WO2012019000A2 - Biomarkers for the identification monitoring and treatment of ovarian cancer - Google Patents

Biomarkers for the identification monitoring and treatment of ovarian cancer Download PDF

Info

Publication number
WO2012019000A2
WO2012019000A2 PCT/US2011/046581 US2011046581W WO2012019000A2 WO 2012019000 A2 WO2012019000 A2 WO 2012019000A2 US 2011046581 W US2011046581 W US 2011046581W WO 2012019000 A2 WO2012019000 A2 WO 2012019000A2
Authority
WO
WIPO (PCT)
Prior art keywords
dnarmarkers
ovarian cancer
subject
group
dnarmarker
Prior art date
Application number
PCT/US2011/046581
Other languages
French (fr)
Other versions
WO2012019000A3 (en
Inventor
William E. Pierceall
Kam Marie Sprott
David T. Weaver
Original Assignee
On-Q-ity
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 On-Q-ity filed Critical On-Q-ity
Publication of WO2012019000A2 publication Critical patent/WO2012019000A2/en
Publication of WO2012019000A3 publication Critical patent/WO2012019000A3/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
    • G01N33/57449Specifically defined cancers of ovaries
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • 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
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/54Determining the risk of relapse

Definitions

  • This invention relates generally to methods of methods of diagnosing and treating ovarian cancer. More specifically, the invention relates to methods of assessing the responsiveness of an ovarian cancer cell to a therapeutic compound.
  • DNA repair refers to a collection of processes by which a cell identifies and corrects damage to the DNA molecules that encode its genome.
  • both normal metabolic activities and environmental factors such as UV light can cause DNA damage, resulting in as many as 1 million individual molecular lesions per cell per day.
  • Many of these lesions cause structural damage to the DNA molecule and can alter or eliminate the cell's ability to transcribe the gene that the affected DNA encodes.
  • Other lesions induce potentially harmful mutations in the cell's genome, which will affect the survival of its daughter cells after it undergoes mitosis. Consequently, the DNA repair process must be constantly active so it can respond rapidly to any damage in the DNA structure.
  • the rate of DNA repair is dependent on many factors, including the cell type, the age of the cell, and the extracellular environment.
  • a cell that has accumulated a large amount of DNA damage, or one that no longer effectively repairs damage incurred by its DNA, can enter one of three possible states: an irreversible state of dormancy, known as senescence; cell suicide, also known as apoptosis or programmed cell death or unregulated cell division, which can lead to the formation or perpetuation of a tumor.
  • a platinum response predictor could better inform the relative weight of chemotherapy in a multidisciplinary treatment regimen.
  • a platinum response predictor has direct relevance due to the wide range of chemotherapies available.
  • Ovarian cancer is the seventh most common cancer in the United States. It occurs most frequently in patients who are between 40 and 65 years of age. The lifetime risk of developing ovarian cancer is 1.4 to 1.8 percent in the United States. There are several different types of cancer that can arise in the ovary, but epithelial ovarian cancer (usually synonomous as ovarian cancer) is the most common.
  • the first step in treating ovarian cancer usually involves surgical removal of cancerous tissue (surgical debulking or cytoreduction). Achieving optimal debulking improves outcomes/prognoses and may also influence the choice of chemotherapy treatment.
  • neoadjuvant chemotherapy may be recommended as a first step in the treatment process.
  • Neoadjuvant chemotherapy is a reasonable approach for patients who could not tolerate a lengthy operation and recovery period. In such cases, initial chemotherapy can shrink the tumor and improve a patient's overall medical condition, making her a better candidate for later debulking surgery.
  • chemotherapeutic agents most commonly used in the treatment of ovarian cancer are microtubule disrupting agents (e.g. paclitaxel and docetaxel), and platinating compounds (e.g. carboplatin and cisplatin). Research suggests that chemotherapy regimens that contain these drugs are effective in preventing recurrence of ovarian cancer, and improve survival.
  • microtubule disrupting agents e.g. paclitaxel and docetaxel
  • platinating compounds e.g. carboplatin and cisplatin.
  • stage IA or IB ovarian cancers that appear to be fairly nonaggressive (described as low-grade or grade 1) do well without chemotherapy, and observation alone is a standard approach.
  • patients with higher grade (grade 2 or 3), stage IA or IB ovarian cancers, and all patients with stage IC ovarian cancer are usually advised to have adjuvant chemotherapy.
  • the standard regimen includes a platinum-based combination such as paclitaxel and carboplatin.
  • a potentially less toxic alternative to combination therapy is single agent therapy with carboplatin alone (ICON Group, 2002). This approach is more often used in Europe, and is considered controversial in the United States.
  • stage II disease patients with stage II disease have a prognosis that is slightly worse than those with stage I disease, but better than those with stage III or IV disease.
  • stage III or IV disease Although there are few trials validating this approach, many groups suggest the use of six cycles of intravenous paclitaxel plus carboplatin, similar to the regimen used for patients with stage III/IV disease.
  • An acceptable alternative is to use a combination of intraperitoneal and intravenous chemotherapy.
  • IP chemotherapy after surgery for stage II ovarian cancer is controversial since the data supporting this treatment was derived from studies of patients with stage III disease.
  • Adjuvant therapy is also an important clinical activity for ovarian cancer patients, as there is a demonstrated benefit to this process post-therapy.
  • the standard course of chemotherapy after surgical debulking for stage III or IV ovarian cancer includes six cycles of paclitaxel plus carboplatin. Each cycle given over three weeks and treatment usually begins within two to six weeks after surgery. However, the preferred treatment for all patients is enrollment in a clinical trial.
  • IP intraperitoneal
  • IV intravenous
  • IP chemotherapy cisplatin and paclitaxel
  • IP chemotherapy was given in combination with intravenous paclitaxel and compared to the standard regimen of all IV treatment with paclitaxel plus cisplatin.
  • treatment-related side effects include both catheter-related complications (infection, blockage or leakage of the catheter) as well as non- catheter-related problems (low white and red blood cell counts, nausea and vomiting, abdominal pain, and neurologic side effects such as numbness and tingling of the fingers and toes).
  • a different non-platinum-containing regimen may be considered.
  • a single drug rather than combination chemotherapy is recommended in these cases.
  • agents including pegylated liposomal doxorubicin, topotecan, docetaxel, oral etoposide, gemcitabine, vinorelbine, if osf amide, leucovorin- modulated 5-fluorouracil, or tamoxifen.
  • VEGF vascular endothelial growth factor
  • Bevacizumab interacts with a protein called vascular endothelial growth factor (VEGF).
  • VEGF vascular endothelial growth factor
  • Bevacizumab interferes with the blood supply of the tumor by interacting with VEGF, causing the tumors to shrink.
  • Bevacizumab also enhances the antitumor effect of conventional chemotherapy drugs, and is used in combination chemotherapy regimens for the treatment of other advanced tumors, such as colorectal, ovarian, lung, and renal cell cancer.
  • Bevacizumab is effective for the treatment of ovarian cancer, and it may be considered for patients with platinum-resistant ovarian cancer, either alone or in combination with chemotherapy.
  • DNA repair genes and/or proteins are already known to provide meaningful data relative to risk and prognosis, as well as offer valuable insights at decision inflection points for predictive therapeutic response in personalized medicine. While risk and prognostic significance should not be discounted, informations leading to correct decisions in patient management should prove most beneficial relative to actionable information leading to improved patient care and survival.
  • DNARMARKERS biological markers
  • proteins such as proteins, nucleic acids, polymorphisms, metabolites, protein modifications, nucleic acid modifications,
  • chromosomes and other analytes, as well as certain physiological conditions and states, are present or altered in subjects with an increased risk of developing a recurrent ovarian cancer.
  • the present invention also provides methods of assessing the likelihood of a local and/or distant recurrence or overall survival or disease free survival or response to therapy by a patient with ovarian cancer by measuring the level of an effective amount of one or more DNARMARKERS in a sample from the subject.
  • the sample can contain ovarian tumor or normal ovarian tissue or a blood sample.
  • the tissue is for example a paraffin embedded tissue, a fresh tissue, or a frozen tissue sample.
  • An increased risk of developing a recurrence of ovarian cancer in the subject is determined by measuring a clinically significant alteration in the level of the
  • DNARMARKER in the sample is determined by comparing the level of the effective amount of DNARMARKER to a reference value.
  • the reference value is an index.
  • the invention provides a method with a predetermined level of predictability for assessing the progression of a ovarian cancer in a
  • the first sample is taken from the subject prior to being treated for the ovarian cancer and the second sample is taken from the subject after being
  • Treatment can include chemotherapy, or radiation therapy.
  • the invention provides a method with a predetermined level of predictability for monitoring the effectiveness of treatment or selecting a treatment regimen for ovarian cancer by detecting the level of an effective amount of
  • DNARMARKERS in a first sample from the subject at a first period of time
  • detecting the level of an effective amount of DNARMARKERS in a second sample from the subject at a second period of time is compared to the level detected at the second period of time or alternatively a reference value. Effectiveness of treatment is monitored by a change in the level of the effective amount of DNARMARKERS from the subject.
  • Treatment is for example, chemotherapy and /or radiotherapy.
  • Chemotherapeutic agents include a platinating agent (e.g., carpoplatin or oxiplatin), taxane, a PARP inhibitor and any combination of these.
  • a DNARMARKER includes for example MLH 1 , pMAPKAPK2, PAR, PARP 1 , XPF, pH2A2and FAND2.
  • One, two, three, four, five, ten or more DNARMARKERS are measured.
  • Ki67 is measured.
  • any other biomarker listed on Tables 1,2 or 3 are measured.
  • the DNARMARKER is XPF, FANCD2, pMK2, PAR, MLH1, PARP1, BRCA1, RAD51, POL H, ATM, or ERCC1.
  • DNARMARKERS are DNA repair proteins belonging to different DNA repair pathways.
  • three or more DNARMARKERS are DNARMARKERS.
  • DNARMARKERS belonging to two or more different DNA repair pathways The level of a DNARMARKER is measured electrophoretically, or immunochemically, or genetically. For example the level of the DNARMARKER is detected by immunohistochemistry, radioimmunoassay, immunofluorescence assay, by an enzyme-linked immunosorbent assay, by genotyping, or by fluorescence in situ hybridization.
  • the subject has ovarian cancer, or a recurrent ovarian cancer.
  • the sample is taken for a subject that has previously been treated for ovarian cancer.
  • the sample is taken from the subject prior to being treated for ovarian cancer.
  • the sample is a tumor biopsy such as a core biopsy, a needle biopsy, an excisional tissue biopsy or an incisional tissue biopsy.
  • the sample is a tumor cell from blood, lymph nodes or a bodily fluid.
  • FIG. 1 shows MLH1 IHC staining and patient distributions based on image analysis and quantification.
  • MLH1 IHC nuclear- localized staining for both positive and negative samples is depicted in the panels to the top.
  • a TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear staining intensity of individual cells.
  • a broad dynamic range of expression is achieved with this biomarker which may assist in separating patient groups into responders and non-responders.
  • FIG. 2 shows phosphoMAPKAPK2 IHC staining and patient distribution based on image analysis and quantification.
  • phosphoMAPKAPK2 IHC nuclear and IHC cytoplasmic -localized staining for both positive and negative samples are depicted in the panels to the top.
  • a TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear and cytoplasmic -staining intensity of individual cells.
  • two separate metrics may be utilized, both nuclear and cytoplasmic intensities.
  • the figure further depicts that many cells stain not merely as nuclear or cytoplasmic, but that many have both nuclear and cytoplasmic staining.
  • FIG. 3 shows PAR IHC staining and patient distribution based on image analysis and quantification. PAR IHC nuclear- localized staining for both positive and negative samples is depicted in the panels to the top.
  • a TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear staining intensity of individual cells.
  • FIG. 4 shows PARP1 IHC staining and patient distribution based on image analysis and quantification.
  • PARP1 IHC nuclear- localized staining for both positive and negative samples is depicted in the panels to the top.
  • a TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear staining intensity of individual cells.
  • FIG. 5 shows XPF IHC staining and patient distribution based on image analysis and quantification.
  • XPF IHC nuclear-localized staining for both positive and negative samples is depicted in the panels to the top.
  • a TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear staining intensity of individual cells.
  • FIG. 6 show Ki67 IHC staining and patient distribution based on image analysis and quantification. Ki67 IHC nuclear- localized staining for both positive and negative samples is depicted in the panels to the top.
  • a TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear staining intensity of individual cells.
  • Figure 7 shows FancD2 IHC Staining. FancD2 IHC nuclear-localized staining for both positive and negative samples is depicted in the panels to the top. Note that the staining for this biomarker may be represented by broad nuclear localization or more granular foci presumably occurring as a micro-localization of FancD2 to repair sites at specific DNA loci.
  • FancD2 staining may be scored by quantifying general nuclear intensity of IHC stain or by enumeration of FancD2 foci. Note that regarding patient ID, that these 2 metrics are not necessarily correlative and may provide independent metrics from a single marker by which to delineate therapeutic response in ovarian cancer treatment.
  • Figure 9 shows phosphoH2AX staining and patient distribution based on image analysis and quantification.
  • staining for phosphoH2AX IHC may be represented by broad nuclear localization or more granular foci presumably occurring as a micro-localization of repair sites at specific DNA loci.
  • phosphoH2AX staining may be scored by quantifying general nuclear intensity (here by binning the data as 1+, 2+, 3+) of IHC stain or by enumeration of phosphoH2AX foci. Note that regarding patient ID, that these 2 metrics are not necessarily correlative and may provide independent metrics from a single marker by which to delineate therapeutic response in ovarian cancer treatment.
  • Figure 10 shows the comparison of FancD2 and phosphoH2AX Scoring: Nuclear IHC and Foci Enumeration. Comparison of staining for H2AX and FancD2 by either nuclear intensity with qualtity of cells staining as positive and % of cells exhibiting nuclear foci indicate that the two markers are not redundant with one another and may provide ortholgous metrics by which to separate patients based on response.
  • Figure 11 shows patient IHC staining differences for phosphoMAPKAPK2 displayed in TMA Format. Representative core samples were taken from ovarian tumors and arranged on Tissue Microarrays. In this format many samples can be stained and analyzed in a more time efficient fashion while at the same time limiting the effects of slide-to-slide and day-to-day variability.
  • Figure 12. shows TMA-based Rank-ordered Patient Group Distribution Plots for PAR, XPF, PARP1, and MLH1. Dynamic ranges for biomarker expressions are generated from TMA-based studies. The rank ordering shown here can be utilized to establish univariate analysis cut-points for responders versus non-responders. The error bars for individual patient specimens are reflections of core-to-core sampling and may represent heterogeneity present within the tumor.
  • Figure 13 shows tumor heterogeneity determined by two DNA repair biomarkers.
  • the pMK2 and FANCD2 biomarkers were examined on serial FFPE sections from the same serous ovarian cancer tumor. Two zones are delineated in the figure. In one case (dashed circle), the region of the tumor is positive for both markers. In the other case (solid circle), the tumor cells are positive for FANCD2 staining and nuclear foci, but are determined to be negative for pMK2. Insert region, the pMK2 staining pattern is distinctively different in the two zones. In one case (cytoplasmic), there is predominantly cytoplasmic staining. In the other case (mitotic), there is punctuate nuclear staining at a small fraction of the tumor cells consistent with a mitotic cell pattern.
  • the present invention relates to the identification of biomarkers associated with ovarian cancer. Specifically, these biomarkers are proteins associated in DNA repair pathways. DNA repair pathways are important to the cellular response network to chemotherapy and radiation.
  • the invention relates to the observation that tumor cells have altered DNA repair and DNA damage response pathways and that loss of one of these pathways renders the cancer more sensitive to a particular class of DNA damaging agents. Cancer therapy procedures such as chemotherapy and radiotherapy work by overwhelming the capacity of the cell to repair DNA damage, resulting in cell death.
  • NER Nucleotide Excision Repair
  • MMR Mismatch Repair
  • HR/FA Recombination/Fanconi Anemia pathway
  • NHEJ Non-Homologous Endjoining
  • TLS Translesion DNA Synthesis repair
  • BER, NER and MMR repair single strand DNA damage.
  • the other strand can be used as a template to guide the correction of the damaged strand.
  • excision repair mechanisms that remove the damaged nucleotide and replace it with an undamaged nucleotide complementary to that found in the undamaged DNA strand.
  • BER repairs damage due to a single nucleotide caused by oxidation, alkylation, hydrolysis, or deamination.
  • NER repairs damage affecting longer strands of 2-30 bases.
  • TCR Transcription-Coupled Repair
  • NHEJ and HR repair double stranded DNA damage double stranded damage is particularly hazardous to dividing cells.
  • the NHEJ pathway operates when the cell has not yet replicated the region of DNA on which the lesion has occurred. The process directly joins the two ends of the broken DNA strands without a template, losing sequence information in the process. Thus, this repair mechanism is necessarily mutagenic. However, if the cell is not dividing and has not replicated its DNA, the NHEJ pathway is the cell's only option. NHEJ relies on chance pairings, or microhomologies, between the single-stranded tails of the two DNA fragments to be joined. There are multiple independent "failsafe" pathways for NHEJ in higher eukaryotes.
  • Recombinational repair requires the presence of an identical or nearly identical sequence to be used as a template for repair of the break.
  • the enzymatic machinery responsible for this repair process is nearly identical to the machinery responsible for chromosomal crossover during meiosis.
  • This pathway allows a damaged chromosome to be repaired using the newly created sister chromatid as a template, i.e. an identical copy that is also linked to the damaged region via the centromere.
  • Double- stranded breaks repaired by this mechanism are usually caused by the replication machinery attempting to synthesize across a single-strand break or unrepaired lesion, both of which result in collapse of the replication fork.
  • Translesion synthesis is an error-prone bypass method where a DNA lesion is left unrepaired during S phase, and is repaired later in the cell cycle.
  • the DNA replication machinery cannot continue replicating past a site of DNA damage, so the advancing replication fork will stall on encountering a damaged base.
  • the translesion synthesis pathway is mediated by specific DNA polymerases that insert alternative bases at the site of damage and thus allow replication to bypass the damaged base to continue with chromosome duplication.
  • the bases inserted by the translesion synthesis machinery are template- independent, but not arbitrary; for example, one human polymerase inserts adenine bases when synthesizing past a thymine dimer. If this residue is not repaired at a later step, the process is mutagenic.
  • Cancer cells accumulate high levels of DNA damage. This damage may result from their heightened proliferative activity or from exposure to chemotherapy or ionizing radiation.
  • MMR mismatch repair
  • BRCA status may be an indicator of susceptibility to treatment with paclitaxel with vinorelbine or radiation and resistance to etoposide and bleomycin (Quinn et al., 2003; Zhou et al., 2003).
  • FanD2/BRCAl node of the FA/HR pathway leads to cisplatin resistance (Taniguchi et al., 2009) and that inhibition of FancD2 (Chirnomas et al., 2006) or deficiency of FancD2 (wang et al., 2006) may lead to restored cisplatin sensitivity and irofulven activity, respectively, in anti-ovarian cancer activities.
  • Fanc-deficient cells are susceptible to treatment with inhibitors to ATM (Kennedy et al., 2007).
  • Biomarkers of DNA repair may guide therapy because these are pathways that respond to damage to DNA caused by chemotherapy and radiation. Single pathways are not adequate to describe the DNA repair responses to even single chemotherapy agents for several reasons. First, chemotherapy causing DNA damage leads to many forms of DNA damage necessitating different biochemical mechanisms to repair. Second, the central DNA repair pathways can at least partially complement each other for cell survival. Even though the second pathway may not be as effective, it provides a salvage mechanism for the cell to return to cell division and growth. For a tumor cell, this is a mechanism for diminishing the strength of and/or evading cell cycle checkpoints.
  • the DNARMARKER is XPF, FANCD2, pMK2, PAR, MLHl, PARPl, BRCAl, RAD51, POL H, ATM, or ERCC1.
  • TP true positives
  • TN true negatives
  • FP false negatives
  • FN false negatives
  • Biomarker in the context of the present invention encompasses, without limitation, proteins, nucleic acids, and metabolites, together with their polymorphisms, mutations, variants, modifications, subunits, fragments, protein-ligand complexes, and degradation products, protein-ligand complexes, elements, related metabolites, and other analytes or sample-derived measures. Biomarkers can also include mutated proteins or mutated nucleic acids. Biomarkers also encompass non-blood borne factors or non-analyte physiological markers of health status, such as "clinical parameters” defined herein, as well as “traditional laboratory risk factors”, also defined herein.
  • Biomarkers also include any calculated indices created mathematically or combinations of any one or more of the foregoing measurements, including temporal trends and differences. Where available, and unless otherwise described herein, determinants which are gene products are identified based on the official letter abbreviation or gene symbol assigned by the international Human Genome Organization Naming Committee (HGNC) and listed at the date of this filing at the US National Center for Biotechnology Information (NCBI) web site
  • “Clinical parameters” encompasses all non-sample or non-analyte biomarkers of subject health status or other characteristics, such as, without limitation, age (Age), ethnicity (RACE), gender (Sex), or family history (FamHX).
  • FN is false negative, which for a disease state test means classifying a disease subject incorrectly as non-disease or normal.
  • FP is false positive, which for a disease state test means classifying a normal subject incorrectly as having disease.
  • a “formula,” “algorithm,” or “model” is any mathematical equation, algorithmic, analytical or programmed process, or statistical technique that takes one or more continuous or categorical inputs (herein called “parameters”) and calculates an output value, sometimes referred to as an "index” or “index value.”
  • Parameters continuous or categorical inputs
  • Non-limiting examples of “formulas” include sums, ratios, and regression operators, such as coefficients or exponents, biomarker value transformations and normalizations (including, without limitation, those normalization schemes based on clinical parameters, such as gender, age, or ethnicity), rules and guidelines, statistical classification models, and neural networks trained on historical populations.
  • DNARMARKERS and other biomarkers are linear and nonlinear equations and statistical classification analyses to determine the relationship between levels of DNARMARKERS detected in a subject sample and the subject's responsivenss to chemotherapy.
  • structural and synactic statistical classification algorithms, and methods of risk index construction utilizing pattern recognition features, including established techniques such as cross-correlation, Principal Components Analysis (PCA), factor rotation, Logistic Regression (LogReg), Linear Discriminant Analysis (LDA), Eigengene Linear Discriminant Analysis (ELD A), Support Vector Machines (SVM), Random Forest (RF), Recursive Partitioning Tree (RPART), as well as other related decision tree classification techniques, Shrunken Centroids (SC), StepAIC, Kth-Nearest Neighbor, Boosting, Decision Trees, Neural Networks, Bayesian Networks, Support Vector Machines, and Hidden Markov Models, among others.
  • PCA Principal Components Analysis
  • LogReg Logistic Regression
  • LDA Linear Discriminant Analysis
  • ELD A Eigengene Line
  • DNARMARKER selection technique such as forward selection, backwards selection, or stepwise selection, complete enumeration of all potential panels of a given size, genetic algorithms, or they may themselves include biomarker selection methodologies in their own technique.
  • AIC Akaike's Information Criterion
  • BIC Bayes Information Criterion
  • the resulting predictive models may be validated in other studies, or cross-validated in the study they were originally trained in, using such techniques as Bootstrap, Leave-One-Out (LOO) and 10-Fold cross-validation (10-Fold CV).
  • LEO Leave-One-Out
  • 10-Fold cross-validation 10-Fold CV.
  • false discovery rates may be estimated by value permutation according to techniques known in the art.
  • a "health economic utility function" is a formula that is derived from a combination of the expected probability of a range of clinical outcomes in an idealized applicable patient population, both before and after the introduction of a diagnostic or therapeutic intervention into the standard of care.
