WO2010042525A9 - Ovarian cancer biomarkers and uses thereof - Google Patents

Ovarian cancer biomarkers and uses thereof Download PDF

Info

Publication number
WO2010042525A9
WO2010042525A9 PCT/US2009/059706 US2009059706W WO2010042525A9 WO 2010042525 A9 WO2010042525 A9 WO 2010042525A9 US 2009059706 W US2009059706 W US 2009059706W WO 2010042525 A9 WO2010042525 A9 WO 2010042525A9
Authority
WO
WIPO (PCT)
Prior art keywords
biomarker
ovarian cancer
individual
biomarkers
biological sample
Prior art date
Application number
PCT/US2009/059706
Other languages
French (fr)
Other versions
WO2010042525A1 (en
Inventor
Larry Gold
Marty Stanton
Edward N. Brody
Rachel M. Ostroff
Dominic Zichi
Alex A.E. Stewart
Original Assignee
Somalogic, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Somalogic, Inc. filed Critical Somalogic, Inc.
Priority to AU2009302537A priority Critical patent/AU2009302537A1/en
Priority to EP09819761A priority patent/EP2350917A4/en
Priority to CA2737004A priority patent/CA2737004A1/en
Publication of WO2010042525A1 publication Critical patent/WO2010042525A1/en
Publication of WO2010042525A9 publication Critical patent/WO2010042525A9/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
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B40/00ICT specially adapted for biostatistics; ICT specially adapted for bioinformatics-related machine learning or data mining, e.g. knowledge discovery or pattern finding
    • G16B40/20Supervised data analysis
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B40/00ICT specially adapted for biostatistics; ICT specially adapted for bioinformatics-related machine learning or data mining, e.g. knowledge discovery or pattern finding

