tumors can predict clinical outcome (van't Veer, LJ. et al., Nature, 415:530-536 (2002); Shipp, M. et al., Nature Medicine, 8(l):68-74
(2002); Glinsky, G. et al., J. Clin. Invest, 113(6):913-923 (2004)). These findings bring hope that cancer treatment will be vastly improved by better predicting and monitoring the response of individual tumors to therapy. [0008] Microtubule-stabilizing agents, such as ixabepilone (IXEMPRA™) and paclitaxel (TAXOL®), are commonly used for the treatment of many types of cancer, including breast and lung cancer.
[0009] Needed are new and alternative methods and procedures to determine drug sensitivity or monitor response in patients to allow the development of individualized diagnostics which are necessary to treat diseases and disorders based on patient response at a molecular level, particularly for breast and lung cancer patients.
SUMMARY OF THE INVENTION [0010] The invention provides methods and procedures for determining patient sensitivity to one or more microtubule-stabilizing agents.
[0011] In one aspect, the invention relates to a method for treating cancer comprising identifying a mammal that overexpresses breast cancer resistance protein; and administering to said mammal a pharmaceutical composition comprising a therapeutically effective amount of ixabepilone, either alone or in combination with another agent. In one aspect, the mammal is not administered an agent that is susceptible to breast cancer resistance protein overexpression resistance. In another aspect, the cancer is breast and/or lung cancer. In yet another aspect, the mammal further overexpresses at least one of BRCP (ABCG2), beta-tubulin UI (TUBB3), MDRl, MRPl, and a beta-tubulin mutant. In one aspect, the mammal is a human. [0012] The present invention provides a method of screening a biological sample, for example cells that do not respond, or that have stopped responding, or that have a diminished response, to one or more microtubule-stabilizing agents. For example, the present invention provides a method of screening cells from an individual suffering from cancer who is either being treated with one or more microtubule-stabilizing agents or is naϊve to said agents, and whose cells do not respond or have stopped responding or that have a diminished response to one or more microtubule-stabilizing agents, for overexpression of breast cancer resistance protein relative to a standard. If breast cancer resistance protein overexpression is present, administration of a therapeutically acceptable amount of ixabepilone, alone or in combination with one or more microtubule-stabilizing agents and/or other agent, such as a CTLA4 antagonist, is warranted to inhibit proliferation of said cells. Wherein said cancer is breast and/or lung cancer.
[0013] The present invention provides a method of screening a biological sample, for example cells that do not respond, or that have stopped responding, or that have a diminished response, to one or more microtubule-stabilizing agents. For example, the present invention provides a method of screening cells from an individual suffering from cancer who is either being treated with one or more microtubule-stabilizing agents or is naϊve to said agents, and whose cells do not respond or have stopped responding or that have a diminished response to one or more microtubule-stabilizing agents, for overexpression of breast cancer resistance protein and beta-tubulin III relative to a standard. If breast cancer resistance protein and beta-tubulin HI overexpression is present, administration of a therapeutically acceptable amount of ixabepilone, alone or in combination with one or more microtubule-stabilizing agents and/or other agent, such as a CTLA4 antagonist, is warranted to inhibit proliferation of said cells. Wherein said cancer is breast and/or lung cancer. [0014] The present invention provides a method of screening a biological sample, for example cells that do not respond, or that have stopped responding, or that have a diminished response, to one or more microtubule-stabilizing agents. For example, the present invention provides a method of screening cells from an individual suffering from cancer who is either being treated with one or more microtubule-stabilizing agents or is naϊve to said agents, and whose cells do not respond or have stopped responding or that have a diminished response to one or more microtubule-stabilizing agents, for overexpression of breast cancer resistance protein and MDRl relative to a standard. If breast cancer resistance protein and MDRl overexpression is present, administration of a therapeutically acceptable amount of ϊxabepilone, alone or in combination with one or more microtυbule-stabilizing agents and/or other agent, such as a CTLA4 antagonist, is warranted to inhibit proliferation of said cells. Wherein said cancer is breast and/or lung cancer. [0015] The present invention provides a method of screening a biological sample, for example cells that do not respond, or that have stopped responding, or that have a diminished response, to one or more microtubule-stabilizing agents. For example, the present invention provides a method of screening cells from an individual suffering from cancer who is either being treated with one or more microtubule-stabilizing agents or is naϊve to said agents, and whose cells do not respond or have stopped responding or that have a diminished response to one or more microtubule-stabilizing agents, for overexpression of breast cancer resistance protein and MRPl relative to a standard. If breast cancer resistance protein and MRPl overexpression is present, administration of a therapeutically acceptable amount of ixabepilone, alone or in combination with one or more microtubule-stabilizing agents and/or other agent, such as a CTLA4 antagonist, is warranted to inhibit proliferation of said cells. Wherein said cancer is breast and/or lung cancer.
[0016] The present invention provides a method of screening a biological sample, for example cells that do not respond, or that have stopped responding, or that have a diminished response, to one or more microtubule-stabilizing agents. For example, the present invention provides a method of screening cells from an individual suffering from cancer who is either being treated with one or more microtubule-stabilizing agents or is naϊve to said agents, and whose cells do not respond or have stopped responding or that have a diminished response to one or more microtubule-stabilizing agents, for overexpression of breast cancer resistance protein and beta-tubulin mutant relative to a standard. If breast cancer resistance protein and beta-tubulin mutant overexpression is present, administration of a therapeutically acceptable amount of ixabepilone, alone or in combination with one or more microtubule-stabilizing agents and/or other agent, such as a CTLA4 antagonist, is warranted to inhibit proliferation of said cells. Wherein said cancer is breast and/or lung cancer.
[0017] The diagnostic methods of the invention can be, for example, an in vitro method wherein the step of measuring in the mammal the level of at least one biomarker comprises taking a biological sample from the mammal and then measuring the level of the biomarker(s) in the biological sample. The biological sample can comprise, for example, at least one of serum, whole fresh blood, peripheral blood mononuclear cells, frozen whole blood, fresh plasma, frozen plasma, urine, saliva, skin, hair follicle, bone marrow, or tumor tissue.
[0018] The level of the at least one biomarker can be, for example, the level of protein and/or mRNA transcript of the biomarker(s).
[0019] The invention also provides an isolated TUBB3 biomarker, an isolated BRCP (ABCG2) biomarker, an isolated MDRl (ABCBl) biomarker, an isolated MRPl (ABCCl) biomarker, and tubulin mutation biomarkers. The biomarkers of the invention include nucleotide and amino acid sequences of full-length TUBB3, BRCP (ABCG2), MDRl (ABCBl), MRPl (ABCCl), and beta-tubulin mutations, as well as fragments and variants thereof. [0020] The invention also provides a biomarker set comprising two or more biomarkers of the invention.
[0021] The invention also provides kits for measuring overexpression of breast cancer resistance protein, and uses thereof. The invention also provides antibodies, including polyclonal or monoclonal, directed breast cancer resistance protein, and uses thereof. [0022] The invention will be better understood upon a reading of the detailed description of the invention when considered in connection with the accompanying Figures.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 shows a schematic of the structure and mechanism of ixabepilone.
Figure 2 illustrates the issue of drug resistance for taxanes as well as providing examples of several mechanisms of how such resistance may arise.
Figure 3 shows the ability of ixabepilone to overcome multiple mechanisms of taxane-resi stance. Figure 4 provides an illustration of ABC drug resistance transporters.
Figure S shows the ability of ixabepilone to effectively overcome the BCRP overexpression-induced resistance to microtubulin stabilizers. BCRP overexpressing HEK293 cells are denoted as "BCRP/HEK 293 Cells", while control HEK293 cells are denoted as "HEK 293 Cells".
Figure 6 shows the IC50 of paclitaxei, mitotropine, and ixabepilone in BCRP overexpressing HEK293 cells. As shown, ixabepilone had a very low IC50.
Figure 7 shows the detection of BCRP overexpressing cells using Hoescht 33342 dye and flow cytometry, and the abolition of BCRP overexpressing cells subsequent to the administration of the BCRP inhibitor, Fumitremorgin C.
Figure 8 shows the increased ability of ixabepilone to overcome BCRP- overexpression dependent resistance to taxanes in a human lung carcinoma cell line, H441.
DETAILED DESCRIPTION OF THE INVENTION [0023] As is known in the art, ixabepilone refers to a compound having the following structure (I):
Figure imgf000009_0001
O).