  • a cost and/or value measurement associated with each outcome, which may be derived from actual health system costs of care (services, supplies, devices and drugs, etc.) and/or as an estimated acceptable value per quality adjusted life year (QALY) resulting in each outcome.
  • the sum, across all predicted outcomes, of the product of the predicted population size for an outcome multiplied by the respective outcome's expected utility is the total health economic utility of a given standard of care.
  • the difference between (i) the total health economic utility calculated for the standard of care with the intervention versus (ii) the total health economic utility for the standard of care without the intervention results in an overall measure of the health economic cost or value of the intervention.
  • This may itself be divided amongst the entire patient group being analyzed (or solely amongst the intervention group) to arrive at a cost per unit intervention, and to guide such decisions as market positioning, pricing, and assumptions of health system acceptance.
  • Such health economic utility functions are commonly used to compare the cost-effectiveness of the intervention, but may also be transformed to estimate the acceptable value per QALY the health care system is willing to pay, or the acceptable cost-effective clinical performance characteristics required of a new intervention.
  • a health economic utility function may preferentially favor sensitivity over specificity, or PPV over NPV based on the clinical situation and individual outcome costs and value, and thus provides another measure of health economic performance and value which may be different from more direct clinical or analytical performance measures.
  • Measurement or “measurement,” or alternatively “detecting” or “detection,” means assessing the presence, absence, quantity or amount (which can be an effective amount) of either a given substance within a clinical or subject-derived sample, including the derivation of qualitative or quantitative concentration levels of such substances, or otherwise evaluating the values or categorization of a subject's non-analyte clinical parameters.
  • NDV Neuronal predictive value
  • hazard ratios and absolute and relative risk ratios within subject cohorts defined by a test are a further measurement of clinical accuracy and utility. Multiple methods are frequently used to defining abnormal or disease values, including reference limits, discrimination limits, and risk thresholds.
  • Performance is a term that relates to the overall usefulness and quality of a diagnostic or prognostic test, including, among others, clinical and analytical accuracy, other analytical and process characteristics, such as use characteristics (e.g., stability, ease of use), health economic value, and relative costs of components of the test. Any of these factors may be the source of superior performance and thus usefulness of the test, and may be measured by appropriate "performance metrics," such as AUC, time to result, shelf life, etc. as relevant.
  • PSV Positive predictive value
  • “Risk” in the context of the present invention relates to the probability that an event will occur over a specific time period, as in the responsiveness to treatmnet, and can mean a subject's "absolute” risk or “relative” risk.
  • Absolute risk can be measured with reference to either actual observation post-measurement for the relevant time cohort, or with reference to index values developed from statistically valid historical cohorts that have been followed for the relevant time period.
  • Relative risk refers to the ratio of absolute risks of a subject compared either to the absolute risks of low risk cohorts or an average population risk, which can vary by how clinical risk factors are assessed.
  • Odds ratios the proportion of positive events to negative events for a given test result, are also commonly used (odds are according to the formula p/(l-p) where p is the probability of event and (1- p) is the probability of no event) to no-conversion.
  • Risk evaluation in the context of the present invention encompasses making a prediction of the probability, odds, or likelihood that an event or disease state may occur, the rate of occurrence of the event or conversion from one disease state. Risk evaluation can also comprise prediction of future clinical parameters, traditional laboratory risk factor values, or other indices of cancer, either in absolute or relative terms in reference to a previously measured population.
  • the methods of the present invention may be used to make continuous or categorical measurements of the responsiveness to treatment thus diagnosing and defining the risk spectrum of a category of subjects defined as being at responders or non-responders. In the categorical scenario, the invention can be used to discriminate between normal and other subject cohorts at higher risk for responding. Such differing use may require different DNARMARKER combinations and individualized panels, mathematical algorithms, and/or cut-off points, but be subject to the same aforementioned measurements of accuracy and performance for the respective intended use.
  • sample in the context of the present invention is a biological sample isolated from a subject and can include, by way of example and not limitation, tissue biopies, whole blood, serum, plasma, blood cells, endothelial cells, lymphatic fluid, ascites fluid, interstitital fluid (also known as "extracellular fluid” and encompasses the fluid found in spaces between cells, including, inter alia, gingival crevicular fluid), bone marrow, cerebrospinal fluid (CSF), saliva, mucous, sputum, sweat, urine, or any other secretion, excretion, or other bodily fluids.
  • tissue biopies whole blood, serum, plasma, blood cells, endothelial cells, lymphatic fluid, ascites fluid
  • interstitital fluid also known as "extracellular fluid” and encompasses the fluid found in spaces between cells, including, inter alia, gingival crevicular fluid
  • bone marrow also known ascites fluid
  • CSF cerebrospinal fluid
  • “Specificity” is calculated by TN/(TN+FP) or the true negative fraction of non- disease or normal subjects.
  • Statistically significant it is meant that the alteration is greater than what might be expected to happen by chance alone (which could be a "false positive”).
  • Statistical significance can be determined by any method known in the art. Commonly used measures of significance include the p-value, which presents the probability of obtaining a result at least as extreme as a given data point, assuming the data point was the result of chance alone. A result is often considered highly significant at a p-value of 0.05 or less.
  • a "subject" in the context of the present invention is preferably a mammal.
  • the mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples. Mammals other than humans can be advantageously used as subjects that represent animal models of cancer.
  • a subject can be male or female.
  • TN is true negative, which for a disease state test means classifying a non- disease or normal subject correctly.
  • TP i s true positive, which for a disease state test means correctly classifying a disease subject.
  • biomarkers associated with DNA repair and DNA damage response are useful in monitoring and predicting the response to a therapeutic compound.
  • the invention features methods for identifying subjects who either are or are pre-disposed to developing resistance or are sensitive to a therapeutic compound, e.g., a chemotherapeutic drug by detection of the biomarkers disclosed herein. These biomarkers are also useful for monitoring subjects undergoing treatments and therapies for cancer and cell proliferative disorders, and for selecting therapies and treatments that would be efficacious in subjects having cancer and cell proliferative disorders.
  • biomarker in the context of the present invention encompasses, without limitation, proteins, nucleic acids, polymorphisms of proteins and nucleic acids, elements, metabolites, and other analytes. Biomarkers can also include mutated proteins or mutated nucleic acids.
  • analyte as used herein can mean any substance to be measured and can encompass electrolytes and elements, such as calcium.
  • Proteins, nucleic acids, polymorphisms, and metabolites whose levels are changed in subjects who have resistance or sensitivity to therapeutic compound, or are predisposed to developing resistance or sensitivity to therapeutic compound are summarized in Table 1 and are collectively referred to herein as, inter alia, "DNA Repair and DNA Damage Response proteins or DNARMARKER".
  • Table 2 summarizes DNARMARKERS associated with Ovarian Cancer.
  • Table 3 summarizes other biomarkers useful in the methods of the invention
  • DNARMARKERS is determined at the protein or nucleic acid level using any method known in the art. For example, at the nucleic acid level Northern hybridization analysis using probes which specifically recognize one or more of these sequences can be used to determine gene expression. Alternatively, expression is measured using reverse-transcription-based PCR assays, e.g., using primers specific for the
  • differentially expressed sequence of genes is also determined at the protein level, i.e. , by measuring the levels of peptides encoded by the gene products described herein, or activities thereof. Such methods are well known in the art and include, e.g. ,
  • immunoassays based on antibodies to proteins encoded by the genes, aptamers or molecular imprints. Any biological material can be used for the detection/quantification of the protein or its activity. Alternatively, a suitable method can be selected to determine the activity of proteins encoded by the marker genes according to the activity of each protein analyzed.
  • the DNARMARKER proteins are detected in any suitable manner, but is typically detected by contacting a sample from the patient with an antibody which binds the DNARMARKER protein and then detecting the presence or absence of a reaction product.
  • the antibody may be monoclonal, polyclonal, chimeric, or a fragment of the foregoing, as discussed in detail above, and the step of detecting the reaction product may be carried out with any suitable immunoassay.
  • the sample from the subject is typically a biological fluid as described above, and may be the same sample of biological fluid used to conduct the method described above.
  • the sample may also be in the form of a tissue specimen from a patient where the specimen is suitable for immunohistochemistry in a variety of formats such as paraffin-embedded tissue, frozen sections of tissue, and freshly isolated tissue.
  • the immunodetection methods are antibody-based but there are numerous additional techniques that allow for highly sensitive determinations of binding to an antibody in the context of a tissue. Those skilled in the art will be familiar with various immunohistochemistry strategies.
  • Immunoassays carried out in accordance with the present invention may be homogeneous assays or heterogeneous assays.
  • the immunological reaction usually involves the specific antibody (e.g., anti- DNARMARKER protein antibody), a labeled analyte, and the sample of interest.
  • the signal arising from the label is modified, directly or indirectly, upon the binding of the antibody to the labeled analyte.
  • Both the immunological reaction and detection of the extent thereof are carried out in a homogeneous solution.
  • Immunochemical labels which may be employed include free radicals, radioisotopes, fluorescent dyes, enzymes, bacteriophages, or coenzymes.
  • the reagents are usually the sample, the antibody, and means for producing a detectable signal.
  • Samples as described above may be used.
  • the antibody is generally immobilized on a support, such as a bead, plate or slide, and contacted with the specimen suspected of containing the antigen in a liquid phase.
  • the support is then separated from the liquid phase and either the support phase or the liquid phase is examined for a detectable signal employing means for producing such signal.
  • the signal is related to the presence of the analyte in the sample.
  • Means for producing a detectable signal include the use of radioactive labels, fluorescent labels, or enzyme labels.
  • an antibody which binds to that site can be conjugated to a detectable group and added to the liquid phase reaction solution before the separation step.
  • the presence of the detectable group on the solid support indicates the presence of the antigen in the test sample.
  • suitable immunoassays are radioimmunoassays, immunofluorescence methods, chemilumenescence methods, electrochemilumenescence or enzyme-linked immunoassays.
  • Antibodies are conjugated to a solid support suitable for a diagnostic assay (e.g., beads, plates, slides or wells formed from materials such as latex or polystyrene) in accordance with known techniques, such as passive binding.
  • Antibodies as described herein may likewise be conjugated to detectable groups such as radiolabels (e.g., 35 S, 125 I, 131 I), enzyme labels (e.g., horseradish peroxidase, alkaline phosphatase), and fluorescent labels (e.g., fluorescein) in accordance with known techniques.
  • radiolabels e.g., 35 S, 125 I, 131 I
  • enzyme labels e.g., horseradish peroxidase, alkaline phosphatase
  • fluorescent labels e.g., fluorescein
  • nucleic acid probes e.g., oligonucleotides, aptamers, siRNAs against any of the DNARMARKERS in Table 1.
  • the invention also includes a DNARMARKER-detection reagent, e.g., nucleic acids that specifically identify one or more DNARMARKER nucleic acids by having homologous nucleic acid sequences, such as oligonucleotide sequences, complementary to a portion of the DNARMARKER nucleic acids or antibodies to proteins encoded by the DNARMARKER nucleic acids packaged together in the form of a kit.
  • the oligonucleotides are fragments of the DNARMARKER genes.
  • the olignucleotides are 200, 150, 100, 50, 25, 10 or less nucleotides in length.
  • the kit may contain in separate containers a nucleic acid or antibody (either already bound to a solid matrix or packaged separately with reagents for binding them to the matrix) , control formulations (positive and/or negative), and/or a detectable label. Instructions (e.g., written, tape, VCR, CD-ROM, etc.) for carrying out the assay may be included in the kit.
  • the assay may for example be in the form of a Northern hybridization or a sandwich ELISA as known in the art.
  • DNARMARKER detection reagent is immobilized on a solid matrix such as a porous strip to form at least one DNARMARKER detection site.
  • the measurement or detection region of the porous strip may include a plurality of sites containing a nucleic acid.
  • a test strip may also contain sites for negative and/or positive controls. Alternatively, control sites are located on a separate strip from the test strip.
  • the different detection sites may contain different amounts of immobilized nucleic acids, i.e., a higher amount in the first detection site and lesser amounts in subsequent sites.
  • the number of sites displaying a detectable signal provides a quantitative indication of the amount of DNARMARKER present in the sample.
  • the detection sites may be configured in any suitably detectable shape and are typically in the shape of a bar or dot spanning the width of a test strip.
  • the kit contains a nucleic acid substrate array comprising one or more nucleic acid sequences.
  • the nucleic acids on the array specifically identify one or more nucleic acid sequences represented by DNARMARKER 1-259.
  • the expression of 2, 3,4, 5, 6, 7,8, 9, 10, 15, 20, 25, 40 or 50 or more of the sequences represented by DNARMARKER 1-259. are identified by virtue of binding to the array.
  • the substrate array can be on, e.g. , a solid substrate, e.g. , a "chip" as described in U.S. Patent No.5,744,305.
  • the substrate array can be a solution array, e.g., Luminex, Cyvera, Vitra and Quantum Dots' Mosaic.
  • the kit contains antibodies for the detection of DNARMARKERS.
  • Responsiveness e.g., resistance or sensitivity
  • Responsiveness of a cell to an agent is determined by measuring an effective amount of a DNARMARKER proteins, nucleic acids,
  • the cell is for example a cancer cell.
  • the cancer is a ovarian cancer.
  • the DNARMARKER includes for example MLH1, pMAPKAPK2, PAR, PARP1, XPF, pH2A2and FAND2. Additionally any other biomarker listed on Tables 1 ,2 or 3 are measured. In another embodimentthe
  • DNARMARKER is XPF, FANCD2, pMK2, PAR, MLH1, PARP1, BRCA1, RAD51, POL H, ATM, or ERCC1.
  • Ki67 is measured.
  • resistance means that the failure of a cell to respond to an agent.
  • resistance to a chemotherapeutic drug means the cell is not damaged or killed by the drug.
  • sensitivity is meant that the cell responds to an agent.
  • sensitivity to a chemotherapeutic drug means the cell is damaged or killed by the drug.
  • responsiveness of a cell to a chemotherapeutic agent identified by identifying a decrease in expression or activity of one or more Ovarian DNARMARKERS For example, responsiveness of a cell to a chemotherapeutic agent identified by identifying a decrease in expression or activity of one or more Ovarian DNARMARKERS.
  • the presence of a deficiency in DNARMARKER indicates that the cell is sensitive to a chemotherapeutic agent.
  • the absence of a deficiency indicates that the cell is resistant to a chemotherapeutic agent.
  • the methods are useful to treat, alleviate the symptoms of, monitor the progression of or delay the onset of cancer in a subject.
  • DNARMARKER proteins, nucleic acids or metabolites also allows for the course of treatment of cancer or a cell proliferative disorder to be monitored.
  • a biological sample is provided from a subject undergoing treatment, e.g., chemotherapeutic treatment, for cancer or a cell proliferative disorder. If desired, biological samples are obtained from the subject at various time points before, during, or after treatment. Expression of an effective amount of DNARMARKER proteins, nucleic acids or metabolites is then determined and compared to a reference, e.g. a control individual or population whose cancer or a cell proliferative disorder state is known or an index value.
  • a reference e.g. a control individual or population whose cancer or a cell proliferative disorder state is known or an index value.
  • the reference sample or index value may be taken or derived from one or more individuals who have been exposed to the treatment.
  • the reference sample or index value may be taken or derived from one or more individuals who have not been exposed to the treatment.
  • samples may be collected from subjects who have received initial treatment for cancer or a cell proliferative disorder and subsequent treatment for diabetes to monitor the progress of the treatment.
  • the amount of the DNARMARKER protein, nucleic acid, polymorphism, metabolite, or other analyte can be measured in a test sample and compared to the "normal control level," utilizing techniques such as reference limits, discrimination limits, or risk defining thresholds to define cutoff points and abnormal values.
  • Such normal control level and cutoff points may vary based on whether a DNARMARKER is used alone or in a formula combining with other DNARMARKERS into an index.
  • the normal control level can be a database of DNARMARKER patterns from previously tested subjects who responded to chemotherapy over a clinically relevant time horizon.
  • the present invention may be used to make continuous or categorical measurements of the response to chemotherapy or cancer survival, thus diagnosing and defining the risk spectrum of a category of subjects defined as at risk for not responding to chemotherapy.
  • the methods of the present invention can be used to discriminate between treatment responsive and treatment non-responsive subject cohorts.
  • the present invention may be used so as to discriminate those who have an improved survival potential. Such differing use may require different
  • Identifying the subject who will be responsive to therapy enables the selection and initiation of various therapeutic interventions or treatment regimens in order increase the individuals survival potential.
  • Levels of an effective amount of DNARMARKER proteins, nucleic acids, polymorphisms, metabolites, or other analytes also allows for the course of treatment of a metastatic disease or metastatic event to be monitored.
  • a biological sample can be provided from a subject undergoing treatment regimens, e.g., drug treatments, for cancer. If desired, biological samples are obtained from the subject at various time points before, during, or after treatment.
  • Levels of an effective amount of DNARMARKER proteins, nucleic acids, polymorphisms, metabolites, or other analytes can then be determined and compared to a reference value, e.g. a control subject or population whose therapeutic responsiveness is known or an index value or baseline value.
  • the reference sample or index value or baseline value may be taken or derived from one or more subjects who have been exposed to the treatment, or may be taken or derived from one or more subjects who are at low risk of surviving the cancer, or may be taken or derived from subjects who have shown
  • the reference sample or index value or baseline value may be taken or derived from one or more subjects who have not been exposed to the treatment.
  • samples may be collected from subjects who have received initial treatment for cancer or and subsequent treatment for cancer or a metastatic event to monitor the progress of the treatment.
  • a reference value can also comprise a value derived from risk prediction algorithms or computed indices from population studies such as those disclosed herein.
  • the DNARMARKERS of the present invention can thus be used to generate a "reference DNARMARKER profile" of those subjects who would or would not be expected respond to cancer treatment.
  • the DNARMARKERS disclosed herein can also be used to generate a "subject DNARMARKER profile" taken from subjects who are responsive cancer treatment.
  • the subject DNARMARKER profiles can be compared to a reference
  • DNARMARKER profile to diagnose or identify subjects at risk for developing resistance to chemotherapy, to monitor the progression of disease, as well as the rate of progression of disease, and to monitor the effectiveness of treatment modalities.
  • the reference and subject DNARMARKER profiles of the present invention can be contained in a machine-readable medium, such as but not limited to, analog tapes like those readable by a VCR, CD-ROM, DVD-ROM, USB flash media, among others.
  • Such machine-readable media can also contain additional test results, such as, without limitation, measurements of clinical parameters and traditional laboratory risk factors.
  • the machine-readable media can also comprise subject information such as medical history and any relevant family history.
  • the machine-readable media can also contain information relating to other disease- risk algorithms and computed indices such as those described herein.
  • Differences in the genetic makeup of subjects can result in differences in their relative abilities to metabolize various drugs, which may modulate the symptoms or risk factors of cancer or metastatic events.
  • Subjects that have cancer, or at risk for developing cancer or a metastatic event can vary in age, ethnicity, and other parameters. Accordingly, use of the DNARMARKERS disclosed herein, both alone and together in combination with known genetic factors for drug metabolism, allow for a pre-determined level of predictability that a putative therapeutic or prophylactic to be tested in a selected subject will be suitable for treating or preventing cancer in the subject.
  • the pattern of DNARMARKER expression in the test sample is measured and compared to a reference profile, e.g., a therapeutic compound reference expression profile. Comparison can be performed on test and reference samples measured concurrently or at temporally distinct times.
  • a reference profile e.g., a therapeutic compound reference expression profile.
  • Comparison can be performed on test and reference samples measured concurrently or at temporally distinct times.
  • An example of the latter is the use of compiled expression information, e.g. , a sequence database, which assembles information about expression levels of DNARMARKERS.
  • the reference sample e.g., a control sample is from cells that are sensitive to a therapeutic compound then a similarity in the amount of the DNARMARKER proteins in the test sample and the reference sample indicates that treatment with that therapeutic compound will be efficacious. However, a change in the amount of the DNARMARKER in the test sample and the reference sample indicates treatment with that compound will result in a less favorable clinical outcome or prognosis. In contrast, if the reference sample, e.g., a control sample is from cells that are resistant to a therapeutic compound then a similarity in the amount of the DNARMARKER proteins in the test sample and the reference sample indicates that the treatment with that compound will result in a less favorable clinical outcome or prognosis. However, a change in the amount of the DNARMARKER in the test sample and the reference sample indicates that treatment with that therapeutic compound will be efficacious.
  • efficacious is meant that the treatment leads to a decrease in the amount of a DNARMARKER protein, or a decrease in size, prevalence, or metastatic potential of cancer in a subject.
  • effcacious means that the treatment retards or prevents cancer or a cell proliferative disorder from forming. Assessment of cancer and cell proliferative disorders is made using standard clinical protocols.
  • the subject is preferably a mammal.
  • the mammal is, e.g. , a human, non- human primate, mouse, rat, dog, cat, horse, or cow.
  • the subject has been previously diagnosed as having cancer or a cell proliferative disorder, and possibly has already undergone treatment for the cancer or a cell proliferative disorder.
  • the subject is suffering from or at risk of developing ovarian cancer.
  • Subjects suffering from or at risk of developing ovarian cancer are identified by methods known in the art.
  • the deficiency is determined by measuring the expression (e.g. increase or decrease relative to a control), detecting a sequence variation or posttranslational modification of one or more DNARMARKERS described herein.
  • Posttranslational modification include for example, phosphorylation,
  • ubiquitination sumo-ylation, acetylation, alkylation, methylation, glycylation, glycosylation, isoprenylation, lipoylation, phosphopantetheinylation, sulfation, selenation and C-terminal amidation.
  • a deficiency in the Homologous Recombination/FA pathway is determined by detecting the monoubiquitination of FANCD2.
  • responsiveness of cancer cell to a MAP2KAP2 inhibitor is determined by detecting phosphorylation of a MAP2KAP2 protein. Phosphorylation indicates the cell is sensitive to a MAP2KAP2 inhibitor. In contrast the absence of phosphorylation indicates the cell is resistant to a MAP2KAP2 inhibitor.
  • Sequence variations such as mutations and polymorphisms may include a deletion, insertion or substitution of one or more nucleotides, relative to the wild- type nucleotide sequence.
  • the one or more variations may be in a coding or non-coding region of the nucleic acid sequence and, may reduce or abolish the expression or function of the DNA repair pathway component polypeptide.
