MX2012012986A - Methods for predicting sensitivity to treatment with a targeted tyrosine kinase inhibitor. - Google Patents

Methods for predicting sensitivity to treatment with a targeted tyrosine kinase inhibitor.

Info

Publication number
MX2012012986A
MX2012012986A MX2012012986A MX2012012986A MX2012012986A MX 2012012986 A MX2012012986 A MX 2012012986A MX 2012012986 A MX2012012986 A MX 2012012986A MX 2012012986 A MX2012012986 A MX 2012012986A MX 2012012986 A MX2012012986 A MX 2012012986A
Authority
MX
Mexico
Prior art keywords
level
sample
subject
marker
cancer
Prior art date
Application number
MX2012012986A
Other languages
Spanish (es)
Inventor
Barry L Dowell
Ke Zhang
Evelyn M Mckeegan
Peter Ansell
Viswanath Devanarayan
Original Assignee
Abbvie Inc
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 Abbvie Inc filed Critical Abbvie Inc
Publication of MX2012012986A publication Critical patent/MX2012012986A/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • 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

Abstract

Methods and kits for predicting the sensitivity of a cancer to treatment with a targeted tyrosine kinase inhibitor are disclosed.

Description

METHODS TO PREDICT SENSITIVITY TO TREATMENT WITH A TIROSINE KINASE INHIBITOR ADDRESSED Related Request Information The present application claims the priority of the US Patent Application Serial No. 61 / 332,545 filed on May 7, 2010, and that it is incorporated in its entirety to the present invention as a reference.
Field of the Invention The present disclosure relates generally to the evaluation and / or treatment of a subject who has or is suspected to have a neoplastic condition, and in particular, to the use of biomarkers to identpatients receptive to a certain drug therapy, and they allow the monitoring of the patient's response to said therapy.
Background of the Invention The genetic homogeneity of cancer is a factor that complicates the development of effective cancer drugs. Cancers that are considered to be a simple disease entity, according to classical histopathological classification, often reveal multiple genomic subtypes when subjected to molecular profiling. In certain cases, certain genomic subtypes seem to have functional relevance for the efficacy of certain drugs. For example, the efficacy of certain targeted cancer drugs has been correlated with the presence of a genomic characteristic, such as an amplification of a gene. (See, for example, TJ Lynch et al., "Activation of mutations in the underlying response of epidermal growth receptor receptor for non-small cell lung cancer to gefitinib", N. Engl. J. Med., 350: 2129-2139, 2004.) Clinical studies have also identified certain plasma and serum markers that can be used to subclasspatients with lung cancer. The National Association of Clinical Biochemistry (National Association of Clinical Biochemistry) has published guidelines and practice recommendations for the use of tumor markers in the clinic. (See, for example, the Guidelines and Recommendations of the NACB Practice for the Use of Tumor Markers in the Clinic, Section 3P Lung Cancer). A pattern of tumor marker release has been correlated with the histologic history of the tumor, and may reveal mixed histological components. Table 1 summarizes the correlation of the CYFRA 21-1, CEA, NSE and ProGRP markers with tumor histology.
Table 1 neuron-specific enolase; ProGRP, progastrin release peptide; SCCA, squamous cell carcinoma antigen.
Although the correlation of certain markers with certain subclasses of lung cancer may be useful in distinguishing between different histological subtypes, the functional importance of these markers is generally not well understood.
ABT-869 (Linifanib) [N- (4- (3-amino-1 H -indazol-4-yl) phenyl) - N '- (2-fiuoro-5-methylphenyl) urea]), is a kinase inhibitor of multiple-directed receptor tyrosine that has been shown to inhibit all members of the VEGF and PDGF receptor families (eg, IC50 value of 4 nM KDR), and to have less activity (IC50 values> 1 μ?) against kinases of unrelated receptor tyrosine, soluble tyrosine kinases and serine / threonine kinases. In addition, it exhibits powerful effects antiproliferative and apoptotic in tumor cells that depend on the constitutively active and mutant kinase FLT3 and KIT. Despite its potent antitumor activity, many types of malignant cells are refractory to ABT-869. The cause of resistance is unknown.
Due to the potential therapeutic use of ABT-869 against various cancers, adjuvant diagnostic assays are needed that could identthe patients most receptive to ABT-869 therapy. In addition, there is a need for diagnostic methods that can be used to monitor the effectiveness of therapy with ABT-869. There is an additional need for complementary assays using markers that can be measured in easily obtainable tissue samples, such as blood or a blood plasma fraction.
Brief Description of the Invention • Levels of neuron-specific enolase (NSE) markers, soluble fragments in serum of cytokeratin 19 (CYFRA 21-1), cancer antigen 125 (CA 125) and embryonic carcinoma antigen (CEA), have been discovered as indicative of the sensitivity of the cancer of a subject to the administration of the drug ABT-869. The methods and equipment described herein are based in part on the discovery that any one or more of: an NSE level below a predetermined level for NSE, a level of CA125 below a predetermined level for CA125, a level of CEA above a predetermined level for CEA, and a level of CYFRA 21-1 below a predetermined level for CYFRA 21-1, or any combination thereof, indicates increased sensitivity of the subject's cancer to the administration of ABT-869, in relative form to a subject that does not have a comparable level of any of the markers.
Accordingly, in one aspect, the present disclosure provides a method for predicting the sensitivity of a cancer in a subject to the administration of ABT-869 to the subject, wherein the method comprises the steps of: determining in a sample obtained from the subject , a level of at least one marker selected from the group consisting of: neuron-specific enolase (NSE), soluble fragments in serum of cytokeratin 19 (CYFRA 21-1), cancer antigen 125 (CA 125) and embryonic carcinoma antigen (CEA), wherein any of: a level of NSE below a predetermined level for NSE, a level of CA125 below a predetermined level for CA125, a level of CEA above a predetermined level for CEA, a level of CYFRA 21 -1 below a predetermined level for CYFRA 21-1 or any combination thereof, indicates increased sensitivity of the subject's cancer to the administration of ABT-869, relative to a subject with a level of NSE, CYFRA 21-1 or CA 125 above the predetermined level for each marker, or to a subject with a CEA level below the predetermined level for each marker. The cancer can be non-small cell lung cancer. The method may comprise, for example, determining the levels of at least two markers selected from the group consisting of: NSE, CA125, CYFRA21-1 and CEA. The method may comprise determining the levels of NSE, CA125, CYFRA21-1 and CEA. The method may further comprise, for example, generating a marker signature for the subject from the levels of two or more markers, wherein a marker signature having a predetermined pattern indicates an increased sensitivity of the subject to the administration of ABT-869, relative to a marker signature that lacks the predetermined pattern. The method may further comprise comparing the levels of two or more markers in the sample with levels of the same markers in a control sample, by applying a classification tree analysis. The classification tree analysis can be carried out through a computer process.
In another aspect, the present disclosure provides a method for predicting the sensitivity of a cancer in a subject to the administration of ABT-869, wherein the method comprises: determining in a sample obtained from the subject marker levels in a marker panel comprising NSE, CA125, CYFRA 21-1 and CEA, and comparing the level of each marker in the sample, with a predetermined level of each marker, wherein the level of each marker in the sample relative to the predetermined level for each marker, indicates the sensitivity of the cancer to the administration of ABT-869 to the subject. In the method, comparing the level of each marker in the sample with a predetermined level for each marker, comprises comparing the marker levels with a level of each of the markers in a reference sample, wherein the reference sample contains each of the markers at a level that corresponds to the predetermined level of each marker. The cancer can be non-small cell lung cancer. In the method, the NSE level in the subject sample may be, for example, below the predetermined level for NSE, the level of CA125 in the subject sample may be below the predetermined level for CA125, the level of CYFRA 21-1 in the sample of the subject may be below the predetermined level for CYFRA 21-1, or the CEA level in the subject's sample is above the predetermined level for CEA, or any combination of the four conditions may be present. The method may further comprise generating a marker signature for the subject, from the levels of the one or more markers, wherein a marker signature having a predetermined pattern indicates an increased sensitivity of the subject to the administration of ABT-869, relative to a subject having a marker number that lacks the predetermined pattern. The method may further comprise comparing the levels of the markers in the subject sample, with levels of the markers in the reference sample, by applying a classification tree analysis. The classification tree analysis can be carried out, for example, through a computer process.