Compound (I) can also be referred to as (1S,3S,7S,1OR,11S,12S,16R)-7,11 - d ihydroxy-8 , 8, 10,12,16-pentamethyl-3-[( 1 E)- 1 -m ethyl-2-(2-methyI-4- thiazolyl)ethenyl]- 17-oxa-4-azabicyclo[ 14.1.0]heptadecane-5 ,9-dione in accordance with IUPAC nomenclature. Use of the term "(1S,3S,7S51OR,11S,12S,16R)-7,11- dihydroxy-8,8,10J12,16-pentamethyl-3-[(lE)-l-methyl-2-(2-methyI-4- thiazolyi)ethenyi]-l 7-oxa-4-azabicyclo[ 14, 1 ,0]heptadecane-5,9-dione" encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of the compound (T) or its salts, such as the forms of (ϊ) described in U.S. Patent No. 6,605,599, issued August 12, 2003, incorporated herein by reference in its entirety and for all purposes. Pharmaceutical compositions Of (IS5SSJS5IOR11 18,128,16R)-7S 11- dihydroxy-8,8,10, 12, 16-ρentamethyl-3-[( IE)-I -methyl-2-(2-methyl-4- thiazolylJethenylj-π-oxa^-azabicyclotM.l.OJheptadecane-S^-dione include all pharmaceutically acceptable compositions comprising (IS,3S,7S,1OR,11S,12S,16R)- 7,1 l-dihydroxy-8,8, 10, 12, 16-pentamethyl-3-[(l E)-I -methyl-2-(2-methyl-4- thiazolyl)ethenyl]-17-oxa-4-azabicyclo[14.1.0]heptadecane-5,9-dione and one or more diluents, vehicles and/or excipients One example of a pharmaceutical composition comprising (lS>3S,7S,10R,l lS,12S,16R)-7,l l-dihydroxy-8,8,10,12,16-pentamethyl- 3-[(IE)-I -methyl-2 -(2-methyl-4-thiazolyl)ethenyI] - 17-oxa-4- azabicycIo[14.1.0]heptadecane-5,9-dione is IXEMPRA™ (Bristol-Myers Squibb Company). IXEMPRA™ comprises (lS,3S57S,10R,l lS,12S!16R)-7,n-dihydroxy- 8,8,10,12,16-pentamethyl-3-[(lE)-l-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-17-oxa- 4-azabicyclo[14.1.0]heptadecane-5,9-dione as the active ingredient, also referred to as ixabepilone, for IV infusion including inactive ingredients in the form of a diluent consisting of a sterile, non-pyrogenic of 52.8% (w/v) purified polyoxyethylated castor oil and 39.8% (w/v) dehydrated alcohol, USP. [0024] Non-limiting examples of other epothϋones for use in the methods and compositions of the present invention are encompassed by formula II:
Figure imgf000011_0001
wherein:
Q is selected from the group consisting of:
Figure imgf000011_0002
G is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, heterocyclo,
Figure imgf000011_0003
W is O or N R!5;
X is O or H, H;
Y is selected from the group consisting of O; H, ORie; ORn, OR]7; NOR] «; H, NHORi9; H, NR20R2S; H, H; and CHR22; wherein OR!7, ORi7 can be a cyclic ketal;
Zi and Z2 are independently selected from the group consisting of CH2, O, NR23, S, and SO2, wherein only one of Zi and Z2 can be a heteroatom; Bi and B2 are independently selected from the group consisting of OR24, OCOR25, and
Figure imgf000012_0001
and when Bi is H and Y is OH, H, they can form a six-membered ring ketal or acetal;
D is selected from the group consisting of NR28R29, NR30COR31 and saturated heterocycle;
Ru R2, R3, R4, R5, R6, R?, Ri3, Rw, Ris, Rw, R2o» R21, R22, R26 and R27 are independently selected from the group consisting of H, alkyl, substituted alkyl, and aryl, and when Rj and R2 are alkyl can be joined to form a cycloalkyl, and when R3 and R4 are alkyl can be joined to form a cycloalkyl;
R9, Rio, RIO, Rj7, R24> R25 and R31 are independently selected from the group consisting of H, alkyl, and substituted alkyl;
Rs, R11, R i 2, R28, R3o, R32, and R33 are independently selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl and heterocyclo; and
Ri 5, R23 and R29 are independently selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, heterocyclo, R32C=O, R33SO2, hydroxy, O-alkyl or O-substituted alkyl; and pharmaceutically acceptable salts thereof and any hydrates, solvates or geometric, optical and stereoisomers thereof.
[0025] Formula III provides another example of an epothilone suitable for use in the methods and compositions of the present invention:
Figure imgf000012_0002
(III) wherein: P-Q is a C, C double bond or an epoxide; G is
Figure imgf000013_0001
R is selected from the group of H, alkyl, and substituted alkyl; R1 is selected from the group consisting of:
Figure imgf000013_0002
R2 is
Figure imgf000013_0003
G1 is selected from the group of H, halogen, CN, alkyl and substituted alkyl; G2 is selected from the group of H, alkyl, and substituted alkyl;
G3 is selected from the group of O, S, and NZ1;
G4 is selected from the group of H, alkyl, substituted alkyl, OZ2, NZ2Z3, Z2C=O, Z4SO2, and optionally substituted glycosyi;
G5 is selected from the group of halogen, N3, NCS, SH, CN, NC, N(Z1J3 + and heteroaryl;
G6 is selected from the group of H, alkyl, substituted alkyl, CF3, OZ5, SZ5, and NZ5Z6;
G7 is CZ7 or N;
G8 is selected from the group of H, halogen, alkyl, substituted alkyl, OZ i0, SZ10, NZ10Z11;
G9 is selected from the group of O, S, -NH-NH- and -N=N-;
G10 is N or CZ12;
G11 is selected from the group of H2N, substituted H2N, alkyl, substituted alkyl, aryl, and substituted aryl; Z1, Z6, Z9, and Zn are independently selected from the group H, alkyl, substituted alkyl, acyl, and substituted acyl;
Z2 is selected from the group of H, alkyl, substituted alkyl, aryl, substituted aryl, and heterocycle; Z3, Z5, Z8, and Z10 are independently selected from the group H, alkyl, substituted alkyl, acyl, substituted acyl, aryl, and substituted aryl;
Z4 is selected from the group of alkyl, substituted alkyl, aryl, substituted aryl, and heterocycle;
Z7 is selected from the group of H, halogen, alkyl, substituted alkyl, aryl, substituted aryl, OZ8, SZ8, and NZ8Z9; and
Z12 is selected from the group of H, halogen, alkyl, substituted alkyl, aryl, and substituted aryl; with the proviso that when R1 is
Figure imgf000014_0001
G1 S G2, G3 and G4 cannot simultaneously have the following meanings:
G1 and G2 = H, G3 = O and G4 = H or Z2C=O where Z2 = alkyl group.
[0026] A preferred compound of Formula III of the invention is Formula Ilia:
R
Figure imgf000014_0002
(HIa) wherein the symbols have the following meaning: P-Q is a C,C double bond or an epoxide; R is a H atom or a methyl group; G1 is an H atom, an alkyl group, a substituted alkyl group or a halogen atom; G2 is an H atom, an alkyl group or a substituted alkyl group;
G3 is an O atom, an S atom or an NZ1 group with Z1 being an H atom, an alkyl group, a substituted alkyl group, an acyl group, or a substituted acyl group;
G4 is an H atom, an alkyl group, a substituted alkyl group, an OZ2 group, an NZ2Z3 group, a Z2C=O group, a Z4SO2 group or an optionally substituted glycosyl group with Z2 being a H atom, an alkyl group, a substituted alkyl group, an aryl group, a substituted aryl group or a heterocyclic group;
Z3 an H atom, an alkyl group, a substituted alkyl group, an acyl group or a substituted acyl group; and Z4 an alkyl, a substituted alkyl, an aryl, a substituted aryl or a heterocyclic group, with the proviso that G1, G2, G3 and G4 cannot have simultaneously the following meanings: G1 and G2 = H atom, G3 = O atom and G4 - H atom or Z2C=O with Z2 = alkyl group.
[0027] A particularly preferred compound of Formula III is [IS- [lR*,3R*(E),7R*,10S*,l lR*>12R*,16S*]]-3-[2-[2-(aminomethyl)-4-thiazolyl]-l- methylethenyl]-7s 11 -dihydroxy-8,8, 10, 12, 16-pentamethyl-4, 17- dioxabicyclo[14.1.0]heptadecane-5,9-dione (Compound 4) and pharmaceutically acceptable salts thereof. [0028] The phrase "microtubule modulating agent" is meant to refer to agents that either stabilize microtubule or destabilize microtubulin synthesis and/or polymerization.
[0029] Microtubulin modulatory agents either agonize or inhibit a cells ability to maintain proper microtubulin assemblies. In the case of paclitaxel (marketed as TAXOL®) causes mitotic abnormalities and arrest, and promotes microtubule assembly into calcium-stable aggregated structures resulting in inhibition of cell replication.
[0030] Epothilones mimic the biological effects of TAXOL®, (Bollag et al, Cancer Res., 55:2325-2333 (1995), and in competition studies act as competitive inhibitors of TAXOL® binding to microtubules. However, epothilones enjoy a significant advantage over TAXOL® in that epothilones exhibit a much lower drop in potency compared to TAXOL® against a multiple drug-resistant cell line (Bollag et al. (1995)). Furthermore, epothilones are considerably less efficiently exported from the cells by P-glycoprotein than is TAXOL® (Gerth et al. (1996)). [0031] Ixabepilone is a semi-synthetic lactam analogue of patupilone that binds to tubulin and promotes tubulin polymerisation and microtubule stabilisation, thereby arresting cells in the G2/M phase of the cell cycle and inducing tumour cell apoptosis. [0032] Thus, in a preferred embodiment, the therapeutic method of the invention comprises the administration of Formulas I, π, ED, and/or Ilia or analogs thereof. [0033] A preferred epothilone analog for use in the methods of the invention is a compound of Formula II:
Figure imgf000016_0001
wherein:
Q is selected from the group consisting of:
Figure imgf000016_0002
G is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, heterocyclo,
Figure imgf000017_0001
W is O or N Ri5;
X is O or H, H; Y is selected from the group consisting of O; H, ORi6; OR]7, ORi7; NOR] g; H,
NHOR19; H, NR20R21; H, H; and CHR22; wherein ORn, OR17 can be a cyclic ketal;
Z\ and Z2 are independently selected from the group consisting of CH2, O, NR23, S, and SO2, wherein only one of Zi and Z2 can be a heteroatom;
B] and B2 are independently selected from the group consisting of OR24, OCOR25, and 0-Ct=O)-NR26R27, and when B, is H and Y is OH, H, they can form a six-membered ring ketal or acetal;
D is selected from the group consisting Of NR2SR2S, NR30COR31 and saturated heterocycle;
Ri, R2, R3, R4, Rs, Re, R7, Ri3> Rw, Ri s, Ri9> R2o, R21, R22, R26 and R27 are independently selected from the group consisting of H, alkyl, substituted alkyl, and aryl, and when Ri and R2 are alkyl can be joined to form a cycloalkyl, and when R3 and R4 are alkyl can be joined to form a cycloalkyl;
R9, Rio, Riό, Rn, R24, R25 and R3] are independently selected from the group consisting of H, alkyl, and substituted alkyl; Rz, Rii, R12, R2S, R3o> R32j and R33 are independently selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl and heterocyclo;
Ri5, R23 and R29 are independently selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, heterocyclo, R32C=O, R^SO2, hydroxy, O-alkyl or O-substituted aϊkyl; and pharmaceutically acceptable salts thereof and any hydrates, solvates or geometric, optical and stereoisomers thereof. [0034] Another preferred epothiione for use in the present invention is a compound of Formula HI:
Figure imgf000018_0001
(III) wherein:
P-Q is a C5 C double bond or an epoxide; G is
Figure imgf000018_0002
R is selected from the group of H, alkyl, and substituted alkyl; R1 is selected from the group consisting of:
Figure imgf000018_0003
R2 is
Figure imgf000018_0004
G1 is selected from the group of H3 halogen, CN, alkyl and substituted alkyl; G2 is selected from the group of Hf alkyl, and substituted alkyl; G3 is selected from the group of O, S, and NZ1;
G4 is selected from the group of H, alkyl, substituted alkyl, OZ2, NZ2Z3, Z2C=O, Z4SO2, and optionally substituted glycosyl; G3 is selected from the group of halogen, N3, NCS, SH, CN, NC, N(Z1J3 + and heteroaryl;
G6 is selected from the group of H, alkyl, substituted alkyl, CF3, OZ5, SZ5, and NZ5Z6; G7 is CZ7 or N;
G8 is selected from the group of H, halogen, alkyl, substituted alkyl, OZ10, SZ10, NZ10Z11;
G9 is selected from the group of O, S, -NH-NH- and -N=N-;
G10 is N or CZ12; G1Ms selected from the group of H2N, substituted H2N, alkyl, substituted alkyl, aryl, and substituted aryl;
Z1, Z6, Z9, and Z11 are independently selected from the group H, alkyl, substituted alkyl, acyl, and substituted acyl;
Z2 is selected from the group of H, alkyl, substituted alkyl, aryl, substituted aryl, and heterocycle;
Z3, Z5, Z8, and Z10 are independently selected from the group H, alkyl, substituted alkyl, acyl, substituted acyl, aryl, and substituted aryl;