  • the variant nucleic acid may encode a variant polypeptide which has reduced or abolished activity or may encode a wild-type polypeptide which has little or no expression within the cell, for example through the altered activity of a regulatory element.
  • a variant nucleic acid may have one, two, three, four or more mutations or polymorphisms relative to the wild-type sequence.
  • the presence of one or more variations in a nucleic acid which encodes a component of a DNA repair pathway is determined for example by detecting, in one or more cells of a test sample, the presence of an encoding nucleic acid sequence which comprises the one or more mutations or polymorphisms, or by detecting the presence of the variant component polypeptide which is encoded by the nucleic acid sequence.
  • sequence information can be retained and subsequently searched without recourse to the original nucleic acid itself.
  • scanning a database of sequence information using sequence analysis software may identify a sequence alteration or mutation.
  • Methods according to some aspects of the present invention may comprise determining the binding of an oligonucleotide probe to nucleic acid obtained from the sample, for example, genomic DNA, RNA or cDNA.
  • the probe may comprise a nucleotide sequence which binds specifically to a nucleic acid sequence which contains one or more mutations or polymorphisms and does not bind specifically to the nucleic acid sequence which does not contain the one or more mutations or polymorphisms, or vice versa.
  • the oligonucleotide probe may comprise a label and binding of the probe may be determined by detecting the presence of the label.
  • a method may include hybridization of one or more (e.g. two) oligonucleotide probes or primers to target nucleic acid. Where the nucleic acid is double- stranded DNA, hybridization will generally be preceded by denaturation to produce single-stranded DNA.
  • the hybridization may be as part of a PCR procedure, or as part of a probing procedure not involving PCR. An example procedure would be a combination of PCR and low stringency hybridization.
  • Binding of a probe to target nucleic acid may be measured using any of a variety of techniques at the disposal of those skilled in the art.
  • probes may be radioactively, fluorescently or enzymatically labeled.
  • Other methods not employing labeling of probe include examination of restriction fragment length polymorphisms, amplification using PCR, RN'ase cleavage and allele specific oligonucleotide probing.
  • Probing may employ the standard Southern blotting technique. For instance, DNA may be extracted from cells and digested with different restriction enzymes. Restriction fragments may then be separated by electrophoresis on an agarose gel, before denaturation and transfer to a nitrocellulose filter. Labeled probe may be hybridized to the DNA fragments on the filter and binding determined.
  • Suitable selective hybridization conditions for oligonucleotides of 17 to 30 bases include hybridization overnig ht at 42.°C in 6x SSC and washing in 6.x SSC at a series of increasing temperatures from 42°C to 65°C.
  • Other suitable conditions and protocols are described in Molecular Cloning: a Laboratory Manual: 3rd edition, Sambrook & Russell (2001) Cold Spring Harbor Laboratory Press NY and Current Protocols in Molecular Biology, Ausubel et al. eds. John Wiley & Sons (1992).
  • Nucleic acid which may be genomic DNA, RNA or cDNA, or an amplified region thereof, may be sequenced to identify or determine the presence of polymorphism or mutation therein.
  • a polymorphism or mutation may be identified by comparing the sequence obtained with the database sequence of the component, as set out above. In particular, the presence of one or more polymorphisms or mutations that cause abrogation or loss of function of the polypeptide component, and thus the DNA repair pathway as a whole, may be determined.
  • Sequencing may be performed using any one of a range of standard techniques. Sequencing of an amplified product may, for example, involve precipitation with
  • Extension products may be electrophoresed on an ABI 377 DNA sequencer and data analyzed using Sequence Navigator software.
  • a specific amplification reaction such as PCR using one or more pairs of primers may conveniently be employed to amplify the region of interest within the nucleic acid sequence, for example, the portion of the sequence suspected of containing mutations or polymorphisms.
  • the amplified nucleic acid may then be sequenced as above, and/or tested in any other way to determine the presence or absence of a mutation or polymorphism which reduces or abrogates the expression or activity of the DNA repair pathway component.
  • Suitable amplification reactions include the polymerase chain reaction (PCR) (reviewed for instance in "PCR protocols; A Guide to Methods and Applications", Eds. Innis et al, 1990, Academic Press, New York, Mullis et al, Cold Spring Harbor Symp. Quant.
  • Mutations and polymorphisms associated with cancer may also be detected at the protein level by detecting the presence of a variant (i.e. a mutant or allelic variant) polypeptide.
  • a method of identifying a cancer cell in a sample from an individual as deficient in DNA repair may include contacting a sample with a specific binding member directed against a variant (e.g. a mutant) polypeptide component of the pathway, and determining binding of the specific binding member to the sample. Binding of the specific binding member to the sample may be indicative of the presence of the variant polypeptide component of the DNA repair pathway in a cell within the sample.
  • Preferred specific binding molecules for use in aspects of the present invention include antibodies and fragments or derivatives thereof ("antibody molecules").
  • the reactivities of a binding member such as an antibody on normal and test samples may be determined by any appropriate means. Tagging with individual reporter molecules is one possibility.
  • the reporter molecules may directly or indirectly generate detectable, and preferably measurable, signals.
  • the linkage of reporter molecules may be directly or indirectly, covalently, e.g. via a peptide bond or non-covalently. Linkage via a peptide bond may be as a result of recombinant expression of a gene fusion encoding binding molecule (e.g. antibody) and reporter molecule.
  • the performance and thus absolute and relative clinical usefulness of the invention may be assessed in multiple ways as noted above.
  • the invention is intended to provide accuracy in clinical diagnosis and prognosis.
  • the accuracy of a diagnostic or prognostic test, assay, or method concerns the ability of the test, assay, or method to distinguish between subjects responsive to chemotherapeutic treatment and those that are not, is based on whether the subjects have an "effective amount” or a "significant alteration" in the levels of a DNARMARKER.
  • an appropriate number of DNARMARKERS (which may be one or more) is different than the predetermined cut-off point (or threshold value) for that DNARMARKER(S) and therefore indicates that the subject responsiveness to therapy for which the DNARMARKER(S) is a determinant.
  • the difference in the level of DNARMARKER between normal and abnormal is preferably statistically significant. As noted below, and without any limitation of the invention, achieving statistical significance, and thus the preferred analytical and clinical accuracy, generally but not always requires that combinations of several DNARMARKERS be used together in panels and combined with mathematical algorithms in order to achieve a statistically significant DNARMARKER index.
  • an "acceptable degree of diagnostic accuracy” is herein defined as a test or assay (such as the test of the invention for determining the clinically significant presence of DNARMARKERS, which thereby indicates the presence of cancer and/or a risk of having a metastatic event) in which the AUC (area under the ROC curve for the test or assay) is at least 0.60, desirably at least 0.65, more desirably at least 0.70, preferably at least 0.75, more preferably at least 0.80, and most preferably at least 0.85.
  • a "very high degree of diagnostic accuracy” it is meant a test or assay in which the AUC (area under the ROC curve for the test or assay) is at least 0.80, desirably at least 0.85, more desirably at least 0.875, preferably at least 0.90, more preferably at least 0.925, and most preferably at least 0.95.
  • the predictive value of any test depends on the sensitivity and specificity of the test, and on the prevalence of the condition in the population being tested. This notion, based on Bayes' theorem, provides that the greater the likelihood that the condition being screened for is present in an individual or in the population (pre-test probability), the greater the validity of a positive test and the greater the likelihood that the result is a true positive. Thus, the problem with using a test in any population where there is a low likelihood of the condition being present is that a positive result has limited value (i.e., more likely to be a false positive). Similarly, in populations at very high risk, a negative test result is more likely to be a false negative.
  • ROC and AUC can be misleading as to the clinical utility of a test in low disease prevalence tested populations (defined as those with less than 1% rate of occurrences (incidence) per annum, or less than 10% cumulative prevalence over a specified time horizon).
  • absolute risk and relative risk ratios as defined elsewhere in this disclosure can be employed to determine the degree of clinical utility.
  • Populations of subjects to be tested can also be categorized into quartiles by the test's measurement values, where the top quartile (25% of the population) comprises the group of subjects with the highest relative risk for therapeutic unresponsiveness, and the bottom quartile comprising the group of subjects having the lowest relative risk for therapeutic unresponsiveness
  • the top quartile (25% of the population) comprises the group of subjects with the highest relative risk for therapeutic unresponsiveness
  • the bottom quartile comprising the group of subjects having the lowest relative risk for therapeutic unresponsiveness
  • values derived from tests or assays having over 2.5 times the relative risk from top to bottom quartile in a low prevalence population are considered to have a "high degree of diagnostic accuracy," and those with five to seven times the relative risk for each quartile are considered to have a "very high degree of diagnostic accuracy.”
  • values derived from tests or assays having only 1.2 to 2.5 times the relative risk for each quartile remain clinically useful are widely used as risk factors for a disease; such is the case with total cholesterol and for many inflammatory biomark
  • a health economic utility function is an yet another means of measuring the performance and clinical value of a given test, consisting of weighting the potential categorical test outcomes based on actual measures of clinical and economic value for each.
  • Health economic performance is closely related to accuracy, as a health economic utility function specifically assigns an economic value for the benefits of correct classification and the costs of misclassification of tested subjects.
  • As a performance measure it is not unusual to require a test to achieve a level of performance which results in an increase in health economic value per test (prior to testing costs) in excess of the target price of the test.
  • diagnostic accuracy is commonly used for continuous measures, when a disease category or risk category has not yet been clearly defined by the relevant medical societies and practice of medicine, where thresholds for therapeutic use are not yet established, or where there is no existing gold standard for diagnosis of the pre-disease.
  • measures of diagnostic accuracy for a calculated index are typically based on curve fit and calibration between the predicted continuous value and the actual observed values (or a historical index calculated value) and utilize measures such as R squared, Hosmer- Lemeshow P-value statistics and confidence intervals.
  • DNARMARKERS In general, by defining the degree of diagnostic accuracy, i.e., cut points on a ROC curve, defining an acceptable AUC value, and determining the acceptable ranges in relative concentration of what constitutes an effective amount of the DNARMARKERS of the invention allows for one of skill in the art to use the DNARMARKERS to identify, diagnose, or prognose subjects with a pre-determined level of predictability and performance.
  • a "panel” within the context of the present invention means a group of biomarkers (whether they are
  • DNARMARKERS clinical parameters, or traditional laboratory risk factors
  • a panel can also comprise additional biomarkers, e.g., clinical parameters, traditional laboratory risk factors, known to be present or associated with responsiveness to chemotherapeutic treatment, in combination with a selected group of the DNARMARKERS listed in Table 1 or Table 2.
  • biomarkers listed in Table 3 are meausured
  • DNARMARKERS As noted above, many of the individual DNARMARKERS, clinical parameters, and traditional laboratory risk factors listed, when used alone and not as a member of a multi- biomarker panel of DNARMARKERS, have little or no clinical use in reliably distinguishing individuals that are responsive to therapeutic treatment and those that are not and thus cannot reliably be used alone in classifying any subject between those two states. Even where there are statistically significant differences in their mean measurements in each of these populations, as commonly occurs in studies which are sufficiently powered, such biomarkers may remain limited in their applicability to an individual subject, and contribute little to diagnostic or prognostic predictions for that subject.
  • a common measure of statistical significance is the p- value, which indicates the probability that an observation has arisen by chance alone; preferably, such p-values are 0.05 or less, representing a 5% or less chance that the observation of interest arose by chance. Such p-values depend significantly on the power of the study performed.
  • DNARMARKER performance Despite this individual DNARMARKER performance, and the general performance of formulas combining only the traditional clinical parameters and few traditional laboratory risk factors, the present inventors have noted that certain specific combinations of two or more DNARMARKERS can also be used as multi-biomarker panels comprising combinations of DNARMARKERS that are known to be involved in one or more physiological or biological pathways, and that such information can be combined and made clinically useful through the use of various formulae, including statistical classification algorithms and others, combining and in many cases extending the performance
  • DNARMARKERS are combined into novel and more useful combinations for the intended indications, is a key aspect of the invention.
  • Multiple biomarkers can often yield better performance than the individual components when proper mathematical and clinical algorithms are used; this is often evident in both sensitivity and specificity, and results in a greater AUC.
  • the suboptimal performance in terms of high false positive rates on a single biomarker measured alone may very well be an indicator that some important additional information is contained within the biomarker results - information which would not be elucidated absent the combination with a second biomarker and a mathematical formula.
  • DNARMARKERS can be advantageously used. Pathway informed seeding of such statistical classification techniques also may be employed, as may rational approaches based on the selection of individual DNARMARKERS based on their participation across in particular pathways or physiological functions.
  • formula such as statistical classification algorithms can be directly used to both select DNARMARKERS and to generate and train the optimal formula necessary to combine the results from multiple DNARMARKERS into a single index.
  • techniques such as forward (from zero potential explanatory parameters) and backwards selection (from all available potential explanatory parameters) are used, and information criteria, such as AIC or BIC, are used to quantify the tradeoff between the performance and diagnostic accuracy of the panel and the number of DNARMARKERS used.
  • information criteria such as AIC or BIC
  • any formula may be used to combine DNARMARKER results into indices useful in the practice of the invention.
  • indices may indicate, among the various other indications, the probability, likelihood, absolute or relative chance of responding to chemotherapy. This may be for a specific time period or horizon, or for remaining lifetime risk, or simply be provided as an index relative to another reference subject population.
  • model and formula types beyond those mentioned herein and in the definitions above are well known to one skilled in the art.
  • the actual model type or formula used may itself be selected from the field of potential models based on the performance and diagnostic accuracy characteristics of its results in a training population.
  • the specifics of the formula itself may commonly be derived from DNARMARKER results in the relevant training population.
  • such formula may be intended to map the feature space derived from one or more DNARMARKER inputs to a set of subject classes (e.g. useful in predicting class membership of subjects as normal, responders and non-responders), to derive an estimation of a probability function of risk using a Bayesian approach (e.g. the risk of cancer or a metastatic event), or to estimate the class-conditional probabilities, then use Bayes' rule to produce the class probability function as in the previous case.
  • subject classes e.g. useful in predicting class membership of subjects as normal, responders and non-responders
  • a probability function of risk e.g. the risk of cancer or
  • Preferred formulas include the broad class of statistical classification algorithms, and in particular the use of discriminant analysis.
  • the goal of discriminant analysis is to predict class membership from a previously identified set of features.
  • LDA linear discriminant analysis
  • features can be identified for LDA using an eigengene based approach with different thresholds (ELD A) or a stepping algorithm based on a multivariate analysis of variance (MANOVA). Forward, backward, and stepwise algorithms can be performed that minimize the probability of no separation based on the Hotelling-Lawley statistic.
  • Eigengene-based Linear Discriminant Analysis is a feature selection technique developed by Shen et al. (2006). The formula selects features (e.g. biomarkers) in a multivariate framework using a modified eigen analysis to identify features associated with the most important eigenvectors. "Important” is defined as those eigenvectors that explain the most variance in the differences among samples that are trying to be classified relative to some threshold.
  • a support vector machine is a classification formula that attempts to find a hyperplane that separates two classes.
  • This hyperplane contains support vectors, data points that are exactly the margin distance away from the hyperplane.
  • the dimensionality is expanded greatly by projecting the data into larger dimensions by taking non-linear functions of the original variables (Venables and Ripley, 2002).
  • filtering of features for SVM often improves prediction.
  • Features e.g., biomarkers
  • KW non-parametric Kruskal-Wallis
  • a random forest (RF, Breiman, 2001) or recursive partitioning (RPART, Breiman et al., 1984) can also be used separately or in combination to identify biomarker combinations that are most important. Both KW and RF require that a number of features be selected from the total. RPART creates a single classification tree using a subset of available biomarkers.
  • an overall predictive formula for all subjects, or any known class of subjects may itself be recalibrated or otherwise adjusted based on adjustment for a population's expected prevalence and mean biomarker parameter values, according to the technique outlined in D'Agostino et al, (2001) JAMA 286: 180-187, or other similar normalization and recalibration techniques.
  • epidemiological adjustment statistics may be captured, confirmed, improved and updated continuously through a registry of past data presented to the model, which may be machine readable or otherwise, or occasionally through the retrospective query of stored samples or reference to historical studies of such parameters and statistics.
  • numeric result of a classifier formula itself may be transformed post-processing by its reference to an actual clinical population and study results and observed endpoints, in order to calibrate to absolute risk and provide confidence intervals for varying numeric results of the classifier or risk formula.
  • An example of this is the presentation of absolute risk, and confidence intervals for that risk, derivied using an actual clinical study, chosen with reference to the output of the recurrence score formula in the Oncotype Dx product of Genomic Health, Inc. (Redwood City, CA).
  • a further modification is to adjust for smaller sub-populations of the study based on the output of the classifier or risk formula and defined and selected by their Clinical Parameters, such as age or sex.
  • Tumor specimens from patients were prepared by FFPE practice and serial sections of the tumor were prepared onto microscope slides.
  • DNA repair biomarkers are evaluated with antibodies directed against specific constituents of the DNA repair pathways. These proteins are either evaluated singly, or in a multiplexed fashion depending on the detection strategies employed.
  • IHC and developed sections were prepared with colorimetric detection strategies such that the stained slides are viewed by light microscopy, and imaged on the Aperio Scanscope Digital Pathology platform. There are many comparable platforms that allow rapid scanning of slides and conversion of the slide into a digital image file.
  • Tumor zones within a complex pathological specimen were identified by a trained pathologist.
  • computer assisted histopathology and morphologic criteria were included to discriminate tumor from non-tumor.
  • a computer-guided scoring strategy was directed specifically to the annotated regions.
  • Annotated tumor regions were scored via automated algorithms written and employed versus the specific stains. There are many alternative algorithms that are capable of representing the different features of a digital image file of a pathology specimen. The key features of the analysis were:
  • a weighted scoring system is utilized whereby the % of cells scored as 3 is weighted more heavily than those scored as 2 and those scored as 2 weighted more heavily than the % scored as 1.
  • One of the key metrics that the IHC staining for a given biomarker is a suitable dynamic range (scorable QIM) for utility in the ability to separate patient groups based on response and non-response in ovarian cancer predictive treatment based on biomarker panel assembly.
  • Increased or decreased activity of a pathway may impart sensitivity to DNA- or mitotic-targeted chemotherapies, or to enzyme/protein targeted chemotherapies, such as PARP inhibitors.
  • FIGS 1 through 6 highlight the scorable dynamic ranges for biomarkers MLH1, pMAPKAPK2, PAR, PARP1, XPF, and KI67, respectively.
  • biomarkers MLH1, pMAPKAPK2, PAR, PARP1, XPF, and KI67 For each of these markers with roles in DNA repair, one can focus on the dynamic ranges that each displays at the point residing at the nuclear localization. In the case of some biomarkers, their cellular localization may actually change depending on characteristics such as post-translational modification state of the protein, cell-cycle point, engagement state of DNA repair mechanisms, proliferative state of the cell, etc. Such is the case for pMAPKAPK2 (Figure 2) that displays both nuclear and cytoplasmic localization - independent metrics from which to separate responders from non-responders in ovarian cancer treatment regimen efficacy prediction modeling. It is also interesting to note here that these dynamic ranges of QIM scores can be obtained for ovarian cells of different origins including normal, carcinoma, and regional and distal metastases.
  • biomarkers such as FancD2 and pH2AX in addition to nuclear IHC staining may additionally display intense nuclear staining in the form of foci, most likely representing a granular depiction at a site of DNA repair in which the protein itself has focused its location and action (Figure 7).
  • Figure 9 shows that pH2AX is another such biomarker whose expression can be represented by measurement of either nuclear IHC staining or enumeration of foci.
  • Figure 10 compares FancD2 and pH2AX IHC expression as well as enumeration of foci in independent patients and illustrates that these two biomarkers display expression patterns that move independently of one another and as such are not prone to redundancy when utilized together in mutimarker modeling of ovarian cancer therapeutic response.
  • Example 2 Evalaution of Ovarian Cancer Patient Specimens By Tissue Micro Arrays
  • Tissue Micro Arrays Tissue Micro Arrays
  • Figure 11 shows one such tissue microarray.
  • multiple cores from each patient's FFPE tumor usually three or four
  • Slide-to-slide variability is minimized in the staining and scoring of large numbers of samples as more information with direct comparability from a single stained slide may be obtained in an efficient manner.
  • Results from multiple cores of individual patients are scored and assembled into ranked distribution plots as shown in Figure 12 indicating what the dynamic range for a given biomarker
  • An average QIM score is generated in plots such as these is comprised of several cores derived from the same patient that may display heterogeneity in expression for a given biomarker as demonstrated by the error bars represented with each of the given biomarker distribution plots.
  • DNA repair biomarkers expression patterns can clearly be impacted by tumor heterogeneity.
  • two markers are evaluated in serial FFPE sections from the tumor of a patient with serous ovarian cancer (Figure 13). FANCD2 and pMK2 IHC determinations are illustrated. The demarcated zones illustrate the marker heterogeity in the tumor from a single patient. Although these proteins are both in the FA/HR DNA repair pathway and the DNA damage response network, their expression is divergent from the perspective of a single patients tumor. Other molecular analysis criteria, such as RNA expression and DNA analysis, as well as proteomic analysis from tumor isolates, do not adequately represent the diversity exhibited within a single tumor that might be relevant to therapy selection and responsiveness. Scores for heterogeneity are another means to determine differences between patients with regard to biomarker panels and response algorithms.
  • APLF aprataxin- and PNK- 164. DDR like factor
  • TDP1 245.
  • Non-classified TOPOl 255.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pathology (AREA)
  • Organic Chemistry (AREA)
  • Analytical Chemistry (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Biophysics (AREA)
  • General Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