In another aspect, the present disclosure provides a method for classifying one or more subjects having each, 0 is suspected of having cancer, for the anticipated efficacy of the administration of ABT-869 for the treatment of cancer, wherein the method comprises determining in a sample of each subject, the level of at least one marker selected from the group consists of: NSE, CYFRA 21-1, CA125 and CEA, where either of: a reduced level of NSE relative to the NSE level in a reference sample, a reduced level of CA125 relative to the level of CA125 in the reference sample , a reduced level of CYFRA 21-1 relative to the level of CYFRA 21-1 in the reference sample, a high level of CEA relative to the level of CEA in the reference sample, or any combination thereof, indicates the sensitivity of the cancer to the administration of ABT-869 to the subject. The method may further comprise classifying each subject as sensitive to treatment with ABT-869, based on the level of at least one of NSE, CA125, CYFRA 21-1 and CEA. In the method, the subject or subjects may have or are suspected of having non-small cell lung cancer. According to the method, for example, the level of NSE in the sample of the subject can be reduced relative to the level of NSE in the reference sample. The level of CA125 in the sample of the subject can be reduced relative to the level of CA125 in the reference sample. The level of CYFRA 21-1 in the subject's sample can be reduced relative to the level of CYFRA 21-1 in the reference sample. The level of CEA in the sample of the subject may be elevated relative to the level of CEA in the reference sample. The method may further comprise generating a marker signature for each subject from the levels of one or more markers, wherein a marker signature having a predetermined pattern indicates an increased sensitivity of the subject to the administration of ABT-869, relative to a subject that has a marker signature that lacks the default pattern. The method can further comprise comparing the levels of the markers in each sample of the subject, with levels of the same markers in the reference sample, by applying a classification tree analysis, which can be carried out through a computer process. . In any of the above methods, the sample may be a blood sample, including a serum or plasma sample. In any of the above methods, the sample can be a blood sample, including a serum or plasma sample. Any of the above methods may further comprise the step of obtaining the sample from the subject. In any of the above methods, the level of each marker can be determined for example by immunohistochemistry or immunoassay.
In another aspect, the present disclosure provides a kit for predicting the sensitivity of a cancer in a subject to the administration of ABT-869 to the subject, wherein the method comprises: a) a formation comprising one or more binding reagents, in wherein each binding reagent has independent binding specificity for at least one marker selected from the group consisting of NSE, CA125, CYFRA 21-1 and CEA, wherein each binding reagent is linked independently to a separate location in at least a substrate; and b) a control sample containing a predetermined level of the marker or markers in the formation, wherein the predetermined level of each marker is a relative level at which a level for said marker indicates a sensitivity of the subject's cancer to the administration of ABT-869. The cancer for which the equipment is configured to predict administration sensitivity of ABT-869 may be non-small cell lung cancer. In the equipment, the level of NSE in the control sample can be, for example, a level below which a Level of NSE in a sample of the subject is indicative of the cancer sensitivity of the subject to the administration of ABT-869. The level of CA125 in the control sample may be a level below which a level of CA125 in a subject sample is indicative of the cancer sensitivity of the subject to the administration of ABT-869. The level of CYFRA 21-1 in the control sample may be a level below which a level of CYFRA 21-1 in a subject's sample is indicative of the cancer sensitivity of the subject to the administration of ABT-869. The level of CEA in the control sample, may be a level above which a CEA level in a subject's sample is indicative of the subject's cancer sensitivity to the administration of ABT-869. In the equipment, one or more substrates each may comprise a solid support coupled to a detectable label. The detectable label may comprise, for example, a fluorescent compound. The equipment may further comprise instructions for determining the level of each marker in a sample of the subject. The sample of the subject may be a blood sample, including a plasma sample or a serum sample.
In another aspect, the present disclosure provides a kit for predicting the sensitivity of a cancer of a subject to the administration of ABT-869 to the subject, wherein the kit comprises: a) a microformation of markers comprising one or more selected from the group consisting of NSE, CA125, CYFRA 21-1, CEA and the truncated forms thereof, and b) a control sample containing a predetermined level of the marker or markers, wherein the predetermined level of each marker is a level relative to the which a level for said marker, indicates a sensitivity of the subject's cancer to the administration of ABT-869. The cancer for which the equipment is configured to predict the sensitivity of ABT-869 administration may be non-small cell lung cancer. In the equipment, the level of NSE in the control sample can be, for example, a level below which a level of NSE in a subject's sample is indicative of the cancer sensitivity of the subject to the administration of ABT- 869. The level of CA125 in the control sample may be a level below which a level of CA125 in a subject's sample is indicative of the cancer sensitivity of the subject to the administration of ABT-869. The level of CYFRA 21-1 in the control sample may be a level below which a level of CYFRA 21-1 in a subject sample is indicative of the cancer sensitivity of the subject to the administration of ABT-869. The level of CEA in the control sample may be a level above which a CEA level in the subject's sample is indicative of the cancer sensitivity of the subject to the administration of ABT-869. In the equipment, one or more substrates may each comprise a solid support coupled to a detectable label. The detectable label may comprise, for example, a fluorescent compound. The equipment may further comprise instructions for determining the level of each marker in a sample of the subject. The sample of the subject may be a blood sample, including a plasma sample or a serum sample.
Brief Description of the Drawings Figure 1 is a Kaplan Meier plot showing General Survival (OS) in days for three different cohorts of patients in stage ¾ NSCLC treated with or without ABT-869.
Figure 2 is a set of Kaplan-Meier tracings for a cohort of patients (M05-780) treated with Alimta® with or without ABT-751, where the OS of each of the eight evaluated markers is plotted, according to the baseline plasma level of marker compared to an average NSCLC threshold value.
Figure 3 shows two Kaplan Meier tracings based on the analysis of a group of patients ("Cluster 2") characterized by an increased OS after treatment with ABT-869 relative to patients in the group not treated with ABT-869 .
Detailed description of the invention A. Definitions Section headers, as used in this section and throughout the description, are not intended to be limiting. a) As used in the present invention, the singular forms "a", "one, one" and "the" include plural referents unless the context clearly indicates otherwise. For the mention of the numerical ranges of this document, each intervention number that exists between them with the same degree of precision, is contemplated explicitly. For example, for the range 6-9, the numbers 7 and 8 are contemplated in addition to 6 and 9, and for the range 6.0-7.0, they are explicitly referred to the numbers 6.0, 6.1, 6.2, 6.3, 6.4, 6.5 , 6.6, 6.7, 6.8, 6.9 and 7.0. b) Neuron-specific enolase ("NSE") As used interchangeably herein, the terms "neuron-specific enolase" and "NSE" refer to a protein encoded by the human gene also known as enolase 2 (official symbol EN02), and conservative variants of the same. As used in the present invention, the term "official symbol" refers to that used in the EntrezGene database maintained by the National Center for Biotechnology Information of the United States (United States National Center for Biotechnology Information). c) Cancer Antigen 125 ("CA125") As used interchangeably herein, the terms "Cancer Antigen 125" and "CA125", refer to a carbohydrate antigen recognized as a tumor marker for ovarian cancer, and derived from Mucin 16, associated in the cell surface, also known as MUC16, which is a protein encoded by the human UC16 gene (official symbol MUC16), and the conservative variants of CA125. d) Soluble fragments in serum of cytokeratin 19 ("CYFRA 21-1") As used interchangeably in the present invention, the terms "Soluble serum fragments of cytokeratin 19" and "CYFRA 21-1" refer to an antigen recognized as a tumor marker for multiple cancers including lung cancer, and derivatives of cytokeratin 19, which is a protein encoded by the human 19 keratin gene (official symbol K T19), and the conservative variants of KT19. e) Carcinoembryonic antigen ("CEA") As used interchangeably herein, the terms "Carcinoembryonic antigen" and "CEA" refer