Definitions

  • the present application relates generally to the detection of biomarkers and the diagnosis of cancer in an individual and, more specifically, to one or more biomarkers, methods, devices, reagents, systems, and kits for diagnosing cancer, more particularly ovarian cancer, in an individual.
  • Ovarian cancer is the eighth most common cancer in women and the fifth leading cause of cancer-related deaths in women in the United States. Of all females born in the United States, one of every 70 will develop ovarian cancer and one of every 100 will die from this disease.
  • the American Cancer Society estimates that approximately 21,550 women will be diagnosed with ovarian cancer in 2009 (American Cancer Society. Cancer Facts & Figures 2009. Atlanta: American Cancer Society; 2009). It is estimated that 14,600 women will die from this disease in 2009.
  • CA- 125 cancer antigen 125
  • serum biomarker for ovarian cancer.
  • Serum concentrations of CA- 125 are elevated (>35 U/ml) in 75-80% of patients with advanced-stage disease and this marker is routinely used to follow response to treatment and disease progression in patients from whom CA- 125 -secreting tumors have been resected.
  • levels of CA- 125 are correlated with tumor volume, only 50% of patients with early-stage disease have elevated levels, indicating that the sensitivity of CA- 125 as a screening tool for early stage disease is limited.
  • CA- 125 screening is further limited by the high frequency of false-positive results associated with a variety of benign conditions, including endometriosis, pregnancy, menstruation, pelvic inflammatory disease, peritonitis, pancreatitis, and liver disease.
  • Classification of cancers determines appropriate treatment and helps determine the prognosis of the patient.
  • Ovarian cancers are classified according to histology (i.e., "grading") and extent of the disease (i.e., "staging”) using recognized grade and stage systems. In grade I, the tumor tissue is well differentiated. In grade II, tumor tissue is moderately well differentiated. In grade III, the tumor tissue is poorly differentiated. Grade III correlates with a less favorable prognosis than either grade I or II.
  • Stage I is generally confined within the capsule surrounding one (stage IA) or both (stage IB) ovaries, although in some stage I (i.e. stage IC) cancers, malignant cells may be detected in ascites, in peritoneal rinse fluid, or on the surface of the ovaries.
  • Stage II involves extension or metastasis of the tumor from one or both ovaries to other pelvic structures.
  • stage HA the tumor extends or has metastasized to the uterus, the fallopian tubes, or both.
  • Stage HB involves metastasis of the tumor to the pelvis.
  • Stage HC is stage HA or HB with the added requirement that malignant cells may be detected in ascites, in peritoneal rinse fluid, or on the surface of the ovaries.
  • the tumor comprises at least one malignant extension to the small bowel or the omentum, has formed extra-pelvic peritoneal implants of microscopic (stage IIIA) or macroscopic ( ⁇ 2 centimeter diameter, stage MB; >2 centimeter diameter, stage IIIC) size, or has metastasized to a retroperitoneal or inguinal lymph node (an alternate indicator of stage IIIC).
  • stage IV distant (i.e. non-peritoneal) metastases of the tumor can be detected.
  • Treatment options include surgery, chemotherapy, and occasionally radiation therapy. Surgery usually involves removal of one or both ovaries, fallopian tubes (salpingoophorectomy), and the uterus (hysterectomy).
  • the 1- and 5-year relative survival of ovarian cancer patients is 75% and 46%, respectively. If diagnosed at the localized stage, the 5-year survival rate is 93%; however, only 19% of all cases are detected at this stage, usually fortuitously during another medical procedure. The majority of cases (67%) are diagnosed at distant stage. For women with regional and distant disease, 5-year survival rates are 71% and 31%, respectively; the chance of recurrence in these women is 20-85%. The 10-year relative survival rate for all stages combined is 39%. Therefore, ovarian cancer tends to be diagnosed too late to save women's lives. Detecting recurrence and predicting and monitoring response to therapy is important for making informed decisions on appropriate treatment throughout the care of these patients.
  • a blood screening test that can reliably detect early stage ovarian cancer will save thousands of lives each year. Where methods of early diagnosis in cancer exist, the benefits are generally accepted by the medical community. Cancers for which widely utilized screening protocols exist have the highest 5-year survival rates, such as breast cancer (88%) and colon cancer (65%) versus 46% for ovarian cancer. However, fortuitous detection of early stage ovarian cancer is associated with a substantial increase in 5-year survival (>95%). Therefore, early detection could significantly impact long-term survival. This demonstrates the clear need for diagnostic methods that can reliably detect early-stage ovarian cancer.
  • Biomarker selection for a specific disease state involves first the identification of markers that have a measurable and statistically significant difference in a disease population compared to a control population for a specific medical application.
  • Biomarkers can include secreted or shed molecules that parallel disease development or progression and readily diffuse into the blood stream from ovarian tissue or from surrounding tissues and circulating cells in response to a tumor. The biomarker or set of biomarkers identified are generally clinically validated or shown to be a reliable indicator for the original intended use for which it was selected.
  • Biomarkers can include small molecules, peptides, proteins, and nucleic acids.
  • biomarker discovery and detection methods using these technologies have serious limitations for the identification of diagnostic biomarkers. These limitations include an inability to detect low-abundance biomarkers, an inability to consistently cover the entire dynamic range of the proteome, ⁇ reproducibility in sample processing and fractionation, and overall ⁇ reproducibility and lack of robustness of the method. Further, these studies have introduced biases into the data and not adequately addressed the complexity of the sample populations, including appropriate controls, in terms of the distribution and randomization required to identify and validate biomarkers within a target disease population.
  • biochemical pathways culminate in or are started by secreted proteins that work locally within the pathology, for example growth factors are secreted to stimulate the replication of other cells in the pathology, and other factors are secreted to ward off the immune system, and so on. While many of these secreted proteins work in a paracrine fashion, some operate distally in the body.
  • One skilled in the art with a basic understanding of biochemical pathways would understand that many pathology-specific proteins ought to exist in blood at concentrations below (even far below) the detection limits of 2D gels and mass spectrometry. What must precede the identification of this relatively abundant number of disease biomarkers is a proteomic platform that can analyze proteins at concentrations below those detectable by 2D gels or mass spectrometry.
  • biomarkers, methods, devices, reagents, systems, and kits that enable (a) the differentiation of benign pelvic masses from ovarian cancer; (b) referral to a gynecologic oncology surgeon rather than a general gynecologic surgeon to surgically treat cases of ovarian cancer; (c) the detection of ovarian cancer biomarkers; and (d) the diagnosis of ovarian cancer.
  • the present application includes biomarkers, methods, reagents, devices, systems, and kits for the detection and diagnosis of cancer and more particularly, ovarian cancer.
  • the biomarkers of the present application were identified using a multiplex aptamer- based assay, which is described in detail in Example 1.
  • this application describes a surprisingly large number of ovarian cancer biomarkers that are useful for the detection and diagnosis of ovarian cancer.
  • identifying these biomarkers over 800 proteins from hundreds of individual samples were measured, some of which were at concentrations in the low femtomolar range. This is about four orders of magnitude lower than biomarker discovery experiments done with 2D gels or mass spectrometry.
  • ovarian cancer biomarkers are useful alone for detecting and diagnosing ovarian cancer
  • methods are described herein for the grouping of multiple subsets of the ovarian cancer biomarkers that are useful as a panel of biomarkers. Once an individual biomarker or subset of biomarkers has been identified, the detection or diagnosis of ovarian cancer in an individual can be accomplished using any assay platform or format that is capable of measuring differences in the levels of the selected biomarker or biomarkers in a biological sample.
  • one or more biomarkers are provided for use either alone or in various combinations to diagnose ovarian cancer or permit the differential diagnosis of pelvic masses as benign or malignant.
  • Exemplary embodiments include the biomarkers provided in Table 1, which as noted above, were identified using a multiplex aptamer-based assay, as described in Examples 1 and 2. The markers provided in Table 1 are useful in distinguishing benign pelvic masses from ovarian cancer.
  • certain of the described ovarian cancer biomarkers are useful alone for detecting and diagnosing ovarian cancer, methods are also described herein for the grouping of multiple subsets of the ovarian cancer biomarkers that are each useful as a panel of three or more biomarkers.
  • N biomarkers wherein N is at least two biomarkers.
  • N is selected to be any number from 2-42 biomarkers.
  • N is selected to be any number from 2-7, 2-10, 2-
  • N is selected to be any number from 3-7, 3-10, 3-15, 3-20, 3-25, 3-30, 3-35, 3-40, or 3-42. In other embodiments, N is selected to be any number from 4-7, 4-10, 4-15, 4-20, 4-25, 4-30, 4-35, 4-40, or 4-42. In other embodiments, N is selected to be any number from 5-7, 5-10, 5-15, 5-20, 5-25, 5-30, 5- 35, 5-40, or 5-42. In other embodiments, N is selected to be any number from 6-10, 6-15, 6- 20, 6-25, 6-30, 6-35, 6-40, or 6-42.
  • N is selected to be any number from 7-10, 7-15, 7-20, 7-25, 7-30, 7-35, 7-40, or 7-42. In other embodiments, N is selected to be any number from 8-10, 8-15, 8-20, 8-25, 8-30, 8-35, 8-40, or 8-42. In other embodiments, N is selected to be any number from 9-15, 9-20, 9-25, 9-30, 9-35, 9-40, or 9-42. In other embodiments, N is selected to be any number from 10-15, 10-20, 10-25, 10-30, 10-35, 10-40, or 10-42. It will be appreciated that N can be selected to encompass similar, but higher order, ranges.
  • a method for diagnosing ovarian cancer in an individual including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers provided in Table 1, wherein the individual is classified as having ovarian cancer based on the at least one biomarker value.
  • a method for diagnosing ovarian cancer in an individual including detecting, in a biological sample from an individual, biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers set forth in Table 1, wherein the likelihood of the individual having ovarian cancer is determined based on the biomarker values.
  • a method for differentiating an individual having a benign pelvic mass from an individual having ovarian cancer including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as having ovarian cancer, or the likelihood of the individual having ovarian cancer is determined, based on the at least one biomarker value.
  • a method for diagnosing that an individual does not have ovarian cancer including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as not having ovarian cancer based on the at least one biomarker value.
  • a method for diagnosing ovarian cancer including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of biomarkers selected from the group of panels set forth in Tables 2-14, wherein a classification of the biomarker values indicates that the individual has ovarian cancer.
  • a method for diagnosing an absence of ovarian cancer including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of biomarkers selected from the group of panels provided in Tables 2-14, wherein a classification of the biomarker values indicates an absence of ovarian cancer in the individual.
  • a computer-implemented method for indicating a likelihood of ovarian cancer.
  • the method comprises: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises biomarker values that each correspond to one of at least N biomarkers, wherein N is as defined above, selected from the group of biomarkers set forth in Table 1 ; performing with the computer a classification of each of the biomarker values; and indicating a likelihood that the individual has ovarian cancer based upon a plurality of classifications.
  • a computer-implemented method is provided for classifying an individual as either having or not having ovarian cancer.
  • the method comprises: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers provided in Table 1 ; performing with the computer a classification of each of the biomarker values; and indicating whether the individual has ovarian cancer based upon a plurality of classifications.
  • a computer program product for indicating a likelihood of ovarian cancer.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers, wherein N is as defined above, in the biological sample selected from the group of biomarkers set forth in Table 1 ; and code that executes a classification method that indicates a likelihood that the individual has ovarian cancer as a function of the biomarker values.
  • a computer program product for indicating an ovarian cancer status of an individual.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers in the biological sample selected from the group of biomarkers provided in Table 1; and code that executes a classification method that indicates an ovarian cancer status of the individual as a function of the biomarker values.
  • a computer-implemented method is provided for indicating a likelihood of ovarian cancer.
  • the method comprises retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers set forth in Table 1; performing with the computer a classification of the biomarker value; and indicating a likelihood that the individual has ovarian cancer based upon the classification.
  • a computer-implemented method is provided for classifying an individual as either having or not having ovarian cancer.
  • the method comprises retrieving, from a computer, biomarker information for an individual, wherein the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers provided in Table 1; performing with the computer a classification of the biomarker value; and indicating whether the individual has ovarian cancer based upon the classification.
  • a computer program product for indicating a likelihood of ovarian cancer.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers set forth in Table 1; and code that executes a classification method that indicates a likelihood that the individual has ovarian cancer as a function of the biomarker value.
  • a computer program product for indicating an ovarian cancer status of an individual.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers provided in Table 1; and code that executes a classification method that indicates an ovarian cancer status of the individual as a function of the biomarker value.
  • Figure IA is a flowchart for an exemplary method for detecting ovarian cancer in a biological sample.
  • Figure IB is a flowchart for an exemplary method for detecting ovarian cancer in a biological sample using a na ⁇ ve Bayes classification method.
  • Figure 2 shows a ROC curve for a single biomarker, BAFF Receptor, using a na ⁇ ve Bayes classifier for a test that detects ovarian cancer in women with pelvis masses.
  • Figure 3 shows ROC curves for biomarker panels of from one to ten biomarkers using na ⁇ ve Bayes classifiers for a test that detects ovarian cancer in women with pelvis masses.
  • Figure 4 illustrates the increase in the classification score (specificity + sensitivity) as the number of biomarkers is increased from one to ten using na ⁇ ve Bayes classification for an ovarian cancer panel.
  • Figure 5 shows the measured biomarker distributions for BAFF Receptor as a cumulative distribution function (cdf) in RFU for the benign control group (solid line) and the ovarian cancer disease group (dotted line) along with their curve fits to a normal cdf (dashed lines) used to train the na ⁇ ve Bayes classifiers.
  • cdf cumulative distribution function
  • Figure 6 illustrates an exemplary computer system for use with various computer- implemented methods described herein.
  • Figure 7 is a flowchart for a method of indicating the likelihood that an individual has ovarian cancer in accordance with one embodiment.
  • Figure 8 is a flowchart for a method of indicating the likelihood that an individual has ovarian cancer in accordance with one embodiment.
  • Figure 9 illustrates an exemplary aptamer assay that can be used to detect one or more ovarian cancer biomarkers in a biological sample.
  • Figure 10 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between ovarian cancer and benign pelvic masses from an aggregated set of potential biomarkers.
  • Figure 11 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between ovarian cancer and benign pelvic masses from a site-consistent set of potential biomarkers.
  • Figure 12 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between ovarian cancer and benign pelvic masses from a set of potential biomarkers resulting from a combination of aggregated and site-consistent markers.
  • Figure 13 shows gel images resulting from pull-down experiments that illustrate the specificity of aptamers as capture reagents for the proteins LBP, C9 and IgM.
  • lane 1 is the eluate from the Streptavidin-agarose beads
  • lane 2 is the final eluate
  • lane is a MW marker lane (major bands are at 110, 50, 30, 15, and 3.5 kDa from top to bottom).
  • Figure 14A shows a pair of histograms summarizing all possible single protein na ⁇ ve Bayes classifier scores (sensitivity + specificity) using the biomarkers set forth in Table
  • Figure 14B shows a pair of histograms summarizing all possible two-protein protein na ⁇ ve Bayes classifier scores (sensitivity + specificity) using the biomarkers set forth in Table 1 (solid) and a set of random non-markers (dotted).
  • Figure 14C shows a pair of histograms summarizing all possible three-protein na ⁇ ve Bayes classifier scores (sensitivity + specificity) using the biomarkers set forth in Table
  • Figure 15 shows the sensitivity + specificity score for na ⁇ ve Bayes classifiers using from 2-10 markers selected from the full panel (•) and the scores obtained by dropping the best 5 ( ⁇ ), 10 (A) and 15 ( ⁇ ) markers during classifier generation.
  • Figure 16A shows a set of ROC curves modeled from the data in Table 18 for panels of from one to five markers.
  • Figure 16B shows a set of ROC curves computed from the training data for panels of from one to five markers as in Figure 16A.
  • the term "about” represents an insignificant modification or variation of the numerical value such that the basic function of the item to which the numerical value relates is unchanged.
  • the present application includes biomarkers, methods, devices, reagents, systems, and kits for the detection and diagnosis of ovarian cancer.
  • one or more biomarkers are provided for use either alone or in various combinations to diagnose ovarian cancer, permit the differential diagnosis of pelvic masses as benign or malignant, monitor ovarian cancer recurrence, or address other clinical indications.
  • exemplary embodiments include the biomarkers provided in Table 1, which were identified using a multiplex aptamer-based assay, as described generally in Example 1 and more specifically in Example 2.
  • Table 1 sets forth the findings obtained from analyzing blood samples from
  • Table 1 lists the biomarkers found to be useful in distinguishing samples obtained from individuals with ovarian cancer from "control" samples obtained from individuals with benign pelvic masses. Using a multiplex aptamer assay, forty-two biomarkers were discovered that distinguished samples obtained from individuals with ovarian cancer from samples obtained from people who had benign pelvic masses (see Table
  • ovarian cancer biomarkers are useful alone for detecting and diagnosing ovarian cancer
  • methods are also described herein for the grouping of multiple subsets of the ovarian cancer biomarkers, where each grouping or subset selection is useful as a panel of three or more biomarkers, interchangeably referred to herein as a "biomarker panel" and a panel.
  • biomarker panel a panel of three or more biomarkers
  • various embodiments of the instant application provide combinations comprising N biomarkers, wherein N is at least two biomarkers. In other embodiments, N is selected from 2-42 biomarkers.
  • N is selected to be any number from 2-7, 2-10, 2-
  • N is selected to be any number from 3-7, 3-10, 3-15, 3-20, 3-25, 3-30, 3-35, 3-40, or 3-42. In other embodiments, N is selected to be any number from 4-7, 4-10, 4-15, 4-20, 4-25, 4-30, 4-35, 4-40, or 4-42. In other embodiments, N is selected to be any number from 5-7, 5-10, 5-15, 5-20, 5-25, 5-30, 5- 35, 5-40, or 5-42. In other embodiments, N is selected to be any number from 6-10, 6-15, 6- 20, 6-25, 6-30, 6-35, 6-40, or 6-42.
  • N is selected to be any number from 7-10, 7-15, 7-20, 7-25, 7-30, 7-35, 7-40, or 7-42. In other embodiments, N is selected to be any number from 8-10, 8-15, 8-20, 8-25, 8-30, 8-35, 8-40, or 8-42. In other embodiments, N is selected to be any number from 9-15, 9-20, 9-25, 9-30, 9-35, 9-40, or 9-42. In other embodiments, N is selected to be any number from 10-15, 10-20, 10-25, 10-30, 10-35, 10-40, or 10-42. It will be appreciated that N can be selected to encompass similar, but higher order, ranges.
  • the number of biomarkers useful for a biomarker subset or panel is based on the sensitivity and specificity value for the particular combination of biomarker values.
  • sensitivity and “specificity” are used herein with respect to the ability to correctly classify an individual, based on one or more biomarker values detected in their biological sample, as having ovarian cancer or not having ovarian cancer.
  • Sensitivity indicates the performance of the biomarker(s) with respect to correctly classifying individuals that have ovarian cancer.
  • Specificity indicates the performance of the biomarker(s) with respect to correctly classifying individuals who do not have ovarian cancer.
  • 85% specificity and 90% sensitivity for a panel of markers used to test a set of control samples and ovarian cancer samples indicates that 85% of the control samples were correctly classified as control samples by the panel, and 90% of the ovarian cancer samples were correctly classified as ovarian cancer samples by the panel.
  • the desired or preferred minimum value can be determined as described in Example 3.
  • Representative panels are set forth in Tables 2-14, which set forth a series of 100 different panels of 3-15 biomarkers, which have the indicated levels of specificity and sensitivity for each panel. The total number of occurrences of each marker in each of these panels is indicated at the bottom of each Table.
  • ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to at least one of the biomarkers SLPI, C9, HGF and RGM-C and at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarkers SLPI, C9, HGF and RGM-C and one of at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or 13.
  • ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker SLPI and one of at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker C9and one of at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker HGF and one of at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker RGM-C and one of at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • the ovarian cancer biomarkers identified herein represent a relatively large number of choices for subsets or panels of biomarkers that can be used to effectively detect or diagnose ovarian cancer. Selection of the desired number of such biomarkers depends on the specific combination of biomarkers chosen. It is important to remember that panels of biomarkers for detecting or diagnosing ovarian cancer may also include biomarkers not found in Table 1, and that the inclusion of additional biomarkers not found in Table 1 may reduce the number of biomarkers in the particular subset or panel that is selected from Table 1. The number of biomarkers from Table 1 used in a subset or panel may also be reduced if additional biomedical information is used in conjunction with the biomarker values to establish acceptable sensitivity and specificity values for a given assay.
  • biomarkers to be used in a subset or panel of biomarkers Another factor that can affect the number of biomarkers to be used in a subset or panel of biomarkers is the procedures used to obtain biological samples from individuals who are being evaluated for ovarian cancer. In a carefully controlled sample procurement environment, the number of biomarkers necessary to meet desired sensitivity and specificity values will be lower than in a situation where there can be more variation in sample collection, handling and storage. In developing the list of biomarkers set forth in Table 1, two sample collection sites were utilized to collect data for classifier training. [0092] One aspect of the instant application can be described generally with reference to Figures IA and B. A biological sample is obtained from an individual or individuals of interest.
  • the biological sample is then assayed to detect the presence of one or more (N) biomarkers of interest and to determine a biomarker value for each of said N biomarkers (referred to in Figure IB as marker RFU (relative fluorescence units)).
  • N one or more biomarkers of interest
  • RFU relative fluorescence units
  • Bio sample refers to any material, biological fluid, tissue, or cell obtained or otherwise derived from an individual.
  • a blood sample can be fractionated into serum or into fractions containing particular types of blood cells, such as red blood cells or white blood cells (leukocytes).
  • a sample can be a combination of samples from an individual, such as a combination of a tissue and fluid sample.
  • biological sample also includes materials containing homogenized solid material, such as from a stool sample, a tissue sample, or a tissue biopsy, for example.
  • biological sample also includes materials derived from a tissue culture or a cell culture.
  • any suitable methods for obtaining a biological sample can be employed; exemplary methods include, e.g., phlebotomy, swab (e.g., buccal swab), and a fine needle aspirate biopsy procedure. Samples can also be collected, e.g., by micro dissection (e.g., laser capture micro dissection (LCM) or laser micro dissection (LMD)), bladder wash, smear (e.g., a PAP smear), or ductal lavage.
  • a "biological sample” obtained or derived from an individual includes any such sample that has been processed in any suitable manner after being obtained from the individual.
  • a biological sample can be derived by taking biological samples from a number of individuals and pooling them or pooling an aliquot of each individual's biological sample.
  • the pooled sample can be treated as a sample from a single individual and if the presence of cancer is established in the pooled sample, then each individual biological sample can be re-tested to determine which individuals have ovarian cancer.
  • the phrase "data attributed to a biological sample from an individual” is intended to mean that the data in some form derived from, or were generated using, the biological sample of the individual.
  • the data may have been reformatted, revised, or mathematically altered to some degree after having been generated, such as by conversion from units in one measurement system to units in another measurement system; but, the data are understood to have been derived from, or were generated using, the biological sample.
  • Target refers to any molecule of interest that may be present in a biological sample.
  • a "molecule of interest” includes any minor variation of a particular molecule, such as, in the case of a protein, for example, minor variations in amino acid sequence, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component, which does not substantially alter the identity of the molecule.
  • a “target molecule”, “target”, or “analyte” is a set of copies of one type or species of molecule or multi-molecular structure.
  • Target molecules refer to more than one such set of molecules.
  • target molecules include proteins, polypeptides, nucleic acids, carbohydrates, lipids, polysaccharides, glycoproteins, hormones, receptors, antigens, antibodies, affybodies, antibody mimics, viruses, pathogens, toxic substances, substrates, metabolites, transition state analogs, cofactors, inhibitors, drugs, dyes, nutrients, growth factors, cells, tissues, and any fragment or portion of any of the foregoing.
  • polypeptide As used herein, “polypeptide,” “peptide,” and “protein” are used interchangeably herein to refer to polymers of amino acids of any length.
  • the polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids.
  • the terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component.
  • polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids, etc.
  • Polypeptides can be single chains or associated chains. Also included within the definition are preproteins and intact mature proteins; peptides or polypeptides derived from a mature protein; fragments of a protein; splice variants; recombinant forms of a protein; protein variants with amino acid modifications, deletions, or substitutions; digests; and post-translational modifications, such as glycosylation, acetylation, phosphorylation, and the like.
  • thrombin refers to thrombin, prothrombin, or both thrombin and prothrombin.
  • marker and “biomarker” are used interchangeably to refer to a target molecule that indicates or is a sign of a normal or abnormal process in an individual or of a disease or other condition in an individual. More specifically, a “marker” or “biomarker” is an anatomic, physiologic, biochemical, or molecular parameter associated with the presence of a specific physiological state or process, whether normal or abnormal, and, if abnormal, whether chronic or acute. Biomarkers are detectable and measurable by a variety of methods including laboratory assays and medical imaging.
  • a biomarker is a protein
  • biomarker value As used herein, “biomarker value”, “value”, “biomarker level”, and “level” are used interchangeably to refer to a measurement that is made using any analytical method for detecting the biomarker in a biological sample and that indicates the presence, absence, absolute amount or concentration, relative amount or concentration, titer, a level, an expression level, a ratio of measured levels, or the like, of, for, or corresponding to the biomarker in the biological sample.
  • the exact nature of the "value” or “level” depends on the specific design and components of the particular analytical method employed to detect the biomarker.
  • a biomarker indicates or is a sign of an abnormal process or a disease or other condition in an individual
  • that biomarker is generally described as being either over- expressed or under-expressed as compared to an expression level or value of the biomarker that indicates or is a sign of a normal process or an absence of a disease or other condition in an individual.
  • Up-regulation “up-regulated”, “over-expression”, “over-expressed”, and any variations thereof are used interchangeably to refer to a value or level of a biomarker in a biological sample that is greater than a value or level (or range of values or levels) of the biomarker that is typically detected in similar biological samples from healthy or normal individuals.
  • the terms may also refer to a value or level of a biomarker in a biological sample that is greater than a value or level (or range of values or levels) of the biomarker that may be detected at a different stage of a particular disease.
  • Down-regulation Down-regulated
  • under-expression under-expressed
  • any variations thereof are used interchangeably to refer to a value or level of a biomarker in a biological sample that is less than a value or level (or range of values or levels) of the biomarker that is typically detected in similar biological samples from healthy or normal individuals.
  • the terms may also refer to a value or level of a biomarker in a biological sample that is less than a value or level (or range of values or levels) of the biomarker that may be detected at a different stage of a particular disease.
  • a biomarker that is either over-expressed or under-expressed can also be referred to as being “differentially expressed” or as having a “differential level” or “differential value” as compared to a "normal” expression level or value of the biomarker that indicates or is a sign of a normal process or an absence of a disease or other condition in an individual.
  • "differential expression” of a biomarker can also be referred to as a variation from a "normal” expression level of the biomarker.
  • differential gene expression and “differential expression” are used interchangeably to refer to a gene (or its corresponding protein expression product) whose expression is activated to a higher or lower level in a subject suffering from a specific disease, relative to its expression in a normal or control subject.
  • the terms also include genes (or the corresponding protein expression products) whose expression is activated to a higher or lower level at different stages of the same disease. It is also understood that a differentially expressed gene may be either activated or inhibited at the nucleic acid level or protein level, or may be subject to alternative splicing to result in a different polypeptide product.
  • Differential gene expression may include a comparison of expression between two or more genes or their gene products; or a comparison of the ratios of the expression between two or more genes or their gene products; or even a comparison of two differently processed products of the same gene, which differ between normal subjects and subjects suffering from a disease; or between various stages of the same disease.
  • Differential expression includes both quantitative, as well as qualitative, differences in the temporal or cellular expression pattern in a gene or its expression products among, for example, normal and diseased cells, or among cells which have undergone different disease events or disease stages.
  • individual refers to a test subject or patient.
  • the individual can be a mammal or a non-mammal.
  • the individual is a mammal.
  • a mammalian individual can be a human or non-human.
  • the individual is a human.
  • a healthy or normal individual is an individual in which the disease or condition of interest (including, for example, ovarian diseases, ovarian -associated diseases, or other ovarian conditions) is not detectable by conventional diagnostic methods.
  • Diagnose refers to the detection, determination, or recognition of a health status or condition of an individual on the basis of one or more signs, symptoms, data, or other information pertaining to that individual.
  • the health status of an individual can be diagnosed as healthy / normal (i.e., a diagnosis of the absence of a disease or condition) or diagnosed as ill / abnormal (i.e., a diagnosis of the presence, or an assessment of the characteristics, of a disease or condition).
  • diagnosis encompass, with respect to a particular disease or condition, the initial detection of the disease; the characterization or classification of the disease; the detection of the progression, remission, or recurrence of the disease; and the detection of disease response after the administration of a treatment or therapy to the individual.
  • diagnosis of ovarian cancer includes distinguishing individuals who have cancer from individuals who do not. It further includes distinguishing benign pelvic masses from ovarian cancer.
  • Prognose refers to the prediction of a future course of a disease or condition in an individual who has the disease or condition (e.g., predicting patient survival), and such terms encompass the evaluation of disease response after the administration of a treatment or therapy to the individual.
  • "Evaluate”, “evaluating”, “evaluation”, and variations thereof encompass both
  • diagnose and "prognose” and also encompass determinations or predictions about the future course of a disease or condition in an individual who does not have the disease as well as determinations or predictions regarding the likelihood that a disease or condition will recur in an individual who apparently has been cured of the disease.
  • the term "evaluate” also encompasses assessing an individual's response to a therapy, such as, for example, predicting whether an individual is likely to respond favorably to a therapeutic agent or is unlikely to respond to a therapeutic agent (or will experience toxic or other undesirable side effects, for example), selecting a therapeutic agent for administration to an individual, or monitoring or determining an individual's response to a therapy that has been administered to the individual.
  • "evaluating" ovarian cancer can include, for example, any of the following: prognosing the future course of ovarian cancer in an individual; predicting the recurrence of ovarian cancer in an individual who apparently has been cured of ovarian cancer; or determining or predicting an individual's response to an ovarian cancer treatment or selecting an ovarian cancer treatment to administer to an individual based upon a determination of the biomarker values derived from the individual's biological sample.
  • "evaluating" ovarian cancer initially detecting the presence or absence of ovarian cancer; determining a specific stage, type or sub-type, or other classification or characteristic of ovarian cancer; determining whether a pelvic mass is benign or malignant; or detecting or monitoring ovarian cancer progression (e.g., monitoring ovarian tumor growth or metastatic spread), remission, or recurrence.
  • additional biomedical information refers to one or more evaluations of an individual, other than using any of the biomarkers described herein, that are associated with ovarian cancer risk.
  • Additional biomedical information includes any of the following: physical descriptors of an individual; physical descriptors of a pelvic mass observed by MRI, abdominal ultrasound, or CT imaging; the height and/or weight of an individual; change in weight; the ethnicity of an individual; occupational history; family history of ovarian cancer (or other cancer); the presence of a genetic marker(s) correlating with a higher risk of ovarian cancer in the individual or a family member; the presence of a pelvic mass; size of mass; location of mass; morphology of mass and associated pelvic region (e.g., as observed through imaging); clinical symptoms such as bloating, abdominal pain, or sudden weight gain or loss; and the like.
  • Additional biomedical information can be obtained from an individual using routine techniques known in the art, such as from the individual themselves by use of a routine patient questionnaire or health history questionnaire, etc., or from a medical practitioner, etc.
  • additional biomedical information can be obtained from routine imaging techniques, including abdominal or transvaginal ultrasound, MRI, CT imaging, and PET-CT.
  • Testing of biomarker levels in combination with an evaluation of any additional biomedical information, including other laboratory tests (e.g., CA- 125 testing) may, for example, improve sensitivity, specificity, and/or AUC for detecting ovarian cancer (or other ovarian cancer-related uses) as compared to biomarker testing alone or evaluating any particular item of additional biomedical information alone (e.g., ultrasound imaging alone).
  • AUC area under the curve
  • ROC receiver operating characteristic
  • the feature data across the entire population e.g., the cases and controls
  • the true positive and false positive rates for the data are calculated.
  • the true positive rate is determined by counting the number of cases above the value for that feature and then dividing by the total number of cases.
  • the false positive rate is determined by counting the number of controls above the value for that feature and then dividing by the total number of controls.
  • ROC curves can be generated for a single feature as well as for other single outputs, for example, a combination of two or more features can be mathematically combined (e.g., added, subtracted, multiplied, etc.) to provide a single sum value, and this single sum value can be plotted in a ROC curve. Additionally, any combination of multiple features, in which the combination derives a single output value, can be plotted in a ROC curve. These combinations of features may comprise a test.
  • the ROC curve is the plot of the true positive rate (sensitivity) of a test against the false positive rate (1 -specificity) of the test.
  • detecting or “determining” with respect to a biomarker value includes the use of both the instrument required to observe and record a signal corresponding to a biomarker value and the material/s required to generate that signal.
  • the biomarker value is detected using any suitable method, including fluorescence, chemiluminescence, surface plasmon resonance, surface acoustic waves, mass spectrometry, infrared spectroscopy, Raman spectroscopy, atomic force microscopy, scanning tunneling microscopy, electrochemical detection methods, nuclear magnetic resonance, quantum dots, and the like.
  • Solid support refers herein to any substrate having a surface to which molecules may be attached, directly or indirectly, through either covalent or non-covalent bonds.
  • a “solid support” can have a variety of physical formats, which can include, for example, a membrane; a chip (e.g., a protein chip); a slide (e.g., a glass slide or coverslip); a column; a hollow, solid, semi-solid, pore- or cavity- containing particle, such as, for example, a bead; a gel; a fiber, including a fiber optic material; a matrix; and a sample receptacle.
  • Exemplary sample receptacles include sample wells, tubes, capillaries, vials, and any other vessel, groove or indentation capable of holding a sample.
  • a sample receptacle can be contained on a multi-sample platform, such as a microtiter plate, slide, microfluidics device, and the like.
  • a support can be composed of a natural or synthetic material, an organic or inorganic material. The composition of the solid support on which capture reagents are attached generally depends on the method of attachment (e.g., covalent attachment).
  • Other exemplary receptacles include microdroplets and microfluidic controlled or bulk oil/aqueous emulsions within which assays and related manipulations can occur.
  • Suitable solid supports include, for example, plastics, resins, polysaccharides, silica or silica-based materials, functionalized glass, modified silicon, carbon, metals, inorganic glasses, membranes, nylon, natural fibers (such as, for example, silk, wool and cotton), polymers, and the like.
  • the material composing the solid support can include reactive groups such as, for example, carboxy, amino, or hydroxyl groups, which are used for attachment of the capture reagents.
  • Polymeric solid supports can include, e.g., polystyrene, polyethylene glycol tetraphthalate, polyvinyl acetate, polyvinyl chloride, polyvinyl pyrrolidone, polyacrylonitrile, polymethyl methacrylate, polytetrafluoroethylene, butyl rubber, styrenebutadiene rubber, natural rubber, polyethylene, polypropylene, (poly)tetrafluoroethylene, (poly)vinylidenefluoride, polycarbonate, and polymethylpentene.
  • Suitable solid support particles that can be used include, e.g., encoded particles, such as Luminex ® -type encoded particles, magnetic particles, and glass particles.
  • methods are provided for diagnosing ovarian cancer in an individual by detecting one or more biomarker values corresponding to one or more biomarkers that are present in the circulation of an individual, such as in serum or plasma, by any number of analytical methods, including any of the analytical methods described herein.
  • biomarkers are, for example, differentially expressed in individuals with ovarian cancer as compared to individuals without ovarian cancer.
  • Detection of the differential expression of a biomarker in an individual can be used, for example, to permit the early diagnosis of ovarian cancer, to distinguish between a benign pelvic mass and ovarian cancer (such as, for example, a mass observed on an abdominal ultrasound or computed tomography (CT) scan), to monitor ovarian cancer recurrence, or for other clinical indications.
  • a benign pelvic mass and ovarian cancer such as, for example, a mass observed on an abdominal ultrasound or computed tomography (CT) scan
  • CT computed tomography
  • any of the biomarkers described herein may be used in a variety of clinical indications for ovarian cancer, including any of the following: detection of ovarian cancer (such as in a high-risk individual or population); characterizing ovarian cancer (e.g., determining ovarian cancer type, sub-type, or stage), such as by determining whether a pelvic mass is benign or malignant; determining ovarian cancer prognosis; monitoring ovarian cancer progression or remission; monitoring for ovarian cancer recurrence; monitoring metastasis; treatment selection (e.g., pre- or post-operative chemotherapy selection); monitoring response to a therapeutic agent or other treatment; combining biomarker testing with additional biomedical information, such as CA- 125 level, the presence of a genetic marker(s) indicating a higher risk for ovarian cancer, etc., or with mass size, morphology, presence of ascites, etc.
  • additional biomedical information such as CA- 125 level, the presence of a genetic marker(s) indicating
  • Biomarker testing may improve positive predictive value (PPV) over CA- 125 testing and imaging alone.
  • the described biomarkers may also be useful in permitting certain of these uses before indications of ovarian cancer are detected by imaging modalities or other clinical correlates, or before symptoms appear.
  • differential expression of one or more of the described biomarkers in an individual who is not known to have ovarian cancer may indicate that the individual has ovarian cancer, thereby enabling detection of ovarian cancer at an early stage of the disease when treatment is most effective, perhaps before the ovarian cancer is detected by other means or before symptoms appear.
  • Increased differential expression from "normal” (since some biomarkers may be down-regulated with disease) of one or more of the biomarkers during the course of ovarian cancer may be indicative of ovarian cancer progression, e.g., an ovarian tumor is growing and/or metastasizing (and thus indicate a poor prognosis), whereas a decrease in the degree to which one or more of the biomarkers is differentially expressed (i.e., in subsequent biomarker tests, the expression level in the individual is moving toward or approaching a "normal” expression level) may be indicative of ovarian cancer remission, e.g., an ovarian tumor is shrinking (and thus indicate a good or better prognosis).
  • an increase in the degree to which one or more of the biomarkers is differentially expressed may indicate that the ovarian cancer is progressing and therefore indicate that the treatment is ineffective
  • a decrease in differential expression of one or more of the biomarkers during the course of ovarian cancer treatment may be indicative of ovarian cancer remission and therefore indicate that the treatment is working successfully.
  • an increase or decrease in the differential expression of one or more of the biomarkers after an individual has apparently been cured of ovarian cancer may be indicative of ovarian cancer recurrence.
  • the individual can be re- started on therapy (or the therapeutic regimen modified such as to increase dosage amount and/or frequency, if the individual has maintained therapy) at an earlier stage than if the recurrence of ovarian cancer was not detected until later.
  • a differential expression level of one or more of the biomarkers in an individual may be predictive of the individual's response to a particular therapeutic agent.
  • changes in the biomarker expression levels may indicate the need for repeat imaging, such as to determine ovarian cancer activity or to determine the need for changes in treatment.
  • Detection of any of the biomarkers described herein may be particularly useful following, or in conjunction with, ovarian cancer treatment, such as to evaluate the success of the treatment or to monitor ovarian cancer remission, recurrence, and/or progression (including metastasis) following treatment.
  • Ovarian cancer treatment may include, for example, administration of a therapeutic agent to the individual, performance of surgery (e.g., surgical resection of at least a portion of a pelvic mass), administration of radiation therapy, or any other type of ovarian cancer treatment used in the art, and any combination of these treatments.
  • any of the biomarkers may be detected at least once after treatment or may be detected multiple times after treatment (such as at periodic intervals), or may be detected both before and after treatment.
  • Differential expression levels of any of the biomarkers in an individual over time may be indicative of ovarian cancer progression, remission, or recurrence, examples of which include any of the following: an increase or decrease in the expression level of the biomarkers after treatment compared with the expression level of the biomarker before treatment; an increase or decrease in the expression level of the biomarker at a later time point after treatment compared with the expression level of the biomarker at an earlier time point after treatment; and a differential expression level of the biomarker at a single time point after treatment compared with normal levels of the biomarker.
  • the biomarker levels for any of the biomarkers described herein can be determined in pre-surgery and post-surgery (e.g., 2-8 weeks after surgery) serum or plasma samples.
  • An increase in the biomarker expression level(s) in the post-surgery sample compared with the pre-surgery sample can indicate progression of ovarian cancer (e.g., unsuccessful surgery), whereas a decrease in the biomarker expression level(s) in the post-surgery sample compared with the pre-surgery sample can indicate regression of ovarian cancer (e.g., the surgery successfully removed the ovarian tumor).
  • Similar analyses of the biomarker levels can be carried out before and after other forms of treatment, such as before and after radiation therapy or administration of a therapeutic agent or cancer vaccine.
  • biomarker levels can also be done in conjunction with determination of SNPs or other genetic lesions or variability that are indicative of increased risk of susceptibility of disease. (See, e.g., Amos et al., Nature Genetics 40, 616-622 (2009)).
  • biomarker levels can also be done in conjunction with relevant symptoms or abdominal ultrasound and CT imaging.
  • Detection of any of the biomarkers described herein may be useful after a pelvic mass has been observed through imaging to aid in the diagnosis of ovarian cancer and guide appropriate clinical care of the individual, including care by an appropriate surgical specialist.
  • biomarkers In addition to testing biomarker levels in conjunction with relevant symptoms or abdominal ultrasound or CT imaging, information regarding the biomarkers can also be evaluated in conjunction with other types of data, particularly data that indicates an individual's risk for ovarian cancer (e.g., patient clinical history, symptoms, family history of cancer, risk factors such as the presence of a genetic marker(s), and/or status of other biomarkers, etc.). These various data can be assessed by automated methods, such as a computer program/software, which can be embodied in a computer or other apparatus/device. [00123] Any of the described biomarkers may also be used in imaging tests.
  • an imaging agent can be coupled to any of the described biomarkers, which can be used to aid in ovarian cancer diagnosis, to monitor disease progression/remission or metastasis, to monitor for disease recurrence, or to monitor response to therapy, among other uses.
  • a biomarker value for the biomarkers described herein can be detected using any of a variety of known analytical methods.
  • a biomarker value is detected using a capture reagent.
  • a capture agent or “capture reagent” refers to a molecule that is capable of binding specifically to a biomarker.
  • the capture reagent can be exposed to the biomarker in solution or can be exposed to the biomarker while the capture reagent is immobilized on a solid support.
  • the capture reagent contains a feature that is reactive with a secondary feature on a solid support.
  • the capture reagent can be exposed to the biomarker in solution, and then the feature on the capture reagent can be used in conjunction with the secondary feature on the solid support to immobilize the biomarker on the solid support.
  • the capture reagent is selected based on the type of analysis to be conducted.
  • Capture reagents include but are not limited to aptamers, antibodies, adnectins, ankyrins, other antibody mimetics and other protein scaffolds, autoantibodies, chimeras, small molecules, an F(ab') 2 fragment, a single chain antibody fragment, an Fv fragment, a single chain Fv fragment, a nucleic acid, a lectin, a ligand-binding receptor, affybodies, nanobodies, imprinted polymers, avimers, peptidomimetics, a hormone receptor, a cytokine receptor, and synthetic receptors, and modifications and fragments of these.
  • a biomarker value is detected using a biomarker/capture reagent complex.
  • the biomarker value is derived from the biomarker/capture reagent complex and is detected indirectly, such as, for example, as a result of a reaction that is subsequent to the biomarker/capture reagent interaction, but is dependent on the formation of the biomarker/capture reagent complex.
  • the biomarker value is detected directly from the biomarker in a biological sample.
  • the biomarkers are detected using a multiplexed format that allows for the simultaneous detection of two or more biomarkers in a biological sample.
  • capture reagents are immobilized, directly or indirectly, covalently or non-covalently, in discrete locations on a solid support.
  • a multiplexed format uses discrete solid supports where each solid support has a unique capture reagent associated with that solid support, such as, for example quantum dots.
  • an individual device is used for the detection of each one of multiple biomarkers to be detected in a biological sample.
  • Individual devices can be configured to permit each biomarker in the biological sample to be processed simultaneously.
  • a microtiter plate can be used such that each well in the plate is used to uniquely analyze one of multiple biomarkers to be detected in a biological sample.
  • a fluorescent tag can be used to label a component of the biomarker/capture complex to enable the detection of the biomarker value.
  • the fluorescent label can be conjugated to a capture reagent specific to any of the biomarkers described herein using known techniques, and the fluorescent label can then be used to detect the corresponding biomarker value.
  • Suitable fluorescent labels include rare earth chelates, fluorescein and its derivatives, rhodamine and its derivatives, dansyl, allophycocyanin, PBXL-3, Qdot 605, Lissamine, phycoerythrin, Texas
  • the fluorescent label is a fluorescent dye molecule.
  • the fluorescent dye molecule includes at least one substituted indolium ring system in which the substituent on the 3-carbon of the indolium ring contains a chemically reactive group or a conjugated substance.
  • the dye molecule includes an AlexFluor molecule, such as, for example, AlexaFluor 488, AlexaFluor 532, AlexaFluor 647, AlexaFluor 680, or AlexaFluor 700.
  • the dye molecule includes a first type and a second type of dye molecule, such as, e.g., two different AlexaFluor molecules.
  • the dye molecule includes a first type and a second type of dye molecule, and the two dye molecules have different emission spectra.
  • Fluorescence can be measured with a variety of instrumentation compatible with a wide range of assay formats. For example, spectrofluorimeters have been designed to analyze microtiter plates, microscope slides, printed arrays, cuvettes, etc. See Principles of Fluorescence Spectroscopy, by J. R. Lakowicz, Springer Science + Business Media, Inc., 2004. See Bioluminescence & Chemiluminescence: Progress & Current Applications; Philip E. Stanley and Larry J. Kricka editors, World Scientific Publishing Company, January 2002.
  • a chemiluminescence tag can optionally be used to label a component of the biomarker/capture complex to enable the detection of a biomarker value.
  • Suitable chemiluminescent materials include any of oxalyl chloride, Rodamin 6G, Ru(bipy) 3 2+ , TMAE (tetrakis(dimethylamino)ethylene), Pyrogallol (1,2,3-trihydroxibenzene), Lucigenin, peroxyoxalates, Aryl oxalates, Acridinium esters, dioxetanes, and others.
  • the detection method includes an enzyme/substrate combination that generates a detectable signal that corresponds to the biomarker value.
  • the enzyme catalyzes a chemical alteration of the chromogenic substrate which can be measured using various techniques, including spectrophotometry, fluorescence, and chemiluminescence.
  • Suitable enzymes include, for example, luciferases, luciferin, malate dehydrogenase, urease, horseradish peroxidase (HRPO), alkaline phosphatase, beta- galactosidase, glucoamylase, lysozyme, glucose oxidase, galactose oxidase, and glucose-6- phosphate dehydrogenase, uricase, xanthine oxidase, lactoperoxidase, microperoxidase, and the like.
  • HRPO horseradish peroxidase
  • alkaline phosphatase beta- galactosidase
  • glucoamylase lysozyme
  • glucose oxidase galactose oxidase
  • glucose-6- phosphate dehydrogenase uricase
  • xanthine oxidase lactoperoxidase
  • microperoxidase and the like
  • the detection method can be a combination of fluorescence, chemiluminescence, radionuclide or enzyme/substrate combinations that generate a measurable signal.
  • Multimodal signaling could have unique and advantageous characteristics in biomarker assay formats.
  • biomarker values for the biomarkers described herein can be detected using known analytical methods including, singleplex aptamer assays, multiplexed aptamer assays, singleplex or multiplexed immunoassays, mRNA expression profiling, miRNA expression profiling, mass spectrometric analysis, histological/cytological methods, etc. as detailed below.
  • Assays directed to the detection and quantification of physiologically significant molecules in biological samples and other samples are important tools in scientific research and in the health care field.
  • One class of such assays involves the use of a microarray that includes one or more aptamers immobilized on a solid support.
  • the aptamers are each capable of binding to a target molecule in a highly specific manner and with very high affinity. See, e.g., U.S. Patent No. 5,475,096 entitled "Nucleic Acid Ligands"; see also, e.g., U.S. Patent No. 6,242,246, U.S. Patent No. 6,458,543, and U.S. Patent No.
  • an "aptamer” refers to a nucleic acid that has a specific binding affinity for a target molecule. It is recognized that affinity interactions are a matter of degree; however, in this context, the "specific binding affinity" of an aptamer for its target means that the aptamer binds to its target generally with a much higher degree of affinity than it binds to other components in a test sample.
  • An “aptamer” is a set of copies of one type or species of nucleic acid molecule that has a particular nucleotide sequence.
  • An aptamer can include any suitable number of nucleotides, including any number of chemically modified nucleotides. "Aptamers" refers to more than one such set of molecules.
  • aptamers can have either the same or different numbers of nucleotides.
  • Aptamers can be DNA or RNA or chemically modified nucleic acids and can be single stranded, double stranded, or contain double stranded regions, and can include higher ordered structures.
  • An aptamer can also be a photoaptamer, where a photoreactive or chemically reactive functional group is included in the aptamer to allow it to be covalently linked to its corresponding target. Any of the aptamer methods disclosed herein can include the use of two or more aptamers that specifically bind the same target molecule. As further described below, an aptamer may include a tag.
  • an aptamer can be identified using any known method, including the SELEX process. Once identified, an aptamer can be prepared or synthesized in accordance with any known method, including chemical synthetic methods and enzymatic synthetic methods.
  • SELEX and “SELEX process” are used interchangeably herein to refer generally to a combination of (1) the selection of aptamers that interact with a target molecule in a desirable manner, for example binding with high affinity to a protein, with (2) the amplification of those selected nucleic acids.
  • the SELEX process can be used to identify aptamers with high affinity to a specific target or biomarker.
  • SELEX generally includes preparing a candidate mixture of nucleic acids, binding of the candidate mixture to the desired target molecule to form an affinity complex, separating the affinity complexes from the unbound candidate nucleic acids, separating and isolating the nucleic acid from the affinity complex, purifying the nucleic acid, and identifying a specific aptamer sequence.
  • the process may include multiple rounds to further refine the affinity of the selected aptamer.
  • the process can include amplification steps at one or more points in the process. See, e.g., U.S. Patent No. 5,475,096, entitled "Nucleic Acid Ligands".
  • the SELEX process can be used to generate an aptamer that covalently binds its target as well as an aptamer that non-covalently binds its target. See, e.g., U.S. Patent No. 5,705,337 entitled “Systematic Evolution of Nucleic Acid Ligands by Exponential Enrichment: Chemi-SELEX.”
  • the SELEX process can be used to identify high-affinity aptamers containing modified nucleotides that confer improved characteristics on the aptamer, such as, for example, improved in vivo stability or improved delivery characteristics. Examples of such modifications include chemical substitutions at the ribose and/or phosphate and/or base positions. SELEX process -identified aptamers containing modified nucleotides are described in U.S. Patent No. 5,660,985, entitled "High Affinity Nucleic Acid Ligands Containing Modified Nucleotides", which describes oligonucleotides containing nucleotide derivatives chemically modified at the 5'- and 2'-positions of pyrimidines. U.S. Patent No.
  • SELEX can also be used to identify aptamers that have desirable off -rate characteristics. See U.S. Patent Application Publication 20090004667, entitled “Method for Generating Aptamers with Improved Off-Rates", which describes improved SELEX methods for generating aptamers that can bind to target molecules. Methods for producing aptamers and photoaptamers having slower rates of dissociation from their respective target molecules are described.
  • the methods involve contacting the candidate mixture with the target molecule, allowing the formation of nucleic acid-target complexes to occur, and performing a slow off -rate enrichment process wherein nucleic acid- target complexes with fast dissociation rates will dissociate and not reform, while complexes with slow dissociation rates will remain intact. Additionally, the methods include the use of modified nucleotides in the production of candidate nucleic acid mixtures to generate aptamers with improved off -rate performance. [00143] A variation of this assay employs aptamers that include photoreactive functional groups that enable the aptamers to covalently bind or "photocrosslink" their target molecules. See, e.g., U.S. Patent No.
  • the photoaptamers are photoactivated, and the solid support is washed to remove any non-specifically bound molecules. Harsh wash conditions may be used, since target molecules that are bound to the photoaptamers are generally not removed, due to the covalent bonds created by the photoactivated functional group(s) on the photoaptamers. In this manner, the assay enables the detection of a biomarker value corresponding to a biomarker in the test sample.
  • the aptamers are immobilized on the solid support prior to being contacted with the sample. Under certain circumstances, however, immobilization of the aptamers prior to contact with the sample may not provide an optimal assay. For example, pre-immobilization of the aptamers may result in inefficient mixing of the aptamers with the target molecules on the surface of the solid support, perhaps leading to lengthy reaction times and, therefore, extended incubation periods to permit efficient binding of the aptamers to their target molecules. Further, when photoaptamers are employed in the assay and depending upon the material utilized as a solid support, the solid support may tend to scatter or absorb the light used to effect the formation of covalent bonds between the photoaptamers and their target molecules.
  • immobilization of the aptamers on the solid support generally involves an aptamer-preparation step (i.e., the immobilization) prior to exposure of the aptamers to the sample, and this preparation step may affect the activity or functionality of the aptamers.
  • Patent Application Publication 20090042206 entitled “Multiplexed Analyses of Test Samples”).
  • the described aptamer assay methods enable the detection and quantification of a non- nucleic acid target (e.g., a protein target) in a test sample by detecting and quantifying a nucleic acid (i.e., an aptamer).
  • the described methods create a nucleic acid surrogate (i.e, the aptamer) for detecting and quantifying a non-nucleic acid target, thus allowing the wide variety of nucleic acid technologies, including amplification, to be applied to a broader range of desired targets, including protein targets.
  • Aptamers can be constructed to facilitate the separation of the assay components from an aptamer biomarker complex (or photoaptamer biomarker covalent complex) and permit isolation of the aptamer for detection and/or quantification.
  • these constructs can include a cleavable or releasable element within the aptamer sequence.
  • additional functionality can be introduced into the aptamer, for example, a labeled or detectable component, a spacer component, or a specific binding tag or immobilization element.
  • the aptamer can include a tag connected to the aptamer via a cleavable moiety, a label, a spacer component separating the label, and the cleavable moiety.
  • a cleavable element is a photocleavable linker.
  • the photocleavable linker can be attached to a biotin moiety and a spacer section, can include an NHS group for derivatization of amines, and can be used to introduce a biotin group to an aptamer, thereby allowing for the release of the aptamer later in an assay method.
  • a method for signal generation takes advantage of anisotropy signal change due to the interaction of a fluorophore-labeled capture reagent with its specific biomarker target.
  • binding events may be used to quantitatively measure the biomarkers in solutions.
  • Other methods include fluorescence polarization assays, molecular beacon methods, time resolved fluorescence quenching, chemiluminescence, fluorescence resonance energy transfer, and the like.
  • An exemplary solution-based aptamer assay that can be used to detect a biomarker value corresponding to a biomarker in a biological sample includes the following: (a) preparing a mixture by contacting the biological sample with an aptamer that includes a first tag and has a specific affinity for the biomarker, wherein an aptamer affinity complex is formed when the biomarker is present in the sample; (b) exposing the mixture to a first solid support including a first capture element, and allowing the first tag to associate with the first capture element; (c) removing any components of the mixture not associated with the first solid support; (d) attaching a second tag to the biomarker component of the aptamer affinity complex; (e) releasing the aptamer affinity complex from the first solid support; (f) exposing the released aptamer affinity complex to a second solid support that includes a second capture element and allowing the second tag to associate with the second capture element; (g) removing any non-complexed aptamer
  • Immunoassay methods are based on the reaction of an antibody to its corresponding target or analyte and can detect the analyte in a sample depending on the specific assay format.
  • monoclonal antibodies are often used because of their specific epitope recognition.
  • Polyclonal antibodies have also been successfully used in various immunoassays because of their increased affinity for the target as compared to monoclonal antibodies.
  • Immunoassays have been designed for use with a wide range of biological sample matrices. Immunoassay formats have been designed to provide qualitative, semiquantitative, and quantitative results.
  • Quantitative results are generated through the use of a standard curve created with known concentrations of the specific analyte to be detected.
  • the response or signal from an unknown sample is plotted onto the standard curve, and a quantity or value corresponding to the target in the unknown sample is established.
  • ELISA or EIA can be quantitative for the detection of an analyte. This method relies on attachment of a label to either the analyte or the antibody and the label component includes, either directly or indirectly, an enzyme. ELISA tests may be formatted for direct, indirect, competitive, or sandwich detection of the analyte. Other methods rely on labels such as, for example, radioisotopes (I 125 ) or fluorescence.
  • Additional techniques include, for example, agglutination, nephelometry, turbidimetry, Western blot, immunoprecipitation, immunocytochemistry, immunohistochemistry, flow cytometry, Luminex assay, and others (see ImmunoAssay: A Practical Guide, edited by Brian Law, published by Taylor & Francis, Ltd., 2005 edition).
  • Exemplary assay formats include enzyme-linked immunosorbent assay
  • ELISA radioimmunoassay
  • FRET fluorescence resonance energy transfer
  • TR-FRET time resolved- FRET
  • biomarkers include biomarker immunoprecipitation followed by quantitative methods that allow size and peptide level discrimination, such as gel electrophoresis, capillary electrophoresis, planar electrochromatography, and the like.
  • Methods of detecting and/or quantifying a detectable label or signal generating material depend on the nature of the label.
  • the products of reactions catalyzed by appropriate enzymes can be, without limitation, fluorescent, luminescent, or radioactive or they may absorb visible or ultraviolet light.
  • detectors suitable for detecting such detectable labels include, without limitation, x-ray film, radioactivity counters, scintillation counters, spectrophotometers, colorimeters, fluorometers, luminometers, and densitometers.
  • Any of the methods for detection can be performed in any format that allows for any suitable preparation, processing, and analysis of the reactions. This can be, for example, in multi-well assay plates (e.g., 96 wells or 384 wells) or using any suitable array or microarray. Stock solutions for various agents can be made manually or robotically, and all subsequent pipetting, diluting, mixing, distribution, washing, incubating, sample readout, data collection and analysis can be done robotically using commercially available analysis software, robotics, and detection instrumentation capable of detecting a detectable label.
  • Measuring mRNA in a biological sample may be used as a surrogate for detection of the level of the corresponding protein in the biological sample.
  • any of the biomarkers or biomarker panels described herein can also be detected by detecting the appropriate RNA.
  • mRNA expression levels are measured by reverse transcription quantitative polymerase chain reaction (RT-PCR followed with qPCR).
  • RT-PCR is used to create a cDNA from the mRNA.
  • the cDNA may be used in a qPCR assay to produce fluorescence as the DNA amplification process progresses. By comparison to a standard curve, qPCR can produce an absolute measurement such as number of copies of mRNA per cell.
  • Northern blots, microarrays, Invader assays, and RT-PCR combined with capillary electrophoresis have all been used to measure expression levels of mRNA in a sample. See Gene Expression Profiling: Methods and Protocols, Richard A. Shimkets, editor, Humana Press, 2004.
  • miRNA molecules are small RNAs that are non-coding but may regulate gene expression. Any of the methods suited to the measurement of mRNA expression levels can also be used for the corresponding miRNA. Recently many laboratories have investigated the use of miRNAs as biomarkers for disease. Many diseases involve wide-spread transcriptional regulation, and it is not surprising that miRNAs might find a role as biomarkers. The connection between miRNA concentrations and disease is often even less clear than the connections between protein levels and disease, yet the value of miRNA biomarkers might be substantial.
  • RNA biomarkers have similar requirements, although many potential protein biomarkers are secreted intentionally at the site of pathology and function, during disease, in a paracrine fashion. Many potential protein biomarkers are designed to function outside the cells within which those proteins are synthesized.
  • any of the described biomarkers may also be used in molecular imaging tests.