This present provides methods of treating cancer and methods of accessing/monitoring the responsiveness of a cancer cell to a therapeutic compound.

Description

BLOMARKERS FOR THE IDENTIFICATION MONITORING AND
TREATMENT OF OVARIAN CANCER
RELATED APPLICATIONS
[0001] This application claims the benefits of U.S. provisional application No.
61/370,546, filed August 4, 2010, which is incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
[0002] This invention relates generally to methods of methods of diagnosing and treating ovarian cancer. More specifically, the invention relates to methods of assessing the responsiveness of an ovarian cancer cell to a therapeutic compound.
BACKGROUND OF THE INVENTION
[0003] DNA repair refers to a collection of processes by which a cell identifies and corrects damage to the DNA molecules that encode its genome. In human cells, both normal metabolic activities and environmental factors such as UV light can cause DNA damage, resulting in as many as 1 million individual molecular lesions per cell per day. Many of these lesions cause structural damage to the DNA molecule and can alter or eliminate the cell's ability to transcribe the gene that the affected DNA encodes. Other lesions induce potentially harmful mutations in the cell's genome, which will affect the survival of its daughter cells after it undergoes mitosis. Consequently, the DNA repair process must be constantly active so it can respond rapidly to any damage in the DNA structure.
[0004] The rate of DNA repair is dependent on many factors, including the cell type, the age of the cell, and the extracellular environment. A cell that has accumulated a large amount of DNA damage, or one that no longer effectively repairs damage incurred by its DNA, can enter one of three possible states: an irreversible state of dormancy, known as senescence; cell suicide, also known as apoptosis or programmed cell death or unregulated cell division, which can lead to the formation or perpetuation of a tumor.
[0005] Generally speaking with regards to management of oncology patients, tailored chemotherapeutic regimens that may or may not be combined with surgery and/or radiation can be placed into defined functional categories: (1) Microtubule disrupting agents
(Docetaxel Paclitaxel, Vinorelbine) (2) DNA adduct-forming platinating compounds (Carboplatin, Cisplatin, Oxaliplatin) (3) Base synthesis inhibitors (Pemetrexed) (4) Modified bases (gemicitibine) and (5) Tyrosine kinase inhibitors (cetuximab, gefitinib - used in tumors identified as EGFR driven). For more aggressive tumors, combinatorial approaches among several regimens may be undertaken. For instance, adenocarcinomas are indicated to benefit from microtubule disrupting agents in combination with platinating compound therapy. Squamous cell tumors may benefit from combined platinating compounds in unison with gemcitibine. In addressing the combination of chemo and surgery (in some cancer instance also combined with radiation) with improved patient response and survival as the goal, a platinum response predictor could better inform the relative weight of chemotherapy in a multidisciplinary treatment regimen. A platinum response predictor has direct relevance due to the wide range of chemotherapies available.
[0006] Ovarian cancer is the seventh most common cancer in the United States. It occurs most frequently in patients who are between 40 and 65 years of age. The lifetime risk of developing ovarian cancer is 1.4 to 1.8 percent in the United States. There are several different types of cancer that can arise in the ovary, but epithelial ovarian cancer (usually synonomous as ovarian cancer) is the most common.
[0007] The first step in treating ovarian cancer usually involves surgical removal of cancerous tissue (surgical debulking or cytoreduction). Achieving optimal debulking improves outcomes/prognoses and may also influence the choice of chemotherapy treatment.
[0008] For selected patients with stage IA and IB disease, surgery alone is effective in treating the disease, and no additional therapy is recommended. However, in most other cases, chemotherapy is recommended post surgery. Occasionally, it is too risky to perform initial surgery because of the extensive nature of the cancer and/or the debilitated condition of the patient. In such cases, neoadjuvant chemotherapy may be recommended as a first step in the treatment process.
[0009] There is controversy as to whether the long-term outcomes in patients who are given neoadjuvant chemotherapy before surgery are as favorable as those in patients who have aggressive surgery followed by chemotherapy (Bristow and Chi, 2006). In general, initial surgery is preferred if optimal debulking surgery is possible and the patient can safely tolerate it. Neoadjuvant chemotherapy is a reasonable approach for patients who could not tolerate a lengthy operation and recovery period. In such cases, initial chemotherapy can shrink the tumor and improve a patient's overall medical condition, making her a better candidate for later debulking surgery.
[00010] Among the chemotherapeutic agents most commonly used in the treatment of ovarian cancer are microtubule disrupting agents (e.g. paclitaxel and docetaxel), and platinating compounds (e.g. carboplatin and cisplatin). Research suggests that chemotherapy regimens that contain these drugs are effective in preventing recurrence of ovarian cancer, and improve survival.
[00011] At least three research studies support the superiority of paclitaxel-containing chemotherapy over other combinations that do not contain paclitaxel (McGuire et al, 1996; Muggia et al., 2000; Piccart et al., 2000). As a result, the combination of a platinum-type drug (usually carboplatin) and paclitaxel is standard therapy for the first-line treatment of patients with ovarian cancer who require chemotherapy. Carboplatin appears to be as effective as cisplatin, and has fewer side effects.
[00012] Following surgery, patients with stage IA or IB ovarian cancers that appear to be fairly nonaggressive (described as low-grade or grade 1) do well without chemotherapy, and observation alone is a standard approach. In contrast, patients with higher grade (grade 2 or 3), stage IA or IB ovarian cancers, and all patients with stage IC ovarian cancer (regardless of the grade or level of aggressiveness) are usually advised to have adjuvant chemotherapy. The standard regimen includes a platinum-based combination such as paclitaxel and carboplatin. A potentially less toxic alternative to combination therapy is single agent therapy with carboplatin alone (ICON Group, 2002). This approach is more often used in Europe, and is considered controversial in the United States.
[00013] Patients with stage II disease have a prognosis that is slightly worse than those with stage I disease, but better than those with stage III or IV disease. Although there are few trials validating this approach, many groups suggest the use of six cycles of intravenous paclitaxel plus carboplatin, similar to the regimen used for patients with stage III/IV disease. An acceptable alternative is to use a combination of intraperitoneal and intravenous chemotherapy. However, the use of IP chemotherapy after surgery for stage II ovarian cancer is controversial since the data supporting this treatment was derived from studies of patients with stage III disease.
[00014] Adjuvant therapy is also an important clinical activity for ovarian cancer patients, as there is a demonstrated benefit to this process post-therapy. For Ovarian cancers in the United States, the standard course of chemotherapy after surgical debulking for stage III or IV ovarian cancer includes six cycles of paclitaxel plus carboplatin. Each cycle given over three weeks and treatment usually begins within two to six weeks after surgery. However, the preferred treatment for all patients is enrollment in a clinical trial.
[00015] In contrast to almost all other cancers, ovarian cancer typically does not spread through the bloodstream. Instead, tumor growth is often limited to the abdominal (peritoneal) cavity, even in advanced cases. Because the bulk of cancerous tissue is found within the peritoneal cavity, treatment approaches give chemotherapy directly into this body cavity, an approach that is called intraperitoneal (IP) chemotherapy. The advantage of IP therapy compared to the intravenous (IV) route is that higher doses of the drugs can be given directly to the areas at highest risk for tumor involvement.
[00016] In a study conducted in several American hospitals (Armstrong et al., 2006), IP chemotherapy (cisplatin and paclitaxel) was given in combination with intravenous paclitaxel and compared to the standard regimen of all IV treatment with paclitaxel plus cisplatin. Participants included patients with optimally debulked stage III ovarian cancer. Patients who were given IP chemotherapy had a 16 month longer survival as compared to patients who were given all of their chemotherapy IV, even though most of the patients did not take the entire six courses of IP treatment because of side effects. The benefits here are
counterbalanced by a higher number of treatment-related side effects. These include both catheter-related complications (infection, blockage or leakage of the catheter) as well as non- catheter-related problems (low white and red blood cell counts, nausea and vomiting, abdominal pain, and neurologic side effects such as numbness and tingling of the fingers and toes).
[00017] The likelihood of ovarian cancer recurrence after initial surgery and chemotherapy has fostered efforts to improve outcomes by increasing intensity and/or duration of chemotherapy. In general, research studies do not support any benefit from increasing the chemotherapy dose (ie, high-dose chemotherapy, sometimes referred to as a "bone marrow transplant").
[00018] The value of giving "maintenance" chemotherapy after the patient achieves a complete response to first-line chemotherapy has been debated. In an early study, patients who were given 12 additional months of paclitaxel alone had a 7 month delay in disease progression compared to those who were given 3 additional months (Markman et al., 2003). However, this study did not prove that more patients were cured by the longer course of chemotherapy. In addition, the benefit of the seven month delay was achieved at the cost of additional chemotherapy side effects. Furthermore, a preliminary report of a second study, the Italian After-6 protocol, failed to show any benefit for a shorter course (6 rather than 12 months) of maintenance paclitaxel in patients who obtained a complete response after surgery and six cycles of paclitaxel and carboplatin (Conte etal, 2007). Compared to observation alone, maintenance therapy did not reduce the risk of disease relapse or improve survival.
[00019] At the end of treatment (both surgery and chemotherapy), "complete response" is achieved if there is no evidence of cancer on imaging studies (such as a CT scan), and the blood levels of CA-125 are normal. However, even when these criteria are met, microscopic residual cancer can still be present. To monitor for recurrent cancer, follow-up blood tests, physical examinations, and imaging tests are recommended for at least five years after treatment ends.
[00020] Patients with recurrent ovarian cancer following an initial complete response, and those who do not respond well to initial chemotherapy are candidates for second- line chemotherapy. The choice of chemotherapy agents for second-line treatment depends upon whether, and how well, the patient responded to first-line treatment with paclitaxel and either carboplatin or cisplatin. If the patient had a complete response, and the response lasted for at least six to 12 months, research has shown that a good response is possible with a second course of the same regimen. In fact, if the initial response lasted longer than 24 months, up to one-fourth of patients will have a complete response to retreatment with the same regimen. Alternately, a regimen including carboplatin with either gemcitabine or pegylated liposomal doxorubicin (PLD) may be used.
[00021] If the patient did not respond well to first- line therapy with paclitaxel and a platinum agent or if she relapses within six months of completing such therapy, a different non-platinum-containing regimen may be considered. Usually, a single drug rather than combination chemotherapy is recommended in these cases. A variety of agents may be considered, including pegylated liposomal doxorubicin, topotecan, docetaxel, oral etoposide, gemcitabine, vinorelbine, if osf amide, leucovorin- modulated 5-fluorouracil, or tamoxifen.
[00022] Molecularly-targeted therapy may also be of benefit for second- line therapy. The drug bevacizumab interacts with a protein called vascular endothelial growth factor (VEGF). VEGF is involved in the development of a blood supply within a growing cancer; this blood supply is essential for the tumor to grow and spread. Bevacizumab interferes with the blood supply of the tumor by interacting with VEGF, causing the tumors to shrink. Bevacizumab also enhances the antitumor effect of conventional chemotherapy drugs, and is used in combination chemotherapy regimens for the treatment of other advanced tumors, such as colorectal, ovarian, lung, and renal cell cancer. Bevacizumab is effective for the treatment of ovarian cancer, and it may be considered for patients with platinum-resistant ovarian cancer, either alone or in combination with chemotherapy.
[00023] The utility and need for molecular diagnostic tests are twofold relative to application to ovarian cancer management. First, patient populations are amalgams of many different global expression profiles. Treatment regimens and drug approvals are guided by a determination of what is likely to be beneficial to a group of patients, and are often not strictly associated with individual patient stratification. The expression of statistical trends during treatment effect, such as Kaplan-Meyer survival, response, or time to failure representations, will identify survival of a patient population following treatment with a given therapeutic regimen. However, as each of the constituent patients' tumors within this population may be expected to have a distinct proteomic profile or tumor biology, one might expect that the patients responses to therapeutic regimens might segregate along specific expression patterns for defined sets of markers. Indeed separating the patient populations from Kaplan- Meyer survival curves based upon these defined discriminators into responders and non-responders may be imminently applicable in allowing physicians to prescribe a regimen with added survival benefit. Indeed, as chemotherapy by definition is toxic (poison), patient non-responders defined by biomarkers may allow a decision to withhold
chemotherapy over prescribing the chemotherapy as the best strategy to maximize a survival benefit.
[00024] Second, in clinical studies comprising drug discovery efficacy assessments a candidate therapeutic is tested against a patient population and then some level of response is measured. But, as noted above in Kaplan- Meyer survival curves, the patient population is an amalgam of many different molecular profile subtypes. Therefore, the likelihood that the patient population will respond universally to the candidate therapeutic is small. Such was the case with Iressa (gefitinib). A tyrosine kinase inhibitor targeting EGFR in Lung, clinical trials efficacy hovered around 15% response for the test population. Faced with the data of an 85% ineffective application to the total patient population, the drugs approval was called into question. Further research showed that that in patients comprising the 15% response group, that EGFR was modified (driver) for all patients within that group. The application of similar proteomic expression profiles will allow for better stratified patient selection and clinical trial design as well as more targeted and streamlined testing, approval and release of new therapeutics with real benefit to select individuals with the correct proteomic
background.
[00025] DNA repair genes and/or proteins are already known to provide meaningful data relative to risk and prognosis, as well as offer valuable insights at decision inflection points for predictive therapeutic response in personalized medicine. While risk and prognostic significance should not be discounted, informations leading to correct decisions in patient management should prove most beneficial relative to actionable information leading to improved patient care and survival.
[00026] Multiple reports in the literature already indicate that DNA repair biomarker expression or lack thereof is strongly correlative patient subgroups more or less likely to respond to a specific prescribed chemotherapy. The rationale behind use of platinating compounds either following or preceding surgery is that the most actively dividing cells will incur the greatest DNA damage and thus suffer the most toxicity at the hands of the affecting agent(s). In theory, the cells that are most prone to induced lethality would have less efficient DNA repair mechanisms and those with efficient mechanisms would prove resistant to the agent of insult. While multiple DNA repair pathways are employed in cell maintenance, many of these already hold predicative value to chemotherapeutic susceptibility/resistance modeling studies relative to constituent biomarker expression profiles.
Summary of the Invention
[00027] The present invention relates in part to the discovery that certain biological markers (referred to herein as "DNARMARKERS"), such as proteins, nucleic acids, polymorphisms, metabolites, protein modifications, nucleic acid modifications,
chromosomes, and other analytes, as well as certain physiological conditions and states, are present or altered in subjects with an increased risk of developing a recurrent ovarian cancer.
[00028] The present invention also provides methods of assessing the likelihood of a local and/or distant recurrence or overall survival or disease free survival or response to therapy by a patient with ovarian cancer by measuring the level of an effective amount of one or more DNARMARKERS in a sample from the subject.
[00029] Risk of early local and/or distant recurrence or decreased overall survival or disease free survival or response to therapy by a patient with ovarian cancer by measuring the level of an effective amount of DNARMARKERs in a sample from the subject. The sample can contain ovarian tumor or normal ovarian tissue or a blood sample. The tissue is for example a paraffin embedded tissue, a fresh tissue, or a frozen tissue sample.
[00030] An increased risk of developing a recurrence of ovarian cancer in the subject is determined by measuring a clinically significant alteration in the level of the
DNARMARKER in the sample. Alternatively, an increased risk of developing a recurrence of ovarian cancer in the subject is determined by comparing the level of the effective amount of DNARMARKER to a reference value. In some aspects the reference value is an index.
[00031] In another aspect the invention provides a method with a predetermined level of predictability for assessing the progression of a ovarian cancer in a
subject by detecting the level of an effective amount a DNARMARKERS in a first sample from the subject at a first period of time, detecting the level of an effective amount of DNARMARKERS in a second sample from the subject at a second period of time and comparing the level of the DNARMARKERS detected to a reference value.
In some aspects the first sample is taken from the subject prior to being treated for the ovarian cancer and the second sample is taken from the subject after being
treated for the cancer. Treatment can include chemotherapy, or radiation therapy.
[00011] In a further aspect the invention provides a method with a predetermined level of predictability for monitoring the effectiveness of treatment or selecting a treatment regimen for ovarian cancer by detecting the level of an effective amount of
DNARMARKERS in a first sample from the subject at a first period of time and
optionally detecting the level of an effective amount of DNARMARKERS in a second sample from the subject at a second period of time. The level of the effective amount of DNARMARKERS detected at the first period of time is compared to the level detected at the second period of time or alternatively a reference value. Effectiveness of treatment is monitored by a change in the level of the effective amount of DNARMARKERS from the subject.
[00032] Treatment is for example, chemotherapy and /or radiotherapy. Chemotherapeutic agents include a platinating agent (e.g., carpoplatin or oxiplatin), taxane, a PARP inhibitor and any combination of these.
[00033] A DNARMARKER includes for example MLH 1 , pMAPKAPK2, PAR, PARP 1 , XPF, pH2A2and FAND2. One, two, three, four, five, ten or more DNARMARKERS are measured. Optionally Ki67 is measured. Additionally any other biomarker listed on Tables 1,2 or 3 are measured. In another embodimentthe DNARMARKER is XPF, FANCD2, pMK2, PAR, MLH1, PARP1, BRCA1, RAD51, POL H, ATM, or ERCC1.
[00034] In a further aspect the DNARMARKERS are DNA repair proteins belonging to different DNA repair pathways. Alternatively three or more DNARMARKERS are
[00035] measured where DNARMARKERS belonging to two or more different DNA repair pathways. The level of a DNARMARKER is measured electrophoretically, or immunochemically, or genetically. For example the level of the DNARMARKER is detected by immunohistochemistry, radioimmunoassay, immunofluorescence assay, by an enzyme-linked immunosorbent assay, by genotyping, or by fluorescence in situ hybridization.
[00036] The subject has ovarian cancer, or a recurrent ovarian cancer. In some aspects the sample is taken for a subject that has previously been treated for ovarian cancer.
Alternatively, the sample is taken from the subject prior to being treated for ovarian cancer. The sample is a tumor biopsy such as a core biopsy, a needle biopsy, an excisional tissue biopsy or an incisional tissue biopsy. Additionally, the sample is a tumor cell from blood, lymph nodes or a bodily fluid.
[00037] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are expressly incorporated by reference in their entirety. In cases of conflict, the present specification, including definitions, will control.
[00038] In addition, the materials, methods, and examples described herein are illustrative only and are not intended to be limiting.
[00039] Other features and advantages of the invention will be apparent from and encompassed by the following detailed description and claims.
BRIEF DESCRIPTION OF THE DRAWINGS
[00040] Figure 1 shows MLH1 IHC staining and patient distributions based on image analysis and quantification. MLH1 IHC nuclear- localized staining for both positive and negative samples is depicted in the panels to the top. A TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear staining intensity of individual cells. A broad dynamic range of expression is achieved with this biomarker which may assist in separating patient groups into responders and non-responders.
[00041] Figure 2 shows phosphoMAPKAPK2 IHC staining and patient distribution based on image analysis and quantification. As shown in the figure phosphoMAPKAPK2 IHC nuclear and IHC cytoplasmic -localized staining for both positive and negative samples are depicted in the panels to the top. A TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear and cytoplasmic -staining intensity of individual cells. Here two separate metrics may be utilized, both nuclear and cytoplasmic intensities. The figure further depicts that many cells stain not merely as nuclear or cytoplasmic, but that many have both nuclear and cytoplasmic staining.
[00042] Figure 3 shows PAR IHC staining and patient distribution based on image analysis and quantification. PAR IHC nuclear- localized staining for both positive and negative samples is depicted in the panels to the top. A TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear staining intensity of individual cells.
[00043] Figure 4 shows PARP1 IHC staining and patient distribution based on image analysis and quantification. PARP1 IHC nuclear- localized staining for both positive and negative samples is depicted in the panels to the top. A TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear staining intensity of individual cells.
[00044] Figure 5 shows XPF IHC staining and patient distribution based on image analysis and quantification. XPF IHC nuclear-localized staining for both positive and negative samples is depicted in the panels to the top. A TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear staining intensity of individual cells.
[00045] Figure 6 show Ki67 IHC staining and patient distribution based on image analysis and quantification. Ki67 IHC nuclear- localized staining for both positive and negative samples is depicted in the panels to the top. A TMA comprising normal ovarian tissue, serous ovarian carcinoma, lymph node metastasis and distal metastasis were scored using tumor recognition and digital pathology for nuclear staining intensity of individual cells. [00046] Figure 7 shows FancD2 IHC Staining. FancD2 IHC nuclear-localized staining for both positive and negative samples is depicted in the panels to the top. Note that the staining for this biomarker may be represented by broad nuclear localization or more granular foci presumably occurring as a micro-localization of FancD2 to repair sites at specific DNA loci.
[00047] Figure 8 demonstrates patient variability in FancD2 expression and nuclear foci. FancD2 staining may be scored by quantifying general nuclear intensity of IHC stain or by enumeration of FancD2 foci. Note that regarding patient ID, that these 2 metrics are not necessarily correlative and may provide independent metrics from a single marker by which to delineate therapeutic response in ovarian cancer treatment.
[00048] Figure 9 shows phosphoH2AX staining and patient distribution based on image analysis and quantification. Like FancD2, staining for phosphoH2AX IHC may be represented by broad nuclear localization or more granular foci presumably occurring as a micro-localization of repair sites at specific DNA loci. phosphoH2AX staining may be scored by quantifying general nuclear intensity (here by binning the data as 1+, 2+, 3+) of IHC stain or by enumeration of phosphoH2AX foci. Note that regarding patient ID, that these 2 metrics are not necessarily correlative and may provide independent metrics from a single marker by which to delineate therapeutic response in ovarian cancer treatment.
[00049] Figure 10 shows the comparison of FancD2 and phosphoH2AX Scoring: Nuclear IHC and Foci Enumeration. Comparison of staining for H2AX and FancD2 by either nuclear intensity with qualtity of cells staining as positive and % of cells exhibiting nuclear foci indicate that the two markers are not redundant with one another and may provide ortholgous metrics by which to separate patients based on response.
[00050] Figure 11 shows patient IHC staining differences for phosphoMAPKAPK2 displayed in TMA Format. Representative core samples were taken from ovarian tumors and arranged on Tissue Microarrays. In this format many samples can be stained and analyzed in a more time efficient fashion while at the same time limiting the effects of slide-to-slide and day-to-day variability.
[00051] Figure 12. shows TMA-based Rank-ordered Patient Group Distribution Plots for PAR, XPF, PARP1, and MLH1. Dynamic ranges for biomarker expressions are generated from TMA-based studies. The rank ordering shown here can be utilized to establish univariate analysis cut-points for responders versus non-responders. The error bars for individual patient specimens are reflections of core-to-core sampling and may represent heterogeneity present within the tumor.
[00052] Figure 13 shows tumor heterogeneity determined by two DNA repair biomarkers. The pMK2 and FANCD2 biomarkers were examined on serial FFPE sections from the same serous ovarian cancer tumor. Two zones are delineated in the figure. In one case (dashed circle), the region of the tumor is positive for both markers. In the other case (solid circle), the tumor cells are positive for FANCD2 staining and nuclear foci, but are determined to be negative for pMK2. Insert region, the pMK2 staining pattern is distinctively different in the two zones. In one case (cytoplasmic), there is predominantly cytoplasmic staining. In the other case (mitotic), there is punctuate nuclear staining at a small fraction of the tumor cells consistent with a mitotic cell pattern.
DETAILED DESCRIPTION OF THE INVENTION
[00053] The present invention relates to the identification of biomarkers associated with ovarian cancer. Specifically, these biomarkers are proteins associated in DNA repair pathways. DNA repair pathways are important to the cellular response network to chemotherapy and radiation.
[00054] More specifically, the invention relates to the observation that tumor cells have altered DNA repair and DNA damage response pathways and that loss of one of these pathways renders the cancer more sensitive to a particular class of DNA damaging agents. Cancer therapy procedures such as chemotherapy and radiotherapy work by overwhelming the capacity of the cell to repair DNA damage, resulting in cell death.
[00055] There are six major DNA repair pathways distinguishable by several criteria which can be divided into three groups those that repair single strand damage and those that repair double stand damage. These pathways include Base-Excision Repair (BER);
Nucleotide Excision Repair (NER); Mismatch Repair (MMR); Homologous
Recombination/Fanconi Anemia pathway (HR/FA); Non-Homologous Endjoining (NHEJ), and Translesion DNA Synthesis repair (TLS).