to the human protein having the amino acid sequence under GenBank Access No. CAE75559, and conservative variants thereof. f) Detectable Label As used herein, the term "detectable label" refers to any portion that generates a measurable signal by optical, electrical or other physical indication, of a change in the state of a molecule or molecules coupled to the portion. Said physical indicators comprise spectroscopic, photochemical, biochemical, immunochemical, electromagnetic, radiochemical and chemical means, such as but not limited to fluorescence, chemofluorescence, chemiluminescence and the like. g) Subject As used in the present invention, the terms "subject" and "patient" are used interchangeably regardless of whether the subject has passed, or is currently going through any form of treatment. As used in the present invention, the terms "subject" and "subjects" refer to any vertebrate, including but not limited to a mammal (e.g., cow, pig, camel, llama, horse, goat, rabbit, sheep) , hamster, guinea pig, cat, dog, rat and mouse, a non-human primate (for example, a monkey such as a cynomolgus monkey, chimpanzee, etc.) and a human). Preferably, the subject is a human. h) Test sample As used in the present invention, the term "test sample" refers generally to a biological material that is tested for, and / or suspected of containing, one or more cancer markers. The biological material can be derived from any biological source. Examples of biological materials include, but are not limited to, a peripheral blood sample, a tumor, or a tissue suspected of having a tumor, a cytological thin-film sample, a sample of fine-needle aspiration, a sample of bone marrow. , a lymph node sample, a urine sample, an ascites sample, a washed sample, an esophageal brushing sample, a bladder or lung wash sample, a spinal fluid sample, a brain fluid sample, a sample of ductal aspirate, a nipple discharge sample, a pleural effusion sample, a fresh frozen tissue sample, a tissue sample embedded in paraffin or an extract or processed sample produced from any peripheral blood sample, a serum fraction or plasma of a blood sample. The test sample can be used directly as obtained from the biological source, or after pretreatment to modify the character of the sample. For example, said pretreatment may include preparing blood plasma, diluting viscous fluids, etc. The pretreatment methods can also involve filtration, precipitation, dilution, distillation, mixing, concentration, inactivation of interference components, addition of reagents, lysate, etc. If such pretreatment methods are employed with respect to the test sample, said pretreatment methods are such that the cancer cells remain in the test sample.
B. Predictive Markers of Cancer Sensitivity to ABT-869 The methods and equipment described herein are based in part on the surprising discovery that the levels of certain markers (or "biomarkers") found in a test sample obtained from a subject are predictive of a subject's cancer sensitivity to the administration of BT-869. These predictive markers include NSE, CA125, CYFRA 21-1 and CEA.
The inventive methods are particularly useful with the compound ABT-869 (Linifanib; [N- (4- (3-amino-1 H -indazol-4-yl) phenyl) -N '- (2-fluoro-5-methylphenyl) urea]), which is a competitive receptor-tyrosine kinase (RTK) inhibitor-ATP, which is a potent inhibitor of the members of the vascular endothelial growth factor (VEGF) receptor families and platelet-derived growth factor. (PDGF). See the Publication of Shankar D.B. et al, Blood, April 15: 109 (8), 3400-8 (2007). The chemical structure of ABT-869 is: Other synthetic methods for ABT-869 have been described (see for example the Publication of A. Kruger et al, Org. Process Res. Dev. 13 (6), 1419-25 (2009)). The pharmaceutical compositions containing ABT-869 and routes and methods for their administration for cancer therapy, are known and describe in detail, for example, in US Patent Application Serial No. 11 / 636,189 (US 2007/0135387), the entire description of which is incorporated herein by reference.
A predictive marker is any marker that can be found and measured in a test sample from a subject, such as a blood sample that can be a plasma or serum sample, wherein the level (ie the amount) of the marker in the sample is correlated with the response of a cancer to a compound and / or class of specific therapeutic compounds. As described in the present invention, the NSE, CA125, CYFRA 21-1 and CEA markers have been found to be predictive of a subject's sensitivity, or rather more specifically, the sensitivity of a cancer of the subject to the treatment with ABT-869, through which the administration of ABT-869 is understood. To determine the correlations of markers with clinical outcome, and more specifically with sensitivity to ABT-869, marker concentrations in subjects having a particular cancer of interest, are measured for example at a time point of departure for a measure of baseline, and subsequently at a second time point in approximately three weeks, for example on day 21 or 22 after the start of a treatment regimen. Marker threshold values or "slices" can be established for example as the average for the particular cancer type, or using any statistical method through which said cut-off value can be selected within a distribution of values. For each marker, subjects are categorized as having a marker level above or below the threshold value. Survival, such as General Survival, is subsequently determined as a function of the treatment class, and compared for each marker and treatment.
Therefore, for each marker, a predetermined cut level is identified and a reference level is provided which can subsequently be used according to the methods and equipment described herein. More specifically, and as described elsewhere in the present invention, a level of NSE below a predetermined level for NSE, a level of CYFRA 21-1 below a predetermined level for CYFRA 21-1, a level of CA125 below a predetermined level for CA 125, a CEA level above a predetermined level for CEA, or any combination thereof, indicates increased sensitivity of the subject's cancer to the administration of ABT-869, relative to a subject with a level of NSE, CA125 or CYFRA 21-1 above the predetermined level for each marker, or for a subject with a CEA level below the predetermined level for CEA.
Normally the level of each marker in the test sample of the subject is determined using an immunohistochemical or immunoassay technique, such as for example an enzyme immunoassay (EIA), and for which there are equipment commercially available from a number of commercial providers. An exemplary microparticle enzyme immunoassay technology is the AXSYM® System available from Abbott Laboratories. The assay can involve a multiplex technique, through which the levels of two or more markers can be determined from the production of a simple assay process. The marker level of any two or more of NSE, CA125, CYFRA 21-1 and CEA in a test sample, can be combined to produce a marker signature (sometimes referred to as a "biomarker profile"), the which is characterized by a pattern composed of at least two or more marking levels. For e] emp \ o, one of said e] emp \ o patterns is composed of a level of NSE due \ o of a predetermined cut for NSE, together with one or more of a level of CA125 below the default cut for CfKMb , a level of CYFRA 21-1 below the default cut for CYFRA 21-1, and a CEA level above the default cut for CEA. The marker signature may include the level of one or more markers in addition to NSE, CA125, CYFRA 21-1 and CEA. A marker signature having a predetermined pattern, that is, satisfying certain criteria such as a cutoff criterion for each of at least two markers, indicates an increased sensitivity of the subject to the administration of ABT-869, relative to a marker number that lacks the default pattern. The use of these markers in the methods and equipment of the present disclosure provides a basis for developing a targeted cancer therapy using ABT-869. The methods may be especially useful, for example, as a basis for adjunctive trials for ABT-869 therapy, which is administered to a subject either as a monotherapy or as part of a combination therapy with other chemotherapy, such as conventional chemotherapy. . The methods can be carried out in relation to any type of cancer for which, it is determined that the marker levels are predictive of the sensitivity of the cancer to the administration of ABT-869. One of the exemplary cancers is any carcinoma, such as non-small cell lung cancer, or any solid tumor.
C. Methods The methods for predicting the sensitivity of a cancer in a subject to the administration of ABT-869 to the subject, involve determining the level of at least one of the predictive markers as described herein, ie, neuron-specific enolase (NSE) , cancer antigen 125 (CA125), soluble fragments in serum of cytokeratin 19 (CYFRA 21-1) and embryonic carcinoma antigen (CEA). Any one or more of: 1) an NSE level below a predetermined level for NSE, 2) a level of CA125 below the predetermined level for CA125, CYFRA 21-1, below a predetermined level for CYFRA 21-1 and 3) a level of CEA above a predetermined level for CEA, or any combination thereof, indicates increased sensitivity of the subject's cancer to the administration of ABT-869 compared to a subject having a level of NSE, CA125 or CYFRA 21-1 above the predetermined level for each marker, or to a subject with a CEA level below the predetermined level for CEA. For example, methods may include determining the level of the four NSEs, CA125, CYFRA 21-1 and CEA. The cancers addressed by the present disclosure comprise any cancer for which antiangiogenic therapy such as ABT-869 therapy is contemplated, and especially any solid tumor including breast tumors and carcinomas including hepatocellular carcinoma, renal cell carcinoma, small cell carcinomas. and large cell and combinations thereof, and include for example, non-small cell lung cancer (NSCLC).
