NZ611525B2 - Use of glu-tubulin as a biomarker of drug response to furazanobenzimidazoles - Google Patents

Use of glu-tubulin as a biomarker of drug response to furazanobenzimidazoles Download PDF

Info

Publication number
NZ611525B2
NZ611525B2 NZ611525A NZ61152512A NZ611525B2 NZ 611525 B2 NZ611525 B2 NZ 611525B2 NZ 611525 A NZ611525 A NZ 611525A NZ 61152512 A NZ61152512 A NZ 61152512A NZ 611525 B2 NZ611525 B2 NZ 611525B2
Authority
NZ
New Zealand
Prior art keywords
cancer
compound
tubulin
glu
use according
Prior art date
Application number
NZ611525A
Other versions
NZ611525A (en
Inventor
Felix Bachmann
Heidi Alexandra Lane
Original Assignee
Basilea Pharmaceutica Ag
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 Basilea Pharmaceutica Ag filed Critical Basilea Pharmaceutica Ag
Priority claimed from PCT/EP2012/050814 external-priority patent/WO2012098203A1/en
Publication of NZ611525A publication Critical patent/NZ611525A/en
Publication of NZ611525B2 publication Critical patent/NZ611525B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/44Multiple drug resistance
    • 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
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5026Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects on cell morphology
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels

Abstract

Disclosed is the use of a glu-tubulin as a biomarker for predicting the response to a compound, wherein the compound is a compound of general formula I, and formula I is as defined in the specification.