  • an imaging agent can be coupled to any of the described biomarkers, which can be used to aid in ovarian cancer diagnosis, to monitor disease progression/remission or metastasis, to monitor for disease recurrence, or to monitor response to therapy, among other uses.
  • In vivo imaging technologies provide non-invasive methods for determining the state of a particular disease in the body of an individual. For example, entire portions of the body, or even the entire body, may be viewed as a three dimensional image, thereby providing valuable information concerning morphology and structures in the body. Such technologies may be combined with the detection of the biomarkers described herein to provide information concerning the cancer status, in particular the ovarian cancer status, of an individual.
  • the contrast agent may be bound to or associated with a capture reagent, such as an aptamer or an antibody, for example, and/or with a peptide or protein, or an oligonucleotide (for example, for the detection of gene expression), or a complex containing any of these with one or more macro molecules and/or other particulate forms.
  • a capture reagent such as an aptamer or an antibody, for example, and/or with a peptide or protein, or an oligonucleotide (for example, for the detection of gene expression), or a complex containing any of these with one or more macro molecules and/or other particulate forms.
  • the contrast agent may also feature a radioactive atom that is useful in imaging. Suitable radioactive atoms include technetium-99m or iodine- 123 for scintigraphic studies.
  • MRI magnetic resonance imaging
  • iodine- 123 again, iodine-131, indium-I l l, fluorine-19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron.
  • MRI magnetic resonance imaging
  • Standard imaging techniques include but are not limited to magnetic resonance imaging, contrast-enhanced abdominal or transvaginal ultrasound, computed tomography (CT) scanning, positron emission tomography (PET), single photon emission computed tomography (SPECT), and the like.
  • CT computed tomography
  • PET positron emission tomography
  • SPECT single photon emission computed tomography
  • the type of detection instrument available is a major factor in selecting a given contrast agent, such as a given radionuclide and the particular biomarker that it is used to target (protein, mRNA, and the like).
  • the radionuclide chosen typically has a type of decay that is detectable by a given type of instrument.
  • its half-life should be long enough to enable detection at the time of maximum uptake by the target tissue but short enough that deleterious radiation of the host is minimized.
  • Exemplary imaging techniques include but are not limited to PET and SPECT, which are imaging techniques in which a radionuclide is synthetically or locally administered to an individual. The subsequent uptake of the radiotracer is measured over time and used to obtain information about the targeted tissue and the biomarker. Because of the high-energy (gamma-ray) emissions of the specific isotopes employed and the sensitivity and sophistication of the instruments used to detect them, the two-dimensional distribution of radioactivity may be inferred from outside of the body.
  • PET and SPECT are imaging techniques in which a radionuclide is synthetically or locally administered to an individual. The subsequent uptake of the radiotracer is measured over time and used to obtain information about the targeted tissue and the biomarker. Because of the high-energy (gamma-ray) emissions of the specific isotopes employed and the sensitivity and sophistication of the instruments used to detect them, the two-dimensional distribution of radioactivity may be inferred from outside of the body.
  • Commonly used positron-emitting nuclides in PET include, for example, carbon-11, nitrogen-13, oxygen-15, and fluorine-18.
  • Isotopes that decay by electron capture and/or gamma-emission are used in SPECT and include, for example iodine- 123 and technetium-99m.
  • An exemplary method for labeling amino acids with technetium-99m is the reduction of pertechnetate ion in the presence of a chelating precursor to form the labile technetium-99m-precursor complex, which, in turn, reacts with the metal binding group of a bifunctionally modified chemotactic peptide to form a technetium-99m-chemotactic peptide conjugate.
  • Antibodies are frequently used for such in vivo imaging diagnostic methods.
  • Labeled antibodies which specifically bind any of the biomarkers in Table 1 can be injected into an individual suspected of having a certain type of cancer (e.g., ovarian cancer), detectable according to the particular biomarker used, for the purpose of diagnosing or evaluating the disease status of the individual.
  • the label used will be selected in accordance with the imaging modality to be used, as previously described. Localization of the label permits determination of the spread of the cancer. The amount of label within an organ or tissue also allows determination of the presence or absence of cancer in that organ or tissue.
  • aptamers may be used for such in vivo imaging diagnostic methods.
  • an aptamer that was used to identify a particular biomarker described in Table 1 may be appropriately labeled and injected into an individual suspected of having ovarian cancer, detectable according to the particular biomarker, for the purpose of diagnosing or evaluating the ovarian cancer status of the individual.
  • the label used will be selected in accordance with the imaging modality to be used, as previously described. Localization of the label permits determination of the spread of the cancer.
  • the amount of label within an organ or tissue also allows determination of the presence or absence of cancer in that organ or tissue.
  • Aptamer-directed imaging agents could have unique and advantageous characteristics relating to tissue penetration, tissue distribution, kinetics, elimination, potency, and selectivity as compared to other imaging agents.
  • Such techniques may also optionally be performed with labeled oligonucleotides, for example, for detection of gene expression through imaging with antisense oligonucleotides. These methods are used for in situ hybridization, for example, with fluorescent molecules or radionuclides as the label. Other methods for detection of gene expression include, for example, detection of the activity of a reporter gene.
  • optical imaging Another general type of imaging technology is optical imaging, in which fluorescent signals within the subject are detected by an optical device that is external to the subject. These signals may be due to actual fluorescence and/or to bioluminescence.
  • Improvements in the sensitivity of optical detection devices have increased the usefulness of optical imaging for in vivo diagnostic assays.
  • in vivo molecular biomarker imaging is increasing, including for clinical trials, for example, to more rapidly measure clinical efficacy in trials for new cancer therapies and/or to avoid prolonged treatment with a placebo for those diseases, such as multiple sclerosis, in which such prolonged treatment may be considered to be ethically questionable.
  • tissue samples may be used in histological or cytological methods. Sample selection depends on the primary tumor location and sites of metastases. For example, fine needle aspirates, cutting needles, and core biopsies can be used for histology. Ascites can be used for cyotology. While cytological analysis is still used in the diagnosis of ovarian cancer, histological methods are known to provide better sensitivity for the detection of cancer. Any of the biomarkers identified herein that were shown to be up-regulated (see Table 15) in the individuals with ovarian cancer can be used to stain a histological specimen as an indication of disease.
  • one or more capture reagents specific to the corresponding biomarker is used in a cytological evaluation of an ovarian cell sample and may include one or more of the following: collecting a cell sample, fixing the cell sample, dehydrating, clearing, immobilizing the cell sample on a microscope slide, permeabilizing the cell sample, treating for analyte retrieval, staining, destaining, washing, blocking, and reacting with one or more capture reagent/s in a buffered solution.
  • the cell sample is produced from a cell block.
  • one or more capture reagents specific to the corresponding biomarker is used in a histological evaluation of an ovarian tissue sample and may include one or more of the following: collecting a tissue specimen, fixing the tissue sample, dehydrating, clearing, immobilizing the tissue sample on a microscope slide, permeabilizing the tissue sample, treating for analyte retrieval, staining, destaining, washing, blocking, rehydrating, and reacting with capture reagent/s in a buffered solution.
  • fixing and dehydrating are replaced with freezing.
  • the one or more aptamers specific to the corresponding biomarker is reacted with the histological or cytological sample and can serve as the nucleic acid target in a nucleic acid amplification method.
  • Suitable nucleic acid amplification methods include, for example, PCR, q-beta replicase, rolling circle amplification, strand displacement, helicase dependent amplification, loop mediated isothermal amplification, ligase chain reaction, and restriction and circularization aided rolling circle amplification.
  • the one or more capture reagent/s specific to the corresponding biomarkers for use in the histological or cytological evaluation are mixed in a buffered solution that can include any of the following: blocking materials, competitors, detergents, stabilizers, carrier nucleic acid, polyanionic materials, etc.
  • a "cytology protocol” generally includes sample collection, sample fixation, sample immobilization, and staining.
  • Cell preparation can include several processing steps after sample collection, including the use of one or more slow off -rate aptamers for the staining of the prepared cells.
  • Sample collection can include directly placing the sample in an untreated transport container, placing the sample in a transport container containing some type of media, or placing the sample directly onto a slide (immobilization) without any treatment or fixation.
  • Sample immobilization can be improved by applying a portion of the collected specimen to a glass slide that is treated with polylysine, gelatin, or a silane. Slides can be prepared by smearing a thin and even layer of cells across the slide. Care is generally taken to minimize mechanical distortion and drying artifacts.
  • Liquid specimens can be processed in a cell block method. Or, alternatively, liquid specimens can be mixed 1 : 1 with the fixative solution for about 10 minutes at room temperature.
  • Cell blocks can be prepared from residual effusions, sputum, urine sediments, gastrointestinal fluids, cell scraping, ascites, or fine needle aspirates. Cells are concentrated or packed by centrifugation or membrane filtration. A number of methods for cell block preparation have been developed. Representative procedures include the fixed sediment, bacterial agar, or membrane filtration methods. In the fixed sediment method, the cell sediment is mixed with a fixative like Bouins, picric acid, or buffered formalin and then the mixture is centrifuged to pellet the fixed cells. The supernatant is removed, drying the cell pellet as completely as possible. The pellet is collected and wrapped in lens paper and then placed in a tissue cassette.
  • a fixative like Bouins, picric acid, or buffered formalin
  • the tissue cassette is placed in ajar with additional fixative and processed as a tissue sample.
  • Agar method is very similar but the pellet is removed and dried on paper towel and then cut in half. The cut side is placed in a drop of melted agar on a glass slide and then the pellet is covered with agar making sure that no bubbles form in the agar. The agar is allowed to harden and then any excess agar is trimmed away. This is placed in a tissue cassette and the tissue process completed.
  • the pellet may be directly suspended in 2% liquid agar at 65°C and the sample centrifuged. The agar cell pellet is allowed to solidify for an hour at 4°C. The solid agar may be removed from the centrifuge tube and sliced in half. The agar is wrapped in filter paper and then the tissue cassette. Processing from this point forward is as described above. Centrifugation can be replaced in any these procedures with membrane filtration. Any of these processes may be used to generate a "cell block sample”.
  • Cell blocks can be prepared using specialized resin including Lowicryl resins,
  • LR White, LR Gold, Unicryl, and MonoStep These resins have low viscosity and can be polymerized at low temperatures and with ultra violet (UV) light.
  • UV ultra violet
  • the embedding process relies on progressively cooling the sample during dehydration, transferring the sample to the resin, and polymerizing a block at the final low temperature at the appropriate UV wavelength.
  • Cell block sections can be stained with hematoxylin-eosin for cytomorphological examination while additional sections are used for examination for specific markers.
  • the sample may be fixed prior to additional processing to prevent sample degradation.
  • This process is called "fixation" and describes a wide range of materials and procedures that may be used interchangeably.
  • the sample fixation protocol and reagents are best selected empirically based on the targets to be detected and the specific cell/tissue type to be analyzed.
  • Sample fixation relies on reagents such as ethanol, polyethylene glycol, methanol, formalin, or isopropanol.
  • the samples should be fixed as soon after collection and affixation to the slide as possible.
  • the fixative selected can introduce structural changes into various molecular targets making their subsequent detection more difficult.
  • fixation and immobilization processes and their sequence can modify the appearance of the cell and these changes must be anticipated and recognized by the cytotechnologist.
  • Fixatives can cause shrinkage of certain cell types and cause the cytoplasm to appear granular or reticular.
  • Many fixatives function by crosslinking cellular components. This can damage or modify specific epitopes, generate new epitopes, cause molecular associations, and reduce membrane permeability.
  • Formalin fixation is one of the most common cytological and histological approaches. Formalin forms methyl bridges between neighboring proteins or within proteins. Precipitation or coagulation is also used for fixation and ethanol is frequently used in this type of fixation.
  • a combination of crosslinking and precipitation can also be used for fixation.
  • a strong fixation process is best at preserving morphological information while a weaker fixation process is best for the preservation of molecular targets.
  • a representative fixative is 50% absolute ethanol, 2 mM polyethylene glycol
  • PEG 1.85% formaldehyde. Variations on this formulation include ethanol (50% to 95%), methanol (20% - 50%), and formalin (formaldehyde) only.
  • Another common fixative is 2% PEG 1500, 50% ethanol, and 3% methanol. Slides are place in the fixative for about 10 to 15 minutes at room temperature and then removed and allowed to dry. Once slides are fixed they can be rinsed with a buffered solution like PBS.
  • a wide range of dyes can be used to differentially highlight and contrast or
  • stain cellular, sub-cellular, and tissue features or morphological structures.
  • Hematoylin is used to stain nuclei a blue or black color.
  • Orange G-6 and Eosin Azure both stain the cell's cytoplasm.
  • Orange G stains keratin and glycogen containing cells yellow.
  • Eosin Y is used to stain nucleoli, cilia, red blood cells, and superficial epithelial squamous cells.
  • Romanowsky stains are used for air dried slides and are useful in enhancing pleomorphism and distinguishing extracellular from intracytoplasmic material.
  • the staining process can include a treatment to increase the permeability of the cells to the stain.
  • Treatment of the cells with a detergent can be used to increase permeability.
  • fixed samples can be further treated with solvents, saponins, or non-ionic detergents. Enzymatic digestion can also improve the accessibility of specific targets in a tissue sample.
  • the sample is dehydrated using a succession of alcohol rinses with increasing alcohol concentration.
  • the final wash is done with xylene or a xylene substitute, such as a citrus terpene, that has a refractive index close to that of the coverslip to be applied to the slide. This final step is referred to as clearing.
  • a mounting medium is applied. The mounting medium is selected to have a refractive index close to the glass and is capable of bonding the coverslip to the slide.
  • the final evaluation of the ovarian cytological specimen is made by some type of microscopy to permit a visual inspection of the morphology and a determination of the marker's presence or absence.
  • exemplary microscopic methods include brightfield, phase contrast, fluorescence, and differential interference contrast.
  • the coverslip may be removed and the slide destained. Destaining involves using the original solvent systems used in staining the slide originally without the added dye and in a reverse order to the original staining procedure. Destaining may also be completed by soaking the slide in an acid alcohol until the cells are colorless. Once colorless the slides are rinsed well in a water bath and the second staining procedure applied.
  • specific molecular differentiation may be possible in conjunction with the cellular morphological analysis through the use of specific molecular reagents such as antibodies or nucleic acid probes or aptamers. This improves the accuracy of diagnostic cytology.
  • Micro-dissection can be used to isolate a subset of cells for additional evaluation, in particular, for genetic evaluation of abnormal chromosomes, gene expression, or mutations.
  • Preparation of a tissue sample for histological evaluation involves fixation, dehydration, infiltration, embedding, and sectioning.
  • the fixation reagents used in histology are very similar or identical to those used in cytology and have the same issues of preserving morphological features at the expense of molecular ones such as individual proteins.
  • Time can be saved if the tissue sample is not fixed and dehydrated but instead is frozen and then sectioned while frozen. This is a more gentle processing procedure and can preserve more individual markers.
  • freezing is not acceptable for long term storage of a tissue sample as subcellular information is lost due to the introduction of ice crystals.
  • Ice in the frozen tissue sample also prevents the sectioning process from producing a very thin slice and thus some microscopic resolution and imaging of subcellular structures can be lost.
  • osmium tetroxide is used to fix and stain phospholipids (membranes).
  • Dehydration of tissues is accomplished with successive washes of increasing alcohol concentration. Clearing employs a material that is miscible with alcohol and the embedding material and involves a stepwise process starting at 50:50 alcohol:clearing reagent and then 100% clearing agent (xylene or xylene substitute). Infiltration involves incubating the tissue with a liquid form of the embedding agent (warm wax, nitrocellulose solution) first at 50:50 embedding agent: clearing agent and the 100% embedding agent. Embedding is completed by placing the tissue in a mold or cassette and filling with melted embedding agent such as wax, agar, or gelatin. The embedding agent is allowed to harden.
  • the hardened tissue sample may then be sliced into thin section for staining and subsequent examination.
  • the tissue section Prior to staining, the tissue section is dewaxed and rehydrated. Xylene is used to dewax the section, one or more changes of xylene may be used, and the tissue is rehydrated by successive washes in alcohol of decreasing concentration. Prior to dewax, the tissue section may be heat immobilized to a glass slide at about 80°C for about 20 minutes.
  • Laser capture micro-dissection allows the isolation of a subset of cells for further analysis from a tissue section.
  • tissue section or slice can be stained with a variety of stains.
  • a large menu of commercially available stains can be used to enhance or identify specific features.
  • analyte retrieval uses high temperature heating of a fixed sample. This method is also referred to as heat-induced epitope retrieval or HIER. A variety of heating techniques have been used, including steam heating, microwaving, autoclaving, water baths, and pressure cooking or a combination of these methods of heating.
  • Analyte retrieval solutions include, for example, water, citrate, and normal saline buffers. The key to analyte retrieval is the time at high temperature but lower temperatures for longer times have also been successfully used. Another key to analyte retrieval is the pH of the heating solution.
  • the section is first dewaxed and hydrated.
  • the slide is then placed in 1OmM sodium citrate buffer pH 6.0 in a dish or jar.
  • a representative procedure uses an 110OW microwave and microwaves the slide at 100% power for 2 minutes followed by microwaving the slides using 20% power for 18 minutes after checking to be sure the slide remains covered in liquid.
  • the slide is then allowed to cool in the uncovered container and then rinsed with distilled water.
  • HIER may be used in combination with an enzymatic digestion to improve the reactivity of the target to immunochemical reagents.
  • One such enzymatic digestion protocol uses proteinase K.
  • a 20 ⁇ g/ml concentration of proteinase K is prepared in 50 mM Tris Base, ImM EDTA, 0.5% Triton X- 100, pH 8.0 buffer. The process first involves dewaxing sections in 2 changes of xylene, 5 minutes each. Then the sample is hydrated in 2 changes of 100% ethanol for 3 minutes each, 95% and 80% ethanol for 1 minute each, and then rinsed in distilled water. Sections are covered with Proteinase K working solution and incubated 10-20 minutes at 37°C in humidified chamber (optimal incubation time may vary depending on tissue type and degree of fixation).
  • the sections are cooled at room temperature for 10 minutes and then rinsed in PBS Tween 20 for 2x2 min. If desired, sections can be blocked to eliminate potential interference from endogenous compounds and enzymes.
  • the section is then incubated with primary antibody at appropriate dilution in primary antibody dilution buffer for 1 hour at room temperature or overnight at 4°C.
  • the section is then rinsed with PBS Tween 20 for 2x2 min. Additional blocking can be performed, if required for the specific application, followed by additional rinsing with PBS Tween 20 for 3x2 min and then finally the immuno staining protocol completed.
  • Blocking reactions may include the need to do any of the following, either alone or in combination: reduce the level of endogenous biotin; eliminate endogenous charge effects; inactivate endogenous nucleases; and inactivate endogenous enzymes like peroxidase and alkaline phosphatase.
  • Endogenous nucleases may be inactivated by degradation with proteinase K, by heat treatment, use of a chelating agent such as EDTA or EGTA, the introduction of carrier DNA or RNA, treatment with a chaotrope such as urea, thiourea, guanidine hydrochloride, guanidine thiocyanate, lithium perchlorate, etc, or diethyl pyrocarbonate.
  • Alkaline phosphatase may be inactivated by treated with 0. IN HCl for 5 minutes at room temperature or treatment with 1 mM levamisole. Peroxidase activity may be eliminated by treatment with 0.03% hydrogen peroxide.
  • Endogenous biotin may be blocked by soaking the slide or section in an avidin (streptavidin, neutravidin may be substituted) solution for at least 15 minutes at room temperature. The slide or section is then washed for at least 10 minutes in buffer. This may be repeated at least three times. Then the slide or section is soaked in a biotin solution for 10 minutes. This may be repeated at least three times with a fresh biotin solution each time. The buffer wash procedure is repeated.
  • Blocking protocols should be minimized to prevent damaging either the cell or tissue structure or the target or targets of interest but one or more of these protocols could be combined to "block" a slide or section prior to reaction with one or more slow off-rate aptamers. See Basic Medical Histology: the Biology of Cells, Tissues and Organs, authored by Richard G. Kessel, Oxford University Press, 1998.
  • mass spectrometers can be used to detect biomarker values.
  • Several types of mass spectrometers are available or can be produced with various configurations.
  • a mass spectrometer has the following major components: a sample inlet, an ion source, a mass analyzer, a detector, a vacuum system, and instrument- control system, and a data system. Difference in the sample inlet, ion source, and mass analyzer generally define the type of instrument and its capabilities.
  • an inlet can be a capillary-column liquid chromatography source or can be a direct probe or stage such as used in matrix-assisted laser desorption.
  • Common ion sources are, for example, electrospray, including nanospray and microspray or matrix-assisted laser desorption.
  • Common mass analyzers include a quadrupole mass filter, ion trap mass analyzer and time- of-flight mass analyzer. Additional mass spectrometry methods are well known in the art (see Burlingame et al. Anal. Chem. 70:647 R-716R (1998); Kinter and Sherman, New York (2000)).
  • Protein biomarkers and biomarker values can be detected and measured by any of the following: electrospray ionization mass spectrometry (ESI-MS), ESI-MS/MS, ESI- MS/(MS)n, matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS), surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF-MS), desorption/ionization on silicon (DIOS), secondary ion mass spectrometry (SIMS), quadrupole time-of-flight (Q-TOF), tandem time-of-flight (TOF/TOF) technology, called ultraflex III TOF/TOF, atmospheric pressure chemical ionization mass spectrometry (APCI-MS), APCI-MS/MS, APCI-(MS) N , atmospheric pressure photoionization mass spectrometry (APPI-MS), APPI-MS),
  • Sample preparation strategies are used to label and enrich samples before mass spectroscopic characterization of protein biomarkers and determination biomarker values.
  • Labeling methods include but are not limited to isobaric tag for relative and absolute quantitation (iTRAQ) and stable isotope labeling with amino acids in cell culture (SILAC).
  • Capture reagents used to selectively enrich samples for candidate biomarker proteins prior to mass spectroscopic analysis include but are not limited to aptamers, antibodies, nucleic acid probes, chimeras, small molecules, an F(ab') 2 fragment, a single chain antibody fragment, an Fv fragment, a single chain Fv fragment, a nucleic acid, a lectin, a ligand-binding receptor, affybodies, nanobodies, ankyrins, domain antibodies, alternative antibody scaffolds (e.g.
  • the foregoing assays enable the detection of biomarker values that are useful in methods for diagnosing ovarian cancer, where the methods comprise detecting, in a biological sample from an individual, at least N biomarker values that each correspond to a biomarker selected from the group consisting of the biomarkers provided in Table 1, wherein a classification, as described in detail below, using the biomarker values indicates whether the individual has ovarian cancer. While certain of the described ovarian cancer biomarkers are useful alone for detecting and diagnosing ovarian cancer, methods are also described herein for the grouping of multiple subsets of the ovarian cancer biomarkers that are each useful as a panel of three or more biomarkers.
  • N is at least three biomarkers.
  • N is selected to be any number from 2-42 biomarkers. It will be appreciated that N can be selected to be any number from any of the above described ranges, as well as similar, but higher order, ranges.
  • biomarker values can be detected and classified individually or they can be detected and classified collectively, as for example in a multiplex assay format.
  • methods for detecting an absence of ovarian cancer, the methods comprising detecting, in a biological sample from an individual, at least N biomarker values that each correspond to a biomarker selected from the group consisting of the biomarkers provided in Table 1, wherein a classification, as described in detail below, of the biomarker values indicates an absence of ovarian cancer in the individual. While certain of the described ovarian cancer biomarkers are useful alone for detecting and diagnosing the absence of ovarian cancer, methods are also described herein for the grouping of multiple subsets of the ovarian cancer biomarkers that are each useful as a panel of three or more biomarkers.
  • N is at least three biomarkers.
  • N is selected to be any number from 2-42 biomarkers. It will be appreciated that N can be selected to be any number from any of the above described ranges, as well as similar, but higher order, ranges.
  • biomarker values can be detected and classified individually or they can be detected and classified collectively, as for example in a multiplex assay format.
  • a biomarker "signature" for a given diagnostic test contains a set of markers, each marker having different levels in the populations of interest. Different levels, in this context, may refer to different means of the marker levels for the individuals in two or more groups, or different variances in the two or more groups, or a combination of both. For the simplest form of a diagnostic test, these markers can be used to assign an unknown sample from an individual into one of two groups, either diseased or not diseased. The assignment of a sample into one of two or more groups is known as classification, and the procedure used to accomplish this assignment is known as a classifier or a classification method. Classification methods may also be referred to as scoring methods.
  • classification methods There are many classification methods that can be used to construct a diagnostic classifier from a set of biomarker values.
  • classification methods are most easily performed using supervised learning techniques where a data set is collected using samples obtained from individuals within two (or more, for multiple classification states) distinct groups one wishes to distinguish. Since the class (group or population) to which each sample belongs is known in advance for each sample, the classification method can be trained to give the desired classification response. It is also possible to use unsupervised learning techniques to produce a diagnostic classifier.
  • training data includes samples from the distinct groups (classes) to which unknown samples will later be assigned.
  • samples collected from individuals in a control population and individuals in a particular disease population can constitute training data to develop a classifier that can classify unknown samples (or, more particularly, the individuals from whom the samples were obtained) as either having the disease or being free from the disease.
  • the development of the classifier from the training data is known as training the classifier. Specific details on classifier training depend on the nature of the supervised learning technique.
  • An illustrative example of the development of a diagnostic test using a set of biomarkers includes the application of a na ⁇ ve Bayes classifier, a simple probabilistic classifier based on Bayes theorem with strict independent treatment of the biomarkers.
  • Each biomarker is described by a class-dependent probability density function (pdf) for the measured RFU values or log RFU (relative fluorescence units) values in each class.
  • n j l where the individual JCJ s are the measured biomarker levels in RFU or log RFU.
  • the classification assignment for an unknown is facilitated by calculating the probability of being diseased p(d I x) having measured x compared to the probability of being disease free (control) p(c I x) for the same measured values. The ratio of these probabilities is computed
  • log likelihood ratio This form is known as the log likelihood ratio and simply states that the log likelihood of being free of the particular disease versus having the disease and is primarily composed of the sum of individual log likelihood ratios of the n individual biomarkers.
  • an unknown sample or, more particularly, the individual from whom the sample was obtained
  • the class -dependent biomarker pdfs p(x [ I c) and p(x[ I d) are assumed to be normal or log-normal distributions in the measured RFU values x[ , i.e.
  • the Bayes classifier is fully determined and may be used to classify unknown samples with measured values x .
  • the performance of the na ⁇ ve Bayes classifier is dependent upon the number and quality of the biomarkers used to construct and train the classifier.
  • a single biomarker will perform in accordance with its KS-distance (Kolmogorov-Smirnov), as defined in Example 3, below. If a classifier performance metric is defined as the sum of the sensitivity (fraction of true positives, fjp ) and specificity (one minus the fraction of false positives, 1 - fpp ), a perfect classifier will have a score of two and a random classifier, on average, will have a score of one.
  • a greedy algorithm is any algorithm that follows the problem solving metaheuristic of making the locally optimal choice at each stage with the hope of finding the global optimum.
  • Example 4 The algorithm approach used here is described in detail in Example 4. Briefly, all single analyte classifiers are generated from a table of potential biomarkers and added to a list. Next, all possible additions of a second analyte to each of the stored single analyte classifiers is then performed, saving a predetermined number of the best scoring pairs, say, for example, a thousand, on a new list. All possible three-marker classifiers are explored using this new list of the best two-marker classifiers, again saving the best thousand of these. This process continues until the score either plateaus or begins to deteriorate as additional markers are added. Those high scoring classifiers that remain after convergence can be evaluated for the desired performance for an intended use.
  • classifiers with a high sensitivity and modest specificity may be more desirable than modest sensitivity and high specificity.
  • classifiers with a high specificity and a modest sensitivity may be more desirable.
  • the desired level of performance is generally selected based upon a trade-off that must be made between the number of false positives and false negatives that can each be tolerated for the particular diagnostic application. Such trade-offs generally depend on the medical consequences of an error, either false positive or false negative.
  • Various other techniques are known in the art and may be employed to generate many potential classifiers from a list of biomarkers using a na ⁇ ve Bayes classifier.
  • a genetic algorithm can be used to combine different markers using the fitness score as defined above. Genetic algorithms are particularly well suited to exploring a large diverse population of potential classifiers.
  • so-called ant colony optimization can be used to generate sets of classifiers.
  • Other strategies that are known in the art can also be employed, including, for example, other evolutionary strategies as well as simulated annealing and other stochastic search methods. Metaheuristic methods, such as, for example, harmony search may also be employed.
  • Exemplary embodiments use any number of the ovarian cancer biomarkers listed in Table 1 in various combinations to produce diagnostic tests for detecting ovarian cancer (see Example 2 for a detailed description of how these biomarkers were identified).
  • a method for diagnosing ovarian cancer uses a na ⁇ ve Bayes classification method in conjunction with any number of the ovarian cancer biomarkers listed in Table 1.
  • the simplest test for detecting ovarian cancer from a population of women with pelvic masses can be constructed using a single biomarker, for example, BAFF Receptor which is down-regulated in ovarian cancer with a KS -distance of
  • the score for a classifier constructed of two biomarkers is not a simple sum of the KS-distances; KS- distances are not additive when combining biomarkers, and it takes many more weak markers to achieve the same level of performance as a strong marker.
  • Adding additional biomarkers, such as, for example, SLPI, C9, ⁇ 2- Antiplasmin, SAP, MMP-7, MCP-3, and HSP90 ⁇ produces a series of ovarian cancer tests summarized in Table 17 and displayed as a series of ROC curves in Figure 3.
  • the score of the classifiers as a function of the number of analytes used in classifier construction is shown in Figure 4.
  • This exemplary ten-marker classifier has a sensitivity of 97% and a specificity of 88% with an AUC of 0.94.
  • markers listed in Table 1 can be combined in many ways to produce classifiers for diagnosing ovarian cancer.
  • panels of biomarkers are comprised of different numbers of analytes depending on a specific diagnostic performance criterion that is selected. For example, certain combinations of biomarkers will produce tests that are more sensitive (or more specific) than other combinations.
  • a panel is defined to include a particular set of biomarkers from Table 1 and a classifier is constructed from a set of training data
  • the definition of the diagnostic test is complete.
  • the procedure used to classify an unknown sample is outlined in Figure IA.
  • the procedure used to classify an unknown sample is outlined in Figure IB.
  • the biological sample is appropriately diluted and then run in one or more assays to produce the relevant quantitative biomarker levels used for classification.
  • the measured biomarker levels are used as input for the classification method that outputs a classification and an optional score for the sample that reflects the confidence of the class assignment.
  • Table 1 identifies 42 biomarkers that are useful for diagnosing ovarian cancer.
  • classifiers can be constructed such that particular biomarkers may be substituted for other biomarkers in a manner that reflects the redundancies that undoubtedly pervade the complexities of the underlying disease processes. That is to say, the information about the disease contributed by any individual biomarker identified in Table 1 overlaps with the information contributed by other biomarkers, such that it may be that no particular biomarker or small group of biomarkers in Table 1 must be included in any classifier.
  • Exemplary embodiments use na ⁇ ve Bayes classifiers constructed from the data in Table 18 to classify an unknown sample.
  • the procedure is outlined in Figures IA and B.
  • the biological sample is optionally diluted and run in a multiplexed aptamer assay.
  • the data from the assay are normalized and calibrated as outlined in Example 3, and the resulting biomarker levels are used as input to a Bayes classification scheme.
  • the log-likelihood ratio is computed for each measured biomarker individually and then summed to produce a final classification score, which is also referred to as a diagnostic score.
  • the resulting assignment as well as the overall classification score can be reported.
  • the individual log-likelihood risk factors computed for each biomarker level can be reported as well.
  • the details of the classification score calculation are presented in Example 3.
  • any combination of the biomarkers of Table 1 can be detected using a suitable kit, such as for use in performing the methods disclosed herein.
  • a suitable kit such as for use in performing the methods disclosed herein.
  • any kit can contain one or more detectable labels as described herein, such as a fluorescent moiety, etc.
  • a kit includes (a) one or more capture reagents (such as, for example, at least one aptamer or antibody) for detecting one or more biomarkers in a biological sample, wherein the biomarkers include any of the biomarkers set forth in Table 1, and optionally (b) one or more software or computer program products for classifying the individual from whom the biological sample was obtained as either having or not having ovarian cancer or for determining the likelihood that the individual has ovarian cancer, as further described herein.
  • one or more instructions for manually performing the above steps by a human can be provided.
  • kits The combination of a solid support with a corresponding capture reagent and a signal generating material is referred to herein as a "detection device" or “kit”.
  • the kit can also include instructions for using the devices and reagents, handling the sample, and analyzing the data. Further the kit may be used with a computer system or software to analyze and report the result of the analysis of the biological sample.
  • the kits can also contain one or more reagents (e.g., solubilization buffers, detergents, washes, or buffers) for processing a biological sample. Any of the kits described herein can also include, e.g., buffers, blocking agents, mass spectrometry matrix materials, antibody capture agents, positive control samples, negative control samples, software and information such as protocols, guidance and reference data.
  • kits for the analysis of ovarian cancer status include PCR primers for one or more biomarkers selected from Table 1.
  • the kit may further include instructions for use and correlation of the biomarkers with ovarian cancer.
  • the kit may also include any of the following, either alone or in combination: a DNA array containing the complement of one or more of the biomarkers selected from Table 1, reagents, and enzymes for amplifying or isolating sample DNA.
  • the kits may include reagents for real-time PCR, such as, for example, TaqMan probes and/or primers, and enzymes.
  • a kit can comprise (a) reagents comprising at least capture reagent for quantifying one or more biomarkers in a test sample, wherein said biomarkers comprise the biomarkers set forth in Table 1, or any other biomarkers or biomarkers panels described herein, and optionally (b) one or more algorithms or computer programs for performing the steps of comparing the amount of each biomarker quantified in the test sample to one or more predetermined cutoffs and assigning a score for each biomarker quantified based on said comparison, combining the assigned scores for each biomarker quantified to obtain a total score, comparing the total score with a predetermined score, and using said comparison to determine whether an individual has ovarian cancer.
  • one or more instructions for manually performing the above steps by a human can be provided.
  • a method for diagnosing an individual can comprise the following: 1) collect or otherwise obtain a biological sample; 2) perform an analytical method to detect and measure the biomarker or biomarkers in the panel in the biological sample; 3) perform any data normalization or standardization required for the method used to collect biomarker values; 4) calculate the marker score; 5) combine the marker scores to obtain a total diagnostic score; and 6) report the individual's diagnostic score.
  • the diagnostic score may be a single number determined from the sum of all the marker calculations that is compared to a preset threshold value that is an indication of the presence or absence of disease.
  • the diagnostic score may be a series of bars that each represent a biomarker value and the pattern of the responses may be compared to a pre-set pattern for determination of the presence or absence of disease.
  • FIG. 6 An example of a computer system 100 is shown in Figure 6. With reference to Figure 6, system 100 is shown comprised of hardware elements that are electrically coupled via bus 108, including a processor 101, input device 102, output device 103, storage device 104, computer-readable storage media reader 105a, communications system 106 processing acceleration (e.g., DSP or special-purpose processors) 107 and memory 109.
  • processing acceleration e.g., DSP or special-purpose processors
  • Computer-readable storage media reader 105a is further coupled to computer-readable storage media 105b, the combination comprehensively representing remote, local, fixed and/or removable storage devices plus storage media, memory, etc. for temporarily and/or more permanently containing computer-readable information, which can include storage device 104, memory 109 and/or any other such accessible system 100 resource.
  • System 100 also comprises software elements (shown as being currently located within working memory 191) including an operating system 192 and other code 193, such as programs, data and the like.
  • system 100 has extensive flexibility and configurability.
  • a single architecture might be utilized to implement one or more servers that can be further configured in accordance with currently desirable protocols, protocol variations, extensions, etc.
  • embodiments may well be utilized in accordance with more specific application requirements.
  • one or more system elements might be implemented as sub- elements within a system 100 component (e.g., within communications system 106).
  • Customized hardware might also be utilized and/or particular elements might be implemented in hardware, software or both.
  • the system can comprise a database containing features of biomarkers characteristic of ovarian cancer.
  • the biomarker data (or biomarker information) can be utilized as an input to the computer for use as part of a computer implemented method.
  • the biomarker data can include the data as described herein.
  • system further comprises one or more devices for providing input data to the one or more processors.
  • the system further comprises a memory for storing a data set of ranked data elements.
  • the device for providing input data comprises a detector for detecting the characteristic of the data element, e.g., such as a mass spectrometer or gene chip reader.
  • the system additionally may comprise a database management system.
  • User requests or queries can be formatted in an appropriate language understood by the database management system that processes the query to extract the relevant information from the database of training sets.
  • the system may be connectable to a network to which a network server and one or more clients are connected.
  • the network may be a local area network (LAN) or a wide area network (WAN), as is known in the art.
  • the server includes the hardware necessary for running computer program products (e.g., software) to access database data for processing user requests.
  • the system may include an operating system (e.g., UNIX or Linux) for executing instructions from a database management system.
  • the operating system can operate on a global communications network, such as the internet, and utilize a global communications network server to connect to such a network.
  • the system may include one or more devices that comprise a graphical display interface comprising interface elements such as buttons, pull down menus, scroll bars, fields for entering text, and the like as are routinely found in graphical user interfaces known in the art.
  • Requests entered on a user interface can be transmitted to an application program in the system for formatting to search for relevant information in one or more of the system databases.
  • Requests or queries entered by a user may be constructed in any suitable database language.
  • the graphical user interface may be generated by a graphical user interface code as part of the operating system and can be used to input data and/or to display inputted data.
  • the result of processed data can be displayed in the interface, printed on a printer in communication with the system, saved in a memory device, and/or transmitted over the network or can be provided in the form of the computer readable medium.
  • the system can be in communication with an input device for providing data regarding data elements to the system (e.g., expression values).
  • the input device can include a gene expression profiling system including, e.g., a mass spectrometer, gene chip or array reader, and the like.
  • the methods and apparatus for analyzing ovarian cancer biomarker information may be implemented in any suitable manner, for example, using a computer program operating on a computer system.
  • a conventional computer system comprising a processor and a random access memory, such as a remotely- accessible application server, network server, personal computer or workstation may be used.
  • Additional computer system components may include memory devices or information storage systems, such as a mass storage system and a user interface, for example a conventional monitor, keyboard and tracking device.
  • the computer system may be a stand-alone system or part of a network of computers including a server and one or more databases.
  • the ovarian cancer biomarker analysis system can provide functions and operations to complete data analysis, such as data gathering, processing, analysis, reporting and/or diagnosis.
  • the computer system can execute the computer program that may receive, store, search, analyze, and report information relating to the ovarian cancer biomarkers.
  • the computer program may comprise multiple modules performing various functions or operations, such as a processing module for processing raw data and generating supplemental data and an analysis module for analyzing raw data and supplemental data to generate an ovarian cancer status and/or diagnosis.
  • Diagnosing ovarian cancer status may comprise generating or collecting any other information, including additional biomedical information, regarding the condition of the individual relative to the disease, identifying whether further tests may be desirable, or otherwise evaluating the health status of the individual.
  • biomarker information can be retrieved for an individual.
  • the biomarker information can be retrieved from a computer database, for example, after testing of the individual's biological sample is performed.
  • a computer can be utilized to classify each of the biomarker values.
  • a determination can be made as to the likelihood that an individual has ovarian cancer based upon a plurality of classifications.
  • the indication can be output to a display or other indicating device so that it is viewable by a person. Thus, for example, it can be displayed on a display screen of a computer or other output device.
  • a computer can be utilized to retrieve biomarker information for an individual.
  • the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers provided in Table 1.
  • a classification of the biomarker value can be performed with the computer.
  • an indication can be made as to the likelihood that the individual has ovarian cancer based upon the classification.
  • the indication can be output to a display or other indicating device so that it is viewable by a person. Thus, for example, it can be displayed on a display screen of a computer or other output device.
  • a computer program product may include a computer readable medium having computer readable program code embodied in the medium for causing an application program to execute on a computer with a database.
  • a "computer program product” refers to an organized set of instructions in the form of natural or programming language statements that are contained on a physical media of any nature (e.g., written, electronic, magnetic, optical or otherwise) and that may be used with a computer or other automated data processing system. Such programming language statements, when executed by a computer or data processing system, cause the computer or data processing system to act in accordance with the particular content of the statements.
  • Computer program products include without limitation: programs in source and object code and/or test or data libraries embedded in a computer readable medium.
  • the computer program product that enables a computer system or data processing equipment device to act in pre-selected ways may be provided in a number of forms, including, but not limited to, original source code, assembly code, object code, machine language, encrypted or compressed versions of the foregoing and any and all equivalents.
  • a computer program product for indicating a likelihood of ovarian cancer.
  • a computer program product for indicating a likelihood of ovarian cancer.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers provided in Table 1; and code that executes a classification method that indicates an ovarian disease status of the individual as a function of the biomarker value.
  • the embodiments may be embodied as code stored in a computer-readable memory of virtually any kind including, without limitation, RAM, ROM, magnetic media, optical media, or magneto-optical media. Even more generally, the embodiments could be implemented in software, or in hardware, or any combination thereof including, but not limited to, software running on a general purpose processor, microcode, PLAs, or ASICs.
  • embodiments could be accomplished as computer signals embodied in a carrier wave, as well as signals (e.g., electrical and optical) propagated through a transmission medium.
  • signals e.g., electrical and optical
  • the various types of information discussed above could be formatted in a structure, such as a data structure, and transmitted as an electrical signal through a transmission medium or stored on a computer readable medium.
  • many of the structures, materials, and acts recited herein can be recited as means for performing a function or step for performing a function. Therefore, it should be understood that such language is entitled to cover all such structures, materials, or acts disclosed within this specification and their equivalents, including the matter incorporated by reference.
  • the multiplexed analysis utilized 811 aptamers, each unique to a specific target.
  • aptamers without a photo-cleavable biotin linker custom stock aptamer solutions for 10%, 1% and 0.03% plasma were prepared at 8x concentration in Ix SB17, 0.05% Tween-20 with appropriate photo-cleavable, biotinylated primers, where the resultant primer concentration was 3 times the relevant aptamer concentration.
  • the primers hybridized to all or part of the corresponding aptamer.
  • Tween-20 (1500 ⁇ L of 8x stock into 4500 ⁇ L of lxSB17, 0.05% Tween-20) to achieve a 2x concentration.
  • Each diluted aptamer master mix was then split, 1500 ⁇ L each, into 4, 2 mL screw cap tubes and brought to 95°C for 5 minutes, followed by a 37°C incubation for 15 minutes. After incubation, the 4, 2 mL tubes corresponding to a particular aptamer master mix were combined into a reagent trough, and 55 ⁇ L of a 2x aptamer mix (for all three mixes) was manually pipetted into a 96-well Hybaid plate and the plate foil sealed.
  • the final result was 3, 96-well, foil-sealed Hybaid plates.
  • the individual aptamer concentration was 0.5 nM.
  • a 20% sample solution was prepared by transferring 16 ⁇ L of sample using a
  • sample and aptamer solutions were mixed on the robot by pipetting up and down.
  • MAHVN4550 were equilibrated with 100 ⁇ L of Ix SB 17, 0.05% Tween-20 for at least 10 minutes.
  • the equilibration buffer was then filtered through the plate and 133.3 ⁇ L of a 7.5% Streptavidin-agarose bead slurry (in Ix SB 17, 0.05% Tween-20) was added into each well.
  • the bead solution was manually mixed with a 200 ⁇ L, 12-channel pipettor, 15 times. After the beads were distributed across the 3 filter plates, a vacuum was applied to remove the bead supernatant.
  • Plates were blotted to remove droplets using an on-deck blot station and then incubated with orbital shakers at 800 rpm for 10 minutes at 25°C.
  • the robot removed this wash via vacuum filtration and blotted the bottom of the filter plate to remove droplets using the on-deck blot station.
  • the NHS-PEO4- biotin reagent was dissolved at 100 mM concentration in anhydrous DMSO and had been stored frozen at -20 0 C.
  • the diluted NHS-PEO4-biotin reagent was manually added to an on-deck trough and the robot program was manually re-initiated to dispense 100 ⁇ L of the NHS-PEO4-biotin into each well of each Catch 1 filter plate. This solution was allowed to incubate with Catch 1 beads shaking at 800 rpm for 5 minutes on the obital shakers.
  • the tagging reaction was quenched by the addition of 150 ⁇ L of 20 mM glycine in Ix SB 17, 0.05% Tween-20 to the Catch 1 plates while still containing the NHS tag. The plates were then incubated for 1 minute on orbital shakers at 800 rpm. The NHS- tag/glycine solution was removed via vacuum filtration. Next, 190 ⁇ L 20 mM glycine (Ix
  • Thermoshaker (Thermo Fisher Scientific, Inc., Waltham, MA ) under the BlackRay (Ted Pella, Inc., Redding, CA) light sources, and irradiated for 10 minutes while shaking at 800 rpm.
  • the robot transferred all of the photo-cleaved eluate from the 1 mL deep-well plate onto the Hybaid plate containing the previously prepared catch 2 MyOne magnetic beads (after removal of the MyOne buffer via magnetic separation).
  • the robot transferred the plate to the on deck magnetic separator station.
  • the plate was incubated on the magnet for 90 seconds before removal and discarding of the supernatant.
  • the catch 2 plate was then placed onto the magnetic separator for 90 seconds prior to transferring 90 ⁇ L of the eluate to a new 96-well plate containing 10 ⁇ L of 500 mM
  • a gasket slide was placed into an Agilent hybridization chamber and 40 ⁇ L of each of the samples containing hybridization and blocking solution was manually pipetted into each gasket.
  • An 8 -channel variable spanning pipettor was used in a manner intended to minimize bubble formation.
  • Custom Agilent microarray slides (Agilent Technologies, Inc.,
  • the assembled hybridization chambers were incubated in an Agilent hybridization oven for 19 hours at 60 0 C rotating at 20 rpm.
  • Agilent Wash Buffer 1 Approximately 400 mL Agilent Wash Buffer 1 was placed into each of two separate glass staining dishes. One of the staining dishes was placed on a magnetic stir plate and a slide rack and stir bar were placed into the buffer. [00310] A staining dish for Agilent Wash 2 was prepared by placing a stir bar into an empty glass staining dish.
  • a fourth glass staining dish was set aside for the final acetonitrile wash.
  • Each of six hybridization chambers was disassembled. One-by-one, the slide/backing sandwich was removed from its hybridization chamber and submerged into the staining dish containing Wash 1. The slide/backing sandwich was pried apart using a pair of tweezers, while still submerging the microarray slide. The slide was quickly transferred into the slide rack in the Wash 1 staining dish on the magnetic stir plate.
  • the slide rack was gently raised and lowered 5 times.
  • the magnetic stirrer was turned on at a low setting and the slides incubated for 5 minutes.
  • microarray slides were placed into Agilent scanner slide holders and loaded into the Agilent Microarray scanner according to the manufacturer's instructions.
  • Example 2 Biomarker Identification
  • the identification of potential ovarian cancer biomarkers was performed for diagnosis of ovarian cancer in women with pelvic masses. Enrollment criteria for this study were women scheduled for laparotomy or pelvic surgery for suspicion of ovarian cancer. The primary criteria for exclusion were women suffering from chronic infectious (e.g. hepatitis B, Hepatitis C or HIV), autoimmune, or inflammatory conditions or women being treated for malignancy (other than basal or squamous cell carcinomas of the skin) within the last two years. Plasma samples were collected from two different clinical sites and included 142 cases and 195 benign controls. Table 19 summarizes the site sample information.
  • the multiplexed ap tamer affinity assay was used to measure and report the RFU value for 811 analytes in each of these 337 samples. Since the plasma samples were obtained from two independent sites under similar protocols, an examination of site differences prior to the analysis for biomarkers discovery was performed. Each of the two populations, benign pelvic mass and ovarian cancer, was separately compared between sites by generating within- site, class-dependent cumulative distribution functions (cdfs) for each of the 811 analytes. The KS -test was then applied to each analyte between both site pairs within a common class to identify those analytes that differed not by class but rather by site. In both site comparisons among the two classes, statistically significant site-dependent differences were observed.
  • cdfs class-dependent cumulative distribution functions
  • biomarkers can be used to build classifiers that assign samples to either a control or disease group.
  • many such classifiers were produced from these sets of biomarkers and the frequency with which any biomarker was used in good scoring classifiers determined. Those biomarkers that occurred most frequently among the top scoring classifiers were the most useful for creating a diagnostic test.
  • Bayesian classifiers were used to explore the classification space but many other supervised learning techniques may be employed for this purpose.
  • the scoring fitness of any individual classifier was gauged by summing the sensitivity and specificity of the classifier at the Bayesian surface assuming a disease prevalence of 0.5. This scoring metric varies from zero to two, with two being an error-free classifier.
  • Example 3 The details of constructing a Bayesian classifier from biomarker population measurements are described in Example 3.
  • KS -distances were computed for all analytes using the class -dependent cdfs aggregated across all sites. Using a KS-distance threshold of 0.4, fifty-nine potential biomarkers for diagnosing malignant ovarian cancer from benign pelvic masses were identified.
  • Method (2) focused on consistency of potential biomarker changes between the control and case groups among the individual sites.
  • the class-dependent cdfs were constructed for all analytes within each site separately and from these cdfs the KS -distances were computed to identify potential biomarkers.
  • Sixty- three analytes were found to have a KS-distance greater than 0.4 in all the sites.
  • Using these Sixty-three analytes to build potential 10-analyte Bayesian classifiers there were 2031 classifiers that had a score of 1.75 or better. Twenty- four analytes occurred with a frequency greater than 5% among these classifiers and are presented in Table 21 and shown in Figure 11.
  • a set of potential biomarkers were produced by requiring an analyte to have a KS distance of 0.4 or better in the aggregated set as well as the two site comparisons. Forty-five analytes satisfy these requirements and are referred to as a blended set of potential biomarkers. For a classification score of 1.75 or better, a total of 1563 Bayesian classifiers were built from this set of potential biomarkers and twenty-seven biomarkers were identified from this set of classifiers using a frequency cut-off of 5%. These analytes are displayed in Table 22 and Figure 12 is a frequency plot for the identified biomarkers.
  • a final list of biomarkers is obtained by combining the three sets of biomarkers identified above with frequencies greater than 5% in high scoring classifiers, Tables 20-22. From these sets of twenty- five, twenty-four, and twenty-seven biomarkers, forty- two unique biomarkers were identified and are shown in Table 1.
  • Table 15 includes a dissociation constant for the aptamer used to identify the biomarker, the limit of quantification for the marker in the multiplex aptamer assay, and whether the marker was up- regulated or down-regulated in the disease population relative to the control population.
  • n 10 here.
  • Each of the terms in the summation is a log-likelihood ratio for an individual marker and the total log-likelihood ratio of a sample x being free from the disease of interest versus having the disease (i.e. in this case, ovarian cancer) is simply the sum of these individual terms plus a term that accounts for the prevalence of the disease.
  • Four of the ten biomarker values have likelihoods more consistent with the disease group (log likelihood ⁇ 0) while the remaining six biomarkers favor the control group, the largest by a factor of 3.5: 1. Multiplying the likelihoods together gives the same result as that shown above; an aggregate likelihood of 7: 1 that the unknown sample is free from the disease. In fact, this sample came from the control population in the training set.
  • This example describes the selection of biomarkers from Table 1 to form panels that can be used as classifiers in any of the methods described herein. Subsets of the biomarkers in Table 1 were selected to construct classifiers with good performance. This method was also used to determine which potential markers were included as biomarkers in Example 2.
  • the measure of classifier performance used here is the sum of the sensitivity and specificity; a performance of 1.0 is the baseline expectation for a random (coin toss) classifier, a classifier worse than random would score between 0.0 and 1.0, a classifier with better than random performance would score between 1.0 and 2.0. A perfect classifier with no errors would have a sensitivity of 1.0 and a specificity of 1.0, therefore a performance of 2.0 (1.0+1.0).
  • One can apply other common measures of performance such as area under the ROC curve, the F-measure, or the product of sensitivity and specificity.
  • any weighting scheme which results in a single performance measure can be used. Different applications will have different benefits for true positive and true negative findings, and will have different costs associated with false positive findings from false negative findings. For example, screening and the differential diagnosis of benign pelvic masses will not in general have the same optimal trade-off between specificity and sensitivity. The different demands of the two tests will in general require setting different weighting to positive and negative misclassifications, which will be reflected in the performance measure. Changing the performance measure will in general change the exact subset of markers selected from Table 1 for a given set of data.
  • the classifier was completely parameterized by the distributions of biomarkers in the disease and non-disease training samples, and the list of biomarkers was chosen from Table 1; that is to say, the subset of markers chosen for inclusion determined a classifier in a one-to-one manner given a set of training data.
  • the greedy method employed here was used to search for the optimal subset of markers from Table 1. For small numbers of markers or classifiers with relatively few markers, every possible subset of markers was enumerated and evaluated in terms of the performance of the classifier constructed with that particular set of markers (see Example 4, Part T).
  • Each marker subset currently on the list was extended by adding any marker from Table 1 not already part of that classifier, and which would not, on its addition to the subset, duplicate an existing subset (these are termed "permissible markers"). Every existing marker subset was extended by every permissible marker from the list. Clearly, such a process would eventually generate every possible subset, and the list would run out of space. Therefore, all the generated classifiers were kept only while the list was less than some predetermined size (often enough to hold all three marker subsets). Once the list reached the predetermined size limit, it became elitist; that is, only those classifiers which showed a certain level of performance were kept on the list, and the others fell off the end of the list and were lost.
  • biomarkers selected in Table 1 gave rise to classifiers that perform better than classifiers built with "non-markers" (i.e., proteins having signals that did not meet the criteria for inclusion in Table 1 (as described in Example 2)).
  • Figure 14 shows histograms of the performance of all possible one, two, and three-marker classifiers built from the biomarker parameters in Table 18 for biomarkers that can discriminate between benign pelvic masses and ovarian cancer and compares these classifiers with all possible one, two, and three-marker classifiers built using the 42 "non- marker" aptamer RFU signals.
  • Figure 14A shows the histograms of single marker classifier performance
  • Figure 14B shows the histogram of two-marker classifier performance
  • Figure 14C shows the histogram of three-marker classifier performance.
  • the solid lines represent the histograms of the classifier performance of all one, two, and three-marker classifiers using the biomarker data for benign pelvic masses and ovarian cancer in Table 18.
  • the dotted lines are the histograms of the classifier performance of all one, two, and three-marker classifiers using the data for benign pelvic masses and ovarian cancer but using the set of random non-marker signals.
  • the classifiers built from the markers listed in Table 1 form a distinct histogram, well separated from the classifiers built with signals from the "non-markers" for all one-marker, two-marker, and three-marker comparisons.
  • the performance and AUC score of the classifiers built from the biomarkers in Table 1 also increase at a higher rate as markers are added than do the classifiers built from the non-markers.
  • the separation of performance increases between the marker and non-marker classifiers as the number of markers per classifier increases. All classifiers built using the biomarkers listed in Table 1 perform distinctly better than classifiers built using the "non-markers".
  • Figure 15 demonstrates that classifiers constructed without the best markers perform well, implying that the performance of the classifiers was not due to some small core group of markers and that the changes in the underlying processes associated with disease are reflected in the activities of many proteins.
  • Many subsets of the biomarkers in Table 1 performed close to optimally, even after removing the top 15 of the 42 markers from Table 1. After dropping the 15 top-ranked markers (ranked by KS-distance) from Table 1, the classifier performance increased with the number of markers selected from the table to reach almost 1.80 (sensitivity + specificity), close to the performance of the optimal classifier score of 1.87 selected from the full list of biomarkers.
  • Figure 16 shows how the ROC performance of typical classifiers constructed from the list of parameters in Table 18 according to Example 3.
  • a five analyte classifier was constructed with TIMP-2, MCP-3, Cadherin-5, SLPI, and C9.
  • Figure 16A shows the performance of the model, assuming independence of these markers, as in Example 3, and
  • Figure 16B shows the empirical ROC curves generated from the study data set used to define the parameters in Table 18. It can be seen that the performance for a given number of selected markers was qualitatively in agreement, and that quantitative agreement was generally quite good, as evidenced by the AUCs, although the model calculation tends to overestimate classifier performance.
  • Example 5 Aptamer Specificity Demonstration in a Pull-down Assay
  • the final readout on the multiplex assay is based on the amount of aptamer recovered after the successive capture steps in the assay.
  • the multiplex assay is based on the premise that the amount of aptamer recovered at the end of the assay is proportional to the amount of protein in the original complex mixture (e.g., plasma).
  • the original complex mixture e.g., plasma
  • This assay can be used to visually demonstrate that a desired protein is in fact pulled out from plasma after equilibration with an aptamer as well as to demonstrate that aptamers bound to their intended protein targets can survive as a complex through the kinetic challenge steps in the assay.
  • recovery of protein at the end of this pull-down assay requires that the protein remain non-covalently bound to the aptamer for nearly two hours after equilibration.
  • non-specifically bound proteins dissociate during these steps and do not contribute significantly to the final signal. It should be noted that the pull-down procedure described in this example includes all of the key steps in the multiplex assay described above.
  • Plasma samples were prepared by diluting 50 ⁇ L EDTA-plasma to 100 ⁇ L in
  • This step will capture proteins bound to aptamers as well as proteins that may have dissociated from aptamers since the initial equilibration.
  • the beads were washed as described in Example 1. Proteins were eluted from the MyOne Streptavidin beads by incubating with 50 mM DTT in SB17T for 25 minutes at 37 0 C with shaking. The eluate was then transferred to MyOne beads coated with a sequence complimentary to the 3' fixed region of the aptamer and incubated for 25 minutes at 37 0 C with shaking. This step captures all of the remaining aptamer. The beads were washed 2x with 100 ⁇ L SB17T for 1 minute and Ix with 100 ⁇ L SB19T for 1 minute.
  • Aptamer was eluted from these final beads by incubating with 45 ⁇ L 20 mM NaOH for 2 minutes with shaking to disrupt the hybridized strands. 40 ⁇ L of this eluate was neutralized with 10 ⁇ L 80 mM HCl containing 0.05% Tween-20. Aliquots representing 5% of the eluate from the first set of beads (representing all plasma proteins bound to the aptamer) and 20% of the eluate from the final set of beads (representing all plasma proteins remaining bound at the end of our clinical assay) were run on a NuPAGE 4-12% Bis-Tris gel (Invitrogen) under reducing and denaturing conditions. Gels were imaged on an Alpha Innotech FluorChem Q scanner in the Cy5 channel to image the proteins.
  • B. Pull-down gels for aptamers were selected against LBP ( ⁇ lxl ⁇ ⁇ 7 M in plasma, polypeptide MW -60 kDa), C9 ( ⁇ lxl ⁇ ⁇ 6 M in plasma, polypeptide MW -60 kDa), and IgM ( ⁇ 9xlO ⁇ 6 M in plasma, MW -70 kDa and 23 kDa), respectively. (See Figure 13).
  • lane 1 is the eluate from the Streptavidin-agarose beads
  • lane 2 is the final eluate
  • lane 3 is a MW marker lane (major bands are at 110, 50, 30, 15, and 3.5 kDa from top to bottom).
  • biomarkers of Table 1 can be specifically excluded either as an individual biomarker or as a biomarker from any panel.
  • Table 18 Parameters derived from training set for na ⁇ ve Bayes classifier.