[00056] BER, NER and MMR repair single strand DNA damage. When only one of the two strands of a double helix has a defect, the other strand can be used as a template to guide the correction of the damaged strand. In order to repair damage to one of the two paired molecules of DNA, there exist a number of excision repair mechanisms that remove the damaged nucleotide and replace it with an undamaged nucleotide complementary to that found in the undamaged DNA strand. BER repairs damage due to a single nucleotide caused by oxidation, alkylation, hydrolysis, or deamination. NER repairs damage affecting longer strands of 2-30 bases. This process recognizes bulky, helix-distorting changes such as thymine dimers as well as single- strand breaks (repaired with enzymes such UvrABC endonuclease). A specialized form of NER known as Transcription-Coupled Repair (TCR) deploys high-priority NER repair enzymes to genes that are being actively transcribed. MMR corrects errors of DNA replication and recombination that result in mispaired nucleotides following DNA replication.
[00057] NHEJ and HR repair double stranded DNA damage. Double stranded damage is particularly hazardous to dividing cells. The NHEJ pathway operates when the cell has not yet replicated the region of DNA on which the lesion has occurred. The process directly joins the two ends of the broken DNA strands without a template, losing sequence information in the process. Thus, this repair mechanism is necessarily mutagenic. However, if the cell is not dividing and has not replicated its DNA, the NHEJ pathway is the cell's only option. NHEJ relies on chance pairings, or microhomologies, between the single-stranded tails of the two DNA fragments to be joined. There are multiple independent "failsafe" pathways for NHEJ in higher eukaryotes. Recombinational repair requires the presence of an identical or nearly identical sequence to be used as a template for repair of the break. The enzymatic machinery responsible for this repair process is nearly identical to the machinery responsible for chromosomal crossover during meiosis. This pathway allows a damaged chromosome to be repaired using the newly created sister chromatid as a template, i.e. an identical copy that is also linked to the damaged region via the centromere. Double- stranded breaks repaired by this mechanism are usually caused by the replication machinery attempting to synthesize across a single-strand break or unrepaired lesion, both of which result in collapse of the replication fork.
[00058] Translesion synthesis is an error-prone bypass method where a DNA lesion is left unrepaired during S phase, and is repaired later in the cell cycle. The DNA replication machinery cannot continue replicating past a site of DNA damage, so the advancing replication fork will stall on encountering a damaged base. The translesion synthesis pathway is mediated by specific DNA polymerases that insert alternative bases at the site of damage and thus allow replication to bypass the damaged base to continue with chromosome duplication. The bases inserted by the translesion synthesis machinery are template- independent, but not arbitrary; for example, one human polymerase inserts adenine bases when synthesizing past a thymine dimer. If this residue is not repaired at a later step, the process is mutagenic.
[00059] Cancer cells accumulate high levels of DNA damage. This damage may result from their heightened proliferative activity or from exposure to chemotherapy or ionizing radiation.
[00060] The literature indicates that biomarker expression from multiple DNA repair pathways may hold prognostic and/or predictive information in the management and treatment of ovarian cancer. Brustmann, (2007) suggests that the base excision repair enzymes (e.g PARP1) may serve as a marker for aggressive serous ovarian tumors while Fong et al. (2009) show relational crosstalk between BER and FA/HR by indicating that a PARP inhibitor exhibits anti-tumor activity in BRCA1 mutant ovarian cancers.
[00061] Several studies have now shown that high ERCCl expression (Nucleotide excision repair, NER) can be correlated with cisplatin resistance and potential better response to other classes of therapy. Smith, et al. (2007) have shown that patients with high ERCCl may benefit from paclitaxel +cisplatin and patients with low ERCCl are likely to benefit from cisplatin alone while Steffensen et al, (2009). Show that ERCCl expression correlated with carboplatin + paclitaxel resistance. Lin et al. (2008) have shown that ERCCl and XPD expression are associated with resistance to platinum-based chemotherapy. Other reports also show correlation for high levels of expression of XPF and XPG with respect to cisplatin resistance, sometimes accompanied by poor prognosis (Stevens et al 2008, Planchard et al. 2009, Bartolucci et al. 2009.)
[00062] Regarding the mismatch repair (MMR) DNA repair pathway, both MLH1 and MSH2 have been shown to undergo loss of expression following exposure to cisplatin in the treatment of ovarian cancer. Further, this loss of expression is correlative with improved overall survivial following chemotherapeutic treatment. (Materna et al, 2007; Scortozzi et al., 2003; Samimi et al 2000.)
[00063] From the Fanconi's Anmeia/Homologous recombination DNA repair pathway, the literature provides evidence for predictive value relative to response as well as prognostic guidance for at least 3 markers including BRCA1, FancD2, and ATM. Swisher et al. (2009) have shown that recurrent ovarian cancer displays BRCA1 elevation and cisplatin resistance while Husain et al.(1998) report similar findings with BRCA1 overexpression in cisplatin- resistant cells. In fact, BRCA status may be an indicator of susceptibility to treatment with paclitaxel with vinorelbine or radiation and resistance to etoposide and bleomycin (Quinn et al., 2003; Zhou et al., 2003). The importance of the pathway in general in cisplatin response is highlighted by further reports suggesting that a functional FanD2/BRCAl node of the FA/HR pathway leads to cisplatin resistance (Taniguchi et al., 2009) and that inhibition of FancD2 (Chirnomas et al., 2006) or deficiency of FancD2 (wang et al., 2006) may lead to restored cisplatin sensitivity and irofulven activity, respectively, in anti-ovarian cancer activities. Lastly, Fanc-deficient cells are susceptible to treatment with inhibitors to ATM (Kennedy et al., 2007).
[00064] Biomarkers of DNA repair may guide therapy because these are pathways that respond to damage to DNA caused by chemotherapy and radiation. Single pathways are not adequate to describe the DNA repair responses to even single chemotherapy agents for several reasons. First, chemotherapy causing DNA damage leads to many forms of DNA damage necessitating different biochemical mechanisms to repair. Second, the central DNA repair pathways can at least partially complement each other for cell survival. Even though the second pathway may not be as effective, it provides a salvage mechanism for the cell to return to cell division and growth. For a tumor cell, this is a mechanism for diminishing the strength of and/or evading cell cycle checkpoints.
[00065] Alterations in DNA repair genes and proteins have been identified in ovarian cancer patients and in some cases have been associated with clinical outcome or treatment efficacy. Profiling proteins from multiple pathways is likely to be a more informative approach to predicting the progression of disease or efficacy of the treatment.
[00066] In this study described herein, representatives from several of these pathways were investigated for associations with clinical outcome of individuals with ovarian cancer. Selected DNA repair protein epitopes in ovarian cancer patient samples were evaluated by immunohistochemistry. The DNA repair protein epitopes evaluated included MLHl, pMAPKAPK2, PAR, PARPl, XPF, pH2A2 and FAND2. Ki67 was also measured.
Additionally any other biomarker listed on Tables 1 ,2 or 3 are measured. In another embodiment the DNARMARKER is XPF, FANCD2, pMK2, PAR, MLHl, PARPl, BRCAl, RAD51, POL H, ATM, or ERCC1.
[00067] Definitions
[00068] "Accuracy" refers to the degree of conformity of a measured or calculated quantity (a test reported value) to its actual (or true) value. Clinical accuracy relates to the proportion of true outcomes (true positives (TP) or true negatives (TN) versus misclassified outcomes (false positives (FP) or false negatives (FN)), and may be stated as a sensitivity, specificity, positive predictive values (PPV) or negative predictive values (NPV), or as a likelihood, odds ratio, among other measures.
[00069] "Biomarker" in the context of the present invention encompasses, without limitation, proteins, nucleic acids, and metabolites, together with their polymorphisms, mutations, variants, modifications, subunits, fragments, protein-ligand complexes, and degradation products, protein-ligand complexes, elements, related metabolites, and other analytes or sample-derived measures. Biomarkers can also include mutated proteins or mutated nucleic acids. Biomarkers also encompass non-blood borne factors or non-analyte physiological markers of health status, such as "clinical parameters" defined herein, as well as "traditional laboratory risk factors", also defined herein. Biomarkers also include any calculated indices created mathematically or combinations of any one or more of the foregoing measurements, including temporal trends and differences. Where available, and unless otherwise described herein, determinants which are gene products are identified based on the official letter abbreviation or gene symbol assigned by the international Human Genome Organization Naming Committee (HGNC) and listed at the date of this filing at the US National Center for Biotechnology Information (NCBI) web site
[00070] "Clinical parameters" encompasses all non-sample or non-analyte biomarkers of subject health status or other characteristics, such as, without limitation, age (Age), ethnicity (RACE), gender (Sex), or family history (FamHX).
[00071] "FN" is false negative, which for a disease state test means classifying a disease subject incorrectly as non-disease or normal.
[00072] "FP" is false positive, which for a disease state test means classifying a normal subject incorrectly as having disease.
[00073] A "formula," "algorithm," or "model" is any mathematical equation, algorithmic, analytical or programmed process, or statistical technique that takes one or more continuous or categorical inputs (herein called "parameters") and calculates an output value, sometimes referred to as an "index" or "index value." Non-limiting examples of "formulas" include sums, ratios, and regression operators, such as coefficients or exponents, biomarker value transformations and normalizations (including, without limitation, those normalization schemes based on clinical parameters, such as gender, age, or ethnicity), rules and guidelines, statistical classification models, and neural networks trained on historical populations. Of particular use in combining DNARMARKERS and other biomarkers are linear and nonlinear equations and statistical classification analyses to determine the relationship between levels of DNARMARKERS detected in a subject sample and the subject's responsivenss to chemotherapy. In panel and combination construction, of particular interest are structural and synactic statistical classification algorithms, and methods of risk index construction, utilizing pattern recognition features, including established techniques such as cross-correlation, Principal Components Analysis (PCA), factor rotation, Logistic Regression (LogReg), Linear Discriminant Analysis (LDA), Eigengene Linear Discriminant Analysis (ELD A), Support Vector Machines (SVM), Random Forest (RF), Recursive Partitioning Tree (RPART), as well as other related decision tree classification techniques, Shrunken Centroids (SC), StepAIC, Kth-Nearest Neighbor, Boosting, Decision Trees, Neural Networks, Bayesian Networks, Support Vector Machines, and Hidden Markov Models, among others. Other techniques may be used in survival and time to event hazard analysis, including Cox, Weibull, Kaplan-Meier and Greenwood models well known to those of skill in the art. Many of these techniques are useful either combined with a DNARMARKER selection technique, such as forward selection, backwards selection, or stepwise selection, complete enumeration of all potential panels of a given size, genetic algorithms, or they may themselves include biomarker selection methodologies in their own technique. These may be coupled with information criteria, such as Akaike's Information Criterion (AIC) or Bayes Information Criterion (BIC), in order to quantify the tradeoff between additional biomarkers and model improvement, and to aid in minimizing overfit. The resulting predictive models may be validated in other studies, or cross-validated in the study they were originally trained in, using such techniques as Bootstrap, Leave-One-Out (LOO) and 10-Fold cross-validation (10-Fold CV). At various steps, false discovery rates may be estimated by value permutation according to techniques known in the art. A "health economic utility function" is a formula that is derived from a combination of the expected probability of a range of clinical outcomes in an idealized applicable patient population, both before and after the introduction of a diagnostic or therapeutic intervention into the standard of care. It encompasses estimates of the accuracy, effectiveness and performance characteristics of such intervention, and a cost and/or value measurement (a utility) associated with each outcome, which may be derived from actual health system costs of care (services, supplies, devices and drugs, etc.) and/or as an estimated acceptable value per quality adjusted life year (QALY) resulting in each outcome. The sum, across all predicted outcomes, of the product of the predicted population size for an outcome multiplied by the respective outcome's expected utility is the total health economic utility of a given standard of care. The difference between (i) the total health economic utility calculated for the standard of care with the intervention versus (ii) the total health economic utility for the standard of care without the intervention results in an overall measure of the health economic cost or value of the intervention. This may itself be divided amongst the entire patient group being analyzed (or solely amongst the intervention group) to arrive at a cost per unit intervention, and to guide such decisions as market positioning, pricing, and assumptions of health system acceptance. Such health economic utility functions are commonly used to compare the cost-effectiveness of the intervention, but may also be transformed to estimate the acceptable value per QALY the health care system is willing to pay, or the acceptable cost-effective clinical performance characteristics required of a new intervention.
[00074] For diagnostic (or prognostic) interventions of the invention, as each outcome (which in a disease classifying diagnostic test may be a TP, FP, TN, or FN) bears a different cost, a health economic utility function may preferentially favor sensitivity over specificity, or PPV over NPV based on the clinical situation and individual outcome costs and value, and thus provides another measure of health economic performance and value which may be different from more direct clinical or analytical performance measures. These different measurements and relative trade-offs generally will converge only in the case of a perfect test, with zero error rate (a.k.a., zero predicted subject outcome misclassifications or FP and FN), which all performance measures will favor over imperfection, but to differing degrees.
[00075] "Measuring" or "measurement," or alternatively "detecting" or "detection," means assessing the presence, absence, quantity or amount (which can be an effective amount) of either a given substance within a clinical or subject-derived sample, including the derivation of qualitative or quantitative concentration levels of such substances, or otherwise evaluating the values or categorization of a subject's non-analyte clinical parameters.
[00076] "Negative predictive value" or "NPV" is calculated by TN/(TN + FN) or the true negative fraction of all negative test results. It also is inherently impacted by the prevalence of the disease and pre-test probability of the population intended to be tested.
[00077] See, e.g., O'Marcaigh AS, Jacobson RM, "Estimating The Predictive Value Of A Diagnostic Test, How To Prevent Misleading Or Confusing Results," Clin. Ped. 1993, 32(8): 485-491, which discusses specificity, sensitivity, and positive and negative predictive values of a test, e.g., a clinical diagnostic test. Often, for binary disease state classification approaches using a continuous diagnostic test measurement, the sensitivity and specificity is summarized by Receiver Operating Characteristics (ROC) curves according to Pepe et al, "Limitations of the Odds Ratio in Gauging the Performance of a Diagnostic, Prognostic, or Screening Marker," Am. J. Epidemiol 2004, 159 (9): 882-890, and summarized by the Area Under the Curve (AUC) or c-statistic, an indicator that allows representation of the sensitivity and specificity of a test, assay, or method over the entire range of test (or assay) cut points with just a single value. See also, e.g., Shultz, "Clinical Interpretation Of Laboratory Procedures," chapter 14 in Teitz, Fundamentals of Clinical Chemistry, Burtis and Ashwood (eds.), 4th edition 1996, W.B. Saunders Company, pages 192-199; and Zweig et al., "ROC Curve Analysis: An Example Showing The Relationships Among Serum Lipid And
Apolipoprotein Concentrations In Identifying Subjects With Coronory Artery Disease," Clin. Chem., 1992, 38(8): 1425-1428. An alternative approach using likelihood functions, odds ratios, information theory, predictive values, calibration (including goodness-of-fit), and reclassification measurements is summarized according to Cook, "Use and Misuse of the Receiver Operating Characteristic Curve in Risk Prediction," Circulation 2007, 115: 928-935.
[00078] Finally, hazard ratios and absolute and relative risk ratios within subject cohorts defined by a test are a further measurement of clinical accuracy and utility. Multiple methods are frequently used to defining abnormal or disease values, including reference limits, discrimination limits, and risk thresholds.
[00079] "Analytical accuracy" refers to the reproducibility and predictability of the measurement process itself, and may be summarized in such measurements as coefficients of variation, and tests of concordance and calibration of the same samples or controls with different times, users, equipment and/or reagents. These and other considerations in evaluating new biomarkers are also summarized in Vasan, 2006.
[00080] "Performance" is a term that relates to the overall usefulness and quality of a diagnostic or prognostic test, including, among others, clinical and analytical accuracy, other analytical and process characteristics, such as use characteristics (e.g., stability, ease of use), health economic value, and relative costs of components of the test. Any of these factors may be the source of superior performance and thus usefulness of the test, and may be measured by appropriate "performance metrics," such as AUC, time to result, shelf life, etc. as relevant.
[00081] "Positive predictive value" or "PPV" is calculated by TP/(TP+FP) or the true positive fraction of all positive test results. It is inherently impacted by the prevalence of the disease and pre-test probability of the population intended to be tested.
[00082] "Risk" in the context of the present invention, relates to the probability that an event will occur over a specific time period, as in the responsiveness to treatmnet, and can can mean a subject's "absolute" risk or "relative" risk. Absolute risk can be measured with reference to either actual observation post-measurement for the relevant time cohort, or with reference to index values developed from statistically valid historical cohorts that have been followed for the relevant time period. Relative risk refers to the ratio of absolute risks of a subject compared either to the absolute risks of low risk cohorts or an average population risk, which can vary by how clinical risk factors are assessed. Odds ratios, the proportion of positive events to negative events for a given test result, are also commonly used (odds are according to the formula p/(l-p) where p is the probability of event and (1- p) is the probability of no event) to no-conversion.
[00083] "Risk evaluation," or "evaluation of risk" in the context of the present invention encompasses making a prediction of the probability, odds, or likelihood that an event or disease state may occur, the rate of occurrence of the event or conversion from one disease state. Risk evaluation can also comprise prediction of future clinical parameters, traditional laboratory risk factor values, or other indices of cancer, either in absolute or relative terms in reference to a previously measured population. The methods of the present invention may be used to make continuous or categorical measurements of the responsiveness to treatment thus diagnosing and defining the risk spectrum of a category of subjects defined as being at responders or non-responders. In the categorical scenario, the invention can be used to discriminate between normal and other subject cohorts at higher risk for responding. Such differing use may require different DNARMARKER combinations and individualized panels, mathematical algorithms, and/or cut-off points, but be subject to the same aforementioned measurements of accuracy and performance for the respective intended use.
[00084] A "sample" in the context of the present invention is a biological sample isolated from a subject and can include, by way of example and not limitation, tissue biopies, whole blood, serum, plasma, blood cells, endothelial cells, lymphatic fluid, ascites fluid, interstitital fluid (also known as "extracellular fluid" and encompasses the fluid found in spaces between cells, including, inter alia, gingival crevicular fluid), bone marrow, cerebrospinal fluid (CSF), saliva, mucous, sputum, sweat, urine, or any other secretion, excretion, or other bodily fluids.
[00085] "Sensitivity" is calculated by TP/(TP+FN) or the true positive fraction of disease subjects.
[00086] "Specificity" is calculated by TN/(TN+FP) or the true negative fraction of non- disease or normal subjects. [00087] By "statistically significant", it is meant that the alteration is greater than what might be expected to happen by chance alone (which could be a "false positive"). Statistical significance can be determined by any method known in the art. Commonly used measures of significance include the p-value, which presents the probability of obtaining a result at least as extreme as a given data point, assuming the data point was the result of chance alone. A result is often considered highly significant at a p-value of 0.05 or less.
[00088] A "subject" in the context of the present invention is preferably a mammal. The mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples. Mammals other than humans can be advantageously used as subjects that represent animal models of cancer. A subject can be male or female.
[00089] "TN" is true negative, which for a disease state test means classifying a non- disease or normal subject correctly.
[00090] "TP" is true positive, which for a disease state test means correctly classifying a disease subject.
[00091] DNA REPAIR AND DNA DAMAGE RESPONSE MARKERS
[00092] Patients have varying degrees of responsiveness to therapy and methods are needed to distinguish the capability of the treatment in a dynamic manner. Identification of changes (e.g., active, hyperactive, repressed, downmodulated, or inactive) to the cellular DNA repair pathways are useful in monitoring and predicting the response to a therapeutic compound. Accordingly, included in the invention are biomarkers associated with DNA repair and DNA damage response. The invention features methods for identifying subjects who either are or are pre-disposed to developing resistance or are sensitive to a therapeutic compound, e.g., a chemotherapeutic drug by detection of the biomarkers disclosed herein. These biomarkers are also useful for monitoring subjects undergoing treatments and therapies for cancer and cell proliferative disorders, and for selecting therapies and treatments that would be efficacious in subjects having cancer and cell proliferative disorders.
[00093] The term "biomarker" in the context of the present invention encompasses, without limitation, proteins, nucleic acids, polymorphisms of proteins and nucleic acids, elements, metabolites, and other analytes. Biomarkers can also include mutated proteins or mutated nucleic acids. The term "analyte" as used herein can mean any substance to be measured and can encompass electrolytes and elements, such as calcium.
[00094] Proteins, nucleic acids, polymorphisms, and metabolites whose levels are changed in subjects who have resistance or sensitivity to therapeutic compound, or are predisposed to developing resistance or sensitivity to therapeutic compound are summarized in Table 1 and are collectively referred to herein as, inter alia, "DNA Repair and DNA Damage Response proteins or DNARMARKER". Table 2 summarizes DNARMARKERS associated with Ovarian Cancer. Table 3 summarizes other biomarkers useful in the methods of the invention
[00095] Expression of the DNARMARKERS is determined at the protein or nucleic acid level using any method known in the art. For example, at the nucleic acid level Northern hybridization analysis using probes which specifically recognize one or more of these sequences can be used to determine gene expression. Alternatively, expression is measured using reverse-transcription-based PCR assays, e.g., using primers specific for the
differentially expressed sequence of genes. Expression is also determined at the protein level, i.e. , by measuring the levels of peptides encoded by the gene products described herein, or activities thereof. Such methods are well known in the art and include, e.g. ,
immunoassays based on antibodies to proteins encoded by the genes, aptamers or molecular imprints.. Any biological material can be used for the detection/quantification of the protein or its activity. Alternatively, a suitable method can be selected to determine the activity of proteins encoded by the marker genes according to the activity of each protein analyzed.
[00096] The DNARMARKER proteins are detected in any suitable manner, but is typically detected by contacting a sample from the patient with an antibody which binds the DNARMARKER protein and then detecting the presence or absence of a reaction product. The antibody may be monoclonal, polyclonal, chimeric, or a fragment of the foregoing, as discussed in detail above, and the step of detecting the reaction product may be carried out with any suitable immunoassay. The sample from the subject is typically a biological fluid as described above, and may be the same sample of biological fluid used to conduct the method described above. The sample may also be in the form of a tissue specimen from a patient where the specimen is suitable for immunohistochemistry in a variety of formats such as paraffin-embedded tissue, frozen sections of tissue, and freshly isolated tissue. The immunodetection methods are antibody-based but there are numerous additional techniques that allow for highly sensitive determinations of binding to an antibody in the context of a tissue. Those skilled in the art will be familiar with various immunohistochemistry strategies.
[00097] Immunoassays carried out in accordance with the present invention may be homogeneous assays or heterogeneous assays. In a homogeneous assay the immunological reaction usually involves the specific antibody (e.g., anti- DNARMARKER protein antibody), a labeled analyte, and the sample of interest. The signal arising from the label is modified, directly or indirectly, upon the binding of the antibody to the labeled analyte. Both the immunological reaction and detection of the extent thereof are carried out in a homogeneous solution. Immunochemical labels which may be employed include free radicals, radioisotopes, fluorescent dyes, enzymes, bacteriophages, or coenzymes.
[00098] In a heterogeneous assay approach, the reagents are usually the sample, the antibody, and means for producing a detectable signal. Samples as described above may be used. The antibody is generally immobilized on a support, such as a bead, plate or slide, and contacted with the specimen suspected of containing the antigen in a liquid phase. The support is then separated from the liquid phase and either the support phase or the liquid phase is examined for a detectable signal employing means for producing such signal. The signal is related to the presence of the analyte in the sample. Means for producing a detectable signal include the use of radioactive labels, fluorescent labels, or enzyme labels. For example, if the antigen to be detected contains a second binding site, an antibody which binds to that site can be conjugated to a detectable group and added to the liquid phase reaction solution before the separation step. The presence of the detectable group on the solid support indicates the presence of the antigen in the test sample. Examples of suitable immunoassays are radioimmunoassays, immunofluorescence methods, chemilumenescence methods, electrochemilumenescence or enzyme-linked immunoassays.
[00099] Those skilled in the art will be familiar with numerous specific immunoassay formats and variations thereof which may be useful for carrying out the method disclosed herein. See generally E. Maggio, Enzyme-Immunoassay, (1980) (CRC Press, Inc., Boca Raton, Fla.); see also U.S. Pat. No. 4,727,022 to Skold et al. titled "Methods for Modulating Ligand-Receptor Interactions and their Application," U.S. Pat. No. 4,659,678 to Forrest et al. titled "Immunoassay of Antigens," U.S. Pat. No. 4,376,110 to David et al., titled