Description

Use of glu-tubulin as a biomarker of drug response The present invention generally relates to use of glu-tubulin as a biomarker for ting the response of a disease, such as a neoplastic or autoimmune disease, preferably cancer, to a compound of general formula I, such as 3-(4-{1-[2- (4-amino-phenyl)oxo-ethyl]-1H-benzoimidazolyl}-furazanylamino)- propionitrile (BAL27862). In other aspects it relates to s and kits involving the use of the biomarker. Also described are methods of treatment involving the use of the biomarker.
Microtubules are one of the components of the cell eleton and are composed of heterodimers of alpha and beta n. Agents that target microtubules are among the most effective cytotoxic chemotherapeutic agents having a broad spectrum of activity. ubule destabilising agents (e.g. the vinca-alkaloids such as vincristine, vinblastine and vinorelbine) are used for example in the treatment of several types of hematologic malignancies, such as lymphoblastic leukaemia and ma, as well as solid s, such as lung cancer. Microtubule stabilising agents (e.g. the taxanes such as paclitaxel, docetaxel) are used for example in the ent of solid tumours, ing breast, lung and prostate cancer.
However resistance to these known microtubule targeting agents can occur.
The ance can either be inherent or can be acquired after exposure to these agents. Such resistance therefore impacts t survival rates, as well as choices of treatment regimes. Several potential mechanisms of resistance have been identified, and include defects in the microtubule targets, such as elevated levels of beta-tubulin subtype III and acquired ons in beta-tubulin subtype I that are known to reduce taxane binding. Furthermore, defects in other cell proteins have been suggested to be associated with resistance to certain microtubule targeting agents, such as overexpression of the efflux pump P-glycoprotein (P-gp pump, also known as multi-drug resistance protein 1 or MDR1). Such factors may then be used as biomarkers of resistance to these conventional microtubule targeting agents.
A vely recently ered class of microtubule destabilising agents are compounds encompassed by the formula given below: R3 R2 N R4 N R5 N N wherein R represents phenyl, thienyl or pyridinyl wherein phenyl is optionally substituted by one or two tuents independently selected from alkyl, halo-lower alkyl, hydroxy-lower alkyl, lower alkoxy-lower alkyl, acyloxy-lower alkyl, phenyl, hydroxy, lower alkoxy, hydroxy-lower alkoxy, lower alkoxy-lower alkoxy, phenyl-lower alkoxy, lower alkylcarbonyloxy, amino, monoalkylamino, dialkylamino, lower alkoxycarbonylamino, lower alkylcarbonylamino, substituted amino n the two substituents on nitrogen form together with the nitrogen heterocyclyl, lower alkylcarbonyl, y, lower alkoxycarbonyl, cyano, halogen, and nitro; and wherein two adjacent substituents are methylenedioxy; and n pyridinyl is optionally substituted by lower alkoxy, amino or halogen; X represents a group C=Y, wherein Y stands for oxygen or nitrogen substituted by hydroxy or lower alkoxy; R1 represents hydrogen, lower alkylcarbonyl, hydroxy-lower alkyl or cyano-lower alkyl; R2, R3 and R6 represent hydrogen; R4 and R5, independently of each other, represent hydrogen, lower alkyl or lower alkoxy; or R4 and R5 together ent methylenedioxy; and pharmaceutically acceptable salts f; or wherein R represents phenyl or pyridinyl wherein phenyl is optionally tuted by one or two substituents ndently selected from alkyl, halo-lower alkyl, hydroxy-lower alkyl, lower alkoxy-lower alkyl, acyloxy-lower alkyl, phenyl, hydroxy, lower , hydroxy-lower alkoxy, lower alkoxy-lower alkoxy, phenyl-lower alkoxy, lower alkylcarbonyloxy, amino, monoalkylamino, dialkylamino, lower alkoxycarbonylamino, lower alkylcarbonylamino, substituted amino wherein the two substituents on nitrogen form together with the en heterocyclyl, lower alkylcarbonyl, carboxy, lower alkoxycarbonyl, formyl, cyano, n, and nitro; and wherein two adjacent substituents are methylenedioxy; and wherein pyridinyl is optionally substituted by lower alkoxy, amino or halogen; X represents oxygen; R1 represents hydrogen, lower alkylcarbonyl, hydroxy-lower alkyl or cyano-lower alkyl; R2, R3 and R6 represent hydrogen; R4 and R5, independently of each other, represent hydrogen, lower alkyl or lower alkoxy; or R4 and R5 together represent enedioxy; and pharmaceutically acceptable salts thereof; and wherein the prefix lower denotes a radical having up to and including a maximum of 7, ally up to and including a maximum of 4 carbon atoms.
These compounds are disclosed in WO2004/103994 A1, which is incorporated by cross-reference herein. These compounds have been shown to arrest tumour cell proliferation and induce apoptosis.
The synthesis of compounds of a I is described in WO2004/103994 A1, in general on pages 29-35, and specifically on pages 39-55, which are incorporated herein by cross-reference. They may be prepared as disclosed or by an analogous method to the processes described therein.
One compound falling within this class, known as BAL27862, and shown in WO2004/103994 A1 as example 58, and specifically orated by reference , has the structure and chemical name given below: N N N N O Chemical name: 3-(4-{1-[2-(4-Amino-phenyl)oxo-ethyl]-1H-benzoimidazolyl}-furazanylamino)- propionitrile.
Or herein as Compound A Further compounds ified in WO2004/103994 A1 as examples 50 and 79 respectively, and also specifically incorporated by cross-reference herein, have the structures and chemical names given below: N N N N O Chemical name: 2-[2-(4-Amino-furazanyl)-benzoimidazolyl](4-amino-phenyl)- ethanone or herein as Compound B and N N N N O Chemical name: 3-(4-{1-[2-(6-Amino-pyridinyl)oxo-ethyl]-1H- benzoimidazolyl}-furazanylamino)-propionitrile or herein as Compound C.
BAL27862 has activity across a broad panel of experimental, solid tumour xenograft models. Moreover, activity is retained even against tumour models which were selected for resistance to conventional microtubule targeting agents (including the vinca-alkaloid ubule ilisers and the microtubule stabilisers paclitaxel and lone B). BAL27862 activity is not affected by over-expression of the P-gp pump in any models tested in vitro, nor in human mammary tumour xenografts.
Additionally, BAL27862 retained its activity despite elevated levels of beta-tubulin subtype III and mutations in tubulin subtype I.
Hence, BAL27862 activity is not affected by a number of factors that confer resistance to conventional microtubule targeting agents.
Moreover, it is known that compounds of general formula I have a ent effect on the phenotype of cells compared to other microtubule targeting agents, including other microtubule destabilisers. Treatment with a compound of general formula I induces a consistent microtubule phenotype in tumour cell lines derived from a variety of , for e lung, cervix and breast, as seen in Figure 1.
Staining the microtubules in these cells with an anti-alpha-tubulin antibody shows that rather than the mitotic spindle fibres of untreated cells, only dot-like structures are e in the treated cells. This same effect is also shown using Compounds C and B in Figures 2A and 2B respectively on the lung cancer cell line A549. It is however very distinct from that observed with the conventional microtubule ing agents vinblastine, colchicine, paclitaxel and zole as seen in Figures 3B, 3C, 3D and 4, respectively. The microtubules were stained with an anti-alpha-tubulin antibody and the cells viewed at a 1000 x magnification (Figures 3, 4). For the cells treated with BAL27862, multiple ke structures are e, whereas, in stark st, the other conventional drugs produce filamentous microtubule structures, or dense microtubule aggregate structures. These differences at the phenotypic level, at nd doses considered l in terms of antiproliferative effect indicate a difference in the mode of action at the molecular level.
Furthermore, it is known that BAL27862 elicits a dominant microtubule phenotype in the presence of the other microtubule targeting agents. Treatment with vinblastine, colchicine, paclitaxel or nocodazole alone induced the microtubule phenotypes characteristic of these agents (Figure 5A, 5D, 5G, 6C-6F respectively).
However, combination treatment with BAL27862 for the last 4 hours resulted in disruption of these phenotypes; despite the ued presence of vinblastine, colchicine, paclitaxel, or nocodazole (Figure 5B, 5E, 5H, 6G-6J tively). In contrast, treating first with BAL27862 and subsequently for 4 hours in combination with vinblastine, cine, paclitaxel, or nocodazole had no impact on generation of the phenotype consistent with BAL27862 treatment (Figure 5C, 5F, 5I, 6K-6N respectively).
These data all demonstrate that BAL27862 affects microtubule y in a different manner than conventional microtubule targeting agents.
Thus, from information about conventional microtubule targeting agents, predictions cannot be made concerning if, or how, particular genes are involved in the action of nds of formula I.
An object of the present ion is to identify factors which are associated with response to compounds of formula I or pharmaceutically acceptable tives thereof, for example to identify factors associated with resistance to compounds of general formula I, in ular BAL27862 or pharmaceutically acceptable derivatives thereof, as defined below; and/or to provide the public with a useful .
It has surprisingly been found that glu-tubulin may be used as a ker of response to treatment with a nd of general formula I or pharmaceutically acceptable derivatives thereof, as defined below.
In one preferred embodiment of the invention, relatively high glu-tubulin levels in a tumour sample are associated with inherent and ed resistance to BAL27862, as described below.
Tubulin is subjected to a variety of posttranslational modifications, including detyrosination/tyrosination, acetylation, glutamylation, polyglycylation, phosphorylation of serine residues and phosphorylation of tyrosine residues, making it one of the most ed proteins known. Many alpha tubulin genes bed to date t the presence of a carboxy terminal tyrosine on tubulin. It is currently thought that once tubulin polymerizes and assembles into microtubules, in some cases a carboxypeptidase (tubulin carboxypeptidase or tubulin tyrosine carboxypeptidase, TTCP) acts to remove the C-terminal tyrosine from alpha tubulin exposing the penultimate glutamate. Another enzyme, tubulin tyrosine ligase (TTL), can reattach this tyrosine. The exact function of this detyrosination/tyrosination cycle in the cell has yet to be elucidated.
The form of alpha tubulin which does not have a tyrosine, but rather a glutamate at its C-terminal is known as glu-tubulin or glu tubulin or detyrosinated tubulin. The term glu-tubulin shall be used herein to refer to the form of alpha tubulin with a glutamate as the final amino acid at the inal. The ation glutubulin , shall also encompass forms wherein other post-translational modifications may additionally be present. The alpha n which forms the basis for the glu- n is known to exist in multiple variants, subtypes and isoforms, as well as there being multiple alpha tubulin genes which give rise to these, and all these shall also be included in the designation glu-tubulin, with the proviso that a glutamate is the final amino acid at the C-terminal. Preferably it relates to human variants, subtypes and isoforms of alpha n, with the o that a glutamate is the final amino acid at the C-terminal. In a more preferred embodiment, a glutamate, not a tyrosine, is the final amino acid at the C-terminus of alpha tubulin after post-translational modification. Subtypes of alpha tubulin include, but are not limited to, tubulin alpha 1A, tubulin alpha 1B, tubulin alpha 1C and tubulin alpha 3C/D. The protein sequences of these es are accessible via the ing National Center for Biotechnology Information (NCBI) Reference numbers NP_006000, NP_006073, NP_116093 and NP_005992, tively. These are also listed in Figures 11-14 (NP_006000.2, NP_006073.2, NP_116093.1, NP_005992.1,) as SEQ ID NO. 1-4, respectively. Glu-tubulin, as described above, does not have the tyrosine at the final amino acid at the C-terminus of the sequences presented in SEQ. ID. NO. 1-4.
Accordingly, in one aspect, the invention relates to the use of glu-tubulin as a biomarker for predicting the se to a compound, wherein the compound is a R3 R2 N R4 N R5 N N compound of general formula I wherein R ents phenyl, thienyl or pyridinyl wherein phenyl is optionally substituted by one or two tuents independently ed from alkyl, halo-lower alkyl, hydroxy-lower alkyl, lower alkoxy-lower alkyl, acyloxy-lower alkyl, phenyl, hydroxy, lower alkoxy, hydroxy-lower alkoxy, lower alkoxy-lower alkoxy, phenyl-lower , lower alkylcarbonyloxy, amino, monoalkylamino, lamino, lower alkoxycarbonylamino, lower alkylcarbonylamino, substituted amino wherein the two substituents on nitrogen form together with the nitrogen heterocyclyl, lower alkylcarbonyl, carboxy, lower alkoxycarbonyl, cyano, halogen, and nitro; and wherein two adjacent substituents are methylenedioxy; and wherein pyridinyl is optionally substituted by lower alkoxy, amino or halogen; X represents a group C=Y, wherein Y stands for oxygen or nitrogen substituted by hydroxy or lower alkoxy; R1 represents hydrogen, lower alkylcarbonyl, hydroxy-lower alkyl or cyano-lower alkyl; R2, R3 and R6 represent hydrogen; R4 and R5, independently of each other, represent hydrogen, lower alkyl or lower alkoxy; or R4 and R5 together represent methylenedioxy; and ceutically acceptable salts, solvates, esters and amides of naturally occurring amino acids, small peptides or pegylated hydroxyl acids, salts of such esters and amides, and polymorphs thereof; or wherein R represents phenyl or pyridinyl wherein phenyl is ally substituted by one or two substituents independently ed from alkyl, halo-lower alkyl, hydroxy-lower alkyl, lower alkoxy-lower alkyl, acyloxy-lower alkyl, phenyl, hydroxy, lower alkoxy, hydroxy-lower alkoxy, lower alkoxy-lower alkoxy, phenyl-lower alkoxy, lower alkylcarbonyloxy, amino, monoalkylamino, lamino, lower alkoxycarbonylamino, lower arbonylamino, substituted amino wherein the two tuents on nitrogen form together with the nitrogen heterocyclyl, lower alkylcarbonyl, y, lower alkoxycarbonyl, formyl, cyano, halogen, and nitro; and wherein two adjacent substituents are methylenedioxy; and wherein pyridinyl is optionally substituted by lower alkoxy, amino or halogen; X represents oxygen; R1 ents hydrogen, lower alkylcarbonyl, hydroxy-lower alkyl or cyano-lower alkyl; R2, R3 and R6 represent hydrogen; R4 and R5, independently of each other, represent hydrogen, lower alkyl or lower alkoxy; or R4 and R5 together represent methylenedioxy; and pharmaceutically acceptable salts, solvates, esters and amides of naturally occurring amino acids, small peptides or pegylated hydroxy acids, salts of such esters and amides, and polymorphs thereof, and wherein the prefix lower denotes a l having up to and including a maximum of 7 carbon atoms and, wherein the response is of a disease in a subject and the biomarker glu-tubulin is measured ex vivo in a sample or s taken from the human or animal body.
In another aspect, the invention provides a method for ting in a subject suffering from a cancer the response of that cancer to a nd of general formula I or to a pharmaceutically able salt, solvate, ester or amide of a naturally occurring amino acid, small peptide or pegylated hydroxy acid, or to a salt of such ester or amide, or to a polymorph thereof, as defined in the first aspect of the invention, comprising the steps of: a) measuring ex vivo a level of glu-tubulin in a sample pre-obtained from tumour tissue or circulating tumour cells of the subject to obtain a value or values representing this level; and b) comparing the value or values from step a) to a standard value or set of standard values from subjects with the same cancer type, wherein a higher glu-tubulin level in the sample ve to the standard value or set of standard values is predictive of resistance of the subject's cancer to the compound of formula (I) or to the pharmaceutically acceptable salt, solvate, ester or amide of a naturally occurring amino acid, small peptide or ted hydroxy acid, or salt of such ester or amide, or polymorph thereof.
In another aspect, the invention relates to the use of a compound of l formula I or of a pharmaceutically acceptable salt, solvate, ester or amide of a lly occurring amino acid, small peptide or pegylated hydroxy acid, or rph thereof as defined in the first aspect of the invention, for the preparation of a pharmaceutical composition for ng a cancer in a subject in need thereof, wherein the t is selected for treatment with the compound of l formula I or with the salt, solvate, ester or amide of a naturally occurring amino acid, small peptide or ted hydroxy acid, salt of such ester or amide, or polymorph thereof as defined in the first aspect of the invention, if the level of glu-tubulin, measured ex vivo in a sample taken from the subject, is not higher than a standard value or set of standard values from subjects with the same tumour histotype or from normal cells, tissue or body fluid.