A cancer or a subject (patient) can be described as sensitive to, or resistant to a selected therapeutic drug regimen, which includes the administration of ABT-869, based on the drug's ability to kill cancer cells or decrease the size of tumor and / or reduce the growth or spread of general cancer (metastasis). Cancer cells or tumors that are not sensitive are considered resistant to a therapeutic regimen, and are those that do not respond to the drug regimen, for example those in which the drug regimen fails to significantly decrease the size of the tumor or decrease the growth or spread of the tumor. Cancer cells that are sensitive to therapeutic regimen are those that do not respond to the drug regimen, resulting in decreased tumor size and / or decreased tumor growth or dispersion, and therefore also an increase in overall survival ("YOU"). The monitoring of a response to the drug regimen can be achieved through numerous pathological, clinical and imaging methods, such as those described elsewhere in the present invention, and as are generally known in the medical field. For example, tumor size can be evaluated using any soft tissue imaging technique, such as ultrasound, CT and / or DCE-MRI. It will also be understood that the methods may also involve obtaining the subject's test sample, using any tissue sampling technique including but not limited to blood extraction and finger bite, and tissue biopsy techniques, including needle biopsy.
When the levels of two or more markers are determined, the method may further comprise generating a marker signature for the subject from the levels of two or more markers. The marker signature may include for example two or more marker levels, wherein each level relative to a cutoff value for said marker defines a characteristic of the marker signature and the characteristics together form the signature. A signature that shares a predetermined pattern, i.e. a pattern that reflects marker levels, each having a relative relationship to a cut-off value for each marker, indicates an increased sensitivity of the subject to the administration of ABT-869, relative to the marker signature that lacks the default pattern. For example, a predetermined signature pattern indicative of the subject's increased sensitivity to the administration of ABT-869 and based on the marker levels of the NSE, CA125, CYFRA 21-1 and CEA, is a pattern characterized by 1) a level of NSE that is below a predetermined level for NSE, 2) a level of CYFRA 21-1 that is below a predetermined level for CYFRA 21-1, 3) a level of a level of CA125 that is below a level default level for CA125, and 3) a level of CEA that is above a predetermined level for CEA. Any signature that has all these characteristics of pattern, is an example of a signature that is indicative of the sensitivity of the subject to the administration of ABT-869.
The analysis of marker levels can also involve comparing the levels of at least two markers with levels of the same markers in a control sample, which can be carried out by applying a classification tree analysis. Classification tree analyzes are generally well known and can be easily applied for the analysis of marker levels using a computer process. For example, a 3D reference contour trace may be generated, which reflects the marker levels as described herein, which correlate with the sensitivity of a cancer to treatment with ABT-869. For any given subject, a comparable 3D trace can be generated, and the trace compared to the reference 3D trace to determine if the subject has a marker signature that indicates sensitivity of the subject to the administration of ABT-869. Classification tree analyzes are well suited to analyze marker levels because they are especially suitable for graphical display and are easy to interpret. Nevertheless, it will be understood that any computer-based application comparing multiple marker levels from two different subjects, or from a reference sample and a subject, can be used, and provide a result indicating the sensitivity of a subject to the administration of ABT-869 based on the methods described here.
The methods can be used to classify one or more subjects, where each subject has or is suspected of having a cancer, to evaluate the efficacy of anticipated administration of ABT-869 for the treatment of cancer in the subject. Said method involves determining, in a sample of each subject, the level of at least one of the markers NSE, CA125, CYFRA 21-1 and CEA and comparing the level of each marker with its level in a reference sample. The reference sample contains an amount of each marker corresponding to a predetermined cut-off value for the marker. Any of: 1) a reduced level of NSE relative to the level of NSE in a reference sample, 2) a reduced level of CYFRA 21-1 relative to the level of CYFRA21-1 in the reference sample, a high level of relative CEA At the level of CEA in the reference sample, a reduced level of CA125 relative to the level of CA125 in the reference sample or any combination thereof, indicates cancer sensitivity to administration of ABT-869 to the subject. Therefore, the methods can be used for example to target a patient population where treatment with ABT-869 is likely to produce superior results compared to alternative therapies. D. Teams The present disclosure also provides kits for predicting the sensitivity of a cancer in a subject to the administration of ABT-869 to the subject. The kit may comprise for example a formation of one or more binding reagents, and a control sample containing a predetermined level of the marker or markers, wherein the predetermined level for each marker, is a relative level at which one level for each marker, indicates a sensitivity of the subject's cancer to the administration of ABT-869. The predetermined level for each marker is for example a cut-off or threshold value determined according to a statistical analysis, for example as described elsewhere in this document, such as in the Examples section. Each binding reagent has independent binding specificity for at least one of NSE, CA125, CYFRA 21-1 and CEA. Exemplary linking reagents are antibodies. Alternatively, a piece of equipment may include a formation of two or more of the markers or shapes or truncated fragments thereof.
Antibodies A binding reagent can be for example a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a human antibody, an affinity matured antibody or an antibody fragment. A sandwich immunoassay format wherein both a capture and a detection antibody are used for each marker can be used. Antibodies can be bound, for example conjugated, to a detectable label. Although the monoclonal antibodies are highly specific for the marker / antigen, a polyclonal antibody can preferably be used as a capture antibody to immobilize the marker / antigen as much as possible. A monoclonal antibody with an inherently higher binding specificity for the marker / antigen can subsequently be used preferably as a detection antibody for each marker / antigen. In any case, capture and detection antibodies recognize epitopes without overlap in each marker, preferably without interfering with the binding of the other.
Polyclonal antibodies are raised by injecting (e.g., subcutaneous or intramuscular injection) an immunogen into a suitable non-human mammal (e.g., a mouse or a rabbit). Generally, the immunogen should induce the production of high titers of antibody with relatively high affinity for the target antigen. If desired, the label can be conjugated to a carrier protein by conjugation techniques that are well known in the art. Commonly used transporters include magnetic penetrating hemocyanin (KLH), thyroglobulin, bovine serum albumin (BSA), and tetanus toxoid. Subsequently the conjugate is used to immunize the animal. Subsequently the antibodies are obtained from blood samples taken from the animal. The techniques used subsequently are obtained from blood samples taken from the animal. The techniques used to produce polyclonal antibodies are described extensively in the literature (for example see the Publication of Methods of Enzymology "Production of Antiserum with Small Immunogen Dose: Multiple Intradermal Injections" Langone, et al. (Acad. Press. , 1981)). The polyclonal antibodies produced by the animals can be further purified, for example, by binding to and elution from a matrix to which the target antigen binds. Those skilled in the art will know the various techniques common in the immunology arts for purification and / or concentration of polyclonal as well as monoclonal antibodies (see for example the Coligan Publication, et al. (1991) Unit 9, Current Protocols in Immunology, Wiley I nterscience).
For many applications, monoclonal antibodies (mAbs) are preferred. The general method used for the production of hybridomas that secrete mAbs is well known (Kohler and Milstein (1975) Nature, 256: 495). In summary, as described by Kohler and Milstein, the technique involves isolating lymphocytes from regional extraction lymph nodes of five cancer patients, separated with either melanoma, teratocarcinoma or cancer of the cervix, glioma or lung, (where the samples are obtained from surgical specimens), sample collection, and cell fusion with SHFP-1.