"Immunometric Assays Using Monoclonal Antibodies," U.S. Pat. No. 4,275,149 to Litman et al., titled "Macromolecular Environment Control in Specific Receptor Assays," U.S. Pat. No. 4,233,402 to Maggio et al., titled "Reagents and Method Employing Channeling," and U.S. Pat. No. 4,230,767 to Boguslaski et al., titled "Heterogenous Specific Binding Assay Employing a Coenzyme as Label. "
[000100] Antibodies are conjugated to a solid support suitable for a diagnostic assay (e.g., beads, plates, slides or wells formed from materials such as latex or polystyrene) in accordance with known techniques, such as passive binding. Antibodies as described herein may likewise be conjugated to detectable groups such as radiolabels (e.g., 35 S, 125 I, 131 I), enzyme labels (e.g., horseradish peroxidase, alkaline phosphatase), and fluorescent labels (e.g., fluorescein) in accordance with known techniques.
[000101] The skilled artisan can routinely make antibodies, nucleic acid probes, e.g., oligonucleotides, aptamers, siRNAs against any of the DNARMARKERS in Table 1.
[000102] The invention also includes a DNARMARKER-detection reagent, e.g., nucleic acids that specifically identify one or more DNARMARKER nucleic acids by having homologous nucleic acid sequences, such as oligonucleotide sequences, complementary to a portion of the DNARMARKER nucleic acids or antibodies to proteins encoded by the DNARMARKER nucleic acids packaged together in the form of a kit. The oligonucleotides are fragments of the DNARMARKER genes. For example the olignucleotides are 200, 150, 100, 50, 25, 10 or less nucleotides in length. The kit may contain in separate containers a nucleic acid or antibody (either already bound to a solid matrix or packaged separately with reagents for binding them to the matrix) , control formulations (positive and/or negative), and/or a detectable label. Instructions (e.g., written, tape, VCR, CD-ROM, etc.) for carrying out the assay may be included in the kit. The assay may for example be in the form of a Northern hybridization or a sandwich ELISA as known in the art.
[000103] For example, DNARMARKER detection reagent, is immobilized on a solid matrix such as a porous strip to form at least one DNARMARKER detection site. The measurement or detection region of the porous strip may include a plurality of sites containing a nucleic acid. A test strip may also contain sites for negative and/or positive controls. Alternatively, control sites are located on a separate strip from the test strip.
Optionally, the different detection sites may contain different amounts of immobilized nucleic acids, i.e., a higher amount in the first detection site and lesser amounts in subsequent sites. Upon the addition of test sample, the number of sites displaying a detectable signal provides a quantitative indication of the amount of DNARMARKER present in the sample. The detection sites may be configured in any suitably detectable shape and are typically in the shape of a bar or dot spanning the width of a test strip.
[000104] Alternatively, the kit contains a nucleic acid substrate array comprising one or more nucleic acid sequences. The nucleic acids on the array specifically identify one or more nucleic acid sequences represented by DNARMARKER 1-259. In various embodiments, the expression of 2, 3,4, 5, 6, 7,8, 9, 10, 15, 20, 25, 40 or 50 or more of the sequences represented by DNARMARKER 1-259. are identified by virtue of binding to the array. The substrate array can be on, e.g. , a solid substrate, e.g. , a "chip" as described in U.S. Patent No.5,744,305. Alternatively the substrate array can be a solution array, e.g., Luminex, Cyvera, Vitra and Quantum Dots' Mosaic.
Preferably, the kit contains antibodies for the detection of DNARMARKERS.
[000105] THERAPEUTIC METHODS
[000106] Responsiveness (e.g., resistance or sensitivity) of a cell to an agent is determined by measuring an effective amount of a DNARMARKER proteins, nucleic acids,
polymorphisms, metabolites, and other analytes (which may be two or more) in a test sample (e.g., a subject derived sample), and comparing the effective amounts to reference or index values, often utilizing mathematical algorithms or formula in order to combine information from results of multiple individual DNARMARKERS and from non-analyte clinical parameters into a single measurement or index. The cell is for example a cancer cell.
Optionally, the cancer is a ovarian cancer. The DNARMARKER includes for example MLH1, pMAPKAPK2, PAR, PARP1, XPF, pH2A2and FAND2. Additionally any other biomarker listed on Tables 1 ,2 or 3 are measured. In another embodimentthe
DNARMARKER is XPF, FANCD2, pMK2, PAR, MLH1, PARP1, BRCA1, RAD51, POL H, ATM, or ERCC1. Optionally Ki67 is measured.
[000107] By resistance is meant that the failure of a cell to respond to an agent. For example, resistance to a chemotherapeutic drug means the cell is not damaged or killed by the drug. By sensitivity is meant that the cell responds to an agent. For example, sensitivity to a chemotherapeutic drug means the cell is damaged or killed by the drug.
[000108] For example, responsiveness of a cell to a chemotherapeutic agent identified by identifying a decrease in expression or activity of one or more Ovarian DNARMARKERS. The presence of a deficiency in DNARMARKER indicates that the cell is sensitive to a chemotherapeutic agent. Whereas, the absence of a deficiency indicates that the cell is resistant to a chemotherapeutic agent.
[000109] The methods are useful to treat, alleviate the symptoms of, monitor the progression of or delay the onset of cancer in a subject.
[000110] Expression of an effective amount of DNARMARKER proteins, nucleic acids or metabolites also allows for the course of treatment of cancer or a cell proliferative disorder to be monitored. In this method, a biological sample is provided from a subject undergoing treatment, e.g., chemotherapeutic treatment, for cancer or a cell proliferative disorder. If desired, biological samples are obtained from the subject at various time points before, during, or after treatment. Expression of an effective amount of DNARMARKER proteins, nucleic acids or metabolites is then determined and compared to a reference, e.g. a control individual or population whose cancer or a cell proliferative disorder state is known or an index value. The reference sample or index value may be taken or derived from one or more individuals who have been exposed to the treatment. Alternatively, the reference sample or index value may be taken or derived from one or more individuals who have not been exposed to the treatment. For example, samples may be collected from subjects who have received initial treatment for cancer or a cell proliferative disorder and subsequent treatment for diabetes to monitor the progress of the treatment.
[000111] The amount of the DNARMARKER protein, nucleic acid, polymorphism, metabolite, or other analyte can be measured in a test sample and compared to the "normal control level," utilizing techniques such as reference limits, discrimination limits, or risk defining thresholds to define cutoff points and abnormal values. Such normal control level and cutoff points may vary based on whether a DNARMARKER is used alone or in a formula combining with other DNARMARKERS into an index. Alternatively, the normal control level can be a database of DNARMARKER patterns from previously tested subjects who responded to chemotherapy over a clinically relevant time horizon.
[000112] The present invention may be used to make continuous or categorical measurements of the response to chemotherapy or cancer survival, thus diagnosing and defining the risk spectrum of a category of subjects defined as at risk for not responding to chemotherapy. In the categorical scenario, the methods of the present invention can be used to discriminate between treatment responsive and treatment non-responsive subject cohorts. In other embodiments, the present invention may be used so as to discriminate those who have an improved survival potential. Such differing use may require different
DNARMARKER combinations in individual panel, mathematical algorithm, and/or cut-off points, but be subject to the same aforementioned measurements of accuracy and other performance metrics relevant for the intended use.
[000113] Identifying the subject who will be responsive to therapy enables the selection and initiation of various therapeutic interventions or treatment regimens in order increase the individuals survival potential. Levels of an effective amount of DNARMARKER proteins, nucleic acids, polymorphisms, metabolites, or other analytes also allows for the course of treatment of a metastatic disease or metastatic event to be monitored. In this method, a biological sample can be provided from a subject undergoing treatment regimens, e.g., drug treatments, for cancer. If desired, biological samples are obtained from the subject at various time points before, during, or after treatment.
[000114] Levels of an effective amount of DNARMARKER proteins, nucleic acids, polymorphisms, metabolites, or other analytes can then be determined and compared to a reference value, e.g. a control subject or population whose therapeutic responsiveness is known or an index value or baseline value. The reference sample or index value or baseline value may be taken or derived from one or more subjects who have been exposed to the treatment, or may be taken or derived from one or more subjects who are at low risk of surviving the cancer, or may be taken or derived from subjects who have shown
improvements in as a result of exposure to treatment. Alternatively, the reference sample or index value or baseline value may be taken or derived from one or more subjects who have not been exposed to the treatment. For example, samples may be collected from subjects who have received initial treatment for cancer or and subsequent treatment for cancer or a metastatic event to monitor the progress of the treatment. A reference value can also comprise a value derived from risk prediction algorithms or computed indices from population studies such as those disclosed herein.
[000115] The DNARMARKERS of the present invention can thus be used to generate a "reference DNARMARKER profile" of those subjects who would or would not be expected respond to cancer treatment. The DNARMARKERS disclosed herein can also be used to generate a "subject DNARMARKER profile" taken from subjects who are responsive cancer treatment. The subject DNARMARKER profiles can be compared to a reference
DNARMARKER profile to diagnose or identify subjects at risk for developing resistance to chemotherapy, to monitor the progression of disease, as well as the rate of progression of disease, and to monitor the effectiveness of treatment modalities. The reference and subject DNARMARKER profiles of the present invention can be contained in a machine-readable medium, such as but not limited to, analog tapes like those readable by a VCR, CD-ROM, DVD-ROM, USB flash media, among others. Such machine-readable media can also contain additional test results, such as, without limitation, measurements of clinical parameters and traditional laboratory risk factors. Alternatively or additionally, the machine-readable media can also comprise subject information such as medical history and any relevant family history. The machine-readable media can also contain information relating to other disease- risk algorithms and computed indices such as those described herein. [000116] Differences in the genetic makeup of subjects can result in differences in their relative abilities to metabolize various drugs, which may modulate the symptoms or risk factors of cancer or metastatic events. Subjects that have cancer, or at risk for developing cancer or a metastatic event can vary in age, ethnicity, and other parameters. Accordingly, use of the DNARMARKERS disclosed herein, both alone and together in combination with known genetic factors for drug metabolism, allow for a pre-determined level of predictability that a putative therapeutic or prophylactic to be tested in a selected subject will be suitable for treating or preventing cancer in the subject.
[000117] To identify therapeutic that is appropriate for a specific subject a the expression of one or more of DNARMARKER proteins, nucleic acids or metabolites is in a test sample form the subject is determined .
[000118] The pattern of DNARMARKER expression in the test sample is measured and compared to a reference profile, e.g., a therapeutic compound reference expression profile. Comparison can be performed on test and reference samples measured concurrently or at temporally distinct times. An example of the latter is the use of compiled expression information, e.g. , a sequence database, which assembles information about expression levels of DNARMARKERS.
[000119] If the reference sample, e.g., a control sample is from cells that are sensitive to a therapeutic compound then a similarity in the amount of the DNARMARKER proteins in the test sample and the reference sample indicates that treatment with that therapeutic compound will be efficacious. However, a change in the amount of the DNARMARKER in the test sample and the reference sample indicates treatment with that compound will result in a less favorable clinical outcome or prognosis. In contrast, if the reference sample, e.g., a control sample is from cells that are resistant to a therapeutic compound then a similarity in the amount of the DNARMARKER proteins in the test sample and the reference sample indicates that the treatment with that compound will result in a less favorable clinical outcome or prognosis. However, a change in the amount of the DNARMARKER in the test sample and the reference sample indicates that treatment with that therapeutic compound will be efficacious.
[000120] By "efficacious" is meant that the treatment leads to a decrease in the amount of a DNARMARKER protein, or a decrease in size, prevalence, or metastatic potential of cancer in a subject. When treatment is applied prophylactically, "efficacious" means that the treatment retards or prevents cancer or a cell proliferative disorder from forming. Assessment of cancer and cell proliferative disorders is made using standard clinical protocols.
[000121] The subject is preferably a mammal. The mammal is, e.g. , a human, non- human primate, mouse, rat, dog, cat, horse, or cow. The subject has been previously diagnosed as having cancer or a cell proliferative disorder, and possibly has already undergone treatment for the cancer or a cell proliferative disorder.
[000122] The subject is suffering from or at risk of developing ovarian cancer. Subjects suffering from or at risk of developing ovarian cancer are identified by methods known in the art.
[000123] Alternatively, the deficiency is determined by measuring the expression (e.g. increase or decrease relative to a control), detecting a sequence variation or posttranslational modification of one or more DNARMARKERS described herein.
[000124] Posttranslational modification include for example, phosphorylation,
ubiquitination, sumo-ylation, acetylation, alkylation, methylation, glycylation, glycosylation, isoprenylation, lipoylation, phosphopantetheinylation, sulfation, selenation and C-terminal amidation. For example, a deficiency in the Homologous Recombination/FA pathway is determined by detecting the monoubiquitination of FANCD2. Similarly, responsiveness of cancer cell to a MAP2KAP2 inhibitor is determined by detecting phosphorylation of a MAP2KAP2 protein. Phosphorylation indicates the cell is sensitive to a MAP2KAP2 inhibitor. In contrast the absence of phosphorylation indicates the cell is resistant to a MAP2KAP2 inhibitor.
[000125] Sequence variations such as mutations and polymorphisms may include a deletion, insertion or substitution of one or more nucleotides, relative to the wild- type nucleotide sequence. The one or more variations may be in a coding or non-coding region of the nucleic acid sequence and, may reduce or abolish the expression or function of the DNA repair pathway component polypeptide. In other words, the variant nucleic acid may encode a variant polypeptide which has reduced or abolished activity or may encode a wild-type polypeptide which has little or no expression within the cell, for example through the altered activity of a regulatory element. A variant nucleic acid may have one, two, three, four or more mutations or polymorphisms relative to the wild-type sequence.
[000126] The presence of one or more variations in a nucleic acid which encodes a component of a DNA repair pathway, is determined for example by detecting, in one or more cells of a test sample, the presence of an encoding nucleic acid sequence which comprises the one or more mutations or polymorphisms, or by detecting the presence of the variant component polypeptide which is encoded by the nucleic acid sequence.
[000127] Various methods are available for determining the presence or absence in a sample obtained from an individual of a particular nucleic acid sequence, for example a nucleic acid sequence which has a mutation or polymorphism that reduces or abrogates the expression or activity of a DNA repair pathway component. Furthermore, having sequenced nucleic acid of an individual or sample, the sequence information can be retained and subsequently searched without recourse to the original nucleic acid itself. Thus, for example, scanning a database of sequence information using sequence analysis software may identify a sequence alteration or mutation.
[000128] Methods according to some aspects of the present invention may comprise determining the binding of an oligonucleotide probe to nucleic acid obtained from the sample, for example, genomic DNA, RNA or cDNA. The probe may comprise a nucleotide sequence which binds specifically to a nucleic acid sequence which contains one or more mutations or polymorphisms and does not bind specifically to the nucleic acid sequence which does not contain the one or more mutations or polymorphisms, or vice versa. The oligonucleotide probe may comprise a label and binding of the probe may be determined by detecting the presence of the label.
[000129] A method may include hybridization of one or more (e.g. two) oligonucleotide probes or primers to target nucleic acid. Where the nucleic acid is double- stranded DNA, hybridization will generally be preceded by denaturation to produce single-stranded DNA. The hybridization may be as part of a PCR procedure, or as part of a probing procedure not involving PCR. An example procedure would be a combination of PCR and low stringency hybridization.
[000130] Binding of a probe to target nucleic acid (e.g. DNA) may be measured using any of a variety of techniques at the disposal of those skilled in the art. For instance, probes may be radioactively, fluorescently or enzymatically labeled. Other methods not employing labeling of probe include examination of restriction fragment length polymorphisms, amplification using PCR, RN'ase cleavage and allele specific oligonucleotide probing.
Probing may employ the standard Southern blotting technique. For instance, DNA may be extracted from cells and digested with different restriction enzymes. Restriction fragments may then be separated by electrophoresis on an agarose gel, before denaturation and transfer to a nitrocellulose filter. Labeled probe may be hybridized to the DNA fragments on the filter and binding determined.
[000131] Those skilled in the art are well able to employ suitable conditions of the desired stringency for selective hybridization, taking into account factors such as oligonucleotide length and base composition, temperature and so on. Suitable selective hybridization conditions for oligonucleotides of 17 to 30 bases include hybridization overnig ht at 42.°C in 6x SSC and washing in 6.x SSC at a series of increasing temperatures from 42°C to 65°C. Other suitable conditions and protocols are described in Molecular Cloning: a Laboratory Manual: 3rd edition, Sambrook & Russell (2001) Cold Spring Harbor Laboratory Press NY and Current Protocols in Molecular Biology, Ausubel et al. eds. John Wiley & Sons (1992).
[000132] Nucleic acid, which may be genomic DNA, RNA or cDNA, or an amplified region thereof, may be sequenced to identify or determine the presence of polymorphism or mutation therein. A polymorphism or mutation may be identified by comparing the sequence obtained with the database sequence of the component, as set out above. In particular, the presence of one or more polymorphisms or mutations that cause abrogation or loss of function of the polypeptide component, and thus the DNA repair pathway as a whole, may be determined.
[000133] Sequencing may be performed using any one of a range of standard techniques. Sequencing of an amplified product may, for example, involve precipitation with
isopropanol, resuspension and sequencing using a TaqFS+ Dye terminator sequencing kit. Extension products may be electrophoresed on an ABI 377 DNA sequencer and data analyzed using Sequence Navigator software.
[000134] A specific amplification reaction such as PCR using one or more pairs of primers may conveniently be employed to amplify the region of interest within the nucleic acid sequence, for example, the portion of the sequence suspected of containing mutations or polymorphisms. The amplified nucleic acid may then be sequenced as above, and/or tested in any other way to determine the presence or absence of a mutation or polymorphism which reduces or abrogates the expression or activity of the DNA repair pathway component. Suitable amplification reactions include the polymerase chain reaction (PCR) (reviewed for instance in "PCR protocols; A Guide to Methods and Applications", Eds. Innis et al, 1990, Academic Press, New York, Mullis et al, Cold Spring Harbor Symp. Quant. Biol., 51:263, (1987), Ehrlich (ed), PCR technology, Stockton Press, NY, 1989, and Ehrlich et al, Science, 252: 1643-1650, (1991)). [000135] Mutations and polymorphisms associated with cancer may also be detected at the protein level by detecting the presence of a variant (i.e. a mutant or allelic variant) polypeptide.
[000136] A method of identifying a cancer cell in a sample from an individual as deficient in DNA repair may include contacting a sample with a specific binding member directed against a variant (e.g. a mutant) polypeptide component of the pathway, and determining binding of the specific binding member to the sample. Binding of the specific binding member to the sample may be indicative of the presence of the variant polypeptide component of the DNA repair pathway in a cell within the sample. Preferred specific binding molecules for use in aspects of the present invention include antibodies and fragments or derivatives thereof ("antibody molecules").
[000137] The reactivities of a binding member such as an antibody on normal and test samples may be determined by any appropriate means. Tagging with individual reporter molecules is one possibility. The reporter molecules may directly or indirectly generate detectable, and preferably measurable, signals. The linkage of reporter molecules may be directly or indirectly, covalently, e.g. via a peptide bond or non-covalently. Linkage via a peptide bond may be as a result of recombinant expression of a gene fusion encoding binding molecule (e.g. antibody) and reporter molecule.
[000138] PERFORMANCE AND ACCURACY MEASURES OF THE INVENTION
[000139] The performance and thus absolute and relative clinical usefulness of the invention may be assessed in multiple ways as noted above. Amongst the various assessments of performance, the invention is intended to provide accuracy in clinical diagnosis and prognosis. The accuracy of a diagnostic or prognostic test, assay, or method concerns the ability of the test, assay, or method to distinguish between subjects responsive to chemotherapeutic treatment and those that are not, is based on whether the subjects have an "effective amount" or a "significant alteration" in the levels of a DNARMARKER. By "effective amount" or "significant alteration," it is meant that the measurement of an appropriate number of DNARMARKERS (which may be one or more) is different than the predetermined cut-off point (or threshold value) for that DNARMARKER(S) and therefore indicates that the subject responsiveness to therapy for which the DNARMARKER(S) is a determinant. The difference in the level of DNARMARKER between normal and abnormal is preferably statistically significant. As noted below, and without any limitation of the invention, achieving statistical significance, and thus the preferred analytical and clinical accuracy, generally but not always requires that combinations of several DNARMARKERS be used together in panels and combined with mathematical algorithms in order to achieve a statistically significant DNARMARKER index.
[000140] In the categorical diagnosis of a disease state, changing the cut point or threshold value of a test (or assay) usually changes the sensitivity and specificity, but in a qualitatively inverse relationship. Therefore, in assessing the accuracy and usefulness of a proposed medical test, assay, or method for assessing a subject's condition, one should always take both sensitivity and specificity into account and be mindful of what the cut point is at which the sensitivity and specificity are being reported because sensitivity and specificity may vary significantly over the range of cut points. Use of statistics such as AUC, encompassing all potential cut point values, is preferred for most categorical risk measures using the invention, while for continuous risk measures, statistics of goodness-of-fit and calibration to observed results or other gold standards, are preferred.
[000141] Using such statistics, an "acceptable degree of diagnostic accuracy", is herein defined as a test or assay (such as the test of the invention for determining the clinically significant presence of DNARMARKERS, which thereby indicates the presence of cancer and/or a risk of having a metastatic event) in which the AUC (area under the ROC curve for the test or assay) is at least 0.60, desirably at least 0.65, more desirably at least 0.70, preferably at least 0.75, more preferably at least 0.80, and most preferably at least 0.85.
[000142] By a "very high degree of diagnostic accuracy", it is meant a test or assay in which the AUC (area under the ROC curve for the test or assay) is at least 0.80, desirably at least 0.85, more desirably at least 0.875, preferably at least 0.90, more preferably at least 0.925, and most preferably at least 0.95.
[000143] The predictive value of any test depends on the sensitivity and specificity of the test, and on the prevalence of the condition in the population being tested. This notion, based on Bayes' theorem, provides that the greater the likelihood that the condition being screened for is present in an individual or in the population (pre-test probability), the greater the validity of a positive test and the greater the likelihood that the result is a true positive. Thus, the problem with using a test in any population where there is a low likelihood of the condition being present is that a positive result has limited value (i.e., more likely to be a false positive). Similarly, in populations at very high risk, a negative test result is more likely to be a false negative. [000144] As a result, ROC and AUC can be misleading as to the clinical utility of a test in low disease prevalence tested populations (defined as those with less than 1% rate of occurrences (incidence) per annum, or less than 10% cumulative prevalence over a specified time horizon). Alternatively, absolute risk and relative risk ratios as defined elsewhere in this disclosure can be employed to determine the degree of clinical utility. Populations of subjects to be tested can also be categorized into quartiles by the test's measurement values, where the top quartile (25% of the population) comprises the group of subjects with the highest relative risk for therapeutic unresponsiveness, and the bottom quartile comprising the group of subjects having the lowest relative risk for therapeutic unresponsiveness Generally, values derived from tests or assays having over 2.5 times the relative risk from top to bottom quartile in a low prevalence population are considered to have a "high degree of diagnostic accuracy," and those with five to seven times the relative risk for each quartile are considered to have a "very high degree of diagnostic accuracy." Nonetheless, values derived from tests or assays having only 1.2 to 2.5 times the relative risk for each quartile remain clinically useful are widely used as risk factors for a disease; such is the case with total cholesterol and for many inflammatory biomarkers with respect to their prediction of future events. Often such lower diagnostic accuracy tests must be combined with additional parameters in order to derive meaningful clinical thresholds for therapeutic intervention, as is done with the aforementioned global risk assessment indices.
[000145] A health economic utility function is an yet another means of measuring the performance and clinical value of a given test, consisting of weighting the potential categorical test outcomes based on actual measures of clinical and economic value for each. Health economic performance is closely related to accuracy, as a health economic utility function specifically assigns an economic value for the benefits of correct classification and the costs of misclassification of tested subjects. As a performance measure, it is not unusual to require a test to achieve a level of performance which results in an increase in health economic value per test (prior to testing costs) in excess of the target price of the test.