Z4 is selected from the group of alkyl, substituted alkyl, aryl, substituted aryl, and heterocycle; Z7 is selected from the group of H, halogen, alkyl, substituted alkyl, aryl, substituted aryl, OZ8, SZS, and NZ8Z9; and
Z12 is selected from the group of H, halogen, alkyl, substituted alkyl, aryl, and substituted aryl; with the proviso that when R1 is
Figure imgf000019_0001
G!, G2, G3 and G4 cannot simultaneously have the following meanings:
G1 and G2 = H, G3 = O and G4 = H or Z2C=O where Z2 = alkyl group.
[0035] A preferred compound of Formula III of the invention is Formula Ilia:
Figure imgf000020_0001
(IHa) wherein the symbols have the following meaning:
P-Q is a C,C double bond or an epoxide; R is a H atom or a methyl group;
G1 is an H atom, an alkyl group, a substituted alkyl group or a halogen atom; G2 is an H atom, an alkyl group or a substituted alkyl group; G3 is an O atom, an S atom or an NZ1 group with
Z1 being an H atom, an alkyl group, a substituted alkyl group, an acyl group, or a substituted acyl group, and
G4 is an H atom, an alkyl group, a substituted alkyl group, an OZ2 group, an NZ2Z3 group, a Z2C=O group, a Z4SO2 group or an optionally substituted glycosyl group with Z2 being a H atom, an alkyl group, a substituted alkyl group, an aryl group, a substituted aryl group or a heterocyclic group; Z3 an H atom, an alkyl group, a substituted alkyl group, an acyl group or a substituted acyl group; and
Z4 an alkyl, a substituted alkyl, an aryl, a substituted aryl or a heterocyclic group, with the proviso that G1, G2, G3 and G4 cannot have simultaneously the following meanings: G1 and G2 = H atom, G3 = O atom and G4 = H atom or Z2C=O with Z2 = alkyl group.
[0036] A particularly preferred compound of Formula III is [IS- [lR*,3R*(E),7R*,10S*,HR*,12R*,16S*]]-3-[2-[2-(ammomethyl)-4-thiazolyl]-l- methylethenyl] -7, 11 -dihydroxy-8,8, 10, 12, 16-pentamethyl-4, 17- dioxabicyclo[14.1.0]heptadecane-5,9-dkme (Compound 4) and pharmaceutically acceptable salts thereof.
[0037] When describing the compounds of the present invention, the phrase "lower alkyl" or "lower alk" (as part of another group) refers to an unsubstituted alkyl group of 1 to 6, preferably 1 to 4, carbon atoms.
[0038] The term "aralkyl" refers to an aryl group bonded directly through a lower alkyl group. A preferred araikyl group is benzyl.
[0039] The term "aryl" refers to a monocyclic or bicyclic aromatic hydrocarbon group having 6 to 12 carbon atoms in the ring portion. Exemplary of aryl herein are phenyl, naphthyl and biphenyl groups.
[0040] The term "heterocyclo" refers to a fully saturated or unsaturated, aromatic or nonaromatic cyclic group which is a 4 to 7 membered monocyclic, 7 to 11 membered bicyclic, or 10 to 15 membered tricyclic ring system which has at least one heteroatom in at least one carbon atom-containing ring. Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3 or 4 heteroatoms selected from nitrogen, oxygen and sulfur where the nitrogen and sulfur heteroatoms may also optionally be oxidized and the nitrogen heteroatoms may also optionally be quaternized. The heterocyclo group may be attached at any heteroatom or carbon atom. [004 IJ Exemplary monocyclic heterocyclo groups include pyrrolidinyl, pyrrolyl, indolyl, pyrazolyl, oxetanyl, pyrazolinyl, imidazolyl, imidazolinyl, imidazolidinyl, oxazolyl, oxazolidinyl, isoxazolinyl, isoxazolyl, thiazolyl, thiadiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, furyl, tetrahydrofiiryϊ, thienyl, oxadiazolyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, 2-oxazepinyI, azepinyl, 4-piperidonyl, pyrldyl. N-oxo-pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, tetrahydrothiopyranyl, tetrahydropyranyl, morpholϊny!, thiamorpholinyl, thiamorpholinyl sulfoxide, tetrahydrothϊopyranylsulfone, thiamorpholinyl sulfone, 1,3-dioxolane, tetrahydro-l,l-dioxothienyl; dioxanyl, isothiazolidinyl, thietanyl, thiiranyl, triazinyl, triazolyl, and the like. [0042] Exemplary bicyclic heterocyclo groups include benzothiazolyl, benzoxazolyl, benzothienyl, quinolinyl, quinolinyl-N-oxide, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyi, benzofuryl, chromonyl, coumarinyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl (such as fijro[2,3-c]pyridinyl, furo[3,l-b]pyridinyl or furo[2,3~b]pyridinyl), dihydroisoindolyl, dihydroquinazolinyl (such as 3,4~dihydro-4-oxo-quinazolinyl), benzisothiazolyl, benzisoxazolyl, benzodiazinyl, benzofurazanyl, benzothiopyranyl, benzotriazolyl, benzpyrazolyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, dihydrobenzopyranyl, indolinyl, isochroraanyl, isoindolinyl, naphthyridinyl, phthalazinyl, piperonyl, purinyl, pyridopyridyl, quinazolinyl, tetrahydroquinolinyl, thienofuryl, thienopyridyl, thienothienyl, and the like. [0043] When a group is referred to as being optionally substituted, it may be substituted with one to five, preferably one to three, substituents such as F, Cl, Br, I, trifluoromethyl, trifluoromethoxy, hydroxy, lower alkoxy, cycloaϊkoxy, heterocyclooxy, oxo, lower alkanoyl, aryloxy, lower alkanoyloxy, amino, lower alkylamino, arylamino, aralkylamino, cycloalkylamino, heterocycloamino, disubstituted amines in which the two amino substituents independently are selected from lower alkyl, aryl or aralkyl, lower alkanoylamino, aroylamino, aralkanoylamino, substituted lower alkanoylamino, substituted arylamino, substituted aralkylanoylamino, thiol, lower alkylthio, arylthio, aralkylthio, cycloalkylthio, heterocyclothio, lower alkylthiono, arylthiono, aralkylthiono, lower alkylsulfonyl, arylsulfonyl, aralkylsulfonyl, sulfonamide (e.g., SO2NH2), substituted sulfonamide, nitro, cyano, carboxy, carbamyl (e.g., CONH2), substituted carbamyl (e.g., CONH- lower alkyl, CONH-aryl, CONH-aralkyl or cases where there are two substituents on the nitrogen independently selected from lower alkyl, aryl or aralkyl), lower alkoxycarbonyl, aryl, substituted aryl, guanidino, and heterocyclos (e.g., indolyl, imidazoiyl, fiiryl, thienyl, thiazolyl, pyrrolidyl, pyridyl, pyrimidyl and the like). Where noted above that the substituent is further substituted, it will be substituted with F, Cl, Br, I, optionally substituted lower alkyl, hydroxy, optionally substituted lower alkoxy, optionally substituted aryl, or optionally substituted aralkyl. [0044] Al! stereoisomers of the Formula I, II, III and IV compounds of the instant invention are contemplated, either in admixture or in pure or substantially pure form. The definition of the formula I compounds embraces all possible stereoisomers and their mixtures. The Formula 1, II, IH and IV definitions very particularly embrace the racemic forms and the isolated optical isomers having the specified activity. [0045] A particularly preferred epothilone analog for use in the methods of the invention is Compound 1: [lS-[lR*,3R*(E),7R*,10SMlRM2RM6S*]]-7,π- dihydroxy-8, 8, 10, 12, 16-pentamethyl-3-[ 1 -methyl-2-(2-methyl-4-thiazolyl)ethenyl] -4- aza-17-oxabicyclo[14.1.0]heptadecane-5,9-dione. Another exemplary epothilone is [lS-[lR*,3R*(E),7R*.10S*,HR*,12R*,I6S*]3-3-[2-[2-(aminomethyl)-4-11iiazolyl]-l- methylethenyl] -7, 11 -dihydroxy-8 ,8, 10,12,16-pentamethyl-4517- dioxabicycIo[14.1.0]heptadecane-5,9-dione., Compound 4. [0046] Compound I5 an exemplary epothilone analog of the invention, is a semisynthetic epothilone analog and has a mode of action analogous to paclitaxel (i.e., microtubule stabilization). However, in preclinical pharmacology studies, Compound 1 has demonstrated significant improvement over paclitaxel in several critical aspects. Compound 1 exhibits a very impressive and broad spectrum of antitumor activity against paclitaxel-sensitive (A2780, HCTl 16 and LS174T) and, more importantly, as well as paclitaxel-resistant human colon tumors (HCTl 16/VM46), ovarian carcinoma (Pat-7 and A2780Tax) and breast carcinoma (Pat-21) models. Compound 1 is orally efficacious; the antitumor activity produced after oral administration is comparable to that produced by parenteral administration of the drug. These preclinical efficacy data indicate that Compound 1 demonstrates improved clinical efficacy in TAXOL®- insensitive and sensitive disease types.
[0047] Compound 2: (R)-2?3,4,5-tetrahydro-l-(lH-imidazol-4-ylmethyl)-3- (phenyImethyl)-4-(2-thienylsulfonyl)-lH-l,4-benzodiazepine-7-carbonitriles hydrochloride salt. [0048] Compound 3: A CDK inhibitor is shown below
Figure imgf000024_0001
[0049] Compound 4: lS-[lR*,3R*(E),7R'f,10S*,l lR*>12RM6S:l:]]-3-[2-[2- (AminomethyO^-thiazolyll-l-methylethenylJ-TJ l-dihydroxy-δ^JO,^,^- pentamethyl-4 , 17-dioxabicyclo [ 14, 1.0] heptadecane-5 ,9-dione . [0050] Compound 5: N-[5-[[[5'(l5l-Diraethylelhyl)-2-oxazolyI]methyl]thio]-2- thiazolyl]-4~piperidinecarboxamide.
[0051] Combinations of a microtubulin-stabilizing agent with another agent is contemplated by the present invention, and may include the addition of an antiproliferative cytotoxic agent. Classes of compounds that may be used as anti- proliferative cytotoxic agents include the following: co-stimulatory modulating agents including, without limitation, CTLA4 antagonists, ipϊlimumab, agatolimod, belatacept, blϊnatumomab, CD40 ligand, anti- B7-1 antibody, anti-B7-2 antibody, anti-B7-H4 antibody, AG4263, eritoran, anti- OX40 antibody, ISF-154, and SGN-70; alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes): Uracil mustard, Chlormethine, Cyclophosphamide (CYTOXAN®), Ifosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylene-melamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Streptozocin, Dacarbazme, and Temozolomide; antimetabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors): Methotrexate, 5-Fluorouracil, Floxuridine, Cytarabine, 6-Mercaptopurine, 6- Thioguanine, Fludarabine phosphate, Pentostatine, and Gemcitabine; and natural products and their derivatives (for example, vinca alkaloids, antitumor antibiotics, enzymes, lyniphokines and epipodophyllotoxins): Vinblastine, Vincristines Vindesine, Bleomycin, Dactinomycin, Daunorubicin, Doxorubicin, Epirubicin, Idambicin, Ara-C, paclitaxel (paclitaxel is commercially available as TAXOL®), Mithramycin, Deoxyco-formycin, Mitomycin-C, L-Asparaginase, Interferons (especially IFN-a), Etoposide, and Tenyposide.
[0052] Other anti-proliferative cytotoxic agents contemplated by the present invention are navelbene, CPT-I l, anastrazole, letrazole, capecitabine, reloxafine, cyclophosphamide, ifosamide, and droloxafme.