In another aspect, the invention provides a kit when used for predicting the response to a compound of general formula I or a pharmaceutically acceptable salt, e, ester or amide of a naturally occurring amino acid, small peptide or pegylated hydroxy acid, salt of such ester or amide, or polymorph thereof, as defined in the first aspect of the invention, comprising reagents necessary for measuring a level of bulin in a sample taken from a subject with a cancer, comprising a compound of the following formula N N N N O or a pharmaceutically acceptable salt thereof and further comprising a ator module which ses a standard value or set of standard values of a level of glu-tubulin taken from samples of tumour tissue or circulating tumour cells of subjects with a cancer of the same histotype to which the level of bulin in the sample is compared.
Certain statements that appear below are broader than what appears in the statements of the invention above. These statements are provided in the sts of providing the reader with a better understanding of the invention and its practice. The reader is directed to the accompanying claim set which defines the scope of the invention.
One embodiment relates to use of glu-tubulin as a biomarker for predicting the response to a compound, wherein the compound is a compound of general formula I, R3 R2 N R4 N R5 N N R represents phenyl, thienyl or pyridinyl wherein phenyl is optionally substituted by one or two tuents ndently selected from alkyl, halo-lower alkyl, y-lower alkyl, lower alkoxy-lower alkyl, acyloxy-lower alkyl, phenyl, hydroxy, lower alkoxy, hydroxy-lower alkoxy, lower alkoxy-lower alkoxy, phenyl-lower alkoxy, lower alkylcarbonyloxy, amino, kylamino, dialkylamino, lower alkoxycarbonylamino, lower alkylcarbonylamino, substituted amino wherein the two substituents on nitrogen form together with the nitrogen heterocyclyl, lower arbonyl, carboxy, lower carbonyl, cyano, halogen, and nitro; and wherein two adjacent substituents are methylenedioxy; and wherein pyridinyl is optionally substituted by lower alkoxy, amino or halogen; X represents a group C=Y, wherein Y stands for oxygen or nitrogen substituted by hydroxy or lower alkoxy; R1 represents hydrogen, lower alkylcarbonyl, hydroxy-lower alkyl or cyano-lower alkyl; R2, R3 and R6 represent hydrogen; R4 and R5, independently of each other, represent hydrogen, lower alkyl or lower alkoxy; or R4 and R5 together represent methylenedioxy; and pharmaceutically acceptable derivatives thereof, or wherein R represents phenyl or pyridinyl wherein phenyl is optionally substituted by one or two substituents independently selected from alkyl, halo-lower alkyl, hydroxy-lower alkyl, lower alkoxy-lower alkyl, acyloxy-lower alkyl, phenyl, hydroxy, lower alkoxy, hydroxy-lower alkoxy, lower alkoxy-lower alkoxy, phenyl-lower alkoxy, lower alkylcarbonyloxy, amino, monoalkylamino, lamino, lower alkoxycarbonylamino, lower alkylcarbonylamino, substituted amino wherein the two substituents on nitrogen form together with the nitrogen heterocyclyl, lower alkylcarbonyl, carboxy, lower alkoxycarbonyl, formyl, cyano, halogen, and nitro; and wherein two nt substituents are methylenedioxy; and n pyridinyl is optionally substituted by lower alkoxy, amino or halogen; X ents oxygen; R1 represents hydrogen, lower alkylcarbonyl, hydroxy-lower alkyl or lower alkyl; R2, R3 and R6 represent hydrogen; R4 and R5, independently of each other, represent hydrogen, lower alkyl or lower alkoxy; or R4 and R5 together represent enedioxy; and pharmaceutically acceptable derivatives thereof; and wherein the prefix lower denotes a radical having up to and including a maximum of 7, especially up to and including a maximum of 4 carbon atoms.
Preferably the response may be of a disease in a subject. Also preferably the response may be to ent, i.e. to treatment with the compound of general formula I or pharmaceutically able derivatives thereof.
The biomarker glu-tubulin is measured ex vivo in a sample or samples taken from the human or animal body, preferably taken from the human body.
In a preferred embodiment, described is the use of glu-tubulin as a biomarker for predicting the resistance of a e in a subject to a compound of general formula I or ceutically acceptable derivatives thereof as defined above.
Preferably the pharmaceutically acceptable derivative is selected from the group consisting of a salt, solvate, pro-drug, salt of a pro-drug, polymorph and isomer of a compound of general formula I as defined above. Pro-drugs are preferably ester and amides of naturally occurring amino acids, small peptides or pegylated hydroxy acids. More preferably, the pro-drug is an amide formed from an amino group present within the R group of the compound of general formula I and the carboxy group of glycine, alanine or .
Particularly preferably the compound is N N N N O or a pharmaceutically able salt thereof, preferably a hydrochloride salt thereof, most preferably a dihydrochloride salt thereof.
Another ment relates to a method for predicting the se of a disease in a subject to a compound of general formula I or pharmaceutically acceptable derivatives thereof as defined above, comprising the steps of: a) measuring a level of glu-tubulin in a sample pre-obtained from the subject to obtain a value or values representing this level; and b) comparing the value or values from step a) to a standard value or set of standard values.
Further preferably the response which is predicted is resistance.
The measuring of a level or levels of glu-tubulin is performed ex-vivo in a sample or samples pre-obtained from the t. Pre-obtained refers to the fact that the sample is obtained before it is subjected to any method involving measuring the level of the biomarker, and pre-obtained is not to be understood as in relation to treatment. In a preferred embodiment, a higher level of glu-tubulin in the sample from the subject relative to the standard value or set of rd values predicts resistance.
Also preferably, the e is a neoplastic or autoimmune disease. More preferably the disease is cancer. Especially preferably, the cancer is selected from the group consisting of breast cancer, prostate cancer, cervical , ovarian cancer, gastric cancer, colorectal cancer (i.e. including colon cancer and rectal cancer), pancreatic cancer, liver cancer, brain cancer, neuroendocrine cancer, lung cancer, kidney cancer, hematological malignancies, melanoma and sarcomas. More especially preferably the cancer is selected from the group consisting of breast cancer, cervical cancer, ovarian cancer, colorectal cancer, melanoma and lung cancer. In an especially preferred embodiment the cancer is selected from the group consisting of lung , melanoma, n cancer and colorectal cancer. In a particularly red embodiment, wherein inherent resistance is determined, the cancer is ed from the group consisting of lung cancer, melanoma or colorectal cancer. In another particularly preferred embodiment, n ed resistance is determined, the cancer is lung or ovarian cancer.
Also described is a method of treating a neoplastic or autoimmune disease, preferably cancer, in a subject in need thereof, sing measuring a level of glutubulin in a sample from the subject to obtain a value or values representing this level, and treating the subject with a compound of general formula I or a pharmaceutically acceptable derivative thereof as defined above, if the level of glu- tubulin in said sample is not higher than a standard value or set of standard values.
Also described is glu-tubulin for use in the treatment of a neoplastic or autoimmune disease, preferably cancer, comprising measuring a level of glu-tubulin in a sample from the subject to obtain a value or values representing this level, and treating the subject with a compound of general formula I or a pharmaceutically acceptable derivative thereof as defined above, if the level of glu-tubulin is not higher than a standard value or set of rd values.
The measuring of a level of glu-tubulin is performed ex-vivo in a sample preobtained from the subject.
Also described is a method of treating a neoplastic or autoimmune disease, ably cancer, by first decreasing the level of glu-tubulin in a t that has a sample with a higher level of glu-tubulin compared to a standard level or set of standard levels, then treating the subject with a compound of l formula I or a pharmaceutically acceptable derivative thereof as defined above.
Also described is a kit for predicting the response to a compound of general formula I or a pharmaceutically acceptable derivative thereof, as defined above, sing reagents necessary for ing the level of bulin in a sample.
More preferably the kit also comprises a comparator module which ses a standard value or set of standard values to which the level of bulin in the sample is compared.
More preferably the kit comprises a nd of general formula I or a pharmaceutically acceptable derivative thereof as defined above. In an especially preferred embodiment the kit comprises a compound of the following formula or a pharmaceutically acceptable salt thereof N N N N O Chemical name: S-2,6-Diamino-hexanoic acid [4-(2-{2-[4-(2-cyanoethylamino )-furazanyl]-benzoimidazolyl}-acetyl)-phenyl]-amide In a particularly preferred embodiment the pharmaceutically acceptable salt is a ochloride salt.
Also described is a device for predicting the response to a nd of l formula I or a pharmaceutically acceptable derivative thereof as defined above, comprising reagents necessary for measuring a level of glu-tubulin in a sample and a comparator module which comprises a standard value or set of standard values to which the level of glu-tubulin in the sample is compared.
In a preferred embodiment, the reagents in the kit or device comprise a capture reagent comprising a detector for glu-tubulin, and a detector reagent.
Especially preferably the capture t is an dy. Also preferably, the disease is predicted to be ant to treatment with said compound when glu-tubulin is higher relative to a standard value or set of standard . In a preferred embodiment, the comparator module is included in instructions for use of the kit. In another preferred embodiment the comparator module is in the form of a display device.
Embodiments of the present invention will now be described by way of example with reference to the accompanying figures. The invention however is not to be understood as limited to these embodiments.
Brief Description of the Figures Figure 1: Shows the treatment of human tumour cell lines from different ypes with 50 nM BAL27862. The microtubules of mitotic or G2/M arrested cells were stained after 24 hours ent with 50 nM BAL27862 or vehicle control.
Fig. 1A and 1B: A549 NSCLC cells; Fig. 1C and 1D: HeLa al cancer cells; Fig. 1E and 1F: SKBR3 breast cancer cells Vehicle control treatment: Figures 1A, 1C & 1E, BAL27862 ent: Figures 1B, 1D & 1F.
Figure 2: Shows the treatment of A549 NSCLC cells with the Compounds B and C. The microtubules of mitotic or G2/M arrested A549 NSCLC cells were stained after 24 hours treatment with 80 nM or 20 nM of Compounds B and C, respectively.
The white scale bar represents 10 micrometres.
Fig. 2A: treatment with 20 nM Compound C Fig. 2B: treatment with 80 nM Compound B Figure 3: Shows a comparison of treatment of cells with BAL27862 compared to conventional microtubule targeting agents. Microtubules of c or G2/M arrested A549 NSCLC cells were stained after 24 hours of treatment with 50 nM of A: 62; B: vinblastine; C: colchicine; D: paclitaxel. Stacks of images taken every 1 µm were processed by using ImageJ software.
Figure 4: Shows a ison of treatment of A549 NSCLC cells with BAL27862 compared to nocodazole. Microtubules of mitotic or G2/M arrested cells were stained after 24 h of treatment with various concentrations of nocodazole (B, C & D) and BAL27862 (E, F & G). A: control, B: Nocodazole 50 nM, C: Nocodazole 100 nM, D: Nocodazole 200 nM, E: BAL27862 20 nM; F: 62 30 nM and G: BAL27862 50 nM. The white scale bar represents 10 micrometres. Representative images of the microtubule phenotypes observed are shown.
Figure 5: Shows a combination of treatment with 62 and conventional microtubule-targeting agents. Microtubules of mitotic or G2/M arrested A549 NSCLC cells were stained after treatment for the times indicated below. 50 nM BAL27862, 50 nM vinblastine, 50 nM colchicine and 25 nM paclitaxel were used. The white scale bar ents 10 micrometres.
Fig. 5A: 24 hours stine treatment; Fig. 5B: 24 hours vinblastine treatment with the final 4 hours including BAL27862; Fig. 5C: 24 hours BAL27862 treatment with the final 4 hours including stine.
Fig. 5D: 24 hours colchicine treatment; Fig. 5E: 24 hours colchicine treatment with the final 4 hours including BAL27862; Fig. 5F: 24 hours BAL27862 ent with the final 4 hours including colchicine.
Fig. 5G: 24 hours paclitaxel treatment; Fig. 5H: 24 hours paclitaxel treatment with the final 4 hours including BAL27862; Fig. 5I: 24 hours BAL27862 treatment with the final 4 hours including paclitaxel.
Figure 6: Shows a combination of treatment with BAL27862 and nocodazole. Microtubules of mitotic or G2/M ed A549 NSCLC cells were stained after treatment for the times ted below. 25 nM BAL27862 and nocodazole at the concentrations indicated below were used. The white scale bar represents 10 micrometers.
Fig. 6A: 24 hours control treatment; Fig. 6B: 24 hours of 25 nM BAL27862 treatment; Fig. 6C: 24 hours of 50 nM nocodazole treatment Fig. 6D: 24 hours of 100 nM nocodazole treatment Fig. 6E: 24 hours of 150 nM nocodazole treatment Fig. 6F: 24 hours of 200 nM nocodazole treatment Fig. 6G: 24 hours of 50 nM nocodazole treatment with the final 4 hours including 25 nM BAL27862; Fig. 6H: 24 hours of 100 nM nocodazole treatment with the final 4 hours including 25 nM 62; Fig. 6I: 24 hours of 150 nM nocodazole treatment with the final 4 hours including 25 nM BAL27862; Fig. 6J: 24 hours of 200 nM nocodazole treatment with the final 4 hours including 25 nM BAL27862; Fig. 6K: 24 hours of 25 nM BAL27862 treatment with the final 4 hours including 50 nM nocodazole; Fig. 6L: 24 hours of 25 nM BAL27862 treatment with the final 4 hours including 100 nM nocodazole; Fig. 6M: 24 hours of 25 nM BAL27862 treatment with the final 4 hours including 150 nM nocodazole; Fig. 6N: 24 hours of 25 nM BAL27862 treatment with the final 4 hours ing 200 nM nocodazole.
Figure 7: Shows protein ts prepared from patient-derived colorectal cancer (Fig. 7A), lung cancer (Fig. 7B) and melanoma (Fig. 7C) tumours obtained from subcutaneous xenografted mice, and analysed by immunoblotting for glu-tubulin expression, with actin included as a loading control. Three independent s were analysed in each case (1 – 3). 62, paclitaxel and vinblastine resistance and sensitivity is as defined in Table 1.
Figure 8: Shows by immunohistochemistry that glu-tubulin levels in tumour cells are increased in a patient-derived xenografted colorectal tumour defined as BAL27862 resistant by ex vivo colony outgrowth analysis. Patient-derived tumour afts (maintained in nude mice) were prepared, fixed and stained for glu-tubulin protein sion using immunohistochemistry. BAL27862, paclitaxel and vinblastine resistance and sensitivity is defined in Table 1.
Figure 9: Shows tumour cell lines which were ed for resistance to BAL27862 through in vitro cultivation in the presence of the compound. Based on IC 50 determinations, BAL27862 resistance factors versus parental lines were: A549 (3.0 fold); SKOV3 (7.6 fold – resistant 1 line); H460 (5.3 fold) (see Table 2). Whole cell protein extracts were prepared from parental and resistant lines and analysed by immunoblotting for glu-tubulin sion. Actin levels were included as a loading control.
Figure 10: Shows that increased glu-tubulin protein levels are ined in SKOV3 tumour lines during resistance development. SKOV3 tumour cell lines were selected for resistance to BAL27862 through in vitro cultivation in the presence of BAL27862 for increasing time periods. Based on IC50 determinations, BAL27862 resistance factors versus parental lines were: SKOV3 resistant 1 (7.6 fold), SKOV3 resistant 2 (11.6 fold) (see Table 2). Whole cell n extracts were prepared from parental and resistant lines and analysed by immunoblot for glu-tubulin expression.
Actin levels act as a loading control.