Abstract

The present application includes biomarkers, methods, devices, reagents, systems, and kits for the detection and diagnosis of ovarian cancer. In one aspect, the application provides biomarkers that can be used alone or in various combinations to diagnose ovarian cancer or permit the differential diagnosis of a pelvic mass as benign or malignant. In another aspect, methods are provided for diagnosing ovarian cancer in an individual, where the methods include detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers provided in Table 1, wherein the individual is classified as having ovarian cancer, or the likelihood of the individual having ovarian cancer is determined, based on the at least one biomarker value.

Description

OVARIAN CANCER BIOMARKERS AND USES THEREOF
FIELD OF THE INVENTION
[0001] The present application relates generally to the detection of biomarkers and the diagnosis of cancer in an individual and, more specifically, to one or more biomarkers, methods, devices, reagents, systems, and kits for diagnosing cancer, more particularly ovarian cancer, in an individual.
BACKGROUND
[0002] The following description provides a summary of information relevant to the present application and is not an admission that any of the information provided or publications referenced herein is prior art to the present application. [0003] Ovarian cancer is the eighth most common cancer in women and the fifth leading cause of cancer-related deaths in women in the United States. Of all females born in the United States, one of every 70 will develop ovarian cancer and one of every 100 will die from this disease. The American Cancer Society estimates that approximately 21,550 women will be diagnosed with ovarian cancer in 2009 (American Cancer Society. Cancer Facts & Figures 2009. Atlanta: American Cancer Society; 2009). It is estimated that 14,600 women will die from this disease in 2009.
[0004] The survival rate and quality of patient life are improved the earlier ovarian cancer is detected. There is currently no sufficiently accurate screening test proven to be effective in the early detection of ovarian cancer. Thus, a pressing need exists for sensitive and specific methods for detecting ovarian cancer, particularly early- stage ovarian cancer. [0005] Approximately 7% of the female population is at increased risk for ovarian cancer, based on genetic or family history. The risk for ovarian cancer increases with age. Women who have had breast cancer or who have a family history of breast or ovarian cancer are at increased risk. Inherited mutations in BRCAl or BRC A2 genes increase risk. Ovarian cancer incidence rates are highest in Western industrialized countries.
[0006] Between 75% and 85% of ovarian cancers are diagnosed at an advanced stage.
There is no consistent, reliable, non-invasive test to signal the presence of ovarian cancer. Pelvic examination only occasionally detects ovarian cancer, generally when the disease is advanced. Symptoms are often vague or nonexistent until late stages of the disease. Symptomatic women report frequent (>12 times/month) abdominal pain, bloating, increased girth, difficulty eating or feeling full quickly (Goff et al. Cancer 2007; 109:221). Trans- vaginal ultrasound and serum CA 125 levels have been tested as a screen for ovarian cancer and have not been found satisfactory. A laparotomy is required when ovarian cancer is suspected. The outcome of ovarian cancer patients operated on by a gynecology oncology surgical specialist is improved compared to a general gynecological surgeon, demonstrating that need for differential diagnosis of ovarian cancer from a suspicious pelvic mass prior to surgery. Goff reported on over 10,000 women in nine states undergoing surgery for a suspicious pelvic mass. Among the most important factors for receiving appropriate surgical management were surgeon specialty of gynecologic oncologist and the volume of cases performed by the surgeon annually. There are only 1000 board certified gynecologic oncologists in the United States, mostly in the larger medical centers across the country. Appropriately directing the women who are most likely to benefit from the care of such specialists can be critical for achieving good patient outcomes. [0007] Currently, cancer antigen 125 (CA- 125) is the most widely used serum biomarker for ovarian cancer. Serum concentrations of CA- 125 are elevated (>35 U/ml) in 75-80% of patients with advanced-stage disease and this marker is routinely used to follow response to treatment and disease progression in patients from whom CA- 125 -secreting tumors have been resected. However, because the levels of CA- 125 are correlated with tumor volume, only 50% of patients with early-stage disease have elevated levels, indicating that the sensitivity of CA- 125 as a screening tool for early stage disease is limited. The utility of CA- 125 screening is further limited by the high frequency of false-positive results associated with a variety of benign conditions, including endometriosis, pregnancy, menstruation, pelvic inflammatory disease, peritonitis, pancreatitis, and liver disease. [0008] Classification of cancers determines appropriate treatment and helps determine the prognosis of the patient. Ovarian cancers are classified according to histology (i.e., "grading") and extent of the disease (i.e., "staging") using recognized grade and stage systems. In grade I, the tumor tissue is well differentiated. In grade II, tumor tissue is moderately well differentiated. In grade III, the tumor tissue is poorly differentiated. Grade III correlates with a less favorable prognosis than either grade I or II. Stage I is generally confined within the capsule surrounding one (stage IA) or both (stage IB) ovaries, although in some stage I (i.e. stage IC) cancers, malignant cells may be detected in ascites, in peritoneal rinse fluid, or on the surface of the ovaries. Stage II involves extension or metastasis of the tumor from one or both ovaries to other pelvic structures. In stage HA, the tumor extends or has metastasized to the uterus, the fallopian tubes, or both. Stage HB involves metastasis of the tumor to the pelvis. Stage HC is stage HA or HB with the added requirement that malignant cells may be detected in ascites, in peritoneal rinse fluid, or on the surface of the ovaries. In stage III, the tumor comprises at least one malignant extension to the small bowel or the omentum, has formed extra-pelvic peritoneal implants of microscopic (stage IIIA) or macroscopic (<2 centimeter diameter, stage MB; >2 centimeter diameter, stage IIIC) size, or has metastasized to a retroperitoneal or inguinal lymph node (an alternate indicator of stage IIIC). In stage IV, distant (i.e. non-peritoneal) metastases of the tumor can be detected. [0009] Treatment options include surgery, chemotherapy, and occasionally radiation therapy. Surgery usually involves removal of one or both ovaries, fallopian tubes (salpingoophorectomy), and the uterus (hysterectomy). In younger women with very early stage tumors who wish to have children, only the involved ovary and fallopian tube may be removed. In more advanced disease, surgically removing all abdominal metastases enhances the effect of chemotherapy and helps improve survival. For women with stage III ovarian cancer that has been optimally debulked (removal of as much of the cancerous tissue as possible), studies have shown that chemotherapy administered both intravenously and directly into the peritoneal cavity improves survival. Studies have found that women who are treated by a gynecologic oncologist have more successful outcomes. [0010] Relative survival varies by age; women younger than 65 are about twice as likely to survive 5 years (57%) following diagnosis as women 65 and older (29%). Overall, the 1- and 5-year relative survival of ovarian cancer patients is 75% and 46%, respectively. If diagnosed at the localized stage, the 5-year survival rate is 93%; however, only 19% of all cases are detected at this stage, usually fortuitously during another medical procedure. The majority of cases (67%) are diagnosed at distant stage. For women with regional and distant disease, 5-year survival rates are 71% and 31%, respectively; the chance of recurrence in these women is 20-85%. The 10-year relative survival rate for all stages combined is 39%. Therefore, ovarian cancer tends to be diagnosed too late to save women's lives. Detecting recurrence and predicting and monitoring response to therapy is important for making informed decisions on appropriate treatment throughout the care of these patients. [0011] A blood screening test that can reliably detect early stage ovarian cancer will save thousands of lives each year. Where methods of early diagnosis in cancer exist, the benefits are generally accepted by the medical community. Cancers for which widely utilized screening protocols exist have the highest 5-year survival rates, such as breast cancer (88%) and colon cancer (65%) versus 46% for ovarian cancer. However, fortuitous detection of early stage ovarian cancer is associated with a substantial increase in 5-year survival (>95%). Therefore, early detection could significantly impact long-term survival. This demonstrates the clear need for diagnostic methods that can reliably detect early-stage ovarian cancer. [0012] Biomarker selection for a specific disease state involves first the identification of markers that have a measurable and statistically significant difference in a disease population compared to a control population for a specific medical application. Biomarkers can include secreted or shed molecules that parallel disease development or progression and readily diffuse into the blood stream from ovarian tissue or from surrounding tissues and circulating cells in response to a tumor. The biomarker or set of biomarkers identified are generally clinically validated or shown to be a reliable indicator for the original intended use for which it was selected. Biomarkers can include small molecules, peptides, proteins, and nucleic acids. Some of the key issues that affect the identification of biomarkers include over- fitting of the available data and bias in the data.
[0013] A variety of methods have been utilized in an attempt to identify biomarkers and diagnose disease. For protein-based markers, these include two-dimensional electrophoresis, mass spectrometry, and immunoassay methods. For nucleic acid markers, these include mRNA expression profiles, microRNA profiles, FISH, serial analysis of gene expression (SAGE), methylation profiles, and large scale gene expression arrays. [0014] The utility of two-dimensional electrophoresis is limited by low detection sensitivity; issues with protein solubility, charge, and hydrophobicity; gel reproducibility; and the possibility of a single spot representing multiple proteins. For mass spectrometry, depending on the format used, limitations revolve around the sample processing and separation, sensitivity to low abundance proteins, signal to noise considerations, and inability to immediately identify the detected protein. Limitations in immunoassay approaches to biomarker discovery are centered on the inability of antibody-based multiplex assays to measure a large number of analytes. One might simply print an array of high-quality antibodies and, without sandwiches, measure the analytes bound to those antibodies. (This would be the formal equivalent of using a whole genome of nucleic acid sequences to measure by hybridization all DNA or RNA sequences in an organism or a cell. The hybridization experiment works because hybridization can be a stringent test for identity. Even very good antibodies are not stringent enough in selecting their binding partners to work in the context of blood or even cell extracts because the protein ensemble in those matrices have extremely different abundances.) Thus, one must use a different approach with immunoassay-based approaches to biomarker discovery - one would need to use multiplexed ELISA assays (that is, sandwiches) to get sufficient stringency to measure many analytes simultaneously to decide which analytes are indeed biomarkers. Sandwich immunoassays do not scale to high content, and thus biomarker discovery using stringent sandwich immunoassays is not possible using standard array formats. Lastly, antibody reagents are subject to substantial lot variability and reagent instability. The instant platform for protein biomarker discovery overcomes this problem.
[0015] Many of these methods rely on or require some type of sample fractionation prior to the analysis. Thus the sample preparation required to run a sufficiently powered study designed to identify and discover statistically relevant biomarkers in a series of well- defined sample populations is extremely difficult, costly, and time consuming. During fractionation, a wide range of variability can be introduced into the various samples. For example, a potential marker could be unstable to the process, the concentration of the marker could be changed, inappropriate aggregation or disaggregation could occur, and inadvertent sample contamination could occur and thus obscure the subtle changes anticipated in early disease.
[0016] It is widely accepted that biomarker discovery and detection methods using these technologies have serious limitations for the identification of diagnostic biomarkers. These limitations include an inability to detect low-abundance biomarkers, an inability to consistently cover the entire dynamic range of the proteome, ^reproducibility in sample processing and fractionation, and overall ^reproducibility and lack of robustness of the method. Further, these studies have introduced biases into the data and not adequately addressed the complexity of the sample populations, including appropriate controls, in terms of the distribution and randomization required to identify and validate biomarkers within a target disease population.
[0017] Although efforts aimed at the discovery of new and effective biomarkers have gone on for several decades, the efforts have been largely unsuccessful. Biomarkers for various diseases typically have been identified in academic laboratories, usually through an accidental discovery while doing basic research on some disease process. Based on the discovery and with small amounts of clinical data, papers were published that suggested the identification of a new biomarker. Most of these proposed biomarkers, however, have not been confirmed as real or useful biomarkers; primarily because the small number of clinical samples tested provide only weak statistical proof that an effective biomarker has in fact been found. That is, the initial identification was not rigorous with respect to the basic elements of statistics. In each of the years 1994 through 2003, a search of the scientific literature shows that thousands of references directed to biomarkers were published. During that same time frame, however, the FDA approved for diagnostic use, at most, three new protein biomarkers a year, and in several years no new protein biomarkers were approved. [0018] Based on the history of failed biomarker discovery efforts, mathematical theories have been proposed that further promote the general understanding that biomarkers for disease are rare and difficult to find. Biomarker research based on 2D gels or mass spectrometry supports these notions. Very few useful biomarkers have been identified through these approaches. However, it is usually overlooked that 2D gel and mass spectrometry measure proteins that are present in blood at approximately 1 nM concentrations and higher, and that this ensemble of proteins may well be the least likely to change with disease. Other than the instant biomarker discovery platform, proteomic biomarker discovery platforms that are able to accurately measure protein expression levels at much lower concentrations do not exist.
[0019] Much is known about biochemical pathways for complex human biology.
Many biochemical pathways culminate in or are started by secreted proteins that work locally within the pathology, for example growth factors are secreted to stimulate the replication of other cells in the pathology, and other factors are secreted to ward off the immune system, and so on. While many of these secreted proteins work in a paracrine fashion, some operate distally in the body. One skilled in the art with a basic understanding of biochemical pathways would understand that many pathology-specific proteins ought to exist in blood at concentrations below (even far below) the detection limits of 2D gels and mass spectrometry. What must precede the identification of this relatively abundant number of disease biomarkers is a proteomic platform that can analyze proteins at concentrations below those detectable by 2D gels or mass spectrometry.
[0020] Accordingly, a need exists for biomarkers, methods, devices, reagents, systems, and kits that enable (a) the differentiation of benign pelvic masses from ovarian cancer; (b) referral to a gynecologic oncology surgeon rather than a general gynecologic surgeon to surgically treat cases of ovarian cancer; (c) the detection of ovarian cancer biomarkers; and (d) the diagnosis of ovarian cancer.
SUMMARY
[0021] The present application includes biomarkers, methods, reagents, devices, systems, and kits for the detection and diagnosis of cancer and more particularly, ovarian cancer. The biomarkers of the present application were identified using a multiplex aptamer- based assay, which is described in detail in Example 1. By using the aptamer-based biomarker identification method described herein, this application describes a surprisingly large number of ovarian cancer biomarkers that are useful for the detection and diagnosis of ovarian cancer. In identifying these biomarkers, over 800 proteins from hundreds of individual samples were measured, some of which were at concentrations in the low femtomolar range. This is about four orders of magnitude lower than biomarker discovery experiments done with 2D gels or mass spectrometry.
[0022] While certain of the described ovarian cancer biomarkers are useful alone for detecting and diagnosing ovarian cancer, methods are described herein for the grouping of multiple subsets of the ovarian cancer biomarkers that are useful as a panel of biomarkers. Once an individual biomarker or subset of biomarkers has been identified, the detection or diagnosis of ovarian cancer in an individual can be accomplished using any assay platform or format that is capable of measuring differences in the levels of the selected biomarker or biomarkers in a biological sample.
[0023] However, it was only by using the aptamer-based biomarker identification method described herein, wherein over 800 separate potential biomarker values were individually screened from a large number of individuals who were postoperatively diagnosed as either having or not having ovarian cancer that it was possible to identify the ovarian cancer biomarkers disclosed herein. This discovery approach is in stark contrast to biomarker discovery using conditioned media or lysed cells as it queries a more patient- relevant system that requires no translation to human pathology.
[0024] Thus, in one aspect of the instant application, one or more biomarkers are provided for use either alone or in various combinations to diagnose ovarian cancer or permit the differential diagnosis of pelvic masses as benign or malignant. Exemplary embodiments include the biomarkers provided in Table 1, which as noted above, were identified using a multiplex aptamer-based assay, as described in Examples 1 and 2. The markers provided in Table 1 are useful in distinguishing benign pelvic masses from ovarian cancer. [0025] While certain of the described ovarian cancer biomarkers are useful alone for detecting and diagnosing ovarian cancer, methods are also described herein for the grouping of multiple subsets of the ovarian cancer biomarkers that are each useful as a panel of three or more biomarkers. Thus, various embodiments of the instant application provide combinations comprising N biomarkers, wherein N is at least two biomarkers. In other embodiments, N is selected to be any number from 2-42 biomarkers. [0026] In yet other embodiments, N is selected to be any number from 2-7, 2-10, 2-
15, 2-20, 2-25, 2-30, 2-35, 2-40, or 2-42. In other embodiments, N is selected to be any number from 3-7, 3-10, 3-15, 3-20, 3-25, 3-30, 3-35, 3-40, or 3-42. In other embodiments, N is selected to be any number from 4-7, 4-10, 4-15, 4-20, 4-25, 4-30, 4-35, 4-40, or 4-42. In other embodiments, N is selected to be any number from 5-7, 5-10, 5-15, 5-20, 5-25, 5-30, 5- 35, 5-40, or 5-42. In other embodiments, N is selected to be any number from 6-10, 6-15, 6- 20, 6-25, 6-30, 6-35, 6-40, or 6-42. In other embodiments, N is selected to be any number from 7-10, 7-15, 7-20, 7-25, 7-30, 7-35, 7-40, or 7-42. In other embodiments, N is selected to be any number from 8-10, 8-15, 8-20, 8-25, 8-30, 8-35, 8-40, or 8-42. In other embodiments, N is selected to be any number from 9-15, 9-20, 9-25, 9-30, 9-35, 9-40, or 9-42. In other embodiments, N is selected to be any number from 10-15, 10-20, 10-25, 10-30, 10-35, 10-40, or 10-42. It will be appreciated that N can be selected to encompass similar, but higher order, ranges.
[0027] In another aspect, a method is provided for diagnosing ovarian cancer in an individual, the method including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers provided in Table 1, wherein the individual is classified as having ovarian cancer based on the at least one biomarker value.
[0028] In another aspect, a method is provided for diagnosing ovarian cancer in an individual, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers set forth in Table 1, wherein the likelihood of the individual having ovarian cancer is determined based on the biomarker values.
[0029] In another aspect, a method is provided for diagnosing ovarian cancer in an individual, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as having ovarian cancer based on the biomarker values, and wherein N = 2-10. [0030] In another aspect, a method is provided for diagnosing ovarian cancer in an individual, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers set forth in Table 1, wherein the likelihood of the individual having ovarian cancer is determined based on the biomarker values, and wherein N = 2-10. [0031] In another aspect, a method is provided for differentiating an individual having a benign pelvic mass from an individual having ovarian cancer, the method including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as having ovarian cancer, or the likelihood of the individual having ovarian cancer is determined, based on the at least one biomarker value. [0032] In another aspect, a method is provided for differentiating an individual having a benign pelvic mass from an individual having ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as having ovarian cancer, or the likelihood of the individual having ovarian cancer is determined, based on the biomarker values, wherein N = 2-10.
[0033] In another aspect, a method is provided for diagnosing that an individual does not have ovarian cancer, the method including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as not having ovarian cancer based on the at least one biomarker value.
[0034] In another aspect, a method is provided for diagnosing that an individual does not have ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each corresponding to one of at least N biomarkers selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as not having ovarian cancer based on the biomarker values, and wherein N = 2-10. [0035] In another aspect, a method is provided for diagnosing ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of N biomarkers, wherein the biomarkers are selected from the group of biomarkers set forth in Table 1, wherein a classification of the biomarker values indicates that the individual has ovarian cancer, and wherein N = 3-10. [0036] In another aspect, a method is provided for diagnosing ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of N biomarkers, wherein the biomarkers are selected from the group of biomarkers set forth in Table 1, wherein a classification of the biomarker values indicates that the individual has ovarian cancer, and wherein N = 3-15. [0037] In another aspect, a method is provided for diagnosing ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of biomarkers selected from the group of panels set forth in Tables 2-14, wherein a classification of the biomarker values indicates that the individual has ovarian cancer.
[0038] In another aspect, a method is provided for differentiating an individual having a benign pelvic mass from an individual having ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of N biomarkers, wherein the biomarkers are selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as having ovarian cancer, or the likelihood of the individual having ovarian cancer is determined, based on the biomarker values, and wherein N = 3-10.
[0039] In another aspect, a method is provided for differentiating an individual having a benign pelvic mass from an individual having ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of N biomarkers, wherein the biomarkers are selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as having ovarian cancer, or the likelihood of the individual having ovarian cancer is determined, based on the biomarker values, and wherein N = 3-15.
[0040] In another aspect, a method is provided for diagnosing an absence of ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of N biomarkers, wherein the biomarkers are selected from the group of biomarkers set forth in Table 1, wherein a classification of the biomarker values indicates an absence of ovarian cancer in the individual, and wherein N = 3-10.
[0041] In another aspect, a method is provided for diagnosing an absence of ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of N biomarkers, wherein the biomarkers are selected from the group of biomarkers set forth in Table 1, wherein a classification of the biomarker values indicates an absence of ovarian cancer in the individual, and wherein N = 3-15.
[0042] In another aspect, a method is provided for diagnosing an absence of ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of biomarkers selected from the group of panels provided in Tables 2-14, wherein a classification of the biomarker values indicates an absence of ovarian cancer in the individual. [0043] In another aspect, a method is provided for diagnosing ovarian cancer in an individual, the method including detecting, in a biological sample from an individual, biomarker values that correspond to one of at least N biomarkers selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as having ovarian cancer based on a classification score that deviates from a predetermined threshold, and wherein N=2-10.
[0044] In another aspect, a method is provided for differentiating an individual having a benign pelvic mass from an individual having ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of N biomarkers, wherein the biomarkers are selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as having ovarian cancer, or the likelihood of the individual having ovarian cancer is determined, based on a classification score that deviates from a predetermined threshold, and wherein N = 3-10. [0045] In another aspect, a method is provided for differentiating an individual having a benign pelvic mass from an individual having ovarian cancer, the method including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of N biomarkers, wherein the biomarkers are selected from the group of biomarkers set forth in Table 1, wherein the individual is classified as having ovarian cancer, or the likelihood of the individual having ovarian cancer is determined, based on a classification score that deviates from a predetermined threshold, wherein N = 3-15. [0046] In another aspect, a method is provided for diagnosing an absence of ovarian cancer in an individual, the method including detecting, in a biological sample from an individual, biomarker values that correspond to one of at least N biomarkers selected from the group of biomarkers set forth in Table 1, wherein said individual is classified as not having ovarian cancer based on a classification score that deviates from a predetermined threshold, and wherein N=2-10.
[0047] In another aspect, a computer-implemented method is provided for indicating a likelihood of ovarian cancer. The method comprises: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises biomarker values that each correspond to one of at least N biomarkers, wherein N is as defined above, selected from the group of biomarkers set forth in Table 1 ; performing with the computer a classification of each of the biomarker values; and indicating a likelihood that the individual has ovarian cancer based upon a plurality of classifications. [0048] In another aspect, a computer-implemented method is provided for classifying an individual as either having or not having ovarian cancer. The method comprises: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers provided in Table 1 ; performing with the computer a classification of each of the biomarker values; and indicating whether the individual has ovarian cancer based upon a plurality of classifications.
[0049] In another aspect, a computer program product is provided for indicating a likelihood of ovarian cancer. The computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers, wherein N is as defined above, in the biological sample selected from the group of biomarkers set forth in Table 1 ; and code that executes a classification method that indicates a likelihood that the individual has ovarian cancer as a function of the biomarker values.
[0050] In another aspect, a computer program product is provided for indicating an ovarian cancer status of an individual. The computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers in the biological sample selected from the group of biomarkers provided in Table 1; and code that executes a classification method that indicates an ovarian cancer status of the individual as a function of the biomarker values. [0051] In another aspect, a computer-implemented method is provided for indicating a likelihood of ovarian cancer. The method comprises retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers set forth in Table 1; performing with the computer a classification of the biomarker value; and indicating a likelihood that the individual has ovarian cancer based upon the classification. [0052] In another aspect, a computer-implemented method is provided for classifying an individual as either having or not having ovarian cancer. The method comprises retrieving, from a computer, biomarker information for an individual, wherein the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers provided in Table 1; performing with the computer a classification of the biomarker value; and indicating whether the individual has ovarian cancer based upon the classification.
[0053] In still another aspect, a computer program product is provided for indicating a likelihood of ovarian cancer. The computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers set forth in Table 1; and code that executes a classification method that indicates a likelihood that the individual has ovarian cancer as a function of the biomarker value.
[0054] In still another aspect, a computer program product is provided for indicating an ovarian cancer status of an individual. The computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers provided in Table 1; and code that executes a classification method that indicates an ovarian cancer status of the individual as a function of the biomarker value.
BRIEF DESCRIPTION OF THE DRAWINGS
[0055] Figure IA is a flowchart for an exemplary method for detecting ovarian cancer in a biological sample.
[0056] Figure IB is a flowchart for an exemplary method for detecting ovarian cancer in a biological sample using a naϊve Bayes classification method.
[0057] Figure 2 shows a ROC curve for a single biomarker, BAFF Receptor, using a naϊve Bayes classifier for a test that detects ovarian cancer in women with pelvis masses.
[0058] Figure 3 shows ROC curves for biomarker panels of from one to ten biomarkers using naϊve Bayes classifiers for a test that detects ovarian cancer in women with pelvis masses.
[0059] Figure 4 illustrates the increase in the classification score (specificity + sensitivity) as the number of biomarkers is increased from one to ten using naϊve Bayes classification for an ovarian cancer panel. [0060] Figure 5 shows the measured biomarker distributions for BAFF Receptor as a cumulative distribution function (cdf) in RFU for the benign control group (solid line) and the ovarian cancer disease group (dotted line) along with their curve fits to a normal cdf (dashed lines) used to train the naϊve Bayes classifiers.
[0061] Figure 6 illustrates an exemplary computer system for use with various computer- implemented methods described herein.
[0062] Figure 7 is a flowchart for a method of indicating the likelihood that an individual has ovarian cancer in accordance with one embodiment.
[0063] Figure 8 is a flowchart for a method of indicating the likelihood that an individual has ovarian cancer in accordance with one embodiment.
[0064] Figure 9 illustrates an exemplary aptamer assay that can be used to detect one or more ovarian cancer biomarkers in a biological sample.
[0065] Figure 10 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between ovarian cancer and benign pelvic masses from an aggregated set of potential biomarkers.
[0066] Figure 11 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between ovarian cancer and benign pelvic masses from a site-consistent set of potential biomarkers.
[0067] Figure 12 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between ovarian cancer and benign pelvic masses from a set of potential biomarkers resulting from a combination of aggregated and site-consistent markers.
[0068] Figure 13 shows gel images resulting from pull-down experiments that illustrate the specificity of aptamers as capture reagents for the proteins LBP, C9 and IgM.
For each gel, lane 1 is the eluate from the Streptavidin-agarose beads, lane 2 is the final eluate, and lane is a MW marker lane (major bands are at 110, 50, 30, 15, and 3.5 kDa from top to bottom).
[0069] Figure 14A shows a pair of histograms summarizing all possible single protein naϊve Bayes classifier scores (sensitivity + specificity) using the biomarkers set forth in Table
1 (solid) and a set of random non-markers (dotted).
[0070] Figure 14B shows a pair of histograms summarizing all possible two-protein protein naϊve Bayes classifier scores (sensitivity + specificity) using the biomarkers set forth in Table 1 (solid) and a set of random non-markers (dotted). [0071] Figure 14C shows a pair of histograms summarizing all possible three-protein naϊve Bayes classifier scores (sensitivity + specificity) using the biomarkers set forth in Table
1 (solid) and a set of non-random markers (dotted).
[0072] Figure 15 shows the sensitivity + specificity score for naϊve Bayes classifiers using from 2-10 markers selected from the full panel (•) and the scores obtained by dropping the best 5 (■), 10 (A) and 15 (♦) markers during classifier generation.
[0073] Figure 16A shows a set of ROC curves modeled from the data in Table 18 for panels of from one to five markers.
[0074] Figure 16B shows a set of ROC curves computed from the training data for panels of from one to five markers as in Figure 16A.
DETAILED DESCRIPTION
[0075] Reference will now be made in detail to representative embodiments of the invention. While the invention will be described in conjunction with the enumerated embodiments, it will be understood that the invention is not intended to be limited to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents that may be included within the scope of the present invention as defined by the claims.
[0076] One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in and are within the scope of the practice of the present invention. The present invention is in no way limited to the methods and materials described.
[0077] Unless defined otherwise, 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 belongs. Although any methods, devices, and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, the preferred methods, devices and materials are now described.
[0078] All publications, published patent documents, and patent applications cited in this application are indicative of the level of skill in the art(s) to which the application pertains. All publications, published patent documents, and patent applications cited herein are hereby incorporated by reference to the same extent as though each individual publication, published patent document, or patent application was specifically and individually indicated as being incorporated by reference. [0079] As used in this application, including the appended claims, the singular forms
"a," "an," and "the" include plural references, unless the content clearly dictates otherwise, and are used interchangeably with "at least one" and "one or more." Thus, reference to "an aptamer" includes mixtures of aptamers, reference to "a probe" includes mixtures of probes, and the like.
[0080] As used herein, the term "about" represents an insignificant modification or variation of the numerical value such that the basic function of the item to which the numerical value relates is unchanged.
[0081] As used herein, the terms "comprises," "comprising," "includes," "including,"
"contains," "containing," and any variations thereof, are intended to cover a non-exclusive inclusion, such that a process, method, product-by-process, or composition of matter that comprises, includes, or contains an element or list of elements does not include only those elements but may include other elements not expressly listed or inherent to such process, method, product-by-process, or composition of matter.
[0082] The present application includes biomarkers, methods, devices, reagents, systems, and kits for the detection and diagnosis of ovarian cancer.
[0083] In one aspect, one or more biomarkers are provided for use either alone or in various combinations to diagnose ovarian cancer, permit the differential diagnosis of pelvic masses as benign or malignant, monitor ovarian cancer recurrence, or address other clinical indications. As described in detail below, exemplary embodiments include the biomarkers provided in Table 1, which were identified using a multiplex aptamer-based assay, as described generally in Example 1 and more specifically in Example 2. [0084] Table 1 sets forth the findings obtained from analyzing blood samples from
142 individuals diagnosed with ovarian cancer and blood samples from 195 individuals diagnosed with a benign pelvic mass. The benign pelvic mass group was designed to match the population with which an ovarian cancer diagnostic test can have significant benefit. (These cases and controls were obtained from two clinical sites). The potential biomarkers were measured in individual samples rather than pooling the disease and control blood; this allowed a better understanding of the individual and group variations in the phenotypes associated with the presence and absence of disease (in this case ovarian cancer). Since over 800 protein measurements were made on each sample, and 337 samples from both the disease and the control populations were individually measured, Table 1 resulted from an analysis of an uncommonly large set of data. The measurements were analyzed using the methods described in the section, "Classification of Biomarkers and Calculation of Disease Scores" herein.
[0085] Table 1 lists the biomarkers found to be useful in distinguishing samples obtained from individuals with ovarian cancer from "control" samples obtained from individuals with benign pelvic masses. Using a multiplex aptamer assay, forty-two biomarkers were discovered that distinguished samples obtained from individuals with ovarian cancer from samples obtained from people who had benign pelvic masses (see Table
1).
[0086] While certain of the described ovarian cancer biomarkers are useful alone for detecting and diagnosing ovarian cancer, methods are also described herein for the grouping of multiple subsets of the ovarian cancer biomarkers, where each grouping or subset selection is useful as a panel of three or more biomarkers, interchangeably referred to herein as a "biomarker panel" and a panel. Thus, various embodiments of the instant application provide combinations comprising N biomarkers, wherein N is at least two biomarkers. In other embodiments, N is selected from 2-42 biomarkers.
[0087] In yet other embodiments, N is selected to be any number from 2-7, 2-10, 2-
15, 2-20, 2-25, 2-30, 2-35, 2-40, or 2-42. In other embodiments, N is selected to be any number from 3-7, 3-10, 3-15, 3-20, 3-25, 3-30, 3-35, 3-40, or 3-42. In other embodiments, N is selected to be any number from 4-7, 4-10, 4-15, 4-20, 4-25, 4-30, 4-35, 4-40, or 4-42. In other embodiments, N is selected to be any number from 5-7, 5-10, 5-15, 5-20, 5-25, 5-30, 5- 35, 5-40, or 5-42. In other embodiments, N is selected to be any number from 6-10, 6-15, 6- 20, 6-25, 6-30, 6-35, 6-40, or 6-42. In other embodiments, N is selected to be any number from 7-10, 7-15, 7-20, 7-25, 7-30, 7-35, 7-40, or 7-42. In other embodiments, N is selected to be any number from 8-10, 8-15, 8-20, 8-25, 8-30, 8-35, 8-40, or 8-42. In other embodiments, N is selected to be any number from 9-15, 9-20, 9-25, 9-30, 9-35, 9-40, or 9-42. In other embodiments, N is selected to be any number from 10-15, 10-20, 10-25, 10-30, 10-35, 10-40, or 10-42. It will be appreciated that N can be selected to encompass similar, but higher order, ranges.
[0088] In one embodiment, the number of biomarkers useful for a biomarker subset or panel is based on the sensitivity and specificity value for the particular combination of biomarker values. The terms "sensitivity" and "specificity" are used herein with respect to the ability to correctly classify an individual, based on one or more biomarker values detected in their biological sample, as having ovarian cancer or not having ovarian cancer. "Sensitivity" indicates the performance of the biomarker(s) with respect to correctly classifying individuals that have ovarian cancer. "Specificity" indicates the performance of the biomarker(s) with respect to correctly classifying individuals who do not have ovarian cancer. For example, 85% specificity and 90% sensitivity for a panel of markers used to test a set of control samples and ovarian cancer samples indicates that 85% of the control samples were correctly classified as control samples by the panel, and 90% of the ovarian cancer samples were correctly classified as ovarian cancer samples by the panel. The desired or preferred minimum value can be determined as described in Example 3. Representative panels are set forth in Tables 2-14, which set forth a series of 100 different panels of 3-15 biomarkers, which have the indicated levels of specificity and sensitivity for each panel. The total number of occurrences of each marker in each of these panels is indicated at the bottom of each Table.
[0089] In one aspect, ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to at least one of the biomarkers SLPI, C9, HGF and RGM-C and at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15. In a further aspect, ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarkers SLPI, C9, HGF and RGM-C and one of at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or 13. In a further aspect, ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker SLPI and one of at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15. In a further aspect, ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker C9and one of at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15. In a further aspect, ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker HGF and one of at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15. In a further aspect, ovarian cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker RGM-C and one of at least N additional biomarkers selected from the list of biomarkers in Table 1, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
[0090] The ovarian cancer biomarkers identified herein represent a relatively large number of choices for subsets or panels of biomarkers that can be used to effectively detect or diagnose ovarian cancer. Selection of the desired number of such biomarkers depends on the specific combination of biomarkers chosen. It is important to remember that panels of biomarkers for detecting or diagnosing ovarian cancer may also include biomarkers not found in Table 1, and that the inclusion of additional biomarkers not found in Table 1 may reduce the number of biomarkers in the particular subset or panel that is selected from Table 1. The number of biomarkers from Table 1 used in a subset or panel may also be reduced if additional biomedical information is used in conjunction with the biomarker values to establish acceptable sensitivity and specificity values for a given assay. [0091] Another factor that can affect the number of biomarkers to be used in a subset or panel of biomarkers is the procedures used to obtain biological samples from individuals who are being evaluated for ovarian cancer. In a carefully controlled sample procurement environment, the number of biomarkers necessary to meet desired sensitivity and specificity values will be lower than in a situation where there can be more variation in sample collection, handling and storage. In developing the list of biomarkers set forth in Table 1, two sample collection sites were utilized to collect data for classifier training. [0092] One aspect of the instant application can be described generally with reference to Figures IA and B. A biological sample is obtained from an individual or individuals of interest. The biological sample is then assayed to detect the presence of one or more (N) biomarkers of interest and to determine a biomarker value for each of said N biomarkers (referred to in Figure IB as marker RFU (relative fluorescence units)). Once a biomarker has been detected and a biomarker value assigned each marker is scored or classified as described in detail herein. The marker scores are then combined to provide a total diagnostic score, which indicates the likelihood that the individual from whom the sample was obtained has ovarian cancer.
[0093] "Biological sample", "sample", and "test sample" are used interchangeably herein to refer to any material, biological fluid, tissue, or cell obtained or otherwise derived from an individual. This includes blood (including whole blood, leukocytes, peripheral blood mononuclear cells, buffy coat, plasma, and serum), sputum, tears, mucus, nasal washes, nasal aspirate, breath, urine, semen, saliva, meningeal fluid, amniotic fluid, glandular fluid, lymph fluid, nipple aspirate, bronchial aspirate, synovial fluid, joint aspirate, ascites, cells, a cellular extract, and cerebrospinal fluid. This also includes experimentally separated fractions of all of the preceding. For example, a blood sample can be fractionated into serum or into fractions containing particular types of blood cells, such as red blood cells or white blood cells (leukocytes). If desired, a sample can be a combination of samples from an individual, such as a combination of a tissue and fluid sample. The term "biological sample" also includes materials containing homogenized solid material, such as from a stool sample, a tissue sample, or a tissue biopsy, for example. The term "biological sample" also includes materials derived from a tissue culture or a cell culture. Any suitable methods for obtaining a biological sample can be employed; exemplary methods include, e.g., phlebotomy, swab (e.g., buccal swab), and a fine needle aspirate biopsy procedure. Samples can also be collected, e.g., by micro dissection (e.g., laser capture micro dissection (LCM) or laser micro dissection (LMD)), bladder wash, smear (e.g., a PAP smear), or ductal lavage. A "biological sample" obtained or derived from an individual includes any such sample that has been processed in any suitable manner after being obtained from the individual. [0094] Further, it should be realized that a biological sample can be derived by taking biological samples from a number of individuals and pooling them or pooling an aliquot of each individual's biological sample. The pooled sample can be treated as a sample from a single individual and if the presence of cancer is established in the pooled sample, then each individual biological sample can be re-tested to determine which individuals have ovarian cancer.
[0095] For purposes of this specification, the phrase "data attributed to a biological sample from an individual" is intended to mean that the data in some form derived from, or were generated using, the biological sample of the individual. The data may have been reformatted, revised, or mathematically altered to some degree after having been generated, such as by conversion from units in one measurement system to units in another measurement system; but, the data are understood to have been derived from, or were generated using, the biological sample.
[0096] "Target", "target molecule", and "analyte" are used interchangeably herein to refer to any molecule of interest that may be present in a biological sample. A "molecule of interest" includes any minor variation of a particular molecule, such as, in the case of a protein, for example, minor variations in amino acid sequence, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component, which does not substantially alter the identity of the molecule. A "target molecule", "target", or "analyte" is a set of copies of one type or species of molecule or multi-molecular structure. "Target molecules", "targets", and "analytes" refer to more than one such set of molecules. Exemplary target molecules include proteins, polypeptides, nucleic acids, carbohydrates, lipids, polysaccharides, glycoproteins, hormones, receptors, antigens, antibodies, affybodies, antibody mimics, viruses, pathogens, toxic substances, substrates, metabolites, transition state analogs, cofactors, inhibitors, drugs, dyes, nutrients, growth factors, cells, tissues, and any fragment or portion of any of the foregoing.
[0097] As used herein, "polypeptide," "peptide," and "protein" are used interchangeably herein to refer to polymers of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component. Also included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art. Polypeptides can be single chains or associated chains. Also included within the definition are preproteins and intact mature proteins; peptides or polypeptides derived from a mature protein; fragments of a protein; splice variants; recombinant forms of a protein; protein variants with amino acid modifications, deletions, or substitutions; digests; and post-translational modifications, such as glycosylation, acetylation, phosphorylation, and the like.
[0098] As used herein, "thrombin" refers to thrombin, prothrombin, or both thrombin and prothrombin.
[0099] As used herein, "marker" and "biomarker" are used interchangeably to refer to a target molecule that indicates or is a sign of a normal or abnormal process in an individual or of a disease or other condition in an individual. More specifically, a "marker" or "biomarker" is an anatomic, physiologic, biochemical, or molecular parameter associated with the presence of a specific physiological state or process, whether normal or abnormal, and, if abnormal, whether chronic or acute. Biomarkers are detectable and measurable by a variety of methods including laboratory assays and medical imaging. When a biomarker is a protein, it is also possible to use the expression of the corresponding gene as a surrogate measure of the amount or presence or absence of the corresponding protein biomarker in a biological sample or methylation state of the gene encoding the biomarker or proteins that control expression of the biomarker.
[00100] As used herein, "biomarker value", "value", "biomarker level", and "level" are used interchangeably to refer to a measurement that is made using any analytical method for detecting the biomarker in a biological sample and that indicates the presence, absence, absolute amount or concentration, relative amount or concentration, titer, a level, an expression level, a ratio of measured levels, or the like, of, for, or corresponding to the biomarker in the biological sample. The exact nature of the "value" or "level" depends on the specific design and components of the particular analytical method employed to detect the biomarker.
[00101] When a biomarker indicates or is a sign of an abnormal process or a disease or other condition in an individual, that biomarker is generally described as being either over- expressed or under-expressed as compared to an expression level or value of the biomarker that indicates or is a sign of a normal process or an absence of a disease or other condition in an individual. "Up-regulation", "up-regulated", "over-expression", "over-expressed", and any variations thereof are used interchangeably to refer to a value or level of a biomarker in a biological sample that is greater than a value or level (or range of values or levels) of the biomarker that is typically detected in similar biological samples from healthy or normal individuals. The terms may also refer to a value or level of a biomarker in a biological sample that is greater than a value or level (or range of values or levels) of the biomarker that may be detected at a different stage of a particular disease.
[00102] "Down-regulation", "down-regulated", "under-expression", "under-expressed", and any variations thereof are used interchangeably to refer to a value or level of a biomarker in a biological sample that is less than a value or level (or range of values or levels) of the biomarker that is typically detected in similar biological samples from healthy or normal individuals. The terms may also refer to a value or level of a biomarker in a biological sample that is less than a value or level (or range of values or levels) of the biomarker that may be detected at a different stage of a particular disease.
[00103] Further, a biomarker that is either over-expressed or under-expressed can also be referred to as being "differentially expressed" or as having a "differential level" or "differential value" as compared to a "normal" expression level or value of the biomarker that indicates or is a sign of a normal process or an absence of a disease or other condition in an individual. Thus, "differential expression" of a biomarker can also be referred to as a variation from a "normal" expression level of the biomarker. [00104] The term "differential gene expression" and "differential expression" are used interchangeably to refer to a gene (or its corresponding protein expression product) whose expression is activated to a higher or lower level in a subject suffering from a specific disease, relative to its expression in a normal or control subject. The terms also include genes (or the corresponding protein expression products) whose expression is activated to a higher or lower level at different stages of the same disease. It is also understood that a differentially expressed gene may be either activated or inhibited at the nucleic acid level or protein level, or may be subject to alternative splicing to result in a different polypeptide product. Such differences may be evidenced by a variety of changes including mRNA levels, surface expression, secretion or other partitioning of a polypeptide. Differential gene expression may include a comparison of expression between two or more genes or their gene products; or a comparison of the ratios of the expression between two or more genes or their gene products; or even a comparison of two differently processed products of the same gene, which differ between normal subjects and subjects suffering from a disease; or between various stages of the same disease. Differential expression includes both quantitative, as well as qualitative, differences in the temporal or cellular expression pattern in a gene or its expression products among, for example, normal and diseased cells, or among cells which have undergone different disease events or disease stages.