Hybridomas were classified for the production of an antibody that binds to cancer cell lines. Confirmation of specificity among mAbs can be achieved using routine classification techniques (such as the enzyme-linked immunosorbent assay, or "ELISA"), to determine the elementary reaction pattern of the mAb of interest.
As used herein, the term "antibody" also encompasses antigen binding antibody fragments, eg, single chain antibodies (scFv or others), which can be produced / selected using phage display technology. The ability to express antibody fragments on the surface of viruses infecting bacteria (bacteriophage or phage) makes it possible to isolate a single binding antibody fragment, for example, from a library of more than 1010 unlinked clones. To express the antibody fragments on the phage surface (phage display), an antibody fragment gene is inserted into the gene encoding a phage surface protein (e.g. pIII) and the fragment fusion protein is shown antibody plll on the phage surface (McCafferty et al (1990) Nature, 348: 552-554; Hoogenboom et al. (1991) Nucleic Acids Res. 19: 4133-4137).
Since the antibody fragments on the surface of the phage are functional, the phage-containing antigen binding antibody fragments can be separated from the unbound phage by antigen affinity chromatography (McCafferty et al (1990) Nature, 348 : 552-554). Depending on the affinity of the antibody fragment, enrichment factors of 20 to 1,000,000 fold are obtained for a single round of affinity selection. However, by infecting the bacteria with the eluted phage, one can grow more phage and undergo another round of selection. In this way, an enrichment of 1000 times in one turn can be converted into a 1,000,000-fold increase in two selection rounds (McCafferty et al. (1990) Nature, 348: 552-554). Therefore, even when enrichments are low (Marks et al (1991) J. Mol. Biol. 222: 581-597), multiple turns of affinity selection can lead to the isolation of the rare phage. Since selection of the phage antibody library on an antigen results in enrichment, most clones bind to the antigen after as few as three to four rounds of selection. Therefore only a relatively small number of clones (a few hundred) need to be analyzed to bind the antigen.
Human antibodies can be produced without prior immunization by displaying very large and diverse V-gene repertoires in the phage (Marks et al (1991) J. Mol. Biol. 222: 581-597). In one embodiment, the natural VH and VL repertoires present in human peripheral blood lymphocytes are isolated from donors not immunized by PCR. The V gene repertoires can be divided together randomly using PCR to create a repertoire of scFv gene that can be cloned into a phage vector to create a library of 30 million phage antibodies (Id). From a single "virgin" phage antibody library, binding antibody fragments have been isolated against more than 17 different antigens, including haptens, polysaccharides and proteins (Marks et al. (1991) J. Mol. B io 1 222: 581-597; Marks et al. (1993). Bio / Technology. 10: 779-783; Griffiths et al. (1993) EMBO J. 12: 725-734; Clackson et al. (1991) Nature. 352: 624-628). Antibodies have been produced against the proteins themselves, including thyroglobulin, immunoglobulin, tumor necrosis factor and human CEA (Griffiths et al. (1993) EMBO J. 12: 725-734). Antibody fragments are highly specific for the antigen used for selection and have affinities within the range of 1 nM to 100 nM (Marks et al (1991) J. Mol. Biol. 222: 581-597; Griffiths et al. 1993) EMBO J. 12: 725-734). Larger phage antibody libraries result in the isolation of more antibodies of higher binding affinity to a proportion of antigens.
As will be readily appreciated by those skilled in the art, antibodies can also be prepared through any of a number of commercial services (eg, Berkeley Antibody Laboratories, Bethil Laboratories, Anawa, Eurogenetec, etc.).
Solid phase In equipment according to the present disclosure, each binding reagent can be linked to a solid phase. A solid phase can be any suitable material with sufficient surface affinity to bind an antibody, for example each capture antibody having a specific binding to one of the labels. The solid phase can take any of a number of forms, such as a magnetic particle, granule, test tube, microtitre plate, cuvette, membrane, a scaffold molecule, quartz crystal, film, filter paper, disk or a chip. Useful solid phase materials include: natural polymeric carbohydrates and their synthetically modified, cross-linked or substituted derivatives, such as agar, agarose, cross-linked alginic acid, substituted and cross-linked guar gums, cellulose esters, especially with nitric acid and carboxylic acid, esters of mixed cellulose and cellulose esters; natural polymers containing nitrogen, such as proteins and derivatives including cross-linked or modified gelatins; natural hydrocarbon polymers such as latex and rubber; synthetic polymers such as vinyl polymers, including polyethylene, polypropylene, polystyrene, polyvinylchloride, polyvinylacetate and their partially hydrolyzed derivatives, polyacrylamides, polymethacrylates, copolymers and terpolymers of the above polycondensates, such as polyesters, polyamides, and other polymers, such as polyurethanes or polyepoxides; inorganic materials such as sulfates or carbonates of alkaline earth metals and magnesium, including barium sulfate, calcium sulfate, calcium carbonate, alkali silicates and alkaline earth metals, aluminum and magnesium; and aluminum and silicone oxides or hydrates, such as clays, alumina, talc, kaolin, zeolite, silica gel or glass (these materials can be used as a filter with the above polymeric materials); and mixtures or copolymers of the above classes, such as graft copolymers obtained by initiating the polymerization of synthetic polymers in a pre-existing natural polymer. All of these materials can be used in suitable forms, such as films, sheets, tubes, particulates or plates, or they can be coated, bonded or laminated into suitable inert carriers, such as paper, glass, plastic films, fabrics or the like. Nitrocellulose has excellent absorption and adsorption qualities for a wide variety of reagents including monoclonal antibodies. Nylon also has similar characteristics and is also suitable. Any of the above materials can be used to form a formation, such as a microformation, of one or more specific binding reagents.
Alternatively, the solid phase can constitute microparticles. The microparticles useful in the present disclosure can be selected by one skilled in the art from any suitable type of a particulate material, and include those composed of polystyrene, polymethylacrylate, polypropylene, latex, polytetrafluoroethylene, polyacrylonitrile, polycarbonate, or similar materials . In addition, the microparticles can be magnetic or magnetic microparticles, to facilitate in this way the manipulation of the microparticle within a magnetic field. In an exemplary embodiment, the microparticles are carboxylated magnetic microparticles. The microparticles can be suspended in the mixture of soluble reagents and the test sample, or they can be retained and immobilized by a support material. In the latter case, the microparticles in or within the support material, do not have the ability to move substantially to positions anywhere within the support material. Alternatively, the microparticles can be separated from the suspension in the mixture of soluble reagents and the test sample by sedimentation or centrifugation. When the microparticles are magnetic or paramagnetic, the microparticles can be separated from the suspension in the mixture of soluble reagents and the test sample, through a magnetic field. The methods of the present disclosure can be adapted for use in systems using microparticle technology, including automatic and semiautomatic systems, wherein the solid phase comprises a microparticle. Such systems include those described in pending North American Application No. 425,651, and US Patent No. 5,089,424, which corresponds to published EPO Applications Nos. EP 0 425 633 and EP 0 424 634, respectively, and to US Patent No. 5,006,309 Other considerations that affect the choice of the solid phase include the ability to minimize the non-specific binding of tagged entities and the compatibility with the tagging system employed. For example, solid phases used with fluorescent labels must have background fluorescence low enough to allow signal detection. After binding of a specific capture antibody, the surface of the solid support can be further treated with materials such as serum, proteins or other blocking agents to minimize non-specific binding.
Detection Systems The equipment according to the present disclosure can include one or more detectable labels. One or more specific binding reagents, eg, antibodies, can be linked to a detectable label. Suitable detectable labels to be used include any compound or composition having a portion that is detectable by means of spectroscopy, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Such labels include, for example, an enzyme, oligonucleotide, nanoparticle, chemiluminophore, fluorophore, fluorescence quencher, chemiluminescent quencher or biotin.