Figure 11: Shows the protein sequence of tubulin alpha-1A chain [Homo sapiens] (SEQ. ID. No. 1) Figure 12: Shows the protein ce of n alpha-1B chain [Homo sapiens] (SEQ ID No. 2) Figure 13: Shows the protein sequence of n alpha-1C chain [Homo sapiens] (SEQ. ID. NO. 3) Figure 14: Shows the protein sequence of Tubulin alpha-3C/D chain [Homo sapiens] (SEQ. ID. NO. 4) Detailed Description Compounds of formula I The compounds useful in the invention are represented by general formula I: R3 R2 N R4 N R5 N N n R represents phenyl, l or pyridinyl wherein phenyl is optionally substituted by one or two substituents independently selected from alkyl, ower alkyl, hydroxy-lower alkyl, lower alkoxy-lower alkyl, acyloxy-lower alkyl, phenyl, hydroxy, lower alkoxy, y-lower alkoxy, lower alkoxy-lower alkoxy, -lower alkoxy, lower alkylcarbonyloxy, amino, kylamino, lamino, lower alkoxycarbonylamino, lower alkylcarbonylamino, substituted amino wherein the two substituents on nitrogen form together with the nitrogen heterocyclyl, lower alkylcarbonyl, carboxy, lower alkoxycarbonyl, cyano, halogen, and nitro; and wherein two adjacent substituents are methylenedioxy; and wherein pyridinyl is optionally substituted by lower alkoxy, amino or halogen; X represents a group C=Y, wherein Y stands for oxygen or nitrogen substituted by hydroxy or lower alkoxy; R1 represents hydrogen, lower alkylcarbonyl, hydroxy-lower alkyl or cyano-lower alkyl; R2, R3 and R6 represent hydrogen; R4 and R5, independently of each other, represent hydrogen, lower alkyl or lower alkoxy; or R4 and R5 together represent methylenedioxy; and pharmaceutically acceptable derivatives thereof, or wherein R ents phenyl or pyridinyl wherein phenyl is optionally substituted by one or two substituents ndently selected from alkyl, halo-lower alkyl, hydroxy-lower alkyl, lower -lower alkyl, acyloxy-lower alkyl, phenyl, hydroxy, lower alkoxy, hydroxy-lower alkoxy, lower alkoxy-lower alkoxy, phenyl-lower alkoxy, lower alkylcarbonyloxy, amino, monoalkylamino, dialkylamino, lower alkoxycarbonylamino, lower alkylcarbonylamino, substituted amino wherein the two substituents on nitrogen form together with the nitrogen heterocyclyl, lower alkylcarbonyl, carboxy, lower alkoxycarbonyl, formyl, cyano, halogen, and nitro; and wherein two adjacent substituents are methylenedioxy; and wherein pyridinyl is optionally substituted by lower alkoxy, amino or halogen; X represents oxygen; R1 represents hydrogen, lower alkylcarbonyl, y-lower alkyl or cyano-lower alkyl; R2, R3 and R6 represent hydrogen; R4 and R5, independently of each other, represent hydrogen, lower alkyl or lower alkoxy; or R4 and R5 together represent methylenedioxy; and pharmaceutically able derivatives thereof; and n the prefix lower s a radical having up to and including a maximum of 7, especially up to and including a maximum of 4 carbon atoms. cyclyl designates preferably a saturated, partially saturated or unsaturated, mono- or bicyclic ring containing 4-10 atoms comprising one, two or three heteroatoms selected from en, oxygen and sulfur, which may, unless otherwise specified, be carbon or nitrogen linked, wherein a ring nitrogen atom may optionally be substituted by a group selected from lower alkyl, amino-lower alkyl, aryl, aryl-lower alkyl and acyl, and a ring carbon atom may be substituted by lower alkyl, amino-lower alkyl, aryl, ower alkyl, heteroaryl, lower alkoxy, hydroxy or oxo.
Examples of heterocyclyl are pyrrolidinyl, oxazolidinyl, thiazolidinyl, piperidinyl, linyl, piperazinyl, dioxolanyl and tetrahydropyranyl.
Acyl designates, for example, arbonyl, cyclohexylcarbonyl, arylcarbonyl, aryl-lower alkylcarbonyl, or heteroarylcarbonyl. Lower acyl is preferably lower alkylcarbonyl, in particular propionyl or acetyl.
Preferably, the compound of general formula I is defined as wherein R1 is ed from the group consisting of hydrogen, acetyl, CH2CH 2CN and CH 2CH 2CH 2OH.
In one preferred embodiment, the nd of general formula I is selected from the group consisting of: 4-(1-Phenacyl-1H-benzimidazolyl)-furazanylamine, 4-[1-(4-Bromophenacyl)-1H-benzimidazolyl]-furazanylamine oxime, N-{4-[1-(4-Chlorophenacyl)-1H-benzimidazolyl]-furazanyl}-acetamide, 4-[1-(4-Chlorophenacyl)-1H-benzimidazolyl]-furazanyl-N-(2-cyanoethyl)-amine 4-[1-(4-Chlorophenacyl)-1H-benzimidazolyl]-furazanyl-N-(3-hydroxypropyl)- amine, 4-[1-(3-Aminochlorophenacyl)-1H-benzimidazolyl]-furazanylamine 4-[1-(3-Methoxymethoxymethoxy-phenacyl)-1H-benzimidazolyl]-furazan e, and ceutically acceptable derivatives thereof.
In another preferred embodiment, the compound of general formula I is selected from the group consisting of: N N wherein R, Y and R1 are d as follows : R Y R1 O H NOH H NOMe H O H NOH H NOH H NOMe H O H NOH H NOMe H O H NOH H NOMe H O H NOMe H O H Cl Cl O H NOH H NOMe H O H NOH H NOMe H NOMe H O H Et N O Ac O H O H O H O CN O CH2CH2CN O H O H O CH2OH O H O CH2CH2CN O H O CH2CH2CN O CH2CH2CN O CH2CH2CN O H AcNH O H O H AcHN O H O H O H O H O CN O H O H O H O H O H O H O H O H O H NH N O CH2CH2CN NH N O H O O O H O O O CH2CH2CN MeO N or pharmaceutically acceptable tives thereof.
In yet another preferred embodiment, the compound of general formula I is selected from the group consisting of: 4-(1-Phenoxymethyl-1H-benzimidazolyl)-furazanylamine, 4-[1-(4-Fluorophenoxymethyl)-1H-benzimidazolyl]-furazanylamine, 4-[1-(3,4-Dimethylphenoxymethyl)-1H-benzimidazolyl]-furazanyl-N-(2- cyanoethyl)-amine and compounds ented by the formula: N N N N O wherein R and R1 are as defined below R R1 CH2CH2CN CH2CH2CN CH2CH2CN CH2CH2CH2OH Cl N NH N and pharmaceutically able derivatives thereof.
In still yet another preferred ment the compound of general formula I R4 N N R5 N N wherein R, R4 and R5 are as defined below R R4 R5 Me Me Me Me Me Me Me Me Me Me OMe OMe OMe OMe OMe OMe OMe OMe OMe OMe or pharmaceutically acceptable derivatives f.
More preferably, the compound is a compound of general formula I R3 R2 N R4 N R5 N N wherein R represents phenyl or pyridinyl wherein phenyl is optionally substituted by one or two substituents independently ed from lower alkyl, lower alkoxy, amino, acetylamino, halogen and nitro; and wherein pyridinyl is optionally substituted by amino or halogen; X represents a group C=O; R1 represents hydrogen or cyano-lower alkyl; R2, R3, R4, R5 and R6 represent hydrogen; and pharmaceutically acceptable tives thereof, and wherein the prefix lower s a radical having up to and including a maximum of 7, especially up to and including a maximum of 4 carbon atoms.
Especially preferably, the compound is represented by the following formula N N wherein R, Y and R1 are defined as follows : R Y R1 O H O CH2CH2CN O H NH N O CH2CH2CN NH N or pharmaceutically acceptable tives thereof.
More especially preferably, the compound is represented by the following formula N N wherein R, Y and R1 are defined as follows: R Y R1 O CH2CH2CN O H O CH2CH2CN NH N or pharmaceutically acceptable derivatives thereof.
Particularly preferably, the compound is N N N N O or pharmaceutically acceptable derivatives thereof.
The term tive or derivatives in the phrase “pharmaceutically acceptable derivative” or “pharmaceutically acceptable derivatives” of compounds of general formula I relates to salts, es and xes f and to solvates and complexes of salts thereof, as well as to pro-drugs, polymorphs, and isomers thereof (including l, geometric and tautomeric isomers) and also salts of prodrugs thereof. In a more preferred embodiment, it relates to salts and pro-drugs, as well as to salts of pro-drugs thereof.
Salts are preferably acid addition salts. Salts are formed, preferably with organic or inorganic acids, from compounds of formula (I) with a basic nitrogen atom, especially the pharmaceutically able salts. Suitable inorganic acids are, for example, halogen acids, such as hydrochloric acid, sulfuric acid, or phosphoric acid.
Suitable organic acids are, for example, carboxylic, phosphonic, sulfonic or sulfamic acids, for example acetic acid, nic acid, octanoic acid, decanoic acid, dodecanoic acid, glycolic acid, lactic acid, fumaric acid, succinic acid, adipic acid, pimelic acid, suberic acid, azelaic acid, malic acid, tartaric acid, citric acid, amino acids, such as glutamic acid or ic acid, maleic acid, hydroxymaleic acid, methylmaleic acid, cyclohexanecarboxylic acid, adamantanecarboxylic acid, benzoic acid, salicylic acid, 4-aminosalicylic acid, phthalic acid, phenylacetic acid, mandelic acid, cinnamic acid, methane- or ethane-sulfonic acid, 2-hydroxyethanesulfonic acid, ethane-1,2-disulfonic acid, benzenesulfonic acid, 2-naphthalenesulfonic acid, 1,5- naphthalene-disulfonic acid, 2-, 3- or ylbenzenesulfonic acid, methylsulfuric acid, ethylsulfuric acid, dodecylsulfuric acid, N-cyclohexylsulfamic acid, N-methyl-, N- ethyl- or N-propyl-sulfamic acid, or other organic protonic acids, such as ascorbic acid.
The compound useful in the invention may be administered in the form of a ug which is broken down in the human or animal body to give a compound of the formula I. es of ugs include in vivo hydrolysable esters and amides of a compound of the formula I. Particular ugs considered are ester and amides of naturally ing amino acids and ester or amides of small peptides, in particular small peptides consisting of up to five, preferably two or three amino acids as well as esters and amides of pegylated hydroxy acids, preferably hydroxy acetic acid and lactic acid. Pro-drug esters are formed from the acid function of the amino acid or the C terminal of the e and le hydroxy group(s) in the compound of formula I.
Pro-drug amides are formed from the amino function of the amino acid or the N terminal of the peptide and suitable carboxy group(s) in the compound of formula I, or from the acid function of the amino acid or the C terminal of the peptide and suitable amino group(s) in the compound of a I. Particularly preferably the pro-drug amides are formed from the amino group(s) present within the R group of formula I.
More preferably, the pro-drug is formed by the addition of glycine, alanine or lysine to the compound of formula I.
Even more ably the compound of general formula I is in the form of a pro-drug selected from the compounds of formulae: N N NH N N N N O N N N N N O N N O O O O O H N O N NH2 H H NH2 NH2 H2N , , , N N NH N N O N N N N N N O N N O O O O N N O H N O N NH2 H H NH2 NH2 H2N , , , H2N N N N N N N O N N O N N O O O O O H N O N NH H H NH2 NH H2N , 2 and .
In an especially preferred ment the compound of general formula I is in the form of a pro-drug which has the ing formula N N N N O In a most especially preferred embodiment the compound is a pharmaceutically acceptable salt, preferably a hydrochloride salt, most preferably a dihydrochloride salt, of a compound of the following formula N N N N O The pharmaceutically active metabolite in vivo in this case is BAL27862.
These pro-drugs may be prepared by processes that are known per se, in ular, a process, wherein a nd of formula (II) N N N N O (II) wherein R1 is defined as for formula (I) and Z is CH or N, or a derivative of such a compound comprising functional groups in protected form, or a salt thereof is (1) acylated with an amino acid of formula (III) R11 (III) wherein R10 is selected from en (Gly); methyl (Ala) and protected aminobutyl (Lys) and R11 is a suitable amino protecting group, and (2) any protecting groups in a protected derivative of the ing compound are removed to yield a pro-drug as shown above, and, if so desired, (3) said pro-drug is converted into a salt by treatment with an acid, or a salt of a compound of formula (II) is converted into the ponding free compound of formula (II) or into another salt, and/or a mixture of isomeric product compounds is separated into the individual isomers.
Acylation of a compound of formula (II) with an amino acid of formula (III) is performed in a manner known per se, usually in the presence of a suitable polar or dipolar aprotic solvent, with cooling or heating as required, for example in a temperature range from imately minus 80°C to approximately plus 150°C, more preferably from minus 30°C to plus 120°C, especially in a range from approximately around 0°C to the reflux temperature of the used solvent. ally a suitable base is added, in particularly an aromatic base like ne or collidine or a tertiary amine base such as triethylamine or diisopropylethylamine, or an inorganic basic salt, e.g. potassium or sodium ate.
Acylation may be accomplished under conditions used for amide formation known per se in peptide chemistry, e.g. with activating agents for the carboxy group, such as carbodiimides like N,N’-diethyl-, N,N’-dipropyl-, N,N’-diisopropyl-, N,N’- dicyclohexylcarbodiimide and N-(3-dimethylaminoisopropyl)-N’-ethylcarbodiimidehydrochloride (EDC), or with agents such as 1-hydroxybenzotriazole (HOBt), benzotriazolyloxytris(dimethylamino)-phosphonium hexafluorophosphate (BOP), O-(7-aza-benzotriazolyl)-N,N,N’,N’-tetramethyl-uronium hexafluorophosphate (HATU), 2-(2-oxo(2H)-pyridyl)-1,1,3,3-tetramethyluronium tetrafluoroborate (TPTU), optionally in the presence of suitable bases, sts or co-reagents. The carboxy group may also be ted as acyl halogenide, preferably as acyl chloride, e.g. by on with thionylchloride or chloride, or as symmetrical or unsymmetrical anhydride, e.g. by reaction with halogeno formates like ethyl chloroformate, optionally in the presence of suitable bases, catalysts or co-reagents.
If one or more other functional groups, for example carboxy, hydroxy or amino, are or need to be protected in a compound of formula (II) or (III), because they should not take part in the reaction, these are such protecting groups as are usually applied in the sis of amides like, in particular peptide compounds, cephalosporins, penicillins, nucleic acid derivatives and sugars, which are known to the skilled persons. Suitable protecting groups for amino groups are for example tbutyl ate, benzyl carbamate or 9-fluorenylmethyl ate.
The protecting groups may already be present in precursors and should protect the functional groups concerned against unwanted secondary reactions, such as alkylations, acylations, etherifications, esterifications, ions, solvolysis, and similar reactions. It is a characteristic of protecting groups that they lend themselves readily, i.e. without undesired secondary reactions, to removal, typically by solvolysis, reduction, photolysis or also by enzyme activity, for example under conditions analogous to logical conditions, and that they are not t in the end products. The specialist knows, or can easily ish, which protecting groups are suitable with the reactions mentioned hereinabove and hereinafter.
The protection of such functional groups by such protecting groups, the ting groups themselves, and their removal ons are described for example in standard reference books for peptide synthesis and in special books on protective groups such as J. F. W. McOmie, ctive Groups in Organic Chemistry", Plenum Press, London and New York 1973, in "Methoden der organischen Chemie" (Methods of organic chemistry), Houben-Weyl, 4th edition, Volume 15/I, Georg Thieme , Stuttgart 1974, and in T. W. Greene, G. M. Wuts "Protective Groups in Organic Synthesis", Wiley, New York, 2006.
Disease The compounds of general formula I have been shown to arrest cell proliferation and induce cell death, for e by apoptosis.
Dysregulation of cell proliferation, or lack of appropriate cell death, has wide ranging al implications. A number of diseases associated with such dysregulation involve hyperproliferation, inflammation, tissue remodeling and repair.
Familiar indications in this category include cancers, restenosis, neointimal hyperplasia, angiogenesis, endometriosis, lymphoproliferative disorders, transplantation d pathologies (graft rejection), polyposis, loss of neural function in the case of tissue remodeling and the like.
Cancer is associated with abnormal cell eration and cell death rates. As apoptosis is inhibited or delayed in most types of proliferative, neoplastic diseases, induction of apoptosis is an option for treatment of cancer, especially in cancer types which show resistance to classic chemotherapy, radiation and immunotherapy (Apoptosis and Cancer Chemotherapy, Hickman and Dive, eds., Blackwell Publishing, 1999). Also in autoimmune and transplantation related diseases and ogies compounds inducing apoptosis may be used to restore normal cell death processes and therefore can eradicate the symptoms and might cure the es.
Further applications of compounds inducing apoptosis may be in restenosis, i.e. accumulation of vascular smooth muscle cells in the walls of arteries, and in persistent infections caused by a failure to eradicate ia- and virus-infected cells. Furthermore, apoptosis can be induced or reestablished in epithelial cells, in endothelial cells, in muscle cells, and in others which have lost contact with extracellular matrix.