[00105] As used herein, "individual" refers to a test subject or patient. The individual can be a mammal or a non-mammal. In various embodiments, the individual is a mammal. A mammalian individual can be a human or non-human. In various embodiments, the individual is a human. A healthy or normal individual is an individual in which the disease or condition of interest (including, for example, ovarian diseases, ovarian -associated diseases, or other ovarian conditions) is not detectable by conventional diagnostic methods. [00106] "Diagnose", "diagnosing", "diagnosis", and variations thereof refer to the detection, determination, or recognition of a health status or condition of an individual on the basis of one or more signs, symptoms, data, or other information pertaining to that individual. The health status of an individual can be diagnosed as healthy / normal (i.e., a diagnosis of the absence of a disease or condition) or diagnosed as ill / abnormal (i.e., a diagnosis of the presence, or an assessment of the characteristics, of a disease or condition). The terms "diagnose", "diagnosing", "diagnosis", etc., encompass, with respect to a particular disease or condition, the initial detection of the disease; the characterization or classification of the disease; the detection of the progression, remission, or recurrence of the disease; and the detection of disease response after the administration of a treatment or therapy to the individual. The diagnosis of ovarian cancer includes distinguishing individuals who have cancer from individuals who do not. It further includes distinguishing benign pelvic masses from ovarian cancer.
[00107] "Prognose", "prognosing", "prognosis", and variations thereof refer to the prediction of a future course of a disease or condition in an individual who has the disease or condition (e.g., predicting patient survival), and such terms encompass the evaluation of disease response after the administration of a treatment or therapy to the individual. [00108] "Evaluate", "evaluating", "evaluation", and variations thereof encompass both
"diagnose" and "prognose" and also encompass determinations or predictions about the future course of a disease or condition in an individual who does not have the disease as well as determinations or predictions regarding the likelihood that a disease or condition will recur in an individual who apparently has been cured of the disease. The term "evaluate" also encompasses assessing an individual's response to a therapy, such as, for example, predicting whether an individual is likely to respond favorably to a therapeutic agent or is unlikely to respond to a therapeutic agent (or will experience toxic or other undesirable side effects, for example), selecting a therapeutic agent for administration to an individual, or monitoring or determining an individual's response to a therapy that has been administered to the individual. Thus, "evaluating" ovarian cancer can include, for example, any of the following: prognosing the future course of ovarian cancer in an individual; predicting the recurrence of ovarian cancer in an individual who apparently has been cured of ovarian cancer; or determining or predicting an individual's response to an ovarian cancer treatment or selecting an ovarian cancer treatment to administer to an individual based upon a determination of the biomarker values derived from the individual's biological sample.
[00109] Any of the following examples may be referred to as either "diagnosing" or
"evaluating" ovarian cancer: initially detecting the presence or absence of ovarian cancer; determining a specific stage, type or sub-type, or other classification or characteristic of ovarian cancer; determining whether a pelvic mass is benign or malignant; or detecting or monitoring ovarian cancer progression (e.g., monitoring ovarian tumor growth or metastatic spread), remission, or recurrence.
[00110] As used herein, "additional biomedical information" refers to one or more evaluations of an individual, other than using any of the biomarkers described herein, that are associated with ovarian cancer risk. "Additional biomedical information" includes any of the following: physical descriptors of an individual; physical descriptors of a pelvic mass observed by MRI, abdominal ultrasound, or CT imaging; the height and/or weight of an individual; change in weight; the ethnicity of an individual; occupational history; family history of ovarian cancer (or other cancer); the presence of a genetic marker(s) correlating with a higher risk of ovarian cancer in the individual or a family member; the presence of a pelvic mass; size of mass; location of mass; morphology of mass and associated pelvic region (e.g., as observed through imaging); clinical symptoms such as bloating, abdominal pain, or sudden weight gain or loss; and the like. Additional biomedical information can be obtained from an individual using routine techniques known in the art, such as from the individual themselves by use of a routine patient questionnaire or health history questionnaire, etc., or from a medical practitioner, etc. Alternately, additional biomedical information can be obtained from routine imaging techniques, including abdominal or transvaginal ultrasound, MRI, CT imaging, and PET-CT. Testing of biomarker levels in combination with an evaluation of any additional biomedical information, including other laboratory tests (e.g., CA- 125 testing), may, for example, improve sensitivity, specificity, and/or AUC for detecting ovarian cancer (or other ovarian cancer-related uses) as compared to biomarker testing alone or evaluating any particular item of additional biomedical information alone (e.g., ultrasound imaging alone).
[00111] The term "area under the curve" or "AUC" refers to the area under the curve of a receiver operating characteristic (ROC) curve, both of which are well known in the art. AUC measures are useful for comparing the accuracy of a classifier across the complete data range. Classifiers with a greater AUC have a greater capacity to classify unknowns correctly between two groups of interest (e.g., ovarian cancer samples and normal or control samples). ROC curves are useful for plotting the performance of a particular feature (e.g., any of the biomarkers described herein and/or any item of additional biomedical information) in distinguishing between two populations (e.g., cases having ovarian cancer and controls without ovarian cancer). Typically, the feature data across the entire population (e.g., the cases and controls) are sorted in ascending order based on the value of a single feature. Then, for each value for that feature, the true positive and false positive rates for the data are calculated. The true positive rate is determined by counting the number of cases above the value for that feature and then dividing by the total number of cases. The false positive rate is determined by counting the number of controls above the value for that feature and then dividing by the total number of controls. Although this definition refers to scenarios in which a feature is elevated in cases compared to controls, this definition also applies to scenarios in which a feature is lower in cases compared to the controls (in such a scenario, samples below the value for that feature would be counted). ROC curves can be generated for a single feature as well as for other single outputs, for example, a combination of two or more features can be mathematically combined (e.g., added, subtracted, multiplied, etc.) to provide a single sum value, and this single sum value can be plotted in a ROC curve. Additionally, any combination of multiple features, in which the combination derives a single output value, can be plotted in a ROC curve. These combinations of features may comprise a test. The ROC curve is the plot of the true positive rate (sensitivity) of a test against the false positive rate (1 -specificity) of the test.
[00112] As used herein, "detecting" or "determining" with respect to a biomarker value includes the use of both the instrument required to observe and record a signal corresponding to a biomarker value and the material/s required to generate that signal. In various embodiments, the biomarker value is detected using any suitable method, including fluorescence, chemiluminescence, surface plasmon resonance, surface acoustic waves, mass spectrometry, infrared spectroscopy, Raman spectroscopy, atomic force microscopy, scanning tunneling microscopy, electrochemical detection methods, nuclear magnetic resonance, quantum dots, and the like.
[00113] "Solid support" refers herein to any substrate having a surface to which molecules may be attached, directly or indirectly, through either covalent or non-covalent bonds. A "solid support" can have a variety of physical formats, which can include, for example, a membrane; a chip (e.g., a protein chip); a slide (e.g., a glass slide or coverslip); a column; a hollow, solid, semi-solid, pore- or cavity- containing particle, such as, for example, a bead; a gel; a fiber, including a fiber optic material; a matrix; and a sample receptacle. Exemplary sample receptacles include sample wells, tubes, capillaries, vials, and any other vessel, groove or indentation capable of holding a sample. A sample receptacle can be contained on a multi-sample platform, such as a microtiter plate, slide, microfluidics device, and the like. A support can be composed of a natural or synthetic material, an organic or inorganic material. The composition of the solid support on which capture reagents are attached generally depends on the method of attachment (e.g., covalent attachment). Other exemplary receptacles include microdroplets and microfluidic controlled or bulk oil/aqueous emulsions within which assays and related manipulations can occur. Suitable solid supports include, for example, plastics, resins, polysaccharides, silica or silica-based materials, functionalized glass, modified silicon, carbon, metals, inorganic glasses, membranes, nylon, natural fibers (such as, for example, silk, wool and cotton), polymers, and the like. The material composing the solid support can include reactive groups such as, for example, carboxy, amino, or hydroxyl groups, which are used for attachment of the capture reagents. Polymeric solid supports can include, e.g., polystyrene, polyethylene glycol tetraphthalate, polyvinyl acetate, polyvinyl chloride, polyvinyl pyrrolidone, polyacrylonitrile, polymethyl methacrylate, polytetrafluoroethylene, butyl rubber, styrenebutadiene rubber, natural rubber, polyethylene, polypropylene, (poly)tetrafluoroethylene, (poly)vinylidenefluoride, polycarbonate, and polymethylpentene. Suitable solid support particles that can be used include, e.g., encoded particles, such as Luminex®-type encoded particles, magnetic particles, and glass particles.
Exemplary Uses of Biomarkers
[00114] In various exemplary embodiments, methods are provided for diagnosing ovarian cancer in an individual by detecting one or more biomarker values corresponding to one or more biomarkers that are present in the circulation of an individual, such as in serum or plasma, by any number of analytical methods, including any of the analytical methods described herein. These biomarkers are, for example, differentially expressed in individuals with ovarian cancer as compared to individuals without ovarian cancer. Detection of the differential expression of a biomarker in an individual can be used, for example, to permit the early diagnosis of ovarian cancer, to distinguish between a benign pelvic mass and ovarian cancer (such as, for example, a mass observed on an abdominal ultrasound or computed tomography (CT) scan), to monitor ovarian cancer recurrence, or for other clinical indications.
[00115] Any of the biomarkers described herein may be used in a variety of clinical indications for ovarian cancer, including any of the following: detection of ovarian cancer (such as in a high-risk individual or population); characterizing ovarian cancer (e.g., determining ovarian cancer type, sub-type, or stage), such as by determining whether a pelvic mass is benign or malignant; determining ovarian cancer prognosis; monitoring ovarian cancer progression or remission; monitoring for ovarian cancer recurrence; monitoring metastasis; treatment selection (e.g., pre- or post-operative chemotherapy selection); monitoring response to a therapeutic agent or other treatment; combining biomarker testing with additional biomedical information, such as CA- 125 level, the presence of a genetic marker(s) indicating a higher risk for ovarian cancer, etc., or with mass size, morphology, presence of ascites, etc. (such as to provide an assay with increased diagnostic performance compared to CA-125 testing or other biomarker testing alone); facilitating the diagnosis of a pelvic mass as malignant or benign; facilitating clinical decision making once a pelvic mass is observed through imaging; and facilitating decisions regarding clinical follow-up (e.g., whether to refer an individual to a surgical specialist, such as a gynecologic oncology surgeon). Biomarker testing may improve positive predictive value (PPV) over CA- 125 testing and imaging alone. Furthermore, the described biomarkers may also be useful in permitting certain of these uses before indications of ovarian cancer are detected by imaging modalities or other clinical correlates, or before symptoms appear.
[00116] As an example of the manner in which any of the biomarkers described herein can be used to diagnose ovarian cancer, differential expression of one or more of the described biomarkers in an individual who is not known to have ovarian cancer may indicate that the individual has ovarian cancer, thereby enabling detection of ovarian cancer at an early stage of the disease when treatment is most effective, perhaps before the ovarian cancer is detected by other means or before symptoms appear. Increased differential expression from "normal" (since some biomarkers may be down-regulated with disease) of one or more of the biomarkers during the course of ovarian cancer may be indicative of ovarian cancer progression, e.g., an ovarian tumor is growing and/or metastasizing (and thus indicate a poor prognosis), whereas a decrease in the degree to which one or more of the biomarkers is differentially expressed (i.e., in subsequent biomarker tests, the expression level in the individual is moving toward or approaching a "normal" expression level) may be indicative of ovarian cancer remission, e.g., an ovarian tumor is shrinking (and thus indicate a good or better prognosis). Similarly, an increase in the degree to which one or more of the biomarkers is differentially expressed (i.e., in subsequent biomarker tests, the expression level in the individual is moving further away from a "normal" expression level) during the course of ovarian cancer treatment may indicate that the ovarian cancer is progressing and therefore indicate that the treatment is ineffective, whereas a decrease in differential expression of one or more of the biomarkers during the course of ovarian cancer treatment may be indicative of ovarian cancer remission and therefore indicate that the treatment is working successfully. Additionally, an increase or decrease in the differential expression of one or more of the biomarkers after an individual has apparently been cured of ovarian cancer may be indicative of ovarian cancer recurrence. In a situation such as this, for example, the individual can be re- started on therapy (or the therapeutic regimen modified such as to increase dosage amount and/or frequency, if the individual has maintained therapy) at an earlier stage than if the recurrence of ovarian cancer was not detected until later. Furthermore, a differential expression level of one or more of the biomarkers in an individual may be predictive of the individual's response to a particular therapeutic agent. In monitoring for ovarian cancer recurrence or progression, changes in the biomarker expression levels may indicate the need for repeat imaging, such as to determine ovarian cancer activity or to determine the need for changes in treatment.
[00117] Detection of any of the biomarkers described herein may be particularly useful following, or in conjunction with, ovarian cancer treatment, such as to evaluate the success of the treatment or to monitor ovarian cancer remission, recurrence, and/or progression (including metastasis) following treatment. Ovarian cancer treatment may include, for example, administration of a therapeutic agent to the individual, performance of surgery (e.g., surgical resection of at least a portion of a pelvic mass), administration of radiation therapy, or any other type of ovarian cancer treatment used in the art, and any combination of these treatments. For example, any of the biomarkers may be detected at least once after treatment or may be detected multiple times after treatment (such as at periodic intervals), or may be detected both before and after treatment. Differential expression levels of any of the biomarkers in an individual over time may be indicative of ovarian cancer progression, remission, or recurrence, examples of which include any of the following: an increase or decrease in the expression level of the biomarkers after treatment compared with the expression level of the biomarker before treatment; an increase or decrease in the expression level of the biomarker at a later time point after treatment compared with the expression level of the biomarker at an earlier time point after treatment; and a differential expression level of the biomarker at a single time point after treatment compared with normal levels of the biomarker.
[00118] As a specific example, the biomarker levels for any of the biomarkers described herein can be determined in pre-surgery and post-surgery (e.g., 2-8 weeks after surgery) serum or plasma samples. An increase in the biomarker expression level(s) in the post-surgery sample compared with the pre-surgery sample can indicate progression of ovarian cancer (e.g., unsuccessful surgery), whereas a decrease in the biomarker expression level(s) in the post-surgery sample compared with the pre-surgery sample can indicate regression of ovarian cancer (e.g., the surgery successfully removed the ovarian tumor). Similar analyses of the biomarker levels can be carried out before and after other forms of treatment, such as before and after radiation therapy or administration of a therapeutic agent or cancer vaccine.
[00119] In addition to testing biomarker levels as a stand-alone diagnostic test, biomarker levels can also be done in conjunction with determination of SNPs or other genetic lesions or variability that are indicative of increased risk of susceptibility of disease. (See, e.g., Amos et al., Nature Genetics 40, 616-622 (2009)). [00120] In addition to testing biomarker levels as a stand-alone diagnostic test, biomarker levels can also be done in conjunction with relevant symptoms or abdominal ultrasound and CT imaging.
[00121] Detection of any of the biomarkers described herein may be useful after a pelvic mass has been observed through imaging to aid in the diagnosis of ovarian cancer and guide appropriate clinical care of the individual, including care by an appropriate surgical specialist.
[00122] In addition to testing biomarker levels in conjunction with relevant symptoms or abdominal ultrasound or CT imaging, information regarding the biomarkers can also be evaluated in conjunction with other types of data, particularly data that indicates an individual's risk for ovarian cancer (e.g., patient clinical history, symptoms, family history of cancer, risk factors such as the presence of a genetic marker(s), and/or status of other biomarkers, etc.). These various data can be assessed by automated methods, such as a computer program/software, which can be embodied in a computer or other apparatus/device. [00123] Any of the described biomarkers may also be used in imaging tests. For example, an imaging agent can be coupled to any of the described biomarkers, which can be used to aid in ovarian cancer diagnosis, to monitor disease progression/remission or metastasis, to monitor for disease recurrence, or to monitor response to therapy, among other uses.
Detection and Determination of Biomarkers and Biomarker Values
[00124] A biomarker value for the biomarkers described herein can be detected using any of a variety of known analytical methods. In one embodiment, a biomarker value is detected using a capture reagent. As used herein, a "capture agent" or "capture reagent" refers to a molecule that is capable of binding specifically to a biomarker. In various embodiments, the capture reagent can be exposed to the biomarker in solution or can be exposed to the biomarker while the capture reagent is immobilized on a solid support. In other embodiments, the capture reagent contains a feature that is reactive with a secondary feature on a solid support. In these embodiments, the capture reagent can be exposed to the biomarker in solution, and then the feature on the capture reagent can be used in conjunction with the secondary feature on the solid support to immobilize the biomarker on the solid support. The capture reagent is selected based on the type of analysis to be conducted. Capture reagents include but are not limited to aptamers, antibodies, adnectins, ankyrins, other antibody mimetics and other protein scaffolds, autoantibodies, chimeras, small molecules, an F(ab')2 fragment, a single chain antibody fragment, an Fv fragment, a single chain Fv fragment, a nucleic acid, a lectin, a ligand-binding receptor, affybodies, nanobodies, imprinted polymers, avimers, peptidomimetics, a hormone receptor, a cytokine receptor, and synthetic receptors, and modifications and fragments of these.
[00125] In some embodiments, a biomarker value is detected using a biomarker/capture reagent complex.
[00126] In other embodiments, the biomarker value is derived from the biomarker/capture reagent complex and is detected indirectly, such as, for example, as a result of a reaction that is subsequent to the biomarker/capture reagent interaction, but is dependent on the formation of the biomarker/capture reagent complex.
[00127] In some embodiments, the biomarker value is detected directly from the biomarker in a biological sample.
[00128] In one embodiment, the biomarkers are detected using a multiplexed format that allows for the simultaneous detection of two or more biomarkers in a biological sample.
In one embodiment of the multiplexed format, capture reagents are immobilized, directly or indirectly, covalently or non-covalently, in discrete locations on a solid support. In another embodiment, a multiplexed format uses discrete solid supports where each solid support has a unique capture reagent associated with that solid support, such as, for example quantum dots.
In another embodiment, an individual device is used for the detection of each one of multiple biomarkers to be detected in a biological sample. Individual devices can be configured to permit each biomarker in the biological sample to be processed simultaneously. For example, a microtiter plate can be used such that each well in the plate is used to uniquely analyze one of multiple biomarkers to be detected in a biological sample.
[00129] In one or more of the foregoing embodiments, a fluorescent tag can be used to label a component of the biomarker/capture complex to enable the detection of the biomarker value. In various embodiments, the fluorescent label can be conjugated to a capture reagent specific to any of the biomarkers described herein using known techniques, and the fluorescent label can then be used to detect the corresponding biomarker value. Suitable fluorescent labels include rare earth chelates, fluorescein and its derivatives, rhodamine and its derivatives, dansyl, allophycocyanin, PBXL-3, Qdot 605, Lissamine, phycoerythrin, Texas
Red, and other such compounds.
[00130] In one embodiment, the fluorescent label is a fluorescent dye molecule. In some embodiments, the fluorescent dye molecule includes at least one substituted indolium ring system in which the substituent on the 3-carbon of the indolium ring contains a chemically reactive group or a conjugated substance. In some embodiments, the dye molecule includes an AlexFluor molecule, such as, for example, AlexaFluor 488, AlexaFluor 532, AlexaFluor 647, AlexaFluor 680, or AlexaFluor 700. In other embodiments, the dye molecule includes a first type and a second type of dye molecule, such as, e.g., two different AlexaFluor molecules. In other embodiments, the dye molecule includes a first type and a second type of dye molecule, and the two dye molecules have different emission spectra. [00131] Fluorescence can be measured with a variety of instrumentation compatible with a wide range of assay formats. For example, spectrofluorimeters have been designed to analyze microtiter plates, microscope slides, printed arrays, cuvettes, etc. See Principles of Fluorescence Spectroscopy, by J. R. Lakowicz, Springer Science + Business Media, Inc., 2004. See Bioluminescence & Chemiluminescence: Progress & Current Applications; Philip E. Stanley and Larry J. Kricka editors, World Scientific Publishing Company, January 2002. [00132] In one or more of the foregoing embodiments, a chemiluminescence tag can optionally be used to label a component of the biomarker/capture complex to enable the detection of a biomarker value. Suitable chemiluminescent materials include any of oxalyl chloride, Rodamin 6G, Ru(bipy)3 2+ , TMAE (tetrakis(dimethylamino)ethylene), Pyrogallol (1,2,3-trihydroxibenzene), Lucigenin, peroxyoxalates, Aryl oxalates, Acridinium esters, dioxetanes, and others.
[00133] In yet other embodiments, the detection method includes an enzyme/substrate combination that generates a detectable signal that corresponds to the biomarker value. Generally, the enzyme catalyzes a chemical alteration of the chromogenic substrate which can be measured using various techniques, including spectrophotometry, fluorescence, and chemiluminescence. Suitable enzymes include, for example, luciferases, luciferin, malate dehydrogenase, urease, horseradish peroxidase (HRPO), alkaline phosphatase, beta- galactosidase, glucoamylase, lysozyme, glucose oxidase, galactose oxidase, and glucose-6- phosphate dehydrogenase, uricase, xanthine oxidase, lactoperoxidase, microperoxidase, and the like.
[00134] In yet other embodiments, the detection method can be a combination of fluorescence, chemiluminescence, radionuclide or enzyme/substrate combinations that generate a measurable signal. Multimodal signaling could have unique and advantageous characteristics in biomarker assay formats.
[00135] More specifically, the biomarker values for the biomarkers described herein can be detected using known analytical methods including, singleplex aptamer assays, multiplexed aptamer assays, singleplex or multiplexed immunoassays, mRNA expression profiling, miRNA expression profiling, mass spectrometric analysis, histological/cytological methods, etc. as detailed below.
Determination of Biomarker Values using Aptamer-Based Assays [00136] Assays directed to the detection and quantification of physiologically significant molecules in biological samples and other samples are important tools in scientific research and in the health care field. One class of such assays involves the use of a microarray that includes one or more aptamers immobilized on a solid support. The aptamers are each capable of binding to a target molecule in a highly specific manner and with very high affinity. See, e.g., U.S. Patent No. 5,475,096 entitled "Nucleic Acid Ligands"; see also, e.g., U.S. Patent No. 6,242,246, U.S. Patent No. 6,458,543, and U.S. Patent No. 6,503,715, each of which is entitled "Nucleic Acid Ligand Diagnostic Biochip". Once the microarray is contacted with a sample, the aptamers bind to their respective target molecules present in the sample and thereby enable a determination of a biomarker value corresponding to a biomarker.
[00137] As used herein, an "aptamer" refers to a nucleic acid that has a specific binding affinity for a target molecule. It is recognized that affinity interactions are a matter of degree; however, in this context, the "specific binding affinity" of an aptamer for its target means that the aptamer binds to its target generally with a much higher degree of affinity than it binds to other components in a test sample. An "aptamer" is a set of copies of one type or species of nucleic acid molecule that has a particular nucleotide sequence. An aptamer can include any suitable number of nucleotides, including any number of chemically modified nucleotides. "Aptamers" refers to more than one such set of molecules. Different aptamers can have either the same or different numbers of nucleotides. Aptamers can be DNA or RNA or chemically modified nucleic acids and can be single stranded, double stranded, or contain double stranded regions, and can include higher ordered structures. An aptamer can also be a photoaptamer, where a photoreactive or chemically reactive functional group is included in the aptamer to allow it to be covalently linked to its corresponding target. Any of the aptamer methods disclosed herein can include the use of two or more aptamers that specifically bind the same target molecule. As further described below, an aptamer may include a tag. If an aptamer includes a tag, all copies of the aptamer need not have the same tag. Moreover, if different aptamers each include a tag, these different aptamers can have either the same tag or a different tag. [00138] An aptamer can be identified using any known method, including the SELEX process. Once identified, an aptamer can be prepared or synthesized in accordance with any known method, including chemical synthetic methods and enzymatic synthetic methods. [00139] The terms "SELEX" and "SELEX process" are used interchangeably herein to refer generally to a combination of (1) the selection of aptamers that interact with a target molecule in a desirable manner, for example binding with high affinity to a protein, with (2) the amplification of those selected nucleic acids. The SELEX process can be used to identify aptamers with high affinity to a specific target or biomarker.
[00140] SELEX generally includes preparing a candidate mixture of nucleic acids, binding of the candidate mixture to the desired target molecule to form an affinity complex, separating the affinity complexes from the unbound candidate nucleic acids, separating and isolating the nucleic acid from the affinity complex, purifying the nucleic acid, and identifying a specific aptamer sequence. The process may include multiple rounds to further refine the affinity of the selected aptamer. The process can include amplification steps at one or more points in the process. See, e.g., U.S. Patent No. 5,475,096, entitled "Nucleic Acid Ligands". The SELEX process can be used to generate an aptamer that covalently binds its target as well as an aptamer that non-covalently binds its target. See, e.g., U.S. Patent No. 5,705,337 entitled "Systematic Evolution of Nucleic Acid Ligands by Exponential Enrichment: Chemi-SELEX."
[00141] The SELEX process can be used to identify high-affinity aptamers containing modified nucleotides that confer improved characteristics on the aptamer, such as, for example, improved in vivo stability or improved delivery characteristics. Examples of such modifications include chemical substitutions at the ribose and/or phosphate and/or base positions. SELEX process -identified aptamers containing modified nucleotides are described in U.S. Patent No. 5,660,985, entitled "High Affinity Nucleic Acid Ligands Containing Modified Nucleotides", which describes oligonucleotides containing nucleotide derivatives chemically modified at the 5'- and 2'-positions of pyrimidines. U.S. Patent No. 5,580,737, see supra, describes highly specific aptamers containing one or more nucleotides modified with 2'-amino (2'-NH2), 2'-fluoro (2'-F), and/or 2'-O-methyl (2'-OMe). See also, U.S. Patent Application Publication 20090098549, entitled "SELEX and PHOTOSELEX", which describes nucleic acid libraries having expanded physical and chemical properties and their use in SELEX and photoSELEX.
[00142] SELEX can also be used to identify aptamers that have desirable off -rate characteristics. See U.S. Patent Application Publication 20090004667, entitled "Method for Generating Aptamers with Improved Off-Rates", which describes improved SELEX methods for generating aptamers that can bind to target molecules. Methods for producing aptamers and photoaptamers having slower rates of dissociation from their respective target molecules are described. The methods involve contacting the candidate mixture with the target molecule, allowing the formation of nucleic acid-target complexes to occur, and performing a slow off -rate enrichment process wherein nucleic acid- target complexes with fast dissociation rates will dissociate and not reform, while complexes with slow dissociation rates will remain intact. Additionally, the methods include the use of modified nucleotides in the production of candidate nucleic acid mixtures to generate aptamers with improved off -rate performance. [00143] A variation of this assay employs aptamers that include photoreactive functional groups that enable the aptamers to covalently bind or "photocrosslink" their target molecules. See, e.g., U.S. Patent No. 6,544,776 entitled "Nucleic Acid Ligand Diagnostic Biochip". These photoreactive aptamers are also referred to as photoaptamers. See, e.g., U.S. Patent No. 5,763,177, U.S. Patent No. 6,001,577, and U.S. Patent No. 6,291,184, each of which is entitled "Systematic Evolution of Nucleic Acid Ligands by Exponential Enrichment: Photo selection of Nucleic Acid Ligands and Solution SELEX"; see also, e.g., U.S. Patent No. 6,458,539, entitled "Photoselection of Nucleic Acid Ligands". After the microarray is contacted with the sample and the photoaptamers have had an opportunity to bind to their target molecules, the photoaptamers are photoactivated, and the solid support is washed to remove any non-specifically bound molecules. Harsh wash conditions may be used, since target molecules that are bound to the photoaptamers are generally not removed, due to the covalent bonds created by the photoactivated functional group(s) on the photoaptamers. In this manner, the assay enables the detection of a biomarker value corresponding to a biomarker in the test sample.
[00144] In both of these assay formats, the aptamers are immobilized on the solid support prior to being contacted with the sample. Under certain circumstances, however, immobilization of the aptamers prior to contact with the sample may not provide an optimal assay. For example, pre-immobilization of the aptamers may result in inefficient mixing of the aptamers with the target molecules on the surface of the solid support, perhaps leading to lengthy reaction times and, therefore, extended incubation periods to permit efficient binding of the aptamers to their target molecules. Further, when photoaptamers are employed in the assay and depending upon the material utilized as a solid support, the solid support may tend to scatter or absorb the light used to effect the formation of covalent bonds between the photoaptamers and their target molecules. Moreover, depending upon the method employed, detection of target molecules bound to their aptamers can be subject to imprecision, since the surface of the solid support may also be exposed to and affected by any labeling agents that are used. Finally, immobilization of the aptamers on the solid support generally involves an aptamer-preparation step (i.e., the immobilization) prior to exposure of the aptamers to the sample, and this preparation step may affect the activity or functionality of the aptamers. [00145] Aptamer assays that permit an aptamer to capture its target in solution and then employ separation steps that are designed to remove specific components of the aptamer-target mixture prior to detection have also been described (see U.S. Patent Application Publication 20090042206, entitled "Multiplexed Analyses of Test Samples"). The described aptamer assay methods enable the detection and quantification of a non- nucleic acid target (e.g., a protein target) in a test sample by detecting and quantifying a nucleic acid (i.e., an aptamer). The described methods create a nucleic acid surrogate (i.e, the aptamer) for detecting and quantifying a non-nucleic acid target, thus allowing the wide variety of nucleic acid technologies, including amplification, to be applied to a broader range of desired targets, including protein targets.
[00146] Aptamers can be constructed to facilitate the separation of the assay components from an aptamer biomarker complex (or photoaptamer biomarker covalent complex) and permit isolation of the aptamer for detection and/or quantification. In one embodiment, these constructs can include a cleavable or releasable element within the aptamer sequence. In other embodiments, additional functionality can be introduced into the aptamer, for example, a labeled or detectable component, a spacer component, or a specific binding tag or immobilization element. For example, the aptamer can include a tag connected to the aptamer via a cleavable moiety, a label, a spacer component separating the label, and the cleavable moiety. In one embodiment, a cleavable element is a photocleavable linker. The photocleavable linker can be attached to a biotin moiety and a spacer section, can include an NHS group for derivatization of amines, and can be used to introduce a biotin group to an aptamer, thereby allowing for the release of the aptamer later in an assay method. [00147] Homogenous assays, done with all assay components in solution, do not require separation of sample and reagents prior to the detection of signal. These methods are rapid and easy to use. These methods generate signal based on a molecular capture or binding reagent that reacts with its specific target. For ovarian cancer, the molecular capture reagents would be an aptamer or an antibody or the like and the specific target would be an ovarian cancer biomarker of Table 1. [00148] In one embodiment, a method for signal generation takes advantage of anisotropy signal change due to the interaction of a fluorophore-labeled capture reagent with its specific biomarker target. When the labeled capture reacts with its target, the increased molecular weight causes the rotational motion of the fluorophore attached to the complex to become much slower changing the anisotropy value. By monitoring the anisotropy change, binding events may be used to quantitatively measure the biomarkers in solutions. Other methods include fluorescence polarization assays, molecular beacon methods, time resolved fluorescence quenching, chemiluminescence, fluorescence resonance energy transfer, and the like.
[00149] An exemplary solution-based aptamer assay that can be used to detect a biomarker value corresponding to a biomarker in a biological sample includes the following: (a) preparing a mixture by contacting the biological sample with an aptamer that includes a first tag and has a specific affinity for the biomarker, wherein an aptamer affinity complex is formed when the biomarker is present in the sample; (b) exposing the mixture to a first solid support including a first capture element, and allowing the first tag to associate with the first capture element; (c) removing any components of the mixture not associated with the first solid support; (d) attaching a second tag to the biomarker component of the aptamer affinity complex; (e) releasing the aptamer affinity complex from the first solid support; (f) exposing the released aptamer affinity complex to a second solid support that includes a second capture element and allowing the second tag to associate with the second capture element; (g) removing any non-complexed aptamer from the mixture by partitioning the non-complexed aptamer from the aptamer affinity complex; (h) eluting the aptamer from the solid support; and (i) detecting the biomarker by detecting the aptamer component of the aptamer affinity complex.
Determination of Biomarker Values using Immunoassays
[00150] Immunoassay methods are based on the reaction of an antibody to its corresponding target or analyte and can detect the analyte in a sample depending on the specific assay format. To improve specificity and sensitivity of an assay method based on immuno-reactivity, monoclonal antibodies are often used because of their specific epitope recognition. Polyclonal antibodies have also been successfully used in various immunoassays because of their increased affinity for the target as compared to monoclonal antibodies. Immunoassays have been designed for use with a wide range of biological sample matrices. Immunoassay formats have been designed to provide qualitative, semiquantitative, and quantitative results.
[00151] Quantitative results are generated through the use of a standard curve created with known concentrations of the specific analyte to be detected. The response or signal from an unknown sample is plotted onto the standard curve, and a quantity or value corresponding to the target in the unknown sample is established.
[00152] Numerous immunoassay formats have been designed. ELISA or EIA can be quantitative for the detection of an analyte. This method relies on attachment of a label to either the analyte or the antibody and the label component includes, either directly or indirectly, an enzyme. ELISA tests may be formatted for direct, indirect, competitive, or sandwich detection of the analyte. Other methods rely on labels such as, for example, radioisotopes (I125) or fluorescence. Additional techniques include, for example, agglutination, nephelometry, turbidimetry, Western blot, immunoprecipitation, immunocytochemistry, immunohistochemistry, flow cytometry, Luminex assay, and others (see ImmunoAssay: A Practical Guide, edited by Brian Law, published by Taylor & Francis, Ltd., 2005 edition).
[00153] Exemplary assay formats include enzyme-linked immunosorbent assay
(ELISA), radioimmunoassay, fluorescent, chemiluminescence, and fluorescence resonance energy transfer (FRET) or time resolved- FRET (TR-FRET) immunoassays. Examples of procedures for detecting biomarkers include biomarker immunoprecipitation followed by quantitative methods that allow size and peptide level discrimination, such as gel electrophoresis, capillary electrophoresis, planar electrochromatography, and the like. [00154] Methods of detecting and/or quantifying a detectable label or signal generating material depend on the nature of the label. The products of reactions catalyzed by appropriate enzymes (where the detectable label is an enzyme; see above) can be, without limitation, fluorescent, luminescent, or radioactive or they may absorb visible or ultraviolet light. Examples of detectors suitable for detecting such detectable labels include, without limitation, x-ray film, radioactivity counters, scintillation counters, spectrophotometers, colorimeters, fluorometers, luminometers, and densitometers.
[00155] Any of the methods for detection can be performed in any format that allows for any suitable preparation, processing, and analysis of the reactions. This can be, for example, in multi-well assay plates (e.g., 96 wells or 384 wells) or using any suitable array or microarray. Stock solutions for various agents can be made manually or robotically, and all subsequent pipetting, diluting, mixing, distribution, washing, incubating, sample readout, data collection and analysis can be done robotically using commercially available analysis software, robotics, and detection instrumentation capable of detecting a detectable label.
Determination of Biomarker Values using Gene Expression Profiling [00156] Measuring mRNA in a biological sample may be used as a surrogate for detection of the level of the corresponding protein in the biological sample. Thus, any of the biomarkers or biomarker panels described herein can also be detected by detecting the appropriate RNA.
[00157] mRNA expression levels are measured by reverse transcription quantitative polymerase chain reaction (RT-PCR followed with qPCR). RT-PCR is used to create a cDNA from the mRNA. The cDNA may be used in a qPCR assay to produce fluorescence as the DNA amplification process progresses. By comparison to a standard curve, qPCR can produce an absolute measurement such as number of copies of mRNA per cell. Northern blots, microarrays, Invader assays, and RT-PCR combined with capillary electrophoresis have all been used to measure expression levels of mRNA in a sample. See Gene Expression Profiling: Methods and Protocols, Richard A. Shimkets, editor, Humana Press, 2004. [00158] miRNA molecules are small RNAs that are non-coding but may regulate gene expression. Any of the methods suited to the measurement of mRNA expression levels can also be used for the corresponding miRNA. Recently many laboratories have investigated the use of miRNAs as biomarkers for disease. Many diseases involve wide-spread transcriptional regulation, and it is not surprising that miRNAs might find a role as biomarkers. The connection between miRNA concentrations and disease is often even less clear than the connections between protein levels and disease, yet the value of miRNA biomarkers might be substantial. Of course, as with any RNA expressed differentially during disease, the problems facing the development of an in vitro diagnostic product will include the requirement that the miRNAs survive in the diseased cell and are easily extracted for analysis, or that the miRNAs are released into blood or other matrices where they must survive long enough to be measured. Protein biomarkers have similar requirements, although many potential protein biomarkers are secreted intentionally at the site of pathology and function, during disease, in a paracrine fashion. Many potential protein biomarkers are designed to function outside the cells within which those proteins are synthesized.
Detection of Biomarkers Using In Vivo Molecular Imaging Technologies [00159] Any of the described biomarkers (see Table 1) may also be used in molecular imaging tests. For example, an imaging agent can be coupled to any of the described biomarkers, which can be used to aid in ovarian cancer diagnosis, to monitor disease progression/remission or metastasis, to monitor for disease recurrence, or to monitor response to therapy, among other uses.
[00160] In vivo imaging technologies provide non-invasive methods for determining the state of a particular disease in the body of an individual. For example, entire portions of the body, or even the entire body, may be viewed as a three dimensional image, thereby providing valuable information concerning morphology and structures in the body. Such technologies may be combined with the detection of the biomarkers described herein to provide information concerning the cancer status, in particular the ovarian cancer status, of an individual.
[00161] The use of in vivo molecular imaging technologies is expanding due to various advances in technology. These advances include the development of new contrast agents or labels, such as radiolabels and/or fluorescent labels, which can provide strong signals within the body; and the development of powerful new imaging technology, which can detect and analyze these signals from outside the body, with sufficient sensitivity and accuracy to provide useful information. The contrast agent can be visualized in an appropriate imaging system, thereby providing an image of the portion or portions of the body in which the contrast agent is located. The contrast agent may be bound to or associated with a capture reagent, such as an aptamer or an antibody, for example, and/or with a peptide or protein, or an oligonucleotide (for example, for the detection of gene expression), or a complex containing any of these with one or more macro molecules and/or other particulate forms. [00162] The contrast agent may also feature a radioactive atom that is useful in imaging. Suitable radioactive atoms include technetium-99m or iodine- 123 for scintigraphic studies. Other readily detectable moieties include, for example, spin labels for magnetic resonance imaging (MRI) such as, for example, iodine- 123 again, iodine-131, indium-I l l, fluorine-19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron. Such labels are well known in the art and could easily be selected by one of ordinary skill in the art. [00163] Standard imaging techniques include but are not limited to magnetic resonance imaging, contrast-enhanced abdominal or transvaginal ultrasound, computed tomography (CT) scanning, positron emission tomography (PET), single photon emission computed tomography (SPECT), and the like. For diagnostic in vivo imaging, the type of detection instrument available is a major factor in selecting a given contrast agent, such as a given radionuclide and the particular biomarker that it is used to target (protein, mRNA, and the like). The radionuclide chosen typically has a type of decay that is detectable by a given type of instrument. Also, when selecting a radionuclide for in vivo diagnosis, its half-life should be long enough to enable detection at the time of maximum uptake by the target tissue but short enough that deleterious radiation of the host is minimized.
[00164] Exemplary imaging techniques include but are not limited to PET and SPECT, which are imaging techniques in which a radionuclide is synthetically or locally administered to an individual. The subsequent uptake of the radiotracer is measured over time and used to obtain information about the targeted tissue and the biomarker. Because of the high-energy (gamma-ray) emissions of the specific isotopes employed and the sensitivity and sophistication of the instruments used to detect them, the two-dimensional distribution of radioactivity may be inferred from outside of the body.
[00165] Commonly used positron-emitting nuclides in PET include, for example, carbon-11, nitrogen-13, oxygen-15, and fluorine-18. Isotopes that decay by electron capture and/or gamma-emission are used in SPECT and include, for example iodine- 123 and technetium-99m. An exemplary method for labeling amino acids with technetium-99m is the reduction of pertechnetate ion in the presence of a chelating precursor to form the labile technetium-99m-precursor complex, which, in turn, reacts with the metal binding group of a bifunctionally modified chemotactic peptide to form a technetium-99m-chemotactic peptide conjugate.
[00166] Antibodies are frequently used for such in vivo imaging diagnostic methods.
The preparation and use of antibodies for in vivo diagnosis is well known in the art. Labeled antibodies which specifically bind any of the biomarkers in Table 1 can be injected into an individual suspected of having a certain type of cancer (e.g., ovarian cancer), detectable according to the particular biomarker used, for the purpose of diagnosing or evaluating the disease status of the individual. The label used will be selected in accordance with the imaging modality to be used, as previously described. Localization of the label permits determination of the spread of the cancer. The amount of label within an organ or tissue also allows determination of the presence or absence of cancer in that organ or tissue. [00167] Similarly, aptamers may be used for such in vivo imaging diagnostic methods.
For example, an aptamer that was used to identify a particular biomarker described in Table 1 (and therefore binds specifically to that particular biomarker) may be appropriately labeled and injected into an individual suspected of having ovarian cancer, detectable according to the particular biomarker, for the purpose of diagnosing or evaluating the ovarian cancer status of the individual. The label used will be selected in accordance with the imaging modality to be used, as previously described. Localization of the label permits determination of the spread of the cancer. The amount of label within an organ or tissue also allows determination of the presence or absence of cancer in that organ or tissue. Aptamer-directed imaging agents could have unique and advantageous characteristics relating to tissue penetration, tissue distribution, kinetics, elimination, potency, and selectivity as compared to other imaging agents.
[00168] Such techniques may also optionally be performed with labeled oligonucleotides, for example, for detection of gene expression through imaging with antisense oligonucleotides. These methods are used for in situ hybridization, for example, with fluorescent molecules or radionuclides as the label. Other methods for detection of gene expression include, for example, detection of the activity of a reporter gene.
[00169] Another general type of imaging technology is optical imaging, in which fluorescent signals within the subject are detected by an optical device that is external to the subject. These signals may be due to actual fluorescence and/or to bioluminescence.
Improvements in the sensitivity of optical detection devices have increased the usefulness of optical imaging for in vivo diagnostic assays.
[00170] The use of in vivo molecular biomarker imaging is increasing, including for clinical trials, for example, to more rapidly measure clinical efficacy in trials for new cancer therapies and/or to avoid prolonged treatment with a placebo for those diseases, such as multiple sclerosis, in which such prolonged treatment may be considered to be ethically questionable.
[00171] For a review of other techniques, see N. Blow, Nature Methods, 6, 465-469,
2009.
Determination of Biomarker Values using Histology or Cytology Methods
[00172] For evaluation of ovarian cancer, a variety of tissue samples may be used in histological or cytological methods. Sample selection depends on the primary tumor location and sites of metastases. For example, fine needle aspirates, cutting needles, and core biopsies can be used for histology. Ascites can be used for cyotology. While cytological analysis is still used in the diagnosis of ovarian cancer, histological methods are known to provide better sensitivity for the detection of cancer. Any of the biomarkers identified herein that were shown to be up-regulated (see Table 15) in the individuals with ovarian cancer can be used to stain a histological specimen as an indication of disease. [00173] In one embodiment, one or more capture reagents specific to the corresponding biomarker is used in a cytological evaluation of an ovarian cell sample and may include one or more of the following: collecting a cell sample, fixing the cell sample, dehydrating, clearing, immobilizing the cell sample on a microscope slide, permeabilizing the cell sample, treating for analyte retrieval, staining, destaining, washing, blocking, and reacting with one or more capture reagent/s in a buffered solution. In another embodiment, the cell sample is produced from a cell block.