Therefore, for example, in an immunoassay equipment configured to employ an optical signal, the optical signal is measured as a concentration of material for analysis that depends on the change in chemiluminescence, fluorescence, phosphorescence, and the chemistry I umiscence, Ultra iolet absorption, visible absorption, infrared absorption, refraction, surface plasmon resonance. In an immunoassay equipment configured to employ an electrical signal, the electrical signal is measured as a concentration of material for analysis that depends on the change in current, resistance, potential, mass to charge ratio or ion count. In an immunoassay kit configured to employ a state change signal, the state change signal is measured as a concentration of material for analysis that depends on the change in size, solubility, mass or resonance.
Useful labels according to the present disclosure include magnetic beads (e.g., Dynabeads ™), fluorescent inks (e.g., fluorescein, Texas Red, rhodamine, green fluorescent protein) and the like (see for example the Molecular Probes Publication, Eugene, Oreg., USA), chemiluminescent compounds such as acridinium (eg, acridinium-9-carboxamide), phenanthridinium, dioxetanes, luminol and the like, radiolabels (e.g., 3H, 1251, 35S, 14C or 32P), catalysts such as enzymes (e.g., horseradish peroxidase, alkaline phosphatase, beta-galactosidase and others commonly used in an ELISA), and colorimetric labels such as colloidal gold (for example, gold particles in a dispersion green light with a diameter range of 40 to 80 nm with high efficiency) or colored glass or plastic beads (for example, po i ie sti re no, polypropylene, latex, etc.). Patents that teach the use of such labels include U.S. Patent Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.
The label can be linked to each antibody, for example a detection antibody in a sandwich immunoassay format, before, or during, or after contact with the biological sample. So-called "direct labels" are detectable labels that directly bind to or are incorporated into the antibody before being used in the assay. Direct tags can be linked or incorporated into the detection antibody through any of a number of well-known means for experts in the art.
In contrast, so-called "indirect labels" usually bind to each antibody at some point during the assay. Often, the indirect label links to a portion that is attached or incorporated into the detection agent before being used. Therefore, for example, each antibody can be biotinylated before being used in an assay. During the assay, an avidin-conjugated fluorophore may bind to the biotin-containing detection agent to provide a label that is readily detected.
In another example of indirect labeling, polypeptides with the ability to specifically bind immunoglobulin constant regions, such as polypeptide A or G polypeptide, can also be used as labels for detection antibodies. These polypeptides are normal constituents of the cell walls of the streptococcal bacteria. They exhibit a strong non-immunogenic reactivity with constant immunoglobulin regions from a variety of species (see generally Kronval, et al., (1973) J. Immunol., 111: 1401-1406, and Akerstrom (1985) J. Immunol, 135: 2589-2542). Said polypeptides can therefore be labeled and added to the test mixture, where they will bind to each capture and detection antibody, as well as to the antibodies, labeling everything and providing a compound signal attributable to the material for analysis and to the antibody present. in the mix.
Some labels may require the use of an additional reagent (s) to produce a detectable signal. In an ELISA, for example, an enzyme tag (eg, beta-galactosidase) will require the addition of a substrate (eg, X-gal) to produce a detectable signal. In an immunoassay kit configured to use an acridinium compound as the direct label, a basic solution and a source of hydrogen peroxide may also be included in the kit.
Test kits according to the present disclosure preferably include instructions for determining the level of each marker in a sample of the subject, for example carrying out one or more immunoassays. The instructions may further include instructions for analyzing a test sample of a specific type, such as a blood sample, or more specifically a serum sample or a plasma sample. The instructions included in the equipment of the present description can be attached to the packaging material or can be included as a package insert. Although the instructions are usually written or printed materials, they are not limited to them. In the present description, any means with the ability to store said instructions and communicate them to an end user is contemplated. Such means include, but are not limited to, electronic storage media (e.g., magnetic disks, tapes, cartridges, chips), optical media (e.g., CD ROM), and the like. As used in the present invention, the term "instructions" may include the address of an internet site that provides the instructions.
Adaptations of the Methods of the Present Description One skilled in the art will readily appreciate that the biomarkers, oligonucleotides, methods, equipment and related compositions described herein are representative of the exemplary embodiments, and are not intended to be limitations of the scope of the present invention. It will be readily appreciated by those skilled in the art that various substitutions and modifications may be made to the present description herein described, without departing from the scope and spirit of the same.
All patents and publications mentioned in the specification indicate the levels of those skilled in the art to which the present invention pertains. All patents and publications are incorporated by reference to the extent as if each individual publication was specifically and individually indicated and incorporated as a reference.
The present description, described in illustrative form, can be carried out in an appropriate manner in the absence of any element or elements, limitation or limitations that are not specifically described in this invention. Therefore, for example, in each case in the present invention any of the terms "comprises", "consists essentially of" and "consists of" can be replaced with any of the other two terms. The terms and expressions that have been used are used as terms of description and not limitation and do not intend to use such terms and expressions, to exclude any equivalents of the characteristics shown and described or parts thereof, but it is recognized that several modifications are possible within the scope of the claimed description. Therefore, it should be understood that although this description has been described specifically to Through preferred embodiments and optional features, those skilled in the art may resort to modifications and variations of the concepts described herein, and such modifications and variations are considered to be within the scope of the present invention, as defined by the appended claims. .
EXAMPLE By way of example, and not limitation, the examples of this description are given below.
Example 1: Correlation of Markers with Clinical Outcome Based on Data Through Multiple Tests NSCLC with Different Therapeutics Three cohorts of patients distinguished by the treatment regimen for overall survival were evaluated. All patients were diagnosed with stage 3/4 NSCLC. The marker concentrations were measured by a baseline immunoassay in the NSCLC tests. The NSCLC subjects were assigned to one of three cohorts as indicated: M05-780 (N = 83) where the subjects received pemetrexed (Alimta® available from Eli Lilly and Company, Indianapolis, IN) with or without ABT-751 (( N- [2 - [(4-Hydroxyphenyl) amino] -3-pyridinyl] -4-methoxybenzenesulfonamide, available from Abbott Laboratories, Abbott Park, IL); M05-782 (N = 21), wherein the subjects received Docetaxel with or without ABT-751, and M06-880 (N = 103), where subjects received only ABT-869.
Patients were categorized as having a marker level above or below the threshold marker level. Survival as a classification function was compared for each marker and treatment. Marker threshold values were evaluated through multiple methods including but not limited to, determination of average value, statistical modeling for optimal threshold values, values determined in the community as predictive of NSCLC versus benign lung disease and comparison of concentration of the marker in patients with stable disease versus rapid progress in therapy with ABT-869.
Figure 1 is a Kaplan Meier trace showing General Survival (OS-DUR) in days for the three different patient NSCLC stage 3/4 cohorts, showing the results for 05-780 in red, for M05-782 in green and for M06-880 in blue. Figure 2 is a set of Kaplan-Meier tracings for the patient cohort (M05-780) that was treated with Alimta® with or without ABT-751, plotting OS for each of the eight plasma markers evaluated, according to with the baseline plasma level of each marker compared to an average threshold value of NSCLC. Table 2 provides a summary of the observed levels of unprocessed marker and the threshold values of seven of the eight markers in Figure 2 (Cyfra21-1, NSE, CEA, SCC, ProGRP, CA 15-3 and CA125), in patients treated with ABT-869 or treated with ABT-751.
Figure 3 shows two Kaplan Meier tracings, both based on additional analysis of a group of patients identified as Group 2. As can be seen in Figure 3, the patients in Group 2 were those who underwent the NSCLC tests. who showed a pronounced increase in OS after treatment with ABT-869, when compared with patients treated with Alimta® with or without ABT-751. Patients in cluster 2 were characterized in terms of baseline plasma marker levels and all showed one or more of an NSE level below the threshold value for NSE, a level of CYFRA 21-1 below the value threshold for CYFRA 21-1, one level of CA125 below the threshold value for CA125, one level of CEA above the threshold value for CEA.
Table 2:

Claims (60)

1. A method for predicting the sensitivity of a cancer in a subject to the administration of ABT-869 to the subject, wherein the method comprises the steps of: determining in a sample obtained from the subject, a level of at least one marker selected from the group consisting of: neuron-specific enolase (NSE), cancer antigen 125 (CA125), CYFRA 21-1 and embryonic carcinoma antigen (CEA), where any of: a level of NSE below a predetermined level for NSE, a level of CYFRA 21-1 below a predetermined level for CYFRA 21-1, a level of CA125 below a predetermined level for CA125, a level of CEA above a predetermined level for CEA, or any combination thereof, indicates an increased sensitivity of the subject's cancer to the administration of ABT-869, relative to a subject with a level of NSE, CA125 or CYFRA 21-1 above the default level for each marker, or to a subject with a CEA level below a predetermined level of each marker.
2. The method as described in the claim 1, characterized in that the cancer is non-small cell lung cancer.
3. The method as described in claim 1, characterized in that the sample is a blood sample.
4. The method as described in claim 1, characterized in that the sample is a serum or plasma sample.
5. The method as described in claim 1, characterized in that the method further comprises obtaining the sample from the subject.
6. The method as described in claim 1, characterized in that the level of each marker is determined by immunohistochemistry or immunoassay.
7. The method as described in claim 1, characterized in that it comprises determining the levels of at least two markers selected from the group consisting of: NSE, CYFRA 21-1, CA125 and CEA.
8. The method as described in claim 1, characterized in that it comprises determining the levels of NSE, CA125, CYFRA 21-1 and CEA.
9. The method as described in any of claims 7 or 8, characterized in that the method further comprises generating a marker signature for the subject from the levels of two or more markers, wherein a marker signature having a pattern predetermined, indicates an increased sensitivity of the subject to the administration of ABT-869, relative to a marker signature lacking the predetermined pattern.
10. The method as described in any of claims 7 or 8, characterized in that the method further comprises comparing the levels of two or more markers in the sample, with levels of the same markers in a control sample, by applying a tree analysis of classification.
11. The method as described in the claim 10, characterized in that the classification tree analysis is carried out through a computer process.
12. A method for predicting the sensitivity of a cancer in a subject to the administration of ABT-869, wherein the method comprises the steps of: determining in a sample obtained from the subject, marker levels in a marker panel comprising NSE, CA125, CYFRA 21-1 and CEA, and comparing the level of each marker in the sample with a predetermined level of each marker, wherein the level of each marker in the sample relative to the predetermined level of each marker, indicates the sensitivity of the cancer to the administration of ABT-869 to the subject.
13. The method as described in claim 12, characterized in that comparing the level of each marker in the sample with a predetermined level for each marker comprises comparing the marker levels with a level of each of the markers in a sample of reference, wherein the reference sample contains each of the markers at a level corresponding to a predetermined level for each marker.
14. The method as described in claim 12, characterized in that the cancer is non-small cell lung cancer.
15. The method as described in the claim 12, characterized in that the level of NSE in the sample of the subject is below the predetermined level for NSE.
16. The method as described in claim 12, characterized in that the level of CYFRA 21-1 in the subject's sample is below the predetermined level for CYFRA 21-1.
17. The method as described in rei indication 12, characterized in that the level of CEA in the sample of the subject is above the predetermined level for CEA.
18. The method as described in the claim 12, characterized in that the level of CA125 in the sample of the subject is below the predetermined level for CA125.
19. The method as described in claim 12, characterized in that the sample of the subject is a blood sample.
20. The method as described in claim 12, characterized in that the sample of the subject is a serum or plasma sample.
21. The method as described in claim 12, characterized in that the method further comprises obtaining the sample from the subject.
22. The method as described in claim 12, characterized in that the level of each marker in the sample of the subject is determined by immunohistochemistry or immunoassay.
23. The method as described in claim 12, characterized in that the method further comprises generating a marker signature for the subject from the levels of the markers, wherein a marker signature having a predetermined pattern indicates an increased sensitivity of the subject to the administration of ABT-869, relative to a subject having a marker number that lacks the predetermined pattern.
24. The method as described in claim 12, characterized in that the method further comprises comparing the levels of the markers in the sample of the subject, with levels of the markers in the reference sample, by applying a classification tree analysis.
25. The method as described in claim 24, characterized in that the classification tree analysis is carried out through a computer process.
26. A method for classifying one or more subjects who have or are suspected of having cancer, with respect to the anticipated efficacy of the administration of ABT-869, for the treatment of cancer, wherein the method comprises determine in a sample of each subject, the level of at least one marker selected from the group consisting of: NSE, CA125, CYFRA 21-1 and CEA, where either of: a reduced level of NSE relative to the level of NSE in a Reference sample, a reduced level of CYFRA 21-1 relative to the level of CYFRA21-1 in the reference sample, a high level of CEA relative to the CEA level in the reference sample, a reduced level of CA125 relative to the level of CA125 in the reference sample, or any combination thereof, indicates the sensitivity of the cancer to the administration of ABT-869 to the subject.
27. The method as described in claim 26, characterized in that the method further comprises classifying each subject as sensitive to treatment with ABT-869, based on the level of at least one of NSE, CYFRA 21-1, CA125 and CEA .
28. The method as described in claim 26, characterized in that the subject or subjects have, or are suspected to have, a non-small cell lung cancer.
29. The method as described in claim 26, characterized in that the level of NSE in the sample of the subject is reduced relative to the level of NSE in the reference sample.
30. The method as described in claim 26, characterized in that the level of CYFRA 21-1 in the subject sample is reduced relative to the level of CYFRA 21-1 in the reference sample.
31. The method as described in claim 26, characterized in that the level of CEA in the subject sample rises relative to the level of CEA in the reference sample.
32. The method as described in claim 26, characterized in that the level of CA125 in the sample of the subject is reduced relative to the level of CA125 in the reference sample.
33. The method as described in claim 26, characterized in that the sample is a blood sample.
34. The method as described in claim 26, characterized in that the sample is a serum or plasma sample.
35. The method as described in claim 26, characterized in that the method further comprises obtaining the sample from each subject.
36. The method as described in the claim 26, characterized in that the level of each marker is determined by immunohistochemistry or immunoassay.
37. The method as described in claim 26, characterized in that the method further comprises generating a marker signature for each subject from the levels of one or more markers, wherein a marker signature having a predetermined pattern indicates a sensitivity increased of the subject to the administration of ABT-869, relative to a subject having a marker number that lacks the predetermined pattern.
38. The method as described in claim 26, characterized in that the method further comprises comparing the levels of the markers in each sample of the subject, with the levels of the same markers in the reference sample, by applying a classification tree analysis.
39. A device for predicting the sensitivity of a cancer in a subject to the administration of ABT-869 to the subject, wherein the equipment comprises: to. a formation comprising one or more binding reagents, wherein each binding reagent has an independent binding specificity for at least one marker selected from the group consisting of NSE, CYFRA 21-1, CA125 and CEA, wherein each reagent link is linked independently to a separate location on at least one substrate; Y b. a control sample containing a predetermined level of the marker or markers in the formation, wherein the predetermined level for each marker is a relative level at which a level for said marker indicates a sensitivity of the subject's cancer to the administration of ABT -869.
40. The equipment as described in claim 39, characterized in that the cancer is non-small cell lung cancer.
41. The equipment as described in the claim 39, characterized in that the level of NSE in the control sample is a level below which, a level of NSE in a subject's sample is indicative of the cancer sensitivity of the subject to the administration of ABT-869.
42. The equipment as described in the claim 39, characterized in that the level of CYFRA 21-1 in the control sample is a level below which a level of CYFRA 21-1 in a subject's sample is indicative of the cancer sensitivity of the subject to the administration of ABT -869.
43. The equipment as described in the claim 39, characterized in that the level of CA125 in the control sample is a level below which, a level of CA125 in a subject's sample is indicative of the cancer sensitivity of the subject to the administration of ABT-869.
44. The equipment as described in the claim 39, characterized in that the level of CEA in the control sample is a level above which a CEA level in a subject's sample is indicative of the cancer sensitivity in the subject to the administration of ABT-869
45. The equipment as described in claim 39, characterized in that one or more substrates, each comprising a solid support coupled to a detectable label.
46. The equipment as described in claim 45, characterized in that the detectable label comprises a fluorescent compound.
47. The equipment as described in claim 39, characterized in that it also comprises instructions for determining the level of each marker in a sample of the subject.
48. The equipment as described in claim 47, characterized in that the sample of the subject is a blood sample.
49. The equipment as described in claim 47, characterized in that the sample of the subject is a plasma sample.
50. The equipment as described in the claim 47, characterized in that the sample of the subject is a serum sample.
51. A device for predicting the sensitivity of a cancer in a subject to the administration of ABT-869 to the subject, wherein the equipment comprises: to. a microformation of markers comprising one or more selected from the group consisting of NSE, CYFRA 21-1, CA125, CEA and truncated forms thereof, and b. a control sample containing a predetermined level of the marker or markers, wherein the predetermined level for each marker is a level relative to which, a level for said marker, indicates a sensitivity of the subject's cancer to the administration of ABT-869.
52. The equipment as described in claim 51, characterized in that the cancer is a non-small cell lung cancer.
53. The equipment as described in claim 51, characterized in that the level of NSE in the control sample is a level below which, a level of NSE in the sample of a subject is indicative of the cancer sensitivity of the subject to the administration of ABT-869.
54. The equipment as described in claim 51, characterized in that the level of CYFRA 21-1 in the control sample, is a level below which, a CYFRA 21-1 level in a subject's sample is indicative of the Cancer sensitivity to the administration of ABT-869.
55. The equipment as described in claim 51, characterized in that the level of CA125 in the control sample is a level below which, a level of CA125 in the sample of a subject is indicative of the sensitivity of the cancer to the administration of ABT-869.
56. The equipment as described in claim 51, characterized in that the level of CEA in the control sample is a level below which, a CEA level in a subject's sample is indicative of the sensitivity of the cancer to the administration of ABT-869.
57. The equipment as described in claim 51, characterized in that it also comprises instructions for determining the level of each marker in a sample of the subject.
58. The equipment as described in the claim 51, characterized in that the sample of the subject is a blood sample.
59. The equipment as described in claim 51, characterized in that the sample of the subject is a plasma sample.
60. The equipment as described in claim 51, characterized in that the sample of the subject is a serum sample.
MX2012012986A 2010-05-07 2011-05-04 Methods for predicting sensitivity to treatment with a targeted tyrosine kinase inhibitor. MX2012012986A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US33254510P 2010-05-07 2010-05-07
PCT/US2011/035213 WO2011140234A1 (en) 2010-05-07 2011-05-04 Methods for predicting sensitivity to treatment with a targeted tyrosine kinase inhibitor