A compound according to general formula I or pharmaceutically able derivatives thereof may be used for the lactic or ally therapeutic treatment of the human or animal body, in particular for ng a stic disease, autoimmune disease, lantation related pathology and/or degenerative e.
Examples of such neoplastic diseases include, but are not d to, epithelial neoplasms, squamous cell neoplasms, basal cell neoplasms, transitional cell papillomas and carcinomas, adenomas und adenocarcinomas, adnexal and skin appendage neoplasms, mucoepidermoid neoplasms, cystic neoplasms, mucinous and serous neoplasms, ducal-, lobular and medullary neoplasms, acinar cell sms, complex epithelial neoplasms, specialized gonadal neoplasms, paragangliomas and glomus tumours, naevi and melanomas, soft tissue tumours and sarcomas, fibromatous neoplasms, myxomatous neoplasms, lipomatous neoplasms, myomatous sms, complex mixed and stromal neoplasms, fibroepithelial neoplasms, synovial like neoplasms, mesothelial neoplasms, germ cell neoplasms, trophoblastic neoplasms, mesonephromas, blood vessel tumours, lymphatic vessel s, osseous and chondromatous neoplasms, giant cell tumours, laneous bone tumours, odontogenic tumours, gliomas, neuroepitheliomatous neoplasms, meningiomas, nerve sheath tumours, granular cell tumours and alveolar soft part sarcomas, Hodgkin's and non-Hodgkin's lymphomas, other lymphoreticular neoplasms, plasma cell tumours, mast cell tumours, proliferative diseases, leukemias, miscellaneous myeloproliferative ers, lymphoproliferative disorders and myelodysplastic syndromes.
The compounds of general a I or pharmaceutically acceptable derivatives thereof may be used to treat autoimmune diseases. Examples of such autoimmune diseases include, but are not limited to, systemic, discoid or subacute cutaneous lupus matosus, rheumatoid arthritis, antiphospholipid syndrome, CREST, progressive systemic sclerosis, mixed connective tissue disease (Sharp syndrome), Reiter's syndrome, juvenile arthritis, cold agglutinin disease, essential mixed cryoglobulinemia, tic fever, sing spondylitis, chronic polyarthritis, myasthenia , multiple sclerosis, chronic inflammatory demyelinating polyneuropathy, Guillan-Barre syndrome, dermatomyositis/ polymyositis, autoimmune hemolytic anemia, thrompocytopenic purpura, neutropenia, type I diabetes mellitus, thyroiditis (including Hashimoto's and Grave'disease), Addison's disease, andular syndrome, pemphigus (vulgaris, foliaceus, sebaceous and vegetans), bullous and cicatricial pemphigoid, pemphigoid gestationis, epidermolysis bullosa acquisita, linear IgA disease, lichen sclerosus et atrophicus, morbus Duhring, psoriasis is, guttate, generalized ar and localized pustular psoriasis, vitiligo, alopecia areata, primary biliary cirrhosis, autoimmune hepatitis, all forms of glomerulonephritis, pulmonal hemorrhage (goodpasture syndrome), IgA pathy, pernicious anemia and mune gastritis, inflammatory bowel diseases (including colitis ulcerosa and morbus Crohn), Behcet's disease, Celic- Sprue disease, mune s, mune myocarditis, granulomatous orchitis, aspermatogenesis without orchitis, idiopatic and secondary pulmonary fibrosis, inflammatory diseases with a possibility of mune pathogensesis, such as pyoderma gangrensosum, lichen ruber, sarcoidosis (including Lofgren and cutaneous/subcutaneous type), granuloma anulare, allergic type I and type IV immunolgical reaction, asthma bronchiale, pollinosis, atopic, contact and airborne dermatitis, large vessel itis (giant cell and Takayasu's arteritis), medium sized vessel vasculitis (polyarteritis nodosa, ki disease), small vessel vasculitis (Wegener's granulomatosis, Churg Strauss syndrome, microscopic polangiitis, HenochSchoenlein purpura, essential cryoglobulinemic vasculitis, cutaneous leukoklastic angiitis), hypersensitivity syndromes, toxic epidermal necrolysis (Stevens-Johnson syndrome, erythema multiforme), es due to drug side effects, all forms of cutaneous, organ- specific and systemic effects due to type l-vu (Coombs fication) immunologic forms of reaction, transplantation related pathologies, such as acute and chronic graft versus host and host versus graft disease, involving all organs (skin, heart, kidney, bone marrow, eye, liver, spleen, lung, muscle, central and peripheral nerve system, connective tissue, bone, blood and lymphatic vessel, genito-urinary system, ear, cartillage, primary and secondary lymphatic system including bone marrow, lymph node, thymus, gastrointestinal tract, including oro-pharynx, esophageus, stomach, small intestine, colon, and rectum, including parts of above mentioned organs down to single cell level and substructures, e. g. stem cells).
Particularly preferably, the disease is a neoplastic or autoimmune disease.
In an especially preferred embodiment the e is cancer.
Examples of cancers in terms of the organs and parts of the body affected include, but are not limited to, the breast, cervix, ovaries, colon, rectum, (including colon and rectum i.e. colorectal cancer), lung, ding small cell lung cancer, non- small cell lung cancer, large cell lung cancer and mesothelioma), bone, endocrine system, adrenal gland, thymus, liver, stomach, intestine, (including gastric cancer), pancreas, bone marrow, hematological ancies, (such as lymphoma, leukemia, myeloma or lymphoid ancies), bladder, urinary tract, s, skin, thyroid, brain, head, neck, te and testis. ably the cancer is ed from the group consisting of breast cancer, prostate cancer, al , ovarian cancer, gastric cancer, colorectal cancer, pancreatic cancer, liver cancer, brain cancer, neuroendocrine cancer, lung cancer, kidney cancer, hematological malignancies, melanoma and sarcomas. Especially preferably the cancer is selected from the group ting of breast cancer, al cancer, ovarian cancer, colorectal cancer, melanoma and lung cancer. More especially ably the cancer is selected from the group consisting of lung cancer, melanoma, ovarian cancer and colorectal cancer. In another preferred embodiment, for the case when the resistance predicted is acquired ance, the cancer is lung cancer or ovarian cancer. In yet another preferred embodiment, for the case where the resistance predicted is inherent resistance, the cancer is selected from the group consisting of colorectal cancer, lung cancer or melanoma.
Samples The ement of the level of glu-tubulin may be performed in vitro, on a sample of ical tissue derived from the subject. The sample may be any biological material separated from the body such as, for example, normal tissue, tumour tissue, cell lines, whole blood, serum, plasma, cerebrospinal fluid, lymph fluid, circulating tumour cells, cell lysate, tissue lysate, urine and aspirates. Preferably the sample is derived from normal tissue, tumour tissue, or circulating tumour cells. More preferably the sample is derived from tumour tissue or circulating tumour cells. In one particularly preferred embodiment the sample is derived from tumour tissue. For example, the level of glu-tubulin may be measured in a fresh, frozen or formalin fixed/paraffin embedded tumour tissue .
The sample is pre-obtained from the subject before the sample is subjected to the method steps involving measuring the level of the ker. The methods for l of the sample are well known in the art, and it may for example be d from the subject by biopsy, for e by punch biopsy, core biopsy or aspiration fine needle biopsy, endoscopic biopsy, or surface biopsy. Blood may be collected by venipuncture and further processed according to standard ques. Circulating tumour cells may also be obtained from blood based on, for example, size (e.g. ISET - Isolation by Size of Epithelial Tumour cells) or immunomagnetic cell enrichment. (e.g. Cellsearch®, Veridex, Raritan, NJ) Sample comparison The t may be human or animal. ably the subject is human.
The biomarker glu-tubulin is measured ex vivo in a sample or samples taken from the human or animal body, preferably taken from the human body. The sample or samples are pre-obtained from the human or animal body, ably pre-obtained from the human body before the sample is subjected to the method steps involving measuring the level of the biomarker.
A biomarker is in general a substance that is used as an indicator of a biological response, ably as an indicator of the susceptibility to a given treatment, which in the present application is treatment with a compound of general formula I or a pharmaceutically acceptable derivative thereof.
In a particularly preferred ment, higher glu-tubulin levels in the sample relative to a standard value or set of rd values predicts resistance.
As used herein, an se or relatively high or high or higher levels relative to a standard level or set of standard levels means the amount or concentration of the biomarker in a sample is detectably greater in the sample relative to the standard level or set of standard levels. This encompasses at least an increase of, or higher level of, about 1% ve to the standard, preferably at least an increase of about % relative to the standard. More preferably it is an increase of, or higher level of, at least about 10% relative to the standard. More ularly preferably it is an increase of, or higher level of, at least about 20% relative to the standard. For example, such an increase of, or higher level of, may include, but is not limited to, at least about 1%, about 10%, about 20%, about 30%, about 50%, about 70%, about 80%, about 100%, about 150% or about 200% or more relative to the standard.
Preferably, higher glu-tubulin levels in a sample or samples i) relative to a standard value or set of standard values from subjects with the same tumour histotype; or ii) taken after treatment initiation and compared to a sample or samples taken from the same subject before treatment initiation; or iii) ve to a rd value or set of standard values from normal cells or are predictive of resistance.
The measuring of a level of glu-tubulin is performed ex-vivo in a sample preobtained from the subject. Further preferably the response which is to be predicted is resistance.
More preferably, higher glu-tubulin levels in a sample or samples i) relative to a rd value or set of standard values from subjects with the same tumour histotype; or ii) taken after treatment initiation and compared to a sample or samples taken from the same t before treatment initiation; are predictive of resistance.
Especially preferably, higher glu-tubulin levels in a sample or samples ve to a rd value or set of standard values from subjects with the same tumour histotype are predictive of ance.
In one preferred embodiment, for the case i) where the measurement is compared in a sample or samples relative to a standard value or set of standard values from samples from subjects with the same tumour histotype as the sample to which it is to be compared, the standard value or set of standard values are established from samples from a population of subjects with that cancer type. The samples from these standard subjects may for example be derived from the tumour tissue, circulating tumour cells or blood, as long as the origin of the sample is consistent between the standard and the sample to be compared.
In r preferred embodiment, for the case ii) where the measurement is compared in a sample or s taken after ent initiation and compared to a sample or samples taken from the same subject before treatment initiation, it is measured preferably to predict acquired resistance. The samples are ed to cells or tissue from the same biological origin. The prediction of ed ance would then indicate that the treatment with the compound should be discontinued.
The biomarker is thus used to monitor whether further treatment with the compound is likely to give the required response (e.g. reduction of abnormal cells), or whether the cells have become non-responsive or resistant to such treatment.
In yet another preferred embodiment, for the case iii) where the ement is compared in a sample or samples ve to a standard value or set of standard values from normal cells or tissue, the standard value or set of standard values may be established from a sample of normal (e.g. non-tumourous) cells or tissue or body fluid. Such data may be gathered from a population of subjects in order to develop the standard value or set of standard values.
The standard value or set of standard values are established ex-vivo from pre-obtained samples which may be from cell lines, or preferably biological material from at least one subject and more preferably from an average of subjects (e.g., n=2 to 1000 or more). The standard value or set of standard values may then be correlated with the response data of the same cell lines, or same subjects, to treatment with a compound of general formula I or a pharmaceutically able derivative thereof. From this correlation a comparator , for example in the form of a relative scale or scoring system, optionally including cut-off or threshold values, can be established which indicates the levels of biomarker associated with a spectrum of response levels to the compound of formula I or a pharmaceutically acceptable derivative thereof. The spectrum of response levels may comprise relative sensitivity to the therapeutic activity of the compound, (e.g. high sensitivity to low sensitivity), as well as resistance to the therapeutic activity. In a preferred ment this comparator module ses a f value or set of values which predicts resistance to treatment.
For example, if an immunohistochemical method is used to measure the level of glu-tubulin in a , standard values may be in the form of a scoring system. Such a system might take into account the percentage of cells in which staining for glu-tubulin is present. The system may also take into account the relative intensity of staining in the individual cells. The standard values or set of standard values of the level of glu-tubulin may then be correlated with data indicating the response, especially resistance, of the subject or tissue or cell line to the eutic activity of a compound of formula I or a pharmaceutically acceptable derivative f. Such data may then form part of a comparator module.
Response is the on of the cell lines, or preferably of the subject, or more preferably of the disease in a subject, to the eutic activity of a compound of general formula I or a ceutically acceptable derivative thereof. The spectrum of response levels may comprise relative sensitivity to the eutic activity of the compound, (e.g. high sensitivity to low ivity), as well as resistance to the therapeutic activity. The se data may for example be monitored in terms of: objective response rates, time to disease progression, progression free survival, and overall survival.
The response of a cancerous e may be evaluated by using criteria well known to a person in the field of cancer treatment, for example but not restricted Response Evaluation Criteria in Solid Tumors (RECIST) Guidelines, Source: Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer.2009; -47; RANO Criteria for High-Grade Gliomas, Source: Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, Galanis E, Degroot J, Wick W, Gilbert MR, Lassman AB, Tsien C, Mikkelsen T, Wong ET, Chamberlain MC, Stupp R, Lamborn KR, Vogelbaum MA, van den Bent MJ, Chang SM. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 8(11):1963-72; CA-125 Rustin Criteria for Ovarian Cancer Response, Source: Rustin GJ, Quinn M, Thigpen T, du Bois A, Pujade-Lauraine E, Jakobsen A, Eisenhauer E, Sagae S, Greven K, e I, Cervantes A, Vermorken J. Re: New guidelines to evaluate the response to treatment in solid tumors (ovarian cancer). J Natl Cancer Inst. 2004; 96(6):487-8; PSA Working Group 2 Criteria for Prostate Cancer Response, Source: Scher HI, Halabi S, Tannock I, Morris M, Sternberg CN, Carducci MA, Eisenberger MA, Higano C, Bubley GJ, Dreicer R, Petrylak D, Kantoff P, Basch E, Kelly WK, Figg WD, Small EJ, Beer TM, Wilding G, Martin A, Hussain M; Prostate Cancer Clinical Trials Working Group. Design and end points of clinical trials for patients with progressive te cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J Clin Oncol. 2008;26(7):1148-59.
Resistance is associated with there not being an observable and/or measurable reduction in, or absence of, one or more of the following: reduction in the number of abnormal cells, preferably cancerous cells; or absence of the abnormal cells, preferably cancerous cells; for ous diseases: reduction in tumour size; inhibition (i.e., slowed to some extent and preferably stopped) of r tumour ; reduction in the levels of tumour markers such as PSA and CA-125, inhibition (i.e., slowed to some extent and ably stopped) of cancer cell infiltration into other organs (including the spread of cancer into soft tissue and bone); inhibition (i.e., slowed to some extent and ably stopped) of tumour asis; alleviation of one or more of the symptoms associated with the specific cancer; and reduced morbidity and mortality.