[0053] Suitable anti-CTLA4 antagonist agents for use in the methods of the invention, include, without limitation, anti~CTLA4 antibodies, human anti-CTLA4 antibodies, mouse anti-CTLA4 antibodies, mammalian anti-CTLA4 antibodies, humanized anti-CTLA4 antibodies, monoclonal anti-CTLA4 antibodies, polyclonal anti-CTLA4 antibodies, chimeric anti-CTLA4 antibodies, MDX-010 (ipilimumab), tremelimumab, antϊ-CD28 antibodies, anti-CTLA4 adnectins, anti-CTLA4 domain antibodies, single chain anti-CTLA4 fragments, heavy chain anti-CTLA4 fragments, light chain anti-CTLA4 fragments, inhibitors of CTLA4 that agonize the co- stimuiatory pathway, the antibodies disclosed in PCT Publication No. WO 2001/014424, the antibodies disclosed in PCT Publication No. WO 2004/035607, the antibodies disclosed in U.S. Publication No. 2005/0201994, and the antibodies disclosed in granted European Patent No. EP1212422B1. Additional CTLA-4 antibodies are described in U.S. Patent Nos. 5,811,097, 5,855,887, 6,051,227, and 6,984,720; in PCT Publication Nos. WO 01/14424 and WO 00/37504; and in U.S. Publication Nos. 2002/0039581 and 2002/086014. Other anti-CTLA-4 antibodies that can be used in a method of the present invention include, for example, those disclosed in: WO 98/42752; U.S. Patent Nos. 6,682,736 and 6,207,156; Hurwitz et al., Proc. Natl. Acad. ScL USA, 95(17):10067-10071 (1998); Camacho et al.s J. Clin. Oncology, 22(145):Abstract No. 2505 (2004) (antibody CP-675206); Mokyr et al., Cancer Res, 58:5301-5304 (1998), U.S. Patent Nos. 5,977,318, 6,682,736, 7,109,003, and 7,132,281. [0054] Additional anti-CTLA4 antagonists include, but are not limited to, the following: any inhibitor that is capable of disrupting the ability of CD28 antigen to bind to its cognate ligand, to inhibit the ability of CTLA4 to bind to its cognate ligand, to augment T cell responses via the co-stimulatory pathway, to disrupt the ability of B7 to bind to CD28 and/or CTLA4, to disrupt the ability of B7 to activate the co- stimulatory pathway, to disrupt the ability of CD80 to bind to CD28 and/or CTLA4, to disrupt the ability of CD 80 to activate the co-stimulatory pathway, to disrupt the ability of CD86 to bind to CD28 and/or CTLA4, to disrupt the ability of CD86 to activate the co-stimu!atory pathway, and to disrupt the co-stimulatory pathway, in general from being activated. This necessarily includes small molecule inhibitors of CD28, CD80, CD86, CTLA4, among other members of the co-stimulatory pathway; antibodies directed to CD28, CD80, CD86, CTLA4, among other members of the co- stimulatory pathway; antisense molecules directed against CD28, CD80, CD86, CTLA4, among other members of the co-stimulatory pathway; adnectins directed against CD28, CD80, CD86, CTLA4, among other members of the co-stimulatory pathway, RNAi inhibitors (both single and double stranded) of CD28, CD80, CD86, CTLA4, among other members of the co-stimulatory pathway, among other anti- CTLA4 antagonists. [0055] As is known in the art, Ipilimumab refers to an anti-CTLA-4 antibody, and is a fully human IgGiM antibody derived from transgenic mice having human genes encoding heavy and light chains to generate a functional human repertoire. Ipilimumab can also be referred to by its CAS Registry No. 477202-00-9, and is disclosed as antibody IODI in PCT Publication No. WO01/14424, incorporated herein by reference in its entirety and for all purposes. Specifically, Ipilimumab describes a human monoclonal antibody or antigen-binding portion thereof that specifically binds to CTLA4, comprising a light chain variable region and a heavy chain variable region having a light chain variable region comprised of SEQ ID NO:5, and comprising a heavy chain region comprised of SEQ ID NO:6. Pharmaceutical compositions of Ipilimumab include all pharmaceutically acceptable compositions comprising Ipilimumab and one or more diluents, vehicles and/or excipients. Examples of a pharmaceutical composition comprising Ipilimumab are provided in PCT Publication No. WO2007/67959. Impiϊimumab may be administered by LV. Light Chain Variable Region for lmpilimumab:
EIVLTQSPGTLSLSPGERATLSCRASQSVGSSYLA WYQQKPGQAPRLLIYGAFS RATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYGSSPWTFGQGTKVEIK (SEQ ID NO: 1)
Heavy Chain Variable Region for Impiliraumab:
QVQLVESGGGWQPGRSLRLSCAASGFTFSSYTMHWVRQAPGKGLEWVTFIS
YDGNNKYΎADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAΓYYCARTGWLG PFDYWGQGTLVTVSS (SEQ ID NO:2)
[0056] As noted elsewhere herein, the administration of one or more anti-CTLA4 antagonists may be administered either alone or in combination with a peptide antigen (e.g., gplOO), in addition to an antiproliferative agent disclosed herein. A non- limiting example of a peptide antigen would be a gplOO peptide comprising, or alternatively consisting of, the sequence selected from the group consisting of: IMDQVPFSV (SEQ ID NO:3), and YLEPGPVTV (SEQ ID NO:4). Such a peptide may be administered orally, or preferably by injection sx. at I mg emulsified in incomplete Freund's adjuvant (IFA) injected s.c. in one extremity, and 1 mg of either the same or a different peptide emulsified in IFA may be injected in another extremity.
[0057] The present invention also encompasses a pharmaceutical composition useful in the treatment of cancer, comprising the administration of a therapeutically effective amount of a microtubulin-stabilizing agent, either alone or in combination with another agent, with or without pharmaceutically acceptable carriers or diluents. The compositions of the present invention may further comprise one or more pharmaceutically acceptable additional ingredient(s) such as alum, stabilizers, antimicrobial agents, buffers, coloring agents, flavoring agents, adjuvants, and the like. The Formula I, H5 HI, and/or IHa, or analogs thereof compounds, CTLA4 antagonist agents, antineoplastic agents, and compositions of the present invention may be administered orally or parenterally including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration. [0058] For oral use, the antineoplastic agents, Formulas I, π, UI, and/or HIa or analogs thereof compounds and compositions of this invention may be administered, for example, in the form of tablets or capsules, powders, dispersible granules, or cachets, or as aqueous solutions or suspensions. In the case of tablets for oral use, carriers which are commonly used include lactose, corn starch, magnesium carbonate, talc, and sugar, and lubricating agents such as magnesium stearate are commonly added. For oral administration in capsule form, useful carriers include lactose, corn starch, magnesium carbonate, talc, and sugar. When aqueous suspensions are used for oral administration, emulsifying and/or suspending agents are commonly added. [0059] In addition, sweetening and/or flavoring agents may be added to the oral compositions. For intramuscular, intraperitoneal, subcutaneous and intravenous use, sterile solutions of the active ingredient(s) are usually employed, and the pH of the solutions should be suitably adjusted and buffered. For intravenous use, the total concentration of the solute(s) should be control !ed in order to render the preparation isotonic.
[0060] For preparing suppositories according to the invention, a low melting wax such as a mixture of fatty acid glycerides or cocoa butter is first melted, and the active ingredient is dispersed homogeneously in the wax, for example by stirring. The molten homogeneous mixture is then poured into conveniently sized molds and allowed to cool and thereby solidify.
[0061] Liquid preparations include solutions, suspensions and emulsions. Such preparations are exemplified by water or water/propylene glycol solutions for parenteral injection. Liquid preparations may also include solutions for intranasal administration. [0062] Aerosol preparations suitable for inhalation may include solutions and solids in powder form, which may be in combination with a pharmaceutically acceptable carrier, such as an inert compressed gas.
[0063] Also included are solid preparations which are intended for conversion, shortly before use, to liquid preparations for either oral or parenteral administration. Such liquid forms include solutions, suspensions and emulsions.
[0064] The compounds of Formulas I, II, IB, and/or Ilia or analogs thereof, as well as the anti-CTLA4 agents and antineoplastic agents, described herein may also be delivered transdermally. The transdermal compositions can take the form of creams, lotions, aerosols and/or emulsions and can be included in a transdermal patch of the matrix or reservoir type as are conventional in the art for this purpose. [0065] The combinations of the present invention may also be used in conjunction with other well known therapies that are selected for their particular usefulness against the condition that is being treated.
[0066] If formulated as a fixed dose, the active ingredient(s) of the microtubulin- stabilizing agents, or combination compositions, of this invention are employed within the dosage ranges described below. Alternatively, the anti-CTLA4 agent, and Formulas I, II, 133, and/or Ilia or analogs thereof compounds may be administered separately in the dosage ranges described below. In a preferred embodiment of the present invention, the anti-CTLA4 agent is administered in the dosage range described below following or simultaneously with administration of the Formulas I, II, III, and/or Ilia or analogs thereof compound in the dosage range described below. [0067] The following sets forth preferred therapeutic combinations and exemplary dosages for use in the methods of the present invention. Where "Compound of Formula II" appears, any of the variations of Formula II or Formula III set forth herein are contemplated for use in the chemotherapeutϊc combinations. Preferably, Compound 1 or Compound 4 is employed.
Figure imgf000029_0001
[0068] While this table provides exemplary dosage ranges of the Formula I, Formula O, Formula III and Formula Ma compounds and certain anticancer agents of the invention, when formulating the pharmaceutical compositions of the invention the clinician may utilize preferred dosages as warranted by the condition of the patient being treated. For example, the compound of Formula I may preferably be administered at about 40 mg/m2 every 3 weeks. Compound I may preferably be administered at about 25-60 mg/m2 every 3 weeks. Compound 2, may preferably be administered at a dosage ranging from about 25-500 mg/m2 every three weeks for as long as treatment is required. [0069] The anti-CTLA4 antibody may preferably be administered at about 0.3 - 10 mg/kg, or the maximum tolerated dose. In an embodiment of the invention, a dosage of CTLA-4 antibody is administered about every three weeks. Alternatively, the CTLA-4 antibody may be administered by an escalating dosage regimen including administering a first dosage of CTLA-4 antibody at about 3 mg/kg, a second dosage of CTLA-4 antibody at about 5 mg/kg, and a third dosage of CTLA-4 antibody at about 9 mg/kg.
[0070] In another specific embodiment, the escalating dosage regimen includes administering a first dosage of CTLA-4 antibody at about 5 mg/kg and a second dosage of CTLA-4 antibody at about 9 mg/kg. [0071] Further, the present invention provides an escalating dosage regimen, which includes administering an increasing dosage of CTLA-4 antibody about every six weeks.