Publications (1)

Publication Number Publication Date
MX2012012986A true MX2012012986A (en) 2013-03-05

Family

ID=44121156

Family Applications (1)

Application Number Title Priority Date Filing Date
MX2012012986A MX2012012986A (en) 2010-05-07 2011-05-04 Methods for predicting sensitivity to treatment with a targeted tyrosine kinase inhibitor.

Country Status (6)

Country Link
EP (1) EP2567232A1 (en)
JP (1) JP2013525814A (en)
CN (1) CN103038638A (en)
CA (1) CA2798441A1 (en)
MX (1) MX2012012986A (en)
WO (1) WO2011140234A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120190563A1 (en) * 2010-05-07 2012-07-26 Abbott Laboratories Methods for predicting sensitivity to treatment with a targeted tyrosine kinase inhibitor
CN108369234B (en) * 2015-10-23 2020-12-01 豪夫迈·罗氏有限公司 Methods of identifying individuals to be treated by chemotherapy based on marker molecules and related uses
US20200025766A1 (en) * 2017-02-09 2020-01-23 Board Of Regents, The University Of Texas System Methods for the detection and treatment of lung cancer

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL154598B (en) 1970-11-10 1977-09-15 Organon Nv PROCEDURE FOR DETERMINING AND DETERMINING LOW MOLECULAR COMPOUNDS AND PROTEINS THAT CAN SPECIFICALLY BIND THESE COMPOUNDS AND TEST PACKAGING.
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US5006309A (en) 1988-04-22 1991-04-09 Abbott Laboratories Immunoassay device with liquid transfer between wells by washing
US5089424A (en) 1988-06-14 1992-02-18 Abbott Laboratories Method and apparatus for heterogeneous chemiluminescence assay
DK0425633T3 (en) 1989-04-14 1994-11-28 Procedes Petroliers Petrochim Process for steam phase cracking of hydrocarbons
CA1330545C (en) * 1989-09-15 1994-07-05 Hanna Sikorska Characteristics and use of the anti-cea antibody produced by the hybridoma having the atcc number hbl0029
US20040137538A1 (en) * 2003-01-10 2004-07-15 Bradford Sherry A. Cancer comprehensive assay kit for identifying cancer protein patterns
EP1968979A2 (en) 2005-12-08 2008-09-17 Abbott Laboratories 9-membered heterobicyclic compounds as inhibitors of protein kinases
US9347945B2 (en) * 2005-12-22 2016-05-24 Abbott Molecular Inc. Methods and marker combinations for screening for predisposition to lung cancer
RU2010147643A (en) * 2008-04-23 2012-05-27 Хелтлинкс Лимитед (Au) ANALYSIS FOR GYNECOLOGICAL STATE DETECTION

Also Published As

Publication number Publication date
WO2011140234A1 (en) 2011-11-10
EP2567232A1 (en) 2013-03-13
CN103038638A (en) 2013-04-10
JP2013525814A (en) 2013-06-20
CA2798441A1 (en) 2011-11-10

Similar Documents

Publication Publication Date Title
KR102204279B1 (en) Biomarkers for predicting and assessing responsiveness of endometrial cancer subjects to lenvatinib compounds
JP5786020B2 (en) Methods and reagents for diagnosing rheumatoid arthritis
WO2011005893A2 (en) Biomarkers and methods for detecting alzheimer's disease
CA2942605C (en) Methods for identifying a biomarker indicative of a reduced drug response using a thermal shift assay
EP2596354B1 (en) Co-coupling to control reactivity of reagents in immunoassays
US20120190563A1 (en) Methods for predicting sensitivity to treatment with a targeted tyrosine kinase inhibitor
EP0430642A1 (en) Methods of diagnosing and monitoring rheumatic diseases
MX2012012986A (en) Methods for predicting sensitivity to treatment with a targeted tyrosine kinase inhibitor.
CN107615071B (en) Biomarkers for combination therapy comprising lenvatinib and everolimus
SI24638A (en) Fluorometric immunoassay for the determination of antibodies against double-stranded DNA
US20120115745A1 (en) Methods for predicting sensitivity to treatment with a targeted tyrosine kinase inhibitor
US9081016B2 (en) Assay for measurement of antibodies binding to a therapeutic monoclonal antibody
ES2939143T3 (en) Biomarkers for a Combination Therapy Comprising Lenvatinib and Everolimus
US20200018750A1 (en) Methods and compositions for the prediction and treatment of focal segmental glomerulosclerosis
TW201930883A (en) Biomarkers for lung cancer stem cells
GB2502223A (en) Identification of cancer-related autoantibodies by analysis of gene expression in cancer cells

Legal Events

Date Code Title Description
FA Abandonment or withdrawal