In a preferred embodiment resistance means there is no observable and/or measurable ion in, or absence of, one or more of the following ia: reduction in tumour size; inhibition of further tumour growth, inhibition of cancer cell infiltration into other organs; and inhibition of tumour metastasis.
In a more preferred embodiment resistance refers to one or more of the following criteria: no reduction in tumour size; no inhibition of further tumour growth, no inhibition of cancer cell infiltration into other organs; and no inhibition of tumour metastasis. ement of the aforementioned ance criteria is according to clinical guidelines well known to a person in the field of cancer treatment, such as those listed above for measuring the response of a cancerous disease.
Response may also be established in vitro by assessing cell proliferation and/or cell death. For example, effects on cell death or proliferation may be assessed in vitro by one or more of the following well established assays: A) Nuclear staining with Hoechst 33342 dye providing information about nuclear morphology and DNA fragmentation which are rks of apoptosis. B) AnnexinV binding assay which reflects the phosphatidylserine content of the outer lipid bilayer of the plasma membrane. This event is considered an early hallmark of sis. C) TUNEL assay (Terminal deoxynucleotidyl transferase mediated dUTP Nick End Labeling assay), a scence method for evaluating cells undergoing apoptosis or necrosis by measuring DNA fragmentation by labeling the terminal end of nucleic acids. D) MTS eration assay measuring the lic activity of cells. Viable cells are metabolically active whereas cells with a mised respiratory chain show a reduced ty in this test. E) Crystal violet staining assay, where effects on cell number are monitored through direct staining of cellular components. F) Proliferation assay monitoring DNA synthesis through incorporation of bromodeoxyuridine (BrdU).
Inhibitory effects on /proliferation can be directly determined. G) YO-PRO assay which involves a membrane impermeable, fluorescent, monomeric cyanine, c acid stain, which permits analysis of dying (e.g. apoptotic) cells without interfering with cell viability. Overall effects on cell number can also be analysed after cell permeabilisation. H) Propidium iodide staining for cell cycle distribution which shows alterations in distribution among the different phases of the cell cycle. Cell cycle arresting points can be determined. I) Anchorage-independent growth assays, such as colony outgrowth assays which assess the ability of single cell suspensions to grow into colonies in soft agar.
In a preferred embodiment relating to determination of resistance in vitro, resistance means there is no decrease in the proliferation rate of abnormal cells and/or reduction in the number of abnormal cells. More preferably resistance means there is no decrease in the proliferation rate of cancerous cells and/or no reduction in the number of cancerous cells. The ion in the number of abnormal, preferably cancerous, cells may occur through a variety of programmed and non-programmed cell death mechanisms. Apoptosis, caspase-independent programmed cell death and autophagic cell death are examples of programmed cell death. r the cell death criteria involved in embodiments of the invention are not to be taken as limited to any one cell death mechanism.
Glu-tubulin Preferred examples of the protein ce of alpha tubulin (human alpha tubulin) are listed in SEQ. ID No. 1-4, Figures 11-14. Alpha tubulin is a sor of bulin. As described previously, the glu-tubulin itself has the C-terminal tyrosine removed. The term bulin also encompasses homologues, mutant forms, allelic ts, isoforms, splice variants and equivalents of the sequences ented by SEQ ID NO 1-4, with the proviso that a glutamate is the final amino acid at the C- terminal. More preferably it encompasses sequences having at least about 75% identity, especially preferably at least about 85% identity, particularly ably at least about 95% identity, and more particularly ably about 99% identity to said sequences, with in each case the proviso that a glutamate is the final amino acid at the C-terminal.
Level of glu-tubulin The level of bulin may be assayed in the sample by protein analysis techniques well known to a skilled person. Examples of methods known in the art which are suitable to measure the level of glu-tubulin at the protein level include, but are not limited to, i) immunohistochemistry (IHC) analysis, ii) western blotting iii) immunoprecipitation iv) enzyme linked immunosorbant assay (ELISA) v) radioimmunoassy vi) Fluorescence ted cell sorting (FACS) vii) mass spectrometry, including matrix assisted laser tion/ionisation (MALDI, e.g.
MALDI-TOF) and electrospray ionisation mass-spectrometry (ESI—MS).
The antibodies involved in some of the above methods may be monoclonal or polyclonal antibodies, antibody fragments, and/or various types of synthetic antibodies, including ic dies. The antibody may be labelled to enable it to be detected or capable of ion following reaction with one or more further s, for example using a secondary dy that is labelled or capable of producing a detectable result. Antibodies specific to the glu-tubulin form of alpha tubulin are available commercially from Milipore or can be prepared via conventional antibody generation methods well known to a skilled person.
Preferred methods of protein analysis are ELISA, mass spectrometry techniques, immunohistochemistry and western blotting, more ably western blotting and immunohistochemistry. In western blotting, also known as immunoblotting, ed antibodies may be used to assess levels of protein, where the intensity of the signal from the detectable label corresponds to the amount of protein, and can be quantified for example by ometry.
Immunohistochemistry again uses labelled antibodies to detect the ce and relative amount of the biomarker. It can be used to assess the percentage of cells for which the biomarker is present. It can also be used to assess the localisation or relative amount of the biomarker in individual cells, the latter is seen as a function of the intensity of staining.
ELISA stands for enzyme linked immunosorbant assay, since it uses an enzyme linked to an antibody or antigen for the detection of a specific protein. ELISA is lly performed as follows (although other variations in methodology exist): a solid ate such as a 96 well plate is coated with a primary antibody, which recognises the biomarker. The bound biomarker is then recognised by a secondary antibody specific for the biomarker. This may be directly joined to an enzyme or a third anti-immunoglobulin antibody may be used which is joined to an enzyme. A substrate is added and the enzyme catalyses a reaction, yielding a specific colour.
By measuring the optical density of this colour, the presence and amount of the biomarker can be determined.
Uses of biomarker The biomarker may be used to t inherent resistance of the disease in a subject to the compound of general formula I or a pharmaceutically acceptable derivative thereof as defined above.
The biomarker may be used to predict acquired resistance of the disease in a subject to the compound of general formula I or a pharmaceutically acceptable derivative thereof as defined above.
The biomarker may be used to select subjects suffering or predisposed to suffering from a disease, preferably cancer, for treatment with a nd of general formula I or a ceutically acceptable derivative thereof as d above. The levels of such a biomarker may be used to identify subjects likely to respond or to not respond or to continue to respond or to not continue to respond to treatment with such agents. Stratification of subjects may be made in order to avoid unnecessary treatment regimes. In particular the biomarker may be used to fy subjects from whom a sample or samples do not display a higher level of bulin, relative to a standard level or set of standard levels, whereupon such ts may then be selected for treatment with the compound of formula I or a pharmaceutically acceptable derivative thereof as d above.
The biomarker may also be used to assist in the determination of treatment regimes, regarding amounts and schedules of dosing. onally, the biomarker may be used to assist in the selection of a combination of drugs to be given to a subject, including a compound or nds of general formula I or a pharmaceutically acceptable derivative thereof, and another chemotherapeutic (cytotoxic) agent or . Furthermore, the biomarker may be used to assist in the determination of therapy gies in a subject including whether a compound of general formula I or a pharmaceutically acceptable derivative thereof is to be stered in combination with ed therapy, endocrine therapy, radiotherapy, immunotherapy or surgical intervention, or a combination of these.
Glu-tubulin may also be used in combination with other biomarkers to predict the response to a nd of general formula I or a pharmaceutically acceptable tive thereof and to determine treatment regimes. It may furthermore be used in combination with chemo-sensitivity testing to predict resistance and to determine treatment regimes. Chemo-sensitivity testing involves directly applying a compound of general formula I to cells taken from the subject, for example from a subject with haematological malignancies or accessible solid tumours, for example breast, head and neck cancers or melanomas, to determine the response of the cells to the compound.
Method of treatment Also described is a method of ent and glu-tubulin for use in a method of treatment, wherein the level of bulin is first ished relative to a standard level or set of standard levels or pre-treatment initiation levels and then a compound of general formula I or a ceutically able derivative thereof as defined above, is stered if the level of glu-tubulin in said sample is not higher than a standard value or set of standard values or has not increased relative to pre- treatment initiation levels respectively. The compound of formula I or a pharmaceutically acceptable derivative thereof may be administered in a pharmaceutical composition, as is well known to a person skilled in the art. Suitable compositions and dosages are for example sed in A1 pages -39, which are specifically incorporated by reference herein. Compositions for enteral administration, such as nasal, buccal, rectal or, especially, oral administration, and for parenteral administration, such as intravenous, intramuscular or subcutaneous administration, to warm-blooded animals, especially humans, are especially preferred. More particularly, compositions for intravenous administration are preferred.
The compositions comprise the active ingredient and a pharmaceutically acceptable carrier. An example of a composition includes, but is not limited to, the following: 5000 soft gelatin capsules, each comprising as active ingredient 0.05 g of one of the nds of general a (I), are prepared as follows: 250 g pulverized active ingredient is suspended in 2 liter Lauroglykol (propylene glycol laurate, Gattefossé S.A., Saint Priest, France) and ground in a wet pulverizer to produce a particle size of about 1 to 3 µm. 0.419 g portions of the mixture are then introduced into soft gelatin capsules using a capsule-filling machine.
Also bed is a method of treating a neoplastic or autoimmune disease, preferably cancer, by first decreasing the level of glu-tubulin in a subject that has a sample with a higher level of glu-tubulin compared to a standard level or set of standard levels or pre-treatment initiation , then treating the subject with a nd of general a I or a ceutically acceptable derivative as defined above. The level of glu-tubulin may be decreased by direct or indirect chemical or genetic means. Examples of such methods are treatment with a drug that results in reduced glu-tubulin expression, ed delivery of viral, plasmid or e constructs, or antibody or siRNA or antisense to downregulate the level of bulin. For example siRNA may be used to reduce the level of TTCP or delivery of a plasmid may be used to increase the expression of TTL, and thereby reduce the level of bulin in the cell. The subject may then be treated with a compound of general formula I or a pharmaceutically acceptable derivative thereof.
A compound of general formula I or a pharmaceutically acceptable derivative thereof can be administered alone or in combination with one or more other eutic agents. le combination therapy may take the form of fixed combinations, or the administration of a compound as described herein and one or more other therapeutic agents which are staggered or given independently of one r, or the combined administration of fixed combinations and one or more other therapeutic agents.
A compound of general formula I or a pharmaceutically acceptable derivative thereof can, besides or in addition, be administered especially for tumour therapy in combination with herapy (cytotoxic therapy), targeted therapy, endocrine therapy, radiotherapy, immunotherapy, surgical intervention, or a combination of these. Long-term therapy is equally possible as is adjuvant therapy in the context of other treatment strategies, as described above. Other possible treatments are therapy to maintain the patient's status after tumour regression, or even preventive therapy, for example in patients at risk.
Kit and device One embodiment relates to a kit and another to a device for predicting the response, preferably of a disease in a subject, to a compound of general formula I or a ceutically acceptable derivative thereof as defined above, comprising reagents necessary for measuring the level of bulin in a sample. Preferably, the reagents comprise a e t comprising a detector for glu-tubulin and a detector reagent.
The kit and device may also preferably comprise a comparator module which comprises a standard value or set of standard values to which the level of glutubulin in the sample is compared. In a preferred embodiment, the comparator module is included in ctions for use of the kit. In another preferred embodiment the comparator module is in the form of a display device, for example a strip of colour or cally coded al which is designed to be placed next to the t of the sample measurement to indicate resistance levels. The rd value or set of standard values may be determined as bed above.
The reagents are ably antibodies or antibody fragments which selectively bind to glu-tubulin. These may for example be in the form of one specific primary antibody which binds to glu-tubulin and a secondary antibody which binds to the primary antibody, and which is itself labelled for detection. Alternatively, the primary antibody may also be labelled for direct detection. The kits or devices may optionally also contain a wash solution(s) that selectively allows retention of the bound biomarker to the capture reagent as compared with other biomarkers after g. Such kits can then be used in ELISA, western blotting, flow cytometry, immunohistochemistry or other immunochemical methods to detect the level of the biomarker.
More preferably the kit comprises a compound of general formula I, or a pharmaceutically acceptable derivative thereof as defined above. This compound may then be administered to the subject, in accordance with the level of the ker in the sample from the subject, as measured by the reagents comprised in the kit. Therefore the kit according to the invention may be used in the method of treatment described herein, as defined above. In an especially preferred embodiment the kit comprises a compound of the ing formula or a pharmaceutically acceptable salt f N N N N O In a particularly preferred embodiment of the kit the pharmaceutically acceptable salt is a dihydrochloride salt. Another embodiment relates to the use of such a kit as described above. rmore the device may comprise imaging s or measurement devices (for example, but not restricted to, measurement of fluorescence) which further process the measured signals and transfer them into a scale in a comparator module.
In the present specification the words “comprise” or ises” or “comprising” are to be understood as to imply the inclusion of a stated item or group of items, but not the exclusion of any other item or group of items.