[0072] In an aspect of the present invention, a stepwise escalating dosage regimen is provided, which includes administering a first CTLA-4 antibody dosage of about 3 mg/kg, a second CTLA-4 antibody dosage of about 3 mg/kg, a third CTLA-4 antibody dosage of about 5 mg/kg, a fourth CTLA-4 antibody dosage of about 5 mg/kg, and a fifth CTLA-4 antibody dosage of about 9 mg/kg. In another aspect of the present invention, a stepwise escalating dosage regimen is provided, which includes administering a first dosage of 5 mg/kg, a second dosage of 5 mg/kg, and a third dosage of 9 mg/kg.
[0073] The actual dosage employed may be varied depending upon the requirements of the patient and the severity of the condition being treated. Determination of the proper dosage for a particular situation is within the skill of the art. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small amounts until the optimum effect under the circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day if desired. Intermittent therapy (e.g., one week out of three weeks or three out of four weeks) may also be used.
[0074] Certain cancers can be treated effectively with compounds of Formulas I, II, III, and/or IEEa and a one or more anti-CTLA4 agents. Such triple and quadruple combinations can provide greater efficacy. When used in such triple and quadruple combinations the dosages set forth above can be utilized.
[0Θ75] When employing the methods or compositions of the present invention, other agents used in the modulation of tumor growth or metastasis in a clinical setting, such as antiemetics, can also be administered as desired. [0076] The present invention encompasses a method for the synergistic treatment of cancer wherein anti-CTLA4 agent and a Formulas I, II, ITI, and/or Ilia compound are administered simultaneously or sequentially. Thus, while a pharmaceutical formulation comprising anti-CTLA4 agent(s) and a Formulas I, H, III, and/or EUa compound may be advantageous for administering the combination for one particular treatment, prior administration of the anti-CTLA4 agent(s) may be advantageous in another treatment. It is also understood that the instant combination of anti-CTLA4 agent(s) and Formulas I, II, III, and/or Ilia compound may be used in conjunction with other methods of treating cancer (preferably cancerous tumors) including, but not limited to, radiation therapy and surgery. It is further understood that a cytostatic or quiescent agent, if any, may be administered sequentially or simultaneously with any or all of the other synergistic therapies.
[0077] The combinations of the instant invention may also be co-administered with other well known therapeutic agents that are selected for their particular usefulness against the condition that is being treated. Combinations of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when a multiple combination formulation is inappropriate. [0078] The chemotherapeutic agent(s) and/or radiation therapy can be administered according to therapeutic protocols well known in the art. It will be apparent to those skilled in the art that the administration of the chemotherapeutic agent(s) and/or radiation therapy can be varied depending on the disease being treated and the known effects of the chemotherapeutic agent(s) and/or radiation therapy on that disease. Also, in accordance with the knowledge of the skilled clinician, the therapeutic protocols (e.g., dosage amounts and times of administration) can be varied in view of the observed effects of the administered therapeutic agents (i.e., anti- CTLA4 agent(s)) on the patient, and in view of the observed responses of the disease to the administered therapeutic agents.
[0079J In the methods of this invention, a compound of Formula I, π, ID or Formula ma is administered simultaneously or sequentially with an anti-CTLA4 agent. Thus, it is not necessary that the anti-CTLA4 therapeutic agent(s) and compound of Formulas I, II, HE, and/or IHa, be administered simultaneously or essentially simultaneously. The advantage of a simultaneous or essentially simultaneous administration is well within the determination of the skilled clinician. [0080] Also, in general, the compound of Formulas I, II, IH, and/or HIa, and anti- CTLA4 agent(s) do not have to be administered in the same pharmaceutical composition, and may, because of different physical and chemical characteristics, have to be administered by different routes. For example, the compound of Formula I, π, HI, or IV may be administered intravenously to generate and maintain good blood levels thereof, while the anti-CTLA4 agent(s) may also be administered intravenously. Alternatively, the compound of Formula I, II, HI, or IV may be administered orally to generate and maintain good blood levels thereof, while the anti-CTLA4 agent(s) may also be administered intravenously. Alternatively, the compound of Formula I, H, IH, or rV may be administered intravenously to generate and maintain good blood levels thereof, while the anti-CTLA4 agent(s) may also be administered orally. The determination of the mode of administration and the advisability of administration, where possible, in the same pharmaceutical composition, is well within the knowledge of the skilled clinician. The initial administration can be made according to established protocols known in the art, and then, based upon the observed effects, the dosage, modes of administration and times of administration can be modified by the skilled clinician.
[0081] The particular choice of compound of Formulas I, II, III, and/or Ola or analogs thereof and anti-CTLA4 agent(s) will depend upon the diagnosis of the attending physicians and their judgment of the condition of the patient and the appropriate treatment protocol.
[0082] If the compound of Formula I, II, III, and/or πia and the anti-CTLA4 agent(s) are not administered simultaneously or essentially simultaneously, then the initial order of administration of the compound of Formulas I, π, III, and/or Ilia, and the anti-CTLA4 agent(s) may be varied. Thus, for example, the compound of Formulas I, II, in, and/or IUa or analogs thereof may be administered first followed by the administration of the anti-CTLA4 agent(s); or the anti-CTLA4 agent(s) may be administered first followed by the administration of the compound of Formulas I, II, III, and/or Ilia. This alternate administration may be repeated during a single treatment protocol. The determination of the order of administration, and the number of repetitions of administration of each therapeutic agent during a treatment protocol, is well within the knowledge of the skilled physician after evaluation of the disease being treated and the condition of the patient. For example, the aπtϊ-CTLA4 agent(s) may be administered initially. The treatment is then continued with the administration of the compound of Formulas I, II, III, and/or IHa or analogs thereof and optionally followed by administration of a cytostatic agent, if desired, until the treatment protocol is complete. Alternatively, the administration of the compound of Formulas I, II, III, and/or Ilia or analogs thereof and optionally followed by administration of a cytostatic agent may be administered initially. The treatment is then continued with the administration of the anti-CTLA4 agent(s), until the treatment protocol is complete.
[0083] Thus, in accordance with experience and knowledge, the practicing physician can modify each protocol for the administration of a component (therapeutic agent - i.e., compound of Formulas I, π, III, and/or Ilia or analogs thereof, anti-CTLA4 agent(s)) of the treatment according to the individual patient's needs, as the treatment proceeds. [0084] The attending clinician, in judging whether treatment is effective at the dosage administered, will consider the general well-being of the patient as well as more definite signs such as relief of disease-related symptoms, inhibition of tumor growth, actual shrinkage of the tumor, or inhibition of metastasis. Size of the tumor can be measured by standard methods such as radiological studies, e.g., CAT or MRI scan, and successive measurements can be used to judge whether or not growth of the tumor has been retarded or even reversed. Relief of disease-related symptoms such as pain, and improvement in overall condition can also be used to help judge effectiveness of treatment. [0085] Thus, the present invention provides methods for the treatment of a variety of cancers, including, but not limited to, the following: carcinoma including that of the bladder (including accelerated and metastatic bladder cancer), breast, colon (including colorectal cancer), kidney, liver, lung (including small and non-small cell lung cancer and lung adenocarcinoma), ovary, prostate, testes, genitourinary tract, lymphatic system, rectum, larynx, pancreas (including exocrine pancreatic carcinoma), esophagus, stomach, gall bladder, cervix, thyroid, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-ceϊl lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, histiocytic lymphoma, and Burketts lymphoma; hematopoietic tumors of myeloid lineage including acute and chronic myelogenous leukemias, myelodysplastic syndrome, myeloid leukemia, and promyelocytic leukemia; tumors of the central and peripheral nervous system including astrocytoma, neuroblastoma, glioma, and schwannomas; tumors of mesenchymal origin including fibrosarcoma, rhabdomyoscarcoma, and osteosarcoma; other tumors including melanoma, xenoderma pigmentosum, keratoactanthoma, seminoma, thyroid follicular cancer, and teratocarcinoma; melanoma, unresectable stage III or IV malignant melanoma, squamous cell carcinoma, small-cell lung cancer, non-small cell lung cancer, glioma, gastrointestinal cancer, renal cancer, ovarian cancer, liver cancer, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, stomach cancer, bladder cancer, hepatoma, breast cancer, colon carcinoma, and head and neck cancer, gastric cancer, genu cell tumor, bone cancer, bone tumors, adult malignant fibrous histiocytoma of bone; childhood malignant fibrous histiocytoma of bone, sarcoma, pediatric sarcoma, sinonasal natural killer, neoplasms, plasma cell neoplasm; myelodysplastic syndromes; neuroblastoma; testicular germ cell tumor, intraocular melanoma, myelodysplastic syndromes; myelodysplastic/myeloproliferative diseases, synovial sarcoma, chronic myeloid leukemia, acute lymphoblastic leukemia, Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ ALL), multiple myeloma, acute myelogenous leukemia, chronic lymphocytic leukemia, mastocytosis and any symptom associated with mastocytosis, and any metastasis thereof. In addition, disorders include urticaria pigmentosa, mastocytosises such as diffuse cutaneous mastocytosis, solitary mastocytoma in human, as well as dog mastocytoma and some rare subtypes like bullous, erythrodermic and teleangiectatϊc mastocytosis, mastocytosis with an associated hematological disorder, such as a myeloproliferative or myelodysplastic syndrome, or acute leukemia, myeloproliferative disorder associated with mastocytosis, mast cell leukemia,, in addition to other cancers. Other cancers are also included within the scope of disorders including, but are not limited to, the following: carcinoma, including that of the bladder, urothelial carcinoma, breast, colon, kidney, liver, lung, ovary, pancreas, stomach, cervix, thyroid, testis, particularly testicular seminomas, and skin; including squamous cell carcinoma; gastrointestinal stromal tumors ("GIST"); hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B- cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma and Burketts lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias and promyelocytic leukemia; tumors of mesenchymal origin, including fibrosarcoma and rhabdomyoscarcoma; other tumors, including melanoma, seminoma, tetratocarcinoma, neuroblastoma and glioma; tumors of the central and peripheral nervous system, including astrocytoma, neuroblastoma, glioma, and schwannomas; tumors of mesenchymal origin, including fibrosarcoma, rhabdomyoscaroma, and osteosarcoma; and other tumors, including melanoma, xenoderma pigmentosum, keratoactanthoma, seminoma, thyroid follicular cancer, teratocarcinoma, chemotherapy refractory non-seminomatous germ-cell tumors, and Kaposi's sarcoma, and any metastasis thereof.