[00174] In another embodiment, one or more capture reagents specific to the corresponding biomarker is used in a histological evaluation of an ovarian tissue sample and may include one or more of the following: collecting a tissue specimen, fixing the tissue sample, dehydrating, clearing, immobilizing the tissue sample on a microscope slide, permeabilizing the tissue sample, treating for analyte retrieval, staining, destaining, washing, blocking, rehydrating, and reacting with capture reagent/s in a buffered solution. In another embodiment, fixing and dehydrating are replaced with freezing. [00175] In another embodiment, the one or more aptamers specific to the corresponding biomarker is reacted with the histological or cytological sample and can serve as the nucleic acid target in a nucleic acid amplification method. Suitable nucleic acid amplification methods include, for example, PCR, q-beta replicase, rolling circle amplification, strand displacement, helicase dependent amplification, loop mediated isothermal amplification, ligase chain reaction, and restriction and circularization aided rolling circle amplification.
[00176] In one embodiment, the one or more capture reagent/s specific to the corresponding biomarkers for use in the histological or cytological evaluation are mixed in a buffered solution that can include any of the following: blocking materials, competitors, detergents, stabilizers, carrier nucleic acid, polyanionic materials, etc. [00177] A "cytology protocol" generally includes sample collection, sample fixation, sample immobilization, and staining. "Cell preparation" can include several processing steps after sample collection, including the use of one or more slow off -rate aptamers for the staining of the prepared cells.
[00178] Sample collection can include directly placing the sample in an untreated transport container, placing the sample in a transport container containing some type of media, or placing the sample directly onto a slide (immobilization) without any treatment or fixation. [00179] Sample immobilization can be improved by applying a portion of the collected specimen to a glass slide that is treated with polylysine, gelatin, or a silane. Slides can be prepared by smearing a thin and even layer of cells across the slide. Care is generally taken to minimize mechanical distortion and drying artifacts. Liquid specimens can be processed in a cell block method. Or, alternatively, liquid specimens can be mixed 1 : 1 with the fixative solution for about 10 minutes at room temperature.
[00180] Cell blocks can be prepared from residual effusions, sputum, urine sediments, gastrointestinal fluids, cell scraping, ascites, or fine needle aspirates. Cells are concentrated or packed by centrifugation or membrane filtration. A number of methods for cell block preparation have been developed. Representative procedures include the fixed sediment, bacterial agar, or membrane filtration methods. In the fixed sediment method, the cell sediment is mixed with a fixative like Bouins, picric acid, or buffered formalin and then the mixture is centrifuged to pellet the fixed cells. The supernatant is removed, drying the cell pellet as completely as possible. The pellet is collected and wrapped in lens paper and then placed in a tissue cassette. The tissue cassette is placed in ajar with additional fixative and processed as a tissue sample. Agar method is very similar but the pellet is removed and dried on paper towel and then cut in half. The cut side is placed in a drop of melted agar on a glass slide and then the pellet is covered with agar making sure that no bubbles form in the agar. The agar is allowed to harden and then any excess agar is trimmed away. This is placed in a tissue cassette and the tissue process completed. Alternatively, the pellet may be directly suspended in 2% liquid agar at 65°C and the sample centrifuged. The agar cell pellet is allowed to solidify for an hour at 4°C. The solid agar may be removed from the centrifuge tube and sliced in half. The agar is wrapped in filter paper and then the tissue cassette. Processing from this point forward is as described above. Centrifugation can be replaced in any these procedures with membrane filtration. Any of these processes may be used to generate a "cell block sample".
[00181] Cell blocks can be prepared using specialized resin including Lowicryl resins,
LR White, LR Gold, Unicryl, and MonoStep. These resins have low viscosity and can be polymerized at low temperatures and with ultra violet (UV) light. The embedding process relies on progressively cooling the sample during dehydration, transferring the sample to the resin, and polymerizing a block at the final low temperature at the appropriate UV wavelength. [00182] Cell block sections can be stained with hematoxylin-eosin for cytomorphological examination while additional sections are used for examination for specific markers.
[00183] Whether the process is cytologoical or histological, the sample may be fixed prior to additional processing to prevent sample degradation. This process is called "fixation" and describes a wide range of materials and procedures that may be used interchangeably. The sample fixation protocol and reagents are best selected empirically based on the targets to be detected and the specific cell/tissue type to be analyzed. Sample fixation relies on reagents such as ethanol, polyethylene glycol, methanol, formalin, or isopropanol. The samples should be fixed as soon after collection and affixation to the slide as possible. However, the fixative selected can introduce structural changes into various molecular targets making their subsequent detection more difficult. The fixation and immobilization processes and their sequence can modify the appearance of the cell and these changes must be anticipated and recognized by the cytotechnologist. Fixatives can cause shrinkage of certain cell types and cause the cytoplasm to appear granular or reticular. Many fixatives function by crosslinking cellular components. This can damage or modify specific epitopes, generate new epitopes, cause molecular associations, and reduce membrane permeability. Formalin fixation is one of the most common cytological and histological approaches. Formalin forms methyl bridges between neighboring proteins or within proteins. Precipitation or coagulation is also used for fixation and ethanol is frequently used in this type of fixation. A combination of crosslinking and precipitation can also be used for fixation. A strong fixation process is best at preserving morphological information while a weaker fixation process is best for the preservation of molecular targets.
[00184] A representative fixative is 50% absolute ethanol, 2 mM polyethylene glycol
(PEG), 1.85% formaldehyde. Variations on this formulation include ethanol (50% to 95%), methanol (20% - 50%), and formalin (formaldehyde) only. Another common fixative is 2% PEG 1500, 50% ethanol, and 3% methanol. Slides are place in the fixative for about 10 to 15 minutes at room temperature and then removed and allowed to dry. Once slides are fixed they can be rinsed with a buffered solution like PBS.
[00185] A wide range of dyes can be used to differentially highlight and contrast or
"stain" cellular, sub-cellular, and tissue features or morphological structures. Hematoylin is used to stain nuclei a blue or black color. Orange G-6 and Eosin Azure both stain the cell's cytoplasm. Orange G stains keratin and glycogen containing cells yellow. Eosin Y is used to stain nucleoli, cilia, red blood cells, and superficial epithelial squamous cells. Romanowsky stains are used for air dried slides and are useful in enhancing pleomorphism and distinguishing extracellular from intracytoplasmic material.
[00186] The staining process can include a treatment to increase the permeability of the cells to the stain. Treatment of the cells with a detergent can be used to increase permeability. To increase cell and tissue permeability, fixed samples can be further treated with solvents, saponins, or non-ionic detergents. Enzymatic digestion can also improve the accessibility of specific targets in a tissue sample.
[00187] After staining, the sample is dehydrated using a succession of alcohol rinses with increasing alcohol concentration. The final wash is done with xylene or a xylene substitute, such as a citrus terpene, that has a refractive index close to that of the coverslip to be applied to the slide. This final step is referred to as clearing. Once the sample is dehydrated and cleared, a mounting medium is applied. The mounting medium is selected to have a refractive index close to the glass and is capable of bonding the coverslip to the slide.
It will also inhibit the additional drying, shrinking, or fading of the cell sample.
[00188] Regardless of the stains or processing used, the final evaluation of the ovarian cytological specimen is made by some type of microscopy to permit a visual inspection of the morphology and a determination of the marker's presence or absence. Exemplary microscopic methods include brightfield, phase contrast, fluorescence, and differential interference contrast.
[00189] If secondary tests are required on the sample after examination, the coverslip may be removed and the slide destained. Destaining involves using the original solvent systems used in staining the slide originally without the added dye and in a reverse order to the original staining procedure. Destaining may also be completed by soaking the slide in an acid alcohol until the cells are colorless. Once colorless the slides are rinsed well in a water bath and the second staining procedure applied.
[00190] In addition, specific molecular differentiation may be possible in conjunction with the cellular morphological analysis through the use of specific molecular reagents such as antibodies or nucleic acid probes or aptamers. This improves the accuracy of diagnostic cytology. Micro-dissection can be used to isolate a subset of cells for additional evaluation, in particular, for genetic evaluation of abnormal chromosomes, gene expression, or mutations.
[00191] Preparation of a tissue sample for histological evaluation involves fixation, dehydration, infiltration, embedding, and sectioning. The fixation reagents used in histology are very similar or identical to those used in cytology and have the same issues of preserving morphological features at the expense of molecular ones such as individual proteins. Time can be saved if the tissue sample is not fixed and dehydrated but instead is frozen and then sectioned while frozen. This is a more gentle processing procedure and can preserve more individual markers. However, freezing is not acceptable for long term storage of a tissue sample as subcellular information is lost due to the introduction of ice crystals. Ice in the frozen tissue sample also prevents the sectioning process from producing a very thin slice and thus some microscopic resolution and imaging of subcellular structures can be lost. In addition to formalin fixation, osmium tetroxide is used to fix and stain phospholipids (membranes).
[00192] Dehydration of tissues is accomplished with successive washes of increasing alcohol concentration. Clearing employs a material that is miscible with alcohol and the embedding material and involves a stepwise process starting at 50:50 alcohol:clearing reagent and then 100% clearing agent (xylene or xylene substitute). Infiltration involves incubating the tissue with a liquid form of the embedding agent (warm wax, nitrocellulose solution) first at 50:50 embedding agent: clearing agent and the 100% embedding agent. Embedding is completed by placing the tissue in a mold or cassette and filling with melted embedding agent such as wax, agar, or gelatin. The embedding agent is allowed to harden. The hardened tissue sample may then be sliced into thin section for staining and subsequent examination. [00193] Prior to staining, the tissue section is dewaxed and rehydrated. Xylene is used to dewax the section, one or more changes of xylene may be used, and the tissue is rehydrated by successive washes in alcohol of decreasing concentration. Prior to dewax, the tissue section may be heat immobilized to a glass slide at about 80°C for about 20 minutes. [00194] Laser capture micro-dissection allows the isolation of a subset of cells for further analysis from a tissue section.
[00195] As in cytology, to enhance the visualization of the microscopic features, the tissue section or slice can be stained with a variety of stains. A large menu of commercially available stains can be used to enhance or identify specific features.
[00196] To further increase the interaction of molecular reagents with cytological or histological samples, a number of techniques for "analyte retrieval" have been developed. The first such technique uses high temperature heating of a fixed sample. This method is also referred to as heat-induced epitope retrieval or HIER. A variety of heating techniques have been used, including steam heating, microwaving, autoclaving, water baths, and pressure cooking or a combination of these methods of heating. Analyte retrieval solutions include, for example, water, citrate, and normal saline buffers. The key to analyte retrieval is the time at high temperature but lower temperatures for longer times have also been successfully used. Another key to analyte retrieval is the pH of the heating solution. Low pH has been found to provide the best immunostaining but also gives rise to backgrounds that frequently require the use of a second tissue section as a negative control. The most consistent benefit (increased immunostaining without increase in background) is generally obtained with a high pH solution regardless of the buffer composition. The analyte retrieval process for a specific target is empirically optimized for the target using heat, time, pH, and buffer composition as variables for process optimization. Using the microwave analyte retrieval method allows for sequential staining of different targets with antibody reagents. But the time required to achieve antibody and enzyme complexes between staining steps has also been shown to degrade cell membrane analytes. Microwave heating methods have improved in situ hybridization methods as well.
[00197] To initiate the analyte retrieval process, the section is first dewaxed and hydrated. The slide is then placed in 1OmM sodium citrate buffer pH 6.0 in a dish or jar. A representative procedure uses an 110OW microwave and microwaves the slide at 100% power for 2 minutes followed by microwaving the slides using 20% power for 18 minutes after checking to be sure the slide remains covered in liquid. The slide is then allowed to cool in the uncovered container and then rinsed with distilled water. HIER may be used in combination with an enzymatic digestion to improve the reactivity of the target to immunochemical reagents.
[00198] One such enzymatic digestion protocol uses proteinase K. A 20 μg/ml concentration of proteinase K is prepared in 50 mM Tris Base, ImM EDTA, 0.5% Triton X- 100, pH 8.0 buffer. The process first involves dewaxing sections in 2 changes of xylene, 5 minutes each. Then the sample is hydrated in 2 changes of 100% ethanol for 3 minutes each, 95% and 80% ethanol for 1 minute each, and then rinsed in distilled water. Sections are covered with Proteinase K working solution and incubated 10-20 minutes at 37°C in humidified chamber (optimal incubation time may vary depending on tissue type and degree of fixation). The sections are cooled at room temperature for 10 minutes and then rinsed in PBS Tween 20 for 2x2 min. If desired, sections can be blocked to eliminate potential interference from endogenous compounds and enzymes. The section is then incubated with primary antibody at appropriate dilution in primary antibody dilution buffer for 1 hour at room temperature or overnight at 4°C. The section is then rinsed with PBS Tween 20 for 2x2 min. Additional blocking can be performed, if required for the specific application, followed by additional rinsing with PBS Tween 20 for 3x2 min and then finally the immuno staining protocol completed.
[00199] A simple treatment with 1% SDS at room temperature has also been demonstrated to improve immunohistochemical staining. Analyte retrieval methods have been applied to slide mounted sections as well as free floating sections. Another treatment option is to place the slide in ajar containing citric acid and 0.1 Nonident P40 at pH 6.0 and heating to 95°C. The slide is then washed with a buffer solution like PBS. [00200] For immunological staining of tissues it may be useful to block non - specific association of the antibody with tissue proteins by soaking the section in a protein solution like serum or non-fat dry milk.
[00201] Blocking reactions may include the need to do any of the following, either alone or in combination: reduce the level of endogenous biotin; eliminate endogenous charge effects; inactivate endogenous nucleases; and inactivate endogenous enzymes like peroxidase and alkaline phosphatase. Endogenous nucleases may be inactivated by degradation with proteinase K, by heat treatment, use of a chelating agent such as EDTA or EGTA, the introduction of carrier DNA or RNA, treatment with a chaotrope such as urea, thiourea, guanidine hydrochloride, guanidine thiocyanate, lithium perchlorate, etc, or diethyl pyrocarbonate. Alkaline phosphatase may be inactivated by treated with 0. IN HCl for 5 minutes at room temperature or treatment with 1 mM levamisole. Peroxidase activity may be eliminated by treatment with 0.03% hydrogen peroxide. Endogenous biotin may be blocked by soaking the slide or section in an avidin (streptavidin, neutravidin may be substituted) solution for at least 15 minutes at room temperature. The slide or section is then washed for at least 10 minutes in buffer. This may be repeated at least three times. Then the slide or section is soaked in a biotin solution for 10 minutes. This may be repeated at least three times with a fresh biotin solution each time. The buffer wash procedure is repeated. Blocking protocols should be minimized to prevent damaging either the cell or tissue structure or the target or targets of interest but one or more of these protocols could be combined to "block" a slide or section prior to reaction with one or more slow off-rate aptamers. See Basic Medical Histology: the Biology of Cells, Tissues and Organs, authored by Richard G. Kessel, Oxford University Press, 1998.
Determination of Biomarker Values using Mass Spectrometry Methods [00202] A variety of configurations of mass spectrometers can be used to detect biomarker values. Several types of mass spectrometers are available or can be produced with various configurations. In general, a mass spectrometer has the following major components: a sample inlet, an ion source, a mass analyzer, a detector, a vacuum system, and instrument- control system, and a data system. Difference in the sample inlet, ion source, and mass analyzer generally define the type of instrument and its capabilities. For example, an inlet can be a capillary-column liquid chromatography source or can be a direct probe or stage such as used in matrix-assisted laser desorption. Common ion sources are, for example, electrospray, including nanospray and microspray or matrix-assisted laser desorption. Common mass analyzers include a quadrupole mass filter, ion trap mass analyzer and time- of-flight mass analyzer. Additional mass spectrometry methods are well known in the art (see Burlingame et al. Anal. Chem. 70:647 R-716R (1998); Kinter and Sherman, New York (2000)).
[00203] Protein biomarkers and biomarker values can be detected and measured by any of the following: electrospray ionization mass spectrometry (ESI-MS), ESI-MS/MS, ESI- MS/(MS)n, matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS), surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF-MS), desorption/ionization on silicon (DIOS), secondary ion mass spectrometry (SIMS), quadrupole time-of-flight (Q-TOF), tandem time-of-flight (TOF/TOF) technology, called ultraflex III TOF/TOF, atmospheric pressure chemical ionization mass spectrometry (APCI-MS), APCI-MS/MS, APCI-(MS)N, atmospheric pressure photoionization mass spectrometry (APPI-MS), APPI-MS/MS, and APPI-(MS)N, quadrupole mass spectrometry, Fourier transform mass spectrometry (FTMS), quantitative mass spectrometry, and ion trap mass spectrometry.
[00204] Sample preparation strategies are used to label and enrich samples before mass spectroscopic characterization of protein biomarkers and determination biomarker values. Labeling methods include but are not limited to isobaric tag for relative and absolute quantitation (iTRAQ) and stable isotope labeling with amino acids in cell culture (SILAC). Capture reagents used to selectively enrich samples for candidate biomarker proteins prior to mass spectroscopic analysis include but are not limited to aptamers, antibodies, nucleic acid probes, chimeras, small molecules, an F(ab')2 fragment, a single chain antibody fragment, an Fv fragment, a single chain Fv fragment, a nucleic acid, a lectin, a ligand-binding receptor, affybodies, nanobodies, ankyrins, domain antibodies, alternative antibody scaffolds (e.g. diabodies etc) imprinted polymers, avimers, peptidomimetics, peptoids, peptide nucleic acids, threose nucleic acid, a hormone receptor, a cytokine receptor, and synthetic receptors, and modifications and fragments of these.
[00205] The foregoing assays enable the detection of biomarker values that are useful in methods for diagnosing ovarian cancer, where the methods comprise detecting, in a biological sample from an individual, at least N biomarker values that each correspond to a biomarker selected from the group consisting of the biomarkers provided in Table 1, wherein a classification, as described in detail below, using the biomarker values indicates whether the individual has ovarian cancer. While certain of the described ovarian cancer biomarkers are useful alone for detecting and diagnosing ovarian cancer, methods are also described herein for the grouping of multiple subsets of the ovarian cancer biomarkers that are each useful as a panel of three or more biomarkers. Thus, various embodiments of the instant application provide combinations comprising N biomarkers, wherein N is at least three biomarkers. In other embodiments, N is selected to be any number from 2-42 biomarkers. It will be appreciated that N can be selected to be any number from any of the above described ranges, as well as similar, but higher order, ranges. In accordance with any of the methods described herein, biomarker values can be detected and classified individually or they can be detected and classified collectively, as for example in a multiplex assay format. [00206] In another aspect, methods are provided for detecting an absence of ovarian cancer, the methods comprising detecting, in a biological sample from an individual, at least N biomarker values that each correspond to a biomarker selected from the group consisting of the biomarkers provided in Table 1, wherein a classification, as described in detail below, of the biomarker values indicates an absence of ovarian cancer in the individual. While certain of the described ovarian cancer biomarkers are useful alone for detecting and diagnosing the absence of ovarian cancer, methods are also described herein for the grouping of multiple subsets of the ovarian cancer biomarkers that are each useful as a panel of three or more biomarkers. Thus, various embodiments of the instant application provide combinations comprising N biomarkers, wherein N is at least three biomarkers. In other embodiments, N is selected to be any number from 2-42 biomarkers. It will be appreciated that N can be selected to be any number from any of the above described ranges, as well as similar, but higher order, ranges. In accordance with any of the methods described herein, biomarker values can be detected and classified individually or they can be detected and classified collectively, as for example in a multiplex assay format.
Classification of Biomarkers and Calculation of Disease Scores [00207] A biomarker "signature" for a given diagnostic test contains a set of markers, each marker having different levels in the populations of interest. Different levels, in this context, may refer to different means of the marker levels for the individuals in two or more groups, or different variances in the two or more groups, or a combination of both. For the simplest form of a diagnostic test, these markers can be used to assign an unknown sample from an individual into one of two groups, either diseased or not diseased. The assignment of a sample into one of two or more groups is known as classification, and the procedure used to accomplish this assignment is known as a classifier or a classification method. Classification methods may also be referred to as scoring methods. There are many classification methods that can be used to construct a diagnostic classifier from a set of biomarker values. In general, classification methods are most easily performed using supervised learning techniques where a data set is collected using samples obtained from individuals within two (or more, for multiple classification states) distinct groups one wishes to distinguish. Since the class (group or population) to which each sample belongs is known in advance for each sample, the classification method can be trained to give the desired classification response. It is also possible to use unsupervised learning techniques to produce a diagnostic classifier. [00208] Common approaches for developing diagnostic classifiers include decision trees; bagging + boosting + forests; rule inference based learning; Parzen Windows; linear models; logistic; neural network methods; unsupervised clustering; K-means; hierarchical ascending/ descending; semi-supervised learning; prototype methods; nearest neighbor; kernel density estimation; support vector machines; hidden Markov models; Boltzmann Learning; and classifiers may be combined either simply or in ways which minimize particular objective functions. For a review, see, e.g., Pattern Classification, R.O. Duda, et al., editors, John Wiley & Sons, 2nd edition, 2001; see also, The Elements of Statistical Learning - Data Mining, Inference, and Prediction, T. Hastie, et al., editors, Springer Science+Business Media, LLC, 2nd edition, 2009; each of which is incorporated by reference in its entirety.
[00209] To produce a classifier using supervised learning techniques, a set of samples called training data are obtained. In the context of diagnostic tests, training data includes samples from the distinct groups (classes) to which unknown samples will later be assigned. For example, samples collected from individuals in a control population and individuals in a particular disease population can constitute training data to develop a classifier that can classify unknown samples (or, more particularly, the individuals from whom the samples were obtained) as either having the disease or being free from the disease. The development of the classifier from the training data is known as training the classifier. Specific details on classifier training depend on the nature of the supervised learning technique. For purposes of illustration, an example of training a naϊve Bayesian classifier will be described below (see, e.g., Pattern Classification, R. O. Duda, et al., editors, John Wiley & Sons, 2nd edition, 2001; see also, The Elements of Statistical Learning - Data Mining, Inference, and Prediction, T. Hastie, et al., editors, Springer Science+Business Media, LLC, 2nd edition, 2009). [00210] Since typically there are many more potential biomarker values than samples in a training set, care must be used to avoid over- fitting. Over- fitting occurs when a statistical model describes random error or noise instead of the underlying relationship. Over-fitting can be avoided in a variety of way, including, for example, by limiting the number of markers used in developing the classifier, by assuming that the marker responses are independent of one another, by limiting the complexity of the underlying statistical model employed, and by ensuring that the underlying statistical model conforms to the data. [00211] An illustrative example of the development of a diagnostic test using a set of biomarkers includes the application of a naϊve Bayes classifier, a simple probabilistic classifier based on Bayes theorem with strict independent treatment of the biomarkers. Each biomarker is described by a class-dependent probability density function (pdf) for the measured RFU values or log RFU (relative fluorescence units) values in each class. The joint pdfs for the set of markers in one class is assumed to be the product of the individual class- dependent pdfs for each biomarker. Training a naϊve Bayes classifier in this context amounts to assigning parameters ("parameterization") to characterize the class dependent pdfs. Any underlying model for the class -dependent pdfs may be used, but the model should generally conform to the data observed in the training set. [00212] Specifically, the class-dependent probability of measuring a value X1 for biomarker i in the disease class is written as p(xf I d) and the overall naϊve Bayes probability of observing n markers with values x = (x[,x2,...xn) is written as
n
Figure imgf000054_0001
j=l where the individual JCJ s are the measured biomarker levels in RFU or log RFU. The classification assignment for an unknown is facilitated by calculating the probability of being diseased p(d I x) having measured x compared to the probability of being disease free (control) p(c I x) for the same measured values. The ratio of these probabilities is computed
from the class -dependent pdfs by application of Bayes theorem, i.e., p(c \ x) p(x I c)(l - P(d)) p(d \ x) p(x \ d)P(d)
where P(d) is the prevalence of the disease in the population appropriate to the test. Taking the logarithm of both sides of this ratio and substituting the naϊve Bayes class-dependent probabilities from above gives
Figure imgf000055_0001
This form is known as the log likelihood ratio and simply states that the log likelihood of being free of the particular disease versus having the disease and is primarily composed of the sum of individual log likelihood ratios of the n individual biomarkers. In its simplest form, an unknown sample (or, more particularly, the individual from whom the sample was obtained) is classified as being free of the disease if the above ratio is greater than zero and having the disease if the ratio is less than zero. [00213] In one exemplary embodiment, the class -dependent biomarker pdfs p(x[ I c) and p(x[ I d) are assumed to be normal or log-normal distributions in the measured RFU values x[ , i.e.
i-Mc,02
9
1 2σ~ . p(xi \ c) = -j= e
J2πσcj
2 with a similar expression for p(xι I d) with //j / and σ , ■ . Parameterization of the model
requires estimation of two parameters for each class-dependent pdf, a mean μ and a variance CT, from the training data. This may be accomplished in a number of ways, including, for example, by maximum likelihood estimates, by least-squares, and by any other methods known to one skilled in the art. Substituting the normal distributions for p(x[ I c) and p(x[ I d) into the log-likelihood ratio defined above gives the following expression: i In
Figure imgf000056_0001
Once a set of μs and σs have been defined for each pdf in each class from the training data and the disease prevalence in the population is specified, the Bayes classifier is fully determined and may be used to classify unknown samples with measured values x .
[00214] The performance of the naϊve Bayes classifier is dependent upon the number and quality of the biomarkers used to construct and train the classifier. A single biomarker will perform in accordance with its KS-distance (Kolmogorov-Smirnov), as defined in Example 3, below. If a classifier performance metric is defined as the sum of the sensitivity (fraction of true positives, fjp ) and specificity (one minus the fraction of false positives, 1 - fpp ), a perfect classifier will have a score of two and a random classifier, on average, will have a score of one. Using the definition of the KS-distance, that value x which maximizes the difference in the cdf functions can be found by solving dKS = d(cdfc(x) -cdfd (x)) = Q dx dx for x which leads to p(x* I c) = p(x* I d) , i.e, the KS distance occurs where the class- dependent pdfs cross. Substituting this value of x into the expression for the KS-distance yields the following definition for KS
KS = cdfc(x*) - cdfd (x*)
x X
= \ p(x \ c)dx - \ p(x \ d)dx
Figure imgf000056_0002
= l ~fFP -fFN > the KS distance is one minus the total fraction of errors using a test with a cut-off at x , essentially a single analyte Bayesian classifier. Since we define a score of sensitivity + specificity = 2 - fpp - fpjy , combining the above definition of the KS-distance we see that sensitivity + specificity = 1 + KS . We select biomarkers with a statistic that is inherently suited for building naϊve Bayes classifiers. [00215] The addition of subsequent markers with good KS distances (>0.3, for example) will, in general, improve the classification performance if the subsequently added markers are independent of the first marker. Using the sensitivity plus specificity as a classifier score, it is straightforward to generate many high scoring classifiers with a variation of a greedy algorithm. (A greedy algorithm is any algorithm that follows the problem solving metaheuristic of making the locally optimal choice at each stage with the hope of finding the global optimum.)
[00216] The algorithm approach used here is described in detail in Example 4. Briefly, all single analyte classifiers are generated from a table of potential biomarkers and added to a list. Next, all possible additions of a second analyte to each of the stored single analyte classifiers is then performed, saving a predetermined number of the best scoring pairs, say, for example, a thousand, on a new list. All possible three-marker classifiers are explored using this new list of the best two-marker classifiers, again saving the best thousand of these. This process continues until the score either plateaus or begins to deteriorate as additional markers are added. Those high scoring classifiers that remain after convergence can be evaluated for the desired performance for an intended use. For example, in one diagnostic application, classifiers with a high sensitivity and modest specificity may be more desirable than modest sensitivity and high specificity. In another diagnostic application, classifiers with a high specificity and a modest sensitivity may be more desirable. The desired level of performance is generally selected based upon a trade-off that must be made between the number of false positives and false negatives that can each be tolerated for the particular diagnostic application. Such trade-offs generally depend on the medical consequences of an error, either false positive or false negative.
[00217] Various other techniques are known in the art and may be employed to generate many potential classifiers from a list of biomarkers using a naϊve Bayes classifier. In one embodiment, what is referred to as a genetic algorithm can be used to combine different markers using the fitness score as defined above. Genetic algorithms are particularly well suited to exploring a large diverse population of potential classifiers. In another embodiment, so-called ant colony optimization can be used to generate sets of classifiers. Other strategies that are known in the art can also be employed, including, for example, other evolutionary strategies as well as simulated annealing and other stochastic search methods. Metaheuristic methods, such as, for example, harmony search may also be employed. [00218] Exemplary embodiments use any number of the ovarian cancer biomarkers listed in Table 1 in various combinations to produce diagnostic tests for detecting ovarian cancer (see Example 2 for a detailed description of how these biomarkers were identified). In one embodiment, a method for diagnosing ovarian cancer uses a naϊve Bayes classification method in conjunction with any number of the ovarian cancer biomarkers listed in Table 1. In an illustrative example (see Example 3), the simplest test for detecting ovarian cancer from a population of women with pelvic masses can be constructed using a single biomarker, for example, BAFF Receptor which is down-regulated in ovarian cancer with a KS -distance of
0.39 ( 1 + KS = 1.39 ). Using the parameters μc ; , σ ■ , //j ; and σ , . for BAFF Receptor
from Table 16 and the equation for the log-likelihood described above, a diagnostic test with a sensitivity of 0.74 and specificity of 0.56 ( sensitivity + specificity = 1.31 ) can be produced, see Table 17. The ROC curve for this test is displayed in Figure 2 and has an AUC of 0.70. [00219] Addition of biomarker RGM-C, for example, with a KS-distance of 0.43, significantly improves the classifier performance to a sensitivity of 82% and specificity of 0.73% ( sensitivity + specificity = 1.51 ) and an AUC = 0.81. Note that the score for a classifier constructed of two biomarkers is not a simple sum of the KS-distances; KS- distances are not additive when combining biomarkers, and it takes many more weak markers to achieve the same level of performance as a strong marker. Adding a third marker, HGF, for example, boosts the classifier performance to 83% sensitivity and 74% specificity and AUC = 0.84. Adding additional biomarkers, such as, for example, SLPI, C9, α2- Antiplasmin, SAP, MMP-7, MCP-3, and HSP90α, produces a series of ovarian cancer tests summarized in Table 17 and displayed as a series of ROC curves in Figure 3. The score of the classifiers as a function of the number of analytes used in classifier construction is shown in Figure 4. This exemplary ten-marker classifier has a sensitivity of 97% and a specificity of 88% with an AUC of 0.94.
[00220] The markers listed in Table 1 can be combined in many ways to produce classifiers for diagnosing ovarian cancer. In some embodiments, panels of biomarkers are comprised of different numbers of analytes depending on a specific diagnostic performance criterion that is selected. For example, certain combinations of biomarkers will produce tests that are more sensitive (or more specific) than other combinations.
[00221] Once a panel is defined to include a particular set of biomarkers from Table 1 and a classifier is constructed from a set of training data, the definition of the diagnostic test is complete. In one embodiment, the procedure used to classify an unknown sample is outlined in Figure IA. In another embodiment the procedure used to classify an unknown sample is outlined in Figure IB. The biological sample is appropriately diluted and then run in one or more assays to produce the relevant quantitative biomarker levels used for classification. The measured biomarker levels are used as input for the classification method that outputs a classification and an optional score for the sample that reflects the confidence of the class assignment.
[00222] Table 1 identifies 42 biomarkers that are useful for diagnosing ovarian cancer.
This is a surprisingly larger number than expected when compared to what is typically found during biomarker discovery efforts and may be attributable to the scale of the described study, which encompassed over 800 proteins measured in hundreds of individual samples, in some cases at concentrations in the low femtomolar range. Presumably, the large number of discovered biomarkers reflects the diverse biochemical pathways implicated in both tumor biology and the body's response to the tumor's presence; each pathway and process involves many proteins. The results show that no single protein of a small group of proteins is uniquely informative about such complex processes; rather, that multiple proteins are involved in relevant processes, such as apoptosis or extracellular matrix repair, for example. [00223] Given the numerous biomarkers identified during the described study, one would expect to be able to derive large numbers of high-performing classifiers that can be used in various diagnostic methods. To test this notion, tens of thousands of classifiers were evaluated using the biomarkers in Table 1. As described in Example 4, many subsets of the biomarkers presented in Table 1 can be combined to generate useful classifiers. By way of example, descriptions are provided for classifiers containing 1, 2, and 3 biomarkers for the diagnosis of ovarian cancer, particularly, the diagnosis of ovarian cancer in individuals who have a pelvic mass that is detectable by CT. As described in Example 4, all classifiers that were built using the biomarkers in Table 1 perform distinctly better than classifiers that were built using "non-markers".
[00224] The performance of ten-marker classifiers obtained by excluding the "best" individual markers from the ten-marker aggregation was tested. As described in Example 4, Part 3, classifiers constructed without the "best" markers in Table 1 performed well. Many subsets of the biomarkers listed in Table 1 performed close to optimally, even after removing the top 15 of the markers listed in the Table. This implies that the performance characteristics of any particular classifier are likely not due to some small core group of biomarkers and that the disease process likely impacts numerous biochemical pathways, which alters the expression level of many proteins. [00225] The results from Example 4 suggest certain possible conclusions: First, the identification of a large number of biomarkers enables their aggregation into a vast number of classifiers that offer similarly high performance. Second, classifiers can be constructed such that particular biomarkers may be substituted for other biomarkers in a manner that reflects the redundancies that undoubtedly pervade the complexities of the underlying disease processes. That is to say, the information about the disease contributed by any individual biomarker identified in Table 1 overlaps with the information contributed by other biomarkers, such that it may be that no particular biomarker or small group of biomarkers in Table 1 must be included in any classifier.
[00226] Exemplary embodiments use naϊve Bayes classifiers constructed from the data in Table 18 to classify an unknown sample. The procedure is outlined in Figures IA and B. In one embodiment, the biological sample is optionally diluted and run in a multiplexed aptamer assay. The data from the assay are normalized and calibrated as outlined in Example 3, and the resulting biomarker levels are used as input to a Bayes classification scheme. The log-likelihood ratio is computed for each measured biomarker individually and then summed to produce a final classification score, which is also referred to as a diagnostic score. The resulting assignment as well as the overall classification score can be reported. Optionally, the individual log-likelihood risk factors computed for each biomarker level can be reported as well. The details of the classification score calculation are presented in Example 3.
Kits
[00227] Any combination of the biomarkers of Table 1 (as well as additional biomedical information) can be detected using a suitable kit, such as for use in performing the methods disclosed herein. Furthermore, any kit can contain one or more detectable labels as described herein, such as a fluorescent moiety, etc.
[00228] In one embodiment, a kit includes (a) one or more capture reagents (such as, for example, at least one aptamer or antibody) for detecting one or more biomarkers in a biological sample, wherein the biomarkers include any of the biomarkers set forth in Table 1, and optionally (b) one or more software or computer program products for classifying the individual from whom the biological sample was obtained as either having or not having ovarian cancer or for determining the likelihood that the individual has ovarian cancer, as further described herein. Alternatively, rather than one or more computer program products, one or more instructions for manually performing the above steps by a human can be provided. [00229] The combination of a solid support with a corresponding capture reagent and a signal generating material is referred to herein as a "detection device" or "kit". The kit can also include instructions for using the devices and reagents, handling the sample, and analyzing the data. Further the kit may be used with a computer system or software to analyze and report the result of the analysis of the biological sample. [00230] The kits can also contain one or more reagents (e.g., solubilization buffers, detergents, washes, or buffers) for processing a biological sample. Any of the kits described herein can also include, e.g., buffers, blocking agents, mass spectrometry matrix materials, antibody capture agents, positive control samples, negative control samples, software and information such as protocols, guidance and reference data.
[00231] In one aspect, the invention provides kits for the analysis of ovarian cancer status. The kits include PCR primers for one or more biomarkers selected from Table 1. The kit may further include instructions for use and correlation of the biomarkers with ovarian cancer. The kit may also include any of the following, either alone or in combination: a DNA array containing the complement of one or more of the biomarkers selected from Table 1, reagents, and enzymes for amplifying or isolating sample DNA. The kits may include reagents for real-time PCR, such as, for example, TaqMan probes and/or primers, and enzymes.
[00232] For example, a kit can comprise (a) reagents comprising at least capture reagent for quantifying one or more biomarkers in a test sample, wherein said biomarkers comprise the biomarkers set forth in Table 1, or any other biomarkers or biomarkers panels described herein, and optionally (b) one or more algorithms or computer programs for performing the steps of comparing the amount of each biomarker quantified in the test sample to one or more predetermined cutoffs and assigning a score for each biomarker quantified based on said comparison, combining the assigned scores for each biomarker quantified to obtain a total score, comparing the total score with a predetermined score, and using said comparison to determine whether an individual has ovarian cancer. Alternatively, rather than one or more algorithms or computer programs, one or more instructions for manually performing the above steps by a human can be provided.
Computer Methods and Software
[00233] Once a biomarker or biomarker panel is selected, a method for diagnosing an individual can comprise the following: 1) collect or otherwise obtain a biological sample; 2) perform an analytical method to detect and measure the biomarker or biomarkers in the panel in the biological sample; 3) perform any data normalization or standardization required for the method used to collect biomarker values; 4) calculate the marker score; 5) combine the marker scores to obtain a total diagnostic score; and 6) report the individual's diagnostic score. In this approach, the diagnostic score may be a single number determined from the sum of all the marker calculations that is compared to a preset threshold value that is an indication of the presence or absence of disease. Or the diagnostic score may be a series of bars that each represent a biomarker value and the pattern of the responses may be compared to a pre-set pattern for determination of the presence or absence of disease. [00234] At least some embodiments of the methods described herein can be implemented with the use of a computer. An example of a computer system 100 is shown in Figure 6. With reference to Figure 6, system 100 is shown comprised of hardware elements that are electrically coupled via bus 108, including a processor 101, input device 102, output device 103, storage device 104, computer-readable storage media reader 105a, communications system 106 processing acceleration (e.g., DSP or special-purpose processors) 107 and memory 109. Computer-readable storage media reader 105a is further coupled to computer-readable storage media 105b, the combination comprehensively representing remote, local, fixed and/or removable storage devices plus storage media, memory, etc. for temporarily and/or more permanently containing computer-readable information, which can include storage device 104, memory 109 and/or any other such accessible system 100 resource. System 100 also comprises software elements (shown as being currently located within working memory 191) including an operating system 192 and other code 193, such as programs, data and the like.
[00235] With respect to Figure 6, system 100 has extensive flexibility and configurability. Thus, for example, a single architecture might be utilized to implement one or more servers that can be further configured in accordance with currently desirable protocols, protocol variations, extensions, etc. However, it will be apparent to those skilled in the art that embodiments may well be utilized in accordance with more specific application requirements. For example, one or more system elements might be implemented as sub- elements within a system 100 component (e.g., within communications system 106). Customized hardware might also be utilized and/or particular elements might be implemented in hardware, software or both. Further, while connection to other computing devices such as network input/output devices (not shown) may be employed, it is to be understood that wired, wireless, modem, and/or other connection or connections to other computing devices might also be utilized. [00236] In one aspect, the system can comprise a database containing features of biomarkers characteristic of ovarian cancer. The biomarker data (or biomarker information) can be utilized as an input to the computer for use as part of a computer implemented method.
The biomarker data can include the data as described herein.
[00237] In one aspect, the system further comprises one or more devices for providing input data to the one or more processors.
[00238] The system further comprises a memory for storing a data set of ranked data elements.
[00239] In another aspect, the device for providing input data comprises a detector for detecting the characteristic of the data element, e.g., such as a mass spectrometer or gene chip reader.
[00240] The system additionally may comprise a database management system. User requests or queries can be formatted in an appropriate language understood by the database management system that processes the query to extract the relevant information from the database of training sets.
[00241] The system may be connectable to a network to which a network server and one or more clients are connected. The network may be a local area network (LAN) or a wide area network (WAN), as is known in the art. Preferably, the server includes the hardware necessary for running computer program products (e.g., software) to access database data for processing user requests.
[00242] The system may include an operating system (e.g., UNIX or Linux) for executing instructions from a database management system. In one aspect, the operating system can operate on a global communications network, such as the internet, and utilize a global communications network server to connect to such a network.
[00243] The system may include one or more devices that comprise a graphical display interface comprising interface elements such as buttons, pull down menus, scroll bars, fields for entering text, and the like as are routinely found in graphical user interfaces known in the art. Requests entered on a user interface can be transmitted to an application program in the system for formatting to search for relevant information in one or more of the system databases. Requests or queries entered by a user may be constructed in any suitable database language.
[00244] The graphical user interface may be generated by a graphical user interface code as part of the operating system and can be used to input data and/or to display inputted data. The result of processed data can be displayed in the interface, printed on a printer in communication with the system, saved in a memory device, and/or transmitted over the network or can be provided in the form of the computer readable medium. [00245] The system can be in communication with an input device for providing data regarding data elements to the system (e.g., expression values). In one aspect, the input device can include a gene expression profiling system including, e.g., a mass spectrometer, gene chip or array reader, and the like.
[00246] The methods and apparatus for analyzing ovarian cancer biomarker information according to various embodiments may be implemented in any suitable manner, for example, using a computer program operating on a computer system. A conventional computer system comprising a processor and a random access memory, such as a remotely- accessible application server, network server, personal computer or workstation may be used. Additional computer system components may include memory devices or information storage systems, such as a mass storage system and a user interface, for example a conventional monitor, keyboard and tracking device. The computer system may be a stand-alone system or part of a network of computers including a server and one or more databases. [00247] The ovarian cancer biomarker analysis system can provide functions and operations to complete data analysis, such as data gathering, processing, analysis, reporting and/or diagnosis. For example, in one embodiment, the computer system can execute the computer program that may receive, store, search, analyze, and report information relating to the ovarian cancer biomarkers. The computer program may comprise multiple modules performing various functions or operations, such as a processing module for processing raw data and generating supplemental data and an analysis module for analyzing raw data and supplemental data to generate an ovarian cancer status and/or diagnosis. Diagnosing ovarian cancer status may comprise generating or collecting any other information, including additional biomedical information, regarding the condition of the individual relative to the disease, identifying whether further tests may be desirable, or otherwise evaluating the health status of the individual.
[00248] Referring now to Figure 7, an example of a method of utilizing a computer in accordance with principles of a disclosed embodiment can be seen. In Figure 7, a flowchart 3000 is shown. In block 3004, biomarker information can be retrieved for an individual. The biomarker information can be retrieved from a computer database, for example, after testing of the individual's biological sample is performed. The biomarker information can comprise biomarker values that each correspond to one of at least N biomarkers selected from a group consisting of the biomarkers provided in Table 1, wherein N = 2-42. In block 3008, a computer can be utilized to classify each of the biomarker values. And, in block 3012, a determination can be made as to the likelihood that an individual has ovarian cancer based upon a plurality of classifications. The indication can be output to a display or other indicating device so that it is viewable by a person. Thus, for example, it can be displayed on a display screen of a computer or other output device.