Experimental methodology Immunofluorescent staining of cultured cells A549 human non-small cell lung cancer (NSCLC, ATCC reference number CCL-185) cells, HeLa cervical cancer cells (ATCC reference number CCL-2) and SKBR3 breast carcinoma cells (ATCC reference number HTB-30) were seeded at densities of 50% on round microscope coverslips and ed for 24 hours in RPMI- 1640 containing 10 % FCS (also referred to as FBS) at 37°C, 5% CO2. Compounds to be tested were dissolved in DMSO. The cell culture medium was replaced with medium ning the diluted compound(s) (paclitaxel, vinblastine, colchicine and nocodazole were purchased from Sigma-Aldrich) or e. After treatment for the times indicated in the Brief Description of the Figures, lips were washed and cells were fixed in methanol/acetone (1:1) for 5 minutes at room temperature and subsequently incubated in blocking buffer (0.5% BSA and 0.1% TX-100 in PBS) for s at room temperature. Specimens were then ted with anti-alpha- tubulin (Sigma, 1:2000) for 1 hour at room temperature in blocking buffer. After several washing steps cells were ted with AlexaFluor-488 goat-anti-mouse IgG (Molecular Probes, 1:3000) for 1 hour at room temperature followed by several washing steps with blocking buffer. Specimens were then mounted with ProLong Gold antifade (Molecular Probes, sealed with nail polish and examined with a Leica immunofluorescence microscope. Images were captured with a cooled mera and processed by ImageJ software.
Colony Outgrowth Assay: Single cell suspensions of patient-derived tumour xenografts (maintained in nude mice) were ed. For colony outgrowth assays, cells were plated in soft agar in 24-well plates according to the assay introduced by Hamburger & Salmon (Primary bioassay of human tumour stem cells, Science , 1977,197:461-463). 2x104- 6x10 4 cells in 0.2 mL medium containing 0.4% agar were plated out on a bottom layer of 0.75% agar. Test compounds were applied in 0.2 mL culture medium. Every 24-well plate ned untreated ls and samples in triplicates. Cultures were incubated at 37°C and 7.5% CO2 for 5 - 28 days. 24 hours prior to analysis, vital colonies were d with a solution of metabolisable tetrazolium salt (Alley MC et al, Life Sci. 1982, 31:3071-3078) and were counted with an automatic image analysis system (Omnicon 3600, Biosys GmbH). ve drug effects were expressed by the ratio of the mean number of colonies in the treated wells and the control wells. IC70 -values were determined by plotting compound concentrations versus relative colony counts.
Generation and Crystal Violet Assay of BAL27862-Resistant Cell Lines BAL27862-resistant sublines of human non-small cell lung cancer (H460 ATCC reference HTB-177; A549 ATCC reference CCL-185), ovarian cancer (SKOV3 ATCC reference HTB-77) lines were ted by long-term selection in complete cell culture medium (RPMI-1640 ning 10% FCS; Sigma-Aldrich) by stepwise increasing concentrations of BAL27862. Dependent on the cell line, the selection process was carried out for 8-12 months in order to achieve resistance factors (ratio of IC50 of resistant cell line and appropriate wild-type cell line) between 3 and 11.6.
The resistant sublines were expanded at the highest tolerated 62 concentration and subsequently frozen and stored in liquid nitrogen.
Cells were seeded in 96 well plates at the following densities: A549: 2000, H460: 1000, SKOV3: 2000 and, after 24 hours incubation, were incubated for 72 hours with DMSO, BAL27862, colchicine, nocodazole, paclitaxel or vinblastine diluted in te medium (final concentration DMSO max. 0.5 %). After medium was d, cells were fixed and stained by adding 50 µl Crystal Violet Staining (0.2 % Crystal Violet in 50 % Methanol) per well. Plates were incubated for 1 hour at room temperature. Subsequently the stain was decanted and plates were washed 4 times with double-distilled water. Plates were air-dried for several hours. Stain was dissolved by adding 100 µl buffer (0.1 M Tris pH 7.5, 0.2 % SDS, 20 % Ethanol) per well and shaking the . Absorbance at 590 nm was measured using a SpectraMax M2e plate reader (Molecular s). Anti-proliferative IC50 values were calculated from concentration response curves using ad Prism software. Resistance factors were calculated as a ratio of 62 IC50 in the resistant line variant versus the IC50 in the parental line.
Protein Extraction Tumour extraction: Tumours were extracted in ice-cold lysis buffer containing 50 mM HEPES (pH 7.5), 150 mM NaCl, 25 mM β-glycerophosphate, 25 mM NaF, 5 mM EGTA, 1 mM EDTA, 0.1% NP40, 15 mM pyrophosphate, 2 mM sodium orthovanadate, 10 mM sodium molybdate, leupeptin (10 µg/mL), aprotinin (10 µg/mL) and 1 mM phenylmethylsulphonyl fluoride (PMSF)(1 mL extraction volume per 45 mg tumour). After homogenisation by on, lysates were adjusted to 1% NP40 and incubated on ice for 20 min. Lysates were clarified by centrifugation and frozen at -80°C.
Tumour cell line extraction: Cells were washed with ice-cold PBS containing 1 mM PMSF and with ice-cold lysis buffer (see above) without NP40. Cells were extracted in the same lysis buffer containing 1% NP40. After homogenisation, lysates were clarified by centrifugation and frozen at -80°C.
Immunoblotting/Western Blotting Immunoblotting was performed using 20 µg of total protein per lane. Total n concentration was ined with the BCA Protein Assay (Pierce). Protein was separated on a 10% SDS-gel and transferred to a PVDF membrane using Semidry Blotting (90 min, 50 mA/gel). The primary dies used for immunoblotting were as follows: Glu-tubulin antibody (available from Milipore, reference number AB3201), rabbit polyclonal, dilution 1:1000, buffer conditions: 3% BSA in PBS/0.1% Tween Actin antibody (available from Chemicon, nce number MAB1501), mouse monoclonal, dilution 1:5000, buffer conditions: 3% BSA in PBS/0.1% Tween The secondary antibodies used for immunoblotting were peroxidase- conjugated goat anti-rabbit or goat anti-mouse (available from Jackson ImmunoResearch Laboratories INC: reference number 111144 JIR and 115- 035-146 JIR), dilution , buffer conditions: 0.5% milk in PBS/0,1% Tween.
Labelled bands were revealed using a t Stella 3200 High Performance Imaging System.
Immunohistochemistry Fixation of patient-derived tumour xenografts (maintained in nude mice) was performed in 10 % neutral-buffered in containing 4 % formaldehyde for 20 – 28 hours at room ature. Fixed specimens were kept in a on of 70 % ethanol for a maximum of one week prior to dehydration and paraffin embedding according to a standard procedure, using the conditions listed below: Sequential Treatment time (hours) 70% EtOH 1 80% EtOH 2 99% EtOH 1 100% Isopropanol 0.5 100% Isopropanol 1 Xylol 0.5 Xylol 1 Xylol 1 Paraffin 1 Paraffin 2 Paraffin 2 Paraffin sections of approximately 2 µm were cut and processed by using the automated immunostainer Benchmark XT® (Roche) g the standard processing steps. The visualisation of the ic antibody staining was done with DAB (3,3-diaminobenzidine) as chromogenic substrate at a concentration of 5 mg/ml.
The following primary antibody and processing conditions were used for staining: Antibody Specification Processing Anti-Glu-Tubulin, Millipore, #AB3201, Cell conditioning 1 buffer from Roche for rabbit onal 90 minutes, antibody incubation at 37°C for 32 minutes at a dilution of 1:50 Detailed examples Example 1: A ct Mitotic Phenotype Induced by nds of general a I Treatment with compound A (BAL27862) or with compound B or compound C, induced a highly reproducible and distinct microtubule phenotype in all tumour cell lines tested (shown for compound A in A549, HeLa and SKBR3 cells in Figure 1, and for compound B and compound C in A549 cells in Figure 2). In dividing cells an apparent fragmentation of the mitotic spindle occurred, resulting in the formation of dot-like structures (Figure 1). This ype was shown to be ct from that observed with conventional microtubule targeting , such as the microtubule stabiliser paclitaxel and the microtubule destabilisers vinblastine and colchicine (Figure 3) and nocodazole (Figure 4).
Example 2: BAL27862 Overcomes Microtubule Phenotype Induced by Conventional Microtubule-targeting Drugs in a Dominant Fashion In order to show the uniqueness of its activity on microtubules, BAL27862 was tested in combination with vinblastine, colchicine and paclitaxel e 5) and nocodazole (Figure 6) using A549 cells. Treatment with vinblastine, colchicine, paclitaxel or nocodazole alone induced the mitotic microtubule phenotypes teristic of these agents. However, combination treatment with BAL27862 for the last 4 hours resulted in disruption of the ubule structures; creating a phenotype consistent with treatment of BAL27862 alone, despite the continued presence of vinblastine, colchicine, paclitaxel or nocodazole. In contrast, ng first with BAL27862 and subsequently for 4 hours in combination with vinblastine, colchicine, paclitaxel or nocodazole had no impact on the ed microtubule phenotype that was consistent with treatment with BAL27862.
These data demonstrate that compounds of formula I affect microtubule biology consistently, but in a different manner than conventional microtubule ing .
Detailed Examples according to the invention and/or disclosure Example 3: Association of high glu-tubulin expression levels with patientderived tumour cells resistant to BAL27862 treatment.
Based on colony wth assays, using tumour cells derived from 6 patient-derived tumours maintained as xenografts in mice, BAL27862-sensitive or relatively resistant tumour cells were identified from melanoma and colorectal and lung cancer (see Table 1). Concentrations at which 70% growth inhibition was observed versus controls (IC70 ) are shown in Table 1. In this table, BAL27862- ive tumour cells have IC70 values in the low nanomolar range, while BAL27862- resistant tumour cells are defined by IC70 values >600 nanomolar. Paclitaxel and stine data, using the same ex vivo assay, was available for 5 of the 6 tumour models. Of these 5 , all were resistant to treatment with paclitaxel, while 4 of 5 of these tumours were sensitive to treatment with vinblastine.
Table 1 Cancer type name Response IC70 Response to Response to to BAL27862 paclitaxel vinblastine BAL27862 [microM] Colorectal CXF 1103 Sensitive 0.022 Resistant Resistant cancer CXF 243 Resistant 0.696 Resistant Sensitive Lung LXFE 211 Sensitive 0.021 Resistant Sensitive cancer LXFE 397 Resistant > 3.5 Not known Not known Melanoma MEXF 1341 Sensitive 0.025 Resistant ive MEXF 276 Resistant > 3.5 Resistant Sensitive Immunoblotting analysis was performed in order to measure glu-tubulin levels in the same tumours ined as xenografts, using the Milipore dy e 7). The actin levels were included on the blot as a loading control.
Analysis of glu-tubulin levels indicated that glu-tubulin expression varied dramatically across all the tumours measured (Figure 7).
Based on the colony outgrowth assay and the same IC70 criteria, there was no association between paclitaxel or vinblastine resistance and high glu-tubulin levels. This is t since, for example, for the melanoma tumour type, both models were resistant to paclitaxel and yet for MEXF 1341 the glu-tubulin levels were clearly lower than in MEXF 276. The same lack of association was true for the vinca alkaloid, stine in the melanoma model, since both these tumours were sensitive to stine. Thus glu-tubulin levels were shown to be unsuitable as a reliable biomarker of resistance to the conventional microtubule agents paclitaxel and vinblastine in patient-derived tumour models.
Surprisingly, in contrast, when the BAL27862 resistance data is compared with the glu-tubulin level, glu-tubulin is shown to be higher only in the resistant s and not the sensitive tumours derived from the same tumour histotype.
Increased levels were therefore consistently indicative of resistance to BAL27862.
Thus glu-tubulin levels were shown to be a biomarker of ance for the compound described herein, BAL27862. e 4: Immunohistochemical analysis of colorectal tumour xenografts Immunohistocehmical analysis was performed on the colorectal tumour xenografts (Figure 8), ing a high level of glu-tubulin in the tumour model CXF 243. Again a clear ation was seen between high levels of glu-tubulin and resistance to BAL27862 (tumour model CXF 243 was BAL27862-resistant, while tumour model CXF 1103 was BAL27862-sensitive; as d by the colony outgrowth assay). Thus glu-tubulin levels were again shown to be a biomarker of resistance for the compound described herein, BAL27862.
Example 5: Higher glu-tubulin expression is observed in tumour lines selected for resistance to a compound of general formula I In vitro selection for resistance to BAL27862 resulted in the generation of three relatively resistant tumour cell lines, with the following resistance factors versus parental lines (based on IC50 determinations using the l Violet assay): A549 (3.0 fold); SKOV3 resistant 1 (7.6 fold); SKOV3 resistant 2 (11.6 fold); H460 (5.3 Table 2).
Table 2: Resistance factors (ratio of IC50 BAL27862-resistant cell line Treatment variant and IC50 al cell line) compound SKOV3 SKOV3 A549 H460 resistant 1 resistant 2 BAL27862 3.0 5.3 7.6 11.6 Colchicine 0.9 1.6 2.0 2.8 zole 1.6 1.3 3.6 3.9 Vinblastine 2.3 4.6 15.7 17.8 Paclitaxel 0.06 0.3 0.4 0.5 In general these BAL27862-resistant cells exhibited a different level of response to other microtubule destabilising agents, such as cine, nocodazole and vinblastine, as compared to BAL27862; and indeed increased sensitivity to the microtubule stabiliser paclitaxel was observed in all lines (Table 2).
Extraction and immunoblot analysis of these lines to measure the glu-tubulin levels, followed by comparison to BAL27862 resistance data, again shows that glutubulin is higher in the ant lines, as compared to the parental lines (Figure 9).
This was maintained hout resistance development in the SKOV3 cells (Figure ). These data show the association of increased glu-tubulin expression levels with acquired resistance to BAL27862.
List of abbreviations A549 human all cell lung cancer cell line AnnexinV phosphatidylserine-binding protein BCA bicinchoninic acid Bcl-2 B-cell lymphoma 2 protein BRCA1 breast cancer type 1 susceptibility protein BrdU bromodeoxyuridine BSA bovine serum albumin CA-125 cancer antigen 125 cDNA complementary deoxyribonucleic acid CREST limited scleroderma syndrome CO2 carbon dioxide CXF 243 t-derived colorectal tumour CXF 1103 t-derived colorectal tumour DAB 3,3-diaminobenzidine DMSO dimethylsulphoxide DNA deoxyribonucleic acid dUTP 2´-Deoxyuridine 5´-Triphosphate ELISA -linked immunosorbent assay ErbB-2 human epidermal growth factor or 2 ESI-MS electrospray ionisation mass-spectrometry EtOH Ethanol FACS fluorescence activated cell scan/sorting FCS/FBS foetal calf / foetal bovine serum G2/M transition from G2 to the c phase in the cell cycle HeLa human squamous cell cancer cell line HEPES 4-(2-Hydroxyethyl)piperazineethanesulphonic acid Hoe33342 2'-(4'-Ethoxyphenyl)(4-methylpiperazinyl)-2,5'-bis-1H- benzimidazole trihydrochloride trihydrate H460 human non-small cell lung cancer cell line IgG immunoglobulin G IHC Immunohistochemistry LXFE 211 Patient-derived lung tumour LXFE 397 Patient-derived lung tumour MALDI matrix-assisted-laser-desorption/ionisation massspectrometry MALDI-TOF matrix-assisted-laser-desorption/ionisation–time-of-flightmass-spectrometry MEXF 276 patient-derived melanoma MEXF 1341 patient-derived melanoma mRNA messenger ribonucleic acid MTS 3-(4,5-dimethylthiazolyl)(3-carboxymethoxyphenyl) (4-sulphophenyl)-2H-tetrazolium NaCl Sodium chloride NaF Sodium fluoride NCBI National Center for Biotechnology Information NSCLC non-small cell lung cancer NP40 Nonidet P40 PBS phosphate buffered saline PCR polymerase chain on P-gp P-glycoprotein PMSF phenylmethylsulphonyl fluoride PSA prostate-specific antigen PVDF polyvinylidene fluoride RANO response assessment for high-grade gliomas RECIST response tion criteria in solid tumours RNA ribonucleic acid 640 cell culture medium used for culturing transformed and non-transformed eukaryotic cells and cell lines SDS sodium dodecyl sulphate SEQ. ID No. sequence identification number siRNA small inhibitory ribonucleic acid SKBR3 human mammary carcinoma cell line SKOV3 human ovarian carcinoma cell line TTCP tubulin ne carboxypeptidase TTL tubulin tyrosine ligase TUNEL terminal deoxynucleotidyl transferase dUTP nick end labeling Tween-20 detergent, Polyoxyethylene sorbitan monolaurate TX-100 Triton-X100 YO-PRO fluorescent, monomeric cyanine, nucleic acid stain The term ‘comprising’ as used in this specification and claims means ‘consisting at least in part of’. When interpreting statements in this specification and claims which includes the ‘comprising’, other features besides the features ed by this term in each statement can also be present. Related terms such as ‘comprise’ and ‘comprised’ are to be interpreted in similar .
In this specification where reference has been made to patent specifications, other al nts, or other sources of information, this is generally for the purpose of providing a t for discussing the features of the invention. Unless specifically stated otherwise, reference to such external documents is not to be construed as an admission that such documents, or such sources of information, in any jurisdiction, are prior art, or form part of the common general knowledge in the art.