[0086] Most preferably, the invention is used to treat accelerated or metastatic cancers of the breast and/or lung.
BIOMARKERS AND BIOMARKER SETS
[0087] The invention includes individual biomarkers and biomarker sets having both diagnostic and prognostic value in proliferative disease areas in which microtubulin status is of importance, e.g.', in cancers or tumors, or in disease states in which cell signaling and/or cellular proliferation controls are abnormal or aberrant. The biomarker sets comprise a plurality of biomarkers that highly correlate with resistance or sensitivity to one or more microtubulin-stabilizing agents. [0088] The biomarkers and biomarker sets of the invention enable one to predict or reasonably foretell the likely effect of one or more microtubulin-stabilizing agents in different biological systems or for cellular responses merely based upon whether one or more of the biomarkers of the present invention are overexpressed relative to normal. The biomarkers and biomarker sets can be used in in vitro assays of cellular proliferation by sample cells to predict in vivo outcome. In accordance with the invention, the various biomarkers and biomarker sets described herein, or the combination of these biomarker sets with other biomarkers or markers, can be used, for example, to predict and monitor how patients with cancer might respond to therapeutic intervention with one or more microtubulin-stabilizing agents. [0089] In specific embodiments of the present invention, overexpression of TUBB3, BCRP (also referred to as ABCG2), MDRl (also referred to as ABCBl), MRPl (also referred to as ABCCl), and/or β-tubulin mutations correlated with response to microtubulin-stabiiizing agents. Specifically, overexpression of TUBB3 resulted in xenografts that were resistant to docetaxel and vinorelbine, yielding antitumor efficacy ranging 0.2-0.9 and 0.1-0.3 LCK, respectively. In contrast, ixabepilone was active in all 4 tumors in which TUBB3 was overexpressed, yielding 1.6-4.2 LCK (see Table 1 ) when tested at their maximum tolerated doses (MTD). The BCRP overexpressing HEK293/BCRP cell line demonstrated resistance to paclitaxel and mitoxantrone by 9.8-fold (IC50=8.7 nM) and 4.1-fold (IC50=26.4 nM), respectively, in comparison with the vector-transfected control line. This resistance could be reversed by fumitremorgin C, a selective inhibitor of BCRP. In contrast, ixabepϊlone was far less susceptible to the BCRP-mediated resistance, resulting in a resistance factor of only 1.9 fold (IC50=4.1 nM). Together, these results suggest ixabepilone may offer breast and/or lung cancer patients a potentially valuable treatment option.
[0090] Measuring the level of expression of a biomarker and biomarker set provides a useful tool for screening one or more tumor samples before treatment of a patient with the microtubulin-stabilizing agents. The screening allows a prediction of whether the cells of a tumor sample will respond favorably to the microtubulin- stabilizing agents, based on the presence or absence of over-expression - such a prediction provides a reasoned assessment as to whether or not the tumor, and hence a patient harboring the tumor, will or will not respond to treatment with the microtubulin-stabilizing agents. [009 ϊ] A difference in the level of the biomarker that is sufficient to indicate whether the mammal will or will not respond therapeutically to the method of treating cancer can be readily determined by one of skill in the art using known techniques. The increase or decrease in the level of the biomarker can be correlated to determine whether the difference is sufficient to identify a mammal that will respond therapeutically. The difference in the level of the biomarker that is sufficient can, in one aspect, be predetermined prior to determining whether the mammal will respond therapeutically to the treatment. In one aspect, the difference in the level of the biomarker is a difference in the mRNA level (measured, for example, by RT-PCR or a microarray), such as at least about a two-fold difference, at least about a three-fold difference, or at least about a four-fold difference in the level of expression, or more. In another aspect, the difference in the level of the biomarker is determined at the protein level by mass spectral methods or by FISH or by IHC. In another aspect, the difference in the level of the biomarker refers to a p-value of <0.05 in Anova analysis. In yet another aspect, the difference is determined in an ELISA assay. [0092] The biomarker or biomarker set can also be used as described herein for monitoring the progress of disease treatment or therapy in those patients undergoing treatment for a disease involving a microtubulin-stabilizing agent. [0093] The biomarkers also serve as targets for the development of therapies for disease treatment. Such targets may be particularly applicable to treatment of cancer, such as, for example, breast and/or lung cancer.
[0094] Indeed, because these biomarkers are differentially expressed in sensitive and resistant cells, their expression patterns are correlated with relative intrinsic sensitivity of cells to treatment with microtubulin-stabilizing agents. Accordingly, the biomarkers over expressed in resistant cells may serve as targets for the development of new therapies for the tumors which are resistant to microtubulin-stabilizing agents. The level of biomarker protein and/or mRNA can be determined using methods well known to those skilled in the art. For example, quantification of protein can be carried out using methods such as ELISA, 2-dimensional SDS PAGE5 Western blot, immunoprecipitation, immunohistochemistry, fluorescence activated cell sorting (FACS), or flow cytometry. Quantification of mRNA can be carried out using methods such as PCR5 array hybridization, Northern blot, in~situ hybridization, dot- blot, TAQMAN®, or RNAse protection assay.
[0095] The present invention encompasses the use of any one or more of the following as a biomarker for use in predicting microtubulin-stabilizing agent response: TUBB3, BRCP, MDRl, MRPl, and beta-tubulin mutations. [0096 J The present invention also encompasses any combination of the aforementioned biomarkers, including, but not limited to: TUBB3, BRCP, MDRl, MRPl, and beta-tubulin mutations; TUBB3, BRCP, MDRl, MEPl; TUBB3, BRCP, MDRl; BRCP, MDRl, MEPl, and beta-tubulin mutations; BRCP, MDRl, MRPl; MDRl, MRPl5 and beta-tubulin mutations; TUBB3 and BRCP; TUBB3 and MDRl; TUBB3 and MRPl; TUBB3 and beta-tubulin mutations; BRCP and MDRl; BRCP and MRPl; BRCP and beta-tubulin mutations; MDRl and MRPl; MDRl and beta- tubulin mutations; and/or MRPl and beta-tubulin mutations.
[0097] Identification of biomarkers that provide rapid and accessible readouts of efficacy, drug exposure, or clinical response is increasingly important in the clinical development of drug candidates. Embodiments of the invention include measuring changes in the levels of mRNA and/or protein in a sample to determine whether said sample contains increased expression of TUBB 3, BRCP, MDRl, MPRl, and/or beta- tubulin mutations. In one aspect, said samples serve as surrogate tissue for biomarker analysis. These biomarkers can be employed for predicting and monitoring response to one or more microtubulin-stabilizing agents. In one aspect, the biomarkers of the invention are one or more of the following: TUBB3, BRCP9 MDRl, MPRl, and/or beta-tubulin mutations, including both polynucleotide and polypeptide sequences. In another aspect, the biomarkers of the invention are nucleotide sequences that, due to the degeneracy of the genetic code, encodes for a polypeptide sequence provided in the sequence listing.
[0098] The biomarkers serve as useful molecular tools for predicting and monitoring response to microtubulin-stabilizing agents. [0099] Methods of measuring the level of any given marker described herein may be performed using methods well known in the art, which include, but are not limited to PCR; RT-PCR; FISH; IHC; immuno-detection methods; immunoprecipitation; Western Blots; ELISA; radioimmunoassays; PET imaging; HPLC; surface plasmon resonance, and optical spectroscopy; and mass spectrometry, among others. [00100] The biomarkers of the invention may be quantified using any immunospecific binding method known in the art. The immunoassays which can be used include but are not limited to competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, to name but a few. Such assays are routine and well known in the art (see, e.g., Ausubel et al., eds., Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York (1994), which is incorporated by reference herein in its entirety). Exemplary immunoassays are described briefly below (but are not intended by way of limitation). [00101] Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X- 100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2, 1% TRASYLOL®) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding the antibody of interest (i.e., one directed to a biomarker of the present invention) to the cell lysate, incubating for a period of time (e.g., 1-4 hours) at 4° C, adding protein A and/or protein G SEPHAROSE® beads to the cell lysate, incubating for about an hour or more at 4° C, washing the beads in lysis buffer and resuspending the beads in SDS/sample buffer. The ability of the antibody of interest to immunoprecipitate a particular antigen can be assessed by, e.g., western blot analysis. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the antibody to an antigen and decrease the background (e.g., pre-clearing the cell lysate with SEPHAROSE® beads). For further discussion regarding immunoprecipitation protocols see, e.g., Ausubel et al., eds., Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.16.1 (1994).