[00249] Referring now to Figure 8, an alternative method of utilizing a computer in accordance with another embodiment can be illustrated via flowchart 3200. In block 3204, a computer can be utilized to retrieve biomarker information for an individual. The biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers provided in Table 1. In block 3208, a classification of the biomarker value can be performed with the computer. And, in block 3212, an indication can be made as to the likelihood that the individual has ovarian cancer based upon the classification. The indication can be output to a display or other indicating device so that it is viewable by a person. Thus, for example, it can be displayed on a display screen of a computer or other output device.
[00250] Some embodiments described herein can be implemented so as to include a computer program product. A computer program product may include a computer readable medium having computer readable program code embodied in the medium for causing an application program to execute on a computer with a database.
[00251] As used herein, a "computer program product" refers to an organized set of instructions in the form of natural or programming language statements that are contained on a physical media of any nature (e.g., written, electronic, magnetic, optical or otherwise) and that may be used with a computer or other automated data processing system. Such programming language statements, when executed by a computer or data processing system, cause the computer or data processing system to act in accordance with the particular content of the statements. Computer program products include without limitation: programs in source and object code and/or test or data libraries embedded in a computer readable medium. Furthermore, the computer program product that enables a computer system or data processing equipment device to act in pre-selected ways may be provided in a number of forms, including, but not limited to, original source code, assembly code, object code, machine language, encrypted or compressed versions of the foregoing and any and all equivalents.
[00252] In one aspect, a computer program product is provided for indicating a likelihood of ovarian cancer. The computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers in the biological sample selected from the group of biomarkers provided in Table 1, wherein N = 2-42; and code that executes a classification method that indicates an ovarian disease status of the individual as a function of the biomarker values.
[00253] In still another aspect, a computer program product is provided for indicating a likelihood of ovarian cancer. The computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers provided in Table 1; and code that executes a classification method that indicates an ovarian disease status of the individual as a function of the biomarker value.
[00254] While various embodiments have been described as methods or apparatuses, it should be understood that embodiments can be implemented through code coupled with a computer, e.g., code resident on a computer or accessible by the computer. For example, software and databases could be utilized to implement many of the methods discussed above. Thus, in addition to embodiments accomplished by hardware, it is also noted that these embodiments can be accomplished through the use of an article of manufacture comprised of a computer usable medium having a computer readable program code embodied therein, which causes the enablement of the functions disclosed in this description. Therefore, it is desired that embodiments also be considered protected by this patent in their program code means as well. Furthermore, the embodiments may be embodied as code stored in a computer-readable memory of virtually any kind including, without limitation, RAM, ROM, magnetic media, optical media, or magneto-optical media. Even more generally, the embodiments could be implemented in software, or in hardware, or any combination thereof including, but not limited to, software running on a general purpose processor, microcode, PLAs, or ASICs.
[00255] It is also envisioned that embodiments could be accomplished as computer signals embodied in a carrier wave, as well as signals (e.g., electrical and optical) propagated through a transmission medium. Thus, the various types of information discussed above could be formatted in a structure, such as a data structure, and transmitted as an electrical signal through a transmission medium or stored on a computer readable medium. [00256] It is also noted that many of the structures, materials, and acts recited herein can be recited as means for performing a function or step for performing a function. Therefore, it should be understood that such language is entitled to cover all such structures, materials, or acts disclosed within this specification and their equivalents, including the matter incorporated by reference.
EXAMPLES
[00257] The following examples are provided for illustrative purposes only and are not intended to limit the scope of the application as defined by the appended claims. All examples described herein were carried out using standard techniques, which are well known and routine to those of skill in the art. Routine molecular biology techniques described in the following examples can be carried out as described in standard laboratory manuals, such as Sambrook et al., Molecular Cloning: A Laboratory Manual, 3rd. ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., (2001).
Example 1. Multiplexed Aptamer Analysis of Samples For Ovarian Cancer Biomarker Selection
[00258] This example describes the multiplex aptamer assay used to analyze the samples and controls for the identification of the biomarkers set forth in Table 1 (see Figure
9). In this case, the multiplexed analysis utilized 811 aptamers, each unique to a specific target.
[00259] In this method, pipette tips were changed for each solution addition.
[00260] Also, unless otherwise indicated, most solution transfers and wash additions used the 96-well head of a Beckman Biomek Fxp. Method steps manually pipetted used a twelve channel P200 Pipetteman (Rainin Instruments, LLC, Oakland, CA), unless otherwise indicated. A custom buffer referred to as SB 17 was prepared in-house, comprising 4OmM
HEPES, 10OmM NaCl, 5mM KCl, 5mM MgCl2, ImM EDTA at pH7.5. All steps were performed at room temperature unless otherwise indicated.
[00261] 1. Preparation of Aptamer Stock Solution
[00262] For aptamers without a photo-cleavable biotin linker, custom stock aptamer solutions for 10%, 1% and 0.03% plasma were prepared at 8x concentration in Ix SB17, 0.05% Tween-20 with appropriate photo-cleavable, biotinylated primers, where the resultant primer concentration was 3 times the relevant aptamer concentration. The primers hybridized to all or part of the corresponding aptamer.
[00263] Each of the 3, 8x aptamer solutions were diluted separately 1:4 into IxSB 17,
0.05% Tween-20 (1500 μL of 8x stock into 4500 μL of lxSB17, 0.05% Tween-20) to achieve a 2x concentration. Each diluted aptamer master mix was then split, 1500 μL each, into 4, 2 mL screw cap tubes and brought to 95°C for 5 minutes, followed by a 37°C incubation for 15 minutes. After incubation, the 4, 2 mL tubes corresponding to a particular aptamer master mix were combined into a reagent trough, and 55 μL of a 2x aptamer mix (for all three mixes) was manually pipetted into a 96-well Hybaid plate and the plate foil sealed.
The final result was 3, 96-well, foil-sealed Hybaid plates. The individual aptamer concentration was 0.5 nM.
[00264] 2. Assay Sample Preparation
[00265] Frozen aliquots of 100% plasma, stored at -800C, were placed in 25°C water bath for 10 minutes. Thawed samples were placed on ice, gently vortexed (set on 4) for 8 seconds and then replaced on ice.
[00266] A 20% sample solution was prepared by transferring 16 μL of sample using a
50 μL 8-channel spanning pipettor into 96-well Hybaid plates, each well containing 64 μL of the appropriate sample diluent at 4°C (0.8x SB 17, 0.05% Tween-20, 2 μM Z-block_2, 0.6 mM MgCl2 for plasma). This plate was stored on ice until the next sample dilution steps were initiated.
[00267] To commence sample and aptamer equilibration, the 20% sample plate was briefly centrifuged and placed on the Beckman FX where it was mixed by pipetting up and down with the 96-well pipettor. A 2% sample was then prepared by diluting 10 μL of the
20% sample into 90 μL of IxSB 17, 0.05% Tween-20. Next, dilution of 6 μL of the resultant
2% sample into 194 μL of IxSB 17, 0.05% Tween-20 made a 0.06% sample plate. Dilutions were done on the Beckman Biomek Fxp. After each transfer, the solutions were mixed by pipetting up and down. The 3 sample dilution plates were then transferred to their respective aptamer solutions by adding 55 μL of the sample to 55 μL of the appropriate 2x aptamer mix.
The sample and aptamer solutions were mixed on the robot by pipetting up and down.
[00268] 3. Sample Equilibration binding
[00269] The sample/ap tamer plates were foil sealed and placed into a 370C incubator for 3.5 hours before proceeding to the Catch 1 step.
[00270] 4. Preparation of Catch 2 bead plate [00271] An 11 niL aliquot of MyOne (Invitrogen Corp., Carlsbad, CA) Streptavidin Cl beads was washed 2 times with equal volumes of 20 mM NaOH (5 minute incubation for each wash), 3 times with equal volumes of Ix SB 17, 0.05% Tween-20 and resuspended in 11 mL Ix SB 17, 0.05% Tween-20. Using a 12-span multichannel pipettor, 50 μL of this solution was manually pipetted into each well of a 96-well Hybaid plate. The plate was then covered with foil and stored at 4°C for use in the assay. [00272] 5. Preparation of Catch 1 bead plates
[00273] Three 0.45 μm Millipore HV plates (Durapore membrane, Cat#
MAHVN4550) were equilibrated with 100 μL of Ix SB 17, 0.05% Tween-20 for at least 10 minutes. The equilibration buffer was then filtered through the plate and 133.3 μL of a 7.5% Streptavidin-agarose bead slurry (in Ix SB 17, 0.05% Tween-20) was added into each well. To keep the streptavidin-agarose beads suspended while transferring them into the filter plate, the bead solution was manually mixed with a 200 μL, 12-channel pipettor, 15 times. After the beads were distributed across the 3 filter plates, a vacuum was applied to remove the bead supernatant. Finally, the beads were washed in the filter plates with 200 μL Ix SB 17, 0.05% Tween-20 and then resuspended in 200 μL Ix SB 17, 0.05% Tween-20. The bottoms of the filter plates were blotted and the plates stored for use in the assay. [00274] 6. Loading the Cytomat
[00275] The cytomat was loaded with all tips, plates, all reagents in troughs (except
NHS-biotin reagent which was prepared fresh right before addition to the plates), 3 prepared catch 1 filter plates and 1 prepared MyOne plate. [00276] 7. Catch 1
[00277] After a 3.5 hour equilibration time, the sample/ap tamer plates were removed from the incubator, centrifuged for about 1 minute, foil removed, and placed on the deck of the Beckman Biomek Fxp. The Beckman Biomek Fxp program was initiated. All subsequent steps in Catch 1 were performed by the Beckman Biomek Fxp robot unless otherwise noted. Within the program, the vacuum was applied to the Catch 1 filter plates to remove the bead supernatant. One hundred microlitres of each of the 10%, 1% and 0.03% equilibration binding reactions were added to their respective Catch 1 filtration plates, and each plate was mixed using an on-deck orbital shaker at 800 rpm for 10 minutes.
[00278] Unbound solution was removed via vacuum filtration. The catch 1 beads were washed with 190 μL of 100 μM biotin in Ix SB 17, 0.05% Tween-20 followed by 190 μL of Ix SB 17, 0.05% Tween-20 by dispensing the solution and immediately drawing a vacuum to filter the solution through the plate. [00279] Next, 190 μL Ix SB 17, 0.05% Tween-20 was added to the Catch 1 plates.
Plates were blotted to remove droplets using an on-deck blot station and then incubated with orbital shakers at 800 rpm for 10 minutes at 25°C.
[00280] The robot removed this wash via vacuum filtration and blotted the bottom of the filter plate to remove droplets using the on-deck blot station.
[00281] 8. Tagging
[00282] A NHS-PEO4-biotin aliquot was thawed at 370C for 6 minutes and then diluted 1: 100 with tagging buffer (SB 17 at pH=7.25 0.05% Tween-20). The NHS-PEO4- biotin reagent was dissolved at 100 mM concentration in anhydrous DMSO and had been stored frozen at -200C. Upon a robot prompt, the diluted NHS-PEO4-biotin reagent was manually added to an on-deck trough and the robot program was manually re-initiated to dispense 100 μL of the NHS-PEO4-biotin into each well of each Catch 1 filter plate. This solution was allowed to incubate with Catch 1 beads shaking at 800 rpm for 5 minutes on the obital shakers.
[00283] 9. Kinetic Challenge and Photo-cleavage
[00284] The tagging reaction was quenched by the addition of 150 μL of 20 mM glycine in Ix SB 17, 0.05% Tween-20 to the Catch 1 plates while still containing the NHS tag. The plates were then incubated for 1 minute on orbital shakers at 800 rpm. The NHS- tag/glycine solution was removed via vacuum filtration. Next, 190 μL 20 mM glycine (Ix
SB 17, 0.05% Tween-20) was added to each plate and incubated for 1 minute on orbital shakers at 800 rpm before removal by vacuum filtration.
[00285] 190 μL of Ix SB 17, 0.05% Tween-20 was added to each plate and removed by vacuum filtration.
[00286] The wells of the Catch 1 plates were subsequently washed three times by adding 190 μL Ix SB 17, 0.05% Tween-20, placing the plates on orbital shakers for 1 minute at 800 rpm followed by vacuum filtration. After the last wash the plates were placed on top of a 1 mL deep-well plate and removed from the deck. The Catch 1 plates were centrifuged at 1000 rpm for 1 minute to remove as much extraneous volume from the agarose beads before elution as possible.
[00287] The plates were placed back onto the Beckman Biomek Fxp and 85 μL of 10 mM DxSO4 in Ix SB 17, 0.05% Tween-20 was added to each well of the filter plates.
[00288] The filter plates were removed from the deck, placed onto a Variomag
Thermoshaker (Thermo Fisher Scientific, Inc., Waltham, MA ) under the BlackRay (Ted Pella, Inc., Redding, CA) light sources, and irradiated for 10 minutes while shaking at 800 rpm.
[00289] The photocleaved solutions were sequentially eluted from each Catch 1 plate into a common deep well plate by first placing the 10% Catch 1 filter plate on top of a 1 mL deep-well plate and centrifuging at 1000 rpm for 1 minute. The 1% and 0.03% catch 1 plates were then sequentially centrifuged into the same deep well plate.
[00290] 10. Catch 2 bead capture
[00291] The 1 mL deep well block containing the combined eluates of catch 1 was placed on the deck of the Beckman Biomek Fxp for catch 2.
[00292] The robot transferred all of the photo-cleaved eluate from the 1 mL deep-well plate onto the Hybaid plate containing the previously prepared catch 2 MyOne magnetic beads (after removal of the MyOne buffer via magnetic separation).
[00293] The solution was incubated while shaking at 1350 rpm for 5 minutes at 25°C on a Variomag Thermoshaker (Thermo Fisher Scientific, Inc., Waltham, MA).
[00294] The robot transferred the plate to the on deck magnetic separator station. The plate was incubated on the magnet for 90 seconds before removal and discarding of the supernatant.
[00295] 11. 37°C 30% glycerol washes
[00296] The catch 2 plate was moved to the on-deck thermal shaker and 75 μL of Ix
SB 17, 0.05% Tween-20 was transferred to each well. The plate was mixed for 1 minute at
1350 rpm and 37°C to resuspend and warm the beads. To each well of the catch 2 plate, 75 μL of 60% glycerol at 370C was transferred and the plate continued to mix for another minute at 1350 rpm and 37°C. The robot transferred the plate to the 37°C magnetic separator where it was incubated on the magnet for 2 minutes and then the robot removed and discarded the supernatant. These washes were repeated two more times.
[00297] After removal of the third 30% glycerol wash from the catch 2 beads, 150 μL of Ix SB 17, 0.05% Tween-20 was added to each well and incubated at 37°C, shaking at 1350 rpm for 1 minute, before removal by magnetic separation on the 37°C magnet.
[00298] The catch 2 beads were washed a final time using 150 μL Ix SB 19, 0.05%
Tween-20 with incubation for 1 minute while shaking at 1350 rpm, prior to magnetic separation.
[00299] 12. Catch 2 Bead Elution and Neutralization [00300] The ap tamers were eluted from catch 2 beads by adding 105 μL of 100 mM
CAPSO with 1 M NaCl, 0.05% Tween-20 to each well. The beads were incubated with this solution with shaking at 1300 rpm for 5 minutes.
[00301] The catch 2 plate was then placed onto the magnetic separator for 90 seconds prior to transferring 90 μL of the eluate to a new 96-well plate containing 10 μL of 500 mM
HCl, 500 mM HEPES, 0.05% Tween-20 in each well. After transfer, the solution was mixed robotically by pipetting 90 μL up and down five times.
[00302] 13. Hybridization
[00303] The Beckman Biomek Fxp transferred 20 μL of the neutralized catch 2 eluate to a fresh Hybaid plate, and 5 μL of 10x Agilent Block, containing a 10x spike of hybridization controls, was added to each well. Next, 25 μL of 2x Agilent Hybridization buffer was manually pipetted to the each well of the plate containing the neutralized samples and blocking buffer and the solution was mixed by manually pipetting 25 μL up and down 15 times slowly to avoid extensive bubble formation. The plate was spun at 1000 rpm for 1 minute.
[00304] A gasket slide was placed into an Agilent hybridization chamber and 40 μL of each of the samples containing hybridization and blocking solution was manually pipetted into each gasket. An 8 -channel variable spanning pipettor was used in a manner intended to minimize bubble formation. Custom Agilent microarray slides (Agilent Technologies, Inc.,
Santa Clara, CA), with their Number Barcode facing up, were then slowly lowered onto the gasket slides (see Agilent manual for detailed description).
[00305] The top of the hybridization chambers were placed onto the slide/backing sandwich and clamping brackets slid over the whole assembly. These assemblies were tightly clamped by turning the screws securely.
[00306] Each slide/backing slide sandwich was visually inspected to assure the solution bubble could move freely within the sample. If the bubble did not move freely the hybridization chamber assembly was gently tapped to disengage bubbles lodged near the gasket.
[00307] The assembled hybridization chambers were incubated in an Agilent hybridization oven for 19 hours at 600C rotating at 20 rpm.
[00308] 14. Post Hybridization Washing
[00309] Approximately 400 mL Agilent Wash Buffer 1 was placed into each of two separate glass staining dishes. One of the staining dishes was placed on a magnetic stir plate and a slide rack and stir bar were placed into the buffer. [00310] A staining dish for Agilent Wash 2 was prepared by placing a stir bar into an empty glass staining dish.
[00311] A fourth glass staining dish was set aside for the final acetonitrile wash.
[00312] Each of six hybridization chambers was disassembled. One-by-one, the slide/backing sandwich was removed from its hybridization chamber and submerged into the staining dish containing Wash 1. The slide/backing sandwich was pried apart using a pair of tweezers, while still submerging the microarray slide. The slide was quickly transferred into the slide rack in the Wash 1 staining dish on the magnetic stir plate.
[00313] The slide rack was gently raised and lowered 5 times. The magnetic stirrer was turned on at a low setting and the slides incubated for 5 minutes.
[00314] When one minute was remaining for Wash 1, Wash Buffer 2 pre-warmed to
37°C in an incubator was added to the second prepared staining dish. The slide rack was quickly transferred to Wash Buffer 2 and any excess buffer on the bottom of the rack was removed by scraping it on the top of the stain dish. The slide rack was gently raised and lowered 5 times. The magnetic stirrer was turned on at a low setting and the slides incubated for 5 minutes.
[00315] The slide rack was slowly pulled out of Wash 2, taking approximately 15 seconds to remove the slides from the solution.
[00316] With one minute remaining in Wash 2 acetonitrile (ACN) was added to the fourth staining dish. The slide rack was transferred to the acetonitrile stain dish. The slide rack was gently raised and lowered 5 times. The magnetic stirrer was turned on at a low setting and the slides incubated for 5 minutes.
[00317] The slide rack was slowly pulled out of the ACN stain dish and placed on an absorbent towel. The bottom edges of the slides were quickly dried and the slide was placed into a clean slide box.
[00318] 15. Microarray Imaging
[00319] The microarray slides were placed into Agilent scanner slide holders and loaded into the Agilent Microarray scanner according to the manufacturer's instructions.
[00320] The slides were imaged in the Cy3-channel at 5 μm resolution at the 100%
PMT setting and the XRD option enabled at 0.05. The resulting tiff images were processed using Agilent feature extraction software version 10.5.
Example 2. Biomarker Identification [00321] The identification of potential ovarian cancer biomarkers was performed for diagnosis of ovarian cancer in women with pelvic masses. Enrollment criteria for this study were women scheduled for laparotomy or pelvic surgery for suspicion of ovarian cancer. The primary criteria for exclusion were women suffering from chronic infectious (e.g. hepatitis B, Hepatitis C or HIV), autoimmune, or inflammatory conditions or women being treated for malignancy (other than basal or squamous cell carcinomas of the skin) within the last two years. Plasma samples were collected from two different clinical sites and included 142 cases and 195 benign controls. Table 19 summarizes the site sample information. The multiplexed ap tamer affinity assay was used to measure and report the RFU value for 811 analytes in each of these 337 samples. Since the plasma samples were obtained from two independent sites under similar protocols, an examination of site differences prior to the analysis for biomarkers discovery was performed. Each of the two populations, benign pelvic mass and ovarian cancer, was separately compared between sites by generating within- site, class-dependent cumulative distribution functions (cdfs) for each of the 811 analytes. The KS -test was then applied to each analyte between both site pairs within a common class to identify those analytes that differed not by class but rather by site. In both site comparisons among the two classes, statistically significant site-dependent differences were observed.
[00322] Such site-dependent effects tend to obscure the ability to identify specific control-disease differences. In order to minimize such effects and identify key disease dependent biomarkers, three distinct strategies were employed for biomarker discovery, namely (1) aggregated class-dependent cdfs across sites, (2) comparison of within-site class- dependent cdfs, and (3) blending methods (1) with (2). Details of these three methodologies and their results follow.
[00323] These three sets of potential biomarkers can be used to build classifiers that assign samples to either a control or disease group. In fact, many such classifiers were produced from these sets of biomarkers and the frequency with which any biomarker was used in good scoring classifiers determined. Those biomarkers that occurred most frequently among the top scoring classifiers were the most useful for creating a diagnostic test. In this example, Bayesian classifiers were used to explore the classification space but many other supervised learning techniques may be employed for this purpose. The scoring fitness of any individual classifier was gauged by summing the sensitivity and specificity of the classifier at the Bayesian surface assuming a disease prevalence of 0.5. This scoring metric varies from zero to two, with two being an error-free classifier. The details of constructing a Bayesian classifier from biomarker population measurements are described in Example 3. [00324] By aggregating the class-dependent samples across all sites in method (1), those analyte measurements that showed large site-to-site variation, on average, failed to exhibit class -dependent differences due to the large site-to-site differences. Such analytes were automatically removed from further analysis. However, those analytes that did show class -dependent differences across the sites are robust biomarkers that were relatively insensitive to sample collection and sample handling variability. KS -distances were computed for all analytes using the class -dependent cdfs aggregated across all sites. Using a KS-distance threshold of 0.4, fifty-nine potential biomarkers for diagnosing malignant ovarian cancer from benign pelvic masses were identified.
[00325] Using the fifty- nine potential biomarkers identified above, a total of 1966 10- analyte classifiers were found with a score of 1.75 or better (>87.5% sensitivity and >87.5% specificity, on average) for diagnosing ovarian cancer from a control group with benign pelvic masses using measurements from both sites. From this set of classifiers, a total of twenty- five biomarkers were found to be present in 5.0% or more of the high scoring classifiers. Table 20 provides a list of these potential biomarkers and Figure 10 is a frequency plot for the identified biomarkers. This completed the biomarker identification using method
(1).
[00326] Method (2) focused on consistency of potential biomarker changes between the control and case groups among the individual sites. The class-dependent cdfs were constructed for all analytes within each site separately and from these cdfs the KS -distances were computed to identify potential biomarkers. Sixty- three analytes were found to have a KS-distance greater than 0.4 in all the sites. Using these Sixty-three analytes to build potential 10-analyte Bayesian classifiers, there were 2031 classifiers that had a score of 1.75 or better. Twenty- four analytes occurred with a frequency greater than 5% among these classifiers and are presented in Table 21 and shown in Figure 11.
[00327] Finally, by combining the criteria for potential biomarker selection described for method (1) and (2) above, a set of potential biomarkers were produced by requiring an analyte to have a KS distance of 0.4 or better in the aggregated set as well as the two site comparisons. Forty-five analytes satisfy these requirements and are referred to as a blended set of potential biomarkers. For a classification score of 1.75 or better, a total of 1563 Bayesian classifiers were built from this set of potential biomarkers and twenty-seven biomarkers were identified from this set of classifiers using a frequency cut-off of 5%. These analytes are displayed in Table 22 and Figure 12 is a frequency plot for the identified biomarkers.
[00328] A final list of biomarkers is obtained by combining the three sets of biomarkers identified above with frequencies greater than 5% in high scoring classifiers, Tables 20-22. From these sets of twenty- five, twenty-four, and twenty-seven biomarkers, forty- two unique biomarkers were identified and are shown in Table 1. Table 15 includes a dissociation constant for the aptamer used to identify the biomarker, the limit of quantification for the marker in the multiplex aptamer assay, and whether the marker was up- regulated or down-regulated in the disease population relative to the control population.
Example 3. Naϊve Bayesian Classification for Ovarian Cancer
[00329] From the list of biomarkers identified as useful for discriminating between benign pelvic masses and ovarian malignancies, a panel of ten biomarkers was selected and a naϊve B ayes classifier was constructed, see Table 18. The class-dependent probability density functions (pdfs), p(x[ I c) and p(x[ I d) , where x[ is the measured RFU value for biomarker i , and c and d refer to the control and disease populations, were modeled as normal distribution functions characterized by a mean μ and variance σ . The parameters for pdfs of the ten biomarkers are listed in Table 18 and an example of the raw data along with the model fit to a normal cdf is shown in Figure 5 for biomarker BAFF Receptor. The underlying assumption appears to fit the data quite well as evidenced by Figure 5. [00330] The naϊve Bayes classification for such a model is given by the following equation, where P(d) is the prevalence of the disease in the population
Figure imgf000076_0001
appropriate to the test and n = 10 here. Each of the terms in the summation is a log-likelihood ratio for an individual marker and the total log-likelihood ratio of a sample x being free from the disease of interest versus having the disease (i.e. in this case, ovarian cancer) is simply the sum of these individual terms plus a term that accounts for the prevalence of the disease.
For simplicity, we assume P(d) = 0.5 so that = 0.
Figure imgf000076_0002
[00331] Given an unknown sample measurement in RFU for each of the ten biomarkers of x = (701, 34158, 182792, 19531, 170310, 896, 3207, 22545, 733, 12535) , the calculation of the classification is detailed in Table 23. The individual components comprising the log likelihood ratio for control versus disease class are tabulated and can be computed from the parameters in Table 18 and the values of x . The sum of the individual log
likelihood ratios is 1.965, or a likelihood of being free from the disease versus having the disease of 7: 1, where likelihood = e ' = 7.14 . Four of the ten biomarker values have likelihoods more consistent with the disease group (log likelihood < 0) while the remaining six biomarkers favor the control group, the largest by a factor of 3.5: 1. Multiplying the likelihoods together gives the same result as that shown above; an aggregate likelihood of 7: 1 that the unknown sample is free from the disease. In fact, this sample came from the control population in the training set.
Example 4. Greedy Algorithm for Selecting Biomarker Panels for Classifiers.
Part 1
[00332] This example describes the selection of biomarkers from Table 1 to form panels that can be used as classifiers in any of the methods described herein. Subsets of the biomarkers in Table 1 were selected to construct classifiers with good performance. This method was also used to determine which potential markers were included as biomarkers in Example 2.
[00333] The measure of classifier performance used here is the sum of the sensitivity and specificity; a performance of 1.0 is the baseline expectation for a random (coin toss) classifier, a classifier worse than random would score between 0.0 and 1.0, a classifier with better than random performance would score between 1.0 and 2.0. A perfect classifier with no errors would have a sensitivity of 1.0 and a specificity of 1.0, therefore a performance of 2.0 (1.0+1.0). One can apply other common measures of performance such as area under the ROC curve, the F-measure, or the product of sensitivity and specificity. Specifically one might want to treat sensitivity and specificity with differing weight, in order to select those classifiers that perform with higher specificity at the expense of some sensitivity, or to select those classifiers which perform with higher sensitivity at the expense of some specificity. Since the method described here only involves a measure of "performance", any weighting scheme which results in a single performance measure can be used. Different applications will have different benefits for true positive and true negative findings, and will have different costs associated with false positive findings from false negative findings. For example, screening and the differential diagnosis of benign pelvic masses will not in general have the same optimal trade-off between specificity and sensitivity. The different demands of the two tests will in general require setting different weighting to positive and negative misclassifications, which will be reflected in the performance measure. Changing the performance measure will in general change the exact subset of markers selected from Table 1 for a given set of data.
[00334] For the Bayesian approach to the discrimination of ovarian cancer samples from control samples described in Example 3, the classifier was completely parameterized by the distributions of biomarkers in the disease and non-disease training samples, and the list of biomarkers was chosen from Table 1; that is to say, the subset of markers chosen for inclusion determined a classifier in a one-to-one manner given a set of training data. [00335] The greedy method employed here was used to search for the optimal subset of markers from Table 1. For small numbers of markers or classifiers with relatively few markers, every possible subset of markers was enumerated and evaluated in terms of the performance of the classifier constructed with that particular set of markers (see Example 4, Part T). (This approach is well known in the field of statistics as "best subset selection"; see, e.g., Hastie et al, supra). However, for the classifiers described herein, the number of combinations of multiple markers can be very large, and it was not feasible to evaluate every possible set of 10 markers, for example, from the list of 42 markers (Table 1) (i.e., 1,471,442,973 combinations). Because of the impracticality of searching through every subset of markers, the single optimal subset may not be found; however, by using this approach, many excellent subsets were found, and, in many cases, any of these subsets may represent an optimal one.
[00336] Instead of evaluating every possible set of markers, a "greedy" forward stepwise approach may be followed (see, e.g., Dabney AR, Storey JD (2007) Optimality Driven Nearest Centroid Classification from Genomic Data. PLoS ONE 2(10): el002. doi: 10.1371/journal.pone.0001002). Using this method, a classifier is started with the best single marker (based on KS -distance for the individual markers) and is grown at each step by trying, in turn, each member of a marker list that is not currently a member of the set of markers in the classifier. The one marker that scores the best in combination with the existing classifier is added to the classifier. This is repeated until no further improvement in performance is achieved. Unfortunately, this approach may miss valuable combinations of markers for which some of the individual markers are not all chosen before the process stops. [00337] The greedy procedure used here was an elaboration of the preceding forward stepwise approach, in that, to broaden the search, rather than keeping just a single candidate classifier (marker subset) at each step, a list of candidate classifiers was kept. The list was seeded with every single marker subset (using every marker in the table on its own). The list was expanded in steps by deriving new classifiers (marker subsets) from the ones currently on the list and adding them to the list. Each marker subset currently on the list was extended by adding any marker from Table 1 not already part of that classifier, and which would not, on its addition to the subset, duplicate an existing subset (these are termed "permissible markers"). Every existing marker subset was extended by every permissible marker from the list. Clearly, such a process would eventually generate every possible subset, and the list would run out of space. Therefore, all the generated classifiers were kept only while the list was less than some predetermined size (often enough to hold all three marker subsets). Once the list reached the predetermined size limit, it became elitist; that is, only those classifiers which showed a certain level of performance were kept on the list, and the others fell off the end of the list and were lost. This was achieved by keeping the list sorted in order of classifier performance; new classifiers which were at least as good as the worst classifier currently on the list were inserted, forcing the expulsion of the current bottom underachiever. One further implementation detail is that the list was completely replaced on each generational step; therefore, every classifier on the list had the same number of markers, and at each step the number of markers per classifier grew by one.
[00338] Since this method produced a list of candidate classifiers using different combinations of markers, one may ask if the classifiers can be combined in order to avoid errors that might be made by the best single classifier, or by minority groups of the best classifiers. Such "ensemble" and "committee of experts" methods are well known in the fields of statistical and machine learning and include, for example, "Averaging", "Voting", "Stacking", "Bagging" and "Boosting" (see, e.g., Hastie et al., supra). These combinations of simple classifiers provide a method for reducing the variance in the classifications due to noise in any particular set of markers by including several different classifiers and therefore information from a larger set of the markers from the biomarker table, effectively averaging between the classifiers. An example of the usefulness of this approach is that it can prevent outliers in a single marker from adversely affecting the classification of a single sample. The requirement to measure a larger number of signals may be impractical in conventional "one marker at a time" antibody assays but has no downside for a fully multiplexed aptamer assay. Techniques such as these benefit from a more extensive table of biomarkers and use the multiple sources of information concerning the disease processes to provide a more robust classification.
Part 2
[00339] The biomarkers selected in Table 1 gave rise to classifiers that perform better than classifiers built with "non-markers" (i.e., proteins having signals that did not meet the criteria for inclusion in Table 1 (as described in Example 2)).
[00340] For classifiers containing only one, two, and three markers, all possible classifiers obtained using the biomarkers in Table 1 were enumerated and examined for the distribution of performance compared to classifiers built from a similar table of randomly selected non-markers signals.
[00341] In Figure 14, the sum of the sensitivity and specificity was used as the measure of performance; a performance of 1.0 is the baseline expectation for a random (coin toss) classifier. The histogram of classifier performance was compared with the histogram of performance from a similar exhaustive enumeration of classifiers built from a "non-marker" table of 42 non-marker analytes; the 42 analytes were randomly chosen from 387 aptamer measurements that did not demonstrate differential signaling between control and disease populations (KS -distance < 0.2).
[00342] Figure 14 shows histograms of the performance of all possible one, two, and three-marker classifiers built from the biomarker parameters in Table 18 for biomarkers that can discriminate between benign pelvic masses and ovarian cancer and compares these classifiers with all possible one, two, and three-marker classifiers built using the 42 "non- marker" aptamer RFU signals. Figure 14A shows the histograms of single marker classifier performance, Figure 14B shows the histogram of two-marker classifier performance, and Figure 14C shows the histogram of three-marker classifier performance. [00343] In Figure 14, the solid lines represent the histograms of the classifier performance of all one, two, and three-marker classifiers using the biomarker data for benign pelvic masses and ovarian cancer in Table 18. The dotted lines are the histograms of the classifier performance of all one, two, and three-marker classifiers using the data for benign pelvic masses and ovarian cancer but using the set of random non-marker signals. [00344] The classifiers built from the markers listed in Table 1 form a distinct histogram, well separated from the classifiers built with signals from the "non-markers" for all one-marker, two-marker, and three-marker comparisons. The performance and AUC score of the classifiers built from the biomarkers in Table 1 also increase at a higher rate as markers are added than do the classifiers built from the non-markers. The separation of performance increases between the marker and non-marker classifiers as the number of markers per classifier increases. All classifiers built using the biomarkers listed in Table 1 perform distinctly better than classifiers built using the "non-markers".
Part 3
[00345] The distributions of classifier performance show that there are many possible multiple-marker classifiers that can be derived from the set of analytes in Table 1. Although some biomarkers are better than others on their own, as evidenced by the distribution of classifier scores and AUCs for single analytes, it was desirable to determine whether such biomarkers are required to construct high performing classifiers. To make this determination, the behavior of classifier performance was examined by leaving out some number of the best biomarkers. Figure 15 compares the performance of classifiers built with the full list of biomarkers in Table 1 with the performance of classifiers built with subsets of biomarkers from Table 1 that excluded top-ranked markers.
[00346] Figure 15 demonstrates that classifiers constructed without the best markers perform well, implying that the performance of the classifiers was not due to some small core group of markers and that the changes in the underlying processes associated with disease are reflected in the activities of many proteins. Many subsets of the biomarkers in Table 1 performed close to optimally, even after removing the top 15 of the 42 markers from Table 1. After dropping the 15 top-ranked markers (ranked by KS-distance) from Table 1, the classifier performance increased with the number of markers selected from the table to reach almost 1.80 (sensitivity + specificity), close to the performance of the optimal classifier score of 1.87 selected from the full list of biomarkers.
[00347] Finally, Figure 16 shows how the ROC performance of typical classifiers constructed from the list of parameters in Table 18 according to Example 3. A five analyte classifier was constructed with TIMP-2, MCP-3, Cadherin-5, SLPI, and C9. Figure 16A shows the performance of the model, assuming independence of these markers, as in Example 3, and Figure 16B shows the empirical ROC curves generated from the study data set used to define the parameters in Table 18. It can be seen that the performance for a given number of selected markers was qualitatively in agreement, and that quantitative agreement was generally quite good, as evidenced by the AUCs, although the model calculation tends to overestimate classifier performance. This is consistent with the notion that the information contributed by any particular biomarker concerning the disease processes is redundant with the information contributed by other biomarkers provided in Table 1 while the model calculation assumes complete independence. Figure 16 thus demonstrates that Table 1 in combination with the methods described in Example 3 enable the construction and evaluation of a great many classifiers useful for the discrimination of ovarian cancer from benign pelvic masses.
Example 5. Aptamer Specificity Demonstration in a Pull-down Assay [00348] The final readout on the multiplex assay is based on the amount of aptamer recovered after the successive capture steps in the assay. The multiplex assay is based on the premise that the amount of aptamer recovered at the end of the assay is proportional to the amount of protein in the original complex mixture (e.g., plasma). In order to demonstrate that this signal is indeed derived from the intended analyte rather than from non-specifically bound proteins in plasma, we developed a gel-based pull-down assay in plasma. This assay can be used to visually demonstrate that a desired protein is in fact pulled out from plasma after equilibration with an aptamer as well as to demonstrate that aptamers bound to their intended protein targets can survive as a complex through the kinetic challenge steps in the assay. In the experiments described in this example, recovery of protein at the end of this pull-down assay requires that the protein remain non-covalently bound to the aptamer for nearly two hours after equilibration. Importantly, in this example we also provide evidence that non-specifically bound proteins dissociate during these steps and do not contribute significantly to the final signal. It should be noted that the pull-down procedure described in this example includes all of the key steps in the multiplex assay described above. [00349] A. Plasma Pull-down Assay
[00350] Plasma samples were prepared by diluting 50 μL EDTA-plasma to 100 μL in
SB 18 with 0.05% Tween-20 (SB 18T) and 2 μM Z-Block. The plasma solution was equilibrated with 10 pmoles of a PBDC-aptamer in a final volume of 150 μL for 2 hours at 37 0C. After equilibration, complexes and unbound aptamer were captured with 133 μL of a 7.5% Streptavidin-agarose bead slurry by incubating with shaking for 5 minutes at RT in a Durapore filter plate. The samples bound to beads were washed with biotin and with buffer under vacuum as described in Example 1. After washing, bound proteins were labeled with 0.5 mM NHS-S-S-biotin, 0.25 mM NHS-Alexa647 in the biotin diluent for 5 minutes with shaking at RT. This staining step allows biotinylation for capture of protein on streptavidin beads as well as highly sensitive staining for detection on a gel. The samples were washed with glycine and with buffer as described in Example 1. Aptamers were released from the beads by photocleavage using a Black Ray light source for 10 minutes with shaking at RT. At this point, the biotinylated proteins were captured on 0.5 mg MyOne Streptavidin beads by shaking for 5 minutes at RT. This step will capture proteins bound to aptamers as well as proteins that may have dissociated from aptamers since the initial equilibration. The beads were washed as described in Example 1. Proteins were eluted from the MyOne Streptavidin beads by incubating with 50 mM DTT in SB17T for 25 minutes at 37 0C with shaking. The eluate was then transferred to MyOne beads coated with a sequence complimentary to the 3' fixed region of the aptamer and incubated for 25 minutes at 37 0C with shaking. This step captures all of the remaining aptamer. The beads were washed 2x with 100 μL SB17T for 1 minute and Ix with 100 μL SB19T for 1 minute. Aptamer was eluted from these final beads by incubating with 45 μL 20 mM NaOH for 2 minutes with shaking to disrupt the hybridized strands. 40 μL of this eluate was neutralized with 10 μL 80 mM HCl containing 0.05% Tween-20. Aliquots representing 5% of the eluate from the first set of beads (representing all plasma proteins bound to the aptamer) and 20% of the eluate from the final set of beads (representing all plasma proteins remaining bound at the end of our clinical assay) were run on a NuPAGE 4-12% Bis-Tris gel (Invitrogen) under reducing and denaturing conditions. Gels were imaged on an Alpha Innotech FluorChem Q scanner in the Cy5 channel to image the proteins.
[00351] B. Pull-down gels for aptamers were selected against LBP (~lxlθ~7 M in plasma, polypeptide MW -60 kDa), C9 (~lxlθ~6 M in plasma, polypeptide MW -60 kDa), and IgM (~9xlO~6 M in plasma, MW -70 kDa and 23 kDa), respectively. (See Figure 13). [00352] For each gel, lane 1 is the eluate from the Streptavidin-agarose beads, lane 2 is the final eluate, and lane 3 is a MW marker lane (major bands are at 110, 50, 30, 15, and 3.5 kDa from top to bottom). It is evident from these gels that there is a small amount nonspecific binding of plasma proteins in the initial equilibration, but only the target remains after performing the capture steps of the assay. It is clear that the single aptamer reagent is sufficient to capture its intended analyte with no up-front depletion or fractionation of the plasma. The amount of remaining aptamer after these steps is then proportional to the amount of the analyte in the initial sample. [00353] The foregoing embodiments and examples are intended only as examples. No particular embodiment, example, or element of a particular embodiment or example is to be construed as a critical, required, or essential element or feature of any of the claims. Further, no element described herein is required for the practice of the appended claims unless expressly described as "essential" or "critical." Various alterations, modifications, substitutions, and other variations can be made to the disclosed embodiments without departing from the scope of the present application, which is defined by the appended claims. The specification, including the figures and examples, is to be regarded in an illustrative manner, rather than a restrictive one, and all such modifications and substitutions are intended to be included within the scope of the application. Accordingly, the scope of the application should be determined by the appended claims and their legal equivalents, rather than by the examples given above. For example, steps recited in any of the method or process claims may be executed in any feasible order and are not limited to an order presented in any of the embodiments, the examples, or the claims. Further, in any of the aforementioned methods, one or more biomarkers of Table 1 can be specifically excluded either as an individual biomarker or as a biomarker from any panel.
Table 1. Biomarkers for Ovarian Cancer
Figure imgf000085_0001
Table 1 cont.
Figure imgf000086_0001
Table 1 cont.
Figure imgf000087_0001
Figure imgf000088_0001
Table 1 cont.
Figure imgf000089_0001
Table 2
Figure imgf000090_0001
Table 2 (cont.)
Figure imgf000091_0001
Table 2 (cont.)
Figure imgf000092_0001
Figure imgf000092_0002
Figure imgf000092_0003
Table 3
100 Panels of 4 Biomarkers for Diagnosing Ovarian Cancer from Benign Pelvic Masses
Figure imgf000093_0001
Table 3 (cont.)
Figure imgf000094_0001
Table 3 (cont.)
Figure imgf000095_0001
Figure imgf000095_0002
Figure imgf000095_0003
Table 4
100 Panels of 5 Biomarkers for Dia nosin Ovarian Cancer from Beni n Pelvic Masses
Figure imgf000096_0001
Table 4 (cont.)
Figure imgf000097_0001
Table 4 (cont.)
Figure imgf000098_0001
Figure imgf000098_0002
Table 5
VO
Figure imgf000099_0001
Table 5 (cont.)
Figure imgf000100_0001
Table 5 (cont.)
O O
Figure imgf000101_0001
Table 5 (cont.)
Figure imgf000102_0001
Table 5 (cont.)
Z/l t"d KJ
H
Figure imgf000103_0001
Table 5 (cont.)
Z/l
W w
H
Figure imgf000104_0001
Table 5 (cont.)
Figure imgf000105_0001
Figure imgf000105_0002
Table 6
100 Panels of 7 Biomarkers for Diagnosing Ovarian Cancer from Benign Pelvic Masses o en
Figure imgf000106_0001
Table 6 (cont.)
H
H
§
Figure imgf000107_0001
Figure imgf000107_0002
Table 6 (cont.)
H
H z/i
W o
Figure imgf000108_0001
Table 6 (cont.)
H
H z/i w i
Figure imgf000109_0001
Table 6 (cont.)
H
H
§
Figure imgf000110_0001
Figure imgf000110_0002
Table 6 (cont.)
H
H
Figure imgf000111_0001
Figure imgf000111_0002
Figure imgf000111_0003
Table 7
100 Panels of 8 Biomarkers for Dia nosin Ovarian Cancer from Beni n Pelvic Masses
Figure imgf000112_0001
Table 7 (cont.)
Figure imgf000113_0001
Table 7 (cont.)
w
Figure imgf000114_0001
Table 7 (cont.)
Figure imgf000115_0001
Table 7 (cont.)
Figure imgf000116_0001
Table 7 (cont.)
Figure imgf000117_0001
Figure imgf000117_0002
Figure imgf000117_0003
Table 8
100 Panels of 9 Biomarkers for Dia nosin Ovarian Cancer from Beni n Pelvic Masses
Figure imgf000118_0001
Table 8 (cont.)
00
Figure imgf000119_0001
Table 8 (cont.)
VO
Figure imgf000120_0001
Table 8 (cont.)
O
Figure imgf000121_0001
Table 8 (cont.)
Figure imgf000122_0001
Table 8 (cont.)
H
Figure imgf000123_0001
H Z/l
H
Figure imgf000123_0002
Figure imgf000123_0003
Table 9
100 Panels of 10 Biomarkers for Diagnosing Ovarian Cancer from Benign Pelvic Masses
Figure imgf000124_0001
Table 9 (cont.)
Figure imgf000125_0001
Table 9 (cont.)
CO
CO
H
CO en
W H
Figure imgf000126_0001
Table 9 (cont.)
CO
CO
H
CO
W H
Figure imgf000127_0001
Table 9 (cont.)
CO
CO
H
CO
W H
Figure imgf000128_0001
Table 9 (cont.)
CO
CO
H
CO
00
W H
Figure imgf000129_0001
Figure imgf000129_0002
Table 10
100 Panels of 11 Biomarkers for Dia nosin Ovarian Cancer from Beni n Pelvic Masses
Figure imgf000130_0001
Table 10 (cont.)
Figure imgf000131_0001
o
H
Figure imgf000131_0002
Table 10 (cont.)
W
W H
Figure imgf000132_0001
Table 10 (cont.)
Figure imgf000133_0001
H
Figure imgf000133_0002
Table 10 (cont.)
W
Figure imgf000134_0001
H
Figure imgf000134_0002
Figure imgf000134_0003
Table 11
100 Panels of 12 Biomarkers for Dia nosin Ovarian Cancer from Beni n Pelvic Masses
Figure imgf000135_0001
Table 11 (cont.)
Figure imgf000136_0001
Table 11 (cont.)
Figure imgf000137_0001
Table 11 (cont.)
Figure imgf000138_0001
Table 11 (cont.)
Figure imgf000139_0001
OO
Figure imgf000139_0002
Figure imgf000139_0003
Table 12
100 Panels of 13 Biomarkers for Diagnosing Ovarian Cancer from Benign Pelvic Masses
Figure imgf000140_0001
Table 12 (cont.)
W
ZΛ d
V i
H
Figure imgf000141_0001
Table 12 (cont.)
W
cβ E
H
Figure imgf000142_0001
Table 12 (cont.)
W
ZΛ d
& S
H
Figure imgf000143_0001
Table 12 (cont.)
W
ZΛ d
∞ E — C
H
Figure imgf000144_0001
Table 12 (cont.)
CO
CO
H
CO
W H
Figure imgf000145_0001
Table 12 (cont.)
CO
CO
H
CO
Ul
W H
Figure imgf000146_0001
Table 12 (cont.)
Figure imgf000147_0001
S
!τ!
H
Figure imgf000147_0002
Figure imgf000147_0003
Table 13
100 Panels of 14 Biomarkers for Dia nosin Ovarian Cancer from Beni n Pelvic Masses
Figure imgf000148_0001
Table 13 (cont.)
00
Figure imgf000149_0001
Table 13 (cont.)
H
H z/i
4-
H
Figure imgf000150_0001
Table 13 (cont.)
H
H o
Figure imgf000151_0001
Table 13 (cont.)
H
H z/i
H
Figure imgf000152_0001
Table 13 (cont.)
κ>
Figure imgf000153_0001
Table 13 (cont.)
Figure imgf000154_0002
Figure imgf000154_0001
Z/l
H
Figure imgf000154_0003
Figure imgf000154_0004
Table 14
100 Panels of 15 Biomarkers for Dia nosin Ovarian Cancer from Beni n Pelvic Masses
Figure imgf000155_0001
Table 14 (cont.)
H
Ui
Figure imgf000156_0001
Table 14 (cont.)
H
H W
Figure imgf000157_0001
Table 14 (cont.)
CO
CO
H
CO
W H
Figure imgf000158_0001
Table 14 (cont.)
W
ZΛ a
00
H
Figure imgf000159_0001
Table 14 (cont.)
CO
CO
H
CO
Figure imgf000160_0001
Table 14 (cont.)
CO
CO
Figure imgf000161_0001
H
CO
9\ O
W H
Figure imgf000161_0002
Figure imgf000161_0003
Table 15
Figure imgf000162_0001
Table 15 cont.
Figure imgf000163_0001
Table 16
Figure imgf000164_0001
Table 17. Sensitivity & Specificity for Exemplary Combinations of BAFF Receptors
Figure imgf000165_0001
Table 18. Parameters derived from training set for naϊve Bayes classifier.
Figure imgf000166_0001
Table 19
Number of Samples by Site
Benign Cancer
Site 1 114 87
Site 2 81 55
TOTAL 195 142
Table 20. Biomarkers of Ovarian Cancer from All Site Analysis (Aggregated Data)
Figure imgf000168_0001
Table 21. Biomarkers of Ovarian Cancer Within Sites
Figure imgf000169_0001
Table 22. Biomarkers of Ovarian Cancer from Blended Data Analysis
Figure imgf000170_0001
W
H
O
W
Figure imgf000171_0001
H