Claims (25)

We claim:
1. Use of glu-tubulin as a biomarker for predicting the response to a compound, wherein the compound is a compound of general formula I R3 R2 N R4 N R5 N N 5 wherein R represents phenyl, l or pyridinyl wherein phenyl is optionally substituted by one or two substituents independently selected from alkyl, halo-lower alkyl, hydroxy-lower alkyl, lower alkoxy-lower alkyl, 10 y-lower alkyl, phenyl, hydroxy, lower alkoxy, hydroxy-lower alkoxy, lower -lower alkoxy, phenyl-lower alkoxy, lower alkylcarbonyloxy, amino, monoalkylamino, dialkylamino, lower alkoxycarbonylamino, lower alkylcarbonylamino, substituted amino wherein the two substituents on nitrogen form er with the nitrogen heterocyclyl, lower alkylcarbonyl, y, lower 15 alkoxycarbonyl, cyano, halogen, and nitro; and wherein two adjacent substituents are methylenedioxy; and wherein nyl is optionally tuted by lower alkoxy, amino or halogen; X represents a group C=Y, wherein Y stands for oxygen or nitrogen substituted by 20 hydroxy or lower alkoxy; R1 represents hydrogen, lower alkylcarbonyl, hydroxy-lower alkyl or cyano-lower alkyl; R2, R3 and R6 represent hydrogen; 25 R4 and R5, independently of each other, represent hydrogen, lower alkyl or lower alkoxy; or R4 and R5 together represent methylenedioxy; and pharmaceutically acceptable salts, solvates, esters and amides of naturally occurring amino acids, small es or pegylated hydroxyl acids, salts of such 5 esters and amides, and polymorphs thereof; or wherein R represents phenyl or pyridinyl wherein phenyl is optionally substituted by one or two substituents independently 10 selected from alkyl, ower alkyl, hydroxy-lower alkyl, lower alkoxy-lower alkyl, acyloxy-lower alkyl, phenyl, y, lower alkoxy, hydroxy-lower alkoxy, lower alkoxy-lower alkoxy, phenyl-lower alkoxy, lower alkylcarbonyloxy, amino, kylamino, dialkylamino, lower alkoxycarbonylamino, lower alkylcarbonylamino, substituted amino wherein the two substituents on nitrogen form 15 together with the nitrogen heterocyclyl, lower alkylcarbonyl, carboxy, lower alkoxycarbonyl, formyl, cyano, halogen, and nitro; and wherein two adjacent substituents are enedioxy; and wherein pyridinyl is optionally substituted by lower , amino or halogen; 20 X represents oxygen; R1 represents hydrogen, lower alkylcarbonyl, hydroxy-lower alkyl or lower alkyl; R2, R3 and R6 ent hydrogen; 25 R4 and R5, independently of each other, represent hydrogen, lower alkyl or lower alkoxy; or R4 and R5 together represent methylenedioxy; and pharmaceutically acceptable salts, solvates, esters and amides of naturally occurring amino acids, small peptides or pegylated hydroxy acids, salts of 30 such esters and amides, and polymorphs thereof, and wherein the prefix lower denotes a radical having up to and including a m of 7 carbon atoms and, wherein the response is of a disease in a subject and the biomarker glu-tubulin is measured ex vivo in a sample or samples taken from the human or animal body.
2. Use according to claim 1, wherein the sample or samples are taken from the human body. 5 3. Use according to claim 1 or claim 2, wherein in the compound of general formula I R represents phenyl or pyridinyl; wherein phenyl is optionally substituted by one or two substituents independently selected from lower alkyl, lower alkoxy, amino, acetylamino, halogen and nitro; 10 and wherein pyridinyl is optionally tuted by amino or halogen; X represents a group C=O; R1 represents hydrogen or cyano-lower alkyl; 15 R2, R
3, R4, R5 and R6 represent hydrogen; and ceutically acceptable salts, solvates, esters and amides of naturally occurring amino acids, small peptides or pegylated y acids, salts of such esters and amides, and polymorphs f, and wherein the prefix lower s a radical having up to and including a maximum of 7 carbon atoms.
4. Use according to any one of claims 1 to 3, wherein the compound is represented by the following formula N N wherein R, Y and R1 are defined as follows: R Y R1 O CH2CH2CN O H O CH2CH2CN NH N or pharmaceutically acceptable salts, solvates, esters and amides of naturally 5 occurring amino acids, small peptides or pegylated y acids, salts of such esters and amides, and polymorphs thereof.
5. Use according to any one of claims 1 to 4, wherein the compound is N N N N O or pharmaceutically acceptable salts, solvates, esters and amides of naturally ing amino acids, small peptides or pegylated hydroxy acids, salts of such esters and amides, and polymorphs thereof.
6. Use according to any one of claims 1 to 5, wherein the prefix lower 5 denotes a radical having up to and including a maximum of 4 carbon atoms.
7. Use according to any one of claims 1 to 6, wherein an amide of the compound of formula I with glycine, alanine or lysine is used as a pro-drug, the amide being formed from an amino group present within the R group of the compound of general formula I as defined in any one of claims 1 to 5 and the carboxy 10 group of e, alanine or lysine.
8. Use according to any one of claims 1 to 7, wherein the compound is N N N N O 15 or a pharmaceutically acceptable salt thereof.
9. Use according to claim 8, wherein the pharmaceutically acceptable salt is a hloride salt or dihydrochloride salt.
10. Use according to any one of claims 1 to 9, for predicting the resistance of the disease in the t to said compound.
11. Use according to any one of claims 1 to 10, wherein the disease is a stic disease or mune disease.
12. Use according to claim 11, n the disease is cancer.
13. Use according to any one of claims 1 to 11, wherein the disease is 5 selected from the group consisting of breast cancer, prostate cancer, cervical cancer, ovarian cancer gastric cancer, colorectal cancer, pancreatic cancer, liver cancer, brain cancer, neuroendocrine cancer, lung cancer, kidney cancer, logical malignancies, melanoma and sarcomas.
14. Use according to claim 13, wherein the disease is ed from the 10 group consisting of breast cancer, cervical cancer, ovarian cancer, colorectal cancer, lung cancer and melanoma.
15. Use according to claim 14, n the disease is selected from the group consisting of ovarian cancer, colorectal cancer, lung cancer and melanoma.
16. Use according to any one of claims 1 to 15, wherein a higher level of 15 glu-tubulin in the sample from the subject relative to a standard value or set of standard values predicts resistance.
17. Use according to claim 16, wherein higher glu-tubulin levels in a sample or samples i) relative to a standard value or set of standard values from subjects with 20 the same tumour histotype; or ii) taken after treatment initiation and ed to a sample or samples taken from the same subject before treatment tion; or iii) ve to a standard value or set of standard values from normal cells or tissue; are predictive of resistance.
18. Use according to any one of claims 1 to 17, wherein the ker is used to select subjects suffering or predisposed to suffering from a disease for treatment with a nd of general formula I or with a pharmaceutically 5 acceptable salt, solvate, ester or amide of a naturally occurring amino acid, small peptide or pegylated hydroxy acid, or with a salt of such ester or amide or with a polymorph thereof as defined in any one of claims 1 to 9.
19. Use according to claim 18, wherein the disease is cancer.
20. Use according to any one of claims 1 to 19, wherein the sample is 10 derived from normal tissue, tumour tissue, or circulating tumour cells.
21. Use according to claim 20, wherein the sample is derived from tumour tissue.
22. A method for predicting in a t suffering from a cancer the response of that cancer to a compound of general a I or to a pharmaceutically 15 acceptable salt, solvate, ester or amide of a naturally occurring amino acid, small peptide or pegylated hydroxy acid, or to a salt of such ester or amide, or to a polymorph f, as defined in any one of claims 1 to 9, comprising the steps of: a) measuring ex vivo a level of glu-tubulin in a sample pre-obtained from tumour tissue or circulating tumour cells of the subject to obtain a value 20 or values representing this level; and b) comparing the value or values from step a) to a standard value or set of standard values from ts with the same cancer type, wherein a higher glu-tubulin level in the sample relative to the standard value or set of standard values is tive of resistance of the subject's cancer to the 25 compound of formula (I) or to the pharmaceutically acceptable salt, solvate, ester or amide of a naturally occurring amino acid, small peptide or pegylated hydroxy acid, or salt of such ester or amide, or polymorph thereof.
23. Use of a compound of general formula I or of a pharmaceutically acceptable salt, solvate, ester or amide of a naturally ing amino acid, small peptide or pegylated hydroxy acid, or polymorph thereof as defined in any one of 5 claims 1 to 9, for the preparation of a pharmaceutical composition for treating a cancer in a t in need thereof, wherein the subject is selected for treatment with the nd of general formula I or with the salt, solvate, ester or amide of a naturally occurring amino acid, small peptide or pegylated hydroxy acid, salt of such ester or amide, or polymorph thereof as defined in any one of claims 1 to 9, if the 10 level of glu-tubulin, measured ex vivo in a sample taken from the subject, is not higher than a standard value or set of rd values from subjects with the same tumour histotype or from normal cells, tissue or body fluid.
24. A kit when used for predicting the response to a compound of general formula I or a pharmaceutically acceptable salt, solvate, ester or amide of a naturally 15 occurring amino acid, small peptide or pegylated hydroxy acid, salt of such ester or amide, or polymorph thereof, as defined in any one of claims 1 to 9, comprising reagents necessary for measuring a level of glu-tubulin in a sample taken from a subject with a cancer, comprising a compound of the ing formula N N N N O 20 or a pharmaceutically acceptable salt f and further comprising a comparator module which comprises a rd value or set of standard values of a level of glu-tubulin taken from samples of tumour tissue or circulating tumour cells of subjects with a cancer of the same histotype to which the level of bulin in the sample is compared. 5
25. The kit according to claim 24, wherein the reagents comprise a capture reagent comprising a detector for glu-tubulin and a detector reagent.
26. The kit according to claim 25, n the capture reagent is an antibody.
27. The kit according to any one of claims 24 to 26, wherein the 10 pharmaceutically acceptable salt of the compound is the dihydrochloride salt.
28. A use as d in any one claims 1 to 21 or 23, substantially as herein described with reference to any example f.
29. A method as claimed in claim 22, substantially as herein described with 15 reference to any example thereof.
31. A kit as claimed in any one of claims 24 to 27, substantially as herein described with reference to any example thereof.
NZ611525A 2011-01-21 2012-01-19 Use of glu-tubulin as a biomarker of drug response to furazanobenzimidazoles NZ611525B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP11151681 2011-01-21
EP11151681.1 2011-01-21
PCT/EP2012/050814 WO2012098203A1 (en) 2011-01-21 2012-01-19 Use of glu-tubulin as a biomarker of drug response to furazanobenzimidazoles

Publications (2)

Publication Number Publication Date
NZ611525A NZ611525A (en) 2015-06-26
NZ611525B2 true NZ611525B2 (en) 2015-09-29

Family

ID=

Similar Documents

Publication Publication Date Title
US10067120B2 (en) Use of acetylated tubulin as a biomarker of drug response to furazanobenzimidazoles
US10656162B2 (en) Use of glu-tubulin as a biomarker of drug response to furazanobenzimidazoles
US10724072B2 (en) Use of phospho-Akt as a biomarker of drug response
EP2666015B1 (en) Use of stathmin as a biomarker of drug response to furazanobenzimidazoles
AU2012208520A1 (en) Use of BUBR1 as a biomarker of drug response to furazanobenzimidazoles
AU2012208521B9 (en) Use of stathmin as a biomarker of drug response to furazanobenzimidazoles
NZ611525B2 (en) Use of glu-tubulin as a biomarker of drug response to furazanobenzimidazoles
NZ613022B2 (en) Use of acetylated tubulin as a biomarker of drug response to furazanobenzimidazoles
NZ614718B2 (en) Use of phospho-akt as a biomarker of drug response
NZ611805B2 (en) Use of bubr1 as a biomarker of drug response to furazanobenzimidazoles