[000146] In general, alternative methods of determining diagnostic accuracy are commonly used for continuous measures, when a disease category or risk category has not yet been clearly defined by the relevant medical societies and practice of medicine, where thresholds for therapeutic use are not yet established, or where there is no existing gold standard for diagnosis of the pre-disease. For continuous measures of risk, measures of diagnostic accuracy for a calculated index are typically based on curve fit and calibration between the predicted continuous value and the actual observed values (or a historical index calculated value) and utilize measures such as R squared, Hosmer- Lemeshow P-value statistics and confidence intervals. It is not unusual for predicted values using such algorithms to be reported including a confidence interval (usually 90% or 95% CI) based on a historical observed cohort's predictions, as in the test for risk of future ovarian cancer recurrence commercialized by Genomic Health, Inc. (Redwood City, California).
[000147] In general, by defining the degree of diagnostic accuracy, i.e., cut points on a ROC curve, defining an acceptable AUC value, and determining the acceptable ranges in relative concentration of what constitutes an effective amount of the DNARMARKERS of the invention allows for one of skill in the art to use the DNARMARKERS to identify, diagnose, or prognose subjects with a pre-determined level of predictability and performance.
[000148] CONSTRUCTION OF DNARMARKER PANELS
[000149] Groupings of DNARMARKERS can be included in "panels." A "panel" within the context of the present invention means a group of biomarkers (whether they are
DNARMARKERS, clinical parameters, or traditional laboratory risk factors) that includes more than one DNARMARKER. A panel can also comprise additional biomarkers, e.g., clinical parameters, traditional laboratory risk factors, known to be present or associated with responsiveness to chemotherapeutic treatment, in combination with a selected group of the DNARMARKERS listed in Table 1 or Table 2. Optionally, biomarkers listed in Table 3 are meausured
[000150] As noted above, many of the individual DNARMARKERS, clinical parameters, and traditional laboratory risk factors listed, when used alone and not as a member of a multi- biomarker panel of DNARMARKERS, have little or no clinical use in reliably distinguishing individuals that are responsive to therapeutic treatment and those that are not and thus cannot reliably be used alone in classifying any subject between those two states. Even where there are statistically significant differences in their mean measurements in each of these populations, as commonly occurs in studies which are sufficiently powered, such biomarkers may remain limited in their applicability to an individual subject, and contribute little to diagnostic or prognostic predictions for that subject. A common measure of statistical significance is the p- value, which indicates the probability that an observation has arisen by chance alone; preferably, such p-values are 0.05 or less, representing a 5% or less chance that the observation of interest arose by chance. Such p-values depend significantly on the power of the study performed. [000151] Despite this individual DNARMARKER performance, and the general performance of formulas combining only the traditional clinical parameters and few traditional laboratory risk factors, the present inventors have noted that certain specific combinations of two or more DNARMARKERS can also be used as multi-biomarker panels comprising combinations of DNARMARKERS that are known to be involved in one or more physiological or biological pathways, and that such information can be combined and made clinically useful through the use of various formulae, including statistical classification algorithms and others, combining and in many cases extending the performance
characteristics of the combination beyond that of the individual DNARMARKERS. These specific combinations show an acceptable level of diagnostic accuracy, and, when sufficient information from multiple DNARMARKERS is combined in a trained formula, often reliably achieve a high level of diagnostic accuracy transportable from one population to another.
[000152] The general concept of how two less specific or lower performing
DNARMARKERS are combined into novel and more useful combinations for the intended indications, is a key aspect of the invention. Multiple biomarkers can often yield better performance than the individual components when proper mathematical and clinical algorithms are used; this is often evident in both sensitivity and specificity, and results in a greater AUC. Secondly, there is often novel unperceived information in the existing biomarkers, as such was necessary in order to achieve through the new formula an improved level of sensitivity or specificity. This hidden information may hold true even for biomarkers which are generally regarded to have suboptimal clinical performance on their own. In fact, the suboptimal performance in terms of high false positive rates on a single biomarker measured alone may very well be an indicator that some important additional information is contained within the biomarker results - information which would not be elucidated absent the combination with a second biomarker and a mathematical formula.
[000153] Several statistical and modeling algorithms known in the art can be used to both assist in DNARMARKER selection choices and optimize the algorithms combining these choices. Statistical tools such as factor and cross-biomarker correlation/covariance analyses allow more rationale approaches to panel construction. Mathematical clustering and classification tree showing the Euclidean standardized distance between the
DNARMARKERS can be advantageously used. Pathway informed seeding of such statistical classification techniques also may be employed, as may rational approaches based on the selection of individual DNARMARKERS based on their participation across in particular pathways or physiological functions.
[000154] Ultimately, formula such as statistical classification algorithms can be directly used to both select DNARMARKERS and to generate and train the optimal formula necessary to combine the results from multiple DNARMARKERS into a single index. Often, techniques such as forward (from zero potential explanatory parameters) and backwards selection (from all available potential explanatory parameters) are used, and information criteria, such as AIC or BIC, are used to quantify the tradeoff between the performance and diagnostic accuracy of the panel and the number of DNARMARKERS used. The position of the individual DNARMARKER on a forward or backwards selected panel can be closely related to its provision of incremental information content for the algorithm, so the order of contribution is highly dependent on the other constituent DNARMARKERS in the panel.
[000155] CONSTRUCTION OF CLINICAL ALGORITHMS
[000156] Any formula may be used to combine DNARMARKER results into indices useful in the practice of the invention. As indicated above, and without limitation, such indices may indicate, among the various other indications, the probability, likelihood, absolute or relative chance of responding to chemotherapy. This may be for a specific time period or horizon, or for remaining lifetime risk, or simply be provided as an index relative to another reference subject population.
[000157] Although various preferred formula are described here, several other model and formula types beyond those mentioned herein and in the definitions above are well known to one skilled in the art. The actual model type or formula used may itself be selected from the field of potential models based on the performance and diagnostic accuracy characteristics of its results in a training population. The specifics of the formula itself may commonly be derived from DNARMARKER results in the relevant training population. Amongst other uses, such formula may be intended to map the feature space derived from one or more DNARMARKER inputs to a set of subject classes (e.g. useful in predicting class membership of subjects as normal, responders and non-responders), to derive an estimation of a probability function of risk using a Bayesian approach (e.g. the risk of cancer or a metastatic event), or to estimate the class-conditional probabilities, then use Bayes' rule to produce the class probability function as in the previous case.
[000158] Preferred formulas include the broad class of statistical classification algorithms, and in particular the use of discriminant analysis. The goal of discriminant analysis is to predict class membership from a previously identified set of features. In the case of linear discriminant analysis (LDA), the linear combination of features is identified that maximizes the separation among groups by some criteria. Features can be identified for LDA using an eigengene based approach with different thresholds (ELD A) or a stepping algorithm based on a multivariate analysis of variance (MANOVA). Forward, backward, and stepwise algorithms can be performed that minimize the probability of no separation based on the Hotelling-Lawley statistic.
[000159] Eigengene-based Linear Discriminant Analysis (ELD A) is a feature selection technique developed by Shen et al. (2006). The formula selects features (e.g. biomarkers) in a multivariate framework using a modified eigen analysis to identify features associated with the most important eigenvectors. "Important" is defined as those eigenvectors that explain the most variance in the differences among samples that are trying to be classified relative to some threshold.
[000160] A support vector machine (SVM) is a classification formula that attempts to find a hyperplane that separates two classes. This hyperplane contains support vectors, data points that are exactly the margin distance away from the hyperplane. In the likely event that no separating hyperplane exists in the current dimensions of the data, the dimensionality is expanded greatly by projecting the data into larger dimensions by taking non-linear functions of the original variables (Venables and Ripley, 2002). Although not required, filtering of features for SVM often improves prediction. Features (e.g., biomarkers) can be identified for a support vector machine using a non-parametric Kruskal-Wallis (KW) test to select the best univariate features. A random forest (RF, Breiman, 2001) or recursive partitioning (RPART, Breiman et al., 1984) can also be used separately or in combination to identify biomarker combinations that are most important. Both KW and RF require that a number of features be selected from the total. RPART creates a single classification tree using a subset of available biomarkers.
[000161] Other formula may be used in order to pre-process the results of individual DNARMARKER measurement into more valuable forms of information, prior to their presentation to the predictive formula. Most notably, normalization of biomarker results, using either common mathematical transformations such as logarithmic or logistic functions, as normal or other distribution positions, in reference to a population's mean values, etc. are all well known to those skilled in the art. Of particular interest are a set of normalizations based on Clinical Parameters such as age, gender, race, or sex, where specific formula are used solely on subjects within a class or continuously combining a Clinical Parameter as an input. In other cases, analyte-based biomarkers can be combined into calculated variableswhich are subsequently presented to a formula.
[000162] In addition to the individual parameter values of one subject potentially being normalized, an overall predictive formula for all subjects, or any known class of subjects, may itself be recalibrated or otherwise adjusted based on adjustment for a population's expected prevalence and mean biomarker parameter values, according to the technique outlined in D'Agostino et al, (2001) JAMA 286: 180-187, or other similar normalization and recalibration techniques. Such epidemiological adjustment statistics may be captured, confirmed, improved and updated continuously through a registry of past data presented to the model, which may be machine readable or otherwise, or occasionally through the retrospective query of stored samples or reference to historical studies of such parameters and statistics. Additional examples that may be the subject of formula recalibration or other adjustments include statistics used in studies by Pepe, M.S. et al, 2004 on the limitations of odds ratios; Cook, N.R., 2007 relating to ROC curves. Finally, the numeric result of a classifier formula itself may be transformed post-processing by its reference to an actual clinical population and study results and observed endpoints, in order to calibrate to absolute risk and provide confidence intervals for varying numeric results of the classifier or risk formula. An example of this is the presentation of absolute risk, and confidence intervals for that risk, derivied using an actual clinical study, chosen with reference to the output of the recurrence score formula in the Oncotype Dx product of Genomic Health, Inc. (Redwood City, CA). A further modification is to adjust for smaller sub-populations of the study based on the output of the classifier or risk formula and defined and selected by their Clinical Parameters, such as age or sex.
EXAMPLES
[000163] Example 1: Immunohistochemical Evaluation of Ovarian Cancer Patient Specimens
[000164] Tumor specimens from patients were prepared by FFPE practice and serial sections of the tumor were prepared onto microscope slides. DNA repair biomarkers are evaluated with antibodies directed against specific constituents of the DNA repair pathways. These proteins are either evaluated singly, or in a multiplexed fashion depending on the detection strategies employed. [000165] IHC and developed sections were prepared with colorimetric detection strategies such that the stained slides are viewed by light microscopy, and imaged on the Aperio Scanscope Digital Pathology platform. There are many comparable platforms that allow rapid scanning of slides and conversion of the slide into a digital image file.
[000166] Tumor zones within a complex pathological specimen were identified by a trained pathologist. In addition, computer assisted histopathology and morphologic criteria were included to discriminate tumor from non-tumor. By imparting this selective annotation, a computer-guided scoring strategy was directed specifically to the annotated regions.
[000167] Annotated tumor regions were scored via automated algorithms written and employed versus the specific stains. There are many alternative algorithms that are capable of representing the different features of a digital image file of a pathology specimen. The key features of the analysis were:
Machine learning of biomarker pattern recognition [1 process per biomarker] IHC Nuclear image analysis algorithms [1 process per biomarker] Candidate calibrator assessment and comparison for the xenograft set Distribution and partition ranking per biomarker
Algorithm definition and testing
Biomarker panel assembly and subgrouping
Statistical analysis on biomarker algorithms
[000168] To score the annotated regions, a weighted scoring system is utilized whereby the % of cells scored as 3 is weighted more heavily than those scored as 2 and those scored as 2 weighted more heavily than the % scored as 1.
3 X (% of 3+ staining cells) + 2 X (% of 2+ staining cells) + 1 X (% of 1+ staining cells) = QIM
[000169] Thus, a samples maximum achievable QIM score is 300. Algorithms were written to achieve a good representative sampling of tumor nuclei and that complete representation is unnecessary and unlikely by our experience to substantially change the overall QIM score.
[000170] One of the key metrics that the IHC staining for a given biomarker is a suitable dynamic range (scorable QIM) for utility in the ability to separate patient groups based on response and non-response in ovarian cancer predictive treatment based on biomarker panel assembly. Increased or decreased activity of a pathway may impart sensitivity to DNA- or mitotic-targeted chemotherapies, or to enzyme/protein targeted chemotherapies, such as PARP inhibitors.
[000171] Figures 1 through 6 highlight the scorable dynamic ranges for biomarkers MLH1, pMAPKAPK2, PAR, PARP1, XPF, and KI67, respectively. For each of these markers with roles in DNA repair, one can focus on the dynamic ranges that each displays at the point residing at the nuclear localization. In the case of some biomarkers, their cellular localization may actually change depending on characteristics such as post-translational modification state of the protein, cell-cycle point, engagement state of DNA repair mechanisms, proliferative state of the cell, etc. Such is the case for pMAPKAPK2 (Figure 2) that displays both nuclear and cytoplasmic localization - independent metrics from which to separate responders from non-responders in ovarian cancer treatment regimen efficacy prediction modeling. It is also interesting to note here that these dynamic ranges of QIM scores can be obtained for ovarian cells of different origins including normal, carcinoma, and regional and distal metastases.
[000172] Further, biomarkers such as FancD2 and pH2AX in addition to nuclear IHC staining may additionally display intense nuclear staining in the form of foci, most likely representing a granular depiction at a site of DNA repair in which the protein itself has focused its location and action (Figure 7). The differences in the ways these foci-inducing markers are scored, such as either gross nuclear expression or enumeration of foci, and the dynamic range that either may exhibit on display in Figure 8. Figure 9 shows that pH2AX is another such biomarker whose expression can be represented by measurement of either nuclear IHC staining or enumeration of foci. These two independent measurements are not coincidental representations of the same value and again represent two independent metrics that can be utilized in the separation of responders and non-responders relative to ovarian cancer treatments. Figure 10 compares FancD2 and pH2AX IHC expression as well as enumeration of foci in independent patients and illustrates that these two biomarkers display expression patterns that move independently of one another and as such are not prone to redundancy when utilized together in mutimarker modeling of ovarian cancer therapeutic response.
[000173] Example 2: Evalaution of Ovarian Cancer Patient Specimens By Tissue Micro Arrays
[000174] In the analysis and assembly of univariate and multimarker modeling studies, one can survey more samples and generate models through the utilization of Tissue Micro Arrays (TMAs). Figure 11 shows one such tissue microarray. In a standard fashion, multiple cores from each patient's FFPE tumor (usually three or four) wer assembled onto microarrays and then sectioned. Slide-to-slide variability is minimized in the staining and scoring of large numbers of samples as more information with direct comparability from a single stained slide may be obtained in an efficient manner. Results from multiple cores of individual patients are scored and assembled into ranked distribution plots as shown in Figure 12 indicating what the dynamic range for a given biomarker An average QIM score is generated in plots such as these is comprised of several cores derived from the same patient that may display heterogeneity in expression for a given biomarker as demonstrated by the error bars represented with each of the given biomarker distribution plots.
[000175] Example 3: Evaluation of Tumor Heterogeneity using DNA repair biomarkers expression patterns
[000176] DNA repair biomarkers expression patterns can clearly be impacted by tumor heterogeneity. In one example, two markers are evaluated in serial FFPE sections from the tumor of a patient with serous ovarian cancer (Figure 13). FANCD2 and pMK2 IHC determinations are illustrated. The demarcated zones illustrate the marker heterogeity in the tumor from a single patient. Although these proteins are both in the FA/HR DNA repair pathway and the DNA damage response network, their expression is divergent from the perspective of a single patients tumor. Other molecular analysis criteria, such as RNA expression and DNA analysis, as well as proteomic analysis from tumor isolates, do not adequately represent the diversity exhibited within a single tumor that might be relevant to therapy selection and responsiveness. Scores for heterogeneity are another means to determine differences between patients with regard to biomarker panels and response algorithms.
[000177] TABLE 1: DNA Repair and DNA Damage Response Markers
Figure imgf000044_0001
hNEIL3 5. BER
AAG 6. BER
UNG1 7. BER
TDG 8. BER
MUTY 9. BER
MTH1 10. BER
MBD4 11. BER
APE1 12. BER
XPG 13. BER
DNAPOLP 14. BER
XRCC1 15. BER
PARP1 16. BER
DNAPOL51 17. BER
DNAPOL52 18. BER
DNAPOL53 19. BER
DNAPOL54 20. BER
DNAPOL55 21. BER
DNAPOLel 22. BER
DNAPOL82 23. BER
DNAPOL83 24. BER
DNAPOL84 25. BER
DNAPOL85 26. BER
DNALigasel 27. BER
PCNA 28. BER
UBC13 29. BER
MMS2 30. BER FEN1 31. BER
RFC1 32. BER
RFC2 33. BER
PAR 34. BER
RFC4 35. BER
RFC5 36. BER
DNALigasel 37. BER
DNAligase3 38. BER
Aprataxin (Aptx) 39. BER
XRCC1 40. HR
PARP1 41. HR
FEN1 42. HR
DNA ligasel 43. HR
SNM1 44. HR
H2A 45. HR
RPA1 46. HR
RPA2 47. HR
RPA3 48. HR
RAD51 49. HR
XRCC2 50. HR
XRCC3 51. HR
RAD51L1 52. HR
RAD51L2 53. HR
RAD51L3 54. HR
DMC1 55. HR
RAD52 56. HR RAD54 57. HR
MUS81 58. HR
MMS4 59. HR
EMSY 60. HR
BRCA1 61. HR
BARD1 62. HR
BLM 63. HR
BLAP75 64. HR
SRS2 65. HR
SAE2 66. HR
ERCC1 67. HR
TRF2 68. HR/FA
BRCA2/FANCD 1 69. HR/FA
FANCA 70. HR/FA
FANCB 71. HR/FA
FANCC 72. HR/FA
FANCD1 73. HR/FA
FANCD2 74. HR/FA
FANCE 75. HR/FA
FANCF 76. HR/FA
FANCG 77. HR/FA
FANCJ 78. HR/FA
FANCL 79. HR/FA FANCM 80. HR/FA hHefl 81. HR/FA
FANCI 82. HR/FA
USP1 83. HR FA
PALB2/FANCN 84. HR/FA
DNMT1 85. MMR hMLHl 86. MMR hPMS2 87. MMR hPMSl 88. MMR
GTBP (hMSH6) 89. MMR hMSH2 90. MMR hMSH3 91. MMR
HMGB1 92. MMR
MSH4 93. MMR
MSH5 94. MMR
EXOl 95. MMR
DNAPOL51 96. MMR
DNAPOL52 97. MMR
DNAPOL53 98. MMR
DNAPOL54 99. MMR
DNAPOL55 100. MMR
DNAPOLel 101. MMR DNAPOL82 102. MMR
DNAPOL83 103. MMR
DNAPOL84 104. MMR
DNAPOL85 105. MMR
DNA Ligase I 106. MMR
PCNA 107. MMR
RPA1 108. MMR
RPA2 109. MMR
RPA3 110. MMR
MUTY 111. MMR
MRE11 112. DDR
RAD50 113. DDR
NBS1 114. DDR
H2A 115. DDR
ATM 116. DDR
P53 117. DDR
SMC1 118. DDR
ATF2 119. DDR
CHK1 120. DDR
CHK2 121. DDR
MAPKAP Kinase2 122. DDR RPA1 123. DDR
RPA2 124. DDR
RPA3 125. DDR
RAD 17 126. DDR
RFC1 127. DDR
RFC2 128. DDR
RFC3 129. DDR
RFC4 130. DDR
RFC5 131. DDR
RAD9 132. DDR
RAD1 133. DDR
HUS1 134. DDR
ATRIP 135. DDR
ATR 136. DDR
MDC1 137. DDR
CLASPIN 138. DDR
TOPB1 139. DDR
BRCC36 140. DDR
BLM 141. DDR
SRS2 142. DDR
SAE2 143. DDR P53BP1 144. DDR
ING1 145. DDR
ING2 146. DDR
SMC1 147. DDR
BLAP75 148. DDR
BACH1 149. DDR
BRCA1 150. DDR
BRCA2 151. DDR
BARD1 152. DDR
RAP80 153. DDR
Abraxas 154. DDR
CDT1 155. DDR
RPB8 156. DDR
PPM1D 157. DDR
GADD45 158. DDR
DTL/CDT2 159. DDR
HCLK2 160. DDR
CTIP 161. DDR
BAAT1 162. DDR
HDM2/MDM2 163. DDR
APLF (aprataxin- and PNK- 164. DDR like factor)
14-3-3 σ 165. DDR
Cdc25A 166. DDR
Cdc25B 167. DDR Cdc25C 168. DDR
PBIP1 169. DDR
H2A 170. NER
XPC 171. NER
HR23A 172. NER
HR23B 173. NER
DDB1 174. NER
DDB2 175. NER
XPD 176. NER
XPB 177. NER
XPG 178. NER
CSA 179. NER
CSB 180. NER
XPA 181. NER
XPF 182. NER
ERCC1 183. NER
RNAPolymerase2 184. NER
GTF2H1 185. NER
GTF2H2 186. NER
GTF2H3 187. NER
GTF2H4 188. NER
GTF2H5 189. NER MNAT1 190. NER
MAT1 191. NER
CDK7 192. NER
CyclinH 193. NER
PCNA 194. NER
RFC1 195. NER
RFC2 196. NER
RFC3 197. NER
RFC4 198. NER
RFC5 199. NER
DNAPOL51 200. NER
DNAPOL52 201. NER
DNAPOL53 202. NER
DNAPOL54 203. NER
DNAPOL55 204. NER
DNAPOLel 205. NER
DNAPOL82 206. NER
DNAPOL83 207. NER
DNAPOL84 208. NER
DNAPOL85 209. NER
DNALigasel 210. NER ϋΝΑΡΟίη 211. TLS
DNAPOLi 212. TLS
DNAPOLK 213. TLS
REV1 214. TLS
DNAPOLC 215. TLS
DNAPOL6 216. TLS
PCNA 217. TLS
UBC13 218. TLS
MMS2 219. TLS
RAD5 220. TLS hRAD6A 221. TLS hRAD6B 222. TLS
RAD 18 223. TLS
WRN 224. TLS
USP1 225. TLS
SIRT6 226. NHEJ
H2A 227. NHEJ
ARP4 228. NHEJ
ARP8 229. NHEJ
Ino80 230. NHEJ
SWR1 231. NHEJ
KU70 232. NHEJ KU80 233. NHEJ
DNAPKcs 234. NHEJ
Artemis 235. NHEJ
PS02 236. NHEJ
XRCC4 237. NHEJ
DNA LIGASE4 238. NHEJ
XLF 239. NHEJ
DNAPOI 240. NHEJ
PNK 241. NHEJ
METNASE 242. NHEJ
TRF2 243. NHEJ
MGMT 244. Non-classified
TDP1 245. Non-classified ϋΝΑΡΟΕμ 246. Non-classified hABHl 247. Non-classified hABH2 248. Non-classified hABH3 249. Non-classified hABH4 250. Non-classified hABH5 251. Non-classified hABH6 252. Non-classified hABH7 253. Non-classified hABH8 254. Non-classified TOPOl 255. Non-classified
TOPOII 256. Non-classified
UBC9 257. Non-classified
UBL1 258. Non-classified
MMS21 259. Non-classified
Table 2 Ovarian Cancer DNA Repair and DNA Damage Response Markers
Figure imgf000057_0001
Table 3 Ovarian Cancer Biomarkers for Use in Combined Algorithms with DNA Repair and DNA Damage Response Markers
Figure imgf000058_0001
β-catenin 290 Signal Transducer β-tubulin 2 291 Microtubule structure and regulation
References
Armstrong, DK, et al. (2006) Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med 354:34.
Bartolucci, R. et al. (2009) XPG mRNA expression levels modulate prognosis in resected non-small-cell lung cancer in conjunction with BRCAl and ERCC1 expression. Clin Lung Cancer. Jan;10(l):47-52.
Bristow, R.E. and Chi, D.S (2006) Platinum-based neoadjuvant chemotherapy and interval surgical cytoreduction for advanced ovarian cancer: A meta-analysis. Gynecol Oncol;
103: 1070.
Brustmann, H. (2007) Poly(adenosine diphosphate-ribose) polymerase expression in serous ovarian carcinoma: correlation with p53, MIB-1, and outcome. Int J Gynecol Pathol. 26, 147-53.
Chirnomas, D. et al. (2006) Chemosensitization to cisplatin by inhibitors of the Fanconi anemia/BRCA pathway. Mol Cancer Ther. 5, 952-961. Fong P.C. et al. (2009) Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009 361, 123-34. Epub 2009 Jun 2.
Conte, PF, et al. (2007) Final results of After-6 protocol 1 : A phase III trial of observation versus 6 courses of paclitaxel (Pac) in advanced ovarian cancer patients in complete response (CR) after platinum-paclitaxel chemotherapy (abstract). J Clin Oncol 25:275s.
Husain A., et al.(1998) BRCAl up-regulation is associated with repair-mediated resistance to cisdiamminedichloroplatinum (II). Cancer Res 58, 1120-1123.
The International Collaborative Ovarian Neoplasm (ICON) group (2002) Paclitaxel plus carboplatin versus standard chemotherapy with either single agent carboplatin or cyclophosphamide, doxorubicin, and cisplatin in women with ovarian cancer: the ICON3 randomised trial.. Lancet 360:505.
Kennedy, R.D. et al. (2007) Fanconi anemia pathway-deficient tumor cells are
hypersensitive to inhibition of ataxia telangiectasia mutated. J Clin Invest.117, 1440-1449. Lin, K. et al. (2008) Protein expression levels of excision repair cross-complementation group 1 and xeroderma pigmentosum D correlate with response to platinum-based chemotherapy in the patients with advanced epithelial ovarian cancer. Int J Gynecol Cancer.18, 1007-1012.
Markman, M, et al. (2003) Phase III randomized trial of 12 versus 3 months of maintenance paclitaxel in patients with advanced ovarian cancer after complete response to platinum and paclitaxel-based chemotherapy: a Southwest Oncology Group and Gynecologic Oncology Group trial. J Clin Oncol 21 :2460.
Materna, V. et al (2007) Expression of factors involved in regulation of DNA mismatch repair- and apoptosis pathways in ovarian cancer patients. Oncol Rep. 17, 505-516.
McGuire, WP, et al. (1996) Cyclophosphamide and cisplatin compared with paclitaxel and cisplatin in patients with stage III and stage IV ovarian cancer. N Engl J Med 334: 1. Muggia, FM, et al. (2000) Phase III randomized study of cisplatin versus paclitaxel versus cisplatin and paclitaxel in patients with suboptimal stage III or IV ovarian cancer: A
Gynecologic Oncology Group study. J Clin Oncol 2000; 18: 106.
Piccart, MJ, et al. (2000) Randomized intergroup trial of cisplatin-paclitaxel versus cisplatin- cyclophosphamide in women with advancd epithelial ovarian cancer: three- year results. J Natl Cancer Inst 92:699.
Planchard, D. et al. (2009) The NER proteins are differentially expressed in ever smokers and in never smokers with lung adenocarcinoma. Ann Oncol. Jul;20(7): 1257-63. Epub 2009 Mar 17.
Quinn J.E., et al. (2003) BRCA1 functions as a differential modulator of chemotherapy- induced apoptosis. Cancer Res 63, 6221-6228.
Samimi, G. et al (2000) Analysis of MLH1 and MSH2 expression in ovarian cancer before and after platinum drug-based chemotherapy. Clinical Cancer Research 6, 1415-1421. Scartozzi, M. et al (2003) Loss of hMLHl expression correlates with improved survival in stage III- IV ovarian cancer patients. Eur J Cancer. 39, 1144-1149.
Smith, S. et al. (2007) ERCC1 genotype and phenotype in epithelial ovarian cancer identify patients likely to benefit from paclitaxel treatment in addition to platinum-based therapy. J Clin Oncol. 25, 5172-5179.
Steffensen, K.D. et al, (2009). The relationship of platinum resistance and ERCC1 protein expression in epithelial ovarian cancer. Int J Gynecol Cancer. 19, 820-825.
Stevens, E.V. et al. (2008) Predicting cisplatin and trabectedin drug sensitivity in ovarian and colon cancers. Mol Cancer Ther. 7, 10-18.
Swisher, E.M. et al. (2009) Methylation and protein expression of DNA repair genes:
association with chemotherapy exposure and survival in sporadic ovarian and peritoneal carcinomas. Molecular Cancer 8, 1-11.
Taniguchi, T., et al. (2003): Disruption of the Fanconi anemia-BRCA pathway in cisplatin- sensitive ovarian tumors. Nat Med 2003, 9:568-574.
Wang, Y. et al. (2006) Fanconi anemia D2 protein confers chemoresistance in response to the anticancer agent, irofulven. Mol Cancer Ther. 5, 3153-3161.
Zhou C. et al. (2003): Role of BRCA1 in cellular resistance to paclitaxel and ionizing radiation in an ovarian cancer cell line carrying a defective BRCA1. Oncogene 22,2396- 2404.