Claims

What is claimed is:
1. A method for diagnosing that an individual does or does not have ovarian cancer, the method comprising: detecting, in a biological sample from an individual, biomarker values that each correspond to one of at least N biomarkers selected from Table 1, wherein said individual is classified as having or not having ovarian cancer based on said biomarker values, and wherein N = 2 - 42.
2. A computer-implemented method for indicating a likelihood of ovarian cancer, the method comprising: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises biomarker values that each correspond to one of at least N biomarkers selected from Table 1 ; performing with the computer a classification of each of said biomarker values; and indicating a likelihood that said individual has ovarian cancer based upon a plurality of classifications, and wherein N = 2 - 42.
3. A computer program product for indicating a likelihood of ovarian cancer, the computer program product comprising: a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers selected from Table 1, wherein said biomarkers were detected in the biological sample; and code that executes a classification method that indicates an ovarian cancer status of the individual as a function of said biomarker values; and wherein N = 2 - 42.
4. The computer program product of claim 3, wherein said classification method uses a probability density function.
5. The computer program product of claim 4, wherein said classification method uses two or more classes.
6. The method of claim 2, wherein indicating the likelihood that the individual has ovarian cancer comprises displaying the likelihood on a computer display.
7. A method for diagnosing that an individual does or does not have ovarian cancer, the method comprising:
detecting, in a biological sample from an individual, biomarker values that each correspond to a panel of biomarkers selected from Table 1, wherein said individual is classified as having or not having ovarian cancer, and wherein the panel of biomarkers has a sensitivity + specificity value of 1.64 or greater.
8. The method of claim 7, wherein the panel has a sensitivity + specificity value of 1.69 or greater.
9. The method of any of claims 1 - 8, wherein N = 2 - 15.
10. The method of any of claims 1 - 8, wherein N = 2 - 10.
11. The method of any of claims 1 - 8, wherein N = 3 - 10.
12. The method of any of claims 1 - 8, wherein N = 4 - 10.
13. The method of any of claims 1 - 8, wherein N = 5 - 10.
14. A method for ovarian cancer in an individual, the method comprising: detecting, in a biological sample from an individual, a biomarker value corresponding to a biomarker selected from Table 1, wherein said individual is classified as having ovarian cancer based on said biomarker value.
15. A computer-implemented method for indicating a likelihood of ovarian cancer, the method comprising: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises a biomarker value corresponding to a biomarker selected from Table 1 ; performing with the computer a classification of said biomarker value; and indicating a likelihood that said individual has ovarian cancer based upon said classification.
16. The method of claim 15, wherein indicating the likelihood that the individual has ovarian cancer comprises displaying the likelihood on a computer display.
17. A computer program product for indicating a likelihood of ovarian cancer, the computer program product comprising: a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker selected from Table 1 ; and code that executes a classification method that indicates an ovarian cancer status of the individual as a function of said biomarker value.
18. The method of any of claims 1 - 17, wherein detecting the biomarker values comprises performing an in vitro assay.
19. The method of claim 18, wherein said in vitro assay comprises at least one capture reagent corresponding to each of said biomarkers, and further comprising selecting said at least one capture reagent from the group consisting of aptamers, antibodies, and a nucleic acid probe.
20. The method of claim 19, wherein said at least one capture reagent is an aptamer.
21. The method of claim 18, wherein the in vitro assay is selected from the group consisting of an immunoassay, an aptamer-based assay, a histological or cytological assay, and an mRNA expression level assay.
22. The method of any of claims 1 - 21, wherein each biomarker value is evaluated based on a predetermined value or a predetermined range of values.
23. The method of any of claims 1 - 22, wherein the biological sample is ovarian tissue and wherein the biomarker values derive from a histological or cytological analysis of said ovarian tissue.
24. The method of any of claims 1 - 22, wherein the biological sample is selected from the group consisting of whole blood, plasma, and serum.
25. The method of any of claims 1 - 22, wherein the biological sample is plasma.
26. The method of any of claims 1 - 25, wherein the individual is a human.
27. The method of any of claims 1 - 26, wherein the individual has a pelvic mass.
PCT/US2009/059706 2008-10-06 2009-10-06 Ovarian cancer biomarkers and uses thereof WO2010042525A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU2009302537A AU2009302537A1 (en) 2008-10-06 2009-10-06 Ovarian cancer biomarkers and uses thereof
EP09819761A EP2350917A4 (en) 2008-10-06 2009-10-06 Ovarian cancer biomarkers and uses thereof
CA2737004A CA2737004A1 (en) 2008-10-06 2009-10-06 Ovarian cancer biomarkers and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10314908P 2008-10-06 2008-10-06
US61/103,149 2008-10-06

Publications (2)

Publication Number Publication Date
WO2010042525A1 WO2010042525A1 (en) 2010-04-15
WO2010042525A9 true WO2010042525A9 (en) 2010-11-18

Family

ID=42076103

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/059706 WO2010042525A1 (en) 2008-10-06 2009-10-06 Ovarian cancer biomarkers and uses thereof

Country Status (4)

Country Link
EP (1) EP2350917A4 (en)
AU (1) AU2009302537A1 (en)
CA (1) CA2737004A1 (en)
WO (1) WO2010042525A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX350533B (en) 2010-08-13 2017-09-08 Somalogic Inc Pancreatic cancer biomarkers and uses thereof.
US9322051B2 (en) 2013-10-07 2016-04-26 General Electric Company Probing of biological samples
GB2543728B (en) 2014-08-12 2019-04-17 Nextgen Jane Inc Medical kit and method for processing a biological sample
EP3442706A4 (en) 2016-04-13 2020-02-19 NextGen Jane, Inc. Sample collection and preservation devices, systems and methods
CN110244733B (en) * 2019-06-20 2020-06-30 西南交通大学 Mobile robot path planning method based on improved ant colony algorithm

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1739185A1 (en) * 2002-12-18 2007-01-03 Ciphergen Biosystems, Inc. Serum biomarkers in lung cancer
CA2535805A1 (en) * 2003-08-15 2005-02-24 University Of Pittsburgh Of The Commnonwealth System Of Higher Education Multifactorial assay for cancer detection
CA2949753A1 (en) * 2005-12-22 2007-07-05 Abbott Molecular Inc. Methods and marker combinations for screening for predisposition to lung cancer
CN101410715A (en) * 2006-01-27 2009-04-15 三路影像公司 Methods for identifying patients with an increased likelihood of having ovarian cancer and compositions therefor

Also Published As

Publication number Publication date
EP2350917A4 (en) 2012-06-06
CA2737004A1 (en) 2010-04-15
AU2009302537A1 (en) 2010-04-15
EP2350917A1 (en) 2011-08-03
WO2010042525A1 (en) 2010-04-15

Similar Documents

Publication Publication Date Title
US11041866B2 (en) Pancreatic cancer biomarkers and uses thereof
AU2015249113B2 (en) Lung cancer biomarkers and uses thereof
EP3029153B1 (en) Mesothelioma biomarkers and uses thereof
US20100086948A1 (en) Ovarian Cancer Biomarkers and Uses Thereof
US20120101002A1 (en) Lung Cancer Biomarkers and Uses Thereof
EP2771692B1 (en) Lung cancer biomarkers and uses thereof
US20120143805A1 (en) Cancer Biomarkers and Uses Thereof
WO2011031344A1 (en) Cancer biomarkers and uses thereof
US20120165217A1 (en) Cancer Biomarkers and Uses Thereof
US20150160225A1 (en) Renal Cell Carcinoma Biomarkers and Uses Thereof
WO2010042525A9 (en) Ovarian cancer biomarkers and uses thereof
US20220065872A1 (en) Lung Cancer Biomarkers and Uses Thereof

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: 09819761

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2009302537

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2737004

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2009302537

Country of ref document: AU

Date of ref document: 20091006

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009819761

Country of ref document: EP