[00102 J Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%- 20% SDS- PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or nonfat milk), washing the membrane in washing buffer (e.g., PBS-Tween 20), blocking the membrane with primary antibody (the antibody of interest) diluted in blocking buffer, washing the membrane in washing buffer, blocking the membrane with a secondary antibody (which recognizes the primary antibody, e.g., an anti-human antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., 32P or 1251) diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected and to reduce the background noise. For further discussion regarding western blot protocols see, e.g., Ausubel et al, eds., Current Protocols in Molecular Biology, Vol. I5 John Wiley & Sons, Inc., New York at 10.8.1 (1994).
[00103] ELISAs comprise preparing antigen, coating the well of a 96 well microtiter plate with the antigen, adding the antibody of interest conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the well and incubating for a period of time, and detecting the presence of the antigen. In ELISAs the antibody of interest does not have to be conjugated to a detectable compound; instead, a second antibody (which recognizes the antibody of interest) conjugated to a detectable compound may be added to the well. Further, instead of coating the well with the antigen, the antibody may be coated to the well. In this case, a second antibody conjugated to a detectable compound may be added following the addition of the antigen of interest to the coated well. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art. For further discussion regarding ELISAs see, e.g., Ausubel et al., eds., Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 11.2.1 (1994).
[00104] Alternatively, identifying the relative quantitation of the biomarker polypeptide(s) may be performed using tandem mass spectrometry; or single or multi dimensional high performance liquid chromatography coupled to tandem mass spectrometry. The method takes into account the fact that an increased number of fragments of an identified protein isolated using single or roulti dimensional high performance liquid chromatography coupled to tandem mass spectrometry directly correlates with the level of the protein present in the sample. Such methods are well known to those skilled in the art and described in numerous publications, for example, 2-D Proteome Analysis Protocols, AJ. Link, ed., Humana Press (1999), ISBN: 0896035247; Mass Spectrometry of Proteins and Peptides, J.R. Chapman, ed., Humana Press (2000), ISBN: 089603609X.
[00105] As used herein the terms "modulate" or "modulates" or "modulators" refer to an increase or decrease in the amount, quality or effect of a particular activity, or the level of DNA, RNA, or protein detected in a sample. [00106] In order to facilitate a further understanding of the invention, the following examples are presented primarily for the purpose of illustrating more specific details thereof. The scope of the invention should not be deemed limited by the examples, but to encompass the entire subject matter defined by the claims. EXAMPLE 1 - METHOD OF ASSESSING THE RESPONSE OF TUBB3 OVEREXPRESSING TUMOR XENOGRAFTS TO THE ADMINISTRATION
OF MICROTUBULIN STABILIZING AGENTS /JV VIVO Methods
[00107] Cancer cell lines overexpressing TUBB3 were evaluated in vivo in mice for sensitivity to ixabepilone, docetaxel and vinorelbine. These include DU4475 and PAT21 breast, as well as Hl 155 and LX-I lung cancer lines. BCRP overexpressing HEK-293 cell line was studied in vitro for sensitivity to ixabepilone, paclitaxel and mitoxantrone.
[00108] Compounds and reagents. Ixabepilone, docetaxel and mitoxantrone were solubilized in 100% DMSO at 10 mg/ml for in vitro studies. [00109] Cell culture: HEK and HEK/BCRP cells were maintained in RPMI-1640 (Gibco) supplemented with 10% heat-inactivated fetal bovine serum and 25 mM HEPES at 37°C/5% CO2.
[00110] Cell growth assays. Cell growth assays were conducted in 6-well culture plates. Cells were plated at a density of 4 x 104 cells/well overnight. Compounds were then added (total DMSO content not exceeding 0.1%). Cell growth was determined by the direct counting of cell number, following trypsinization, using a Coulter Channelyzer.
(00111] Determination of BCRP expression by flow cytometry.
[00112] Cells were acquired using CellQuest Pro on a FACSCalibur (BD) and analyzed using Flow Jo software.
[00113] Immunohistochemistry Methods. Tumors were fixed overnight in 10% neutral buffered formalin and processed for paraffin embedding. Specimens were sectioned at 5 microns. Immunohistochemistry was performed on sections utilizing the Dako Envision Mouse Polymer kit with an antibody against Class EEI Beta- Tubulin, clone TUJl (Covance #MMS-435P), and counterstained with Gill 2 Hematoxylin. Images were taken at 20Ox magnification using ImagePro Plus software on an Olympus BX-60 microscope.
Study Design [00114] In vivo efficacy of ixabepilone was evaluated in TUBB 3 overexpressing human breast (DU4475 and PAT21) and lung (Hl 155 and LX-I) tumor xenograft models
• Tumor response was determined by measurement of tumors by caliper twice a week
• Tumor weights were estimated using the formula
- Tumor weight = (length x width2) / 2
• Tumor response endpoint was expressed in terms of log cell kill (LCK) expressed as - LCK = (T-C) ÷ (3.32 X TVDT)
- T-C, the difference in the time (days) required for the treated tumors (T) to reach a predetermined target size compared with those of the control group (C)
- Tumor volume doubling time = TVDT : Median time (days) for control tumors to reach target size - Median time (days) for control tumors to reach half the target size
• Activity was defined as achievement of LCK > 1
• MTD - the dose level immediately above which excessive toxicity (i.e., more then one death) occurred * Statistical evaluations were performed using Gehan's generalized Wilcoxon test
Results
[00115] Efficacy evaluation in nude mice demonstrated that the 4 xenografts overexpressing TUBB3 were resistant to docetaxel and vinorelbine, yielding antitumor efficacy ranging 0.2-0.9 and 0.1-0.3 LCK3 respectively (see Fig. 5). In contrast, ixabepilone was active in all 4 tumors, yielding 1.6-4.2 LCK (Table 1) when tested at their maximum tolerated doses (MTD). The BCRP overexpressing HEK293/BCRP cell line demonstrated resistance to paclitaxel and mitoxantrone by 9.8-fold (ΪC50=8.7 nM) and 4.1-fold (IC50=26.4 nM), respectively, in comparison with the vector-transfected control line (see Fig. 6). This resistance can be reversed by fumitremorgϊn C, a selective inhibitor of BCRP (see Fig. 7). In contrast, ixabepilone was far less susceptible io the BCRP-mediated resistance, resulting in a resistance factor of only 1.9 fold (IC50=4.1 nM) (see Fig. 8).
Conclusion [00116] Compared with agents commonly used in breast cancer - paclitaxel, docetaxel, mitoxantrone, and vinorelbine - ixabepilone has markedly lower susceptibility to multiple resistance mechanisms that affect these agents (see Fig. 3). These include overexpression of TUBB3, BCRP (also referred to as ABCG2), MDRl (also referred to as ABCBl), and MRPl (also referred to as ABCCl), and β-tubulin mutations.
[00117] In vitro and in vivo models demonstrated that ixabepilone is minimally affected by overexpression of TUBB3, BCRP, MDRl and MRPl, and β-tubulin mutations.
[00118] Ixabepilone's clinical activity has been demonstrated in metastatic breast cancer patients who developed resistance to other chemotherapy regimens,, including anthracyclines and taxanes.
[00119] As a result of this activity, ixabepilone was recently FDA-approved in the United States combination with capecitabine for the treatment of patients with metastatic or locally advanced breast cancer after failure of an anthracycline and a taxane, and as monotherapy for the treatment of metastatic or locally advanced breast cancer in patients after failure of an anthracycline, a taxane, and capecitabine. [00120] The results from the present study add to accumulating data supporting the inclusion of ixabepilone as a key component of breast cancer treatment. Moreover, the results demonstrate the utility of diagnosing patients for the presence of BCRP- overexpression, in addition to MDRl, MRPl, other transporters, TUBB3 overexpressϊon, and tubulin mutations, who may benefit from the administration of ixabepilone in the efficacious treatment of cancer, including breast and lung cancer. [00121] TABLE 1
Comparison of the Antitumor Efficacy of Ixabepilone, Docetaxel and Vinorelbine in 4 Human Tumor Xenografts Overexpressing TUBB3
Figure imgf000045_0001
EXAMPLE 2 - METHOD TO ASSESS MULTIPLE RESISTANCE EXPRESSION PROFILE USING MRNA FROM TISSUE AND CELL
SOURCES
[00122] Total RNA may be purified using RNEASY® system (Qiagen, CA5 USA). Mixed Oligo-d(T)is primers may be used to generate single-stranded cDNAs using the SUPERSCRIPT® First-strand Synthesis kit (Invitrogen, CA, USA). Levels for each gene of interest and GAPDH transcripts may be analyzed using an Applied Biosystems 7900HT Sequence Detection System. Mixed primer/probe sets for each transcript of interest (TUBB3, catalog # Hs00964962^gl; BCRP, catalog # HsOOl 84979_ml; MDR-I9 catalog # HSOOl 8449 ljn I; MRP-I, catalog # Hs00219905_ml ; GAPDH, catalog # 4326317E) may be obtained from Applied Biosystems and used according to the manufacturer's instructions. [00123] Expression levels of transcripts of interest may then be normalized to endogenous GAPDH transcripts. Comparisons may be made between samples by ΔΔCt comparative analysis using manufacturer's software (Applied Biosystems). Briefly, ΔCT = (MDR CT)-(GAPDH CT); ΔΔCT = (ΔCTProbel - ΔCTProbe2); and Fold change = — 2 oΔΔC
[00124] The present invention is not to be limited in scope by the embodiments disclosed herein, which are intended as single illustrations of individual aspects of the invention, and any that are functionally equivalent are within the scope of the invention. Various modifications to the models and methods of the invention, in addition to those described herein, will become apparent to those skilled in the art from the foregoing description and teachings, and are similarly intended to fall within the scope of the invention. Such modifications or other embodiments can be practiced without departing from the true scope and spirit of the invention. [00125] The entire disclosure of each document cited (including patents, patent applications, journal articles, abstracts, laboratory manuals, books, GENBANK® Accession numbers, SWISS-PROT® Accession numbers, or other disclosures) in the Background of the Invention, Detailed Description, Brief Description of the Figures, and Examples is hereby incorporated herein by reference in their entirety. Further, the hard copy of the Sequence Listing submitted herewith, in addition to its corresponding Computer Readable Form, are incorporated herein by reference in their entireties.