Claims

What is claimed is:
1. A method of determining the sensitivity of a ovarian cancer to a chemotherapeutic agent comprising identifying an alteration in at least one DNARMARKER selected from the group consisting of MLHl, pMAPKAPK2, PAR, PARPl, XPF, pH2AX
and FAND2 wherein the presence of said alteration indicates said cell is sensitive to a chemotherapeutic agent.
2. A method of determining the resistance of a ovarian cancer to a chemotherapeutic agent comprising identifying an alteration in at least one DNARMARKER selected from the group consisting of MLHl, pMAPKAPK2, PAR, PARPl, XPF, pH2AX
and FAND2 wherein the absence of said alteration indicates said cell is resistant to a chemotherapeutic agent.
3. A method of predicting the effectiveness of a chemotherapeutic agent or combination of chemotherapeutic agents of a subject having a ovarian cancer comprising
a) measuring the level of an effective amount of one or more DNARMARKERS selected from the group consisting of selected from the group consisting of MLHl, pMAPKAPK2, PAR, PARPl, XPF, pH2AX and FAND2 in a sample from the subject, and
b) comparing the level of the effective amount of the one or more
DNARMARKERS to a reference value.
4. A method of monitoring the chemotherapeutic agent treatment of a subject with ovarian cancer comprising
a) detecting the level of an effective amount of one or more DNARMARKERS selected from the group consisting of DNARMARKERS selected from the group consisting of MLHl, pMAPKAPK2, PAR, PARPl, XPF, pH2AX and FAND2 in a first sample from the subject at a first period of time;
b) detecting the level of an effective amount of one or more DNARMARKERS in a second sample from the subject at a second period of time;
c) comparing the level of the effective amount of one or more DNARMARKERS detected in step (a) to the amount detected in step (b), or to a reference value.
5. A method of predicting a ovarian cancer patient's likelihood to have an event regardless of treatment comprising
a) measuring the level of an effective amount of one or more
DNARMARKERS selected from the group consisting of DNARMARKERS selected from the group consisting of MLH1, pMAPKAPK2, PAR, PARP1, XPF, pH2AX
and FAND2 in a sample from the subject, and
b) comparing the level of the effective amount of the one or more
DNARMARKERS to a reference value.
6. The method of any one of claims 1-5, further comprising measuring Ki67
7. The method of any one of claims 1-6, further comprising determining an alteration in a DNARMARKER selected from Table 1, Table 2 or Table 3.
8. The method of any one of claims 1-7, wherein said alteration is determined by protein levels, post-translational protein modifications, gene copy levels, chromosome copy levels, polymorphisms, nucleic acid modifications.
9. The method claim 8, wherein said post-translational modification is selected from the group consisting of phosphorylation, ubiquitination, sumo-ylation, acetylation, alkylation, methylation, glycylation, glycosylation, hydroxylation, isoprenylation, lipoylation, phosphopantetheinylation, sulfation, selenation and C-terminal amidation.
10. The method of any one of claims 1-9, wherein said alteration is an increase or decrease in protein expression of said DNARMARKER.
11. The method of any one of claims 1-10, wherein said chemotherapeutic agent is selected from the group consisting of PARP inhibitor, taxane or a platinum drug or any combination of therof.
The method of claim 11 , wherein said platinum drug is carboplatin
13 The method of claim 5, wherein said event is selected from the group consisting of distant recurrence, local recurrence, overall survival, relapse free disease, pathological complete response, partial response, no response.
14. The method any one of claims 1-13, wherein said sample is selected from the group consisting of ovarian tumor tissue, normal ovarian tissue, and blood.
15. An algorithm that is derived from the list of biomarkers in Table 1, Table 2 or Table 3 which specifies how the biomarkers are associated in relation to the other biomarkers in the panel, such that the biomarker algorithm indicates a predictive or prognostic value in treatment response of ovarian cancer.
PCT/US2011/046581 2010-08-04 2011-08-04 Biomarkers for the identification monitoring and treatment of ovarian cancer WO2012019000A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US37054610P 2010-08-04 2010-08-04
US61/370,546 2010-08-04

Publications (2)

Publication Number Publication Date
WO2012019000A2 true WO2012019000A2 (en) 2012-02-09
WO2012019000A3 WO2012019000A3 (en) 2012-05-24

Family

ID=45560069

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/046581 WO2012019000A2 (en) 2010-08-04 2011-08-04 Biomarkers for the identification monitoring and treatment of ovarian cancer

Country Status (1)

Country Link
WO (1) WO2012019000A2 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014160080A1 (en) * 2013-03-14 2014-10-02 Children's Medical Center Corporation Cancer diagnosis, treatment selection and treatment
WO2015042115A1 (en) * 2013-09-17 2015-03-26 The Board Of Trustees Of The Leland Stanford Junior University Biomarkers for ovarian cancer
US9279156B2 (en) 2011-06-17 2016-03-08 Myriad Genetics, Inc. Methods and materials for assessing allelic imbalance
US9388472B2 (en) 2011-12-21 2016-07-12 Myriad Genetics, Inc. Methods and materials for assessing loss of heterozygosity
US9512485B2 (en) 2010-08-24 2016-12-06 Dana-Farber Cancer Institute. Inc. Methods for predicting anti-cancer response
US10190160B2 (en) 2012-02-23 2019-01-29 Children's Medical Center Corporation Methods for predicting anti-cancer response
US10400287B2 (en) 2014-08-15 2019-09-03 Myriad Genetics, Inc. Methods and materials for assessing homologous recombination deficiency
US11091808B2 (en) 2012-06-07 2021-08-17 Institut Curie Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors
US11149316B2 (en) 2013-12-09 2021-10-19 Institut Curie Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors
US20230197223A1 (en) * 2016-03-31 2023-06-22 OM1, Inc. Health care information system providing additional data fields in patient data

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050059013A1 (en) * 2002-08-06 2005-03-17 The Johns Hopkins University Use of biomarkers for detecting ovarian cancer
US20070141582A1 (en) * 2005-12-15 2007-06-21 Weiwei Li Method and kit for detection of early cancer or pre-cancer using blood and body fluids
US20070292883A1 (en) * 2006-06-12 2007-12-20 Ossovskaya Valeria S Method of treating diseases with PARP inhibitors
US20080038723A1 (en) * 2004-06-07 2008-02-14 Bitter Grant A Functional assessment of DNA mismatch repair gene variants
US20100068818A1 (en) * 2006-10-04 2010-03-18 The Johns Hopkins University Algorithms for multivariant models to combine a panel of biomarkers for assessing the risk of developing ovarian cancer

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050059013A1 (en) * 2002-08-06 2005-03-17 The Johns Hopkins University Use of biomarkers for detecting ovarian cancer
US20080038723A1 (en) * 2004-06-07 2008-02-14 Bitter Grant A Functional assessment of DNA mismatch repair gene variants
US20070141582A1 (en) * 2005-12-15 2007-06-21 Weiwei Li Method and kit for detection of early cancer or pre-cancer using blood and body fluids
US20070292883A1 (en) * 2006-06-12 2007-12-20 Ossovskaya Valeria S Method of treating diseases with PARP inhibitors
US20100068818A1 (en) * 2006-10-04 2010-03-18 The Johns Hopkins University Algorithms for multivariant models to combine a panel of biomarkers for assessing the risk of developing ovarian cancer

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10577662B2 (en) 2010-08-24 2020-03-03 Dana-Farber Cancer Institute, Inc. Methods for predicting anti-cancer response
US9512485B2 (en) 2010-08-24 2016-12-06 Dana-Farber Cancer Institute. Inc. Methods for predicting anti-cancer response
US9574229B2 (en) 2011-06-17 2017-02-21 Myriad Genetics, Inc. Methods and materials for assessing allelic imbalance
US11225685B2 (en) 2011-06-17 2022-01-18 Myriad Genetics, Inc. Methods and materials for assessing allelic imbalance
US9279156B2 (en) 2011-06-17 2016-03-08 Myriad Genetics, Inc. Methods and materials for assessing allelic imbalance
US10626449B2 (en) 2011-06-17 2020-04-21 Myriad Genetics, Inc. Methods and materials for assessing allelic imbalance
US9388472B2 (en) 2011-12-21 2016-07-12 Myriad Genetics, Inc. Methods and materials for assessing loss of heterozygosity
US10612098B2 (en) 2011-12-21 2020-04-07 Myriad Genetics, Inc. Methods and materials for assessing loss of heterozygosity
US10190160B2 (en) 2012-02-23 2019-01-29 Children's Medical Center Corporation Methods for predicting anti-cancer response
US11299782B2 (en) 2012-02-23 2022-04-12 Children's Medical Center Corporation Methods for predicting anti-cancer response
US11091808B2 (en) 2012-06-07 2021-08-17 Institut Curie Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors
US10308986B2 (en) 2013-03-14 2019-06-04 Children's Medical Center Corporation Cancer diagnosis, treatment selection and treatment
WO2014160080A1 (en) * 2013-03-14 2014-10-02 Children's Medical Center Corporation Cancer diagnosis, treatment selection and treatment
CN105531590A (en) * 2013-09-17 2016-04-27 利兰斯坦福初级大学董事会 Biomarkers for ovarian cancer
WO2015042115A1 (en) * 2013-09-17 2015-03-26 The Board Of Trustees Of The Leland Stanford Junior University Biomarkers for ovarian cancer
US11149316B2 (en) 2013-12-09 2021-10-19 Institut Curie Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors
US10400287B2 (en) 2014-08-15 2019-09-03 Myriad Genetics, Inc. Methods and materials for assessing homologous recombination deficiency
US20230197223A1 (en) * 2016-03-31 2023-06-22 OM1, Inc. Health care information system providing additional data fields in patient data

Also Published As

Publication number Publication date
WO2012019000A3 (en) 2012-05-24

Similar Documents

Publication Publication Date Title
WO2012019000A2 (en) Biomarkers for the identification monitoring and treatment of ovarian cancer
US20230117133A1 (en) Methods and materials for assessing homologous recombination deficiency
US20210310079A1 (en) Methods and materials for assessing homologous recombination deficiency
US20110217713A1 (en) Biomarkers For The Identification, Monitoring, And Treatment Of Non-Small Cell Lung Cancer (NSCLC)
US20090239229A1 (en) DNA Repair Proteins Associated With Triple Negative Breast Cancers and Methods of Use Thereof
AU2009246256A1 (en) Biomarkers for the identification, monitoring, and treatment of head and neck cancer
US9593377B2 (en) Signatures and determinants associated with cancer and methods of use thereof
US20150111970A1 (en) Genetic markers and diagnostic methods for resistance of breast cancer to hormonal therapies
Lu et al. Discovery of a biomarker candidate for surgical stratification in high-grade serous ovarian cancer
Ratovomanana et al. Prediction of response to immune checkpoint blockade in patients with metastatic colorectal cancer with microsatellite instability
JP2023548419A (en) Method for Determining Sensitivity to PARP inhibitor or genotoxic drugs based on non-function l transcripts}
Noushmehr et al. Detection of glioma and prognostic subtypes by non-invasive circulating cell-free DNA methylation markers
US20240141435A1 (en) Methods for detecting and predicting cancer
WO2021211057A1 (en) Method of predicting the responsiveness to a cancer therapy
WO2011143337A1 (en) Biomarkers for the identification monitoring and treatment of breast cancer
US20230212653A1 (en) Methods and materials for assessing homologous recombination deficiency in breast cancer subtypes
Raman et al. Update on genetic tests currently available for clinical use in common cancers
Ding Mature B-Cell Neoplasms
Mabonga et al. Molecular Profiling and Personalized Medicine in Drug-Resistant Breast Cancer Check for updates
Styczynskı et al. Microarray technology in pediatric malignancies: an insight towards individualized therapy

Legal Events

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

Ref document number: 11815317

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11815317

Country of ref document: EP

Kind code of ref document: A2