Claims

WHAT IS CLAIMED IS:
1. A method for predicting the likelihood a patient will respond therapeutically to a cancer treatment comprising the administration of a microtubul in- stabilizing agent, wherein said prediction method comprises the steps of: (a) measuring the level of a biomarker in a sample from said patient; and
(b) comparing the level of said biomarker in said sample relative to a standard to permit assignment of said sample to either being a member of an overexpression positive class or an overexpression negative class, wherein an overexpression positive sample member indicates an increased likelihood said patient will respond therapeutically to said cancer treatment, wherein said cancer treatment is the administration of ixabepilone.
2. The method according to Claim 1, wherein said cancer is breast or lung cancer.
3. The method according to Claim 1, wherein said biomarker is selected from the group consisting of: TUBB3, BRCP, MDRl, MPRl, and beta-tubulin mutations.
4. The method according to Claim 3, wherein said method further comprises measuring the expression level of an additional biomarker selected from the group consisting of: TUBB3, BRCP, MDRl, MPRl, and beta-tubulin mutations.
5. A method for treating a patient with cancer comprising the steps of:
(a) measuring the level of a biomarker in a sample from said patient; and
(b) comparing the level of said biomarker in said sample relative to a standard to permit assignment of said sample to either being a member of an overexpression positive class or an overexpression negative class, wherein an overexpression positive sample member indicates an increased likelihood said patient will respond therapeutically to said cancer treatment, wherein said cancer treatment is the administration of ixabepilone.
6. The method according to Claim 5, wherein said cancer is breast or lung cancer.
7. The method according to Claim 5, wherein said biomarker is selected from the group consisting of: TUBB3, BRCP, MDRl, MPRl, and beta-tubulin mutations.
8. The method according to Claim 7, wherein said method further comprises measuring the expression level of an additional biomarker selected from the group consisting of: TUBB3, BRCP5 MDRl, MPRl, and beta-tubulin mutations.
9. A kit for use in treating a patient with cancer, comprising: (a) a means for determining whether a sample from said patient is positive for overexpression of a biomarker;
(b) a therapeutically effective amount of a ixabepilone, or a pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutically acceptable salt or hydrate or solvate thereof; and (c) instructions for use of said kit.
10. The kit according to Claim 9, wherein said cancer is breast or lung cancer.
11. The kit according to Claim 9, wherein said biomarker is selected from the group consisting of: TUBB3, BRCP, MDRl, MPRl, and beta-tubulin mutations.
12. The method according to Claim 11, wherein said method further comprises measuring the expression level of an additional biomarker selected from the group consisting of: TUBB3, BRCP, MDRl, MPRl, and beta-tubulin mutations.
B. The method according to Claims 1, 5, or 9, wherein said measurement is performed using a method selected from the group consisting of: (a) PCR; (b) RT- PCR; (c) FISH; (d) IHC; (e) immunodetection methods; (f) Western Blot; (g) ELISA; (h) radioimmuno assays; (i) immunoprecipitation ; (j) PET imaging; (k) HPLC; (1) surface plasmon resonance; (m) optical spectroscopy; and (i) mass spectrometry.
PCT/US2009/031052 2008-01-15 2009-01-15 Methods for treating cancer in patients having breast cancer resistance protein overexpression WO2009091859A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/812,777 US20110112155A1 (en) 2008-01-15 2009-01-15 Methods for treating cancer in patients having breast cancer resistance protein overexpression
EP09702397A EP2238454A1 (en) 2008-01-15 2009-01-15 Methods for treating cancer in patients having breast cancer resistance protein overexpression

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US2114908P 2008-01-15 2008-01-15
US61/021,149 2008-01-15

Publications (1)

Publication Number Publication Date
WO2009091859A1 true WO2009091859A1 (en) 2009-07-23

Family

ID=40527470

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/031052 WO2009091859A1 (en) 2008-01-15 2009-01-15 Methods for treating cancer in patients having breast cancer resistance protein overexpression

Country Status (3)

Country Link
US (1) US20110112155A1 (en)
EP (1) EP2238454A1 (en)
WO (1) WO2009091859A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115398011A (en) * 2020-01-31 2022-11-25 亚拉勒提治疗欧洲私人有限公司 Methods for predicting ixabepilone responsiveness in cancer patients

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2002153274A (en) * 2000-11-17 2002-05-28 Kainosu:Kk Kit for quantitatively measuring expression level of mdr1 gene
US20060121511A1 (en) * 2004-11-30 2006-06-08 Hyerim Lee Biomarkers and methods for determining sensitivity to microtubule-stabilizing agents
WO2007117439A2 (en) * 2006-03-31 2007-10-18 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to microtubule-stabilizing agents

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5569588A (en) * 1995-08-09 1996-10-29 The Regents Of The University Of California Methods for drug screening
US5972626A (en) * 1997-07-30 1999-10-26 University Of Massachusetts Cancer detection by centrosome abnormality

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2002153274A (en) * 2000-11-17 2002-05-28 Kainosu:Kk Kit for quantitatively measuring expression level of mdr1 gene
US20060121511A1 (en) * 2004-11-30 2006-06-08 Hyerim Lee Biomarkers and methods for determining sensitivity to microtubule-stabilizing agents
WO2007117439A2 (en) * 2006-03-31 2007-10-18 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to microtubule-stabilizing agents

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BASELGA J ET AL: "Predicting response to ixabepilone: genomics study in patients receiving single agent ixabepilone as neoadjuvant treatment for breast cancer (BC)", BREAST CANCER RESEARCH AND TREATMENT, SPRINGER, NEW YORK, NY, vol. 94, no. Suppl. 1, 1 January 2005 (2005-01-01), pages S31 - S32, XP002457557, ISSN: 0167-6806 *
DOYLE L A ET AL: "A MULTIDRUG RESISTANCE TRANSPORTER FROM HUMAN MCF-7 BREAST CANCER CELLS", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE, WASHINGTON, DC.; US, vol. 95, 1 December 1998 (1998-12-01), pages 15665 - 15670, XP002919721, ISSN: 0027-8424 *
FERLINI CRISTIANO ET AL: "Looking at drug resistance mechanisms for microtubule interacting drugs: Does TUBB3 work?", CURRENT CANCER DRUG TARGETS, BENTHAM SCIENCE PUBLISHERS, HILVERSUM, NL, vol. 7, no. 8, 1 December 2007 (2007-12-01), pages 704 - 712, XP009115400, ISSN: 1568-0096 *
PUSZTAI L: "Markers predicting clinical benefit in breast cancer from microtubule-targeting agents.", ANNALS OF ONCOLOGY : OFFICIAL JOURNAL OF THE EUROPEAN SOCIETY FOR MEDICAL ONCOLOGY / ESMO DEC 2007, vol. 18 Suppl 12, December 2007 (2007-12-01), pages xii15 - xii20, XP002524121, ISSN: 1569-8041 *

Also Published As

Publication number Publication date
EP2238454A1 (en) 2010-10-13
US20110112155A1 (en) 2011-05-12

Similar Documents

Publication Publication Date Title
US7932031B2 (en) Methods for determining sensitivity to microtubule-stabilizing agents comprising ixabepilone by measuring the level of estrogen receptor 1
AU2013226323B2 (en) Cancer patient selection for administration of Wnt signaling inhibitors using RNF43 mutation status
JP2008533053A (en) Histone deacetylase inhibitors sensitize cancer cells to epidermal growth factor receptor inhibitors
JP2010527232A (en) Biological markers predicting anticancer responses to epidermal growth factor receptor kinase inhibitors
WO2009015233A2 (en) Gefitinib sensitivity-related gene expression and products and methods related thereto
KR20140026537A (en) Biomarkers for hedgehog inhibitor therapy
JP5774473B2 (en) Irinotecan sensitivity determination method and use thereof
US20120220594A1 (en) Methods for treating cancer in patients having igf-1r inhibitor resistance
MX2015005441A (en) Synthetic lethality and the treatment of cancer.
EP1599606A1 (en) Protein tyrosine phosphatase - prl-1 a a marker and therapeutic target for pancreatic cancer
US20110166154A1 (en) Identification of predictive markers of response to dasatinib in human colon cancer
MX2014000661A (en) Synthetic lethality and the treatment of cancer.
US20110112155A1 (en) Methods for treating cancer in patients having breast cancer resistance protein overexpression
US20060204980A1 (en) Colorectal cancer therapies
EP3696545A1 (en) Method for predicting the resistance of melanoma to a treatment with a braf inhibitor
US20140309229A1 (en) Methods for selecting and treating cancer in patients with igf-1r/ir inhibitors
US20050100897A1 (en) NFAT transcription factors in tumor progression
WO2016112302A1 (en) Method of predicting sensitivity of tumor cell growth to combination therapy of fgfr inhibitor
WO2012034076A2 (en) Etv1 as a diagnostic, prognostic and therapeutic target for gastrointestinal stromal tumors
AU2014244593A1 (en) Predicting response to EGFR inhibitors
US20240156800A1 (en) Ep300 degrader and uses thereof in neuroblastoma
US9823256B2 (en) CXCR1 as a predictor of response to treatment with epidermal growth factor receptor therapeutic
US20090099197A1 (en) Methods of Identifying and Treating Individuals Exhibiting MDR-1 Overexpression With Protein Tyrosine Kinase Inhibitors and Combinations Thereof
KR20220145688A (en) Biomarker for colon cancer diagnosis and pharmaceutical composition for preventing or treating colon cancer
JP2023535280A (en) Method for screening colon cancer metastasis inhibitor

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09702397

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 12812777

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009702397

Country of ref document: EP